WO2024006392A1 - Analyse d'acides nucléiques et de protéines à l'aide de sondes - Google Patents

Analyse d'acides nucléiques et de protéines à l'aide de sondes Download PDF

Info

Publication number
WO2024006392A1
WO2024006392A1 PCT/US2023/026504 US2023026504W WO2024006392A1 WO 2024006392 A1 WO2024006392 A1 WO 2024006392A1 US 2023026504 W US2023026504 W US 2023026504W WO 2024006392 A1 WO2024006392 A1 WO 2024006392A1
Authority
WO
WIPO (PCT)
Prior art keywords
probe
sequence
nucleic acid
molecule
tissue
Prior art date
Application number
PCT/US2023/026504
Other languages
English (en)
Inventor
Katherine Pfeiffer
Andrew Scott KOHLWAY
Andrew John HILL
Paul Eugene LUND
Jawad N. ABOUSOUD
Sharmila CHATTERJEE BHATTACHARJEE
Ryan STOTT
Original Assignee
10X Genomics, Inc.
Priority date (The priority date 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 date listed.)
Filing date
Publication date
Application filed by 10X Genomics, Inc. filed Critical 10X Genomics, Inc.
Priority to US18/217,310 priority Critical patent/US20240002914A1/en
Priority to US18/370,826 priority patent/US20240019422A1/en
Publication of WO2024006392A1 publication Critical patent/WO2024006392A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING 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
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6841In situ hybridisation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING 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
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6816Hybridisation assays characterised by the detection means
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70539MHC-molecules, e.g. HLA-molecules
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1037Screening libraries presented on the surface of microorganisms, e.g. phage display, E. coli display
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1055Protein x Protein interaction, e.g. two hybrid selection
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1065Preparation or screening of tagged libraries, e.g. tagged microorganisms by STM-mutagenesis, tagged polynucleotides, gene tags
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1075Isolating an individual clone by screening libraries by coupling phenotype to genotype, not provided for in other groups of this subclass
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING 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
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6804Nucleic acid analysis using immunogens
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING 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
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6806Preparing nucleic acids for analysis, e.g. for polymerase chain reaction [PCR] assay
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING 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
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6816Hybridisation assays characterised by the detection means
    • C12Q1/6818Hybridisation assays characterised by the detection means involving interaction of two or more labels, e.g. resonant energy transfer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING 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
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6827Hybridisation assays for detection of mutation or polymorphism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING 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
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6881Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for tissue or cell typing, e.g. human leukocyte antigen [HLA] probes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N1/00Sampling; Preparing specimens for investigation
    • G01N1/28Preparing specimens for investigation including physical details of (bio-)chemical methods covered elsewhere, e.g. G01N33/50, C12Q
    • G01N1/30Staining; Impregnating ; Fixation; Dehydration; Multistep processes for preparing samples of tissue, cell or nucleic acid material and the like for analysis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • G01N33/5032Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects on intercellular interactions
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/505Cells of the immune system involving T-cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/5302Apparatus specially adapted for immunological test procedures
    • G01N33/5304Reaction vessels, e.g. agglutination plates
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/5306Improving reaction conditions, e.g. reduction of non-specific binding, promotion of specific binding
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/5308Immunoassay; Biospecific binding assay; Materials therefor for analytes not provided for elsewhere, e.g. nucleic acids, uric acid, worms, mites
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/54306Solid-phase reaction mechanisms
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/54366Apparatus specially adapted for solid-phase testing
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/544Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals the carrier being organic
    • G01N33/548Carbohydrates, e.g. dextran
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56966Animal cells
    • G01N33/56977HLA or MHC typing
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/10Applications; Uses in screening processes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING 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
    • C12Q2537/00Reactions characterised by the reaction format or use of a specific feature
    • C12Q2537/10Reactions characterised by the reaction format or use of a specific feature the purpose or use of
    • C12Q2537/164Methylation detection other then bisulfite or methylation sensitive restriction endonucleases
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING 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
    • C12Q2563/00Nucleic acid detection characterized by the use of physical, structural and functional properties
    • C12Q2563/179Nucleic acid detection characterized by the use of physical, structural and functional properties the label being a nucleic acid
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING 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
    • C12Q2563/00Nucleic acid detection characterized by the use of physical, structural and functional properties
    • C12Q2563/185Nucleic acid dedicated to use as a hidden marker/bar code, e.g. inclusion of nucleic acids to mark art objects or animals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING 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
    • C12Q2565/00Nucleic acid analysis characterised by mode or means of detection
    • C12Q2565/10Detection mode being characterised by the assay principle
    • C12Q2565/101Interaction between at least two labels
    • C12Q2565/1015Interaction between at least two labels labels being on the same oligonucleotide
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B30/00Methods of screening libraries
    • C40B30/04Methods of screening libraries by measuring the ability to specifically bind a target molecule, e.g. antibody-antigen binding, receptor-ligand binding
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B50/00Methods of creating libraries, e.g. combinatorial synthesis
    • C40B50/06Biochemical methods, e.g. using enzymes or whole viable microorganisms
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B70/00Tags or labels specially adapted for combinatorial chemistry or libraries, e.g. fluorescent tags or bar codes

Definitions

  • a sample may be processed for various purposes, such as identification of a type of moiety within the sample.
  • the sample may be a biological sample.
  • Biological samples may be processed, such as for detection of a disease (e.g., cancer) or identification of a particular species.
  • PCR polymerase chain reaction
  • Biological samples may be processed within various reaction environments, such as partitions.
  • Partitions may be wells or droplets.
  • Droplets or wells may be employed to process biological samples in a manner that enables the biological samples to be partitioned and processed separately.
  • droplets may be fluidically isolated from other droplets, enabling accurate control of respective environments in the droplets.
  • Biological samples in partitions may be subjected to various processes, such as chemical processes or physical processes. Samples in partitions may be subjected to heating or cooling, or chemical reactions, such as to yield species that may be qualitatively or quantitatively processed.
  • Bio molecules such as nucleic acids and proteins, within biological samples may be probed and/or processed for quantitative or qualitative assessment.
  • the present disclosure provides methods for use in sample processing and analysis.
  • the methods provided herein may involve hybridizing a probe to a molecule of interest (e.g., target protein, target nucleic acid molecule) and processing the probe-molecule complex.
  • processing can include barcoding the probe, the probe-molecule complex, or the molecule, and/or performing a nucleic acid reaction.
  • the probe may comprise a nucleic acid molecule, and further processing can include extension, denaturation, and amplification processes to provide nucleic acid molecules comprising a sequence the same or substantially the same as or complementary to that of a target region of a nucleic acid molecule of interest (e.g., target nucleic acid molecule).
  • a method may comprise hybridizing a first probe and a second probe to first and second target regions of the nucleic acid molecule, linking the first and second probes to provide a probe-linked nucleic acid molecule, and barcoding the probe-linked nucleic acid molecule.
  • a method may comprise hybridizing a first probe to a first target region of a nucleic acid molecule, barcoding the probe, and hybridizing a second probe to a second target region of the nucleic acid molecule to generate a barcoded, probe-linked nucleic acid molecule.
  • the method may comprise hybridizing a probe to a nucleic acid molecule attached to a feature-binding moiety to provide a probe-binding moiety complex and barcoding the probe.
  • One or more processes of the methods provided herein may be performed within a partition such as a droplet or well.
  • the methods of the present disclosure be useful, for example, in controlled analysis and processing of analytes such as biological particles, nucleic acids, and proteins.
  • One or more of the methods described herein may allow for genomic, transcriptomic, or exomic profiling with higher sensitivity.
  • the methods of the present disclosure may be useful in detecting variants and characterizing nucleic acid molecules, e.g., for assessment of single nucleotide polymorphisms (SNPs), alternative splice junctions, insertions, deletions, V(D)J rearrangements, etc.
  • the methods of the present disclosure may be useful for multiplexed analysis of nucleic acids and proteins while minimizing reagent usage, e.g., by decreasing the number of unoccupied partitions for analysis.
  • a method for multiplexed nucleic acid assays comprising: (a) contacting a cell, nucleus, or cell bead with a first probe, a second probe, and a third probe under conditions sufficient to generate a first probe-associated molecule and a second probe-associated molecule, wherein the cell, nucleus, or cell bead comprises (i) a nucleic acid molecule comprising a first target region and a second target region and (ii) a feature coupled to a feature binding group, wherein the feature binding group comprises (i) a reporter oligonucleotide associated with the feature and (ii) a feature probe binding sequence, wherein the first probe comprises (i) a first probe sequence complementary to the first target region and (ii) a probe capture sequence, wherein the second probe comprises a second probe sequence complementary to the second target region, wherein the third probe comprises a (i) third probe sequence complementary to the feature probe binding sequence and (ii)
  • the first target region and the second target region are on a same strand of the nucleic acid molecule.
  • the probe capture sequence is common to a plurality of first probes including the first probe, wherein one or more additional partitions of the first set of partitions comprise one or more additional probe-associated nucleic acid molecules, wherein each of the one or more additional probe-associated nucleic acid molecules comprises the probe capture sequence.
  • the second probe comprises a second probe capture sequence complementary to a capture sequence of the plurality of capture molecules, and wherein (c) comprises hybridizing the second probe capture sequence to the capture sequence.
  • the barcode molecules comprise a capture binding sequence complementary to a capture sequence of the plurality of capture molecules, and wherein (c) comprises hybridizing the capture binding sequence to the capture sequence.
  • the first set of partitions are a plurality of wells.
  • the second set of partitions are a plurality of droplets.
  • the second set of partitions are a plurality of wells.
  • the plurality of capture molecules is coupled to a particle.
  • the particle is a bead.
  • the bead is a gel bead.
  • each capture molecule of the plurality of capture molecules coupled to the gel bead comprises the second barcode sequence.
  • one or more additional partitions of the second set of partitions comprise one or more additional gel beads of a plurality of gel beads, and wherein the second barcode sequence is unique to the gel bead among the plurality of gel beads.
  • a capture molecule of the plurality of capture molecules comprises a third barcode sequence unique to the capture molecule among the plurality of capture molecules.
  • one or more additional partitions of the second set of partitions comprise one or more additional capture molecules, and wherein the second barcode sequence is unique to the second partition among the second set of partitions.
  • (a) comprises hybridizing the first probe and the second probe to the first target region and the second target region, respectively.
  • the method further comprises subjecting the first probe-associated molecule to conditions sufficient to yield a probe-linked nucleic acid molecule comprising the first probe linked to the second probe.
  • the probe-linked nucleic acid molecule is generated via chemical or enzymatic ligation of the first probe and the second probe.
  • the chemical or enzymatic ligation occurs subsequent to (b).
  • the first target region and the second target region are adjacent.
  • the first target region and the second target region are non-adjacent, and the method further comprises (i) extending the first probe or the second probe annealed to the first target region or the second target region, respectively, towards the second target region or the first target region, respectively, to generate an extended probe, and (ii) ligating the extended probe to the second probe or the first probe, respectively.
  • (a) comprises contacting the first probe and the second probe to the nucleic acid molecule inside the cell or nucleus.
  • the first partition comprises a plurality of cells, cell beads, or nuclei.
  • the cell or nucleus is permeabilized.
  • the cell or nucleus is fixed.
  • the method further comprises releasing the first probe-associated molecule, or derivative thereof, from the cell, nucleus, or cell bead. In some embodiments, the releasing comprises lysing the cell. In some embodiments, the reporter oligonucleotide comprises the feature probe binding sequence. In some embodiments, the method further comprises, subsequent to (b) and prior to (c), pooling the first barcoded nucleic acid molecule, the second barcoded nucleic acid molecule, additional first barcoded nucleic acid molecules from the first set of partitions, and additional second barcoded nucleic acid molecules form the first set of partitions.
  • the method further comprises subsequent to (c) and prior to sequencing, pooling the third barcoded nucleic acid molecule, the fourth barcoded nucleic acid molecule, additional third barcoded nucleic acid molecules from the second set of partitions, and additional fourth barcoded nucleic acid molecules from the second set of partitions.
  • the probe capture sequence is from 8 to 50 bp.
  • a method comprising: (a) contacting a nucleic acid molecule with a first probe to generate a probe-associated nucleic acid molecule, wherein the nucleic acid molecule comprises a first target region and a second target region not adjacent to the first target region, wherein the first probe comprises a first probe sequence complementary to the first target region; (b) extending the first probe under conditions sufficient to generate an extended probe molecule comprising a sequence complementary to the second target region; (c) in a partition of a plurality of partitions, providing the extended probe molecule, a second probe, and a barcode molecule and a probe binding molecule, under conditions sufficient to generate a barcoded molecule, wherein the second probe comprises a second probe sequence corresponding to the second target region, wherein the first probe or the second probe comprises a probe capture sequence, wherein the barcode molecule comprises (i) a barcode capture sequence and (ii) a barcode sequence, wherein the probe
  • (c) comprises ligating the second probe and the barcode molecule.
  • the ligating comprises chemical or enzymatic ligation.
  • the particle is a bead.
  • the bead is a gel bead.
  • the cell or nucleus is permeabilized.
  • the cell or nucleus is fixed.
  • the first probe comprises the probe capture sequence.
  • the method further comprises, subsequent to (b), releasing the extended probe molecule from the nucleic acid molecule.
  • the releasing comprises freeing a ribonucleic acid (RNA) strand using RNase.
  • the releasing comprises heat cycling.
  • (c) comprises (i) hybridizing the second probe sequence of the second probe to the sequence complementary to the second target region and (ii) extending the second probe.
  • the barcode molecule and the probe binding molecule are provided as a pre-annealed complex, wherein the barcode capture sequence is annealed to the barcode binding sequence in the pre-annealed complex.
  • the first probe comprises the probe capture sequence
  • (c) comprises (i) annealing the probe binding sequence and the barcode binding sequence to the probe capture sequence and the barcode capture sequence, respectively (ii) ligating the barcode molecule and the extended probe molecule, to generate a first barcoded molecule and (iii) annealing the second probe to the first barcoded molecule and initiating an extension reaction to generate the barcoded molecule.
  • (ii) and (iii) are preformed outside the partition.
  • the second probe comprises the probe capture sequence
  • (c) comprises (i) annealing the second probe to the extended probe molecule and initiating an extension reaction to generate an extension molecule, (ii) annealing the probe binding sequence and the barcode binding sequence to the probe capture sequence and the barcode capture sequence, and (iii) ligating the barcode molecule and the extension molecule.
  • a method of analyzing a sample comprising: (a) providing: (i) a feature-binding group bound to at least a portion of the sample, wherein the feature-binding group comprises a reporter oligonucleotide, wherein the reporter oligonucleotide comprises a reporter barcode sequence, a first target region and a second target region, wherein the first target region and the second target region are disposed on a same strand of the reporter oligonucleotide; (ii) a first probe comprising a first probe sequence, wherein the first probe sequence of the first probe is complementary to the first target region of the reporter oligonucleotide; and (iii) a second probe comprising a second probe sequence, wherein the second probe sequence of the second probe is complementary to the second target region of the reporter oligonucleotide; (b) subjecting the sample to conditions sufficient to (i) hybridize the first probe sequence of the first probe to the first
  • the method further comprises (d) attaching a barcode molecule to the probe-linked nucleic acid molecule. In some embodiments, (d) occurs in a partition. In some embodiments, the partition is a droplet or a well. In some embodiments, the sample comprises a nucleic acid molecule, and wherein (d) further comprises attaching an additional barcode molecule to the nucleic acid molecule or derivative thereof. In some embodiments, the feature-binding group is an antibody. In some embodiments, the first probe or the second probe comprises an additional probe sequence. In some embodiments, the method further comprises attaching a barcode sequence to the additional probe sequence.
  • the method further comprises (d) providing a barcode molecule and a probe binding molecule comprising (i) a first sequence complementary to the additional probe sequence and (ii) a second sequence complementary to a capture sequence of the barcode molecule. In some embodiments, the method further comprises providing conditions sufficient to hybridize the first sequence to the additional probe sequence and the second sequence to the capture sequence of the barcode molecule, thereby generating a barcoded probe-associated complex. In some embodiments, (d) occurs in a partition among a plurality of partitions.
  • the method further comprises pooling the barcoded probe-associated complex from the partition with other barcoded probe-associated complexes from other partitions of the plurality of partitions to generate a pooled set of barcoded probe-associated complexes.
  • the method further comprises (i) partitioning the pooled set of barcoded probe- associated complexes into a plurality of additional partitions, wherein an additional partition of the plurality of additional partitions comprises the barcoded probe-associated complex and an additional barcode molecule comprising an additional barcode sequence and (ii) attaching the additional barcode molecule to the barcoded probe-associated complex.
  • the additional barcode molecule is coupled to a bead.
  • the bead is a gel bead.
  • the additional barcode molecule is releasably coupled to the bead.
  • (d) occurs prior to (c).
  • (d) occurs subsequent to (c).
  • the at least the portion of the sample comprises a feature.
  • the feature is a protein.
  • the protein is a cell surface receptor or an intracellular protein.
  • the sample comprises a cell or cell bead.
  • the cell is a formalin fixed, paraffin-embedded cell.
  • (c) occurs in a partition.
  • the partition is among a plurality of partitions.
  • (c) comprises enzymatic or chemical ligation. In some embodiments, the ligation is performed in the absence of adenosine triphosphate. In some embodiments, the first probe or the second probe comprises an adenylated end, a phosphorylated end, a ribonucleotide, a dideoxynucleotide, or a flap sequence. In some embodiments, the first target region and the second target region are separated by a gap region disposed between the first target region and the second target region. In some embodiments, (c) comprises performing an extension reaction to fill the gap region to yield the probe-linked nucleic acid molecule.
  • (c) comprises providing a third probe comprising a third probe sequence complementary to the gap region, hybridizing the third probe sequence to the gap region, and providing conditions sufficient to yield the probe-linked nucleic acid molecule comprising the first probe linked to the second probe via the third probe.
  • the present disclosure provides a method for analyzing a nucleic acid molecule of an embedded, fixed tissue, comprising: (a) providing the embedded, fixed tissue, wherein: (i) the embedded, fixed tissue comprises a plurality of cells, (ii) a cell of the plurality of cells comprises the nucleic acid molecule, and (iii) the embedded, fixed tissue is embedded in a solid medium; (b) removing at least a portion of the solid medium from the embedded, fixed tissue, thereby obtaining a fixed tissue comprising the plurality of cells; (c) dissociating the fixed tissue into cells of the plurality of cells, wherein the cells comprise the cell comprising the nucleic acid molecule; and (d) in the cell, using a barcode sequence to generate a barcoded nucleic acid molecule comprising a barcode sequence and a sequence corresponding to the nucleic acid molecule.
  • the fixed tissue is fixed at least about 1 year prior to (a). In some embodiments, the fixed tissue is fixed at least about 5 years prior to (a). In some embodiments, the method further comprises, in (b), adding a first solvent to the embedded, fixed tissue, thereby dissolving the solid medium, thereby obtaining a fixed tissue.
  • the first solvent is a non-polar solvent.
  • the non-polar solvent comprises xylene. In some embodiments, the non-polar solvent comprises neo-clear® xylene alternative. In some embodiments, the method further comprises removing the first solvent from the fixed tissue. In some embodiments, the method further comprises adding a second solvent to the fixed tissue.
  • the second solvent is a polar solvent. In some embodiments, the polar solvent comprises ethanol. In some embodiments, the method further comprises removing the second solvent from the fixed tissue. In some embodiments, the method further comprises adding a rehydration agent to the fixed tissue. In some embodiments, the rehydration agent comprises water. In some embodiments, the method further comprises removing the rehydration agent from the tissue. In some embodiments, the method further comprises adding buffer to the fixed tissue. In some embodiments, the method further comprises removing the buffer from the fixed tissue. In some embodiments, the method further comprises, in (c), dissociating the tissue. In some embodiments, the method further comprises resuspending the tissue in a supernatant.
  • the method further comprises filtering the tissue. In some embodiments, the method further comprises washing the tissue. In some embodiments, the method further comprises resuspending the tissue. In some embodiments, the dissociating the tissue is performed using an automated dissociator. In some embodiments, said automated dissociator is a gentleMACSTM Octo Dissociator. In some embodiments, the dissociating the tissue is performed using a manual pulverizer. In some embodiments, (b) and (c) provide a yield of cells of at least about 1 x 10 5 cells per two 25 micrometer sections of the fixed tissue. In some embodiments, the fixed tissue comprises fixed human tissue.
  • the fixed tissue comprises one or more of fixed connective tissue, fixed epithelial tissue, fixed organ tissue, fixed muscle tissue, fixed ligaments, fixed tendons, fixed a skin tissue, fixed breast tissue, fixed bladder, fixed kidney tissue, fixed liver tissue, fixed colon tissue, fixed thyroid tissue, fixed cervical tissue, fixed prostate tissue, fixed lung tissue, fixed cardiac tissue, fixed heart tissue, fixed muscle tissue, fixed pancreas tissue, fixed anal tissue, fixed bile duct tissue, fixed a bone tissue, fixed bone marrow, fixed uterine tissue, fixed ovarian tissue, fixed endometrial tissue, fixed vaginal tissue, fixed vulvar tissue, fixed stomach tissue, fixed ocular tissue, fixed nasal tissue, fixed sinus tissue, fixed penile tissue, fixed salivary gland tissue, fixed gut tissue, fixed gallbladder tissue, fixed gastrointestinal tissue, fixed bladder tissue, fixed brain tissue, fixed spinal tissue, fixed neurons, fixed cells representative of a blood-brain barrier, fixed blood, fixed hair, fixed nails, fixed keratin, or fixed collagen.
  • (d) comprises hybridizing the barcode sequence to the nucleic acid molecule.
  • the method further comprises extending the barcode to form the barcoding nucleic acid molecule.
  • the extending comprises the use of an enzyme.
  • the extending comprises ligating the barcode sequence to a probe.
  • the probe comprises a portion at least partially complementary to the nucleic acid molecule.
  • the method further comprises, prior to (d), partitioning the nucleic acid molecule into a partition.
  • the partition is a well of a plurality of wells.
  • the partition is a droplet of a plurality of droplets.
  • the plurality of droplets comprises an oil droplet.
  • the partition comprises a support.
  • the support comprises a bead.
  • the barcode sequence is attached to the support.
  • the solid medium comprises paraffin.
  • the fixed tissue comprises a formalin fixed tissue embedded in the paraffin.
  • the nucleic acid molecule comprises a ribonucleic acid (RNA) molecule.
  • the nucleic acid molecule comprises a messenger RNA molecule.
  • the present disclosure provides a method for analyzing a nucleic acid molecule of an embedded, fixed tissue, comprising: (a) providing the embedded, fixed tissue, wherein: (i) the embedded, fixed tissue comprises a plurality of cells, (ii) a cell of the plurality of cells comprises the nucleic acid molecule, and (iii) the embedded, fixed tissue is embedded in a solid medium; (b) removing at least a portion of the solid medium from the embedded, fixed tissue, by adding a first solvent to the embedded, fixed tissue, thereby dissolving the solid medium thereby obtaining a fixed tissue comprising the plurality of cells; (c) removing the first solvent from the fixed tissue; (d) adding a second solvent to the fixed tissue; (e) removing the second solvent from the fixed tissue; (f) adding a rehydrated agent to the fixed tissue; (g) removing the rehydration agent from the fixed tissue; (h) adding buffer to the fixed tissue; (i) removing the buffer from the fixed tissue
  • the solid medium comprises paraffin.
  • the first solvent comprises xylene.
  • the second solvent comprises ethanol.
  • the rehydration agent comprises water.
  • the buffer comprises phosphate buffered saline.
  • dissociating the tissue is performed using an automated dissociator or a manual pulverizer.
  • the strainer has a pore size of about 70 micrometers.
  • Another aspect of the present disclosure provides a non-transitory computer readable medium comprising machine executable code that, upon execution by one or more computer processors, implements any of the methods above or elsewhere herein.
  • Another aspect of the present disclosure provides a system comprising one or more computer processors and computer memory coupled thereto.
  • the computer memory comprises machine executable code that, upon execution by the one or more computer processors, implements any of the methods above or elsewhere herein.
  • FIG. 1 shows an example of a microfluidic channel structure for partitioning individual biological particles.
  • FIG. 2 shows an example of a microfluidic channel structure for the controlled partitioning of beads into discrete droplets.
  • FIG. 3 illustrates an example of a barcode carrying bead.
  • FIG. 4 illustrates another example of a barcode carrying bead.
  • FIG. 5 schematically illustrates an example microwell array.
  • FIG. 6 schematically illustrates an example workflow for processing nucleic acid molecules.
  • FIG. 7 schematically illustrates another example workflow for processing nucleic acid molecules.
  • FIG. 8 schematically illustrates another example workflow for processing nucleic acid molecules.
  • FIG. 9 schematically illustrates another example workflow for processing nucleic acid molecules.
  • FIG. 10 schematically illustrates an example workflow for analyzing cells, nuclei or cell beads.
  • FIG. 11 schematically illustrates example labelling agents with nucleic acid molecules attached thereto.
  • FIG. 12A schematically shows an example of labelling agents.
  • FIG. 12B schematically shows another example workflow for processing nucleic acid molecules.
  • FIG. 12C schematically shows another example workflow for processing nucleic acid molecules.
  • FIG. 13 schematically shows another example of a barcode-carrying bead.
  • FIG. 14 shows a computer system that is programmed or otherwise configured to implement methods provided herein.
  • FIG. 15 shows an example processed nucleic acid molecule described herein.
  • FIG. 16A shows an example workflow for processing multiple analytes in a partition.
  • FIG. 16B shows another example workflow for processing multiple analytes in a partition.
  • FIG. 17 schematically shows a feature-binding group described herein.
  • FIG. 18 shows example data from a workflow described herein.
  • FIG. 19 shows additional example data from a workflow described herein.
  • FIG. 20 shows additional example data from a workflow described herein.
  • FIG. 21A shows example data comparing fixed cells and unfixed cells.
  • FIG. 21B shows additional example data comparing fixed cells and unfixed cells.
  • FIG. 21C shows additional example data comparing fixed cells and unfixed cells.
  • FIG. 22 schematically shows an example workflow for assaying two different analyte types.
  • FIG. 23 shows example data of a barcoding approach described herein.
  • FIG. 24 shows example data of different analyte types using the barcoding approaches described herein.
  • FIG. 25 schematically shows an example method for processing nucleic acid molecules.
  • FIG. 26 shows another example method for processing nucleic acid molecules
  • FIG. 27 shows an example workflow for generating probe-linked nucleic acid molecules.
  • FIG. 28 shows another example workflow for generating probe-linked nucleic acid molecules.
  • FIG. 29 shows an example workflow for processing cells according to the methods described herein.
  • FIG. 30A shows example protein expression data resulting from barcoding of multiple analytes using different sample preparation parameters.
  • FIG. 30B shows additional protein expression data resulting from barcoding of multiple analytes using different sample preparation parameters.
  • FIG. 31 shows example gene expression data resulting from barcoding of multiple analytes using different sample preparation parameters.
  • FIGs. 32A-C shows example data of multiple analyte probing for a negative control group.
  • FIG. 32A shows example data showing different immune cell clusters.
  • FIG. 32B shows example data of gene expression of GZMB gene.
  • FIG. 32C shows example data of protein expression resulting from antibody staining.
  • FIGs. 33A-C shows example data of multiple analyte probing for an experimental group.
  • FIG. 33A shows example data showing different immune cell clusters.
  • FIG. 33B shows example data of gene expression of GZMB gene.
  • FIG. 33C shows example data of protein expression resulting from antibody staining.
  • FIGs. 34A-C shows example data of multiple analyte probing for an experimental group.
  • FIG. 34A shows example data showing different immune cell clusters.
  • FIG. 34B shows example data of gene expression of GZMB gene.
  • FIG. 34C shows example data of protein expression resulting from antibody staining.
  • FIGs. 35A-C shows example data of multiple analyte probing for an experimental group.
  • FIG. 35A shows example data showing different immune cell clusters.
  • FIG. 35B shows example data of gene expression of GZMB gene.
  • FIG. 35C shows example data of protein expression resulting from antibody staining.
  • FIGs. 36A-C shows example data of multiple analyte probing for an experimental group.
  • FIG. 35A shows example data showing different immune cell clusters.
  • FIG. 36B shows example data of gene expression of GZMB gene.
  • FIG. 36C shows example data of protein expression resulting from antibody staining.
  • FIGs. 37A-C shows example data of multiple analyte probing for an experimental group.
  • FIG. 37A shows example data showing different immune cell clusters.
  • FIG. 37B shows example data of gene expression of GZMB gene.
  • FIG. 37C shows example data of protein expression resulting from antibody staining.
  • FIG. 38 shows another example workflow for assaying two different analyte types.
  • FIGs. 39A-39E are an example of time series data for fixed samples.
  • FIG. 40 shows a flowchart of a method for analyzing a nucleic acid molecule of an embedded, fixed tissue, according to some embodiments.
  • FIG. 41 shows an example of a nucleic acid profiling workflow, according to some embodiments.
  • FIG. 42 shows an example of a nucleic acid profiling workflow, according to some embodiments.
  • FIG. 43 show an example of a plurality of probe molecules, according to some embodiments.
  • FIGs. 44 - 46 show examples of methods of preparation for fixed samples, according to some embodiments.
  • FIG. 47 provides a table of the pass percentages and other properties for the various procedures of the present example, according to some embodiments.
  • FIG. 48 shows examples of various tissues processed by the workflows of FIGs. 44 - 46, according to some embodiments.
  • FIGs. 49A-49D show example cell type clustering for single cell cancer fixed tissues.
  • FIGs. 50A and 50B show example tissue dissociation using pestle dissociation.
  • FIG. 51 shows an example gentleMACSTM program for tissue dissociation.
  • FIG. 52A shows an example of representative cells after dissociation.
  • FIG. 52B shows an example of representative cells after post-hybridization wash.
  • FIG. 53 shows example cell yield from fixed tissue sections.
  • barcode generally refers to a label, or identifier, that conveys or is capable of conveying information about an analyte.
  • a barcode can be part of an analyte.
  • a barcode can be independent of an analyte.
  • a barcode can be a tag attached to an analyte (e.g., nucleic acid molecule) or a combination of the tag in addition to an endogenous characteristic of the analyte (e.g., size of the analyte or end sequence(s)).
  • a barcode may be unique. Barcodes can have a variety of different formats.
  • barcodes can include: polynucleotide barcodes; random nucleic acid and/or amino acid sequences; and synthetic nucleic acid and/or amino acid sequences.
  • a barcode can be attached to an analyte in a reversible or irreversible manner.
  • a barcode can be added to, for example, a fragment of a deoxyribonucleic acid (DNA) or ribonucleic acid (RNA) sample before, during, and/or after sequencing of the sample. Barcodes can allow for identification and/or quantification of individual sequencing-reads.
  • real time can refer to a response time of less than about 1 second, a tenth of a second, a hundredth of a second, a millisecond, or less. The response time may be greater than 1 second. In some instances, real time can refer to simultaneous or substantially simultaneous processing, detection or identification.
  • subject generally refers to an animal, such as a mammal (e.g., human, mouse, rat) or avian (e.g., bird), or other organism, such as a plant.
  • the subject can be a vertebrate, such as a mammal, a rodent (e.g., a mouse), a primate, a simian or a human.
  • Animals may include, but are not limited to, farm animals, sport animals, and pets.
  • a subject can be a healthy or asymptomatic individual, an individual that has or is suspected of having a disease (e g., cancer) or a pre-disposition to the disease, and/or an individual that is in need of therapy or suspected of needing therapy.
  • a subject can be a patient.
  • a subject can be a microorganism or microbe (e.g., bacteria, fungi, archaea, viruses).
  • genomic information generally refers to genomic information from a subject, which may be, for example, at least a portion or an entirety of a subject’s hereditary information.
  • a genome can be encoded either in DNA or in RNA.
  • a genome can comprise coding regions (e.g., that code for proteins) as well as non-coding regions.
  • a genome can include the sequence of all chromosomes together in an organism.
  • the human genome ordinarily has a total of 46 chromosomes. The sequence of all of these together may constitute a human genome.
  • adaptor(s) can be used synonymously.
  • An adaptor or tag can be coupled to a polynucleotide sequence to be “tagged” by any approach, including ligation, hybridization, or other approaches.
  • sequence of nucleotide bases in one or more polynucleotides generally refers to methods and technologies for determining the sequence of nucleotide bases in one or more polynucleotides.
  • the polynucleotides can be, for example, nucleic acid molecules such as deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), including variants or derivatives thereof (e.g., single stranded DNA). Sequencing can be performed by various systems currently available, such as, without limitation, a sequencing system by Illumina®, Pacific Biosciences (PacBio®), Oxford Nanopore®, or Life Technologies (Ion Torrent®).
  • sequencing may be performed using nucleic acid amplification, polymerase chain reaction (PCR) (e.g., digital PCR, quantitative PCR, or real time PCR), or isothermal amplification.
  • PCR polymerase chain reaction
  • Such systems may provide a plurality of raw genetic data corresponding to the genetic information of a subject (e g., human), as generated by the systems from a sample provided by the subject.
  • sequencing reads also “reads” herein).
  • a read may include a string of nucleic acid bases corresponding to a sequence of a nucleic acid molecule that has been sequenced.
  • systems and methods provided herein may be used with proteomic information.
  • the term “bead,” as used herein, generally refers to a particle.
  • the bead may be a solid or semi-solid particle.
  • the bead may be a gel bead.
  • the gel bead may include a polymer matrix (e g , matrix formed by polymerization or cross-linking).
  • the polymer matrix may include one or more polymers (e.g., polymers having different functional groups or repeat units). Polymers in the polymer matrix may be randomly arranged, such as in random copolymers, and/or have ordered structures, such as in block copolymers. Cross-linking can be via covalent, ionic, or inductive, interactions, or physical entanglement.
  • the bead may be a macromolecule.
  • the bead may be formed of nucleic acid molecules bound together.
  • the bead may be formed via covalent or non-covalent assembly of molecules (e g., macromolecules), such as monomers or polymers.
  • Such polymers or monomers may be natural or synthetic.
  • Such polymers or monomers may be or include, for example, nucleic acid molecules (e.g., DNA or RNA).
  • the bead may be formed of a polymeric material.
  • the bead may be magnetic or non-magnetic.
  • the bead may be rigid.
  • the bead may be flexible and/or compressible.
  • the bead may be disruptable or dissolvable.
  • the bead may be a solid particle (e g., a metal-based particle including but not limited to iron oxide, gold or silver) covered with a coating comprising one or more polymers. Such coating may be disruptable or dissolvable.
  • barcoded nucleic acid molecule generally refers to a nucleic acid molecule that results from, for example, the processing of a nucleic acid barcode molecule with a nucleic acid sequence (e g., nucleic acid sequence complementary to a nucleic acid primer sequence encompassed by the nucleic acid barcode molecule).
  • the nucleic acid sequence may be a targeted sequence or a non-targeted sequence.
  • hybridization and reverse transcription of a nucleic acid molecule e g., a messenger RNA (mRNA) molecule
  • a nucleic acid barcode molecule e.g., a nucleic acid barcode molecule containing a barcode sequence and a nucleic acid primer sequence complementary to a nucleic acid sequence of the mRNA molecule
  • a barcoded nucleic acid molecule that has a sequence corresponding to the nucleic acid sequence of the mRNA and the barcode sequence (or a reverse complement thereof).
  • a barcoded nucleic acid molecule may serve as a template, such as a template polynucleotide, that can be further processed (e.g., amplified) and sequenced to obtain the target nucleic acid sequence.
  • a barcoded nucleic acid molecule may be further processed (e.g., amplified) and sequenced to obtain the nucleic acid sequence of the mRNA.
  • sample generally refers to a biological sample of a subject.
  • the biological sample may comprise any number of macromolecules, for example, cellular macromolecules.
  • the sample may be a cell sample.
  • the sample may be a cell line or cell culture sample.
  • the sample can include one or more cells or nuclei.
  • the sample can include one or more microbes.
  • the biological sample may be a nucleic acid sample or protein sample.
  • the biological sample may also be a carbohydrate sample or a lipid sample.
  • the biological sample may be derived from another sample.
  • the sample may be a tissue sample, such as a biopsy, core biopsy, needle aspirate, or fine needle aspirate.
  • the tissue sample may be a fresh tissue sample, a frozen tissue sample (e.g., flash frozen, lyophilized, cryo-sectioned, etc.), or a fixed tissue sample (e.g., a formalin-fixed and paraffin-embedded tissue sample).
  • the sample may be a fluid sample, such as a blood sample, urine sample, or saliva sample.
  • the sample may be a skin sample.
  • the sample may be a cheek swab.
  • the sample may be a plasma or serum sample.
  • the sample may be a cell-free or cell free sample.
  • a cell-free sample may include extracellular polynucleotides. Extracellular polynucleotides may be isolated from a bodily sample that may be selected from the group consisting of blood, plasma, serum, urine, saliva, mucosal excretions, sputum, stool and tears.
  • the term “biological particle,” as used herein, generally refers to a discrete biological system derived from a biological sample.
  • the biological particle may be a macromolecule.
  • the biological particle may be a small molecule.
  • the biological particle may be a virus.
  • the biological particle may be a cell or derivative of a cell.
  • the biological particle may be an organelle. Examples of an organelle from a cell include, without limitation, a nucleus, a ribosome, a Golgi apparatus, an endoplasmic reticulum, a chloroplast, an endocytic vesicle, an exocytic vesicle, a vacuole, and a lysosome.
  • the biological particle may be a rare cell from a population of cells.
  • the biological particle may be any type of cell, including without limitation prokaryotic cells, eukaryotic cells, bacterial, fungal, plant, mammalian, or other animal cell type, mycoplasmas, normal tissue cells, tumor cells, or any other cell type, whether derived from single cell or multicellular organisms.
  • the biological particle may be a constituent of a cell.
  • the biological particle may be or may include DNA, RNA, organelles, proteins, or any combination thereof.
  • the biological particle may be or may include a matrix (e.g., a gel or polymer matrix) comprising a cell or one or more constituents from a cell (e.g., cell bead), such as DNA, RNA, organelles, proteins, or any combination thereof, from the cell.
  • the biological particle may be obtained from a tissue of a subject (e.g., a human, a mouse, a rat, or other mammal).
  • the biological particle may be a hardened cell. Such hardened cell may or may not include a cell wall or cell membrane.
  • the biological particle may include one or more constituents of a cell, but may not include other constituents of the cell. An example of such constituents is a nucleus or an organelle.
  • a cell may be a live cell.
  • the live cell may be capable of being cultured, for example, being cultured when enclosed in a gel or polymer matrix, or cultured when comprising a gel or polymer matrix.
  • the term “macromolecular constituent,” as used herein, generally refers to a macromolecule contained within or from a biological particle.
  • the macromolecular constituent may comprise a nucleic acid.
  • the biological particle may be a macromolecule.
  • the macromolecular constituent may comprise DNA.
  • the macromolecular constituent may comprise RNA.
  • the RNA may be coding or non-coding.
  • the RNA may be messenger RNA (mRNA), ribosomal RNA (rRNA) or transfer RNA (tRNA), for example.
  • the RNA may be a transcript.
  • the RNA may be small RNA that are less than 200 nucleic acid bases in length, or large RNA that are greater than 200 nucleic acid bases in length.
  • Small RNAs may include 5.8S ribosomal RNA (rRNA), 5S rRNA, transfer RNA (tRNA), microRNA (miRNA), small interfering RNA (siRNA), small nucleolar RNA (snoRNAs), Piwi-interacting RNA (piRNA), tRNA-derived small RNA (tsRNA) and small rDNA-derived RNA (srRNA).
  • the RNA may be double-stranded RNA or single-stranded RNA.
  • the RNA may be circular RNA.
  • the macromolecular constituent may comprise a protein.
  • the macromolecular constituent may comprise a peptide.
  • the macromolecular constituent may comprise a polypeptide.
  • the term “molecular tag,” as used herein, generally refers to a molecule capable of binding to a macromolecular constituent.
  • the molecular tag may bind to the macromolecular constituent with high affinity.
  • the molecular tag may bind to the macromolecular constituent with high specificity.
  • the molecular tag may comprise a nucleotide sequence.
  • the molecular tag may comprise a nucleic acid sequence.
  • the nucleic acid sequence may be at least a portion or an entirety of the molecular tag.
  • the molecular tag may be a nucleic acid molecule or may be part of a nucleic acid molecule.
  • the molecular tag may be an oligonucleotide or a polypeptide.
  • the molecular tag may comprise a DNA aptamer.
  • the molecular tag may be or comprise a primer.
  • the molecular tag may be, or comprise, a protein.
  • the molecular tag may comprise a polypeptide.
  • the molecular tag may be a barcode.
  • partition refers to a space or volume that may be suitable to contain one or more species or conduct one or more reactions.
  • a partition may be a physical compartment, such as a droplet or well. The partition may isolate space or volume from another space or volume.
  • the droplet may be a first phase (e.g., aqueous phase) in a second phase (e.g., oil) immiscible with the first phase.
  • the droplet may be a first phase in a second phase that does not phase separate from the first phase, such as, for example, a capsule or liposome in an aqueous phase.
  • a partition may comprise one or more other (inner) partitions.
  • a partition may be a virtual compartment that can be defined and identified by an index (e.g., indexed libraries) across multiple and/or remote physical compartments.
  • a physical compartment may comprise a plurality of virtual compartments.
  • solid medium generally refers to a non-gaseous or nonliquid material.
  • the solid medium may comprise a long chain hydrophobic compound, glycerin, gels, hydrogels, epoxy, resin, or any combination thereof.
  • a long chain hydrophobic compound may include a wax, such as paraffin wax.
  • a method of the present disclosure may comprise barcoding one or more types of biomolecules (e.g., a nucleic acid molecule, a protein, a lipid, a carbohydrate, or a combination thereof).
  • the biomolecule may be, for instance, a nucleic acid molecule (e.g., a ribonucleic acid (RNA) molecule) or a protein.
  • RNA ribonucleic acid
  • Such a method may involve attaching one or more probes (e.g., nucleic acid probes) to the biomolecules and subsequently attaching a nucleic acid barcode molecule comprising a barcode sequence to the one or more probes.
  • the nucleic acid barcode molecule may attach to an overhanging sequence of a probe or to the end of a probe. Extension from an end of the probe to an end of the nucleic acid barcode molecule may form an extended nucleic acid molecule comprising both a sequence complementary to the barcode sequence and a sequence complementary to a target region of the nucleic acid molecule. The extended nucleic acid molecule may then be denatured from the nucleic acid barcode molecule and the nucleic acid molecule may be duplicated. One or more processes of the method may be carried out within a partition such as a droplet or well.
  • the present disclosure also provides a method of processing a sample (e.g., a cell sample or a tissue sample) that provides a barcoded nucleic acid molecule having linked probe molecules attached thereto.
  • the method may comprise providing a sample comprising a nucleic acid molecule (e.g., an RNA molecule) having a first and second target region; a first probe having a (i) first probe sequence that is complementary to the first target region and (ii) an additional probe sequence; and a second probe having a second probe sequence that is complementary to the second target region.
  • the first target region and the second target region are adjacent.
  • the first and second probe sequences may also comprise first and second reactive moieties, respectively.
  • the reactive moieties may be adjacent to one another. Subsequent reaction between the adjacent reactive moieties under sufficient conditions may link the first and second probes to yield a probe-linked nucleic acid molecule.
  • the probe-linked nucleic acid molecule may also be referred to as a probe-ligated nucleic acid molecule.
  • the first target region and the second target region are not adjacent, and a nucleic acid reaction (e g., a nucleic acid extension reaction, a gap-filling reaction) may be performed to yield a probe-linked nucleic acid molecule.
  • a nucleic acid reaction e g., a nucleic acid extension reaction, a gap-filling reaction
  • the probe-linked nucleic acid molecule may be barcoded with a barcode sequence of a nucleic acid barcode molecule to provide a barcoded probe-linked nucleic acid molecule. Barcoding may be achieved by hybridizing a binding sequence of the nucleic acid barcode molecule to the additional probe sequence of the first probe of the probe-linked nucleic acid molecule.
  • the barcoded probe linked-nucleic acid molecule may be subjected to amplification reactions to yield an amplified product comprising the first and second target regions and the barcode sequence or sequences complementary to these sequences. Accordingly, the method may provide amplified products without the use of reverse transcription.
  • One or more processes may be performed within a partition such as a droplet or well.
  • the present disclosure also provides a method of generating barcoded, probe-linked nucleic acid molecules.
  • the method may comprise providing a sample comprising a nucleic acid molecule (e.g., an RNA molecule) having a first target region and a second target region; a first probe having a first probe sequence that is complementary to the first target region and optionally an additional probe sequence; and a second probe having a second probe sequence that is complementary to the second target region.
  • the additional probe sequence of the first probe may comprise a probe capture sequence.
  • the second probe may comprise a probe capture sequence.
  • the first probe sequence of the first probe may hybridize to the first target region of the nucleic acid molecule, generating a probe-associated nucleic acid molecule, and a nucleic acid reaction (e.g., a nucleic acid extension reaction using a polymerase or reverse transcriptase) may be performed to generate an extended nucleic acid molecule comprising a sequence complementary to the second target region.
  • a nucleic acid reaction e.g., a nucleic acid extension reaction using a polymerase or reverse transcriptase
  • the second probe may hybridize to the nucleic acid molecule (or extended nucleic acid molecule, or complement thereof), and optionally, a nucleic acid extension reaction may be performed.
  • the extended nucleic acid molecule may be barcoded, such as by (a) hybridization of a barcode binding sequence of the nucleic acid barcode molecule to the first probe (e g., the additional probe sequence of the first probe) or the second probe (e.g., a probe capture sequence of the second probe), or (b) via a probe binding molecule (also referred to herein as a “splint molecule” or “splint oligonucleotide”), in which the probe binding molecule comprises (i) a probe binding sequence complementary to the additional probe sequence of the first probe (which may comprise the probe capture sequence) and/or a capture sequence of the second probe and a (ii) barcode binding sequence complementary to a sequence (e g., a common sequence) of the barcode molecule.
  • a probe binding molecule also referred to herein as a “splint molecule” or “splint oligonucleotide”
  • the probe binding molecule comprises (i) a probe binding
  • the barcoding may be performed prior to hybridization of the second probe to the second target region.
  • the barcoded nucleic acid molecule may be subjected to conditions sufficient for hybridization of the second probe sequence of the second probe to the second target region of the nucleic acid molecule (or barcoded nucleic acid molecule).
  • a nucleic acid reaction e.g., nucleic acid extension
  • Another aspect of the present disclosure provides a method of barcoding multiple analytes, such as the probe-linked nucleic acid molecules described herein, as well as other types of biomolecules (e.g., proteins).
  • the method may comprise providing (i) a sample comprising a nucleic acid molecule (e.g., an RNA molecule) having first and second target regions and (ii) a feature-binding moiety comprising a reporter oligonucleotide comprising a capture sequence; (iii) a first probe having a first probe sequence that is complementary to the first target region and an additional probe sequence; (iv) a second probe having a second probe sequence that is complementary to the second target region; and (v) a third probe having a third probe sequence that is complementary to a sequence of the reporter oligonucleotide.
  • a nucleic acid molecule e.g., an RNA molecule
  • a feature-binding moiety comprising a reporter oligonucleotide
  • the first probe and the second probe may be subjected to conditions sufficient to hybridize to the first target region and the second target region, respectively, and to generate a probe-linked nucleic acid molecule.
  • the third probe sequence of the third probe may be subjected to conditions sufficient to hybridize to the capture sequence of the reporter oligonucleotide, generating a probe-binding moiety complex.
  • the probe-linked nucleic acid molecule and the probe-binding moiety complex may be subjected to conditions sufficient for barcoding, thereby generating a barcoded probe-linked nucleic acid molecule and a barcoded probe-binding moiety complex.
  • the barcoded probe- linked molecule may be subjected to amplification reactions to yield an amplified product comprising the first and second target regions and the barcode sequence or sequences complementary to these sequences.
  • the barcoded probe-binding moiety complex may similarly be subjected to amplification reactions to yield an amplified product comprising the fourth probe sequence and the barcode sequence.
  • One or more processes may be performed within a cell bead and/or a partition, such as a droplet or well.
  • the methods described herein may be useful in indexing cells, nuclei, or cell beads to partitions; such indexing may be useful in partitions occupied by more than one cell and identifying the cell, nucleus, cell bead or partition from which an analyte was derived.
  • a sample may be a fixed sample.
  • a sample may comprise a plurality of fixed samples, such as a plurality of fixed cells or fixed nuclei.
  • a sample may comprise a fixed tissue. Fixation of cell or cellular constituent, or a tissue comprising a plurality of cells or nuclei, may comprise application of a chemical species or chemical stimulus.
  • the term “fixed” as used herein with regard to biological samples generally refers to the state of being preserved from decay and/or degradation.
  • fixation generally refers to a process that results in a fixed sample, and in some instances can include contacting the biomolecules within a biological sample with a fixative (or fixation reagent) for some amount of time, whereby the fixative results in covalent bonding interactions such as crosslinks between biomolecules in the sample.
  • a “fixed biological sample” may generally refer to a biological sample that has been contacted with a fixation reagent or fixative. For example, a formaldehyde- fixed biological sample has been contacted with the fixation reagent formaldehyde.
  • “Fixed cells”, “fixed nuclei” or “fixed tissues” refer to cells/nuclei or tissues that have been in contact with a fixative under conditions sufficient to allow or result in the formation of intra- and inter- molecular covalent crosslinks between biomolecules in the biological sample.
  • a fixation reagent e.g., paraformaldehyde or PF A
  • the fixation reagent, formaldehyde may result in covalent aminal crosslinks within RNA, DNA, and/or protein molecules.
  • the widely used fixative reagent paraformaldehyde or PF A, fixes tissue samples by catalyzing crosslink formation between basic amino acids in proteins, such as lysine and glutamine. Both intra-molecular and inter-molecular crosslinks can form in the protein. These crosslinks can preserve protein secondary structure and also eliminate enzymatic activity in the preserved tissue sample.
  • fixation reagents include but are not limited to aldehyde fixatives (e g., formaldehyde, also commonly referred to as “paraformaldehyde,” “PF A,” and “formalin”; glutaraldehyde; etc.), imidoesters, NHS (N-Hydroxysuccinimide) esters, and the like.
  • aldehyde fixatives e g., formaldehyde, also commonly referred to as “paraformaldehyde,” “PF A,” and “formalin”; glutaraldehyde; etc.
  • imidoesters e g., imidoesters, NHS (N-Hydroxysuccinimide) esters, and the like.
  • the fixative or fixation reagent useful for fixing samples is formaldehyde.
  • formaldehyde when used in the context of a fixative may also refer to “paraformaldehyde” (or “PF A”) and “formalin”, both of which are terms with specific meanings related to the formaldehyde composition (e.g., formalin is a mixture of formaldehyde and methanol).
  • PF A paraformaldehyde
  • formalin is a mixture of formaldehyde and methanol
  • a formaldehyde-fixed biological sample may also be referred to as formalin- fixed or PFA-fixed. Protocols and methods for the use of formaldehyde as a fixation reagent to prepare fixed biological samples are well known in the art and can be used in the methods and compositions of the present disclosure.
  • suitable ranges of formaldehyde concentrations for use in preparing a fixed biological sample is 0.1 to 10%, 1-8%, 1-4%, 1-2%, 3-5%, or 3.5-4.5%.
  • the biological sample is fixed using a final concentration of 1% formaldehyde, 4% formaldehyde, or 10% formaldehyde.
  • the formaldehyde can be diluted from a more concentrated stock solution - e.g., a 35%, 25%, 15%, 10%, 5% PFA stock solution.
  • fixatives include, for example, organic solvents such as alcohols (e.g., methanol or ethanol), ketones (e.g., acetone), and aldehydes (e.g., paraformaldehyde, formaldehyde (e.g., formalin), or glutaraldehyde).
  • organic solvents such as alcohols (e.g., methanol or ethanol), ketones (e.g., acetone), and aldehydes (e.g., paraformaldehyde, formaldehyde (e.g., formalin), or glutaraldehyde).
  • cross-linking agents may also be used for fixation including, without limitation, disuccinimidyl suberate (DSS), dimethylsuberimidate (DMS), formalin, and dimethyladipimidate (DMA), dithio-bis(- succinimidyl propionate) (DSP), disuccinimidyl tartrate (DST), and ethylene glycol bis(succinimidyl succinate) (EGS).
  • a cross-linking agent may be a cleavable cross-linking agent (e.g., thermally cleavable, photocleavable, etc.).
  • fixation reagent can be used in combination when preparing a fixed biological sample.
  • a first fixation agent such as an organic solvent
  • a second fixation agent such as a cross-linking agent.
  • the organic solvent may be an alcohol (e.g., ethanol or methanol), ketone (e.g., acetone), or aldehyde (e.g., paraformaldehyde, formaldehyde, or glutaraldehyde).
  • the cross-linking agent may be selected from the group consisting of disuccinimidyl suberate (DSS), dimethylsuberimidate (DMS), formalin, and dimethyladipimidate (DMA), dithio-bis(- succinimidyl propionate) (DSP), disuccinimidyl tartrate (DST), and ethylene glycol bis(succinimidyl succinate) (EGS).
  • DSS disuccinimidyl suberate
  • DMS dimethylsuberimidate
  • DMA dimethyladipimidate
  • DSP dithio-bis(- succinimidyl propionate)
  • DST disuccinimidyl tartrate
  • ELS ethylene glycol bis(succinimidyl succinate)
  • a first fixation agent may be provided to or brought into contact with the cell or nucleus to bring about a change in a first characteristic or set of characteristics of the cell/nucleus
  • a fixation agent may be provided to or brought into contact with the cell or nucleus to bring about a change in a second characteristic or set of characteristics of the cell or nucleus.
  • a first fixation agent may be provided to or brought into contact with a cell or nucleus to bring about a change in a dimension of the cell (e.g., a reduction in cross-sectional diameter, see, e.g., U.S. Pat. Pub. No.
  • a second fixation agent may be provided to or brought into contact with a cell or nucleus to bring about a change in a second characteristic or set of characteristics of the cell (e.g., forming crosslinks within and/or surrounding the cell or nucleus).
  • the first and second fixation agents may be provided to or brought into contact with the cell or nucleus at the same or different times.
  • Other suitable fixing agents include those disclosed in, e.g., International PCT App. No. PCT/US2020/066705, which is incorporated herein by reference in its entirety.
  • a first fixation agent that is an organic solvent may be provided to a cell to change a first characteristic (e.g., cell size) and a second fixation agent that is a crosslinking agent may be provided to a cell to change a second characteristic (e.g., cell fluidity or rigidity).
  • the first fixation agent may be provided to the cell before the second fixation agent.
  • biomolecules e.g., biological samples such as tissue specimens
  • a fixation reagent containing both formaldehyde and glutaraldehyde
  • the contacted biomolecules can include fixation crosslinks resulting both from formaldehyde induced fixation and glutaraldehyde induced fixation.
  • a suitable concentration of glutaraldehyde can be used for use as a fixation reagent can be 0.1 to 1%.
  • Fixation and wash reagents may also include commercially available products, e.g., BioLegend® Fixation Buffer (420801) and Permeabilization Wash Buffer (421002).
  • Changes to a characteristic or a set of characteristics of a cell or cellular constituents may be at least partially reversible (e.g., via rehydration or de-crosslinking).
  • changes to a characteristic or set of characteristics of a cell or cellular constituents may be substantially irreversible.
  • a sample e.g., a cell sample
  • a fixation process at any useful point in time.
  • cells, nuclei and/or cellular/nuclear constituents of a sample may be subjected to a fixation process involving one or more fixation agents (e.g., as described herein) prior to commencement of any subsequent processing, such as for storage.
  • Cells, nuclei and/or cellular/nuclear constituents such as cells, nuclei and/or cellular/nuclear constituents of a tissue sample, subjected to a fixation process prior to storage, may be stored in an aqueous solution, optionally in combination with one or more preserving agents configured to preserve morphology, size, or other features of the cells and/or cellular components.
  • Fixed cells, nuclei and/or cellular/nuclear constituents may be stored below room temperature, such as in a freezer.
  • cells, nuclei and/or cellular/nuclear constituents of a sample may be subjected to a fixation process involving one or more fixation agents subsequent to one or more other processes, such as filtration, centrifugation, agitation, selective precipitation, purification, permeabilization, isolation, heating, etc.
  • fixation process involving one or more fixation agents subsequent to one or more other processes, such as filtration, centrifugation, agitation, selective precipitation, purification, permeabilization, isolation, heating, etc.
  • fixation process e.g., as described herein.
  • a sample comprising a plurality of cells including a plurality of cells of a given type may be subjected to a positive separation process to provide a sample enriched in the plurality of cells of the given type.
  • the enriched sample may then be subjected to a fixation process involving one or more fixation agents (e.g., as described herein) to provide an enriched sample comprising a plurality of fixed cells.
  • a fixation process may be performed in a bulk solution.
  • fixed samples e.g., fixed cells, fixed nuclei, and/or cellular/nuclear constituents
  • partitions e.g., droplets or wells
  • fixed samples may undergo additional processing, such as partial or complete reversal of a fixation process by, for example, rehydration or de-crosslinking, prior to partitioning and any subsequent processing.
  • fixed samples may undergo partial or complete reversal of a fixation process within a plurality of partitions (e.g., prior to or concurrent with additional processing described elsewhere herein).
  • a tissue specimen comprising a plurality of cells, nuclei and/or cellular/nuclear constituents may be processed to provide formalin-fixed paraffin-embedded (FFPE) tissue.
  • FFPE formalin-fixed paraffin-embedded
  • a tissue specimen may be contacted (e.g., saturated) with formalin and then embedded in a solid medium.
  • the solid medium may be paraffin wax.
  • FFPE processing may facilitate preservation of a tissue sample (e.g., prior to subsequent processing and analysis).
  • a tissue sample, including an FFPE tissue sample may additionally or alternatively be subjected to storage in a low-temperature freezer.
  • Cells, nuclei and/or cellular/nuclear constituents may be dissociated from a tissue sample (e.g., FFPE tissue sample) prior to undergoing subsequent processing.
  • tissue sample e.g., FFPE tissue sample
  • individual cells, nuclei and/or cellular/nuclear constituents of a tissue sample such as an FFPE tissue sample may be optically detected, labeled, or otherwise processed prior to any such dissociation.
  • detection, labeling, or other processing may be performed according to a 2- or 3 -dimensional array and optionally according to a pre-determined pattern.
  • a tissue specimen may be embedded in other materials such as, for example, optimal cutting temperature (OCT) compound, crosslinking-based supports (e.g., polymers), or the like.
  • OCT optimal cutting temperature
  • crosslinking-based supports e.g., polymers
  • the tissue specimen may have been fixed at least about 1 day (d), 2 d, 3 d, 4 d, 5 d, 6 d, 1 week (wk), 2 wk, 3 wk, 1 month (m), 2 m, 3 m, 4 m, 5 m, 6 m, 7 m, 8 m, 9 m, 10 m, 11 m, 1 year (y), 2 y, 3 y, 4 y, 5 y, 6 y, 7 y, 8 y, 9 y, 10 y, 15 y, 20 y, 25 y, 30 y, 35 y, 40 y, 45 y, 50 y, or longer before use in the methods and systems described elsewhere herein.
  • preparation of a fixed sample may comprise securing and sectioning at least a portion of a fixed sample (e.g., via microtomy, ultramicrotomy, etc.).
  • the fixed sample may be sectioned into one or more (e.g., a plurality) of scrolls.
  • the fixed tissue e.g., a scroll
  • the fixed tissue may be at least about 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, or more micrometers in thickness.
  • the sectioning and/or securing may be performed at ambient temperature.
  • the sectioning and/or securing may be performed at temperatures above or below ambient temperature.
  • the fixed tissue may have a mass of at least about 100 micrograms (pg), 150 pg, 200 pg 250 pg, 300 pg, 350 pg, 400 pg, 450 pg, 500 pg, 550 pg, 600 pg, 650 pg, 700 pg, 750 pg, 800 pg, 850 pg, 900 pg, 950 pg, 1 milligram (mg), 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 150 mg, 200 mg, 250 mg, 300 mg, 350 mg, 400 mg, 450 mg, 500 mg, 550 mg, 600 mg, 650 mg, 700 mg, 750 mg, 800 mg, 95 mg, 100 mg, 150 mg
  • the fixed tissue may have a mass of at most about 50 g, 45 g, 40 g, 35 g, 30 g, 25 g, 20 g, 15 g, 10 g, 5 g, 4.75 g, 4.5 g, 4.25 g, 4 g, 3.75 g, 3.5 g, 3.25 g, 3 g, 2.75 g, 2.5 g, 2.25 g, 2 g, 1.75 g, 1.5 g, 1.25 g, 1 g, 950 mg, 900 mg, 850 mg, 800 mg, 750 mg, 700 mg, 650 mg, 600 mg, 550 mg, 500 mg, 450 mg, 400 mg, 350 mg, 300 mg, 250 mg, 200 mg, 150 mg, 100 mg, 95 mg, 90 mg, 85 mg, 80 mg, 75 mg, 70 mg, 65 mg, 60 mg, 55 mg, 50 mg, 45 mg, 40 mg, 35 mg, 30 mg, 25 mg, 20 mg, 15 mg, 10 mg, 9 mg, 8
  • the scroll can be mechanically and/or enzymatically dissociated.
  • the mechanical dissociation may comprise sonication (e.g., sonication at below ambient temperatures).
  • the sonication may comprise a sonication at a power of at least about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 65, 80, 85, 90, 95, or 100 percent power.
  • the sonication may comprise sonication at a power of at most about 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45, 40, 35, 30, 25, 20, 15, 10, 5, or less percent power.
  • the sonication may be for at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 30, 45, 60, or more minutes.
  • the sonication may be for at most about 60, 45, 30, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, or fewer minutes.
  • the mechanical dissociation may comprise use of a shake plate.
  • the sample can be placed in a sample tube, and the sample tube can be shaken on a shake plate.
  • the mechanical dissociation may comprise stirring the sample.
  • the fixed sample may be processed to remove one or more fixatives and/or supports (e.g., solid medium).
  • the fixed sample can be deparaffinized.
  • the fixed sample may not be processed to remove the one or more fixatives and/or supports (e.g., solid medium).
  • a fixed sample can be used as sectioned (e.g., a scroll). Examples of processes include use of one or more non-polar solvents (e g., linear alkanes, cyclic alkanes, benzene, xylenes, neo-clear, orange oil, other substituted or non-substituted alkanes, or the like, or any combination thereof).
  • non-polar solvents e g., linear alkanes, cyclic alkanes, benzene, xylenes, neo-clear, orange oil, other substituted or non-substituted alkanes, or the like, or any combination thereof.
  • the non-polar solvent is xylene.
  • the removal of the one or more fixatives and/or supports may be repeated (e.g., for more complete removal of the one or more fixatives and/or supports).
  • the sample may be rehydrated (e.g., via water addition, ethanol rehydration, gaseous rehydration, or the like, or any combination thereof).
  • ethanolic solutions of water with increasing water concentration can be used to rehydrate the sample.
  • the sample can be analyzed without rehydration.
  • the sample may be washed using a polar (e.g., aqueous) solution to remove additional impurities.
  • an aqueous solution of phosphate buffered saline can be used to remove impurities from a sample.
  • One or more dissociation solutions comprising, for example, liberase with low thermolysis (TL), liberase with medium thermolysis (TM), liberase with high thermolysis (TH), collagenases, or the like, or any combination thereof may be added to process a sample.
  • the dissociation sample may be added at ambient (e g., room) temperature.
  • the dissociation solution be heated prior to the addition.
  • the dissociation solution may be cooled prior to the addition.
  • the dissociation sample may be at a temperature of at least about 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 34, 36, 37, 38, 39, 40, or more degrees Celsius when added.
  • the dissociation solution may be at a temperature of at most about 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, or fewer degrees Celsius when added.
  • the dissociation solution may be added at a temperature in a range as defined by any two of the proceeding values.
  • the sample may be titrated at least about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, or more times to form a cellular suspension.
  • impurities e.g., paraffin
  • impurities can be removed by allowing the suspension to rest, and the impurities can precipitate from the solution, and the purified solution can be removed and further processed.
  • a liberase can comprise one or more enzymes configured to degrade at least a portion of a biological molecule.
  • the liberase can comprise one or more collagenases.
  • the collagenases can comprise one or more isoforms of collagenase, collagenase I, collagenase II, or the like, or any combination thereof.
  • the collagenase isoforms may be present in ratios between the various isoforms of at least about 1 :20, 1 :19, 1:18, 1 : 17, 1 :16, 1:15, 1: 14, 1 :13, 1:12, 1: 11, 1:10, 1:9, 1:8, 1 :7, 1:6, 1 :5, 1:4, 1 :3, 1:2, 1 :1, 2: 1, 3:1, 4: 1, 5:1, 6:1, 7: 1, 8:1, 9: 1, 10: 1, 11:1, 12:1, 13:1, 14: 1, 15:1, 16:1, 17: 1, 18:1, 19:1, 20: 1, or more.
  • a composition comprising the one or more enzymes can comprise a dispase.
  • the dispase may be a protease (e g., a neutral protease, etc.).
  • the dispase may be a non-clostridial dispase.
  • a composition comprising the one or more enzymes may comprise thermoysin.
  • the thermoysin can be a protease (e g., a neutral protease, etc.).
  • the thermolysin can be a non-clostridial thermoysin.
  • a composition comprising the one or more enzymes may comprise a plurality of additional components as described elsewhere herein (e.g., a dispase, a termoysin, etc.).
  • the sample may be filtered one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) times.
  • the filtration may comprise use of one or more different sizes (e.g., pore sizes) of filter.
  • the filter may comprise a pore size of at most about 500, 400, 300, 200, 100, 90, 80, 70, 60, 50, 40, 30, 20, 10, 5, 1, or less micrometers.
  • a first filtration with a 70 micrometer pore size filter can be performed.
  • a second filtration with a 30 micrometer filtration can be performed, which can reduce debris (e g., undissolved paraffin or other support) without reducing cellular recovery from the sample.
  • the filtrate and the cellular suspension may be combined and subsequently centrifuged.
  • the centrifugation may occur at a value of at least about 100, 200, 300, 400, 500, 600, 700, 850, 900, 950, 1,000, 1,100, 1,200, 1,300, 1,400, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,100, 2,200, 2,300, 2,400, 2,500, 3,000, or more reciprocal centrifugal force (ref).
  • the centrifugation may occur at a value of at most about 3,000, 2,500, 2,400, 2,300, 2,200, 2,100, 2,000, 1,900, 1,800, 1,700, 1,600, 1,500, 1,400, 1,300, 1,200, 1,100, 1,000, 950, 900, 850, 800, 700, 600 ,500, 400, 300, 200, 100, or fewer ref.
  • the centrifugation may be performed at a value within a range as defined by any two of the proceeding values. For example, the centrifugation may be performed at a value of about 850 to about 2,000 ref.
  • the solution can be removed from the centrifuged pellet (e.g., without disturbing the pellet).
  • the pellet can be resuspended into solution.
  • the pellet can be resuspended into a buffer solution.
  • the resuspended solution can then be analyzed (e g., to determine cellular concentration).
  • cellular concentration determination systems include, but are not limited to the Countess II FL Automated Cell Counter, Cellaca MX High-Throughput Automated Cell Counter, or the like using a fluorescent dye (e.g., ethidium homodimer-1, ect.) or AO/PI staining solution, or the like.
  • the resuspended solution may then be used as a sample for the methods and systems described elsewhere herein (e.g., RNA profiling, etc.).
  • a fixed sample e.g., an FFPE sample
  • the fixation process may capture ephemeral states of the cells of the sample and/or ephemeral types of cells in the sample that may not be captured in fresh samples.
  • cellular processes can be investigated in different ways by use of fixed samples.
  • use of the methods and systems described elsewhere herein on fixed samples may provide unexpected improvements to the analysis of the fixed samples, such as improved sensitivity versus other analysis methods as well as the aforementioned analysis of different ephemeral states/types within the sample.
  • the use of fixed samples may permit time series analysis of samples (e.g., samples can be fixed at different times and later analyzed). Such a time series analysis may provide information related to the evolution of states and/or cell types within the samples.
  • the present disclosure provides a method for barcoding nucleic acid molecules.
  • the method may generally comprise contacting a nucleic acid molecule with a pair of probes and a barcode molecule to generate a barcoded molecule (e g., a barcoded probe-linked molecule).
  • the nucleic acid molecule may comprise a sequence corresponding to a target sequence or a template sequence.
  • One or more nucleic acid reactions e.g., a ligation, a nucleic acid extension reaction, amplification, etc. may be performed to generate the barcoded molecule.
  • the method comprises: contacting a nucleic acid molecule with a first probe to generate a probe-associated nucleic acid molecule, wherein the nucleic acid molecule comprises a first target region and a second target region, wherein the first probe comprises a first probe sequence complementary to the first target region; performing a nucleic acid reaction (e g., a nucleic acid extension reaction, e.g., by using a polymerase or reverse transcriptase, etc.) to generate an extended probe molecule comprising a sequence complementary to the second target region; providing (i) a second probe comprising a second probe sequence corresponding to or complementary to the second target region and (ii) a nucleic acid barcode molecule; and subjecting the extended probe molecule or derivative thereof to conditions sufficient to generate a barcoded molecule.
  • a nucleic acid reaction e g., a nucleic acid extension reaction, e.g., by using a polymerase or reverse transcriptase, etc.
  • the first target region and the second target region may be disposed adjacent to one another or may be separate from one another (e.g., disposed on opposite ends of a gap region).
  • barcoding may be facilitated by providing a probe binding molecule (also referred to herein as a “splint molecule” or in some instances, a “splint oligonucleotide”).
  • the first probe and/or the second probe may comprise a probe capture sequence
  • the probe-binding molecule may comprise a probe-binding sequence complementary to the probe capture sequence.
  • the nucleic acid barcode molecule may comprise a barcode sequence and a barcode capture sequence
  • the probe-binding molecule may comprise a barcode binding sequence complementary to the barcode capture sequence.
  • the probe-binding molecule may be pre-annealed to the nucleic acid barcode molecule.
  • Barcoding may comprise hybridization of the probe binding molecule to the probe capture sequence (or complement thereof) of the first probe and/or second probe and to the barcode capture sequence of the nucleic acid barcode molecule.
  • the barcoded molecule may comprise a sequence corresponding to the first target region, a sequence corresponding to the second target region, a sequence corresponding to the probe capture sequence, and a sequence corresponding to the barcode sequence.
  • One or more operations may be performed within a partition (e.g., droplet or well).
  • the methods described herein may facilitate gene expression profiling with singlecell, single-nucleus or single-cell bead resolution using, for example, nucleic acid extension reactions, probe hybridization, chemical or enzymatic ligation, barcoding, amplification, and sequencing.
  • the methods described herein may allow for gene expression analysis while avoiding the use of specialized imaging equipment and, in certain instances, reverse transcription, which may be highly error prone and inefficient.
  • the methods may be used to analyze a pre-determined panel of target genes in a population of single cells, nuclei, or cell beads in a sensitive and accurate manner.
  • the methods described herein may also be useful in detecting or characterizing genetic variants, for example, in instances where the sequence of a region disposed between the target regions (e.g., a gap region) is not known.
  • the methods described herein may be useful in analyzing a single nucleotide polymorphism (SNP), an alternative-spliced junction, an insertion, a mutation, a deletion, a gene rearrangement (e.g., V(D)J rearrangements), a transposon, or other genetic element or variants.
  • SNP single nucleotide polymorphism
  • V(D)J rearrangements e.g., V(D)J rearrangements
  • the nucleic acid molecule analyzed by the methods described herein may comprise a fusion gene (e.g., a hybrid gene generated via translocation, interstitial deletion, or chromosomal inversion).
  • the methods described herein may be useful in analyzing genomic, transcriptomic, exomic and/or proteomic elements in cells, nuclei, cell beads, tissue samples, spatial arrays of cells, nuclei or tissues, etc.
  • the nucleic acid molecule analyzed by the methods described herein may be a singlestranded or a double-stranded nucleic acid molecule.
  • a double-stranded nucleic acid molecule may be completely or partially denatured to provide access to a target region (e.g., a target sequence) of a strand of the nucleic acid molecule.
  • Denaturation may be achieved by, for example, adjusting the temperature or pH of a solution comprising the nucleic acid molecule; using a chemical agent such as formamide, guanidine, sodium salicylate, dimethyl sulfoxide, propylene glycol, urea, or an alkaline agent (e.g., NaOH); or using mechanical agitation (e.g., centrifuging or vortexing a solution including the nucleic acid molecule).
  • a chemical agent such as formamide, guanidine, sodium salicylate, dimethyl sulfoxide, propylene glycol, urea, or an alkaline agent (e.g., NaOH); or using mechanical agitation (e.g., centrifuging or vortexing a solution including the nucleic acid molecule).
  • the nucleic acid molecule may be a target nucleic acid molecule.
  • the target nucleic acid molecule may be an RNA molecule.
  • the RNA molecule may be, for example, a transfer RNA (tRNA) molecule, ribosomal RNA (rRNA) molecule, mitochondrial RNA (mtRNA) molecule, messenger RNA (mRNA) molecule, non-coding RNA molecule, synthetic RNA molecule, or another type of RNA molecule.
  • the RNA molecule may be an mRNA molecule.
  • the nucleic acid molecule may be a viral or pathogenic RNA.
  • the nucleic acid molecule may be a synthetic nucleic acid molecule previously introduced into or onto a cell.
  • the nucleic acid molecule may comprise a plurality of barcode sequences, and two or more barcode sequences may be target regions of the nucleic acid molecule.
  • the nucleic acid molecule is a guide RNA (gRNA), which may be exogenously introduced in a cell or cell bead.
  • the nucleic acid molecule is an RNA molecule derived from an exogenously introduced nucleic acid molecule, e.g., an RNA derived from a plasmid, an integrated DNA sequence (e g. using viral transduction in a cell), a gRNA from a CRISPR genetic element, etc.
  • the nucleic acid molecule may comprise one or more features selected from the group consisting of a 5’ cap structure, an untranslated region (UTR), a 5’ triphosphate moiety, a 5’ hydroxyl moiety, a Kozak sequence, a Shine-Dalgamo sequence, a coding sequence, a codon, an intron, an exon, an open reading frame, a regulatory sequence, an enhancer sequence, a silencer sequence, a promoter sequence, and a poly(A) sequence (e.g., a poly(A) tail).
  • UTR untranslated region
  • 5’ triphosphate moiety e.g., a 5’ triphosphate moiety
  • 5’ hydroxyl moiety e.g., a 5’ hydroxyl moiety
  • Kozak sequence e.g., a Shine-Dalgamo sequence
  • a coding sequence e.g., a codon, an intron, an exon, an open reading frame
  • the nucleic acid molecule may comprise one or more features selected from the group consisting of a 5’ cap structure, an untranslated region (UTR), a Kozak sequence, a Shine-Dalgarno sequence, a coding sequence, and a poly(A) sequence (e.g., a poly (A) tail).
  • a 5’ cap structure an untranslated region (UTR)
  • UTR untranslated region
  • Kozak sequence e.g., a Shine-Dalgarno sequence
  • a coding sequence e.g., a poly (A) tail
  • a 5’ cap structure may comprise one or more nucleoside moi eties joined by a linker such as a triphosphate (ppp) linker.
  • a 5’ cap structure may comprise naturally occurring nucleoside and/or non- naturally occurring (e.g., modified) nucleosides.
  • a 5’ cap structure may comprise a guanine moiety or a modified (e.g., alkylated, reduced, or oxidized) guanine moiety such as a 7- methylguanylate (m 7 G) cap.
  • Examples of 5’ cap structures include, but are not limited to, m 7 GpppG, m 7 Gpppm 7 G, m 7 GpppA, m 7 GpppC, GpppG, m 2,7 GpppG, m 2,2 ’ 7 GpppG, and antireverse cap analogs such as m 7,2 Ome GpppG, m 7,2 d GpppG, m 7,3 Ome GpppG, and m 7,3 d GpppG.
  • An untranslated region may be a 5’ UTR or a 3’ UTR.
  • a UTR may include any number of nucleotides.
  • a UTR may comprise at least 3, 5, 7, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, or more nucleotides. In some cases, a UTR may comprise fewer than 20 nucleotides. In other cases, a UTR may comprise at least 100 nucleotides, such as more than 200, 300, 400, 500, 600, 700, 800, 900, or 1000 nucleotides. Similarly, a coding sequence may include any number of nucleotides, such as at least 3, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, or more nucleotides. A UTR, coding sequence, or other sequence of a nucleic acid molecule may have any nucleotide or base content or arrangement.
  • a sequence of a nucleic acid molecule may comprise any number or concentration of guanine, cytosine, uracil, and adenine bases.
  • a nucleic acid molecule may also include non-naturally occurring (e.g., modified) nucleosides.
  • a modified nucleoside may comprise one or more modifications (e.g., alkylations, hydroxylation, oxidation, or other modification) in its nucleobase and/or sugar moieties.
  • the nucleic acid molecule may comprise one or more target regions.
  • a target region may correspond to a gene or a portion thereof. Each region may have the same or different sequences.
  • the nucleic acid molecule may comprise two target regions having the same sequence located at different positions along a strand of the nucleic acid molecule.
  • the nucleic acid molecule may comprise two or more target regions having different sequences. Different target regions may be interrogated by different probes. Target regions may be located adjacent to one another or may be spatially separated along a strand of the nucleic acid molecule. The target regions may be located on the same strand or different strands.
  • adjacent may mean that the entities directly next to one other (e.g., contiguous) or in proximity to one another.
  • a first target region may be directly next to a second target region (e.g., having no other entity disposed between the first and second target regions) or in proximity to a second target region (e.g., having an intervening sequence or molecule between the first and second target regions).
  • a double-stranded nucleic acid molecule may comprise a target region in each strand that may be the same or different.
  • the methods described herein may be performed for one or more target regions at a time.
  • a single target region of the multiple target regions may be analyzed (e.g., as described herein) or two or more target regions may be analyzed at the same time. Analyzing two or more target regions may involve providing two or more probes, where a first probe has a sequence that is complementary to the first target region, a second probe has a sequence that is complementary to the second target region, etc.
  • Each probe may further comprise one or more additional sequences (e.g., additional probe sequences, unique molecular identifiers (UMIs), a barcode sequence, a primer sequence, a capture sequence, or other functional sequence).
  • additional probe sequences e.g., additional probe sequences, unique molecular identifiers (UMIs), a barcode sequence, a primer sequence, a capture sequence, or other functional sequence.
  • UMIs unique molecular identifiers
  • the first probe and/or the second probe may comprise the same or different barcode sequences.
  • the first probe and the second probe may be configured to hybridize to one or more nucleic acid barcode molecules.
  • the first probe and/or the second probe may comprise a probe capture sequence, which may be configured to hybridize to a nucleic acid barcode molecule or to a probe binding molecule (e.g., a splint oligonucleotide) that is configured to hybridize to a nucleic acid barcode molecule (e g., via a barcode binding sequence that is complementary to a capture sequence of the nucleic acid barcode molecule).
  • the probe capture sequence may be any useful length; for example, the probe capture sequence may be about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 20, about 30, about 40, about 50, about 60, about 70, about 80, about 90, about 100 or more nucleotides in length.
  • the probe capture sequence may be at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90, at least 100 or more nucleotides in length.
  • the probe capture sequence may be at most 100, at most 90, at most 80, at most 70, at most 60, at most 50, at most 40, at most 30, at most 20, at most 10, at most 9, at most 8, at most 7, at most 6, at most 5, at most 4, at most 3, at most 2, or at most 1 nucleotide in length.
  • a range of lengths of the probe capture sequence such as from about 8 to about 50 nucleotides in length, etc.
  • the probe capture sequence length may be varied based on any useful application and properties, e.g., melting temperature, annealing temperature, annealing strength (e.g., GC content), hybridization stringency, etc.
  • the probe binding molecule and nucleic acid barcode molecule may further comprise one or more additional sequences (e.g., unique molecular identifiers (UMIs), a barcode sequence, a primer sequence, a capture sequence, or other functional sequence).
  • UMIs unique molecular identifiers
  • the probe binding molecule or barcode molecule may comprise a functional sequence, a primer sequence (e.g., sequencing primer sequence or partial sequencing primer sequence), a UMI, etc.
  • the probe binding molecule and the nucleic acid barcode molecule may be any useful length; for example, either or both may be about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 20, about 30, about 40, about 50, about 60, about 70, about 80, about 90, about 100 or more nucleotides in length.
  • the probe binding molecule or the barcode molecule may be at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90, at least 100 or more nucleotides in length.
  • the probe capture binding molecule or the barcode molecule may be at most 100, at most 90, at most 80, at most 70, at most 60, at most 50, at most 40, at most 30, at most 20, at most 10, at most 9, at most 8, at most 7, at most 6, at most 5, at most 4, at most 3, at most 2, or at most 1 nucleotide in length.
  • a range of lengths of the probe binding molecule or barcode molecule may be used, such as from about 16 to about 100 nucleotides in length, etc.
  • the probe binding molecule or barcode molecule length may be varied based on any useful application and properties, e.g., melting temperature, annealing temperature, etc.
  • the first target region and the second target region of the nucleic acid molecule are not adjacent.
  • the first target region and the second target region may be separated by one or more gap regions disposed between the first target region and the second target region.
  • the gap region may comprise, for example, at least one nucleotide base, at least about 2, at least about 3, at least about 4, at least about 5, at least about 6, at least about 7, at least about 8, at least about 9, at least about 10, at least about 20, at least about 30, at least about 40, at least about 50, at least about 60, at least about 70, at least about 80, at least about 90, at least about 100, at least about 200, at least about 300, at least about 400, at least about 500, or more bases.
  • the gap region may comprise at most about 1000, at most about 500, at most about 400, at most about 300, at most about 200, at most about 100, at most about 90, at most about 80, at most about 70, at most about 60, at most about 50, at most about 40, at most about 30, at most about 20, at most about 10, or at most about 5 bases.
  • the gap region may comprise a range of number of bases, such as between about 1 and 30 bases.
  • a target region of the nucleic acid molecule may have one or more useful characteristics. For example, a target region may have any useful length, base content, sequence, melting point, or other characteristic.
  • a target region may comprise, for example, at least 10 bases, such as at least about 20, 25, 30, 35, 40, 45, 50, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 350, 400, 450, 500, or more bases.
  • a target region may have any useful base content and any useful sequence and combination of bases.
  • a target region may comprise one or more adenine, thymine, uracil, cytosine, and/or guanine bases (e.g., natural or canonical bases).
  • a target region may also comprise one or more derivatives or modified versions of a natural or canonical base, such as an oxidized, alkylated (e.g., methylated), hydroxylated, or otherwise modified base.
  • a target region may comprise ribose or deoxyribose moieties and phosphate moieties or derivatives or modified versions thereof.
  • a target region of the nucleic acid molecule may comprise one or more sequences or features, or portions thereof, of the nucleic acid molecule.
  • a target region may comprise all or a portion of a UTR (e.g., a 3’ UTR or a 5’ UTR), a Kozak sequence, a Shine- Dalgamo sequence, a coding sequence, a polyA sequence, a cap structure, an intron, an exon, or any other sequence or feature of the nucleic acid molecule.
  • the nucleic acid molecule (e.g., RNA molecule, such as an mRNA molecule) of a sample may be included within a cell, nucleus or cell bead.
  • the sample may comprise a cell or nucleus comprising the nucleic acid molecule.
  • the cell, nucleus, or cell bead may comprise additional nucleic acid molecules that may be the same as or different from the nucleic acid molecule of interest.
  • the sample may comprise a plurality of cells, and each cell may contain one or more nucleic acid molecules.
  • the cell may be, for example, a human cell, an animal cell, or a plant cell. In some cases, the cell may be derived from a tissue or fluid, as described herein.
  • the cell may be a prokaryotic cell or a eukaryotic cell.
  • the cell may be a lymphocyte such as a B cell or T cell.
  • the cell may be comprised within a bead, such as those disclosed in U.S. Pat. No. 10,428,326, which is incorporated by reference herein in its entirety.
  • the cell is comprised within a tissue sample and may be fixed to a substrate.
  • the cell may be a cell of a formalin-fixed, paraffin-embedded (FFPE) sample, as described above.
  • FFPE formalin-fixed, paraffin-embedded
  • the method may comprise additional operations for preparing the cell or nucleic acid molecule comprised therein, e.g., deparaffinization, staining (e g., using immunological agents) or destaining, decrosslinking, washing, enzymatic treatment, etc. Additional examples of treating FFPE samples prior to and following hybridization of probes are included in PCT/US2020/066720, which is included by reference herein in its entirety.
  • Additional examples of treating FFPE samples prior to and following hybridization of probes are included in PCT/US2020/066720, which is included by reference herein in its entirety.
  • Access to a nucleic acid molecule included in a cell, nucleus or cell bead may be provided by lysing or permeabilizing the cell or nucleus.
  • Lysing the cell, nucleus or cell bead may release the nucleic acid molecule contained therein from the cell, nucleus or cell bead.
  • a cell or nucleus may be lysed using a lysis agent such as a bioactive agent.
  • a bioactive agent useful for lysing a cell or nucleus may be, for example, an enzyme (e.g., as described herein).
  • An enzyme used to lyse a cell or nucleus may or may not be capable of carrying out additional functions such as degrading, extending, reverse transcribing, or otherwise altering a nucleic acid molecule.
  • an ionic or non-ionic surfactant such as TritonX-100, Tween 20, sarcosyl, or sodium dodecyl sulfate may be used to lyse a cell or nucleus.
  • Cell/nucleus lysis may also be achieved using a cellular disruption method such as an electroporation or a thermal, acoustic, or mechanical disruption method.
  • a cell or nucleus may be permeabilized to provide access to a nucleic acid molecule included therein. Permeabilization may involve partially or completely dissolving or disrupting a cell/nuclear membrane or a portion thereof.
  • Permeabilization may be achieved by, for example, contacting a cell membrane with an organic solvent (e.g., methanol) or a detergent such as Triton X-100 or NP-40.
  • an organic solvent e.g., methanol
  • a detergent such as Triton X-100 or NP-40.
  • the cell, nucleus or cell bead may be fixed, as described elsewhere herein.
  • the cell may be lysed within the cell bead, and a subset of the intracellular contents may associate with the bead.
  • the cell bead may comprise thioacrydite-modified nucleic acid molecules that can hybridize with nucleic acids from the cell.
  • a poly-T nucleic acid sequence may be thioacrydite-modified and bound to the cell bead matrix.
  • the cellular nucleic acids e.g., mRNA
  • the retained intracellular/intranuclear contents may be released, for example, by addition of a reducing agent, e.g., DTT, TCEP, etc. The release may occur at any convenient step, such as before or after partitioning.
  • the nucleic acid molecule or probe-associated nucleic acid molecule may be subjected to conditions sufficient to generate a probe-linked molecule.
  • the first target region may be adjacent to the second target region, and the first probe and the second probe may hybridize to the first target region and the second target region, respectively.
  • the first probe may comprise a first reactive moiety
  • the second probe may comprise a second reactive moiety.
  • the first reactive moiety of the first probe is adjacent to the second reactive moiety of the second probe.
  • the reactive moi eties may then be subjected to conditions sufficient to cause them to react to yield a probe-linked nucleic acid molecule comprising the first probe linked to the second probe.
  • the reactive moieties may be joined together via click chemistry or enzymatic ligation, such as those disclosed in in U.S. Pat. Pub. No. 2020/0239874, International Pub. No. WO 2019/165318, and International Pat. Pub. No. WO2021/237087, each of which is incorporated by reference herein in its entirety.
  • the first probe or the second probe may comprise an adenylated oligonucleotide or moiety (e.g., an adenylated phosphate group), which may be useful in reducing non-specific ligation reactions.
  • the linking of the probes may be performed in substantially ATP-free conditions, optionally using an enzyme (e.g., ligase) that does not require ATP (e.g., truncated T4 RNA ligase) or that is pre-activated (e.g., a preactivated T4 DNA ligase).
  • an enzyme e.g., ligase
  • ligase that does not require ATP
  • pre-activated e.g., a preactivated T4 DNA ligase.
  • Additional examples of such ligation schemes can be found in PCT/US2020/066720 and International Pat. App. No. PCT/US2021/33649, filed May 21, 2021, which is incorporated by reference herein in its entirety.
  • the first target region of the nucleic acid molecule may not be adjacent to the second target region.
  • the nucleic acid molecule may be subjected to conditions sufficient for hybridization of the first probe sequence of the first probe to the first target region to generate a probe-associated nucleic acid molecule.
  • the probe-associated nucleic acid molecule may be subjected to a nucleic acid reaction (e g., a nucleic acid extension reaction, reverse transcription, etc.) to generate an extended probe molecule comprising a sequence complementary to the second target region.
  • a second probe comprising a second probe sequence may hybridize to the extended probe molecule (or complement thereof) and subjected to conditions sufficient (e.g., nucleic acid extension, amplification, hybridization of additional probe molecules, ligation, etc.) to generate a probe- linked molecule comprising a sequence corresponding to the first target region and a sequence corresponding to the second target region.
  • the first probe and the second probe may be provided simultaneously, and following hybridization of the first probe sequence and the second probe sequence to the first target region and the second target region, respectively, to generate a dual-probe-associated nucleic acid molecule, the gap (e.g., the region disposed between the first target region and the second region) may be filled (e.g., via a nucleic acid extension or gap-fill reaction and/or hybridization of additional probe molecules that hybridize to at least a portion of the gap region).
  • the gap e.g., the region disposed between the first target region and the second region
  • the gap may be filled (e.g., via a nucleic acid extension or gap-fill reaction and/or hybridization of additional probe molecules that hybridize to at least a portion of the gap region).
  • one or both probes may comprise an overhang or flap sequence (e.g., at a 5’ end) that is recognizable or cleavable by an enzyme (e.g., an endonuclease such as FEN1 endonuclease).
  • the second probe may comprise a 5’ flap sequence that is cleaved by FEN1 endonuclease if at least a specific portion of the second probe hybridizes to the nucleic acid molecule (e.g., target molecule).
  • an endonuclease e.g., FEN1
  • FEN1 an endonuclease
  • the gap region may be filled, followed by cleavage of the flap sequence.
  • the first probe or the second probe and the gap-filled region may be ligated, e.g., chemically or enzymatically. Additional examples of systems and methods for generating probe-linked nucleic acid molecules and gap-filling reactions can be found, for example in U.S. Pat. Pub. No. 2020/0239874, International Pub. No. WO 2019/165318, and International Pat. Pub. No. WO2021/237087, each of which is incorporated by reference herein in its entirety.
  • the probe-linked nucleic acid molecule may be barcoded to provide a barcoded probe-linked nucleic acid molecule, or barcoding may occur prior to generation of the probe- linked nucleic acid molecule. Barcoding may be performed using a variety of techniques.
  • the first probe or the second probe may comprise a probe capture sequence.
  • the nucleic acid barcode molecule may comprise a barcode capture sequence capable of hybridizing to the probe capture sequence.
  • barcoding may be mediated by a probe binding molecule (e.g., a splint oligonucleotide) comprising (i) a probe binding sequence, which may be complementary to the probe capture sequence of the first probe or the second probe, and (ii) a barcode binding sequence, which may be complementary to the barcode capture sequence of the nucleic acid barcode molecule.
  • a probe binding molecule e.g., a splint oligonucleotide
  • a barcode binding sequence which may be complementary to the barcode capture sequence of the nucleic acid barcode molecule.
  • the barcoding may be followed by ligation, e.g., chemically or enzyme-mediated, to covalently link the nucleic acid barcode molecule to the probe (or to the probe binding sequence, and the probe binding sequence may be ligated to the probe).
  • Examples of chemical ligation of nucleic acid molecules may include “click chemistry” approaches, e.g., reaction of azide and alkyne moi eties, as described in U.S. Pat. Pub. No. 2020/0239874, which is incorporated by reference herein in its entirety.
  • the first probe may comprise a first probe sequence and a probe capture sequence, and the first probe may be subjected to conditions sufficient to hybridize the first probe sequence to the first target region, thereby generating a probe-associated nucleic acid molecule.
  • the probe-associated nucleic acid molecule may be subjected to washing or other conditions to remove unannealed probes from a mixture.
  • the probe-associated nucleic acid molecule may be extended from an end of the first probe towards an end of the nucleic acid molecule to which it is hybridized (towards the end which is proximal to the second target region) to provide an extended nucleic acid molecule.
  • the extended nucleic acid barcode molecule may comprise the first probe sequence and a complement to the second target region.
  • the extended nucleic acid molecule may be barcoded, e.g., by hybridizing the barcode capture sequence of the nucleic acid barcode molecule to the probe capture sequence, or by hybridizing (i) a probe-binding molecule comprising a probe binding sequence and a barcode binding sequence to the probe capture sequence and (ii) the barcode capture sequence of the nucleic acid barcode molecule to the barcode binding sequence of the probe binding molecule.
  • the probe-binding molecule may be provided pre-annealed to the nucleic acid barcode molecule. Subsequently, a second probe comprising a second probe sequence may be provided.
  • the barcoded, extended nucleic acid molecule may be subjected to conditions sufficient to hybridize the second probe sequence to the second target region or complement thereof.
  • a nucleic acid extension reaction may be performed, thereby generating a barcoded molecule (e.g., barcoded probe-linked molecule) comprising a sequence corresponding to the first target region, a sequence corresponding to the second target region, a sequence corresponding to the probe capture sequence, and a sequence corresponding to the barcode sequence.
  • FIG. 7 schematically shows a method for generating a barcoded nucleic acid molecule, as described herein.
  • a nucleic acid molecule (e.g., RNA molecule) 700 comprising a first target region 702 and a second target region 704 may be provided.
  • the nucleic acid molecule 700 may be contacted with a first probe 706 comprising a first probe sequence 708 and, optionally, a functional sequence 710, thereby generating a probe-associated nucleic acid molecule.
  • the first probe sequence 708 may be complementary to the first target region 702.
  • the functional sequence 710 may comprise, for instance, a probe capture sequence used for downstream barcoding, or it may comprise a different functional sequence, such as a primer sequence, a partial primer sequence, a barcode sequence, a sequencing primer sequence, etc.
  • the probe-associated nucleic acid molecule may be subjected to conditions sufficient to extend the first probe 706, thereby generating an extended probe molecule 712 comprising a sequence complementary to the second target region 704.
  • the extended probe molecule 712 may be released from the nucleic acid molecule 700, e.g., via denaturing and/or degrading the nucleic acid molecule 700 (e g., using an RNAse, increased temperature or heat cycling, pH, etc ).
  • a nucleic acid barcode molecule may be provided.
  • the nucleic acid barcode molecule may be partially double-stranded and may comprise a first strand 720 comprising a barcode sequence, and a second strand 722 comprising a sequence 724 at least partially complementary to the barcode sequence and a probe binding sequence 726, which may be at least partially complementary to the functional sequence (e.g., probe capture sequence) 710 of the first probe 706.
  • the nucleic acid barcode molecule is single-stranded and comprises only first strand 720 comprising the barcode sequence and a barcode capture sequence.
  • a probe binding molecule (e.g., a splint oligonucleotide) 722 may be provided, comprising barcodebinding sequence 724, which is at least partially complementary to the barcode capture sequence, and the probe binding sequence 726.
  • the probe binding molecule and the nucleic acid barcode molecule may be provided as a pre-annealed complex.
  • the nucleic acid barcode molecule (or the pre-annealed complex) may be coupled to a bead, such as a gel bead, as described herein, and may comprise additional functional sequences, including, but not limited to, a unique molecular identifier (UMI), a capture sequence, a primer sequence (e.g., a R1/R2 sequence).
  • UMI unique molecular identifier
  • a capture sequence e.g., a R1/R2 sequence
  • the extended probe molecule may be barcoded by hybridizing the probe binding sequence 726 to the functional sequence (e.g., probe capture sequence 710).
  • the nucleic acid barcode molecule may be covalently linked to the extended probe molecule (e.g., via the probe capture sequence), e.g., enzymatically (e.g., using a ligase) or chemically (e.g., using click chemistry).
  • a second probe molecule 716 may be provided.
  • operation 707 may also include a denaturation of the doublestranded molecule.
  • the second probe molecule 716 may comprise a second probe sequence 714 corresponding to the second target region 704 and optionally a functional sequence 718, which may comprise a probe capture sequence, a barcode sequence, a primer sequence, a sequencing primer sequence, etc.
  • a nucleic acid extension reaction may be performed, e.g., using a polymerase, to extend the second probe 716 along the extended probe molecule, thereby generating a barcoded molecule comprising a sequence corresponding to the first target region 702, the second target region 704, a sequence corresponding to the probe capture sequence 710, and a sequence corresponding to the barcode sequence 720.
  • the first probe and the second probe may be linked (e.g., by chemical ligation or enzymatic extension and/or ligation) prior to barcoding.
  • the first probe may be hybridized to the nucleic acid molecule (e.g., via hybridization of the first probe sequence to the first target region) to generate a probe-associated nucleic acid molecule.
  • the probe-associated nucleic acid molecule may be extended from an end of the first probe to an end of the nucleic acid molecule to which it is hybridized, to provide an extended nucleic acid molecule.
  • the extended molecule may be subjected to conditions sufficient to hybridize the second probe to the second target region or complement thereof (e.g., via hybridization of the second probe sequence to the second target region or complement thereof).
  • An additional nucleic acid extension reaction may be performed, to generate an extended, and the resultant extension product may be barcoded, generating a barcoded molecule.
  • the barcoded molecule may comprise a sequence corresponding to the first target region, a sequence corresponding to the second target region, a sequence corresponding to the probe capture sequence, and a sequence corresponding to the barcode sequence.
  • the nucleic acid barcode molecule (or the probe binding molecule) may be chemically linked to the first probe or the second probe, such as by ligation or click chemistry.
  • the nucleic acid barcode molecule may comprise a first reactive moiety, and the first or the second probe may comprise a second reactive moiety; the first reactive moiety may be configured to react with the second reactive moiety to generate a covalent linkage.
  • Barcoded nucleic acid molecules or derivatives thereof may then be optionally further processed and analyzed by any suitable technique, including nucleic acid sequencing (e.g., Illumina sequencing).
  • FIG. 8 schematically shows another method for generating a barcoded nucleic acid molecule, as described herein.
  • a nucleic acid molecule (e.g., RNA molecule) 800 comprising a first target region 802 and a second target region 804 may be provided.
  • the nucleic acid molecule 800 may be contacted with a first probe 806 comprising a first probe sequence 808 and, optionally, a functional sequence 810, thereby generating a probe-associated nucleic acid molecule.
  • the first probe sequence 808 may be complementary to the first target region 802.
  • the functional sequence 810 may comprise, for instance, a probe capture sequence used for downstream barcoding, or it may comprise a different functional sequence, such as a primer sequence, a partial primer sequence, a barcode sequence, a sequencing primer sequence, etc.
  • the probe-associated nucleic acid molecule may be subjected to conditions sufficient to extend the first probe 806, thereby generating an extended probe molecule 812 comprising a sequence complementary to the second target region 804.
  • the extended probe molecule 812 may be released from the nucleic acid molecule 800, e.g., via denaturing and/or degrading the nucleic acid molecule 800 (e g., using an RNAse, increased temperature or heat cycling, pH, etc ).
  • a nucleic acid barcode molecule and a second probe 816 may be provided.
  • the second probe 816 may comprise a second probe sequence 814 corresponding to the second target region 804 and optionally a functional sequence 818, which may comprise a probe capture sequence.
  • the nucleic acid barcode molecule may be partially double-stranded and may comprise a first strand 820 comprising a barcode sequence, and a second strand 822 comprising a sequence 824 complementary to the barcode sequence and a probe binding sequence 826, which may be complementary to the functional sequence (e.g., probe capture sequence) 818 of the second probe 816.
  • the nucleic acid barcode molecule is single-stranded and comprises only first strand 820 comprising the barcode sequence and a barcode capture sequence.
  • a probe binding molecule e.g., a splint oligonucleotide
  • the probe binding molecule and the nucleic acid barcode molecule may be provided as a pre-annealed complex.
  • the nucleic acid barcode molecule (or the pre-annealed complex) may be coupled to a bead, such as a gel bead, as described herein, and may comprise additional functional sequences, including, but not limited to, a unique molecular identifier (UMI), a capture sequence, a primer sequence (e.g., a R1/R2 sequence).
  • UMI unique molecular identifier
  • the second probe 816 may hybridize to the extended probe molecule 812 (e.g., via hybridization of the second probe sequence 814 to the second target region 804 or complement thereof), and the nucleic acid barcode molecule may be attached or coupled to the second probe 816, e.g., via hybridization of the probe binding sequence 826 to the probe capture sequence 818.
  • the nucleic acid barcode molecule or the probe binding molecule may be ligated to the second probe 816, e.g., using a ligase or via chemical linkage, such as click chemistry.
  • a nucleic acid extension reaction may be performed, e g., using a polymerase (e.g., DNA polymerase, Hot Start polymerase, etc.), to extend the nucleic acid barcode molecule and the second probe 816 along the extended probe molecule, thereby generating a barcoded molecule comprising a sequence corresponding to the first target region 802, the second target region 804, a sequence corresponding to the probe capture sequence 818, and a sequence corresponding to the barcode sequence 820.
  • Barcoded nucleic acid molecules or derivatives thereof may then be optionally further processed and analyzed by any suitable technique, including nucleic acid sequencing (e.g., Illumina sequencing).
  • FIG. 9 schematically shows another method for generating a barcoded nucleic acid molecule, similar to that shown in FIG. 8.
  • a nucleic acid molecule (e.g., RNA molecule) 900 comprising a first target region 902 and a second target region 904 may be provided.
  • the nucleic acid molecule 900 may be contacted with a first probe 906 comprising a first probe sequence 908 and, optionally, a functional sequence 910, thereby generating a probe-associated nucleic acid molecule.
  • the first probe sequence 908 may be complementary to the first target region 902.
  • the functional sequence 910 may comprise, for instance, a probe capture sequence, or it may comprise a different functional sequence, such as a primer sequence, a partial primer sequence, a barcode sequence, a sequencing primer sequence, etc.
  • the probe-associated nucleic acid molecule may be subjected to conditions sufficient to extend the first probe 906, thereby generating an extended probe molecule 912 comprising a sequence complementary to the second target region 906.
  • the extended probe molecule 912 may be released from the nucleic acid molecule 900, e.g., via denaturing and/or degrading the nucleic acid molecule 900 (e g., using an RNAse, increased temperature or heat cycling, pH, etc ).
  • a second probe 916 may be provided.
  • the second probe 916 may comprise a second probe sequence 914 corresponding to the second target region 904 and optionally a functional sequence 918, which may comprise a probe capture sequence.
  • a nucleic acid extension reaction may be performed, e.g., using a polymerase, to extend the nucleic acid barcode molecule and the second probe 916 along the extended probe molecule, thereby generating a probe-linked molecule comprising a sequence corresponding to the first target region 902 and the second target region 904.
  • a nucleic acid barcode molecule may also be provided with the second probe.
  • the nucleic acid barcode molecule may be partially doublestranded and may comprise a first strand 920 comprising a barcode sequence, and a second strand 922 comprising a sequence 924 complementary to the barcode sequence and a probe binding sequence 926, which may be complementary to the functional sequence (e.g., probe capture sequence) 918 of the second probe 916.
  • the nucleic acid barcode molecule is single-stranded and comprises only first strand 920 comprising the barcode sequence and a barcode capture sequence.
  • a probe binding molecule (e.g., a splint oligonucleotide) 922 may be provided, comprising barcode-binding sequence 924 that is complementary to the barcode capture sequence, and the probe binding sequence 926.
  • the probe binding molecule and the nucleic acid barcode molecule may be provided as a pre-annealed complex.
  • the nucleic acid barcode molecule (or the pre-annealed complex) may be coupled to a bead, such as a gel bead, as described herein, and may comprise additional functional sequences, including, but not limited to, a unique molecular identifier (UMI), a capture sequence, a primer sequence (e.g., a R1/R2 sequence).
  • UMI unique molecular identifier
  • a capture sequence e.g., a R1/R2 sequence
  • the nucleic acid barcode molecule may be attached or coupled to the second probe 916, e.g., via hybridization of the probe binding sequence 926 to the probe capture sequence 918.
  • the resultant barcoded product may comprise a sequence corresponding to the first target region 902, the second target region 904, a sequence corresponding to the probe capture sequence 918, and a sequence corresponding to the barcode sequence 920.
  • the nucleic acid barcode molecule may be covalently linked to the extended probe molecule (e.g., via the probe capture sequence 918), e.g., enzymatically (e.g., using a ligase) or chemically (e.g., using click chemistry). Barcoded nucleic acid molecules or derivatives thereof may then be optionally further processed and analyzed by any suitable technique, including nucleic acid sequencing (e.g., Illumina sequencing).
  • the methods of the present disclosure may comprise generating probe-associated nucleic acid molecules, and barcoding the probe-associated nucleic acid molecules, optionally with a linking operation (e.g., prior to or subsequent to barcoding of the probe-associated nucleic acid molecules).
  • a nucleic acid molecule e.g., RNA molecule
  • the nucleic acid molecule may be contacted with (i) a first probe comprising a first probe sequence complementary to the first target region and (ii) a second probe comprising a second probe sequence complementary to the second target region, thereby generating a probe-associated nucleic acid molecule.
  • the probe-associated nucleic acid molecule may be subjected to conditions sufficient to link the first probe to the second probe (e.g., enzymatically, such as with a polymerase, reverse transcriptase, and/or ligase, or chemically), thereby generating a probe-linked nucleic acid molecule.
  • the probe-associated nucleic acid molecule or the probe-linked molecule may subsequently be barcoded (e.g., in a partition) to generate a barcoded nucleic acid molecule.
  • FIG. 25 schematically shows an example method for generating a probe-linked nucleic acid molecule, which may subsequently be barcoded, e.g., in a partition, to generate a barcoded nucleic acid molecule.
  • a nucleic acid molecule (e.g., RNA molecule) 2500 comprising a first target region 2502 and a second target region 2504 may be provided. In some instances, the first target region is adjacent to the second target region.
  • the nucleic acid molecule 2500 may be contacted, in operation 2501, with a first probe 2506 comprising a first probe sequence 2508 complementary to the first target region 2502 and a second probe 2516 comprising a second probe sequence 2514 complementary to the second target region 2504, thereby generating a probe-associated nucleic acid molecule.
  • the first probe 2506 and/or the second probe 2516 may comprise a functional sequence, e.g., a probe capture sequence, a primer sequence, a partial primer sequence, a barcode sequence, a sequencing primer sequence, etc.
  • one of the probes comprises a flap or overhang sequence 2530, which may be recognized by an endonuclease (e.g., FEN1) upon annealing of the second probe sequence 2514 to the second target region 2504.
  • the second probe 2516 may comprise a 5’ flap sequence 2530, and subsequent to annealing of the first probe 2506 and the second probe 2516 to the nucleic acid molecule 2500, the flap sequence may be adjacent to an end of the first probe (e g., a 3’ end) as well as an end of the second probe (e g., a 5’ end).
  • an endonuclease e.g., FEN1 may be used to remove the flap sequence 2530 i leaving a ligatable end (e.g., 5 ’phosphorylated end) of the second probe 2516.
  • a ligation reaction may be performed (e.g., using a ligase) to link the first probe to the second probe, thereby generating a probe-linked nucleic acid molecule.
  • the probe-linked nucleic acid molecule may subsequently be barcoded, e.g., in partitions, as is described elsewhere herein. In some instances, the probe-associated nucleic acid molecules may be barcoded and linked (e.g., in partitions).
  • FIG. 26 shows another example workflow, similar to that shown in FIG. 25, in which the target regions of the nucleic acid molecule are not adjacent.
  • a workflow may comprise an additional gap-fill reaction to generate the probe-associated molecule.
  • the first target region 2602 of nucleic acid molecule 2600 may not be adjacent to the second target region 2604.
  • the a gap region may be disposed between the first target region and the second target region.
  • the first probe 2606 may anneal to the first target region 2602 and the second probe 2616 may anneal to the second target region 2604.
  • an extension reaction (e.g., using a polymerase, reverse transcriptase, etc.) may be performed to fill in the gap region between the first probe 2606 and the second probe 2616, yielding a gap-filled nucleic acid molecule.
  • the second probe 2616 comprises a flap sequence 2630.
  • an endonuclease e.g., FEN1 may be used to remove the flap sequence 2630, leaving a ligatable end (e.g., 5 ’phosphorylated end) of the second probe 2616.
  • a ligation reaction may be performed (e g., using a ligase) to link the first probe to the second probe, thereby generating a probe-linked nucleic acid molecule.
  • the probe-linked nucleic acid molecule, or alternatively, the un-linked molecule, may be barcoded, e.g., in a partition.
  • FIG. 27 shows an additional scheme of generating a probe-linked nucleic acid molecule by performing a gap-filling reaction using a third probe.
  • a first probe 2706 and a second probe 2716 anneal (e.g., via a first probe sequence and a second probe sequence, respectively) to a first target region 2702 and a second target region 2704 of nucleic acid molecule 2700 to generate a probe-associated nucleic acid molecule.
  • a gap sequence may be disposed between the first target region 2702 and the second target region 2704.
  • Third probe molecules 2770 may be provided (illustrated as two different probe molecules, which may be used for SNP detection), which may anneal to the gap sequence (FIG. 27 Panel B).
  • FIG. 27 Panel A a first probe 2706 and a second probe 2716 anneal (e.g., via a first probe sequence and a second probe sequence, respectively) to a first target region 2702 and a second target region 2704 of nucleic acid molecule 2700 to generate
  • the first probe, the third probe, and the second probe may be ligated (e.g., using a ligase) to generate a probe-linked nucleic acid molecule.
  • the probe-linked nucleic acid molecule, or alternatively, the probe-associated nucleic acid molecule may be barcoded, e.g., in a partition.
  • FIG. 28 shows an example of a ligation scheme used to generate probe-linked nucleic acid molecules.
  • the probe molecules may hybridize to the nucleic acid molecule.
  • the first probe may be ligated to the second probe, optionally with a gap-fill operation, as described above, using an enzyme.
  • the enzyme may be a preactivated enzyme, e.g., a preactivated T4 DNA ligase, and the ligation may occur under ATP- reduced or ATP-removed conditions, e.g. using Apyrase.
  • the nucleic acid barcode molecule may be attached (e g., via hybridization) to either the first probe and/or the second probe (e.g., via a probe capture sequence comprised in the first probe or the second probe).
  • the first probe and the second probe may comprise any useful functional sequences, such as primer sequences, barcode sequences, unique molecular identifier (UMI) sequences, flow cell attachment sequences, primer-binding sequences, capture sequences, etc.
  • the first probe may hybridize to the left-hand side (e g., a 3’ end) of a nucleic acid molecule (e g., 700, 800, or 900) or to the right-hand side (e.g., a 5’ end).
  • the second probe may hybridize to the left-hand side or to the right-hand side of the nucleic acid molecule.
  • one or more extension reactions may be performed on the probe- hybridized nucleic acid molecules.
  • the probe may be extended from an end of the probe to an end of the nucleic acid barcode molecule, or a second probe may be extended from an end of the second probe to an end of the first probe of a probe-associated nucleic acid molecule.
  • Extension may comprise the use of an enzyme (e.g., a polymerase, reverse transcriptase) to add one or more nucleotides to the end of the probe.
  • Extension may provide an extended nucleic acid molecule comprising sequences complementary to the target region of the nucleic acid molecule of interest, the barcode sequence, and optionally, one or more additional sequences of the nucleic acid barcode molecule such as one or more binding sequences.
  • appropriate conditions and or chemical agents e.g., as described herein
  • one or more processes may involve the use of thermosensitive agents. For example, in some cases, probes may be annealed or hybridized under one set of temperature conditions, and extension may occur under a different set of temperature conditions. In some cases, a Warm or Hot Start polymerase may be used.
  • hybridization of the nucleic acid barcode molecule to one or more of the probes may precede hybridization of the probe to the target region of the nucleic acid molecule.
  • the barcoded nucleic acid molecule may be duplicated or amplified by, for example, one or more amplification reactions.
  • the amplification reactions may comprise polymerase chain reactions (PCR) and may involve the use of one or more primers or polymerases.
  • PCR polymerase chain reactions
  • the extension, denaturation, and/or amplification processes may take place within a partition, or in bulk.
  • the extended nucleic acid molecule or derivatives thereof may be duplicated or amplified within a partition to provide an amplified product.
  • the barcoded product, or a complement thereof e.g., an amplified product
  • the nucleic acid molecule or a derivative thereof e g., a probe-linked nucleic acid molecule, a nucleic acid molecule having one or more probes hybridized thereto, a barcoded probe-linked nucleic acid molecule, or an extended nucleic acid molecule or complement thereof
  • a cell or cell bead comprising the nucleic acid molecule or a derivative thereof may be provided within a partition such as a well or droplet, e.g., as described herein.
  • One or more reagents may be co-partitioned with a nucleic acid molecule or a derivative thereof or a cell comprising the nucleic acid molecule or a derivative thereof.
  • a nucleic acid molecule or a derivative thereof or a cell comprising the nucleic acid molecule or a derivative thereof may be co-partitioned with one or more reagents selected from the group consisting of lysis agents or buffers, permeabilizing agents, enzymes (e.g., enzymes capable of digesting one or more RNA molecules, extending one or more nucleic acid molecules, reverse transcribing an RNA molecule, permeabilizing or lysing a cell, or carrying out other actions), fluorophores, oligonucleotides, primers, probes, barcodes, nucleic acid barcode molecules (e.g., nucleic acid barcode molecules comprising one or more barcode sequences), buffers, deoxynucleotide triphosphates, detergents, reducing agents, chelating agents, oxidizing agents, nanoparticles, beads, and antibodies.
  • one or more reagents selected from the group consisting of lysis agents or buffers, permeabilizing agents, enzymes
  • a nucleic acid molecule or a derivative thereof, or a cell comprising the nucleic acid molecule or a derivative thereof may be copartitioned with one or more reagents selected from the group consisting of temperaturesensitive enzymes, pH-sensitive enzymes, light-sensitive enzymes, reverse transcriptases, proteases, ligase, polymerases, restriction enzymes, nucleases, protease inhibitors, exonucleases, and nuclease inhibitors.
  • a nucleic acid molecule or a derivative thereof or a cell comprising the nucleic acid molecule or a derivative thereof may be co-partitioned with a polymerase and nucleotide molecules.
  • Partitioning a nucleic acid molecule or a derivative thereof or a cell comprising the nucleic acid molecule or a derivative thereof and one or more reagents may comprise flowing a first phase comprising an aqueous fluid, the cell, and the one or more reagents and a second phase comprising a fluid that is immiscible with the aqueous fluid toward a junction.
  • a discrete droplet of the first phase comprising the nucleic acid molecule or a derivative thereof or a cell comprising the nucleic acid molecule or a derivative thereof (e.g., a cell bead) and the one or more reagents may be formed.
  • the partition may comprise a single cell.
  • the cell may be lysed or permeabilized within the partition (e.g., droplet) to provide access to the nucleic acid molecule of the cell.
  • One or more processes may be carried out within a partition (e.g., droplet, well, etc.).
  • the nucleic acid molecule, or a cell or cell bead comprising the nucleic acid molecule may be co-partitioned with one or more reagents (e.g., as described herein) at any useful stage of the method.
  • the probe-associated nucleic acid molecule e.g., the nucleic acid molecule with the first probe hybridized thereto
  • the extended probe molecule may be subsequently partitioned in a partition among a plurality of partitions.
  • the partition may comprise the second probe and a nucleic acid barcode molecule and optionally, a probe binding molecule.
  • the second probe may hybridize (e.g., via the second probe sequence) to the second target region or complement thereof of the probe- associated molecule.
  • the partition may comprise additional reagents for performing a nucleic acid reaction (e.g., digestion, ligation, extension, amplification).
  • the probe- associated nucleic acid molecule may comprise or be hybridized to the nucleic acid molecule
  • the partition may comprise a degrading enzyme (e.g., RNAse), which may be useful in digesting or removing the template strand (e.g., the nucleic acid molecule, such as an RNA molecule) from the extended probe molecule.
  • the partition may comprise a polymerase, which may be used to extend the second probe hybridized to the extended probe molecule.
  • the partition comprises a linking enzyme (e.g., ligase), which may be used to ligate the nucleic acid barcode molecule to the first probe or the second probe (e.g., via a probe capture sequence).
  • the ligase may in some instances be used to ligate the probe binding molecule to the probe capture sequence of the first probe or the second probe.
  • the probe binding molecule, the probe capture sequence, and/or the barcode capture sequence comprises one or more reactive moieties, which may be used to chemically or enzymatically link the nucleic acid barcode molecule to the probe capture sequence, or complement thereof.
  • the resultant barcoded product may comprise a sequence corresponding to the first target region, a sequence corresponding to the second target region, a sequence corresponding to the probe capture sequence, and a sequence corresponding to the barcode sequence.
  • operation 701 may be performed in bulk (e.g., outside a partition), while operations 703, 705 may be performed in a partition.
  • Operations 707 and 709 may be performed in bulk or within the partition.
  • operation 801 may be performed in bulk, while operation 803 may be performed in a partition.
  • Operation 805 may be performed in bulk or in a partition.
  • operation 901 may be performed in bulk, while operations 903, 905, and 907 may be performed in a partition. It will be appreciated that any of the operations may be performed in bulk or in partitions at any convenient step and that the order of the operations may be changed for a suitable or useful purpose.
  • the nucleic acid molecule or the cell or cell bead comprising the nucleic acid molecule, or derivatives thereof may be released from a partition at any useful stage of the method.
  • the extended probe molecule may be hybridized to the second probe and released from the partition subsequent to hybridization of the barcode capture sequence of the nucleic acid barcode molecule to the first probe, the second probe, or the probe binding molecule.
  • the extended probe molecule may be released from the partition subsequent to (i) hybridization of the second probe and nucleic acid barcode molecule and (ii) extension of the second probe to generate the barcoded molecule comprising a sequence corresponding to the first target region, a sequence corresponding to the second target region, a sequence corresponding to the probe capture sequence, and a sequence corresponding to the barcode sequence.
  • Duplication and/or amplification of the extended nucleic acid molecule may be carried out within the partition or in bulk, e.g., within a solution. In some cases, the solution may comprise additional extended nucleic acid molecules generated through the same process carried out in different partitions.
  • Each extended nucleic acid molecule may comprise a different barcode sequence, and the barcode sequence may be useful in identifying the partition or cell from whence the extended nucleic acid molecules originated.
  • the solution may comprise a pooled mixture comprising the contents of two or more partitions (e.g., droplets).
  • Additional processes or operations may be performed within a partition, including, but not limited to: lysis, permeabilization, denaturation, hybridization, extension, duplication, and amplification of one or more components of a sample. In some cases, multiple processes are carried out within a partition.
  • Hybridization of the probe sequences to the target regions of the nucleic acid molecule may be performed within or outside of a partition. In some cases, hybridization may be preceded by denaturation of a double-stranded nucleic acid molecule to provide a singlestranded nucleic acid molecule or by lysis or permeabilization of a cell. In some cases, the hybridization may occur in a cell bead comprising a cell.
  • the sequence of the probe that is complementary to the target region may be situated at an end of the probe. Alternatively, this sequence may be disposed between other sequences such that when the probe sequence is hybridized to the target region, additional probe sequences extend beyond the hybridized sequence in one or more directions.
  • the probe sequence that hybridizes to the target region of the nucleic acid molecule may be of the same or different length as the target region.
  • the probe sequence may be shorter than the target region and may only hybridize to a portion of the target region.
  • the probe sequence may be longer than the target region and may hybridize to the entirety of the target region and extend beyond the target region in one or more directions.
  • the probe may comprise one or more additional probe sequences.
  • the probe may comprise the probe sequence complementary to the target region and a second probe sequence.
  • the second probe sequence may have any useful length and other characteristics.
  • the probe may comprise one or more additional sequences or moieties, such as one or more barcode sequences or unique molecule identifier (UMI) sequences, adapter sequences, functional sequences (e.g., primer sequences, sequencing primer sequences, etc.).
  • UMI unique molecule identifier
  • one or more probe sequences of the probe may comprise a detectable moiety such as a fluorophore or a fluorescent moiety.
  • the first probe or the second probe may comprise a reactive moiety, as described elsewhere herein.
  • the first probe or the second probe may comprise an azide moiety, an alkyne moiety, a phosphorothioate moiety, an iodide moiety, an amine moiety, a phosphate moiety, or a combination thereof.
  • the first probe may comprise a first reactive moiety and the second probe may comprise a second reactive moiety, and reaction of the first reactive moiety and the second reactive moiety may be sufficient to yield a probe-linked molecule comprising the first probe linked to the second probe.
  • the first reactive moiety and the second reactive moiety is linked via ligation.
  • the first probe or the second probe may comprise one or more moieties or modified nucleotides to facilitate ligation, e.g., one or more ribonucleotides or dideoxynucleotides (ddNTPs), which may be ligated to a phosphorylated end of the second probe using a ligase (e.g., T4 DNA ligase, SplintR ligase).
  • a ligase e.g., T4 DNA ligase, SplintR ligase
  • the probe e.g., the first probe or the second probe
  • ligases Other suitable enzymes, e.g., ligases, may be used, for example, the enzymes and ligases disclosed in U.S. Provisional App. No. 63/171,031, filed April 5, 2021, which is incorporated herein by reference in its entirety.
  • a probe sequence of the probe may be capable of hybridizing with a sequence of a nucleic acid barcode molecule or a probe binding molecule (e.g., splint oligonucleotide).
  • a nucleic acid barcode molecule may comprise a first binding sequence (e.g., a barcode capture sequence) that is complementary to a probe sequence of the probe (e.g., a probe capture sequence).
  • the nucleic acid barcode molecule may comprise one or more additional functional sequences , e.g., primer sequences, primer annealing sequences, and immobilization sequences.
  • the binding sequences may have any useful length and other characteristics.
  • the binding sequence (e g., barcode capture sequence) that is complementary to a probe sequence of the probe may be the same length as the probe sequence.
  • the binding sequence may be a different length of the probe sequence.
  • the binding sequence may be shorter than the probe sequence and may only hybridize to a portion of the probe sequence.
  • the binding sequence may be longer than the probe sequence and may hybridize to the entirety of the probe sequence and extend beyond the probe sequence in one or more directions.
  • the binding sequence (e.g., barcode capture sequence) of the nucleic acid barcode molecule may be the same length as the barcode binding sequence of the probe-binding molecule, or the binding sequence may be longer or shorter than the barcode binding sequence.
  • One or more processes described herein may be performed in a cell, nucleus or cell bead.
  • a plurality of cells, nuclei or cell beads may comprise a plurality of nucleic acid molecules.
  • the cells, nuclei or cell beads may be alive or fixed and/or permeabilized.
  • the first probes may be provided to the cells, nuclei or cell beads, such as in a bulk solution.
  • the cells, nuclei or cell beads may be washed to remove unbound first probes, and the nucleic acid extension reaction, as described herein, may be performed. Subsequently, the cells, nuclei or cell beads comprising the plurality of nucleic acid molecules (or the extended, probe nucleic acid molecules) may be partitioned into a plurality of separate partitions, where at least a subset of the plurality of separate partitions comprises a single cell, single nucleus, or single cell bead.
  • Access to a target nucleic acid molecule contained within a cell, nucleus or cell bead in a partition may be provided by lysing or permeabilizing the nucleus or cell (e.g., as described herein), which may be performed prior to or during partitioning.
  • access to a target nucleic acid molecule contained within a cell, nucleus, or cell bead may be performed in a bulk solution using the methods described herein.
  • Barcoding of the target nucleic acid may be performed within a cell, within a cell bead, or within a partition comprising the cell or cell bead.
  • the target nucleic acid molecule may be barcoded in a partition and outside the cell, cell nucleus, or cell bead.
  • Additional probe hybridization e.g., providing of the second probe
  • barcoding may comprise using a nucleic acid barcode molecule to attach or hybridize to the target nucleic acid molecule or derivative thereof (e.g., the extended probe molecule, or complement thereof).
  • Nucleic acid barcode molecules provided within each partition of the plurality of separate partitions may be provided attached to beads.
  • the nucleic acid barcode molecule may be releasably attached to a bead (e.g., via a labile bond).
  • Each partition (or a subset of partitions) of the plurality of separate partitions may comprise a bead comprising a plurality of nucleic acid barcode molecules attached thereto (e g., as described herein).
  • the plurality of nucleic acid barcode molecules attached to each bead may comprise a unique barcode sequence, such that each partition of the plurality of separate partitions comprises a different barcode sequence.
  • the barcoded molecules arising from a single cell, single nucleus, or single cell bead may have a same barcode sequence (e.g., a common barcode sequence), such that each barcoded nucleic acid molecule can be traced to a given partition and/or, in some instances, a given cell, nucleus or cell bead.
  • the methods described herein may comprise additional barcoding operations, which may be useful, for example, in indexing nucleic acid molecules to a cell, nucleus, cell bead, a sample, a partition, or a plurality of partitions. Such indexing may be useful in situations when a single partition is occupied by multiple cells, nuclei, or cell beads. In some instances, it may be beneficial to overload partitions such that a partition comprises more than a single cell, single nucleus, or single cell bead; for example, it may be useful in certain situations to overload partitions, e.g., to overcome Poisson loading statistics in partitions and/or to prevent reagent waste (e.g., from unoccupied partitions). Accordingly, such indexing may be useful in attributing cells, nuclei or nucleic acid molecules in multiply-occupied partitions to the originating cell, nucleus, cell bead, partition, sample, etc.
  • a barcoded molecule such as the barcoded molecules generated using the methods described herein (e.g., in FIGs. 7-9, FIGs. 25-28, as well as barcoded, probe-linked nucleic acid molecules described in U.S. Pat. Pub. No. 2020/0239874 and International Pub. No. WO 2019/165318, each of which is incorporated by reference herein) may be provided.
  • the barcoded molecule may comprise, as described herein, a sequence corresponding to the first target region, a sequence corresponding to the second target region, a sequence corresponding to the probe capture sequence (which may be disposed on the first probe or the second probe), and a sequence corresponding to the barcode sequence of the nucleic acid barcode molecule.
  • a barcode sequence may be specific to the partition and may differ from other barcode sequences of other partitions and thus may be used to identify a partition from which a nucleic acid molecule (or derivative thereof) originated.
  • some of the partitions may comprise a single cell, single nucleus, or single cell bead and thus the nucleic acid barcode molecule or barcode sequence may be used to identify a cell, nucleus, or cell bead from which a nucleic acid molecule (or derivative thereof) originated.
  • the barcoded molecule may be subjected to an additional barcoding operation, e.g., in partitions or in bulk.
  • the barcoded molecule may be re-partitioned in a partition among a plurality of partitions comprising a plurality of additional nucleic acid barcode molecules.
  • the plurality of additional nucleic acid barcode molecules may comprise additional barcode sequences that differ across the partitions.
  • the barcoded molecules may be subjected to conditions sufficient to barcode the barcoded molecules to generate a combinatorially barcoded molecule comprising two barcode sequences. As each barcode sequence pertains to a unique partition, the combination of barcodes may be useful in generating a greater diversity of barcoded molecules, as well as for identifying the originating partitions of the combinatorially barcoded molecule.
  • combinatorial assembly of barcode segments may be performed using, e.g., a split-pool approach.
  • the probe-linked nucleic acid molecules may be combinatorially barcoded using a split pool approach.
  • a plurality of permeabilized cells (or permeabilized nuclei or cell beads) comprising, e.g., probe- linked nucleic acid molecule, which may optionally be barcoded (e.g., the product following operation 709 of FIG. 7, 805 of FIG. 8, or 905 or 907 of FIG.
  • each partition of the plurality of partitions comprises a different (e.g., unique) barcode sequence segment.
  • the plurality of permeabilized cells or permeabilized nuclei or cell beads
  • the different barcode sequence segments delivered to the respective partitions containing the cells, nuclei, and/or cell beads.
  • cells can be collected from the plurality of partitions, pooled, and partitioned into an additional plurality of partitions (e.g., a plurality of wells) wherein each partition of the additional plurality of partitions comprises a different (e.g., unique) second barcode sequence segment.
  • additional plurality of partitions e.g., a plurality of wells
  • each partition of the additional plurality of partitions comprises a different (e.g., unique) second barcode sequence segment.
  • Combinatorial barcoding as described herein may comprise at least 1, 2, 3, 4, 5, 6, 7, 8 or more operations (e.g., split-pool cycles).
  • Combinatorial barcoding comprising multiple operations may be useful, for example, in generation of greater barcode diversity and to synthesize a unique barcode sequence on nucleic acid molecules derived from each single cell, nucleus, or cell bead of a plurality of cells, nuclei, cell beads.
  • combinatorial barcoding comprising three operations, each comprising attachment of a unique nucleic acid sequence in each of 96 partitions, will yield up to 884,736 unique barcode combinations.
  • up to M N unique barcode combinations may be generated.
  • Cells or nuclei or cell beads may be partitioned such that at least one cell (or nuclei or cell bead) is present in each partition of a plurality of partitions.
  • Cells, nuclei, or cell beads may be partitioned such that at least 1; 2; 3; 4; 5; 10; 20; 50; 100; 500; 1,000; 5,000; 10,000; 100,000; 1,000,000; or more cells, nuclei, or cell beads are present in a single partition.
  • Cells, nuclei, or cell beads may be partitioned such that at most 1,000,000; 100,000; 10,000; 5,000; 1,000; 500; 100; 50; 20; 10; 5; 4; 3; 2; or 1 cell (or nucleus or cell bead) is present in a single partition.
  • Cells, nuclei, and /or cell beads may be partitioned in a random configuration.
  • the additional barcoding operations may be performed prior to some of the operations described herein.
  • it may be beneficial to combinatorially barcode the first probe in a bulk solution e.g., prior to or following generation of the extended probe molecule or probe-linked molecule.
  • the nucleic acid molecule may be contacted, e g., in bulk, with a first probe to generate a probe-associated molecule.
  • the probe- associated molecule may optionally be extended, e.g., using the methods described herein, to generate an extended probe molecule.
  • the probe-associated molecule or the extended probe molecule may then be subjected to combinatorial barcoding, e.g., in partitions, as described above, to generate a combinatorially barcoded molecule.
  • the combinatorially barcoded molecule may then be partitioned with a second probe and a nucleic acid barcode molecule, which, as described herein, may attach to either the first probe (or combinatorially barcoded probe), the second probe, or both probes.
  • a plurality of the combinatorially barcoded molecules may be traced back to the individual partitions from which they originated.
  • the combinatorial barcoding may be useful in generating greater probe diversity.
  • the combinatorial barcoding of the first probe may be particularly useful when combined with the second probe and nucleic acid barcode molecule, which may comprise a barcode sequence that is specific to the partition.
  • the presence of the probespecific barcode(s) and the partition-specific barcode sequence may allow for indexing of individual cells (or nuclei or cell beads) within a partition.
  • partitions comprising cell/nucleus/cell bead multiplets e.g., cell doublets, triplets, etc.
  • cells, nuclei, or cell beads may be “overloaded” into partitions using conditions such that a higher probability of cell/nucleus/cell bead multiplets (2, 3, 4, 5+ cells, nuclei, or cell beads per partition) are formed, wherein target libraries of these cell multiplets may be computationally deconvolved into single cells, nuclei, or cell beads.
  • FIG. 10 schematically shows an example workflow of barcoding nucleic acid molecules in partitions comprising cell/nucleus/cell bead multiplets.
  • one or more populations of cells/nuclei/cell beads may be subjected to barcoding, as described herein (e.g., using processes shown and described in FIGs. 7-9 and FIGs. 15-16).
  • a first population of cells (or nuclei or cell beads) 1002 (comprising a first plurality of nucleic acid molecules) may be subjected to barcoding in a first subset of a first plurality of partitions, generating a first plurality of barcoded nucleic acid molecules comprising a first barcode sequence.
  • a second population of cells (or nuclei or cell beads) 1004 may be barcoded in a second subset of the first plurality of partitions, generating a second plurality of barcoded nucleic acid molecules comprising a second barcode sequence.
  • the first barcode sequence may be different than the second barcode sequence.
  • the first population of cells (or nuclei or cell beads) 1002 may be pooled together with the second population of cells (or nuclei or cell beads) 1004 to generate a mixture of cells.
  • the mixture of cells (or nuclei or cell beads) may be partitioned into a second plurality of partitions. In some instances, the mixture of cells/nuclei/cell beads may be partitioned into the second plurality of partitions such that some partitions of the second plurality of partitions comprises more than one cell (e g., a cell multiplet partition).
  • a partition 1035 of the second plurality of partitions may comprise a cell, nucleus, or cell bead (“Cell A”) from the first population of cells 1002 and a cell, nucleus, or cell bead (“Cell B”) from the second population of cells 1004.
  • the partition 1035 may comprise an additional barcode sequence, which may be unique to the partition.
  • the cells/nuclei/cell beads in each partition may be subjected to an additional barcoding operation to append the additional barcode sequence on the barcoded nucleic acid molecules.
  • the barcoded nucleic acid molecules may be deconvolved, using the different barcode sequences (e.g., the first barcode sequence, the second barcode sequence, and the additional barcode sequences), to identify the originating cell/nucleus/cell bead.
  • the barcoded nucleic acid molecule comprising the additional barcode sequence from partition 1035 and the first barcode sequence from the first population of cells (or nuclei or cell beads) 1002 may be used to identify that barcoded nucleic acid molecule as originating from Cell A.
  • a barcoded nucleic acid molecule comprising the additional barcode sequence from partition 1035 and the second barcode sequence from the second populations of cells (or nuclei or cell beads) 1004 may be used to identify that barcoded nucleic acid molecule as originating from Cell B.
  • the contents of the partitions may be pooled and the barcoded molecules (e.g., barcoded probe-linked nucleic acid molecules) may be duplicated or amplified by, for example, one or more amplification reactions, which may in some instances be isothermal.
  • the amplification reactions may comprise polymerase chain reactions (PCR) and may involve the use of one or more primers or polymerases.
  • the one or more primers may comprise one or more functional sequences (e g., a primer sequence/primer binding sequence, a sequencing primer sequence (e g., R1 or R2), a partial sequencing primer sequence (e.g., partial R1 or partial R2), a sequence configured to attach to the flow cell of a sequencer (e.g., P5 or P7, or partial sequences thereof), etc.) and may facilitate addition of said one or more functional sequences to the extended nucleic acid molecule.
  • the barcoded molecules, or derivatives thereof may be detected via nucleic acid sequencing (e.g., as described herein).
  • systems useful for barcoding nucleic acid molecules may comprise any of the components described herein, e.g., a plurality of partitions (e.g., droplets, wells), which may be provided in any useful format, e.g., a microfluidic device, a multi-well array or plate, etc.
  • the systems may include nucleic acid barcode molecules, optionally coupled to supports (e g., particles, beads, gel beads, etc ).
  • the systems may comprise any of the probes described herein, such as a first probe or plurality of first probes, a second probe or plurality of second probes, and any useful reaction components (e.g., for performing a nucleic acid reaction, e.g., extension, ligation, amplification, etc.).
  • useful reaction components can include, in non-limiting examples, enzymes (e.g., ligases, polymerases, reverse transciptases, restriction enzymes, etc.), nucleotides bases, etc.
  • compositions useful for systems and methods for barcoding nucleic acid molecules may comprise any of the probes described herein.
  • a composition may comprise a plurality of first probes, a plurality of second probes, and/or a plurality of first probes and a plurality of second probes.
  • a probe or a set of probes may be designed to target a specific sequence or a set of specific sequences. Such probes may be designed to have the same or different sequences within different partitions.
  • a first composition may comprise a first probe and a second probe designed to target two regions of a first gene
  • a second composition may comprise a first probe and a second probe designed to target two regions of a second gene, which second gene is different than the first gene.
  • a composition may comprise nucleic acid barcode molecules, and/or probe binding molecules, which may optionally be provided coupled to a support (e.g., particle, bead).
  • a composition may be a part of or comprise a reaction mixture, which can include reaction components or reagents, e.g., enzymes, nucleotide bases, catalysts, buffers etc.
  • the present disclosure provides methods for performing multiplexed assays.
  • a multiplexed assay may comprise assaying or analyzing one or more biomolecules (e g., nucleic acid molecules, proteins, lipids, carbohydrates, etc.).
  • a method may comprise using one or more probes and a nucleic acid barcode molecule to barcode a nucleic acid molecule of a cell/nucleus/cell bead, thereby generating a first barcoded nucleic acid molecule; attaching or coupling a feature-binding group to a feature of the cell/nucleus/cell bead, wherein the feature-binding group comprises a reporter oligonucleotide comprising a reporter sequence that identifies the feature-binding group; using an additional nucleic acid barcode molecule, and optionally, an additional probe, to barcode the reporter sequence to generate a second barcoded nucleic acid molecule; and optionally barcoding the first barcoded nucleic acid molecule and the second barcoded nucleic acid molecule to generate a third barcoded nucleic acid molecule and a fourth barcoded nucleic acid molecule.
  • One or more operations may be performed within a partition (e.g., droplet or well).
  • the methods described herein may facilitate profiling of one or more biomolecules with single-cell/single nucleus/single cell bead resolution, using, for example, probe hybridization, feature binding groups (e.g., antibodies, antibody fragments, epitope-binding groups, etc.), barcoding, amplification, and sequencing.
  • the methods may be useful in providing genomic, transcriptomic, proteomic, exomic, or other “-omic” information from a single cell/nucleus/cell bead.
  • the methods may be used to analyze a predetermined panel of target genes and a pre-determined panel of target features (e.g., proteins, peptides, or other biomolecules) in a sensitive and accurate manner.
  • the methods may be used to analyze whole genomic, whole transcriptomic, whole exomic, etc. characteristics of a cell.
  • the methods comprise contacting a cell/nucleus/cell bead with a first probe, a second probe, and a third probe under conditions sufficient to generate a first probe- associated molecule and a second probe-associated molecule.
  • the cell/nucleus/cell bead may comprise (i) a nucleic acid molecule (e.g., a target nucleic acid molecule such as RNA or DNA) comprising a first target region and a second target region and (ii) a feature (e.g., protein, peptide, or other biomolecule) coupled to a feature-binding group.
  • the feature binding group may comprise or be coupled to (i) a reporter oligonucleotide comprising a reporter sequence, which may be associated with the feature or may be used to identify the feature, and (ii) a feature probe-binding sequence.
  • the first probe may comprise a first probe sequence complementary to the first target region of the nucleic acid molecule and, optionally, an additional probe sequence, such as a probe capture sequence or other functional sequence.
  • the second probe may comprise a second probe sequence complementary to the second target region and, optionally, a probe capture sequence or functional sequence.
  • the third probe may comprise (i) a third probe sequence complementary to the feature probe-binding sequence and (ii) a probe capture sequence or functional sequence, which may be the same sequence as the probe capture sequence of the first probe and/or second probe.
  • the first probe-associated molecule may comprise the nucleic acid molecule, the first probe, the second probe, or combinations or complements thereof.
  • the second probe-associated molecule may comprise the reporter oligonucleotide (which comprises the reporter sequence) and the third probe, or complements thereof.
  • the method comprises providing the first probe-associated molecule and the second probe-associated molecule, and barcoding the first probe-associated molecule and the second probe-associated molecules.
  • Such barcoding operations may occur in a first set of partitions (e.g., droplets or wells).
  • Such an example method may comprise contacting the first probe-associated molecule and the second-probe-associated molecule with probe binding molecules (e.g., a splint oligonucleotide) and barcode molecules (e.g., nucleic acid barcode molecules) under conditions sufficient to generate a first barcoded nucleic acid molecule and a second barcoded nucleic acid molecule.
  • probe binding molecules e.g., a splint oligonucleotide
  • barcode molecules e.g., nucleic acid barcode molecules
  • the barcode molecules may comprise (i) a barcode capture sequence, e.g., a common sequence that is common to a plurality of barcode molecules and (ii) a first barcode sequence.
  • the first barcode sequence may be unique to a first partition of a first set of partitions, and the barcode molecules within the first partition may share the same first barcode sequence.
  • the probe-binding molecule may comprise (i) a probe-binding sequence complementary to the probe capture sequence (of the first probe, the second probe, and/or the third probe) and (ii) a barcode binding sequence complementary to the barcode capture sequence (e.g., common sequence) of the plurality of barcode molecules.
  • barcoding of the first probe-associated molecule and the second probe-associated molecule may comprise hybridization of the probe binding molecule to (i) the probe capture sequence (or complement thereof) of the first probe, the second probe, and/or the third probe, and (ii) the barcode capture sequence (or common sequence) of the nucleic acid barcode molecule.
  • the first barcoded nucleic acid molecule comprises a sequence corresponding to the first probe sequence, a sequence corresponding to the second probe sequence, and a sequence corresponding to the first barcode sequence.
  • the second barcoded nucleic acid molecule may comprise a sequence corresponding to the reporter sequence, a sequence corresponding to the third probe sequence, and a sequence corresponding to the first barcode sequence.
  • the method may further comprise providing a second set of partitions, and in a second partition of the second set of partitions, (i) contacting the first barcoded nucleic acid molecule, or derivative thereof (e.g., complements, amplicons, extension products thereof), to a first capture molecule of a plurality of capture molecules under conditions sufficient to generate a third barcoded nucleic acid molecule, and (ii) contacting the second barcoded nucleic acid molecule, or derivative thereof, to a second capture molecule of the plurality of capture molecules under conditions sufficient to generate a fourth barcoded nucleic acid molecule.
  • the first barcoded nucleic acid molecule, or derivative thereof e.g., complements, amplicons, extension products thereof
  • the plurality of capture molecules may each comprise a second barcode sequence, which may be the same or different than the first barcode sequence from the first set of partitions.
  • the second barcode sequence may be unique to the partition (e.g., differ across partitions).
  • the third barcoded nucleic acid molecule and the fourth barcoded molecule may each comprise a sequence corresponding to the first barcode sequence and a sequence corresponding to the second barcode sequence.
  • the third barcoded nucleic acid molecule may comprise a sequence corresponding to the first target region, a sequence corresponding to the second target region, a sequence corresponding to a probe capture sequence, the first barcode sequence and the second barcode sequence.
  • the fourth barcoded nucleic acid molecule may comprise a sequence corresponding to the reporter sequence, a sequence corresponding to the feature probe binding sequence, a sequence corresponding to the third probe, the first barcode sequence and the second barcode sequence.
  • the feature binding group may comprise a labelling agent, as described elsewhere herein. Accordingly, the feature binding group may comprise, in some examples, an antibody or antibody fragment, an epitope binding moiety, a protein, a peptide, a lipophilic moiety (such as cholesterol), a cell surface receptor binding molecule, a receptor ligand, a small molecule, a bispecific antibody, a bi-specific T-cell engager, a T-cell receptor engager, a B-cell receptor engager, a pro-body, an aptamer, a monobody, an affimer, a darpin, and a protein scaffold, or any combination thereof.
  • an antibody or antibody fragment an epitope binding moiety, a protein, a peptide, a lipophilic moiety (such as cholesterol), a cell surface receptor binding molecule, a receptor ligand, a small molecule, a bispecific antibody, a bi-specific T-cell engager, a T-cell receptor engager, a B-cell
  • the probe capture sequence of the first probe may be common to a plurality of first probes (or second probes), a plurality of partitions, and/or a plurality of cells/nuclei/cell beads.
  • the first set of partitions may comprise one or more additional partitions that comprise additional probe-associated nucleic acid molecules.
  • the additional probe-associated nucleic acid molecules may comprise identical sequences (e.g., first probe sequence, second probe sequence) to the probe-associated nucleic acid molecule of the first partition, or the additional probe-associated nucleic acid molecules of the additional partitions may comprise different sequences (e.g., different probe sequences) than the probe- associated nucleic acid molecule of the first partition.
  • each of the one or more additional probe-associated nucleic acid molecules comprises a probe capture sequence, which may be identical or different across the first set of partitions.
  • the probe-associated molecules may be a probe-linked molecule.
  • the probe-associated molecules may be the probe-associated molecules or barcoded molecules described herein (e.g., in FIGS. 7-9), or a probe-linked molecule, such as those described in U.S. Pat. Pub. No. 2020/0239874 and International Pub. No. WO 2019/165318, each of which is incorporated by reference herein in its entirety.
  • two sets of probe-associated molecules may be generated, in which: (i) a first probe-associated molecule comprises the nucleic acid molecule, with the first probe and the second probe hybridized thereto (e.g., via hybridization of the first probe sequence to the first target region and the second probe sequence to the second target region) and (ii) a second probe-associated molecule comprises the reporter oligonucleotide (which comprises the reporter sequence), with the third probe hybridized thereto.
  • the first probe, the second probe, and/or the third probe may comprise a probe capture sequence.
  • the probe capture sequence on the first probe may be the same or different than the probe capture sequence of the second probe or the third probe.
  • the probe capture sequence of the second probe may be the same or different than the probe capture sequence of the third probe. Accordingly, the barcoding operations described herein may occur on the first probe, the second probe, the third probe, or any combination thereof.
  • a first barcode molecule comprising the first barcode sequence (“BC1”) may hybridize (e.g., directly or via a probe-binding molecule) to the first probe to generate a first barcoded nucleic acid molecule, and subsequently, a capture molecule comprising a second barcode sequence (“BC2”) may be annealed to a region of the first barcode molecule, thereby generating a molecule comprising a sequence, or complementary sequences, of BC2-BCl-probe 1-probe 2.
  • the first barcode molecule comprising the first barcode sequence (“BC1”) may hybridize (e.g., directly or via a probe-binding molecule) to the second probe to generate a first barcoded nucleic acid molecule, and subsequently, a capture molecule comprising the second barcode sequence (“BC2”) may be annealed to a region of the first barcode molecule, thereby generating a molecule comprising a sequence of probe 1-probe 2-BC1-BC2.
  • the barcode molecules and the capture molecules may be annealed to different probes.
  • the first barcode molecule comprising the first barcode sequence (“BC1”) may hybridize (e.g., directly or via a probe-binding molecule) to the first probe to generate a first barcoded nucleic acid molecule, and subsequently, a capture molecule comprising the second barcode sequence (“BC2”) may be annealed to the second probe, thereby generating a molecule comprising a sequence of BC1- probe 1-probe 2-BC2.
  • the first barcode molecule comprising the first barcode sequence (“BC1”) may hybridize (e.g., directly or via a probe-binding molecule) to the second probe to generate a first barcoded nucleic acid molecule, and subsequently, a capture molecule comprising the second barcode sequence (“BC2”) may be annealed to the first probe, thereby generating a molecule comprising a sequence of BC2-probe 1-probe 2-BC1.
  • a capture molecule comprising the second barcode sequence (“BC2”) may be annealed to the first probe, thereby generating a molecule comprising a sequence of BC2-probe 1-probe 2-BC1.
  • the barcode molecules may comprise a capture-binding sequence complementary to a capture sequence of the plurality of capture molecules.
  • the first probe may comprise a probe capture sequence which may hybridize to a probe binding molecule, which may mediate hybridization of the barcode molecule (e.g., via hybridization of the barcode binding sequence of the probe binding molecule to the barcode capture sequence (e.g., common sequence) of the barcode molecule).
  • the barcode molecule may additionally comprise the capture-binding sequence, which may allow for hybridization of the capture sequence of the capture molecules to the barcode molecule.
  • FIG. 15 schematically illustrates an example barcoded nucleic acid molecule as described herein.
  • a nucleic acid molecule e.g., RNA molecule
  • the nucleic acid molecule 1500 may be contacted with a first probe 1506 comprising a first probe sequence 1508 and, optionally, a first probe capture sequence 1510.
  • the first probe sequence 1508 may be complementary to the first target region 1502.
  • the first probe capture sequence 1510 may additionally, in some instances, comprise a functional sequence, such as a primer sequence, a partial primer sequence, a barcode sequence, a sequencing primer sequence, etc.
  • the nucleic acid molecule 1500 may also be contacted with a second probe 1516 comprising a second probe sequence 1514 and, optionally, a second probe capture sequence 1518.
  • the second probe sequence 1514 may be complementary to the second target region 1504.
  • the second probe capture sequence 1518 may additionally comprise a functional sequence. Hybridization of the first probe 1506 and the second probe 1516 to the nucleic acid molecule 1500 may generate a probe-associated molecule.
  • the probe-associated molecule may be subjected to one or more barcoding operations.
  • a barcoding operation may occur in one or more partitions (e.g., a first set of partitions) and may include hybridizing a probe binding molecule 1517 and a barcode molecule 1519 comprising a barcode capture sequence (e.g., a common sequence), to the probe- associated molecule.
  • the probe binding molecule 1517 and the barcode molecule 1519 may be provided as a pre-annealed complex, or they may be provided as separate molecules.
  • the barcode capture sequence (e.g., common sequence) may be a sequence that is common to the plurality of barcode molecules in the first set of partitions, or the common sequence may be unique to the barcode molecules in only a single first partition (e.g., the common sequence differs across partitions of the first set of partitions).
  • the probe binding molecule 1517 may comprise a probe binding sequence complementary to the probe capture sequence 1518 of the second probe 1516, as well as a barcode binding sequence complementary to a sequence of the barcode molecule 1519.
  • the probe-associated molecule may be subjected to conditions sufficient to generate a first barcoded nucleic acid molecule, which can include annealing of the probe-binding molecule 1517 to (i) the probe capture sequence 1518 and (ii) the barcode capture sequence (e.g., common sequence) of the barcode molecule 1519.
  • the barcoding process may comprise additional operations, such as ligation, which may be performed chemically or enzymatically, as described elsewhere herein.
  • the first barcoded nucleic acid molecule or derivatives thereof may be subjected to a second barcoding operation.
  • a second barcoding operation may occur in a second set of partitions.
  • the first barcoded nucleic acid molecule may be removed from the first set of partitions, pooled (e.g., with other barcoded nucleic acid molecules from other first partitions of the first set of partitions), and partitioned in a second partition of a second set of partitions.
  • the second partition may comprise a capture molecule 1520.
  • the capture molecule 1520 may comprise a second barcode sequence and a sequence complementary to the probe capture sequence 1510 of the first probe 1506.
  • the second barcode sequence may be a sequence that is common to the plurality of capture molecules in the second set of partitions, or the barcode sequence may be unique to the capture molecules in only the second partition (e.g., differ across partitions).
  • the capture molecule 1520 may hybridize to the probe capture sequence 1510 to generate an additional barcoded molecule (also referred to herein as a “third barcoded nucleic acid molecule”).
  • the additional barcoded molecule may comprise a sequence corresponding to the first barcode sequence (of the barcode molecule 1519), and a sequence corresponding to the second barcode sequence (of the capture molecule 1520).
  • Panel B of FIG. 15 schematically illustrates another example barcoded molecule in which the capture molecule 1520 is hybridized to the barcode molecule 1519.
  • the nucleic acid molecule (e.g., RNA molecule) 1500 comprising a first target region 1502 and a second target region 1504 may be provided.
  • the nucleic acid molecule 1500 may be contacted with a first probe 1506 comprising a first probe sequence 1508 and a probe capture sequence 1510.
  • the first probe sequence 1508 may be complementary to the first target region 1502.
  • the probe capture sequence 1510 may additionally comprise a functional sequence, such as a primer sequence, a partial primer sequence, a barcode sequence, a sequencing primer sequence, etc.
  • the nucleic acid molecule 1500 may also be contacted with a second probe 1516 comprising a second probe sequence 1514 and, optionally, an additional sequence 1518.
  • the second probe sequence 1514 may be complementary to the second target region 1504.
  • the additional sequence 1518 may comprise, for instance, a probe capture sequence, or a functional sequence (e.g., primer, primer binding site, sequencing primer sequence, etc.). Hybridization of the first probe 1506 and the second probe 1516 to the nucleic acid molecule 1500 may generate a probe-associated molecule.
  • the probe-associated molecule may be contacted with one or more barcode molecules. Such barcoding operations, as described herein, may occur in a plurality of partitions (e g., a first partition of a first set of partitions and/or a second partition of a second set of partitions).
  • the probe-associated molecule may be contacted with a probe binding molecule 1517 and a barcode molecule 1519, which may comprise a first barcode capture sequence (e.g., a common sequence) and a second barcode capture sequence 1521 (also referred to herein as “capture binding sequence”).
  • the probe binding molecule 1517 and the barcode molecule 1519 may be provided as a pre-annealed complex or as separate molecules.
  • the first barcode capture sequence (e.g., common sequence) may be a sequence that is common to the plurality of barcode molecules in the first set of partitions, or the common sequence may be unique to the barcode molecules in only the first partition (e.g., differ across partitions).
  • the probe binding molecule 1517 may comprise a probe binding sequence complementary to the probe capture sequence 1510 as well as a barcode binding sequence complementary to the first barcode capture sequence (e.g., common sequence) of the barcode molecule 1519.
  • the probe- associated molecule may be subjected to conditions sufficient to generate a first barcoded nucleic acid molecule, which can include annealing of the probe-binding molecule 1517 to (i) the probe capture sequence 1510 and (ii) the first barcode capture sequence (e g., common sequence) of the barcode molecule 1519.
  • the barcoding process may comprise additional operations, such as ligation, which may be performed chemically or enzymatically, as described elsewhere herein.
  • the first barcoded nucleic acid molecule or derivatives thereof may be subjected to a second barcoding operation.
  • a second barcoding operation may occur in a second set of partitions.
  • the first barcoded nucleic acid molecule may be removed from the first partition and partitioned in a second partition of a second set of partitions (e g., droplets).
  • the second partition may comprise a capture molecule 1520.
  • the capture molecule 1520 may comprise a second barcode sequence and a sequence complementary to the second barcode capture sequence 1521 of the barcode molecule 1519.
  • the second barcode sequence may be a sequence that is common to the plurality of capture molecules in the second set of partitions, or the barcode sequence may be unique to the capture molecules in only the second partition (e.g., differ across partitions).
  • the capture molecule may hybridize to the second barcode capture sequence 1521 to generate an additional barcoded molecule (also referred to herein as a “third barcoded nucleic acid molecule”).
  • the additional barcoded molecule may comprise a sequence corresponding to the first barcode sequence (of the barcode molecule 1519), and a sequence corresponding to the second barcode sequence (of the capture molecule 1520).
  • Panel C of FIG. 15 illustrates another example barcoded nucleic acid molecule.
  • a nucleic acid molecule (e.g., RNA molecule) 1500 comprising a first target region 1502 and a second target region 1504 may be provided.
  • the nucleic acid molecule 1500 may be contacted with a first probe 1506 comprising a first probe sequence 1508 and, optionally, a first probe capture sequence 1510.
  • the first probe sequence 1508 may be complementary to the first target region 1502.
  • the first probe or first probe capture sequence 1510 may additionally, in some instances, comprise a functional sequence, such as a primer sequence, a partial primer sequence, a barcode sequence, a sequencing primer sequence, etc.
  • the nucleic acid molecule 1500 may also be contacted with a second probe 1516 comprising a second probe sequence 1514 and, optionally, a second probe capture sequence 1518.
  • the second probe sequence 1514 may be complementary to the second target region 1504.
  • the second probe capture sequence 1518 may additionally comprise a functional sequence. Hybridization of the first probe 1506 and the second probe 1516 to the nucleic acid molecule 1500 may generate a probe-associated molecule or complex.
  • the probe-associated molecule may be subjected to one or more barcoding operations.
  • a barcoding operation may occur in one or more partitions (e.g., a first set of partitions) and may include hybridizing a probe binding molecule 1517 and a barcode molecule 1519 comprising a barcode capture sequence (e.g., a common sequence), to the probe- associated molecule or complex.
  • the probe binding molecule 1517 and the barcode molecule 1519 are provided as a pre-annealed complex (e.g., a partially double-stranded molecule comprising the probe binding molecule 1517 and the barcode molecule 1519), or they may be provided as separate molecules, which may separately anneal to the probe-associated molecule or complex (e.g., the probe binding molecule 1517 may hybridize to the probe- associated molecule or complex, e.g., via the second probe capture sequence 1518, and the barcode molecule 1519 may hybridize to the probe binding molecule 1517).
  • a pre-annealed complex e.g., a partially double-stranded molecule comprising the probe binding molecule 1517 and the barcode molecule 1519
  • the probe binding molecule 1517 may hybridize to the probe- associated molecule or complex, e.g., via the second probe capture sequence 1518, and the barcode molecule 1519 may hybridize to the probe binding molecule 1517.
  • the barcode capture sequence (e.g., common sequence) may be a sequence that is common to the plurality of barcode molecules in the first set of partitions, or the common sequence may be unique to the barcode molecules in only a single first partition (e.g., the common sequence differs across partitions of the first set of partitions).
  • the probe binding molecule 1517 may comprise a probe binding sequence complementary to the probe capture sequence 1518 of the second probe 1516, as well as a barcode binding sequence complementary to a sequence of the barcode molecule 1519.
  • the probe binding molecule 1517 and/or the barcode molecule 1519 comprise an additional sequence, e.g., an adapter sequence, a primer sequence (e.g., sequencing primer sequence or partial sequencing primer sequence), a UMI, a sample index sequence, etc.
  • the probe binding molecule 1517 comprises the entire sequence of the barcode molecule 1519, such that no overhang remains.
  • the probe binding molecule 1517 and barcode molecule 1519 comprise a sample index sequence, which may be useful in identifying the partition, cell, nucleus, or cell bead from which the target nucleic acid molecule 1500 originates.
  • the probe-associated molecule may be subjected to conditions sufficient to generate a first barcoded nucleic acid molecule, which can include annealing of the probebinding molecule 1517 to (i) the probe capture sequence 1518 and (ii) the barcode capture sequence (e.g., common sequence) of the barcode molecule 1519.
  • the barcoding process may comprise additional operations, such as ligation (e.g., ligation of the barcode molecule 1519 to the probe capture sequence 1518), which may be performed chemically or enzymatically, as described elsewhere herein.
  • the first barcoded nucleic acid molecule or derivatives thereof may be subjected to a second barcoding operation.
  • a second barcoding operation may occur in a second set of partitions.
  • the first barcoded nucleic acid molecule may be removed from the first set of partitions, pooled (e.g., with other barcoded nucleic acid molecules from other first partitions of the first set of partitions), and partitioned in a second partition of a second set of partitions.
  • the second partition may comprise a capture molecule 1520.
  • the capture molecule 1520 may comprise a second barcode sequence and a sequence complementary to the probe capture sequence 1510 of the first probe 1506 (and/or the second probe 1516).
  • the second barcode sequence may be a sequence that is common to the plurality of capture molecules in the second set of partitions, or the barcode sequence may be unique to the capture molecules in only the second partition (e.g., differ across partitions).
  • the capture molecule 1520 may hybridize to the probe capture sequence 1510 to generate an additional barcoded molecule (also referred to herein as a “third barcoded nucleic acid molecule”).
  • the additional barcoded molecule may comprise a sequence corresponding to the first barcode sequence (of the barcode molecule 1519), and a sequence corresponding to the second barcode sequence (of the capture molecule 1520).
  • the present disclosure provides for methods of multiplexed analysis, e.g., processing of additional biomolecule types, such as proteins and peptides.
  • the method may comprise providing a feature-binding group (e.g., antibody, protein, binding moiety, etc.), which may couple to or bind to a feature (e.g., protein, peptide) of a cell, nucleus or cell bead.
  • a feature-binding group e.g., antibody, protein, binding moiety, etc.
  • Such a method may comprise providing a cell, nucleus or cell bead having a feature of interest (e g., protein) and contacting the cell, nucleus or cell bead with the feature-binding group.
  • the feature-binding group may couple to the feature of interest
  • the feature-binding group may comprise a reporter oligonucleotide comprising a reporter sequence coupled thereto, which may be specific for a particular feature and thus be used to identify the feature.
  • the feature-binding group may be an antibody and the reporter oligonucleotide may comprise a reporter sequence that identifies the antigen or binding moiety (e.g., epitope, epitope fragment) to which the antibody couples or binds.
  • the feature binding group may comprise a feature probe binding sequence, which may be used for downstream probe-binding and/or barcoding. Following the contacting of the cell (nucleus or cell bead) with the feature binding group, the cell/nucleus/cell bead may comprise the feature coupled to the feature binding group.
  • the methods described herein may additionally comprise: providing a cell, nucleus or cell bead comprising (i) the nucleic acid molecule comprising the first target region and the second target region and (ii) the feature coupled to the feature binding group and contacting the cell, nucleus or cell bead with a plurality of probes.
  • the cell/nucleus/cell bead may be contacted (e.g., in a first partition) with a first probe, a second probe, and a third probe.
  • the first probe and the second probe may associate with the first target region and the second target region of the nucleic acid molecule, thereby generating a first probe-associated molecule.
  • the third probe may associate with (e g., via hybridization) with the feature binding group, thereby generating a second probe-associated molecule.
  • the third probe may comprise a third probe sequence that is complementary to the feature probe binding sequence, and in some instances, the third probe may additionally comprise a probe capture sequence.
  • the first probe and/or the second comprise may also comprise a probe capture sequence, which may be the same or different than the probe capture sequence of the third probe.
  • the first probe-associated molecule e.g., the nucleic acid molecule with the first probe and the second probe associated therewith
  • the second-probe- associated molecule e.g., the feature binding group with the third probe associated therewith
  • Such a barcoding operation may comprise, for example, providing barcode molecules comprising a first barcode sequence and a barcode-capture sequence such as a common sequence, which may hybridize directly with the first probe-associated molecule and the second probe-associated molecule, e.g., via the probe capture sequences.
  • the barcode molecules may be provided with probe-binding molecules which comprise (i) a probe binding sequence complementary to the probe capture sequence of the first probe, the second probe, and/or the third probe and (ii) a barcode binding sequence, which may be complementary to the common sequence of the barcode molecules.
  • probe binding molecules and the barcode molecules may be provided as a pre-annealed complex.
  • Barcoding of the first probe-associated molecule and the second probe-associated molecule may include hybridization of the barcode molecules (e.g., the barcode capture sequence such as a common sequence) to a portion (e.g., the probe capture sequence) of the first probe-associated molecule and the second probe-associated molecule, or the barcoding may include hybridization of the barcode molecules to the probe binding molecule and hybridization of the probe binding molecule to the first probe-associated molecule or the second probe-associated molecule. Additional operations such as ligation (e.g., enzymatic or chemical ligation) may be performed to generate the first barcoded molecule and the second barcoded molecule.
  • ligation e.g., enzymatic or chemical ligation
  • the first barcoded molecule and the second barcoded molecule may be subjected to additional barcoding operations, e.g., in a second set of partitions.
  • additional barcoding operations may include: contacting the first barcoded nucleic acid molecule or derivative thereof to a first capture molecule of a plurality of capture molecules to generate a third barcoded nucleic acid molecule and contacting the second barcoded nucleic acid molecule or derivative thereof to a second capture molecule of the plurality of capture molecules to generate a fourth barcoded nucleic acid molecule.
  • the capture molecules within a partition may each comprise a second barcode sequence, which may be unique to the partition (e g., differ across partitions). Accordingly, both the third barcoded nucleic acid molecule and the fourth barcoded nucleic acid molecule may comprise a first barcode sequence (or complement thereof) and a second barcode sequence (or complement thereof).
  • FIG. 16A schematically illustrates an example workflow for barcoding multiple analytes of a cell, nucleus or cell bead.
  • the cell, nucleus or cell bead 1600 may comprise a nucleic acid molecule (e.g., RNA molecule or other target nucleic acid molecule) 1601 comprising a first target region 1602 and a second target region 1604.
  • the cell, nucleus or cell bead may additionally comprise a feature (e.g., a protein, such as a cell surface receptor (or nuclear membrane protein) or an intracellular/intranuclear protein) 1650.
  • a feature e.g., a protein, such as a cell surface receptor (or nuclear membrane protein) or an intracellular/intranuclear protein
  • the cell, nucleus or cell bead 1600 may be processed, e.g., fixed, permeabilized, treated with a treatment, etc.
  • processing may include providing one or more feature binding groups (e.g., antibodies, antibody fragments, etc.) 1652, which may couple to the feature 1650.
  • the feature binding group 1652 may comprise or be coupled to a reporter oligonucleotide 1657, which may comprise a reporter sequence 1654.
  • the reporter sequence 1654 may be indicative of the feature binding group 1652 or feature 1650.
  • the reporter sequence 1654 may be pre-indexed or assigned to a particular antibody or other feature binding group, such that presence of the reporter sequence 1654 indicates presence of the particular feature 1650 in a sample.
  • the feature binding group 1652 or the reporter oligonucleotide 1657 may also comprise or be coupled to feature probe binding sequence 1656.
  • the cell, nucleus or cell bead 1600 may be contacted with the feature binding group 1652 and fixed, e.g., either in addition to or alternatively to a fixation and permeabilization operation before the contacting.
  • a permeabilized (and optionally fixed) cell (or nucleus) may be contacted with (i) one or more feature binding groups (or labeling agents) that are configured to couple to intracellular proteins (or intranuclear proteins) and/or (ii) one or more feature binding groups (or labeling agents) that are configured to couple to cell membrane proteins (or nuclear membrane proteins).
  • permeabilization may involve partially or completely dissolving or disrupting a cell membrane (or nuclear membrane) or a portion thereof.
  • Permeabilization may be achieved by, for example, contacting a cell membrane (or a nuclear membrane) with an organic solvent (e.g., methanol) or a detergent such as Triton X-100 or NP-40.
  • an organic solvent e.g., methanol
  • a detergent such as Triton X-100 or NP-40.
  • the cell, nucleus, or cell bead may be fixed, as described elsewhere herein.
  • a second feature binding group (or labeling agent) similar to 1652 (not shown) can be used to couple to an intracellular feature, such as an intracellular protein, and comprise or be coupled to a second reporter oligonucleotide, which may comprise a second reporter sequence.
  • the second reporter sequence may be indicative of the second feature binding group or the intracellular feature.
  • the second reporter sequence may be pre-indexed or assigned to a particular antibody or other feature binding group, such that presence of the second reporter sequence indicates presence of the particular intracellular feature in a sample.
  • the second feature binding group or the second reporter oligonucleotide may also comprise or be coupled to a second feature probe binding sequence, similar to that of 1656.
  • the cell, nucleus or cell bead 1600 may be contacted with a first probe 1606, a second probe 1616, and a third probe 1658, under conditions sufficient to generate a first probe- associated molecule (or probe-associated complex) 1630 and a second probe-associated molecule (or probe-associated complex) 1665.
  • the first probe-associated molecule 1630 may be or comprise a probe-linked molecule, as described elsewhere herein.
  • the first probe-associated molecule 1630 (or probe-linked molecule) may be any of the probe-associated molecules or probe-linked molecules described herein (e.g., generated from an extended probe, a barcoded extended probe, etc.).
  • the first probe 1606 may comprise a first probe sequence 1608 and, optionally, a probe capture sequence 1610.
  • the first probe sequence 1608 may be complementary to the first target region 1602.
  • the second probe 1616 may comprise a second probe sequence 1615 and, optionally, a probe capture sequence 1618.
  • the second probe sequence 1615 may be complementary to the second target region 1604.
  • the third probe 1658 may comprise a third probe sequence 1660 and a probe capture sequence 1662.
  • the third probe sequence 1660 may be complementary to the feature probe binding sequence 1656.
  • the probe capture sequence 1662 is the same probe capture sequence as the probe capture sequences 1610, 1618 of the first probe and/or the second probe, respectively.
  • the cell, cell bead or nucleus 1600 may be further contacted with additional probes under conditions to generate additional probe-associated molecules or probe- associated complexes.
  • the additional probe-associated molecule(s) may be or comprise a probe- linked molecule, as described elsewhere herein.
  • the additional probe-associated molecule(s) or probe-linked molecule(s) may be any of the probe-associated molecules or probe- linked molecules described herein (e.g., generated from an extended probe, a barcoded extended probe, etc.).
  • the cell (or cell bead or nucleus) 1600 may be further contacted with a fourth probe (not shown) similar to 1658 which comprises (i) a fourth probe sequence similar to 1660 and (ii) a fourth probe capture sequence similar to 1662.
  • the fourth probe sequence may be complementary to the second feature probe binding sequence, as further described herein.
  • the fourth probe capture sequence is the same probe capture sequence as the probe capture sequences 1610, 1618 of the first probe and/or the second probe, respectively.
  • the cell, nucleus or cell bead 1600 may be partitioned into a first partition of a first set of partitions prior to any processing operations described above including, without limitation, fixing, permeabilizing, contacting with probes, and generating probe- associated or probe-linked molecules.
  • the cell, nucleus or cell bead 1600 may be fixed and optionally permeabilized prior to partitioning in the first partition and then subsequently processed in the first partition, e.g., contacting with probes and generating molecules.
  • the cell, nucleus or cell bead 1600 comprising the first probe- associated molecule 1630 and the second probe-associated molecule 1665 may be partitioned into a first partition of a first set of partitions or further processed in the first partition.
  • the cell, cell bead or nucleus 1600 may further comprise additional probe- associated molecules or complexes.
  • 1600 may comprise a third probe-associated complex (not shown) that is similar to 1665 but comprises (i) a fourth probe comprising a fourth probe sequence complementary to the second feature probe binding sequence and (ii) a reporter oligonucleotide (similar to 1657) as further described herein.
  • the reporter oligonucleotide may be provided as part of or coupled to the second feature binding group, e.g., a feature binding group configured to couple to an intracellular protein.
  • the cell, nucleus or cell bead 1600 may be subjected to processing within the partition, such as lysis, to release the cellular/nuclear components (e.g., the first probe-associated molecule and the second probe-associated molecule) within the partition.
  • the cell, nucleus or cell bead 1600 may remain intact.
  • the cell bead is processed to release cellular components while keeping the cell bead intact.
  • a probe binding molecule 1617 and a barcode molecule 1619 may be provided within the first partition.
  • the first probe- associated molecule 1630 and the second probe-associated molecule 1665 may be contacted with one or more probe binding molecules 1617 and barcode molecules 1619.
  • the first partition further comprises one or more additional probe-associated molecules or complexes similar to 1665 (not shown).
  • the additional probe-associated complex may comprise the third probe-associated complex described above, which comprises a fourth probe and a reporter oligonucleotide for a second feature binding group, e.g., a feature binding group configured to couple to an intracellular protein.
  • Additional probe-associated complexes, such as the third probe-associated complex may be contacted with one or more probe binding molecules 1617 and barcode molecules 1619.
  • the contacting of a cell, nucleus or cell bead 1600 in the first partition with one or more probe binding molecules may be simultaneously as the contacting with the probes (e.g., 1606, 1616, 1658 and optionally the fourth probe) as described above.
  • the barcode molecules 1619 may comprise a barcode capture sequence or a common sequence common to a plurality of barcode molecules and a first barcode sequence common to the first partition of the first set of partitions.
  • the nucleic acid barcode molecule may, in some instances, be coupled to a bead, such as a gel bead, or other support, as described herein, and can comprise additional functional sequences, including, but not limited to, a unique molecular identifier (UMI), a capture sequence, a primer sequence (e.g., a R1/R2 sequence), additional barcode sequence segments, etc..
  • UMI unique molecular identifier
  • the probe binding molecules 1617 may comprise a probe binding sequence complementary to any or a combination of the probe capture sequences 1610, 1618, 1662, a fourth probe capture sequence, and a barcode binding sequence complementary to the common sequence of the barcode molecule 1619.
  • the probe binding molecules 1617 and the barcode molecules 1619 may be provided as a preannealed complex.
  • the probe binding molecules 1617 and the barcode molecules 1619 may hybridize to the first probe-associated molecule 1630 and the second probe-associated molecule 1665 and/or an additional probe-associated complex, such as the third probe-associated complex (e g., via hybridization of the probe binding molecules 1617 to the probe capture sequences 1610, 1618, 1662, and the fourth probe capture sequence), thereby generating a first barcoded nucleic acid molecule and a second barcoded nucleic acid molecule, and optionally additional barcoded nucleic acid molecules.
  • Additional processing may occur within the first partition, e.g., ligation of the barcode molecules 1619 to the probes (1606, 1616, 1658 or the fourth probe).
  • the additional barcoded nucleic acid molecule is generating using an additional probe-associated complex, e.g., the third probe-associated complex (not shown), probe binding molecules 1617 and barcode molecules 1619.
  • the contents of each partition or a subset of the first set of partitions may be collected from the first set of partitions, e.g., from operation 1670, and repartitioned into a second set of partitions.
  • the contents of the first set of partitions may comprise the cell, nucleus or cell bead 1600 and/or the processed cellular or nuclear components, e.g., the first barcoded nucleic acid molecule, the second barcoded nucleic acid molecule, and optionally the additional barcoded nucleic acid molecule(s).
  • the contents of the partitions of the first set of partitions may be pooled together and re-distributed to a second set of partitions.
  • a second partition of the second set of partitions may comprise the cell, nucleus or cell bead 1600 and/or the processed cellular/nuclear components.
  • the cell, nucleus or cell bead 1600 may be subjected to processing within the second partition, such as lysis, to release the cellular/nuclear components (e.g., the first barcoded nucleic acid molecule, the second barcoded nucleic acid molecule, and optionally the additional barcoded nucleic acid molecule(s)) within the second partition.
  • the cell, nucleus or cell bead 1600 may remain intact.
  • a plurality of capture molecules 1620 may be provided within the second partition.
  • the plurality of capture molecules 1620 may be coupled to a support (e.g., a particle, bead, gel bead, etc.). In some instances, the plurality of capture molecules 1620 may be releasably coupled to the support and the plurality of capture molecules 1620 may be released in the second partition.
  • the capture molecules 1620 may each comprise a second barcode sequence, which may be the same sequence or a different sequence as the first barcode sequence (of the barcode molecule 1619). The second barcode sequence may be unique to the second partition and differ from the second barcode sequences of other partitions of the second set of partitions.
  • the first barcoded nucleic acid molecule and the second barcoded nucleic acid molecule may each be contacted with a capture molecule 1620.
  • the capture molecules 1620 may comprise a second barcode capture sequence, which may be complementary to a sequence of the barcode molecule 1619. Hybridization of the capture molecules 1620 to the first barcoded molecule and the second barcoded nucleic acid molecule may be sufficient to generate a third barcoded nucleic acid molecule and a fourth barcoded nucleic acid molecule.
  • hybridization of capture molecules 1620 to the additional barcoded nucleic acid molecule(s), e.g., from additional reporter oligonucleotides 1657 on additional feature binding groups 1652, may be sufficient to generate a fifth barcoded nucleic acid molecule.
  • hybridization of the capture molecules 1620 to the first barcoded molecule and the second barcoded nucleic acid molecule may be sufficient to couple the capture molecule (comprising the second barcode sequence) to both the first barcoded molecule and the second barcoded nucleic acid molecule.
  • hybridization of a capture molecule 1620 to the additional barcoded nucleic acid molecule may be sufficient to couple the capture molecule (comprising the second barcode sequence) to the additional barcoded nucleic acid molecule.
  • further processing may be performed, e.g., ligation of the capture molecules 1620 to the first barcoded nucleic acid molecule and the second barcode nucleic acid molecule (and optionally the additional barcoded nucleic acid molecule).
  • the first and second barcoded nucleic acid molecule may comprise the capture molecule 1620.
  • the third barcoded nucleic acid molecule, the fourth barcoded nucleic acid molecule, and the fifth barcoded nucleic acid molecule may each comprise a sequence corresponding to the first barcode sequence and a sequence corresponding to the second barcode sequence.
  • an extension reaction is performed (e.g., from the capture molecule 1620 toward the reporter oligonucleotide sequence 1657) to generate the fourth barcoded molecule and/or the fifth barcoded nucleic acid molecule.
  • FIG. 16B schematically illustrates another example workflow for barcoding multiple analytes of a cell, nucleus or cell bead.
  • the workflow for processing a nucleic acid molecule may be substantially similar to that depicted in FIG. 16A, but the workflow for processing a feature (e.g., protein) may differ.
  • the feature binding group 1652 or the reporter oligonucleotide 1657 may comprise a binding sequence that is capable of hybridizing to a probe binding molecule 1617 and/or barcode molecule 1619.
  • a permeabilized (and optionally fixed) cell or nucleus may be contacted with one or more feature binding groups 1652, which may (a) comprise the reporter oligonucleotide 1657 and (b) be configured to couple to (i) an intracellular protein (or an intranuclear protein) or (ii) a cell membrane protein (or nuclear membrane protein).
  • feature binding groups 1652 may (a) comprise the reporter oligonucleotide 1657 and (b) be configured to couple to (i) an intracellular protein (or an intranuclear protein) or (ii) a cell membrane protein (or nuclear membrane protein).
  • the one or more feature binding groups 1652 includes (i) a first feature binding group that comprises the reporter oligonucleotide 1657 and is configured to couple to an intracellular (or an intranuclear protein) and (ii) a second feature binding group that comprises the reporter oligonucleotide 1657 and is configured to couple to a cell membrane protein (or a nuclear membrane protein).
  • the cell, nucleus or cell bead 1600 comprising the first probe- associated molecule 1630 and the one or more feature binding group 1652 may be partitioned into a first partition of a first set of partitions or further processed in the first partition.
  • a probe binding molecule 1617 and a barcode molecule 1619 may be provided.
  • the feature binding group 1652 (e.g., one or more feature binding groups configured to couple to an intracellular protein or an intranuclear protein) coupled to the reporter oligonucleotide 1657 may be contacted with one or more probe binding molecules 1617 and barcode molecules 1619.
  • a barcode molecule 1619 may comprise a barcode capture sequence or a common sequence common to a plurality of barcode molecules and a first barcode sequence common to the first partition of the first set of partitions.
  • the nucleic acid barcode molecule may, in some instances, be coupled to a bead, such as a gel bead, or other support, as described herein, and can comprise additional functional sequences, including, but not limited to, a unique molecular identifier (UMI), a capture sequence, a primer sequence (e.g., a R1/R2 sequence), additional barcode sequence segments, etc..
  • UMI unique molecular identifier
  • the probe binding molecules 1617 may comprise a probe binding sequence complementary to a sequence of the reporter oligonucleotide 1657.
  • the probe binding molecules 1617 and the barcode molecules 1619 may be provided as a preannealed complex.
  • the probe binding molecules 1617 and the barcode molecules 1619 may hybridize to the first probe-associated molecule 1630 (as described above) and the reporter oligonucleotide 1657 (e.g., via hybridization of the probe binding molecules 1617 to a sequence of the reporter oligonucleotide 1657), thereby generating a first barcoded nucleic acid molecule and a second barcoded nucleic acid molecule.
  • Additional barcoded nucleic acid molecules may be generated using additional reporter oligonucleotides 1657 from additional feature binding groups 1652 (e.g., configured to couple to cell or nuclear membrane proteins and/or intracellular or intranuclear proteins). Additional processing may occur within the first partition, e g., ligation of the barcode molecules 1619 to the probes (1606, 1616) or to the reporter oligonucleotide 1657
  • the contents of each partition or a subset of the first set of partitions may be collected from the first set of partitions, e.g., from operation 1670, and repartitioned into a second set of partitions.
  • the contents of the first set of partitions may comprise the cell, nucleus or cell bead 1600 and/or the processed cellular/nuclear components, e.g., the first barcoded nucleic acid molecule, the second barcoded nucleic acid molecule, and optionally the additional barcoded nucleic acid molecule(s).
  • the contents of the partitions of the first set of partitions may be pooled together and re-distributed to a second set of partitions.
  • a second partition of the second set of partitions may comprise the cell, nucleus or cell bead 1600 and/or the processed cellular/nuclear components (e.g., barcoded products).
  • the cell, nucleus or cell bead 1600 may be subjected to processing within the second partition, such as lysis, to release the cellular/nuclear components (e.g., the first barcoded nucleic acid molecule, the second barcoded nucleic acid molecule, and optionally the additional barcoded nucleic acid molecule(s)) within the second partition.
  • the cell, nucleus or cell bead 1600 may remain intact.
  • a plurality of capture molecules 1620 may be provided within the second partition.
  • the plurality of capture molecules 1620 may be coupled to a support (e.g., a particle, bead, gel bead, etc.).
  • the plurality of capture molecules 1620 may be releasably coupled to the support and the plurality of capture molecules 1620 may be released in the second partition.
  • the capture molecules 1620 may each comprise a second barcode sequence, which may be the same sequence or a different sequence as the first barcode sequence (of the barcode molecule 1619).
  • the second barcode sequence may be unique to the second partition and differ from the second barcode sequences of other partitions of the second set of partitions.
  • the first barcoded nucleic acid molecule and the second barcoded nucleic acid molecule may each be contacted with a capture molecule 1620.
  • the capture molecules 1620 may comprise a second barcode capture sequence, which may be complementary to a sequence of the barcode molecule 1619.
  • the capture molecules 1620 may comprise a sequence complementary to an additional probe-binding molecule (e.g., splint oligonucleotide, not shown), and the probe-binding molecule may comprise a sequence complementary to a sequence of the barcode molecule 1619.
  • Hybridization of the capture molecules 1620 to the first barcoded molecule and the second barcoded nucleic acid molecule (or to the additional probe-binding molecule, which may hybridize to the first barcoded molecule and the second barcoded molecule) may be sufficient to generate a third barcoded nucleic acid molecule and a fourth barcoded nucleic acid molecule.
  • hybridization of 1620 to the additional barcoded nucleic acid molecule(s), e.g., from additional reporter oligonucleotides 1657 on additional feature binding groups 1652, may be sufficient to generate a fifth barcoded nucleic acid molecule.
  • hybridization of the capture molecules 1620 to the first barcoded molecule and the second barcoded nucleic acid molecule may be sufficient to couple the capture molecule (comprising the second barcode sequence) to both the first barcoded molecule and the second barcoded nucleic acid molecule.
  • hybridization of 1620 to the additional barcoded nucleic acid molecule may be sufficient to couple the capture molecule (comprising the second barcode sequence) to the additional barcoded nucleic acid molecule e.g., generated from additional reporter oligonucleotides 1657 on additional feature binding groups 1652.
  • further processing may be performed, e.g., performing an extension reaction, ligation of the capture molecules 1620 to the first barcoded nucleic acid molecule, the second barcode nucleic acid molecule, and optionally the additional barcoded nucleic acid molecule.
  • the first and second barcoded nucleic acid molecule may comprise the capture molecule 1620.
  • the third barcoded nucleic acid molecule, the fourth barcoded nucleic acid molecule, and the fifth barcoded nucleic acid molecule may each comprise a sequence corresponding to the first barcode sequence and a sequence corresponding to the second barcode sequence.
  • an extension reaction is performed (e.g., from the capture molecule 1620 toward the reporter oligonucleotide sequence 1657) to generate the fourth barcoded molecule and/or the fifth barcoded nucleic acid molecule.
  • the reporter oligonucleotide (comprising the reporter sequence) of the feature binding group may be contacted with a plurality of probes.
  • the reporter oligonucleotide comprises one or more feature probe binding sequences, which may comprise sequences complementary to the pair of probes. For example, referring to FIG.
  • a cell, nucleus or cell bead 1700 may comprise a feature (e.g., a protein such as a cell/nuclear membrane protein or an intracellular/intranuclear protein) 1750.
  • a feature binding group 1752 may be coupled to the feature 1750.
  • the feature binding group 1752 may comprise or be coupled to an oligonucleotide comprising a reporter oligonucleotide (comprising a reporter sequence) 1754 and, in some instances, additional functional sequences, such as primer sequences, sequencing primer sequences, UMIs, etc., as described elsewhere herein.
  • the reporter oligonucleotide 1754 may comprise any number of target regions.
  • the reporter oligonucleotide 1754 may comprise two target regions to which a first probe 1757 and a second probe 1758 may hybridize.
  • the two target regions may be adjacent or non-adj acent, and they may be disposed on the same strand of the reporter oligonucleotide 1754.
  • the probes may comprise sequences that are complementary to the target regions of the reporter oligonucleotide 1754, and each probe may comprise other useful sequences.
  • a probe (e.g., the first probe 1757 or the second probe 1758) may comprise (i) a probe sequence (e g., 1760) complementary to a target region of the reporter oligonucleotide 1754, and (ii) a probe capture sequence 1762, which may be complementary to a sequence of a probe binding molecule 1717 (also referred to as a splint or splint oligonucleotide).
  • the probe binding molecule 1717 may also comprise a sequence complementary to a sequence (e.g., capture sequence) of a barcode molecule 1719.
  • barcoding e.g.
  • hybridization of the probe binding molecule 1717 and barcode molecule 1719 to the probe capture sequence 1762) may occur in bulk or in a partition.
  • barcoding may be performed without a probe binding molecule.
  • the barcode molecule 1719 may comprise a sequence complementary to the probe capture sequence 1762 and directly anneal to the probe.
  • the feature binding group 1752 is subjected to conditions sufficient for hybridization of the probe molecules to the reporter oligonucleotide 1754, thereby generating a probe-associated reporter oligonucleotide complex.
  • the coupling of the probes to the reporter oligonucleotide 1754 may occur in bulk or in a partition.
  • the probes may be linked together (e.g., enzymatically or chemically), thereby generating a probe-linked nucleic acid molecule (or complex).
  • the first probe 1757 may comprise a first reactive moiety and the second probe 1758 may comprise a second reactive moiety.
  • the reactive moieties may be positioned such that, following hybridization of the first probe 1757 and the second probe 1758 to the reporter oligonucleotide 1754, the reactive moieties are adjacent.
  • the reactive moieties may then be subjected to conditions sufficient to cause them to react to yield a probe-linked nucleic acid molecule (or complex) comprising the first probe 1757 linked to the second probe 1758.
  • the probes comprise “click chemistry” moieties.
  • the first probe may be enzymatically linked (e.g., via ligation) to the second probe.
  • a gap region (not shown) may be disposed between the first probe 1757 and the second probe 1758, following hybridization of the probes to the reporter oligonucleotide 1754.
  • the first probe 1757 may be linked to the second probe 1758 using a gap-fill approach, such as those described above.
  • the probe-linked nucleic acid molecule may then be subjected to barcoding (e.g., contacting with the probe binding molecule 1717 and the barcode molecule 1719), which may occur in a partition.
  • the barcoding may occur prior to the linking of the probes.
  • the reporter oligonucleotide 1754 may be hybridized to the probes, partitioned, barcoded, and then the probes may be linked.
  • the reporter oligonucleotide 1754 may be hybridized to the probes, linked, partitioned, then barcoded.
  • the reporter oligonucleotide 1754 may be hybridized to the probes, partitioned, linked, then barcoded.
  • the operations described herein may occur at any useful process, or in any useful order.
  • multiple partitioning operations maybe performed, e.g., for combinatorial barcoding.
  • the reporter oligonucleotide may comprise the same target sequences (e.g., 702, 704, 802, 804, 902, 904, 1502, 1504, 1602, 1604, etc.) as the nucleic acid molecule (e.g., RNA molecule).
  • the nucleic acid molecule e.g., RNA molecule.
  • the first probe may have a first sequence that is complementary to both the first target sequence of a nucleic acid molecule (e.g., 702, 802, 902, 1502, 1602) and a first sequence of the reporter oligonucleotide 1754
  • the second probe may have a second sequence that is complementary to both the second target sequence of a nucleic acid molecule (e.g., 704, 804, 904, 1504, and 1604) and a second sequence of the reporter oligonucleotide 1754.
  • the provision of just two probe types e.g., a first probe and a second probe
  • a first probe and a second probe may be sufficient to generate the first barcoded molecule (e.g., generated from the nucleic acid molecule, e g., RNA molecule)
  • the second barcoded molecule e.g , generated from the reporter oligonucleotide of the feature binding group, such as a group configured to couple to a cell/nuclear membrane protein
  • additional barcoded molecules e.g., generated from the reporter oligonucleotide of an additional feature binding group, such as a group configured to couple to an intracellular/intranuclear protein.
  • each of the probes may be capable of or configured to hybridize to a barcode molecule (e.g., in the first partition) and/or a capture molecule.
  • each of the probes may be multiplexed or combinatorially barcoded, such that multiplet partitions (e.g., partitions comprising more than one cell, one nucleus or cell bead) may be deconvolved, for example to determine the originating partition or sample of each cell, nucleus or cell bead within a partition (see, e.g., FIG. 10).
  • the barcoded molecules may be used to determine the origin of different analyte types (e g., proteins, nucleic acid molecule, etc ); for example, two analyte types may be attributed to the same originating cell, nucleus, cell bead, sample, or partition(s).
  • the reporter oligonucleotide comprises two or more target sequences which are different than the target sequences of the nucleic acid molecule (e.g., RNA molecule).
  • a first probe and a second probe may hybridize to a first target region and a second target region of a nucleic acid molecule
  • a third probe and a fourth probe may hybridize to target regions of a reporter oligonucleotide (e.g., a reporter oligonucleotide from a feature binding group, such as a feature binding group configured to couple to a cell/nuclear membrane protein).
  • a reporter oligonucleotide e.g., a reporter oligonucleotide from a feature binding group, such as a feature binding group configured to couple to a cell/nuclear membrane protein
  • Additional probe types may be provided, such as a fifth probe and a sixth probe, that hybridize to target regions of an additional reporter oligonucleotide (e.g., a reporter oligonucleotide from a feature binding group, such as a feature binding group configured to couple to an intracellular/intranuclear protein).
  • an additional reporter oligonucleotide e.g., a reporter oligonucleotide from a feature binding group, such as a feature binding group configured to couple to an intracellular/intranuclear protein.
  • Each of the probes or a combination of the probes may comprise probe capture sequences, which may be used for subsequent barcoding.
  • each of the probes may be capable of or configured to hybridize to a barcode molecule (e.g., in the first partition) and/or a capture molecule (e.g., in a second partition).
  • each of the probes may be multiplexed or combinatorially barcoded, such that multiplet partitions (e.g., partitions comprising more than one cell, nucleus or cell bead) may be deconvolved, for example to determine the originating partition or sample of each cell, nucleus or cell bead within a partition (see, e.g., FIG. 10).
  • the barcoded molecules may be used to determine the origin of different analyte types (e.g., proteins, nucleic acid molecules, etc ); for example, two analyte types may be attributed to the same originating cell, nucleus, cell bead, sample, or partition(s).
  • the nucleic acid molecules e.g., from a cell, a nucleus or cell bead, or a reporter oligonucleotide
  • the one or more target regions may correspond to a gene or a portion thereof, or another known sequence.
  • the target regions may have the same or different sequences, and may be located within the same strand or on different strands.
  • the target regions may be located adjacent to one another or may be spatially separated along a strand of the nucleic acid molecule.
  • the target regions may be located on the same strand or different strands. Analyzing two or more target regions may involve providing two or more probes, where a first probe has a sequence that is complementary to the first target region, a second probe has a sequence that is complementary to the second target region, etc.
  • the nucleic acid molecule may be a target nucleic acid molecule and may comprise any number of nucleic acid features or nucleotides.
  • any of the probes may comprise any number of additional adaptor or functional sequences, such as an additional probe sequence, a unique molecule identifier, a barcode sequence, a primer sequence, a capture sequence, a sequencing primer sequence, etc.
  • one or more operations may be performed within a partition, such as a droplet or well.
  • the nucleic acid molecule e.g., RNA molecule
  • the feature e.g., protein
  • a cell, nucleus or cell bead comprising the nucleic acid molecule and feature
  • the probe-linked or probe-associated nucleic acid molecule optionally comprised within or on a cell, nucleus or cell bead, may be generated in a bulk solution or in a partition.
  • the cell, nucleus or cell bead may be contacted with a feature binding group in a bulk solution or in a partition. Provision of the probes (e.g., the first probe, the second probe, and the third probe) may occur in the bulk solution or in individual partitions.
  • a partition e.g., a first partition of a first set of partitions
  • Different partitions within the first set of partitions may comprise the same or different probes (e.g., for different target sequences or different reporter sequences).
  • the probe binding molecules and the nucleic acid barcode molecules may be provided in a partition.
  • the cell, nucleus or cell bead comprising the feature and the nucleic acid molecule may be contacted with the probes in bulk, and partitioned into a first set of partitions.
  • the first set of partitions may comprise the probe binding molecule and the nucleic acid barcode molecules comprising a common sequence.
  • Different partitions among the first set of partitions may comprise barcode molecules with different barcode sequences; for instance, an additional partition of the first set of partitions may comprise numerous barcode molecules that each have a barcode sequence that is unique to the partition (e.g. differs across partitions).
  • the partition may comprise additional reagents for performing a nucleic acid reaction (e.g., digestion, ligation, extension, amplification).
  • the partition may comprise a linking enzyme (e.g., ligase), which may be used to ligate the nucleic acid barcode molecule to the first probe, the second probe, or the third probe (e.g., via the probe capture sequence of each probe).
  • a linking enzyme e.g., ligase
  • the probe binding molecule, the probe capture sequence, and/or the barcode capture sequence comprises one or more reactive moieties, which may be used to chemically link the nucleic acid barcode molecule to the probe capture sequence.
  • the resultant barcoded products may comprise: a first barcoded product comprising a sequence corresponding to the first target region, a sequence corresponding to the second target region, a sequence corresponding to the probe capture sequence of the first probe or the second probe, and a sequence corresponding to the barcode sequence; and a second barcoded product comprising a sequence corresponding to the reporter sequence, the probe capture sequence of the third probe (which may be the same or different than that of the first probe or second probe), and the barcode sequence.
  • one or more processes described herein may be performed in a cell (e.g., a cell in solution, or a cell comprised within a tissue sample), nucleus or cell bead.
  • a plurality of cells, nuclei or cell beads may comprise a plurality of nucleic acid molecules and features.
  • the cells, nuclei or cell beads may be alive or fixed and/or permeabilized.
  • the cells, nuclei or cell beads may be contacted with a feature binding group comprising a reporter sequence.
  • the first probe, the second probe, and the third probe may also be provided to the cells, nuclei or cell beads, in bulk solution or in a partition to generate the first probe-associated molecule and the second probe-associated molecule.
  • the cells, nuclei or cell beads may be washed to remove unbound probes. Subsequently, the cells, nuclei or cell beads comprising the probe-associated molecules may be partitioned into a plurality of separate partitions, where at least a subset of the plurality of separate partitions comprises a single cell, single nucleus, or single cell bead. Barcoding may be performed within the separate partitions. Barcoding, as described herein, may comprise attaching or hybridizing a nucleic acid barcode molecule to the first probe-associated molecule and the second probe-associated molecule. The nucleic acid barcode molecules provided within each partition of the plurality of separate partitions may be provided attached to beads.
  • the nucleic acid barcode molecule may be releasably attached to a bead (e.g., via a labile bond).
  • Each partition (or a subset of partitions) of the plurality of separate partitions may comprise a bead comprising a plurality of nucleic acid barcode molecules attached thereto (e g., as described herein).
  • the plurality of nucleic acid barcode molecules attached to each bead may comprise a unique barcode sequence, such that each partition of the plurality of separate partitions comprises a different barcode sequence.
  • the barcoded molecules arising from a single cell, single nucleus, or single cell bead may have a same barcode sequence (e.g., a common barcode sequence), such that each barcoded nucleic acid molecule can be traced to a given partition and/or, in some instances, a single cell, a single nucleus, or a single cell bead.
  • the released components may then be partitioned, as described herein, in a second set of partitions comprising capture molecules with a second barcode sequence, such that different partitions of the second set of partitions have a unique second barcode sequence.
  • the cells, nuclei, or cell beads described herein may be processed either prior to, during, or following barcoding.
  • the cells, nuclei, or cell beads may be fixed or permeabilized at any useful point in time.
  • the cells, nuclei, or cell beads may be fixed and permeabilized prior to or following hybridization of the probes, or prior to or following contact with the feature binding groups.
  • the cells, nuclei, or cell beads may be fixed and permeabilized prior to contact with the feature binding groups, and then contacted with the probes.
  • the fixation or permeabilization process may be repeated.
  • a cell, nucleus, or cell bead may be fixed and permeabilized, contacted with the probes and the feature binding groups (either simultaneously or in a step-wise fashion), and then fixed again.
  • the cells, nuclei, or cell beads may be stored for a duration of time prior to further processing, e.g., contacting the cells, nuclei, or cell beads with the probes and/or feature binding groups.
  • the cells, nuclei, or cell beads may be fixed and/or permeabilized and then contacted with the probes and/or feature binding groups after about 1 minute, 10 minutes, 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, 18 hours, 19 hours, 20 hours, 21 hours, 22 hours, 23 hours, 24 hours or more.
  • the cells, nuclei, or cell beads may be fixed and/or permeabilized and then contacted with the probes and/or feature binding groups after about 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days or more.
  • the cells, nuclei, or cell beads may be fixed and/or permeabilized and then contacted with the probes and/or feature binding groups after about 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 20 weeks, 30 weeks, 40 weeks, 50 weeks or more.
  • the cells, nuclei, or cell beads may be fixed and/or permeabilized and then contacted with the probes and/or feature binding groups after about 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more.
  • the cells, nuclei, or cell beads may be fixed and/or permeabilized and then contacted with the probes and/or feature binding groups at any useful time, which may fall within a range of times, e.g., after about 2-5 weeks, after about 3-6 months, after about 1-2 years, etc.
  • the cells, nuclei, or cell beads may be frozen, e.g., subsequent to fixation and/or permeabilization. Such freezing of the cells, nuclei, or cell beads may be useful in storage of samples for longer durations, e.g., if a sample is to be stored for greater than 1-2 weeks prior to contacting the sample with the probes and/or feature binding groups.
  • the cells, nuclei, or cell beads may be fixed, optionally permeabilized, and then frozen for any useful duration of time, followed by contacting of the cells, nuclei, or cell beads with the probes and/or feature binding groups.
  • the cells, nuclei, or cell beads may be fixed, frozen, and permeabilized, either prior to or following contacting of the cells, nuclei or cell beads with the probes and/or feature binding groups.
  • the freezing operation may be performed at any useful or convenient time, e.g., prior to, concurrently with, or following fixation, permeabilization, contacting with probes, contacting with feature binding groups, etc.
  • the cells, nuclei, or cell beads may be contacted with the probes and feature binding groups at any useful time, in partitions or in bulk.
  • the cells, nuclei, or cell beads may be contacted with the probes prior to, during, or following contact with the feature binding groups. Contact with the probes and/or feature binding groups may occur in bulk or in partitions (e g., droplets, wells).
  • the cells, nuclei, or cell beads may be contacted with the probes and feature binding groups (either simultaneously, or in a step-wise fashion), and then barcoded in partitions.
  • the cells, nuclei, or cell beads may be contacted with the probes and feature binding groups in partitions.
  • FIG. 29 shows an example workflow of processing cells, according to the methods described herein.
  • a cell may be fixed and permeabilized, e.g., in 4% formaldehyde and 0.01% Tween-20 or a commercially available fixation and permeabilization buffer (e g., commercially available BioLegend® fixation and permeabilization buffer).
  • the fixed and permeabilized cell may be incubated with a first probe and a second probe to generate a first probe-associated molecule (e.g., a probe-associated RNA molecule).
  • the cell may then be contacted with a feature binding group (e.g., antibody) comprising a reporter oligonucleotide to generate a cell comprising a feature coupled to a feature-binding group.
  • a feature binding group e.g., antibody
  • the fixed and permeabilized cell may be incubated with a feature binding group, optionally fixed again, and then contacted with a first probe and a second probe to generate a probe-associated molecule (e.g., a probe-associated RNA molecule).
  • the fixed and permeabilized cell may be incubated with the first probe and the second probe to generate a probe-associated molecule, and then contacted with the feature binding groups.
  • Subsequent barcoding may be performed, e.g., in partitions.
  • a cell may be fixed, contacted with the probe and/or feature binding group, then subsequently permeabilized. It will be appreciated that any order of operations of fixation, permeabilization, probe hybridization, contacting with the feature binding groups, etc., may be performed at any convenient or useful step and in any order, and that any of the processes may be repeated.
  • a cell, nucleus, or cell bead may be contacted with the feature binding groups, fixed and/or permeabilized, contacted with additional feature binding groups, which may be beneficial for assaying extracellular and intracellular peptides, polypeptides, or proteins, and optionally, fixed again.
  • the cell, nucleus, or cell bead may be fixed and/or permeabilized, then contacted with feature binding groups (e.g. for intracellular and/or extracellular analytes) and optionally, fixed again.
  • the cell, nucleus, or cell bead may be contacted with the sets of probes (e.g., first probe, second probe, and/or third probe). See also, Examples 8 and 9.
  • the methods, compositions, kits, and systems of the present disclosure may comprise providing methods for processing fixed biological particles (e.g., a cell, nucleus, or cell bead).
  • the method comprises a) fixing and permeabilizing a biological particle or providing a fixed and permeabilized biological particle.
  • the method may further comprise b) contacting the fixed and permeabilized biological particle with a first reagent configured to couple to an analyte of the biological particle.
  • the analyte is an intracellular analyte, such as a nucleic acid or a polypeptide, and the biological particle is a cell.
  • the analyte is an intranuclear analyte, such as a nucleic acid or a polypeptide, and the biological particle is a nucleus.
  • the first reagent configured to couple to an analyte may be (i) a first reagent configured to couple to a nucleic acid (such as one or more nucleic acid probes as described herein) or (ii) a first reagent configured to couple to a peptide, polypeptide, or protein (such as one or more feature binding groups as described herein).
  • b) provides a fixed and permeabilized biological particle, e.g., cell or nucleus, comprising the first reagent coupled to the analyte, e.g., nucleic acid or polypeptide, of the biological particle.
  • the method may further comprise c) performing an additional fixation of the biological particle from b).
  • c) comprises additional fixation of the biological particle from b), wherein the biological particle from b) comprises the first reagent configured to couple to an analyte of the biological particle.
  • the first reagent may be coupled to the analyte (nucleic acid or polypeptide) of the biological particle (e.g., cell or nucleus).
  • the first reagent may be a reagent configured to couple to a nucleic acid analyte or a reagent configured to couple to a polypeptide.
  • c) comprises additional fixation of the biological particle, such as a cell, wherein the cell comprises a first reagent coupled to a polypeptide.
  • the polypeptide is an intracellular polypeptide.
  • the method may further comprise d) comprising contacting the biological particle (e g., cell or nucleus) from c) (which has been initially fixed and permeabilized, contacted with the first reagent or comprises the first reagent, and additionally fixed) with a second reagent configured to couple to an analyte (e.g., a nucleic acid or polypeptide), wherein the second reagent is different from the first reagent and/or the second reagent is configured to couple to an analyte that is different than the analyte that the first reagent is configured to couple to.
  • a biological particle e.g., cell or nucleus
  • an analyte e.g., a nucleic acid or polypeptide
  • the first reagent is configured to couple to a polypeptide (such as one or more feature binding groups as described herein) and the second reagent is configured to couple to a nucleic acid (such as one or more nucleic acid probes as described herein).
  • the biological particle of d) may comprise the first reagent coupled to a polypeptide and the second reagent coupled to a nucleic acid.
  • any number of barcoding operations may be performed for a given nucleic acid molecule and/or feature binding group, e.g., using a combinatorial barcoding (e.g., split-pool) approaches.
  • additional barcoding operations may be useful, for example, in indexing nucleic acid molecules and features (e.g., proteins) to a cell, a nucleus, a cell bead, a sample, a partition, or a plurality of partitions. Such indexing may be useful in situations when a single partition is occupied by multiple cells, nuclei, or cell beads.
  • a partition comprises more than one cell, nucleus or cell bead; for example, it may be useful in certain situations to overload partitions, e.g., to overcome Poisson loading statistics in partitions and/or to prevent reagent waste (e.g., from unoccupied partitions). Accordingly, such indexing may be useful in attributing (i) nucleic acid molecules and (ii) features (e.g., proteins) in multiply-occupied partitions to the originating cell, nucleus, cell bead, partition, sample, etc., as is described elsewhere herein.
  • features e.g., proteins
  • the workflow provided in FIG. 10 may be performed for nucleic acid molecules and features (e.g., proteins) within a population of cells, nuclei or cell beads.
  • a first population of cells, nuclei or cell beads 1002 may be contacted with the first probe, the second probe, and optionally, the third probe (e g., as shown in FIG. 16A and FIG. 16B).
  • the first probe and the second probe may hybridize to the nucleic acid molecule, generating a first probe-associated molecule (or complex), and optionally, the third probe may hybridize to a reporter oligonucleotide (comprising a reporter sequence) or feature probe-binding sequence of a feature binding group (e.g., a group configured to couple to a cell/nuclear membrane protein) to generate a second probe-associated molecule (or complex).
  • a reporter oligonucleotide comprising a reporter sequence
  • feature probe-binding sequence of a feature binding group e.g., a group configured to couple to a cell/nuclear membrane protein
  • Additional probe(s) may be provided to hybridize to additional reporter oligonucleotide(s) or feature probe-binding sequence(s) of an additional feature binding group (e.g., a group configured to couple to an intracellular/intranuclear protein) of the first population of cells, nuclei or cell beads to generate additional probe-associated molecule(s).
  • additional feature binding group e.g., a group configured to couple to an intracellular/intranuclear protein
  • a second population of cells, nuclei or cell beads 1004 may be also be treated in the same way, e.g., with a fourth probe, a fifth probe, and optionally a sixth probe.
  • the fourth probe and the fifth probe may hybridize to the nucleic acid molecule of the second population of cells, nuclei or cell beads to generate a third-probe-associated molecule, and optionally, the sixth probe may hybridize to a reporter oligonucleotide or feature probe-binding sequence of a feature binding group of the second population of cells, nuclei or cell beads to generate a fourth probe-associated molecule.
  • Additional probe(s) may be provided to hybridize to additional reporter oligonucleotide(s) or feature probe-binding sequence(s) of an additional feature binding group (e.g., a group configured to couple to an intracellular/intranuclear protein) of the second population of cells, nuclei or cell beads to generate additional probe-associated molecule(s).
  • an additional feature binding group e.g., a group configured to couple to an intracellular/intranuclear protein
  • the first population of cells 1002 (or nuclei or cell beads) and the second population of cells 1004 (or nuclei or cell beads) may be barcoded with a first barcode sequence, as described herein, such that the first population of cells (or components therein, such as the first probe-associated molecule and the second-probe-associated molecule) 1002 has a different first barcode sequence than the second population of cells (or nuclei or cell beads or components within the cell, nuclei or cell beads, such as the third probe-associated molecule and the fourth probe-associated molecule) 1004.
  • the first population of cells 1002 may be pooled together with the second population of cells 1004 (or nuclei or cell beads) to generate a mixture of cells (or nuclei or cell beads).
  • the mixture of cells may be partitioned into a second plurality of partitions. In some instances, the mixture of cells (or nuclei or cell beads) may be partitioned into the second plurality of partitions such that some partitions of the second plurality of partitions comprises more than one cell (e.g., a cell, nucleus or cell bead multiplet partition).
  • a partition 1035 of the second plurality of partitions may comprise a cell, nucleus, or cell bead (“Cell A”) from the first population of cells 1002 (or nuclei or cell beads) and a cell, nucleus, or cell bead (“Cell B”) from the second population of cells 1004 (or nuclei or cell beads).
  • the partition 1035 may comprise an additional barcode sequence, which may be unique to the partition.
  • the cells (or nuclei or cell beads) in each partition may be subjected to an additional barcoding operation to append the additional barcode sequence on the barcoded nucleic acid molecules.
  • the barcoded nucleic acid molecules may be deconvolved, using the different barcode sequences (e.g., the first barcode sequence, the second barcode sequence, and the additional barcode sequences), to identify the originating cell, nucleus, or cell bead.
  • a barcoded nucleic acid molecule comprising the additional barcode sequence from partition 1035 and the first barcode sequence from the first population of cells 1002 may be used to identify that barcoded nucleic acid molecule as originating from Cell A.
  • a barcoded nucleic acid molecule comprising the additional barcode sequence from partition 1035 and the second barcode sequence from the second populations of cells 1004 may be used to identify that barcoded nucleic acid molecule from originating from Cell B.
  • the feature binding group(s) may be pre-indexed to a partition.
  • the feature binding group may be provided in the partitions in a pre-indexed manner, e.g., using a barcode sequence unique to the partition.
  • the feature binding group may be provided at a later operation of the method, subsequent to barcoding of the nucleic acid molecules within the cell.
  • the feature binding group may be provided and contacted with the feature 1650 of the cell, nucleus or cell bead (or released from the cell, nucleus or cell bead in the second partition).
  • the feature binding group may comprise or be hybridized to a barcode sequence that is specific to the second partition and that differs across the second partitions.
  • the barcode sequence can be used to index the feature binding group to the particular partition and back to the originating cell or cell bead, instead of using the first barcode sequence and the second barcode sequence from the first partition and second partition, respectively, to identify the partition, cell, nucleus, or cell bead.
  • the feature binding group(s) may be indexed to a partition by attaching or coupling a partition-specific barcode sequence directly to the feature binding group, thus obviating the usage of a third probe.
  • the feature binding group may comprise or be coupled to a reporter oligonucleotide comprising the reporter sequence and an attachment sequence, which may be used to attach a barcode molecule directly to the feature binding group.
  • the feature binding group may comprise a probe capture sequence (e g., 1662), thereby obviating the need for a third probe comprising the probe capture sequence.
  • the probe capture sequence may subsequently be barcoded, e.g., with the first barcode sequence of the barcode molecule within the first partition and with the second barcode sequence of the capture molecule within the second partition.
  • the attachment sequence may be used to hybridize a probe-binding molecule (e.g., splint molecule or splint oligonucleotide), which may be partially complementary to the barcode molecule (as described herein).
  • the attachment sequence of the reporter oligonucleotide may be used to hybridize the probe- binding molecule, which may hybridize (or be pre-annealed) to the barcode molecule, e.g., in a first partition.
  • a second barcode sequence from the capture molecule may be provided in the first partition or in a different (e.g., second) partition, which may anneal to a portion of the first barcode molecule.
  • additional operations are performed, e.g., extension, ligation, etc. to generate a barcoded molecule comprising sequences corresponding to the first barcode sequence, the second barcode sequence, and the reporter sequence.
  • the contents of the partitions may be pooled and the barcoded molecules may be duplicated or amplified by, for example, one or more amplification reactions, which may in some instances be isothermal.
  • the amplification reactions may comprise polymerase chain reactions (PCR) and may involve the use of one or more primers or polymerases.
  • the one or more primers may comprise one or more functional sequences (e.g., a primer sequence/primer binding sequence, a sequencing primer sequence (e g., R1 or R2), a partial sequencing primer sequence (e g., partial R1 or partial R2), a sequence configured to attach to the flow cell of a sequencer (e.g., P5 or P7, or partial sequences thereof), etc.) and may facilitate addition of said one or more functional sequences to the extended nucleic acid molecule.
  • the barcoded molecules, or derivatives thereof may be detected via nucleic acid sequencing (e.g., as described herein).
  • the systems may comprise any of the components described herein, e.g., a plurality of partitions (e.g., droplets, wells), which may be provided in any useful format, e g., a microfluidic device, a multi-well array or plate, etc.
  • the system may comprise a first set of partitions and a second set of partitions.
  • the first set of partitions may be the same or different types of partitions as the second set of partitions.
  • the first set of partitions may comprise microwells and the second set of partitions may comprise droplets.
  • both the first set of partitions and the second set of partitions may comprise droplets.
  • the systems may include nucleic acid barcode molecules, optionally coupled to supports (e.g., particles, beads, gel beads, etc ).
  • the systems may comprise any of the probes described herein, such as a first probe or plurality of first probes, a second probe or plurality of second probes, a third probe or plurality of third probes, and any useful reaction components (e.g., for performing a nucleic acid reaction, e.g., extension, ligation, amplification, etc ).
  • the systems may comprise one or more feature-binding groups.
  • the feature binding groups may be the same or different across partitions; for example, the feature binding groups may comprise a variety of antibodies that bind to different epitopes within a single partition, or the partitions may comprise different feature binding groups that bind to different epitopes or moieties.
  • the systems may include reaction components that are useful, such as, in non-limiting examples, enzymes (e.g., ligases, polymerases, reverse transcriptases, restriction enzymes, etc.), nucleotides bases, etc.
  • compositions useful for systems and methods for barcoding multiple analytes e.g., nucleic acid molecules and proteins (e.g., via a nucleic acid molecule, such as a reporter oligonucleotide, comprised in or coupled to a feature binding group).
  • a composition may comprise any of the probes described herein.
  • a composition may comprise a plurality of first probes, a plurality of second probes, a plurality of third probes, and/or a plurality of first probes, a plurality of second probes, and a plurality of third probes.
  • a probe or a set of probes may be designed to target a specific sequence or a set of specific sequences.
  • Such probes may be designed to have the same or different sequences within different partitions.
  • a first composition may comprise a first probe and a second probe designed to target two regions of a first gene
  • a second composition may comprise a first probe and a second probe designed to target two regions of a second gene, which second gene is different than the first gene.
  • the third probe (or pair of probes) may be designed to target a region of the reporter oligonucleotide (comprising the reporter sequence) or feature probe-binding sequence, which may be the same or different across partitions.
  • a composition may comprise nucleic acid barcode molecules, and/or probe binding molecules, which may optionally be provided coupled to a support (e.g., particle, bead).
  • a composition may comprise capture molecules, optionally coupled to a support.
  • a composition may be a part of or comprise a reaction mixture, which can include reaction components or reagents, e.g., enzymes, nucleotide bases, catalysts, etc.
  • the systems and methods described herein provide for the compartmentalization, depositing, or partitioning of one or more particles (e.g., biological particles, macromolecular constituents of biological particles, beads, reagents, etc.) into discrete compartments or partitions (referred to interchangeably herein as partitions), where each partition maintains separation of its own contents from the contents of other partitions.
  • the partition can be a droplet in an emulsion or a well.
  • a partition may comprise one or more other partitions.
  • a partition may include one or more particles.
  • a partition may include one or more types of particles.
  • a partition of the present disclosure may comprise one or more biological particles and/or macromolecular constituents thereof.
  • a partition may comprise one or more beads.
  • a partition may comprise one or more gel beads.
  • a partition may comprise one or more cell beads.
  • a partition may include a single gel bead, a single cell bead, or both a single cell bead and single gel bead.
  • a partition may include one or more reagents.
  • a partition may be unoccupied.
  • a partition may not comprise a bead.
  • a cell bead can be a biological particle and/or one or more of its macromolecular constituents encased inside of a gel or polymer matrix, such as via polymerization of a droplet containing the biological particle and precursors capable of being polymerized or gelled.
  • Unique identifiers such as barcodes, may be injected into the droplets previous to, subsequent to, or concurrently with droplet generation, such as via a support (e.g., bead), as described elsewhere herein.
  • the methods and systems of the present disclosure may comprise methods and systems for generating one or more partitions such as droplets.
  • the droplets may comprise a plurality of droplets in an emulsion.
  • the droplets may comprise droplets in a colloid.
  • the emulsion may comprise a microemulsion or a nanoemulsion.
  • the droplets may be generated with aid of a microfluidic device and/or by subjecting a mixture of immiscible phases to agitation (e.g., in a container).
  • a combination of the mentioned methods may be used for droplet and/or emulsion formation.
  • Droplets can be formed by creating an emulsion by mixing and/or agitating immiscible phases.
  • Mixing or agitation may comprise various agitation techniques, such as vortexing, pipetting, tube flicking, or other agitation techniques.
  • mixing or agitation may be performed without using a microfluidic device.
  • the droplets may be formed by exposing a mixture to ultrasound or sonication.
  • Microfluidic devices or platforms comprising microfluidic channel networks can be utilized to generate partitions such as droplets and/or emulsions as described herein.
  • Methods and systems for generating partitions such as droplets, methods of encapsulating biological particle methods of increasing the throughput of droplet generation, and various geometries, architectures, and configurations of microfluidic devices and channels are described in U.S. Patent Publication Nos. 2019/0367997 and 2019/0064173, each of which is entirely incorporated herein by reference for all purposes.
  • individual particles can be partitioned to discrete partitions by introducing a flowing stream of particles in an aqueous fluid into a flowing stream or reservoir of a non-aqueous fluid, such that droplets may be generated at the junction of the two streams/reservoir, such as at the junction of a microfluidic device provided elsewhere herein.
  • the methods of the present disclosure may comprise generating partitions and/or encapsulating particles, such as biological particles, in some cases, individual biological particles such as single cells, nuclei or cell beads.
  • reagents may be encapsulated and/or partitioned (e.g., co-partitioned with biological particles) in the partitions.
  • Various mechanisms may be employed in the partitioning of individual particles.
  • An example may comprise porous membranes through which aqueous mixtures of cells may be extruded into fluids (e.g., nonaqueous fluids).
  • the partitions can be flowable within fluid streams.
  • the partitions may comprise, for example, micro-vesicles that have an outer barrier surrounding an inner fluid center or core.
  • the partitions may comprise a porous matrix that is capable of entraining and/or retaining materials within its matrix.
  • the partitions can be droplets of a first phase within a second phase, wherein the first and second phases are immiscible.
  • the partitions can be droplets of aqueous fluid within a non-aqueous continuous phase (e.g., oil phase).
  • the partitions can be droplets of a non-aqueous fluid within an aqueous phase.
  • the partitions may be provided in a water-in-oil emulsion or oil-in-water emulsion.
  • a variety of different vessels are described in, for example, U.S. Patent Application Publication No. 2014/0155295, which is entirely incorporated herein by reference for all purposes.
  • Emulsion systems for creating stable droplets in non-aqueous or oil continuous phases are described in, for example, U.S. Patent Application Publication No. 2010/0105112, which is entirely incorporated herein by reference for all purposes.
  • Fluid properties e.g., fluid flow rates, fluid viscosities, etc
  • particle properties e.g., volume fraction, particle size, particle concentration, etc.
  • microfluidic architectures e.g., channel geometry, etc.
  • partition occupancy can be controlled by providing the aqueous stream at a certain concentration and/or flow rate of particles.
  • the relative flow rates of the immiscible fluids can be selected such that, on average, the partitions may contain less than one biological particle per partition in order to ensure that those partitions that are occupied are primarily singly occupied.
  • partitions among a plurality of partitions may contain at most one biological particle (e g., bead, DNA, cell or cellular material).
  • the various parameters e.g., fluid properties, particle properties, microfluidic architectures, etc.
  • the flows and channel architectures can be controlled as to ensure a given number of singly occupied partitions, less than a certain level of unoccupied partitions and/or less than a certain level of multiply occupied partitions.
  • FIG. 1 shows an example of a microfluidic channel structure 100 for partitioning individual biological particles.
  • the channel structure 100 can include channel segments 102, 104, 106 and 108 communicating at a channel junction 110.
  • a first aqueous fluid 112 that includes suspended biological particles (or cells) 114 may be transported along channel segment 102 into junction 110, while a second fluid 116 that is immiscible with the aqueous fluid 112 is delivered to the junction 110 from each of channel segments 104 and 106 to create discrete droplets 118, 120 of the first aqueous fluid 112 flowing into channel segment 108, and flowing away from junction 110.
  • the channel segment 108 may be fluidically coupled to an outlet reservoir where the discrete droplets can be stored and/or harvested.
  • a discrete droplet generated may include an individual biological particle 114 (such as droplets 118).
  • a discrete droplet generated may include more than one individual biological particle 114 (not shown in FIG. 1).
  • a discrete droplet may contain no biological particle 114 (such as droplet 120).
  • Each discrete partition may maintain separation of its own contents (e.g., individual biological particle 114) from the contents of other partitions.
  • the second fluid 116 can comprise an oil, such as a fluorinated oil, that includes a fluorosurfactant for stabilizing the resulting droplets, for example, inhibiting subsequent coalescence of the resulting droplets 118, 120.
  • an oil such as a fluorinated oil
  • fluorosurfactant for stabilizing the resulting droplets, for example, inhibiting subsequent coalescence of the resulting droplets 118, 120.
  • the channel segments described herein may be coupled to any of a variety of different fluid sources or receiving components, including reservoirs, tubing, manifolds, or fluidic components of other systems.
  • the microfluidic channel structure 100 may have other geometries.
  • a microfluidic channel structure can have more than one channel junction.
  • a microfluidic channel structure can have 2, 3, 4, or 5 channel segments each carrying particles (e g., biological particles, cell beads, and/or gel beads) that meet at a channel junction. Fluid may be directed to flow along one or more channels or reservoirs via one or more fluid flow units.
  • a fluid flow unit can comprise compressors (e.g., providing positive pressure), pumps (e.g., providing negative pressure), actuators, and the like to control flow of the fluid. Fluid may also or otherwise be controlled via applied pressure differentials, centrifugal force, electrokinetic pumping, vacuum, capillary or gravity flow, or the like.
  • the generated droplets may comprise two subsets of droplets: (1) occupied droplets 118, containing one or more biological particles 114, and (2) unoccupied droplets 120, not containing any biological particles 114.
  • Occupied droplets 118 may comprise singly occupied droplets (having one biological particle) and multiply occupied droplets (having more than one biological particle).
  • the majority of occupied partitions can include no more than one biological particle per occupied partition and some of the generated partitions can be unoccupied (of any biological particle). In some cases, though, some of the occupied partitions may include more than one biological particle.
  • the partitioning process may be controlled such that fewer than about 25% of the occupied partitions contain more than one biological particle, and in many cases, fewer than about 20% of the occupied partitions have more than one biological particle, while in some cases, fewer than about 10% or even fewer than about 5% of the occupied partitions include more than one biological particle per partition.
  • the Poissonian distribution may expectedly increase the number of partitions that include multiple biological particles. As such, where singly occupied partitions are to be obtained, at most about 95%, 90%, 85%, 80%, 75%, 70%, 65%, 60%, 55%, 50%, 45%, 40%, 35%, 30%, 25%, 20%, 15%, 10%, 5% or less of the generated partitions can be unoccupied.
  • the flow of one or more of the biological particles (e.g., in channel segment 102), or other fluids directed into the partitioning junction (e.g., in channel segments 104, 106) can be controlled such that, in many cases, no more than about 50% of the generated partitions, no more than about 25% of the generated partitions, or no more than about 10% of the generated partitions are unoccupied.
  • These flows can be controlled so as to present a non- Poissonian distribution of single-occupied partitions while providing lower levels of unoccupied partitions.
  • the above noted ranges of unoccupied partitions can be achieved while still providing any of the single occupancy rates described above.
  • the use of the systems and methods described herein can create resulting partitions that have multiple occupancy rates of less than about 25%, less than about 20%, less than about 15%, less than about 10%, and in many cases, less than about 5%, while having unoccupied partitions of less than about 50%, less than about 40%, less than about 30%, less than about 20%, less than about 10%, less than about 5%, or less.
  • the above-described occupancy rates are also applicable to partitions that include both biological particles and additional reagents, including, but not limited to, supports such as beads (e.g., gel beads) carrying barcoded nucleic acid molecules (e.g., oligonucleotides) (described in relation to FIG. 2).
  • the occupied partitions e.g., at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99% of the occupied partitions
  • a support e.g., bead
  • biological particles may be encapsulated within a support that comprises an outer shell, layer or porous matrix in which is entrained one or more individual biological particles or small groups of biological particles.
  • the support may include other reagents.
  • Encapsulation of biological particles may be performed by a variety of processes. Such processes may combine an aqueous fluid containing the biological particles with a polymeric precursor material that may be capable of being formed into a gel or other solid or semi-solid matrix upon application of a particular stimulus to the polymer precursor.
  • Such stimuli can include, for example, thermal stimuli (e.g., either heating or cooling), photo-stimuli (e.g., through photo-curing), chemical stimuli (e g., through crosslinking, polymerization initiation of the precursor (e.g., through added initiators)), mechanical stimuli, or a combination thereof.
  • thermal stimuli e.g., either heating or cooling
  • photo-stimuli e.g., through photo-curing
  • chemical stimuli e.g., through crosslinking, polymerization initiation of the precursor (e.g., through added initiators)
  • mechanical stimuli e.g., mechanical stimuli, or a combination thereof.
  • Preparation of supports comprising biological particles may be performed by a variety of methods.
  • air knife droplet or aerosol generators may be used to dispense droplets of precursor fluids into gelling solutions in order to form beads (e.g., gel beads) that include individual biological particles or small groups of biological particles.
  • membrane-based encapsulation systems may be used to generate beads comprising encapsulated biological particles as described herein.
  • Microfluidic systems of the present disclosure such as that shown in FIG. 1, may be readily used in encapsulating biological particles (e.g., cells) as described herein.
  • non-aqueous fluid 116 may also include an initiator (not shown) to cause polymerization and/or crosslinking of the polymer precursor to form the porous matrix that includes the entrained biological particles.
  • polymer precursor/initiator pairs examples include those described in U.S. Patent Application Publication No. 2014/0378345, which is entirely incorporated herein by reference for all purposes.
  • encapsulated biological particles can be selectively releasable from the support, such as through passage of time or upon application of a particular stimulus, that degrades the microcapsule sufficiently to allow the biological particles (e.g., cell), or its other contents to be released from the support, such as into a partition (e.g., droplet).
  • a partition e.g., droplet
  • the biological particle can be subjected to other conditions sufficient to polymerize or gel the precursors.
  • the conditions sufficient to polymerize or gel the precursors may comprise exposure to heating, cooling, electromagnetic radiation, and/or light.
  • the conditions sufficient to polymerize or gel the precursors may comprise any conditions sufficient to polymerize or gel the precursors.
  • a polymer or gel may be formed around the biological particle.
  • the polymer or gel may be diffusively permeable to chemical or biochemical reagents.
  • the polymer or gel may be diffusively impermeable to macromolecular constituents of the biological particle.
  • the polymer or gel may act to allow the biological particle to be subjected to chemical or biochemical operations while spatially confining the macromolecular constituents to a region of the droplet defined by the polymer or gel.
  • the polymer or gel may include one or more of disulfide cross-linked polyacrylamide, agarose, alginate, polyvinyl alcohol, polyethylene glycol (PEG)-diacrylate, PEG-acrylate, PEG-thiol, PEG-azide, PEG-alkyne, other acrylates, chitosan, hyaluronic acid, collagen, fibrin, gelatin, or elastin.
  • the polymer or gel may comprise any other polymer or gel.
  • the polymer or gel may be functionalized to bind to targeted analytes, such as nucleic acids, proteins, carbohydrates, lipids or other analytes.
  • the polymer or gel may be polymerized or gelled via a passive mechanism.
  • the polymer or gel may be stable in alkaline conditions or at elevated temperature.
  • the polymer or gel may have mechanical properties similar to the mechanical properties of the bead. For instance, the polymer or gel may be of a similar size to the bead.
  • the polymer or gel may have a mechanical strength (e.g. tensile strength) similar to that of the bead.
  • the polymer or gel may be of a lower density than an oil.
  • the polymer or gel may be of a density that is roughly similar to that of a buffer.
  • the polymer or gel may have a tunable pore size.
  • the pore size may be chosen to, for instance, retain denatured nucleic acids.
  • the pore size may be chosen to maintain diffusive permeability to exogenous chemicals such as sodium hydroxide (NaOH) and/or endogenous chemicals such as inhibitors.
  • the polymer or gel may be biocompatible.
  • the polymer or gel may maintain or enhance cell viability.
  • the polymer or gel may be biochemically compatible.
  • the polymer or gel may be polymerized and/or depolymerized thermally, chemically, enzymatically, and/or optically.
  • the polymer may comprise poly(acrylamide-co-acrylic acid) crosslinked with disulfide linkages.
  • the preparation of the polymer may comprise a two-step reaction.
  • poly(acrylamide-co-acrylic acid) may be exposed to an acylating agent to convert carboxylic acids to esters.
  • the poly(acrylamide-co-acrylic acid) may be exposed to 4-(4,6-dimethoxy-l,3,5-triazin-2-yl)-4-methylmorpholinium chloride (DMTMM).
  • DTMM 4-(4,6-dimethoxy-l,3,5-triazin-2-yl)-4-methylmorpholinium chloride
  • the polyacrylamide-co-acrylic acid may be exposed to other salts of 4-(4,6-dimethoxy-l,3,5- triazin-2-yl)-4-methylmorpholinium.
  • the ester formed in the first step may be exposed to a disulfide crosslinking agent.
  • the ester may be exposed to cystamine (2,2’-dithiobis(ethylamine)).
  • the biological particle may be surrounded by polyacrylamide strands linked together by disulfide bridges. In this manner, the biological particle may be encased inside of or comprise a gel or matrix (e.g., polymer matrix) to form a “cell bead.”
  • a cell bead can contain biological particles (e.g., a cell) or macromolecular constituents (e.g., RNA, DNA, proteins, etc.) of biological particles.
  • a cell bead may include a single cell or multiple cells, or a derivative of the single cell or multiple cells. For example after lysing and washing the cells, inhibitory components from cell lysates can be washed away and the macromolecular constituents can be bound as cell beads.
  • Systems and methods disclosed herein can be applicable to both cell beads (and/or droplets or other partitions) containing biological particles and cell beads (and/or droplets or other partitions) containing macromolecular constituents of biological particles.
  • Cell beads may be or include a cell, cell derivative, cellular material and/or material derived from the cell in, within, or encased in a matrix, such as a polymeric matrix.
  • a cell bead may comprise a live cell.
  • the live cell may be capable of being cultured when enclosed in a gel or polymer matrix, or of being cultured when comprising a gel or polymer matrix.
  • the polymer or gel may be diffusively permeable to certain components and diffusively impermeable to other components (e.g., macromolecular constituents).
  • a partition which may be a well.
  • the well may be a well of a plurality of wells of a substrate, such as a microwell of a microwell array or plate, or the well may be a microwell or microchamber of a device (e.g., microfluidic device) comprising a substrate.
  • the well may be a well of a well array or plate, or the well may be a well or chamber of a device (e.g., fluidic device).
  • the wells or microwells may assume an “open” configuration, in which the wells or microwells are exposed to the environment (e.g., contain an open surface) and are accessible on one planar face of the substrate, or the wells or microwells may assume a “closed” or “sealed” configuration, in which the microwells are not accessible on a planar face of the substrate.
  • the wells or microwells may be configured to toggle between “open” and “closed” configurations.
  • an “open” microwell or set of microwells may be “closed” or “sealed” using a membrane (e.g., semi-permeable membrane), an oil (e.g., fluorinated oil to cover an aqueous solution), or a lid, as described elsewhere herein.
  • a membrane e.g., semi-permeable membrane
  • an oil e.g., fluorinated oil to cover an aqueous solution
  • a lid e.g., a lid
  • the well may have a volume of less than 1 milliliter (mL).
  • the well may be configured to hold a volume of at most 1000 microliters (pL), at most 100 pL, at most 10 pL, at most 1 pL, at most 100 nanoliters (nL), at most 10 nL, at most 1 nL, at most 100 picoliters (pL), at most 10 (pL), or less.
  • the well may be configured to hold a volume of about 1000 pL, about 100 pL, about 10 pL, about 1 pL, about 100 nL, about 10 nL, about 1 nL, about 100 pL, about 10 pL, etc.
  • the well may be configured to hold a volume of at least 10 pL, at least 100 pL, at least 1 nL, at least 10 nL, at least 100 nL, at least 1 pL, at least 10 pL, at least 100 pL, at least 1000 pL, or more.
  • the well may be configured to hold a volume in a range of volumes listed herein, for example, from about 5 nL to about 20 nL, from about 1 nL to about 100 nL, from about 500 pL to about 100 pL, etc.
  • the well may be of a plurality of wells that have varying volumes and may be configured to hold a volume appropriate to accommodate any of the partition volumes described herein.
  • a microwell array or plate comprises a single variety of microwells.
  • a microwell array or plate comprises a variety of microwells.
  • the microwell array or plate may comprise one or more types of microwells within a single microwell array or plate.
  • the types of microwells may have different dimensions (e.g., length, width, diameter, depth, cross-sectional area, etc.), shapes (e.g., circular, triangular, square, rectangular, pentagonal, hexagonal, heptagonal, octagonal, nonagonal, decagonal, etc.), aspect ratios, or other physical characteristics.
  • the microwell array or plate may comprise any number of different types of microwells.
  • the microwell array or plate may comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000 or more different types of microwells.
  • a well may have any dimension (e g., length, width, diameter, depth, cross-sectional area, volume, etc.), shape (e.g., circular, triangular, square, rectangular, pentagonal, hexagonal, heptagonal, octagonal, nonagonal, decagonal, other polygonal, etc.), aspect ratios, or other physical characteristics described herein with respect to any well.
  • the microwell array or plate comprises different types of microwells that are located adjacent to one another within the array or plate. For instance, a microwell with one set of dimensions may be located adjacent to and in contact with another microwell with a different set of dimensions. Similarly, microwells of different geometries may be placed adjacent to or in contact with one another.
  • the adjacent microwells may be configured to hold different articles; for example, one microwell may be used to contain a cell, cell bead, or other sample (e.g., cellular components, nucleic acid molecules, etc.) while the adjacent microwell may be used to contain a support (e.g., a bead such as a gel bead), droplet, or other reagent.
  • the adjacent microwells may be configured to merge the contents held within, e.g., upon application of a stimulus, or spontaneously, upon contact of the articles in each microwell.
  • a plurality of partitions may be used in the systems, compositions, and methods described herein.
  • any suitable number of partitions e g., wells or droplets
  • wells at least about 1,000 wells, at least about 5,000 wells, at least about 10,000 wells, at least about 50,000 wells, at least about 100,000 wells, at least about 500,000 wells, at least about 1,000,000 wells, at least about 5,000,000 wells at least about 10,000,000 wells, at least about 50,000,000 wells, at least about 100,000,000 wells, at least about 500,000,000 wells, at least about 1,000,000,000 wells, or more wells can be generated or otherwise provided.
  • the plurality of wells may comprise both unoccupied wells (e.g., empty wells) and occupied wells.
  • a well may comprise any of the reagents described herein, or combinations thereof. These reagents may include, for example, barcode molecules, enzymes, adapters, and combinations thereof.
  • the reagents may be physically separated from a sample (e.g., a cell, cell bead, or cellular components, e.g., proteins, nucleic acid molecules, etc.) that is placed in the well. This physical separation may be accomplished by containing the reagents within, or coupling to, a support (e.g., a bead such as a gel bead) that is placed within a well.
  • a support e.g., a bead such as a gel bead
  • the physical separation may also be accomplished by dispensing the reagents in the well and overlaying the reagents with a layer that is, for example, dissolvable, meltable, or permeable prior to introducing the polynucleotide sample into the well.
  • This layer may be, for example, an oil, wax, membrane (e.g., semi-permeable membrane), or the like.
  • the well may be sealed at any point, for example, after addition of the support (e.g., bead), after addition of the reagents, or after addition of either of these components.
  • the sealing of the well may be useful for a variety of purposes, including preventing escape of beads or loaded reagents from the well, permitting select delivery of certain reagents (e.g., via the use of a semi-permeable membrane), for storage of the well prior to or following further processing, etc.
  • a well may comprise free reagents and/or reagents encapsulated in, or otherwise coupled to or associated with, supports (e.g., beads) or droplets. Any of the reagents described in this disclosure may be encapsulated in, or otherwise coupled to, a support (e.g., bead) or droplet, with any chemicals, particles, and elements suitable for sample processing reactions involving biomolecules, such as, but not limited to, nucleic acid molecules and proteins.
  • a bead or droplet used in a sample preparation reaction for DNA sequencing may comprise one or more of the following reagents: enzymes, restriction enzymes (e.g., multiple cutters), ligase, polymerase, fluorophores, oligonucleotide barcodes, adapters, buffers, nucleotides (e.g., dNTPs, ddNTPs) and the like.
  • reagents include, but are not limited to: buffers, acidic solution, basic solution, temperature-sensitive enzymes, pH-sensitive enzymes, light-sensitive enzymes, metals, metal ions, magnesium chloride, sodium chloride, manganese, aqueous buffer, mild buffer, ionic buffer, inhibitor, enzyme, protein, polynucleotide, antibodies, saccharides, lipid, oil, salt, ion, detergents, ionic detergents, non-ionic detergents, oligonucleotides, nucleotides, deoxyribonucleotide triphosphates (dNTPs), dideoxyribonucleotide triphosphates (ddNTPs), DNA, RNA, peptide polynucleotides, complementary DNA (cDNA), double stranded DNA (dsDNA), single stranded DNA (ssDNA), plasmid DNA, cosmid DNA, chromosomal DNA, genomic DNA, viral DNA, bacterial DNA,
  • one or more reagents in the well may be used to perform one or more reactions, including but not limited to: cell lysis, cell fixation, permeabilization, nucleic acid reactions, e.g., nucleic acid extension reactions, amplification, reverse transcription, transposase reactions (e.g., tagmentation), etc.
  • kits may comprise instructions for use, a microwell array or device, and reagents (e.g., beads).
  • the kit may comprise any useful reagents for performing the processes described herein, e.g., nucleic acid reactions, barcoding of nucleic acid molecules, sample processing (e.g., for cell lysis, fixation, and/or permeabilization).
  • a well comprises a support (e g., a bead), or droplet that comprises a set of reagents that has a similar attribute (e.g., a set of enzymes, a set of minerals, a set of oligonucleotides, a mixture of different barcode molecules, a mixture of identical barcode molecules).
  • a support or droplet comprises a heterogeneous mixture of reagents.
  • the heterogeneous mixture of reagents can comprise all components necessary to perform a reaction.
  • such mixture can comprise all components necessary to perform a reaction, except for 1, 2, 3, 4, 5, or more components necessary to perform a reaction.
  • such additional components are contained within, or otherwise coupled to, a different support or droplet, or within a solution within a partition (e.g., microwell) of the system.
  • FIG. 5 schematically illustrates an example of a microwell array.
  • the array can be contained within a substrate 500.
  • the substrate 500 comprises a plurality of wells 502.
  • the wells 502 may be of any size or shape, and the spacing between the wells, the number of wells per substrate, as well as the density of the wells on the substrate 500 can be modified, depending on the particular application.
  • a sample molecule 506 which may comprise a cell or cellular components (e.g., nucleic acid molecules) is co-partitioned with a bead 504, which may comprise a nucleic acid barcode molecule coupled thereto.
  • the wells 502 may be loaded using gravity or other loading technique (e.g., centrifugation, liquid handler, acoustic loading, optoelectronic, etc.). In some instances, at least one of the wells 502 contains a single sample molecule 506 (e.g., cell) and a single bead 504.
  • gravity or other loading technique e.g., centrifugation, liquid handler, acoustic loading, optoelectronic, etc.
  • at least one of the wells 502 contains a single sample molecule 506 (e.g., cell) and a single bead 504.
  • Reagents may be loaded into a well either sequentially or concurrently.
  • reagents are introduced to the device either before or after a particular operation.
  • reagents (which may be provided, in certain instances, in supports or droplets) are introduced sequentially such that different reactions or operations occur at different steps.
  • the reagents (or supports or droplets) may also be loaded at operations interspersed with a reaction or operation step.
  • supports comprising reagents for fragmenting polynucleotides (e.g., restriction enzymes) and/or other enzymes (e.g., transposases, ligases, polymerases, etc.) may be loaded into the well or plurality of wells, followed by loading of supports or droplets comprising reagents for attaching nucleic acid barcode molecules to a sample nucleic acid molecule.
  • Reagents may be provided concurrently or sequentially with a sample, e.g., a cell or cellular components (e.g., organelles, proteins, nucleic acid molecules, carbohydrates, lipids, etc.). Accordingly, use of wells may be useful in performing multi-step operations or reactions.
  • the nucleic acid barcode molecules and other reagents may be contained within a support (e.g., a bead), or droplet. These supports, or droplets may be loaded into a partition (e.g., a microwell) before, after, or concurrently with the loading of a cell, such that each cell is contacted with a different support or droplet.
  • This technique may be used to attach a unique nucleic acid barcode molecule to nucleic acid molecules obtained from each cell.
  • the sample nucleic acid molecules may be attached to a support.
  • the partition (e.g., microwell) may comprise a bead which has coupled thereto a plurality of nucleic acid barcode molecules.
  • the sample nucleic acid molecules, or derivatives thereof, may couple or attach to the nucleic acid barcode molecules on the support.
  • the resulting barcoded nucleic acid molecules may then be removed from the partition, and in some instances, pooled and sequenced.
  • the nucleic acid barcode sequences may be used to trace the origin of the sample nucleic acid molecule.
  • polynucleotides with identical barcodes may be determined to originate from the same cell or partition, while polynucleotides with different barcodes may be determined to originate from different cells, nuclei, cell beads, or partitions.
  • the samples or reagents may be loaded in the wells or microwells using a variety of approaches.
  • the samples e.g., a cell, cell bead, or cellular component
  • reagents as described herein
  • the samples may be loaded into the well or microwell using an external force, e.g., gravitational force, electrical force, magnetic force, or using mechanisms to drive the sample or reagents into the well, e.g., via pressure-driven flow, centrifugation, optoelectronics, acoustic loading, electrokinetic pumping, vacuum, capillary flow, etc.
  • a fluid handling system may be used to load the samples or reagents into the well.
  • the loading of the samples or reagents may follow a Poissonian distribution or a non-Poissonian distribution, e.g., super Poisson or subPoisson.
  • the geometry, spacing between wells, density, and size of the microwells may be modified to accommodate a useful sample or reagent distribution; for instance, the size and spacing of the microwells may be adjusted such that the sample or reagents may be distributed in a super-Poissonian fashion.
  • the microwell array or plate comprises pairs of microwells, in which each pair of microwells is configured to hold a droplet (e.g., comprising a single cell) and a single bead (such as those described herein, which may, in some instances, also be encapsulated in a droplet).
  • the droplet and the bead (or droplet containing the bead) may be loaded simultaneously or sequentially, and the droplet and the bead may be merged, e.g., upon contact of the droplet and the bead, or upon application of a stimulus (e.g., external force, agitation, heat, light, magnetic or electric force, etc.).
  • a stimulus e.g., external force, agitation, heat, light, magnetic or electric force, etc.
  • the loading of the droplet and the bead is super-Poissonian.
  • the wells are configured to hold two droplets comprising different reagents and/or samples, which are merged upon contact or upon application of a stimulus.
  • the droplet of one microwell of the pair can comprise reagents that may react with an agent in the droplet of the other microwell of the pair.
  • one droplet can comprise reagents that are configured to release the nucleic acid barcode molecules of a bead contained in another droplet, located in the adjacent microwell.
  • the nucleic acid barcode molecules may be released from the bead into the partition (e.g., the microwell or microwell pair that are in contact), and further processing may be performed (e.g., barcoding, nucleic acid reactions, etc.).
  • the partition e.g., the microwell or microwell pair that are in contact
  • further processing e.g., barcoding, nucleic acid reactions, etc.
  • one of the droplets may comprise lysis reagents for lysing the cell upon droplet merging.
  • a droplet or support (e.g., a bead) may be partitioned into a well.
  • the droplets may be selected or subjected to pre-processing prior to loading into a well.
  • the droplets may comprise cells, and only certain droplets, such as those containing a single cell (or at least one cell), may be selected for use in loading of the wells.
  • Such a pre-selection process may be useful in efficient loading of single cells, such as to obtain a non-Poissonian distribution, or to pre-filter cells for a selected characteristic prior to further partitioning in the wells.
  • the technique may be useful in obtaining or preventing cell doublet or multiplet formation prior to or during loading of the microwell.
  • the wells can comprise nucleic acid barcode molecules attached thereto.
  • the nucleic acid barcode molecules may be attached to a surface of the well (e g., a wall of the well).
  • the nucleic acid barcode molecule (e.g., a partition barcode sequence) of one well may differ from the nucleic acid barcode molecule of another well, which can permit identification of the contents contained with a single partition or well.
  • the nucleic acid barcode molecule can comprise a spatial barcode sequence that can identify a spatial coordinate of a well, such as within the well array or well plate.
  • the nucleic acid barcode molecule can comprise a unique molecular identifier for individual molecule identification.
  • the nucleic acid barcode molecules may be configured to attach to or capture a nucleic acid molecule within a sample or cell distributed in the well.
  • the nucleic acid barcode molecules may comprise a capture sequence that may be used to capture or hybridize to a nucleic acid molecule (e.g., RNA, DNA) within the sample.
  • the nucleic acid barcode molecules may be releasable from the microwell.
  • the nucleic acid barcode molecules may comprise a chemical cross-linker which may be cleaved upon application of a stimulus (e.g., photo-, magnetic, chemical, biological, stimulus).
  • the released nucleic acid barcode molecules which may be hybridized or configured to hybridize to a sample nucleic acid molecule, may be collected and pooled for further processing, which can include nucleic acid processing (e.g., amplification, extension, reverse transcription, etc.) and/or characterization (e.g., sequencing).
  • nucleic acid processing e.g., amplification, extension, reverse transcription, etc.
  • characterization e.g., sequencing
  • the unique partition barcode sequences may be used to identify the cell or partition from which a nucleic acid molecule originated.
  • Characterization of samples within a well may be performed. Such characterization can include, in non-limiting examples, imaging of the sample (e.g., cell, cell bead, or cellular components) or derivatives thereof. Characterization techniques such as microscopy or imaging may be useful in measuring sample profiles in fixed spatial locations.
  • imaging of each microwell and the contents contained therein may provide useful information on cell doublet formation (e.g., frequency, spatial locations, etc.), cell-bead pair efficiency, cell viability, cell size, cell morphology, expression level of a biomarker (e.g., a surface marker, a fluorescently labeled molecule therein, etc ), cell or bead loading rate, number of cell-bead pairs, etc.
  • a biomarker e.g., a surface marker, a fluorescently labeled molecule therein, etc
  • imaging may be used to characterize live cells in the wells, including, but not limited to: dynamic live-cell tracking, cell-cell interactions (when two or more cells are co-partitioned), cell proliferation, etc.
  • imaging may be used to characterize a quantity of amplification products in the well.
  • a well may be loaded with a sample and reagents, simultaneously or sequentially.
  • the well may be subjected to washing, e.g., to remove excess cells (or nuclei or cell beads) from the well, microwell array, or plate. Similarly, washing may be performed to remove excess beads or other reagents from the well, microwell array, or plate.
  • the cells may be lysed in the individual partitions to release the intracellular components or cellular analytes. Alternatively, the cells may be fixed or permeabilized in the individual partitions.
  • the intracellular components or cellular analytes may couple to a support, e.g., on a surface of the microwell, on a solid support (e.g., bead), or they may be collected for further downstream processing. For instance, after cell lysis, the intracellular components or cellular analytes may be transferred to individual droplets or other partitions for barcoding.
  • the intracellular components or cellular analytes may couple to a bead comprising a nucleic acid barcode molecule; subsequently, the bead may be collected and further processed, e.g., subjected to nucleic acid reaction such as reverse transcription, amplification, or extension, and the nucleic acid molecules thereon may be further characterized, e.g., via sequencing.
  • the intracellular components or cellular analytes may be barcoded in the well (e.g., using a bead comprising nucleic acid barcode molecules that are releasable or on a surface of the microwell comprising nucleic acid barcode molecules).
  • the barcoded nucleic acid molecules or analytes may be further processed in the well, or the barcoded nucleic acid molecules or analytes may be collected from the individual partitions and subjected to further processing outside the partition. Further processing can include nucleic acid processing (e.g., performing an amplification, extension) or characterization (e.g., fluorescence monitoring of amplified molecules, sequencing).
  • the well or microwell array or plate
  • the well may be sealed (e.g., using an oil, membrane, wax, etc.), which enables storage of the assay or selective introduction of additional reagents.
  • the well may be subjected to conditions for further processing of a biological particle (e.g., a cell, a cell bead or a nucleus) in the well.
  • a biological particle e.g., a cell, a cell bead or a nucleus
  • reagents in the well may allow further processing of the biological particle, e.g., lysis of the cell or nucleus, as further described herein.
  • the well (or wells such as those of a well-based array) comprising the biological particle may be subjected to freezethaw cycling to process the biological particle(s), e.g., lysis of a cell or nucleus.
  • the well containing the biological particle may be subjected to freezing temperatures (e.g., 0°C, below 0°C, -5°C, -10°C, -15°C, -20°C, -25°C, -30°C, -35°C, -40°C, - 45°C, -50°C, -55°C, -60°C, -65°C, -70°C, -80°C, or -85°C). Freezing may be performed in a suitable manner, e.g., sub-zero freezer or a dry ice/ethanol bath.
  • freezing temperatures e.g., 0°C, below 0°C, -5°C, -10°C, -15°C, -20°C, -25°C, -30°C, -35°C, -40°C, - 45°C, -50°C, -55°C, -60°C, -65°C, -70°C, -80°C,
  • the well (or wells) comprising the biological particle(s) may be subjected to freeze thaw cycles to lyse biological parti cle(s).
  • the initially frozen well (or wells) are thawed to a temperature above freezing (e.g., room temperature or 25°C).
  • the freezing is performed for less than 10 minutes (e g., 5 minutes or 7 minutes) followed by thawing at room temperature for less than 10 minutes (e g., 5 minutes or 7 minutes).
  • This freeze-thaw cycle may be repeated a number of times, e.g., 2, 3, or 4 times, to obtain lysis of the biological particle(s) (e.g., cell(s), cell bead(s), nucleus, or nuclei) in the well (or wells).
  • the freezing, thawing and/or freeze/thaw cycling is performed in the absence of a lysis buffer.
  • FIG. 6 schematically shows an example workflow for processing nucleic acid molecules within a sample.
  • a substrate 600 comprising a plurality of microwells 602 may be provided.
  • a sample 606 which may comprise a cell, cell bead, cellular components or analytes (e g., proteins and/or nucleic acid molecules) can be co-partitioned, in a plurality of microwells 602, with a plurality of beads 604 comprising nucleic acid barcode molecules.
  • the sample 606 may be processed within the partition. For instance, in the case of live cells, the cell may be subjected to conditions sufficient to lyse the cells or nuclei and release the analytes contained therein.
  • the bead 604 may be further processed.
  • processes 620a and 620b schematically illustrate different workflows, depending on the properties of the bead 604.
  • the bead comprises nucleic acid barcode molecules that are attached thereto, and sample nucleic acid molecules (e.g., RNA, DNA) may attach, e.g., via hybridization of ligation, to the nucleic acid barcode molecules. Such attachment may occur on the bead.
  • sample nucleic acid molecules e.g., RNA, DNA
  • the beads 604 from multiple wells 602 may be collected and pooled. Further processing may be performed in process 640.
  • one or more nucleic acid reactions may be performed, such as reverse transcription, nucleic acid extension, amplification, ligation, transposition, etc.
  • adapter sequences are ligated to the nucleic acid molecules, or derivatives thereof, as described elsewhere herein.
  • sequencing primer sequences may be appended to each end of the nucleic acid molecule.
  • further characterization such as sequencing may be performed to generate sequencing reads.
  • the sequencing reads may yield information on individual cells or populations of cells, which may be represented visually or graphically, e.g., in a plot 655.
  • the bead comprises nucleic acid barcode molecules that are releasably attached thereto, as described below.
  • the bead may degrade or otherwise release the nucleic acid barcode molecules into the well 602; the nucleic acid barcode molecules may then be used to barcode nucleic acid molecules within the well 602. Further processing may be performed either inside the partition or outside the partition. For example, one or more nucleic acid reactions may be performed, such as reverse transcription, nucleic acid extension, amplification, ligation, transposition, etc. In some instances, adapter sequences are ligated to the nucleic acid molecules, or derivatives thereof, as described elsewhere herein.
  • sequencing primer sequences may be appended to each end of the nucleic acid molecule.
  • further characterization such as sequencing may be performed to generate sequencing reads.
  • the sequencing reads may yield information on individual cells or populations of cells, which may be represented visually or graphically, e.g., in a plot 655.
  • Nucleic acid barcode molecules may be delivered to a partition (e g., a droplet or well) via a solid support or carrier (e.g., a bead).
  • a solid support or carrier e.g., a bead
  • nucleic acid barcode molecules are initially associated with the solid support and then released from the solid support upon application of a stimulus, which allows the nucleic acid barcode molecules to dissociate or to be released from the solid support.
  • nucleic acid barcode molecules are initially associated with the solid support (e.g., bead) and then released from the solid support upon application of a biological stimulus, a chemical stimulus, a thermal stimulus, an electrical stimulus, a magnetic stimulus, and/or a photo stimulus.
  • a nucleic acid barcode molecule may contain a barcode sequence and a functional sequence, such as a nucleic acid primer sequence or a template switch oligonucleotide (TSO) sequence.
  • TSO template switch oligonucleotide
  • the solid support may be a bead.
  • a solid support e.g., a bead, may be porous, non- porous, hollow (e.g., a microcapsule), solid, semi-solid, and/or a combination thereof.
  • Beads may be solid, semi-solid, semi-fluidic, fluidic, and/or a combination thereof.
  • a solid support e.g., a bead
  • a solid support may be at least partially dissolvable, disruptable, and/or degradable.
  • a solid support, e.g., a bead may not be degradable.
  • the solid support e.g., a bead
  • a gel bead may be a hydrogel bead.
  • a gel bead may be formed from molecular precursors, such as a polymeric or monomeric species.
  • a semi-solid support, e g., a bead may be a liposomal bead.
  • Solid supports, e.g., beads may comprise metals including iron oxide, gold, and silver.
  • the solid support, e.g., the bead may be a silica bead.
  • the solid support, e.g., a bead can be rigid.
  • the solid support, e.g., a head may be flexible and/or compressible.
  • a partition may comprise one or more unique identifiers, such as barcodes.
  • Barcodes may be previously, subsequently or concurrently delivered to the partitions that hold the compartmentalized or partitioned biological particle.
  • barcodes may be injected into droplets or deposited in microwells previous to, subsequent to, or concurrently with droplet generation or providing of reagents in the microwells, respectively.
  • the delivery of the barcodes to a particular partition allows for the later attribution of the characteristics of the individual biological particle to the particular partition.
  • Barcodes may be delivered, for example on a nucleic acid molecule (e.g., an oligonucleotide), to a partition via any suitable mechanism.
  • Barcoded nucleic acid molecules can be delivered to a partition via a support (e.g., a bead).
  • a support in some instances, can comprise a bead. Beads are described in further detail below.
  • barcoded nucleic acid molecules can be initially associated with the support (e.g., bead) and then released from the support. Release of the barcoded nucleic acid molecules can be passive (e.g., by diffusion from or out of the support). In addition or alternatively, release from the support can be upon application of a stimulus which allows the barcoded nucleic acid nucleic acid molecules to dissociate or to be released from the support (e g., bead). Such stimulus may disrupt the support, an interaction that couples the barcoded nucleic acid molecules to or within the support, or both.
  • a stimulus which allows the barcoded nucleic acid nucleic acid molecules to dissociate or to be released from the support (e g., bead). Such stimulus may disrupt the support, an interaction that couples the barcoded nucleic acid molecules to or within the support, or both.
  • Such stimulus can include, for example, a thermal stimulus, photo-stimulus, chemical stimulus (e.g., change in pH or use of a reducing agent(s)), a mechanical stimulus, a radiation stimulus; a biological stimulus (e.g., enzyme), or any combination thereof.
  • a thermal stimulus e.g., a thermal stimulus, photo-stimulus, chemical stimulus (e.g., change in pH or use of a reducing agent(s)), a mechanical stimulus, a radiation stimulus; a biological stimulus (e.g., enzyme), or any combination thereof.
  • beads, biological particles, and droplets may flow along channels (e g., the channels of a microfluidic device), in some cases at substantially regular flow profdes (e g., at regular flow rates).
  • Such regular flow profdes may permit a droplet to include a single bead and a single biological particle.
  • Such regular flow profdes may permit the droplets to have an occupancy (e.g., droplets having beads and biological particles) greater than 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 95%.
  • Such regular flow profdes and devices that may be used to provide such regular flow profdes are provided in, for example, U.S. Patent Publication No. 2015/0292988, which is entirely incorporated herein by reference.
  • a bead may be porous, non-porous, solid, semi-solid, semi-fluidic, fluidic, and/or a combination thereof.
  • a bead may be dissolvable, disruptable, and/or degradable.
  • a bead may not be degradable.
  • the bead may be a gel bead.
  • a gel bead may be a hydrogel bead.
  • a gel bead may be formed from molecular precursors, such as a polymeric or monomeric species.
  • a semi-solid bead may be a liposomal bead.
  • Solid beads may comprise metals including iron oxide, gold, and silver.
  • the bead may be a silica bead.
  • the bead can be rigid. In other cases, the bead may be flexible and/or compressible.
  • a bead may be of any suitable shape.
  • bead shapes include, but are not limited to, spherical, non- spherical, oval, oblong, amorphous, circular, cylindrical, and variations thereof.
  • Beads may be of uniform size or heterogeneous size.
  • the diameter of a bead may be at least about 10 nanometers (nm), 100 nm, 500 nm, 1 micrometer (pm), 5pm, 10pm, 20pm, 30pm, 40pm, 50pm, 60pm, 70pm, 80pm, 90pm, 100pm, 250pm, 500pm, 1mm, or greater.
  • a bead may have a diameter of less than about 10 nm, 100 nm, 500 nm, 1pm, 5pm, 10pm, 20pm, 30pm, 40pm, 50pm, 60pm, 70pm, 80pm, 90pm, 100pm, 250pm, 500pm, 1mm, or less.
  • a bead may have a diameter in the range of about 40- 75pm, 30-75pm, 20-75pm, 40-85pm, 40-95pm, 20-100pm, 10-100pm, l-100pm, 20-250pm, or 20-500pm.
  • beads can be provided as a population or plurality of beads having a relatively monodisperse size distribution. Where it may be desirable to provide relatively consistent amounts of reagents within partitions, maintaining relatively consistent bead characteristics, such as size, can contribute to the overall consistency.
  • the beads described herein may have size distributions that have a coefficient of variation in their cross- sectional dimensions of less than 50%, less than 40%, less than 30%, less than 20%, and in some cases less than 15%, less than 10%, less than 5%, or less.
  • a bead may comprise natural and/or synthetic materials.
  • a bead can comprise a natural polymer, a synthetic polymer or both natural and synthetic polymers.
  • natural polymers include proteins and sugars such as deoxyribonucleic acid, rubber, cellulose, starch (e.g., amylose, amylopectin), proteins, enzymes, polysaccharides, silks, polyhydroxyalkanoates, chitosan, dextran, collagen, carrageenan, ispaghula, acacia, agar, gelatin, shellac, sterculia gum, xanthan gum, Com sugar gum, guar gum, gum karaya, agarose, alginic acid, alginate, or natural polymers thereof.
  • proteins and sugars such as deoxyribonucleic acid, rubber, cellulose, starch (e.g., amylose, amylopectin), proteins, enzymes, polysaccharides, silks, polyhydroxyalkanoates, chitosan, dextran, collagen, carrageenan, ispaghula, acacia, agar, gelatin, shellac, ster
  • Examples of synthetic polymers include acrylics, nylons, silicones, spandex, viscose rayon, polycarboxylic acids, polyvinyl acetate, polyacrylamide, polyacrylate, polyethylene glycol, polyurethanes, polylactic acid, silica, polystyrene, polyacrylonitrile, polybutadiene, polycarbonate, polyethylene, polyethylene terephthalate, poly(chlorotrifluoroethylene), poly(ethylene oxide), poly(ethylene terephthalate), polyethylene, polyisobutylene, poly(methyl methacrylate), poly(oxymethylene), polyformaldehyde, polypropylene, polystyrene, poly(tetrafluoroethylene), poly(vinyl acetate), poly(vinyl alcohol), poly(vinyl chloride), poly(vinylidene dichloride), poly(vinylidene difluoride), poly(vinyl fluoride) and/or combinations (e.g., co-polymers) thereof.
  • the bead may contain molecular precursors (e.g., monomers or polymers), which may form a polymer network via polymerization of the molecular precursors.
  • a precursor may be an already polymerized species capable of undergoing further polymerization via, for example, a chemical cross-linkage.
  • a precursor can comprise one or more of an acrylamide or a methacrylamide monomer, oligomer, or polymer.
  • the bead may comprise prepolymers, which are oligomers capable of further polymerization.
  • polyurethane beads may be prepared using prepolymers.
  • the bead may contain individual polymers that may be further polymerized together.
  • beads may be generated via polymerization of different precursors, such that they comprise mixed polymers, co-polymers, and/or block co-polymers.
  • the bead may comprise covalent or ionic bonds between polymeric precursors (e.g., monomers, oligomers, linear polymers), nucleic acid molecules (e.g., oligonucleotides), primers, and other entities.
  • the covalent bonds can be carbon-carbon bonds, thioether bonds, or carbonheteroatom bonds.
  • Cross-linking may be permanent or reversible, depending upon the particular crosslinker used. Reversible cross-linking may allow for the polymer to linearize or dissociate under appropriate conditions. In some cases, reversible cross-linking may also allow for reversible attachment of a material bound to the surface of a bead. In some cases, a cross-linker may form disulfide linkages. In some cases, the chemical cross-linker forming disulfide linkages may be cystamine or a modified cystamine.
  • disulfide linkages can be formed between molecular precursor units (e.g., monomers, oligomers, or linear polymers) or precursors incorporated into a bead and nucleic acid molecules (e.g., oligonucleotides).
  • Cystamine is an organic agent comprising a disulfide bond that may be used as a crosslinker agent between individual monomeric or polymeric precursors of a bead.
  • Polyacrylamide may be polymerized in the presence of cystamine or a species comprising cystamine (e.g., a modified cystamine) to generate polyacrylamide gel beads comprising disulfide linkages (e.g., chemically degradable beads comprising chemically-reducible cross-linkers).
  • the disulfide linkages may permit the bead to be degraded (or dissolved) upon exposure of the bead to a reducing agent.
  • chitosan a linear polysaccharide polymer
  • Crosslinking of chitosan polymers may be achieved by chemical reactions that are initiated by heat, pressure, change in pH, and/or radiation.
  • a bead may comprise an acrydite moiety, which in certain aspects may be used to attach one or more nucleic acid molecules (e.g., barcode sequence, barcoded nucleic acid molecule, barcoded oligonucleotide, primer, or other oligonucleotide) to the bead.
  • an acrydite moiety can refer to an acrydite analogue generated from the reaction of acrydite with one or more species, such as, the reaction of acrydite with other monomers and cross-linkers during a polymerization reaction.
  • Acrydite moieties may be modified to form chemical bonds with a species to be attached, such as a nucleic acid molecule (e.g., barcode sequence, barcoded nucleic acid molecule, barcoded oligonucleotide, primer, or other oligonucleotide).
  • Acrydite moieties may be modified with thiol groups capable of forming a disulfide bond or may be modified with groups already comprising a disulfide bond. The thiol or disulfide (via disulfide exchange) may be used as an anchor point for a species to be attached or another part of the acrydite moiety may be used for attachment.
  • attachment can be reversible, such that when the disulfide bond is broken (e.g., in the presence of a reducing agent), the attached species is released from the bead.
  • an acrydite moiety can comprise a reactive hydroxyl group that may be used for attachment.
  • nucleic acid molecules e.g., oligonucleotides
  • Functionalization of beads for attachment of nucleic acid molecules may be achieved through a wide range of different approaches, including activation of chemical groups within a polymer, incorporation of active or activatable functional groups in the polymer structure, or attachment at the pre-polymer or monomer stage in bead production.
  • precursors e.g., monomers, cross-linkers
  • precursors that are polymerized to form a bead may comprise acrydite moieties, such that when a bead is generated, the bead also comprises acrydite moieties.
  • the acrydite moieties can be attached to a nucleic acid molecule (e g., oligonucleotide) that comprises one or more functional sequences, such as a TSO sequence or a primer sequence (e.g., a poly T sequence, or a nucleic acid primer sequence complementary to a target nucleic acid sequence and/or for amplifying a target nucleic acid sequence, a random primer, or a primer sequence for messenger RNA) that is useful for incorporation into the bead, etc.) and/or one or more barcode sequences.
  • a nucleic acid molecule e g., oligonucleotide
  • a primer sequence e.g., a poly T sequence, or a nucleic acid primer sequence complementary to a target nucleic acid sequence and/or for amplifying a target nucleic acid sequence, a random primer, or a primer sequence for messenger RNA
  • the one or more barcode sequences may include sequences that are the same for all nucleic acid molecules coupled to a given bead and/or sequences that are different across all nucleic acid molecules coupled to the given bead.
  • the nucleic acid molecule may be incorporated into the bead.
  • the nucleic acid molecule can comprise a functional sequence, for example, for attachment to a sequencing flow cell, such as, for example, a P5 sequence (or a portion thereof) for Illumina® sequencing.
  • the nucleic acid molecule or derivative thereof e.g., oligonucleotide or polynucleotide generated from the nucleic acid molecule
  • can comprise another functional sequence such as, for example, a P7 sequence (or a portion thereof) for attachment to a sequencing flow cell for Illumina sequencing.
  • the nucleic acid molecule can comprise a barcode sequence.
  • the nucleic acid molecule can further comprise a unique molecular identifier (UMI).
  • UMI unique molecular identifier
  • the nucleic acid molecule can comprise an R1 primer sequence for Illumina sequencing. In some cases, the nucleic acid molecule can comprise an R2 primer sequence for Illumina sequencing. Examples of such nucleic acid molecules (e.g., oligonucleotides, polynucleotides, etc.) and uses thereof, as may be used with compositions, devices, methods and systems of the present disclosure, are provided in U.S. Patent Pub. Nos. 2014/0378345 and 2015/0376609, each of which is entirely incorporated herein by reference.
  • the nucleic acid molecule can comprise one or more functional sequences.
  • a functional sequence can comprise a sequence for attachment to a sequencing flow cell, such as, for example, a P5 sequence for Illumina® sequencing.
  • the nucleic acid molecule or derivative thereof e.g., oligonucleotide or polynucleotide generated from the nucleic acid molecule
  • the functional sequence can comprise a barcode sequence or multiple barcode sequences.
  • the functional sequence can comprise a unique molecular identifier (UMI).
  • the functional sequence can comprise a primer sequence (e.g., an R1 primer sequence for Illumina sequencing, an R2 primer sequence for Illumina sequencing, etc ).
  • a functional sequence can comprise a partial sequence, such as a partial barcode sequence, partial anchoring sequence, partial sequencing primer sequence (e.g., partial R1 sequence, partial R2 sequence, etc.), a partial sequence configured to attach to the flow cell of a sequencer (e.g., partial P5 sequence, partial P7 sequence, etc.), or a partial sequence of any other type of sequence described elsewhere herein.
  • a partial sequence may contain a contiguous or continuous portion or segment, but not all, of a full sequence, for example.
  • a downstream procedure may extend the partial sequence, or derivative thereof, to achieve a full sequence of the partial sequence, or derivative thereof.
  • nucleic acid molecules e.g., oligonucleotides, polynucleotides, etc.
  • uses thereof as may be used with compositions, devices, methods and systems of the present disclosure, are provided in U.S. Patent Pub. Nos. 2014/0378345 and 2015/0376609, each of which is entirely incorporated herein by reference.
  • FIG. 3 illustrates an example of a barcode carrying bead.
  • a nucleic acid molecule 302 such as an oligonucleotide, can be coupled to a bead 304 by a releasable linkage 306, such as, for example, a disulfide linker.
  • the same bead 304 may be coupled (e.g., via releasable linkage) to one or more other nucleic acid molecules 318, 320.
  • the nucleic acid molecule 302 may be or comprise a barcode. As noted elsewhere herein, the structure of the barcode may comprise a number of sequence elements.
  • the nucleic acid molecule 302 may comprise a functional sequence 308 that may be used in subsequent processing.
  • the functional sequence 308 may include one or more of a sequencer specific flow cell attachment sequence (e g., a P5 sequence for Illumina® sequencing systems) and a sequencing primer sequence (e.g., a R1 primer for Illumina® sequencing systems), or partial sequence(s) thereof.
  • the nucleic acid molecule 302 may comprise a barcode sequence 310 for use in barcoding the sample (e.g., DNA, RNA, protein, etc.).
  • the barcode sequence 310 can be bead-specific such that the barcode sequence 310 is common to all nucleic acid molecules (e.g., including nucleic acid molecule 302) coupled to the same bead 304.
  • the barcode sequence 310 can be partition-specific such that the barcode sequence 310 is common to all nucleic acid molecules coupled to one or more beads that are partitioned into the same partition.
  • the nucleic acid molecule 302 may comprise a specific priming sequence 312, such as an mRNA specific priming sequence (e.g., poly-T sequence), a targeted priming sequence, and/or a random priming sequence.
  • the nucleic acid molecule 302 may comprise an anchoring sequence 314 to ensure that the specific priming sequence 312 hybridizes at the sequence end (e.g., of the mRNA).
  • the anchoring sequence 314 can include a random short sequence of nucleotides, such as a 1-mer, 2-mer, 3-mer or longer sequence, which can ensure that a poly-T segment is more likely to hybridize at the sequence end of the poly-A tail of the mRNA.
  • the nucleic acid molecule 302 may comprise a unique molecular identifying sequence 316 (e.g., unique molecular identifier (UMI)).
  • the unique molecular identifying sequence 316 may comprise from about 5 to about 8 nucleotides.
  • the unique molecular identifying sequence 316 may compress less than about 5 or more than about 8 nucleotides.
  • the unique molecular identifying sequence 316 may be a unique sequence that varies across individual nucleic acid molecules (e.g., 302, 318, 320, etc.) coupled to a single bead (e.g., bead 304).
  • the unique molecular identifying sequence 316 may be a random sequence (e.g., such as a random N-mer sequence).
  • the UMI may provide a unique identifier of the starting mRNA molecule that was captured, in order to allow quantitation of the number of original expressed RNA.
  • FIG. 3 shows three nucleic acid molecules 302, 318, 320 coupled to the surface of the bead 304, an individual bead may be coupled to any number of individual nucleic acid molecules, for example, from one to tens to hundreds of thousands or even millions of individual nucleic acid molecules.
  • the respective barcodes for the individual nucleic acid molecules can comprise both common sequence segments or relatively common sequence segments (e g., 308, 310, 312, etc.) and variable or unique sequence segments (e.g., 316) between different individual nucleic acid molecules coupled to the same bead.
  • a biological particle e.g., cell, DNA, RNA, etc.
  • the nucleic acid barcode molecules 302, 318, 320 can be released from the bead 304 in the partition.
  • the poly-T segment e.g., 312
  • one of the released nucleic acid molecules e.g., 302
  • Reverse transcription may result in a cDNA transcript of the mRNA, but which transcript includes each of the sequence segments 308, 310, 316 of the nucleic acid molecule 302.
  • the nucleic acid molecule 302 comprises an anchoring sequence 3144
  • all of the cDNA transcripts of the individual mRNA molecules may include a common barcode sequence segment 310.
  • the transcripts made from the different mRNA molecules within a given partition may vary at the unique molecular identifying sequence 312 segment (e g., UMI segment).
  • UMI segment unique molecular identifying sequence
  • the number of different UMIs can be indicative of the quantity of mRNA originating from a given partition, and thus from the biological particle (e.g., cell).
  • the transcripts can be amplified, cleaned up and sequenced to identify the sequence of the cDNA transcript of the mRNA, as well as to sequence the barcode segment and the UMI segment. While a poly-T primer sequence is described, other targeted or random priming sequences may also be used in priming the reverse transcription reaction. Likewise, although described as releasing the barcoded oligonucleotides into the partition, in some cases, the nucleic acid molecules bound to the bead (e.g., gel bead) may be used to hybridize and capture the mRNA on the solid phase of the bead, for example, in order to facilitate the separation of the RNA from other cell contents.
  • the nucleic acid molecules bound to the bead e.g., gel bead
  • the nucleic acid molecules bound to the bead may be used to hybridize and capture the mRNA on the solid phase of the bead, for example, in order to facilitate the separation of the RNA from other cell contents.
  • RNA molecules on the beads may be subjected to reverse transcription or other nucleic acid processing, additional adapter sequences may be added to the barcoded nucleic acid molecules, or other nucleic acid reactions (e.g., amplification, nucleic acid extension) may be performed.
  • the beads or products thereof e.g., barcoded nucleic acid molecules
  • the beads comprising nucleic acid barcode molecules may be introduced into a partition (e.g., well or droplet) prior to, during, or following introduction of a sample into the partition.
  • the nucleic acid molecules of a sample may be subjected to barcoding, which may occur on the bead (in cases where the nucleic acid molecules remain coupled to the bead) or following release of the nucleic acid barcode molecules into the partition.
  • the beads from various partitions may be collected, pooled, and subjected to further processing (e g., reverse transcription, adapter attachment, amplification, clean up, sequencing).
  • the processing may occur in the partition.
  • conditions sufficient for barcoding, adapter attachment, reverse transcription, or other nucleic acid processing operations may be provided in the partition and performed prior to clean up and sequencing.
  • a bead may comprise a capture sequence or binding sequence configured to bind to a corresponding capture sequence or binding sequence.
  • a bead may comprise a plurality of different capture sequences or binding sequences configured to bind to different respective corresponding capture sequences or binding sequences.
  • a bead may comprise a first subset of one or more capture sequences each configured to bind to a first corresponding capture sequence, a second subset of one or more capture sequences each configured to bind to a second corresponding capture sequence, a third subset of one or more capture sequences each configured to bind to a third corresponding capture sequence, and etc.
  • a bead may comprise any number of different capture sequences.
  • a bead may comprise at least 2, 3, 4, 5, 6, 7, 8, 9, 10 or more different capture sequences or binding sequences configured to bind to different respective capture sequences or binding sequences, respectively.
  • a bead may comprise at most about 10, 9, 8, 7, 6, 5, 4, 3, or 2 different capture sequences or binding sequences configured to bind to different respective capture sequences or binding sequences.
  • the different capture sequences or binding sequences may be configured to facilitate analysis of a same type of analyte.
  • the different capture sequences or binding sequences may be configured to facilitate analysis of different types of analytes (with the same bead).
  • the capture sequence may be designed to attach to a corresponding capture sequence.
  • such corresponding capture sequence may be introduced to, or otherwise induced in, a biological particle (e.g., cell, cell bead, etc.) for performing different assays in various formats (e.g., barcoded antibodies comprising the corresponding capture sequence, barcoded MHC dextramers comprising the corresponding capture sequence, barcoded guide RNA molecules comprising the corresponding capture sequence, etc.), such that the corresponding capture sequence may later interact with the capture sequence associated with the bead.
  • a biological particle e.g., cell, cell bead, etc.
  • a biological particle e.g., cell, cell bead, etc.
  • formats e.g., barcoded antibodies comprising the corresponding capture sequence, barcoded MHC dextramers comprising the corresponding capture sequence, barcoded guide RNA molecules comprising the corresponding capture sequence, etc.
  • a capture sequence coupled to a bead may be configured to attach to a linker molecule, such as a splint molecule, wherein the linker molecule is configured to couple the bead (or other support) to other molecules through the linker molecule, such as to one or more analytes or one or more other linker molecules.
  • a linker molecule such as a splint molecule
  • FIG. 4 illustrates another example of a barcode carrying bead.
  • a nucleic acid molecule 405, such as an oligonucleotide, can be coupled to a bead 404 by a releasable linkage 406, such as, for example, a disulfide linker.
  • the nucleic acid molecule 405 may comprise a first capture sequence 460.
  • the same bead 404 may be coupled (e.g., via releasable linkage) to one or more other nucleic acid molecules 403, 407 comprising other capture sequences.
  • the nucleic acid molecule 405 may be or comprise a barcode.
  • the structure of the barcode may comprise a number of sequence elements, such as a functional sequence 408 (e.g., flow cell attachment sequence, sequencing primer sequence, etc ), a barcode sequence 410 (e.g., bead-specific sequence common to bead, partition-specific sequence common to partition, etc.), and a unique molecular identifier 412 (e.g., unique sequence within different molecules attached to the bead), or partial sequences thereof.
  • the capture sequence 460 may be configured to attach to a corresponding capture sequence 465.
  • the corresponding capture sequence 465 may be coupled to another molecule that may be an analyte or an intermediary carrier. For example, as illustrated in FIG.
  • the corresponding capture sequence 465 is coupled to a guide RNA molecule 462 comprising a target sequence 464, wherein the target sequence 464 is configured to attach to the analyte.
  • Another oligonucleotide molecule 407 attached to the bead 404 comprises a second capture sequence 480 which is configured to attach to a second
  • the second corresponding capture sequence 485 is coupled to an antibody 482.
  • the antibody 482 may have binding specificity to an analyte (e.g., surface protein). Alternatively, the antibody 482 may not have binding specificity.
  • Another oligonucleotide molecule 403 attached to the bead 404 comprises a third capture sequence 470 which is configured to attach to a second corresponding capture sequence 475. As illustrated in FIG. 4, the third corresponding capture sequence 475 is coupled to a molecule 472.
  • the molecule 472 may or may not be configured to target an analyte.
  • the other oligonucleotide molecules 403, 407 may comprise the other sequences (e g., functional sequence, barcode sequence, UMI, etc.) described with respect to oligonucleotide molecule 405. While a single oligonucleotide molecule comprising each capture sequence is illustrated in FIG. 4, it will be appreciated that, for each capture sequence, the bead may comprise a set of one or more oligonucleotide molecules each comprising the capture sequence. For example, the bead may comprise any number of sets of one or more different capture sequences. Alternatively or in addition, the bead 404 may comprise other capture sequences.
  • the bead 404 may comprise fewer types of capture sequences (e.g., two capture sequences).
  • the bead 404 may comprise oligonucleotide molecule(s) comprising a priming sequence, such as a specific priming sequence such as an mRNA specific priming sequence (e.g., poly-T sequence), a targeted priming sequence, and/or a random priming sequence, for example, to facilitate an assay for gene expression.
  • the barcoded oligonucleotides may be released (e g., in a partition), as described elsewhere herein.
  • the nucleic acid molecules bound to the bead e.g., gel bead
  • analytes e.g., one or more types of analytes
  • precursors comprising a functional group that is reactive or capable of being activated such that it becomes reactive can be polymerized with other precursors to generate gel beads comprising the activated or activatable functional group.
  • the functional group may then be used to attach additional species (e.g., disulfide linkers, primers, other oligonucleotides, etc.) to the gel beads.
  • additional species e.g., disulfide linkers, primers, other oligonucleotides, etc.
  • some precursors comprising a carboxylic acid (COOH) group can co-polymerize with other precursors to form a gel bead that also comprises a COOH functional group.
  • acrylic acid a species comprising free COOH groups
  • acrylamide acrylamide
  • bis(acryloyl)cystamine can be co-polymerized together to generate a gel bead comprising free COOH groups.
  • the COOH groups of the gel bead can be activated (e.g., via l-Ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC) and N- Hydroxysuccinimide (NHS) or 4-(4,6-Dimethoxy-l,3,5-triazin-2-yl)-4-methylmorpholinium chloride (DMTMM)) such that they are reactive (e.g., reactive to amine functional groups where EDC/NHS or DMTMM are used for activation).
  • EDC l-Ethyl-3-(3-dimethylaminopropyl)carbodiimide
  • NHS N- Hydroxysuccinimide
  • DTMM 4-(4,6-Dimethoxy-
  • the activated COOH groups can then react with an appropriate species (e.g., a species comprising an amine functional group where the carboxylic acid groups are activated to be reactive with an amine functional group) comprising a moiety to be linked to the bead.
  • an appropriate species e.g., a species comprising an amine functional group where the carboxylic acid groups are activated to be reactive with an amine functional group
  • Beads comprising disulfide linkages in their polymeric network may be functionalized with additional species via reduction of some of the disulfide linkages to free thiols.
  • the disulfide linkages may be reduced via, for example, the action of a reducing agent (e g., DTT, TCEP, etc.) to generate free thiol groups, without dissolution of the bead.
  • Free thiols of the beads can then react with free thiols of a species or a species comprising another disulfide bond (e.g., via thiol-disulfide exchange) such that the species can be linked to the beads (e.g., via a generated disulfide bond).
  • free thiols of the beads may react with any other suitable group.
  • free thiols of the beads may react with species comprising an acrydite moiety.
  • the free thiol groups of the beads can react with the acrydite via Michael addition chemistry, such that the species comprising the acrydite is linked to the bead.
  • uncontrolled reactions can be prevented by inclusion of a thiol capping agent such as N- ethylmalieamide or iodoacetate.
  • Activation of disulfide linkages within a bead can be controlled such that only a small number of disulfide linkages are activated. Control may be exerted, for example, by controlling the concentration of a reducing agent used to generate free thiol groups and/or concentration of reagents used to form disulfide bonds in bead polymerization.
  • a low concentration e.g., molecules of reducing agent: gel bead ratios of less than or equal to about 1:100,000,000,000, less than or equal to about 1: 10,000,000,000, less than or equal to about 1:1,000,000,000, less than or equal to about 1 :100,000,000, less than or equal to about 1:10,000,000, less than or equal to about 1:1,000,000, less than or equal to about 1 :100,000, less than or equal to about 1 : 10,000
  • reducing agent may be used for reduction.
  • Controlling the number of disulfide linkages that are reduced to free thiols may be useful in ensuring bead structural integrity during functionalization.
  • optically-active agents such as fluorescent dyes may be coupled to beads via free thiol groups of the beads and used to quantify the number of free thiols present in a bead and/or track a bead.
  • addition of moieties to a gel bead after gel bead formation may be advantageous.
  • addition of an oligonucleotide (e g., barcoded oligonucleotide) after gel bead formation may avoid loss of the species during chain transfer termination that can occur during polymerization.
  • smaller precursors e.g., monomers or cross linkers that do not comprise side chain groups and linked moieties
  • functionalization after gel bead synthesis can minimize exposure of species (e g., oligonucleotides) to be loaded with potentially damaging agents (e.g., free radicals) and/or chemical environments.
  • the generated gel may possess an upper critical solution temperature (UCST) that can permit temperature driven swelling and collapse of a bead.
  • UCT upper critical solution temperature
  • Such functionality may aid in oligonucleotide (e.g., a primer) infdtration into the bead during subsequent functionalization of the bead with the oligonucleotide.
  • Post-production functionalization may also be useful in controlling loading ratios of species in beads, such that, for example, the variability in loading ratio is minimized.
  • Species loading may also be performed in a batch process such that a plurality of beads can be functionalized with the species in a single batch.
  • a bead injected or otherwise introduced into a partition may comprise releasably, cleavably, or reversibly attached barcodes.
  • a bead injected or otherwise introduced into a partition may comprise activatable barcodes.
  • a bead injected or otherwise introduced into a partition may be degradable, disruptable, or dissolvable beads.
  • Barcodes can be releasably, cleavably or reversibly attached to the beads such that barcodes can be released or be releasable through cleavage of a linkage between the barcode molecule and the bead, or released through degradation of the underlying bead itself, allowing the barcodes to be accessed or be accessible by other reagents, or both.
  • cleavage may be achieved through reduction of di-sulfide bonds, use of restriction enzymes, photo-activated cleavage, or cleavage via other types of stimuli (e.g., chemical, thermal, pH, enzymatic, etc.) and/or reactions, such as described elsewhere herein.
  • Releasable barcodes may sometimes be referred to as being activatable, in that they are available for reaction once released.
  • an activatable barcode may be activated by releasing the barcode from a bead (or other suitable type of partition described herein).
  • Other activatable configurations are also envisioned in the context of the described methods and systems.
  • the beads may be degradable, disruptable, or dissolvable spontaneously or upon exposure to one or more stimuli (e.g., temperature changes, pH changes, exposure to particular chemical species or phase, exposure to light, reducing agent, etc.).
  • a bead may be dissolvable, such that material components of the beads are solubilized when exposed to a particular chemical species or an environmental change, such as a change temperature or a change in pH.
  • a gel bead can be degraded or dissolved at elevated temperature and/or in basic conditions.
  • a bead may be thermally degradable such that when the bead is exposed to an appropriate change in temperature (e.g., heat), the bead degrades.
  • Degradation or dissolution of a bead bound to a species e.g., a nucleic acid molecule, e.g., barcoded oligonucleotide
  • a species e.g., a nucleic acid molecule, e.g., barcoded oligonucleotide
  • the degradation of a bead may refer to the disassociation of a bound or entrained species from a bead, both with and without structurally degrading the physical bead itself.
  • the degradation of the bead may involve cleavage of a cleavable linkage via one or more species and/or methods described elsewhere herein.
  • entrained species may be released from beads through osmotic pressure differences due to, for example, changing chemical environments.
  • alteration of bead pore sizes due to osmotic pressure differences can generally occur without structural degradation of the bead itself.
  • an increase in pore size due to osmotic swelling of a bead can permit the release of entrained species within the bead.
  • osmotic shrinking of a bead may cause a bead to better retain an entrained species due to pore size contraction.
  • a degradable bead may be introduced into a partition, such as a droplet of an emulsion or a well, such that the bead degrades within the partition and any associated species (e g., oligonucleotides) are released within the droplet when the appropriate stimulus is applied.
  • the free species e.g., oligonucleotides, nucleic acid molecules
  • a polyacrylamide bead comprising cystamine and linked, via a disulfide bond, to a barcode sequence, may be combined with a reducing agent within a droplet of a water-in-oil emulsion.
  • the reducing agent can break the various disulfide bonds, resulting in bead degradation and release of the barcode sequence into the aqueous, inner environment of the droplet.
  • heating of a droplet comprising a bead-bound barcode sequence in basic solution may also result in bead degradation and release of the attached barcode sequence into the aqueous, inner environment of the droplet.
  • any suitable number of molecular tag molecules can be associated with a bead such that, upon release from the bead, the molecular tag molecules (e.g., primer, e.g., barcoded oligonucleotide) are present in the partition at a pre-defined concentration.
  • the pre-defined concentration may be selected to facilitate certain reactions for generating a sequencing library, e.g., amplification, within the partition.
  • the pre-defined concentration of the primer can be limited by the process of producing nucleic acid molecule (e.g., oligonucleotide) bearing beads.
  • beads can be non-covalently loaded with one or more reagents.
  • the beads can be non-covalently loaded by, for instance, subjecting the beads to conditions sufficient to swell the beads, allowing sufficient time for the reagents to diffuse into the interiors of the beads, and subjecting the beads to conditions sufficient to de-swell the beads.
  • the swelling of the beads may be accomplished, for instance, by placing the beads in a thermodynamically favorable solvent, subjecting the beads to a higher or lower temperature, subjecting the beads to a higher or lower ion concentration, and/or subjecting the beads to an electric field.
  • the swelling of the beads may be accomplished by various swelling methods.
  • the de-swelling of the beads may be accomplished, for instance, by transferring the beads in a thermodynamically unfavorable solvent, subjecting the beads to lower or high temperatures, subjecting the beads to a lower or higher ion concentration, and/or removing an electric field.
  • the de-swelling of the beads may be accomplished by various de-swelling methods. Transferring the beads may cause pores in the bead to shrink. The shrinking may then hinder reagents within the beads from diffusing out of the interiors of the beads. The hindrance may be due to steric interactions between the reagents and the interiors of the beads.
  • the transfer may be accomplished microfluidically. For instance, the transfer may be achieved by moving the beads from one coflowing solvent stream to a different co-flowing solvent stream.
  • the swellability and/or pore size of the beads may be adjusted by changing the polymer composition of the bead.
  • an acrydite moiety linked to a precursor, another species linked to a precursor, or a precursor itself can comprise a labile bond, such as chemically, thermally, or photo-sensitive bond e.g., disulfide bond, UV sensitive bond, or the like.
  • a labile bond such as chemically, thermally, or photo-sensitive bond e.g., disulfide bond, UV sensitive bond, or the like.
  • the bead may also comprise the labile bond.
  • the labile bond may be, for example, useful in reversibly linking (e.g., covalently linking) species (e.g., barcodes, primers, etc.) to a bead.
  • a thermally labile bond may include a nucleic acid hybridization based attachment, e.g., where an oligonucleotide is hybridized to a complementary sequence that is attached to the bead, such that thermal melting of the hybrid releases the oligonucleotide, e.g., a barcode containing sequence, from the support (e.g., a bead such as a gel bead).
  • a nucleic acid hybridization based attachment e.g., where an oligonucleotide is hybridized to a complementary sequence that is attached to the bead, such that thermal melting of the hybrid releases the oligonucleotide, e.g., a barcode containing sequence, from the support (e.g., a bead such as a gel bead).
  • labile bonds may result in the generation of a bead capable of responding to varied stimuli.
  • Each type of labile bond may be sensitive to an associated stimulus (e g., chemical stimulus, light, temperature, enzymatic, etc.) such that release of species attached to a bead via each labile bond may be controlled by the application of the appropriate stimulus.
  • an associated stimulus e g., chemical stimulus, light, temperature, enzymatic, etc.
  • Such functionality may be useful in controlled release of species from a gel bead.
  • another species comprising a labile bond may be linked to a gel bead after gel bead formation via, for example, an activated functional group of the gel bead as described above.
  • barcodes that are releasably, cleavably or reversibly attached to the beads described herein include barcodes that are released or releasable through cleavage of a linkage between the barcode molecule and the bead, or that are released through degradation of the underlying bead itself, allowing the barcodes to be accessed or accessible by other reagents, or both.
  • a species e.g., oligonucleotide molecules comprising barcodes
  • a solid support e.g., a bead
  • the U-excising element may comprise a singlestranded DNA (ssDNA) sequence that contains at least one uracil.
  • the species may be attached to a solid support via the ssDNA sequence containing the at least one uracil.
  • the species may be released by a combination of uracil-DNA glycosylase (e.g., to remove the uracil) and an endonuclease (e.g., to induce an ssDNA break). If the endonuclease generates a 5’ phosphate group from the cleavage, then additional enzyme treatment may be included in downstream processing to eliminate the phosphate group, e.g., prior to ligation of additional sequencing handle elements, e.g., Illumina full P5 sequence, partial P5 sequence, full R1 sequence, and/or partial R1 sequence.
  • additional enzyme treatment may be included in downstream processing to eliminate the phosphate group, e.g., prior to ligation of additional sequencing handle elements, e.g., Illumina full P5 sequence, partial P5 sequence, full R1 sequence, and/or partial R1 sequence.
  • the barcodes that are releasable as described herein may sometimes be referred to as being activatable, in that they are available for reaction once released.
  • an activatable barcode may be activated by releasing the barcode from a bead (or other suitable type of partition described herein).
  • Other activatable configurations are also envisioned in the context of the described methods and systems.
  • labile bonds that may be coupled to a precursor or bead include an ester linkage (e.g., cleavable with an acid, a base, or hydroxylamine), a vicinal diol linkage (e g., cleavable via sodium periodate), a Diels-Alder linkage (e g., cleavable via heat), a sulfone linkage (e.g., cleavable via a base), a silyl ether linkage (e g., cleavable via an acid), a glycosidic linkage (e g., cleavable via an amylase), a peptide linkage (e.g., cleavable via a protease), or a phosphodiester linkage (e.g., cleavable via a nuclease), or a phosphodiester linkage (e.g., cleavable via a nuclel link
  • Species may be encapsulated in beads during bead generation (e.g., during polymerization of precursors). Such species may or may not participate in polymerization. Such species may be entered into polymerization reaction mixtures such that generated beads comprise the species upon bead formation. In some cases, such species may be added to the gel beads after formation.
  • Such species may include, for example, nucleic acid molecules (e.g., oligonucleotides), reagents for a nucleic acid amplification reaction (e.g., primers, polymerases, dNTPs, co-factors (e.g., ionic co-factors), buffers) including those described herein, reagents for enzymatic reactions (e.g., enzymes, co-factors, substrates, buffers), reagents for nucleic acid modification reactions such as polymerization, ligation, or digestion, and/or reagents for template preparation (e.g., tagmentation) for one or more sequencing platforms (e.g., Nextera® for Illumina®).
  • nucleic acid molecules e.g., oligonucleotides
  • reagents for a nucleic acid amplification reaction e.g., primers, polymerases, dNTPs, co-factors (e.g., i
  • Such species may include one or more enzymes described herein, including without limitation, polymerase, reverse transcriptase, restriction enzymes (e g., endonuclease), transposase, ligase, proteinase K, DNAse, etc.
  • Such species may include one or more reagents described elsewhere herein (e.g., lysis agents, inhibitors, inactivating agents, chelating agents, stimulus). Trapping of such species may be controlled by the polymer network density generated during polymerization of precursors, control of ionic charge within the gel bead (e.g., via ionic species linked to polymerized species), or by the release of other species.
  • Encapsulated species may be released from a bead upon bead degradation and/or by application of a stimulus capable of releasing the species from the bead.
  • species may be partitioned in a partition (e.g., droplet) during or subsequent to partition formation.
  • a partition e.g., droplet
  • Such species may include, without limitation, the abovementioned species that may also be encapsulated in a bead.
  • a degradable bead may comprise one or more species with a labile bond such that, when the bead/species is exposed to the appropriate stimuli, the bond is broken and the bead degrades.
  • the labile bond may be a chemical bond (e g., covalent bond, ionic bond) or may be another type of physical interaction (e.g., van der Waals interactions, dipole-dipole interactions, etc ).
  • a crosslinker used to generate a bead may comprise a labile bond. Upon exposure to the appropriate conditions, the labile bond can be broken and the bead degraded.
  • a degradable bead may be useful in more quickly releasing an attached species (e.g., a nucleic acid molecule, a barcode sequence, a primer, etc.) from the bead when the appropriate stimulus is applied to the bead as compared to a bead that does not degrade.
  • an attached species e.g., a nucleic acid molecule, a barcode sequence, a primer, etc.
  • the species may have greater mobility and accessibility to other species in solution upon degradation of the bead.
  • a species may also be attached to a degradable bead via a degradable linker (e.g., disulfide linker).
  • the degradable linker may respond to the same stimuli as the degradable bead or the two degradable species may respond to different stimuli.
  • a barcode sequence may be attached, via a disulfide bond, to a polyacrylamide bead comprising cystamine. Upon exposure of the barcoded-bead to a reducing agent, the bead degrades and the barcode sequence is released upon breakage of both the disulfide linkage between the barcode sequence and the bead and the disulfide linkages of the cystamine in the bead.
  • degradation may refer to the disassociation of a bound or entrained species from a bead, both with and without structurally degrading the physical bead itself.
  • entrained species may be released from beads through osmotic pressure differences due to, for example, changing chemical environments.
  • alteration of bead pore sizes due to osmotic pressure differences can generally occur without structural degradation of the bead itself.
  • an increase in pore size due to osmotic swelling of a bead can permit the release of entrained species within the bead.
  • osmotic shrinking of a bead may cause a bead to better retain an entrained species due to pore size contraction.
  • degradable beads it may be beneficial to avoid exposing such beads to the stimulus or stimuli that cause such degradation prior to a given time, in order to, for example, avoid premature bead degradation and issues that arise from such degradation, including for example poor flow characteristics and aggregation.
  • beads comprise reducible cross-linking groups, such as disulfide groups
  • reducing agents e g., DTT or other disulfide cleaving reagents.
  • treatment to the beads described herein will, in some cases be provided free of reducing agents, such as DTT.
  • reducing agent free (or DTT free) enzyme preparations in treating the beads described herein.
  • enzymes include, e.g., polymerase enzyme preparations, reverse transcriptase enzyme preparations, ligase enzyme preparations, as well as many other enzyme preparations that may be used to treat the beads described herein.
  • the terms “reducing agent free” or “DTT free” preparations can refer to a preparation having less than about 1/1 Oth, less than about l/50th, or even less than about 1/100th of the lower ranges for such materials used in degrading the beads.
  • the reducing agent free preparation can have less than about 0.01 millimolar (mM), 0.005 mM, 0.001 mM DTT, 0.0005 mM DTT, or even less than about 0.0001 mM DTT. In many cases, the amount of DTT can be undetectable.
  • Numerous chemical triggers may be used to trigger the degradation of beads. Examples of these chemical changes may comprise pH-mediated changes to the integrity of a component within the bead, degradation of a component of a bead via cleavage of cross-linked bonds, and depolymerization of a component of a bead.
  • a bead may be formed from materials that comprise degradable chemical crosslinkers, such as BAG or cystamine. Degradation of such degradable crosslinkers may be accomplished through a number of mechanisms.
  • a bead may be contacted with a chemical degrading agent that may induce oxidation, reduction or other chemical changes.
  • a chemical degrading agent may be a reducing agent, such as dithiothreitol (DTT).
  • reducing agents may include P-mercaptoethanol, (2S)-2-amino-l,4-dimercaptobutane (dithiobutylamine or DTBA), tris(2-carboxyethyl) phosphine (TCEP), or combinations thereof
  • a reducing agent may degrade the disulfide bonds formed between gel precursors forming the bead, and thus, degrade the bead.
  • a change in pH of a solution such as an increase in pH, may trigger degradation of a bead.
  • exposure to an aqueous solution, such as water may trigger hydrolytic degradation, and thus degradation of the bead.
  • any combination of stimuli may trigger degradation of a bead.
  • a change in pH may enable a chemical agent (e.g., DTT) to become an effective reducing agent.
  • Beads may also be induced to release their contents upon the application of a thermal stimulus.
  • a change in temperature can cause a variety of changes to a bead. For example, heat can cause a solid bead to liquefy. A change in heat may cause melting of a bead such that a portion of the bead degrades. In other cases, heat may increase the internal pressure of the bead components such that the bead ruptures or explodes. Heat may also act upon heat-sensitive polymers used as materials to construct beads.
  • any suitable agent may degrade beads.
  • changes in temperature or pH may be used to degrade thermo-sensitive or pH-sensitive bonds within beads.
  • chemical degrading agents may be used to degrade chemical bonds within beads by oxidation, reduction or other chemical changes.
  • a chemical degrading agent may be a reducing agent, such as DTT, wherein DTT may degrade the disulfide bonds formed between a crosslinker and gel precursors, thus degrading the bead.
  • a reducing agent may be added to degrade the bead, which may or may not cause the bead to release its contents.
  • reducing agents may include dithiothreitol (DTT), P-mercaptoethanol, (2S)-2-amino-l,4-dimercaptobutane (dithiobutylamine or DTBA), tris(2- carboxyethyl) phosphine (TCEP), or combinations thereof.
  • the reducing agent may be present at a concentration of about O.lmM, 0.5mM, ImM, 5mM, lOmM.
  • the reducing agent may be present at a concentration of at least about 0. ImM, 0.5mM, ImM, 5mM, lOmM, or greater than 10 mM.
  • the reducing agent may be present at concentration of at most about lOmM, 5mM, ImM, 0.5mM, O.lmM, or less.
  • any suitable number of molecular tag molecules can be associated with a bead such that, upon release from the bead, the molecular tag molecules (e.g., primer, e.g., barcoded oligonucleotide) are present in the partition at a pre-defined concentration.
  • the molecular tag molecules e.g., primer, e.g., barcoded oligonucleotide
  • the pre-defined concentration of the primer can be limited by the process of producing oligonucleotide bearing beads.
  • a partition of the plurality of partitions may comprise a single biological particle (e.g., a single cell or a single nucleus of a cell).
  • a partition of the plurality of partitions may comprise multiple biological particles.
  • Such partitions may be referred to as multiply occupied partitions, and may comprise, for example, two, three, four or more cells and/or supports (e.g., beads) comprising barcoded nucleic acid molecules (e g., oligonucleotides) within a single partition.
  • the flow characteristics of the biological particle and/or bead containing fluids and partitioning fluids may be controlled to provide for such multiply occupied partitions.
  • the flow parameters may be controlled to provide a given occupancy rate at greater than about 50% of the partitions, greater than about 75%, and in some cases greater than about 80%, 90%, 95%, or higher.
  • additional supports e.g., beads
  • additional supports can be used to deliver additional reagents to a partition.
  • the flow and frequency of the different beads into the channel or junction may be controlled to provide for a certain ratio of supports from each source, while ensuring a given pairing or combination of such beads into a partition with a given number of biological particles (e.g., one biological particle and one bead per partition).
  • the partitions described herein may comprise small volumes, for example, less than about 10 microliters (pL), 5pL, IpL, 900 picoliters (pL), 800 pL, 700 pL, 600 pL, 500 pL, 400pL, 300 pL, 200 pL, lOOpL, 50 pL, 20 pL, 10 pL, 1 pL, 500 nanoliters (nL), 100 nL, 50 nL, or less.
  • small volumes for example, less than about 10 microliters (pL), 5pL, IpL, 900 picoliters (pL), 800 pL, 700 pL, 600 pL, 500 pL, 400pL, 300 pL, 200 pL, lOOpL, 50 pL, 20 pL, 10 pL, 1 pL, 500 nanoliters (nL), 100 nL, 50 nL, or less.
  • the droplets may have overall volumes that are less than about 1000 pL, 900 pL, 800 pL, 700 pL, 600 pL, 500 pL, 400pL, 300 pL, 200 pL, lOOpL, 50 pL, 20 pL, 10 pL, 1 pL, or less.
  • sample fluid volume e.g., including co-partitioned biological particles and/or beads
  • the sample fluid volume within the partitions may be less than about 90% of the above described volumes, less than about 80%, less than about 70%, less than about 60%, less than about 50%, less than about 40%, less than about 30%, less than about 20%, or less than about 10% of the above described volumes.
  • partitioning species may generate a population or plurality of partitions.
  • any suitable number of partitions can be generated or otherwise provided. For example, at least about 1,000 partitions, at least about 5,000 partitions, at least about 10,000 partitions, at least about 50,000 partitions, at least about 100,000 partitions, at least about 500,000 partitions, at least about 1,000,000 partitions, at least about 5,000,000 partitions at least about 10,000,000 partitions, at least about 50,000,000 partitions, at least about 100,000,000 partitions, at least about 500,000,000 partitions, at least about 1,000,000,000 partitions, or more partitions can be generated or otherwise provided.
  • the plurality of partitions may comprise both unoccupied partitions (e.g., empty partitions) and occupied partitions.
  • a single or integrated process workflow may permit the processing, identification, and/or analysis of more or multiple analytes, more or multiple types of analytes, and/or more or multiple types of analyte characterizations.
  • one or more labelling agents capable of binding to or otherwise coupling to one or more cell features may be used to characterize biological particles and/or cell features.
  • cell features include cell surface features.
  • Cell surface features may include, but are not limited to, a receptor, an antigen, a surface protein, a transmembrane protein, a cluster of differentiation protein, a protein channel, a protein pump, a carrier protein, a phospholipid, a glycoprotein, a glycolipid, a cell-cell interaction protein complex, an antigen-presenting complex, a major histocompatibility complex, an engineered T-cell receptor, a T-cell receptor, a B-cell receptor, a chimeric antigen receptor, a gap junction, an adherens junction, or any combination thereof.
  • cell features may include intracellular analytes, such as proteins, protein modifications (e.g., phosphorylation status or other post-translational modifications), nuclear proteins, nuclear membrane proteins, or any combination thereof.
  • a labelling agent may include, but is not limited to, a protein, a peptide, an antibody (or an epitope binding fragment thereof), a lipophilic moiety (such as cholesterol), a cell surface receptor binding molecule, a receptor ligand, a small molecule, a bispecific antibody, a bi-specific T-cell engager, a T-cell receptor engager, a B-cell receptor engager, a pro-body, an aptamer, a monobody, an affimer, a darpin, and a protein scaffold, or any combination thereof.
  • the labelling agents can include (e.g., are attached to) a reporter oligonucleotide that is indicative of the cell surface feature to which the binding group binds.
  • the reporter oligonucleotide may comprise a barcode sequence that permits identification of the labelling agent.
  • a labelling agent that is specific to one type of cell feature e.g., a first cell surface feature
  • a labelling agent that is specific to a different cell feature e.g., a second cell surface feature
  • a different reporter oligonucleotide coupled thereto e.g., a second cell surface feature
  • a library of potential cell feature labelling agents or binding groups may be provided, where the respective cell feature labelling agents are associated with nucleic acid reporter molecules (or reporter oligonucleotides), such that a different reporter oligonucleotide sequence is associated with each labelling agent capable of binding to a specific cell feature.
  • different members of the library may be characterized by the presence of a different oligonucleotide sequence label.
  • an antibody capable of binding to a first protein may have associated with it a first reporter oligonucleotide sequence
  • an antibody capable of binding to a second protein may have a different reporter oligonucleotide sequence associated with it.
  • the presence of the particular oligonucleotide sequence may be indicative of the presence of a particular antibody or cell feature which may be recognized or bound by the particular antibody.
  • Labelling agents capable of binding to or otherwise coupling to one or more biological particles may be used to characterize a biological particle as belonging to a particular set of biological particles.
  • labeling agents may be used to label a sample of cells, nuclei, or cell beads, or a group of cells, nuclei, or cell beads.
  • a group of cells may be labeled as different from another group of cells (or nuclei or cell beads).
  • a first group of cells may originate from a first sample and a second group of cells may originate from a second sample.
  • Labelling agents may allow the first group and second group to have a different labeling agent (or reporter oligonucleotide associated with the labeling agent). This may, for example, facilitate multiplexing, where cells of the first group and cells of the second group may be labeled separately and then pooled together for downstream analysis. The downstream detection of a label may indicate analytes as belonging to a particular group.
  • a reporter oligonucleotide may be linked to an antibody or an epitope binding fragment thereof, and labeling a biological particle may comprise subjecting the antibody-linked barcode molecule or the epitope binding fragment-linked barcode molecule to conditions suitable for binding the antibody to a molecule present on a surface of the biological particle.
  • the binding affinity between the antibody or the epitope binding fragment thereof and the molecule present on the surface may be within a useful range to ensure that, the antibody or the epitope binding fragment thereof remains bound to the molecule.
  • the binding affinity may be within a useful range to ensure that the antibody or the epitope binding fragment thereof remains bound to the molecule during various sample processing steps, such as partitioning and/or nucleic acid amplification or extension.
  • a dissociation constant (Kd) between the antibody or an epitope binding fragment thereof and die molecule to which it binds may be less than about 100 pM, 90 pM, 80 pM, 70 uM, 60 pM, 50 pM, 40 uM, 30 pM, 20 pM, 10 pM, 9 pM, 8 pM, 7 pM, 6 pM, 5 pM, 4 pM, 3 pM, 2 pM, 1 uM, 900 nM ; 800 nM, 700 nM, 600 nM, 500 nM, 400 nM, 300 nM, 200 nM, 100 nM, 90 nM, 80 nM, 70 nM, 60 nM, 50 n
  • a reporter oligonucleotide may be coupled to a cell “penetrating peptide (CPP), and labeling cells may comprise delivering the CPP coupled reporter oligonucleotide into a biological particle.
  • Labeling biological particles may comprise delivering the CPP conjugated oligonucleotide into a cell and/or cell bead by the cell-penetrating peptide.
  • a cell-penetrating peptide that can be used in the methods provided herein can comprise at least one non-functional cysteine residue, which may be either free or derivatized to form a disulfide link with an oligonucleotide that has been modified for such linkage.
  • Non-limiting examples of cell -penetrating peptides that can be used in embodiments herein include penetratin, transportan, plsl, TAT(48-60), pVEC, MTS, and MAP.
  • Cell -penetrating peptides useful in the methods provided herein can have the capability of inducing cell penetration for at least about 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% of cells of a cell population.
  • the cell-penetrating peptide may be an arginine-rich peptide transporter.
  • the cell-penetrating peptide may be Penetratin or the Tai peptide.
  • a reporter oligonucleotide may be coupled to a fluorophore or dye, and labeling cells (or nuclei or cell beads) may comprise subjecting the fluorophore-linked barcode molecule to conditions suitable for binding the fluorophore to the surface of the biological particle.
  • fluorophores can interact strongly with lipid bilayers and labeling biological particles may comprise subjecting the fluorophore-linked barcode molecule to conditions such that the fluorophore binds to or is inserted into a membrane of the biological particle.
  • the fluorophore is a water-soluble, organic fluorophore.
  • the fluorophore is Alexa 532 maleimide, tetramethylrhodamine-5-maleimide (TMR maleimide), BOD IPY-TMR maleimide, Sulfo-Cy3 maleimide, Alexa 546 carboxylic acid/succinimidyl ester, Atto 550 maleimide, Cy3 carboxylic acid/succinimidyl ester, Cy3B carboxylic acid/succinimidyl ester, Atto 565 biotin, Sulforhodamine B, Alexa 594 maleimide, Texas Red maleimide, Alexa 633 maleimide, Abberior STAR 635P azide, Atto 647N maleimide, Atto 647 SE, or Sulfo-Cy5 maleimide. See, e.g., Hughes L D, et ah PLoS One. 2014 Feb. 4; 9(2):e87649, which is hereby incorporated by reference in its entirety for all
  • a reporter oligonucleotide may be coupled to a lipophilic molecule, and labeling biological particles may comprise delivering the nucleic acid barcode molecule to a membrane of the biological particle or a nuclear membrane by the lipophilic molecule.
  • Lipophilic molecules can associate with and/or insert into lipid membranes such as cell membranes and nuclear membranes. In some cases, the insertion can be reversible. In some cases, the association between the lipophilic molecule and biological particle may be such that the biological particle retains the lipophilic molecule (e g., and associated components, such as nucleic acid barcode molecules, thereof) during subsequent processing (e.g., partitioning, cell permeabilization, amplification, pooling, etc.).
  • the reporter nucleotide may enter into the intracellular space and/or a cell nucleus.
  • a reporter oligonucleotide may be part of a nucleic acid molecule comprising any number of functional sequences, as described elsewhere herein, such as a target capture sequence, a random primer sequence, and the like, and coupled to another nucleic acid molecule that is, or is derived from, the analyte.
  • the cells Prior to, during, or following partitioning, the cells (or nuclei or cell beads) may be incubated with the library of labelling agents, that may be labelling agents to a broad panel of different cell features, e.g., receptors, proteins, etc., and which include their associated reporter oligonucleotides. Unbound labelling agents may be washed from the cells, and the cells (or nuclei or cell beads) may then be co-partitioned (e.g., into droplets or wells ) along with partition-specific barcode oligonucleotides (e.g., attached to a support, such as a bead or gel bead) as described elsewhere herein. As a result, the partitions may include the cell or cells, as well as the bound labelling agents and their known, associated reporter oligonucleotides.
  • the library of labelling agents that may be labelling agents to a broad panel of different cell features, e.g., receptors, proteins, etc., and which
  • a labelling agent that is specific to a particular cell feature may have a first plurality of the labelling agent (e.g., an antibody or lipophilic moiety) coupled to a first reporter oligonucleotide and a second plurality of the labelling agent coupled to a second reporter oligonucleotide.
  • the first plurality of the labeling agent and second plurality of the labeling agent may interact with different cells, cell populations or samples, allowing a particular report oligonucleotide to indicate a particular cell population (or cell or sample) and cell feature.
  • libraries of labelling agents may be associated with a particular cell feature as well as be used to identify analytes as originating from a particular biological particle, population, or sample.
  • the biological particles may be incubated with a plurality of libraries and a given biological particle may comprise multiple labelling agents.
  • a cell may comprise coupled thereto a lipophilic labeling agent and an antibody.
  • the lipophilic labeling agent may indicate that the cell is a member of a particular cell sample, whereas the antibody may indicate that the cell comprises a particular analyte.
  • the reporter oligonucleotides and labelling agents may allow multi -analyte, multiplexed analyses to be performed.
  • these reporter oligonucleotides may comprise nucleic acid barcode sequences that permit identification of the labelling agent which the reporter oligonucleotide is coupled to.
  • the use of oligonucleotides as the reporter may provide advantages of being able to generate significant diversity in terms of sequence, while also being readily attachable to most biomolecules, e.g., antibodies, etc., as well as being readily detected, e.g., using sequencing or array technologies.
  • Attachment (coupling) of the reporter oligonucleotides to the labelling agents may be achieved through any of a variety of direct or indirect, covalent or non-covalent associations or attachments.
  • oligonucleotides may be covalently attached to a portion of a labelling agent (such a protein, e.g., an antibody or antibody fragment) using chemical conjugation techniques (e.g., Lightning-Link® antibody labelling kits available from Innova Biosciences), as well as other non-covalent attachment mechanisms, e.g., using biotinylated antibodies and oligonucleotides (or beads that include one or more biotinylated linker, coupled to oligonucleotides) with an avidin or streptavidin linker.
  • a labelling agent such as a protein, e.g., an antibody or antibody fragment
  • chemical conjugation techniques e.g., Lightning-Link® antibody labelling kits available from Innova Biosciences
  • other non-covalent attachment mechanisms
  • Antibody and oligonucleotide biotinylation techniques are available. See, e.g., Fang, et al., “Fluoride-Cleavable Biotinylation Phosphoramidite for 5'-end-Labelling and Affinity Purification of Synthetic Oligonucleotides,” Nucleic Acids Res. Jan. 15, 2003; 31 (2):708-715, which is entirely incorporated herein by reference for all purposes. Likewise, protein and peptide biotinylation techniques have been developed and are readily available. See, e.g., U.S. Pat. No. 6,265,552, which is entirely incorporated herein by reference for all purposes.
  • click reaction chemistry such as a Methyltetrazine-PEG5-NHS Ester reaction, a TCO-PEG4-NHS Ester reaction, or the like, may be used to couple reporter oligonucleotides to labelling agents.
  • Commercially available kits such as those from Thunderlink and Abeam, and techniques common in the art may be used to couple reporter oligonucleotides to labelling agents as appropriate.
  • a labelling agent is indirectly (e.g., via hybridization) coupled to a reporter oligonucleotide comprising a barcode sequence that identifies the label agent.
  • the labelling agent may be directly coupled (e g., covalently bound) to a hybridization oligonucleotide that comprises a sequence that hybridizes with a sequence of the reporter oligonucleotide.
  • Hybridization of the hybridization oligonucleotide to the reporter oligonucleotide couples the labelling agent to the reporter oligonucleotide.
  • the reporter oligonucleotides are releasable from the labelling agent, such as upon application of a stimulus.
  • the reporter oligonucleotide may be attached to the labeling agent through a labile bond (e.g., chemically labile, photolabile, thermally labile, etc.) as generally described for releasing molecules from supports elsewhere herein.
  • the reporter oligonucleotides described herein may include one or more functional sequences that can be used in subsequent processing, such as an adapter sequence, a unique molecular identifier (UMI) sequence, a sequencer specific flow cell attachment sequence (such as an P5, P7, or partial P5 or P7 sequence), a primer or primer binding sequence, a sequencing primer or primer biding sequence (such as an Rl, R2, or partial R1 or R2 sequence).
  • UMI unique molecular identifier
  • the labelling agent can comprise a reporter oligonucleotide and a label.
  • a label can be fluorophore, a radioisotope, a molecule capable of a colorimetric reaction, a magnetic particle, or any other suitable molecule or compound capable of detection.
  • the label can be conjugated to a labelling agent (or reporter oligonucleotide) either directly or indirectly (e.g., the label can be conjugated to a molecule that can bind to the labelling agent or reporter oligonucleotide).
  • a label is conjugated to an oligonucleotide that is complementary to a sequence of the reporter oligonucleotide, and the oligonucleotide may be allowed to hybridize to the reporter oligonucleotide.
  • FIG. 11 describes example labelling agents (1110, 1120, 1130) comprising reporter oligonucleotides (1140) attached thereto.
  • Labelling agent 1110 e.g., any of the labelling agents described herein
  • reporter oligonucleotide 1140 may comprise barcode sequence 1142 that identifies labelling agent 1110.
  • Reporter oligonucleotide 1140 may also comprise one or more functional sequences that can be used in subsequent processing, such as an adapter sequence, a unique molecular identifier (UMI) sequence, a sequencer specific flow cell attachment sequence (such as an P5, P7, or partial P5 or P7 sequence), a primer or primer binding sequence, or a sequencing primer or primer biding sequence (such as an Rl, R2, or partial Rl or R2 sequence).
  • UMI unique molecular identifier
  • sequencer specific flow cell attachment sequence such as an P5, P7, or partial P5 or P7 sequence
  • primer or primer binding sequence such as an Rl, R2, or partial Rl or R2 sequence.
  • reporter oligonucleotide 1140 conjugated to a labelling agent comprises a primer sequence 1141, a barcode sequence that identifies the labelling agent (e.g., 1110, 1120, 1130), and functional sequence 1143.
  • Functional sequence 1143 may be configured to hybridize to a complementary sequence, such as a complementary sequence present on a nucleic acid barcode molecule 1190 (not shown), such as those described elsewhere herein.
  • nucleic acid barcode molecule 1190 is attached to a support (e.g., a bead, such as a gel bead), such as those described elsewhere herein.
  • nucleic acid barcode molecule 1190 may be attached to the support via a releasable linkage (e g., comprising a labile bond), such as those described elsewhere herein.
  • reporter oligonucleotide 1140 comprises one or more additional functional sequences, such as those described above.
  • the labelling agent 1110 is a protein or polypeptide (e.g., an antigen or prospective antigen) comprising reporter oligonucleotide 1140.
  • Reporter oligonucleotide 1140 comprises barcode sequence 1142 that identifies polypeptide 1110 and can be used to infer the presence of an analyte, e.g., a binding partner of polypeptide 1110 (e.g., a molecule or compound to which polypeptide 1110 can bind).
  • the labelling agent 1110 is a lipophilic moiety (e.g., cholesterol) comprising reporter oligonucleotide 1140, where the lipophilic moiety is selected such that labelling agent 1110 integrates into a membrane of a cell or nucleus.
  • Reporter oligonucleotide 1140 comprises barcode sequence 1142 that identifies lipophilic moiety 1110 which in some instances is used to tag cells (e.g., groups of cells, cell samples, etc.) and may be used for multiplex analyses as described elsewhere herein.
  • the labelling agent is an antibody 1120 (or an epitope binding fragment thereof) comprising reporter oligonucleotide 1140.
  • Reporter oligonucleotide 1140 comprises barcode sequence 1142 that identifies antibody 1120 and can be used to infer the presence of, e.g., a target of antibody 1120 (e.g., a molecule or compound to which antibody 1120 binds).
  • labelling agent 1130 comprises an MHC molecule 1131 comprising peptide 1132 and reporter oligonucleotide 1140 that identifies peptide 1132.
  • the MHC molecule is coupled to a support 1133.
  • support 1133 may be a polypeptide, such as streptavidin, or a polysaccharide, such as dextran.
  • reporter oligonucleotide 1140 may be directly or indirectly coupled to MHC labelling agent 1130 in any suitable manner.
  • reporter oligonucleotide 1140 may be coupled to MHC molecule 1131, support 1133, or peptide 1132.
  • labelling agent 1130 comprises a plurality of MHC molecules, (e g. is an MHC multimer, which may be coupled to a support (e.g., 1133)).
  • Class I and/or Class II MHC multimers that can be utilized with the compositions, methods, and systems disclosed herein, e.g., MHC tetramers, MHC pentamers (MHC assembled via a coiled-coil domain, e.g., Pro5® MHC Class I Pentamers, (Prolmmune, Ltd.), MHC octamers, MHC dodecamers, MHC decorated dextran molecules (e.g., MHC Dextramer® (Immudex)), etc.
  • labelling agents including antibody and MHC -based labelling agents, reporter oligonucleotides, and methods of use, see, e g., U.S. Pat.
  • FIG. 13 illustrates another example of a barcode carrying bead.
  • analysis of multiple analytes may comprise nucleic acid barcode molecules as generally depicted in FIG. 13.
  • nucleic acid barcode molecules 1310 and 1320 are attached to support 1330 via a releasable linkage 1340 (e.g., comprising a labile bond) as described elsewhere herein.
  • Nucleic acid barcode molecule 1310 may comprise adapter sequence 1311, barcode sequence 1312 and adapter sequence 1313.
  • Nucleic acid barcode molecule 1320 may comprise adapter sequence 1321, barcode sequence 1312, and adapter sequence 1323, wherein adapter sequence 1323 comprises a different sequence than adapter sequence 1313.
  • adapter 1311 and adapter 1321 comprise the same sequence.
  • adapter 1311 and adapter 1321 comprise different sequences.
  • support 1330 is shown comprising nucleic acid barcode molecules 1310 and 1320, any suitable number of barcode molecules comprising common barcode sequence 1312 are contemplated herein.
  • support 1330 further comprises nucleic acid barcode molecule 1350.
  • Nucleic acid barcode molecule 1350 may comprise adapter sequence 1351, barcode sequence 1312 and adapter sequence 1353, wherein adapter sequence 1353 comprises a different sequence than adapter sequence 1313 and 1323.
  • nucleic acid barcode molecules e.g., 1310, 1320, 1350
  • nucleic acid barcode molecules 1310, 1320 or 1350 may interact with analytes as described elsewhere herein, for example, as depicted in FIGs. 12A-C. [00359] Referring to FIG.
  • sequence 1223 may be complementary to an adapter sequence of a reporter oligonucleotide.
  • Cells or nuclei or cell beads
  • labelling agents 1210 e.g., polypeptide, antibody, or others described elsewhere herein.
  • the cells or nuclei or cell beads
  • a cell that is bound to labelling agent 1210 which is conjugated to oligonucleotide 1220 and support 1230 e.g., a bead, such as a gel bead
  • nucleic acid barcode molecule 1290 is partitioned into a partition amongst a plurality of partitions (e.g., a droplet of a droplet emulsion or a well of a microwell array).
  • the partition comprises at most a single cell bound to labelling agent 1210.
  • reporter oligonucleotide 1220 conjugated to labelling agent 1210 comprises a first adapter sequence 1211 (e.g., a primer sequence), a barcode sequence 1212 that identifies the labelling agent 1210 (e g., the polypeptide, antibody, or peptide of a pMHC molecule or complex), and an adapter sequence 1213.
  • Adapter sequence 1213 may be configured to hybridize to a complementary sequence, such as sequence 1223 present on a nucleic acid barcode molecule 1290.
  • oligonucleotide 1220 comprises one or more additional functional sequences, such as those described elsewhere herein.
  • Barcoded nucleic may be generated (e.g., via a nucleic acid reaction, such as nucleic acid extension or ligation) from the constructs described in FIGs. 12A-C.
  • sequence 1213 may then be hybridized to complementary sequence 1223 to generate (e.g., via a nucleic acid reaction, such as nucleic acid extension or ligation) a barcoded nucleic acid molecule comprising cell (e.g., partition specific) barcode sequence 1222 (or a reverse complement thereof) and reporter barcode sequence 1212 (or a reverse complement thereof).
  • Barcoded nucleic acid molecules can then be optionally processed as described elsewhere herein, e.g., to amplify the molecules and/or append sequencing platform specific sequences to the fragments. See, e.g., U.S. Pat. Pub. 2018/0105808, which is hereby entirely incorporated by reference for all purposes. Barcoded nucleic acid molecules, or derivatives generated therefrom, can then be sequenced on a suitable sequencing platform.
  • analysis of multiple analytes may be performed.
  • the workflow may comprise a workflow as generally depicted in any of FIGs. 12A-C, or a combination of workflows for an individual analyte, as described elsewhere herein.
  • the workflow may comprise a workflow as generally depicted in any of FIGs. 12A-C, or a combination of workflows for an individual analyte, as described elsewhere herein.
  • multiple analytes can be analyzed.
  • analysis of an analyte comprises a workflow as generally depicted in FIG. 12A.
  • a nucleic acid barcode molecule 1290 may be co-partitioned with the one or more analytes.
  • nucleic acid barcode molecule 1290 is attached to a support 1230 (e.g., a bead, such as a gel bead), such as those described elsewhere herein.
  • nucleic acid barcode molecule 1290 may be attached to support 1230 via a releasable linkage 1240 (e.g., comprising a labile bond), such as those described elsewhere herein.
  • Nucleic acid barcode molecule 1290 may comprise a barcode sequence 1221 and optionally comprise other additional sequences, for example, a UMI sequence 1222 (or other functional sequences described elsewhere herein).
  • the nucleic acid barcode molecule 1290 may comprise a sequence 1223 that may be complementary to another nucleic acid sequence, such that it may hybridize to a particular sequence. [00363]
  • sequence 1223 may comprise a poly-T sequence and may be used to hybridize to mRNA. Referring to FIG.
  • nucleic acid barcode molecule 1290 comprises sequence 1223 complementary to a sequence of RNA molecule 1260 from a cell.
  • sequence 1223 comprises a sequence specific for an RNA molecule.
  • Sequence 1223 may comprise a known or targeted sequence or a random sequence.
  • a nucleic acid extension reaction may be performed, thereby generating a barcoded nucleic acid product comprising sequence 1223, the barcode sequence 1221, UMI sequence 1222, any other functional sequence, and a sequence corresponding to the RNA molecule 1260.
  • sequence 1223 may be complementary to an overhang sequence or an adapter sequence that has been appended to an analyte.
  • primer 1250 comprises a sequence complementary to a sequence of nucleic acid molecule 1260 (such as an RNA encoding for a BCR sequence) from a biological particle.
  • primer 1250 comprises one or more sequences 1251 that are not complementary to RNA molecule 1260.
  • Sequence 1251 may be a functional sequence as described elsewhere herein, for example, an adapter sequence, a sequencing primer sequence, or a sequence the facilitates coupling to a flow cell of a sequencer.
  • primer 1250 comprises a poly-T sequence.
  • primer 1250 comprises a sequence complementary to a target sequence in an RNA molecule. In some instances, primer 1250 comprises a sequence complementary to a region of an immune molecule, such as the constant region of a TCR or BCR sequence.
  • Primer 1250 is hybridized to nucleic acid molecule 1260 and complementary molecule 1270 is generated (see Panel 1202).
  • complementary molecule 1270 may be cDNA generated in a reverse transcription reaction.
  • an additional sequence may be appended to complementary molecule 1270.
  • the reverse transcriptase enzyme may be selected such that several non-templated bases 1280 (e.g., a poly-C sequence) are appended to the cDNA.
  • Nucleic acid barcode molecule 1290 comprises a sequence 1224 complementary to the non-templated bases, and the reverse transcriptase performs a template switching reaction onto nucleic acid barcode molecule 1290 to generate a barcoded nucleic acid molecule comprising cell (e.g., partition specific) barcode sequence 1222 (or a reverse complement thereof) and a sequence of complementary molecule 1270 (or a portion thereof).
  • sequence 1223 comprises a sequence complementary to a region of an immune molecule, such as the constant region of a TCR or BCR sequence. Sequence 1223 is hybridized to nucleic acid molecule 1260 and a complementary molecule 1270 is generated.
  • complementary molecule 1270 may be generated in a reverse transcription reaction generating a barcoded nucleic acid molecule comprising cell (e g., partition specific) barcode sequence 1222 (or a reverse complement thereof) and a sequence of complementary molecule 1270 (or a portion thereof). Additional methods and compositions suitable for barcoding cDNA generated from mRNA transcripts including those encoding V(D)J regions of an immune cell receptor and/or barcoding methods and composition including a template switch oligonucleotide are described in International Patent Application WO2018/075693, U.S. Patent Publication No. 2018/0105808, U.S. Patent Publication No. 2015/0376609, filed June 26, 2015, and U.S. Patent Publication No. 2019/0367969, , each of which applications is herein entirely incorporated by reference for all purposes.
  • biological particles may be partitioned along with lysis reagents in order to release the contents of the biological particles within the partition.
  • the lysis agents can be contacted with the biological particle suspension concurrently with, or immediately prior to, the introduction of the biological particles into the partitioning junction/droplet generation zone (e.g., junction 210), such as through an additional channel or channels upstream of the channel junction.
  • biological particles may be partitioned along with other reagents, as will be described further below.
  • the methods and systems of the present disclosure may comprise microfluidic devices and methods of use thereof, which may be used for co-partitioning biological particles or biological particles with reagents.
  • microfluidic devices and methods of use thereof may be used for co-partitioning biological particles or biological particles with reagents.
  • Such systems and methods are described in U.S. Patent Publication No. US/20190367997, which is herein incorporated by reference in its entirety for all purposes.
  • the lysis reagents can facilitate the release of the contents of the biological particles within the partition.
  • the contents released in a partition may remain discrete from the contents of other partitions.
  • the channel segments of the microfluidic devices described elsewhere herein may be coupled to any of a variety of different fluid sources or receiving components, including reservoirs, tubing, manifolds, or fluidic components of other systems.
  • the microfluidic channel structures may have various geometries and/or configurations.
  • a microfluidic channel structure can have more than two channel junctions.
  • a microfluidic channel structure can have 2, 3, 4, 5 channel segments or more each carrying the same or different types of beads, reagents, and/or biological particles that meet at a channel junction. Fluid flow in each channel segment may be controlled to control the partitioning of the different elements into droplets.
  • Fluid may be directed flow along one or more channels or reservoirs via one or more fluid flow units.
  • a fluid flow unit can comprise compressors (e.g., providing positive pressure), pumps (e.g., providing negative pressure), actuators, and the like to control flow of the fluid. Fluid may also or otherwise be controlled via applied pressure differentials, centrifugal force, electrokinetic pumping, vacuum, capillary or gravity flow, or the like.
  • lysis agents include bioactive reagents, such as lysis enzymes that are used for lysis of different cell types, e.g., gram positive or negative bacteria, plants, yeast, mammalian, etc., such as lysozymes, achromopeptidase, lysostaphin, labiase, kitalase, lyticase, and a variety of other lysis enzymes available from, e.g., Sigma-Aldrich, Inc. (St Louis, MO), as well as other commercially available lysis enzymes.
  • Other lysis agents may additionally or alternatively be co-partitioned with the biological particles to cause the release of the biological particle’s contents into the partitions.
  • surfactant-based lysis solutions may be used to lyse cells, although these may be less desirable for emulsion based systems where the surfactants can interfere with stable emulsions.
  • lysis solutions may include non-ionic surfactants such as, for example, TritonX-100 and Tween 20.
  • lysis solutions may include ionic surfactants such as, for example, sarcosyl and sodium dodecyl sulfate (SDS).
  • Electroporation, thermal, acoustic or mechanical cellular disruption may also be used in certain cases, e.g., non-emulsion based partitioning such as encapsulation of biological particles that may be in addition to or in place of droplet partitioning, where any pore size of the encapsulate is sufficiently small to retain nucleic acid fragments of a given size, following cellular disruption.
  • non-emulsion based partitioning such as encapsulation of biological particles that may be in addition to or in place of droplet partitioning, where any pore size of the encapsulate is sufficiently small to retain nucleic acid fragments of a given size, following cellular disruption.
  • reagents can also be co-partitioned with the biological particles, including, for example, DNase and RNase inactivating agents or inhibitors, such as proteinase K, chelating agents, such as EDTA, and other reagents employed in removing or otherwise reducing negative activity or impact of different cell lysate components on subsequent processing of nucleic acids.
  • DNase and RNase inactivating agents or inhibitors such as proteinase K
  • chelating agents such as EDTA
  • the biological particles may be exposed to an appropriate stimulus to release the biological particles or their contents from a co-partitioned support (e.g., bead).
  • a chemical stimulus may be co-partitioned along with an encapsulated biological particle to allow for the degradation of the support and release of the cell or its contents into the larger partition.
  • this stimulus may be the same as the stimulus described elsewhere herein for release of nucleic acid molecules (e.g., oligonucleotides) from their respective support (e.g., bead).
  • this may be a different and non- overlapping stimulus, in order to allow an encapsulated biological particle to be released into a partition at a different time from the release of nucleic acid molecules into the same partition.
  • compositions, and systems for encapsulating cells also referred to as a “cell bead”
  • a cell bead For a description of methods, compositions, and systems for encapsulating cells (also referred to as a “cell bead”), see, e.g., U.S. Pat. 10,428,326 and U.S. Pat. Pub. 20190100632, which are each incorporated by reference in their entirety.
  • Additional reagents may also be co-partitioned with the biological particles, such as endonucleases to fragment a biological particle’s DNA, DNA polymerase enzymes and dNTPs used to amplify the biological particle’s nucleic acid fragments and to attach the barcode molecular tags to the amplified fragments.
  • Other enzymes may be co-partitioned, including without limitation, polymerase, transposase, ligase, proteinase K, DNAse, etc.
  • Additional reagents may also include reverse transcriptase enzymes, including enzymes with terminal transferase activity, primers and oligonucleotides, and switch oligonucleotides (also referred to herein as “switch oligos” or “template switching oligonucleotides”) which can be used for template switching.
  • reverse transcriptase enzymes including enzymes with terminal transferase activity
  • primers and oligonucleotides include primers and oligonucleotides, and switch oligonucleotides (also referred to herein as “switch oligos” or “template switching oligonucleotides”) which can be used for template switching.
  • switch oligonucleotides also referred to herein as “switch oligos” or “template switching oligonucleotides” which can be used for template switching.
  • template switching can be used to increase the length of a cDNA.
  • template switching can
  • cDNA can be generated from reverse transcription of a template, e.g., cellular mRNA, where a reverse transcriptase with terminal transferase activity can add additional nucleotides, e.g., polyC, to the cDNA in a template independent manner.
  • Switch oligos can include sequences complementary to the additional nucleotides, e.g., polyG.
  • the additional nucleotides (e.g., polyC) on the cDNA can hybridize to the additional nucleotides (e g., polyG) on the switch oligo, whereby the switch oligo can be used by the reverse transcriptase as template to further extend the cDNA.
  • Template switching oligonucleotides may comprise a hybridization region and a template region.
  • the hybridization region can comprise any sequence capable of hybridizing to the target.
  • the hybridization region comprises a series of G bases to complement the overhanging C bases at the 3’ end of a cDNA molecule.
  • the series of G bases may comprise 1 G base, 2 G bases, 3 G bases, 4 G bases, 5 G bases or more than 5 G bases.
  • the template sequence can comprise any sequence to be incorporated into the cDNA.
  • the template region comprises at least 1 (e.g., at least 2, 3, 4, 5 or more) tag sequences and/or functional sequences.
  • Switch oligos may comprise deoxyribonucleic acids; ribonucleic acids; modified nucleic acids including 2-Aminopurine, 2,6-Diaminopurine (2-Amino-dA), inverted dT, 5-Methyl dC, 2 ’-deoxy Inosine, Super T (5-hydroxybutynl-2’-deoxyuridine), Super G (8-aza- 7-deazaguanosine), locked nucleic acids (LNAs), unlocked nucleic acids (UNAs, e g., UNA-A, UNA-U, UNA-C, UNA-G), Iso-dG, Iso-dC, 2’ Fluoro bases (e.g., Fluoro C, Fluoro U, Fluoro A, and Fluoro G), or any combination. [00372] In some cases, the length of a switch oligo may be at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,
  • the length of a switch oligo may be at most about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35,
  • the macromolecular components e.g., macromolecular constituents of biological particles, such as RNA, DNA, or proteins
  • the macromolecular component contents of individual biological particles can be provided with unique identifiers such that, upon characterization of those macromolecular components they may be attributed as having been derived from the same biological particle or particles.
  • unique identifiers such that, upon characterization of those macromolecular components they may be attributed as having been derived from the same biological particle or particles.
  • the ability to attribute characteristics to individual biological particles or groups of biological particles is provided by the assignment of unique identifiers specifically to an individual biological particle or groups of biological particles.
  • Unique identifiers in the form of nucleic acid barcodes can be assigned or associated with individual biological particles or populations of biological particles, in order to tag or label the biological particle’s macromolecular components (and as a result, its characteristics) with the unique identifiers. These unique identifiers can then be used to attribute the biological particle’s components and characteristics to an individual biological particle or group of biological particles.
  • this is performed by co-partitioning the individual biological particle or groups of biological particles with the unique identifiers, such as described above (with reference to FIG. 2).
  • the unique identifiers are provided in the form of nucleic acid molecules (e.g., oligonucleotides) that comprise nucleic acid barcode sequences that may be attached to or otherwise associated with the nucleic acid contents of individual biological particle, or to other components of the biological particle, and particularly to fragments of those nucleic acids.
  • the nucleic acid molecules are partitioned such that as between nucleic acid molecules in a given partition, the nucleic acid barcode sequences contained therein are the same, but as between different partitions, the nucleic acid molecule can, and do have differing barcode sequences, or at least represent a large number of different barcode sequences across all of the partitions in a given analysis. In some aspects, only one nucleic acid barcode sequence can be associated with a given partition, although in some cases, two or more different barcode sequences may be present.
  • the nucleic acid barcode sequences can include from about 6 to about 20 or more nucleotides within the sequence of the nucleic acid molecules (e.g., oligonucleotides).
  • the nucleic acid barcode sequences can include from about 6 to about 20, 30, 40, 50, 60, 70, 80, 90, 100 or more nucleotides.
  • the length of a barcode sequence may be about 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 nucleotides or longer.
  • the length of a barcode sequence may be at least about 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 nucleotides or longer.
  • the length of a barcode sequence may be at most about 6, 7,
  • nucleotides 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 nucleotides or shorter. These nucleotides may be completely contiguous, e.g., in a single stretch of adjacent nucleotides, or they may be separated into two or more separate subsequences that are separated by 1 or more nucleotides. In some cases, separated barcode subsequences can be from about 4 to about 16 nucleotides in length. In some cases, the barcode subsequence may be about 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16 nucleotides or longer. In some cases, the barcode subsequence may be at least about 4, 5, 6, 7, 8,
  • the co-partitioned nucleic acid molecules can also comprise other functional sequences useful in the processing of the nucleic acids from the co-partitioned biological particles.
  • sequences include, e.g., targeted or random/universal amplification primer sequences for amplifying nucleic acids (e.g., mRNA, the genomic DNA) from the individual biological particles within the partitions while attaching the associated barcode sequences, sequencing primers or primer recognition sites, hybridization or probing sequences, e.g., for identification of presence of the sequences or for pulling down barcoded nucleic acids, or any of a number of other potential functional sequences.
  • nucleic acids e.g., mRNA, the genomic DNA
  • oligonucleotides may also be employed, including, e.g., coalescence of two or more droplets, where one droplet contains oligonucleotides, or microdispensing of oligonucleotides (e.g., attached to a bead) into partitions, e.g., droplets within microfluidic systems.
  • supports such as beads
  • each include large numbers of the above described barcoded nucleic acid molecules (e.g., barcoded oligonucleotides) releasably attached to the beads, where all of the nucleic acid molecules attached to a particular bead will include the same nucleic acid barcode sequence, but where a large number of diverse barcode sequences are represented across the population of beads used.
  • barcoded nucleic acid molecules e.g., barcoded oligonucleotides
  • hydrogel beads e.g., comprising polyacrylamide polymer matrices
  • hydrogel beads are used as a solid support and delivery vehicle for the nucleic acid molecules into the partitions, as they are capable of carrying large numbers of nucleic acid molecules, and may be configured to release those nucleic acid molecules upon exposure to a particular stimulus, as described elsewhere herein.
  • the population of beads provides a diverse barcode sequence library that includes at least about 1,000 different barcode sequences, at least about 5,000 different barcode sequences, at least about 10,000 different barcode sequences, at least about 50,000 different barcode sequences, at least about 100,000 different barcode sequences, at least about 1,000,000 different barcode sequences, at least about 5,000,000 different barcode sequences, or at least about 10,000,000 different barcode sequences, or more.
  • each bead can be provided with large numbers of nucleic acid (e.g., oligonucleotide) molecules attached.
  • the number of molecules of nucleic acid molecules including the barcode sequence on an individual bead can be at least about 1,000 nucleic acid molecules, at least about 5,000 nucleic acid molecules, at least about 10,000 nucleic acid molecules, at least about 50,000 nucleic acid molecules, at least about 100,000 nucleic acid molecules, at least about 500,000 nucleic acids, at least about 1,000,000 nucleic acid molecules, at least about 5,000,000 nucleic acid molecules, at least about 10,000,000 nucleic acid molecules, at least about 50,000,000 nucleic acid molecules, at least about 100,000,000 nucleic acid molecules, at least about 250,000,000 nucleic acid molecules and in some cases at least about 1 billion nucleic acid molecules, or more.
  • Nucleic acid molecules of a given bead can include identical (or common) barcode sequences, different barcode sequences, or a combination of both. Nucleic acid molecules of a given bead can include multiple sets of nucleic acid molecules. Nucleic acid molecules of a given set can include identical barcode sequences. The identical barcode sequences can be different from barcode sequences of nucleic acid molecules of another set.
  • the resulting population of partitions can also include a diverse barcode library that includes at least about 1,000 different barcode sequences, at least about 5,000 different barcode sequences, at least about 10,000 different barcode sequences, at least at least about 50,000 different barcode sequences, at least about 100,000 different barcode sequences, at least about 1,000,000 different barcode sequences, at least about 5,000,000 different barcode sequences, or at least about 10,000,000 different barcode sequences.
  • each partition of the population can include at least about 1,000 nucleic acid molecules, at least about 5,000 nucleic acid molecules, at least about 10,000 nucleic acid molecules, at least about 50,000 nucleic acid molecules, at least about 100,000 nucleic acid molecules, at least about 500,000 nucleic acids, at least about 1,000,000 nucleic acid molecules, at least about 5,000,000 nucleic acid molecules, at least about 10,000,000 nucleic acid molecules, at least about 50,000,000 nucleic acid molecules, at least about 100,000,000 nucleic acid molecules, at least about 250,000,000 nucleic acid molecules and in some cases at least about 1 billion nucleic acid molecules.
  • a mixed, but known set of barcode sequences may provide greater assurance of identification in the subsequent processing, e.g., by providing a stronger address or attribution of the barcodes to a given partition, as a duplicate or independent confirmation of the output from a given partition.
  • the nucleic acid molecules are releasable from the beads upon the application of a particular stimulus to the beads.
  • the stimulus may be a photo-stimulus, e.g., through cleavage of a photo-labile linkage that releases the nucleic acid molecules.
  • a thermal stimulus may be used, where elevation of the temperature of the beads environment will result in cleavage of a linkage or other release of the nucleic acid molecules from the beads.
  • a chemical stimulus can be used that cleaves a linkage of the nucleic acid molecules to the beads, or otherwise results in release of the nucleic acid molecules from the beads.
  • such compositions include the polyacrylamide matrices described above for encapsulation of biological particles, and may be degraded for release of the attached nucleic acid molecules through exposure to a reducing agent, such as DTT.
  • Droplet size may be controlled by adjusting certain geometric features in channel architecture (e.g., microfluidics channel architecture). For example, an expansion angle, width, and/or length of a channel may be adjusted to control droplet size.
  • channel architecture e.g., microfluidics channel architecture
  • FIG. 2 shows an example of a microfluidic channel structure for the controlled partitioning of beads into discrete droplets.
  • a channel structure 200 can include a channel segment 202 communicating at a channel junction 206 (or intersection) with a reservoir 204.
  • the reservoir 204 can be a chamber. Any reference to “reservoir,” as used herein, can also refer to a “chamber.”
  • an aqueous fluid 208 that includes suspended beads 212 may be transported along the channel segment 202 into the junction 206 to meet a second fluid 210 that is immiscible with the aqueous fluid 208 in the reservoir 204 to create droplets 216, 218 of the aqueous fluid 208 flowing into the reservoir 204.
  • droplets can form based on factors such as the hydrodynamic forces at the junction 206, flow rates of the two fluids 208, 210, fluid properties, and certain geometric parameters (e.g., w, ho, a, etc.) of the channel structure 200.
  • a plurality of droplets can be collected in the reservoir 204 by continuously injecting the aqueous fluid 208 from the channel segment 202 through the junction 206.
  • a discrete droplet generated may include a bead (e.g., as in occupied droplets 216). Alternatively, a discrete droplet generated may include more than one bead. Alternatively, a discrete droplet generated may not include any beads (e.g., as in unoccupied droplet 218). In some instances, a discrete droplet generated may contain one or more biological particles, as described elsewhere herein. In some instances, a discrete droplet generated may comprise one or more reagents, as described elsewhere herein.
  • the aqueous fluid 208 can have a substantially uniform concentration or frequency of beads 212.
  • the beads 212 can be introduced into the channel segment 202 from a separate channel (not shown in FIG. 2).
  • the frequency of beads 212 in the channel segment 202 may be controlled by controlling the frequency in which the beads 212 are introduced into the channel segment 202 and/or the relative flow rates of the fluids in the channel segment 202 and the separate channel.
  • the beads can be introduced into the channel segment 202 from a plurality of different channels, and the frequency controlled accordingly.
  • the aqueous fluid 208 in the channel segment 202 can comprise biological particles.
  • the aqueous fluid 208 can have a substantially uniform concentration or frequency of biological particles.
  • the biological particles can be introduced into the channel segment 202 from a separate channel.
  • the frequency or concentration of the biological particles in the aqueous fluid 208 in the channel segment 202 may be controlled by controlling the frequency in which the biological particles are introduced into the channel segment 202 and/or the relative flow rates of the fluids in the channel segment 202 and the separate channel.
  • the biological particles can be introduced into the channel segment 202 from a plurality of different channels, and the frequency controlled accordingly.
  • a first separate channel can introduce beads and a second separate channel can introduce biological particles into the channel segment 202. The first separate channel introducing the beads may be upstream or downstream of the second separate channel introducing the biological particles.
  • the second fluid 210 can comprise an oil, such as a fluorinated oil, that includes a fluorosurfactant for stabilizing the resulting droplets, for example, inhibiting subsequent coalescence of the resulting droplets.
  • an oil such as a fluorinated oil, that includes a fluorosurfactant for stabilizing the resulting droplets, for example, inhibiting subsequent coalescence of the resulting droplets.
  • the second fluid 210 may not be subjected to and/or directed to any flow in or out of the reservoir 204.
  • the second fluid 210 may be substantially stationary in the reservoir 204.
  • the second fluid 210 may be subjected to flow within the reservoir 204, but not in or out of the reservoir 204, such as via application of pressure to the reservoir 204 and/or as affected by the incoming flow of the aqueous fluid 208 at the junction 206.
  • the second fluid 210 may be subjected and/or directed to flow in or out of the reservoir 204.
  • the reservoir 204 can be a channel directing the second fluid 210 from upstream to downstream, transporting the generated droplets.
  • the channel structure 200 at or near the junction 206 may have certain geometric features that at least partly determine the sizes of the droplets formed by the channel structure 200.
  • the channel segment 202 can have a height, ho and width, w, at or near the junction 206.
  • the channel segment 202 can comprise a rectangular cross-section that leads to a reservoir 204 having a wider cross-section (such as in width or diameter).
  • the cross-section of the channel segment 202 can be other shapes, such as a circular shape, trapezoidal shape, polygonal shape, or any other shapes.
  • the top and bottom walls of the reservoir 204 at or near the junction 206 can be inclined at an expansion angle, a.
  • the expansion angle, a allows the tongue (portion of the aqueous fluid 208 leaving channel segment 202 at junction 206 and entering the reservoir 204 before droplet formation) to increase in depth and facilitate decrease in curvature of the intermediately formed droplet.
  • Droplet size may decrease with increasing expansion angle.
  • the resulting droplet radius, Rd may be predicted by the following equation for the aforementioned geometric parameters of ho, w, and a. ( i - w ⁇ ho
  • the predicted droplet size is 121 pm.
  • the predicted droplet size is 123 pm.
  • the predicted droplet size is 124 pm.
  • the expansion angle, a may be between a range of from about 0.5° to about 4°, from about 0.1° to about 10°, or from about 0° to about 90°.
  • the expansion angle can be at least about 0.01°, 0.1°, 0.2°, 0.3°, 0.4°, 0.5°, 0.6°, 0.7°, 0.8°, 0.9°, 1°, 2°, 3°, 4°, 5°, 6°, 7°, 8°, 9°, 10°, 15°, 20°, 25°, 30°, 35°, 40°, 45°, 50°, 55°, 60°, 65°, 70°, 75°, 80°, 85°, or higher.
  • the expansion angle can be at most about 89°, 88°, 87°, 86°, 85°, 84°, 83°, 82°, 81°, 80°, 75°, 70°, 65°, 60°, 55°, 50°, 45°, 40°, 35°, 30°, 25°, 20°, 15°, 10°, 9°, 8°, 7°, 6°, 5°, 4°, 3°, 2°, 1°, 0.1°, 0.01°, or less.
  • the width, w can be between a range of from about 100 micrometers (pm) to about 500 pm. In some instances, the width, w, can be between a range of from about 10 pm to about 200 pm.
  • the width can be less than about 10 pm. Alternatively, the width can be greater than about 500 pm.
  • the flow rate of the aqueous fluid 208 entering the junction 206 can be between about 0.04 microliters (pL)/minute (min) and about 40 pL/min. In some instances, the flow rate of the aqueous fluid 208 entering the junction 206 can be between about 0.01 microliters (pL)/minute (min) and about 100 pL/min. Alternatively, the flow rate of the aqueous fluid 208 entering the junction 206 can be less than about 0.01 pL/min.
  • the flow rate of the aqueous fluid 208 entering the junction 206 can be greater than about 40 pL/min, such as 45 pL/min, 50 pL/min, 55 pL/min, 60 pL/min, 65 pL/min, 70 pL/min, 75 pL/min, 80 pL/min, 85 pL/min, 90 pL/min, 95 pL/min, 100 pL/min, 110 pL/min , 120 pL/min , 130 pL/min , 140 pL/min , 150 pL/min, or greater.
  • the droplet radius may not be dependent on the flow rate of the aqueous fluid 208 entering the junction 206.
  • At least about 50% of the droplets generated can have uniform size. In some instances, at least about 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or greater of the droplets generated can have uniform size. Alternatively, less than about 50% of the droplets generated can have uniform size.
  • the throughput of droplet generation can be increased by increasing the points of generation, such as increasing the number of junctions (e.g., junction 206) between aqueous fluid 208 channel segments (e.g., channel segment 202) and the reservoir 204.
  • the throughput of droplet generation can be increased by increasing the flow rate of the aqueous fluid 208 in the channel segment 202.
  • the methods and systems described herein may be used to greatly increase the efficiency of single cell applications and/or other applications receiving droplet-based input.
  • subsequent operations that can be performed can include generation of amplification products, purification (e.g., via solid phase reversible immobilization (SPRI)), further processing (e.g., shearing, ligation of functional sequences, and subsequent amplification (e.g., via PCR)).
  • SPRI solid phase reversible immobilization
  • further processing e.g., shearing, ligation of functional sequences
  • subsequent amplification e.g., via PCR
  • Additional reagents that may be co-partitioned along with the barcode bearing bead may include oligonucleotides to block ribosomal RNA (rRNA) and nucleases to digest genomic DNA from cells. Alternatively, rRNA removal agents may be applied during additional processing operations.
  • the configuration of the constructs generated by such a method can help minimize (or avoid) sequencing of the poly-T sequence during sequencing and/or sequence the 5’ end of a polynucleotide sequence.
  • the amplification products for example, first amplification products and/or second amplification products, may be subject to sequencing for sequence analysis. In some cases, amplification may be performed using the Partial Hairpin Amplification for Sequencing (PHASE) method.
  • a variety of applications require the evaluation of the presence and quantification of different biological particle or organism types within a population of biological particles, including, for example, microbiome analysis and characterization, environmental testing, food safety testing, epidemiological analysis, e.g., in tracing contamination or the like.
  • FIG. 40 shows a flowchart of a method 4000 for analyzing a nucleic acid molecule of an embedded, fixed tissue, according to some embodiments.
  • the method 4000 may comprise providing the embedded, fixed tissue.
  • the embedded, fixed tissue may comprise a plurality of cells.
  • a cell of the plurality of cells may comprise the nucleic acid molecule.
  • the embedded, fixed tissue may be embedded in a solid medium.
  • the nucleic acid molecule may be as described elsewhere herein.
  • the nucleic acid molecule may comprise a ribonucleic acid (RNA) molecule.
  • the nucleic acid molecule may comprise a messenger RNA molecule.
  • the fixed tissue may be fixed at least about 1 day (d), 2 d, 3 d, 4 d, 5 d, 6 d, 1 week (w), 2 w, 3 w, 1 month (m), 2 m, 3 m, 4 m, 5 m, 6 m, 7 m, 8 m, 9 m, 10 m, 11 m, 1 year (y), 2 y, 3 y, 4 y, 5 y, 10 y, 15 y, 20 y, or more prior to operation 4010.
  • the fixed tissue may be fixed at most about 20 y, 15 y, 10 y, 5 y, 4 y, 3 y, 2 y, 1 y, 11 m, 10 m, 9 m, 8 m, 7 m, 6 m, 5 m, 4 m, 3 m, 2 m, 1 m, 3 w, 2 w, 1 w, 6 d, 5 d, 4 d, 3 d, 2 d, 1 d, or less prior to operation 4010.
  • the fixed tissue may be fixed at a time as defined by a range of any two of the proceeding values.
  • the fixed tissue may comprise one or more of, for example, connective tissue, epithelial tissue, organ tissue, muscle tissue, ligaments, tendons, a skin tissue, breast tissue, bladder, kidney tissue, liver tissue, colon tissue, thyroid tissue, cervical tissue, prostate tissue, lung tissue, cardiac tissue, heart tissue, muscle tissue, pancreas tissue, anal tissue, bile duct tissue, a bone tissue, bone marrow, uterine tissue, ovarian tissue, endometrial tissue, vaginal tissue, vulvar tissue, stomach tissue, ocular tissue, nasal tissue, sinus tissue, penile tissue, salivary gland tissue, gut tissue, gallbladder tissue, gastrointestinal tissue, bladder tissue, brain tissue, spinal tissue, neurons, cells representative of a blood-brain barrier, blood, hair, nails, keratin, collagen, or any combination thereof.
  • the fixed tissue may comprise fixed human tissue.
  • the fixed tissue may comprise fixed cancer tissue (e.g., fixed breast cancer tissue).
  • the solid medium comprises paraffin.
  • the solid medium may comprise, for example, other long chain hydrophobic compounds (e g., waxes, etc.), glycerin, gels, epoxy, resin, or the like, or any combination thereof.
  • the fixed tissue may be fixed using, for example, formaldehyde (e.g., formalin), ethanolic fixatives, polyethylene glycol, glycerol, acetic acid, zinc-based fixatives, khandsari, honey, or the like, or any combination thereof.
  • the method 4000 may comprise removing at least a portion of the solid medium from the embedded, fixed tissue, thereby obtaining a fixed tissue comprising the plurality of cells.
  • operation 4020 may comprise adding a first solvent to the embedded, fixed tissue, thereby dissolving the solid medium, thereby obtaining a fixed tissue.
  • the first solvent comprises a non-polar solvent.
  • non-polar solvents include, but are not limited to, alkanes (e.g., butane, pentane, hexanes, etc.), aromatic compounds (e.g., benzene, toluene, xylenes, etc ), xylene alternative compounds (e.g., neo-clear® xylene alternative, etc.), or the like, or any combination thereof.
  • the non-polar solvent may have no or substantially no dipole moment over the solvent molecule.
  • the non-polar solvent may be configured to dissolve at least a portion of the solid medium.
  • the solid medium may comprise a non-polar portion dissolvable by the first solvent.
  • the operation 4020 may comprise removing the first solvent from the fixed tissue.
  • the removing may comprise removing without damaging the fixed tissue.
  • the first solvent can be removed without physical damage to the fixed tissue.
  • the fixed tissue can remain as a tissue scroll subsequent to the removal of the solid medium and the first solvent.
  • the removing may comprise use of a pipette.
  • the first solvent can be removed via pipetting away the first solvent without disrupting the fixed tissue.
  • the removing may comprise decanting the first solvent away from the fixed tissue.
  • the operation 4020 may further comprise adding a second solvent into the fixed tissue.
  • the second solvent may comprise a polar solvent.
  • polar solvents include, but are not limited to, alcohols (e.g., methanol, ethanol, propanol, iso-propanol, etc.), carbonyl containing compounds (e.g., acetone, ethyl acetate, etc.), haloforms (e.g., chloroform, etc.), dimethyl sulphoxide (DMSO), N-methyl formamide (NMF), or the like, or any combination thereof.
  • the polar solvent is ethanol.
  • the operation 4020 may comprise removing the second solvent from the fixed tissue. The removing may be as described elsewhere herein. For example, the removing may comprise not disrupting the fixed tissue.
  • the operation 4020 may comprise adding a rehydration agent to the fixed tissue.
  • the rehydration agent may comprise water.
  • the rehydration agent can be water.
  • the rehydration agent can comprise a buffer solution.
  • the rehydration agent may comprise one or more of buffers, salts, ions, ore the like, or any combination thereof.
  • the operation 4020 may comprise removing the rehydration agent from the tissue as described elsewhere herein.
  • the operation 4020 may comprise adding a buffer to the fixed tissue.
  • buffers include, but are not limited to, tris buffers, phosphate buffers, glycine buffers, boric acid buffers, carboxylic acid buffers (e.g., malic acid, formic acid, lactic acid, etc.), or the like, or any combination thereof.
  • the buffer may be removed from the fixed tissue. The removing may be as described elsewhere herein.
  • the method 4000 may comprise dissociating the fixed tissue into cells of the plurality of cells.
  • the cells may comprise the cell comprising the nucleic acid molecule.
  • Operation 4030 may comprise dissociating the tissue.
  • the dissociating the tissue may comprise us of one or more of a manual dissociator (e.g., a manual pulverizer, a mortar and pestle, a manual chopper, etc.), an automated dissocator (e.g., a gentleMACSTM Octo Dissociator, a kerafast® PCT shredder, a Covaris® cryoPREP automated dry pulvarizer, etc.), or the like, or any combination thereof.
  • a manual dissociator e.g., a manual pulverizer, a mortar and pestle, a manual chopper, etc.
  • an automated dissocator e.g., a gentleMACSTM Octo Dissociator, a kerafast® PCT shredder, a Covaris® cryoPREP automated dry pulvarizer, etc.
  • Operation 4030 may comprise resuspending the tissue.
  • the resuspending may comprise adding a resuspension solution to the tissue.
  • the resuspension solution may comprise, for example, water, surfactants, buffers, salts, or the like, or any combination thereof.
  • the resuspension solution may be configured to solubilize at least a portion of the tissue and/or components of the tissue (e.g., biological molecules, nucleic acids, etc.).
  • the resuspending may make the contents of the tissue available for the methods and systems described elsewhere herein.
  • the resuspending may provide the tissue for additional process operations.
  • Operation 4030 may comprise washing the tissue.
  • the washing may remove nonpredetermined molecules (e.g., waste molecules, non-target molecules, etc.).
  • the washing may comprise use of one or more solvents (e g., water, non-polar solvents, polar solvents, etc.) as described elsewhere herein.
  • the tissue may be washed at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more times.
  • the tissue may be washed at most about 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, or fewer times.
  • the tissue may be resuspended.
  • the tissue can be resuspended as described elsewhere herein.
  • the method 4000 may provide a yield of cells from two 25 micrometer sections of fixed tissue of at least about 1 x 10 3 (1E3), 5E3, 1E4, 5E4, 1E4, 5E5, 1E5, 5E6, 1E6, 5E7, 1E7, 5E8, 1E8, 5E9, 1E9, 5E10, or more cells.
  • the method 4000 may provide a yield of cells from two 25 micrometer sections of fixed tissue of at most about 5E10, 1E10, 5E10, 1E9, 5E9, 1E8, 5E8, 1E7, 5E7, 1E6, 5E6, 1E5, 5E5, 1E4, 5E4, 1E4, 5E3, 1E3, or fewer cells.
  • Non-limiting examples of experimentally realized cell yields per two 50 micrometer sections include human breast tissue (0.6E6 cells), human lymph node (2.33E6 cells), human lung cancer (5.05E6 cells), human brain (9.36E6 cells), human pancreas (5.45E6 cells), human heart (1.11E6 cells), human testis (1.99E6 cells), and human tonsil (5.6E6 cells).
  • Non-limiting examples of experimentally realized cell yields per two 25 micrometer sections includes normal human pancreas (0.6E6 cells), human brain glioblastoma multiforme (0.14E6 cells), human tonsil chronic tonsillitis with lymphoid hyperplasia (5.6E6 cells), human lung cancer (0.38E6 cells), human liver- hepatocellular carcinoma (0.52E6 cells), normal human thymus (2.56E6 cells), human kidney normal adjacent 2.49E6 cells), human ovarian cancer (0.33E6 cells), normal human spleen (1.89E6 cells), human heart myxoma (0.34E6 cells), human skin melanoma (0.51E6 cells), human prostate diseased (0.3E6 cells), human lymph node adenocarcinoma (0.46E6 cells), and human colon colorectal cancer (1.34E6 cells).
  • the nucleic acid molecule can be partitioned into a partition.
  • the partitioning can occur prior to operation 4040.
  • the partition may be a well of a plurality of wells.
  • the partitioning can comprise placing the nucleic acid molecule into a well of a 96 well plate.
  • the partition may be a droplet of a plurality of droplets, as described elsewhere herein.
  • the droplet can be an oil emulsion droplet of a plurality of oil emulsion droplets.
  • the droplet can be a phase that is immiscible with a surrounding phase.
  • the droplet can be a droplet surrounded by gas (e g., air).
  • the partition may comprise a support.
  • the support may be as described elsewhere herein.
  • the support may comprise a solid support.
  • the support can comprise a bead.
  • the barcode sequence may be attached to the support.
  • the method 4000 may comprise, in the cell, using a barcode sequence to generate a barcoded nucleic acid molecule comprising a barcode sequence and a sequence corresponding to the nucleic acid molecule.
  • the operation 4040 may comprise hybridizing the barcode sequence to the nucleic acid molecule.
  • the hybridizing may be as described elsewhere herein.
  • the barcode can be extended to form the barcoding nucleic acid molecule as described elsewhere herein.
  • the extending can comprise the use of an enzyme.
  • the extending can comprise use of ligation.
  • the extending can comprise ligating the barcode sequence to a probe as described elsewhere herein.
  • the probe can comprise a portion at least partially complementary to the nucleic acid molecule.
  • FIG. 41 shows an example of a nucleic acid profding workflow, according to some embodiments.
  • the nucleic acid profding workflow can be combined with the other portions of the present disclosure such as, for example, fixed tissue preparation methods.
  • the nucleic acid profiling workflow of FIG. 41 may be a singleplex workflow.
  • the nucleic acid profiling workflow can be performed on one or more samples (e.g., cells, nucleic acid
  • the nucleic acid profiling workflow can comprise a staining operation.
  • the staining operation may be as described elsewhere herein.
  • the workflow can comprise fixing and/or permeabilizing the cells.
  • the fixing may comprise fixation as described elsewhere herein.
  • the fixing may comprise embedding.
  • the fixed cells can be fixed and subsequently embedded in a matrix.
  • the permeabilizing the cells may comprise use of one or more permeabilization reagents (e.g., methanol, ethanol, acetone, detergents, etc.).
  • the permeabilizing may comprise an at least partial degradation of the cell wall of a cell of the sample.
  • the permeabilizing may be configured to make accessible the contents of the cell (e.g., nucleic acids).
  • the sample may be stored.
  • the storage may be for at least about 1 day (d), 2 d, 3 d, 4 d, 5 d, 6 d, 1 week (w), 2 w, 3 w, 1 month (m), 2 m, 3 m, 4 m, 5 m, 6 m, 7 m, 8 m, 9 m, 10 m, 11 m, 1 year (y), 2 y, 3 y, 4 y, 5 y, 10 y, 15 y, 20 y, or more.
  • the storage may be for at most about 20 y, 15 y, 10 y, 5 y, 4 y, 3 y, 2 y, 1 y, 11 m, 10 m, 9 m, 8 m, 7 m, 6 m, 5 m, 4 m, 3 m, 2 m, 1 m, 3 w, 2 w, 1 w, 6 d, 5 d, 4 d, 3 d, 2 d, 1 d, or less.
  • the workflow can comprise hybridizing one or more probes to the sample as described elsewhere herein.
  • the hybridizing may comprise use of a probe molecule such as the probe molecule of FIG. 43.
  • the hybridizing may not comprise multiplexing.
  • each sample can be run through the workflow individually.
  • Each sample run through the workflow can comprise a probe barcode
  • the singleplex format can be compatible with a cell labeling scheme (e g., cell surface protein labeling).
  • the barcoded nucleic acids can be portioned as described elsewhere herein.
  • the barcoded nucleic acids can be partitioned into a plurality of droplets.
  • the barcoded nucleic acids can be partitioned into a plurality of wells.
  • ligation and extension can be performed as described elsewhere herein.
  • Library constmction may comprise one or more of construction of a cell surface protein library and a gene expression library as described elsewhere herein. The partitioning may be as described in FIG. 43.
  • FIG. 43 shows an example of droplet partition generation, according to some embodiments.
  • a plurality of barcoded beads 4301 can be provided.
  • the washed fixed sample can be provided to the barcoded beads as shown and partitioned into a plurality of droplets suspended in oil. In this way, the sample can be partitioned such that, on average, less than one sample is present in each partition as shown.
  • the nucleic acid molecule from the sample can be sequenced as described elsewhere herein.
  • the sequencing can be sequencing by synthesis.
  • the data generated by the sequencing can be analyzed as described elsewhere herein.
  • FIG. 42 shows an example of a nucleic acid profiling workflow, according to some embodiments.
  • the nucleic acid profiling workflow of FIG. 42 may be a multiplex workflow (e g., multiple samples can be processed at a same time). In some cases, the multiplexed workflow may not be compatible with cellular labeling.
  • the operations of the nucleic acid profiling workflow can be as described elsewhere herein. For example, cells may be fixed in formaldehyde and nucleic acid molecules from the cells may be hybridized with probe sets. Each probe set may comprise a unique probe barcode to permit sample multiplexing and read level demux. Samples may be hybridized overnight, pooled, washed, and partitioned. The probes may be ligated along with additional barcodes.
  • the nucleic acid profiling workflow can comprise use of a sample as described elsewhere herein.
  • the sample can comprise a nucleic acid.
  • the fixation and/or permeabilization operations may be as described elsewhere herein.
  • the sample may be stored subsequent to the fixation and/or permeabilization.
  • probes can be hybridized to nucleic acid molecules of the sample.
  • different probes can be hybridized to different nucleic acid molecules in the sample.
  • each sample used in the workflow can be correlated to a different barcode probe.
  • the multiplexing may comprise simultaneous hybridizing of at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more samples.
  • the multiplexing may comprise simultaneous hybridizing of at most about 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or fewer samples.
  • the nucleic acid molecules can be stored subsequent to the hybridization of the probes. [00419] Subsequent to the hybridization, the nucleic acid molecules can be pooled and partitioned as described elsewhere herein (e.g., FIG. 43).
  • the barcode molecules can be ligated and/or extended as described elsewhere herein.
  • a library (e.g., a gene expression library) can be constructed, the barcode molecule can be sequenced, and the sequencing data can be analyzed.
  • the pooling operations described elsewhere herein can comprise one or more washing operations.
  • the washing may comprise removal of a supernatant from a centrifuged solution.
  • the removal of the supernatant can be a complete removal (e g., all of the supernatant can be removed).
  • the removal of the supernatant can be a partial removal (e.g., a portion of the supernatant can be left behind).
  • Leaving supernatant can provide benefits for processing samples with low cell numbers (e.g., less than about 500,000 cells in the sample). Incomplete removal of the supernatant can enhance cell recovery without significantly impacting the performance of the assays of the present disclosure.
  • FIG. 43 show an example of a plurality of probe molecules 4301 and 4302, according to some embodiments.
  • the probe can be as described elsewhere herein.
  • the probe can be configured to analyze a gene.
  • the probe can be configured with a barcode portion 4303.
  • the barcode portion can be a barcode as described elsewhere herein.
  • the barcode can be attached to the probe prior to a user using the probe for analysis of a nucleic acid molecule.
  • the barcode can be pre-affixed to the probe.
  • the probe can be supplied to a user with the barcode already a part of the probe.
  • the probe molecule can bind to an analyte nucleic acid 4304. Subsequent to the binding, the probes can be ligated together.
  • the ligation may comprise sealing a nick (e.g., ligating adjacent probes without gap filling).
  • the ligation may comprise a gap filling ligation reaction (e.g., filling a gap of one or more nucleotides between the probes during the ligation).
  • the single, unified probe 4305 can be dissociated from the analyte nucleic acid and captured by a capture region 4306 of a nucleic acid associated with a bead 4307.
  • the probe can then be extended to incorporate one or more barcode molecules, primers, or the like as described elsewhere herein.
  • the probe may then be amplified as described elsewhere herein.
  • the methods and systems of the present disclosure can provide improvements on the efficacy of analysis of fixed samples. Fixation can improve the mechanical resilience of a cellular sample, as well as lock in transient cellular states In this way, the analysis of the cells of a sample can provide information about the state of the cell when fixed, which can provide additional insight about short lived cellular states.
  • the improvements of the present disclosure can also enable larger longitudinal studies. For example, high quality data generated from fixed samples permits study of the samples over longer periods of time. Additionally, modern techniques to be used with historical samples, unlocking new information previously unavailable.
  • the methods and systems of the present disclosure can also reduce transportation and/or storage constraints on samples. For example, fixed and embedded samples can be more resilient to poor storage conditions than unfixed samples.
  • FIG. 14 shows a computer system 1401 that is programmed or otherwise configured to process or analyze sequencing reads.
  • the computer system 1401 can regulate various aspects of the present disclosure, such as, for example, aligning sequencing reads, indexing sequencing reads to a cell, partition, etc.
  • the computer system 1401 can be an electronic device of a user or a computer system that is remotely located with respect to the electronic device.
  • the electronic device can be a mobile electronic device.
  • the computer system 1401 includes a central processing unit (CPU, also “processor” and “computer processor” herein) 1405, which can be a single core or multi core processor, or a plurality of processors for parallel processing.
  • the computer system 1401 also includes memory or memory location 1410 (e g., random-access memory, read-only memory, flash memory), electronic storage unit 1415 (e.g., hard disk), communication interface 1420 (e.g., network adapter) for communicating with one or more other systems, and peripheral devices 1425, such as cache, other memory, data storage and/or electronic display adapters.
  • the memory 1410, storage unit 1415, interface 1420 and peripheral devices 1425 are in communication with the CPU 1405 through a communication bus (solid lines), such as a motherboard.
  • the storage unit 1415 can be a data storage unit (or data repository) for storing data.
  • the computer system 1401 can be operatively coupled to a computer network (“network”) 1430 with the aid of the communication interface 1420.
  • the network 1430 can be the Internet, an internet and/or extranet, or an intranet and/or extranet that is in communication with the Internet.
  • the network 1430 in some cases is a telecommunication and/or data network.
  • the network 1430 can include one or more computer servers, which can enable distributed computing, such as cloud computing.
  • the network 1430 in some cases with the aid of the computer system 1401, can implement a peer-to-peer network, which may enable devices coupled to the computer system 1401 to behave as a client or a server.
  • the CPU 1405 can execute a sequence of machine-readable instructions, which can be embodied in a program or software.
  • the instructions may be stored in a memory location, such as the memory 1410.
  • the instructions can be directed to the CPU 1405, which can subsequently program or otherwise configure the CPU 1405 to implement methods of the present disclosure. Examples of operations performed by the CPU 1405 can include fetch, decode, execute, and writeback.
  • the CPU 1405 can be part of a circuit, such as an integrated circuit.
  • a circuit such as an integrated circuit.
  • One or more other components of the system 1401 can be included in the circuit.
  • the circuit is an application specific integrated circuit (ASIC).
  • ASIC application specific integrated circuit
  • the storage unit 1415 can store files, such as drivers, libraries and saved programs.
  • the storage unit 1415 can store user data, e.g., user preferences and user programs.
  • the computer system 1401 in some cases can include one or more additional data storage units that are external to the computer system 1401, such as located on a remote server that is in communication with the computer system 1401 through an intranet or the Internet.
  • the computer system 1401 can communicate with one or more remote computer systems through the network 1430.
  • the computer system 1401 can communicate with a remote computer system of a user (e.g., operator).
  • remote computer systems include personal computers (e.g., portable PC), slate or tablet PC’s (e.g., Apple® iPad, Samsung® Galaxy Tab), telephones, Smart phones (e.g., Apple® iPhone, Android-enabled device, Blackberry®), or personal digital assistants.
  • the user can access the computer system 1401 via the network 1430.
  • Methods as described herein can be implemented by way of machine (e.g., computer processor) executable code stored on an electronic storage location of the computer system 1401, such as, for example, on the memory 1410 or electronic storage unit 1415.
  • the machine executable or machine readable code can be provided in the form of software.
  • the code can be executed by the processor 1405.
  • the code can be retrieved from the storage unit 1415 and stored on the memory 1410 for ready access by the processor 1405.
  • the electronic storage unit 1415 can be precluded, and machine-executable instructions are stored on memory 1410.
  • the code can be pre-compiled and configured for use with a machine having a processor adapted to execute the code, or can be compiled during runtime.
  • the code can be supplied in a programming language that can be selected to enable the code to execute in a precompiled or as-compiled fashion.
  • aspects of the systems and methods provided herein can be embodied in programming.
  • Various aspects of the technology may be thought of as “products” or “articles of manufacture” typically in the form of machine (or processor) executable code and/or associated data that is carried on or embodied in a type of machine readable medium.
  • Machine-executable code can be stored on an electronic storage unit, such as memory (e g., read-only memory, random-access memory, flash memory) or a hard disk.
  • “Storage” type media can include any or all of the tangible memory of the computers, processors or the like, or associated modules thereof, such as various semiconductor memories, tape drives, disk drives and the like, which may provide non-transitory storage at any time for the software programming. All or portions of the software may at times be communicated through the Internet or various other telecommunication networks. Such communications, for example, may enable loading of the software from one computer or processor into another, for example, from a management server or host computer into the computer platform of an application server.
  • another type of media that may bear the software elements includes optical, electrical and electromagnetic waves, such as used across physical interfaces between local devices, through wired and optical landline networks and over various air-links.
  • a machine readable medium such as computer-executable code
  • a tangible storage medium such as computer-executable code
  • Non-volatile storage media include, for example, optical or magnetic disks, such as any of the storage devices in any computer(s) or the like, such as may be used to implement the databases, etc. shown in the drawings.
  • Volatile storage media include dynamic memory, such as main memory of such a computer platform.
  • Tangible transmission media include coaxial cables; copper wire and fiber optics, including the wires that comprise a bus within a computer system.
  • Carrier-wave transmission media may take the form of electric or electromagnetic signals, or acoustic or light waves such as those generated during radio frequency (RF) and infrared (IR) data communications.
  • RF radio frequency
  • IR infrared
  • Common forms of computer-readable media therefore include for example: a floppy disk, a flexible disk, hard disk, magnetic tape, any other magnetic medium, a CD-ROM, DVD or DVD-ROM, any other optical medium, punch cards paper tape, any other physical storage medium with patterns of holes, a RAM, a ROM, a PROM and EPROM, a FLASH-EPROM, any other memory chip or cartridge, a carrier wave transporting data or instructions, cables or links transporting such a carrier wave, or any other medium from which a computer may read programming code and/or data.
  • Many of these forms of computer readable media may be involved in carrying one or more sequences of one or more instructions to a processor for execution.
  • the computer system 1401 can include or be in communication with an electronic display 1435 that comprises a user interface (UI) 1440 for providing, for example, results of sequencing analysis, etc.
  • UI user interface
  • Examples of UIs include, without limitation, a graphical user interface (GUI) and web-based user interface.
  • GUI graphical user interface
  • Methods and systems of the present disclosure can be implemented by way of one or more algorithms.
  • An algorithm can be implemented by way of software upon execution by the central processing unit 1405.
  • the algorithm can, for example, perform sequencing.
  • Devices, systems, compositions and methods of the present disclosure may be used for various applications, such as, for example, processing a single analyte (e.g., RNA, DNA, or protein) or multiple analytes (e.g., DNA and RNA, DNA and protein, RNA and protein, or RNA, DNA and protein) from a single cell.
  • a biological particle e.g., a cell or cell bead
  • a partition e.g., droplet
  • multiple analytes from the biological particle are processed for subsequent processing.
  • the multiple analytes may be from the single cell. This may enable, for example, simultaneous proteomic, transcriptomic and genomic analysis of the cell.
  • the cell or cell bead may comprise a target RNA molecule for barcoding and/or a feature, which may have a feature binding group comprising a reporter oligonucleotide (comprising a reporter sequence) coupled thereto.
  • the target RNA molecule may be hybridized to a first probe and a second probe; for example, the target RNA molecule may have a first target region and a second target region complementary to a first probe sequence of the first probe and a second probe sequence of the second probe.
  • a probe-linked molecule may be generated, e.g., via ligation of the probes when hybridized to the RNA molecule, or using one or more nucleic acid reactions, e.g., via an extension reaction, and/or enzymatic or chemical ligation.
  • the probe-linked molecule may be barcoded in one or more sets of partitions.
  • cells may be partitioned in a first set of partitions (e.g., microwells or other vessels) and contacted with a hybridization buffer comprising the first probe, the second probe, a probe binding molecule (e.g., a splint oligonucleotide) and a barcode molecule.
  • the hybridization buffer may comprise reagents (e.g., formamide, ethylene carbonate, salts, etc.) to facilitate hybridization of the first probe and the second probe to a target nucleic acid molecule.
  • Cells (or nuclei or cell beads) from multiple partitions may then be pooled and washed, e.g., to remove unhybridized probes.
  • the cells (or nuclei or cell beads) may then be counted and re-partitioned in a second set of partitions, e.g., droplets.
  • a ligation and extension reaction may be performed to generate barcoded nucleic acid molecules.
  • the droplets may additionally comprise a capture molecule comprising an additional barcode sequence. Accordingly, the barcoded nucleic acid molecules within the droplets may comprise two barcode sequences.
  • the cells may be partitioned in a first set of partitions (e.g., microwells) and contacted with a hybridization buffer comprising the first probe, the second probe, a probe binding molecule (e.g., a splint oligonucleotide) and a barcode molecule.
  • a probe binding molecule e.g., a splint oligonucleotide
  • Cells (or nuclei or cell beads) within the partitions may then be washed, e.g., to remove unhybridized probes, then pooled together.
  • the cells (or nuclei or cell beads) may then be counted and re-partitioned in a second set of partitions, e.g., droplets.
  • a ligation and extension reaction may be performed to generate barcoded nucleic acid molecules.
  • the droplets may additionally comprise a capture molecule comprising an additional barcode sequence.
  • the barcoded nucleic acid molecules within the droplets may comprise two barcode sequences.
  • cells may be partitioned in a first set of partitions (e.g., microwells) and contacted with a hybridization buffer comprising the first probe, the second probe, a probe binding molecule (e.g., a splint oligonucleotide) and a barcode molecule.
  • a probe binding molecule e.g., a splint oligonucleotide
  • Cells (or nuclei or cell beads) from multiple partitions may then be pooled and washed, e.g., to remove unhybridized probes.
  • the cells (or nuclei or cell beads) may then be counted and subjected to conditions sufficient to ligate the barcode molecules to the probe- hybridized nucleic acid molecules.
  • the ligated molecules may then be partitioned, e.g., into droplets.
  • an extension reaction may be performed to generate barcoded nucleic acid molecules.
  • the droplets may additionally comprise a capture molecule comprising an additional barcode sequence Accordingly, the barcoded nucleic acid molecules within the droplets may comprise two barcode sequences.
  • the cells may be partitioned in a first set of partitions (e.g., microwells) and contacted with a hybridization buffer comprising the first probe, the second probe, a probe binding molecule (e.g., a splint oligonucleotide) and a barcode molecule.
  • a probe binding molecule e.g., a splint oligonucleotide
  • Cells (or nuclei or cell beads) within the partitions may then be washed, e.g., to remove unhybridized probes, then pooled together.
  • the cells (or nuclei or cell beads) may then be counted and subjected to conditions sufficient to ligate the barcode molecules to the probe- hybridized nucleic acid molecules.
  • the ligated molecules may then be partitioned, e.g., into droplets.
  • an extension reaction may be performed to generate barcoded nucleic acid molecules.
  • the droplets may additionally comprise a capture molecule comprising an additional barcode sequence. Accordingly, the barcoded nucleic acid molecules within the droplets may comprise two barcode sequences.
  • the barcoded nucleic acid molecules may be subjected to additional barcoding operations in additional partitions, e.g., in droplets.
  • additional barcoding operations e.g., in droplets.
  • the contents of the droplets may be pooled and processed downstream for analysis, e.g., via sequencing.
  • cells, nuclei, or cell beads which may optionally be fixed and permeabilized, may be first hybridized to a set of probes and then barcoded (e.g., in partitions).
  • the cells may be contacted, e.g., in a bulk solution, with a hybridization buffer comprising the first probe and the second probe.
  • the cells may then be washed, e.g., to remove unhybridized probes, then partitioned into a first set of partitions (e.g., microwells).
  • the first set of partitions may each comprise a probe binding molecule and a barcode molecule.
  • the cells (or nuclei or cell beads) in the first set of partitions may be subjected to conditions sufficient to hybridize the probe binding molecule and the barcode molecule to the target nucleic acid molecule, the probe molecules, or derivatives thereof (e.g., extended probe-associated molecules, etc.).
  • the contents of the partitions may then be pooled together and optionally, washed.
  • the cells or nuclei or cell beads
  • the cells may then be counted and partitioned into a second set of partitions and subjected to conditions sufficient to extend and/or ligate the barcode molecules to the probe-hybridized nucleic acid molecules, thereby generating barcoded nucleic acid molecules.
  • the droplets may additionally comprise a capture molecule comprising an additional barcode sequence. Accordingly, the barcoded nucleic acid molecules within the droplets may comprise two barcode sequences.
  • the partitions may be washed and then pooled together.
  • the cells may then be counted and partitioned into a second set of partitions and subjected to conditions sufficient to extend and/or ligate the barcode molecules to the probe- hybridized nucleic acid molecules, thereby generating barcoded nucleic acid molecules.
  • the droplets may additionally comprise a capture molecule comprising an additional barcode sequence. Accordingly, the barcoded nucleic acid molecules within the droplets may comprise two barcode sequences.
  • the contents of the partitions may then be pooled together and optionally, washed.
  • the cells or nuclei or cell beads
  • the ligated molecules may be partitioned in a second set of partitions, e.g., droplets, and subjected to conditions sufficient to extend the ligated molecules to generate barcoded nucleic acid molecules.
  • the droplets may additionally comprise a capture molecule comprising an additional barcode sequence. Accordingly, the barcoded nucleic acid molecules within the droplets may comprise two barcode sequences.
  • the partitions may be first washed, then the contents of the partitions may be pooled together.
  • the cells or nuclei or cell beads
  • the ligated molecules may be partitioned in a second set of partitions, e.g., droplets, and subjected to conditions sufficient to extend the ligated molecules to generate barcoded nucleic acid molecules.
  • the droplets may additionally comprise a capture molecule comprising an additional barcode sequence. Accordingly, the barcoded nucleic acid molecules within the droplets may comprise two barcode sequences.
  • the cells, nuclei, or cell beads may be first hybridized to a set of probes, washed, counted, subjected to conditions sufficient to ligate the probes or generate a probe-linked nucleic acid molecule, washed again, and then partitioned.
  • the cells (or nuclei or cell beads) may be partitioned in a first set of partitions (e.g., microwells) with a probe binding molecule and a barcode molecule.
  • the probe binding molecule and the barcode molecule may hybridize to the probe-associated molecule (or probe-linked molecules), pooled, washed (or alternatively, washed in partitions, then pooled), counted and then loaded into a second set of partitions (e.g., droplets).
  • the cells may then be subjected to conditions sufficient to extend and/or ligate the barcode molecules to the probe-associated or probe-linked nucleic acid molecules, thereby generating barcoded nucleic acid molecules.
  • the cells (or nuclei or cell beads) may be ligated in bulk and extended within the second set of partitions.
  • the droplets may additionally comprise a capture molecule comprising an additional barcode sequence.
  • the barcoded nucleic acid molecules within the droplets may comprise two barcode sequences.
  • the cell or cell beads may be contacted with a feature binding group comprising or coupled to a reporter oligonucleotide (comprising a reporter sequence), as described herein.
  • the feature binding group may couple to one or more features (e.g., proteins) of the cell.
  • the cell may also comprise target nucleic acid molecules (e.g., RNA molecules) for assaying.
  • Example Protocol 1 the cells, nuclei, or cell beads having the feature binding groups coupled thereto may be partitioned in a first set of partitions.
  • Each partition of the first set of partitions may comprise, for example, -50,000 cells in a 50 microliter volume.
  • the partitions may each comprise a set of probes (e.g., a first probe, a second probe, and a third probe), which may be provided at a 2 micromolar concentration.
  • Each partition may additionally comprise 5 micromolar of splint oligonucleotides (probe-binding molecules), and 7.5 micromolar barcode molecules.
  • the barcode molecules may differ across the partitions.
  • the probe molecules may be hybridized to (i) the target nucleic acid (e.g., RNA) molecule (e g., via the first and second probes) and (ii) the feature binding group (e.g., via the third probe).
  • the contents of the first set of partitions may then be pooled, washed, and analyzed, e.g., using optical approaches such as absorbance, fluorescence, etc., gel electrophoresis, or via sequencing.
  • Example Protocol 2 In another example protocol, the cells, nuclei, or cell beads having the feature binding groups coupled thereto may hybridized, in bulk, to the first set of probes (e.g., a first probe, a second probe, and a third probe), which may be provided at a 2 micromolar concentration.
  • the cells, nuclei, or cell beads may be subjected to conditions sufficient to hybridize the probe molecules to the target nucleic acid and/or the feature binding group.
  • the cells, nuclei, or cell beads may then be washed and then partitioned in a first set of partitions. Each partition of the first set of partitions may comprise, for example, -50,000 cells in a 50 microliter volume.
  • the partitions may each comprise 1 micromolar of splint oligonucleotides (probe-binding molecules), and 2 micromolar barcode molecules.
  • the barcode molecules may differ across the first set of partitions. Within the first set of partitions, the barcode molecules may hybridize to the probe-associated molecules. The contents of the first set of partitions may then be pooled, washed, and analyzed, e g., using optical approaches such as absorbance, fluorescence, etc., gel electrophoresis, or via sequencing.
  • FIG. 18 shows example data of a barcoding scheme described herein (Example Protocol 1).
  • the left plot shows barcoding performed in peripheral blood mononuclear cells (PBMCs) and the right plot shows barcoding performed in various cell lines.
  • Two peaks can be identified: a 230 bp sequence and 270 base pair sequence.
  • the 270 base pair sequence corresponds to the first target region and the second target region of the target RNA molecule of the first and second probe.
  • FIG. 19 shows example data of DNA gel electrophoresis of the barcoded molecules described herein.
  • the left plots indicate an annealing temperature of 67 degrees Celsius, whereas the right plots indicate an annealing temperature of 63 degrees Celsius.
  • the top plots indicate one method of verifying the barcode products using LED.
  • the bottom plots indicate another method of verifying the barcode products using V2.
  • the first lane (“Lane 0”) in each gel electrophoresis plot is a nucleic acid standard ladder.
  • Lane l is a PBMC without barcode (negative control)
  • Lane 2 is a cell line sample without barcode (negative control)
  • Lane 3 is a PBMC with a synthetic barcode (positive control)
  • Lane 4 is a cell line with a synthetic barcode (positive control)
  • Lane 5 is a PBMC with a splint molecule, performed according to Example Protocol 1
  • Lane 6 is a cell line with a splint molecule, performed according to Example Protocol 7
  • Lane 7 is a PBMC with a splint molecule, performed according to Example Protocol 2
  • Lane 8 is a cell line with a splint, performed according to Example Protocol 2.
  • the 63 degree annealing temperature results in higher yield (darker bands).
  • FIG. 20 shows additional example data of a barcoding scheme described herein.
  • the left plots show fluorescence intensity as a function of sequence length of the PBMC cells barcoded using Example Protocol 7.
  • the right plots show the fluorescence intensity as a function of sequence length of the cell lines barcoded using Example Protocol 1.
  • Example 3 Fixed RNA profiling on a PBMC sample
  • a PBMC sample may be paraformaldehyde-fixed and then stored for 7 days at 4°C.
  • Fixed cells or nuclei or cell beads
  • Sequencing libraries may be prepared, enriched using a 2000-gene immuno-oncology panel and analyzed.
  • FIG. 21A-C shows example data comparing fixed cells and un-fixed control samples.
  • FIG. 21A shows a bar plot and illustrate that fixed cells, when compared to a Day 0 un-fixed control sample, demonstrate stable cell type annotation over seven days of storage.
  • FIG. 21B shows a line plot of the panel reads per cell as a function of the UMIs detected. The data illustrate a comparable median number of genes and UMI counts per cell.
  • 21C shows a log plot of the per-gene UMI counts between the Day 0 and Day 7 sample. An excellent correlation between the per-gene UMI counts between Day 0 control and the Day 7 can be visualized. The results demonstrate that the fixed sample is effectively stabilized for 7 days, which allows for further various manipulations following fixation, e.g., sample collection, storage, shipment, batching with other samples, etc.
  • fixed PBMCs may be processed according to the protocols described herein and the resulting libraries compared to fresh PBMCs processed with the 3’ Single Cell Gene Expression solution (lOx Genomics).
  • UMI detection over the 2000-gene panel illustrate comparable sensitivity between the fresh and fixed workflows.
  • cell type annotation is similar between the two samples as well.
  • Major PBMC cell types can be detected in each of the two samples.
  • Example 4- Multiplexed assay: barcoding of RNA templated ligation product and probe- associated reporter oligonucleotide
  • the cells, nuclei, or cell beads may be contacted with a feature binding group comprising or coupled to a reporter oligonucleotide (comprising a reporter sequence), as described herein.
  • the feature binding group may couple to one or more features (e g., proteins) of the cell.
  • the cell may also comprise target nucleic acid molecules (e.g., RNA molecules) for assaying.
  • cells are contacted with two sets of antibodies, as depicted schematically in FIG. 22.
  • the first set of antibodies (“Antibody A”) 2252 comprises a reporter oligonucleotide comprising two target sequences.
  • the second set of antibodies (“Antibody B”) 2253 comprises a reporter oligonucleotide comprising a capture sequence.
  • the cells e.g., 2200 are then contacted with a pair of probes.
  • the pair of probes (“probe 1” 2206 and “probe 2” 2216) are configured to hybridize to a first target region 2202 and a second target region 2204 of a nucleic acid molecule 2201 in the cell (e.g., mRNA), thereby generating a probe-associated molecule 2230. At least one of the probes of the pair of probes may comprise a capture sequence (e g., 2210 and/or 2218). Additionally, in some instances, the pair of probes 2206, 2216 are configured to hybridize to the two target sequences of Antibody A.
  • an additional pair of probes (“probe 3” and “probe 4”, not shown) that are different from probe 1 and probe 2 may be provided; the additional pair of probes may comprise complementary sequences to the target sequences of Antibody A and may hybridize to the reporter oligonucleotide of Antibody A.
  • Subsequent barcoding may be performed, either in bulk or in partitions (e.g., wells or droplets).
  • a barcode molecule 2220 comprising a first barcode sequence may hybridize, either directly or via a splint molecule, to the (i) probe associated molecule 2230 or derivative thereof (e.g., a complement or amplification product thereof), (ii) Antibody A- probe pair complex comprising the pair of probes (e.g., probe 1 2206 and probe 2 2216, or probe 3 and probe 4 (not shown)) hybridized to the reporter oligonucleotide of Antibody A 2252, and/or (iii) Antibody B 2253 (e g., via the capture sequence of the reporter oligonucleotide).
  • the barcode molecule 2220 may optionally be coupled to a bead.
  • the barcode molecule is shown as hybridizing directly to the probes or capture sequence, but barcode molecule hybridization may be mediated via a splint molecule (e.g., as shown in FIG. 16A). Additional barcoding operations may also be performed (not shown). The barcoded molecules or derivatives thereof are then sequenced.
  • FIG. 23 shows example data resulting from such a barcoding operation as described in FIG. 22.
  • FIG 23 shows gene expression plots of four biomarkers (CD4, CD8, CD3 and CD 14) which are obtained from sequencing of the barcoded RNA products (e.g., the barcoded probe-associated molecule 2230).
  • the intensity of the spots indicate the overlap between the gene expression detected from using the dual probes and the protein expression using either the first set of antibodies (“Antibody A”) or the second set of antibodies (“Antibody B”).
  • the plots indicate that usage of either set of antibodies (Antibody A or Antibody B) have similar coverage in detecting the analyte of interest.
  • either or both sets of antibodies may be used to detect the protein analytes (e.g., CD4, CD8, CD3, CD 14).
  • the first set of Antibodies (“Antibody A”) as the dual probes used for barcoding the reporter oligonucleotide allow for additional multiplexing or combinatorial barcoding, which allows for improved indexing and determination of cell, sample, or partition origin.
  • the use of one or more probes for barcoding the reporter oligonucleotides may be useful for appending additional functional sequences (e.g., primers, capture sequences, UMI, barcode sequences, etc.) to the reporter oligonucleotide, or derivative thereof.
  • the data shown in FIG. 23 may be generated without barcoding of any RNA molecules.
  • the cell may be contacted with (i) the first set of antibodies (“Antibody A”) 2252 comprising a reporter oligonucleotide comprising two target sequences and (ii) the second set of antibodies (“Antibody B”) 2253 comprising a capture sequence.
  • the cells e.g., 2200
  • the cells may then be contacted with a pair of probes.
  • the pair of probes (“probe 1” 2206 and “probe 2” 2216) are configured to hybridize to the target regions of the reporter oligonucleotide of Antibody A. At least one of the probes of the pair of probes may comprise a capture sequence (e.g., 2210 and/or 2218). Barcoding as described above may be performed, resulting in two barcoded products: (i) Antibody A-probe pair comprising the pair of probes (e.g., probe 1 2206 and probe 22216) hybridized to the reporter oligonucleotide of Antibody A 2252, and (ii) Antibody B 2253 (e.g., via the capture sequence).
  • the barcoded products or derivatives thereof may then be sequenced and the sequence reads may be overlapped to generate the plots of FIG. 23.
  • the barcoding efficiency using either approach is similar, and that both or either approach is viable in detecting analytes (e.g., proteins).
  • analytes e.g., proteins.
  • one or more probes for barcoding the reporter oligonucleotides may be useful for appending additional functional sequences (e.g., primers, capture sequences, UMI, barcode sequences, etc.) to the reporter oligonucleotide, or derivative thereof.
  • additional functional sequences e.g., primers, capture sequences, UMI, barcode sequences, etc.
  • Example 5 Multiplexed assay: barcoding of RNA templated ligation product and reporter oligonucleotide of a feature-binding group
  • the cells, nuclei, or cell beads may be contacted with a feature binding group comprising or coupled to a reporter oligonucleotide (comprising a reporter sequence), as described herein.
  • the feature binding group may couple to one or more features (e g., proteins) of the cell.
  • the cell may also comprise target nucleic acid molecules (e.g., RNA molecules) for assaying.
  • cells may be contacted with a set of antibodies (“Antibody B”) 2253, in which the antibodies of the set comprise a capture sequence.
  • the cells may be processed, e.g., subjected to fixation and/or permeabilization, which may occur prior to, subsequent to, or both prior to and subsequent to contacting the cells with the antibodies.
  • the cells are then contacted with a pair of probes
  • the pair of probes (“probe 1” 2206 and “probe 2” 2216) are configured to hybridize to a first target region 2202 and a second target region 2204 of a nucleic acid molecule 2201 in the cell (e.g., mRNA), thereby generating a probe-associated molecule 2230.
  • the probes may optionally be linked to one another (e.g., using an extension reaction, ligation, and/or chemical linkage). At least one of the probes of the pair of probes may comprise a capture sequence (e.g., 2210 and/or 2218).
  • Subsequent barcoding may be performed, either in bulk or in partitions (e.g., wells or droplets).
  • a barcode molecule 2220 comprising a first barcode sequence may hybridize, either directly or via a splint molecule, to the (i) probe associated molecule 2230 or derivative thereof (e g., a complement or amplification product thereof), and/or (ii) Antibody B 2253 (e.g., via the capture sequence).
  • the barcode molecule 2220 may optionally be coupled to a bead.
  • the barcoded molecules or derivatives thereof are then sequenced.
  • FIG. 24 shows example data of gene expression and protein analysis data resulting from the barcoding scheme described above.
  • Each plot shows a plot of a biomarker (CD14, CD8a, CD 19 and CD3) which are obtained from sequencing of the (i) barcoded RNA products (e g., the barcoded probe-associated molecule 2230) and (ii) barcoded Antibody B (e.g., barcoded reporter oligonucleotide).
  • the intensity of the spots indicate the relative expression levels (e.g., gene expression or protein expression) detected from the barcoded products.
  • a biomarker profile may be determined by assaying the biomarker protein (e.g., barcoding a reporter oligonucleotide of a feature binding group which binds to the biomarker protein), or the biomarker profile may be determined by assaying the biomarker gene expression (e.g., barcoding RNA using dual probes for gene expression profiling). In some instances, both the gene expression profile and the protein profile may be useful in characterizing a cell, e.g., to determine a correlation between gene expression and protein expression.
  • Cells may be contacted with feature binding groups comprising reporter oligonucleotides that identify the feature or feature-binding group and one or more probes (e.g., for hybridizing to target regions of a target nucleic acid molecule, e.g., mRNA).
  • feature binding groups comprising reporter oligonucleotides that identify the feature or feature-binding group and one or more probes (e.g., for hybridizing to target regions of a target nucleic acid molecule, e.g., mRNA).
  • the reporter oligonucleotides and/or the one or more probes (or the probe-associated molecules) may be barcoded in a plurality of partitions. Partitions may be overloaded such that fewer partitions of a plurality of partitions are unoccupied. In one non-limiting example, a population of -100,000 cells may be loaded into -80,000 partitions.
  • partitions are overloaded, there may still be many partitions that comprise a single cell .
  • the single-cell partitions and may be identified or filtered.
  • the plurality of partitions may be filtered (e.g., using lOx Genomics CellPlex), such that only singly-occupied partitions are analyzed.
  • the protein information and RNA information may be obtained from the singly-occupied partitions.
  • the protein information may be inferred, e.g., using the gene expression and the protein profile of cells with similar profiles (e.g., obtained from the single-cell analysis).
  • cell overloading may be useful in decreasing reagent waste while providing useful, multiplexed data on gene expression and protein profiles in individual cells.
  • Cells, nuclei, and/or cell beads may be fixed. In some instances, fixation may be performed prior to hybridization of the probe molecules described herein.
  • An example protocol and list of reagents for fixing a sample comprising cells, nuclei, or cell beads is listed below.
  • Example Protocol a. Thaw up to 10 million cells with warm media. b. Centrifuge at 300g for 5 minutes at 4C. c. Remove supernatant without disturbing the cell pellet, and resuspend cell pellet with ImL of cold Cell Resuspension Buffer. d. Transfer to a 1.5-mL tube and measure concentration and viability. If cell suspension has visible debris chunks, Flowmi and count again. e. Centrifuge at 300g for 5 minutes at 4C. f. Remove supernatant without disturbing the cell pellet. g. Using a regular-bore pipette tip, add 1 mL Fixation Buffer to the cell pellet and gently pipette mix 15x. h. Incubate at room temperature for 1 hour. i.
  • RNA and Proteins in Single Cells with Double Fixation may be useful in assaying multiple analytes in single cells.
  • a plurality of cells may be assayed for two analytes: (i) RNA, using a pair of probes (e.g., comprising sequences complementary to a target region of RNA), and (ii) peptides, polypeptides, or proteins, using feature binding groups (e g., antibodies) comprising reporter oligonucleotides.
  • RNA and protein data may be correlated to better understand transcriptomic and proteomic profiles within single cells, e g., by assaying gene and protein expression within a cell.
  • a plurality of cells may be fixed and permeabilized and contacted with (i) a plurality of probes, including a first probe and a second probe and (ii) an antibody comprising a reporter oligonucleotide.
  • the first probe and the second probe may hybridize to a first target region and a second target region of an RNA molecule within the cells to generate a probe-associated molecule, and the antibody may bind to a target protein on or within the cells.
  • barcoding may be performed, e.g., in partitions, to barcode the probe-associated molecule and the reporter oligonucleotide.
  • Barcoded molecules e g., barcoded probe-associated molecules or derivatives thereof and barcoded reporter oligonucleotides or derivatives thereof
  • Barcoded molecules may be sequenced and attributed to single cells based on the barcode sequences.
  • a variety of parameters for preparing the RNA and protein molecules for barcoding within a cell may be tested.
  • it may be advantageous to provide additional fixation operations e.g., after contacting the antibody with the target protein (also referred to herein as “antibody staining”), which may aid in securing the antibody to the target protein during downstream processing, e.g., barcoding.
  • the antibody staining may be performed prior to or following hybridization of the first probe and the second probe (also collectively referred to as “the probes”).
  • the fixation or permeabilization of the cells may be performed using different fixative and permeabilization methods.
  • it may be advantageous to quench an antibody e.g., in a blocking buffer. Such example parameters may be tested experimentally.
  • a plurality of cell fixation schemes may be performed (e.g., as shown in FIG. 29).
  • Multiple experimental groups may be used: 1. Negative control group: cells are contacted with reporter-oligonucleotide conjugated antibodies, fixed and permeabilized, quenched, then contacted with the first and second probes. 2. Group A: cells are fixed and permeabilized and optionally quenched (e.g., in a blocking buffer comprising bovine serum albumin (0.5%) and Tween (0.01%), then contacted with the antibodies, then the probes; 3.
  • Group B cells are fixed and permeabilized and optionally quenched (e.g., in a blocking buffer comprising bovine serum albumin (0.5%) and Tween (0.01%), then contacted with the antibodies, fixed again, quenched again, then contacted with the probes; 4.
  • Group C cells are fixed and permeabilized and optionally quenched (e g., in a blocking buffer comprising bovine serum albumin (0.5%) and Tween (0.01%), then contacted with the antibodies, fixed and permeabilized again, quenched, then contacted with the probes; 5.
  • Group D cells are fixed and permeabilized and optionally quenched (e g., in a blocking buffer comprising bovine serum albumin (0.5%) and Tween (0.01%), then contacted with the probes, rinsed, and then contacted with the antibodies; 6.
  • Group E cells are fixed and permeabilized and optionally quenched (e.g., in a blocking buffer comprising bovine serum albumin (0.5%) and Tween (0.01%), then contacted with the probes, rinsed, and contacted with antibodies in a blocking solution (e.g., 0.5% BSA); 7.
  • Group F cells are fixed and permeabilized using commercially available BioLegend® reagents, washed using BioLegend® Permwash, contacted with the antibodies, then contacted with the probes, 8.
  • Group G cells are fixed and permeabilized using BioLegend® reagents, washed using BioLegend® Permwash, contacted with the antibodies, quenched, fixed and permeabilized again, quenched, then contacted with the probes. All groups may then be subjected to barcoding (e.g.
  • RNA and reporter oligonucleotides are examples of the probes or probe-associated molecules, or derivatives thereof, and reporter oligonucleotides of the antibodies, or derivatives thereof), sequencing, and association of the RNA and reporter oligonucleotides (indicative of a target protein being present in a cell) to single cells.
  • PBMC cells are used.
  • the cells are contacted with reporter oligonucleotide-conjugated antibodies to Perforin (dG9) (ab270703) and Granzyme B (QA18A28) antibodies.
  • dG9 Perforin
  • QA18A28 Granzyme B
  • FIG. 30A shows example data resulting from the experimental groups listed above. From left to right, the samples indicate the fraction of antibody reads that are “usable” (e.g., can be attributed back to a single cell from a barcode sequence) for the following groups: Negative control, Group A, Group B, Group C, Group D, Group E, Group F, Group G. As can be seen in FIG.
  • FIG. 30B shows example data from the same experiment indicating the results of a second fixation operation after antibody staining.
  • the plots indicate a density of antibodies detected, using sequencing and barcode identification, per cell density for two proteins: Perforin (left) and Granzyme (right).
  • Three conditions are categorized: fixation after antibody staining (e g., Groups B, C, and G), negative control, and no second fixation.
  • fixation-after-antibody-staining condition labeled Fix after Ab, 1
  • two peaks are observed, which may indicate two populations of cells, which may be one negative population (e.g., monocytes having background signal or nonspecific staining) and one positive population (e.g., cells having higher signal for the second fixation, e.g., specific staining of Natural Killer and/or cytotoxic T cells) or possibly two positive populations.
  • Further studies may seek to elucidate the specific populations, e.g., by running isotype controls.
  • FIG. 31 shows example gene expression data resulting from the experimental groups listed above. From left to right, the samples indicate the median number of genes detected (e.g., from sequencing the probe-associated molecules or derivatives thereof) for the following groups: Negative control, Group A, Group B, Group C, Group D, Group E, Group F, Group G. As can be seen in FIG. 31, performing an additional fixation operation following antibody staining (Groups B, C, and G) may reduce the number of genes detected (e.g., the sensitivity) of the assay. As such, a tradeoff between antibody sensitivity and gene expression sensitivity when using a second fixation operation may be observed.
  • the negative control group fixing and permeabilization after antibody staining
  • FIGs. 32-37 show example data of gene expression and antibody staining results of some of the experimental groups described above.
  • FIGs. 32A-C show t-SNE plots of the negative control group (cells that are stained with antibodies prior to fixing and permeabilization).
  • FIG. 32A shows a plot of different immune cell clusters, with the oval indicating natural killer and cytotoxic T cell types;
  • FIG. 32B shows the gene expression profile of GZMB in the immune cells (e.g., resulting from barcoding of the probes targeting GZMB, or probe-associated molecules)
  • FIG. 32C shows the antibody staining profile in the immune cells.
  • the GZMB gene expression profile indicates GZMB expression in natural killer and cytotoxic T cells.
  • the antibody staining shows some non-specific staining on monocytes and B cells and limited staining on natural killer cells.
  • FIGs. 33A-C show t-SNE plots for Group D (cells that are fixed and permeabilized, contacted with the probes, then stained with antibodies).
  • FIG. 33A shows a plot of different immune cell clusters, with the oval indicating natural killer and cytotoxic T cell types;
  • FIG. 33B shows the gene expression profile of GZMB in the immune cells (e.g., resulting from barcoding of the probes targeting GZMB, or probe-associated molecules) and
  • FIG. 33C shows the antibody staining profile in the immune cells.
  • the GZMB gene expression profile indicates GZMB expression in natural killer and cytotoxic T cells.
  • the antibody staining shows some non-specific staining on monocytes and B cells, and some specific staining on natural killer cells.
  • FIGs. 34A-C show t-SNE plots for Group B (cells that are fixed and permeabilized, contacted with the antibodies, fixed again, then contacted with the probes).
  • FIG. 34A shows a plot of different immune cell clusters, with the oval indicating natural killer and cytotoxic T cell types;
  • FIG. 34B shows the gene expression profile of GZMB in the immune cells (e.g., resulting from barcoding of the probes targeting GZMB, or probe-associated molecules) and
  • FIG. 34C shows the antibody staining profile in the immune cells.
  • the GZMB gene expression profile indicates GZMB expression in natural killer and cytotoxic T cells.
  • the antibody staining shows some non-specific staining on monocytes, and stronger specific staining on natural killer cells compared to the Group D cells.
  • FIGs. 35A-C show t-SNE plots for Group E (cells that are fixed and permeabilized, contacted with the probes, then stained with antibodies in a blocking solution).
  • FIG. 35A shows a plot of different immune cell clusters, with the oval indicating natural killer and cytotoxic T cell types;
  • FIG. 35B shows the gene expression profile of GZMB in the immune cells (e.g., resulting from barcoding of the probes targeting GZMB, or probe-associated molecules) and
  • FIG. 35C shows the antibody staining profile in the immune cells.
  • the GZMB gene expression profile indicates GZMB expression in natural killer and cytotoxic T cells.
  • the antibody staining shows some non-specific staining.
  • FIGs. 36A-C show t-SNE plots for Group F (cells that are fixed and permeabilized using a commercially available BioLegend® kit, stained with antibodies, then contacted with the probes).
  • FIG. 36A shows a plot of different immune cell clusters, with the oval indicating natural killer and cytotoxic T cell types;
  • FIG. 36B shows the gene expression profile of GZMB in the immune cells (e.g., resulting from barcoding of the probes targeting GZMB, or probe- associated molecules) and
  • FIG. 36C shows the antibody staining profile in the immune cells.
  • the GZMB gene expression profile indicates GZMB expression in natural killer and cytotoxic T cells.
  • the antibody staining shows some non-specific staining.
  • FIGs. 37A-C show t-SNE plots for Group G (cells that are fixed and permeabilized using a commercially available BioLegend® kit, stained with antibodies, fixed again, then contacted with the probes).
  • FIG. 37A shows a plot of different immune cell clusters, with the oval indicating natural killer and cytotoxic T cell types;
  • FIG. 37B shows the gene expression profile of GZMB in the immune cells (e.g., resulting from barcoding of the probes targeting GZMB, or probe-associated molecules) and
  • FIG. 37C shows the antibody staining profile in the immune cells.
  • the GZMB gene expression profile indicates GZMB expression in natural killer and cytotoxic T cells.
  • the antibody staining shows some non-specific staining among monocytes and other cells, but preferential staining on the natural killer cells. More specific staining is observed with the second fixation, compared to without the second fixation.
  • Example 9 Multiplexed assay: barcoding of RNA templated ligation product and reporter oligonucleotide of a feature-binding group
  • the methods described herein may be useful in assaying multiple analytes in a population of cells, nuclei, or cell beads.
  • the cells, nuclei, or cell beads may be contacted with a feature binding group comprising or coupled to a reporter oligonucleotide (comprising a reporter sequence), as described herein.
  • the feature binding group may couple to one or more features (e.g., proteins) of the cell.
  • the cell may also comprise target nucleic acid molecules (e.g., RNA molecules) for assaying.
  • FIG. 38 shows another example multiplexed workflow for assaying cell features (e g., proteins) and target nucleic acid molecules (e.g., RNA molecules).
  • a cell, nucleus, or cell bead may be fixed and permeabilized, e.g., in 4% formaldehyde and 0.01% Tween-20 or a commercially available fixation and permeabilization buffer (e.g., commercially available BioLegend® fixation and permeabilization buffer).
  • the cell, nucleus, or cell bead may be contacted with one or more feature binding groups comprising reporter oligonucleotides.
  • the one or more feature binding groups may specifically bind to cell features (e.g., specific proteins) if present on or within the cell, nucleus, or cell bead.
  • the reporter oligonucleotides may be used to identify the feature binding group and thus the presence or absence of a target cell feature (e.g., specific protein).
  • a plurality of cells may be contacted with a plurality of feature binding groups, which may be the same or different, and may comprise the same or different reporter oligonucleotides.
  • a cell of the plurality of cells may be contacted with different feature binding groups that can bind to different cell features (e.g., different surface or intracellular proteins).
  • each feature binding group comprises a reporter oligonucleotide comprising a barcode sequence that identifies the feature binding group
  • the presence of such different cell features may be assessed (e.g., via sequencing) by the presence of the barcode sequences.
  • the cell, nucleus, or cell bead may be contacted with a first probe and a second probe to generate a probe-associated molecule (e.g., a probe-associated RNA molecule), as described herein.
  • a probe-associated molecule e.g., a probe-associated RNA molecule
  • the cell, nucleus, or cell bead which may optionally be fixed and permeabilized, may comprise a target nucleic acid molecule (e.g., RNA molecule) comprising a first target region and a second target region.
  • the first probe may comprise a first probe sequence that is at least partially complementary to the first target region
  • the second probe may comprise a second probe sequence that is at least partially complementary to the second target region. Hybridization of the first probe sequence to the first target region and the second probe sequence to the second target region may be sufficient to generate the probe-associated molecule.
  • the cell, nucleus, or cell bead may be contacted with a plurality of different probes.
  • the plurality of different probes may specifically hybridize to target regions of target nucleic acid molecules, if present.
  • the probe sequences may comprise probe barcode sequences that may be used to identify the probe.
  • a plurality of cells may be contacted with a plurality of probes, which may be the same or different, and may comprise the same or different sequences (e.g., barcode sequences, probe sequences, adapter sequences).
  • a cell of the plurality of cells may be contacted with different probes that can hybridize to different target regions of a target nucleic acid molecule (e.g., RNA molecule).
  • a target nucleic acid molecule e.g., RNA molecule
  • Each probe may comprise a probe barcode sequence that identifies the probe, and the presence of such different target sequences may be assessed (e.g., via sequencing) by the presence of the probe barcode sequences or the probe sequences.
  • the probe barcode sequences may be used to identify the originating sample or to deconvolve a sequence and identify the sequence as originating from a cell, nucleus, or cell bead (e.g., as shown in FIG. 10).
  • the cell, nucleus, or cell bead may be washed to remove any unbound or non-hybridized probes.
  • the cell, nucleus, or cell bead may then be partitioned (e.g., in a droplet or well) for barcoding, as described herein.
  • the cell, nucleus, or cell bead may be partitioned with a nucleic acid barcode molecule (shown in FIG. 38 as coupled to a bead).
  • the nucleic acid barcode molecule may comprise a barcode sequence and a capture sequence complementary to a sequence of one of the probes (e.g., the first probe or the second probe).
  • the nucleic acid barcode molecule may comprise additional sequences, e.g., a UMI, a primer sequence, a sequencing primer sequence (e.g., P5, P7, Rl, R2 sequences).
  • the capture sequence of the nucleic acid barcode molecule may anneal to the complementary sequence of one of the probes (e.g., the first probe or the second probe), and optionally, an extension reaction may be performed to generate a barcoded nucleic acid molecule comprising the barcode sequence or complement thereof and a sequence of at least one of the probes, or complements thereof.
  • the nucleic acid barcode molecule capture sequence may also anneal to a sequence of the reporter oligonucleotide (not shown in FIG. 38).
  • an extension reaction may be performed to generate an additional barcoded nucleic acid molecule comprising a sequence of the reporter oligonucleotide or complement thereof and the barcode sequence or complement thereof.
  • the barcoded nucleic acid molecule and the additional barcoded nucleic acid molecule may be removed from the partitions and subjected to conditions sufficient for sequencing, e.g., amplification, cleanup, sample-index PCR, etc.
  • Such an example workflow may be useful in obtaining multiplexed information regarding cell features (e.g., proteins) and correlating the features with nucleic acid information, e.g., the presence or genotype of target nucleic acid molecules (e.g., RNA).
  • the processes described herein may be performed in any useful or convenient order.
  • the fixation, permeabilization, contacting with the feature binding groups, and contacting with the first probe and the second probe may occur in any useful order and may be repeated any number of times. Any of these processes, e.g., fixation, permeabilization, contacting with the feature binding groups, and contacting with the first probe and the second probe, may occur in bulk or in partitions.
  • Example 10 -RNA templated ligation for whole transcriptome analysis in tissue samples The methods described herein may be useful in assaying nucleic acid molecules (e.g., mRNA) in tissue samples, e.g., fresh tissue samples, frozen (e g., flash-frozen) tissue samples, etc.
  • whole transcriptome analysis may be performed in tissue samples.
  • a tissue sample may comprise mRNA molecules that can be contacted with a plurality of first probes and second probes.
  • the plurality of first probes and second probes may comprise a set of whole transcriptome analysis probes, such that hundreds, thousands, or millions of RNA targets may be analyzed.
  • the plurality of first probes and second probes may comprise thousands of different first probes and second probes that may hybridize to different target sequences (e.g., coding or non-coding) of mRNA.
  • the plurality of first probes and second probes may have sufficient sequence diversity and coverage to analyze the entire transcriptome of a sample.
  • the plurality of first probes and second probes may comprise gene-specific sequences, which may be species specific (e g., able to distinguish from different animal cell types, e.g., human and mouse).
  • the use of a dual-probe (e.g., using a first probe and a second probe that hybridize to first and second target regions, respectively of an mRNA molecule) approach to conduct mRNA analysis may be advantageous in providing higher analyte sensitivity, improved efficiency of barcoding, and/or discernment of a greater number of barcodes, UMIs, or both, as compared to the use of a single probe (e.g., the 3’ Single Cell Gene Expression solution (lOx Genomics)).
  • Table 1 shows example data of a comparison of the number of UMIs detected in flash-frozen human and mouse tissue samples for whole- transcriptome analysis using either (i) a single probe approach, e.g., as shown and described in FIG.
  • RNA-templated ligation Five different human samples, from the liver, colon, jejunum, ileum, testis, and one mouse sample from the brain are tested. All samples are flash frozen.
  • Each column of the numeric columns of Table 1 illustrate the number of UMIs detected at either 5,000 panel reads per cell (“PRPC”) or 10,000 PRPC in both the RTL (dual-probe whole transcriptome analysis) and SC3P (single-probe whole transcriptome analysis) approaches.
  • PRPC panel reads per cell
  • SC3P single-probe whole transcriptome analysis
  • Table 2 shows example data of a comparison of the number of UMIs detected in fresh mouse tissue samples for whole-transcriptome analysis using either (i) a single probe approach (“SC3P”) or (ii) a dual-probe approach (“RTL”). Five different mouse samples, from the brain, colon, kidney, lung, and liver are tested. All samples are fresh.
  • Each column of the numeric columns of Table 2 illustrate the number of UMIs detected at either 5,000 panel reads per cell (“PRPC”) or 10,000 PRPC in both the RTL (dual-probe whole transcriptome analysis) and SC3P (single-probe whole transcriptome analysis) approaches
  • PRPC panel reads per cell
  • SC3P single-probe whole transcriptome analysis
  • a first workflow for preparation of the sample can comprise securing and sectioning at least a portion of a fixed tissue (e.g., a formalin-fixed paraffin embedded tissue sample).
  • the tissue can be sectioned into one or more 10 to 50 micrometer scrolls, which can then be placed into a sample tube.
  • 3 milliliters (mb) of a non-polar solvent e.g., xylene, neo-clear, orange oil, etc.
  • a non-polar solvent e.g., xylene, neo-clear, orange oil, etc.
  • 3 mL of 100% ethanol can be added to the sample tube and incubated with the one or more scrolls for 30 seconds at ambient temperatures.
  • the ethanol can be aspirated using a Pasteur pipette and discarded. This process can be repeated with 1 mL of 70% ethanol, 1 mL of 50% ethanol, and 1 mL of double distilled water.
  • phosphate buffered saline PBS
  • 1 mL of citrate pH 8.0 crosslinking buffer can be added to the sample tube. If the optional citrate buffer is added, the sample tube can be incubated for 30 minutes to 1 hour at 70 degrees Celsius. The sample can then be centrifuged at 850-2000 relative centrifugal force (ref).
  • a dissociation solution comprising, for example, liberase with low thermolysis (TL), liberase with medium thermolysis (TM), liberase with high thermolysis (TH), collagenases, or the like, or any combination thereof, can be heated at 37 degrees Celsius for 10 minutes. 2 mL of the pre-warmed dissociation solution can be added to the sample and incubated for 20 minutes to 1 hour at 37 degrees Celsius while shaking the sample tube intermittently. Then, using a silanized glass pipette, the scrolls can be triturated 15 to 20 times (e.g., until the solution begins to turn cloudy) to obtain a cell suspension.
  • TL liberase with low thermolysis
  • TM medium thermolysis
  • TH liberase with high thermolysis
  • collagenases or the like, or any combination thereof
  • the dissociated tissue can be passed through a 70 micrometer fdter, and any tissue pieces can be pushed through the filter with the back of a syringe plunger to maximize cell yields and remove debris and undissociated tissue pieces.
  • An additional wash of the 70 micrometer filter can be performed by adding 2 mL of PBS to the filter.
  • the filtrate from the additional wash can be added to the filtrate from the first filtering operation.
  • the combined filtrates can be centrifuged at 850-2000 ref for 5 minutes, the supernatant can be removed without disturbing the pellet formed by centrifugation, and the pellet can be resuspended in 1 mL of chilled PBS.
  • the cell concentration of the resuspended solution can then be determined using a Countess II FL Automated Cell Counter, a Cellaca MX High-Throughput Automated Cell Counter, or the like using a fluorescent dye (e.g., ethidium homodimer- 1, ect.) or AO/PI staining solution, or the like.
  • a fluorescent dye e.g., ethidium homodimer- 1, ect.
  • AO/PI staining solution e.g., AO/PI staining solution, or the like.
  • the resuspended solution may then be used as a sample for the methods and systems described elsewhere herein (e.g., RNA profiling, etc.).
  • a second workflow may comprise securing and sectioning at least a portion of a fixed tissue (e.g., a formalin-fixed paraffin embedded tissue sample).
  • the tissue can be sectioned into one or more 10 to 50 micrometer scrolls, which can then be placed onto a slide.
  • a non-polar solvent e.g., xylene, neo-clear, orange oil, etc.
  • the non-polar solvent can then be removed and discarded, and the process repeated two more times.
  • 1 mL of 100% ethanol can be added to the slide and incubated with the one or more scrolls for 30 seconds at ambient temperatures.
  • the ethanol can be aspirated using a Pasteur pipette and discarded.
  • the ethanol additions may be configured as an ethanol hydration (e.g., the amount of water mixed with the ethanol can increase with each addition.
  • This process can be repeated with 1 mL of 70% ethanol, 1 mL of 50% ethanol, and 1 mL of double distilled water.
  • phosphate buffered saline PBS
  • TL phosphate buffered saline
  • TM medium thermolysis
  • TH liberase with high thermolysis
  • collagenases or the like, or any combination thereof
  • the scrolls can be triturated 15 to 20 times (e.g., until the solution begins to turn cloudy) to obtain a cell suspension.
  • the dissociated tissue can be passed through a 70 micrometer fdter, and any tissue pieces can be pushed through the fdter with the back of a syringe plunger to maximize cell yields and remove debris and undissociated tissue pieces.
  • An additional wash of the 70 micrometer fdter can be performed by adding 2 mL of PBS to the fdter.
  • the fdtrate from the additional wash can be added to the fdtrate from the first filtering operation.
  • the combined filtrates can be centrifuged at 850-2000 ref for 5 minutes, the supernatant can be removed without disturbing the pellet formed by centrifugation, and the pellet can be resuspended in 1 mL of chilled PBS.
  • the cell concentration of the resuspended solution can then be determined using a Countess II FL Automated Cell Counter, a Cellaca MX High-Throughput Automated Cell Counter, or the like using a fluorescent dye (e.g., ethidium homodimer- 1, ect.) or AO/PI staining solution, or the like.
  • the resuspended solution may then be used as a sample for the methods and systems described elsewhere herein (e.g., RNA profiling, etc.).
  • T-cell stimulation with anti-CD3 and anti-CD28 may be used to study antigen specific T-cell response.
  • the use of single-cell transcriptomics may uncover gene-level drives of activation and/or associations with cell surface protein activation markers. Measuring single-cell gene expression on a minute scale time frame, such as that of T-cell stimulation, may be difficult due to the logistics of sampling, counting, and processing cells. Use of fixatives to freeze samples at short time scales may enable single-cell level analysis of these fast processes.
  • two samples of peripheral blood mononuclear cells can be cultured and stimulated using anti-CD3 and anti-CD28, and the resultant stimulated cells can be sampled at 6 different time scales, as shown in FIG. 39A.
  • the samples can be fixed at the various times to freeze the samples in the state of those time frames, multiplexed into an analysis chip, and sequenced.
  • each fixed time sample can be analyzed, and the cells can be prepared as described elsewhere herein (e.g., Example 11).
  • the overlay of FIG. 39B can show clusters of monocytes, T-cells, and B-cells.
  • FIG. 39C shows that, subsequent to T-cell activation, FOS expression can be broadly observed with the T-cell population.
  • CD69 expression can be observed as being upregulated and detected at an RNA level within 15 minutes of stimulation. This may be faster than the protein level detection of CD69, which may take an hour or more for the CD69 to be translated sufficiently for detection.
  • FIG. 39B each fixed time sample can be analyzed, and the cells can be prepared as described elsewhere herein (e.g., Example 11).
  • the overlay of FIG. 39B can show clusters of monocytes, T-cells, and B-cells.
  • FIG. 39C shows that,
  • FIG. 39D the transcripts of Interleukin 2 (IL2) and interferon gamma (fFN-y) can be detected at elevated levels subsequent to stimulation.
  • FIG. 39E shows expression of the VISR gene, which can encode the V-domain Ig suppressor of T-cell activation (VISTA) protein.
  • the protein may be produced at high levels in activated T-cells and tumor infiltrating T-cells.
  • the VISR gene may be transcribed in higher levels after an hour or more of activation in the activated T-cells.
  • a fixed sample can be generated using 4% formaldehyde as the fixation reagent and a 10X Genomics Fixed RNA Sample Preparation Kit.
  • the sample can be fixed for 1 hour at ambient temperature or 16 to 24 hours at 4 degrees Celsius.
  • the same fixation time being used can reduce batch effects observed between samples.
  • each sample can be fixed for 16 hours to reduce batch effects between the samples.
  • the samples can be stored at -80 degrees Celsius until further processing is performed.
  • the samples can be maintained at -80 degrees Celsius for years prior to analysis without significantly decreased performance of a nucleic acid sequencing processes performed on the sample.
  • FIGs. 44 - 46 show examples of methods of preparation for fixed samples, according to some embodiments.
  • the methods of this example can be operated on two 25 micrometer scrolls per sample. In this example, the methods are validated with both expert and novice users.
  • the tissue section transfer and rehydration portion may be similar for various methods of the present example.
  • the tissue rehydration process can be agnostic to the dissociation method used on the tissue sample.
  • the tissue rehydration may be as described elsewhere herein, e.g., Example 11.
  • the dissociation can comprise use of an automated dissociator (e.g., gentleMACS octo dissociator) (FIGs. 44 - 45) or a manual dissociator (FIG. 46).
  • FIG. 44 and 45 can be that FIG. 44 uses the Miltenyi FFPE kit while FIG. 45 uses the methods and systems of the present disclosure.
  • the automated dissociation can comprise addition of 2 mL of a dissociation enzyme mixture (e.g., a 1 milligram/milliliter liberase TH mixture) to a rehydrated sample rehydrated as described elsewhere herein held in a dissociation tube.
  • a dissociation enzyme mixture e.g., a 1 milligram/milliliter liberase TH mixture
  • the sample can be dissociated using the automated dissociator and subsequently centrifuged at 300 ref.
  • the pellet can be resuspended in a supernatant as described elsewhere herein and fdtered through a 70 micrometer strainer.
  • the strainer can be washed with phosphate buffered saline (PBS) to enhance yield before an additional centrifugation at 850 ref for 5 minutes.
  • PBS phosphate buffered saline
  • the supernatant can be removed from the resultant pellet, and the pellet can be resuspended in 0.5 mL of tissue resuspension buffer.
  • the sample can be counted and analyzed as described elsewhere herein.
  • the manual dissociation can comprise adding 100 microliters of a liberase TH 1 mg/mL and using the manual dissociation pestle to dissociate the scrolls. 900 microliters of dissociation enzyme mix can be used to clean the tip of the pestle. The full 1 mL of solution can be incubated for 45 minutes at 37 degrees Celsius (e.g., in a water bath, in a thermomixer at 800 rpm, etc.). Subsequent to the incubation, the sample can be titurated by pipette mixing ten to twenty times before being passed through a 70 micrometer filter.
  • the filter can be washed with 2 mL of buffer to enhance yields before the filtered solution is centrifuged at 850 ref for five minutes, the supernatant is discarded, 0.5 mL of resuspension buffer is added and the sample is ready for further analysis.
  • validation provided a fraction of usable reads of at least about 40%, a fraction of reads mapped to the filtered probe set of at least about 80%, a fraction of reads in cells on high quality preparations of at least about 60%, and at least about 90% valid barcodes.
  • the yield of the examples was at least about 400,000 cells per sample for the 225 micrometer sections, with an observed cells to expected cells recovered of at least about 50%. Additionally, the pseudobulk gene expression versus cytassist assay ratio was at least about 0.7, and the observed targeted gene cell-UMI recovery median was at least about 0.8.
  • FIG. 47 provides a table of the pass percentages and other properties for the various procedures of the present example, according to some embodiments.
  • the table shows the enhanced performance of the methods of the present disclosure versus other sample preparation methods, and this data is further seen in FIG. 48, which shows examples of various tissues processed by the workflows of FIGs. 44 - 46 (labeled as 4801, 4802, and 4803, respectively), according to some embodiments.
  • the table shown in FIG. 47 provides a design input requirement (DIR) which may correspond to a success threshold or validation threshold for an assay.
  • DIR design input requirement
  • the validation threshold for cell yield from two scrolls may be greater than or equal to 400,000 cells.
  • Formalin fixed, paraffin embedded (FFPE) tissue samples may be a large repository of human tumor specimens.
  • Gene expression profiling of FFPE samples may provide insights into disease etiology.
  • the methods described herein may be useful for profiling gene expression in FFPE samples without RNA degradation or with significantly reduced RNA degradation at single cell resolution.
  • the methods described herein may permit single cell FFPE sequencing for detection of whole transcriptome gene expression in dissociated FFPE tissues via in situ hybridization, ligation, and barcoding of RNA-specific probe pairs.
  • FFPE sections from nine different human cancer tissue blocks may be profiled using the methods described herein.
  • FFPE sectioned tissue samples (e.g., scrolls) of each cancer block may be dissociated into a single cell suspension using a combination of enzymatic digestion and mechanical dissociation.
  • the dissociated cells may be processed using the workflow described elsewhere herein, for example, as shown in FIG. 42.
  • Genes and UMIs per cell may demonstrate sensitive of the single cell assay on FFPE tissues at single cell resolution.
  • Cell numbers post dissociation from two 25 pm sectioning of each tissue type may be sufficient for hybridization and partitioning, for example, partitioning into droplets.
  • Table 3 shows example gene expression metrics for the profiled tissues. Gene and UMI metrics may be measured at 10,000 raw reads per cell. Post dissociation cell yields of tissue sections may be listed along with the tissue cross section dimensions.
  • FIGs. 49A-49D show example cell type clustering in single cell FFPE.
  • FIG. 49A shows an example uniform manifold approximation and projection (UMAP) projection of single cells taken from prostate cancer FFPE. Cell types may be manually annotated.
  • FIG. 49B show MYH1 1 gene expression showing higher expression in the perivascular-like (PVL) cell cluster.
  • FIG. 49C shows an example UMAP projection of single cells taken from colorectal cancer FFPE.
  • FIG. 49D shows MZB1 gene expression showing higher expression in the plasma cell cluster.
  • Cells may be isolated from formaldehyde fixed & paraffin embedded (FFPE) tissue sections for use with the methods described elsewhere herein.
  • Cell isolation may include deparaffmizing and rehydrating fixed tissue sections.
  • the deparaffinized and rehydrated tissue sections may be dissociated using a dissociation enzyme mix and a pellet pestle or a gentleMACS Octo Dissociator.
  • the dissociated cells may be resuspended in a tissue resuspension buffer or quenching buffer prior to cell counting. At least 400,000 cells may be used for singleplex cell analysis. At least 100,000 cells may be used for multiplex cell analysis.
  • a pilot multiplex assay may be run using 10,000 cells per probe barcode for a singleplex assay or a four sample multiplex assay. For multiplexing sixteen samples, 8,000 cells may be loaded per probe barcode. Cell isolation may be demonstrated using FFPE tissue samples ranging from one to ten years in age. Each tissue sample may yield different amounts of material and data quality depending on age, tissue type, pre-fixation tissue quality, tissue density, and size and area of tissue in the scrolls.
  • Cell isolation may be demonstrated using two or more 25 micrometer (pm) or 50 pm tissue sections from a diverse spectrum of tissue types. For human tissue, two or more 25 pm sections may be used. For mouse tissue, two or more 50 jam sections may be used. Some tissue types may use more than two sections to yield sufficient cells for further processing. FFPE block properties may also impact yields. In some cases, a single 25 pm or 50 pm tissue section may yield sufficient cells for further processing.
  • Tissue may be sectioned and dissociated using pestle dissociation or gentleMACS Octo Dissociator protocols.
  • tissue sections e.g., scrolls
  • the deparaffmized sample may be dissociated using pestle dissociation.
  • the tissue sections e.g., scrolls
  • the deparaffmized sample may then be processed by the Octo Dissociator.
  • the FFPE block Prior to cell isolation, the FFPE block may be inspected for extend of dehydration.
  • the Tissue black may be faced to remove a portion of the paraffin and expose the tissue.
  • the faced tissue block may be placed for a sufficient time in an ice water back with the block facing the water to rehydrate of the tissue block.
  • the first 5 pm of the tissue may be removed and disposed.
  • the microtome may be used to section the fixed tissue into 25 pm or 50 pm sections (e.g., scrolls). After discarding the first section, additional section(s) from the rehydrated FFPE block may be transferred into a 1.5 mL tube or gentleMACS C tube while maintaining the scrolls intact.
  • the scrolls may be stored at 4 °C for up to one week.
  • One mL of xylene may be added to the 1.5 mL tube or 3 mL of xylene to the gentleMACS tube.
  • the tubes may be incubated for 10 minutes at room temperature. Using a Pasteur pipette, the liquid may be removed from the tubes without braking or otherwise damaging the scrolls.
  • the xylene wash may be repeated for a total of three washes.
  • One mL of 100% ethanol may be added to the 1.5 mL tube or 3 mL of 100% ethanol may be added to the gentleMACS C tube.
  • the tubes may be incubated at room temperature for 30 seconds.
  • the liquid may be removed from the tube using a Pasteur pipette.
  • One mL of 70% ethanol may be added to the 1.5 mL tube or 3 mL of 70% ethanol may be added to the gentleMACS C tube.
  • the tubes may be incubated at room temperature for 30 seconds.
  • the liquid may be removed from the tube using a Pasteur pipette without breaking or damaging the scrolls.
  • One mL of 50% ethanol may be added to the 1.5 mL tube or 3 mL of 50% ethanol may be added to the gentleMACS C tube.
  • the tubes may be incubated at room temperature for 30 seconds.
  • the liquid may be removed from the tube using a Pasteur pipette without breaking or damaging the scrolls.
  • One mL of nuclease-free water may be added to the tube.
  • the tube may be incubated for 30 seconds at room temperature.
  • PBS phosphate buffered saline
  • the PBS buffer may be removed from the 1.5 mL tube using a Pasteur pipette without breaking the scrolls.
  • 100 pL of dissociation enzyme mix may be added to the tube, as shown in FIG. 50A.
  • the tissue scrolls may be dissociated using a 1.5 mL pellet pestle to break the scrolls into smaller pieces, as shown in FIG. 50B.
  • 900 pL of dissociation enzyme mix may be added to the tube while rinsing the pestle tip into the tube to collect any additional tissue pieces sticking to the pestle.
  • the tissue pieces may be mixed with the dissociation enzyme mix using a pipette.
  • the tube may be incubated for 45 minutes at 37 °C in a thermomixer at 800 rpm.
  • the tube may be incubated at 37 °C for 45 minutes.
  • the sample may be mixed at 15 minute intervals via inversion.
  • the tissue pieces may be triturated by pipetting 10 to 20 times.
  • the sample may be aspirated by pushing the tissue pieces and solution through a 23 -gauge needle five or more times to improve cell recovery.
  • the suspension may be passed through a pre-separation filter (30 pm) placed on a 5- or 15-mL tube placed on ice.
  • the original tube and filter may each be rinsed with 1 mL chilled PBS to minimize cell loss.
  • the filtrate may be collected in the 5- or 15-mL tube.
  • the tube comprising the cell suspension may be centrifuged at 850 ref at 4 °C for 5 minutes. The supernatant may be removed without disturbing the pellet.
  • the pellet may be resuspended in 0.5 mL chilled tissue resuspension buffer or quenching buffer. The mixture may be pipetted five or more times to mix and maintained on ice.
  • the cell concentration of the fixed tissue sample may be determined using an automated cell counter (e.g., Countess II or Cellaca MX) or hemocytometer. The sample may immediately be used for analysis described elsewhere herein.
  • the PBS may be removed from the gentleMACS C tube without breaking the scrolls.
  • Two mL of dissociation enzyme mix may be added to the tube and it may be closed securely.
  • the tube may be placed in the gentleMACS Octo Dissociator, applying Heting Units and running the gentleMACS Program 37C FFPE 1, as shown in FIG. 51.
  • the program may be run for about 48 minutes.
  • the tube may be detached from the gentleMACS Octo Dissociator and visually inspected to ensure the scrolls have been dissociated.
  • the tube may be centrifuged at about 300 ref for 1 min and the resultant pellet may be resuspended in the supernatant.
  • the suspension may be passed through a pre-separation filter (30 pm) placed on a 15-mL tube on ice.
  • the original gentleMACS tube and filter may be rinsed with 2 mL of chilled PBS to minimize cell loss.
  • the filtrate may be collected in the tube.
  • the cell suspension may be centrifuged at 850 ref at 4 °C for 5 min. The supernatant may be removed without disturbing the pellet.
  • the pellet may be resuspended in 0.5 mL chilled tissue resuspension buffer or quenching buffer. The pellet may be mixed with the buffer by pipetting the mixture five or more time while on ice.
  • the cell concentration of the fixed tissue sample may be determined using an automated cell counter (e.g., Countess II or Cellaca MX) or hemocytometer. The sample may immediately be used for analysis described elsewhere herein.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Analytical Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Biophysics (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Medicinal Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Plant Pathology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Toxicology (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Virology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

L'invention concerne des systèmes et des procédés de traitement de biomolécules (par exemple, des molécules d'acide nucléique, des protéines) à partir d'un échantillon. Un procédé de traitement de biomolécules peut comprendre l'hybridation d'une molécule de sonde à une région cible d'une molécule d'acide nucléique (par exemple, une molécule d'acide ribonucléique (ARN) et codant pour le complexe sonde-molécule d'acide nucléique ou des dérivés de celui-ci. Un tel procédé peut comprendre l'exécution d'une réaction d'acide nucléique, par exemple l'extension, la dénaturation et l'amplification. Un procédé de traitement d'un échantillon peut comprendre l'hybridation de sondes à (i) des régions cibles d'une molécule d'acide nucléique (par exemple, une molécule d'ARN) et (ii) un oligonucléotide rapporteur d'un groupe de liaison de caractéristiques, ainsi que le codage à barres des molécules associées à une sonde. Un ou plusieurs processus des procédés décrits dans la présente invention peuvent être réalisés à l'intérieur d'un compartiment, telle qu'une gouttelette ou un puits.
PCT/US2023/026504 2022-06-29 2023-06-28 Analyse d'acides nucléiques et de protéines à l'aide de sondes WO2024006392A1 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US18/217,310 US20240002914A1 (en) 2022-06-29 2023-06-30 Probe-based analysis of nucleic acids and proteins
US18/370,826 US20240019422A1 (en) 2022-06-29 2023-09-20 Probe-based analysis of nucleic acids and proteins

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202263356887P 2022-06-29 2022-06-29
US63/356,887 2022-06-29
US202263386135P 2022-12-05 2022-12-05
US63/386,135 2022-12-05

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US18/217,310 Continuation US20240002914A1 (en) 2022-06-29 2023-06-30 Probe-based analysis of nucleic acids and proteins

Publications (1)

Publication Number Publication Date
WO2024006392A1 true WO2024006392A1 (fr) 2024-01-04

Family

ID=87514115

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/026504 WO2024006392A1 (fr) 2022-06-29 2023-06-28 Analyse d'acides nucléiques et de protéines à l'aide de sondes

Country Status (2)

Country Link
US (2) US20240002914A1 (fr)
WO (1) WO2024006392A1 (fr)

Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6265552B1 (en) 1993-07-30 2001-07-24 Affymax Technologies N.V. Biotinylation of proteins
US20100105112A1 (en) 2006-08-07 2010-04-29 Christian Holtze Fluorocarbon emulsion stabilizing surfactants
WO2013192292A1 (fr) * 2012-06-21 2013-12-27 Justin Lamb Analyse de séquence d'acide nucléique spécifique d'un locus multiplexe massivement parallèle
US20140155295A1 (en) 2012-08-14 2014-06-05 10X Technologies, Inc. Capsule array devices and methods of use
US20140378345A1 (en) 2012-08-14 2014-12-25 10X Technologies, Inc. Compositions and methods for sample processing
US20150292988A1 (en) 2014-04-10 2015-10-15 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same
US20150376609A1 (en) 2014-06-26 2015-12-31 10X Genomics, Inc. Methods of Analyzing Nucleic Acids from Individual Cells or Cell Populations
US20180105808A1 (en) 2016-10-19 2018-04-19 10X Genomics, Inc. Methods and systems for barcoding nucleic acid molecules from individual cells or cell populations
US20190064173A1 (en) 2017-08-22 2019-02-28 10X Genomics, Inc. Methods of producing droplets including a particle and an analyte
US20190100632A1 (en) 2017-10-04 2019-04-04 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
US20190177800A1 (en) 2017-12-08 2019-06-13 10X Genomics, Inc. Methods and compositions for labeling cells
WO2019165318A1 (fr) 2018-02-22 2019-08-29 10X Genomics, Inc. Analyse induite par ligature d'acides nucléiques
US10428326B2 (en) 2017-01-30 2019-10-01 10X Genomics, Inc. Methods and systems for droplet-based single cell barcoding
US20190367969A1 (en) 2018-02-12 2019-12-05 10X Genomics, Inc. Methods and systems for analysis of chromatin
US20190367997A1 (en) 2018-04-06 2019-12-05 10X Genomics, Inc. Systems and methods for quality control in single cell processing
US20200033237A1 (en) 2018-06-25 2020-01-30 10X Genomics, Inc. Methods and systems for cell and bead processing
US10550429B2 (en) 2016-12-22 2020-02-04 10X Genomics, Inc. Methods and systems for processing polynucleotides
WO2020176788A1 (fr) * 2019-02-28 2020-09-03 10X Genomics, Inc. Profilage d'analytes biologiques avec des réseaux d'oligonucléotides à codes-barres spatiaux
WO2021237087A1 (fr) 2020-05-22 2021-11-25 10X Genomics, Inc. Analyse spatiale pour détecter des variants de séquence

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110027862A1 (en) * 2009-06-29 2011-02-03 Life Technologies Corporation Sample stabilization
US9273349B2 (en) * 2013-03-14 2016-03-01 Affymetrix, Inc. Detection of nucleic acids
CN106574298A (zh) * 2014-06-26 2017-04-19 10X基因组学有限公司 用于样品分析的方法和组合物
US20210389325A1 (en) * 2016-10-28 2021-12-16 Frontier Diagnostics, Llc Imaging mass spectrometry and uses thereof
US10815525B2 (en) * 2016-12-22 2020-10-27 10X Genomics, Inc. Methods and systems for processing polynucleotides

Patent Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6265552B1 (en) 1993-07-30 2001-07-24 Affymax Technologies N.V. Biotinylation of proteins
US20100105112A1 (en) 2006-08-07 2010-04-29 Christian Holtze Fluorocarbon emulsion stabilizing surfactants
WO2013192292A1 (fr) * 2012-06-21 2013-12-27 Justin Lamb Analyse de séquence d'acide nucléique spécifique d'un locus multiplexe massivement parallèle
US20140155295A1 (en) 2012-08-14 2014-06-05 10X Technologies, Inc. Capsule array devices and methods of use
US20140378345A1 (en) 2012-08-14 2014-12-25 10X Technologies, Inc. Compositions and methods for sample processing
US20150292988A1 (en) 2014-04-10 2015-10-15 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same
US20150376609A1 (en) 2014-06-26 2015-12-31 10X Genomics, Inc. Methods of Analyzing Nucleic Acids from Individual Cells or Cell Populations
US20180105808A1 (en) 2016-10-19 2018-04-19 10X Genomics, Inc. Methods and systems for barcoding nucleic acid molecules from individual cells or cell populations
WO2018075693A1 (fr) 2016-10-19 2018-04-26 10X Genomics, Inc. Procédés et systèmes de codage de molécules d'acide nucléique provenant de cellules individuelles ou de populations de cellules
US10550429B2 (en) 2016-12-22 2020-02-04 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10428326B2 (en) 2017-01-30 2019-10-01 10X Genomics, Inc. Methods and systems for droplet-based single cell barcoding
US20190064173A1 (en) 2017-08-22 2019-02-28 10X Genomics, Inc. Methods of producing droplets including a particle and an analyte
US20190100632A1 (en) 2017-10-04 2019-04-04 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
US20190323088A1 (en) 2017-12-08 2019-10-24 10X Genomics, Inc. Methods and compositions for labeling cells
US20190177800A1 (en) 2017-12-08 2019-06-13 10X Genomics, Inc. Methods and compositions for labeling cells
US20190367969A1 (en) 2018-02-12 2019-12-05 10X Genomics, Inc. Methods and systems for analysis of chromatin
WO2019165318A1 (fr) 2018-02-22 2019-08-29 10X Genomics, Inc. Analyse induite par ligature d'acides nucléiques
US20200239874A1 (en) 2018-02-22 2020-07-30 10X Genomics, Inc. Ligation mediated analysis of nucleic acids
US20190367997A1 (en) 2018-04-06 2019-12-05 10X Genomics, Inc. Systems and methods for quality control in single cell processing
US20200033237A1 (en) 2018-06-25 2020-01-30 10X Genomics, Inc. Methods and systems for cell and bead processing
WO2020176788A1 (fr) * 2019-02-28 2020-09-03 10X Genomics, Inc. Profilage d'analytes biologiques avec des réseaux d'oligonucléotides à codes-barres spatiaux
WO2021237087A1 (fr) 2020-05-22 2021-11-25 10X Genomics, Inc. Analyse spatiale pour détecter des variants de séquence

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
FANG ET AL.: "Fluoride-Cleavable Biotinylation Phosphoramidite for 5'-end-Labelling and Affinity Purification of Synthetic Oligonucleotides", NUCLEIC ACIDS RES., vol. 31, no. 2, 15 January 2003 (2003-01-15), pages 708 - 715
HUGHES L, D ET AL., PLOS ONE, vol. 9, no. 2, 4 February 2014 (2014-02-04), pages e87649

Also Published As

Publication number Publication date
US20240002914A1 (en) 2024-01-04
US20240019422A1 (en) 2024-01-18

Similar Documents

Publication Publication Date Title
US11952626B2 (en) Probe-based analysis of nucleic acids and proteins
US20230167433A1 (en) Methods and systems for increasing cell recovery efficiency
US20230167432A1 (en) Methods and systems for analysis and identification of barcode multiplets
US20240124871A1 (en) Drug screening methods
US20210230584A1 (en) Ligation mediated analysis of nucleic acids
US20220228220A1 (en) Methods and compositions for labeling cells
WO2023076528A2 (fr) Méthodes de préparation d'échantillons
EP4022309B1 (fr) Procédés de détection et d'analyse d'analyte
US20220403375A1 (en) Methods for enriching nucleic acid libraries for target molecules that do not produce artefactual antisense reads
US11852628B2 (en) Methods and systems for characterizing analytes from individual cells or cell populations
US20230167496A1 (en) Compositions and methods for isolating nucleic acid molecules
US20240002914A1 (en) Probe-based analysis of nucleic acids and proteins
US20240209346A1 (en) Nucleic acid processing via circularization
US20240053337A1 (en) Compositions and methods for single cell analyte detection and analysis
US20240068029A1 (en) Compositions and methods for characterization of antigen-binding molecule antigen-binding sites and uses thereof
CN117015617A (zh) 基于探针的核酸和蛋白质分析
US20240191291A1 (en) Compositions, systems and methods for enzyme optimization
WO2024076908A1 (fr) Compositions et procédés d'analyse de sites d'insertion génomique d'acides nucléiques exogènes
WO2023060110A1 (fr) Procédés d'analyse de cellules immunitaires
WO2023225259A1 (fr) Compositions et procédés de caractérisation de molécules de liaison à l'antigène à partir de cellules uniques
WO2022182664A1 (fr) Procédé de compartimentage d'épitope de nouveaux anticorps monoclonaux
WO2024015378A1 (fr) Procédés et systèmes de caractérisation de molécules de liaison à l'antigène exprimées par des cellules immunitaires

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: 23748139

Country of ref document: EP

Kind code of ref document: A1