REVERSIBLE STREPTAVIDIN BASED ANALYTE ENRICHMENT SYSTEM FOR USE IN CROSSLINKING MASS SPECTROMETRY ANALYSIS

Information

  • Patent Application
  • 20230280352
  • Publication Number
    20230280352
  • Date Filed
    June 17, 2021
    2 years ago
  • Date Published
    September 07, 2023
    8 months ago
Abstract
It is provided a reversible streptavidin based analyte enrichment system for use in crosslinking mass spectrometry analysis, in particular for enriching at least parts of crosslinked peptides pairs in mass spectrometry analysis, and a method of enriching at least parts of crosslinked peptides pairs, in particular for use in crosslinking mass spectroscopy analysis.
Description

The disclosure relates to a reversible streptavidin based analyte enrichment system for use in crosslinking mass spectrometry analysis, and a method of enriching at least parts of crosslinked peptides pairs, in particular for use in crosslinking mass spectroscopy analysis.


BACKGROUND

Bioconjugation via the streptavidin--biotin system is thoroughly established as a universal tool in most fields of the biological sciences, including (but not limited to): Affinity chromatography, isolation studies, immobilising agents, selective elimination, flow cytometry, affinity cytochemistry, localisation studies, light microscopy, fluorescent microscopy, electron microscopy, histochemistry, signal amplification, immunoassay, diagnostics, gene probes, bioaffinity studies, epitope mapping, blotting technology, cytological probes, pathological probes, affinity therapy, drug delivery, fusogenic agents, affinity perturbation, affinity targeting, protein purification and crosslinking.


The streptavidin-biotin system is so widely used, partly because of the exceptionally high affinity, specificity and stability of the streptavidin-biotin complex. The streptavidin-biotin interaction is the strongest non-covalent biological interaction known (Kd ~ 10-14 M). However, the strength of the interaction becomes a liability when reversal of the interaction is desired.


Methods attempting to reverse the interaction include extreme denaturing conditions, such as boiling in 6 M guanidinium hydrochloride at pH 1.5. This frustrates the range and scope of its application, where only conjugation is realistically feasible when dealing with sensitive study subjects. Extreme elution conditions can destroy the biological sample of interest and/or replace the undesired sample background with an equally deleterious background deriving from the enrichment procedure. Other problems associated with the use of biotin are its relative hydrophobicity and resultant insolubility, as well as its tendency to oxidise in certain preparative conditions (a particular problem for methods relating to mass spectrometry) (Villamor, J. G. et al. Profiling protein kinases and other ATP binding proteins in Arabidopsis using Acyl-ATP probes. Mol. Cell. Proteomics 12, 2481-2496 (2013)).


Crosslinking mass spectrometry analysis is a field of structural biology that could benefit enormously from the implementation of a truly reversible streptavidin-biotin based enrichment system. Crosslinking mass spectrometry involves the translation of temporal spatial proximity between and within proteins into covalent bonds. Proximal amino acid residues are crosslinked and proteins subsequently digested into complex mixtures of crosslinked and non-crosslinked peptides and peptide pairs, using enzymes. The resulting complex mixture is analysed by mass spectrometry and matched by database search, thus elucidating protein-protein interactions and revealing protein structure. Importantly, one of the largest advantages of crosslinking mass spectrometry technology is that it allows the study of proteins inside their native environment, the cell. A significant challenge to the approach, however, is that crosslinked peptide pairs containing the three-dimensional structural information following crosslinking, are typically of very low abundance, whereas analysis is usually performed on a much greater abundant modified and non-modified linear peptide mixture. Analysis is therefore akin to trying to find the proverbial needle in a haystack.


Currently, standard crosslinking mass spectrometry can routinely generate data on individual proteins and small-to-medium sized complexes, but when sample complexity increases beyond this, enrichment of crosslinked peptide pairs becomes a fundamental necessity, especially when scaling crosslinking upwards to cellular lysates, organelles and even living cells.


Crosslinking can be achieved using exogenous chemical crosslinking reagents, which represent a foundational pillar of crosslinking mass spectrometry. Such crosslinking reagents typically constitute two protein-reactive groups separated by a spacer (the simplest being an alkyl chain of varying length). The spacer can act as a passive distance restraint or can have additional functionality such as reactive or affinity handles grafted onto the crosslinker scaffold, which allow the capability for enrichment of crosslinked peptide pairs following enzymatic digestion of crosslinked protein. Any reactive or affinity handles present on a crosslinker need to be chemically inert, or at least non-reactive with the protein-reactive groups. In addition, crosslinkers should be chemically inert under the electrospray and gas-phase conditions of the mass spectrometer, unless there is the intention of generating specific reporter ions for the purposes of enhanced search strategies.


Analyte enrichment can be achieved in a three-step process. Firstly, the reactive or affinity group grafted on the crosslinker spacer must bind the crosslinked peptide pairs to a solid support. Secondly, the solid-support is washed to remove non-binding, non-crosslinker-modified linear peptides. Thirdly, solid-support bound crosslinked peptides are eluted prior to their analysis by mass spectrometry. For successful enrichment, all three steps must be completed. Crosslinking is one aspect of the broader field of bioconjugation.


In applications where solid-supported streptavidin is used to capture biotinylated moieties, the non-reversibility issue of the streptavidin-biotin system has led to numerous attempts at workarounds, including (but not limited to): using enhanced denaturing conditions, on-bead digestion of bound proteins or by implementing cleavable chemistry for biotin derivatives. Enhanced denaturing conditions can be characterised as either involving: 1. Attempts at competitive elution (Cheah, J. S. & Yamada, S. A simple elution strategy for biotinylated proteins bound to streptavidin conjugated beads using excess biotin and heat. Biochem. Biophys. Res. Commun. 493, 1522--15′-)7 (2017)) or, 2. Attempts to reduce the binding affinity of one part of the system, either the “solid-support” component (with modified avidin molecules) (Vermette, P. et al. immobilisation and surface characterisation of NeutrAvidin biotin-binding protein on different hydrogel interlayers. J. Colloid Interface Sei. 259, 13-26 (2003)) or the “tag” component (with reduced-affinity biotin analogues) (Garret-Flaudy, F. & Freitag, R. Use of the avidin (imino)biotin system as a general approach to affinity precipitation. Biotechnol. Bioeng. 71, 223-234 (2000)). None of these workarounds are fully satisfying when applied.


Competitive elution is an accepted concept and is already applied whereby both biotin and desthiobiotin are used as an eluent. Where excess biotin is currently used for attempting competitive elution of biotinylated bound derivatives, excess heating is also required. In the case of the Strep-tag® system, competitive elution from the affinity column is achieved without extreme heating, using either biotin or the more weakly binding desthiobiotin. The Strep-tag® system is used to purify proteins on a specially engineered solid-supported streptavidin (Strep-Tactin). Strep-Tag II is an 8 amino acid synthetic peptide that is N- or C-tenninally fused to recombinant proteins and has high-specificity for Strep-Tactin, but a binding affinity lower than both desthiobiotin and biotin. DSB-X biotin is a desthiobiotinylated succinimidyl ester reagent that can either be used to reversibly bind tagged moieties to a solid support, or modify the solid-support itself for reversible binding of free protein (Hirsch, J. D. et al. Easily reversible desthiobiotin binding to streptavidin, avidin, and other biotin-binding proteins: uses for protein labeling, detection, and isolation. Anal.Biochem. 308, 343-357 (2002)). Competitive elution is used to reverse binding on desthiobiotin incorporated into chemical probes (desthiobiotin-ATP), for studying nucleotide-binding proteins and grafted to DNA/RNA for the analysis of chromatin composition. In these cases, desthiobiotin is used as an affinity group to link two entities, where reversibility is required at one end.


Both biotin- and non-biotin-based enrichable crosslinking reagents have been developed for crosslinking mass spectrometry. An enrichable crosslinker, using a biotin handle as an affinity group for crosslinking enrichment, even pre-dates the time when crosslinking was routinely being analysed by mass spectrometry (Hatanaka, Y., Hashimoto, M. & Kanaoka, Y. A novel biotinylated heterobifunctional cross-linking reagent bearing an aromatic diazirine. Bioorg. Med. Chem. 2, 1367--1373 (1994)). Since then, multiple biotin-exploiting crosslinkers have followed, whereby biotin is either directly grafted onto a crosslinker spacer region (Fujii, N., Jacobsen, R. B., Wood, N. L., Schoeniger, J. S. & Guy, R. K. A novel protein crosslinking reagent for the determination of moderate resolution protein structures by mass spectrometry (MS3-D). Bioorg. Med. Chem. Lett. 14, 427-429 (2004)), or subsequently added through a bioorthogonal transformation of another crosslinker functionality following crosslinking (Tan, D. et al. Trifunctional cross-linker for mapping protein-protein interaction networks and comparing protein conformational states. Elife 5, el2509 (2016)). Most recently there has been an attempt to move away altogether from a biotin-streptavidin based enrichment system for crosslink enrichment, with the introduction of an IMAC-enrichable phosphonic acid based crosslinking reagent (Steigenberger, B., Pieters, R. J., Heck, A. J. R. & Scheltema, R. A. PhoX: An IMAC-Enrichable Cross-Linking Reagent. ACS Cent Sei 5, 1514-1522 (2019)). Vanishingly few of these crosslinkers have progressed past the proof-of-principle stage, however.


SUMMARY

The object underlying the proposed solution was to provide a tool that allows for an improved analysis of proteins by mass spectroscopy.


According to the proposed solution this object is being solved by a system comprising a crosslinking reagent having features as described herein.


Accordingly, a reversible streptavidin based analyte enrichment system for use in crosslinking mass spectrometry analysis, in particular for enriching at least parts of crosslinked peptides pairs in mass spectrometry analysis, is provided, wherein the system comprises at least one crosslinking agent of the structure of general formulae (I)




embedded image


Wherein

  • R1 is selected from a functional group comprising ester, halogen, aldehyde, isothiocyanate, isocyanate, anhydride, epoxide, acetyl, glyoxal, triazine, hydrazine, disulfide, azide, ketone, phosphine, alkene, amine, N-heterocycle,
  • a = 0-10, preferably 1-5, more preferably 2-3;
  • Y is selected from —O—, —S—, —S—S—, —S(═O)—, —13CH2—, —CD2—, -18O-m, in particular —O—, —S—, —S—S—; most in particular —O—;
  • n = 0-4, preferably 0, 1 or 2,
  • X is selected from a group comprising —N—, —C6H3(NH—), —CH(NH—)—, wherein R2 is attached to N, —CH—, —CH(CO—NH—), wherein R2 is attached to C;
  • R2 is one of the structures of formulae (IV) comprising biotin analogues or formulae (V) comprising or desthiobiotin analogues (V), wherein biotin is excluded:
  • embedded image - (IV)
  • embedded image - (V)

wherein
  • -X1, X2, X3 is selected from —NH2, —NH—, —N—Me, —N—Et, —O—.,
  • R3 is selected from the group consisting of H, optionally substituted C1-C10 alkyl, optionally substituted C3-C10 cycloalkyl, optionally substituted C2-C10 alkenyl, optionally substituted C3-C10 cycloalkenyl, optionally substituted C2-C10 alkynyl, optionally substituted C2-C10 heteroalkyl, optionally substituted C3-C10 heterocycloalkyl, optionally substituted C2-C10 heteroalkenyl, optionally substituted C3-C10 heterocycloalkenyl, optionally substituted C2-C10 heteroalkynyl, optionally substituted C6-C14 aryl, optionally substituted C5-C14 heteroaryl;
  • Z is selected from —CO—, aryl, preferably C5-C6 such as C6-C14 aryl, C5-C14 heteroaryl, alkyl, preferably C1-C4 alkyl, triazoles, alkenes, preferably C2-C4 alkenes, alkyl tetrazines, preferably C1-C4 tetrazines, optionally substituted —CO—NH—(CH2)b—, —CO—NH—(CH2)b—NH—CO—, —CO—NH—(CH2)b—CO—, wherein b = 1-10, preferably 2-6, optionally substituted —CO—NH—(CH2—O—)c—(CH2)d—CO—, CO—NH—(CH2—O—)c—Triazin—(CH2)d—CO—, —CO—NH—(CH2)b—(CH2—O—)c—(CH2)d—NH—CO—(CH2)e—CO— wherein b, c, d, e = 1-10, preferably 2-6.


Thus, the crosslinking agent according to the solution is a trifunctional agent, which comprises at least two functional moieties as terminal moieties and a biotin derivative that is able to bind to streptavidin. An essential feature is that a biotin derivative is used as affinity group, which has a lower affinity to avidin or streptavidin than free biotin. The two terminal moieties (R1) placed on the crosslinking agent can be identical (homobifunctional crosslinkers) or different (heterobifunctional crosslinkers).


In Hirsch et al. desthiobiotin was used in a bifunctional reagent that reacts with proteins for the capture and competitive elution of all proteins in a sample: Total protein binding is followed by total elution. There was no enrichment of labelled proteins over non-labelled proteins. It remained unclear if DTB could be used for the purpose of enrichment at all and if this would suffice to achieve extreme enrichment ratios as are needed in our application. Hirsch et al. does not mention or even discuss the possibility of enrichment using DTB. This is not obviously the case, as DTB has a lower affinity for streptavidin than biotin, which is used commonly for the capture of biomolecules, but has elution problems. Hirsch et al. also shows the retention of proteins that are likely conjugated to multiple copies of DTB which strongly enhances the binding affinity to the stationary phase. It is unclear if peptides conjugated to a single DTB moiety would efficiently be retained and if this would suffice for enrichment. It could also be that DTB might affect the tryptic cleavage of crosslinked proteins into peptides, which is a critical step for mass spectroscopy analysis. Using DTB for labelling requires the synthesis of bifunctional probes.


As presently claimed, a trifunctional structure is needed to (i) crosslink residues between proteins and (ii) enrich crosslinked peptides. The synthetic routes required for trifunctional structures are a significantly greater challenge because they typically necessitate the manipulation of multiple orthogonal protecting groups to achieve the final reagent. It can therefore not be simply concluded that the existence of DTB in a bifunctional probe could be transferred into a trifunctional probe.


Crosslinked peptides are present in a matrix of a much more abundant and very complex mixture of peptides that are not crosslinked. This renders their mass spectrometric detection highly challenging when working with complex biological samples, such as whole cells or organelles. To improve the detection of crosslinked peptides, biotin is used in crosslinkers as a high-affinity capture reagent. Because of the very low abundance of crosslinked peptides and the very high abundance of the matrix one must have a very large difference in binding affinities to a solid support between the crosslinked peptides and the matrix (non-crosslinked peptides) to achieve a separation or at least marked enrichment of crosslinked peptides. This is being achieved by biotin as part of the crosslinker. However, the problem to be solved is how to then efficiently elute the crosslinked peptides under conditions that leave the peptides intact and unmodified. The field has been developing chemistry as part of the crosslinker to cleave off the captured crosslinked peptides. However, this can change the peptides and so far, is not very efficient. The solution represents a means of doing chemistry-free enrichment, since the elution of crosslinked peptides does not require chemically (and/or photo-) cleavable linkers. The presently claimed reagent and method comprises a crosslinker reagent that is chemically unchanged, between the point of the crosslinking reaction and analysis, resulting in greater analysis efficiency.


Because of the lower affinity of DTB compared to biotin it is not obvious that the desired enrichment ratios can be achieved by DTB. It is also not clear a priori that elution is efficient. Finally, it is not a priori clear if the capture and elution can be achieved with the very small quantities typically available in biological samples and analysed by mass spectrometry. The overall efficiency of the process must be very high.


It would also not be clear if DTB is actually compatible with MS fragment analysis. If Biotin-modified peptides can be eluted without chemical cleavage, Biotin gives numerous breakdown products that complicate the MS2 spectra. In addition, biotin oxidation under mild conditions is well-known and complicates the MS analysis. The absence of the thioether group in DTB, solves the oxidation issue. It is unclear how DTB-peptide conjugates would behave under peptide fragmentation conditions in the mass spectrometer and if peptide identification would suffer or even still be possible.


In an embodiment the crosslinking agent is of an asymmetrical structure (II)




embedded image - (II)


or a symmetrical structure (III)




embedded image - (III)


wherein

  • a = 1-10, preferably 2-5, more preferably 2-3.
  • n = 1-4, preferably 1-2, more preferably 1.
  • Y is selected from —O—, —S—, —S—S—, —S(═O)—, —13CH2—, —CD2—, —18O—.


The moiety R1 may be selected from a group comprising N-hydroxysuccinimide esters, imidoesters, isothiocyanates, isocyanates, acyl chlorides, sulfonyl chlorides, aryl sulfonyl fluorides, acyl azides, fluorophenyl esters, anhydrides, fluorobenzene, epoxides, alpha,beta-unsaturated aldehydes, 1,3-ketoaldehydes, 1,2,3-triazines, 1,2-cyclohexanedione, 2-methoxy-3-oxindoles, phenylglyoxal, a-keto-oximes, 2-fluoro-5-nitrotropolone, O-phthalaldehyde, maleimides, halo acetyls, pyridyl disulfides, aryl azides, diazirines, benzophenones, psoralens, phoshines, alkenes, cyclooctynes, tetrazines, hydrazines, alkoxyamines;


In an embodiment the terminal moiety R1 may be ester groups such N-hydroxysuccinimide esters (NHS), imidoesters, isothiocyanates and isocyanates, but also may be acyl chlorides, sulfonyl chlorides, aryl sulfonyl fluorides, acyl azides, anhydrides, fluorobenzene, epoxides, aldehydes, 1,2,3-triazines, 1,2-cyclohexanedione, 2-methoxy-3-oxindoles, phenylglyoxal, a-keto-oximes and 2-fluoro-5-nitrotropolone. These terminal moieties are able to undergo a reaction with amino groups (primary amines —NH2 or secondary amines —NH—) or hydroxyl groups (—OH), in particular on the side chain of Lys, Arg, His, Ser, Thr or Tyr in proteins.


Terminal moieties R1 such as maleimides, halo acetyls and pyridyl disulfides are able to undergo a reaction with sulfhydryl groups (—SH) of Cys in proteins. Photoreactive terminal moieties such as aryl azides, diazirines, benzophenones and psoralens can react unspecifically with amino acid residues within proteins or with nucleic acids, such as DNA/RNA. The aldehyde/ketone groups present on other biomolecules (i.e. oxidised sugars of glycoproteins) can react with hydrazines and alkoxyamines when they are used as reactive sites of a crosslinking agent.


A different approach is the incorporation of noncanonical amino acids that enable site-specific or residue-selective labelling of proteins with alkyl azides, alkenes or trans-cyclooctenes. These bioorthogonal chemical groups allow covalent binding through a crosslinking agent by (a) Staudinger ligation between alkyl azides and phosphine reagents, (b) Cu(I)-catalysed cycloaddition between alkyl azides and alkynes to afford triazole adducts, (c) Cu-free cycloaddition between alkyl azides and activated cyclooctynes, or (d) tetrazine moieties can undergo selective and rapid Diels-Alder reactions with activated alkenes such as trans-cyclooctenes.


In a preferred embodiment said terminal moiety R1 may be ester groups such as succinimide ester (NHS), phthalic-di-aldehyde, diazirine.


The (at least) two functional terminal moieties are separated by a spacer comprising —(CH2)a — X—(CH2)a —. The spacer arm of the general structure (I) can be varied by its length (~7-30 Å, a = 1-10, preferably 2-5, more preferably 2-3). The length of the spacer arm impacts the distance restraint information obtained from crosslinking analysis. Longer spacer arms are able to capture more distance restraints in proteins, but the information can be less informative for structural modelling than that derived from the use of shorter spacer arms.


The spacer arm can also be varied by its composition, which can affect subsequent hydrophobicity/hydrophilicity (i.e. polyethylene glycol chains increase hydrophilicity) or enhance data analysis capabilities through additional functionality (such as isotopically labelled chains for crosslinking quantitation and cleavable groups such as disulfides or sulfoxides).


As mentioned above moiety R2 is one of the following biotin analogues (IV) or desthiobiotin analogues (V):




embedded image - (IV)




embedded image - (V)


It is to be noted that all the stereocenters are selected for both configurations S and R (4-16 stereoisomers in total), preferably configuration 3S, 4S, 6R for biotin analogues and configuration 3S, 4S for desthiobiotin analogues.


In a further embodiment of the present crosslinking agent X1, X2 are in each case NH and X3 is O, NH.


In an embodiment of the present crosslinking agent moiety R2 comprises a biotin derivative wherein the cyclic moiety of biotin is different from that of free biotin such as desthiobiotin, 2-iminobiotin, 3,4-diaminobiotin or chemically modified derivatives thereof such as carbonates or carbamates. It is mostly preferred if R2 comprises desthiobiotin. R2 may be added to the crosslinking reagent before or after crosslinking. Thus, the moiety R2 has a lower affinity to avidin or streptavidin than free biotin to allow competitive elution, but at least Kd 10-5 M to allow selective enrichment. It is to be noted that R2 is attached to the crosslinker without the intention of cleavage for elution.


Optionally, substituents in the valeryl side chain (R3) affects by decreasing the affinity towards Streptavidin, and therefore more suitable for reversible binding. In a preferred embodiment R3 is H or —CH3.


In still another embodiment Z is — CO—, —CO—NH—(CH2)b—, —CO—NH—(CH2)b—NH—CO—, —CO—NH—(CH2)b—CO—, —CO—NH—(CH2—O—)c—(CH2)d—CO—, —CO—NH—(CH2—O—)c—Triazin—(CH2)d—CO—, —CO—NH—(CH2)b—(CH2—O—)c—(CH2)d—NH—CO—(CH2)e—CO— wherein independently from each other b, c, d, e = 1-8, preferably 2-6, more preferably 2-5.


The crosslinking agent may have one of the following structures:




embedded image - 4




embedded image - 5




embedded image - 6




embedded image - 7




embedded image - 8




embedded image - 9




embedded image - 10




embedded image - 11




embedded image - 12




embedded image - 13


In one embodiment a preferred crosslinking agent is synthesised in a method comprising the following steps:

  • a) Reacting a compound of general formulae (Ia)
  • embedded image
  • wherein
    • R1a is an ester —COOR1b with R16 being a C1-C10 alkyl moiety, preferably a C3-C4 alkyl moiety, or another leaving group, preferably —Br, —I, —Cl, —OTs, -OMs, -OCOR, —ONO2, —OPO(OH)2 and
    • X is selected from N, P, —CH—, aliphatic cyclic structures and aromatic cyclic structures;

    with at least one biotin derivative R2 for obtaining a compound of general formulae (Ib).
  • embedded image
  • b) adding at least one acid for hydrolysing agent to obtain a compound of general formulae (Ic)
  • embedded image
  • c) Adding at least one compound of the general formulae (1d)
  • embedded image
  • Wherein
    • Y is a leaving group like —Br, —I, —Cl, —OTs, -OMs, -OCOR, —ONO2, —OPO(OH)2 and
    • Nu is a nucleophile like —NH2, —OH, —SH.

    to obtain the compound of general formulae (I)
  • embedded image


In a preferred embodiment of the present method the compound of the general formulae (1d) is selected from a group comprising ester, halogen, aldehyde, isothiocyanate, isocyanate, anhydride, epoxide, acetyl, glyoxal, triazine, hydrazine, disulfide, azide, ketone, phosphine, alkene, amine.


The compound of the general formulae (1d) may be selected from a group comprising N-Hydroxysuccinimide esters, imidoesters, isothiocyanates, isocyanates, acyl chlorides, sulfonyl chlorides, aryl sulfonyl fluorides, acyl azides, fluorophenyl esters, anhydrides, fluorobenzene, epoxides, alpha,beta-unsaturated aldehydes, 1,3-ketoaldehydes, 1,2,3-triazines, 1,2-cyclohexanedione, 2-methoxy-3-oxindoles, phenylglyoxal, a-keto-oximes, 2-fluoro-5-nitrotropolone, O-phthalaldehyde, maleimides, halo acetyls, pyridyl disulfides, aryl azides, diazirines, benzophenones, psoralens, phosphines, alkenes, cyclooctynes, tetrazines, hydrazines, alkoxyamines.


Further synthesis methods are illustrated in the Figures.


As mentioned above the present crosslinking agent is used for crosslinking mass spectrometry analysis. In particular, the present crosslinking agent is used for enriching at least parts of crosslinked peptides pairs.


Such a method for enriching at least parts of crosslinked peptides pairs comprises the steps of:

  • Providing a mixture of at least one crosslinking agent as described above and at least one protein to be analysed,
  • Digesting the protein by adding at least one proteolytic enzyme to obtain a peptide mixture of crosslinked peptides and linear peptides;
  • Applying the peptide mixture to a streptadivin support, whereby the crosslinked peptides pairs will be bound to the streptadivin support and linear peptides are washed out; and
  • Eluting the crosslinked peptides from the streptavidin support with an excess of biotin to obtain enriched crosslinked peptide pairs.


In a preferred embodiment the crosslinked peptides are eluted from the streptavidin support by using biotin contained in a buffer system with a pH between 6 and 8, in particular at a pH of 6.5 and 7. The crosslinking reaction is carried out in an amine-free aqueous buffer at or close to physiological pH (pH 7.2 - 7.8). Reaction time is typically between 30 minutes and 2 hours, carried out on ice or at room temperature. The crosslinking reaction is quenched by adding NH4HCO3 (ABC) at a concentration of 50 mM, with incubation for 15 minutes at room temperature.


The peptide mixture is applied to a streptadivin support, in particular streptavidin immobilized on Sepharose (highly crosslinked beaded agarose with high chemical stability, GE Healthcare).


Eluting may be done using a suitable buffer system such as PBS buffer system.


For the purpose of conducting the above enrichment process a kit may be provided, wherein the kit comprises

  • at least one crosslinking agent as described above;
  • Streptadivin attached to a solid support;
  • optionally at least one proteolytic enzyme;
  • a mobile phase / eluting agent.


As illustrated in FIG. 4, left site, the present solution provides a biotin-streptavidin system based enrichable crosslinker reagent which solves the problem of poor reversibility of biotin-streptavidin binding with the use of desthiobiotin, a biotin derivative with a weaker (but still substantial) binding affinity to streptavidin. Elution from solid-supported streptavidin is achieved via competitive elution, using an excess of biotin (in PBS buffer, pH 7.4), which acts as a competitive inhibitor for binding between streptavidin and desthiobiotin. The extremely mild elution conditions allowed by the competitive elution concept have few to no negative consequences for subsequent analyses, including by electrospray ionisation mass spectrometry. This approach may be described as crosslinker STAGEcl, for Stop And Go Extraction crosslinker, reflecting a continuation of heritage from the StageTips (Rappsilber, J., Ishihama, Y. & Mann, M. Stop and go extraction tips for matrix-assisted laser desorption/ionization, nanoelectrospray, and LC/MS sample pretreatment in proteomics. Anal. Chem. 75, 663-670 (2003)) widely used for peptide extraction in proteomics.


Thus, the solution provides the synthesis of an enrichable crosslinking reagent (STAGEcl), in a very simple three-step synthesis. STAGEcl comprises two protein reactive (NHS-ester) groups (that react predominantly with proximal lysine residues, but also with N-termini, tyrosine, threonine and serine residues) separated by a spacer, onto which a desthiobiotin moiety is grafted in the middle.


The solution provides also a means to do crosslinking mass spectrometry analysis on proteins inside living cells, organelles and cellular lysates, by enrichment of crosslinked and tagged peptides, where analysis is currently prohibited by the overabundance of linear background peptides.


The solution represents an enrichable, bioorthogonal, chemical reagent for bioconjugation, whereby two linked moieties (two analytes or one analyte and one probe) can be reversibly bound and released from a capture agent. This is in contrast to the systems as described in the prior (see also FIG. 4, right side), wherein a release of the linked moieties is only possible by chemical cleavage at least partially destroying the analyte.


The solution represents further a trifunctional chemical reagent for bioconjugation, whereby one of the functionalities is an enrichable tag and two functionalities are reactive groups capable of forming covalent bonds.


Besides, the solution provides a means of reversible binding by competitive elution (using biotin contained in an extremely mild buffer (PBS, pH 7.4), of crosslinked analytes, including proteins and peptides. The solution comprises a biotin-alternative affinity reagent for use in chemical tags and crosslinking proteomics, which has greater water-solubility, is more chemically inert, less prone to aggregation, simpler and easier to elute from the solid-support.


The solution comprises also a chemistry for grafting a functionality containing a carboxylic acid derivative to a secondary amine on a crosslinker precursor backbone, via a tertiary amide, for the purposes of functionalized crosslinker synthesis.





BRIEF DESCRIPTION OF THE DRAWINGS

The solution is explained in more detail with reference to examples and figures.



FIGS. 1a-j show synthesis of different crosslinkers according to the solution.



FIG. 2 shows GST crosslinking titration using a crosslinker according to the solution.



FIGS. 3A-C show crosslinking mass spectrometry analysis of Human Serum Albumin (HSA) crosslinked wit crosslinker 3 according to the solution.



FIG. 3D shows a crosslinking and enrichment procedure.



FIG. 3E shows enrichment of crosslinked peptide pairs.



FIG. 4 shows a schematic comparison of prior art approach and the approach according to the solution.





DETAILED DESCRIPTION


FIG. 1A shows the three-step synthesis and final structure of one crosslinker 4 according to the solution (called STAGEcl). The finished crosslinking reagent constitutes NHS-ester protein reactive groups (reactive predominantly with the amino groups of lysine residues and protein N-termini, but also with the hydroxyl groups of serine, threonine and tyrosine), separated by a spacer, which has a desthiobiotin moiety grafted in the middle, via a tertiary amide.


Synthesis Protocol of Crosslinker 4
General Methods

Chemicals and solvents were purchased from Fisher Scientific, Sigma-Aldrich, VWR International Ltd or TCI UK Ltd. NMR spectra were recorded at ambient temperature on a 500 MHz Bruker Avance III spectrometer. Chemical shifts are reported in parts per million (ppm) relative to the solvent peak. Rf values were determined on Merck TLC Silica gel 60 F254 plates under a 254 nm UV source. Purification was carried out by flash chromatography using commercially available Silica 60 Å, particle size 40-63 micron under positive pressure. Compounds purity was >95% pure, as measured by LC-MS using an UV-Vis 254 nm detector and Bruker ESI Micro-Tof mass spectrometer. Method was eluent A: water and trifluoroacetic acid (0.4%); eluent B: acetonitrile; A/B = 95:5 to 20:80 in 6 min, isocratic 1 min, 20:80 to 95:5 in 0.1 min, and isocratic 2 min.


Synthesis of Di-tert-butyl 3,3′-((6-(5-methyl-2-oxoimidazolidin-4-yl)hexanoyl)azanediyl) Dipropionate



embedded image


d-desthiobiotin (500 mg, 2.33 mmol) and 1-hydroxybenzotriazole hydrate (HOBt) (530 mg, 3.50 mmol) were dissolved in dry DCM (25 mL) under a nitrogen atmosphere. N,N-Diisopropylethylamine (DIEA) (1.1 mL, 6.99 mmol) was added dropwise and a cloudy solution was formed. Then, N,N′-diisopropylcarbodiimide (DIC) (545 L, 3.50 mmol) was added dropwise and the mixture was stirred for 10 mins. A solution of di-tert-butyl 3,3′-iminodipropionate (1, 650 L, 2.33 mmol) in dry DCM (1 mL) was then added under a nitrogen atmosphere. After the reaction was stirred overnight at room temperature, the mixture was concentrated under reduced pressure. The crude residue was dissolved in DCM and washed with water (3 × 20 mL), HCl 5% 2N (3 × 20 mL) and brine (20 mL). The organic layer was dried using anhydrous MgSO4 and the solvent was evaporated in vacuo. The resulting residue was purified by flash chromatography (2.5% MeOH in DCM) to yield 2 as a pale oil (496 mg, 46% yield). Rf = 0.44 (5% MeOH in DCM). 1H NMR (500 MHz, DMSO) δ 1H NMR (500 MHz, DMSO) δ 6.30 (s, 1H), 6.11 (s, 1H), 3.67 - 3.58 (m, 1H), 3.53 - 3.49 (m, 2H), 3.40 (t, J = 7.2 Hz, 2H), 2.49 (d, J = 7.1 Hz, 2H), 2.40 (t, J = 7.2 Hz, 2H), 2.29 (t, J = 7.4 Hz, 2H), 1.47 (p, J = 7.4 Hz, 2H), 1.40 (d, J = 2.9 Hz, 18H), 1.38 - 1.13 (m, 6H), 1.01 (d, J = 6.5 Hz, 2H), 0.96 (d, J = 6.4 Hz, 2H). 13C NMR (126 MHz, DMSO) δ 172.40, 171.23, 170.96, 163.25, 80.70, 80.33, 55.45, 50.68, 49.07, 43.91, 41.81, 41.14, 35.13, 33.94, 32.49, 29.99, 29.21, 28.18, 28.16, 26.19, 25.17, 23.77, 15.96. HRMS (ESI) m/z [M + H]+ calcd for C24H44N3O6, 470.32246; found 470.32270.


Synthesis of 3,3′-((6-(5-methyl-2-oxoimidazolidin-4-yl)hexanoyl)azanediyl)Dipropionic Acid



embedded image


Compound 2 (496 mg, 1.05 mmol) was treated with a trifluoroacetic acid (TFA) solution (90% in water) for 2 h at room temperature. The mixture was then concentrated under reduced pressure. The crude was purified by SPE Cartridges Chromabond C18, 6 mL/1000mg (30% MeOH in H2O) to yield 3 as a pale oil (292 mg, 78% yield).1H NMR (500 MHz, DMSO) δ 6.29 (s, 1H), 6.10 (s, 1H), 3.60 (ddt, J = 9.6, 6.3, 3.5 Hz, 1H), 3.52 (t, J = 7.4 Hz, 2H), 3.40 (t, J = 7.4 Hz, 2H), 2.53 - 2.51 (m, 1H), 2.47 (s, 1H), 2.40 (t, J = 7.3 Hz, 2H), 2.29 (t, J = 7.4 Hz, 2H), 1.47 (p, J = 7.4 Hz, 2H), 1.37 - 1.16 (m, 6H), 1.00 (d, J = 6.5 Hz, 1H), 0.96 (d, J = 6.4 Hz, 3H). 13C NMR (126 MHz, DMSO) δ 182.56, 182.22, 181.44, 172.32, 64.48, 59.71, 53.11, 51.00, 43.21, 41.97, 41.41, 39.00, 38.22, 35.19, 34.18, 32.78, 24.98. HRMS (ESI) m/z [M + H]+ calcd for C16H28N3O6, 358.19726; found 358.19680.


NHS Activation of 3,3′-((6-(5-methyl-2-oxoimidazolidin-4-yl)hexanoyl)azanediyl) Dipropionic Acid



embedded image


Compound 3 (292 mg, 0.82 mmol) and N-hydroxysuccinimide (190 mg, 1.64 mmol) were dissolved in dry DMF (5 mL) under a nitrogen atmosphere, followed by addition of 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDC·HCl) (289 mg, 1.64 mmol) dissolved in dry DMF (1 mL). The mixture was stirred for 24 h at room temperature. The solvent was evaporated in vacuo and the solid was dissolved in EtOAc (10 mL), which was then washed with sat. NaHCO3 (2 × 10 mL), 10% citric acid (2 × 10 mL) and brine (10 mL). The organic layer was dried using anhydrous MgSO4 and the solvent was evaporated in vacuo to yield 4 as a colourless oil (303 mg, 68%). Rf = 0.50 (10% MeOH in DCM). 1H NMR (500 MHz, DMSO) δ 6.28 (s, 1H), 6.10 (s, 1H), 3.70 (t, J = 6.9 Hz, 2H), 3.60 - 3.56 (m, 2H), 3.51 - 3.47 (m, 1H), 3.07 (t, J = 6.9 Hz, 2H), 2.93 (t, J = 7.2 Hz, 2H), 2.82 (s, 8H), 2.35 (t, J = 7.5 Hz, 2H), 1.49 (p, J = 7.5 Hz, 2H), 1.41 - 1.14 (m, 6H), 1.01 (d, J = 6.5 Hz, 1H), 0.96 (d, J = 6.4 Hz, 3H). 13C NMR (126 MHz, DMSO) δ 172.94, 170.61, 170.57, 168.01, 167.88, 163.26, 162.77, 55.46, 50.69, 43.56, 41.54, 41.14, 36.25, 32.47, 31.25, 30.54, 29.99, 29.39, 29.17, 26.22, 25.92, 25.01, 23.77, 15.97. HRMS (ESI) m/z [M + H]+ calcd for C24H34N5O10, 552.23002; found 552.22970.



FIGS. 1b-j show the synthesis and final structure of further crosslinkers 5-13 according to the solution.



FIG. 2 shows the SDS-PAGE resulting from crosslinking of Glutathione S-Transferase (GST) dimer with increasing amounts of STAGEcl (crosslinker 4). GST runs on SDS-PAGE as a 25 kDa monomer under denaturing conditions. Crosslinked dimer can be observed as a result of crosslinking and increasing the ratio crosslinker:protein results in increased formation of crosslinked GST dimer.


GST in 2 µg aliquots was crosslinked (0.33 µg/µL) in crosslinking buffer (20 mM HEPES, 20 mM NaCl, 5 mM MgCl2, pH 7.8) using different crosslinker:protein ratios (w/w): 0.15:1, 0.44:1, 1.3:1 and 4:1. Crosslinking was carried out for 1 h at room temperature, after which the reaction was quenched with 50 mM ABC, with incubation for 20 mins at room temperature. Crosslinked protein samples were separated by SDS-PAGE on a 1 mm thick NuPAGE 4-12% Bis-Tris SDS-PAGE gel, using MES running buffer and Coomassie blue stain.



FIG. 3A shows a crosslinked network resulting from STAGEcl crosslinking mass spectrometry analysis of human serum albumin (HSA) with crosslinker 4. The outer circular line represents the protein sequence. Links shown are at a 5% FDR level.


HSA (200 µg, 0.5 µg/µL) in crosslinking buffer (20 mM HEPES, 20 mM NaCl, 5 mM MgCl2, pH 7.8) was crosslinked using STAGEcl (crosslinker 4) in a crosslinker:protein (w/w) ratio of 1.64:1, for 1 h at room temperature. The reaction was then quenched with 50 mM ABC. Crosslinked protein was separated by SDS-PAGE on a 1.5 mm thick NuPAGE 4-12% Bis-Tris SDS-PAGE gel using MES running buffer and Coomassie blue stain.



FIG. 3B shows a high-resolution fragmentation spectrum of a matched STAGEcl (crosslinker 4) crosslinked peptide pair. The sequences of the matched crosslinked peptides, Peptide 1 (red, upper line) and Peptide 2 (black, lower line), are given by the single letter amino acid code. The crosslinking site (K-K) is represented by the solid black line between residues, connecting both peptide sequences. Crosslinked peptide pairs are matched by database search according to precursor m/z (obtained by an MS1 (survey) scan), and the combination of m/z fragments identified in the mass spectrometer following MS2 fragmentation. The fragment ions detected for each peptide are indicated by vertical lines between amino acid residues. Fragment ions belong to either a y-series (indicated by a top tick on the vertical line) or b-series (indicated by a bottom tick on the vertical line). The presented fragmentation spectrum shows the m/z of b- and y-ions identified according to the matched peptide sequence.



FIG. 3C shows links (indicated by red lines) between residue pairs (5% FDR) fitted to the solved x-ray crystal structure (PDB 1A06, cartoon representation in grey). The histogram shows the observed Ca-Ca distance distribution (in angstroms) for observed linked residue pairs (red), against the random distance distribution (grey) where all crosslinked distances are considered. STAGEcl has an estimated upper distance constraint for crosslinked K-K residues of 27 Å. The majority of crosslinked residues have Ca-Ca distance <20 Å, falling well within the expected distance distribution. The crosslinked residue pairs that exceed this estimated distance fall within the range of expected false positive matches according to the FDR estimation.



FIG. 3D shows a scheme of the protein crosslinking, digestion and enrichment process. Following the crosslinking reaction, the crosslinked proteins can be proteolytically digested, the resulting peptide mixture applied to solid-supported streptavidin in a PBS buffer, and crosslinked peptides bound to the solid-support with high specificity and efficiency. Linear modified peptides are then washed from the solid-support, and competitive elution with an excess of biotin is achieved, also in PBS buffer, pH 7.4. The resulting enriched crosslinked peptide pairs can then be either analysed directly by mass spectrometry, or subjected to further chromatographic fractionation, including (but not limited to) SCX, SEC and hSAX.



FIG. 3E shows the level of enrichment following the enrichment procedure on a STAGEcl (crosslinker 4) crosslinked E.coli lysate digest. The three columns show the relative intensity of matched MS1 precursors for non-crosslinker and crosslinker modified peptides (comprised of mono-linked peptides and crosslinked peptide pairs) at different stages of enrichment: (1) input to beads (without enrichment), (FT) the flow-through from beads during enrichment and (E) eluate from beads after enrichment. The bars representing the input (1) and flow-through (FT) were the result of the respective samples following an additional SEC fractionation. The bar representing the eluate (E) was the result of enrichment (affinity enrichment-SCX-SEC).


Cell Production

A single clone of E.coli K12 strain (BW25113, DSMZ, Germany) grown on agar plates was selected for inoculation of lysogeny broth (LB)-media. Fermentation was initiated in a Biostat A Plus Bioreactor (Sartorius, Göttingen, Germany) using a preculture aliquot in LB medium, with 0.5% (w/v) glucose, at 37° C. Growth was monitored frequently by taking optical density measurements at 600 nm. Fermentation was stopped at an optical density of 10 by rapidly cooling the culture in stirred ice water followed by biomass harvesting by centrifugation at 5000 g, 4° C. for 15 mins. Cell pellets were snap-frozen in liquid nitrogen and stored at -80° C.


Cell Lysis

Cell pellets were resuspended in ice-cold lysis buffer (50 mM HEPES, pH 7.4 at RT, 50 mM KCl, 50 mM NaCl, 1.5 mM MgCl2, 5% (v/v) glycerol, 1 mM dithiothreitol (DTT), a spatula tip of chicken egg white lysozyme (Sigma-Aldrich, St. Louis, MO, USA) and complete EDTA-free protease inhibitor cocktail (Roche, Basel, Switzerland)). Cells were lysed by sonication on ice at 30% amplitude, 30 s on/off for 10 cycles (total time 5 mins), using a Branson Digital Sonifier. After sonication, 125 units of Benzonase (Merck, Darmstadt, Germany) were added. Liquid was collected in a centrifuge tube (upper foaming with DNA proteins was discarded) and sample was clarified by centrifugation at 15,500 rpm for 30 mins at 4° C. DTT was added to 2 mM. The cleared lysate was then subjected to ultracentrifugation using a 70 Ti fixed-angle rotor for 1h at 106,000 g at 4° C., after which the supernatant was concentrated 10x using ultrafiltration with Amicon spin filters (15 kDa molecular weight cut-off) to achieve a total protein concentration of 10 mg/mL, determined by microBCA protein assay (Thermo Fisher Scientific, Waltham, MA, USA).


E.Coli Lysate Crosslinking

Cell lysate was diluted to 1 mg/mL protein concentration with crosslinking buffer (50 mM HEPES, 50 mM NaCl, 50 mM KCl, 1.5 mM MgCl2, 5% glycerol, pH 7.4). Cell lysate was then crosslinked with 1 mM STAGEcl (crosslinker 4) for 1 h at room temperature. After this time the crosslinking reaction was quenched with 50 mM ABC, with incubation for 30 mins on ice. Crosslinked proteins were acetone-precipitated overnight at -20° C. Protein was solubilised in 6 M urea, 2 M thiourea, 100 mM ABC. Protein sample was then subjected to proteolysis with LysC added at a 1:100 (m/m) ratio followed by incubation for 4 h at 37° C. After 1:5 dilution with 100 mM ABC, trypsin was added at a ratio of 1:25 (m/m) and the digestion allowed to continue for 16 h at 37° C., after which digestion was stopped with the addition of TFA to 1% (v/v). Digests were desalted using SPE cartridges according to the manufacturer’s instruction and eluates dried, aliquoted and stored at -20° C. until further use.

Claims
  • 1. A reversible streptavidin based analyte enrichment system for use in crosslinking mass spectrometry analysis, in particular for enriching at least parts of crosslinked peptides pairs in mass spectrometry analysis, comprising at least one crosslinking agent of the structure of general formulae (I) whereinR1 is selected from a functional group comprising ester, halogen, aldehyde, isothiocyanate, isocyanate, anhydride, epoxide, acetyl, glyoxal, triazine, hydrazine, disulfide, azide, ketone, phosphine, alkene, amine, N-heterocycle,a = 0-10, preferably 1-5, more preferably 2-3;Y is selected from —O—, —S—, —S—S—, —S(═O)—, —13CH2—, —CD2—, —18O—m, in particular —O—, —S—, —S—S—;n = 0-4, preferably 0, 1 or 2,X is selected from a group comprising —N—, —C6H3(NH—), —CH(NH—)—, wherein R2 is attached to N, —CH—, —CH(CO—NH—), wherein R2 is attached to C;R2 is one of the structures of formulae (IV) comprising biotin analogues or formulae (V) comprising or desthiobiotin analogues (V), wherein biotin is excluded: orwhereinX1, X2, X3 is selected from —NH2, —NH—, —N—Me, —N—Et, —O—.,R3 is selected from the group consisting of H, optionally substituted C1-C10 alkyl, optionally substituted C3-C10 cycloalkyl, optionally substituted C2-C10 alkenyl, optionally substituted C3-C10 cycloalkenyl, optionally substituted C2-C10 alkynyl, optionally substituted C2-C10 heteroalkyl, optionally substituted C3-C10 heterocycloalkyl, optionally substituted C2-C10 heteroalkenyl, optionally substituted C3-C10 heterocycloalkenyl, optionally substituted C2-C10 heteroalkynyl, optionally substituted C6-C14 aryl, optionally substituted C5-C14 heteroaryl;Z is selected from —CO—, aryl, preferably C5-C6 such as C6-C14 aryl, C5-C14 heteroaryl, alkyl, preferably C1-C4 alkyl, triazoles, alkenes, preferably C2-C4 alkenes, alkyl tetrazines, preferably C1-C4 tetrazines, optionally substituted —CO—NH—(CH2)b—, —CO—NH—(CH2)b—NH—CO—, —CO—NH—(CH2)b—CO—, wherein b = 1-10, preferably 2-6, optionally substituted —CO—NH—(CH2—O—)c—(CH2)d—CO—, CO—NH—(CH2—O—)c—Triazin—(CH2)d—CO—, —CO—NH—(CH2)b—(CH2—O—)c—(CH2)d—NH—CO—(CH2)e—CO— wherein b, c, d, e = 1-10, preferably 2-6.
  • 2. The enrichment system according to claim 1, wherein the crosslinking agent is of an asymmetrical structure (II) or a symmetrical structure (III) whereina = 1-10, preferably 2-5, more preferably 2-3.n = 1-4, preferably 1-2, more preferably 1.Y is selected from —O—, —S—, —S—S—, —S(═O)—, —13CH2—, —CD2—, —18O—.
  • 3. The enrichment system according to claim 1, wherein R1 is selected from a group comprising N-hydroxysuccinimide esters, imidoesters, isothiocyanates, isocyanates, acyl chlorides, sulfonyl chlorides, aryl sulfonyl fluorides, acyl azides, fluorophenyl esters, anhydrides, fluorobenzene, epoxides, alpha,beta-unsaturated aldehydes, 1,3-ketoaldehydes, 1,2,3-triazines, 1,2-cyclohexanedione, 2-methoxy-3-oxindoles, phenylglyoxal, a-keto-oximes, 2-fluoro-5-nitrotropolone, O-phthalaldehyde, maleimides, halo acetyls, pyridyl disulfides, aryl azides, diazirines, benzophenones, psoralens, phosphines, alkenes, cyclooctynes, tetrazines, hydrazines, alkoxyamines.
  • 4. The enrichment system according to claim 1, wherein R1 is a succinimide ester (NHS), phthalic-di-aldehyde, diazirine.
  • 5. The enrichment system according to claim 1, wherein X1, X2 are in each case NH and X3 is O, NH.
  • 6. The enrichment system according to claim 1, wherein R2 comprises a biotin derivative wherein the cyclic moiety of biotin different from that of biotin such as desthiobiotin, 2-iminobiotin, 3,4-diaminobiotin.
  • 7. The enrichment system according to claim 1, wherein R3 is H or —CH3.
  • 8. The enrichment system according to claim 1, wherein Z is — CO—, —CO—NH—(CH2)b—, —CO—NH—(CH2)b—NH—CO—, —CO—NH—(CH2)b—CO—, —CO—NH—(CH2—O—)c—(CH2)d—CO—, —CO—NH—(CH2—O—)c—Triazin—(CH2)d—CO—, —CO—NH—(CH2)b—(CH2—O—)c—(CH2)d—NH—CO—(CH2)e—CO— wherein independently from each other b, c, d, e = 1-8, preferably 2-6, more preferably 2-5.
  • 9. The enrichment system according to claim 1, wherein the crosslinking agent is of .
  • 10. (canceled)
  • 11. A method of enriching at least parts of crosslinked peptides pairs, in particular for use in crosslinking mass spectroscopy analysis, comprising the steps of: Providing a mixture of at least one crosslinking agent as defined in one of the claims 1-9 and at least one protein to be analysed,Digesting the protein by adding at least one proteolytic enzyme to obtain a peptide mixture of crosslinked peptides and linear peptides;Applying the peptide mixture to a streptavidin support whereby the crosslinked peptides pairs will be bound to the streptavidin support and linear peptides are washed out; andEluting the crosslinked peptides from the streptavidin support with an excess of biotin to obtain enriched crosslinked peptide pairs.
  • 12. The method according to claim 11, wherein the crosslinked peptides are eluted from the streptavidin support by using biotin contained in an buffer system with a pH between 6 and 8, in particular at a pH of 6.5 and 7.5.
  • 13. A kit comprising at least one crosslinking agent as defined in claim 1;Streptavidin attached to a solid support;optionally at least one proteolytic enzyme;a mobile phase / eluting agent.
Priority Claims (1)
Number Date Country Kind
20182274.9 Jun 2020 EP regional
CROSS-REFERENCE TO A RELATED APPLICATION

This application is a National Phase Patent Application of International Patent Application Number PCT/EP2021/066373, filed on Jun. 17, 2021, which claims priority of European Patent Application Number 20 182 274.9, filed on Jun. 25, 2020.

PCT Information
Filing Document Filing Date Country Kind
PCT/EP2021/066373 6/17/2021 WO