WO2024223837A1 - Dna-paint related materials and methods - Google Patents
Dna-paint related materials and methods Download PDFInfo
- Publication number
- WO2024223837A1 WO2024223837A1 PCT/EP2024/061562 EP2024061562W WO2024223837A1 WO 2024223837 A1 WO2024223837 A1 WO 2024223837A1 EP 2024061562 W EP2024061562 W EP 2024061562W WO 2024223837 A1 WO2024223837 A1 WO 2024223837A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- nucleic acid
- molecule
- acid sequence
- target
- imaging
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/68—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
- C12Q1/6813—Hybridisation assays
Definitions
- the present invention relates to the field of detection and quantification of targets, for example via PAINT, and single cell analysis, particularly spatial intermolecular single-cell omics.
- targets for example via PAINT
- single cell analysis particularly spatial intermolecular single-cell omics.
- documents including patent applications and manufacturer’s manuals are cited.
- the disclosure of these documents, while not considered relevant for the patentability of this invention, is herewith incorporated by reference in its entirety. More specifically, all referenced documents are incorporated by reference to the same extent as if each individual document was specifically and individually indicated to be incorporated by reference.
- spatial single-cell omics In what is generally known as “spatial single-cell omics”, there are many technologies, represented by multiple companies (e.g. Akoya, 10x Genomics), which aim at identifying proteins and nucleic acids, by direct hybridization of fluorophore-labeled oligonucleotides, or by enzymatic processes like rolling-circle amplification. However, these known technologies are concerned with cell-to-tissue context and use the single cell as the base unit.
- spatial intermolecular single-cell omics refers to the spatial context of targets within a single cell, i.e. the intermolecular spatial relationships within a single cell.
- the resolution of the known technologies mentioned above cannot be increased in a straightforward manner to reach intermolecular spatial information, due to basic effects like prohibitive probe size, diffusion limits, etc.
- the present invention provides an approach to spatial intermolecular single-cell omics based on fluorescence imaging.
- the fluorescence imaging technique and related molecules, compositions and kits comprise independent inventive aspects that may be used for other imaging applications than spatial intermolecular single-cell omics.
- Major progress has been made in the field of fluorescence imaging over the years.
- Super- resolution techniques, such as STED, STORM, PALM and PAINT were developed to overcome the diffraction limit of light microscopy, generally known to be approximated by Ernst Abbe’s formula.
- DNA-PAINT (with the sub-forms Exchange-PAINT, SPEED-PAINT and qPAINT) is a super-resolution technique that breaks the optical diffraction limit by temporally separating fluorescence signals from targets that are locally unresolvable in non-super-resolution fluorescence microscopy, e.g. confocal microscopy. The signals are localized timely separated, and the image is reconstructed from the localized data. This concept is also known as single-molecule localization microscopy, in short SMLM.
- SPEED-PAINT describes optimized sequences and buffer conditions for up to 100x speed increase compared to non-optimized sequences, meaning the acquisition time is drastically decreased.
- the drawback of SPEED-PAINT is its limitation to only a few (currently 6) suitable imager sequences and thus targets.
- An object of the present invention is to provide increased plexing capabilities in PAINT. Another object of the present invention is PAINT-related speed optimization. Another object of the present invention is to provide techniques and methods for spatial intermolecular single-cell omics.
- the present invention relates in a first aspect to a single-stranded nucleic acid molecule, comprising (a) a first nucleic acid sequence being capable of specifically hybridizing to a target complementary nucleic acid sequence, and (b) a second nucleic acid sequence that differs from the first nucleic acid sequence and is capable of transiently binding to a complementary nucleic acid sequence being labeled by an imaging molecule, wherein the first nucleic acid sequence is capable of stronger associating with its complementary nucleic acid sequence than the second nucleic acid sequence.
- the first nucleic acid sequence may be capable of stronger associating with its complementary nucleic acid sequence (e.g. via specific hybridization) than the second nucleic acid sequence under the same conditions (e.g.
- the term “associating” refers to binding strength between the first or second nucleic acid sequence, respectively, and its complementary nucleic acid sequence.
- the first nucleic acid sequence may be capable of stronger binding to its complementary nucleic acid sequence than the second nucleic acid sequence under the same conditions.
- nucleic acid sequence compositions can be selected by routine means.
- single-stranded nucleic acid molecule in accordance with the present invention may refer to single stranded DNA or RNA.
- DNA deoxyribonucleic acid
- DNA deoxyribonucleic acid
- RNA ribonucleic acid
- the nucleic acid molecule may also be modified by many means known in the art.
- Non-limiting examples of such modifications include methylation, "caps", substitution of one or more of the naturally occurring nucleotides with an analog, and internucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoroamidates, carbamates, etc.) and with charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.).
- Nucleic acid molecules in the following also referred as polynucleotides and/or oligonucleotides, may contain one or more additional covalently linked moieties, such as, for example, proteins (e.g., nucleases, toxins, antibodies, signal peptides, poly-L-lysine, etc.), intercalators (e.g., acridine, psoralen, etc.), chelators (e.g., metals, radioactive metals, iron, oxidative metals, etc.), and alkylators.
- the polynucleotides may be derivatized by formation of a methyl or ethyl phosphotriester or an alkyl phosphoramidate linkage.
- nucleic acid mimicking molecules known in the art such as synthetic or semi-synthetic derivatives of DNA or RNA and mixed polymers.
- nucleic acid mimicking molecules or nucleic acid derivatives according to the invention include phosphorothioate nucleic acid, phosphoramidate nucleic acid, 2’-O- methoxyethyl ribonucleic acid, morpholino nucleic acid, hexitol nucleic acid (HNA), peptide nucleic acid (PNA) and locked nucleic acid (LNA) (see Braasch and Corey, Chem Biol 2001, 8: 1).
- LNA is an RNA derivative in which the ribose ring is constrained by a methylene linkage between the 2’-oxygen and the 4’-carbon.
- nucleic acids containing modified bases for example thio-uracil, thio-guanine and fluoro-uracil.
- the target complementary nucleic acid sequence may be a portion of a target molecule to be detected, e.g., in a single cell. In this case, the single-stranded nucleic acid molecule may directly hybridize to the target molecule.
- the target complementary nucleic acid sequence may be a portion of a primary binder.
- the primary binder is a molecule that specifically binds the target molecule.
- the single-stranded nucleic acid molecule may indirectly bind to the target molecule via the primary binder.
- Hybridization as used herein is the process in which two complementary single-stranded nucleic acid molecules bind together to form a double-stranded molecule. The bonding is dependent on the appropriate base-pairing across the two single-stranded molecules. Whether or not two complementary single-stranded nucleic acid molecules bind together to form a double-stranded molecule also depends on the hybridization condition, that is the sum of environmental factors influencing hybridization. Examples of environmental factors that may affect hybridization include temperature, the concentration of one or more salts and pH. This is also referred to in the prior art as the “stringency” of hybridization.
- the stringency is determined by the hybridization temperature and the salt concentration in the hybridization buffer, whereby high temperature and low salt is more stringent as only perfectly matched hybrids will be stable.
- a pH that is too alkaline may cause the strands to separate; too acidic and they may be forced together.
- the length and the GC content of nucleic acid molecules influence hybridization. Generally, binding occurs under more stringent conditions for long nucleic acid molecules and nucleic acid molecules with a high GC content and binding occurs under less stringent conditions for short nucleic acid molecules and/or nucleic acid molecules with a high AT content.
- the determination of ideal hybridization conditions for two given single-stranded nucleic acid molecules to bind together to form a double-stranded molecule is a matter of routine in the field of molecular biology.
- the ideal hybridization conditions are estimated from the calculation of the melting temperature (Tm) of the double-stranded molecule. At the Tm, half of the sequence is double stranded and half of the sequence is single stranded.
- Tm melting temperature
- the Tm for short probes 14 – 20 base pairs
- Tm 4°C x number of G/C pairs + 2°C x number of A/T pairs
- the hybridization temperature (annealing temp) of oligonucleotide probes is generally approximately 5°C below the melting temperature.
- the requirement that the first nucleic acid sequence is capable of specifically hybridizing to a target complementary nucleic acid sequence means that under the selected or envisioned hybridization conditions the first nucleic acid sequence only hybridizes to the target complementary nucleic acid sequence and to no other nucleic acid molecule that might be present, for example, in a sample.
- the requirement that the second nucleic acid sequence is capable of transiently binding to a complementary nucleic acid sequence means that under the selected or envisioned hybridization conditions the second nucleic acid sequence binds together with the complementary nucleic acid sequence to form a double-stranded molecule transiently, whereby transiently means only momentarily or briefly.
- a binding interaction with a mean duration of shorter than 30 seconds is deemed transient, while a duration of longer than 1 hour is deemed stable.
- the intermediate duration between 30 seconds and 1 hours may be designated “semi-stable”.
- DNA hybridization and de-hybridization is a first order chemical reaction and therefore the binding duration follows an exponential distribution.
- the values for the binding duration given above are the mean values of the distribution. It follows for the above that in the context of the first aspect of the invention the first nucleic acid sequence is capable of stronger associating with its complementary nucleic acid sequence (i.e. via specific hybridization) than the second nucleic acid sequence (i.e. via transient binding only).
- the complementary nucleic acid sequence is labeled by an imaging molecule.
- labeled means that the imaging molecule is connected or linked to the complementary nucleic acid sequence, preferably via covalent chemical bonds.
- the first nucleic acid sequence is capable of stably hybridizing to its target complementary nucleic sequence. “Stably hybridizing” in the context of the present invention has to be held distinct from the above described “transient binding”.
- “Stably hybridizing” means that under the selected or envisioned hybridization conditions the first nucleic acid sequence hybridizes to the target complementary nucleic acid sequence thereby forming a double-stranded molecule that remains stably in the double-stranded form and does not dissociate again into single-strands.
- the present invention relates in a second aspect to a hybridization complex, wherein the nucleic acid molecule is specifically hybridized to a target complementary nucleic acid sequence.
- the first nucleic acid sequence of the single stranded nucleic acid molecule hybridizes to a target complementary nucleic acid sequence thereby forming a double-stranded region within the nucleic acid molecule.
- the target nucleic acid sequence is complementary to the first nucleic acid sequence. This means that it comprises or consists of a nucleic acid sequence that is complementary to the first nucleic acid sequence.
- This complementary nucleic acid sequence may have a length of 6nt to 150nt, preferably 10nt to 50 nt, more preferably 12 nt to 30 nt.
- the target nucleic acid sequence is preferably an exogenous nucleic acid sequence, for example, an in vitro synthesized or produced nucleic acid sequence. However, the target nucleic acid sequence may also be or be a part of a naturally occurring nucleic acid sequence, such as mRNA.
- the target complementary nucleic acid sequence is conjugated to a binding molecule, wherein the binding molecule is preferably a nucleic acid sequence, a small molecule or a protein, wherein the protein is preferably an antibody, antibody mimetic, or aptamer.
- a binding molecule is a compound being capable of binding to a target molecule.
- the binding molecule preferably specifically binds to the target molecule. Specific binding designates that the binding molecule does not or essentially does not bind to other molecules, e.g. in the context of a sample, than the target molecule.
- the target molecule of the binding molecule may be any kind of molecule that is potentially present in a sample, e.g.
- the sample can be, for example, a cellular sample or a body sample, such as a body liquid or tissue sample.
- the cellular sample may be from cultured cells or may have been obtained from a subject, e.g. via a biopsy.
- the body liquid is preferably a blood sample (whole blood, serum or plasma).
- the binding molecule is a nucleic acid sequence it may bind to a target nucleic acid via hybridization.
- the binding of the binding molecule to the target molecule may be directly or indirectly, wherein indirectly means that there may be one or more further binding partners between the binding molecule and the target molecule.
- indirectly means that there may be one or more further binding partners between the binding molecule and the target molecule.
- the binding molecule is an antibody the antibody might be a biotin labelled antibody that binds to a streptavidin labelled antibody and the streptavidin labelled antibody binds to the target molecule.
- the "small molecule” as used herein is preferably an organic molecule.
- Organic molecules relate or belong to the class of chemical compounds having a carbon basis, the carbon atoms linked together by carbon-carbon bonds.
- Organic compounds can be natural or synthetic.
- the organic molecule is preferably an aromatic molecule and more preferably a heteroaromatic molecule.
- aromaticity is used to describe a cyclic (ring-shaped), planar (flat) molecule with a ring of resonance bonds that exhibits more stability than other geometric or connective arrangements with the same set of atoms.
- Aromatic molecules are very stable, and do not break apart easily to react with other substances.
- a heteroaromatic molecule at least one of the atoms in the aromatic ring is an atom other than carbon, e.g.
- the molecular weight is preferably in the range of 200 Da to 1500 Da and more preferably in the range of 300 Da to 1000 Da.
- the "small molecule" in accordance with the present invention may be an inorganic compound. Inorganic compounds are derived from mineral sources and include all compounds without carbon atoms (except carbon dioxide, carbon monoxide and carbonates). Preferably, the small molecule has a molecular weight of less than about 2000 Da, or less than about 1000 Da such as less than about 500 Da, and even more preferably less than about 200 Da, or amu.
- the size of a small molecule can be determined by methods well-known in the art, e.g., mass spectrometry.
- the small molecules may be designed, for example, based on the crystal structure of the target molecule, where sites presumably responsible for the biological activity can be identified and verified in in vivo assays such as in vivo high-throughput screening (HTS) assays.
- the term “protein” as used herein is interchangeably with the term “polypeptide” and describes linear molecular chains of amino acids, including single chain proteins or their fragments. The protein may also be a peptide.
- the term “peptide” as used herein describes a group of molecules consisting of up to 49 amino acids, whereas the term “polypeptide” (also referred to as "protein”) as used herein preferably describes a group of molecules consisting of at least 50 amino acids.
- (poly)peptides may further form oligomers consisting of at least two identical or different molecules.
- the corresponding higher order structures of such multimers are, correspondingly, termed homo- or heterodimers, homo- or heterotrimers etc..
- antibody as used in accordance with the present invention comprises, for example, polyclonal or monoclonal antibodies. Furthermore, also derivatives or fragments thereof, which still retain the binding specificity to the target are comprised in the term "antibody”.
- Antibody fragments or derivatives comprise, inter alia, Fab or Fab’ fragments, Fd, F(ab')2, Fv or scFv fragments, single domain VH or V-like domains, such as VhH or V-NAR-domains, as well as multimeric formats such as minibodies, nanobodies, diabodies, tribodies or triplebodies, tetrabodies or chemically conjugated Fab’-multimers (see, for example, Harlow and Lane “Antibodies, A Laboratory Manual”, Cold Spring Harbor Laboratory Press, 198; Harlow and Lane “Using Antibodies: A Laboratory Manual” Cold Spring Harbor Laboratory Press, 1999; Altshuler EP, Serebryanaya DV, Katrukha AG.2010, Biochemistry (Mosc)., vol.
- the multimeric formats in particular comprise bispecific antibodies that can simultaneously bind to two different types of antigen.
- the first antigen can be found on a protein of interest.
- the second antigen may, for example, be a tumor marker that is specifically expressed on cancer cells or a certain type of cancer cells.
- Bispecific antibodies formats are Biclonics (bispecific, full length human IgG antibodies), DART (Dual-affinity Re- targeting Antibody) and BiTE (consisting of two single-chain variable fragments (scFvs) of different antibodies) molecules (Kontermann and Brinkmann (2015), Drug Discovery Today, 20(7):838-847).
- antibody also includes embodiments such as chimeric (human constant domain, non-human variable domain), single chain and humanised (human antibody with the exception of non-human CDRs) antibodies.
- chimeric human constant domain, non-human variable domain
- single chain humanised (human antibody with the exception of non-human CDRs) antibodies.
- Various techniques for the production of antibodies are well known in the art and described, e.g. in Harlow and Lane (1988) and (1999) and Altshuler et al., 2010, loc. cit.
- polyclonal antibodies can be obtained from the blood of an animal following immunisation with an antigen in mixture with additives and adjuvants and monoclonal antibodies can be produced by any technique which provides antibodies produced by continuous cell line cultures. Examples for such techniques are described, e.g.
- Harlow E and Lane D Cold Spring Harbor Laboratory Press, 1988; Harlow E and Lane D, Using Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, 1999 and include the hybridoma technique originally described by Köhler and Milstein, 1975, the trioma technique, the human B-cell hybridoma technique (see e.g. Kozbor D, 1983, Immunology Today, vol.4, 7; Li J, et al.2006, PNAS, vol.103(10), 3557) and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole et al., 1985, Alan R. Liss, Inc, 77-96).
- recombinant antibodies may be obtained from monoclonal antibodies or can be prepared de novo using various display methods such as phage, ribosomal, mRNA, or cell display.
- a suitable system for the expression of the recombinant (humanised) antibodies may be selected from, for example, bacteria, yeast, insects, mammalian cell lines or transgenic animals or plants (see, e.g., US patent 6,080,560; Holliger P, Hudson PJ.2005, Nat Biotechnol., vol.23(9), 11265).
- techniques described for the production of single chain antibodies see, inter alia, US Patent 4,946,778) can be adapted to produce single chain antibodies specific for an epitope of a target.
- antibody mimetics refers to compounds which, like antibodies, can specifically bind antigens, but which are not structurally related to antibodies. Antibody mimetics are usually artificial peptides or proteins with a molar mass of about 3 to 20 kDa.
- an antibody mimetic may be selected from the group consisting of affibodies, adnectins, anticalins, DARPins, avimers, nanofitins, affilins, Kunitz domain peptides, Fynomers®, trispecific binding molecules and prododies.
- affibody refers to a family of antibody mimetics which is derived from the Z-domain of staphylococcal protein A. Structurally, affibody molecules are based on a three-helix bundle domain which can also be incorporated into fusion proteins. In itself, an affibody has a molecular mass of around 6kDa and is stable at high temperatures and under acidic or alkaline conditions. Target specificity is obtained by randomisation of 13 amino acids located in two alpha-helices involved in the binding activity of the parent protein domain (Feldwisch J, Tolmachev V.; (2012) Methods Mol Biol.899:103-26).
- adnectin (also referred to as “monobody”), as used herein, relates to a molecule based on the 10th extracellular domain of human fibronectin III (10Fn3), which adopts an Ig- like ⁇ -sandwich fold of 94 residues with 2 to 3 exposed loops, but lacks the central disulphide bridge (Gebauer and Skerra (2009) Curr Opinion in Chemical Biology 13:245-255).
- Adnectins with the desired target specificity can be genetically engineered by introducing modifications in specific loops of the protein.
- the term “anticalin”, as used herein, refers to an engineered protein derived from a lipocalin (Beste G, Schmidt FS, Stibora T, Skerra A.
- Anticalins possess an eight-stranded ⁇ -barrel which forms a highly conserved core unit among the lipocalins and naturally forms binding sites for ligands by means of four structurally variable loops at the open end.
- Anticalins although not homologous to the IgG superfamily, show features that so far have been considered typical for the binding sites of antibodies: (i) high structural plasticity as a consequence of sequence variation and (ii) elevated conformational flexibility, allowing induced fit to targets with differing shape.
- DARPin refers to a designed ankyrin repeat domain (166 residues), which provides a rigid interface arising from typically three repeated ⁇ -turns. DARPins usually carry three repeats corresponding to an artificial consensus sequence, wherein six positions per repeat are randomised. Consequently, DARPins lack structural flexibility (Gebauer and Skerra, 2009).
- avimer refers to a class of antibody mimetics which consist of two or more peptide sequences of 30 to 35 amino acids each, which are derived from A-domains of various membrane receptors and which are connected by linker peptides.
- binding of target molecules occurs via the A-domain and domains with the desired binding specificity can be selected, for example, by phage display techniques.
- the binding specificity of the different A- domains contained in an avimer may, but does not have to be identical (Weidle UH, et al., (2013), Cancer Genomics Proteomics; 10(4):155-68).
- a “nanofitin” (also known as affitin) is an antibody mimetic protein that is derived from the DNA binding protein Sac7d of Sulfolobus acidocaldarius.
- Nanofitins usually have a molecular weight of around 7kDa and are designed to specifically bind a target molecule, by randomising the amino acids on the binding surface (Mouratou B, Béhar G, Paillard-Laurance L, Colinet S, Pecorari F., (2012) Methods Mol Biol.; 805:315-31).
- the term “affilin”, as used herein, refers to antibody mimetics that are developed by using either gamma-B crystalline or ubiquitin as a scaffold and modifying amino-acids on the surface of these proteins by random mutagenesis. Selection of affilins with the desired target specificity is effected, for example, by phage display or ribosome display techniques.
- affilins have a molecular weight of approximately 10 or 20kDa.
- the term affilin also refers to di- or multimerised forms of affilins (Weidle UH, et al., (2013), Cancer Genomics Proteomics; 10(4):155-68).
- a “Kunitz domain peptide” is derived from the Kunitz domain of a Kunitz-type protease inhibitor such as bovine pancreatic trypsin inhibitor (BPTI), amyloid precursor protein (APP) or tissue factor pathway inhibitor (TFPI).
- BPTI bovine pancreatic trypsin inhibitor
- APP amyloid precursor protein
- TFPI tissue factor pathway inhibitor
- Kunitz domains have a molecular weight of approximately 6kDA and domains with the required target specificity can be selected by display techniques such as phage display (Weidle et al., (2013), Cancer Genomics Proteomics; 10(4):155-68).
- the term "Fynomer®” refers to a non-immunoglobulin-derived binding polypeptide derived from the human Fyn SH3 domain. Fyn SH3-derived polypeptides are well-known in the art and have been described e.g. in Grabulovski et al. (2007) JBC, 282, p.
- the target complementary nucleic acid sequence may be conjugated to the binding molecule in any suitable manner.
- the conjugation between the target complementary nucleic acid sequence and the binding molecule can be covalent or non-covalent.
- the target complementary nucleic acid sequence is conjugated to the binding molecule via a linker, wherein the linker preferably comprises biotin and one of avidin or streptavidin.
- the target complementary nucleic acid sequence is covalently coupled to the binding molecule via NHS- chemistry or site-specific labeling via click chemistry.
- the specific hybridization between the first nucleic acid sequence and its target complementary nucleic acid sequence is based on the formation of more hydrogen bonds than the transient binding between the second nucleic acid sequence and the complementary nucleic acid sequence being labeled by an imaging molecule.
- a nucleic acid molecule labeled by an imaging molecule, which transiently binds another nucleic acid molecule is known as an ‘imager’ or ‘imager strand’.
- the ‘imager’ or ‘imager strand’ preferably comprises or consists of a complementary nucleotide sequence having a length of 5 to 13 nucleotides that carries the imaging molecule. Any combination of suitable melting temperatures (Tm) that provide for the appropriate difference in association strength of the first nucleic acid sequence with its complementary nucleic acid sequence on the one hand and the second nucleic acid sequence with the complementary nucleic acid sequence being labeled by an imaging molecule on the other hand is contemplated.
- Tm melting temperatures
- the specific hybridization between the first nucleic acid sequence and its target complementary nucleic acid sequence has a melting temperature of between 25°C and 90°C, preferably between 45°C and 85°C and most preferably between 62°C and 78°C.
- the transient binding between the second nucleic acid sequence and the complementary nucleic acid sequence being labeled by an imaging molecule may have a melting temperature of between 8°C and 22°C, preferably 12°C to 18°C and most preferably 14°C and 16° C.
- the specific hybridization between the first nucleic acid sequence and its target complementary nucleic acid sequence may have a melting temperature of between 45°C and 85°C and the transient binding between the second nucleic acid sequence and the complementary nucleic acid sequence being labeled by an imaging molecule may have a melting temperature of 12°C to 18°C.
- the specific hybridization between the first nucleic acid sequence and its target complementary nucleic acid sequence may have a melting temperature of between 62°C and 78°C and the transient binding between the second nucleic acid sequence and the complementary nucleic acid sequence being labeled by an imaging molecule may have a melting temperature of 14°C to 16°C.
- the imaging molecule may be of any suitable type.
- the imaging molecule is a fluorescent molecule.
- Preferred fluorescent molecules are fluorescent proteins or fluorescent dyes.
- the fluorescent dye is preferably a component selected from Atto, Alexa Fluor or Cy dyes.
- the fluorescent protein is preferably GFP or YFP.
- other detectable types of imaging molecules may also be used, for example a radionuclide.
- the radionuclide is preferably either selected from the group of gamma-emitting isotopes, more preferably 99m Tc, 123 I, 111 In, and/or from the group of positron emitters, more preferably 18 F, 64 Cu, 68 Ga, 86 Y, 124 I, and/or from the group of beta-emitter, more preferably 131 I, 90 Y, 177 Lu, 67 Cu, 90 Sr, or from the group of alpha-emitter, preferably 213 Bi, 211 At.
- the nucleic acid sequence being labeled by an imaging molecule may have any suitable length that is suitable to achieve the proviso that the first nucleic acid sequence is capable of stronger associating with its complementary nucleic acid sequence than the second nucleic acid sequence.
- the nucleic acid sequence being labeled by an imaging molecule has a length of 4 to 10 nucleotides. This short length is particularly advantageous for providing achieving the transient binding between the second nucleic acid sequence and the complementary nucleic acid sequence being labeled by an imaging molecule.
- the first nucleic acid sequence may have any suitable length that is suitable to achieve the proviso that the first nucleic acid sequence is capable of stronger associating with its complementary nucleic acid sequence than the second nucleic acid sequence.
- the first nucleic acid sequence may have a length of 4 to 30 nucleotides, preferably 16 to 24 nucleotides. This length is longer than the short length nucleic acid sequence being labeled by an imaging molecule and is particularly advantageous for providing for achieving the specific, preferably stable hybridization of the first nucleic acid sequence to the target complementary nucleic acid sequence.
- the first nucleic acid sequence may have a GC-content of 45%- 55%, preferably 50%. Both the length and the GC-content of the first nucleic acid sequence may be used, individually or in combination, to advantageously provide for the specific, preferably stable hybridization between the first nucleic acid sequence and its target complementary nucleic acid sequence. As discussed above, the binding strength may thus be adjusted as appropriate, in order to ensure that the first nucleic acid sequence is capable of stronger associating with its complementary nucleic acid sequence (i.e. via specific hybridization) than the second nucleic acid sequence (i.e. via transient binding only). As discussed above, appropriate hybridization conditions and nucleic acid sequence compositions can be selected by routine means.
- the second nucleic acid sequence of the single-stranded nucleic acid molecule may have any suitable length, for example a length of 4 to 10 nucleotides for the use of traditional DNA- PAINT probes, or 8-36 nucleotides for the use with speed-optimized DNA-PAINT probes.
- the nucleic acid molecule further comprises a toehold seed, whereby the specific hybridization between the first nucleic acid sequence and its complementary target nucleic acid sequence can be disconnected via toehold mediated strand displacement.
- Toehold mediated strand displacement is generally known in the art; see, for example, Yurke et al.
- protector strand that is hybridized to a complementary nucleic acid sequence (called original strand), with another nucleic acid strand (called invading strand).
- the original strand comprises an overhanging region (called toehold) that is not hybridized to the protector strand.
- the invading strand is complementary to the original strand, including the toehold.
- the invading strand first binds to the toehold. Branch migration of the invading strand causes the replacement of the protector strand.
- an invading strand may be added to the hybridization complex in which the nucleic acid molecule is specifically hybridized to the target complementary nucleic acid sequence, but in which the nucleic acid molecule has an overhanging toehold that is not hybridized to the target complementary nucleic acid sequence.
- the invading strand may replace the target complementary nucleic acid sequence, i.e. may hybridize to the nucleic acid molecule instead of the target complementary nucleic acid sequence.
- the nucleic acid molecule may be separated from the target complementary nucleic acid sequence. This enables, i.a. removing the nucleic acid molecule from a sample, e.g. by washing it out.
- the present invention relates in a third aspect to a plurality of nucleic acid molecules of the first aspect of the invention or hybridization complexes of the second aspect of the invention, wherein the nucleic acid molecules comprise: different first nucleic acid sequences that differ from each other in that they are capable of specifically and stably hybridizing to different target complementary nucleic acid sequences; and/or different second nucleic acid sequences that differ from each other in that they are capable of transiently binding to different complementary nucleic acid sequences optionally being labeled by at least two, at least three, at least four, or at least five different imaging molecules, wherein preferably each of the different target complementary nucleic acid sequences forms a cognate pair with a different imaging molecule.
- each target complementary nucleic acid can be distinguished from all other target complementary nucleic acid sequences in the plurality of target complementary nucleic acid sequences by a distinct imaging molecule when, both, the imaging molecule and target complementary nucleic acid sequence are bound to the nucleic acid molecule or the hybridization complex of the invention.
- each target complementary nucleic acid sequence can be identified by a different imaging molecule.
- the different complementary nucleic acid sequences may be structurally different and/or the different target complementary nucleic acid sequences may be structurally different.
- the second nucleic acid sequences may be capable of transiently binding to the same type of complementary nucleic acid sequences.
- the first nucleic acid sequences may be capable of specifically and stably binding to the same type of target complementary nucleic acid sequences.
- the second nucleic acid sequences and preferably the nucleic acid molecules may be orthogonal.
- orthogonal is used herein for two or more entities that are sufficiently different so that two corresponding binding partners specifically bind to them, i.e. without binding among each other or to the one or more non-corresponding entities.
- Orthogonal is also used herein for two or more entities that are sufficiently different to specifically bind to two corresponding binding partners, i.e. without binding among each other or to the one or more non-corresponding binding partners.
- specificity it may also be used as a complementary term to specificity.
- the second nucleic acid sequences comprise or consist of sequences being selected from (TCC)n, (ACC)n, (CTT)n, (AAC)n, (CT)n, and (AC)n, wherein n is 4 to 12.
- sequences (TCC)n, (ACC)n, (CTT)n, (AAC)n, (CT)n, and (AC)n are known as so-called SPEED-sequences known from the SPEED-PAINT technique; see Strauss and Jungmann (2020), Nature Methods, 17:789–791.
- SPEED-sequences allow a design of tunable hybridization kinetics and demonstrated up to 100-fold faster imaging compared to classical DNA-PAINT. Besides being faster these sequences enable imaging at lower imager concentration, leading to reduced background and thus increased signal-to-noise. Since 6 SPEED sequences are available 6-plex experiments can be designed for multiplexing.
- the present invention relates in a fourth aspect to a kit or composition
- a kit or composition comprising (a) the nucleic acid molecule or the hybridization complex or the plurality of nucleic acid molecules or hybridization complexes of the above aspects of the invention and at least one complementary nucleic acid sequence being labeled by an imaging molecule (also termed herein type A kit or composition); or (b) one or more single-stranded nucleic acid molecules comprising a first nucleic acid sequence being capable of specifically hybridizing to a target complementary nucleic acid sequence and a second nucleic acid sequence that differs from the first nucleic acid sequence and is capable of transiently binding to a complementary nucleic acid sequence being labeled by an imaging molecule, one or more target complementary nucleic acid sequences capable of specifically hybridizing to the first nucleic acid sequence of the one or more single-stranded nucleic acid molecules, and one or more complementary nucleic acid sequences being labeled by an imaging molecule capable of transiently binding to the second nu
- the one or more single stranded nucleic acid molecules may have any of the properties described herein in relation to single-stranded nucleic acid molecules, with the proviso that the first nucleic acid sequence may be capable of reversibly, preferably transiently, binding to a target complementary nucleic acid sequence.
- the type B kit or composition is not necessarily limited by the proviso that the first nucleic acid sequence is capable of stronger associating with its complementary nucleic acid sequence than the second nucleic acid sequence.
- a desired higher amount of single- stranded nucleic acid sequences being bound to the target complementary nucleic acid sequence may be implemented by selecting the higher appropriate amount of single-stranded nucleic acid molecules compared to the amount of complementary nucleic acid sequence being labeled by an imaging molecule, taking into account the particular binding kinetics of all three types of molecules.
- the excess of single-stranded nucleic acid molecules as compared to complementary nucleic acid sequences being labeled by an imaging molecule ensures that more first nucleic acid sequences are hybridized to their target complementary nucleic acid sequences than second nucleic acid sequences being bound to their complementary nucleic acid sequences being labeled by an imaging molecule.
- the excess amount in connection with the type B kit or composition the essentially same technical effect is achieved as by the proviso that first nucleic acid sequence is capable of stronger associating with its complementary nucleic acid sequence than the second nucleic acid sequence in connection with the type A kit or composition.
- Two or more complementary nucleic acid sequences being labeled by an imaging molecule may have different imaging molecules, particularly imaging molecules that may be distinguished in the detection technique.
- the fluorescent imaging molecules may have different excitation/emission-spectra that are distinguishable in fluorescence microscopy.
- the present invention also relates in a fifth aspect to a method of detecting a target molecule in a sample, comprising: a. contacting the sample with the nucleic acid molecule or the kit or composition of the aspects of the invention as described above; b.
- the sample optionally contacting the sample with a target complementary nucleic acid sequence under condition wherein it specifically hybridizes to the first nucleic acid sequence of the nucleic acid molecule or composition of (a), wherein the target complementary nucleic acid sequence is conjugated to a binding molecule that specifically binds the target molecule in the sample; c. contacting the sample with a complementary nucleic acid sequence being labeled by an imaging molecule under conditions wherein it transiently binds to the second nucleic acid sequence of the nucleic acid molecule or composition of (a); and d. detecting the imaging molecule in the sample, thereby detecting the biological target molecule in the sample.
- the order of method steps may be any order unless explicitly stated otherwise.
- steps a), b) and c) of the above method may be any order. It may be advantageous to have the order step b) before step a), and optionally step d) after step c) after step a). It may also be advantageous to have the steps in alphabetical order.
- the target molecule in the sample is a nucleic acid sequence in the sample and the binding molecule is a nucleic acid being complementary to the nucleic acid sequence in the sample
- the target molecule in the sample is a protein or peptide or polysaccharide in the sample and the binding molecule is a small molecule or a protein, wherein the protein is preferably an antibody, antibody mimetic, or aptamer, specifically binding to the protein or peptide or polysaccharide in the sample
- the target molecule in the sample is a nucleic acid sequence comprising a/the target complementary nucleic acid sequence .
- Method step b) may not be required for cases in which the single-stranded nucleic acid molecules directly bind to target molecules, i.e. in which the one or more target complementary nucleic acids are a part of the one or more target molecules.
- the present invention relates in a sixth aspect to a method of detecting two or more target molecules in a sample, comprising a. contacting the sample with a first nucleic acid molecule or composition of any preceding claim and a second nucleic acid molecule or composition of any preceding claim; b.
- the method is carried out sequentially, wherein the sample is first contacted with the first nucleic acid molecule or the composition, the first target complementary nucleic acid sequence, and the first complementary nucleic acid sequence being labeled by an imaging molecule and the imaging molecule is detected, thereby detecting the first target molecule in the sample, and then the sample is contacted with the second nucleic acid molecule or the composition, the second target complementary nucleic acid sequence, and the second complementary nucleic acid sequence being labeled by an imaging molecule and the imaging molecule is detected, thereby detecting the second biological target molecule in the sample, wherein preferably the first complementary nucleic acid sequence being labeled by an imaging molecule is removed before the second complementary nucleic acid sequence being labeled by an imaging molecule is added; (ii) the imaging molecule of the first complementary nucleic acid sequence being labeled by an imaging molecule is different from the imaging molecule of the
- the two or more target molecules in a sample are at least 10 target molecules and the method comprises in step (a) at least 10 different nucleic acid molecules or compositions, in step (b) at least 10 different target complementary nucleic acid sequences, and optionally in step (c) at least 10 different first complementary nucleic acid sequences being labeled by an imaging molecule; preferably at least 30 target molecules and the method comprises in step (a) at least 30 different nucleic acid molecules or compositions, in step (b) at least 30 different target complementary nucleic acid sequences, and optionally in step (c) at least 30 different first complementary nucleic acid sequences being labeled by an imaging molecule; more preferably at least 60 target molecules and the method comprises in step (a) at least 60 different nucleic acid molecules or compositions, in step (b) at least 60 different target complementary nucleic acid sequences, and optionally in step (c) at least 60 different first complementary nucleic acid sequences being labeled by an imaging molecule
- the method is particularly useful for a large number of different target molecules in the sample.
- different target molecules may mean different types of target molecules.
- the at least 10, at least 30, at least 60 or at least 100 target molecules in the sample are distinguished from each other in step (d) by one or more of method options (i) to (iii), preferably option (iii) as described in the above preferred embodiment and further comprising e. mapping the localization of the at least 10, at least 30, at least 60 or at least 100 target molecules based on the localization of the at least 10, at least 30, at least 60 or at least 100 target molecules within the sample, preferably within cells of the sample; and f.
- the methods according to the invention may include labeling all targets with the same type of imager and thus quantifying the number of targets in a localization cluster.
- the codebook may be adaptively provided in response to the number of unresolved target molecules. This may be advantageous for handling cases in which two or more target molecules are so close to each other that assigning the sequential imager binding in the different imaging rounds to the target molecules individually is impossible –only the sum of the two identification sequences will be detected. Hence, there is no way of correct identification.
- a solution is to first determine the largest number of target molecules that cannot be separated in localization, and adapt the codebook accordingly, such that one can still tell the identity of the target molecules present.
- the adaption of the codebook is combinatorics that is generally known in the art.
- one option is to target all target complementary nucleic acids with a single stranded nucleic acid which reversible bind the same imager and use qPAINT as known in the art to determine the number of target complementary nucleic acids in each localization cluster (as described in the section methods/image analysis below), and create a codebook that distinguishes all combinations of target complementary single stranded nucleic acids up to the maximum number of molecules per localization cluster if the analysis is done beforehand, or if the analysis of the number of target complementary single stranded nucleic acids is done after the planned experiment, by analyzing the potentially confused or wrongly assigned codebook entries, and adding rounds of single stranded nucleic acid addition and imaging, thus extending the codes, to disambiguate the analysis.
- targets present may be one of the tuples (2, 3), (2, 4), (3, 4), (4, 4). Therefore, to disambiguate the results, two rounds may be added to identify targets 2-4, to make the full codes for example 1: ‘10000’, 2: ‘01010’, 3: ‘00101’, 4: ‘01100’.
- the present invention also relates to a method including method step iii) related to the codebook including: (v) if applicable: removing the single-stranded nucleic acid molecules from a previous round of the following steps from the target molecules, preferably using buffer conditions, toehold-mediated strand displacement, or heat; (iv) contacting the sample with all single-stranded nucleic acid molecules assigned to one of the N positions of all identification sequences and the complementary nucleic acid sequence(s) being labeled by an imaging molecule; (iiv) identifying and localizing the imaging molecules in the sample; (iiiv) repeating step (v), (iv) and (iiv) for all other N-1 identification sequence positions.
- (ix) for one or more locations in the sample generating a detection sequence having a length of N positions corresponding to the N positions of the identification sequence, wherein each position of the detection sequence is either assigned with an identified imaging molecule of the corresponding round of steps (iv) and (iiv) or, if no imaging molecule was detected, with a gap; (x) for each location of step (ix): by comparing the detection sequence of the location with the identification sequences of the codebook of step c), identifying the target type detected at the location.
- the second nucleic acid sequences of the different single-stranded nucleic acid sequences are orthogonal.
- detecting the imaging molecule(s) and thus imager(s) in the sample may include performing detection steps of DNA-PAINT (Schnitzbauer et al. (2017), Nature Protocols, 12:1198–1228).
- DNA-PAINT the transient association of the fluorophore to a target molecule is mediated by the pairing of short ( ⁇ 10 nucleotides) complementary DNA sequences:
- a docking strand comprising a nucleic acid sequence is coupled to the target molecule, usually through an antibody, nanobody, aptamer or other high affinity probe and an imager strand carries a fluorophore (i.e. the imaging molecule).
- the imaging strand carrying the fluorophore is free to diffuse in the imaging buffer.
- DNA hybridization i.e. binding of the imaging strand to the docking strand
- the fluorophore is transiently immobilized near the target molecule, and thus excited by the laser light, typically in Total Internal Reflection Fluorescence (TIRF) or highly inclined and laminated optical sheet (HiLO) configuration, however light sheet and spinning disk microscopies have been used as well.
- TIRF Total Internal Reflection Fluorescence
- HiLO highly inclined and laminated optical sheet
- the emitted light can then be captured by the camera as a diffraction limited flash.
- any of the nucleic acid probes described above may comprise two, three, four, five or six second nucleic acid sequences. This may enable binding of two, three, four, five or six complementary nucleic acid sequence being labeled by an imaging molecule.
- the second nucleic acid sequences may be the same and thus provide for binding with the same type of imaging molecule, or different thus providing for binding to different complementary nucleic acid sequences being labeled by an imaging molecule, preferably with different imaging molecules. Thus, different combinations of intensities and/or colors of imaging molecules may be created and/or used. Additionally or alternatively, contacting the sample with the different complementary nucleic acid sequences being labeled by an imaging molecule at different times allows for the usage of a larger barcoding space. Additionally or alternatively, no target molecule is labeled with more than five, preferentially two, more preferentially one single-stranded nucleic acid molecule(s).
- one or more single-stranded nucleic acid molecule is capable of binding exactly one type of imaging molecule via its complementary nucleic acid sequence being labeled by this imaging molecule. Additionally or alternatively, different single stranded nucleic-acid molecules may be capable of binding different types of imaging molecules. Alternatively, one or more nucleic acid probes may be capable of binding multiple types of imaging molecules. In some embodiments, one or more single-stranded nucleic acid molecule is capable of binding exactly one type of complementary nucleic acid sequence being labeled by an imaging molecule. Additionally or alternatively, different single stranded nucleic-acid molecules may be capable of binding different types of complementary nucleic acid sequence being labeled an imaging molecule.
- one or more single-stranded nucleic acid molecule is capable of binding multiple types of complementary nucleic acid sequence being labeled an imaging molecule.
- the sample may comprise one or more cells and/or a cell lysate and/or purified components, all e.g. from cell culture, a biopsy, and/or a liquid biopsy, for example primary patient cells (from healthy tissue, immune cells, cancer), fresh frozen (FF) tissue, formalin-fixed paraffin embedded (FFPE) tissue, or extracts from liquid biopsy (e.g. cells or exosomes), or spheroids, or organs-on a chip, all from human or animal.
- the methods may include drift correction.
- the term complementary is used as follows: The complementarity may be 100% or less, for example between 80% and 100%, or more preferably between 90% and 100%.
- the methods may include: determining the binding kinetics of at least one, preferably all, first nucleic acid sequences to their target complementary nucleic acid sequences, and/or second nucleic acid sequences to their complementary nucleic acid sequences being labeled by an imaging molecule; and taking the determined binding kinetic into account for detection step d).
- the target complementary nucleic acid sequence may be a portion of a secondary binder (not to be confused with secondary probe/label).
- the secondary binder is a molecule that specifically binds to a molecule, termed the binding molecule, which may specifically bind to the target molecule.
- the binding molecule may specifically bind to the target molecule.
- the single-stranded nucleic acid molecule may indirectly bind to the target molecule via the secondary binder and the binding molecule.
- Secondary binders may be comprised of a nucleic acid molecule and a secondary binding molecule.
- a secondary binding molecule is a molecule specifically binding a primary binding molecule as described herein above.
- Secondary binding molecules are generally known in the art. For example, they may be secondary antibodies. Secondary antibodies are antibodies which specifically bind to antibodies produced from the respective animals. For example ‘anti-rat’ secondary antibodies bind to antibodies from rats.
- secondary binding molecules may be nanobodies.
- the invention further relates to a hybridization complex comprising the target complementary nucleic acid, the nucleic acid molecule, and a blocking strand.
- a blocking strand also termed “blocker strand” or “blocker” is generally known in the art as a single stranded nucleic acid that may hybridize to a different single stranded nucleic acid molecule with one or more reaction partners that have a lower affinity than the blocking strand to the different single stranded nucleic acid molecule (see e.g. doi: 10.1021/acs.nanolett.9b02565). It can be used to block the interactions of the different single stranded nucleic acid molecule with its reaction partner(s).
- a blocking strand may comprise the complementary sequence to a docking sequence (e.g. the second nucleic acid sequence of the nucleic acid molecule), to prevent the transient interaction of an imager with the second nucleic acid sequence of the nucleic acid molecule.
- a docking sequence e.g. the second nucleic acid sequence of the nucleic acid molecule
- the number of nucleotides of a blocking strand is 2-30 nucleotides, preferably 5-20 nucleotides, more preferably 10-15 nucleotides longer than the competitive interaction partner, and they should cover the whole sequence targetable by the interaction partner. Their total length is 15-40 nucleotides.
- imager strands R1-R6 (SeqID No 3533- 3538) are 6-7 nucleotides long, while the blocking strands used (SeqID No 3498-3526) are 19-20 nucleotides long and cover the complete docking sequences (SeqID No 3527-3532).
- blocking strands may fulfill the following requirements: * blocking strands should cover the whole region targeted by their competitor; * blocking strands should stably bind to the region targeted by their competitor; * blocking strands should not be displaced by their competitors (i.e. bind much more stably and thus be longer).
- the approach of using blocking strands is especially useful when a target molecule is designed to provide signal in only one round of imaging, as described in this example.
- the single stranded nucleic acid molecule can be provided to the sample before the respective imaging round, thereby activating the target molecule, and the blocker can be provided after the imaging round, thereby deactivating the target molecule.
- the present invention also relates to a method of analyzing a sample, comprising: a. localizing with increasing preference at least 10, at least 30, at least 60 or at least 100 target molecules within the sample, preferably within cells of the sample; e.
- Point Pattern Analysis is a collection of methods to extract information from point patterns, especially the spread or distribution of each of the target molecules over the space of the sample, preferably on the cell surface or within the volume of the cells. It provides a number of metrics that describe the type of association and association probability between different target molecules.
- Point pattern analysis is the study of point patterns, the spatial arrangements of points in space, herein generally in 3-dimensional space.
- Nearest Neighbour Analysis measures the spread or distribution of each of the target molecules over the space of the sample, preferably on the cell surface or within the volume of the cells. It provides a numerical value that describes the extent to which the different the target molecules are clustered or uniformly spaced. This in turn results in an interaction pattern of the different target molecules.
- Different target molecules that are found to co-localize or almost co-localize likely bind to each other in the context of the sample, e.g. within cells.
- Such interaction partners are bona fide targets for the modification of biological processes with samples, e.g. within cells or between cells.
- the method further comprises binding molecule evaluation and development.
- binding molecule evaluation and development it is beneficial to first characterize binders that are later used for labelling target molecules. This characterization can then be integrated into the data evaluation as a calibration.
- the method of analyzing a sample uses one or more single-stranded nucleic acid molecules and/or one or more hybridization complexes as defined herein above.
- the method uses hybridization complexes as defined herein above, wherein the target complementary nucleic acid sequences of the hybridization complexes are conjugated to different binding molecules that are capable of specifically binding to the at least 10, at least 30, at least 60 or at least 100 target molecules.
- the hybridization complexes as defined herein above are particularly advantageous for the method of analyzing a sample because different hybridization complexes are conjugated to different binding molecules that are capable of specifically binding to the at least 10, at least 30, at least 60 or at least 100 target molecules, and can be easily designed, thereby achieving a high multiplex level.
- the second nucleic acid sequences of the single-stranded nucleic acid molecules and/or the complexes comprise or consist of sequences being selected from (TCC)n, (ACC)n, (CTT)n, (AAC)n, (CT)n, and (AC)n, wherein n is 4 to 12.
- (TCC)n, (ACC)n, (CTT)n, (AAC)n, (CT)n, and (AC)n, wherein n is 4 to 12, are the six speed- optimized sequences R1-R6 (doi: 10.1038/s41592-020-0869-x). Due the speed-optimization these sequences are ideally suitable for multiplexed applications, such as the method of analyzing a sample.
- the method of analyzing a sample is preferably conducted via multiple subsequent imaging rounds and the speed-optimized sequences are particularly advantageous for achieving the subsequent imaging rounds in short time.
- only one of the second nucleic acid sequences is present per imaging round.
- only two, only three, only four, only five, only six or more different second nucleic acid sequences are present and addressable per imaging round.
- only one type of the first nucleic acid sequences is present per imaging round.
- only two, only three, only four, only five, only six or more different first nucleic acid sequences are present and addressable per imaging round.
- a present single stranded nucleic acid may be turned non-addressable by blocking using a “blocker strand” (also termed “blocking strand”).
- the method comprises binding molecule preselection, preferably bare binding molecule preselection. To preselect probably appropriate binding molecules from a given pool of potential binding molecules to a target molecule of interest, their specificity may be screened. Briefly, this may be done by colocalizing fluorescence signal originating from the target molecule of interest (or a moiety fused to it) with a fluorescence signal related to a secondary binder known to bind the bare binding molecule.
- “Secondary binder” refers to a molecule that specifically binds a binding molecule/primary binder.
- the method comprises binding molecule specificity testing. After successful preselection, a short DNA oligonucleotide can be conjugated onto the bare binding molecule, to make it what is called a primary binder herein. The short DNA oligonucleotide can be conjugated to the binding molecule using site-specific DNA conjugation.
- the method comprises primary binder labelling efficiency testing.
- primary binder labelling efficiency may be tested.
- the method comprises primary binder labelling efficiency determination, e.g. via 2-plex Exchange-PAINT imaging or any of the methods described herein.
- binding molecule characterization e.g. binding molecule specificity testing and/or primary binder labelling efficiency testing
- any cell line may be used, e.g. CHO, BSC1, HeLa.
- the method comprises sample preparation for multiplexed immune receptor DNA-PAINT.
- samples may be prepared for imaging in any suitable way. This can be done in multiple ways and also depends on the sample type.
- the method comprises preparation of functionalized planar supported lipid bilayers (SLBs).
- the method comprises cell preparation for multiplexed immune receptor DNA-PAINT imaging.
- the method comprises data acquisition, e.g. multiplexed molecular imaging of target molecules.
- the method comprises multiplexed cellular imaging of cellular proteins.
- the method comprises data evaluation, e.g.
- the method comprises image analysis, e.g. postprocessing of a/the raw super-resolution imaging data. It may provide for getting from multiple single channel transient binding movies to one multiplexed molecular map, which specifies the localizations of all target molecules detected in the sample.
- the method comprises data analysis, wherein data is aggregated to elucidate the direct interaction patterns present in the sample. This step corresponds to getting from the multiplexed molecular map to one or more direct interaction patterns present in the sample.
- a direct interaction pattern describes a set of target molecules commonly found in close proximity, and optionally probability distributions of their relative distances.
- the method comprises selecting one or more binding molecule candidates having a size of 25nm or less, preferably 12.5nm or less, for example 12.4nm or 4nm.
- the binding molecules may be small enough to allow for generation of direct interaction patterns precise enough for meaningful insights for drug development or diagnostics.
- the method comprises selecting one or more binding molecule candidates having an affinity as measured in bulk measurements (e.g. SPR, Octet, FRET-based assays) of KD ⁇ 200nM, preferably ⁇ 20nM.
- the step of localizing at least 10, at least 30, at least 60 or at least 100 target molecules within the sample, preferably within cells of the sample may comprise super-resolution imaging, preferably super-resolution fluorescence microscopy, preferably DNA-PAINT, optionally including any of its improvements such as Exchange-PAINT, RESI, and/or the methods described herein.
- a sample in any of the embodiments herein may be one or more cell lines, primary patient cells (from healthy tissue, immune cells, cancer), fresh frozen (FF) tissue, formalin-fixed paraffin embedded (FFPE) tissue, or extracts from liquid biopsy (e.g.
- the target complementary nucleic acid sequence e.g. in the form of DNA
- the target complementary nucleic acid sequence may be conjugated to a binding molecule site-specifically, preferably by using sortase, C-terminal cysteine, N-terminal serine, threonine, or artificial aminoacids.
- a molecular density of target-molecule-fused reference molecules may be less than 5000 molecules/ ⁇ m 2 , preferably less than 500 molecules/ ⁇ m 2 , more preferably less than 50 molecules/ ⁇ m 2 .
- target-molecule-fused reference molecules may be more than 0.01 molecules/ ⁇ m 2 , preferably more than 0.1 molecules/ ⁇ m 2 , more preferably more than 1 molecule/ ⁇ m 2 .
- Target-molecule-fused reference molecules are easily-taggable reference molecules, which are genetically fused to target molecules.
- the cells when developing binding molecules for the target molecule CD-80, the cells can be CD-80 non-expressing or knock-out cells and transfected with a GFP-CD80 fusion.
- GFP can serve as a reference molecule, for which a well-characterized anti-GFP nanobody is available and can be used in the characterization of the binding molecules for the target molecule.
- the method is configured for rational drug design and comprises one or a combination of the following: • Indication selection, e.g. melanoma, non-small cell lung cancer (NSLC); • Target and off-target sample selection, e.g.: o Healthy, normal vs cancerous tissue, cancer vs dendritic cell, T cell & DC vs T cell & cancerous tissue; and/or o FACS-sorted samples; • Visualization of target molecule identification, e.g.: o Membrane proteins (e.g.
- CD80 & CD86 may be often ⁇ 15nm apart from each other in the target sample, but rarely in off-target sample, with the total number of CD80 and CD86 being similar in target and off-target samples.
- the method may comprise one or a combination of the following: • Drug development comprising: o Developing mono-, bi- or multivalent primary binder (antibody-based or similar primary binder; optional: biocompatible scaffold for organizing target binders) based on the direct interaction pattern, and use cooperativity for binding to investigate if target proteins are present on pathogenic cells/tissue, such that a single binding domain does not bind stably but the specific neighbourhood configuration (or most of it) is required for prolonged, stable binding, thereby generating specificity between target and off-target o using the neighborhood configuration distances, angles and configurations (hetero- and homo-oligomerisation) as described by the direct interaction pattern to guide the drug molecule design (see, for example, Bila et al, J Am Chem Soc
- the method is configured for hit-to-lead and/or lead optimization, and comprises one or a combination of the following: • Target and off-target sample selection; • Selection of the molecular targets potentially involved o Use Proteins appearing in biochemical hypotheses and general knowledge of the indication involved; Also add candidate drug as a visualization target, preferably each binding domain separately, to elucidate its position in the interaction pattern • creating a direct interaction pattern, optionally including: o Define an optimization metric based on the direct interaction pattern: e.g.
- fraction of colocalized binding domains to target molecules, and drug molecule target vs off- target binding fully characterize recorded protein map consisting of multiple different proteins or protein epitopes and binding epitopes and extract key parameters such as distances, angles, molecular orientations, oligomerisation, cluster contributions (cluster size, cluster shape, cluster density, amount of clusters, protein ratios, protein motifs), reduction of dimensionality (e.g. UMAP) for key parameter elucidation; • Data acquisition and evaluation of samples with multiple optimization candidates as target molecules; • Scoring of optimization candidates, resulting in one optimal candidate; • Based on the direct interaction pattern, optimization of mono- and multivalent binder geometry, e.g.
- Fig.1 schematically illustrate a single-stranded nucleic acid molecule according to the invention and related concepts and methods
- Fig.2 schematically illustrates a single-stranded nucleic acid molecule according to the invention and related concepts and methods
- Fig.3 illustrates a proof of principle experiment
- Fig.4 schematically illustrates aspects of methods including a codebook
- Fig.5 schematically illustrates aspects of methods including a codebook.
- Fig.6 shows a false color image of neurons imaged with a 29-plex imaging method according to the present invention
- Fig.7 shows zoom-ins from a whole-cell view, via DNA-PAINT localization Data, to single target molecule localization data obtained from
- Fig.8 shows Specificity
- Fig.9 shows Labeling Efficiency
- Fig.10 shows preparation steps for functionalized planar glass-supported lipid bilayers
- Fig.11 shows multiplexed single-protein imaging of immune checkpoint receptors
- Fig.12 shows a direct interaction analysis for dendritic cells and that multiplexed spatial receptor pattern analysis reveals novel key interaction motifs in dendritic cells
- Fig.13 shows a direct interaction analysis for cancer cells and that the absence of costimulatory receptors drives formation of PD-L1/MHC-I clusters in B16-F10.
- Fig.14 shows that CD80 presence interferes with MHC-I/PD-L1 clustering irrespective of cell type.
- Fig.15 Workflow of analysis pipeline. Schematic outline of workflow for the analysis of 6- plex receptor point patterns on cells. Following image post-processing, whole-cell datasets were utilized for further analysis. In the first step, global receptor correlations were determined by a modified version of Ripley’s K function. Ripley’s K curve was calculated for all 36 possible pairwise receptor combinations and compared to the results of complete spatial randomness (CSR). The normalized Ripley’s K curves were then integrated and averaged from all individual cells of a single cell type and corresponding condition.
- CSR complete spatial randomness
- a correlation matrix for all 36 possible receptor combinations was generated to distinguish between clustered or dispersed receptor patterns, as well as random receptor distributions.
- NBD nearest neighbor distance
- single channel data was compared to CSR simulations to determine the percentage of homo- interactions.
- Hetero-interactions were evaluated via correlation of cross-channel data to corresponding CSR simulations.
- the percentage of receptor interactions, as well as the corresponding interaction distance were defined.
- global DBSCAN analysis was used to identify receptor motifs within the upper limit of evaluated receptor interaction distances in clustered regions containing at least three receptors.
- DBSCAN was applied to both experimental and simulated 6-plex data, ignoring receptor identities at this stage.
- Detected receptor motifs for experimental and CSR simulated data were sorted based on their receptor identities within each cluster, leading to 63 unique cluster IDs. Comparison of experimental and CSR simulated data allowed for the extraction of multiple different parameters. All readout parameters are highlighted in blue.
- Fig.16 Multiplexed spatial receptor pattern analysis of non-stimulated MutuDCs.
- A DNA-PAINT image of non-stimulated MutuDCs showing receptor positions of the imaged immune checkpoint receptors.
- Correlation matrix for all 36 possible receptor combinations allows classification into “clustered”, “random” or “dispersed” receptor distributions as in Fig.12B.
- C Receptor interactions visualized via a circle plot. Receptor species are positioned at the corners of a hexagon, with the circle size proportional to the average receptor density and connections between interacting receptors marked by lines colored by their corresponding receptor identity with the line thickness being proportional to the average interaction score derived from pairwise correlation analysis.
- D NND analysis yields quantitative information about directly interacting receptor species.
- E Global DBSCAN analysis is used to identify receptor motifs within a 35 nm search radius in clustered regions containing at least three receptors. Overall contributions of “clustered” vs. “non-clustered” areas were compared to a CSR distribution of target receptors on the same cell surface.
- Non-clustered areas were compared to a CSR distribution of target receptors on the same cell surface.
- Receptor motifs were identified by grouping individual clusters based on their receptor species, weighted by underlying area and plotted as normalized histograms (mean +- 95% CI). Respective key receptor motifs are highlighted.
- G Quantitative analysis of the key receptor motifs from (F) reveals 2 distinct motifs (Motif 1 – MHC-I/CD80/PD-L1, Motif 2 – MHC-I/CD86/PD-L1. The motifs represent 2.0% ⁇ 1.3%, 1.9% ⁇ 1.2% of all clusters, respectively.
- FIG.18 Multiplexed spatial receptor pattern analysis of 6 hours stimulated MutuDCs.
- A DNA-PAINT image of 6 h stimulated MutuDCs showing receptor positions of the imaged immune checkpoint receptors.
- B Correlation matrix for all 36 possible receptor combinations allows classification into “clustered”, “random” or “dispersed” receptor distributions.
- C Receptor interactions visualized via a circle plot.
- Receptor species are positioned at the corners of a hexagon, with the circle size proportional to the average receptor density and connections between interacting receptors marked by lines colored by their corresponding receptor identity with the line thickness being proportional to the average interaction score derived from pairwise correlation analysis.
- D NND analysis yields quantitative information about directly interacting receptor species.
- E Global DBSCAN analysis is used to identify receptor motifs within a 35 nm search radius in clustered regions containing at least three receptors. Overall contributions of “clustered” vs. “non-clustered” areas were compared to a CSR distribution of target receptors on the same cell surface.
- (F) Receptor motifs were identified by grouping individual clusters based on their receptor species, weighted by underlying area and plotted as normalized histograms (mean +- 95% CI). Respective key receptor motifs are highlighted.
- (G) Quantitative analysis of the key receptor motifs from (F) reveals 4 distinct motifs (Motif 1 – CD86/CD80/PD-L1, Motif 2 – CD80, Motif 3 – MHC-I/CD80/PD-L1, Motif 4 – MHC-I/CD86/CD80/PD-L1. The motifs represent 5.9% ⁇ 2.1%, 0.7% ⁇ 0.3%, 4.5% ⁇ 1.1%, 5.1% ⁇ 2.9% of all clusters, respectively.
- FIG.19 Multiplexed spatial receptor pattern analysis of 12 hours stimulated MutuDCs.
- A DNA-PAINT image of 12 h stimulated MutuDCs showing receptor positions of the imaged immune checkpoint receptors.
- B Correlation matrix for all 36 possible receptor combinations allows classification into “clustered”, “random” or “dispersed” receptor distributions.
- C Receptor interactions visualized via a circle plot.
- Receptor species are positioned at the corners of a hexagon, with the circle size proportional to the average receptor density and connections between interacting receptors marked by lines colored by their corresponding receptor identity with the line thickness being proportional to the average interaction score derived from pairwise correlation analysis.
- D NND analysis yields quantitative information about directly interacting receptor species.
- E Global DBSCAN analysis is used to identify receptor motifs within a 35 nm search radius in clustered regions containing at least three receptors. Overall contributions of “clustered” vs. “non-clustered” areas were compared to a CSR distribution of target receptors on the same cell surface.
- (F) Receptor motifs were identified by grouping individual clusters based on their receptor species, weighted by underlying area and plotted as normalized histograms (mean +- 95% CI). Respective key receptor motifs are highlighted.
- (G) Quantitative analysis of the key receptor motifs from (F) reveals 3 distinct motifs (Motif 1 – MHC-I/CD80/PD-L1, Motif 2 – MHC-I/CD86/PD-L1, Motif 3 – MHC-II/CD80). The motifs represent 1.7% ⁇ 0.5%, 1.7% ⁇ 0.8%, 1.1% ⁇ 0.9% of all clusters, respectively.
- FIG.20 Multiplexed spatial receptor pattern analysis of 24 hours stimulated MutuDCs.
- A DNA-PAINT image of 24 h stimulated MutuDCs showing receptor positions of the imaged immune checkpoint receptors.
- B Correlation matrix for all 36 possible receptor combinations allows classification into “clustered”, “random” or “dispersed” receptor distributions.
- C Receptor interactions visualized via a circle plot.
- Receptor species are positioned at the corners of a hexagon, with the circle size proportional to the average receptor density and connections between interacting receptors marked by lines colored by their corresponding receptor identity with the line thickness being proportional to the average interaction score derived from pairwise correlation analysis.
- D NND analysis yields quantitative information about directly interacting receptor species.
- E Global DBSCAN analysis is used to identify receptor motifs within a 35 nm search radius in clustered regions containing at least three receptors. Overall contributions of “clustered” vs. “non-clustered” areas were compared to a CSR distribution of target receptors on the same cell surface.
- (F) Receptor motifs were identified by grouping individual clusters based on their receptor species, weighted by underlying area and plotted as normalized histograms (mean +- 95% CI). Respective key receptor motifs are highlighted.
- (G) Quantitative analysis of the key receptor motifs from (F) reveals 6 distinct motifs (Motif 1 – CD86/CD80/PD-L1, Motif 2 – CD80/PD-L1, Motif 3 & Motif 4 – MHC-I/CD80/PD-L1, Motif 5 & Motif 8 – MHC- I/CD86/PD-L1, Motif 6 – MHC-I/CD86/CD80, Motif 7 – MHC-I/CD86/PD-L2).
- the motifs represent 1.9% ⁇ 0.6%, 0.5% ⁇ 0.1%, 2.3% ⁇ 0.7%, 4.5% ⁇ 1.7%, 3.1% ⁇ 0.8%, 0.5% ⁇ 0.2% of all clusters, respectively.
- Data is shown as mean ⁇ 95% confidence interval of three independent experiments and 10 cells, * p ⁇ 0.05; *** p ⁇ 0.001; n.s., not significant)
- Fig.21 Multiplexed spatial receptor pattern analysis of specific peptide-MHC-I complexes on MutuDCs fed ovalbumin protein.
- A DNA-PAINT image of MutuDCs stimulated for 6h with CpG and IFN ⁇ , while being fed ovalbumin, showing receptor positions of the imaged immune checkpoint receptors.
- A DNA-PAINT image of non-stimulated B16-F10 cells showing receptor positions of the imaged immune checkpoint receptors.
- B Correlation matrix for all 36 possible receptor combinations allows classification into “clustered”, “random” or “dispersed” receptor distributions.
- C Receptor interactions visualized via a circle plot. Receptor species are positioned at the corners of a hexagon, with the circle size proportional to the average receptor density and connections between interacting receptors marked by lines colored by their corresponding receptor identity with the line thickness being proportional to the average interaction score derived from pairwise correlation analysis.
- D NND analysis yields quantitative information about directly interacting receptor species.
- Fig.23 Multiplexed spatial receptor pattern analysis of 3 hours stimulated B16-F10 cells.
- A DNA-PAINT image of 3h IFN ⁇ stimulated B16-F10 cells showing receptor positions of the imaged immune checkpoint receptors.
- B Correlation matrix for all 36 possible receptor combinations allows classification into “clustered”, “random” or “dispersed” receptor distributions.
- C Receptor interactions visualized via a circle plot.
- Receptor species are positioned at the corners of a hexagon, with the circle size proportional to the average receptor density and connections between interacting receptors marked by lines colored by their corresponding receptor identity with the line thickness being proportional to the average interaction score derived from pairwise correlation analysis.
- D NND analysis yields quantitative information about directly interacting receptor species.
- E Global DBSCAN analysis is used to identify receptor motifs within a 35 nm search radius in clustered regions containing at least three receptors. Overall contributions of “clustered” vs. “non-clustered” areas were compared to a CSR distribution of target receptors on the same cell surface.
- (F) Receptor motifs were identified by grouping individual clusters based on their receptor species, weighted by underlying area and plotted as normalized histograms (mean +- 95% CI). Respective key receptor motifs are highlighted.
- (G) Quantitative analysis of the key receptor motifs from (F) reveals a single key motif (Motif 1 – MHC-I/PD-L1). The motif represents 29.2% ⁇ 3.9% of all clusters, respectively. (Data is shown as mean ⁇ 95% confidence interval of three independent experiments and 11 cells, *** p ⁇ 0.001; n.s., not significant) Fig.24. Multiplexed spatial receptor pattern analysis of 6 hours stimulated B16-F10 cells.
- A DNA-PAINT image of 6h stimulated B16-F10 cells showing receptor positions of the imaged immune checkpoint receptors.
- B Correlation matrix for all 36 possible receptor combinations allows classification into “clustered”, “random” or “dispersed” receptor distributions.
- C Receptor interactions visualized via a circle plot. Receptor species are positioned at the corners of a hexagon, with the circle size proportional to the average receptor density and connections between interacting receptors marked by lines colored by their corresponding receptor identity with the line thickness being proportional to the average interaction score derived from pairwise correlation analysis.
- D NND analysis yields quantitative information about directly interacting receptor species.
- E Global DBSCAN analysis is used to identify receptor motifs within a 35 nm search radius in clustered regions containing at least three receptors. Overall contributions of “clustered” vs. “non-clustered” areas were compared to a CSR distribution of target receptors on the same cell surface.
- F Receptor motifs were identified by grouping individual clusters based on their receptor species, weighted by underlying area and plotted as normalized histograms (mean +- 95% CI). Respective key receptor motifs are highlighted.
- Non-clustered areas were compared to a CSR distribution of target receptors on the same cell surface.
- Receptor motifs were identified by grouping individual clusters based on their receptor species, weighted by underlying area and plotted as normalized histograms (mean +- 95% CI). Respective key receptor motifs are highlighted.
- G Quantitative analysis of the key receptor motifs from (F) reveals 3 independent receptor motifs (Motif 1 – MHC-I/MHC- II/PD-L1, Motif 2 – MHC-II, Motif 3 – MHC-I/MHC-II/CD86/PD-L1).
- the receptor motifs represent 30.0% ⁇ 3.1%, 3.6% ⁇ 1.1%, 3.3% ⁇ 1.2% of all clusters, respectively.
- Data is shown as mean ⁇ 95% confidence interval of three independent experiments and 10 cells, ** p ⁇ 0.01; *** p ⁇ 0.001; n.s., not significant
- Fig.26 Multiplexed spatial receptor pattern analysis of 24 hours stimulated B16-F10 cells.
- A DNA-PAINT image of 24h stimulated B16-F10 cells showing receptor positions of the imaged immune checkpoint receptors.
- B Correlation matrix for all 36 possible receptor combinations allows classification into “clustered”, “random” or “dispersed” receptor distributions.
- C Receptor interactions visualized via a circle plot.
- Receptor species are positioned at the corners of a hexagon, with the circle size proportional to the average receptor density and connections between interacting receptors marked by lines colored by their corresponding receptor identity with the line thickness being proportional to the average interaction score derived from pairwise correlation analysis.
- D NND analysis yields quantitative information about directly interacting receptor species.
- E Global DBSCAN analysis is used to identify receptor motifs within a 35 nm search radius in clustered regions containing at least three receptors. Overall contributions of “clustered” vs. “non-clustered” areas were compared to a CSR distribution of target receptors on the same cell surface.
- (F) Receptor motifs were identified by grouping individual clusters based on their receptor species, weighted by underlying area and plotted as normalized histograms (mean +- 95% CI). Respective key receptor motifs are highlighted.
- (G) Quantitative analysis of the key receptor motifs from (F) reveals 2 independent receptor motifs (Motif 1 – MHC-I/MHC- II/PD-L1, Motif 2 – MHC-I/MHC-II/CD86/PD-L1). The receptor motifs represent 2.3% ⁇ 1.2%, 0.6% ⁇ 0.5% of all clusters, respectively.
- FIG.27 Multiplexed spatial receptor pattern analysis of specific OVA peptide-MHC-I complexes on B16-F10 cells.
- A DNA-PAINT image of B16-F10 cells that transgenically express OVA protein stimulated for 6h with IFN ⁇ showing receptor positions of the imaged immune checkpoint receptors.
- B Correlation matrix for all 36 possible receptor combinations allows classification into “clustered”, “random” or “dispersed” receptor distributions.
- C Receptor interactions visualized via a circle plot.
- Receptor species are positioned at the corners of a hexagon, with the circle size proportional to the average receptor density and connections between interacting receptors marked by lines colored by their corresponding receptor identity with the line thickness being proportional to the average interaction score derived from pairwise correlation analysis.
- D NND analysis yields quantitative information about directly interacting receptor species.
- E Global DBSCAN analysis is used to identify receptor motifs within a 35 nm search radius in clustered regions containing at least three receptors. Overall contributions of “clustered” vs. “non-clustered” areas were compared to a CSR distribution of target receptors on the same cell surface.
- (F) Receptor motifs were identified by grouping individual clusters based on their receptor species, weighted by underlying area and plotted as normalized histograms (mean +- 95% CI). Respective key receptor motifs are highlighted.
- (G) Quantitative analysis of the key receptor motifs from (F) reveals a single key receptor motif (Motif 1 – MHC-I/PD-L1). The receptor motif represents 29.0% ⁇ 3.5% of all clusters, respectively. (Data is shown as mean ⁇ 95% confidence interval of three independent experiments and 6 cells, *** p ⁇ 0.001; n.s., not significant) Fig.28. Validation of CD80 KO cDC1 cells.
- cDC1 cells were then differentiated from the bone marrow in Flt3 cultures prior to stimulation with 500nM CpG1826 + 100U/ml IFN ⁇ for 6 hours and sorting as in (A) for subsequent imaging experiments. Verification of CD80 deletion on cDC1 cells by flow cytometric staining is shown. Fig.29. Multiplexed spatial receptor pattern analysis of 6 hours stimulated wild-type cDC1 cells.
- A DNA-PAINT image of 6 h stimulated cDC1 cells showing receptor positions of the imaged immune checkpoint receptors.
- B Correlation matrix for all 36 possible receptor combinations allows classification into “clustered”, “random” or “dispersed” receptor distributions.
- C Receptor interactions visualized via a circle plot.
- Receptor species are positioned at the corners of a hexagon, with the circle size proportional to the average receptor density and connections between interacting receptors marked by lines colored by their corresponding receptor identity with the line thickness being proportional to the average interaction score derived from pairwise correlation analysis.
- D NND analysis yields quantitative information about directly interacting receptor species.
- E Global DBSCAN analysis is used to identify receptor motifs within a 35 nm search radius in clustered regions containing at least three receptors. Overall contributions of “clustered” vs. “non-clustered” areas were compared to a CSR distribution of target receptors on the same cell surface.
- (F) Receptor motifs were identified by grouping individual clusters based on their receptor species, weighted by underlying area and plotted as normalized histograms (mean +- 95% CI). Respective key receptor motifs are highlighted.
- (G) Quantitative analysis of the key receptor motifs from (F) reveals 3 distinct receptor motifs (Motif 1 & Motif 2 – CD86/CD80/PD-L1, Motif 3 – MHC-I/CD86/CD80, Motif 4 & Motif 5 – MHC- I/CD86/CD80/PD-L1). The motifs represent 9.3% ⁇ 1.9%, 0.4% ⁇ 0.1%, 19.8% ⁇ 0.6% of all clusters, respectively.
- FIG.30 Multiplexed spatial receptor pattern analysis of 6 hours stimulated CD80 KO cDC1 cells.
- A DNA-PAINT image of 6 h stimulated CD80 KO cDC1 cells showing receptor positions of the imaged immune checkpoint receptors.
- B Correlation matrix for all 36 possible receptor combinations allows classification into “clustered”, “random” or “dispersed” receptor distributions.
- C Receptor interactions visualized via a circle plot.
- Receptor species are positioned at the corners of a hexagon, with the circle size proportional to the average receptor density and connections between interacting receptors marked by lines colored by their corresponding receptor identity with the line thickness being proportional to the average interaction score derived from pairwise correlation analysis.
- D NND analysis yields quantitative information about directly interacting receptor species.
- E Global DBSCAN analysis is used to identify receptor motifs within a 35 nm search radius in clustered regions containing at least three receptors. Overall contributions of “clustered” vs. “non-clustered” areas were compared to a CSR distribution of target receptors on the same cell surface.
- (F) Receptor motifs were identified by grouping individual clusters based on their receptor species, weighted by underlying area and plotted as normalized histograms (mean +- 95% CI). Respective key receptor motifs are highlighted.
- (G) Quantitative analysis of the key receptor motifs from (F) reveals 2 distinct receptor motifs (Motif 1 – MHC-I/PD- L1, Motif 2 – MHC-I/CD86/PD-L1). The motifs represent 46.6% ⁇ 3.0%, 5.3% ⁇ 1.7% of all clusters, respectively. (Data is shown as mean ⁇ 95% confidence interval of two independent experiments and 11 cells, *** p ⁇ 0.001; n.s., not significant) Fig.31.
- A DNA-PAINT image of non-stimulated cDC1 cells showing receptor positions of the imaged immune checkpoint receptors.
- B Correlation matrix for all 36 possible receptor combinations allows classification into “clustered”, “random” or “dispersed” receptor distributions.
- C Receptor interactions visualized via a circle plot. Receptor species are positioned at the corners of a hexagon, with the circle size proportional to the average receptor density and connections between interacting receptors marked by lines colored by their corresponding receptor identity with the line thickness being proportional to the average interaction score derived from pairwise correlation analysis.
- D NND analysis yields quantitative information about directly interacting receptor species.
- E Global DBSCAN analysis is used to identify receptor motifs within a 35 nm search radius in clustered regions containing at least three receptors. Overall contributions of “clustered” vs. “non-clustered” areas were compared to a CSR distribution of target receptors on the same cell surface.
- F Receptor motifs were identified by grouping individual clusters based on their receptor species, weighted by underlying area and plotted as normalized histograms (mean +- 95% CI). Respective key receptor motifs are highlighted.
- A DNA-PAINT image of non-stimulated CD80 KO cDC1 cells showing receptor positions of the imaged immune checkpoint receptors.
- B Correlation matrix for all 36 possible receptor combinations allows classification into “clustered”, “random” or “dispersed” receptor distributions.
- C Receptor interactions visualized via a circle plot. Receptor species are positioned at the corners of a hexagon, with the circle size proportional to the average receptor density and connections between interacting receptors marked by lines colored by their corresponding receptor identity with the line thickness being proportional to the average interaction score derived from pairwise correlation analysis.
- D NND analysis yields quantitative information about directly interacting receptor species.
- Fig.33 Multiplexed spatial receptor pattern analysis of 6 hours stimulated CD80 KO MutuDCs.
- A DNA-PAINT image of 6 h stimulated CD80 KO MutuDCs showing receptor positions of the imaged immune checkpoint receptors.
- B Correlation matrix for all 36 possible receptor combinations allows classification into “clustered”, “random” or “dispersed” receptor distributions.
- C Receptor interactions visualized via a circle plot.
- Receptor species are positioned at the corners of a hexagon, with the circle size proportional to the average receptor density and connections between interacting receptors marked by lines colored by their corresponding receptor identity with the line thickness being proportional to the average interaction score derived from pairwise correlation analysis.
- D NND analysis yields quantitative information about directly interacting receptor species.
- E Global DBSCAN analysis is used to identify receptor motifs within a 35 nm search radius in clustered regions containing at least three receptors. Overall contributions of “clustered” vs. “non-clustered” areas were compared to a CSR distribution of target receptors on the same cell surface.
- (F) Receptor motifs were identified by grouping individual clusters based on their receptor species, weighted by underlying area and plotted as normalized histograms (mean +- 95% CI). Respective key receptor motifs are highlighted.
- (G) Quantitative analysis of the key receptor motifs from (F) reveals 2 distinct receptor motifs (Motif 1 & Motif 2 & Motif 4 – MHC-I/PD-L1, Motif 3 – MHC-I/CD86/PD-L1). The motifs represent 19.3% ⁇ 2.7%, 5.7% ⁇ 2.6% of all clusters, respectively.
- FIG.34 Validation of CD80-overexpressing B16-F10 cell lines.
- B16-F10 cells were retrovirally transduced using MSCV-mCD80 (IRES-mCherry) retroviral vectors to overexpress either wild-type CD80 or mutant CD80-L107E.
- mCherry expressing cells were then sorted by flow cytometry to generate stably transduced cell lines. Representative flow cytometric plots showing mCherry versus CD80 staining are included for all 3 cell lines.
- Fig.35 Validation of CD80-overexpressing B16-F10 cell lines.
- A DNA-PAINT image of 6 h stimulated CD80- overexpressing B16-F10 cell showing receptor positions of the imaged immune checkpoint receptors.
- B Correlation matrix for all 36 possible receptor combinations allows classification into “clustered”, “random” or “dispersed” receptor distributions.
- C Receptor interactions visualized via a circle plot. Receptor species are positioned at the corners of a hexagon, with the circle size proportional to the average receptor density and connections between interacting receptors marked by lines colored by their corresponding receptor identity with the line thickness being proportional to the average interaction score derived from pairwise correlation analysis.
- NND analysis yields quantitative information about directly interacting receptor species.
- E Global DBSCAN analysis is used to identify receptor motifs within a 35 nm search radius in clustered regions containing at least three receptors. Overall contributions of “clustered” vs. “non-clustered” areas were compared to a CSR distribution of target receptors on the same cell surface.
- F Receptor motifs were identified by grouping individual clusters based on their receptor species, weighted by underlying area and plotted as normalized histograms (mean +- 95% CI). Respective key receptor motifs are highlighted.
- NND analysis yields quantitative information about directly interacting receptor species.
- E Global DBSCAN analysis is used to identify receptor motifs within a 35 nm search radius in clustered regions containing at least three receptors. Overall contributions of “clustered” vs. “non-clustered” areas were compared to a CSR distribution of target receptors on the same cell surface.
- F Receptor motifs were identified by grouping individual clusters based on their receptor species, weighted by underlying area and plotted as normalized histograms (mean +- 95% CI). Respective key receptor motifs are highlighted.
- the receptor motif represents 2.1% ⁇ 0.5%, 27.5% ⁇ 3.7%, 14.6% ⁇ 2.3% of all clusters, respectively.
- Data is shown as mean ⁇ 95% confidence interval of two independent experiments and 12 cells, *** p ⁇ 0.001; n.s., not significant
- Fig.38 Multiplexed spatial receptor pattern analysis of Abatacept-treated CD80 KO MutuDCs.
- A DNA-PAINT image of 6h stimulated CD80 KO MutuDCs showing receptor positions of the imaged immune checkpoint receptors. Cells were treated with Abatacept during the last 10 minutes of the overall time.
- C Receptor interactions visualized via a circle plot. Receptor species are positioned at the corners of a hexagon, with the circle size proportional to the average receptor density and connections between interacting receptors marked by lines colored by their corresponding receptor identity with the line thickness being proportional to the average interaction score derived from pairwise correlation analysis.
- D NND analysis yields quantitative information about directly interacting receptor species.
- E Global DBSCAN analysis is used to identify receptor motifs within a 35 nm search radius in clustered regions containing at least three receptors. Overall contributions of “clustered” vs. “non-clustered” areas were compared to a CSR distribution of target receptors on the same cell surface.
- (F) Receptor motifs were identified by grouping individual clusters based on their receptor species, weighted by underlying area and plotted as normalized histograms (mean +- 95% CI). Respective key receptor motifs are highlighted.
- (G) Quantitative analysis of the key receptor motifs from (F) reveals a single key receptor motif (Motif 1 – MHC-I/CD86/PD-L1). The receptor motif represents 35.0% ⁇ 3.9% of all clusters, respectively. (Data is shown as mean ⁇ 95% confidence interval of two independent experiments and 12 cells, *** p ⁇ 0.001; n.s., not significant) Fig.39. Receptor interaction distances.
- DNA origami discs were either non-functionalized (“empty”) or functionalized either with pMHC only or a combination of pMHC and PD-L1. The latter were arranged into clusters that are either closely spaced (“close”, ⁇ 14 ⁇ ⁇ ) or widely (“far”, ⁇ 28 ⁇ ⁇ ) spaced.
- B Agarose gel analysis (2% agarose) of the DNA origami disc library either functionalized (+ Ligands) or non-functionalized (No Ligand) with pMHC and/or PD-L1 molecules showing properly folded and purified samples. Cy5 signal characterizes assembled DNA origami disc and SYBR Safe signal represents all DNA-based samples. A delayed sample migration indicates successful attachment of the ligands.
- the Examples illustrate the present invention. Examples The present invention provides, i.a., nucleic acid based molecules, kits and compositions and methods for target detection, particularly for multiplexed imaging. The present invention provides for an application in a biological environment, particularly a single cell, particularly intermolecular single cell multiomics. Intermolecular single cell multiomics provides for localizing a relatively large number of target molecules of a single cell, particularly via super-resolution fluorescence microscopy, particularly PAINT.
- Figure 1 schematically illustrates a target 1 and a single-stranded nucleic acid molecule 2, the single-stranded nucleic acid molecule 2 comprising a first nucleic acid sequence 4 and a second nucleic acid sequence 6 that differs from the first nucleic acid sequence.
- the first nucleic acid sequence is capable of specifically hybridizing to a target complementary nucleic acid sequence 10 and the second nucleic acid sequence 6 is capable of transiently binding to a complementary nucleic acid sequence being labeled by an imaging molecule 20.
- the single-stranded nucleic acid molecule 2 may comprise a toehold seed 8 as shown, but is not so limited.
- the toehold seed 8 may be used for separating these molecules via toehold mediated strand displacement, i.e. by contacting the hybridized molecules with an invader strand as explained above.
- the target complementary nucleic acid 10 may be part of a primary binder 14.
- the primary binder 14 may further comprise a binding molecule 12.
- the target complementary nucleic acid 10 may be conjugated to the binding molecule 12 in any suitable way, see above.
- the binding molecule 12 may be capable of selectively binding the target molecule 1.
- the target molecule 1 may be a protein and the binding molecule 12 may be a corresponding antibody.
- the target molecule 1 may comprise a nucleic acid sequence, e.g. DNA, and the binding molecule 12 may be a complementary nucleic acid sequence.
- the primary binder 14 may be a nucleic acid strand comprising two domains, one is the target complementary nucleic acid sequence 10 and the other one is the binding molecule 12.
- the target complementary nucleic acid 10 may alternatively be part of the target molecule (not illustrated).
- the single-stranded nucleic acid molecule 2 directly hybridizes to the target molecule via the first nucleic acid sequence 4, without any primary binder 14 in between.
- the complementary nucleic acid sequence being labeled by an imaging molecule 20 may comprise one or more fluorescent imaging molecules 22, for example Alexa488, Cy3b, and/or Atto 647N, and a nucleic acid sequence 24 that is complementary to the second nucleic acid sequence 6.
- the hybridization kinetics between these two sequences (this hybridization being termed HybB) and the hybridization kinetics between the first nucleic acid sequence 4 and the target complementary nucleic acid sequence 10 (this hybridization being termed HybA) may be chosen such that the first nucleic acid sequence is capable of stronger associating with its complementary nucleic acid sequence than the second nucleic acid sequence. This may include an appropriate selection of conditions.
- HybA may be a reversible but stable binding
- HybB is a transient binding
- the melting temperatures, the GC content and/or the length of the sequences i.e. the number of hybridizing base pairs
- the appropriate binding behavior may be achieved.
- HybA and HybB may be selected to have similar binding kinetics when the ratio is 1:1. With such tool at hand, a detection method as explained above may be performed.
- Figure 2 illustrates an exemplary method according to the invention.
- Inset a) of Figure 2 shows a target molecule 1 bound to a primary binder 14 via a binding molecule 12.
- the target molecule 1 may be comprised in a single cell that was appropriately prepared for fluorescence imaging as generally known in the art or an artificially created sample such as a surface with DNA origami, etc. Examples for sample preparation are given below.
- the target molecule 1 may be contacted with the primary binder 14, which may be present in solution.
- the binding of the target molecule 1 to the primary binder 14 as well as the binding molecule 12 to the target complementary nucleic acid strand 10 are stable under the given conditions.
- the single-stranded nucleic acid strand 2 is added and contacted with the complementary primary binder 14.
- the first nucleic acid sequence 4 of the single-stranded nucleic acid strand 2 hybridizes with the target complementary nucleic acid sequence 10 of the primary binder 12, the result being shown in inset b) of Figure 2. Under the given conditions this hybridization (HybA) may be stable.
- An imaging buffer including the complementary nucleic acid sequence being labeled by an imaging molecule 20 is added, resulting in insert c), which shows transient binding of the second nucleic acid strand 6 of the nucleic acid molecule 2 to the complementary nucleic acid sequence being labeled by an imaging molecule 20.
- the binding constant kon may be 10 7 1/Ms, 10 8 1/Ms, 10 9 1/Ms (with the unit 1/Ms being equivalent to liter/(mol*s) ) and the unbinding constant koff may be 3/s, 1/s, 10/s, or 50/s.
- the amount of the added molecules i.e.
- the order of the step of contacting the sample/target molecule 1 with the primary binder and thus the target complementary nucleic acid sequence 10 and binding molecule 12, the step of contacting the sample/target molecule 1 with the single-stranded nucleic acid molecule, and the step of contacting the sample with a complementary nucleic acid sequence being labeled by an imaging molecule 20 under appropriate conditions may be performed in any order.
- DNA-PAINT image acquisition or any other suitable acquisition, may be carried out.
- the imaging molecule 22 provides for a signal in an image when bound to the target molecule 1 (ON-state), here via the nucleic acid molecule 2 and optionally the primary binder 14.
- ON-state the imaging molecule 22
- OFF-state the imaging molecule 22
- Switching between ON- and OFF-states is a stochastic process for each target molecule in the sample.
- the binding may be controlled to enable a sufficiently low number of target molecules 2 in the ON-state in each acquired image to be able to localize the individual target molecules 2 within one image without disturbing signals from neighbors at a distance that is classically unresolvable.
- a sufficiently large number of target molecules 2 will be present as a signal in one of the images.
- all the signals as derived from the individual images may be added and analyzed to localize the individual target molecules 2.
- a final data set which includes the positions of the localized target molecules 2 may be created and visualized as appropriate, e.g. plotted and/or shown in a single image.
- a drift marker may be added to the sample prior to starting the image acquisition.
- a drift marker may be any suitable marker known in the art, for example one or more gold beads, fluorescent beads, or fluorescent dyes immobilized to a fix reference in the sample such as a surface of a cover slip or imaging chamber, channel, or well.
- the drift marker has a shape and/or combination of spectrally distinguishable fluorescence dyes that enables identification and correction for drift in all three dimensions.
- an unbinding step may be performed, e.g. as illustrated in Fig.2 d).
- the unbinding step may comprise any of the techniques mentioned above. Particularly, and as illustrated in Fig.2, it may include adding an invader strand 30 corresponding to the toehold seed 8 and the first nucleic acid sequence 4.
- the invader strand 30 may hybridize with the first nucleic acid sequence 4 and remove the single- stranded nucleic acid molecule 2 from the primary binder 14.
- the invader strand 30 may remove the single-stranded nucleic acid molecule 2 from the target molecule 1.
- the complex of the single-stranded nucleic acid molecule 2 and the invader strand 30 is removed from the sample, e.g. by washing with an appropriate buffer solution.
- the unbinding step may include applying heat and/or buffer conditions to the sample that support and/or enable dissociation of the single-stranded nucleic acid molecule 2 from the corresponding primary binder 14 and/or target molecule 1.
- An unbinding step may also be done before the first detection round in the sample with an unspecific technique, e.g. heat and/or buffer conditions, to ensure that the target molecules 1 are free to bind the single-stranded nucleic acid molecules according to the invention in the first detection round.
- an unspecific technique e.g. heat and/or buffer conditions
- Another option is to use different imaging molecules 22 that are distinguishable in the applied detection method, e.g. have distinguishable fluorescence spectra. This enables detecting the different target molecules 1 in the same detection round.
- the different complementary nucleic acid sequences being labeled by an imaging molecule 20 may specifically bind to the different single stranded nucleic acid sequences 2 for the different target molecules 1.
- the different complementary nucleic acid sequences being labeled by an imaging molecule 20 may be orthogonal and the second nucleic acid strands 6 of the different single stranded nucleic acid sequences 2 may be orthogonal.
- the primary binders 14 need to be orthogonal with respect to their binding molecule 12 and with respect to their target complementary nucleic acid sequence 10 in order to allow for specific detection of the two or more target molecules 1.
- Contacting the sample with the two or more orthogonal primary binders 14 may be a single step in which all orthogonal primary binders 14 are added to the sample at once, preferably in a step upstream the cycle of detection rounds. This means that the sample may first be contacted with all primary binders 14, and then the cycle of M detection rounds starts with the first detection round by contacting the sample with the first single-stranded nucleic acid molecule as described above.
- adding one or more but not all orthogonal primary binders 14 may be a step upstream of the detection cycle, and the rest of the orthogonal primary binders 14 may be added to the sample as part of one or more detection rounds as described above.
- the orthogonal primary binders 14 may be grouped for addition to the sample according to reaction condition requirements and/or constraints that are given by the way of coding the labeling of the different target molecules 1 (see also explanations relating to codebook below).
- Primary binders 14 may not be required for target molecules 1 that may be directly bound by single-stranded nucleic acid molecules 2, for example for target molecules comprising the same type of nucleic acid as the single-stranded nucleic acid molecule 2.
- the target complementary nucleic acid sequence 10 is a domain of the target molecule 2. Accordingly, the step of contacting such target molecules 1 with primary binders 14 may be omitted. Alternatively, it is also possible to have one or more additional intermediate binding molecules (not shown) between the primary binder 14 and the single-stranded nucleic acid molecule 2. In this respect reference is made to the above provided example of biotin and streptavidin-labelled antibodies.
- the step of adding a single-stranded nucleic acid molecule 2 to the sample may be performed at the same time as adding the target complementary nucleic acid sequence 14. Particularly, the nucleic acid molecule 2 may be contacted with the target complementary nucleic acid sequence 14 before adding both to the sample.
- the nucleic acid molecule 2 may be hybridized to the target complementary nucleic acid sequence 14 when it is added to the sample.
- the complementary nucleic acid sequences being labeled by an imaging molecule 20 may be added to the sample together with the corresponding nucleic acid molecules 2 and/or primary binders. However, it is preferred to add the complementary nucleic acid sequences being labeled by an imaging molecule 20 after the optional addition of the corresponding primary binders 14 and the corresponding nucleic acid molecules 2, particularly after some incubation time in order to allow for the primary binders 14 and the nucleic acid molecules 2 to stably bind.
- the first nucleic acid sequence 4 of the single-stranded nucleic acid molecule 2 may have a length that is appropriate for the respective application.
- the length may be as short as possible to avoid unnescessary long reaction time, and as long as necessary to provide for the required specificity and number of orthogonal sequences.
- the first nucleic acid sequence 4 may have a length of 4 to 30 nucleotides, preferably 16 to 24 nucleotides.
- the first nucleic acid sequence may have a GC-content of 45%- 55%, preferably 50%. The GC-content may be used to set the binding kinetics as appropriate.
- the second nucleic acid sequence 6 of the single-stranded nucleic acid molecule 2 may have a length of
- the target nucleic acid sequence in a primary binder may have a length of 6nt to 150nt, preferably 10nt to 50 nt, more preferably 12 to 20 nt, for example 21 nt.
- the nucleic acid sequence being labeled by an imaging molecule may have any suitable length.
- the nucleic acid sequence being labeled by an imaging molecule may have a length of 4 to 10 nucleotides.
- the length of any of the above mentioned nucleic acid sequences may also be used, within the constraints of specificity and orthogonality, for setting the binding kinetics to the complementary nucleic acid sequence.
- the present invention provides for several advantages.
- the invention uses two hybridizations, HybA and HybB.
- HybA of the first nucleic acid sequence 4 of the single stranded nucleic acid molecule 2 to the target complementary nucleic acid sequence 10 of or bound to the target molecule 1 provides for a high degree of multiplexing.
- the number of available orthogonal first sequences 4 is high enough to enable detection of a large number of different target molecules 1 in one sample, e.g. proteins in a single cell.
- the imaging molecule 22 is indirectly bound to the target molecule 1 via the complementary nucleic acid sequence being labeled by an imaging molecule 20 and the second nucleic acid sequence 6 of the single-stranded nucleic acid molecule 2, i.e. HybB.
- the degree of multiplexing is decoupled form the binding kinetics of the complementary nucleic acid sequence being labeled by an imaging molecule 20 via HybB.
- the second nucleic acid sequences 6 comprise or consist of sequences being selected from (TCC)n, (ACC)n, (CTT)n, (AAC)n, (CT)n, and (AC)n, wherein n is 4 to 12.
- the sequences known from the SPEED- PAINT technique for example the sequences known from the SPEED- PAINT technique.
- the complementary nucleic acid sequences being labeled by an imaging molecule 20 have complementary sequences. This enables speed-optimized acquisition of images in the detection rounds. Nevertheless, the multiplexing may be 20 to 30, i.e.
- the single-stranded nucleic acid molecule 2 may comprise more than 1 second nucleic acid sequences 6.
- the single-stranded nucleic acid molecule 2 may comprise 2 or more,3 or more, 4 or more, 5 or more, or 6 or more second nucleic acid sequences 6. This may enable binding of a corresponding number of complementary nucleic acid sequences being labeled by an imaging molecule 20.
- At least some of the second nucleic acid sequences of one single-stranded nucleic acid molecule 2 may be orthogonal. This may enable binding of orthogonal complementary nucleic acid sequences being labeled by an imaging molecule 20 and thus different imaging molecules 22.
- the same type of imaging molecules 22 may be bound to one nucleic acid molecule 2 via the same type of second nucleic acid sequence 6 or via different types of second nucleic acid sequence 6. Additionally or alternatively, multiplexing may be increased by binding more than one primary binder 14 per target molecule 1. Particularly, 2 or more, 3 or more, 4 or more, 5 or more, or 6 or more primary binders 14 and thus corresponding single stranded nucleic acid molecules 2 may be used per target molecule 1. However, for both cases multiple second nucleic acid sequences 6 per single-stranded nucleic acid molecule and multiple primary binder 14 per target molecule 1 care needs to be taken not to corrupt the binding kinetics of HybA and/or HybB.
- the singe-stranded nucleic acid molecules 2 according to the invention provide for another advantage: Using them as imager targets and washing them off after a detection round stabilizes imager accessibility of the target molecule 1. Nucleic acid strands that hybridize to complementary nucleic acid sequences being labeled by an imaging molecule 20 tend to get corrupted by photodamage in prolonged DNA-PAINT experiments [doi:10.3390/molecules23123165]. Therefore, replacing the single-stranded nucleic acid molecules in between detection rounds provides for fresh single-stranded nucleic acid molecules in the subsequent detection round, thus increasing the reliability.
- Figure 3 illustrates a proof of principle experiment. Insert A shows the design of an experiment according to the present invention. Insert B shows an exemplary image of the respective experiment.
- DNA origami is a technique generally known in the art. The term is used for DNA that is artificially designed to fold into a specific structure or shape. In short, a long scaffold strand and multiple small so called staple strands are designed. When the staple strands bind to the scaffold strand the scaffold is forced into a desired shape. It is useful to know that DNA origami may provide for very well defined structures with defined lengths and distances in between certain points, which can be simply addressed with DNA strands by extending staple strands, such that single-stranded nucleic acid sequences are present at the respective positions. Therefore, DNA origami is often used as a microscopy standard or ruler.
- DNA-origami was chosen to have an overall shape approximating a cuboid, with a significant number of helices lying essentially parallel, and a thickness of only one layer of helices.
- the comic in figure 3 shows a top view of a DNA-origami cuboid 40 including the addressable staple strand positions 42 of the structure, represented by hexagons (only one of them is referenced with reference sign 42). Some of the positions 42 were designed to be a first type of target molecules 1. Some of the positions 42, represented by dotted hexagons, were designed to be regular DNA-PAINT targets as known in the art. A pattern was created by providing the target molecules 1 of the first type at 12 positions 42 with a spacing of 20 nm, indicated by cross hatch, and the regular DNA-PAINT targets at clusters of positions 42 in the four corners of the shown top of the cuboid 40.
- a second type of these DNA origami were created, using different extension sequences at the cross hatch positions (second type of target molecules 1), acting as target complementary nucleic acid sequences as described herein below.
- a first DNA origami structure was designed to comprise a first target complementary nucleic acid sequence 10 at the cross hatched positions and a second DNA origami structure was designed to comprise a second target complementary nucleic acid sequence 10 at the cross hatched positions.
- the DNA-origami were immobilized on a surface of a cover slip of a reaction chamber.
- Labelling (i.e. in this case attaching a docking strand) of the regular DNA-PAINT targets (in this case the respective positions of the scaffold) at the dotted positions 42 for standard DNA-PAINT was done during DNA origami folding by extending the respective staple strand sequences with a docking strand sequence.
- the first and second DNA origami comprising the first and second target complementary nucleic acid sequences 10 were mixed and immobilized on a surface of a cover slip of a reaction chamber. Detection occurred in three detection rounds, a first detection round using standard DNA-PAINT to localize the DNA origami using the regular DNA-PAINT targets (dotted positions in Fig.3), and a second and third detection round according to the present invention for the first and second type target molecule 1 represented by cross hatched positions.
- the sample i.e.
- the first and second DNA origami which already incorporated standard DNA PAINT docking strands at the dotted positions and a first and second primary binder 14, respectively, at the cross hatched positions, was contacted in the first imaging round with an imager capable of transiently binding the standard DNA PAINT docking strands and a standard DNA PAINT dataset was recorded.
- a first single-stranded nucleic acid molecule comprising (a) a first nucleic acid sequence being capable of specifically hybridizing to the first target complementary nucleic acid sequence 10 and (b) a second nucleic acid sequence that differs from the first nucleic acid sequence and is capable of transiently binding to a complementary nucleic acid sequence being labeled by an imaging molecule, wherein the first nucleic acid sequence is capable of stronger associating with its complementary nucleic acid sequence than the second nucleic acid sequence, was added to the sample. Then, an imaging buffer comprising the complementary nucleic acid sequence being labeled by an imaging molecule 20 was added. Then, detection of the imaging molecule 22 was performed according to PAINT.
- a third detection round was performed for the second target complementary nucleic acid sequence 10 with the second nucleic acid molecule 2 but with the same type of complementary nucleic acid sequence being labeled by an imaging molecule 20.
- the two DNA origami structures and their different target molecule types 1 were distinguished via their imaging time (second detection round or third detection round). The result of the experiment is shown in the false color picture in panel B of Fig.3. The top row shows signal from a first DNA origami structure and the bottom row shows signal from a second DNA origami structure.
- the left, second to left and second to right columns show the signal for the first, second and third imaging round, respectively, and the right column shows the overlayed signal.
- the duration of an experiment with the technique described so far still increases for every additional target molecule 1, i.e. the more target molecules to be detected the longer the overall detection time.
- another aspect of the present invention is related to a specific way of barcoding the target molecules 1, which will be described in the following with reference to Fig.4.
- Figure 4 shows a scheme for up to 1820-plex single protein imaging in 16 sequential detection rounds.
- the numbers are only exemplary and depend on the length of the used barcodes and whether a single type of imager and primary binders with exactly one target complementary nucleic acid sequence 10 are used (as in the scheme), or more, which would of course further increase the plexing-number (see above).
- the concept is as follows: To every type of target molecule 1 a unique barcode (i.e. identification sequence) having M ordered positions is assigned. Every position represents one detection round and is assigned with either “to be imaged” or “not to be imaged”. Thus, the identification sequence defines for every one of the M detection rounds, whether the corresponding type of target molecule 1 is to be labeled and detected according to the invention described above or not.
- the identification sequences assigned to the target molecules 1 are referred to as a codebook.
- “to be imaged” is represented by “1”
- “not to be imaged” is represented by “0”.
- a unique single-stranded nucleic acid molecule 2 is used, as represented by the library in Fig.4B.
- the types of single-stranded nucleic acid molecules 2 have orthogonal first nucleic acid sequences 4 but the same second nucleic acid sequence and are consequently configured to bind the same nucleic acid sequence labeled by an imaging molecule 20, i.e. the same type of imaging molecule 22.
- FIG.4C the pictures schematically illustrate a portion of a cell is schematically illustrated by the thick black curved lines (note that this is an exemplary environment).
- Target molecules 1 of different types are represented by hexagons with different numbers (T1, T4, T5 and T8).
- Figure 4C depicts three of 16 detection rounds, namely round 1 in the left column, round 2 in the middle column and round 16 in the right column.
- the top row of Fig.4C schematically depicts the nucleic acid strands that are present/bound.
- the bottom row shows schematic illustrations of what is detected in imaging.
- detection round M all target molecules 1 that have been assigned with “to be imaged” in that detection round M as set out in their identification sequence are detected as described herein.
- detection round 1 left column of fig.4C
- target molecules T5 and T8 are labelled, as indicated by the black stars representing an imaging molecule 22.
- Target molecules of type T5 and T8 are bound by corresponding primary binders (represented by the short black straight line extending from the hexagons; note the different degrees of abstraction of the drawn molecules in different figures as explained in Fig.4A), which present target specific target complementary nucleic acid sequences.
- Secondary binders (long black line) with first nucleic acid sequences corresponding to the respective target complementary nucleic acid sequences have been added to the sample and are hybridized to the target complementary nucleic acid sequence.
- the secondary probes present the same second nucleic acid sequences capable of transiently binding to the complementary nucleic acid sequence being labelled by an imaging molecule (short black line with black star). It must be noted that, due to this transient binding, the “labelling” is still of the stochastic nature that is the basic principle of the PAINT technique. After imaging, the result of which is represented in the bottom left comic of Fig.
- the secondary probes and the nucleic acid sequences being labelled by an imaging molecule of the previous round are removed via toehold mediated replacement and washing.
- primary binders are added such that all target molecule types are bound to corresponding primary binders before starting the imaging rounds and adding the first secondary probes. As already explained, this is optional; the primary binders may also be added individually in the individual detection rounds, e.g. together with the secondary probes.
- the imaging data collected in the different imaging rounds is processed, particularly aligned.
- the imaging data in combination with the knowledge, which target types are labelled in which imaging round i.e. the data from the codebook
- the individual target types may not only imaged in high resolution but also be identified.
- the maximum ratio of identification sequence positions being “1” over the total number of identification sequence positions may be chosen to allow for error correction.
- the ratio may be from 1/16 to 10/16, or from 1/10 to 7/10, or from 1 ⁇ 4 to 3/4.
- specific rules may be set, defining the minimum number of “0”-positions between neighboring “1”-positions, for example at least 1, at least 2, or at least 3 “0”-postions between neighboring “1”-positions or vice versa at least 1, at least 2, or at least 3 “1”-postions between neighboring “0”-positions. This allows for certain error correction because false negative or false positive detection events may be excluded.
- the present invention relates to a method of mapping the localization of different target molecules within a sample, preferably within cells of the sample.
- an interaction pattern of the different target molecules based on their localization may be performed, preferably by nearest neighbor-based analysis.
- this aspect of the present invention is an additional and independent invention. It is independent of the specific imaging technique and tools, particularly of the imaging techniques and tools mentioned above. Any imaging technique that provides for the required imaging quality may be used. Nevertheless, the imaging techniques and tools mentioned above are among the preferred ones. Molecularly resolved, multiplexed image data may be used to rationally inform the choice and further development of drugs.
- the image data may not only be used for improving quantification of the target molecules by counting instead of using fluorescence intensity as a metric, but also the key information of target molecule maps such as distances, angles, molecular orientations, oligomerisation, cluster contributions (cluster size, cluster shape, protein ratios, protein motifs) may be used for characterizing differences between samples for better patient stratification as well as developing multivalent drugs that specifically target one type of molecular key motif, thereby increasing specificity and sensitivity in treated patients while reducing/inhibiting undesired side effects as well as significantly reducing development time and costs.
- target molecule maps such as distances, angles, molecular orientations, oligomerisation, cluster contributions (cluster size, cluster shape, protein ratios, protein motifs) may be used for characterizing differences between samples for better patient stratification as well as developing multivalent drugs that specifically target one type of molecular key motif, thereby increasing specificity and sensitivity in treated patients while reducing/inhibiting undesired side effects as well as significantly reducing development time
- the present invention provides to-date unprecedented biomedical understanding and insight into basic principles of cell-cell communication and thus not only in the development of novel multi-specific drugs but also in the characterization of the mechanism of action of already existing as well as novel drugs by being able to characterize the spatial organization of key target molecules (e.g. proteins) at molecular resolution, determining specificity and efficiency of target drug, and molecular reorganization at the nanoscale in response to different drug doses. Thus, it may guide the design of dosing, toxicity and drug combination studies, as well as for patient stratification.
- This aspect of the present invention is most relevant to drugs that specifically bind to target molecules, e.g. therapeutic and diagnostic antibodies or other binders (e.g. nanobody, aptamer, scF V ).
- the present invention may enable study potential reorganization of the molecular architecture, thus enabling mode of action or biomarker investigations.
- Indications most relevant to the present invention for drug development are those targetable with therapeutic antibodies, such as cancer, autoimmune diseases, migraine and various infectious diseases (e.g. HIV, HPV, Ebola, Alzheimer’s disease, Crohn’s disease, multiple sclerosis, rheumatic arthritis, as well as cancers like melanoma, non-small cell lung cancer, breast cancer, and others).
- DNA origami sample preparation was done in a 6-channel u-slide (Ibidi Cat.: 80607).
- the chamber was washed with 5 ml of 2xSSC buffer and 1 ml of B+ buffer until 1 ml of imager solution was applied for and between imaging rounds. After all imaging rounds for one set of secondary labels, the chamber was incubated with dehybridization buffer for 15 min. More explicitly, after immobilization of the 42 DNA origami in one chamber and one DNA origami each in 42 chambers and washing all chambers with buffers B+ and 2xSSC, secondary labels 1, 2, 3, 4, 5, and 6 were incubated at a concentration of 100 nM each in hybridization buffer for 15 min.
- the chamber was washed with 5ml of 2xSSC and 1ml B+ buffer, and flushed with 1ml R1 imager solution (500 pM R1 in Buffer B+ with 7 ⁇ g/ml PCD (Sigma-Aldrich cat. P8279-25UN), 0,4 mg/ml PCA (Sigma-Aldrich 37580-25g-f), and 0,3 mg/ml Trolox (Sigma-Aldrich 238813-1G)).
- DNA-PAINT imaging (10000 frames at exposure of 100 ms with an illumination of 25 mW at 561nm) of this imager, the chamber was washed with 3ml B+ buffer, and 1ml R2 imager solution was applied.
- the secondary labels (single stranded nucleic acid molecules) were stripped off by flushing 1ml of 100nm secondary label-specific toehold strands (as listed in Table 1, 600nm in total) in dehybridization buffer into the chamber and incubating for 15 min, followed by injecting 1ml of new secondary labels at 100 nM each in hybridization buffer and incubating for 15 min. Subsequently, the chamber was washed with 5ml of 2xSSC and 1ml B+ buffer, and flushed with 1ml R1 imager solution, repeating the rounds of imagers.
- Tables 1 and 2 list the sequences and codes used, and Figure 5 shows representative results, where the single- DNA origami samples could be used to confirm the correct identification of DNA origami structures based on their codes.
- a code for a given location is created by a binary number with as many digits as DNA-PAINT imaging rounds are performed (in this case 42), where a ‘0’ denotes no imager binding in the respective location in the respective imaging round, while a ‘1’ denotes registered imager binding at that location.
- DNA-PAINT imaging rounds are taken in chronological order, so for example the third DNA-PAINT imaging round (of imager R3 in the present case) of the second secondary labeling round (II) is the 9 th DNA- PAINT imaging round in total, so it is represented in the 9 th digit of the code.
- 210 different DNA origamis were designed using 1220nm spaced positions of target complementary sequences, as described herein above. Secondary labels (single-stranded nucleic acid molecules) for a target complementary sequence were used in four secondary label hybridization rounds each, while they were absent in six secondary label hybridization rounds, leading to codebook entries as described in Table 3.
- the sample was washed three times with 1 ml PBS, and a 1:3 dilution of Gold nanoparticles (90nm Standard Gold Nanoparticles, Cytodiagnostics, Inc., cat.: G-90-100) were incubated for 5 min as fiducial markers for drift correction.
- Gold nanoparticles 90nm Standard Gold Nanoparticles, Cytodiagnostics, Inc., cat.: G-90-100
- Target complementary nucleic acids were conjugated to the C-terminus of their respective secondary binding molecules (in this case secondary nanobodies) according to table 4 and 5 (Mouse: FluoTag- X2 anti-Mouse Ig kappa light chain, NanoTag Biotechnologies; Rabbit: FluoTag-X2 anti- Rabbit IgG, NanoTag Biotechnologies; Synaptotagmin: FluoTag-X2 anti-Synaptotagmin 1, NanoTag Biotechnologies, PSD95: sdAb anti-PSD95, NanoTag Biotechnologies) to form secondary binders: First, buffer was exchanged to 1 ⁇ PBS + 5 mM EDTA, pH 7.0 using Amicon centrifugal filters (10k Da molecular weight cut-off) and free cysteines were reacted with 20-fold molar excess of bifunctional maleimide-DBCO linker (Sigma Aldrich, cat: 760668) for 2-3 hours on ice.
- Unreacted linker was removed by buffer exchange to PBS using Amicon centrifugal filters. Azide-functionalized DNA was added with 3-5 molar excess to the DBCO-nanobody and reacted overnight at 4°C. Unconjugated nanobody and free azide-DNA was removed by anion exchange using an ⁇ KTA Pure liquid chromatography system equipped with a Resource Q 1 ml column. Nanobody-DNA concentration was adjusted to 5 ⁇ M (in 1xPBS, 50% glycerol, 0.05% NaN3) and stored at -20°C.
- Target molecule labeling with these secondary binders was done by performing a preincubation of the antibody (binding molecule) with their respective secondary binder in 10 ul antibody incubation buffer at room temperature for 2 h, for each target molecule separately. After the preincubation time, a large excess (molar ratio of 1:2) of unlabeled secondary nanobody was introduced and incubated for 5 min. Subsequently, the six antibody-to-secondary-binder complexes corresponding to the first round of secondary label (single-stranded nucleic acid molecule) hybridization were pooled in 300 ul antibody incubation buffer and incubated in the neuron sample for 90 min.
- the sample was washed five times with 1ml PBS and once with 1 ml Buffer C followed by a postfixation with 2.4% paraformaldehyde in PBS for 7 min. Afterwards, the sample was washed three times with 1ml PBS and once with 1ml 2xSSC buffer and the secondary label (nucleic acid molecule) hybridization for barcoding round 1 was carried out according to table 4 with 100 nM of each secondary label for 20 min (totalling 600 nM). Finally, the sample was washed five times with 1ml 2xSSC buffer and once with 1ml buffer C.
- the sample was then flushed with 1ml R1 imager solution (concentration as described in Table 4, in buffer C with 7 ⁇ g/ml PCD (Sigma-Aldrich cat. P8279-25UN), 0,4 mg/ml PCA (Sigma-Aldrich 37580-25g-f), and 0,3 mg/ml Trolox (Sigma-Aldrich 238813-1G)).
- 1ml R1 imager solution concentration as described in Table 4, in buffer C with 7 ⁇ g/ml PCD (Sigma-Aldrich cat. P8279-25UN), 0,4 mg/ml PCA (Sigma-Aldrich 37580-25g-f), and 0,3 mg/ml Trolox (Sigma-Aldrich 238813-1G)).
- DNA-PAINT imaging 15 mW at 561nm HILO illumination
- the chamber was washed with 1ml buffer C, and 1ml R2 imager solution was applied.
- the secondary labels (single-stranded nucleic acid molecules) were blocked by flushing 1ml 100 nM blocking strands per second nucleic acid sequence to block binding of the complementary nucleic acid sequences being labeled by an imager (corresponding to the six imagers previously imaged, totalling 600 nM) in hybridization buffer into the chamber and incubating for 15 min, followed by injecting 1ml of new secondary labels at 100 nM each in hybridization buffer and incubating for 15 min.
- Fig 6 shows the results of the described 29-plex SUM-PAINT experiment mapping the 3D protein distribution of a single neuron with all the 29 channels color-coded and overlayed.
- Fig 6 B The gallery with the respective protein distribution and localization precision can be seen in Fig 6 B. Summing up the acquisition time of single channels and adding the transition time between barcoding rounds the whole 29-plex single molecule atlas can be acquired in only 30h of total experimental time. Vendor Cat.
- T cells may be very relevant in other applications, especially for characterization of the mode of action of drugs, or for biomarker discovery and validation. Other target molecules than proteins are also contemplated.
- One of the most important interfaces for cell-cell communication is the cell surface.
- the receptor-ligand interactions engaged during a cell-cell encounter can trigger signalling cascades that have a profound downstream impact upon cell behaviour and differentiation. This is particularly important in the immune system, where the outcome of cellular interactions can dictate whether an immune response is initiated, the nature of the ensuing response, and whether an existing response is sustained or extinguished.
- DCs dendritic cells
- Surface encounter of specific peptide-MHC antigen complexes alongside co-stimulatory molecules, such as CD80 and CD86, can drive both initial T cell differentiation into effector cells, and subsequent expansion and differentiation of an established effector T cell response.
- antigen encounter in the context of excessive immune checkpoint molecules, such as PD-L1 and PD-L2 can both block initial T cell priming, and restrain effector T cell function and differentiation.
- T cell stimulatory capacity of a given cell type has classically been defined by the absolute cell surface levels of key immunomodulatory ligands.
- the native, single protein spatial organisation of immune regulatory molecules on the cell surface, and how this spatial organisation contributes to T cell control remains poorly understood. This represents a major knowledge gap, particularly given that protein clustering is a well-defined determinant of surface protein signalling and function. Progress has been limited in this area due to a lack of technologies capable of concurrent spatial mapping of multiple proteins at the single molecule level.
- Target molecule of interest
- Binding molecule the affinity molecule/antibody/antibody mimetic/.... to tag the target molecule
- Secondary binder an affinity molecule tagging the binding molecule, either directly fluorescently labeled, or labeled with a DNA-PAINT docking sequence
- Binding molecule evaluation and development is an optional aspect of the present invention. This also applies to all of its sub-aspects. As mentioned above, for optimal sample analysis, it may be beneficial to first characterize binding molecules and/or primary binders that are later used for labelling target molecules. This characterization can then be integrated into the data evaluation as a calibration.
- a protein fusion of the target molecule of interest e.g. MHC-I, MHC-II, CD86, CD80, PD-L1, PDL2
- reference targets for reference labelling e.g. epitope tags like ALFA-tag, Halo- tag, SNAP tag, SPOT tag, FLAG-tag, His-Tag, sortase tag or fluorescent proteins
- unnatural amino acid labeling or gene editing e.g. CRISPR
- binding molecule and secondary binder binding according to version 1 (preferably if the cells express a single target molecule): o Incubate a binding molecule with the cell sample to let it attach to the target molecule; o Incubate a secondary binder against the binding molecule in the cell sample, where the secondary binder comprises a modification for stable or transient fluorescence (e.g. fluorophore modification, or DNA oligomer conjugation comprising a docking sequence for a complementary nucleic acid sequence being labelled by an imaging molecule, preferably a DNA-PAINT imager).
- a modification for stable or transient fluorescence e.g. fluorophore modification, or DNA oligomer conjugation comprising a docking sequence for a complementary nucleic acid sequence being labelled by an imaging molecule, preferably a DNA-PAINT imager.
- Binding molecule and secondary binder binding according to version 2 (preferably if the cells express multiple target molecules each with a unique reference target) – as an alternative to step 5, version 1: o Incubate a secondary binder against the binding molecule in bulk solution, where the secondary binder comprises a modification for stable or transient fluorescence (e.g. fluorophore modification, or DNA oligomer conjugation comprising a docking sequence for a complementary nucleic acid sequence being labelled by an imaging molecule, preferably a DNA-PAINT imager). O Incubate the complex of secondary binder and binding molecule against the sample, to let it attach to the target molecule. Optional steps: 1. Add fiducial markers, e.g. gold nanoparticles. 2.
- CHO cells were transfected with a single receptor construct (one of the following set in each sample: mEGFP-ALFA-MHC-I, mEGFPALFA-MHC-II, mEGFP-ALFA-CD86, mEGFP-ALFA-CD80, meGFP-ALFA-PD-L1, meGFP- ALFA-PDL2) for binding molecule characterization using Lipofectamine LTX as specified by the manufacturer.
- CHO cells were allowed to express mEGFP-ALFA-receptors for 16–24 h.
- Binding molecules (one each for the corresponding samples: CD80, CD86, MHC-I, MHC-II, PD-L1, PD-L2) and ALFA-tag nanobody (as reference binders) were diluted in blocking buffer and added at a final concentration of 50nM each for 90min at 24°C. Unbound binding molecules and reference binders were removed by washing with Buffer Y, followed by washing once with Buffer X for 5 min.
- Fluorescently labelled secondary antibodies targeting binding molecules were dissolved in blocking buffer and added at a final concentration of 100nM each for 60min at 24°C (to the corresponding sample: CD80, CD86, MHC-I, MHC-II, PD-L1, PD-L2). Unbound secondary antibodies were removed by washing with Buffer Y, followed by washing once with Buffer X for 5 min. Post- fixation was performed with 2% paraformaldehyde in PBS for 5 min. Prior addition of gold fiducials, samples were washed with PBS.
- Binding molecule preselection Imaging Required steps: 1. Perform at least two-plex fluorescence imaging of the sample cells, by using two-plex DNA-PAINT or optionally by chromatic splitting using dichroic mirrors or filters, preferentially using light sheet, confocal, epifluorescence or TIRF microscopy. Continuation of the Specific Example: Prior to image acquisition, all fluorophores (e.g.
- CHO-mEGFP-ALFA-MHC-I, CHO-mCherry- CD80 were deactivated by a high intensity bleach pulse (488 nm, 150 mW at the sample plane, for 1 minute).
- Cellular imaging was conducted via two subsequent conventional DNA- PAINT imaging rounds (i.e. detection rounds) using distinct imagers for each binding molecule with only one of the imagers present at a time.
- Cy3B as the imaging molecule
- Cy3B-conjugated imagers were dissolved in Buffer Z and imager solution was added to the sample to perform conventional DNA-PAINT measurements. In between imaging rounds, the sample was washed with PBS until no residual signal from the previous imager solution was detected, followed by incubation of Buffer X for 5min.
- Binding molecule preselection determination Required steps: 1. Quantify the number of secondary binder molecules (e.g. secondary nanobody) bound to binding molecules, which in turn are bound to their target molecules. This can be done with various accuracies and precisions, e.g.
- o Using diffraction-limited fluorescence intensity of secondary binder in the test sample as a proxy for secondary binder molecule numbers, optionally in the image region confirmed to be covered by a cell; o Using diffraction-limited fluorescence intensity of the corresponding imager of secondary binder in the test sample, colocalizing with fluorescent protein signal as a proxy for secondary binder molecule numbers; o Performing conventional DNA-PAINT image analysis by identifying and localizing imager binding events and aggregating to secondary binder molecule localizations. Using secondary binder molecule counts as a proxy for secondary binder molecule numbers, optionally only those that are in an image region confirmed to be covered by a cell.
- o Using diffraction-limited fluorescence intensity of secondary binder in the control sample as a metric, optionally in the image region confirmed to be covered by a cell; o Using diffraction-limited fluorescence intensity of the corresponding imager of secondary binder in the test sample as a proxy, optionally in the image region confirmed to be covered by a cell; o Performing DNA-PAINT image analysis by identifying and localizing imager binding events and aggregating to secondary binder molecule localizations. Using secondary binder molecule counts as a proxy, optionally only those that are in an image region confirmed to be covered by a cell. 3. Calculate a metric for bare binding molecule selection, e.g.
- binding molecule density was evaluated by correlating respective integrated bulk intensity values to the background-corrected intensity value of a clearly defined monomeric single fluorophore and further calculating corresponding ratios between control and test sample as well as ratios between secondary binder and reference binder density in test sample only.
- binding molecule selection may be done via conventional DNA-PAINT.
- DNA-PAINT based determination of binding molecule selection raw fluorescence data were subjected to super-resolution reconstruction using the Picasso software package (latest version available at https://github.com/jungmannlab/picasso). Drift correction was performed with a redundant cross-correlation and gold particles as fiducials for cellular experiments.
- DNA-PAINT data were analysed using the Picasso clustering algorithm (latest version available at https://github.com/jungmannlab/picasso) for each target individually. Circular clusters of localizations centered around local maxima were identified and grouped (assigned a unique identification number). Binding molecule selection was evaluated by counting the number of secondary binder signals in control and test sample within the cell or optionally, in the image region confirmed to be covered by a cell, determining underlying secondary binder density and further corresponding ratios between control and test sample as well as ratios between secondary binder and reference binder density in test sample only. 1.2.
- a short DNA oligo target complementary nucleic acid
- a short DNA oligo target complementary nucleic acid
- the primary binder shows little off-target binding. This can be characterized by primary binder specificity testing.
- 1.2.1 Sample preparation for primary binder specificity imaging (Version 1). Required steps: 1. Seed cells that do not endogenously express proteins of interest on a microscopy slide. Split into control and test sample. 2. Transfect cells in test sample with a receptor construct, leading them to express a protein fusion of the target molecule (e.g.
- an epitope tag e.g. Alfa-tag, Halo-tag, FLAG-tag, His-Tag, sortase
- a fluorescent protein – or CRISPR edit e.g. Alfa-tag, Halo-tag, FLAG-tag, His-Tag, sortase
- a fluorescent protein – or CRISPR edit e.g. Alfa-tag, Halo-tag, FLAG-tag, His-Tag, sortase
- a fluorescent protein – or CRISPR edit e.g. Alfa-tag, Halo-tag, FLAG-tag, His-Tag, sortase
- a fluorescent protein – or CRISPR edit e.g. Alfa-tag, Halo-tag,
- meGFP-ALFA-MHC-I meGFP-ALFA-MHC-I
- meGFP-ALFA-MHC-II meGFP-ALFA-CD86
- meGFP-ALFA-CD80 meGFP-ALFA-PD-L1, meGFP-ALFA-PD-L2
- Lipofectamine LTX Lipofectamine LTX as specified by the manufacturer.
- CHO cells were allowed to express meGFP-ALFA-receptors for 16–24 h. Then, the medium was replaced with fresh F-12K Medium + 10% FBS + 100U/ml Penicilin and 100 ⁇ g/ml Streptomycin followed by fixation.4% PFA solution was preheated to 37°C before addition to the cells.
- Non-transfected CHO cells served as a reference.
- DNA-conjugated antibodies CD80, CD86, MHC-I, MHC-II, PD-L1, PD-L2
- primary binders were dissolved in blocking buffer and added at a final concentration of 100nM each for 90min at 24°C. Unbound primary binders were removed by washing with Buffer Y, followed by washing once with Buffer X for 5 min.
- MutuDC 1940 wt were seeded on ibidi 8 Well high Glass Bottom chambers at a density of 20000 cells per cm 2 several hours prior fixation.
- MutuDC1940 KO (MutuDC1940 MHC-I KO, MutuDC1940 MHC-II KO, MutuDC1940 CD86 KO, MutuDC1940 CD80 KO, MutuDC1940 PD-L1 KO, MutuDC1940 PD-L2 KO) cells served as a reference.
- DNA-conjugated antibodies (CD80, CD86, MHC-I/MHC-I OVA, MHC-II, PD-L1, PD-L2) were dissolved in blocking buffer and added at a final concentration of 100nM each for 90min at 24°C. Unbound antibodies were removed by washing with Buffer Y, followed by washing once with Buffer X for 5 min. Post-fixation was performed with 2% paraformaldehyde in PBS for 5 min. Prior addition of gold fiducials, samples were washed with PBS. Subsequently, 250 ⁇ l of 90 nm standard gold nanoparticles, diluted 1:3 in PBS, were added and incubated for 5 min before washing with PBS.
- DNA-PAINT imaging for primary binder specificity determination (Version 1). This step may be performed after Version 1 of sample preparation for primary binder specificity imaging.
- meGFP-ALFA-MHC-I meGFP-ALFA-MHC-I
- meGFP-ALFA-MHC-II meGFP- ALFA-CD86, meGFP-ALFA-CD80, meGFP-ALFA-PD-L1, meGFP-ALFA-PD-L2
- was conducted via imaging single target receptors using distinct imagers for each primary binder Prior to imaging a high intensity bleach pulse was applied until no residual signal from meGFP was observable. Cy3b-conjugated imager strands were dissolved in Buffer Z and imager solution was added to the sample to perform DNA-PAINT measurements. 1.2.4 DNA-PAINT imaging for primary binder specificity determination (Version 2). This step may be performed after Version 2 of sample preparation for primary binder specificity imaging.
- Imager Imager Cluster Table 6 Imaging parameters for primary binder specificity imaging.
- Underlying primary binder density was evaluated by correlating respective integrated and background corrected bulk intensity values to the background-corrected intensity value of a clearly defined monomeric single fluorophore and further calculating corresponding ratios between control and test sample.
- raw fluorescence data were subjected to super-resolution reconstruction using the Picasso software package (latest version available at https://github.com/jungmannlab/picasso).
- Drift correction was performed with a redundant cross-correlation and gold particles as fiducials for cellular experiments.
- gold particles were also used to align all rounds for 2-plex Exchange-PAINT experiments.
- DNA-PAINT data were analysed using the Picasso clustering algorithm (latest version available at https://github.com/jungmannlab/picasso) for each target individually. Circular clusters of localizations centred around local maxima were identified and grouped (assigned a unique identification number). Primary binder specificity was evaluated by counting the number of binder signals in control and test sample within the cell or alternatively, in the image region confirmed to be covered by a cell, determining underlying binder density and further corresponding ratios between control and test sample. 1.3. Primary binder labelling efficiency testing For sample analysis, it may be beneficial to correct the observed data for incomplete binding of primary binders to target molecules. For example, primary binder labelling efficiency may be tested.
- Sample preparation for primary binder labelling efficiency imaging Required steps: 1. Seed cells that do not endogenously express proteins of interest (i.e. target molecules) on a microscopy slide. 2. Transfect cells with a receptor construct which leads them to express a protein fusion of the target molecule of interest (e.g. MHI-I, MHC-II, CD86, CD80, PD-L1, PD-L2), and one or more targets for reference labeling (e.g. epitope tags like Alfa-tag, Halo-tag, FLAG-tag, His- Tag, sortase or fluorescent proteins) – or CRISPR edit. 3. Incubate cells until target receptor construct will be expressed. 4. Cell fixation and permeabilization. 5. Incubate primary binders. 6.
- targets for reference labeling e.g. epitope tags like Alfa-tag, Halo-tag, FLAG-tag, His- Tag, sortase or fluorescent proteins
- the one or more reference targets in either case being conjugated to a DNA-oligo comprising a docking sequence for a complementary nucleic acid sequence being labeled by an imaging molecule orthogonal to other complementary nucleic acid sequence being labeled by imaging molecules used, where the different epitope tags and or fluorescent proteins may be tagged with the same or different docking sequences; those must be orthogonal to the docking sequence used in 5.
- CHO cells were seeded on ibidi 8 Well high Glass Bottom chambers the day prior to transfection at a density of 15000 cells per cm2.
- CHO cells were transfected with a single receptor construct (meGFP-ALFA-MHC-I, meGFP-ALFA-MHC-II, meGFP-ALFA-CD86, meGFPALFA-CD80, meGFP-ALFA-PD-L1, meGFP-ALFA-PD-L2) for primary binder characterization using Lipofectamine LTX as specified by the manufacturer.
- CHO cells were allowed to express eGFP-ALFA-receptors for 16–24 h. Then, the medium was replaced with fresh F-12K Medium + 10% FBS + 100U/ml Penicilin and 100 ⁇ g/ml Streptomycin followed by fixation.4% PFA solution was preheated to 37°C before addition to the cells. Cells were fixed in 4% PFA for 15 minutes and washed with PBS. Cells were permeabilized in 0.125% TritonX- 100 dissolved in PBS for 5 minutes, washed with PBS followed by surface passivation with blocking buffer for 60min at 24°C.
- DNA-conjugated antibodies (CD80, CD86, MHC-I, MHC-II, PD-L1, PD-L2) – primary binders – and ALFA-tag nanobody – reference binders – were dissolved in blocking buffer and added at a final concentration of 100nM each for 90min at 24°C. Unbound primary and reference binders were removed by washing with Buffer Y, followed by washing once with Buffer X for 5 min. Post-fixation was performed with 2% paraformaldehyde in PBS for 5 min. Prior addition of gold fiducials, samples were washed with PBS.
- Cellular imaging was conducted via two subsequent imaging rounds using distinct imagers for each primary binder (Table 7) with only one of the imagers present at a time. Cyb3-labeled imagers were dissolved in Buffer Z and imager solution was added to the sample to perform DNA-PAINT measurements. In between imaging rounds, the sample was washed with PBS until no residual signal from the previous imager solution was detected followed by incubation of Buffer X for 5min. Then, the next imager solution was introduced. Imaging parameters are listed in detail in Table 7.
- Image Clust Clust Table 7 Imaging parameters for binder labelling efficiency imaging.
- primary binders will be selected which specifically tag target molecules.
- the number of primary binders found to be bound to target molecules can be corrected for the labelling efficiency of the reference labels (this is more beneficial if only one reference label is used; with more reference labels, the overall labelling efficiency gets closer to 100%, which renders the correction less beneficial).
- Diffraction-limited analysis option For Diffraction-limited based determination of binder labelling efficiency, background corrected integrated fluorescence intensity values were determined from raw fluorescence data for cellular experiments. Prior to integration, fiducial markers (e.g. gold particles) were used to align channels of target binder and reference in the test sample of 2-plex experiments.
- DNA-PAINT analysis alternative For DNA-PAINT based determination of primary binder labelling efficiency, raw fluorescence data were subjected to super-resolution reconstruction using the Picasso software package (latest version available at https://github.com/jungmannlab/picasso). Drift correction was performed with a redundant cross-correlation and gold particles as fiducials for cellular experiments. Further gold particles were used to align all subsequent imaging rounds for 2- plex Exchange-PAINT experiments.
- DNA-PAINT data were analysed using the Picasso clustering algorithm (latest version available at https://github.com/jungmannlab/picasso) for each target individually. Circular clusters of localizations centered around local maxima were identified and grouped (assigned a unique identification number).
- Primary binder labelling efficiency was determined as follows: 1. Cross-nearest neighbour distances were determined for primary binder and reference binder, corresponding histograms were plotted and colocalizing contributions of primary binder and reference binder were determined based on comparison of experimental and simulated data. Underlying labelling efficiency was extracted by simulating different contributions of randomly distributed monomers (reference) and dimers (target + reference) and fitting respective contributions to optimally fit distribution of experimental data.
- Respective cut-off distance was based on the molecular size of used target primary binder and reference primary binder accounting for the underlying binding epitope of both, target and reference primary binder.
- Cross-nearest neighbour distances were determined for primary and reference binders, corresponding histograms were plotted and colocalizing contributions of primary and reference binders were determined based on comparison of experimental and simulated data. Underlying labelling efficiency was extracted by a multi-gaussian fit of simulated CSR distribution of dimers only (target + reference) or monomers only (reference) to optimally fit experimental data.
- Off-reference and Off-target binding was accounted for by randomly removing signals according to percentage of non-specific binding from respective region of interest with the labeling efficiencies determined beforehand. 2.
- Sample preparation for multiplexed immune receptor DNA-PAINT For the analysis of target molecule patterns, samples have to be prepared for imaging. This can be done in multiple ways and also depends on the sample type.
- Vesicle refers to a spheroid compartment of aqueous solution with a lipid bilayer as a boundary and a diameter smaller than 100 ⁇ m.
- Glass coverslips were plasma cleaned for 3 min and attached to the bottom side of an 8-well chamber slide. Coverslips were incubated with a fivefold diluted vesicle solution for 10 min at 24°C, before they were extensively rinsed with PBS. For specific and efficient cell attachment sequential two-step incubation procedure is required. First, SLBs were incubated with DNA-modified lipids (e.g. cholesterol) at a final concentration of 100nM for 60min at 30°C followed by washing excessive lipids off with PBS.
- DNA-modified lipids e.g. cholesterol
- FIG. 10 shows the general concept of the preparation steps according to a preparation method, exemplarily performed in the Specific Example: 1. Contacting a coverslip, preferentially a glass coverslip, with an aqueous solution comprising SUVs. 2. This results in a supported lipid bilayer (SLB) supported by the coverslip. 3.
- histidine-tagged adhesion proteins e.g. His 10 -tag ICAM-1
- HBSS HBSS supplemented with 2% FBS, 2mM CaCl2 and 2mM MgCl2 prior cell seeding.
- Figure 10 shows the general concept of the preparation steps according to a preparation method, exemplarily performed in the Specific Example: 1. Contacting a coverslip, preferentially a glass coverslip, with an aqueous solution comprising SUVs. 2. This results in a supported lipid bilayer (SLB) supported by the coverslip. 3.
- SLB supported lipid bilayer
- Incubate sample with primary binders against multiple target molecules Incubate sample with primary binders against multiple target molecules.
- stimulatory reagents e.g. CpG1826, IFN ⁇ , ovalbumin
- fiducial markers e.g. gold nanoparticles
- Post-fixation was performed for 5min at ambient temperatures using distinct post-fixation buffers for different kinds of binding molecules (e.g.2% paraformaldehyde in PBS for antibody-based imaging).
- samples Prior to addition of gold fiducials, samples were washed with PBS. Subsequently, 250 ⁇ l of 90 nm standard gold nanoparticles, diluted 1:3 in PBS, were added and incubated for 5 min before washing with PBS.
- 3 Data acquisition describes the imaging procedure for multiplexed molecular imaging of targets.
- 3.1 Multiplexed cellular imaging of immune receptors Required steps: 1. Flush imager solution for the current target molecule into the sample.
- Imager concentration 1pM to 10nM, more preferably 50pM to 500pM; in Buffer, preferably buffer Z, B+ or C+, most preferably buffer Z. 2. Acquire movie of transient imager binding events; exposure time 1ms to 1s, more preferably 50ms-200ms; number of frames 5.000-100.000, more preferably 10.000-30.000. 3. Repeat steps 1&2 for all target molecules. Optional steps: 1. Deactivation of remaining fluorophores, e.g. GFP, for example via high-energy epifluorescent illumination, such as 150 mW at 488 nm for 1 min. 2. Wash sample between imaging rounds, e.g. to deplete sample of imagers of preceding imaging round. 3.
- Cy3b) imager strands (also called complementary nucleic acid sequence being labeled by an imager) were dissolved in Buffer Z and the imager solution was added to the sample to perform DNA- PAINT measurements.
- the result of DNA-PAINT measurements was raw imaging data in the 5 form of transient binding movies. In between imaging rounds, the sample was washed with PBS until no residual signal from the previous imager solution was detected followed by incubation of Buffer X for 5min. Then, the next imager solution was introduced. Imaging parameters for DNA-PAINT cell experiments are listed in detail in Table 3. 10 Power (at Ima er Ima er Ima er Cluster AGAGAGA 20 20 a e : mmune ecep o magng pa ame e s. 4.
- the raw imaging data needs to be processed. This is described here.
- 4.1 Image analysis Here, the raw super-resolution imaging data is postprocessed.
- the step describes getting from multiple single channel transient binding movies to one multiplexed molecular map, which specifies the localizations of all target molecules detected in the sample.
- Circular clusters of localizations centered around local maxima were identified and grouped (assigned a unique identification number). Subsequently, the centers of the localization groups were calculated as weighted mean by employing the squared inverse localization precisions as weights. These centers represent the single-protein positions of the respective imaging round. Merging localizations of all rounds yields the final multiplexed DNA-PAINT data/image (multiplexed molecular map).
- the data needs to be aggregated to elucidate the direct interaction patterns present in the sample. This step describes getting from the multiplexed molecular map to one or more direct interaction patterns present in the sample.
- a direct interaction pattern describes a set of target molecules commonly found in close proximity, and optionally probability distributions of their relative distances.
- Ripley The interaction of receptor molecules was assessed via modified version of Ripley’s K function [as disclosed, e.g., at https://rss.onlinelibrary.wiley.com/doi/10.1111/j.2517- 6161.1977.tb01615.x].
- Ripley K function as ⁇ ⁇ ⁇ where ⁇ is the density of points, ⁇ ⁇ ⁇ ⁇ points within radius ⁇ of the i-th point ⁇ ⁇ , and the sum is taken over ⁇ points.
- the cell area was determined via the total area of bins constituting the cell, and the density by the total number of points divided by the cell area.
- For normalization of Ripley’s K curves we subtracted the obtained mean from each curve, and normalized the data such that the 2.5 and 97.5 percentiles corresponded to values of –1 and 1, respectively.
- the 95% confidence interval of the integral for complete spatial randomness is given by [ ⁇ ⁇ , ⁇ ], where ⁇ is the length of the integration interval.
- the patternbar in the corresponding Figures indicates integral values scaling from 2000 (max. clustering) to -2000 (max. dispersion).
- NND Nearest-Neighbor Distance
- the surface density of each receptor species is calculated from the number of receptors found on the cell surface area. The latter is measured via masking the multiplexed DNA-PAINT image. Then, a completely spacially randomly (CSR) distributed dataset is generated for each receptor species at the respective experimental protein density. The data points of the CSR distribution are placed within an area defined by the cell outline. Subsequently, the DBSCAN analysis is performed on the “in silico” dataset in the same way as for the experimental data. Finally, the properties of protein clusters in the cell and in the CSR scenario can be compared.
- Dominating receptor motifs were identified by grouping individual clusters based on their receptor species, weighted by underlying area and plotted as normalized histograms (mean ⁇ 95% CI). Frequencies of all 63 receptor motifs were then compared to respective CSR distribution and tested for significance. In analogy, receptor contributions for all significant receptor motifs were quantified, compared to respective CSR distributions and tested for significance.
- FIG.11B shows an Exchange-PAINT schematic.
- Exchange-PAINT uses orthogonal strands linked to target molecules 1 (here: receptors) and sequential imaging and washing of complementary imagers for multiplexed super-resolution; see Jungmann et al., Nature Methods, 11:313–318 (2014).
- a legend of the symbols indicating the different types of receptors is provided in Figs.11D and 11E.
- the multiplexed Exchange-PAINT imaging was used to simultaneously map the MHC class I (H2-K b protein) and MHC class II molecules, the co-stimulatory receptors CD86 and CD80 and the inhibitory checkpoint receptors PD-L1 and PD-L2 on the surface of individual mouse conventional DCs (MutuDC cDC1 cell line) and melanoma cells (B16-F10 cells) at the single protein level.
- sequential imaging with different imager strands labelled with the same fluorophore (Cy3b) was utilized to visualize respective immune checkpoint receptors simultaneously on target cells, overcoming the classical diffraction limit using DNA-PAINT as a single-molecule localization technique.
- FIG.11C shows the 6-plex Exchange-PAINT image of a MutuDC stimulated for 6 h with CpG and IFN ⁇ .
- the zoom-ins in the right column depict diffraction- limited vs. super-resolution representation (top vs. middle).
- Subsequent spot analysis allows digitization of receptor molecules and reveals their molecular arrangement (bottom).
- individual receptors could be clearly identified in the respective DNA-PAINT image.
- Figure 11D shows morphological changes of MutuDC stimulated with CpG and IFN ⁇ over 24 h at molecular resolution.
- Zoom-ins (bottom row) at multiple different time-points (0 h, 3 h, 6 h, 12 h, 24 h) reveal the spatial details of key receptor interactions.
- Quantitative analysis of morphology and the underlying receptors enabled measurement of absolute cell surface area15 (Fig.11E, table 9) and absolute receptor density (Fig.11F, table 9) over time (24 h stimulation), with a peak overall receptor density observed at 6 hours after beginning of stimulation in MutuDC (for the mean over multiple experiments).
- (G) Global DBSCAN analysis is used to identify receptor motifs within a 35 nm search radius in clustered regions containing at least three receptors. A dominating exemplary motif is shown in the zoom-in.
- Figure 12C shows a correlation matrix for all 36 possible receptor combinations, which allows classification into “clustered”, “random” or “dispersed” receptor distributions.
- Fig.12D which shows 10 different interactions.
- nearest neighbor distance analysis See Fig.12E: Representative whole-cell analysis of first nearest neighbor distances (short: NND) of MHC-I to CD80 receptors (black) and PD-L2 (white) are shown in histograms with a total area of 1. The fit of simulations to these data is shown as solid and dashed lines, respectively.
- Fig.12F This allowed us to determine the percentage of molecular interactions for each receptor pair (Fig.12F), accounting for the underlying labeling efficiency of the target binders (fig.9).
- the numbers represent the fraction of receptors in the group that interact with the receptors on the x-axis. For example, 49% of the MHC-I molecules (left-most cluster in Fig. 12F) are in an interaction cluster with CD80. Prior to stimulation most molecules were not clustered and only minor CD86/CD86 and CD80/CD80 interactions were observed (Fig.16). Starting at 3 h and becoming most prominent at 6h stimulation, there was a substantial remodelling of receptor interactions, particularly interactions involving CD80 (Fig.12F, Fig.16 - 20).
- PD-L2 was either randomly distributed or mildly dispersed from other molecules, while MHC-II was strongly dispersed away from all other imaged molecules. This may imply that MHC-II and PD-L2 are genuinely dispersed away from protein-protein interactions, or that they are recruited into other complexes containing factors that are not being imaged. Nevertheless, small MHC- II clusters were observed at 12h after stimulation (Fig. 19), although they dispersed again at 24h (Fig.20). We next wished to determine if clusters containing 3 or more proteins could be identified. By applying DBSCAN to the receptor centers in the multiplexed DNA-PAINT data independent of the receptor identity, key receptor motifs could be identified (Fig.12G, Fig.18).
- FIG.12G Global DBSCAN analysis is used to identify receptor motifs within a 35 nm search radius in clustered regions containing at least three receptors.
- a dominating exemplary motif is shown in the zoom-in of fig.12G.
- Quantification of receptor motifs identified CD86/CD80/PD-L1 hetero-trimers and MHC-I/CD86/CD80/PD-L1 hetero-tetramers as dominating interaction motifs on dendritic cells stimulated for 6 hours (see Figure 12H, the motif represents 5.9% ⁇ 2.1% of all clusters; black filled bars represent the number of respective receptors expected to occur in the area of the motive due to a spatially random distribution based on their observed density, without motif interaction; white (empty) bars represent the observed receptors in the cluster; only contributors that have statistically significant values are included in the motif.
- MutuDCs were stimulated for 6h with CpG and IFN ⁇ in the presence of the model protein antigen ovalbumin, and instead of imaging total MHC-I, we specifically measured MHC-I (H-2K b protein) bound to the SIINFEKL ovalbumin peptide antigen using a specific detection antibody. Similar receptor motifs were identified containing peptide antigen-bound MHC-I molecules, indicating that specific, cross-presented antigens acquired by DCs can be incorporated into these surface motifs (Fig. 21). Collectively, these data support a surface architecture on DCs that favours T cell activation, and identify a large number of previously unknown surface protein interactions of key immune molecules. 5.3.
- FIG.13A shows morphological changes of stimulated B16-F10 cells (see table 9 for the number of cells analyzed) over 24 h. Zoom-ins (bottom row) at multiple different time-points (0 h, 3 h, 6 h, 12 h, 24 h) reveal key receptor interaction patterns.
- Figure 13C shows the dynamics of overall receptor density and respective species contributions in B16-F10s (solid outline) vs. MutuDCs (dashed outline) over 24 h stimulation. Overall receptor density is almost identical for dendritic and cancer cells, significant differences can be observed for individual receptor densities. Consistent with this visible difference in surface architecture, the pairwise clustering patterns between proteins on melanoma cells (Fig. 13D: Correlation matrix of all 36 possible receptor combinations allows identification of key receptor interactions in B16-F10 cells.) was noticeably different from that of DCs (Fig.12C), with a strong enrichment of PD-L1 and MHC- I clustering and a paucity of CD80 interactions (likely due to its low surface abundance) (Fig.13D).
- FIG.13E is a visualization of corresponding receptor interactions in B16-F10 cells (top) using a river plot, which reveals homo-interactions of MHC-I, MHC-II and PD-L1. Most significant however are MHC-I/PD-L1 hetero-interactions.
- a comparison to MutuDCs (bottom) reveals downregulation of costimulatory receptors (mainly CD80) as driving force for formation of MHC-I/PD-L1 clusters.
- FIG.13F,G shows the quantification of dominating receptor interactions in B16- F10 and a comparison to MutuDC (if possible) shows significant differences in MHC-I, MHC-II and PD-L1 interactions.
- Figure 13G shows exemplary zoom-ins of key receptor interactions in B16-F10. Data are shown as the mean ⁇ 95% confidence interval. *** p ⁇ 0.001.
- MHC-I and PD-L1 homodimers were seen on melanoma cells, and unlike on DCs where MHC-II was exclusively non-clustered, MHC-II showed pronounced self-association on melanoma cells.
- Quantification of key receptor motifs similarly identified MHC-I/PD-L1 clusters as the dominant receptor motif on melanoma cells. Strikingly, even on non-stimulated B16-F10 cells, MHC-I/PD-L1 heteroclusters accounted for 26.4% of all detected clusters, and at 6h this increased to almost 60% of all identified receptor motifs detected on the cell surface (Fig.22-24).
- MHC-II was recruited to MHC-I/PD-L1 clusters (Fig. 25-26).
- B16-F10 line used for imaging transgenically expressed ovalbumin we could again determine if specific peptide antigen-MHC-I complexes similarly incorporate into these structures, and we found comparable PD-L1 clustering with antigen-bound MHC-I as for total MHC-I (Fig. 27).
- Fig. 27 there are profound differences in surface organisation of immune checkpoint molecules on melanoma cells relative to DCs, with structures that favour PD-L1 recruitment into the synapse upon antigen recognition enriched on cancer cells.
- FIG 14A top row, schematic sketches of our understanding of relevant receptor interactions (MHC-I - CD80 - PD- L1; with the receptor identified by signs according to the legend) as revealed by the analysis presented herein are illustrated for cDC1, cDC1 CD80 knock out, B16-F10 and CD80 overexpressing B16-F10 either expressing the physiological costimulatory receptor CD80 or the L107E mutant CD80, lacking its PD-L1 binding domain.
- Crossed-out receptors indicate knock-out, upward-facing arrows annotated with ‘High’ indicate overexpression, downward- facing arrows annotated with ‘Low’ indicate a low expression level, an encircled minus indicates a noninteracting mutant.
- Horizontal arrows indicate interaction where one-sided arrows indicate that the interaction is driven by one receptor (e.g. in the case of CD80 overexpression, the interaction between CD80 and PD-L1 is driven by CD80).
- Fig.14A morphological differences are shown for all cells together with corresponding zoom-ins to illustrate key receptor interactions at molecular resolution at 6 h stimulation. Strikingly, CD80 deletion was sufficient to precipitate MHC-I/PD-L1 clustering on DCs at levels that were comparable to those observed on melanoma cells (Fig.14B, Fig.29, 30).
- FIG 14B receptor interactions are visualized via a circle plot; Receptor species are positioned at the corners of a hexagon, with the circle size proportional to the average receptor density and connections between interacting receptors marked by lines with the line thickness being proportional to the average interaction score derived from pairwise correlation analysis), with this pattern already detectable prior to DC stimulation (Fig.31-32). Similar patterns were seen in wild-type (Fig.16, 18, 21) and CD80 knock-out MutuDCs (Fig. 33). Conversely, retroviral over-expression of CD80 on melanoma cells (Fig. 34) completely disrupted MHC-I/PD-L1 clustering alongside reinstating PD-L1/CD80 interactions (Fig.14B).
- CTLA4-Ig fusion reagents such as Abatacept
- CD80-blocking immunosuppressive reagents that are used in the clinic to treat autoinflammatory conditions, such as rheumatoid arthritis, juvenile idiopathic arthritis, and psoriatic arthritis. While the predominant mode of action is presumed to be due to CD80 blockade, CTLA4-Ig fusion proteins are also known to disrupt the interaction between CD80 and PD-L1, suggesting that PD-L1 liberation may contribute to clinical efficacy.
- CD80 strongly interacts with MHC-I and PD-L1 in untreated dendritic cells (as shown on the left side in a cartoon, as a representative cluster, and in the interaction network, where the interaction arrows between PD-L1 and CD80 as well as MHC-I and CD80 are strong).
- PD-L1 is released from CD80 and clusters with MHC-I (as shown on the rights side in a cartoon, as a representative cluster, and in the interaction network, where the interaction of PD-L1 with CD80 is much lower than on the left, but a strong interaction of PD-L1 with MHC- I can be observed).
- MHC-I/PD-L1 clustering While DCs showed relatively low levels of MHC-I/PD-L1 clustering (typical distance between MHC-I and PD-L1 of about 30 nm), B16-F10 cells displayed a significant increase in MHC/PD-L1 heterodimers (typical distance between MHC-I and PD-L1 of about 15 nm) which could limit T cell activation (left).
- MHC-I and PD- L1 were arranged into clusters separated by either a small (close, 10-15 nm) or larger (far, 25- 30 nm) distance, as observed on B16-F10 and DCs, respectively (middle).
- DNA origami structures presenting no (empty) or only stimulatory ligands (pMHCs) were included for comparison (right).), and interrogated the primary T cell response.
- MHC-II molecular family member MHC-I, where clustering supports prolonged signal propagation in CD8+ T-cells.
- the direct interaction map shows strong interactions between PD-L1 and MHC-I in the cancer cell, while this interaction is weak in the dendritic cell due to the presence of CD80.
- PD-L1 and CD86 and CD80 interact strongly in the dendritic cell and hardly in the cancer cell, even though all molecules are present on the membrane of both cells.
- cytotoxic bi- or multispecific compounds against PD-L1 and MHC-I together with MHC-II could be developed to specifically kill cancer cells by recruiting not only CD4+ Helper T-cells but also CD8+ Killer T-cells followed by further recruitment of multiple different immune cells.
- Discussion The higher order clustering of immune-regulatory ligands on the cell surface likely plays a critically important role in guiding T cell immunity, but our understanding of this aspect of immune cell biology is very limited.
- melanoma cells were enriched for higher order immunosuppressive MHC-I/PD-L1 aggregates, and other motifs that were not present at significant levels on DCs.
- CD80 was the key determinant of this surface architecture switch, as it was both necessary and sufficient to prevent MHC-I/PD-L1 aggregation.
- similar surface restructuring could be precipitated by a clinically approved immunosuppressive agent that disrupts the CD80/PD-L1 interaction (Abatacept), implicating surface remodelling as a contributing factor to clinical efficacy. Overall, this implicates changes in surface architecture as an important regulator of T cell immunity.
- Nanoscale surface organisation has typically been inferred through biochemical approaches, like co-immunoprecipitation, indirect methods such as Fluorescence Energy Resonance Transfer (FRET), or through structural studies. While these existing methodologies are powerful tools for probing protein-protein association, they are unable to spatially resolve complex clustering at the single protein level on the cell surface, particularly those clusters involving 3 or more species.
- Super-resolution imaging has the potential to address this knowledge gap, however prior methodologies were not sufficiently resolved or multiplexed. While current state-of-the -art super-resolution techniques have made major effort to break the molecular resolution barrier, the present invention overcame these limitations, and the value of higher-plex single protein imaging was illustrated.
- PD-1 engagement inhibits T cells through recruitment of the phosphatase SHP2, which can inhibit signalling downstream of both CD28 and TCR engagement (see https://pubmed.ncbi.nlm.nih.gov/16227604/; https://pubmed.ncbi.nlm.nih.gov/22641383/; https://pubmed.ncbi.nlm.nih.gov/28280247/; https://pubmed.ncbi.nlm.nih.gov/28280249/).
- CD80 as a central regulator of MHC-I/PD-L1 interactions that disrupts clustering by binding to PD-L1.
- CD80-bound PD-L1 is sequestered away from other proteins, which in turn has significant downstream effects on surface organisation.
- CD80 stripping may also contribute to regulatory T cell-mediated immunosuppression.
- CD80 in addition to co-stimulation through CD28, and inhibition of PD-L1, CD80 also remodels the cell surface into a configuration that favours T cell activation. More broadly, our results highlight the importance of considering spatial organisation in drug design. We demonstrate that the cell surface is in a finely tuned equilibrium, where small perturbations can drastically remodel surface architecture. We find that blocking reagents, such as those used for immunotherapy in autoimmunity and cancer, can mimick these effects. In the case of Abatacept, the surface changes precipitated by target binding will likely augment the intended immunosuppressive activity of this clinical agent. 7.
- Diagnostics development The results from the data analysis may be further used for drug development.
- DNA oligonucleotides modified with DBCO-PEG4 and Cy3B were ordered from IDT and MWG Eurofins.
- Sticky-slide 8 well chambers (80808) were purchased from Ibidi and glass slides (10756991) were purchased from Marienfeld. Double-sided tape (665D) was ordered from Scotch. FBS Good (P40-37500) was purchased from PAN Biotech. FBS Advanced (FBS-11A, heat inactivated) was ordered from Capricorn Scientific. His 10 -tag ICAM-1 (50440-M08H) was purchased from Biozol.
- EndoFit TM ovalbumin (vac-pova) and Primocin® (ant-pm-05) were obtained from InvivoGen.
- CpG1826 was ordered from Pfizer.
- Recombinant murine IFN ⁇ (315-05) was purchased from PeproTech.
- Ninety- nanometer gold nanoparticles (G-90-100) were ordered from Cytodiagnostics.
- High-binding 96-well plates (3361) were obtained from Corning.
- EasySep TM Mouse CD8+ T Cell Isolation Kit (19853) was purchased from STEMCELL Technologies. Buffer Recipes: • Buffer X: 1x PBS, 500mM NaCl. • Buffer Y: 1x PBS, 1mM EDTA, 0.01% Tween-20.
- Buffer Z 1x PBS, 1 mM EDTA, 500 mM NaCl (pH 7.4), 0.01%Tween-20 supplemented with 1x Trolox.
- Blocking buffer 1x PBS, 1mM EDTA, 0.02% Tween-20, 0.05% NaN3, 2% BSA, 0.05 mg/ml sheared salmon sperm DNA.
- Trolox 100x Trolox was made by adding 100 mg Trolox to 430 ⁇ l of 100% methanol and 345 ⁇ l of 1 M NaOH in 3.2 ml water.
- Hybridization buffer 4xSSC 10% Dextrane sulfate 10% ethylanecarbonate 0.04% Tween20 ⁇
- Dehybridization buffer 2xSSC 10% Dextrane sulfate 20% ethylanecarbonate 0.04% Tween20 ⁇
- Blocking Buffer 2 1x PBS, 3% BSA 0.25% Triton 0.05 mg/ml sheared salmon sperm DNA (DNA-PAINT) microscope setup: Fluorescence imaging was carried out on an inverted microscope (Nikon Instruments, Eclipse Ti2) with the Perfect Focus System, applying an objective-type TIRF configuration equipped with an oil-immersion objective (Nikon Instruments, Apo SR TIRF 100x, NA 1.49, Oil).
- a 560- nm laser (MPB Communications, 1 W) was used for excitation.
- the laser beam was passed through a cleanup filter (Chroma Technology, ZET561/10) and coupled into the microscope objective using a beam splitter (Chroma Technology, ZT561rdc). Fluorescence was spectrally filtered with an emission filter (Chroma Technology, ET600/50m and ET575lp) and imaged on an sCMOS camera (Andor, Zyla 4.2 Plus) without further magnification, resulting in an effective pixel size of 130 nm (after 2 x 2 binning). The readout rate was set to 540 MHz. Images were acquired by choosing a region of interest with a size of 512 x 512 pixels.
- SiteClickTM antibody-DNA conjugation Prior functionalization unconjugated antibodies (MHC-I, MHC-I OVA, MHC-II, PD-L2) were concentrated to 1mg/ml in Tris, pH 7.0, by using Amicon centrifugal filters (50,000 MWCO). For each conjugation 200 ⁇ g of respective antibody were used. Azide-modified antibodies were produced following manufacturer’s protocol. Azido-modified antibodies were reacted with 10x molar excess of DBCO- functionalized DNA (R2 - MHC-II, R5 - MHC-I/MHC-I OVA and R6 - PD-L2) in Tris, pH 7.0, overnight at 25°C, 300rpm.
- DBCO- functionalized DNA R2 - MHC-II, R5 - MHC-I/MHC-I OVA and R6 - PD-L2
- Unreacted DNA was removed by buffer exchange to PBS using Amicon centrifugal filters (50,000 MWCO). Unconjugated antibody were removed by anion exchange chromatography using an ⁇ KTA pure system equipped with a Resource Q 1-ml column and antibody concentration was adjusted to 5 ⁇ M. DNA-conjugated antibodies were stored at 4°C. Enzymatic antibody-DNA conjugation:Prior functionalization unconjugated antibodies (CD80, CD86, PD-L1) were concentrated to 1mg/ml in TBS + 0.05% Tween20 by using Amicon centrifugal filters (50,000 MWCO). For each conjugation 100 ⁇ g of respective antibody were used.
- PNGase 0.6 U
- mTG 1.2 U
- an 80-fold molar excess of bifunctional azide-PEG3- amine linker were added to the antibody and reacted for 16h at 37°C, 300rpm.
- Enzymes and excessive linker were removed by buffer exchange to PBS using Amicon centrifugal filters (50,000 MWCO).
- Azido-modified antibodies were reacted with 10x molar excess of DBCO- functionalized DNA (R1 – CD86, R3 – CD80 and R4 - PD-L1) overnight at 25°C, 300rpm.
- Unreacted DNA and unconjugated antibody were removed by anion exchange chromatography using an ⁇ KTA pure system equipped with a Resource Q 1-ml column and antibody concentration was adjusted to 5 ⁇ M.
- DNA-conjugated antibodies were stored at 4°C.
- ALFA-tag nanobody-DNA conjugation ALFA-tag nanobodies were conjugated as described previously1. Unconjugated nanobodies were thawed on ice, then 20-fold molar excess of bifunctional DBCO-PEG4-Maleimide linker was added and reacted for 2 h on ice. Unreacted linker was removed by buffer exchange to PBS using Amicon centrifugal filters (10,000 MWCO).
- the DBCO-modified nanobodies were reacted with 5xmolar excess of azide- functionalized DNA (R3, R4) overnight at 4°C. Unconjugated protein and free DNA were removed by anion exchange chromatography using an ⁇ KTA pure system equipped with a Resource Q 1-ml column. Preparation of functionalized planar SLBs.
- Vesicles containing 98% 1-palmitoyl-2-oleoyl-sn- glycero-3-phosphocholine (POPC) and 2% 1,2-dioleoyl-sn-glycero-3-[N(5-amino-1- carboxypentyl)iminodiaceticacid]succinyl[nickel salt] (Ni-DOGS NTA) were prepared at a total lipid concentration of 0.5mg ml-1 as described ( J. B. Huppa, et al., TCR-peptide-MHC interactions in situ show accelerated kinetics and increased affinity.
- PBS phosphate-buffered saline
- MutuDC1940 KO MHC-I KO, MHC-II KO, CD86 KO, CD80 KO, PD-L1 KO, PD-L2 KO cells served as a reference.
- Cells were stimulated using 500 nM CpG1826 + 100 U/ml IFN ⁇ ⁇ ovalbumin for 6 hours at 37°C.
- the 4% PFA solution was preheated to 37°C before addition to the cells. Cells were fixed in 4% PFA for 15 minutes and washed with PBS.
- Target binding molecules (anti-MHC-I/MHC-I OVA antibody, anti-MHC-II antibody, anti-CD86 antibody, anti- CD80 antibody, anti-PD-L1 antibody, anti-PD-L2 antibody) were dissolved in blocking buffer and added at a final concentration of 100 nM each overnight at 4°C. Unbound antibodies (binding molecules) were removed by washing with Buffer Y, followed by washing once with Buffer X for 5 min. Post-fixation was performed with 2% paraformaldehyde in PBS for 5 min.
- DNA origami preparation For folding DNA origami, 10 nM single-stranded DNA scaffold from M13p18 bacteriophages (SeqID No 3541, Tilibit GmbH cat.
- the folded origami structures were then purified from excess staples using 100 kDa MWCO centrifugal filters.
- Purified origami structures were stored in buffer C (PBS, 500 mM NaCl) at -20 °C until usage.
- buffer C PBS, 500 mM NaCl
- Sample preparation for binding molecule labeling efficiency imaging CHO cells were seeded on ibidi 8 Well high Glass Bottom chambers (Cat.No: 80807) the day prior to transfection at a density of 15 ⁇ 10 4 cells/well.
- CHO cells were transfected with a single receptor construct (mEGFP-ALFA-MHC-I, mEGFP-ALFA-MHC-II, mEGFP- ALFA-CD86, mEGFP-ALFA-CD80, mEGFP-ALFA-PD-L1, mEGFP-ALFA-PD-L2) at a time for binding molecule characterization using Lipofectamine LTX as specified by the manufacturer.
- CHO cells were allowed to express mEGFP-ALFA-receptors for 16–24 h. Then, the medium was replaced with fresh F-12K Medium + 10% FBS + 100 U/ml Penicillin + 100 ⁇ g/ml Streptomycin followed by fixation.
- Target binding molecules (anti-MHC-I/MHC-I OVA antibody, anti-MHC-II antibody, anti-CD86 antibody, anti-CD80 antibody, anti-PD-L1 antibody, anti-PD-L2 antibody) were dissolved in blocking buffer and added at a final concentration of 100 nM each overnight at 4°C followed by addition of ALFA-tag nanobody, dissolved in blocking buffer and added at a final concentration of 500 pM for 60 min at 24°C.. Unbound binders were removed by washing with Buffer Y, followed by washing once with Buffer X for 5 min. Post-fixation was performed with 2% paraformaldehyde in PBS for 5 min. Prior to the addition of gold fiducials, samples were washed with PBS.
- DNA-conjugated antibodies (anti-MHC-I/MHC-I OVA antibody, anti-MHC-II antibody, anti-CD86 antibody, anti-CD80 antibody, anti-PD-L1 antibody, anti-PD-L2 antibody) were dissolved in blocking buffer and added at a final concentration of 100 nM each overnight at 4°C. Unbound antibodies were removed by washing with Buffer Y, followed by washing once with Buffer X for 5 min. Post-fixation was performed with 2% paraformaldehyde in PBS for 5 min. Prior to the addition of gold fiducials, samples were washed with PBS.
- DNA origami discs (Fig. S32) were assembled as previously [https://pubmed.ncbi.nlm.nih.gov/34613711/].
- the staples corresponding to the functionalization points were extended at their 3′ end with a 21-nucleotide overhang of docking strands (SeqID No 3527-3532).
- Cy3B-conjugated imager strands were dissolved in Buffer Z and 600 ⁇ l of the imager solution was added to the sample to perform DNA-PAINT measurements. Imaging parameters are listed in detail in table 6. 2-plex Exchange-PAINT imaging for binding molecule labeling efficiency determination. Prior image acquisition, all fluorophores (e.g. CHO-mEGFP-ALFA-MHC-I) were deactivated by a high intensity bleach pulse. Cellular imaging was conducted via two subsequent imaging rounds using distinct imagers for each binding molecule (table 12) with only one of the imagers present at a time. Cy3B-conjugated imager strands were dissolved in Buffer Z and 600 ⁇ l of the imager solution was added to the sample to perform DNA-PAINT measurements.
- fluorophores e.g. CHO-mEGFP-ALFA-MHC-I
- Imaging parameters are listed in detail in table 7. Multiplexed cellular imaging of immune receptors. Prior image acquisition, all fluorophores (B16-F10 CD80- mCherry, B16-F10 CD80 (L107E)-mCherry) were deactivated by a high intensity bleach pulse.
- Multiplexed cellular imaging was conducted via six subsequent imaging rounds using the six imagers R1-R6 (as published in https://pubmed.ncbi.nlm.nih.gov/32601424/ ) with only one of the imagers present at a time. Cy3B- conjugated imager strands were dissolved in Buffer Z and 600 ⁇ l of the imager solution was added to the sample to perform DNA-PAINT measurements. In between imaging rounds, the sample was washed with 2 ml PBS until no residual signal from the previous imager solution was detected followed by incubation of Buffer X for 5 min. Then, the next imager solution was introduced. Imaging parameters for DNA-PAINT cell experiments are listed in detail in table 8. Microscope setup.
- Fluorescence imaging was carried out on an inverted microscope (Nikon Instruments, Eclipse Ti2) with the Perfect Focus System, applying an objective-type TIRF configuration equipped with an oil- immersion objective (Nikon Instruments, Apo SR TIRF ⁇ 100, NA 1.49, Oil).
- a 560-nm laser MPB Communications, 1 W was used for excitation.
- the laser beam was passed through a cleanup filter (Chroma Technology, ZET561/10) and coupled into the microscope objective using a beam splitter (Chroma Technology, ZT561rdc).
- the readout rate was set to 540 MHz. Images were acquired by choosing a region of interest with a size of 512 ⁇ 512 pixels. Detailed imaging conditions for the respective experiments are shown in table 6-8. Image analysis. Raw fluorescence data were subjected to super-resolution reconstruction using the Picasso software package (latest version available at https://github.com/jungmannlab/picasso).
- Drift correction was performed with a redundant cross-correlation and gold particles as fiducials for cellular experiments. Gold particles were also used to align all rounds for multiplexed Exchange-PAINT experiments.
- DNA- PAINT data were analyzed using the Picasso clustering algorithm (latest version available at https://github.com/jungmannlab/picasso) for each target individually. Circular clusters of localizations centered around local maxima were identified and grouped (assigned a unique identification number). Subsequently, the centers of the localization groups were calculated as weighted mean by employing the squared inverse localization precisions as weights. Merging localizations of all rounds yields the final multiplexed DNA-PAINT image. Data analysis – Binding molecule specificity.
- Binding molecule specificity was evaluated by counting the number of binding molecule signals (circular clusters of localizations centered around local maxima) in stimulated wild-type MutuDC1940 and MutuDC1940 KO samples within the cell area, determining underlying binding molecule density and further corresponding ratios between MutuDC1940 and MutuDC1940 KO samples. Data analysis - Labeling efficiency.
- NND nearest neighbor distance
- the algorithm of the simulation can be summarized as follows: 1. Parameters. Density of target monomers: number of target monomers per unit area. Density of reference monomers: number of reference monomers per unit area. Density of target-reference dimers: number of dimers per unit area. Dimer distance: expected distance between reference and target molecule including the labeling construct. Uncertainty: variability in the position of each molecule due to labeling and localization errors.
- the total density for target and reference is set to match the respective experimentally observed densities in each channel.
- Simulation of monomers a set of spatial coordinates with CSR distribution and given density are drawn.
- Simulation of dimers a set of spatial coordinates with CSR distribution are drawn, representing the center of each dimer. For each dimer center two positions are generated with a random orientation and a distance with expected value Dimer distance. The position of each pair of molecules are drawn taking into account the Uncertainty parameter (drawn from a gaussian distribution).
- NND are calculated on the subset of detectable molecules.
- CHO cells CCL-61, ATCC were cultured in GibcoTM Ham's F-12K (Kaighn's) Medium, supplemented with 10% Fetal Bovine Serum (FBS) (11573397, Gibco), 100 U/ml Penicillin, 100 ⁇ g/ml Streptomycin.
- FBS Fetal Bovine Serum
- Murine dendritic cell line (MutuDC 1940) was cultured in IMDM supplemented with 10% FBS, 100 ⁇ M ⁇ - Mercaptoethanol (Gibco), 100U/ml Penicilin, 100 ⁇ g/ml Streptomycin and 1.32 mM Glutamax (Gibco).
- Murine melanoma B16-F10melanoma cell line was cultured in Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% FBS, 100 U/ml Penicillin and 100 ⁇ g/ml Streptomycin. Cells were maintained and passaged using Accutase solution. Cloning.
- DMEM Modified Eagle Medium
- Receptor c-terminally tagged with ALFA-tag and mEGFP (MHC-I-ALFA-mEGFP, MHC-II-ALFA- mEGFP, CD86-ALFA-mEGFP, CD80-ALFA-mEGFP, PD-L1-ALFA-mEGFP, PD-L2-ALFA-mEGFP) were individually cloned into pcDNATM3.1 (+) Mammalian Expression Vector (Invitrogen Cat. No. V79020).
- Retroviral Transduction of B16-F10 The retroviral transduction involved the production of murine stem cell virus (MSCV) followed by the transduction of the B16-F10 cell line.
- MSCV murine stem cell virus
- MSCV vectors were produced in HEK293GP cells by co-transfection of packaging plasmids containing mCD80 L107E (IRES-mCherry), mCD80 (IRES- mCherry) and empty vector with the VSV-G envelope glycoprotein, using Lipofectamine 2000 (Invitrogen) for 6 hours. Following five days of culture in DMEM supplemented with 10% FBS, 100 U/ml Penicillin, 100 ⁇ g/ml Streptomycin and 1.32 mM Glutamax (Gibco), virus-containing supernatant was collected, filtered and frozen at -20°C.5 ⁇ 10 4 B16-F10 cells were seeded in each well of 6-well plate.
- the cells were transduced with 2ml of viral supernatant and 8 ug/ml polybrene (Sigma Aldrich) followed by 1-hour centrifugation at 500G. This was done twice a day for three days for a total of five viral hits.
- CD80-mCherry expression was validated by FACS and mCherry positive B16-F10 cells were sorted using FACSAria Fusion Flow Cytometer (BD Biosciences). Flow cytometry data was analyzed using FlowJo (v10.8.1, Treestar). CRISPR/Cas9 gene editing.
- the CRISPR editing protocol for the MutuDC1940, B16-F10 and primary bone marrow dendritic cells was adapted from https://pubmed.ncbi.nlm.nih.gov/32152070/.
- sgRNAs targeting the murine MHC-I, MHC-II, CD86, CD80, PD-L1, PD-L2 and non-targeting Ctrl genes were obtained from Integrated DNA Technology (sequences of each target sgRNA is described in table 11).
- 5 ⁇ 10 6 cells were electroporated for the MutuDC1940 cell lines (MHC-I, MHC-II, CD86, CD80, PD-L1, PD-L2 and non-targeting Ctrl), for the B16-F10 cell lines (CD80 and non-targeting Ctrl) and for the primary bone marrow cells (CD80 and non-targeting Ctrl), and 100 ⁇ 10 6 cells were electroporated for the primary bone marrow cells.
- sgRNA/Cas9 RNP complex formation Cas9 protein and sgRNA were combined and incubated at room temperature for 10 minutes.
- Bone marrow cells were collected from the femurs and tibia of C57BL/6 mice. Red blood cells were lysed using RBC lysis buffer (Ammonium Chloride) for 2 minutes and washed in media. Cells were filtered and spun down at 200G for 7 minutes. Cells were counted and divided for KO experiments using sgRNAs targeting the murine CD80 and nontargeting Ctrl sgRNA (Integrated DNA Technologies).
- primary bone marrow cells were cultured at 1.5 ⁇ 10 6 cells/ml for 8 days in RPMI 1640 media (ThermoFisher Scientific) supplemented with 1.32 mM Glutamax, 10% FBS, 90 ⁇ M ⁇ -mercaptoethanol, 100 U/ml Penicillin, 100 ⁇ g/ml Streptomycin and 150 ng/ml Flt3L (BioXCell). Following 8 days of culture, cells were stimulated using 500 nM CpG1826 (Pfizer) + 100 U/ml IFN (Peprotech) for 6 hours in suspension.
- both unstimulated and stimulated cells were incubated in Fc Block (BD Biosciences) for 10 minutes on ice, stained for 30 minutes using B220 (PE, Clone R836B2, BD #553090), SIRP ⁇ (BV510, Clone P84, BD #740159), CD11c (BV785, Clone N418, BioLegend #117336), CD24 (BUV395, Clone M1/69, BD #744471) antibodies.
- B220- CD11c high CD24 high SIRP ⁇ low cDC1s were then sorted using FACSAria Fusion Flow Cytometer (BD Biosciences).
- CD80 deletion was validated on a separate sample using CD80 (APC, Clone 16-10A1, BioLegend #104714) antibody. Following the sort, the cells were washed in media and resuspended in Hank’s Balanced Salt Solution (HBSS) ((H8264-500ML, Sigma Aldrich).) supplemented with 2% FBS, 2 mM CaCl2 and 2 mM MgCl2 and seeded onto the SLB. I m r S Imager name Imager Table 12. Imager sequences used for binder characterization. It is emphasized that in the examples for a method of mapping the localization of different target molecules within a sample DNA-PAINT was used as the imaging method.
- HBSS Hank’s Balanced Salt Solution
- DNA-PAINT shows advantageous characteristics for the analyzing of direct interaction patterns, due the combination of molecular resolution and high degree of multiplexing. Nonetheless, other super-resolution fluorescence imaging techniques, such as dSTORM, may be used as well. It is also emphasized that not only the conventional DNA-PAINT techniques may be used but also the inventive concepts presented in this disclosure. With respect to molecular resolution it is stressed that the analysis in the methods of mapping the localization of different target molecules within a sample according to the present invention analysis is preferably based on data with such a high resolution that proteins that touch each other can be spatially separated, i.e. the resolution is about 5 nm. This means that imaging data can be interpreted as and transformed into primary binder localizations or target molecule positions. Nonetheless, cluster analysis of lower-resolution imaging data and/or analysis of imager localizations may also be used and yield usable results.
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Microbiology (AREA)
- Immunology (AREA)
- Physics & Mathematics (AREA)
- Molecular Biology (AREA)
- Biotechnology (AREA)
- Biophysics (AREA)
- Analytical Chemistry (AREA)
- Biochemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Genetics & Genomics (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Abstract
Description
Claims
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| EP23170811 | 2023-04-28 | ||
| EP23170811.6 | 2023-04-28 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024223837A1 true WO2024223837A1 (en) | 2024-10-31 |
Family
ID=86282689
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/EP2024/061562 Pending WO2024223837A1 (en) | 2023-04-28 | 2024-04-26 | Dna-paint related materials and methods |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2024223837A1 (en) |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2025132913A1 (en) * | 2023-12-22 | 2025-06-26 | Massive Photonics Gmbh | Bispecific compound for dna-paint and method |
Citations (8)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US4946778A (en) | 1987-09-21 | 1990-08-07 | Genex Corporation | Single polypeptide chain binding molecules |
| US6080560A (en) | 1994-07-25 | 2000-06-27 | Monsanto Company | Method for producing antibodies in plant cells |
| WO2008022759A2 (en) | 2006-08-21 | 2008-02-28 | Eidgenoessische Technische Hochschule Zürich | Specific and high affinity binding proteins comprising modified sh3 domains of fyn kinase |
| US20100129796A1 (en) * | 2008-11-24 | 2010-05-27 | Micah Halpern | Dye probe fluorescence resonance energy transfer genotyping |
| US20120088246A1 (en) * | 2010-10-07 | 2012-04-12 | Samsung Techwin Co., Ltd. | Real time pcr detection of single nucleotide polymorphisms |
| US20200087707A1 (en) * | 2018-09-14 | 2020-03-19 | The Broad Institute, Inc. | Oligonucleotide-coupled antibodies for single cell or single complex protein measurements |
| EP3670666A1 (en) * | 2018-12-20 | 2020-06-24 | Freie Universität Berlin | Method for optical imaging a target molecule in a sample, a protein-oligonuleotide conjugate for use in the method and a kit for carrying out the method |
| WO2023172734A1 (en) * | 2022-03-11 | 2023-09-14 | President And Fellows Of Harvard College | Multiplex fluorescent cellular and tissue imaging with dna encoded thermal channels and uses thereof |
-
2024
- 2024-04-26 WO PCT/EP2024/061562 patent/WO2024223837A1/en active Pending
Patent Citations (8)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US4946778A (en) | 1987-09-21 | 1990-08-07 | Genex Corporation | Single polypeptide chain binding molecules |
| US6080560A (en) | 1994-07-25 | 2000-06-27 | Monsanto Company | Method for producing antibodies in plant cells |
| WO2008022759A2 (en) | 2006-08-21 | 2008-02-28 | Eidgenoessische Technische Hochschule Zürich | Specific and high affinity binding proteins comprising modified sh3 domains of fyn kinase |
| US20100129796A1 (en) * | 2008-11-24 | 2010-05-27 | Micah Halpern | Dye probe fluorescence resonance energy transfer genotyping |
| US20120088246A1 (en) * | 2010-10-07 | 2012-04-12 | Samsung Techwin Co., Ltd. | Real time pcr detection of single nucleotide polymorphisms |
| US20200087707A1 (en) * | 2018-09-14 | 2020-03-19 | The Broad Institute, Inc. | Oligonucleotide-coupled antibodies for single cell or single complex protein measurements |
| EP3670666A1 (en) * | 2018-12-20 | 2020-06-24 | Freie Universität Berlin | Method for optical imaging a target molecule in a sample, a protein-oligonuleotide conjugate for use in the method and a kit for carrying out the method |
| WO2023172734A1 (en) * | 2022-03-11 | 2023-09-14 | President And Fellows Of Harvard College | Multiplex fluorescent cellular and tissue imaging with dna encoded thermal channels and uses thereof |
Non-Patent Citations (27)
| Title |
|---|
| ALTSHULER EPSEREBRYANAYA DVKATRUKHA AG., BIOCHEMISTRY (MOSC)., vol. 75, no. 13, 2010, pages 1584 |
| BERTSCHINGER ET AL., PROTEIN ENG DES SEL, vol. 20, no. 2, 2007, pages 57 - 68 |
| BESTE GSCHMIDT FSSTIBORA TSKERRA A., PROC NATL ACAD SCI USA., vol. 96, no. 5, 1999, pages 1898 - 903 |
| BILA ET AL., J AM CHEM SOC., vol. 144, no. 47, 30 November 2022 (2022-11-30), pages 21576 - 21586 |
| BRAASCHCOREY, CHEM BIOL, vol. 8, 2001, pages 1 |
| CHRISTOPH SPAHN ET AL: "Protein-Specific, Multicolor and 3D STED Imaging in Cells with DNA-Labeled Antibodies", ANGEWANDTE CHEMIE, WILEY - V C H VERLAG GMBH & CO. KGAA, DE, vol. 131, no. 52, 7 November 2019 (2019-11-07), pages 19011 - 19014, XP071379588, ISSN: 0044-8249, DOI: 10.1002/ANGE.201910115 * |
| COLE ET AL., ALAN R. LISS, INC, 1985, pages 77 - 96 |
| GEBAUERSKERRA, CURR OPINION IN CHEMICAL BIOLOGY, vol. 13, 2009, pages 245 - 255 |
| GRABULOVSKI ET AL., JBC, vol. 282, 2007, pages 3196 - 3204 |
| HOLLIGER PHUDSON PJ., NAT BIOTECHNOL., vol. 23, no. 9, 2005, pages 11265 |
| J. B. HUPPA ET AL.: "TCR-peptide-MHC interactions in situ show accelerated kinetics and increased affinity", NATURE, vol. 463, 2010, pages 963 - 967, XP055629814, DOI: 10.1038/nature08746 |
| JUNGMANN ET AL., NATURE METHODS, vol. 11, 2014, pages 313 - 318 |
| KONTERMANNBRINKMANN, DRUG DISCOVERY TODAY, vol. 20, no. 7, 2015, pages 838 - 847 |
| KOZBOR D, IMMUNOLOGY TODAY, vol. 4, 1983, pages 7 |
| LI J ET AL., PNAS, vol. 103, no. 10, 2006, pages 3557 |
| MOERTELMAIER, M.BRAMESHUBER, M.LINIMEIER, M.SCHUTZ, G. J.STOCKINGER, H.: "Thinning out Clusters While Conserving Stoichiometry of Labeling", APPL. PHYS. LETT., vol. 87, no. 26, 2005, pages 1 - 3, XP012077129, DOI: 10.1063/1.2158031 |
| MOURATOU BBEHAR GPAILLARD-LAURANCE LCOLINET SPECORARI F., METHODS MOL BIOL., vol. 805, 2012, pages 315 - 31 |
| OLA SÖDERBERG ET AL: "Characterizing proteins and their interactions in cells and tissues using the in situ proximity ligation assay", METHODS, ACADEMIC PRESS, NL, vol. 45, no. 3, 1 July 2008 (2008-07-01), pages 227 - 232, XP002638191, ISSN: 1046-2023, DOI: 10.1016/J.YMETH.2008.06.014 * |
| SCHLATTER, MABS, vol. 4, no. 4, 2012, pages 1 - 12 |
| SCHMIDT, T.SCHUTZ, G. J.GRUBER, H. J.SCHINDLER, H.: "Local Stoichiometries Determined by Counting Individual Molecules", ANAL. CHEM., vol. 68, no. 24, 1996, pages 4397 - 4401, XP002131497, DOI: 10.1021/ac960710g |
| SCHNITZBAUER ET AL., NATURE PROTOCOLS, vol. 12, 2017, pages 1198 - 1228 |
| SIMMEL ET AL., CHEM. REV., vol. 119, no. 10, 2019, pages 6326 - 6369 |
| STEINHAUER ET AL., ANGEWANDTE CHEMIE INT., vol. 48, no. 47, 2009, pages 8870 - 8873 |
| STRAUSS SEBASTIAN ET AL: "Up to 100-fold speed-up and multiplexing in optimized DNA-PAINT", NATURE METHODS, NATURE PUBLISHING GROUP US, NEW YORK, vol. 17, no. 8, 29 June 2020 (2020-06-29), pages 789 - 791, XP037208335, ISSN: 1548-7091, [retrieved on 20200629], DOI: 10.1038/S41592-020-0869-X * |
| STRAUSSJUNGMANN, NATURE METHODS, vol. 17, 2020, pages 789 - 791 |
| WEIDLE UH ET AL., CANCER GENOMICS PROTEOMICS, vol. 10, no. 4, 2013, pages 155 - 68 |
| YURKE ET AL., NATURE, vol. 406, no. 6796, 2000, pages 605 - 608 |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2025132913A1 (en) * | 2023-12-22 | 2025-06-26 | Massive Photonics Gmbh | Bispecific compound for dna-paint and method |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US20250346947A1 (en) | Multiplexed single molecule rna visualization with a two-probe proximity ligation system | |
| Savino et al. | The nucleolar antigen Nop52, the human homologue of the yeast ribosomal RNA processing RRP1, is recruited at late stages of nucleologenesis | |
| CN106170564B (en) | Proximity assays based on detection of Hybrid Chain Reaction (HCR) | |
| JP2023524733A (en) | Compositions, systems and methods for creating, identifying and characterizing effector domains for activating and silencing gene expression | |
| US20100212040A1 (en) | Isolation of living cells and preparation of cell lines based on detection and quantification of preselected cellular ribonucleic acid sequences | |
| JP7456637B2 (en) | Lipid-modified oligonucleotides and methods of use thereof | |
| JP2024541968A (en) | Systems and methods for identifying MHC-associated antigens for therapeutic intervention - Patents.com | |
| WO2024223837A1 (en) | Dna-paint related materials and methods | |
| Huang et al. | CRISPR-dCas13-tracing reveals transcriptional memory and limited mRNA export in developing zebrafish embryos | |
| AU2018304396B2 (en) | Methods and compositions for viral Nano-FISH | |
| EP3821033B1 (en) | Use of droplet single cell epigenome profiling for patient stratification | |
| US20220281931A1 (en) | Chemically inducible polypeptide polymerization | |
| Wang et al. | A subcellular selective APEX2-based proximity labeling used for identifying mitochondrial G-quadruplex DNA binding proteins | |
| US7838243B2 (en) | Protein forming complex with c-Jun protein, nucleic acid encoding the same and method of using the same | |
| EP2186827A1 (en) | Surrogate marker directed cDNA cloning of selectively induced mRNAs | |
| Fraser et al. | Quantitative studies of ribosome conformational dynamics | |
| JP5430090B2 (en) | Antibody fragment screening method and assay system | |
| JP4478772B2 (en) | Protein that forms a complex with c-Fos protein, nucleic acid encoding the same, and methods for using them | |
| AU2011203213A1 (en) | Selection and Isolation of Living Cells Using mRNA-Binding Probes | |
| Sakong et al. | Electrostatic properties of disordered regions control transcription factor search and pioneer activity | |
| Bellone et al. | Structural Characterization of Protein–Nucleic Acid Complexes: An Overview of the Recent Innovation in the Analytic Methods | |
| Mukherjee et al. | Decoding the base flipping mechanism of the SET-and RING-associated (SRA) domain of the epigenetic UHRF1 protein | |
| HK40107624A (en) | Multiplexed single molecule rna visualization with a two-probe proximity ligation system | |
| Willemin | Multiplexed Immunodetection by Sequencing for Protein Quantification In Genome Architecture Mapping Technologies | |
| JP5753134B2 (en) | Selection and isolation of viable cells using mRNA binding probes |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24722251 Country of ref document: EP Kind code of ref document: A1 |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2024722251 Country of ref document: EP |
|
| ENP | Entry into the national phase |
Ref document number: 2024722251 Country of ref document: EP Effective date: 20251128 |
|
| ENP | Entry into the national phase |
Ref document number: 2024722251 Country of ref document: EP Effective date: 20251128 |
|
| ENP | Entry into the national phase |
Ref document number: 2024722251 Country of ref document: EP Effective date: 20251128 |