Prokaryotic expression of human complement regulator ...1147592/FULLTEXT01.pdf · Master Thesis...

24
INOM EXAMENSARBETE BIOTEKNIK, AVANCERAD NIVÅ, 30 HP , STOCKHOLM SVERIGE 2017 Prokaryotic expression of human complement regulator factor H domains and their interaction with Pneumococcal surface protein PspC. NILS LINDSTRÖM KTH SKOLAN FÖR BIOTEKNOLOGI

Transcript of Prokaryotic expression of human complement regulator ...1147592/FULLTEXT01.pdf · Master Thesis...

  • INOM EXAMENSARBETE BIOTEKNIK,AVANCERAD NIVÅ, 30 HP

    , STOCKHOLM SVERIGE 2017

    Prokaryotic expression of human complement regulator factor H domains and their interaction with Pneumococcal surface protein PspC.

    NILS LINDSTRÖM

    KTHSKOLAN FÖR BIOTEKNOLOGI

  • Royal Institute of Technology

    Prokaryotic expression of human complement regulator factor H domains and their interaction with Pneumococcal surface protein PspC. Master Thesis Project Report

    Nils Lindström 2017-09-21

    Abstract

    Virulent strains of S. pneumoniae are known to evade the alternative pathway of

    complement immunity by means of a surface bound protein called PspC that recruits the

    human complement regulator Factor H. Factor H is a self surface marker that inhibits the

    activity of the alternative pathway of complement immunity and is comprised of 20

    Complement Control Protein (CCP) domains in a “bead on a string” fashion. It has been

    concluded that PspC can use two different mechanisms of binding Factor H. The PspC

    allele of the TIGR4 strain of S. pneumoniae has been shown to have affinity for the 9th

    CCP of Factor H through a “lock and key” mechanism mediated by a critical Tyr90

    residue. However, PspC from the D39 strain of S. pneumoniae does not posess Tyr90 and

    it hasn’t been conclusively shown which CCP it binds to. In an effort to elucidate this

    mechanism, individual CCP domains were expressed as fusion proteins with Maltose

    Binding Protein in an E. coli based expression system. The fusion proteins were used in

    experiments with recombinant PspC cloned from the BHN_418 strain of S. pneumoniae

    which is homologous to that of D39 PspC. Affinity interactions were investigated with a

    pulldown assay, copurification, microscale thermophoresis and ligand tracer assays. The

    results are inconclusive. The recombinant PspC is shown to bind full length Factor H but

    not any of the individual CCP-MBP fusion proteins, most likely due to the CCPs failing

    to achieve proper tertiary structure during expression.

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    1

    Introduction Community acquired bacterial pneumonia is

    still one of the most deadly infectious diseases

    in the world and in almost 50% of cases

    Streptococcus pneumoniae can be isolated as

    the causative pathogen.- 1. S. pneumoniae is a

    gram positive, facultative anaerobic and

    commensal bacterium that often colonizes the

    mucosal surfaces of the respiratory tract and

    nasopharyngeal space.2 It is responsible for 30-

    60% of acute otitis media worldwide 3 as well

    as meningitis, sinusitis, sepsis and other acute

    infections in immunocompromised individuals

    like HIV-patients 4. Estimates from 2009

    indicate that circa 800 000 children under the

    age of five die each year from S. pneumoniae

    infections 5. An important virulence factor of S.

    pneumoniae is Pneumococcal Surface Protein C

    (PspC), also known as SpsA, Hic or CbpA.

    Apart from acting as an adhesion molecule It is

    known to recruit the human complement

    regulator factor H, (fH) on the bacterial surface

    in order to evade complement deposition. fH is

    a 155kDa protein comprised of 20 domains

    known varyingly as complement control

    proteins (CCPs), Short Consensus Repeats or

    sushi domains. It has an unusual structure where

    the 20 CCP domains are connected with short

    peptide linkers in a “bead on a string” fashion,

    where the different domains of the protein have

    different functions in vivo.6 S. pneumoniae

    utilizes the affinity for fH to mediate adhesion

    to host epithelial cells and to circumvent the

    complement system of immunity. 7 The latter

    property is enabled because of the regulatory

    functionality of fH on the alternative pathway

    of the complement immune system. The

    alternative pathway of complement immunity is

    in a state of constant equilibrium between a

    positive feedback loop that generates

    opsonizing C3b and the repressive effect that fH

    has on this loop. Complement protein C3 is

    spontaneously cleaved in small amounts in the

    bloodstream and when this occurs in close

    proximity to a cell surface the resulting

    fragment C3b is covalently bound to it by its

    revealed nucleophilic thioester domain. Surface

    bound C3b recruits factor B to produce the C3

    convertase or C3bBb complex which acts as a

    potent catalyst for further cleavage of C3. This

    action constitutes a positive feedback loop

    which will eventually result in all nearby

    surfaces being opsonized by C3b which leads to

    the formation of the Membrane attack complex,

    cell lysis and phagocytosis. To prevent this to

    occur on self surfaces fH acts as catalyst for

    removing the Factor Bb subunit of the C3-

    convertase and form a complex called C3b-

    Factor H which possesses affinity for Factor I

    which further cleaves the complex to produce

    iC3b and C3c which can take no further part in

    the amplification loop.8,9

    Figure 1 - The alternative pathway of immune system activation with available structure data. CVF-Factor B is homologous

    to C3b-Factor B. Graphics by Serruto et.al. 2010 Nature Reviews .8

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    2

    Thus the pathway does not rely on anti-bodies

    having specific affinity towards a pathogen to

    activate. Instead, it is constantly on the verge of

    activating everywhere and is relying on self-

    surface markers like fH to repress its activity.

    This key regulatory role of fH in protecting self-

    surfaces from attack by the innate immune

    system makes it an attractive target for

    microbial hijack. A strategy used by several

    pathogenic bacteria such as Staphylococcus

    aureus, Yersinia enterocolitica, Borrelia

    burgdorferi and Streptococcus pneumoniae.8 In

    2002 Ianelli et. al. identified 11 allelic variants

    of PspC. They all sharing a similar domain

    organization of an N-terminal fH binding

    region, a helical region, a proline rich region

    and a C-terminal anchor sequence. Ianelli et. al.

    proposed to classify these variants into two

    families A and B, distinct in the way they attach

    to the bacterial outer membrane. The suggestion

    was that Family A is defined by being anchored

    with a C-terminal choline binding domain

    whereas PspC in Family B is defined by being

    anchored through a sortase dependent LPxTG

    motif. 10 Since then, further study of PspC has

    revealed that alleles of PspC can differ from

    each other not only in the mechanism of

    anchoring to the bacterial membrane but also in

    the binding mechanism towards fH. In 2015 two

    important articles were published illustrating

    this fact. Achila et. al. were able to show that

    PspC from the S. pneumoniae strain TIGR4

    binds to the 9th CCP domain of fH through a

    hydrophobic “lock and key” mechanism

    mediated by the Tyr90 residue. 11 The same year,

    Herbert et. al. showed that PspC from strain

    D39 which lacks the Tyr90 residue binds the

    same region of CCPs 8-10 as TIGR4 PspC,

    despite sharing only 50% sequence similarity

    and 6 out of 10 critical interface residues in the

    binding site.12 This is a strong indication that

    D39 PspC utilizes a different binding

    mechanism than TIGR4 PspC, a mechanism

    which is to current date unknown. Since the

    both types of PspC, with and without Tyr90,

    contribute to virulence it is of great interest to

    elucidate the binding mechanisms of both.

    Some strains of S. pneumoniae express more

    than one allele of PspC where one such strain is

    S. pneuminae_BHN418. Unpublished results by

    Anuj Pathak of Birgitta Henrique Normark’s

    Lab show that this strain expresses two PspC

    alleles that are very similar to those expressed

    by D39 and TIGR4 respectively. This property

    makes it a good system for studying the

    different functions of the two alleles and has

    been the strain used for cloning PspC in this

    project.

    Figure 2 - PspC and its

    interaction with Factor H.

    A: General domain

    organization of PspC and

    protein sequence alignment

    of [Tigr4]PspC and

    [BHN418]PspC2 which

    possess Tyr90 as contrasted

    with [D39]PspC and

    [BHN418]PspC1 which

    lacks Tyr90. Sequence

    alignments were performed

    by the PROMALS3D web

    server. 18 B: Factor H

    structure model prediction

    by the PHYRE2 server and

    CCP domain organization.

    C: N-terminal domain of

    [TIGR4]PspC and its

    interaction with fH CCP9

    through a hydrophobic

    “lock and key” mechanism

    mediated by Tyr90.

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    3

    Anuj Pathak’s unpublished results reveal a

    linear binding motif in [BHN418]PspC1

    composed of residues K131-K139 and E149-

    A154 in the N-terminal domain of

    [BHN418]PspC1. Thus proving that the binding

    site for fH is located on the N-terminal domain.

    This master thesis report presents a project on

    PspC1 isolated from S.pneumoniae strain

    BHN418. From this point in this document

    PspC1 refers to the PspC-allele of BHN418 that

    is similar to [D39]PspC while PspC2 will refer

    to the PspC-allele similar to [TIGR4]PspC in

    their N terminal fH binding domain sequence.

    The purpose of the project has been to elucidate

    the mechanism of binding that provides PspC1

    with affinity for fH. The method to accomplish

    this has been to express recombinant PspC1 in

    E. coli and to express individual fH CCP

    domains as fusion proteins with Maltose

    Binding Protein (MBP) in a prokaryote, E. coli

    based expression system. Forcing expression of

    eukaryote proteins in prokaryote systems is

    often problematic, however this was the

    approach used in the 2015 Achila. et. al study

    which reports success in this endeavor.13

    The long term potential of this research lies in

    the possibility to increase understanding of

    potential future protein vaccines against

    S.pneumoniae. Purified PspC along with other

    surface proteins like PspA has been shown to

    have potential as antigens in protein vaccines

    against pneumococcal infections. Even though

    efforts to develop vaccines against

    S.pneumoniae have been largely successful,

    more than 90 distinct serotypes of S.

    pneumoniae have been identified and current

    vaccines are not effective against all of them.

    Further efforts to understand and characterize

    the bacterium is important in this effort.14

    Materials and methods

    Production of Recombinant Proteins All cloning was performed using the Sequence

    and Ligation Independent Cloning (SLIC)

    technique.15 In this technique genes are PCR-

    amplified with primers designed to have

    overhangs of around 20 bp that are homologous

    to the intended vector sequence. All primer used

    are indexed in Supplementary 1: Primers. The

    vector construct was amplified as a linear

    stretch of DNA. PCR-products were purified on

    agarose gel with QIAquick PCR-cleanup kit by

    Qiagen. Inserts and vectors were digested with

    T4 DNA polymerase for 30 min to form 5’

    sticky ends after which the digest was halted by

    addition of a single dNTP. The vector and the

    construct were incubated together in equimolar

    amounts in 37°C for one hour before

    transformation of the non-ligated intermediary

    construct. Ligation and gap-filling occurs in in-

    vivo and a completed vector was extracted with

    QIAprep Spin Miniprep kit according to

    protocol.

    Transformation of the non-ligated intermediary

    constructs was done with Mix’n’Go Competent

    E.coli by Zymore Research, amplification of the

    vector construct was done with XL-1 Blue

    Figure 3 - SLIC-Procedure. Graphic adapted from 2007

    M. Li and S. Elledge, Nature Methods.15

    competent E.coli by Agilent. All plasmids were

    controlled by sequencing before transformation

    of the amplification and expression systems.

    Protein production was performed in 1 liter

    cultures of Terrific Broth supplemented with

    100µg/ml ampicillin. Expression was induced

    with 0.4mM Isopropyl β-D-1-

    thiogalactopyranoside at OD600 = 0.4. Cell lysis

    was performed by liquid homogenization. All

    purified proteins were flash frozen in 10%

    Glycerol and 20mM Hepes Buffer at pH 7.5 for

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    4

    storage and were thawed directly before use in

    assays. Standard chemical were obtained from

    Sigma Aldrich, AppliChem, VWR and

    MERCK.

    Recombinant PspC1

    PspC1 was cloned from genomic DNA from S.

    pneumoniae strain BHN_418. The gene was

    cloned into a pET21d vector providing

    ampicillin resistance, Lac-operon, a His6

    purification tag and a TEV-cleaving site at the

    N-terminal domain. The full length

    PspC1_CBD construct was used for further

    cloning to produce truncated fragments N1, N2

    and N3. A chart detailing the recombinant

    proteins can be seen in Figure 4.

    Cell pellets were re-suspended in Lysis Buffer

    B before cell disruption, buffer compositions

    are detailed in Supplementary 2: Buffer

    compositions. His-tagged recombinant PspC1

    was purified on a 5 ml HisTrap FF column by

    GE Healthcare, and eluted with 500mM

    Imidazole according to protocol. Further

    purification was performed using a 5 ml HiTrap

    SP FF Ion Exchange Chromatography column

    by GE Healthcare for proteins N1 and N2 due

    to the theoretical pI of these constructs being 9.2

    and 9.26 respectively. The longer N3 variant

    was purified using size exclusion

    chromatography on a HiLoad 16/600 Superdex

    75 pg column by GE Healthcare since it had a

    theoretical pI of 7.8.

    fH CCP-MBP fusion proteins A vector construct of human fH synthesized by

    Eurofins Genomics with optimized codons for

    E.coli expression was used as the template for

    cloning of the recombinant MBP-CCP fusion

    constructs into a pET21d vector carrying an

    ampicillin resistance module, Lac-operon,

    Twin-Strep-tag purification tags by iba,

    Maltose Binding Protein (MBP) and a TEV site

    at the N-terminal domain. Four different CCP-

    constructs were designed based on the results

    from Achila et.al. 11 Each construct was made in

    two versions, one with and one without a MalE

    signal peptide for periplasmic expression at the

    N-terminal domain. Cloning of additional CCP-

    constructs comprising CCPS 1:7, 6:8, 8:10,

    10:12, 13:15, 16:18 and 19:20 were also

    prepared but were never transformed into an

    expression system or used in any assays.

    Small Scale Expression Test

    Protein expression was tested in 2ml Terrific

    Broth with 100µg/ml Ampicillin. Cultures were

    incubated for 4 hours before induction with 0.4

    mM IPTG at OD600= 0.7 and incubation over

    night at 22°C. The entire culture volume was

    pelleted and lysed with detergent based Lysis

    Buffer B. The Protein was purified with

    Figure 4 - Schematic view of produced PspC1 constructs. N signifies that the construct comprises the N-terminal domain

    while CBD signifies Choline Binding Domain.

    Figure 5 - Schematic overview detailing the CCP constructs. Eight constructs were produced, where four contained the

    MalE signal peptide for periplasmic expression, hence the parenthesis around MalE in the schematic.

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    5

    MagStrep “type 3” XT beads by iba according

    to protocol. SDS samples of the eluates were

    run in parallel to compare the expression results

    as seen in Figure 8. Expression was tested in

    two strains of E.coli. SHuffle T7 Competent

    E.coli (C3026) and SHuffle T7 Express

    competent E.coli (C3029) by NEB. Both strains

    constitutively express disulfide bond

    reshuffling protein DsbC. At the point of testing

    expression the CCP9 construct were not

    completed since a completed CCP8:9 construct

    was needed as a template. Hence expression

    was tested for twelve cultures in total. Once the

    expression strategy was chosen the CCP9

    construct was cloned and expressed using the

    same method. Large scale purification of the

    fusion proteins was performed by resuspending

    the cell pellet in Lysis Buffer A before cell

    disruption followed by affinity chromatography

    using a 5 ml Gravity flow Strep-Tactin XT

    Superflow column by iba according to protocol.

    Final purification was performed by Size

    Exclusion Chromatography using a HiLoad

    16/600 Superdex 75 pg column by GE

    Healthcare.

    Experiments

    Pulldown assay

    A preliminary assay of binding between CCP-

    MBP fusion proteins and PspC1_N2 was

    performed with a pulldown assay. Cell lysate

    containing the recombinant MBP-CCP

    constructs was incubated with MagStrep “type

    3” XT beads by iba and washed according to

    protocol but omitting the elution step resulting

    in magnetic beads coated with CCP-MBP

    fusion proteins. The bead slurry was divided

    into two aliquots. One aliquot was eluted with

    2.5mM Desthiobiotin in TBS and used as a

    negative control while the other aliquot was

    incubated with 1mg/ml His6-PspC1_N2 for one

    hour before washing in TBS and elution. 1µl of

    samples, negative control and positive control

    (10ng/ml pure PspC1_N2) was blotted on

    nitrocellulose membrane and allowed to air dry

    for one hour before blocking over night with 1%

    BSA. The membrane was washed with PBST

    three times for 10 minutes each before

    incubation with 1:4000 diluted HRP-coupled

    Anti-6X His Tag antibody by abcam. An

    identical blot was made for incubating with

    1:4000 diluted HRP-coupled Anti-Strep-tag II

    antibody by abcam. The membranes were

    developed by coating the membranes in 750 µl

    Pierce ECL Western Blotting Substrate by

    Thermo Fisher according to protocol. Signal

    was recorded with a BioRad ChemiDoc system.

    Size Exclusion Chromatography Co-

    purification

    Recombinant MBP-CCP8 was cleaved by

    overnight incubation in 4°C using TEV-

    protease in Storage Buffer with addition of 1

    mM EDTA, 3 mM Glutathione and 0.3 mM

    Gluthathione disulfide to activate the TEV-

    protease. The cleaved construct was purified

    using on a Superdex 75 HR column by GE

    Healthcare, and the fractions containing CCP8

    were pooled and concentrated. CCP8 was

    incubated with PspC1 constructs in equimolar

    amounts and run over Superdex 75 HR column.

    The resulting chromatogram was compared

    with runs of PspC1 and CCP8 run separately at

    the same respective concentrations. The assay

    was performed with PspC1_N1 and PspC1_N2

    constructs.

    Figure 6 - Pulldown Assay. 1. Purification of CCP-MBP fusion proteins by affinity chromatography on MagStrep “type 3” XT

    beads. 2. Addition of purified His6PspC1. 3. Elution from beads by addition of desthiobiotin. 4. Blotting on membrane and

    signal detection by HRP-coupled antibodies.

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    6

    Figure 7 – Co-purification with Immobilized Metal Ion Chromatography and StrepTactin Affinity Chromatography. 1.

    Recombinant PspC1_N3 was loaded onto the IMAC column and washed in 3x TBS. 2. Pure CCP-MBP fusion protein was

    loaded onto the same IMAC column and washed in 3x TBS. The column was eluted with 500 mM imidazole. 3. The IMAC eluate

    was loaded onto a Gravity flow Strep-Tactin XT Superflow column and washed in 3x TBS. 4. Strep-Tactin column was eluted

    with addition of 2.5 mM desthiobiotin. Samples for analysis by SDS-PAGE were taken throughout experiment.

    IMAC/StrepTag Co-purification

    20µmol of His6-PspC1_N3 was loaded onto a

    5ml Column Volume HisTrap FF by GE

    Healthcare and washed with five column

    volumes of TBS containing 25mM Imidazole

    after which 15µmol of MBP-CCP8 in 800µl

    HBS was loaded unto the same column and

    washed with 5 CV. Elution was performed

    using TBS containing 500mM Imidazole and

    the eluate was collected. All fractions were

    loaded onto a 5 ml Gravity flow Strep-Tactin

    XT Superflow column by iba which was

    washed in TBS 5x CV before eluting with

    2.5mM Desthiobiotin in TBS. Flowthrough and

    eluted fractions from the Strep-Tactin column

    were pooled and concentrated 10 times before

    analysis by SDS-PAGE. A schematic detailing

    the procedure can be seen in Figure 7.

    Ligand Tracer

    The Ligand Tracer by Ridgeview Instruments is

    a platform for measuring affinity interactions

    between a receptor and a ligand. Measurements

    take place on disposable petri-dishes onto which

    the receptor is immobilized along with a

    negative control and a blank at discrete

    positions. The dish is placed at an angle in the

    machine and the ligand is titrated into solution

    at the bottom of the tilted dish. Measurements

    take place at the top of the dish after it has been

    rotated so the immobilized receptor passes

    through the ligand solution. The ligand needs

    fluorescent activity to be measured. In the ideal

    case the fluorescent intensity measured at the

    receptor position increases for every rotation

    through the ligand-solution, and for every

    increase in ligand concentration until the

    receptor is saturated. At this point the ligand

    solution is replaced with pure buffer and as the

    measurements continue a dissociation curve can

    be observed. 16

    Samples of MBP-CCP8, MBP-CCP9 and full

    length fH were spotted on petri dishes coated

    with polydopamine and incubated overnight in

    4°C. Care was taken not to let the spots flow

    together. The spots were washed three times in

    PBS and blocked with 1% BSA in PBS or

    Pierce protein free blocking buffer by Thermo

    Fisher, the dish was placed in the ligand tracer

    instrument and filled with 3 ml PBS. Sequential

    additions of fluorescently labeled PspC1_N3

    was added with increasing concentrations at

    approximately 100 minute intervals after which

    the PBS solution containing PspC1_N3 was

    replaced with pure PBS to measure any

    dissociation curve. Binding activity was

    monitored continuously throughout the

    experiment at 488nm wavelength.

    Microscale Thermophoresis

    Microscale thermophoresis is a technique that

    utilizes the thermophoresis phenomenon to

    study particle interactions. Thermophoresis is

    the phenomenon where particles migrate

    directionally in small temperature gradients.

    This migration depends on the size, charge and

    hydration shell of the particle and is useful for

    studying proteins since the thermophoretic

    properties of a protein change as a function of

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    7

    conformational changes and affinity

    interactions. The technique is made possible by

    recent advances in optics and laser-physics

    which allow very precise temperature gradients

    to be induced by means of an infrared laser. The

    thermophoretic movement is observed as

    fluctuations of a fluorescent signal recorded at

    the same point where the temperature gradient

    is induced. This means that the particles

    included in the experiment must have

    fluorescent activity for a signal to be recorded.

    In the case of making affinity interaction

    measurements only one of the ligand or the

    receptor needs to possess this activity. 17

    Four measurements were taken; PspC1_N3

    against fH twice, MBP-CCP8 against

    PspC1_N3 and MBP-CCP9 against PspC1_N3.

    The PspC1_N3 was titrated as a twelve point

    1:2 dilution series starting at 125 nM and a

    separate measurement was made with a three

    point 1:2 dilution series in triplicate. Both

    measurements were performed with a constant

    concentration of 500 nM fH. MBP-CCP8 and

    MBP-CCP9 was measured by titrating MBP-

    CCPs in a twelve point 1:2 dilution series

    starting at 500 nM, also with a constant

    concentration of 500 nM fH. The experimental

    set-up used the fluorescent activity of the CCPs

    tryptophan residue.

    Results

    Expression and purification

    MBP-CCP small scale expression test

    Based on the expression test cytoplasmic

    expression was chosen since it generally

    produces better yields when compared to

    periplasmic expression. Also, periplasmic

    expression failed for CCP8 and CCP8:9 in the

    C3026 strain. Theoretical molecular weights

    correspond to results as analyzed by SDS-

    PAGE. Degradation of fusion protein constructs

    is evident by the presence of multiple bands.

    MBP-CCP8

    A population of proteins of similar molecular

    weight are visible in both StrepTag AC and SEC

    fractions. This indicates cleavage of the fusion

    protein in vivo since the theoretical weight of

    the CCP8 domain is 6.8kDa. The second lane in

    the SEC-fractions after StrepTag Affinity

    Chromatography gel is a sample from the void

    volume. These are likely aggregated MBP-

    CCP8 that have not been cleaved. The void

    volume samples were also used in assays since

    these were the only fractions containing pure,

    un-cleaved MBP-CCP8.

    Figure 8 - Expression test of

    CCP constructs. The lanes

    marked with yellow stars

    correspond to the expression

    strategy that was chosen.

    SHuffle T7 Competent E.coli

    (C3026) and SHuffle T7

    Express competent E.coli

    (C3029) by NEB were tested.

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    8

    Figure 9 - MBP-CCP8 purification results.

    Figure 10 - MBP-CCP9 purification results.

    Figure 11 –MBP-CCP8:9 purification results.

    MBP-CCP9

    The same population can be seen here as were

    seen for CCP8. The first three lanes after the

    ladder on the StrepTag AC elution fractions gel

    are the complete lysate, the cleared lysate and

    the flowthrough on the column. The first lane

    after the ladder on the SEC fractions after Strep-

    Tactin AC gel is the void volume. The same

    tendency for aggregation can be seen for MBP-

    CCP9 as for MBP-CCP8 although the band is

    not as strong, indicating that the tendency for

    aggregation is not as great.

    MBP-CCP8:9

    The same population is visible here as for MBP-

    CCP8 and 9. The first two lanes after the ladder

    on the StrepTag AC fractions gel are the lysate

    and the column flow through.

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    9

    PspC1

    Production of the truncated recombinant PspC1

    was successful. The theoretical weight of

    PspC1_N2 is 14.9 kDa and 42.1 kDa for

    PspC1_N3. Analysis of produced proteins

    match this within reason. A band corresponding

    in weight to a dimer is present in purifications

    of PspC1_N2. Purification of PspC1_N1

    succeeded but no gels exist available for

    presentation.

    Figure 12 - PspC1 purification results. PspC1_N2: Both lanes after the ladder are samples of the pooled protein containing

    fractions of the eluate from the respective columns. PspC1_N3: Lane 2 in the SEC-fractions after IMAC is the void volume.

    Figure 13 - Pulldown

    Assay Results

    .

    Pulldown Assay The results indicate binding between PspC1 and

    CCP8 and not CCP9 as previously reported by

    Achila et. al. 11 Binding to CCP10 could not be

    ruled out since the Streptavidin positive control

    gave a weak signal. The PspC1_N2 positive

    control did not give any signal when developed

    with Anti-His6 Ab, this might be due to the

    concentration being very low. No signal was

    observed from the CCP8:9 construct.

    SEC Co-purification No significant decrease in the area of the peak

    corresponding to CCP8 can be observed

    coupled to an increase in area of any of the

    PspC1 peaks for neither PspC1_N1, N2 nor N3.

    No binding can thus be observed by analyzing

    the overlayed chromatograms.

    Figure 14 - SEC-Binding assay results.

    PspC1_N2

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    10

    IMAC and StrepTag AC Copurification

    Figure 15 –SDS-Page analysis of the copurification results.

    Ligand Tracer

    Figure 16 - Ligand Tracer results. On the left is the result from blocking with BSA and on the right is the result of protein free

    block. Both plots show fluorescent signal with background subtracted.

    Results indicate that PspC1_N3 has affinity for the full length Factor H. No strong conclusions can be

    made due to poor signal to noise ratio, especially as PspC1_N3 concentration approaches 100nM. Before

    this point, fH signal is consistently higher than both MBP-CCP8 and MBP-CCP9. Indications for

    binding to CCP8 are also present since MBP-CCP8 is higher than MBP-CCP9. Protein free block

    resulted in lower background and thus better signal to noise ratio, but still not good enough to make

    draw strong conlusions.

    PspC1_N3 does not show strong affinity for

    MBP-CCP8 as is evident by the strong

    signal from MBP-CCP8 in the flow through

    of the IMAC as well as there being barely

    any signal of any of the proteins in the

    StrepTag AC eluate. Note that the eluate

    and the flow through of the StrepTactin

    column is concentrated 10x before loading

    on the gel.

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    11

    Microscale Thermophoresis

    Figure 17 –MST-results. Cap signifies the capillary number. Samples prepared as 12 point 1:2 dilution series and were loaded

    from high to low concentration. Measurements with the MBP-CCPs start at 500nM PspC1_N3 and a constant concentration

    of 250nM MBP-CCP. The measurements with fH are made with a higher constant concentration of 500nM fH. Upper left

    illustrates PspC1_N3 and MBP-CCP8. MBP-CCP8. No affinity interactions can be observed, capillary 1 can be regarded as

    an outlier. Upper right illustrates PspC1_N3 and full length fH. Clear signs of affinity interaction can be observed since

    different concentration of PspC1_N3 give different signals. Lower left illustrates PspC1_N3 and MBP-CCP9. No affinity

    interaction can be observed. Lower right illustrates the signal from PspC1_N3 by itself, capillary 1 can be considered an

    outlier.

    Affinity measurement calculations were

    performed for the PspC1_N3 interaction with

    full length fH resulting in a Kd of 248.6+70.4

    nM, far from the single-digit nanomolar range

    reported by Achila.et.al. 11

    Discussion Expression of recombinant CCP constructs in

    E.coli was a partial success. Peptides of the

    correct weight were produced but it is unclear

    whether the correct tertiary structure was

    achieved by the prokaryote expression system.

    Extensive degradation of the recombinant

    MBP-CCP fusion construct was observed and

    seems to have occurred in vivo. Analysis by

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    12

    SDS-PAGE as seen in Figure 9, Figure 10 and

    Figure 11 are consistent with the degradation

    being the result of hydrolysis of the TEV-site

    since the CCPs have a theoretical molecular

    weight of around 7 kDa. Redesigning the

    constructs with a different protease site might

    rectify this issue. Another good reason for using

    a different protease site is that TEV-protease

    requires a reducing environment to operate and

    reducing environments could cause the

    recombinant CCP to denature due to reducing

    the cysteine bonds that stabilize its tertiary

    structure.

    The initial pulldown assay gave strong

    indications of affinity interactions between

    PspC1 and MBP-CCP8 but this result was not

    reproducible by independent methods leading to

    the conclusion that the pulldown assay result

    was an artifact, despite being internally

    reproducible. This is not entirely surprising

    since neither the positive control of only PspC1

    nor the MBP-CCP8:9 gave any signal. While

    performing the experiments this was speculated

    to be the result of steric hindrance of the binding

    site, but that hypothesis does not explain the

    lack of signal from the positive control.

    However the positive control was heavily

    diluted to a concentration of around 10ng/ml in

    order to avoid its signal occluding the signal

    from the adjacent spots. This concentration

    might have been so low as to wash of and not

    produce any signal whatsoever.

    None of the PspC1 constructs showed affinity

    towards CCP8 when co-purifying over SEC

    with PspC1 or CCP runs at equimolar

    concentration as reference, see Figure 16. The

    first two experiments with PspC1_N1 and N2

    were conducted with CCP8 as resulting from

    treating MBP-CCP8 with TEV-protease. Strong

    suspicions regarding the fold of the CCP8

    domains were raised at this time and therefore a

    third experiment was conducted with the

    PSPC1_N3 construct and uncleaved MBP-

    CCP8 since PspC1_N3 has a higher molecular

    weight of 42.1 kDa as compared to PspC1_N2

    at 14.9 kDa and PspC1_N1 at 12.8 kDa, the

    MBP-CCP8 construct would not have to be

    cleaved to resolve an affinity complex on the

    column. If binding could be demonstrated under

    these conditions one could draw the conclusion

    that the TEV-cleavage denatured the CCP8

    domain. No affinity interactions could be

    observed however, leading to two possible

    explanations. Either the CCP8 domain was

    never properly expressed or PspC1 has affinity

    for some other CCP-domain. In either case one

    must conclude that the pulldown assay results

    amount to an artifact. In retrospect the

    constructs should have been redesigned after

    performing the binding assays on the SEC-

    column and not getting any significant

    indications of binding. At the time the results

    from the pulldown assay was considered so

    convincing that work proceeded with the

    original constructs

    It seems likely that the lack of observable

    affinity between the recombinant PspC1 and the

    MBP-CCPs is due to the CCPs failing to

    achieve proper tertiary structure since the

    PspC1 is shown to have affinity for the full

    length fH but not the CCPs. It seems likely that

    this is due to a failure of the necessary disulfide

    bonds to form. It is tempting to speculate that

    this could also be an explanation for the

    tendency for the CCPs to aggregate in the void

    volume when doing SEC (see Figure 9) since

    the molten-globule that likely results would

    have exposed protein backbone and thus affinity

    for copies of itself.

    The production of PspC1 can be deemed a

    success since binding was observed against full

    length factor H during MST. PspC1 is a

    prokaryote protein without complex tertiary

    structure or cysteine bonds so this should also

    not be problematic in theory. The truncation of

    PspC1 could theoretically cause problems with

    the tertiary structure but this was not observed.

    The question of whether the shorter constructs

    of PspC1, N1 and N2 are valid is still

    unanswered since expression of these proteins

    failed before the deadline for doing the MST-

    measurements. Assessing whether these

    constructs have affinity for full length fH would

    be useful since shorter constructs are preferable

    when producing co-crystals. An attempt to do

    this with a pulldown assay was made by

    utilizing magnetically coupled NHS-beads to

    bind full length factor H. The assay was set up

    according to protocol but the NHS-coupling

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    13

    failed for unknown reasons. It was decided best

    to abort the experiment and not spend time

    optimizing since the protocol was rather time-

    consuming.

    The Ligand Tracer assays gave indications for

    PspC1_N3 binding to full length fH. There was

    also some indications for binding to MBP-

    CCP8, but these suspicions were proven

    unfounded by MST. It seems likely that

    PspC1_N3 is very sticky since the background

    noise was consistently higher than the signal.

    This could also explain why it seemed like there

    was affinity for MBP-CCP8. At higher

    concentrations of PspC1_N3, approaching

    100nM, the background noise became so great

    as to completely occlude the signal, this also

    made it impossible to calculate a wash-off curve

    with any precision.

    During Microscale Thermophoresis affinity

    interactions were observed between PspC1_N3

    and the full length fH but not between

    PspC1_N3 and the two MBP-CCP constructs

    that were tested, CCP8 and CCP9. All the runs

    that included full length fH showed a strange

    behavior in the beginning of the experiment

    before the point of T-Jump where the

    fluorescence signal was rapidly increasing. In

    the typical case the signal stays constant before

    T-Jump. One possible explanation for this

    phenomenon that resembles photo bleaching in

    reverse is that the fH aggregated at the capillary

    walls rather than flowing freely in solution. All

    experiments also produced noisy signals, which

    might be due to the signal being dependent on

    the fluorescent activity of tryptophan in the

    CCPs and not a stronger fluorophore.

    To proceed with the project from this point there

    are several avenues to pursue. Trying to assay

    binding with a Surface Plasmon Resonance

    platform such as those provided by Biacore

    seems reasonable since this type of

    measurement might produce better data than

    MST for this particular experimental set-up. It

    might be a good idea to try the disulfide bond

    reshuffling with DsbC in vitro as performed by

    Achila et. al.11 on the produced constructs. The

    T7 Shuffle Competent E.coli used as the

    expression system constitutively expresses

    DsbC, but results might differ if disulfide bond

    reshuffling is performed ex-vivo. It also seems

    reasonable to try the periplasmic expression

    constructs since these are cloned and ready for

    transformation, however the periplasmic

    constructs also have the TEV cleavage site. If

    neither of these approaches work it seems sound

    to proceed by trying a eukaryote expression

    system based on insect or leichmania cells.

    While maintaining the cell culture is more

    arduous as compared to a prokaryote cell

    culture it might be a better option since it is

    more likely to be able to express the

    recombinant CCPs without an MBP-fusion.

    This would reduce the work necessary to

    produce the pure CCPs and also reduce the

    metabolic strain on the expression system,

    which in theory should increase the yield.

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    14

    References 1. Anevlavis S, Bouros D. Community acquired bacterial pneumonia. Expert Opin Pharmacother.

    2010;11(3):361-374. doi:10.1517/14656560903508770.

    2. Pneumococcal Disease | Clinical | Streptococcus pneumoniae | CDC.

    https://www.cdc.gov/pneumococcal/clinicians/streptococcus-pneumoniae.html. Accessed May

    4, 2017.

    3. Rodgers GL, Arguedas A, Cohen R, Dagan R. Global serotype distribution among Streptococcus

    pneumoniae isolates causing otitis media in children: Potential implications for pneumococcal

    conjugate vaccines. Vaccine. 2009;27(29):3802-3810. doi:10.1016/j.vaccine.2009.04.021.

    4. French N, Gordon SB, Mwalukomo T, et al. A Trial of a 7-Valent Pneumococcal Conjugate

    Vaccine in HIV-Infected Adults. N Engl J Med. 2010;362(9):812-822.

    doi:10.1056/NEJMoa0903029.

    5. O’Brien KL, Wolfson LJ, Watt JP, et al. Burden of disease caused by Streptococcus pneumoniae

    in children younger than 5 years: global estimates. Lancet. 2009;374(9693):893-902.

    doi:10.1016/S0140-6736(09)61204-6.

    6. Schmidt CQ, Herbert AP, Kavanagh D, et al. A New Map of Glycosaminoglycan and C3b

    Binding Sites on Factor H. J Immunol. 2008;181(4).

    7. Dave S, Brooks-Walter A, Pangburn MK, McDaniel LS. PspC, a pneumococcal surface protein,

    binds human factor H. Infect Immun. 2001;69(5):3435-3437. doi:10.1128/IAI.69.5.3435-

    3437.2001.

    8. Serruto D, Rappuoli R, Scarselli M, Gros P, Strijp JAG Van. Molecular mechanisms of

    complement evasion: learning from staphylococci and meningococci. 2010.

    http://dx.doi.org/10.1038/nrmicro2366.

    9. Parham P. The Immune System.; 2014.

    10. Iannelli F, Oggioni MR, Pozzi G. Allelic variation in the highly polymorphic locus pspC of

    Streptococcus pneumoniae. Gene. 2002. doi:10.1016/S0378-1119(01)00896-4.

    11. Achila D, Liu A, Banerjee R, et al. Structural determinants of host specificity of complement

    Factor H recruitment by Streptococcus pneumoniae. Biochem J. 2015;465(2):325-335.

    doi:10.1042/BJ20141069.

    12. Herbert AP, Makou E, Chen ZA, et al. Complement Evasion Mediated by Enhancement of

    Captured Factor H: Implications for Protection of Self-Surfaces from Complement. J Immunol.

    2015;195(10). http://www.jimmunol.org/content/195/10/4986. Accessed May 17, 2017.

    13. Achila D, Liu A, Banerjee R, et al. Structural determinants of host specificity of complement

    Factor H recruitment by Streptococcus pneumoniae. Biochem J. 2015;465(2):325-335.

    doi:10.1042/BJ20141069.

    14. KALIN M. Pneumococcal serotypes and their clinical relevance. Thorax. 1998;53(3):159 LP-

    162. http://thorax.bmj.com/content/53/3/159.abstract.

    15. Li MZ, Elledge SJ. Harnessing homologous recombination in vitro to generate recombinant

    DNA via SLIC. Nat Methods. 2007;4(3):251-256. doi:10.1038/nmeth1010.

    16. Understanding how your ligand interacts with living cells.

    http://www.ligandtracer.com/docs/LigandTracer Technology Note 1.6.pdf. Accessed August 31,

    2017.

    17. Jerabek-Willemsen M, André T, Wanner R, et al. MicroScale Thermophoresis: Interaction

    analysis and beyond. J Mol Struct. 2014;1077:101-113. doi:10.1016/j.molstruc.2014.03.009.

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    15

    18. Pei J, Kim B-H, Grishin N V. PROMALS3D: a tool for multiple protein sequence and structure

    alignments. Nucleic Acids Res. 2008;36(7):2295-2300. doi:10.1093/nar/gkn072.

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    16

    Appendix

    Supplementary 1: Primers Primers used for cloning are detailed here.

    Melting temperatures were predicted by:

    http://insilico.ehu.es/tm.php or

    http://eu.idtdna.com/analyzer/Applications/OligoAnalyzer/

    Description Comment Overhang (homologous

    recombination, 54°C, >

    50°C?)

    Iidentical to target Tm

    of 60°C

    Tmelt

    (> 59°C)

    Vec_R ACCCTGGAAGTAC

    AGGTTTTCG

    Vec_F TAACTCGAGGATC

    CGGCTG

    PspC1_F CCTGTACTTCCAGG

    GT

    GAAGAGGTTGGTG

    GTAGGA

    52.8

    PspC1_F2 CCTGTACTTCCAGG

    GT

    GAAGAGGTTGGTG

    GTAGGAATACC

    56.8

    PspC1N_REV1 404bp CCGGATCCTCGAGT

    TA

    GCGATCTTCTTCTT

    TTTGATCCTCG

    57

    PspC1cbd_REV1 2075bp CCGGATCCTCGAGT

    TA

    GTTTACCCATTCAC

    CATTGGCATTG

    57.8

    PspC1cbd_REV1 CCGGATCCTCGAGT

    TA

    GTTTACCCATTCAC

    CATTGG

    51.6

    PspC1N2_REV1 1100bp CCGGATCCTCGAGT

    TA

    TTCTTCTGCTGCTT

    TTCGTTTAGC

    57

    PspC_4k12_F CCTGTACTTCCAGG

    GT

    GGGCAAGATATAT

    CGAAGAAGTATGC

    56.1

    PspC_4k12_R 290bp CCGGATCCTCGAGT

    TA

    TTCTGCAACTTTCT

    TCTCAGCTTCTG

    58.2

    MBPcyto_F 1000bp GAAGAAGGTAAAC

    TGGTAATCTGGAT

    TAAC

    56

    MBPcyto_R AGTACAGGTTTTCT

    GA

    GTTATTGTTGTTGT

    TGTTCGAGCTC

    56

    twSTII:vec_R 5600bp CCAGTTTACCTTCTT

    C

    GGAACCGCCACCG

    GAC

    58

    http://insilico.ehu.es/tm.phphttp://eu.idtdna.com/analyzer/Applications/OligoAnalyzer/

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    17

    twSTII:vec_F TCAGAAAACCTGT

    ACTTCCAGGG

    56.5

    twSTIIMBP:vec_

    R

    CATATGTTTCCTCC

    TTTTGGATCTGATA

    AATTG

    twSTIIMBP:vec_

    F

    6760bp GGCTCTCGCCAAAA

    TC

    AGCGCGTGGAGCC

    ATC

    Peri_F 100bp AAGGAGGAAACATA

    TG

    AAAATAAAAACAG

    GTGCACGCATCC

    Peri_R GATTTTGGCGAGA

    GCCGAGG

    pET_MBP_R GCCCTGGAAGTAC

    AGGTTTTC

    55.7

    pET_MBP_F TAACTCGAGGATC

    CGGCTG

    55.8

    fHCCP1:7_F 1370bp CCTGTACTTCCAGG

    GC

    CGCCTGCTGGCTA

    AAATCATTTG

    58

    fHCCP1:7_R CCGGATCCTCGAGT

    TA

    AGTTTTCACACGA

    ATGCAGCG

    57

    fHCCP6:8_F 596bp CCTGTACTTCCAGG

    GC

    ACGCTGAAACCAT

    GCGACTATC

    58

    fHCCP6:8_R CCGGATCCTCGAGT

    TA

    GCTTTTGATACAG

    GTCGGTTGTG

    57

    fHCCP8:10_F 575bp CCTGTACTTCCAGG

    GC

    AAAACTTGTAGCA

    AAAGCTCTATTGA

    CATC

    57

    fHCCP8:10_R CCGGATCCTCGAGT

    TA

    TTGTTCTTTACAGA

    TCGGCAAATCAG

    57

    fHCCP10:12_F 398bp CCTGTACTTCCAGG

    GC

    CAAAGTTGTGGCC

    CTCCG

    56

    fHCCP10:12_R CCGGATCCTCGAGT

    TA

    CTTATCGATGGCA

    ACGCATTG

    55.4

    fHCCP13:15_F 566bp CCTGTACTTCCAGG

    GC

    AAGAAATGCAAAT

    CTTCTAACCTTATC

    ATCC

    56

    fHCCP13:15_R CCGGATCCTCGAGT

    TA

    ACCTTCGCACTGT

    GGCG

    58

    fHCCP16:18_F 569bp CCTGTACTTCCAGG

    GC

    GAAGGTTTGCCCT

    GCAAATC

    55

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    18

    fHCCP16:18_R CCGGATCCTCGAGT

    TA

    GGAGTCTTTGCAT

    TGCGGC

    57.5

    fHCCP19:20_F 413bp CCTGTACTTCCAGG

    GC

    TCCACAGGCAAAT

    GCGG

    55

    fHCCP19:20_R CCGGATCCTCGAGT

    TA

    CCGTTTCGCACAA

    GTCGG

    57

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    19

    Supplementary 2: Buffer compositions

    Lysis Buffer A

    Gycerol 10% by volume

    pH 7.5 Hepes 50 mM

    NaCl 500 mM

    MgCl2 1 mM

    DNAse 40 µg/ml

    Lysozyme 500 µg/ml

    PMSF 1 mM

    1 tablet cOmplete Ultra

    protease inhibitor.

    In 50ml total volume

    dH20 To 50ml

    Lysis Buffer B

    B-PER II Bacterial Protein

    Extraction Reagent by

    Thermo Fisher.

    To 50ml

    DNAse 40 µg/ml

    Lysozyme 500 µg/ml

    PMSF 1 mM

    1 tablet cOmplete Ultra

    protease inhibitor.

    In 50ml total volume

    Storage Buffer

    Glycerol 10% by volume

    pH 7.5 Hepes 20 mM

    NaCl 300 mM

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    20

    Supplementary 2: Protein sequences of produced constructs.

    Orange – StrepTag or His6-Tag, Grey – Peptide linker, Blue – Maltose Binding Protein, Red – TEV-

    site, Black – GOI.

    twSTII-MBP-TEV-CCP8

    SAWSHPQFEKGGGSGGGSGSAWSHPQFEKSGGGSEEGKLVIWINGDKGYNGLAEVGKKFEK

    DTGIKVTVEHPDKLEEKFPQVAATGDGPDIIFWAHDRFGGYAQSGLLAEITPDKAFQDKLYPF

    TWDAVRYNGKLIAYPIAVEALSLIYNKDLLPNPPKTWEEIPALDKELKAKGKSALMFNLQEPY

    FTWPLIAADGGYAFKYENGKYDIKDVGVDNAGAKAGLTFLVDLIKNKHMNADTDYSIAEAA

    FNKGETAMTINGPWAWSNIDTSKVNYGVTVLPTFKGQPSKPFVGVLSAGINAASPNKELAKE

    FLENYLLTDEGLEAVNKDKPLGAVALKSYEEELAKDPRIAATMENAQKGEIMPNIPQMSAFW

    YAVRTAVINAASGRQTVDEALKDAQTNSSSNNNNNNSENLYFQGKTCSKSSIDIENGFISESQ

    YTYALKEKAKYQCKLGYVTADGETSGSITCGKDGWSAQPTCIKS

    twSTII-MBP-TEV-CCP8:9

    SAWSHPQFEKGGGSGGGSGSAWSHPQFEKSGGGSEEGKLVIWINGDKGYNGLAEVGKKFEK

    DTGIKVTVEHPDKLEEKFPQVAATGDGPDIIFWAHDRFGGYAQSGLLAEITPDKAFQDKLYPF

    TWDAVRYNGKLIAYPIAVEALSLIYNKDLLPNPPKTWEEIPALDKELKAKGKSALMFNLQEPY

    FTWPLIAADGGYAFKYENGKYDIKDVGVDNAGAKAGLTFLVDLIKNKHMNADTDYSIAEAA

    FNKGETAMTINGPWAWSNIDTSKVNYGVTVLPTFKGQPSKPFVGVLSAGINAASPNKELAKE

    FLENYLLTDEGLEAVNKDKPLGAVALKSYEEELAKDPRIAATMENAQKGEIMPNIPQMSAFW

    YAVRTAVINAASGRQTVDEALKDAQTNSSSNNNNNNSENLYFQGKTCSKSSIDIENGFISESQ

    YTYALKEKAKYQCKLGYVTADGETSGSITCGKDGWSAQPTCIKSCDIPVFMNARTKNDFTWF

    KLNDTLDYECHDGYESNTGSTTGSIVCGYNGWSDLPICYERE

    twSTII-MBP-TEV-CCP9

    SAWSHPQFEKGGGSGGGSGSAWSHPQFEKSGGGSEEGKLVIWINGDKGYNGLAEVGKKFEK

    DTGIKVTVEHPDKLEEKFPQVAATGDGPDIIFWAHDRFGGYAQSGLLAEITPDKAFQDKLYPF

    TWDAVRYNGKLIAYPIAVEALSLIYNKDLLPNPPKTWEEIPALDKELKAKGKSALMFNLQEPY

    FTWPLIAADGGYAFKYENGKYDIKDVGVDNAGAKAGLTFLVDLIKNKHMNADTDYSIAEAA

    FNKGETAMTINGPWAWSNIDTSKVNYGVTVLPTFKGQPSKPFVGVLSAGINAASPNKELAKE

    FLENYLLTDEGLEAVNKDKPLGAVALKSYEEELAKDPRIAATMENAQKGEIMPNIPQMSAFW

    YAVRTAVINAASGRQTVDEALKDAQTNSSSNNNNNNSENLYFQGSCDIPVFMNARTKNDFT

    WFKLNDTLDYECHDGYESNTGSTTGSIVCGYNGWSDLPICYERE

    twSTII-MBP-TEV-CCP10

    SAWSHPQFEKGGGSGGGSGSAWSHPQFEKSGGGSEEGKLVIWINGDKGYNGLAEVGKKFEK

    DTGIKVTVEHPDKLEEKFPQVAATGDGPDIIFWAHDRFGGYAQSGLLAEITPDKAFQDKLYPF

    TWDAVRYNGKLIAYPIAVEALSLIYNKDLLPNPPKTWEEIPALDKELKAKGKSALMFNLQEPY

    FTWPLIAADGGYAFKYENGKYDIKDVGVDNAGAKAGLTFLVDLIKNKHMNADTDYSIAEAA

    FNKGETAMTINGPWAWSNIDTSKVNYGVTVLPTFKGQPSKPFVGVLSAGINAASPNKELAKE

    FLENYLLTDEGLEAVNKDKPLGAVALKSYEEELAKDPRIAATMENAQKGEIMPNIPQMSAFW

  • Master Thesis Report Royal Institute of Technology Nils Lindström

    21

    YAVRTAVINAASGRQTVDEALKDAQTNSSSNNNNNNSENLYFQGERECELPKIDVHLVPDRK

    KDQYKVGEVLKFSCKPGFTIVGPNSVQCYHFGLSPDLPICKEQ

    His6-TEV-PspC1_N1

    HHHHHHENLYFQGGQDISKKYADEVESHLKKILSEIQTQLDRKRHTKTVALINELQDIKKTYL

    YNLNVLKEKSELPSKIKAKLEVAFDQFKKDTLKPGEKVAEAEKKVAE

    His6-TEV-PspC1_N2

    HHHHHHENLYFQGEEVGGRNTPTVTSSGQDISKKYADEVESHLKKILSEIQTQLDRKRHTKTV

    ALINELQDIKKTYLYNLNVLKEKSELPSKIKAKLEVAFDQFKKDTLKPGEKVAEAEKKVAEA

    KKKAEDQKEEDR

    His6-TEV-PspC1_N3

    HHHHHHENLYFQGEEVGGRNTPTVTSSGQDISKKYADEVESHLKKILSEIQTQLDRKRHTKTV

    ALINELQDIKKTYLYNLNVLKEKSELPSKIKAKLEVAFDQFKKDTLKPGEKVAEAEKKVAEA

    KKKAEDQKEEDRRNYPTNTYKTLELEIAESDVKVKEAELELVNEEAKPGNEEKIKKAKAKVE

    SEKAEAIRLEEIKTDREEAKRKADAKLKEAVENNAATSEQGEPKRRVKRGVLGEPATPDKKE

    NDAKSSDSSVGEETLPSPSLKPEKKVAEAEKKAKDQKEEDRRNYPTNTYKTLELEIAESDVKV

    KEAELELVKEEAKESRNEEKVKQAKAKVESKKAEATRLEKIKTDRKKAEEAKRKAAEE

    His6-TEV-PspC1_CBD

    HHHHHHENLYFQGEEVGGRNTPTVTSSGQDISKKYADEVESHLKKILSEIQTQLDRKRHTKTV

    ALINELQDIKKTYLYNLNVLKEKSELPSKIKAKLEVAFDQFKKDTLKPGEKVAEAEKKVAEA

    KKKAEDQKEEDRRNYPTNTYKTLELEIAESDVKVKEAELELVNEEAKPGNEEKIKKAKAKVE

    SEKAEAIRLEEIKTDREEAKRKADAKLKEAVENNAATSEQGEPKRRVKRGVLGEPATPDKKE

    NDAKSSDSSVGEETLPSPSLKPEKKVAEAEKKAKDQKEEDRRNYPTNTYKTLELEIAESDVKV

    KEAELELVKEEAKESRNEEKVKQAKAKVESKKAEATRLEKIKTDRKKAEEAKRKAAEEDKV

    KEKPAEQPQPAPAPQPEKPAPKPEKPAPAPKPENPAEQPKAEKPADQQAEEDYARRSEEEYNR

    LTQQQPPKTEKPAQPSTPKTGWKQENGMWYFYNTDGSMATGWLQNNGSWYYLNSNGAMA

    TGWLQNNGSWYYLNANGSMATGWLQNNGSWYYLNANGSMATGWLQNNGSWYYLNANG

    SMATGWLQNNGSWYYLNANGSMATGWLQYNGSWYYLNANGSMATGWLQYNGSWYYLN

    SNGAMVTGWLQNNGSWYYLNANGSMATDWVKDGDTWYYLEASGAMKASQWFKVSDKW

    YYVNGSGALAVNTTVDSYRVNANGEWVN

  • www.kth.se

    kth-cover3218715292565489387 nilsNils Lindström Master Thesis Report 170921kth-cover3218715292565489387 nils