US20180282804A1 - Methods and systems for processing polynucleotides - Google Patents

Methods and systems for processing polynucleotides Download PDF

Info

Publication number
US20180282804A1
US20180282804A1 US16/000,803 US201816000803A US2018282804A1 US 20180282804 A1 US20180282804 A1 US 20180282804A1 US 201816000803 A US201816000803 A US 201816000803A US 2018282804 A1 US2018282804 A1 US 2018282804A1
Authority
US
United States
Prior art keywords
fragments
polynucleotide
partitions
partition
nucleotides
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/000,803
Inventor
Benjamin Hindson
Serge Saxonov
Kevin Ness
Paul Hardenbol
Michael Schnall-Levin
Mirna Jarosz
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
10X Genomics Inc
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=58558428&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20180282804(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US14/104,650 external-priority patent/US9567631B2/en
Priority claimed from US14/175,973 external-priority patent/US9388465B2/en
Priority to US16/000,803 priority Critical patent/US20180282804A1/en
Application filed by 10X Genomics Inc filed Critical 10X Genomics Inc
Priority to US16/052,486 priority patent/US10323279B2/en
Priority to US16/052,431 priority patent/US10273541B2/en
Publication of US20180282804A1 publication Critical patent/US20180282804A1/en
Priority to US16/165,389 priority patent/US10533221B2/en
Priority to US16/212,441 priority patent/US10752949B2/en
Priority to US16/231,142 priority patent/US10584381B2/en
Priority to US16/231,185 priority patent/US10400280B2/en
Priority to US16/246,322 priority patent/US10597718B2/en
Priority to US16/294,769 priority patent/US10450607B2/en
Priority to US16/395,090 priority patent/US10669583B2/en
Priority to US16/435,362 priority patent/US10626458B2/en
Priority to US16/435,417 priority patent/US10752950B2/en
Priority to US16/698,740 priority patent/US11473138B2/en
Priority to US16/736,323 priority patent/US20200255894A1/en
Priority to US16/844,141 priority patent/US11441179B2/en
Priority to US16/998,425 priority patent/US11035002B2/en
Priority to US16/998,414 priority patent/US11021749B2/en
Priority to US17/314,526 priority patent/US11359239B2/en
Priority to US17/392,610 priority patent/US20220098659A1/en
Priority to US18/186,088 priority patent/US20240002929A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • C12Q1/6869Methods for sequencing
    • C12Q1/6874Methods for sequencing involving nucleic acid arrays, e.g. sequencing by 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
    • C12Q1/6806Preparing 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
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/6853Nucleic acid amplification reactions using modified primers or templates
    • C12Q1/6855Ligating adaptors
    • 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
    • C12Q1/6869Methods for sequencing

Definitions

  • No. 15/376,582 is also a continuation-in-part of U.S. patent application Ser. No. 14/250,701, filed on Apr. 11, 2014, which is a continuation of U.S. patent application Ser. No. 14/175,973, filed on Feb. 7, 2014, now U.S. Pat. No. 9,388,465, which claims priority to U.S. Provisional Application No. 61/844,804, filed on Jul. 10, 2013, U.S. Provisional Application No. 61/840,403, filed on Jun. 27, 2013, U.S. Provisional Application No. 61/800,223, filed on Mar. 15, 2013, and U.S. Provisional Application No. 61/762,435, filed on Feb. 8, 2013, each of which is entirely incorporated herein by reference for all purposes.
  • polynucleotides and polynucleotide fragments are critical aspects of a wide variety of technologies, including polynucleotide sequencing.
  • Polynucleotide sequencing continues to find more widespread use in medical applications such as genetic screening and genotyping of tumors.
  • Many polynucleotide sequencing methods rely on sample processing techniques solely utilizing random fragmentation of polynucleotides. Such random, uncontrolled fragmentation can introduce several problems in downstream processing. For example, these methods may produce fragments with large variation in length, including a large number or fraction of sequences that are too long to be sequenced accurately. This results in a loss of sequence information.
  • a method provided herein comprises: (a) providing a target polynucleotide; (b) fragmenting said target polynucleotide to generate a plurality of non-overlapping first polynucleotide fragments; (c) partitioning said first polynucleotide fragments to generate partitioned first polynucleotide fragments, wherein at least one partition of said partitioned first polynucleotide fragments comprises a first polynucleotide fragment with a unique sequence within said at least one partition; and (d) fragmenting said partitioned first polynucleotide fragments, to generate a plurality of non-overlapping second polynucleotide fragments.
  • a third and fourth set of polynucleotide fragments are generated by performing the method described above and additionally performing a method comprising: (a) fragmenting said target polynucleotide to generate a plurality of non-overlapping third polynucleotide fragments; (b) partitioning said third polynucleotide fragments to generate partitioned third polynucleotide fragments, wherein at least one partition of said partitioned third polynucleotide fragments comprises a third polynucleotide fragment with a unique sequence within said at least one partition; and (c) fragmenting said partitioned third polynucleotide fragments to generate a plurality of non-overlapping fourth polynucleotide fragments.
  • the third polynucleotide fragments may overlap with the first polynucleotide fragments.
  • the fourth polynucleotide fragments may overlap with the second polynucleotide fragments.
  • the target polynucleotide may be, for example, DNA, RNA, cDNA, or any other polynucleotide.
  • At least one of the first, second, third, and fourth polynucleotide fragments are generated by an enzyme.
  • the enzyme may be a restriction enzyme.
  • the restriction enzyme used to generate the first polynucleotide fragments may be different from the restriction enzyme used to generate the third polynucleotide fragments.
  • the restriction enzyme used to generate the second polynucleotide fragments may be different from the restriction enzyme used to generate the fourth polynucleotide fragments.
  • the restriction enzymes may have a recognition site of at least about six nucleotides in length.
  • the fragments can be of a variety of lengths.
  • the first and/or third polynucleotide fragments may have a median length of least about 10,000 nucleotides.
  • the second or fourth polynucleotide fragments may have a median length of less than about 200 nucleotides.
  • the fragments can be attached to barcodes.
  • the second polynucleotide fragments and/or the fourth polynucleotide fragments may be attached to barcodes, to generate barcoded second and/or fourth polynucleotide fragments.
  • the barcodes may be polynucleotide barcodes.
  • the attachment of the barcodes to the polynucleotide fragments may be performed using an enzyme.
  • the enzyme may be a ligase.
  • the barcoded fragments may be pooled. Unpooled or pooled barcoded fragments may be sequenced.
  • one or more steps of the methods described in this disclosure may be performed within a device.
  • the device may comprise at least one well.
  • the well may be a microwell. Any of the partitioning steps described in this disclosure may be performed by dispensing into a microwell.
  • the microwell may comprise reagents. These reagents may be any reagent, including, for example, barcodes, enzymes, adapters, and combinations thereof.
  • the reagents may be physically separated from a polynucleotide sample placed in the microwell. This physical separation may be accomplished by containing the reagents within a microcapsule that is placed within a microwell. The physical separation may also be accomplished by dispensing the reagents in the microwell and overlaying the reagents with a layer that is, for example, dissolvable, meltable, or permeable prior to introducing the polynucleotide sample into the microwell. This layer may be, for example, an oil, wax, membrane, or the like.
  • the microwell may be sealed at any point, for example after addition of the microcapsule, after addition of the reagents, or after addition of either of these components plus a polynucleotide sample.
  • Partitioning may also be performed by a variety of other means, including through the use of fluid flow in microfluidic channels, by emulsification, using spotted arrays, by surface acoustic waves, and by piezoelectric droplet generation.
  • Additional methods of fragmenting nucleic acids that are compatible with the methods provided herein include mechanical disruption, sonication, chemical fragmentation, treatment with UV light, and heating, and combinations thereof. These methods may be used to fragment, for example, the partitioned first or third polynucleotide fragments described above.
  • Partitioning may be done at any time.
  • the first polynucleotide fragments and/or the third polynucleotide fragments may each be further partitioned into two or more partitions before further processing.
  • This disclosure provides methods for pseudo-random fragmentation of polynucleotides.
  • such methods comprise: (a) providing a target polynucleotide; (b) fragmenting said target polynucleotide to generate a plurality of first polynucleotide fragments; (c) partitioning said first polynucleotide fragments to generate partitioned first polynucleotide fragments, such that at least one partition comprises a first polynucleotide fragment with a unique sequence within said at least one partition; and (d) fragmenting said partitioned first polynucleotide fragments with at least one restriction enzyme in at least one partition, to generate a plurality of second polynucleotide fragments, wherein said partitioned first polynucleotide fragment is fragmented with at least two restriction enzymes across all partitions.
  • At least two restriction enzymes are disposed within the same partition. In some cases, at least two restriction enzymes are disposed across a plurality of different partitions.
  • the pseudo-random fragmentation methods can be performed in order to yield fragments of a certain size.
  • at least about 50% of the nucleotides within a target polynucleotide are within about 100 nucleotides of a restriction site of a restriction enzyme used to perform pseudo-random fragmentation.
  • at most about 25% of the nucleotides within a target polynucleotide are within about 50 nucleotides of a restriction site of a restriction enzyme used to perform pseudo-random fragmentation.
  • at most about 10% of the nucleotides within a target polynucleotide are more than about 200 nucleotides from a restriction site a restriction enzyme used to perform pseudo-random fragmentation.
  • a polynucleotide may be treated with two or more restriction enzymes concurrently or sequentially.
  • the pseudo-randomly fragmented polynucleotides may be attached to barcodes, to generate barcoded polynucleotide fragments.
  • the barcoded polynucleotides may be pooled and sequenced.
  • the number of partitions holding the partitioned first polynucleotide fragments may be at least about 1,000 partitions.
  • the volume of these partitions may be less than about 500 nanoliters.
  • Each enzyme may occupy an equivalent number of partitions, or each enzyme may occupy a different number of partitions.
  • This disclosure provides methods for recycling certain unwanted reaction side products back into starting materials that can be used to generate a desired product.
  • these methods comprise: (a) providing a first polynucleotide, a second polynucleotide, a first restriction enzyme, and a second restriction enzyme, wherein said first polynucleotide comprises a target polynucleotide or a fragment thereof; and (b) attaching said first polynucleotide to said second polynucleotide, to generate a polynucleotide product, wherein said first restriction enzyme cuts a polynucleotide generated by attachment of said first polynucleotide to itself, said second restriction enzyme cuts a polynucleotide generated by attachment of said second polynucleotide to itself, and neither said first restriction enzyme nor said second restriction enzyme cuts said polynucleotide product.
  • the first polynucleotide may be generated in the same reaction volume as the polynucleotide product, or in a different reaction volume.
  • the target polynucleotide may be, for example, a fragment of genomic DNA.
  • the second polynucleotide may be generated in the same reaction volume as the polynucleotide product, or in a different reaction volume.
  • the second polynucleotide may be, for example, a barcode or an adapter.
  • the first restriction enzyme may have a recognition site of at most about four nucleotides in length.
  • the second restriction enzyme may have a recognition site of at least about six nucleotides in length.
  • the first restriction enzyme may have a recognition site of about four nucleotides in length.
  • the second restriction enzyme may have a recognition site of at least about five nucleotides in length.
  • the first and second restriction enzymes may generate ligation compatible ends. These ends may have single-stranded overhangs (i.e., “sticky ends”) or be blunt. The sticky ends may match in sequence and orientation, to allow ligation.
  • the attachment step may be performed by ligation.
  • sequence 5′ to the ligation compatible end generated by the first restriction enzyme may be different from the sequence 5′ to the ligation compatible end generated by the second restriction enzyme. This will ensure that the desired product cannot be re-cut by either restriction enzyme.
  • sequence 3′ to the ligation compatible end generated by the first restriction enzyme may be different from the sequence 3′ to the ligation compatible end generated by the second restriction enzyme. This will ensure that the desired product cannot be re-cut by either restriction enzyme. Given the criteria provided throughout this specification, one of ordinary skill in the art will recognize that many pairs of enzymes are suitable for use with this method.
  • the recycling may provide increased yield of the desired product, for example at least about 75% (w/w).
  • polynucleotide fragment generated by any of the methods provided herein, devices for performing the methods provided herein, and systems for performing the methods provided herein.
  • the methods provided in this disclosure may also be used with each other.
  • the non-overlapping fragmentation methods may be used alone and/or with the pseudo-random fragmentation methods and/or with the restriction enzyme-mediated recycling methods.
  • the pseudo-random fragmentation methods may be used alone and/or with the non-overlapping fragmentation methods and/or with the restriction enzyme-mediated recycling methods.
  • the restriction enzyme-mediated recycling methods may be used alone and/or with the non-overlapping fragmentation methods and/or with the pseudo-random fragmentation methods.
  • FIG. 1 is a schematic representation of overlapping and non-overlapping deoxyribonucleic acid (DNA) fragments.
  • FIG. 2 is a schematic representation of methods of generating non-overlapping DNA fragments for DNA sequencing.
  • FIG. 2 discloses SEQ ID NOS 8-10, respectively, in order of appearance.
  • FIG. 3 shows a distribution of DNA fragment size after simulating generation of 1 Mbp random DNA sequences followed by cutting the sequences with a 6 Mer cutter, StuI (AGG/CCT).
  • FIG. 4 shows a distribution of DNA fragment size after simulating generation of 1 Mbp random DNA sequences followed by cutting the sequences with a 4 Mer cutter, CviQI (G/TAC).
  • FIG. 5 shows a distribution of DNA fragment size after simulating the generation of a 1 Mbp random DNA sequence followed by cutting the sequences with seven 4 Mer cutters: (1) CviQI (G/TAC), (2) BfaI (C/TAG), (3) HinP1I (G/CGC), (4) CviAII (C/ATG), (5) Taq ⁇ I (T/CGA), (6) Msel (T/TAA), and (7) MspI (C/CGG).
  • CviQI G/TAC
  • BfaI C/TAG
  • HinP1I G/CGC
  • CviAII C/ATG
  • Taq ⁇ I T/CGA
  • Msel T/TAA
  • MspI C/CGG
  • FIG. 6 shows the generation of unwanted byproducts (“Side products”) during ligation of adapters to genomic DNA fragments and the recycling of the unwanted byproducts into starting materials (“Genomic DNA”, “Adapter 1”, and “Adapter 2”) by paring of appropriate restriction enzymes (here, MspI and NarI).
  • SEQ ID NOS 11 and 11-13 respectively, in order of appearance.
  • FIG. 7A shows exemplary 4 Mer cutter and 6 Mer cutter pairs generating sticky ends.
  • FIG. 7B shows exemplary 4 Mer cutter and 6 Mer cutter pairs generating blunt ends.
  • FIG. 8 shows a capsule containing reagents for barcoding of polynucleotide fragments in a microwell (left) and a microwell containing reagents for barcoding of polynucleotide fragments dispensed in a microwell and sealed to prevent evaporation (right).
  • Polynucleotides sequencing includes the sequencing of whole genomes, detection of specific sequences such as single nucleotide polymorphisms (SNPs) and other mutations, detection of nucleic acid (e.g., deoxyribonucleic acid) insertions, and detection of nucleic acid deletions.
  • SNPs single nucleotide polymorphisms
  • nucleic acid e.g., deoxyribonucleic acid
  • Utilization of the methods, compositions, systems, and devices described herein may incorporate, unless otherwise indicated, conventional techniques of organic chemistry, polymer technology, microfluidics, molecular biology and recombinant techniques, cell biology, biochemistry, and immunology.
  • conventional techniques include microwell construction, microfluidic device construction, polymer chemistry, restriction digestion, ligation, cloning, polynucleotide sequencing, and polynucleotide sequence assembly.
  • suitable techniques are described throughout this disclosure. However, equivalent procedures may also be utilized. Descriptions of certain techniques may be found in standard laboratory manuals, such as Genome Analysis: A Laboratory Manual Series ( Vols.
  • barcode generally refers to a label that may be attached to a polynucleotide, or any variant thereof, to convey information about the polynucleotide.
  • a barcode may be a polynucleotide sequence attached to all fragments of a target polynucleotide contained within a particular partition. This barcode may then be sequenced with the fragments of the target polynucleotide. The presence of the same barcode on multiple sequences may provide information about the origin of the sequence.
  • a barcode may indicate that the sequence came from a particular partition and/or a proximal region of a genome. This may be particularly useful when several partitions are pooled before sequencing.
  • base pairs generally refers to an abbreviation for “base pairs”.
  • the enzyme CviQI has a recognition site of GTAC (4 nucleotides on one strand) and is thus referred to as a “4 Mer cutter.”
  • the enzyme StuI has a recognition site of AGGCCT (6 nucleotides on one strand) and is thus referred to as a “6 Mer cutter.”
  • microwell generally refers to a well with a volume of less than 1 mL. Microwells may be made in various volumes, depending on the application. For example, microwells may be made in a size appropriate to accommodate any of the partition volumes described herein.
  • non-overlapping and “overlapping,” as used to refer to polynucleotide fragments generally refer to a collection of polynucleotide fragments without overlapping sequence or with overlapping sequence, respectively.
  • This genome may be fragmented randomly (e.g., by shearing in a pipette) or non-randomly (e.g., by digesting with a rare cutter). Fragmenting randomly produces overlapping sequences (second set of sequences from top in FIG. 1 , “Fragmented randomly to generate overlap”), because each copy of the genome is cut at different positions.
  • this overlap may be used to determine the linear order of the fragments, thereby enabling assembly of the entire genomic sequence.
  • fragmenting by digesting with a rare cutter produces non-overlapping fragments, because each copy of the (same) genome is cut at the same position (third set of sequences from the top in FIG. 1 , “Fragmented non-randomly using RE-1 to generate non-overlapping fragments”).
  • the linear order may be determined by, for example, fragmenting the genome using a different technique. The fourth set of sequences from the top of FIG.
  • 1 demonstrates the use of a second rare-cutter enzyme to generate a second set of non-overlapping fragments (“Fragmented non-randomly using RE-2 to generate non-overlapping fragments”). Because two different enzymes, for example, are used to generate the two sets of non-overlapping fragments, there is overlap between the fragments generated with the first rare-cutter enzyme (RE-1) and the fragments generated with the second rare-cutter enzyme (RE-2). This overlap may then be used to assemble the linear order of the sequences, and therefore the sequence of the entire genome.
  • RE-1 first rare-cutter enzyme
  • RE-2 fragments generated with the second rare-cutter enzyme
  • partition may be a verb or a noun.
  • the term refers to the fractionation of a substance (e.g., a polynucleotide) between vessels that can be used to sequester one fraction from another. Such vessels are referred to using the noun “partition.”
  • Partitioning may be performed, for example, using microfluidics, dilution, dispensing, and the like.
  • a partition may be, for example, a well, a microwell, a droplet, a test tube, a spot, or any other means of sequestering one fraction of a sample from another.
  • polynucleotides are often partitioned into microwells.
  • polynucleotide or “nucleic acid,” as used herein, are used herein to refer to biological molecules comprising a plurality of nucleotides.
  • exemplary polynucleotides include deoxyribonucleic acids, ribonucleic acids, and synthetic analogues thereof, including peptide nucleic acids.
  • rare-cutter enzyme generally refers to an enzyme with a recognition site that occurs only rarely in a genome.
  • the size of restriction fragments generated by cutting a hypothetical random genome with a restriction enzyme may be approximated by 4 N , where N is the number of nucleotides in the recognition site of the enzyme.
  • N is the number of nucleotides in the recognition site of the enzyme.
  • an enzyme with a recognition site consisting of 7 nucleotides would cut a genome once every 4 7 bp, producing fragments of about 16,384 bp.
  • rare-cutter enzymes have recognition sites comprising 6 or more nucleotides.
  • a rare cutter enzyme may have a recognition site comprising or consisting of 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 nucleotides.
  • rare-cutter enzymes include Notl (GCGGCCGC), XmaIII (CGGCCG), SstII (CCGCGG), SalI (GTCGAC), NruI (TCGCGA), NheI (GCTAGC), Nb.BbvCI (CCTCAGC), BbvCI (CCTCAGC), AscI (GGCGCGCC), AsiSI (GCGATCGC), FseI (GGCCGGCC), PacI (TTAATTAA), PmeI (GTTTAAAC), SbfI (CCTGCAGG), SgrAI (CRCCGGYG), SwaI (ATTTAAAT), BspQI (GCTCTTC), SapI (GCTCTTC), SfiI (GGCCNNNNNGGCC (SEQ ID NO: 1)), CspCI (CAANNNNNGTGG (SEQ ID NO: 2)), AbsI (CCTCGAGG), CciNI (GCGGCCGC), FspAI (RTGCGCAY), MauBI (CGCGCGCG), MreI
  • target polynucleotide generally refers to a polynucleotide to be processed. For example, if a user intends to process genomic DNA into fragments that may be sequenced, the genomic DNA would be the target polynucleotide. If a user intends to process fragments of a polynucleotide, then the fragments of the polynucleotide may be the target polynucleotide.
  • a target polynucleotide 101 such as genomic DNA
  • genomic DNA may be fragmented to generate a plurality of non-overlapping first polynucleotide fragments 102 .
  • This fragmentation may be performed, for example, by digesting the target polynucleotide with a rare-cutter enzyme (e.g., rare-cutter enzyme 1), or an artificial restriction DNA cutter (ARCUT; Yamamoto et al., Nucleic Acids Res., 2007, 35(7), e53).
  • a rare-cutter enzyme e.g., rare-cutter enzyme 1
  • ARCUT artificial restriction DNA cutter
  • the first polynucleotide fragments may then be partitioned, such that at least one partition 103 comprises a first polynucleotide fragment with a unique sequence within that partition and, optionally, an additional first polynucleotide fragment with a different sequence 104 .
  • the partitioned first polynucleotide fragments may then be further fragmented to produce a plurality of non-overlapping second polynucleotide fragments 105 . This fragmentation may be performed, for example, by enzymatic digestion, exposure to ultraviolet (UV) light, ultrasonication, and/or mechanical agitation.
  • the second polynucleotide fragments may be of a size that is appropriate for DNA sequencing, i.e., a size that enables a DNA sequencer to obtain accurate sequence data for the entire fragment.
  • the second fragments may be attached to a barcode, which may be attached to all of the second fragments disposed in a particular partition.
  • the barcode may be, for example, a DNA barcode.
  • the barcoded fragments may be pooled into a partition comprising pooled, barcoded sequences 106 . Three barcodes are depicted as [1], [2], and [3] in 106 .
  • the pooled fragments may be sequenced.
  • Certain methods of genome sequence assembly rely on the presence of overlapping fragments in order to generate higher order sequence data (e.g., whole genome sequences) from sequenced fragments.
  • sequence data e.g., whole genome sequences
  • the methods, compositions, systems, and devices provided herein may also be used to provide overlapping fragments.
  • fragments overlapping with the first and second fragments described above may be generated by generating a plurality of non-overlapping third polynucleotide fragments from the target polynucleotide 107 .
  • the third polynucleotide fragments may be generated, for example, by digesting the target polynucleotide 101 with a rare-cutter enzyme (e.g., rare-cutter enzyme 2; or ARCUT) that is different from the rare-cutter enzyme used to generate the first polynucleotide fragments. If rare-cutter enzymes 1 and 2 are chosen to cut the target polynucleotide sequence at different positions, the third polynucleotide fragments and the first polynucleotide fragments will overlap. The third polynucleotide fragments may then be processed as described above for the first polynucleotide fragments.
  • a rare-cutter enzyme e.g., rare-cutter enzyme 2; or ARCUT
  • the third polynucleotide fragments may be partitioned such that at least one partition 108 comprises a third polynucleotide fragment with a unique sequence within that partition and, optionally, an additional third polynucleotide fragment with a different sequence 109 .
  • These partitioned fragments may then be further fragmented to produce a plurality of non-overlapping fourth polynucleotide fragments 110 .
  • the fourth polynucleotides fragments and the second polynucleotide fragments may overlap.
  • the fourth polynucleotide fragments may be generated by, for example, enzymatic digestion, exposure to ultraviolet (UV) light, ultrasonication, and/or mechanical agitation.
  • the fourth fragments may be of a size that is appropriate for DNA sequencing, i.e., a size that enables a DNA sequencer to obtain accurate sequence data for the entire fragment.
  • the fourth fragments may be attached to a barcode, which may be attached to all of the fourth fragments disposed in a particular partition.
  • the barcode may be, for example, a DNA barcode.
  • the barcoded fragments may be pooled, into a partition comprising pooled, barcoded, sequences 111 . Three barcodes are depicted as [4], [5], and [6] in 111 .
  • the pooled fragments may be sequenced.
  • the overlap between the sequences of the second fragments and the fourth fragments may be used to assemble higher order sequences, such whole genome sequences.
  • microwells for example a microfluidic device.
  • These microwells may be connected to each other, or to a source of reagents, by channels.
  • the first and third fragments may be generated outside of the device and then introduced into the device (or separate devices) for further processing. Partitioning of the first and third fragments may accomplished using fluidic techniques. Generation of the second and fourth fragments may then occur within the microwells of the device or devices.
  • These microwells may contain reagents for barcoding of the second and fourth fragments, such as DNA barcodes, ligase, adapter sequences, and the like.
  • Microwells may feed or be directed into a common outlet, so that barcoded fragments may be pooled or otherwise collected into one or more aliquots which may then be sequenced.
  • the entire process could be performed within a single device.
  • a device could be split into two sections.
  • a first section may comprise a partition comprising rare-cutter enzyme 1 (generating first polynucleotide fragments) and a second section may comprise a partition comprising rare-cutter enzyme 2 (generating third polynucleotide fragments).
  • An aliquot of the target polynucleotide sequence may be placed into each of these partitions.
  • the enzyme may be inactivated and the samples may be partitioned, fragmented, barcoded, pooled, and sequenced as described above.
  • this example has been described using rare-cutter enzymes as the means of generating the first and third fragments. However, this is not intended to be limiting, here or anywhere else in this disclosure.
  • One of ordinary skill in the art will readily recognize that other means of generating non-overlapping, or predominantly non-overlapping, fragments would be just as suitable as the use of rare-cutter enzymes.
  • This disclosure also provides methods, compositions, systems, and devices for fragmenting polynucleotides in a pseudo-random manner. This may be performed by treating partitioned polynucleotides with more than one restriction enzyme. For example, polynucleotides partitioned into microwells may be treated with combinations of restriction enzymes. Within each partition containing a particular combination of enzymes, the cutting is defined and predictable. However, across all of the partitions (through the use of multiple combinations of restriction enzymes in different partitions), the polynucleotide fragments generated approximate those obtained from methods of random fragmentation. However, these polynucleotide fragments are generated in a much more controlled manner than random fragments generated by methods known in the art (e.g., shearing).
  • the partitioned, pseudo-randomly fragmented polynucleotides may be barcoded, as described throughout this disclosure, pooled, and sequenced.
  • the pseudo-random fragmentation methods may be used with the non-overlapping fragmentation methods described herein, or with any other method described herein such as the high yield adapter/barcode attachment method.
  • Pseudo-random fragmentation may occur by exposing a polynucleotide to multiple enzymes simultaneously, sequentially, or simultaneously and sequentially.
  • this disclosure provides methods and systems for processing polynucleotides comprising generating pseudo-random fragments of said polynucleotides. These pseudo random fragments are generated by treating a polynucleotide with more than one restriction enzyme.
  • a polynucleotide may be treated with about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, 50, or more restriction enzymes.
  • a polynucleotide may be treated with at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, 50, or more restriction enzymes.
  • a polynucleotide may be treated with at least 2 but fewer than 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes.
  • a polynucleotide may be treated with about 2-4, 4-6, 6-8, 8-10, 10-12, 12-14, 14-16, 16-18, 18-20, 20-25, 25-30, 35-40, 40-45, or 45-50 restriction enzymes.
  • the restriction enzymes may be chosen in order to maximize the number or fraction of fragments that will provide accurate sequencing data, based on the size of the fragments generated by the pseudo-random fragmentation. For present day sequencing technology, accuracy degrades beyond a read length of about 100 nucleotides. Therefore, fragments of about 200 or fewer nucleotides generally provide the most accurate sequence data since they can be sequenced from either end. Fragments below about 50 nucleotides are generally less desirable because, although the produce accurate sequencing data, they underutilize the read length capacity of current sequencing instruments which are capable of 150 to 200 base reads. Fragments of about 200 to about 400 nucleotides may be sequenced with systematic errors introduced as the read length increases beyond the initial 100 bases from each end.
  • Sequence information from fragments greater than about 400 nucleotides is typically completely lost for those bases greater than 200 bases from either end.
  • sequencing technology is constantly advancing and that the ability to obtain accurate sequence information from longer fragments is also constantly improving.
  • the pseudo-random fragmentation methods presented herein may be used to produce optimal fragment lengths for any sequencing method.
  • fragments may be defined by the distance of their component nucleotides from a restriction site (measured in nucleotides).
  • each nucleotide within a polynucleotide fragment generated by the pseudo-random fragmentation method may be less than about 10, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, 400, 550, 600, 1000, 5000, 10000, or 100000 nucleotides from the restriction site of an enzyme to which the polynucleotide is exposed.
  • Each nucleotide within a polynucleotide fragment may be about 10, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, 400, 550, 600, 1000, 5000, 10000, or 100000 nucleotides from the restriction site of an enzyme to which the polynucleotide is exposed.
  • Each nucleotide within a polynucleotide fragment may be at least about 10, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, 400, 550, 600, 1000, 5000, 10000, or 100000 nucleotides from the restriction site of an enzyme to which the polynucleotide is exposed.
  • At least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%, of the nucleotides comprising a target polynucleotide sequence are within about 10, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, 400, 550, 600, 1000, 5000, 10000, or 100000 nucleotides from the restriction site of an enzyme to which the polynucleotide is exposed. All combinations of these percentages and polynucleotide lengths are contemplated.
  • nucleotides comprising a target polynucleotide sequence are within about 1, 5, 10, 50, 200, 250, 300, 350, 400, 550, 600, 1000, 5000, 10000, or 100000 nucleotides from the restriction site of an enzyme to which the polynucleotide is exposed. All combinations of these percentages and polynucleotide lengths are contemplated.
  • the pseudo-random fragmentation methods may be used to obtain fragments of about 10 to 50 nucleotides, 46 to 210 nucleotides, 50 to 250 nucleotides, 250 to 400 nucleotides, 400 to 550 nucleotides, 550 to 700 nucleotides, 700 to 1000 nucleotides, 1000 to 1300 nucleotides, 1300 to 1600 nucleotides, 1600 to 1900 nucleotides, 1900 to 2200 nucleotides, or 2200 to 3000 nucleotides.
  • the pseudo-random fragmentation methods may be used to obtain fragments with a mean or median of about 40 nucleotides, 60 nucleotides, 80 nucleotides, 100 nucleotides, 120 nucleotides, 130 nucleotides, 140 nucleotides, 160 nucleotides, 180 nucleotides, 200 nucleotides, 250 nucleotides, 300 nucleotides, 400 nucleotides, 500 nucleotides, 600 nucleotides, 700 nucleotides, 800 nucleotides, 900 nucleotides, 1000 nucleotides, 1200 nucleotides, 1400 nucleotides, 1600 nucleotides, 1800 nucleotides, 2000 nucleotides, 2500 nucleotides, 3000 nucleotides, or more.
  • the pseudo-random fragmentation methods may be used to obtain fragments with a mean or median of at least about 40 nucleotides, 60 nucleotides, 80 nucleotides, 100 nucleotides, 120 nucleotides, 130 nucleotides, 140 nucleotides, 160 nucleotides, 180 nucleotides, 200 nucleotides, 250 nucleotides, 300 nucleotides, 400 nucleotides, 500 nucleotides, 600 nucleotides, 700 nucleotides, 800 nucleotides, 900 nucleotides, 1000 nucleotides, 1200 nucleotides, 1400 nucleotides, 1600 nucleotides, 1800 nucleotides, 2000 nucleotides, 2500 nucleotides, 3000 nucleotides, or more.
  • the pseudo-random fragmentation methods may be used to obtain fragments with a mean or median of less than about 40 nucleotides, 60 nucleotides, 80 nucleotides, 100 nucleotides, 120 nucleotides, 130 nucleotides, 140 nucleotides, 160 nucleotides, 180 nucleotides, 200 nucleotides, 250 nucleotides, 300 nucleotides, 400 nucleotides, 500 nucleotides, 600 nucleotides, 700 nucleotides, 800 nucleotides, 900 nucleotides, 1000 nucleotides, 1200 nucleotides, 1400 nucleotides, 1600 nucleotides, 1800 nucleotides, 2000 nucleotides, 2500 nucleotides, or 3000 nucleotides.
  • the pseudo-random fragmentation methods provided herein are used to generate fragments wherein a particular percentage (or fraction) of the fragments generated fall within any of the size ranges described herein. For example, about 0%, 2%, 4%, 6%, 8%, 10%, 12%, 14%, 16%, 18%, 20%, 22%, 24%, 26%, 28%, 30%, 32%, 34%, 36%, 38%, 40%, 42%, 44%, 46%, 48%, 50%, 52%, 54%, 56%, 58%, 60%, 62%, 64%, 66%, 68%, 70%, 72%, 74%, 76%, 78%, 80%, 82%, 84%, 86%, 88%, 90%, 92%, 94%, 96%, 98%, or 100% of the fragments generated may fall within any of the size ranges described herein.
  • multiple 4 Mer cutters may be used to provide a distribution of about 18% of fragments of about 50 nucleotides or less, about 38% of fragments of about 200 nucleotides or less, about 25% of fragments between about 200 and about 400 nucleotides, and about 37% of fragments greater than about 400 nucleotides (e.g., see FIG. 4 ).
  • the pseudo-random fragmentation method may be designed to minimize the percentage of fragments greater than a certain number of nucleotides in length, in order to minimize the loss of sequence information.
  • the method may be designed to yield less than about 0.1%, 0.5%, 1%, 2%, 5%, 10%, 20%, or 50% fragments greater than 100 nucleotides.
  • the method may be designed to yield less than about 0.1%, 0.5%, 1%, 2%, 5%, 10%, 20%, or 50% fragments greater than 150 nucleotides.
  • the method may be designed to yield less than about 0.1%, 0.5%, 1%, 2%, 5%, 10%, 20%, or 50% fragments greater than 200 nucleotides.
  • the method may be designed to yield less than about 0.1%, 0.5%, 1%, 2%, 5%, 10%, 20%, or 50% fragments greater than 300 nucleotides, and so on.
  • the pseudo-random fragmentation methods of the invention may be used to generate sequences suitable for any chosen read length.
  • Enzymes for use with the pseudo-random fragmentation method described herein may be chosen, for example, based on the length of their recognition site and their compatibility with certain buffer conditions (to allow for combination with other enzymes). Enzymes may also be chosen so that their cutting activity is methylation insensitive, or sensitive to methylation. For example, restriction enzymes with shorter recognition sites generally cut polynucleotides more frequently. Thus, cutting a target polynucleotide with a 6 Mer cutter will generally produce more large fragments than cutting the same polynucleotide with a 4 Mer cutter (e.g., compare FIGS. 3 and 4 ). Cutting a target polynucleotide with a plurality of enzymes (e.g.
  • This disclosure also provides methods of selecting a plurality of enzymes for pseudo-random fragmentation of a polynucleotide sequence.
  • a target polynucleotide may be exposed separately to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 restriction enzymes.
  • the size distribution of the target polynucleotide fragments is then determined, for example, by electrophoresis.
  • the combination of enzymes providing the greatest number of fragments that are capable of being sequenced can then be chosen.
  • the method can also be carried out in silico.
  • the enzymes may be disposed within the same partition, or within a plurality of partitions.
  • any of the plurality of enzyme number described herein may be disposed within a single partition, or across partitions.
  • a polynucleotide may be treated with about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, 50, or more restriction enzymes in the same partition, or across partitions.
  • a polynucleotide may be treated with at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, 50, or more restriction enzymes in the same partition, or across partitions.
  • a polynucleotide may be treated with at least 2 but fewer than 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes in the same partition, or across partitions.
  • a polynucleotide may be treated with about 2-4, 4-6, 6-8, 8-10, 10-12, 12-14, 14-16, 16-18, 18-20, 20-25, 25-30, 35-40, 40-45, or 45-50 restriction enzymes in the same partition, or across partitions.
  • each restriction enzyme may be distributed across an equivalent number of partitions, so that the number of partitions occupied by each restriction enzyme is equivalent. For example, if 10 restriction enzymes are used in a device containing 1,000 partitions, each enzyme may be present in 100 partitions. In other cases, each restriction enzyme may be distributed across a non-equivalent number of partitions, so that the number of partitions occupied by each restriction enzyme is not equivalent.
  • restriction enzymes 1-8 may be present in 100 partitions each, enzyme 9 may be present in 50 partitions, and enzyme 10 may be present in 150 partitions.
  • Placement of restriction enzymes in an unequal number of partitions may be beneficial, for example, when an enzyme generates a desired product at a low yield. Placing this low-yield enzyme in more partitions will therefore expose more of the target polynucleotide to the enzyme, increasing the amount of the desired product (e.g., fragment of a certain size or composition) that can be formed from the enzyme.
  • Such an approach may be useful for accessing portions of a target polynucleotide (e.g., a genome) that are not cut by enzymes producing polynucleotide fragments at a higher yield.
  • a target polynucleotide e.g., a genome
  • the restriction site and efficiency of an enzyme, composition of the target polynucleotide, and efficiency and side-products generated by the enzyme may all be among the factors considered when determining how many partitions should receive a particular enzyme.
  • different numbers of restriction enzymes may be used in a single partition and across all partitions. For example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes or more may be used in each partition, while 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes or more may be used across all partitions. All combinations of these numbers are included within the invention.
  • Non-limiting specific examples include the use of 1 restriction enzyme per partition and 2, 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; 2 restriction enzymes per partition and 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; 3 restriction enzymes per partition and 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; 4 restriction enzymes per partition and 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; 5 restriction enzymes per partition and 6, 7, 8, 9, or 10 restriction enzymes across all partitions; 6 restriction enzymes per partition and 7, 8, 9, or 10 restriction enzymes across all partitions; 7 restriction enzymes per partition and 8, 9, or 10 restriction enzymes across all partitions; 8 restriction enzymes per partition and 9 or 10 restriction enzymes across all partitions; and 9 restriction enzymes per partition and 10 or more restriction enzymes across all partitions.
  • At least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes or more may be used in each partition, while at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes or more may be used across all partitions. All combinations of these numbers are included within the invention.
  • Non-limiting specific examples include the use of at least 1 restriction enzyme per partition and at least 2, 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 2 restriction enzymes per partition and at least 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 3 restriction enzymes per partition and at least 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 4 restriction enzymes per partition and at least 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 5 restriction enzymes per partition and at least 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 6 restriction enzymes per partition and at least 7, 8, 9, or 10 restriction enzymes across all partitions; at least 7 restriction enzymes per partition and at least 8, 9, or 10 restriction enzymes across all partitions; at least 8 restriction enzymes per partition and at least 9 or 10 restriction enzymes across all partitions; and at least 9 restriction enzymes per partition and at least 10 or more restriction enzymes across all partitions.
  • restriction enzymes or more may be used in each partition, while at most 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes or more may be used across all partitions. All combinations of these numbers are included within the invention.
  • Non-limiting specific examples include the use of at most 1 restriction enzyme per partition and at most 2, 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 2 restriction enzymes per partition and at most 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 3 restriction enzymes per partition and at most 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 4 restriction enzymes per partition and at most 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 5 restriction enzymes per partition and at most 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 6 restriction enzymes per partition and at most 7, 8, 9, or 10 restriction enzymes across all partitions; at most 7 restriction enzymes per partition and at most 8, 9, or 10 restriction enzymes across all partitions; at most 8 restriction enzymes per partition and at most 9 or 10 restriction enzymes across all partitions; and at most 9 restriction enzymes per partition and at most 10 or more restriction enzymes across all partitions.
  • At least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes or more may be used in each partition, while at most 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes or more may be used across all partitions. All combinations of these numbers are included within the invention.
  • Non-limiting specific examples include the use of at least 1 restriction enzyme per partition and at most 2, 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 2 restriction enzymes per partition and at most 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 3 restriction enzymes per partition and at most 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 4 restriction enzymes per partition and at most 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 5 restriction enzymes per partition and at most 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 6 restriction enzymes per partition and at most 7, 8, 9, or 10 restriction enzymes across all partitions; at least 7 restriction enzymes per partition and at most 8, 9, or 10 restriction enzymes across all partitions; at least 8 restriction enzymes per partition and at most 9 or 10 restriction enzymes across all partitions; and at least 9 restriction enzymes per partition and at most 10 or more restriction enzymes across all partitions.
  • Non-limiting specific examples include the use of at most 1 restriction enzyme per partition and at least 2, 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 2 restriction enzymes per partition and at least 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 3 restriction enzymes per partition and at least 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 4 restriction enzymes per partition and at least 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 5 restriction enzymes per partition and at least 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 6 restriction enzymes per partition and at least 7, 8, 9, or 10 restriction enzymes across all partitions; at most 7 restriction enzymes per partition and at least 8, 9, or 10 restriction enzymes across all partitions; at most 8 restriction enzymes per partition and at least 9 or 10 restriction enzymes across all partitions; and at most 9 restriction enzymes per partition and at least 10 or more restriction enzymes across all partitions.
  • Barcodes may be polynucleotide barcodes, which may be ligated to the fragmented target polynucleotides or added via an amplification reaction.
  • fragmentation of target polynucleotides may be performed using one or more restriction enzymes contained within a partition (e.g., a microwell) where the fragmentation is performed.
  • the partition may also contain a polynucleotide barcode and a ligase, which enables the attachment of the barcode to the fragmented polynucleotide.
  • an adapter may be used to make a fragmented target polynucleotide compatible for ligation with a barcode.
  • the presence of adapters, fragmented target polynucleotide, barcodes, restriction enzymes, and ligases in the same partition may lead to the generation of undesirable side products that decrease the yield of a desired end product.
  • self-ligation may occur between adapters, target polynucleotide fragments, and/or barcodes. These self-ligations reduce the amount of starting material and decrease the yield of the desired product, for example, a polynucleotide fragment properly ligated to a barcode and/or and adapter.
  • This disclosure provides methods, compositions, systems, and devices for addressing this problem and increasing the yield of a desired product.
  • the problem is addressed by pairing a first restriction enzyme and a second restriction enzyme.
  • the two restriction enzymes create compatible termini upon cutting, but each enzyme has a different recognition sequence.
  • Ligation of two pieces of DNA generated after cutting with the first restriction enzyme will regenerate the recognition site for the first restriction enzyme, allowing the first restriction enzyme to re-cut the ligated DNA.
  • ligation of two pieces of DNA generated after cutting with the second restriction enzyme will regenerate the recognition site for the second restriction enzyme, allowing the second restriction enzyme to re-cut the ligated DNA.
  • ligation of one piece of DNA generated after cutting with the first restriction enzyme and one piece of DNA generated after cutting with the second restriction enzyme will result in ligated DNA that is unrecognizable (and therefore uncuttable) by both the first and second enzymes.
  • any multimers of fragmented target polynucleotides are re-cut and any multimers of adapter (or other molecules, e.g., barcodes) are also re-cut.
  • adapter or other molecules, e.g., barcodes
  • the restriction sites for both enzymes are not present and the correctly ligated molecule may not be re-cut by either enzyme.
  • FIG. 6 An example of this method is illustrated in FIG. 6 , and additional pairs of enzymes that may be used with the method are provided in FIGS. 7A-7B .
  • Any pair of enzymes may be used, so long as they meet the following criteria: (1) the enzymes should create identical, or at least similar, ligatable termini upon cutting; and (2) the enzymes should have different recognition sequences.
  • the enzymes may be selected to avoid or minimize cutting of certain polynucleotide sequences such as barcodes, adapters, and other polynucleotide components of a sample processing or preparation platform.
  • the enzymes may be selected for methylation insensitivity or methylation sensitivity.
  • the enzymes may also be selected to be active under s single set of environmental conditions, such as buffer conditions, temperature, etc. Minimizing the cutting of barcodes and adapters may be accomplished by pairing certain enzymes with certain barcodes and/or adapters.
  • This method may be used to increase the yield of any of the barcoding methods described herein.
  • the regeneration of the starting materials allows these starting materials another opportunity to form the desired products (i.e., fragmented target polynucleotides ligated to barcodes, optionally with adapters).
  • the methods described above may be used to achieve about 75%, 85%, 95%, 96%, 97%, 98%, 99%, or 99.5% yield (w/w).
  • the methods may be used to achieve at least about 75%, 85%, 95%, 96%, 97%, 98%, 99%, or 99.5% yield (w/w).
  • the methods described above may use, for example, a pair of restriction enzyme selected from the group consisting of MspI-NarI, BfaI-NarI, BfaI-NdeI, HinP1I-ClaI, Msel-NdeI, CviQI-NdeI, Taq ⁇ I-AcII, RsaI-PmeI, AluI-EcoRV, BstUI-PmeI, DpnI-StuI, HaeIII-PmeI, and HpyCH4V-SfoI.
  • This list of enzymes is provided for purposes of illustration only, and is not meant to be limiting.
  • the methods described above may generally use any two enzymes that create ligatable termini upon cutting but that have different recognition sequences.
  • the method is not limited to ligation.
  • multimers formed after amplification of side products formed by association of compatible ends could also be re-cut using the methods described above.
  • More than one pair of enzymes may also be used.
  • the number of pairs of enzymes chosen will vary depending on the number of undesirable side products formed in a reaction. For example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more pairs of enzymes may be used. Treatment of a polynucleotide with the enzymes may be sequential, simultaneous, or both.
  • Target polynucleotides processed according to the methods provided in this disclosure may be DNA, RNA, peptide nucleic acids, and any hybrid thereof, where the polynucleotide contains any combination of deoxyribo- and ribo-nucleotides.
  • Polynucleotides may be single stranded or double stranded, as specified, or contain portions of both double stranded or single stranded sequence.
  • Polynucleotides may contain any combination of nucleotides, including uracil, adenine, thymine, cytosine, guanine, inosine, xanthine, hypoxanthine, isocytosine, isoguanine and any nucleotide derivative thereof.
  • nucleotide may include nucleotides and nucleosides, as well as nucleoside and nucleotide analogs, and modified nucleotides, including both synthetic and naturally occurring species.
  • Target polynucleotides may be cDNA, mitochondrial DNA (mtDNA), messenger RNA (mRNA), ribosomal RNA (rRNA), transfer RNA (tRNA), nuclear RNA (nRNA), small interfering RNA (siRNA), small nuclear RNA (snRNA), small nucleolar RNA (snoRNA), small Cajal body-specific RNA (scaRNA), microRNA (miRNA), double stranded (dsRNA), ribozyme, riboswitch or viral RNA.
  • mtDNA mitochondrial DNA
  • mRNA messenger RNA
  • rRNA ribosomal RNA
  • tRNA transfer RNA
  • nRNA nuclear RNA
  • siRNA small interfering RNA
  • snRNA small nuclear RNA
  • Target polynucleotides may be contained on a plasmid, cosmid, or chromosome, and may be part of a genome.
  • a target polynucleotide may comprise one or more genes and/or one or more pseudogenes.
  • a pseudogene generally refers to a dysfunctional relative of a gene that has lost its protein coding ability and/or is otherwise no longer expressed in the cell.
  • Target polynucleotides may be obtained from a sample using any methods known in the art.
  • a target polynucleotide processed as described herein may be obtained from whole cells, cell preparations and cell-free compositions from any organism, tissue, cell, or environment.
  • target polynucleotides may be obtained from bodily fluids which may include blood, urine, serum, lymph, saliva, mucosal secretions, perspiration, or semen.
  • polynucleotides may be obtained from environmental samples including air, agricultural products, water, and soil.
  • polynucleotides may be the products of experimental manipulation including, recombinant cloning, polynucleotide amplification (as generally described in PCT/US99/01705), polymerase chain reaction (PCR) amplification, purification methods (such as purification of genomic DNA or RNA), and synthesis reactions.
  • Genomic DNA may be obtained from naturally occurring or genetically modified organisms or from artificially or synthetically created genomes.
  • Target polynucleotides comprising genomic DNA may be obtained from any source and using any methods known in the art.
  • genomic DNA may be isolated with or without amplification.
  • Amplification may include PCR amplification, multiple displacement amplification (MDA), rolling circle amplification and other amplification methods.
  • Genomic DNA may also be obtained by cloning or recombinant methods, such as those involving plasmids and artificial chromosomes or other conventional methods (see Sambrook and Russell, Molecular Cloning: A Laboratory Manual., cited supra.) Polynucleotides may be isolated using other methods known in the art, for example as disclosed in Genome Analysis: A Laboratory Manual Series (Vols. I-IV) or Molecular Cloning: A Laboratory Manual. If the isolated polynucleotide is an mRNA, it may be reverse transcribed into cDNA using conventional techniques, as described in Sambrook and Russell, Molecular Cloning: A Laboratory Manual., cited supra.
  • Target polynucleotides may also be isolated from “target organisms” or “target cells”.
  • target organism and “target cell” refer to an organism or cell, respectively, from which target polynucleotides may be obtained.
  • Target cells may be obtained from a variety of organisms including human, mammal, non-human mammal, ape, monkey, chimpanzee, plant, reptilian, amphibian, avian, fungal, viral or bacterial organisms.
  • Target cells may also be obtained from a variety of clinical sources such as biopsies, aspirates, blood, urine, formalin fixed embedded tissues, and the like.
  • Target cells may comprise a specific cell type, such as a somatic cell, germline cell, wild-type cell, cancer or tumor cells, or diseased or infected cell.
  • a target cell may refer to a cell derived from a particular tissue or a particular locus in a target organism
  • a target cell may comprise whole intact cells, or cell preparations.
  • Target polynucleotides may also be obtained or provided in specified quantities. Amplification may be used to increase the quantity of a target polynucleotide. Target polynucleotides may quantified by mass. For example, target polynucleotides may be provided in a mass ranging from about 1-10, 10-50, 50-100, 100-200, 200-1000, 1000-10000 ng.
  • Target polynucleotides may be provided in a mass of at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, or 10000 ng.
  • Target polynucleotides may be provided in a mass of less than about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, or 10000 ng.
  • Target polynucleotides may also be quantified as “genome equivalents.”
  • a genome equivalent is an amount of polynucleotide equivalent to one haploid genome of an organism from which the target polynucleotide is derived. For example, a single diploid cell contains two genome equivalents of DNA.
  • Target polynucleotides may be provided in an amount ranging from about 1-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, or 100000-1000000 genome equivalents.
  • Target polynucleotides may be provided in an amount of at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, 20000, 30000, 40000, 50000, 60000 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, or 1000000 genome equivalents.
  • Target polynucleotides may be provided in an amount less than about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, 20000, 30000, 40000, 50000, 60000 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, or 1000000 genome equivalents.
  • Target polynucleotide may also be quantified by the amount of sequence coverage provided.
  • the amount of sequence coverage refers to the average number of reads representing a given nucleotide in a reconstructed sequence. Generally, the greater the number of times a region is sequenced, the more accurate the sequence information obtained.
  • Target polynucleotides may be provided in an amount that provides a range of sequence coverage from about 0.1X-10X, 10-X-50X, 50X-100X, 100X-200X, or 200X-500X.
  • Target polynucleotide may be provided in an amount that provides at least about 0.1X, 0.2X, 0.3X, 0.4X, 0.5X, 0.6X, 0.7X, 0.8X, 0.9X, 1.0X, 5X, 10X, 25X, 50X, 100X, 125X, 150X, 175X, or 200X sequence coverage.
  • Target polynucleotide may be provided in an amount that provides less than about 0.2X, 0.3X, 0.4X, 0.5X, 0.6X, 0.7X, 0.8X, 0.9X, 1.0X, 5X, 10X, 25X, 50X, 100X, 125X, 150X, 175X, or 200X sequence coverage.
  • Fragmentation of polynucleotides is used as a step in a variety of processing methods described herein.
  • the size of the polynucleotide fragments may vary depending on the source of the target polynucleotide, the method used for fragmentation, and the desired application.
  • certain methods of the invention are illustrated using a certain number of fragmentation steps, the number of fragmentation steps provided is not meant to be limiting, and any number of fragmentation steps may be used. For example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more fragmentation steps may be used.
  • Fragments generated using the methods described herein may be about 1-10, 10-20, 20-50, 50-100, 50-200, 100-200, 200-300, 300-400, 400-500, 500-1000, 1000-5000, 5000-10000, 10000-100000, 100000-250000, or 250000-500000 nucleotides in length. Fragments generated using the methods described herein may be at least about 10, 20, 100, 200, 300, 400, 500, 1000, 5000, 10000, 100000, 250000, 500000, or more nucleotides in length. Fragments generated using the methods described herein may be less than about 10, 20, 100, 200, 300, 400, 500, 1000, 5000, 10000, 100000, 250000, 500000, nucleotides in length.
  • Fragments generated using the methods described herein may have a mean or median length of about 1-10, 10-20, 20-50, 50-100, 50-200, 100-200, 200-300, 300-400, 400-500, 500-1000, 1000-5000, 5000-10000, 10000-100000, 100000-250000, or 250000-500000 nucleotides. Fragments generated using the methods described herein may have a mean or median length of at least about 10, 20, 100, 200, 300, 400, 500, 1000, 5000, 10000, 100000, 250000, 500000, or more nucleotides. Fragments generated using the methods described herein may have a mean or median length of less than about 10, 20, 100, 200, 300, 400, 500, 1000, 5000, 10000, 100000, 250000, 500000, nucleotides.
  • fragmentation may be performed through physical, mechanical or enzymatic methods.
  • Physical fragmentation may include exposing a target polynucleotide to heat or to UV light.
  • Mechanical disruption may be used to mechanically shear a target polynucleotide into fragments of the desired range.
  • Mechanical shearing may be accomplished through a number of methods known in the art, including repetitive pipetting of the target polynucleotide, sonication and nebulization.
  • Target polynucleotides may also be fragmented using enzymatic methods. In some cases, enzymatic digestion may be performed using enzymes such as using restriction enzymes.
  • this polynucleotide may be a target polynucleotide, such as a genome.
  • this polynucleotide may be a fragment of a target polynucleotide which one wishes to further fragment.
  • still further fragments may be still further fragmented.
  • Any suitable polynucleotide may be fragmented according the methods described herein.
  • a fragment of a polynucleotide generally comprises a portion of the sequence of the targeted polynucleotide from which the fragment was generated.
  • a fragment may comprise a copy of a gene and/or pseudogene, including one included in the original target polynucleotide.
  • a plurality of fragments generated from fragmenting a target polynucleotide may comprise fragments that each comprise a copy of a gene and/or pseudogene.
  • Restriction enzymes may be used to perform specific or non-specific fragmentation of target polynucleotides.
  • the methods of the present disclosure may use one or more types of restriction enzymes, generally described as Type I enzymes, Type II enzymes, and/or Type III enzymes.
  • Type II and Type III enzymes are generally commercially available and well known in the art.
  • Type II and Type III enzymes recognize specific sequences of nucleotide base pairs within a double stranded polynucleotide sequence (a “recognition sequence” or “recognition site”). Upon binding and recognition of these sequences, Type II and Type III enzymes cleave the polynucleotide sequence.
  • cleavage will result in a polynucleotide fragment with a portion of overhanging single stranded DNA, called a “sticky end.” In other cases, cleavage will not result in a fragment with an overhang, creating a “blunt end.”
  • the methods of the present disclosure may comprise use of restriction enzymes that generate either sticky ends or blunt ends.
  • Restriction enzymes may recognize a variety of recognition sites in the target polynucleotide. Some restriction enzymes (“exact cutters”) recognize only a single recognition site (e.g., GAATTC). Other restriction enzymes are more promiscuous, and recognize more than one recognition site, or a variety of recognition sites. Some enzymes cut at a single position within the recognition site, while others may cut at multiple positions. Some enzymes cut at the same position within the recognition site, while others cut at variable positions.
  • the present disclosure provides method of selecting one or more restriction enzymes to produce fragments of a desired length.
  • Polynucleotide fragmentation may be simulated in silico, and the fragmentation may be optimized to obtain the greatest number or fraction of polynucleotide fragments within a particular size range, while minimizing the number or fraction of fragments within undesirable size ranges. Optimization algorithms may be applied to select a combination of two or more enzymes to produce the desired fragment sizes with the desired distribution of fragments quantities.
  • a polynucleotide may be exposed to two or more restriction enzymes simultaneously or sequentially. This may be accomplished by, for example, adding more than one restriction enzyme to a partition, or by adding one restriction enzyme to a partition, performing the digestion, deactivating the restriction enzyme (e.g., by heat treatment) and then adding a second restriction enzyme. Any suitable restriction enzyme may be used alone, or in combination, in the methods presented herein.
  • Fragmenting of a target polynucleotide may occur prior to partitioning of the target polynucleotide or fragments generated from fragmenting.
  • genomic DNA gDNA
  • a target polynucleotide may be entered into a partition along with reagents necessary for fragmentation (e.g., including a restriction enzyme), such that fragmentation of the target polynucleotide occurs within the partition.
  • reagents necessary for fragmentation e.g., including a restriction enzyme
  • gDNA may be fragmented in a partition comprising a restriction enzyme, and the restriction enzyme is used to fragment the gDNA.
  • a plurality of fragments may be generated prior to partitioning, using any method for fragmentation described herein.
  • Some or all of the fragments of the plurality may each comprise a copy of a gene and/or a pseudogene.
  • the fragments can be separated and partitioned such that each copy of the gene or pseudogene is located in a different partition.
  • Each partition for example, can comprise a different barcode sequence such that each copy of the gene and/or pseudogene can be associated with a different barcode sequence, using barcoding methods described elsewhere herein.
  • each gene and/or pseudogene can be counted and/or differentiated during sequencing of the barcoded fragments. Any sequencing method may be used, including those described herein.
  • genomic DNA can be fragmented to generate a plurality of non-overlapping fragments of the gDNA. At least some of the fragments of the plurality may each comprise a copy of a gene and/or a pseudogene. The fragments may be separated and partitioned such that each copy of the gene or pseudogene is located in a different partition. Each partition, for example, can comprise a different barcode sequence such that each copy of the gene and/or pseudogene may be barcoded with a different barcode sequence. Via the different barcode sequences, the genes and/or pseudogenes may be counted and or differentiated after sequencing of the barcoded fragments. Any sequencing method may be used, including those described herein.
  • certain methods, systems, and compositions of the disclosure may utilize partitioning of polynucleotides into separate partitions (e.g., microwells, droplets of an emulsion). These partitions may be used to contain polynucleotides for further processing, such as, for example, cutting, ligating, and/or barcoding.
  • partitions e.g., microwells, droplets of an emulsion.
  • partitions are formed from droplets, emulsions, or spots on a substrate.
  • Weizmann et al. (Nature Methods, 2006, Vol. 3 No. 7 pages 545-550). Suitable methods for forming emulsions, which can be used as partitions or to generate microcapsules, include the methods described in Weitz et al. (U.S. Pub. No. 2012/0211084). Partitions may also be formed through the use of wells, microwells, multi-well plates, and microwell arrays.
  • Partitioning may be performed using piezoelectric droplet generation (e.g., Bransky et al., Lab on a Chip, 2009, 9, 516-520). Partitioning may be performed using surface acoustic waves (e.g., Demirci and Montesano, Lab on a Chip, 2007, 7, 1139-1145).
  • piezoelectric droplet generation e.g., Bransky et al., Lab on a Chip, 2009, 9, 516-520.
  • Partitioning may be performed using surface acoustic waves (e.g., Demirci and Montesano, Lab on a Chip, 2007, 7, 1139-1145).
  • Each partition may also contain, or be contained within any other suitable partition.
  • a well, microwell, hole, a surface of a bead, or a tube may comprise a droplet (e.g., a droplet in an emulsion), a continuous phase in an emulsion, a spot, a capsule, or any other suitable partition.
  • a droplet may comprise a capsule, bead, or another droplet.
  • a capsule may comprise a droplet, bead, or another capsule.
  • a well or microwell comprises a plurality of droplets and a plurality of capsules.
  • a capsule comprises a plurality of capsules and a plurality of droplets. All combinations of partitions are envisioned. Table 1 shows non-limiting examples of partitions that may be combined with each other.
  • a partition may comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, or 50000 partitions.
  • a partition may comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, or 50000 partitions. In some cases, a partition may comprise less than 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, or 50000 partitions. In some cases, each partition may comprise 2-50, 2-20, 2-10, or 2-5 partitions.
  • the number of partitions employed may vary depending on the application.
  • the number of partitions may be about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, or 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000, or more.
  • the number of partitions may be at least about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000, or more.
  • the number of partitions may be less than about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000.
  • the number of partitions may be about 5-10000000, 5-5000000, 5-1,000,000, 10-10,000, 10-5,000, 10-1,000, 1,000-6,000, 1,000-5,000, 1,000-4,000, 1,000-3,000, or 1,000-2,000.
  • Such partitions may be pre-loaded with reagents to perform a particular reaction.
  • a capsule containing one or more reagents may be placed within a microwell. After adding a polynucleotide sample to the well, the capsule may be made to release its contents.
  • the contents of the capsule may include, for example, restriction enzymes, ligases, barcodes, and adapters for processing the polynucleotide sample placed in the microwell.
  • such partitions may be droplets of an emulsion.
  • a droplet of an emulsion may be an aqueous droplet in an oil phase.
  • the droplet may comprise, for example, one or more reagents (e.g., restriction enzymes, ligases, polymerases, reagents necessary for nucleic acid amplification (e.g., primers, DNA polymerases, dNTPs, buffers)), a polynucleotide sample, and a barcode sequence.
  • the barcode sequence, polynucleotide sample, or any reagent may be associated with a solid surface within a droplet. In some cases, the solid surface is a bead.
  • the bead is a gel bead (see e.g., Agresti et al., U.S. Patent Publication No. 2010/0136544).
  • the droplet is hardened into a gel bead (e.g., via polymerization).
  • a species may be contained within a droplet in an emulsion containing, for example, a first phase (e.g., oil or water) forming the droplet and a second (continuous) phase (e.g., water or oil).
  • a first phase e.g., oil or water
  • a second phase e.g., water or oil
  • An emulsion may be a single emulsion, for example, a water-in-oil or an oil-in-water emulsion.
  • An emulsion may be a double emulsion, for example a water-in-oil-in-water or an oil-in-water-in-oil emulsion. Higher-order emulsions are also possible.
  • the emulsion may be held in any suitable container, including any suitable partition described in this disclosure.
  • droplets in an emulsion comprise other partitions.
  • a droplet in an emulsion may comprise any suitable partition including, for example, another droplet (e.g., a droplet in an emulsion), a capsule, a bead, and the like.
  • Each partition may be present as a single partition or a plurality of partitions, and each partition may comprise the same species or different species.
  • a droplet in an emulsion comprises a capsule comprising reagents for sample processing.
  • a capsule may contain one or more capsules, or other partitions.
  • a sample comprising an analyte to be processed is contained within the droplet.
  • a stimulus is applied to cause release of the contents of the capsule into the droplet, resulting in contact between the reagents and the analyte to be processed.
  • the droplet is incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered. While this example describes an embodiment where a reagent is in a capsule and an analyte is in the droplet, the opposite configuration—i.e., reagent in the droplet and analyte in the capsule—is also possible.
  • the droplets in an emulsion may be of uniform size or heterogeneous size.
  • the diameter of a droplet in an emulsion may be about 0.001 ⁇ m, 0.01 ⁇ m, 0.05 ⁇ m, 0.1 ⁇ m, 0.5 ⁇ m, 1 ⁇ m, 5 ⁇ m, 10 ⁇ m, 50 ⁇ m, 100 ⁇ m, 150 ⁇ m, 200 ⁇ m, 300 ⁇ m, 400 ⁇ m, 500 ⁇ m, 600 ⁇ m, 700 ⁇ m, 800 ⁇ m, 900 ⁇ m, or 1 mm.
  • a droplet may have a diameter of at least about 0.001 ⁇ m, 0.01 ⁇ m, 0.05 ⁇ m, 0.1 ⁇ m, 0.5 ⁇ m, 1 ⁇ m, 5 ⁇ m, 10 ⁇ m, 50 ⁇ m, 100 ⁇ m, 150 ⁇ m, 200 ⁇ m, 300 ⁇ m, 400 ⁇ m, 500 ⁇ m, 600 ⁇ m, 700 ⁇ m, 800 ⁇ m, 900 ⁇ m, or 1 mm.
  • a droplet may have a diameter of less than about 0.001 ⁇ m, 0.01 ⁇ m, 0.05 ⁇ m, 0.1 ⁇ m, 0.5 ⁇ m, 1 ⁇ m, 5 ⁇ m, 10 ⁇ m, 50 ⁇ m, 100 ⁇ m, 150 ⁇ m, 200 ⁇ m, 300 ⁇ m, 400 ⁇ m, 500 ⁇ m, 600 ⁇ m, 700 ⁇ m, 800 ⁇ m, 900 ⁇ m, or 1 mm.
  • a droplet may have a diameter of about 0.001 ⁇ m to 1 mm, 0.01 ⁇ m to 900 ⁇ m, 0.1 ⁇ m to 600 ⁇ m, 100 ⁇ m to 200 ⁇ m, 100 ⁇ m to 300 ⁇ m, 100 ⁇ m to 400 ⁇ m, 100 ⁇ m to 500 ⁇ m, 100 ⁇ m to 600 ⁇ m, 150 ⁇ m to 200 ⁇ m, 150 ⁇ m to 300 ⁇ m, or 150 ⁇ m to 400 ⁇ m.
  • Droplets in an emulsion also may have a particular density.
  • the droplets are less dense than an aqueous fluid (e.g., water); in some cases, the droplets are denser than an aqueous fluid.
  • the droplets are less dense than a non-aqueous fluid (e.g., oil); in some cases, the droplets are denser than a non-aqueous fluid.
  • Droplets may have a density of about 0.05 g/cm 3 , 0.1 g/cm 3 , 0.2 g/cm 3 , 0.3 g/cm 3 , 0.4 g/ cm 3 , 0.5 g/cm 3 , 0.6 g/cm 3 , 0.7 g/cm 3 , 0.8 g/cm 3 , 0.81 g/cm 3 , 0.82 g/cm 3 , 0.83 g/cm 3 , 0.84 g/cm 3 , 0.85 g/cm 3 , 0.86 g/cm 3 , 0.87 g/cm 3 , 0.88 g/cm 3 , 0.89 g/cm 3 , 0.90 g/cm 3 , 0.91 g/cm 3 , 0.92 g/cm 3 , 0.93 g/cm 3 , 0.94 g/cm 3 , 0.95 g/cm 3 , 0.96 g/c
  • Droplets may have a density of at least about 0.05 g/cm 3 , 0.1 g/cm 3 , 0.2 g/cm 3 , 0.3 g/cm 3 , 0.4 g/ cm 3 , 0.5 g/cm 3 , 0.6 g/cm 3 , 0.7 g/cm 3 , 0.8 g/cm 3 , 0.81 g/cm 3 , 0.82 g/cm 3 , 0.83 g/cm 3 , 0.84 g/cm 3 , 0.85 g/cm 3 , 0.86 g/cm 3 , 0.87 g/cm 3 , 0.88 g/cm 3 , 0.89 g/cm 3 , 0.90 g/cm 3 , 0.91 g/cm 3 , 0.92 g/cm 3 , 0.93 g/cm 3 , 0.94 g/cm 3 , 0.95 g/cm 3 , 0.96 g
  • droplet densities may be at most about 0.7 g/cm 3 , 0.8 g/cm 3 , 0.81 g/cm 3 , 0.82 g/cm 3 , 0.83 g/cm 3 , 0.84 g/cm 3 , 0.85 g/cm 3 , 0.86 g/cm 3 , 0.87 g/cm 3 , 0.88 g/cm 3 , 0.89 g/cm 3 , 0.90 g/cm 3 , 0.91 g/cm 3 , 0.92 g/cm 3 , 0.93 g/cm 3 , 0.94 g/cm 3 , 0.95 g/cm 3 , 0.96 g/cm 3 , 0.97 g/cm 3 , 0.98 g/cm 3 , 0.99 g/cm 3 , 1.00 g/cm 3 , 1.05 g/cm 3 , 1.1 g/cm 3 ,
  • Polynucleotides may be partitioned using a variety of methods. For example, polynucleotides may be diluted and dispensed across a plurality of partitions. A terminal dilution of a medium comprising polynucleotides may be performed such that the number of partitions or wells exceeds the number of polynucleotides.
  • the ratio of the number of polynucleotides to the number of partitions may range from about 0.1-10, 0.5-10, 1-10, 2-10, 10-100, 100-1000, or more.
  • the ratio of the number of polynucleotides to the number of partitions may be about 0.1, 0.5, 1, 2, 4, 8, 10, 20, 50, 100, or 1000.
  • the ratio of the number of polynucleotides to the number of partitions may be at least about 0.1, 0.5, 1, 2, 4, 8, 10, 20, 50, 100, or 1000.
  • the ratio of the number of polynucleotides to the number of partitions may be less than about 0.1, 0.5, 1, 2, 4, 8, 10, 20, 50, 100, or 1000.
  • the number of partitions employed may vary depending on the application.
  • the number of partitions may be about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, or 10,000, or more.
  • the number of partitions may be at least about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, or 10,000, or more.
  • the number of partitions may be less than about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, or 10,000.
  • the volume of the partitions may vary depending on the application.
  • the volume of the partitions may be about 1000 ⁇ l 900 ⁇ l, 800 ⁇ l, 700 ⁇ l, 600 ⁇ l, 500 ⁇ l, 400 ⁇ l, 300 ⁇ l, 200 ⁇ l, 100 ⁇ l, 50 ⁇ l, 25 ⁇ l, 10 ⁇ l, 5 ⁇ l, 1 ⁇ l, 900 nL, 800 nL, 700 nL, 600 nL, 500 nL, 400 nL, 300 nL, 200 nL, 100 nL, 50 nL, 25 nL, 10 nL, or 5 nL.
  • the volume of the partitions may be at least about 1000 ⁇ l, 900 ⁇ l, 800 ⁇ l, 700 ⁇ l, 600 ⁇ l, 500 ⁇ l, 400 ⁇ l, 300 ⁇ l, 200 ⁇ l, 100 ⁇ l, 50 ⁇ l, 25 ⁇ l, 10 ⁇ l, 5 ⁇ l, 1 ⁇ l, 900 nL, 800 nL, 700 nL, 600 nL, 500 nL, 400 nL, 300 nL, 200 nL, 100 nL, 50 nL, 25 nL, 10 nL, or 5 nL.
  • the volume of the partitions may be less than about 1000 ⁇ l, 900 ⁇ l, 800 ⁇ l, 700 ⁇ l, 600 ⁇ l, 500 ⁇ l, 400 ⁇ l, 300 ⁇ l, 200 ⁇ l, 100 ⁇ l, 50 ⁇ l, 25 ⁇ l, 10 ⁇ l, 5 ⁇ l, 1 ⁇ l, 900 nL, 800 nL, 700 nL, 600 nL, 500 nL, 400 nL, 300 nL, 200 nL, 100 nL, 50 nL, 25 nL, 10 nL, or 5 nL.
  • Species may also be partitioned at a particular density. For example, species may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 100000, or 1000000 species per partition. Species may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 100000, 1000000 or more species per partition. Species may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 100000, or 1000000 species per partition. Species may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, or 100000-1000000 species per partition.
  • Species may be partitioned such that at least one partition comprises a species that is unique within that partition. This may be true for about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more of the partitions. This may be true for at least about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more of the partitions. This may be true for less than about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the partitions.
  • Particular polynucleotides may also be targeted to specific partitions.
  • a capture reagent such as an oligonucleotide probe may be immobilized in a partition to capture specific polynucleotides through hybridization.
  • Polynucleotides may also be partitioned at a particular density. For example, polynucleotides may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, or 100000-1000000 polynucleotides per partition. Polynucleotides may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 100000, 1000000 or more polynucleotides per partition. Polynucleotides may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 100000, or 1000000 polynucleotides per partition. Polynucleotides may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 100000, or 1000000 polynucleotides per partition.
  • Polynucleotides may be partitioned such that at least one partition comprises a polynucleotide sequence with a unique sequence compared to all other polynucleotide sequences contained within the same partition. This may be true for about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more of the partitions. This may be true for less than about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more of the partitions. This may be true for more than about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more of the partitions.
  • Barcodes may be of a variety of different formats, including polynucleotide barcodes. Depending upon the specific application, barcodes may be attached to polynucleotide fragments in a reversible or irreversible manner. Barcodes may also allow for identification and/or quantification of individual polynucleotide fragments during sequencing.
  • Barcodes may be loaded into partitions so that one or more barcodes are introduced into a particular partition.
  • Each partition may contain a different set of barcodes. This may be accomplished by directly dispensing the barcodes into the partitions, enveloping the barcodes (e.g., in a droplet of an emulsion), or by placing the barcodes within a container that is placed in a partition (e.g., a microcapsule).
  • the number of partitions employed may vary depending on the application.
  • the number of partitions may be about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, or 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000, or more.
  • the number of partitions may be at least about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000, or more.
  • the number of partitions may be less than about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000.
  • the number of partitions may be about 5-10000000, 5-5000000, 5-1,000,000, 10-10,000, 10-5,000, 10-1,000, 1,000-6,000, 1,000-5,000, 1,000-4,000, 1,000-3,000, or 1,000-2,000.
  • the number of different barcodes or different sets of barcodes that are partitioned may vary depending upon, for example, the particular barcodes to be partitioned and/or the application. Different sets of barcodes may be, for example, sets of identical barcodes where the identical barcodes differ between each set. Or different sets of barcodes may be, for example, sets of different barcodes, where each set differs in its included barcodes.
  • less than about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 different barcodes or different sets of barcodes may be partitioned. In some examples, about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, 100000-1000000, 10000-1000000, 10000-10000000, or 10000-100000000 barcodes may be partitioned.
  • Barcodes may be partitioned at a particular density. For example, barcodes may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 barcodes per partition.
  • Barcodes may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, 100000000, or more barcodes per partition.
  • Barcodes may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 barcodes per partition.
  • Barcodes may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, 100000-1000000, 10000-1000000, 10000-10000000, or 10000-100000000 barcodes per partition.
  • Barcodes may be partitioned such that identical barcodes are partitioned at a particular density.
  • identical barcodes may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 identical barcodes per partition.
  • Barcodes may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, 100000000, or more identical barcodes per partition.
  • Barcodes may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 identical barcodes per partition.
  • Barcodes may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, 100000-1000000, 10000-1000000, 10000-10000000, or 10000-100000000 identical barcodes per partition.
  • Barcodes may be partitioned such that different barcodes are partitioned at a particular density. For example, different barcodes may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 different barcodes per partition.
  • Barcodes may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, 100000000, or more different barcodes per partition.
  • Barcodes may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 different barcodes per partition.
  • Barcodes may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, 100000-1000000, 10000-1000000, 10000-10000000, or 10000-100000000 different barcodes per partition.
  • the number of partitions employed to partition barcodes may vary, for example, depending on the application and/or the number of different barcodes to be partitioned.
  • the number of partitions employed to partition barcodes may be about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, or 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000 or more.
  • the number of partitions employed to partition barcodes may be at least about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1000000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000 or more.
  • the number of partitions employed to partition barcodes may be less than about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1000000, 2000000, 3000000, 4000000, 5000000, 10000000, or 20000000.
  • the number of partitions employed to partition barcodes may be about 5-10000000, 5-5000000, 5-1,000,000, 10-10,000, 10-5,000, 10-1,000, 1,000-6,000, 1,000-5,000, 1,000-4,000, 1,000-3,000, or 1,000-2,000.As described above, different barcodes or different sets of barcodes (e.g., each set comprising a plurality of identical barcodes or different barcodes) may be partitioned such that each partition comprises a different barcode or different barcode set. In some cases, each partition may comprise a different set of identical barcodes. Where different sets of identical barcodes are partitioned, the number of identical barcodes per partition may vary.
  • each partition comprises a different set of identical barcodes.
  • the number of identical barcodes per set of barcodes may be about 1,000,000 identical barcodes.
  • the number of different sets of barcodes may be equal to or substantially equal to the number of partitions.
  • any suitable number of different barcodes or different barcode sets may be combined to generate a diverse library of partitioned barcodes with high numbers of barcodes per partition.
  • any of the above-described different numbers of barcodes may be provided with any of the above-described barcode densities per partition, and in any of the above-described numbers of partitions.
  • a population of microcapsules may be prepared such that a first microcapsule in the population comprises multiple copies of identical barcodes (e.g., polynucleotide bar codes, etc.) and a second microcapsule in the population comprises multiple copies of a barcode that differs from the barcode within the first microcapsule.
  • the population of microcapsules may comprise multiple microcapsules (e.g., greater than about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 100, 500, 1000, 5000, 10000, 100000, 1000000, 10000000, 100000000, or 1000000000 microcapsules), each containing multiple copies of a barcode that differs from that contained in the other microcapsules.
  • the population may comprise greater than about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 100, 500, 1000, 5000, 10000, 100000, 1000000, 10000000, 100000000, or 1000000000 microcapsules with identical sets of barcodes. In some cases, the population may comprise greater than about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 100, 500, 1000, 5000, 10000, 100000, 1000000, 10000000, 100000000, or 1000000000 microcapsules, wherein the microcapsules each comprise a different combination of barcodes.
  • a first microcapsule may comprise, e.g., barcodes A, B, and C
  • a second microcapsule may comprise barcodes A, B, and D
  • the different combinations do not overlap, such that a first microcapsule may comprise, e.g., barcodes A, B, and C, while a second microcapsule may comprise barcodes D, E, and F.
  • the use of microcapsules is, of course, optional. All of the combinations described above, and throughout this disclosure, may also be generated by dispending barcodes (and other reagents) directly into partitions (e.g., microwells).
  • the barcodes may be loaded into the partitions at an expected or predicted ratio of barcodes per species to be barcoded (e.g., polynucleotide fragment, strand of polynucleotide, cell, etc.). In some cases, the barcodes are loaded into partitions such that more than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, 100, 500, 1000, 5000, 10000, or 200000 barcodes are loaded per species. In some cases, the barcodes are loaded in the partitions so that less than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, 100, 500, 1000, 5000, 10000, or 200000 barcodes are loaded per species.
  • an expected or predicted ratio of barcodes per species to be barcoded e.g., polynucleotide fragment, strand of polynucleotide, cell, etc.
  • the barcodes are loaded into partitions such that more than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, 100, 500, 1000, 5000,
  • the average number of barcodes loaded per species is less than, or greater than, about 0.0001, 0.001, 0.01, 0.1, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, 100, 500, 1000, 5000, 10000, or 200000 barcodes per species.
  • barcodes may be copies of the same barcode, or multiple different barcodes.
  • the attachment process may be designed to attach multiple identical barcodes to a single polynucleotide fragment, or multiple different barcodes to the polynucleotide fragment.
  • the methods provided herein may comprise loading a partition (e.g., a microwell, droplet of an emulsion) with the reagents necessary for the attachment of barcodes to polynucleotide fragments.
  • a partition e.g., a microwell, droplet of an emulsion
  • reagents including restriction enzymes, ligase enzymes, buffers, adapters, barcodes and the like may be loaded into a partition.
  • reagents including primers, DNA polymerases, DNTPs, buffers, barcodes and the like may be loaded into a partition.
  • these reagents may be loaded directly into the partition, or via a container such as a microcapsule. If the reagents are not disposed within a container, they may be loaded into a partition (e.g., a microwell) which may then be sealed with a wax or oil until the reagents are used.
  • Barcodes may be ligated to a polynucleotide fragment using sticky or blunt ends. Barcoded polynucleotide fragments may also be generated by amplifying a polynucleotide fragment with primers comprising barcodes.
  • Microcapsules and microwell capsule array (MCA) devices may be used to perform the polynucleotide processing methods described herein.
  • MCA devices are devices with a plurality of microwells. Microcapsules are introduced into these microwells, before, after, or concurrently with the introduction of a sample.
  • Microwells may comprise free reagents and/or reagents encapsulated in microcapsules. Any of the reagents described in this disclosure may be encapsulated in a microcapsule, including any chemicals, particles, and elements suitable for sample processing reactions involving a polynucleotide.
  • a microcapsule 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, dNTPs, ddNTPs and the like.
  • Additional exemplary reagents include: 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, mtDNA (mitochondrial
  • a microcapsule comprises a set of reagents that have a similar attribute (e.g., a set of enzymes, a set of minerals, a set of oligonucleotides, a mixture of different bar-codes, a mixture of identical bar-codes).
  • a microcapsule comprises a heterogeneous mixture of reagents.
  • the heterogeneous mixture of reagents comprises all components necessary to perform a reaction.
  • such mixture comprises 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 a different microcapsule or within a solution within a partition (e.g., microwell) of the device.
  • microcapsules comprising reagents are introduced.
  • both free reagents and reagents encapsulated in microcapsules are loaded into the device, either sequentially or concurrently.
  • reagents are introduced to the device either before or after a particular step.
  • reagents and/or microcapsules comprising reagents are introduced sequentially such that different reactions or operations occur at different steps.
  • the reagents (or microcapsules) may be also be loaded at steps interspersed with a reaction or operation step.
  • microcapsules comprising reagents for fragmenting polynucleotides (e.g., restriction enzymes) may be loaded into the device, followed by loading of microcapsules comprising reagents for ligating bar-codes and subsequent ligation of the bar-codes to the fragmented molecules.
  • reagents for fragmenting polynucleotides e.g., restriction enzymes
  • Microcapsules may be pre-formed and filled with reagents by injection.
  • the picoinjection methods described in Abate et al. Proc. Natl. Acad. Sci. U.S.A., 2010, 107(45), 19163-19166) and Weitz et al. (U.S. Pub. No. 2012/0132288) may be used to introduce reagents into the interior of microcapsules described herein. These methods can also be used to introduce a plurality of any of the reagents described herein into microcapsules.
  • Microcapsules may be formed by any emulsion technique known in the art.
  • the multiple emulsion technique of Weitz et al. U.S. Pub. No. 2012/0211084
  • the multiple emulsion technique of Weitz et al. U.S. Pub. No. 2012/0211084
  • microcapsules or partitions
  • Numerous chemical triggers may be used to trigger the disruption of partitions (e.g., Plunkett et al., Biomacromolecules, 2005, 6:632-637).
  • Examples of these chemical changes may include, but are not limited to pH-mediated changes to the integrity of a component of a partition, disintegration of a component of a partition via chemical cleavage of crosslink bonds, and triggered depolymerization of a component of a partition.
  • Bulk changes may also be used to trigger disruption of partitions.
  • a change in pH of a solution may trigger disruption of a partition via a number of different mechanisms.
  • the addition of acid may cause degradation or disassembly a portion of a partition through a variety of mechanisms.
  • Addition of protons may disassemble cross-linking of polymers in a component of a partition, disrupt ionic or hydrogen bonds in a component of a partition, or create nanopores in a component of a partition to allow the inner contents to leak through to the exterior.
  • a change in pH may also destabilize an emulsion, leading to release of the contents of the droplets.
  • a partition is produced from materials that comprise acid-degradable chemical cross-linkers, such a ketals.
  • a decrease in pH, particular to a pH lower than 5, may induce the ketal to convert to a ketone and two alcohols and facilitate disruption of the partition.
  • the partitions may be produced from materials comprising one or more polyelectrolytes that are pH sensitive. A decrease in pH may disrupt the ionic- or hydrogen-bonding interactions of such partitions, or create nanopores therein.
  • partitions made from materials comprising polyelectrolytes comprise a charged, gel-based core that expands and contracts upon a change of pH.
  • Disruption of cross-linked materials comprising a partition can be accomplished through a number of mechanisms.
  • a partition can be contacted with various chemicals that induce oxidation, reduction or other chemical changes.
  • a reducing agent such as beta-mercaptoethanol
  • enzymes may be added to cleave peptide bonds in materials forming a partition, thereby resulting in a loss of integrity of the partition.
  • Depolymerization can also be used to disrupt partitions.
  • a chemical trigger may be added to facilitate the removal of a protecting head group.
  • the trigger may cause removal of a head group of a carbonate ester or carbamate within a polymer, which in turn causes depolymerization and release of species from the inside of a partition.
  • a chemical trigger may comprise an osmotic trigger, whereby a change in ion or solute concentration in a solution induces swelling of a material used to make a partition. Swelling may cause a buildup of internal pressure such that a partition ruptures to release its contents. Swelling may also cause an increase in the pore size of the material, allowing species contained within the partition to diffuse out, and vice versa.
  • a partition may also be made to release its contents via bulk or physical changes, such as pressure induced rupture, melting, or changes in porosity.
  • Sequencing may be performed by any available technique. For example, sequencing may be performed by the classic Sanger sequencing method. Sequencing methods may also include: high-throughput sequencing, pyrosequencing, sequencing-by-synthesis, single-molecule sequencing, nanopore sequencing, sequencing-by-ligation, sequencing-by-hybridization, RNA-Seq (Illumina), Digital Gene Expression (Helicon), next generation sequencing, single molecule sequencing by synthesis (SMSS) (Helicos), massively-parallel sequencing, clonal single molecule Array (Solexa), shotgun sequencing, Maxim-Gilbert sequencing, primer walking, and any other sequencing methods known in the art.
  • SMSS single molecule sequencing by synthesis
  • Solexa single molecule sequencing by synthesis
  • shotgun sequencing Maxim-Gilbert sequencing
  • primer walking and any other sequencing methods known in the art.
  • varying numbers of fragments are sequenced. For example, in some cases about 30%-90% of the fragments are sequenced. In some cases, about 35%-85%, 40%-80%, 45%-75%, 50%-70%, 55%-65%, or 50%-60% of the fragments are sequenced. In some cases, at least about 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the fragments are sequenced. In some cases less than about 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the fragments are sequenced.
  • sequences from fragments are assembled to provide sequence information for a contiguous region of the original target polynucleotide that is longer than the individual sequence reads.
  • Individual sequence reads may be about 10-50, 50-100, 100-200, 200-300, 300-400, or more nucleotides in length.
  • the identities of the barcode tags may serve to order the sequence reads from individual fragments as well as to differentiate between haplotypes. For example, during the partitioning of individual fragments, parental polynucleotide fragments may separated into different partitions. With an increase in the number of partitions, the likelihood of a fragment from both a maternal and paternal haplotype contained in the same partition becomes negligibly small. Thus, sequence reads from fragments in the same partition may be assembled and ordered.
  • This disclosure also provides methods and compositions to prepare polynucleotide fragments in such a manner that may enable phasing or linkage information to be generated. Such information may allow for the detection of linked genetic variations in sequences, including genetic variations (e.g., SNPs, mutations, indels, copy number variations, transversions, translocations, inversions, etc.) that are separated by long stretches of polynucleotides.
  • the term “indel” refers to a mutation resulting in a colocalized insertion and deletion and a net gain or loss in nucleotides.
  • a “microindel” is an indel that results in a net gain or loss of 1 to 50 nucleotides.
  • These variations may exist in either a cis or trans relationship. In a cis relationship, two or more genetic variations exist in the same polynucleotide or strand. In a trans relationship, two or more genetic variations exist on multiple polynucleotide molecules or strands.
  • Methods provided herein may be used to determine polynucleotide phasing.
  • a polynucleotide sample e.g., a polynucleotide that spans a given locus or loci
  • the polynucleotide may then be fragmented, barcoded, and sequenced.
  • the sequences may be examined for genetic variation.
  • the detection of genetic variations in the same sequence tagged with two different bar codes may indicate that the two genetic variations are derived from two separate strands of DNA, reflecting a trans relationship.
  • the detection of two different genetic variations tagged with the same bar codes may indicate that the two genetic variations are from the same strand of DNA, reflecting a cis relationship.
  • Phase information may be important for the characterization of a polynucleotide fragment, particularly if the polynucleotide fragment is derived from a subject at risk of, having, or suspected of a having a particular disease or disorder (e.g., hereditary recessive disease such as cystic fibrosis, cancer, etc.).
  • the information may be able to distinguish between the following possibilities: (1) two genetic variations within the same gene on the same strand of DNA and (2) two genetic variations within the same gene but located on separate strands of DNA.
  • Possibility (1) may indicate that one copy of the gene is normal and the individual is free of the disease, while possibility (2) may indicate that the individual has or will develop the disease, particularly if the two genetic variations are damaging to the function of the gene when present within the same gene copy.
  • the phasing information may also be able to distinguish between the following possibilities: (1) two genetic variations, each within a different gene on the same strand of DNA and (2) two genetic variations, each within a different gene but located on separate strands of DNA.
  • Methods provided herein may also be used to prepare polynucleotide contained within cells in a manner that enables cell-specific information to be obtained.
  • the methods enable detection of genetic variations (e.g., SNPs, mutations, indels, copy number variations, transversions, translocations, inversions, etc.) from very small samples, such as from samples comprising about 10-100 cells.
  • about 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 cells may be used in the methods described herein.
  • at least about 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 cells may be used in the methods described herein.
  • at most about 5, 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 cells may be used in the methods described herein.
  • a method comprises partitioning a cellular sample (or crude cell extract) such that at most one cell (or extract of one cell) is present per partition, lysing the cells, fragmenting the polynucleotides contained within the cells by any of the methods described herein, attaching the fragmented polynucleotides to barcodes, pooling, and sequencing.
  • the barcodes and other reagents may be contained within a microcapsule. These microcapsules may be loaded into a partition (e.g., a microwell) before, after, or concurrently with the loading of the cell, such that each cell is contacted with a different microcapsule.
  • This technique may be used to attach a unique barcode to polynucleotides obtained from each cell.
  • the resulting tagged polynucleotides may then be pooled and sequenced, and the barcodes may be used to trace the origin of the polynucleotides. For example, polynucleotides with identical barcodes may be determined to originate from the same cell, while polynucleotides with different barcodes may be determined to originate from different cells.
  • the methods described herein may be used to detect the distribution of oncogenic mutations across a population of cancerous tumor cells.
  • some tumor cells may have a mutation, or amplification, of an oncogene (e.g., HER2, BRAF, EGFR, KRAS) in both alleles (homozygous), others may have a mutation in one allele (heterozygous), and still others may have no mutation (wild-type).
  • an oncogene e.g., HER2, BRAF, EGFR, KRAS
  • the methods described herein may be used to detect these differences, and also to quantify the relative numbers of homozygous, heterozygous, and wild-type cells. Such information may be used, for example, to stage a particular cancer and/or to monitor the progression of the cancer and its treatment over time.
  • this disclosure provides methods of identifying mutations in two different oncogenes (e.g., KRAS and EGFR). If the same cell comprises genes with both mutations, this may indicate a more aggressive form of cancer. In contrast, if the mutations are located in two different cells, this may indicate that the cancer is more benign, or less advanced.
  • oncogenes e.g., KRAS and EGFR
  • a sample may comprise a cell, mRNA, or cDNA reverse transcribed from mRNA.
  • the sample may be a pooled sample, comprising extracts from several different cells or tissues, or a sample comprising extracts from a single cell or tissue.
  • Cells may be placed directly into an partition (e.g., a microwell) and lysed. After lysis, the methods of the invention may be used to fragment and barcode the polynucleotides of the cell for sequencing. Polynucleotides may also be extracted from cells prior to introducing them into a partition used in a method of the invention. Reverse transcription of mRNA may be performed in a partition described herein, or outside of such a partition. Sequencing cDNA may provide an indication of the abundance of a particular transcript in a particular cell over time, or after exposure to a particular condition.
  • partition e.g., a microwell
  • the methods of the invention may be used to fragment and barcode the polynucleotides of the cell for sequencing. Polynucleotides may also be extracted from cells prior to introducing them into a partition used in a method of the invention. Reverse transcription of mRNA may be performed in a partition described herein, or outside of such a partition. Sequencing cDNA may
  • the methods presented throughout this disclosure provide several advantages over current polynucleotide processing methods.
  • Third, the controlled fragmentation of the target polynucleotides allows the user to produce polynucleotide fragments with a defined and appropriate length. This aids in partitioning the polynucleotides and also reduces the amount of sequence information loss due to the present of overly-large fragments.
  • the methods and systems also provide a facile workflow that maintains the integrity of the processed polynucleotide.
  • the use of restriction enzymes enables the user to create DNA overhangs (“sticky ends”) that may be designed for compatibility with adapters and/or barcodes.
  • This example demonstrates a method for the generation of non-overlapping DNA fragments suitable for DNA sequencing and other downstream applications. An implementation of this method is schematically illustrated in FIG. 2 .
  • a target polynucleotide 101 genomic DNA is fragmented with the enzyme Notl, to generate a plurality of non-overlapping first polynucleotide fragments 102 .
  • the first polynucleotide fragments are partitioned into separate microwells 103 in a microdevice such that each microwell comprises a plurality of fragments, but only a single fragment with a particular sequence 104 .
  • the left-hand side of FIG. 2 illustrates three microwells (one is labeled 103 ), each containing three exemplary unique fragments 104 , corresponding to the first polynucleotide fragments 102 . Referring again to the left-hand side of FIG. 2 , the left-most well contains fragments A1, B2, and C3, the middle well contains fragments B1, A2, and A3, and the right-most well contains fragments C1, C2, and B3.
  • each member of the second polynucleotide fragments is designated by its first fragment identifier (e.g., A1, B2, etc.), followed by a “ ⁇ 1” or a “ ⁇ 2”.
  • first fragment A1 is fragmented to produce second fragments A1-1 and A1-2.
  • First fragment B2 is fragmented to produce second fragments B2-1 and B2-2, and so on.
  • only two second fragments are shown for each first fragment. This is, of course, not meant to be limiting, as any number of fragments may be generated at any step of the process.
  • the second set polynucleotide fragments are barcoded, and the barcoded sequences are pooled.
  • the labels [1], [2], and [3] represent three different barcode sequences used to label the second fragments 105 .
  • the labeled sequences are designated 106 .
  • adapter sequences (not shown) are used to make the second fragments 105 compatible for ligation with the barcodes.
  • the barcoding is performed while the fragments are still partitioned, before pooling.
  • the pooled barcoded sequences are then sequenced.
  • the methods described above are then repeated, using a second rare cutter enzyme, XmaIII to digest the genomic DNA and generate a plurality of non-overlapping third polynucleotide fragments 107 .
  • the third polynucleotide fragments and the first polynucleotide fragments are overlapping, because they are generated with different rare-cutter enzymes that cut the target polynucleotides at different sites.
  • the third polynucleotide fragments are partitioned into separate microwells 108 in a microdevice such that each microwell comprises a plurality of fragments, but only a single fragment with a particular sequence 109 .
  • FIG. 2 illustrates three microwells (one is labeled 108 ), each containing three exemplary unique fragments 109 , corresponding to the third polynucleotide fragments 107 .
  • the left-most well contains fragments D1, E2, and F3
  • the middle well contains fragments E1, D2, and D3
  • the right-most well contains fragments F1, F2, and E3.
  • the partitioned fragments are then further fragmented, to generate a plurality of non-overlapping fourth polynucleotide fragments 110 .
  • the fourth polynucleotide fragments and the second polynucleotide fragments are overlapping, because they are generated by fragmenting the third and first fragments, respectively, which were generated with rare-cutter enzymes that cut the target polynucleotide at different sites, as described above.
  • each member of the fourth set of polynucleotide fragments is designated by its third fragment identifier (e.g., D1, E2, etc.), followed by a “ ⁇ 1” or a “ ⁇ 2”.
  • third fragment D1 is fragmented to produce fourth fragments D1-1 and D1-2.
  • Third fragment E2 is fragmented to produce fourth fragments E2-1 and E2-2, and so on.
  • only two fourth fragments are shown for each third fragment. This is, of course, not meant to be limiting, as any number of fragments may be generated.
  • the fourth polynucleotides fragments are barcoded, and the barcoded sequences are pooled.
  • the numbers [4], [5], and [6] represent three different barcode sequences used to label the fourth fragments 110 .
  • the labeled sequences are designated 111 .
  • adapter sequences (not shown) are used to make the fourth fragments 110 compatible for ligation with the barcodes.
  • the barcoding is performed while the fragments are still partitioned, before pooling.
  • the pooled barcoded sequences are then sequenced.
  • the example above describes sequencing the barcoded second fragments separately from the barcoded fourth fragments.
  • the barcoded second fragments and the barcoded fourth fragments may also be combined, and the combined sample may be sequenced.
  • One or more steps of the process may be carried out in a device.
  • the steps carried out in a device may be carried out in the same device or in different devices.
  • sequence contigs are assembled and the overlapping sequences between the second fragments and the fourth fragments are used to assemble the sequence of the genome.
  • FIG. 3 shows the size distribution of a random 1 Mbp DNA sequence cut with the 6 Mer cutter StuI (AGG/CCT). Fragments less than about 50 nucleotides were designated as “low yield,” because they underutilize the read length capacity of sequencing instruments. Fragments less than about 200 nucleotides were designated as fragments likely to provide the most accurate data from today's sequencing technology. As described throughout this disclosure, this size range is in no way meant to be limiting, and the methods exemplified here, and described throughout this disclosure, may be used to generate fragments of any size range. Fragments from about 200 to about 400 nucleotides typically produce sequence data with systematic error for bases more than 100 bases from either fragment end.
  • Fragments of more than about 400 nucleotides typically do not produce any useful sequence information for bases further than 200 bases from a fragment end, using today's sequencing technologies. However, this is expected to change, and the methods presented herein can be used to generate sequences of this size or larger.
  • FIG. 4 shows the results from a second simulation using the 4 Mer cutter CviQI (G/TAC), instead of StuI (the 6 Mer cutter described above) to simulate cutting a random 1 Mbp DNA sequence.
  • G/TAC 4 Mer cutter CviQI
  • StuI the 6 Mer cutter described above
  • the 4 Mer cutters were: (A) CviQI (G/TAC); (B) BfaI (C/TAG); (C) HinP1I (G/CGC); (D) CviAII (C/ATG); (E) Taq ⁇ I (T/CGA); (F) MseI (T/TAA); and (G) MspI (C/CGG).
  • the results of these simulations are shown in FIG. 5 .
  • increasing the number of 4 Mer cutter enzymes, from one to seven increases the number of fragments with nucleotides within 100 nucleotides of a restriction site. Therefore, cutting the randomly generated 1 Mbp DNA sequence with more than one 4 Mer cutter results in more fragments that may be fully sequenced than cutting with a single 4 Mer cutter.
  • the number of enzymes used to cut a sequence can be chosen so that a particular fraction of a target nucleotide (e.g., a genomic) sequence within 100 nucleotides of a restriction enzyme is achieved.
  • a target nucleotide e.g., a genomic
  • the fraction of a random genome within 100 nucleotides of a restriction site for a 4 Mer cutter is equal to 1-0.44 x , where x is the number of independent 4 Mer cutters.
  • the fraction of a random genome within 100 nucleotides of a restriction site for a 5 Mer cutter is equal to 1-0.25 x , where x is the number of independent 5 Mer cutters.
  • the fraction of a random genome within 100 nucleotides of a restriction site is equal to 1-0.95 x , where x is the number of independent 6 Mer cutters.
  • Table 1 shows the percentage of sequences with a length greater than 100 nucleotides for each of the seven enzymatic treatments described above. These sequences are considered those likely to result in missing data. Increasing the number of enzymes decreases the percentage of sequences greater than 100 nucleotides. The number of enzymes and their restriction site recognition length may be chosen in order to minimize the loss of sequence information from sequences greater than 100 nucleotides from a restriction site while also minimizing the generation of sequences less than 50 nucleotides, which are undesirable because the underutilize the read length capacity of sequencing instruments.
  • fragments may be minimized or avoided by selecting restriction enzymes that cut more rarely but at the potential price of reduced sequencing coverage of the DNA (i.e., more fragments may have bases >100 bases from a restriction site). These fragments may also be physically removed by a size selection step. Since these fragments are small and some fraction of the bases represented in the small fragments may be covered in larger fragments from other enzymes, the effect on coverage would likely be minimal.
  • the exemplary 4 Mer cutter methods presented herein are optimized to provide fragments compatible with current DNA sequencing technology, which may achieve accurate read lengths up to about 100 nucleotides from the terminus of a fragment.
  • restriction enzymes e.g., 5 Mer cutters, 6 Mer cutters, etc.
  • the methods presented in this disclosure are, of course generalizable, and may be used to obtain DNA fragments of any size distribution compatible with present or future sequencing technology.
  • polynucleotide barcodes may be used to provide compatible ends for the attachment of a barcode to a polynucleotide fragment (e.g., by ligation or PCR).
  • the desired products may be, for example:
  • [B] represents a barcode
  • [A] represents an adapter
  • [TPF] represents a target polynucleotide fragment.
  • undesirable side products may form, for example, from the self ligation of barcodes, adapters, and/or target polynucleotide fragments. This example demonstrates one solution to this potential problem.
  • FIG. 6 shows a schematic of an implementation of the method described in this example.
  • three polynucleotide starting materials (Genomic DNA; Adapter 1; and Adapter 2) and three enzymes (MspI; NarI; and DNA Ligase) are contained within a partition.
  • the restriction enzyme MspI (C/CGG) recognizes the CCGG sequence occurring within the Genomic DNA sequence and cuts the Genomic DNA sequence to generate a fragment of genomic DNA. If the reaction proceeds as intended, the fragment of genomic DNA is then ligated to Adapter 1 and Adapter 2, to generate a fragment of genomic DNA flanked by ligated adapters ( FIG. 6 , lower-left). This fragment with ligated adapters may then be ligated to DNA barcodes, which may also be present within the same partition (not shown).
  • FIG. 6 illustrates this concept by showing only self-ligation of fragmented genomic DNA and adapters.
  • One unwanted side product is a multimer of genomic DNA fragments. This may occur, for example, if genomic DNA fragments with compatible ends are ligated to each other after cutting.
  • cutting of Genomic DNA with MspI generates compatible ends that may be ligated by the ligase present in the partition.
  • Adapter 1 and Adapter 2 as shown, have compatible ligatable ends, and may also be ligated to form multimers.
  • one solution to this problem is to pair one enzyme (in this example, MspI) with a second enzyme (in this example, NarI).
  • MspI re-cuts genomic DNA multimers produced by self-ligation of genomic DNA fragments. Therefore, MspI recycles unwanted genomic DNA fragment multimers back into genomic DNA fragments, which may then be correctly ligated to the adapters.
  • NarI cuts multimers of Adapter 1 and Adapter 2 into monomers of Adapter 1 and monomers of Adapter 2, which may then be correctly ligated to genomic DNA fragments. This recycles unwanted adapter multimers back into the desired starting materials of Adapter 1 and Adapter 2.
  • the enzymes are chosen such that the desired product (i.e., the genomic DNA fragment with adapters on each end) does not contain a recognition site for either enzyme. Therefore, the product will not be re-cut by any enzyme contained within the partition. This process increases the yield of the desired product, while minimizing the number of unwanted side products and reducing the amount of starting material required to produce a desired amount of a product.
  • a pair of enzymes may be chosen so that one enzyme recognizes one undesirable side-product and regenerates a starting material and another recognizes another undesirable side product and regenerates another starting material, but neither enzyme recognizes the desired product. This can be done for an unlimited number of side products.
  • one strategy for selecting such pairs is to choose two enzymes that create identical (or similar, ligatable) termini after cutting, but have recognition sequences of different lengths.
  • FIG. 7 shows examples of such pairs of enzymes.
  • the enzymes provided in FIG. 7A provide sticky ends, while those provided in FIG. 7B provide blunt ends.
  • the exemplary embodiment shown in FIG. 6 uses Genomic DNA and two adapters (Adapter 1 and Adapter 2) as starting materials. Therefore, in this embodiment, MspI is used not only to regenerate genomic DNA fragments after self-ligation, but also to generate the genomic DNA fragments in the first place, from Genomic DNA. Of course, this is optional, as one may introduce pre-fragmented genomic DNA into the partition and the method is still applicable.
  • Adapter molecules may also be provided as a single polynucleotide sequence which is then cut by an enzyme contained within the partition (in this example, NarI) to generate ligation compatible ends for attachment to the fragmented genomic DNA.
  • NarI an enzyme contained within the partition
  • the method is also applicable to other polynucleotides described throughout this disclosure and to methods of attachment based on techniques other than ligation (e.g., attachment of an adapter or a barcode by PCR).
  • Pseudo-complimentary nucleotides that preferentially bind natural nucleotides over themselves may also be used to minimize or avoid the formation of certain multimers, for example adapter-adapter multimers and barcode-barcode multimers. If adapters and/or barcodes (and/or other polynucleotides are synthesized using pseudo-complimentary nucleotides, they will prefer to hybridize with naturally occurring polynucleotide fragments (e.g., genomic DNA fragments) rather than themselves, therefore leading to a higher yield of the desired product.
  • synthetic nucleotide fragments e.g., genomic DNA fragments
  • the polynucleotide processing methods described herein may involve the treatment of partitioned polynucleotides with a variety of reagents.
  • reagents may include, for example, restriction enzymes, ligases, phosphatases, kinases, barcodes, adapters, or any other reagent useful in polynucleotide processing or in a downstream application, such as sequencing.
  • FIG. 8 shows two exemplary methods of providing reagents. On the left-hand side of FIG. 8 , reagents are provided within a microcapsule. The microcapsule that is shown in FIG.
  • FIG. 8 has an outer shell (“3”), an intermediate non-aqueous layer (“2”) and an inner aqueous drop contained within the intermediate non-aqueous layer (“1ABC+RE”).
  • This droplet is made by a water-oil-water emulsion technique followed by polymerization of the outermost water layer (“3”) to form a shell.
  • Reagents are contained within the inner aqueous phase of the capsule.
  • the left-hand side of FIG. 8 shows an exemplary embodiment with four reagents contained within the aqueous phase of the capsule, namely three barcode reagents (1A, 1B, and 1C), and a restriction enzyme (“RE”).
  • the embodiment shown is merely exemplary.
  • the reagents may be located in any part of the capsule.
  • the capsule is dispensed into a partition (e.g., a microwell).
  • a target polynucleotide and a ligase are then added to the partition.
  • the capsule is made to release its contents by exposure to a stimulus, such as a change in temperature, a solvent, or stirring.
  • the restriction enzyme fragments the target polynucleotide and the ligase attaches the barcode reagents to the target polynucleotide fragments generated by the restriction enzyme.
  • restriction digestion and ligation may proceed according to any of the methods described herein, for example by non-overlapping fragmentation techniques, by pseudo-random fragmentation methods, and/or by pairing of restriction enzymes to recycle unwanted side products into new starting products (e.g., target polynucleotide fragments and barcodes).
  • Adapters may also be included within the microcapsule.
  • the barcodes shown in FIG. 8 are modular. For example, barcode components 1A, 1B, and 1C may ligate to form barcode: [1A]-[1B]-[1C].
  • FIG. 8 shows the same reagents dispensed into a microwell, followed by sealing with sealant (e.g., a wax or oil), to prevent evaporation before use.
  • sealant e.g., a wax or oil
  • This approach may be substituted for the approach described above, where the reagents are placed within microcapsules. Both approaches are used to produce partitions (e.g., microwells) pre-loaded with reagents for DNA fragmentation and barcoding.
  • partitions e.g., microwells
  • a user unseals a partition, and introduces a target polynucleotide and a ligase (or any other reagents applicable for the method the user is conducting).
  • the restriction enzyme fragments the target polynucleotide and the ligase attaches the barcode reagents to the target polynucleotide fragments generated by the restriction enzyme.
  • both approaches may be combined by placing certain reagents in the microwell and others in the microcapsule.

Abstract

The present disclosure provides compositions, methods, systems, and devices for polynucleotide processing. Such polynucleotide processing may be useful for a variety of applications, including polynucleotide sequencing.

Description

    CROSS-REFERENCE
  • This application is a continuation of U.S. application Ser. No. 15/850,241, filed Dec. 21, 2017, which is a continuation of U.S. patent application Ser. No. 15/588,519, filed May 5, 2017, now U.S. Pat. No. 9,856,530, which is a continuation of U.S. patent application Ser. No. 15/376,582, filed Dec. 12, 2016, now U.S. Pat. No. 9,701,998, which is a continuation-in-part of U.S. patent application Ser. No. 14/104,650, filed on Dec. 12, 2013, now U.S. Pat. No. 9,567,631, which claims priority to U.S. Provisional Application No. 61/737,374, filed on Dec. 14, 2012; U.S. patent application Ser. No. 15/376,582 is also a continuation-in-part of U.S. patent application Ser. No. 14/250,701, filed on Apr. 11, 2014, which is a continuation of U.S. patent application Ser. No. 14/175,973, filed on Feb. 7, 2014, now U.S. Pat. No. 9,388,465, which claims priority to U.S. Provisional Application No. 61/844,804, filed on Jul. 10, 2013, U.S. Provisional Application No. 61/840,403, filed on Jun. 27, 2013, U.S. Provisional Application No. 61/800,223, filed on Mar. 15, 2013, and U.S. Provisional Application No. 61/762,435, filed on Feb. 8, 2013, each of which is entirely incorporated herein by reference for all purposes.
  • CROSS-REFERENCE
  • The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Jun. 5, 2018, is named 43487703311SL.txt and is 5 kilobytes in size.
  • BACKGROUND
  • The processing of polynucleotides and polynucleotide fragments is a critical aspect of a wide variety of technologies, including polynucleotide sequencing. Polynucleotide sequencing continues to find more widespread use in medical applications such as genetic screening and genotyping of tumors. Many polynucleotide sequencing methods rely on sample processing techniques solely utilizing random fragmentation of polynucleotides. Such random, uncontrolled fragmentation can introduce several problems in downstream processing. For example, these methods may produce fragments with large variation in length, including a large number or fraction of sequences that are too long to be sequenced accurately. This results in a loss of sequence information. Current methods of processing may also damage polynucleotides, resulting in incorrect sequence information, and/or the loss of sequence information. These, and other, problems may be significantly amplified by relatively minor operator variability. Thus, there is a significant need for improved methods that provide better control over all aspects of polynucleotide fragmentation and processing. In particular, there is need for polynucleotide processing methods that consistently provide fragments of appropriate size and composition for any downstream application, including sequencing.
  • SUMMARY I. Non-Overlapping Fragmentation
  • This disclosure provides methods, compositions, systems, and devices for processing polynucleotides. In one example, a method provided herein comprises: (a) providing a target polynucleotide; (b) fragmenting said target polynucleotide to generate a plurality of non-overlapping first polynucleotide fragments; (c) partitioning said first polynucleotide fragments to generate partitioned first polynucleotide fragments, wherein at least one partition of said partitioned first polynucleotide fragments comprises a first polynucleotide fragment with a unique sequence within said at least one partition; and (d) fragmenting said partitioned first polynucleotide fragments, to generate a plurality of non-overlapping second polynucleotide fragments.
  • In some of the methods provided in this disclosure, a third and fourth set of polynucleotide fragments are generated by performing the method described above and additionally performing a method comprising: (a) fragmenting said target polynucleotide to generate a plurality of non-overlapping third polynucleotide fragments; (b) partitioning said third polynucleotide fragments to generate partitioned third polynucleotide fragments, wherein at least one partition of said partitioned third polynucleotide fragments comprises a third polynucleotide fragment with a unique sequence within said at least one partition; and (c) fragmenting said partitioned third polynucleotide fragments to generate a plurality of non-overlapping fourth polynucleotide fragments.
  • The third polynucleotide fragments may overlap with the first polynucleotide fragments. The fourth polynucleotide fragments may overlap with the second polynucleotide fragments.
  • The target polynucleotide may be, for example, DNA, RNA, cDNA, or any other polynucleotide.
  • In some cases, at least one of the first, second, third, and fourth polynucleotide fragments are generated by an enzyme. The enzyme may be a restriction enzyme. The restriction enzyme used to generate the first polynucleotide fragments may be different from the restriction enzyme used to generate the third polynucleotide fragments. The restriction enzyme used to generate the second polynucleotide fragments may be different from the restriction enzyme used to generate the fourth polynucleotide fragments. The restriction enzymes may have a recognition site of at least about six nucleotides in length.
  • The fragments can be of a variety of lengths. For example, the first and/or third polynucleotide fragments may have a median length of least about 10,000 nucleotides. The second or fourth polynucleotide fragments may have a median length of less than about 200 nucleotides.
  • The fragments can be attached to barcodes. For example, the second polynucleotide fragments and/or the fourth polynucleotide fragments may be attached to barcodes, to generate barcoded second and/or fourth polynucleotide fragments. The barcodes may be polynucleotide barcodes. The attachment of the barcodes to the polynucleotide fragments may be performed using an enzyme. The enzyme may be a ligase. The barcoded fragments may be pooled. Unpooled or pooled barcoded fragments may be sequenced.
  • In some cases, one or more steps of the methods described in this disclosure may be performed within a device. The device may comprise at least one well. The well may be a microwell. Any of the partitioning steps described in this disclosure may be performed by dispensing into a microwell.
  • The microwell (or well) may comprise reagents. These reagents may be any reagent, including, for example, barcodes, enzymes, adapters, and combinations thereof. The reagents may be physically separated from a polynucleotide sample placed in the microwell. This physical separation may be accomplished by containing the reagents within a microcapsule that is placed within a microwell. The physical separation may also be accomplished by dispensing the reagents in the microwell and overlaying the reagents with a layer that is, for example, dissolvable, meltable, or permeable prior to introducing the polynucleotide sample into the microwell. This layer may be, for example, an oil, wax, membrane, or the like. The microwell may be sealed at any point, for example after addition of the microcapsule, after addition of the reagents, or after addition of either of these components plus a polynucleotide sample.
  • Partitioning may also be performed by a variety of other means, including through the use of fluid flow in microfluidic channels, by emulsification, using spotted arrays, by surface acoustic waves, and by piezoelectric droplet generation.
  • Additional methods of fragmenting nucleic acids that are compatible with the methods provided herein include mechanical disruption, sonication, chemical fragmentation, treatment with UV light, and heating, and combinations thereof. These methods may be used to fragment, for example, the partitioned first or third polynucleotide fragments described above.
  • Partitioning may be done at any time. For example, the first polynucleotide fragments and/or the third polynucleotide fragments may each be further partitioned into two or more partitions before further processing.
  • Pseudo-Random Fragmentation
  • This disclosure provides methods for pseudo-random fragmentation of polynucleotides. In some cases, such methods comprise: (a) providing a target polynucleotide; (b) fragmenting said target polynucleotide to generate a plurality of first polynucleotide fragments; (c) partitioning said first polynucleotide fragments to generate partitioned first polynucleotide fragments, such that at least one partition comprises a first polynucleotide fragment with a unique sequence within said at least one partition; and (d) fragmenting said partitioned first polynucleotide fragments with at least one restriction enzyme in at least one partition, to generate a plurality of second polynucleotide fragments, wherein said partitioned first polynucleotide fragment is fragmented with at least two restriction enzymes across all partitions.
  • In some cases, at least two restriction enzymes are disposed within the same partition. In some cases, at least two restriction enzymes are disposed across a plurality of different partitions.
  • The pseudo-random fragmentation methods can be performed in order to yield fragments of a certain size. In some cases, at least about 50% of the nucleotides within a target polynucleotide are within about 100 nucleotides of a restriction site of a restriction enzyme used to perform pseudo-random fragmentation. In some cases, at most about 25% of the nucleotides within a target polynucleotide are within about 50 nucleotides of a restriction site of a restriction enzyme used to perform pseudo-random fragmentation. In some cases, at most about 10% of the nucleotides within a target polynucleotide are more than about 200 nucleotides from a restriction site a restriction enzyme used to perform pseudo-random fragmentation.
  • A polynucleotide may be treated with two or more restriction enzymes concurrently or sequentially.
  • The pseudo-randomly fragmented polynucleotides may be attached to barcodes, to generate barcoded polynucleotide fragments. The barcoded polynucleotides may be pooled and sequenced.
  • The number of partitions holding the partitioned first polynucleotide fragments may be at least about 1,000 partitions. The volume of these partitions may be less than about 500 nanoliters.
  • Each enzyme may occupy an equivalent number of partitions, or each enzyme may occupy a different number of partitions.
  • III. Restriction Enzyme-Mediated Recycling
  • This disclosure provides methods for recycling certain unwanted reaction side products back into starting materials that can be used to generate a desired product. In some cases, these methods comprise: (a) providing a first polynucleotide, a second polynucleotide, a first restriction enzyme, and a second restriction enzyme, wherein said first polynucleotide comprises a target polynucleotide or a fragment thereof; and (b) attaching said first polynucleotide to said second polynucleotide, to generate a polynucleotide product, wherein said first restriction enzyme cuts a polynucleotide generated by attachment of said first polynucleotide to itself, said second restriction enzyme cuts a polynucleotide generated by attachment of said second polynucleotide to itself, and neither said first restriction enzyme nor said second restriction enzyme cuts said polynucleotide product.
  • The first polynucleotide may be generated in the same reaction volume as the polynucleotide product, or in a different reaction volume. The target polynucleotide may be, for example, a fragment of genomic DNA.
  • The second polynucleotide may be generated in the same reaction volume as the polynucleotide product, or in a different reaction volume. The second polynucleotide may be, for example, a barcode or an adapter.
  • The first restriction enzyme may have a recognition site of at most about four nucleotides in length. The second restriction enzyme may have a recognition site of at least about six nucleotides in length. The first restriction enzyme may have a recognition site of about four nucleotides in length. The second restriction enzyme may have a recognition site of at least about five nucleotides in length.
  • The first and second restriction enzymes may generate ligation compatible ends. These ends may have single-stranded overhangs (i.e., “sticky ends”) or be blunt. The sticky ends may match in sequence and orientation, to allow ligation. The attachment step may be performed by ligation.
  • The sequence 5′ to the ligation compatible end generated by the first restriction enzyme may be different from the sequence 5′ to the ligation compatible end generated by the second restriction enzyme. This will ensure that the desired product cannot be re-cut by either restriction enzyme.
  • The sequence 3′ to the ligation compatible end generated by the first restriction enzyme may be different from the sequence 3′ to the ligation compatible end generated by the second restriction enzyme. This will ensure that the desired product cannot be re-cut by either restriction enzyme. Given the criteria provided throughout this specification, one of ordinary skill in the art will recognize that many pairs of enzymes are suitable for use with this method.
  • The recycling may provide increased yield of the desired product, for example at least about 75% (w/w).
  • Also provided by this disclosure is a polynucleotide fragment generated by any of the methods provided herein, devices for performing the methods provided herein, and systems for performing the methods provided herein.
  • The methods provided in this disclosure (and portions thereof) may also be used with each other. For example, the non-overlapping fragmentation methods may be used alone and/or with the pseudo-random fragmentation methods and/or with the restriction enzyme-mediated recycling methods. Likewise, the pseudo-random fragmentation methods may be used alone and/or with the non-overlapping fragmentation methods and/or with the restriction enzyme-mediated recycling methods. Similarly, the restriction enzyme-mediated recycling methods may be used alone and/or with the non-overlapping fragmentation methods and/or with the pseudo-random fragmentation methods.
  • Additional aspects and advantages of the present disclosure will become readily apparent to those skilled in this art from the following detailed description, wherein only illustrative embodiments of the present disclosure are shown and described. As will be realized, the present disclosure is capable of other and different embodiments, and its several details are capable of modifications in various obvious respects, all without departing from the disclosure. Accordingly, the drawings and description are to be regarded as illustrative in nature, and not as restrictive.
  • INCORPORATION BY REFERENCE
  • All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The novel features of methods, compositions, systems, and devices of this disclosure are set forth with particularity in the appended claims. A better understanding of the features and advantages of this disclosure will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the methods, compositions, systems, and devices of this disclosure are utilized, and the accompanying drawings of which:
  • FIG. 1 is a schematic representation of overlapping and non-overlapping deoxyribonucleic acid (DNA) fragments.
  • FIG. 2 is a schematic representation of methods of generating non-overlapping DNA fragments for DNA sequencing. FIG. 2 discloses SEQ ID NOS 8-10, respectively, in order of appearance.
  • FIG. 3 shows a distribution of DNA fragment size after simulating generation of 1 Mbp random DNA sequences followed by cutting the sequences with a 6 Mer cutter, StuI (AGG/CCT).
  • FIG. 4 shows a distribution of DNA fragment size after simulating generation of 1 Mbp random DNA sequences followed by cutting the sequences with a 4 Mer cutter, CviQI (G/TAC).
  • FIG. 5 shows a distribution of DNA fragment size after simulating the generation of a 1 Mbp random DNA sequence followed by cutting the sequences with seven 4 Mer cutters: (1) CviQI (G/TAC), (2) BfaI (C/TAG), (3) HinP1I (G/CGC), (4) CviAII (C/ATG), (5) TaqαI (T/CGA), (6) Msel (T/TAA), and (7) MspI (C/CGG).
  • FIG. 6 shows the generation of unwanted byproducts (“Side products”) during ligation of adapters to genomic DNA fragments and the recycling of the unwanted byproducts into starting materials (“Genomic DNA”, “Adapter 1”, and “Adapter 2”) by paring of appropriate restriction enzymes (here, MspI and NarI). FIG. 6 discloses SEQ ID NOS 11 and 11-13, respectively, in order of appearance.
  • FIG. 7A shows exemplary 4 Mer cutter and 6 Mer cutter pairs generating sticky ends.
  • FIG. 7B shows exemplary 4 Mer cutter and 6 Mer cutter pairs generating blunt ends.
  • FIG. 8 shows a capsule containing reagents for barcoding of polynucleotide fragments in a microwell (left) and a microwell containing reagents for barcoding of polynucleotide fragments dispensed in a microwell and sealed to prevent evaporation (right).
  • DETAILED DESCRIPTION
  • While various embodiments of the invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions may occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed.
  • This disclosure provides methods, compositions, systems, and devices for processing polynucleotides. Applications include processing polynucleotides for polynucleotide sequencing. Polynucleotides sequencing includes the sequencing of whole genomes, detection of specific sequences such as single nucleotide polymorphisms (SNPs) and other mutations, detection of nucleic acid (e.g., deoxyribonucleic acid) insertions, and detection of nucleic acid deletions.
  • Utilization of the methods, compositions, systems, and devices described herein may incorporate, unless otherwise indicated, conventional techniques of organic chemistry, polymer technology, microfluidics, molecular biology and recombinant techniques, cell biology, biochemistry, and immunology. Such conventional techniques include microwell construction, microfluidic device construction, polymer chemistry, restriction digestion, ligation, cloning, polynucleotide sequencing, and polynucleotide sequence assembly. Specific, non-limiting, illustrations of suitable techniques are described throughout this disclosure. However, equivalent procedures may also be utilized. Descriptions of certain techniques may be found in standard laboratory manuals, such as Genome Analysis: A Laboratory Manual Series (Vols. I-IV), Using Antibodies: A Laboratory Manual, Cells: A Laboratory Manual, PCR Primer: A Laboratory Manual, and Molecular Cloning: A Laboratory Manual (all from Cold Spring Harbor Laboratory Press), and “Oligonucleotide Synthesis: A Practical Approach” 1984, IRL Press London, all of which are herein incorporated in their entirety by reference for all purposes.
  • I. Definitions
  • The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.
  • As used herein, the singular forms “a,” “an,” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise. Furthermore, to the extent that the terms “including”, “includes”, “having”, “has”, “with”, “such as”, or variants thereof, are used in either the specification and/or the claims, such terms are not limiting and are intended to be inclusive in a manner similar to the term “comprising”.
  • The term “about,” as used herein, generally refers to a range that is 15% greater than or less than a stated numerical value within the context of the particular usage. For example, “about 10” would include a range from 8.5 to 11.5.
  • The term “barcode”, as used herein, generally refers to a label that may be attached to a polynucleotide, or any variant thereof, to convey information about the polynucleotide. For example, a barcode may be a polynucleotide sequence attached to all fragments of a target polynucleotide contained within a particular partition. This barcode may then be sequenced with the fragments of the target polynucleotide. The presence of the same barcode on multiple sequences may provide information about the origin of the sequence. For example, a barcode may indicate that the sequence came from a particular partition and/or a proximal region of a genome. This may be particularly useful when several partitions are pooled before sequencing.
  • The term “bp,” as used herein, generally refers to an abbreviation for “base pairs”.
  • The term “Mer,” as used herein to refer to restriction enzymes, generally refers to the number of nucleotides in one strand of a restriction enzyme's recognition site. For example, the enzyme CviQI has a recognition site of GTAC (4 nucleotides on one strand) and is thus referred to as a “4 Mer cutter.” The enzyme StuI has a recognition site of AGGCCT (6 nucleotides on one strand) and is thus referred to as a “6 Mer cutter.”
  • The term “microwell,” as used herein, generally refers to a well with a volume of less than 1 mL. Microwells may be made in various volumes, depending on the application. For example, microwells may be made in a size appropriate to accommodate any of the partition volumes described herein.
  • The terms “non-overlapping” and “overlapping,” as used to refer to polynucleotide fragments, generally refer to a collection of polynucleotide fragments without overlapping sequence or with overlapping sequence, respectively. By way of illustration, consider a hypothetical partition containing three copies of a genome (FIG. 1, top set of sequences). This genome may be fragmented randomly (e.g., by shearing in a pipette) or non-randomly (e.g., by digesting with a rare cutter). Fragmenting randomly produces overlapping sequences (second set of sequences from top in FIG. 1, “Fragmented randomly to generate overlap”), because each copy of the genome is cut at different positions. After sequencing of the fragments (which provides “sequence contigs”), this overlap may be used to determine the linear order of the fragments, thereby enabling assembly of the entire genomic sequence. By contrast, fragmenting by digesting with a rare cutter produces non-overlapping fragments, because each copy of the (same) genome is cut at the same position (third set of sequences from the top in FIG. 1, “Fragmented non-randomly using RE-1 to generate non-overlapping fragments”). After sequencing these fragments, it may be difficult to deduce their linear order due to the lack of overlap between the fragments. However, as described in this disclosure, the linear order may be determined by, for example, fragmenting the genome using a different technique. The fourth set of sequences from the top of FIG. 1 demonstrates the use of a second rare-cutter enzyme to generate a second set of non-overlapping fragments (“Fragmented non-randomly using RE-2 to generate non-overlapping fragments”). Because two different enzymes, for example, are used to generate the two sets of non-overlapping fragments, there is overlap between the fragments generated with the first rare-cutter enzyme (RE-1) and the fragments generated with the second rare-cutter enzyme (RE-2). This overlap may then be used to assemble the linear order of the sequences, and therefore the sequence of the entire genome.
  • The term “partition,” as used herein, may be a verb or a noun. When used as a verb (e.g., “partitioning”), the term refers to the fractionation of a substance (e.g., a polynucleotide) between vessels that can be used to sequester one fraction from another. Such vessels are referred to using the noun “partition.” Partitioning may be performed, for example, using microfluidics, dilution, dispensing, and the like. A partition may be, for example, a well, a microwell, a droplet, a test tube, a spot, or any other means of sequestering one fraction of a sample from another. In the methods and systems described herein, polynucleotides are often partitioned into microwells.
  • The terms “polynucleotide” or “nucleic acid,” as used herein, are used herein to refer to biological molecules comprising a plurality of nucleotides. Exemplary polynucleotides include deoxyribonucleic acids, ribonucleic acids, and synthetic analogues thereof, including peptide nucleic acids.
  • The term “rare-cutter enzyme,” as used herein, generally refers to an enzyme with a recognition site that occurs only rarely in a genome. The size of restriction fragments generated by cutting a hypothetical random genome with a restriction enzyme may be approximated by 4N, where N is the number of nucleotides in the recognition site of the enzyme. For example, an enzyme with a recognition site consisting of 7 nucleotides would cut a genome once every 47 bp, producing fragments of about 16,384 bp. Generally rare-cutter enzymes have recognition sites comprising 6 or more nucleotides. For example, a rare cutter enzyme may have a recognition site comprising or consisting of 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 nucleotides. Examples of rare-cutter enzymes include Notl (GCGGCCGC), XmaIII (CGGCCG), SstII (CCGCGG), SalI (GTCGAC), NruI (TCGCGA), NheI (GCTAGC), Nb.BbvCI (CCTCAGC), BbvCI (CCTCAGC), AscI (GGCGCGCC), AsiSI (GCGATCGC), FseI (GGCCGGCC), PacI (TTAATTAA), PmeI (GTTTAAAC), SbfI (CCTGCAGG), SgrAI (CRCCGGYG), SwaI (ATTTAAAT), BspQI (GCTCTTC), SapI (GCTCTTC), SfiI (GGCCNNNNNGGCC (SEQ ID NO: 1)), CspCI (CAANNNNNGTGG (SEQ ID NO: 2)), AbsI (CCTCGAGG), CciNI (GCGGCCGC), FspAI (RTGCGCAY), MauBI (CGCGCGCG), MreI (CGCCGGCG), MssI (GTTTAAAC), PalAI (GGCGCGCC), RgaI (GCGATCGC), RigI (GGCCGGCC), SdaI (CCTGCAGG), SfaAI (GCGATCGC), SgfI (GCGATCGC), SgrDI (CGTCGACG), SgsI (GGCGCGCC), SmiI (ATTTAAAT), SrfI (GCCCGGGC), Sse2321 (CGCCGGCG), Sse83871 (CCTGCAGG), LguI (GCTCTTC), PciSI (GCTCTTC), AarI (CACCTGC), AjuI (GAANNNNNNNTTGG (SEQ ID NO: 3)), AloI (GAACNNNNNNTCC (SEQ ID NO: 4)), BarI (GAAGNNNNNNTAC (SEQ ID NO: 5)), PpiI (GAACNNNNNCTC (SEQ ID NO: 6)), PsrI (GAACNNNNNNTAC (SEQ ID NO: 7)), and others.
  • The term “target polynucleotide,” as used herein, generally refers to a polynucleotide to be processed. For example, if a user intends to process genomic DNA into fragments that may be sequenced, the genomic DNA would be the target polynucleotide. If a user intends to process fragments of a polynucleotide, then the fragments of the polynucleotide may be the target polynucleotide.
  • II. Non-Overlapping Fragmentation
  • This disclosure provides methods, compositions, systems, and devices for the generation of non-overlapping polynucleotide fragments. These fragments may be useful for downstream analyses such as DNA sequencing. For example, with reference to FIG. 2, a target polynucleotide 101, such as genomic DNA, may be fragmented to generate a plurality of non-overlapping first polynucleotide fragments 102. This fragmentation may be performed, for example, by digesting the target polynucleotide with a rare-cutter enzyme (e.g., rare-cutter enzyme 1), or an artificial restriction DNA cutter (ARCUT; Yamamoto et al., Nucleic Acids Res., 2007, 35(7), e53). The first polynucleotide fragments may then be partitioned, such that at least one partition 103 comprises a first polynucleotide fragment with a unique sequence within that partition and, optionally, an additional first polynucleotide fragment with a different sequence 104. The partitioned first polynucleotide fragments may then be further fragmented to produce a plurality of non-overlapping second polynucleotide fragments 105. This fragmentation may be performed, for example, by enzymatic digestion, exposure to ultraviolet (UV) light, ultrasonication, and/or mechanical agitation. The second polynucleotide fragments may be of a size that is appropriate for DNA sequencing, i.e., a size that enables a DNA sequencer to obtain accurate sequence data for the entire fragment.
  • In order to facilitate DNA sequence assembly, the second fragments may be attached to a barcode, which may be attached to all of the second fragments disposed in a particular partition. The barcode may be, for example, a DNA barcode. With continued reference to FIG. 2, after attachment of the barcode, the barcoded fragments may be pooled into a partition comprising pooled, barcoded sequences 106. Three barcodes are depicted as [1], [2], and [3] in 106. The pooled fragments may be sequenced.
  • Certain methods of genome sequence assembly rely on the presence of overlapping fragments in order to generate higher order sequence data (e.g., whole genome sequences) from sequenced fragments. The methods, compositions, systems, and devices provided herein may also be used to provide overlapping fragments. For example, with continued reference to FIG. 2, fragments overlapping with the first and second fragments described above may be generated by generating a plurality of non-overlapping third polynucleotide fragments from the target polynucleotide 107. The third polynucleotide fragments may be generated, for example, by digesting the target polynucleotide 101 with a rare-cutter enzyme (e.g., rare-cutter enzyme 2; or ARCUT) that is different from the rare-cutter enzyme used to generate the first polynucleotide fragments. If rare- cutter enzymes 1 and 2 are chosen to cut the target polynucleotide sequence at different positions, the third polynucleotide fragments and the first polynucleotide fragments will overlap. The third polynucleotide fragments may then be processed as described above for the first polynucleotide fragments.
  • Specifically, the third polynucleotide fragments may be partitioned such that at least one partition 108 comprises a third polynucleotide fragment with a unique sequence within that partition and, optionally, an additional third polynucleotide fragment with a different sequence 109. These partitioned fragments may then be further fragmented to produce a plurality of non-overlapping fourth polynucleotide fragments 110. The fourth polynucleotides fragments and the second polynucleotide fragments may overlap. As for the second polynucleotide fragments, the fourth polynucleotide fragments may be generated by, for example, enzymatic digestion, exposure to ultraviolet (UV) light, ultrasonication, and/or mechanical agitation. The fourth fragments may be of a size that is appropriate for DNA sequencing, i.e., a size that enables a DNA sequencer to obtain accurate sequence data for the entire fragment.
  • In order to facilitate DNA sequencing, the fourth fragments may be attached to a barcode, which may be attached to all of the fourth fragments disposed in a particular partition. The barcode may be, for example, a DNA barcode. After attachment of the barcode, the barcoded fragments may be pooled, into a partition comprising pooled, barcoded, sequences 111. Three barcodes are depicted as [4], [5], and [6] in 111. The pooled fragments may be sequenced. The overlap between the sequences of the second fragments and the fourth fragments may be used to assemble higher order sequences, such whole genome sequences.
  • The steps described above may be performed using a variety of techniques. For example, certain steps of the methods may be performed in a device comprising microwell chambers (microwells), for example a microfluidic device. These microwells may be connected to each other, or to a source of reagents, by channels. The first and third fragments may be generated outside of the device and then introduced into the device (or separate devices) for further processing. Partitioning of the first and third fragments may accomplished using fluidic techniques. Generation of the second and fourth fragments may then occur within the microwells of the device or devices. These microwells may contain reagents for barcoding of the second and fourth fragments, such as DNA barcodes, ligase, adapter sequences, and the like. Microwells may feed or be directed into a common outlet, so that barcoded fragments may be pooled or otherwise collected into one or more aliquots which may then be sequenced.
  • In another example, the entire process could be performed within a single device. For example, a device could be split into two sections. A first section may comprise a partition comprising rare-cutter enzyme 1 (generating first polynucleotide fragments) and a second section may comprise a partition comprising rare-cutter enzyme 2 (generating third polynucleotide fragments). An aliquot of the target polynucleotide sequence may be placed into each of these partitions. Following digestion, the enzyme may be inactivated and the samples may be partitioned, fragmented, barcoded, pooled, and sequenced as described above. For convenience, this example has been described using rare-cutter enzymes as the means of generating the first and third fragments. However, this is not intended to be limiting, here or anywhere else in this disclosure. One of ordinary skill in the art will readily recognize that other means of generating non-overlapping, or predominantly non-overlapping, fragments would be just as suitable as the use of rare-cutter enzymes.
  • III. Pseudo-Random Fragmentation
  • This disclosure also provides methods, compositions, systems, and devices for fragmenting polynucleotides in a pseudo-random manner. This may be performed by treating partitioned polynucleotides with more than one restriction enzyme. For example, polynucleotides partitioned into microwells may be treated with combinations of restriction enzymes. Within each partition containing a particular combination of enzymes, the cutting is defined and predictable. However, across all of the partitions (through the use of multiple combinations of restriction enzymes in different partitions), the polynucleotide fragments generated approximate those obtained from methods of random fragmentation. However, these polynucleotide fragments are generated in a much more controlled manner than random fragments generated by methods known in the art (e.g., shearing). The partitioned, pseudo-randomly fragmented polynucleotides may be barcoded, as described throughout this disclosure, pooled, and sequenced. The pseudo-random fragmentation methods may be used with the non-overlapping fragmentation methods described herein, or with any other method described herein such as the high yield adapter/barcode attachment method. Pseudo-random fragmentation may occur by exposing a polynucleotide to multiple enzymes simultaneously, sequentially, or simultaneously and sequentially.
  • Thus, this disclosure provides methods and systems for processing polynucleotides comprising generating pseudo-random fragments of said polynucleotides. These pseudo random fragments are generated by treating a polynucleotide with more than one restriction enzyme. For example, a polynucleotide may be treated with about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, 50, or more restriction enzymes. A polynucleotide may be treated with at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, 50, or more restriction enzymes. A polynucleotide may be treated with at least 2 but fewer than 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes. A polynucleotide may be treated with about 2-4, 4-6, 6-8, 8-10, 10-12, 12-14, 14-16, 16-18, 18-20, 20-25, 25-30, 35-40, 40-45, or 45-50 restriction enzymes.
  • The restriction enzymes may be chosen in order to maximize the number or fraction of fragments that will provide accurate sequencing data, based on the size of the fragments generated by the pseudo-random fragmentation. For present day sequencing technology, accuracy degrades beyond a read length of about 100 nucleotides. Therefore, fragments of about 200 or fewer nucleotides generally provide the most accurate sequence data since they can be sequenced from either end. Fragments below about 50 nucleotides are generally less desirable because, although the produce accurate sequencing data, they underutilize the read length capacity of current sequencing instruments which are capable of 150 to 200 base reads. Fragments of about 200 to about 400 nucleotides may be sequenced with systematic errors introduced as the read length increases beyond the initial 100 bases from each end. Sequence information from fragments greater than about 400 nucleotides is typically completely lost for those bases greater than 200 bases from either end. One of skill in the art will recognize that sequencing technology is constantly advancing and that the ability to obtain accurate sequence information from longer fragments is also constantly improving. Thus, the pseudo-random fragmentation methods presented herein may be used to produce optimal fragment lengths for any sequencing method.
  • In some cases, fragments may be defined by the distance of their component nucleotides from a restriction site (measured in nucleotides). For example, each nucleotide within a polynucleotide fragment generated by the pseudo-random fragmentation method may be less than about 10, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, 400, 550, 600, 1000, 5000, 10000, or 100000 nucleotides from the restriction site of an enzyme to which the polynucleotide is exposed. Each nucleotide within a polynucleotide fragment may be about 10, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, 400, 550, 600, 1000, 5000, 10000, or 100000 nucleotides from the restriction site of an enzyme to which the polynucleotide is exposed. Each nucleotide within a polynucleotide fragment may be at least about 10, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, 400, 550, 600, 1000, 5000, 10000, or 100000 nucleotides from the restriction site of an enzyme to which the polynucleotide is exposed.
  • In some cases, at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%, of the nucleotides comprising a target polynucleotide sequence are within about 10, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, 400, 550, 600, 1000, 5000, 10000, or 100000 nucleotides from the restriction site of an enzyme to which the polynucleotide is exposed. All combinations of these percentages and polynucleotide lengths are contemplated.
  • In some cases, at less than about 1%, 5%, 10%, 25%, 30%, 35%, 40%, 45%, or 50% of the nucleotides comprising a target polynucleotide sequence are within about 1, 5, 10, 50, 200, 250, 300, 350, 400, 550, 600, 1000, 5000, 10000, or 100000 nucleotides from the restriction site of an enzyme to which the polynucleotide is exposed. All combinations of these percentages and polynucleotide lengths are contemplated.
  • The pseudo-random fragmentation methods may be used to obtain fragments of about 10 to 50 nucleotides, 46 to 210 nucleotides, 50 to 250 nucleotides, 250 to 400 nucleotides, 400 to 550 nucleotides, 550 to 700 nucleotides, 700 to 1000 nucleotides, 1000 to 1300 nucleotides, 1300 to 1600 nucleotides, 1600 to 1900 nucleotides, 1900 to 2200 nucleotides, or 2200 to 3000 nucleotides. The pseudo-random fragmentation methods may be used to obtain fragments with a mean or median of about 40 nucleotides, 60 nucleotides, 80 nucleotides, 100 nucleotides, 120 nucleotides, 130 nucleotides, 140 nucleotides, 160 nucleotides, 180 nucleotides, 200 nucleotides, 250 nucleotides, 300 nucleotides, 400 nucleotides, 500 nucleotides, 600 nucleotides, 700 nucleotides, 800 nucleotides, 900 nucleotides, 1000 nucleotides, 1200 nucleotides, 1400 nucleotides, 1600 nucleotides, 1800 nucleotides, 2000 nucleotides, 2500 nucleotides, 3000 nucleotides, or more. The pseudo-random fragmentation methods may be used to obtain fragments with a mean or median of at least about 40 nucleotides, 60 nucleotides, 80 nucleotides, 100 nucleotides, 120 nucleotides, 130 nucleotides, 140 nucleotides, 160 nucleotides, 180 nucleotides, 200 nucleotides, 250 nucleotides, 300 nucleotides, 400 nucleotides, 500 nucleotides, 600 nucleotides, 700 nucleotides, 800 nucleotides, 900 nucleotides, 1000 nucleotides, 1200 nucleotides, 1400 nucleotides, 1600 nucleotides, 1800 nucleotides, 2000 nucleotides, 2500 nucleotides, 3000 nucleotides, or more. The pseudo-random fragmentation methods may be used to obtain fragments with a mean or median of less than about 40 nucleotides, 60 nucleotides, 80 nucleotides, 100 nucleotides, 120 nucleotides, 130 nucleotides, 140 nucleotides, 160 nucleotides, 180 nucleotides, 200 nucleotides, 250 nucleotides, 300 nucleotides, 400 nucleotides, 500 nucleotides, 600 nucleotides, 700 nucleotides, 800 nucleotides, 900 nucleotides, 1000 nucleotides, 1200 nucleotides, 1400 nucleotides, 1600 nucleotides, 1800 nucleotides, 2000 nucleotides, 2500 nucleotides, or 3000 nucleotides.
  • In some examples, the pseudo-random fragmentation methods provided herein are used to generate fragments wherein a particular percentage (or fraction) of the fragments generated fall within any of the size ranges described herein. For example, about 0%, 2%, 4%, 6%, 8%, 10%, 12%, 14%, 16%, 18%, 20%, 22%, 24%, 26%, 28%, 30%, 32%, 34%, 36%, 38%, 40%, 42%, 44%, 46%, 48%, 50%, 52%, 54%, 56%, 58%, 60%, 62%, 64%, 66%, 68%, 70%, 72%, 74%, 76%, 78%, 80%, 82%, 84%, 86%, 88%, 90%, 92%, 94%, 96%, 98%, or 100% of the fragments generated may fall within any of the size ranges described herein.
  • In some examples multiple 4 Mer cutters may be used to provide a distribution of about 18% of fragments of about 50 nucleotides or less, about 38% of fragments of about 200 nucleotides or less, about 25% of fragments between about 200 and about 400 nucleotides, and about 37% of fragments greater than about 400 nucleotides (e.g., see FIG. 4).
  • Additionally, the pseudo-random fragmentation method may be designed to minimize the percentage of fragments greater than a certain number of nucleotides in length, in order to minimize the loss of sequence information. For example, the method may be designed to yield less than about 0.1%, 0.5%, 1%, 2%, 5%, 10%, 20%, or 50% fragments greater than 100 nucleotides. The method may be designed to yield less than about 0.1%, 0.5%, 1%, 2%, 5%, 10%, 20%, or 50% fragments greater than 150 nucleotides. The method may be designed to yield less than about 0.1%, 0.5%, 1%, 2%, 5%, 10%, 20%, or 50% fragments greater than 200 nucleotides. The method may be designed to yield less than about 0.1%, 0.5%, 1%, 2%, 5%, 10%, 20%, or 50% fragments greater than 300 nucleotides, and so on. As the ability of sequencing technologies to accurately read long DNA fragments increases, the pseudo-random fragmentation methods of the invention may be used to generate sequences suitable for any chosen read length.
  • Enzymes for use with the pseudo-random fragmentation method described herein may be chosen, for example, based on the length of their recognition site and their compatibility with certain buffer conditions (to allow for combination with other enzymes). Enzymes may also be chosen so that their cutting activity is methylation insensitive, or sensitive to methylation. For example, restriction enzymes with shorter recognition sites generally cut polynucleotides more frequently. Thus, cutting a target polynucleotide with a 6 Mer cutter will generally produce more large fragments than cutting the same polynucleotide with a 4 Mer cutter (e.g., compare FIGS. 3 and 4). Cutting a target polynucleotide with a plurality of enzymes (e.g. 2, 3, 4, 5, 6, 7, or more) may produce a greater number or fraction of fragments in the optimal size range for DNA sequencing than cutting with a single enzyme (see FIG. 5). Any restriction enzyme may be used with this method. Many are named in this specification, but others are known in the art.
  • This disclosure also provides methods of selecting a plurality of enzymes for pseudo-random fragmentation of a polynucleotide sequence. For example, a target polynucleotide may be exposed separately to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 restriction enzymes. The size distribution of the target polynucleotide fragments is then determined, for example, by electrophoresis. The combination of enzymes providing the greatest number of fragments that are capable of being sequenced can then be chosen. The method can also be carried out in silico.
  • The enzymes may be disposed within the same partition, or within a plurality of partitions. For example, any of the plurality of enzyme number described herein may be disposed within a single partition, or across partitions. For example, a polynucleotide may be treated with about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, 50, or more restriction enzymes in the same partition, or across partitions. A polynucleotide may be treated with at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, 50, or more restriction enzymes in the same partition, or across partitions. A polynucleotide may be treated with at least 2 but fewer than 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes in the same partition, or across partitions. A polynucleotide may be treated with about 2-4, 4-6, 6-8, 8-10, 10-12, 12-14, 14-16, 16-18, 18-20, 20-25, 25-30, 35-40, 40-45, or 45-50 restriction enzymes in the same partition, or across partitions.
  • The distribution of the restriction enzymes among the partitions will vary depending on the restriction enzymes, the target polynucleotide, and the desired fragment size. In some cases, each restriction enzyme may be distributed across an equivalent number of partitions, so that the number of partitions occupied by each restriction enzyme is equivalent. For example, if 10 restriction enzymes are used in a device containing 1,000 partitions, each enzyme may be present in 100 partitions. In other cases, each restriction enzyme may be distributed across a non-equivalent number of partitions, so that the number of partitions occupied by each restriction enzyme is not equivalent. For example, if 10 restriction enzymes are used in a device containing 1,000 partitions, enzymes 1-8 may be present in 100 partitions each, enzyme 9 may be present in 50 partitions, and enzyme 10 may be present in 150 partitions. Placement of restriction enzymes in an unequal number of partitions may be beneficial, for example, when an enzyme generates a desired product at a low yield. Placing this low-yield enzyme in more partitions will therefore expose more of the target polynucleotide to the enzyme, increasing the amount of the desired product (e.g., fragment of a certain size or composition) that can be formed from the enzyme. Such an approach may be useful for accessing portions of a target polynucleotide (e.g., a genome) that are not cut by enzymes producing polynucleotide fragments at a higher yield. The restriction site and efficiency of an enzyme, composition of the target polynucleotide, and efficiency and side-products generated by the enzyme may all be among the factors considered when determining how many partitions should receive a particular enzyme.
  • In some cases, different numbers of restriction enzymes may be used in a single partition and across all partitions. For example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes or more may be used in each partition, while 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes or more may be used across all partitions. All combinations of these numbers are included within the invention. Non-limiting specific examples include the use of 1 restriction enzyme per partition and 2, 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; 2 restriction enzymes per partition and 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; 3 restriction enzymes per partition and 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; 4 restriction enzymes per partition and 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; 5 restriction enzymes per partition and 6, 7, 8, 9, or 10 restriction enzymes across all partitions; 6 restriction enzymes per partition and 7, 8, 9, or 10 restriction enzymes across all partitions; 7 restriction enzymes per partition and 8, 9, or 10 restriction enzymes across all partitions; 8 restriction enzymes per partition and 9 or 10 restriction enzymes across all partitions; and 9 restriction enzymes per partition and 10 or more restriction enzymes across all partitions.
  • In some cases, at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes or more may be used in each partition, while at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes or more may be used across all partitions. All combinations of these numbers are included within the invention. Non-limiting specific examples include the use of at least 1 restriction enzyme per partition and at least 2, 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 2 restriction enzymes per partition and at least 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 3 restriction enzymes per partition and at least 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 4 restriction enzymes per partition and at least 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 5 restriction enzymes per partition and at least 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 6 restriction enzymes per partition and at least 7, 8, 9, or 10 restriction enzymes across all partitions; at least 7 restriction enzymes per partition and at least 8, 9, or 10 restriction enzymes across all partitions; at least 8 restriction enzymes per partition and at least 9 or 10 restriction enzymes across all partitions; and at least 9 restriction enzymes per partition and at least 10 or more restriction enzymes across all partitions.
  • In some cases, at most 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes or more may be used in each partition, while at most 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes or more may be used across all partitions. All combinations of these numbers are included within the invention. Non-limiting specific examples include the use of at most 1 restriction enzyme per partition and at most 2, 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 2 restriction enzymes per partition and at most 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 3 restriction enzymes per partition and at most 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 4 restriction enzymes per partition and at most 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 5 restriction enzymes per partition and at most 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 6 restriction enzymes per partition and at most 7, 8, 9, or 10 restriction enzymes across all partitions; at most 7 restriction enzymes per partition and at most 8, 9, or 10 restriction enzymes across all partitions; at most 8 restriction enzymes per partition and at most 9 or 10 restriction enzymes across all partitions; and at most 9 restriction enzymes per partition and at most 10 or more restriction enzymes across all partitions.
  • In some cases, at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes or more may be used in each partition, while at most 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes or more may be used across all partitions. All combinations of these numbers are included within the invention. Non-limiting specific examples include the use of at least 1 restriction enzyme per partition and at most 2, 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 2 restriction enzymes per partition and at most 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 3 restriction enzymes per partition and at most 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 4 restriction enzymes per partition and at most 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 5 restriction enzymes per partition and at most 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at least 6 restriction enzymes per partition and at most 7, 8, 9, or 10 restriction enzymes across all partitions; at least 7 restriction enzymes per partition and at most 8, 9, or 10 restriction enzymes across all partitions; at least 8 restriction enzymes per partition and at most 9 or 10 restriction enzymes across all partitions; and at least 9 restriction enzymes per partition and at most 10 or more restriction enzymes across all partitions.
  • In some cases, at most 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes or more may be used in each partition, while at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 45, 45, or 50 restriction enzymes or more may be used across all partitions. All combinations of these numbers are included within the invention. Non-limiting specific examples include the use of at most 1 restriction enzyme per partition and at least 2, 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 2 restriction enzymes per partition and at least 3, 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 3 restriction enzymes per partition and at least 4, 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 4 restriction enzymes per partition and at least 5, 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 5 restriction enzymes per partition and at least 6, 7, 8, 9, or 10 restriction enzymes across all partitions; at most 6 restriction enzymes per partition and at least 7, 8, 9, or 10 restriction enzymes across all partitions; at most 7 restriction enzymes per partition and at least 8, 9, or 10 restriction enzymes across all partitions; at most 8 restriction enzymes per partition and at least 9 or 10 restriction enzymes across all partitions; and at most 9 restriction enzymes per partition and at least 10 or more restriction enzymes across all partitions.
  • IV. Restriction Enzyme-Mediated Recycling
  • As described throughout this disclosure, certain methods of the invention involve the addition of barcodes, adapters, or other sequences to fragmented target polynucleotides. Barcodes may be polynucleotide barcodes, which may be ligated to the fragmented target polynucleotides or added via an amplification reaction. As described throughout this disclosure, fragmentation of target polynucleotides may be performed using one or more restriction enzymes contained within a partition (e.g., a microwell) where the fragmentation is performed. The partition may also contain a polynucleotide barcode and a ligase, which enables the attachment of the barcode to the fragmented polynucleotide. In some cases, an adapter may be used to make a fragmented target polynucleotide compatible for ligation with a barcode. The presence of adapters, fragmented target polynucleotide, barcodes, restriction enzymes, and ligases in the same partition may lead to the generation of undesirable side products that decrease the yield of a desired end product. For example, self-ligation may occur between adapters, target polynucleotide fragments, and/or barcodes. These self-ligations reduce the amount of starting material and decrease the yield of the desired product, for example, a polynucleotide fragment properly ligated to a barcode and/or and adapter.
  • This disclosure provides methods, compositions, systems, and devices for addressing this problem and increasing the yield of a desired product. The problem is addressed by pairing a first restriction enzyme and a second restriction enzyme. The two restriction enzymes create compatible termini upon cutting, but each enzyme has a different recognition sequence.
  • Ligation of two pieces of DNA generated after cutting with the first restriction enzyme will regenerate the recognition site for the first restriction enzyme, allowing the first restriction enzyme to re-cut the ligated DNA. Likewise, ligation of two pieces of DNA generated after cutting with the second restriction enzyme will regenerate the recognition site for the second restriction enzyme, allowing the second restriction enzyme to re-cut the ligated DNA. However, ligation of one piece of DNA generated after cutting with the first restriction enzyme and one piece of DNA generated after cutting with the second restriction enzyme will result in ligated DNA that is unrecognizable (and therefore uncuttable) by both the first and second enzymes. The result is that any multimers of fragmented target polynucleotides are re-cut and any multimers of adapter (or other molecules, e.g., barcodes) are also re-cut. However, when a fragmented target polynucleotide is properly ligated to an adapter (or barcode), the restriction sites for both enzymes are not present and the correctly ligated molecule may not be re-cut by either enzyme.
  • An example of this method is illustrated in FIG. 6, and additional pairs of enzymes that may be used with the method are provided in FIGS. 7A-7B. Any pair of enzymes may be used, so long as they meet the following criteria: (1) the enzymes should create identical, or at least similar, ligatable termini upon cutting; and (2) the enzymes should have different recognition sequences. The enzymes may be selected to avoid or minimize cutting of certain polynucleotide sequences such as barcodes, adapters, and other polynucleotide components of a sample processing or preparation platform. The enzymes may be selected for methylation insensitivity or methylation sensitivity. The enzymes may also be selected to be active under s single set of environmental conditions, such as buffer conditions, temperature, etc. Minimizing the cutting of barcodes and adapters may be accomplished by pairing certain enzymes with certain barcodes and/or adapters.
  • This method may be used to increase the yield of any of the barcoding methods described herein. The regeneration of the starting materials (e.g., fragmented target polynucleotide, adapters, and barcodes) allows these starting materials another opportunity to form the desired products (i.e., fragmented target polynucleotides ligated to barcodes, optionally with adapters). This greatly increases the yield of the reaction and therefore decreases the amount of starting material required to produce the necessary amount of the desired products while limiting the amount of undesirable side products and lost sequence information.
  • The methods described above may be used to achieve about 75%, 85%, 95%, 96%, 97%, 98%, 99%, or 99.5% yield (w/w). The methods may be used to achieve at least about 75%, 85%, 95%, 96%, 97%, 98%, 99%, or 99.5% yield (w/w).
  • The methods described above may use, for example, a pair of restriction enzyme selected from the group consisting of MspI-NarI, BfaI-NarI, BfaI-NdeI, HinP1I-ClaI, Msel-NdeI, CviQI-NdeI, TaqαI-AcII, RsaI-PmeI, AluI-EcoRV, BstUI-PmeI, DpnI-StuI, HaeIII-PmeI, and HpyCH4V-SfoI. This list of enzymes is provided for purposes of illustration only, and is not meant to be limiting.
  • The methods described above may generally use any two enzymes that create ligatable termini upon cutting but that have different recognition sequences. However, the method is not limited to ligation. For example, multimers formed after amplification of side products formed by association of compatible ends could also be re-cut using the methods described above.
  • More than one pair of enzymes may also be used. The number of pairs of enzymes chosen will vary depending on the number of undesirable side products formed in a reaction. For example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more pairs of enzymes may be used. Treatment of a polynucleotide with the enzymes may be sequential, simultaneous, or both.
  • V. Preparation of Target Polynucleotides
  • Target polynucleotides processed according to the methods provided in this disclosure may be DNA, RNA, peptide nucleic acids, and any hybrid thereof, where the polynucleotide contains any combination of deoxyribo- and ribo-nucleotides. Polynucleotides may be single stranded or double stranded, as specified, or contain portions of both double stranded or single stranded sequence. Polynucleotides may contain any combination of nucleotides, including uracil, adenine, thymine, cytosine, guanine, inosine, xanthine, hypoxanthine, isocytosine, isoguanine and any nucleotide derivative thereof. As used herein, the term “nucleotide” may include nucleotides and nucleosides, as well as nucleoside and nucleotide analogs, and modified nucleotides, including both synthetic and naturally occurring species. Target polynucleotides may be cDNA, mitochondrial DNA (mtDNA), messenger RNA (mRNA), ribosomal RNA (rRNA), transfer RNA (tRNA), nuclear RNA (nRNA), small interfering RNA (siRNA), small nuclear RNA (snRNA), small nucleolar RNA (snoRNA), small Cajal body-specific RNA (scaRNA), microRNA (miRNA), double stranded (dsRNA), ribozyme, riboswitch or viral RNA. Target polynucleotides may be contained on a plasmid, cosmid, or chromosome, and may be part of a genome. In some cases, a target polynucleotide may comprise one or more genes and/or one or more pseudogenes. A pseudogene generally refers to a dysfunctional relative of a gene that has lost its protein coding ability and/or is otherwise no longer expressed in the cell.
  • Target polynucleotides may be obtained from a sample using any methods known in the art. A target polynucleotide processed as described herein may be obtained from whole cells, cell preparations and cell-free compositions from any organism, tissue, cell, or environment. In some instances, target polynucleotides may be obtained from bodily fluids which may include blood, urine, serum, lymph, saliva, mucosal secretions, perspiration, or semen. In some instances, polynucleotides may be obtained from environmental samples including air, agricultural products, water, and soil. In other instances polynucleotides may be the products of experimental manipulation including, recombinant cloning, polynucleotide amplification (as generally described in PCT/US99/01705), polymerase chain reaction (PCR) amplification, purification methods (such as purification of genomic DNA or RNA), and synthesis reactions.
  • Genomic DNA may be obtained from naturally occurring or genetically modified organisms or from artificially or synthetically created genomes. Target polynucleotides comprising genomic DNA may be obtained from any source and using any methods known in the art. For example, genomic DNA may be isolated with or without amplification. Amplification may include PCR amplification, multiple displacement amplification (MDA), rolling circle amplification and other amplification methods. Genomic DNA may also be obtained by cloning or recombinant methods, such as those involving plasmids and artificial chromosomes or other conventional methods (see Sambrook and Russell, Molecular Cloning: A Laboratory Manual., cited supra.) Polynucleotides may be isolated using other methods known in the art, for example as disclosed in Genome Analysis: A Laboratory Manual Series (Vols. I-IV) or Molecular Cloning: A Laboratory Manual. If the isolated polynucleotide is an mRNA, it may be reverse transcribed into cDNA using conventional techniques, as described in Sambrook and Russell, Molecular Cloning: A Laboratory Manual., cited supra.
  • Target polynucleotides may also be isolated from “target organisms” or “target cells”. The terms “target organism” and “target cell” refer to an organism or cell, respectively, from which target polynucleotides may be obtained. Target cells may be obtained from a variety of organisms including human, mammal, non-human mammal, ape, monkey, chimpanzee, plant, reptilian, amphibian, avian, fungal, viral or bacterial organisms. Target cells may also be obtained from a variety of clinical sources such as biopsies, aspirates, blood, urine, formalin fixed embedded tissues, and the like. Target cells may comprise a specific cell type, such as a somatic cell, germline cell, wild-type cell, cancer or tumor cells, or diseased or infected cell. A target cell may refer to a cell derived from a particular tissue or a particular locus in a target organism A target cell may comprise whole intact cells, or cell preparations.
  • Target polynucleotides may also be obtained or provided in specified quantities. Amplification may be used to increase the quantity of a target polynucleotide. Target polynucleotides may quantified by mass. For example, target polynucleotides may be provided in a mass ranging from about 1-10, 10-50, 50-100, 100-200, 200-1000, 1000-10000 ng. Target polynucleotides may be provided in a mass of at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, or 10000 ng. Target polynucleotides may be provided in a mass of less than about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, or 10000 ng.
  • Target polynucleotides may also be quantified as “genome equivalents.” A genome equivalent is an amount of polynucleotide equivalent to one haploid genome of an organism from which the target polynucleotide is derived. For example, a single diploid cell contains two genome equivalents of DNA. Target polynucleotides may be provided in an amount ranging from about 1-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, or 100000-1000000 genome equivalents. Target polynucleotides may be provided in an amount of at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, 20000, 30000, 40000, 50000, 60000 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, or 1000000 genome equivalents. Target polynucleotides may be provided in an amount less than about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, 20000, 30000, 40000, 50000, 60000 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, or 1000000 genome equivalents.
  • Target polynucleotide may also be quantified by the amount of sequence coverage provided. The amount of sequence coverage refers to the average number of reads representing a given nucleotide in a reconstructed sequence. Generally, the greater the number of times a region is sequenced, the more accurate the sequence information obtained. Target polynucleotides may be provided in an amount that provides a range of sequence coverage from about 0.1X-10X, 10-X-50X, 50X-100X, 100X-200X, or 200X-500X. Target polynucleotide may be provided in an amount that provides at least about 0.1X, 0.2X, 0.3X, 0.4X, 0.5X, 0.6X, 0.7X, 0.8X, 0.9X, 1.0X, 5X, 10X, 25X, 50X, 100X, 125X, 150X, 175X, or 200X sequence coverage. Target polynucleotide may be provided in an amount that provides less than about 0.2X, 0.3X, 0.4X, 0.5X, 0.6X, 0.7X, 0.8X, 0.9X, 1.0X, 5X, 10X, 25X, 50X, 100X, 125X, 150X, 175X, or 200X sequence coverage.
  • VI. Fragmentation of Target Polynucleotides
  • Fragmentation of polynucleotides is used as a step in a variety of processing methods described herein. The size of the polynucleotide fragments, typically described in terms of length (quantified by the linear number of nucleotides per fragment), may vary depending on the source of the target polynucleotide, the method used for fragmentation, and the desired application. Moreover, while certain methods of the invention are illustrated using a certain number of fragmentation steps, the number of fragmentation steps provided is not meant to be limiting, and any number of fragmentation steps may be used. For example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more fragmentation steps may be used.
  • Fragments generated using the methods described herein may be about 1-10, 10-20, 20-50, 50-100, 50-200, 100-200, 200-300, 300-400, 400-500, 500-1000, 1000-5000, 5000-10000, 10000-100000, 100000-250000, or 250000-500000 nucleotides in length. Fragments generated using the methods described herein may be at least about 10, 20, 100, 200, 300, 400, 500, 1000, 5000, 10000, 100000, 250000, 500000, or more nucleotides in length. Fragments generated using the methods described herein may be less than about 10, 20, 100, 200, 300, 400, 500, 1000, 5000, 10000, 100000, 250000, 500000, nucleotides in length.
  • Fragments generated using the methods described herein may have a mean or median length of about 1-10, 10-20, 20-50, 50-100, 50-200, 100-200, 200-300, 300-400, 400-500, 500-1000, 1000-5000, 5000-10000, 10000-100000, 100000-250000, or 250000-500000 nucleotides. Fragments generated using the methods described herein may have a mean or median length of at least about 10, 20, 100, 200, 300, 400, 500, 1000, 5000, 10000, 100000, 250000, 500000, or more nucleotides. Fragments generated using the methods described herein may have a mean or median length of less than about 10, 20, 100, 200, 300, 400, 500, 1000, 5000, 10000, 100000, 250000, 500000, nucleotides.
  • Numerous fragmentation methods are described herein and known in the art. For example, fragmentation may be performed through physical, mechanical or enzymatic methods. Physical fragmentation may include exposing a target polynucleotide to heat or to UV light. Mechanical disruption may be used to mechanically shear a target polynucleotide into fragments of the desired range. Mechanical shearing may be accomplished through a number of methods known in the art, including repetitive pipetting of the target polynucleotide, sonication and nebulization. Target polynucleotides may also be fragmented using enzymatic methods. In some cases, enzymatic digestion may be performed using enzymes such as using restriction enzymes.
  • While the methods of fragmentation described in the preceding paragraph, and in some paragraphs of the disclosure, are described with reference to “target” polynucleotides, this is not meant to be limiting, above or anywhere else in this disclosure. Any means of fragmentation described herein, or known in the art, can be applied to any polynucleotide used with the invention. In some cases, this polynucleotide may be a target polynucleotide, such as a genome. In other cases, this polynucleotide may be a fragment of a target polynucleotide which one wishes to further fragment. In still other cases, still further fragments may be still further fragmented. Any suitable polynucleotide may be fragmented according the methods described herein.
  • A fragment of a polynucleotide generally comprises a portion of the sequence of the targeted polynucleotide from which the fragment was generated. In some cases, a fragment may comprise a copy of a gene and/or pseudogene, including one included in the original target polynucleotide. In some cases, a plurality of fragments generated from fragmenting a target polynucleotide may comprise fragments that each comprise a copy of a gene and/or pseudogene.
  • Restriction enzymes may be used to perform specific or non-specific fragmentation of target polynucleotides. The methods of the present disclosure may use one or more types of restriction enzymes, generally described as Type I enzymes, Type II enzymes, and/or Type III enzymes. Type II and Type III enzymes are generally commercially available and well known in the art. Type II and Type III enzymes recognize specific sequences of nucleotide base pairs within a double stranded polynucleotide sequence (a “recognition sequence” or “recognition site”). Upon binding and recognition of these sequences, Type II and Type III enzymes cleave the polynucleotide sequence. In some cases, cleavage will result in a polynucleotide fragment with a portion of overhanging single stranded DNA, called a “sticky end.” In other cases, cleavage will not result in a fragment with an overhang, creating a “blunt end.” The methods of the present disclosure may comprise use of restriction enzymes that generate either sticky ends or blunt ends.
  • Restriction enzymes may recognize a variety of recognition sites in the target polynucleotide. Some restriction enzymes (“exact cutters”) recognize only a single recognition site (e.g., GAATTC). Other restriction enzymes are more promiscuous, and recognize more than one recognition site, or a variety of recognition sites. Some enzymes cut at a single position within the recognition site, while others may cut at multiple positions. Some enzymes cut at the same position within the recognition site, while others cut at variable positions.
  • The present disclosure provides method of selecting one or more restriction enzymes to produce fragments of a desired length. Polynucleotide fragmentation may be simulated in silico, and the fragmentation may be optimized to obtain the greatest number or fraction of polynucleotide fragments within a particular size range, while minimizing the number or fraction of fragments within undesirable size ranges. Optimization algorithms may be applied to select a combination of two or more enzymes to produce the desired fragment sizes with the desired distribution of fragments quantities.
  • A polynucleotide may be exposed to two or more restriction enzymes simultaneously or sequentially. This may be accomplished by, for example, adding more than one restriction enzyme to a partition, or by adding one restriction enzyme to a partition, performing the digestion, deactivating the restriction enzyme (e.g., by heat treatment) and then adding a second restriction enzyme. Any suitable restriction enzyme may be used alone, or in combination, in the methods presented herein.
  • Fragmenting of a target polynucleotide may occur prior to partitioning of the target polynucleotide or fragments generated from fragmenting. For example, genomic DNA (gDNA) may be fragmented, using, for example, a restriction enzyme, prior to the partitioning of its generated fragments. In another example, a target polynucleotide may be entered into a partition along with reagents necessary for fragmentation (e.g., including a restriction enzyme), such that fragmentation of the target polynucleotide occurs within the partition. For example, gDNA may be fragmented in a partition comprising a restriction enzyme, and the restriction enzyme is used to fragment the gDNA.
  • In some cases, a plurality of fragments may be generated prior to partitioning, using any method for fragmentation described herein. Some or all of the fragments of the plurality, for example, may each comprise a copy of a gene and/or a pseudogene. The fragments can be separated and partitioned such that each copy of the gene or pseudogene is located in a different partition. Each partition, for example, can comprise a different barcode sequence such that each copy of the gene and/or pseudogene can be associated with a different barcode sequence, using barcoding methods described elsewhere herein. Via the different barcode sequences, each gene and/or pseudogene can be counted and/or differentiated during sequencing of the barcoded fragments. Any sequencing method may be used, including those described herein.
  • For example, using restriction enzymes, genomic DNA (gDNA) can be fragmented to generate a plurality of non-overlapping fragments of the gDNA. At least some of the fragments of the plurality may each comprise a copy of a gene and/or a pseudogene. The fragments may be separated and partitioned such that each copy of the gene or pseudogene is located in a different partition. Each partition, for example, can comprise a different barcode sequence such that each copy of the gene and/or pseudogene may be barcoded with a different barcode sequence. Via the different barcode sequences, the genes and/or pseudogenes may be counted and or differentiated after sequencing of the barcoded fragments. Any sequencing method may be used, including those described herein.
  • IV. Partitioning of Polynucleotides
  • As described throughout the disclosure, certain methods, systems, and compositions of the disclosure may utilize partitioning of polynucleotides into separate partitions (e.g., microwells, droplets of an emulsion). These partitions may be used to contain polynucleotides for further processing, such as, for example, cutting, ligating, and/or barcoding.
  • Any number of devices, systems or containers may be used to hold, support or contain partitions of polynucleotides and their fragments. In some cases, partitions are formed from droplets, emulsions, or spots on a substrate. Weizmann et al. (Nature Methods, 2006, Vol. 3 No. 7 pages 545-550). Suitable methods for forming emulsions, which can be used as partitions or to generate microcapsules, include the methods described in Weitz et al. (U.S. Pub. No. 2012/0211084). Partitions may also be formed through the use of wells, microwells, multi-well plates, and microwell arrays. Partitioning may be performed using piezoelectric droplet generation (e.g., Bransky et al., Lab on a Chip, 2009, 9, 516-520). Partitioning may be performed using surface acoustic waves (e.g., Demirci and Montesano, Lab on a Chip, 2007, 7, 1139-1145).
  • Each partition may also contain, or be contained within any other suitable partition. For example, a well, microwell, hole, a surface of a bead, or a tube may comprise a droplet (e.g., a droplet in an emulsion), a continuous phase in an emulsion, a spot, a capsule, or any other suitable partition. A droplet may comprise a capsule, bead, or another droplet. A capsule may comprise a droplet, bead, or another capsule. These descriptions are merely illustrative, and all suitable combinations and pluralities are also envisioned. For example, any suitable partition may comprise a plurality of the same or different partitions. In one example, a well or microwell comprises a plurality of droplets and a plurality of capsules. In another example, a capsule comprises a plurality of capsules and a plurality of droplets. All combinations of partitions are envisioned. Table 1 shows non-limiting examples of partitions that may be combined with each other.
  • TABLE 1
    Examples of partitions that may be combined with each other.
    Well Spot Droplet Capsule
    Well Well inside Spot inside Droplet Capsule
    well well inside well inside well
    Spot Spot inside Spot inside Droplet Capsule
    well spot inside spot inside spot
    Droplet Droplet Droplet Droplet Droplet
    inside well inside spot inside droplet inside
    capsule
    Capsule
    inside droplet
    Capsule Capsule Capsule Capsule Capsule
    inside well inside spot inside droplet inside
    Spot inside Droplet capsule
    capsule inside
    capsule
    Surface Bead inside Spot on bead Bead inside Bead inside
    of a well Bead inside droplet capsule
    Bead spot
  • Any partition described herein may comprise multiple partitions. For example, a partition may comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, or 50000 partitions. A partition may comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, or 50000 partitions. In some cases, a partition may comprise less than 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, or 50000 partitions. In some cases, each partition may comprise 2-50, 2-20, 2-10, or 2-5 partitions.
  • The number of partitions employed may vary depending on the application. For example, the number of partitions may be about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, or 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000, or more. The number of partitions may be at least about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000, or more. The number of partitions may be less than about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000. The number of partitions may be about 5-10000000, 5-5000000, 5-1,000,000, 10-10,000, 10-5,000, 10-1,000, 1,000-6,000, 1,000-5,000, 1,000-4,000, 1,000-3,000, or 1,000-2,000.
  • Such partitions may be pre-loaded with reagents to perform a particular reaction. For example, a capsule containing one or more reagents may be placed within a microwell. After adding a polynucleotide sample to the well, the capsule may be made to release its contents. The contents of the capsule may include, for example, restriction enzymes, ligases, barcodes, and adapters for processing the polynucleotide sample placed in the microwell.
  • In some cases, such partitions may be droplets of an emulsion. For example, a droplet of an emulsion may be an aqueous droplet in an oil phase. The droplet may comprise, for example, one or more reagents (e.g., restriction enzymes, ligases, polymerases, reagents necessary for nucleic acid amplification (e.g., primers, DNA polymerases, dNTPs, buffers)), a polynucleotide sample, and a barcode sequence. In some cases, the barcode sequence, polynucleotide sample, or any reagent may be associated with a solid surface within a droplet. In some cases, the solid surface is a bead. In some cases, the bead is a gel bead (see e.g., Agresti et al., U.S. Patent Publication No. 2010/0136544). In some cases the droplet is hardened into a gel bead (e.g., via polymerization).
  • A species may be contained within a droplet in an emulsion containing, for example, a first phase (e.g., oil or water) forming the droplet and a second (continuous) phase (e.g., water or oil). An emulsion may be a single emulsion, for example, a water-in-oil or an oil-in-water emulsion. An emulsion may be a double emulsion, for example a water-in-oil-in-water or an oil-in-water-in-oil emulsion. Higher-order emulsions are also possible. The emulsion may be held in any suitable container, including any suitable partition described in this disclosure.
  • In some cases, droplets in an emulsion comprise other partitions. A droplet in an emulsion may comprise any suitable partition including, for example, another droplet (e.g., a droplet in an emulsion), a capsule, a bead, and the like. Each partition may be present as a single partition or a plurality of partitions, and each partition may comprise the same species or different species.
  • In one example, a droplet in an emulsion comprises a capsule comprising reagents for sample processing. As described elsewhere in this disclosure, a capsule may contain one or more capsules, or other partitions. A sample comprising an analyte to be processed is contained within the droplet. A stimulus is applied to cause release of the contents of the capsule into the droplet, resulting in contact between the reagents and the analyte to be processed. The droplet is incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered. While this example describes an embodiment where a reagent is in a capsule and an analyte is in the droplet, the opposite configuration—i.e., reagent in the droplet and analyte in the capsule—is also possible.
  • The droplets in an emulsion may be of uniform size or heterogeneous size. In some cases, the diameter of a droplet in an emulsion may be about 0.001 μm, 0.01 μm, 0.05 μm, 0.1 μm, 0.5 μm, 1 μm, 5 μm, 10 μm, 50 μm, 100 μm, 150 μm, 200 μm, 300 μm, 400 μm, 500 μm, 600 μm, 700 μm, 800 μm, 900 μm, or 1 mm. A droplet may have a diameter of at least about 0.001 μm, 0.01 μm, 0.05 μm, 0.1 μm, 0.5 μm, 1 μm, 5 μm, 10 μm, 50 μm, 100 μm, 150 μm, 200 μm, 300 μm, 400 μm, 500 μm, 600 μm, 700 μm, 800 μm, 900 μm, or 1 mm. In some cases, a droplet may have a diameter of less than about 0.001 μm, 0.01 μm, 0.05 μm, 0.1 μm, 0.5 μm, 1 μm, 5 μm, 10 μm, 50 μm, 100 μm, 150 μm, 200 μm, 300 μm, 400 μm, 500 μm, 600 μm, 700 μm, 800 μm, 900 μm, or 1 mm. In some cases, a droplet may have a diameter of about 0.001 μm to 1 mm, 0.01 μm to 900 μm, 0.1 μm to 600 μm, 100 μm to 200 μm, 100 μm to 300 μm, 100 μm to 400 μm, 100 μm to 500 μm, 100 μm to 600 μm, 150 μm to 200 μm, 150 μm to 300 μm, or 150 μm to 400 μm.
  • Droplets in an emulsion also may have a particular density. In some cases, the droplets are less dense than an aqueous fluid (e.g., water); in some cases, the droplets are denser than an aqueous fluid. In some cases, the droplets are less dense than a non-aqueous fluid (e.g., oil); in some cases, the droplets are denser than a non-aqueous fluid. Droplets may have a density of about 0.05 g/cm3, 0.1 g/cm3, 0.2 g/cm3, 0.3 g/cm3, 0.4 g/ cm3, 0.5 g/cm3, 0.6 g/cm3, 0.7 g/cm3, 0.8 g/cm3, 0.81 g/cm3, 0.82 g/cm3, 0.83 g/cm3, 0.84 g/cm3, 0.85 g/cm3, 0.86 g/cm3, 0.87 g/cm3, 0.88 g/cm3, 0.89 g/cm3, 0.90 g/cm3, 0.91 g/cm3, 0.92 g/cm3, 0.93 g/cm3, 0.94 g/cm3, 0.95 g/cm3, 0.96 g/cm3, 0.97 g/cm3, 0.98 g/cm3, 0.99 g/cm3, 1.00 g/cm3, 1.05 g/cm3, 1.1 g/cm3, 1.2 g/cm3, 1.3 g/cm3, 1.4 g/cm3, 1.5 g/cm3, 1.6 g/cm3, 1.7 g/cm3, 1.8 g/cm3, 1.9 g/cm3, 2.0 g/cm3, 2.1 g/cm3, 2.2 g/cm3, 2.3 g/cm3, 2.4 g/cm3, or 2.5 g/cm3. Droplets may have a density of at least about 0.05 g/cm3, 0.1 g/cm3, 0.2 g/cm3, 0.3 g/cm3, 0.4 g/ cm3, 0.5 g/cm3, 0.6 g/cm3, 0.7 g/cm3, 0.8 g/cm3, 0.81 g/cm3, 0.82 g/cm3, 0.83 g/cm3, 0.84 g/cm3, 0.85 g/cm3, 0.86 g/cm3, 0.87 g/cm3, 0.88 g/cm3, 0.89 g/cm3, 0.90 g/cm3, 0.91 g/cm3, 0.92 g/cm3, 0.93 g/cm3, 0.94 g/cm3, 0.95 g/cm3, 0.96 g/cm3, 0.97 g/cm3, 0.98 g/cm3, 0.99 g/cm3, 1.00 g/cm3, 1.05 g/cm3, 1.1 g/cm3, 1.2 g/cm3, 1.3 g/cm3, 1.4 g/cm3, 1.5 g/cm3, 1.6 g/cm3, 1.7 g/cm3, 1.8 g/cm3, 1.9 g/cm3, 2.0 g/cm3, 2.1 g/cm3, 2.2 g/cm3, 2.3 g/cm3, 2.4 g/cm3, or 2.5 g/cm3. In other cases, droplet densities may be at most about 0.7 g/cm3, 0.8 g/cm3, 0.81 g/cm3, 0.82 g/cm3, 0.83 g/cm3, 0.84 g/cm3, 0.85 g/cm3, 0.86 g/cm3, 0.87 g/cm3, 0.88 g/cm3, 0.89 g/cm3, 0.90 g/cm3, 0.91 g/cm3, 0.92 g/cm3, 0.93 g/cm3, 0.94 g/cm3, 0.95 g/cm3, 0.96 g/cm3, 0.97 g/cm3, 0.98 g/cm3, 0.99 g/cm3, 1.00 g/cm3, 1.05 g/cm3, 1.1 g/cm3, 1.2 g/cm3, 1.3 g/cm3, 1.4 g/cm3, 1.5 g/cm3, 1.6 g/cm3, 1.7 g/cm3, 1.8 g/cm3, 1.9 g/cm3, 2.0 g/cm3, 2.1 g/cm3, 2.2 g/cm3, 2.3 g/cm3, 2.4 g/cm3, or 2.5 g/cm3. Such densities can reflect the density of the capsule in any particular fluid (e.g., aqueous, water, oil, etc.)
  • Polynucleotides may be partitioned using a variety of methods. For example, polynucleotides may be diluted and dispensed across a plurality of partitions. A terminal dilution of a medium comprising polynucleotides may be performed such that the number of partitions or wells exceeds the number of polynucleotides. The ratio of the number of polynucleotides to the number of partitions may range from about 0.1-10, 0.5-10, 1-10, 2-10, 10-100, 100-1000, or more. The ratio of the number of polynucleotides to the number of partitions may be about 0.1, 0.5, 1, 2, 4, 8, 10, 20, 50, 100, or 1000. The ratio of the number of polynucleotides to the number of partitions may be at least about 0.1, 0.5, 1, 2, 4, 8, 10, 20, 50, 100, or 1000. The ratio of the number of polynucleotides to the number of partitions may be less than about 0.1, 0.5, 1, 2, 4, 8, 10, 20, 50, 100, or 1000.
  • The number of partitions employed may vary depending on the application. For example, the number of partitions may be about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, or 10,000, or more. The number of partitions may be at least about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, or 10,000, or more. The number of partitions may be less than about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, or 10,000.
  • The volume of the partitions may vary depending on the application. For example, the volume of the partitions may be about 1000 μl 900 μl, 800 μl, 700 μl, 600 μl, 500 μl, 400 μl, 300 μl, 200 μl, 100 μl, 50 μl, 25 μl, 10 μl, 5μl, 1 μl, 900 nL, 800 nL, 700 nL, 600 nL, 500 nL, 400 nL, 300 nL, 200 nL, 100 nL, 50 nL, 25 nL, 10 nL, or 5 nL. The volume of the partitions may be at least about 1000 μl, 900 μl, 800 μl, 700 μl, 600 μl, 500 μl, 400 μl, 300 μl, 200 μl, 100 μl, 50 μl, 25 μl, 10 μl, 5 μl, 1 μl, 900 nL, 800 nL, 700 nL, 600 nL, 500 nL, 400 nL, 300 nL, 200 nL, 100 nL, 50 nL, 25 nL, 10 nL, or 5 nL. The volume of the partitions may be less than about 1000 μl, 900 μl, 800 μl, 700 μl, 600 μl, 500 μl, 400 μl, 300 μl, 200 μl, 100 μl, 50 μl, 25 μl, 10 μl, 5 μl, 1 μl, 900 nL, 800 nL, 700 nL, 600 nL, 500 nL, 400 nL, 300 nL, 200 nL, 100 nL, 50 nL, 25 nL, 10 nL, or 5 nL.
  • Species may also be partitioned at a particular density. For example, species may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 100000, or 1000000 species per partition. Species may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 100000, 1000000 or more species per partition. Species may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 100000, or 1000000 species per partition. Species may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, or 100000-1000000 species per partition.
  • Species may be partitioned such that at least one partition comprises a species that is unique within that partition. This may be true for about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more of the partitions. This may be true for at least about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more of the partitions. This may be true for less than about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the partitions.
  • Particular polynucleotides may also be targeted to specific partitions. For example, in some cases, a capture reagent such as an oligonucleotide probe may be immobilized in a partition to capture specific polynucleotides through hybridization.
  • Polynucleotides may also be partitioned at a particular density. For example, polynucleotides may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, or 100000-1000000 polynucleotides per partition. Polynucleotides may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 100000, 1000000 or more polynucleotides per partition. Polynucleotides may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 100000, or 1000000 polynucleotides per partition. Polynucleotides may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 100000, or 1000000 polynucleotides per partition.
  • Polynucleotides may be partitioned such that at least one partition comprises a polynucleotide sequence with a unique sequence compared to all other polynucleotide sequences contained within the same partition. This may be true for about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more of the partitions. This may be true for less than about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more of the partitions. This may be true for more than about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more of the partitions.
  • V. Barcoding
  • Downstream applications, for example DNA sequencing, may rely on the barcodes to identify the origin of a sequence and, for example, to assemble a larger sequence from sequenced fragments. Therefore, it may be desirable to add barcodes to the polynucleotide fragments generated by the methods described herein. Barcodes may be of a variety of different formats, including polynucleotide barcodes. Depending upon the specific application, barcodes may be attached to polynucleotide fragments in a reversible or irreversible manner. Barcodes may also allow for identification and/or quantification of individual polynucleotide fragments during sequencing.
  • Barcodes may be loaded into partitions so that one or more barcodes are introduced into a particular partition. Each partition may contain a different set of barcodes. This may be accomplished by directly dispensing the barcodes into the partitions, enveloping the barcodes (e.g., in a droplet of an emulsion), or by placing the barcodes within a container that is placed in a partition (e.g., a microcapsule).
  • The number of partitions employed may vary depending on the application. For example, the number of partitions may be about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, or 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000, or more. The number of partitions may be at least about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000, or more. The number of partitions may be less than about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000. The number of partitions may be about 5-10000000, 5-5000000, 5-1,000,000, 10-10,000, 10-5,000, 10-1,000, 1,000-6,000, 1,000-5,000, 1,000-4,000, 1,000-3,000, or 1,000-2,000.
  • The number of different barcodes or different sets of barcodes that are partitioned may vary depending upon, for example, the particular barcodes to be partitioned and/or the application. Different sets of barcodes may be, for example, sets of identical barcodes where the identical barcodes differ between each set. Or different sets of barcodes may be, for example, sets of different barcodes, where each set differs in its included barcodes. For example, about 1, 5, 10, 50, 100, 1000, 10000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, 100000000, or more different barcodes or different sets of barcodes may be partitioned. In some examples, at least about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, 100000000, or more different barcodes or different sets of barcodes may be partitioned. In some examples, less than about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 different barcodes or different sets of barcodes may be partitioned. In some examples, about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, 100000-1000000, 10000-1000000, 10000-10000000, or 10000-100000000 barcodes may be partitioned.
  • Barcodes may be partitioned at a particular density. For example, barcodes may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 barcodes per partition. Barcodes may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, 100000000, or more barcodes per partition. Barcodes may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 barcodes per partition. Barcodes may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, 100000-1000000, 10000-1000000, 10000-10000000, or 10000-100000000 barcodes per partition.
  • Barcodes may be partitioned such that identical barcodes are partitioned at a particular density. For example, identical barcodes may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 identical barcodes per partition. Barcodes may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, 100000000, or more identical barcodes per partition. Barcodes may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 identical barcodes per partition. Barcodes may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, 100000-1000000, 10000-1000000, 10000-10000000, or 10000-100000000 identical barcodes per partition.
  • Barcodes may be partitioned such that different barcodes are partitioned at a particular density. For example, different barcodes may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 different barcodes per partition. Barcodes may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, 100000000, or more different barcodes per partition. Barcodes may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 different barcodes per partition. Barcodes may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, 100000-1000000, 10000-1000000, 10000-10000000, or 10000-100000000 different barcodes per partition.
  • The number of partitions employed to partition barcodes may vary, for example, depending on the application and/or the number of different barcodes to be partitioned. For example, the number of partitions employed to partition barcodes may be about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, or 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000 or more. The number of partitions employed to partition barcodes may be at least about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1000000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000 or more. The number of partitions employed to partition barcodes may be less than about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1000000, 2000000, 3000000, 4000000, 5000000, 10000000, or 20000000. The number of partitions employed to partition barcodes may be about 5-10000000, 5-5000000, 5-1,000,000, 10-10,000, 10-5,000, 10-1,000, 1,000-6,000, 1,000-5,000, 1,000-4,000, 1,000-3,000, or 1,000-2,000.As described above, different barcodes or different sets of barcodes (e.g., each set comprising a plurality of identical barcodes or different barcodes) may be partitioned such that each partition comprises a different barcode or different barcode set. In some cases, each partition may comprise a different set of identical barcodes. Where different sets of identical barcodes are partitioned, the number of identical barcodes per partition may vary. For example, about 100,000 or more different sets of identical barcodes may be partitioned across about 100,000 or more different partitions, such that each partition comprises a different set of identical barcodes. In each partition, the number of identical barcodes per set of barcodes may be about 1,000,000 identical barcodes. In some cases, the number of different sets of barcodes may be equal to or substantially equal to the number of partitions. Any suitable number of different barcodes or different barcode sets (including numbers of different barcodes or different barcode sets to be partitioned described elsewhere herein), number of barcodes per partition (including numbers of barcodes per partition described elsewhere herein), and number of partitions (including numbers of partitions described elsewhere herein) may be combined to generate a diverse library of partitioned barcodes with high numbers of barcodes per partition. Thus, as will be appreciated, any of the above-described different numbers of barcodes may be provided with any of the above-described barcode densities per partition, and in any of the above-described numbers of partitions.
  • For example, a population of microcapsules may be prepared such that a first microcapsule in the population comprises multiple copies of identical barcodes (e.g., polynucleotide bar codes, etc.) and a second microcapsule in the population comprises multiple copies of a barcode that differs from the barcode within the first microcapsule. In some cases, the population of microcapsules may comprise multiple microcapsules (e.g., greater than about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 100, 500, 1000, 5000, 10000, 100000, 1000000, 10000000, 100000000, or 1000000000 microcapsules), each containing multiple copies of a barcode that differs from that contained in the other microcapsules. In some cases, the population may comprise greater than about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 100, 500, 1000, 5000, 10000, 100000, 1000000, 10000000, 100000000, or 1000000000 microcapsules with identical sets of barcodes. In some cases, the population may comprise greater than about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 100, 500, 1000, 5000, 10000, 100000, 1000000, 10000000, 100000000, or 1000000000 microcapsules, wherein the microcapsules each comprise a different combination of barcodes. For example, in some cases the different combinations overlap, such that a first microcapsule may comprise, e.g., barcodes A, B, and C, while a second microcapsule may comprise barcodes A, B, and D. In another example, the different combinations do not overlap, such that a first microcapsule may comprise, e.g., barcodes A, B, and C, while a second microcapsule may comprise barcodes D, E, and F. The use of microcapsules is, of course, optional. All of the combinations described above, and throughout this disclosure, may also be generated by dispending barcodes (and other reagents) directly into partitions (e.g., microwells).
  • The barcodes may be loaded into the partitions at an expected or predicted ratio of barcodes per species to be barcoded (e.g., polynucleotide fragment, strand of polynucleotide, cell, etc.). In some cases, the barcodes are loaded into partitions such that more than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, 100, 500, 1000, 5000, 10000, or 200000 barcodes are loaded per species. In some cases, the barcodes are loaded in the partitions so that less than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, 100, 500, 1000, 5000, 10000, or 200000 barcodes are loaded per species. In some cases, the average number of barcodes loaded per species is less than, or greater than, about 0.0001, 0.001, 0.01, 0.1, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, 100, 500, 1000, 5000, 10000, or 200000 barcodes per species.
  • When more than one barcode is present per polynucleotide fragment, such barcodes may be copies of the same barcode, or multiple different barcodes. For example, the attachment process may be designed to attach multiple identical barcodes to a single polynucleotide fragment, or multiple different barcodes to the polynucleotide fragment.
  • The methods provided herein may comprise loading a partition (e.g., a microwell, droplet of an emulsion) with the reagents necessary for the attachment of barcodes to polynucleotide fragments. In the case of ligation reactions, reagents including restriction enzymes, ligase enzymes, buffers, adapters, barcodes and the like may be loaded into a partition. In the case barcoding by amplification, reagents including primers, DNA polymerases, DNTPs, buffers, barcodes and the like may be loaded into a partition. As described throughout this disclosure, these reagents may be loaded directly into the partition, or via a container such as a microcapsule. If the reagents are not disposed within a container, they may be loaded into a partition (e.g., a microwell) which may then be sealed with a wax or oil until the reagents are used.
  • Barcodes may be ligated to a polynucleotide fragment using sticky or blunt ends. Barcoded polynucleotide fragments may also be generated by amplifying a polynucleotide fragment with primers comprising barcodes.
  • Barcodes may be assembled combinatorially, from smaller components designed to assemble in a modular format. For example, three modules, 1A, 1B, and 1C may be combinatorially assembled to produce barcode 1ABC. Such combinatorial assembly may significantly reduce the cost of synthesizing a plurality of barcodes. For example, a combinatorial system consisting of 3 A modules, 3 B modules, and 3 C modules may generate 3*3*3=27 possible barcode sequences from only 9 modules.
  • VI. Microcapsules and Microwell Capsule Arrays
  • Microcapsules and microwell capsule array (MCA) devices may be used to perform the polynucleotide processing methods described herein. MCA devices are devices with a plurality of microwells. Microcapsules are introduced into these microwells, before, after, or concurrently with the introduction of a sample.
  • Microwells may comprise free reagents and/or reagents encapsulated in microcapsules. Any of the reagents described in this disclosure may be encapsulated in a microcapsule, including any chemicals, particles, and elements suitable for sample processing reactions involving a polynucleotide. For example, a microcapsule 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, dNTPs, ddNTPs and the like.
  • Additional exemplary reagents include: 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, mtDNA (mitochondrial DNA), mRNA, rRNA, tRNA, nRNA, siRNA, snRNA, snoRNA, scaRNA, microRNA, dsRNA, ribozyme, riboswitch and viral RNA, polymerase, ligase, restriction enzymes, proteases, nucleases, protease inhibitors, nuclease inhibitors, chelating agents, reducing agents, oxidizing agents, fluorophores, probes, chromophores, dyes, organics, emulsifiers, surfactants, stabilizers, polymers, water, small molecules, pharmaceuticals, radioactive molecules, preservatives, antibiotics, aptamers, and pharmaceutical drug compounds.
  • In some cases, a microcapsule comprises a set of reagents that have a similar attribute (e.g., a set of enzymes, a set of minerals, a set of oligonucleotides, a mixture of different bar-codes, a mixture of identical bar-codes). In other cases, a microcapsule comprises a heterogeneous mixture of reagents. In some cases, the heterogeneous mixture of reagents comprises all components necessary to perform a reaction. In some cases, such mixture comprises all components necessary to perform a reaction, except for 1, 2, 3, 4, 5, or more components necessary to perform a reaction. In some cases, such additional components are contained within a different microcapsule or within a solution within a partition (e.g., microwell) of the device.
  • In some cases, only microcapsules comprising reagents are introduced. In other cases, both free reagents and reagents encapsulated in microcapsules are loaded into the device, either sequentially or concurrently. In some cases, reagents are introduced to the device either before or after a particular step. In some cases, reagents and/or microcapsules comprising reagents are introduced sequentially such that different reactions or operations occur at different steps. The reagents (or microcapsules) may be also be loaded at steps interspersed with a reaction or operation step. For example, microcapsules comprising reagents for fragmenting polynucleotides (e.g., restriction enzymes) may be loaded into the device, followed by loading of microcapsules comprising reagents for ligating bar-codes and subsequent ligation of the bar-codes to the fragmented molecules.
  • Microcapsules may be pre-formed and filled with reagents by injection. For example, the picoinjection methods described in Abate et al. (Proc. Natl. Acad. Sci. U.S.A., 2010, 107(45), 19163-19166) and Weitz et al. (U.S. Pub. No. 2012/0132288) may be used to introduce reagents into the interior of microcapsules described herein. These methods can also be used to introduce a plurality of any of the reagents described herein into microcapsules.
  • Microcapsules may be formed by any emulsion technique known in the art. For example, the multiple emulsion technique of Weitz et al. (U.S. Pub. No. 2012/0211084) may be used to form microcapsules (or partitions) for use with the methods disclosed herein.
  • Numerous chemical triggers may be used to trigger the disruption of partitions (e.g., Plunkett et al., Biomacromolecules, 2005, 6:632-637). Examples of these chemical changes may include, but are not limited to pH-mediated changes to the integrity of a component of a partition, disintegration of a component of a partition via chemical cleavage of crosslink bonds, and triggered depolymerization of a component of a partition. Bulk changes may also be used to trigger disruption of partitions.
  • A change in pH of a solution, such as a decrease in pH, may trigger disruption of a partition via a number of different mechanisms. The addition of acid may cause degradation or disassembly a portion of a partition through a variety of mechanisms. Addition of protons may disassemble cross-linking of polymers in a component of a partition, disrupt ionic or hydrogen bonds in a component of a partition, or create nanopores in a component of a partition to allow the inner contents to leak through to the exterior. A change in pH may also destabilize an emulsion, leading to release of the contents of the droplets.
  • In some examples, a partition is produced from materials that comprise acid-degradable chemical cross-linkers, such a ketals. A decrease in pH, particular to a pH lower than 5, may induce the ketal to convert to a ketone and two alcohols and facilitate disruption of the partition. In other examples, the partitions may be produced from materials comprising one or more polyelectrolytes that are pH sensitive. A decrease in pH may disrupt the ionic- or hydrogen-bonding interactions of such partitions, or create nanopores therein. In some cases, partitions made from materials comprising polyelectrolytes comprise a charged, gel-based core that expands and contracts upon a change of pH.
  • Disruption of cross-linked materials comprising a partition can be accomplished through a number of mechanisms. In some examples, a partition can be contacted with various chemicals that induce oxidation, reduction or other chemical changes. In some cases, a reducing agent, such as beta-mercaptoethanol, can be used, such that disulfide bonds of a partition are disrupted. In addition, enzymes may be added to cleave peptide bonds in materials forming a partition, thereby resulting in a loss of integrity of the partition.
  • Depolymerization can also be used to disrupt partitions. A chemical trigger may be added to facilitate the removal of a protecting head group. For example, the trigger may cause removal of a head group of a carbonate ester or carbamate within a polymer, which in turn causes depolymerization and release of species from the inside of a partition.
  • In yet another example, a chemical trigger may comprise an osmotic trigger, whereby a change in ion or solute concentration in a solution induces swelling of a material used to make a partition. Swelling may cause a buildup of internal pressure such that a partition ruptures to release its contents. Swelling may also cause an increase in the pore size of the material, allowing species contained within the partition to diffuse out, and vice versa.
  • A partition may also be made to release its contents via bulk or physical changes, such as pressure induced rupture, melting, or changes in porosity.
  • VII. Polynucleotide Sequencing
  • Generally, the methods and compositions provided herein are useful for preparation of polynucleotide fragments for downstream applications such as sequencing. Sequencing may be performed by any available technique. For example, sequencing may be performed by the classic Sanger sequencing method. Sequencing methods may also include: high-throughput sequencing, pyrosequencing, sequencing-by-synthesis, single-molecule sequencing, nanopore sequencing, sequencing-by-ligation, sequencing-by-hybridization, RNA-Seq (Illumina), Digital Gene Expression (Helicon), next generation sequencing, single molecule sequencing by synthesis (SMSS) (Helicos), massively-parallel sequencing, clonal single molecule Array (Solexa), shotgun sequencing, Maxim-Gilbert sequencing, primer walking, and any other sequencing methods known in the art.
  • In some cases varying numbers of fragments are sequenced. For example, in some cases about 30%-90% of the fragments are sequenced. In some cases, about 35%-85%, 40%-80%, 45%-75%, 50%-70%, 55%-65%, or 50%-60% of the fragments are sequenced. In some cases, at least about 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the fragments are sequenced. In some cases less than about 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the fragments are sequenced.
  • In some cases sequences from fragments are assembled to provide sequence information for a contiguous region of the original target polynucleotide that is longer than the individual sequence reads. Individual sequence reads may be about 10-50, 50-100, 100-200, 200-300, 300-400, or more nucleotides in length.
  • The identities of the barcode tags may serve to order the sequence reads from individual fragments as well as to differentiate between haplotypes. For example, during the partitioning of individual fragments, parental polynucleotide fragments may separated into different partitions. With an increase in the number of partitions, the likelihood of a fragment from both a maternal and paternal haplotype contained in the same partition becomes negligibly small. Thus, sequence reads from fragments in the same partition may be assembled and ordered.
  • VIII. Polynucleotide Phasing
  • This disclosure also provides methods and compositions to prepare polynucleotide fragments in such a manner that may enable phasing or linkage information to be generated. Such information may allow for the detection of linked genetic variations in sequences, including genetic variations (e.g., SNPs, mutations, indels, copy number variations, transversions, translocations, inversions, etc.) that are separated by long stretches of polynucleotides. The term “indel” refers to a mutation resulting in a colocalized insertion and deletion and a net gain or loss in nucleotides. A “microindel” is an indel that results in a net gain or loss of 1 to 50 nucleotides. These variations may exist in either a cis or trans relationship. In a cis relationship, two or more genetic variations exist in the same polynucleotide or strand. In a trans relationship, two or more genetic variations exist on multiple polynucleotide molecules or strands.
  • Methods provided herein may be used to determine polynucleotide phasing. For example, a polynucleotide sample (e.g., a polynucleotide that spans a given locus or loci) may be partitioned such that at most one molecule of polynucleotide is present per partition (e.g., microwell). The polynucleotide may then be fragmented, barcoded, and sequenced. The sequences may be examined for genetic variation. The detection of genetic variations in the same sequence tagged with two different bar codes may indicate that the two genetic variations are derived from two separate strands of DNA, reflecting a trans relationship. Conversely, the detection of two different genetic variations tagged with the same bar codes may indicate that the two genetic variations are from the same strand of DNA, reflecting a cis relationship.
  • Phase information may be important for the characterization of a polynucleotide fragment, particularly if the polynucleotide fragment is derived from a subject at risk of, having, or suspected of a having a particular disease or disorder (e.g., hereditary recessive disease such as cystic fibrosis, cancer, etc.). The information may be able to distinguish between the following possibilities: (1) two genetic variations within the same gene on the same strand of DNA and (2) two genetic variations within the same gene but located on separate strands of DNA. Possibility (1) may indicate that one copy of the gene is normal and the individual is free of the disease, while possibility (2) may indicate that the individual has or will develop the disease, particularly if the two genetic variations are damaging to the function of the gene when present within the same gene copy. Similarly, the phasing information may also be able to distinguish between the following possibilities: (1) two genetic variations, each within a different gene on the same strand of DNA and (2) two genetic variations, each within a different gene but located on separate strands of DNA.
  • IX. Sequencing Polynucleotides from Small Numbers of Cells
  • Methods provided herein may also be used to prepare polynucleotide contained within cells in a manner that enables cell-specific information to be obtained. The methods enable detection of genetic variations (e.g., SNPs, mutations, indels, copy number variations, transversions, translocations, inversions, etc.) from very small samples, such as from samples comprising about 10-100 cells. In some cases, about 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 cells may be used in the methods described herein. In some cases, at least about 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 cells may be used in the methods described herein. In other cases, at most about 5, 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 cells may be used in the methods described herein.
  • In an example, a method comprises partitioning a cellular sample (or crude cell extract) such that at most one cell (or extract of one cell) is present per partition, lysing the cells, fragmenting the polynucleotides contained within the cells by any of the methods described herein, attaching the fragmented polynucleotides to barcodes, pooling, and sequencing.
  • As described elsewhere herein, the barcodes and other reagents may be contained within a microcapsule. These microcapsules may be loaded into a partition (e.g., a microwell) before, after, or concurrently with the loading of the cell, such that each cell is contacted with a different microcapsule. This technique may be used to attach a unique barcode to polynucleotides obtained from each cell. The resulting tagged polynucleotides may then be pooled and sequenced, and the barcodes may be used to trace the origin of the polynucleotides. For example, polynucleotides with identical barcodes may be determined to originate from the same cell, while polynucleotides with different barcodes may be determined to originate from different cells.
  • The methods described herein may be used to detect the distribution of oncogenic mutations across a population of cancerous tumor cells. For example, some tumor cells may have a mutation, or amplification, of an oncogene (e.g., HER2, BRAF, EGFR, KRAS) in both alleles (homozygous), others may have a mutation in one allele (heterozygous), and still others may have no mutation (wild-type). The methods described herein may be used to detect these differences, and also to quantify the relative numbers of homozygous, heterozygous, and wild-type cells. Such information may be used, for example, to stage a particular cancer and/or to monitor the progression of the cancer and its treatment over time.
  • In some examples, this disclosure provides methods of identifying mutations in two different oncogenes (e.g., KRAS and EGFR). If the same cell comprises genes with both mutations, this may indicate a more aggressive form of cancer. In contrast, if the mutations are located in two different cells, this may indicate that the cancer is more benign, or less advanced.
  • X. Analysis of Gene Expression
  • Methods of the disclosure may be applicable to processing samples for the detection of changes in gene expression. A sample may comprise a cell, mRNA, or cDNA reverse transcribed from mRNA. The sample may be a pooled sample, comprising extracts from several different cells or tissues, or a sample comprising extracts from a single cell or tissue.
  • Cells may be placed directly into an partition (e.g., a microwell) and lysed. After lysis, the methods of the invention may be used to fragment and barcode the polynucleotides of the cell for sequencing. Polynucleotides may also be extracted from cells prior to introducing them into a partition used in a method of the invention. Reverse transcription of mRNA may be performed in a partition described herein, or outside of such a partition. Sequencing cDNA may provide an indication of the abundance of a particular transcript in a particular cell over time, or after exposure to a particular condition.
  • The methods presented throughout this disclosure provide several advantages over current polynucleotide processing methods. First, inter-operator variability is greatly reduced. Second, the methods may be carried out in microfluidic devices, which have a low cost and can be easily fabricated. Third, the controlled fragmentation of the target polynucleotides allows the user to produce polynucleotide fragments with a defined and appropriate length. This aids in partitioning the polynucleotides and also reduces the amount of sequence information loss due to the present of overly-large fragments. The methods and systems also provide a facile workflow that maintains the integrity of the processed polynucleotide. Additionally, the use of restriction enzymes enables the user to create DNA overhangs (“sticky ends”) that may be designed for compatibility with adapters and/or barcodes.
  • EXAMPLES Example 1 Generation of Non-Overlapping DNA Fragments for Sequencing
  • This example demonstrates a method for the generation of non-overlapping DNA fragments suitable for DNA sequencing and other downstream applications. An implementation of this method is schematically illustrated in FIG. 2.
  • With reference to FIG. 2, a target polynucleotide 101, genomic DNA, is fragmented with the enzyme Notl, to generate a plurality of non-overlapping first polynucleotide fragments 102. The first polynucleotide fragments are partitioned into separate microwells 103 in a microdevice such that each microwell comprises a plurality of fragments, but only a single fragment with a particular sequence 104. The left-hand side of FIG. 2 illustrates three microwells (one is labeled 103), each containing three exemplary unique fragments 104, corresponding to the first polynucleotide fragments 102. Referring again to the left-hand side of FIG. 2, the left-most well contains fragments A1, B2, and C3, the middle well contains fragments B1, A2, and A3, and the right-most well contains fragments C1, C2, and B3.
  • The partitioned fragments are then further fragmented, to generate a plurality of non-overlapping second polynucleotide fragments 105. Referring again to the left-hand side of FIG. 2, each member of the second polynucleotide fragments is designated by its first fragment identifier (e.g., A1, B2, etc.), followed by a “−1” or a “−2”. For example, first fragment A1 is fragmented to produce second fragments A1-1 and A1-2. First fragment B2 is fragmented to produce second fragments B2-1 and B2-2, and so on. For the sake of simplicity, only two second fragments are shown for each first fragment. This is, of course, not meant to be limiting, as any number of fragments may be generated at any step of the process.
  • The second set polynucleotide fragments are barcoded, and the barcoded sequences are pooled. Referring to the lower left-hand side of FIG. 2, the labels [1], [2], and [3] represent three different barcode sequences used to label the second fragments 105. The labeled sequences are designated 106. Optionally, adapter sequences (not shown) are used to make the second fragments 105 compatible for ligation with the barcodes. The barcoding is performed while the fragments are still partitioned, before pooling. The pooled barcoded sequences are then sequenced.
  • With continued reference to FIG. 2, the methods described above are then repeated, using a second rare cutter enzyme, XmaIII to digest the genomic DNA and generate a plurality of non-overlapping third polynucleotide fragments 107. The third polynucleotide fragments and the first polynucleotide fragments are overlapping, because they are generated with different rare-cutter enzymes that cut the target polynucleotides at different sites. The third polynucleotide fragments are partitioned into separate microwells 108 in a microdevice such that each microwell comprises a plurality of fragments, but only a single fragment with a particular sequence 109. The right-hand side of FIG. 2 illustrates three microwells (one is labeled 108), each containing three exemplary unique fragments 109, corresponding to the third polynucleotide fragments 107. Referring again to the right-hand side of FIG. 2, the left-most well contains fragments D1, E2, and F3, the middle well contains fragments E1, D2, and D3, and the right-most well contains fragments F1, F2, and E3.
  • With continued reference to FIG. 2, The partitioned fragments are then further fragmented, to generate a plurality of non-overlapping fourth polynucleotide fragments 110. The fourth polynucleotide fragments and the second polynucleotide fragments are overlapping, because they are generated by fragmenting the third and first fragments, respectively, which were generated with rare-cutter enzymes that cut the target polynucleotide at different sites, as described above. Referring again to the right-hand side of FIG. 2, each member of the fourth set of polynucleotide fragments is designated by its third fragment identifier (e.g., D1, E2, etc.), followed by a “−1” or a “−2”. For example, third fragment D1 is fragmented to produce fourth fragments D1-1 and D1-2. Third fragment E2 is fragmented to produce fourth fragments E2-1 and E2-2, and so on. For the sake of simplicity, only two fourth fragments are shown for each third fragment. This is, of course, not meant to be limiting, as any number of fragments may be generated.
  • The fourth polynucleotides fragments are barcoded, and the barcoded sequences are pooled. Referring to the lower right-hand side of FIG. 2, the numbers [4], [5], and [6] represent three different barcode sequences used to label the fourth fragments 110. The labeled sequences are designated 111. Optionally, adapter sequences (not shown) are used to make the fourth fragments 110 compatible for ligation with the barcodes. The barcoding is performed while the fragments are still partitioned, before pooling. The pooled barcoded sequences are then sequenced.
  • The example above describes sequencing the barcoded second fragments separately from the barcoded fourth fragments. The barcoded second fragments and the barcoded fourth fragments may also be combined, and the combined sample may be sequenced. One or more steps of the process may be carried out in a device. The steps carried out in a device may be carried out in the same device or in different devices.
  • After sequencing, sequence contigs are assembled and the overlapping sequences between the second fragments and the fourth fragments are used to assemble the sequence of the genome.
  • Example 2 Pseudo-Random Fragmentation of Polynucleotides
  • A simulation was performed to evaluate the size distribution of fragments generated by a 6 Mer cutter (StuI), a 4 Mer cutter (CviQI), and two to seven 4 Mer cutters. Random 1 Mbp DNA sequences were generated in silico and cuts were simulated based on the occurrence of the recognition sites for each of the restriction enzymes within the random sequences.
  • FIG. 3 shows the size distribution of a random 1 Mbp DNA sequence cut with the 6 Mer cutter StuI (AGG/CCT). Fragments less than about 50 nucleotides were designated as “low yield,” because they underutilize the read length capacity of sequencing instruments. Fragments less than about 200 nucleotides were designated as fragments likely to provide the most accurate data from today's sequencing technology. As described throughout this disclosure, this size range is in no way meant to be limiting, and the methods exemplified here, and described throughout this disclosure, may be used to generate fragments of any size range. Fragments from about 200 to about 400 nucleotides typically produce sequence data with systematic error for bases more than 100 bases from either fragment end. Fragments of more than about 400 nucleotides typically do not produce any useful sequence information for bases further than 200 bases from a fragment end, using today's sequencing technologies. However, this is expected to change, and the methods presented herein can be used to generate sequences of this size or larger.
  • As shown in FIG. 3, 3 of 271 fragments (1.5%) were considered low yield since they were 50 bases or smaller. Fourteen fragments (5%) were considered high accuracy since they were 200 bases or smaller (i.e., each base of the fragment is within 100 bases of a restriction site and could be sequenced with high accuracy). Eleven fragments (4%) were between 200 and 400 bases and would generate data that is both accurate (0-100 bases from each end) and inaccurate (100-200 bases from each end). The remaining 246 fragments (91%) were greater than 400 bases and would generate accurate (0-100), inaccurate (100-200) and no (>200 bases from a restriction site) sequence data. Overall only 5% of the 1 Mbp random sequence was within 100 bases from a restriction site and would generate accurate sequence data.
  • FIG. 4 shows the results from a second simulation using the 4 Mer cutter CviQI (G/TAC), instead of StuI (the 6 Mer cutter described above) to simulate cutting a random 1 Mbp DNA sequence. As shown in FIG. 4, the use of a restriction enzyme with a shorter recognition site results in more cuts, and the size distribution of the fragments is therefore shifted toward a smaller size range. In particular, as shown in FIG. 4, 18% of fragments were considered low yield since they were 50 bases or smaller. Thirty-eight percent of fragments were considered high accuracy since they were 200 bases or smaller (i.e., each base of the fragment was within 100 bases of a restriction site and could be sequenced with high accuracy). Twenty five percent of fragments were between 200 and 400 bases and would generate data that is both accurate (0-100 bases from each end) and inaccurate (100-200 bases from each end). The remaining fragments (37%) were greater than 400 bases and would generate accurate (0-100), inaccurate (100-200) and no (>200 bases from a restriction site) sequence data. Overall 56% of the 1 Mbp random sequence was within 100 bases from a restriction site and would generate accurate sequence data. Therefore, cutting the randomly generated 1 Mbp DNA sequence with CviQI resulted in a higher percentage of fragments with nucleotides within 100 nucleotides of a restriction site than cutting with StuI (i.e., 56% vs. 5%, respectively). Cutting with CviQI is therefore expected to provide more fragments that may be fully sequenced.
  • Next, simulated cuts were made in a random 1 Mbp DNA sequence using combinations of one to seven different 4 Mer cutters. The 4 Mer cutters were: (A) CviQI (G/TAC); (B) BfaI (C/TAG); (C) HinP1I (G/CGC); (D) CviAII (C/ATG); (E) TaqαI (T/CGA); (F) MseI (T/TAA); and (G) MspI (C/CGG). The results of these simulations are shown in FIG. 5. As shown in FIG. 5, increasing the number of 4 Mer cutter enzymes, from one to seven, increases the number of fragments with nucleotides within 100 nucleotides of a restriction site. Therefore, cutting the randomly generated 1 Mbp DNA sequence with more than one 4 Mer cutter results in more fragments that may be fully sequenced than cutting with a single 4 Mer cutter.
  • The number of enzymes used to cut a sequence can be chosen so that a particular fraction of a target nucleotide (e.g., a genomic) sequence within 100 nucleotides of a restriction enzyme is achieved. For example, the fraction of a random genome within 100 nucleotides of a restriction site for a 4 Mer cutter is equal to 1-0.44x, where x is the number of independent 4 Mer cutters. Similarly, the fraction of a random genome within 100 nucleotides of a restriction site for a 5 Mer cutter is equal to 1-0.25x, where x is the number of independent 5 Mer cutters. For a 6 Mer cutter, the fraction of a random genome within 100 nucleotides of a restriction site is equal to 1-0.95x, where x is the number of independent 6 Mer cutters.
  • Table 1 shows the percentage of sequences with a length greater than 100 nucleotides for each of the seven enzymatic treatments described above. These sequences are considered those likely to result in missing data. Increasing the number of enzymes decreases the percentage of sequences greater than 100 nucleotides. The number of enzymes and their restriction site recognition length may be chosen in order to minimize the loss of sequence information from sequences greater than 100 nucleotides from a restriction site while also minimizing the generation of sequences less than 50 nucleotides, which are undesirable because the underutilize the read length capacity of sequencing instruments. The presence of these fragments may be minimized or avoided by selecting restriction enzymes that cut more rarely but at the potential price of reduced sequencing coverage of the DNA (i.e., more fragments may have bases >100 bases from a restriction site). These fragments may also be physically removed by a size selection step. Since these fragments are small and some fraction of the bases represented in the small fragments may be covered in larger fragments from other enzymes, the effect on coverage would likely be minimal.
  • The exemplary 4 Mer cutter methods presented herein are optimized to provide fragments compatible with current DNA sequencing technology, which may achieve accurate read lengths up to about 100 nucleotides from the terminus of a fragment. One of ordinary skill in the art will readily recognize that other restriction enzymes (e.g., 5 Mer cutters, 6 Mer cutters, etc.) would be suitable for DNA sequencing technologies capable of accurately reading larger fragments of DNA (e.g., 300-400, or more nucleotides). The methods presented in this disclosure are, of course generalizable, and may be used to obtain DNA fragments of any size distribution compatible with present or future sequencing technology.
  • TABLE 1
    Percentage of random 1Mbp sequence more than 100
    nucleotides from any restriction site. The letters in the first
    row refer to treatment with the following enzymes: (A) CviQI
    (G/TAC); (B) BfaI (C/TAG); (C) HinP1I (G/CGC); (D) CviAII
    (C/ATG); (E) TaqαI (T/CGA); (F) MseI (T/TAA);
    and (G) MspI (C/CGG).
    A AB ABC ABCD ABCDE ABCDEF ABCDEFG
    44.2% 20.1% 9.3% 4.2% 1.7% 0.6% 0.3%
  • Example 3 High Yield Adapter Ligation by Restriction Enzyme-Mediated Recycling of Undesirable Side Products
  • As described elsewhere herein, many downstream applications of the polynucleotide processing methods provided herein may utilize polynucleotide barcodes. An adapter may be used to provide compatible ends for the attachment of a barcode to a polynucleotide fragment (e.g., by ligation or PCR). In these cases, the desired products may be, for example:

  • [B]-[TPF]-[B], or

  • [B]-[A]-[TPF]-[A]-[B], where
  • [B] represents a barcode, [A] represents an adapter, and [TPF] represents a target polynucleotide fragment. However, in some cases, undesirable side products may form, for example, from the self ligation of barcodes, adapters, and/or target polynucleotide fragments. This example demonstrates one solution to this potential problem.
  • FIG. 6 shows a schematic of an implementation of the method described in this example. In the example shown in FIG. 6, three polynucleotide starting materials (Genomic DNA; Adapter 1; and Adapter 2) and three enzymes (MspI; NarI; and DNA Ligase) are contained within a partition. The restriction enzyme MspI (C/CGG) recognizes the CCGG sequence occurring within the Genomic DNA sequence and cuts the Genomic DNA sequence to generate a fragment of genomic DNA. If the reaction proceeds as intended, the fragment of genomic DNA is then ligated to Adapter 1 and Adapter 2, to generate a fragment of genomic DNA flanked by ligated adapters (FIG. 6, lower-left). This fragment with ligated adapters may then be ligated to DNA barcodes, which may also be present within the same partition (not shown).
  • However, the reaction described above may also result in several unwanted side products, including multimers produced by self-ligation of the fragmented genomic DNA and adapters (or other molecules, such as barcodes, which are not shown). For the sake of simplicity, FIG. 6 illustrates this concept by showing only self-ligation of fragmented genomic DNA and adapters.
  • One unwanted side product is a multimer of genomic DNA fragments. This may occur, for example, if genomic DNA fragments with compatible ends are ligated to each other after cutting. In FIG. 6, cutting of Genomic DNA with MspI generates compatible ends that may be ligated by the ligase present in the partition. Similarly, Adapter 1 and Adapter 2, as shown, have compatible ligatable ends, and may also be ligated to form multimers.
  • As indicated in FIG. 6, one solution to this problem is to pair one enzyme (in this example, MspI) with a second enzyme (in this example, NarI). In this example, MspI re-cuts genomic DNA multimers produced by self-ligation of genomic DNA fragments. Therefore, MspI recycles unwanted genomic DNA fragment multimers back into genomic DNA fragments, which may then be correctly ligated to the adapters. Similarly, NarI cuts multimers of Adapter 1 and Adapter 2 into monomers of Adapter 1 and monomers of Adapter 2, which may then be correctly ligated to genomic DNA fragments. This recycles unwanted adapter multimers back into the desired starting materials of Adapter 1 and Adapter 2.
  • The enzymes are chosen such that the desired product (i.e., the genomic DNA fragment with adapters on each end) does not contain a recognition site for either enzyme. Therefore, the product will not be re-cut by any enzyme contained within the partition. This process increases the yield of the desired product, while minimizing the number of unwanted side products and reducing the amount of starting material required to produce a desired amount of a product. As described in this disclosure, a pair of enzymes may be chosen so that one enzyme recognizes one undesirable side-product and regenerates a starting material and another recognizes another undesirable side product and regenerates another starting material, but neither enzyme recognizes the desired product. This can be done for an unlimited number of side products.
  • In general, one strategy for selecting such pairs is to choose two enzymes that create identical (or similar, ligatable) termini after cutting, but have recognition sequences of different lengths. FIG. 7 shows examples of such pairs of enzymes. The enzymes provided in FIG. 7A provide sticky ends, while those provided in FIG. 7B provide blunt ends.
  • The exemplary embodiment shown in FIG. 6 uses Genomic DNA and two adapters (Adapter 1 and Adapter 2) as starting materials. Therefore, in this embodiment, MspI is used not only to regenerate genomic DNA fragments after self-ligation, but also to generate the genomic DNA fragments in the first place, from Genomic DNA. Of course, this is optional, as one may introduce pre-fragmented genomic DNA into the partition and the method is still applicable.
  • Similarly, the embodiment shown in FIG. 6 shows two separate adapter molecules as starting materials. Adapter molecules may also be provided as a single polynucleotide sequence which is then cut by an enzyme contained within the partition (in this example, NarI) to generate ligation compatible ends for attachment to the fragmented genomic DNA. The method is also applicable to other polynucleotides described throughout this disclosure and to methods of attachment based on techniques other than ligation (e.g., attachment of an adapter or a barcode by PCR).
  • Pseudo-complimentary nucleotides that preferentially bind natural nucleotides over themselves (e.g., Biochemistry (1996) 35, 11170-11176; Nucleic Acids Research (1996) 15, 2470-2475), may also be used to minimize or avoid the formation of certain multimers, for example adapter-adapter multimers and barcode-barcode multimers. If adapters and/or barcodes (and/or other polynucleotides are synthesized using pseudo-complimentary nucleotides, they will prefer to hybridize with naturally occurring polynucleotide fragments (e.g., genomic DNA fragments) rather than themselves, therefore leading to a higher yield of the desired product.
  • Example 4 Provision of Reagents in Microcapsules and Directly in Microwells
  • As described throughout this disclosure, the polynucleotide processing methods described herein may involve the treatment of partitioned polynucleotides with a variety of reagents. These reagents may include, for example, restriction enzymes, ligases, phosphatases, kinases, barcodes, adapters, or any other reagent useful in polynucleotide processing or in a downstream application, such as sequencing. FIG. 8 shows two exemplary methods of providing reagents. On the left-hand side of FIG. 8, reagents are provided within a microcapsule. The microcapsule that is shown in FIG. 8 has an outer shell (“3”), an intermediate non-aqueous layer (“2”) and an inner aqueous drop contained within the intermediate non-aqueous layer (“1ABC+RE”). This droplet is made by a water-oil-water emulsion technique followed by polymerization of the outermost water layer (“3”) to form a shell. Reagents are contained within the inner aqueous phase of the capsule. The left-hand side of FIG. 8 shows an exemplary embodiment with four reagents contained within the aqueous phase of the capsule, namely three barcode reagents (1A, 1B, and 1C), and a restriction enzyme (“RE”). The embodiment shown is merely exemplary. The reagents may be located in any part of the capsule.
  • The capsule is dispensed into a partition (e.g., a microwell). A target polynucleotide and a ligase are then added to the partition. The capsule is made to release its contents by exposure to a stimulus, such as a change in temperature, a solvent, or stirring. The restriction enzyme fragments the target polynucleotide and the ligase attaches the barcode reagents to the target polynucleotide fragments generated by the restriction enzyme.
  • The restriction digestion and ligation may proceed according to any of the methods described herein, for example by non-overlapping fragmentation techniques, by pseudo-random fragmentation methods, and/or by pairing of restriction enzymes to recycle unwanted side products into new starting products (e.g., target polynucleotide fragments and barcodes). Adapters may also be included within the microcapsule. The barcodes shown in FIG. 8 are modular. For example, barcode components 1A, 1B, and 1C may ligate to form barcode: [1A]-[1B]-[1C].
  • The right-hand side of FIG. 8 shows the same reagents dispensed into a microwell, followed by sealing with sealant (e.g., a wax or oil), to prevent evaporation before use. This approach may be substituted for the approach described above, where the reagents are placed within microcapsules. Both approaches are used to produce partitions (e.g., microwells) pre-loaded with reagents for DNA fragmentation and barcoding. In order to fragment and barcode DNA using reagents dispensed within a microwell, a user unseals a partition, and introduces a target polynucleotide and a ligase (or any other reagents applicable for the method the user is conducting). As described above, the restriction enzyme fragments the target polynucleotide and the ligase attaches the barcode reagents to the target polynucleotide fragments generated by the restriction enzyme. Of course, both approaches may be combined by placing certain reagents in the microwell and others in the microcapsule. While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.

Claims (20)

What is claimed is:
1. A method for nucleic acid preparation or analysis, comprising:
(a) partitioning a plurality of barcode molecules and a plurality of nucleic acid molecules derived from a cell-free biological sample in a plurality of partitions, wherein said plurality of barcode molecules comprise a plurality of barcode sequences;
(b) in said plurality of partitions, using said plurality of nucleic acid molecules and said plurality of barcode molecules to generate a plurality of barcoded nucleic acid molecules, which plurality of barcoded nucleic acid molecules comprises said plurality of barcode sequences;
(c) recovering or releasing said plurality of barcoded nucleic acid molecules from said plurality of partitions.
2. The method of claim 1, wherein a barcode sequence associated with a given partition of said plurality of partitions is different from a barcode sequence associated with other partitions of said plurality of partitions.
3. The method of claim 1, wherein said plurality of partitions is a plurality of droplets.
4. The method of claim 1, wherein said plurality of partitions is a plurality of wells.
5. The method of claim 1, wherein said plurality of barcode molecules are coupled to beads.
6. The method of claim 5, wherein said beads are solid particles.
7. The method of claim 5, wherein said beads are gel beads.
8. The method of claim 5, further comprising releasing said plurality of barcode molecules from said beads in said plurality of partitions.
9. The method of claim 5, wherein said plurality of barcode molecules is covalently attached to said beads through linkers.
10. The method of claim 1, wherein said plurality of nucleic acid molecules are ribonucleic acid molecules.
11. The method of claim 1, wherein said plurality of nucleic acid molecules are deoxyribonucleic acid molecules.
12. The method of claim 1, wherein said cell-free biological sample is a bodily fluid sample.
13. The method of claim 12, wherein said bodily fluid sample is blood.
14. The method of claim 12, wherein said bodily fluid sample is serum.
15. The method of claim 12, wherein said bodily fluid sample is saliva.
16. The method of claim 12, wherein said bodily fluid sample is urine.
17. The method of claim 1, wherein in (c), said plurality of barcoded nucleic acid molecules is generated by nucleic acid amplification using said plurality of nucleic acid molecules and said plurality of barcode molecules.
18. The method of claim 1, wherein a partition of said plurality of partitions comprises barcode molecules from said plurality of barcode molecules, and wherein said barcode molecules comprise a common barcode sequence.
19. The method of claim 1, further comprising, subsequent to (c), sequencing said plurality of barcoded nucleic acid molecules or derivatives thereof to generate a plurality of sequences, which plurality of sequences comprise sequences of said plurality of nucleic acid molecules derived from said cell-free biological sample and said plurality of barcode sequences.
20. The method of claim 19, further comprising using said plurality of sequences to differentiate between haplotypes in said plurality of nucleic acid molecules derived from said cell-free biological sample.
US16/000,803 2012-08-14 2018-06-05 Methods and systems for processing polynucleotides Abandoned US20180282804A1 (en)

Priority Applications (20)

Application Number Priority Date Filing Date Title
US16/000,803 US20180282804A1 (en) 2012-12-14 2018-06-05 Methods and systems for processing polynucleotides
US16/052,486 US10323279B2 (en) 2012-08-14 2018-08-01 Methods and systems for processing polynucleotides
US16/052,431 US10273541B2 (en) 2012-08-14 2018-08-01 Methods and systems for processing polynucleotides
US16/165,389 US10533221B2 (en) 2012-12-14 2018-10-19 Methods and systems for processing polynucleotides
US16/212,441 US10752949B2 (en) 2012-08-14 2018-12-06 Methods and systems for processing polynucleotides
US16/231,142 US10584381B2 (en) 2012-08-14 2018-12-21 Methods and systems for processing polynucleotides
US16/231,185 US10400280B2 (en) 2012-08-14 2018-12-21 Methods and systems for processing polynucleotides
US16/246,322 US10597718B2 (en) 2012-08-14 2019-01-11 Methods and systems for sample processing polynucleotides
US16/294,769 US10450607B2 (en) 2012-08-14 2019-03-06 Methods and systems for processing polynucleotides
US16/395,090 US10669583B2 (en) 2012-08-14 2019-04-25 Method and systems for processing polynucleotides
US16/435,417 US10752950B2 (en) 2012-08-14 2019-06-07 Methods and systems for processing polynucleotides
US16/435,362 US10626458B2 (en) 2012-08-14 2019-06-07 Methods and systems for processing polynucleotides
US16/698,740 US11473138B2 (en) 2012-12-14 2019-11-27 Methods and systems for processing polynucleotides
US16/736,323 US20200255894A1 (en) 2012-08-14 2020-01-07 Methods and systems for sample processing polynucleotides
US16/844,141 US11441179B2 (en) 2012-08-14 2020-04-09 Methods and systems for processing polynucleotides
US16/998,425 US11035002B2 (en) 2012-08-14 2020-08-20 Methods and systems for processing polynucleotides
US16/998,414 US11021749B2 (en) 2012-08-14 2020-08-20 Methods and systems for processing polynucleotides
US17/314,526 US11359239B2 (en) 2012-08-14 2021-05-07 Methods and systems for processing polynucleotides
US17/392,610 US20220098659A1 (en) 2012-08-14 2021-08-03 Methods and systems for processing polynucleotides
US18/186,088 US20240002929A1 (en) 2012-08-14 2023-03-17 Methods and systems for processing polynucleotides

Applications Claiming Priority (12)

Application Number Priority Date Filing Date Title
US201261737374P 2012-12-14 2012-12-14
US201361762435P 2013-02-08 2013-02-08
US201361800223P 2013-03-15 2013-03-15
US201361840403P 2013-06-27 2013-06-27
US201361844804P 2013-07-10 2013-07-10
US14/104,650 US9567631B2 (en) 2012-12-14 2013-12-12 Methods and systems for processing polynucleotides
US14/175,973 US9388465B2 (en) 2013-02-08 2014-02-07 Polynucleotide barcode generation
US14/250,701 US20140235506A1 (en) 2013-02-08 2014-04-11 Polynucleotide barcode generation
US15/376,582 US9701998B2 (en) 2012-12-14 2016-12-12 Methods and systems for processing polynucleotides
US15/588,519 US9856530B2 (en) 2012-12-14 2017-05-05 Methods and systems for processing polynucleotides
US15/850,241 US10676789B2 (en) 2012-12-14 2017-12-21 Methods and systems for processing polynucleotides
US16/000,803 US20180282804A1 (en) 2012-12-14 2018-06-05 Methods and systems for processing polynucleotides

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US14/316,447 Continuation-In-Part US10221442B2 (en) 2012-08-14 2014-06-26 Compositions and methods for sample processing
US15/850,241 Continuation US10676789B2 (en) 2012-08-14 2017-12-21 Methods and systems for processing polynucleotides

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US16/052,431 Continuation-In-Part US10273541B2 (en) 2012-08-14 2018-08-01 Methods and systems for processing polynucleotides
US16/052,486 Continuation-In-Part US10323279B2 (en) 2012-08-14 2018-08-01 Methods and systems for processing polynucleotides
US16/165,389 Continuation-In-Part US10533221B2 (en) 2012-12-14 2018-10-19 Methods and systems for processing polynucleotides

Publications (1)

Publication Number Publication Date
US20180282804A1 true US20180282804A1 (en) 2018-10-04

Family

ID=58558428

Family Applications (5)

Application Number Title Priority Date Filing Date
US15/376,582 Active US9701998B2 (en) 2012-08-14 2016-12-12 Methods and systems for processing polynucleotides
US15/588,519 Active US9856530B2 (en) 2012-08-14 2017-05-05 Methods and systems for processing polynucleotides
US15/850,241 Active US10676789B2 (en) 2012-08-14 2017-12-21 Methods and systems for processing polynucleotides
US16/000,803 Abandoned US20180282804A1 (en) 2012-08-14 2018-06-05 Methods and systems for processing polynucleotides
US16/852,906 Active US11421274B2 (en) 2012-12-14 2020-04-20 Methods and systems for processing polynucleotides

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US15/376,582 Active US9701998B2 (en) 2012-08-14 2016-12-12 Methods and systems for processing polynucleotides
US15/588,519 Active US9856530B2 (en) 2012-08-14 2017-05-05 Methods and systems for processing polynucleotides
US15/850,241 Active US10676789B2 (en) 2012-08-14 2017-12-21 Methods and systems for processing polynucleotides

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/852,906 Active US11421274B2 (en) 2012-12-14 2020-04-20 Methods and systems for processing polynucleotides

Country Status (1)

Country Link
US (5) US9701998B2 (en)

Cited By (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10150964B2 (en) 2013-02-08 2018-12-11 10X Genomics, Inc. Partitioning and processing of analytes and other species
US10208343B2 (en) 2014-06-26 2019-02-19 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10221436B2 (en) 2015-01-12 2019-03-05 10X Genomics, Inc. Processes and systems for preparation of nucleic acid sequencing libraries and libraries prepared using same
US10221442B2 (en) 2012-08-14 2019-03-05 10X Genomics, Inc. Compositions and methods for sample processing
US10227648B2 (en) 2012-12-14 2019-03-12 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10273541B2 (en) 2012-08-14 2019-04-30 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10323279B2 (en) 2012-08-14 2019-06-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10343166B2 (en) 2014-04-10 2019-07-09 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same
US10345219B2 (en) 2011-08-01 2019-07-09 Celsee Diagnostics, Inc. Cell capture system and method of use
US10350601B2 (en) 2013-03-13 2019-07-16 Celsee Diagnostics, Inc. System and method for capturing and analyzing cells
US10391492B2 (en) 2017-08-29 2019-08-27 Celsee Diagnostics, Inc. System and method for isolating and analyzing cells
US10391490B2 (en) 2013-05-31 2019-08-27 Celsee Diagnostics, Inc. System and method for isolating and analyzing cells
US10400280B2 (en) 2012-08-14 2019-09-03 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10400235B2 (en) 2017-05-26 2019-09-03 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US10428326B2 (en) 2017-01-30 2019-10-01 10X Genomics, Inc. Methods and systems for droplet-based single cell barcoding
US10466160B2 (en) 2011-08-01 2019-11-05 Celsee Diagnostics, Inc. System and method for retrieving and analyzing particles
US10509022B2 (en) 2013-03-13 2019-12-17 Celsee Diagnostics, Inc. System for imaging captured cells
US10533221B2 (en) 2012-12-14 2020-01-14 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10533229B2 (en) 2013-05-31 2020-01-14 Celsee Diagnostics, Inc. System and method for isolating and analyzing cells
US10550429B2 (en) 2016-12-22 2020-02-04 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10633693B1 (en) 2019-04-16 2020-04-28 Celsee Diagnostics, Inc. System and method for leakage control in a particle capture system
US10676789B2 (en) 2012-12-14 2020-06-09 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10697000B2 (en) 2015-02-24 2020-06-30 10X Genomics, Inc. Partition processing methods and systems
US10718007B2 (en) 2013-01-26 2020-07-21 Bio-Rad Laboratories, Inc. System and method for capturing and analyzing cells
US10745742B2 (en) 2017-11-15 2020-08-18 10X Genomics, Inc. Functionalized gel beads
US10752949B2 (en) 2012-08-14 2020-08-25 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10774370B2 (en) 2015-12-04 2020-09-15 10X Genomics, Inc. Methods and compositions for nucleic acid analysis
US10815525B2 (en) 2016-12-22 2020-10-27 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10829815B2 (en) 2017-11-17 2020-11-10 10X Genomics, Inc. Methods and systems for associating physical and genetic properties of biological particles
US10900032B2 (en) 2019-05-07 2021-01-26 Bio-Rad Laboratories, Inc. System and method for automated single cell processing
US11078522B2 (en) 2012-08-14 2021-08-03 10X Genomics, Inc. Capsule array devices and methods of use
US11084036B2 (en) 2016-05-13 2021-08-10 10X Genomics, Inc. Microfluidic systems and methods of use
US11135584B2 (en) 2014-11-05 2021-10-05 10X Genomics, Inc. Instrument systems for integrated sample processing
US11155881B2 (en) 2018-04-06 2021-10-26 10X Genomics, Inc. Systems and methods for quality control in single cell processing
US11273439B2 (en) 2019-05-07 2022-03-15 Bio-Rad Laboratories, Inc. System and method for target material retrieval from microwells
US11274343B2 (en) 2015-02-24 2022-03-15 10X Genomics, Inc. Methods and compositions for targeted nucleic acid sequence coverage
WO2022066992A1 (en) * 2020-09-24 2022-03-31 University Of Cincinnati Continuous sensing with adapters and aptamers
US11504719B2 (en) 2020-03-12 2022-11-22 Bio-Rad Laboratories, Inc. System and method for receiving and delivering a fluid for sample processing
US11591637B2 (en) 2012-08-14 2023-02-28 10X Genomics, Inc. Compositions and methods for sample processing
US11629344B2 (en) 2014-06-26 2023-04-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11724256B2 (en) 2019-06-14 2023-08-15 Bio-Rad Laboratories, Inc. System and method for automated single cell processing and analyses
US11773389B2 (en) 2017-05-26 2023-10-03 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
EP4217493A4 (en) * 2020-09-24 2024-04-10 Univ Cincinnati Solute-phase electrochemical aptamer sensors with rapid time-to-measurement

Families Citing this family (75)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010033200A2 (en) 2008-09-19 2010-03-25 President And Fellows Of Harvard College Creation of libraries of droplets and related species
WO2010115100A1 (en) * 2009-04-03 2010-10-07 L&C Diagment, Inc. Multiplex nucleic acid detection methods and systems
US9902950B2 (en) * 2010-10-08 2018-02-27 President And Fellows Of Harvard College High-throughput single cell barcoding
GB2504240B (en) 2012-02-27 2015-05-27 Cellular Res Inc Compositions and kits for molecular counting of nucleic acids
LT3305918T (en) 2012-03-05 2020-09-25 President And Fellows Of Harvard College Methods for epigenetic sequencing
KR102536833B1 (en) 2013-08-28 2023-05-26 벡톤 디킨슨 앤드 컴퍼니 Massively parallel single cell analysis
US9824068B2 (en) 2013-12-16 2017-11-21 10X Genomics, Inc. Methods and apparatus for sorting data
US20150298091A1 (en) 2014-04-21 2015-10-22 President And Fellows Of Harvard College Systems and methods for barcoding nucleic acids
JP2017532024A (en) 2014-09-09 2017-11-02 ザ・ブロード・インスティテュート・インコーポレイテッド Droplet-based methods and instruments for composite single cell nucleic acid analysis
WO2016069939A1 (en) 2014-10-29 2016-05-06 10X Genomics, Inc. Methods and compositions for targeted nucleic acid sequencing
EP3227684B1 (en) 2014-12-03 2019-10-02 Isoplexis Corporation Analysis and screening of cell secretion profiles
EP3262192B1 (en) 2015-02-27 2020-09-16 Becton, Dickinson and Company Spatially addressable molecular barcoding
EP3268462B1 (en) 2015-03-11 2021-08-11 The Broad Institute, Inc. Genotype and phenotype coupling
EP4180535A1 (en) 2015-03-30 2023-05-17 Becton, Dickinson and Company Methods and compositions for combinatorial barcoding
AU2016248995B2 (en) 2015-04-17 2022-04-28 President And Fellows Of Harvard College Barcoding systems and methods for gene sequencing and other applications
US10619186B2 (en) 2015-09-11 2020-04-14 Cellular Research, Inc. Methods and compositions for library normalization
US11123297B2 (en) 2015-10-13 2021-09-21 President And Fellows Of Harvard College Systems and methods for making and using gel microspheres
WO2017075297A1 (en) 2015-10-28 2017-05-04 The Broad Institute Inc. High-throughput dynamic reagent delivery system
US11092607B2 (en) 2015-10-28 2021-08-17 The Board Institute, Inc. Multiplex analysis of single cell constituents
US11371094B2 (en) 2015-11-19 2022-06-28 10X Genomics, Inc. Systems and methods for nucleic acid processing using degenerate nucleotides
JP6735348B2 (en) 2016-02-11 2020-08-05 10エックス ジェノミクス, インコーポレイテッド Systems, methods and media for de novo assembly of whole genome sequence data
US10301677B2 (en) 2016-05-25 2019-05-28 Cellular Research, Inc. Normalization of nucleic acid libraries
US10640763B2 (en) 2016-05-31 2020-05-05 Cellular Research, Inc. Molecular indexing of internal sequences
US10202641B2 (en) 2016-05-31 2019-02-12 Cellular Research, Inc. Error correction in amplification of samples
SG11201901733PA (en) 2016-09-26 2019-04-29 Cellular Res Inc Measurement of protein expression using reagents with barcoded oligonucleotide sequences
DK3538891T3 (en) 2016-11-11 2022-03-28 Isoplexis Corp COMPOSITIONS AND PROCEDURES FOR CONTEMPORARY GENOMIC, TRANSCRIPTOMIC AND PROTEOMIC ANALYSIS OF SINGLE CELLS
EP3545284A4 (en) 2016-11-22 2020-07-01 Isoplexis Corporation Systems, devices and methods for cell capture and methods of manufacture thereof
US10011872B1 (en) 2016-12-22 2018-07-03 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11319583B2 (en) 2017-02-01 2022-05-03 Becton, Dickinson And Company Selective amplification using blocking oligonucleotides
US10995333B2 (en) 2017-02-06 2021-05-04 10X Genomics, Inc. Systems and methods for nucleic acid preparation
US11072816B2 (en) 2017-05-03 2021-07-27 The Broad Institute, Inc. Single-cell proteomic assay using aptamers
CN117143960A (en) 2017-05-18 2023-12-01 10X基因组学有限公司 Method and system for sorting droplets and beads
US10544413B2 (en) 2017-05-18 2020-01-28 10X Genomics, Inc. Methods and systems for sorting droplets and beads
US10549279B2 (en) 2017-08-22 2020-02-04 10X Genomics, Inc. Devices having a plurality of droplet formation regions
US10837047B2 (en) 2017-10-04 2020-11-17 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
US10590244B2 (en) 2017-10-04 2020-03-17 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
WO2019083852A1 (en) 2017-10-26 2019-05-02 10X Genomics, Inc. Microfluidic channel networks for partitioning
WO2019084043A1 (en) 2017-10-26 2019-05-02 10X Genomics, Inc. Methods and systems for nuclecic acid preparation and chromatin analysis
CN114525273A (en) 2017-10-27 2022-05-24 10X基因组学有限公司 Methods and systems for sample preparation and analysis
WO2019108851A1 (en) 2017-11-30 2019-06-06 10X Genomics, Inc. Systems and methods for nucleic acid preparation and analysis
WO2019157529A1 (en) 2018-02-12 2019-08-15 10X Genomics, Inc. Methods characterizing multiple analytes from individual cells or cell populations
US11639928B2 (en) 2018-02-22 2023-05-02 10X Genomics, Inc. Methods and systems for characterizing analytes from individual cells or cell populations
JP7407128B2 (en) 2018-05-03 2023-12-28 ベクトン・ディキンソン・アンド・カンパニー High-throughput multi-omics sample analysis
WO2019213237A1 (en) 2018-05-03 2019-11-07 Becton, Dickinson And Company Molecular barcoding on opposite transcript ends
US11932899B2 (en) 2018-06-07 2024-03-19 10X Genomics, Inc. Methods and systems for characterizing nucleic acid molecules
US11703427B2 (en) 2018-06-25 2023-07-18 10X Genomics, Inc. Methods and systems for cell and bead processing
US20200032335A1 (en) 2018-07-27 2020-01-30 10X Genomics, Inc. Systems and methods for metabolome analysis
CN112805389A (en) 2018-10-01 2021-05-14 贝克顿迪金森公司 Determination of 5' transcript sequences
JP2022506546A (en) 2018-11-08 2022-01-17 ベクトン・ディキンソン・アンド・カンパニー Single-cell whole transcriptome analysis using random priming
WO2020100079A2 (en) * 2018-11-14 2020-05-22 Foresee Genomic Ltd Multimer for sequencing and methods for preparing and analyzing the same
US11459607B1 (en) 2018-12-10 2022-10-04 10X Genomics, Inc. Systems and methods for processing-nucleic acid molecules from a single cell using sequential co-partitioning and composite barcodes
EP3894552A1 (en) 2018-12-13 2021-10-20 Becton, Dickinson and Company Selective extension in single cell whole transcriptome analysis
TWI725686B (en) 2018-12-26 2021-04-21 財團法人工業技術研究院 Tubular structure for producing droplets and method for producing droplets
US11845983B1 (en) 2019-01-09 2023-12-19 10X Genomics, Inc. Methods and systems for multiplexing of droplet based assays
ES2945227T3 (en) 2019-01-23 2023-06-29 Becton Dickinson Co Antibody Associated Oligonucleotides
SG11202108788TA (en) 2019-02-12 2021-09-29 10X Genomics Inc Methods for processing nucleic acid molecules
US11851683B1 (en) 2019-02-12 2023-12-26 10X Genomics, Inc. Methods and systems for selective analysis of cellular samples
US11467153B2 (en) 2019-02-12 2022-10-11 10X Genomics, Inc. Methods for processing nucleic acid molecules
AU2020220461A1 (en) 2019-02-14 2021-08-05 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Haplotagging - haplotype phasing and single-tube combinatorial barcoding of nucleic acid molecules using bead-immobilized Tn5 transposase
US11655499B1 (en) 2019-02-25 2023-05-23 10X Genomics, Inc. Detection of sequence elements in nucleic acid molecules
CN113767178A (en) 2019-03-11 2021-12-07 10X基因组学有限公司 Systems and methods for processing optically labeled beads
US11939622B2 (en) 2019-07-22 2024-03-26 Becton, Dickinson And Company Single cell chromatin immunoprecipitation sequencing assay
US20210032702A1 (en) * 2019-07-31 2021-02-04 The General Hospital Corporation Lineage inference from single-cell transcriptomes
CA3157359A1 (en) 2019-10-10 2021-04-15 1859, Inc. Methods and systems for microfluidic screening
US11773436B2 (en) 2019-11-08 2023-10-03 Becton, Dickinson And Company Using random priming to obtain full-length V(D)J information for immune repertoire sequencing
JP2023511279A (en) 2020-01-13 2023-03-17 フルーエント バイオサイエンシーズ インコーポレイテッド Single cell sequencing
CN115244184A (en) 2020-01-13 2022-10-25 贝克顿迪金森公司 Methods and compositions for quantifying protein and RNA
EP4090328A4 (en) 2020-01-13 2024-02-14 Fluent Biosciences Inc Emulsion based drug screening
EP4090466A4 (en) 2020-01-13 2024-01-17 Fluent Biosciences Inc Methods and systems for single cell gene profiling
WO2021188500A1 (en) 2020-03-16 2021-09-23 Fluent Biosciences Inc. Multi-omic analysis in monodisperse droplets
US11851700B1 (en) 2020-05-13 2023-12-26 10X Genomics, Inc. Methods, kits, and compositions for processing extracellular molecules
WO2021231779A1 (en) 2020-05-14 2021-11-18 Becton, Dickinson And Company Primers for immune repertoire profiling
US11932901B2 (en) 2020-07-13 2024-03-19 Becton, Dickinson And Company Target enrichment using nucleic acid probes for scRNAseq
EP4247967A1 (en) 2020-11-20 2023-09-27 Becton, Dickinson and Company Profiling of highly expressed and lowly expressed proteins
EP4298244A1 (en) 2021-02-23 2024-01-03 10X Genomics, Inc. Probe-based analysis of nucleic acids and proteins

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120022049A1 (en) * 2002-03-25 2012-01-26 Universite Laval Nicotinic receptor agonists for the treatment of inflammatory diseases
US20150001143A1 (en) * 2013-06-27 2015-01-01 Desmond Bell Oil Filter Conversion Kit

Family Cites Families (793)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2797149A (en) 1953-01-08 1957-06-25 Technicon International Ltd Methods of and apparatus for analyzing liquids containing crystalloid and non-crystalloid constituents
US3047367A (en) 1959-12-01 1962-07-31 Technicon Instr Automatic analysis with fluid segmentation
US3479141A (en) 1967-05-17 1969-11-18 Technicon Corp Method and apparatus for analysis
US4124638A (en) 1977-09-12 1978-11-07 Hansen John N Solubilizable polyacrylamide gels containing disulfide cross-linkages
US4253846A (en) 1979-11-21 1981-03-03 Technicon Instruments Corporation Method and apparatus for automated analysis of fluid samples
GB2097692B (en) 1981-01-10 1985-05-22 Shaw Stewart P D Combining chemical reagents
DE3230289A1 (en) 1982-08-14 1984-02-16 Bayer Ag, 5090 Leverkusen PRODUCTION OF PHARMACEUTICAL OR COSMETIC DISPERSIONS
US4582802A (en) 1983-09-30 1986-04-15 The United States Of America As Represented By The Department Of Health And Human Services Stimulation of enzymatic ligation of DNA by high concentrations of nonspecific polymers
JPS60227826A (en) 1984-04-27 1985-11-13 Sogo Yatsukou Kk Graft capsule responding to ph
KR890003947B1 (en) 1985-12-11 1989-10-13 가부시기가이샤 시마즈세이사구쇼 Apparatus for cell fusion
US4916070A (en) 1986-04-14 1990-04-10 The General Hospital Corporation Fibrin-specific antibodies and method of screening for the antibodies
US5618711A (en) 1986-08-22 1997-04-08 Hoffmann-La Roche Inc. Recombinant expression vectors and purification methods for Thermus thermophilus DNA polymerase
US4872895A (en) 1986-12-11 1989-10-10 American Telephone And Telegraph Company, At&T Bell Laboratories Method for fabricating articles which include high silica glass bodies
US5202231A (en) 1987-04-01 1993-04-13 Drmanac Radoje T Method of sequencing of genomes by hybridization of oligonucleotide probes
US5525464A (en) 1987-04-01 1996-06-11 Hyseq, Inc. Method of sequencing by hybridization of oligonucleotide probes
US5149625A (en) 1987-08-11 1992-09-22 President And Fellows Of Harvard College Multiplex analysis of DNA
US5137829A (en) 1987-10-05 1992-08-11 Washington University DNA transposon TN5SEQ1
US5185099A (en) 1988-04-20 1993-02-09 Institut National De Recherche Chimique Appliquee Visco-elastic, isotropic materials based on water, fluorinate sufactants and fluorinated oils, process for their preparation, and their use in various fields, such as optics, pharmacology and electrodynamics
US5237016A (en) 1989-01-05 1993-08-17 Siska Diagnostics, Inc. End-attachment of oligonucleotides to polyacrylamide solid supports for capture and detection of nucleic acids
US6176962B1 (en) 1990-02-28 2001-01-23 Aclara Biosciences, Inc. Methods for fabricating enclosed microchannel structures
US5756334A (en) 1990-04-26 1998-05-26 New England Biolabs, Inc. Thermostable DNA polymerase from 9°N-7 and methods for producing the same
DK0540693T3 (en) 1990-07-24 1999-09-13 Hoffmann La Roche Reduction of non-specific amplification during in vitro nucleic acid amplification using modified nucleic acid
US5489523A (en) 1990-12-03 1996-02-06 Stratagene Exonuclease-deficient thermostable Pyrococcus furiosus DNA polymerase I
US6582908B2 (en) 1990-12-06 2003-06-24 Affymetrix, Inc. Oligonucleotides
US5270183A (en) 1991-02-08 1993-12-14 Beckman Research Institute Of The City Of Hope Device and method for the automated cycling of solutions between two or more temperatures
US5994056A (en) 1991-05-02 1999-11-30 Roche Molecular Systems, Inc. Homogeneous methods for nucleic acid amplification and detection
JP3340434B2 (en) 1991-07-04 2002-11-05 ダコ アクティーゼルスカブ Water-soluble polymer-based reagents and conjugates comprising components derived from divinyl sulfone
EP0597960B1 (en) 1991-08-10 1999-01-20 Medical Research Council Treatment of cell populations
DK0604552T3 (en) 1991-09-18 1997-08-04 Affymax Tech Nv Process for the synthesis of different assemblies of oligomers
US5413924A (en) 1992-02-13 1995-05-09 Kosak; Kenneth M. Preparation of wax beads containing a reagent for release by heating
AU3816993A (en) 1992-03-19 1993-10-21 Regents Of The University Of California, The Multiple tag labeling method for DNA sequencing
US5587128A (en) 1992-05-01 1996-12-24 The Trustees Of The University Of Pennsylvania Mesoscale polynucleotide amplification devices
ATE174813T1 (en) 1992-05-01 1999-01-15 Univ Pennsylvania POLYNUCLEOTIDE AMPLIFICATION ANALYSIS USING A MICROFABRICATED DEVICE
US5840865A (en) 1992-09-14 1998-11-24 Institute Of Molecular Biology And Biotechnology/Forth Eukaryotic transposable element
US5897783A (en) 1992-09-24 1999-04-27 Amersham International Plc Magnetic separation method
US5569364A (en) 1992-11-05 1996-10-29 Soane Biosciences, Inc. Separation media for electrophoresis
IL108497A0 (en) 1993-02-01 1994-05-30 Seq Ltd Methods and apparatus for dna sequencing
AU6175594A (en) 1993-02-16 1994-09-14 Alliance Pharmaceutical Corporation Method of microemulsifying fluorinated oils
AU6637394A (en) 1993-04-19 1994-11-08 Medisorb Technologies International L.P. Long-acting treatment by slow-release delivery of antisense oligodeoxyribonucleotides from biodegradable microparticles
US5456986A (en) 1993-06-30 1995-10-10 Carnegie Mellon University Magnetic metal or metal carbide nanoparticles and a process for forming same
DE69429038T2 (en) 1993-07-28 2002-03-21 Pe Corp Ny Norwalk Device and method for nucleic acid amplification
EP0711303B2 (en) 1993-07-30 2009-06-10 Affymax, Inc. Biotinylation of proteins
WO1995006652A1 (en) 1993-08-30 1995-03-09 Promega Corporation Nucleic acid purification compositions and methods
US5512131A (en) 1993-10-04 1996-04-30 President And Fellows Of Harvard College Formation of microstamped patterns on surfaces and derivative articles
US20030044777A1 (en) 1993-10-28 2003-03-06 Kenneth L. Beattie Flowthrough devices for multiple discrete binding reactions
US5605793A (en) 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
US5558071A (en) 1994-03-07 1996-09-24 Combustion Electromagnetics, Inc. Ignition system power converter and controller
US5648211A (en) 1994-04-18 1997-07-15 Becton, Dickinson And Company Strand displacement amplification using thermophilic enzymes
CA2189958C (en) 1994-05-11 2005-04-19 Adrian Parton Methods of capturing species from liquids and assay procedures
US5705628A (en) 1994-09-20 1998-01-06 Whitehead Institute For Biomedical Research DNA purification and isolation using magnetic particles
US5846719A (en) 1994-10-13 1998-12-08 Lynx Therapeutics, Inc. Oligonucleotide tags for sorting and identification
US5585069A (en) 1994-11-10 1996-12-17 David Sarnoff Research Center, Inc. Partitioned microelectronic and fluidic device array for clinical diagnostics and chemical synthesis
WO1996029629A2 (en) 1995-03-01 1996-09-26 President And Fellows Of Harvard College Microcontact printing on surfaces and derivative articles
US5700642A (en) 1995-05-22 1997-12-23 Sri International Oligonucleotide sizing using immobilized cleavable primers
EP0832287B1 (en) 1995-06-07 2007-10-10 Solexa, Inc Oligonucleotide tags for sorting and identification
CA2332731A1 (en) 1995-06-07 1996-12-19 Sydney Brenner Oligonucleotide tags for sorting and identification
US5856174A (en) 1995-06-29 1999-01-05 Affymetrix, Inc. Integrated nucleic acid diagnostic device
US6866760B2 (en) 1998-08-27 2005-03-15 E Ink Corporation Electrophoretic medium and process for the production thereof
US5872010A (en) 1995-07-21 1999-02-16 Northeastern University Microscale fluid handling system
US6057149A (en) 1995-09-15 2000-05-02 The University Of Michigan Microscale devices and reactions in microscale devices
US5851769A (en) 1995-09-27 1998-12-22 The Regents Of The University Of California Quantitative DNA fiber mapping
US5736330A (en) 1995-10-11 1998-04-07 Luminex Corporation Method and compositions for flow cytometric determination of DNA sequences
US6051377A (en) 1995-11-30 2000-04-18 Pharmaseq, Inc. Multiplex assay for nucleic acids employing transponders
US6001571A (en) 1995-11-30 1999-12-14 Mandecki; Wlodek Multiplex assay for nucleic acids employing transponders
US5736332A (en) 1995-11-30 1998-04-07 Mandecki; Wlodek Method of determining the sequence of nucleic acids employing solid-phase particles carrying transponders
US6355198B1 (en) 1996-03-15 2002-03-12 President And Fellows Of Harvard College Method of forming articles including waveguides via capillary micromolding and microtransfer molding
WO1997039359A1 (en) 1996-04-15 1997-10-23 Dade International Inc. Apparatus and method for analysis
WO1997045559A1 (en) 1996-05-29 1997-12-04 Cornell Research Foundation, Inc. Detection of nucleic acid sequence differences using coupled ligase detection and polymerase chain reactions
US5846727A (en) 1996-06-06 1998-12-08 Board Of Supervisors Of Louisiana State University And Agricultural & Mechanical College Microsystem for rapid DNA sequencing
DE69707288T2 (en) 1996-07-15 2002-07-18 Calcitech Ltd PRODUCTION OF POWDERS
US5965443A (en) 1996-09-09 1999-10-12 Wisconsin Alumni Research Foundation System for in vitro transposition
US6133436A (en) 1996-11-06 2000-10-17 Sequenom, Inc. Beads bound to a solid support and to nucleic acids
US5900481A (en) 1996-11-06 1999-05-04 Sequenom, Inc. Bead linkers for immobilizing nucleic acids to solid supports
WO1998022625A1 (en) 1996-11-20 1998-05-28 The Regents Of The University Of Michigan Microfabricated isothermal nucleic acid amplification devices and methods
US5958703A (en) 1996-12-03 1999-09-28 Glaxo Group Limited Use of modified tethers in screening compound libraries
US20020172965A1 (en) 1996-12-13 2002-11-21 Arcaris, Inc. Methods for measuring relative amounts of nucleic acids in a complex mixture and retrieval of specific sequences therefrom
US20050042625A1 (en) 1997-01-15 2005-02-24 Xzillion Gmbh & Co. Mass label linked hybridisation probes
US6297006B1 (en) 1997-01-16 2001-10-02 Hyseq, Inc. Methods for sequencing repetitive sequences and for determining the order of sequence subfragments
US20020034737A1 (en) 1997-03-04 2002-03-21 Hyseq, Inc. Methods and compositions for detection or quantification of nucleic acid species
IL131332A (en) 1997-02-12 2003-07-31 Eugene Y Chan Methods and products for analyzing polymers
US6327410B1 (en) 1997-03-14 2001-12-04 The Trustees Of Tufts College Target analyte sensors utilizing Microspheres
US7622294B2 (en) 1997-03-14 2009-11-24 Trustees Of Tufts College Methods for detecting target analytes and enzymatic reactions
US20030027203A1 (en) 1997-03-24 2003-02-06 Fields Robert E. Biomolecular processor
US6391622B1 (en) 1997-04-04 2002-05-21 Caliper Technologies Corp. Closed-loop biochemical analyzers
US6143496A (en) 1997-04-17 2000-11-07 Cytonix Corporation Method of sampling, amplifying and quantifying segment of nucleic acid, polymerase chain reaction assembly having nanoliter-sized sample chambers, and method of filling assembly
CA2287570C (en) 1997-05-02 2008-10-28 Gen-Probe Incorporated Two-step hybridization and capture of a polynucleotide
EP0981408B1 (en) 1997-05-16 2004-04-21 Alberta Research Council Microfluidic system and methods of use
JP4294740B2 (en) 1997-05-23 2009-07-15 ソレクサ・インコーポレイテッド System and apparatus for serial processing of analytes
US6969488B2 (en) 1998-05-22 2005-11-29 Solexa, Inc. System and apparatus for sequential processing of analytes
US20040241759A1 (en) 1997-06-16 2004-12-02 Eileen Tozer High throughput screening of libraries
CA2792122C (en) 1997-07-07 2015-09-08 Medical Research Council In vitro sorting method
GB9714716D0 (en) 1997-07-11 1997-09-17 Brax Genomics Ltd Characterising nucleic acids
FI103809B1 (en) 1997-07-14 1999-09-30 Finnzymes Oy In vitro method for producing templates for DNA sequencing
US6974669B2 (en) 2000-03-28 2005-12-13 Nanosphere, Inc. Bio-barcodes based on oligonucleotide-modified nanoparticles
US20050037397A1 (en) 2001-03-28 2005-02-17 Nanosphere, Inc. Bio-barcode based detection of target analytes
US6368871B1 (en) 1997-08-13 2002-04-09 Cepheid Non-planar microstructures for manipulation of fluid samples
CA2300940A1 (en) 1997-08-15 1999-02-25 Hyseq, Inc. Methods and compositions for detection or quantification of nucleic acid species
WO1999014368A2 (en) 1997-09-15 1999-03-25 Whitehead Institute For Biomedical Research Methods and apparatus for processing a sample of biomolecular analyte using a microfabricated device
US20020092767A1 (en) 1997-09-19 2002-07-18 Aclara Biosciences, Inc. Multiple array microfluidic device units
US7214298B2 (en) 1997-09-23 2007-05-08 California Institute Of Technology Microfabricated cell sorter
WO1999018438A1 (en) 1997-10-02 1999-04-15 Aclara Biosciences, Inc. Capillary assays involving separation of free and bound species
US5842787A (en) 1997-10-09 1998-12-01 Caliper Technologies Corporation Microfluidic systems incorporating varied channel dimensions
US6485944B1 (en) 1997-10-10 2002-11-26 President And Fellows Of Harvard College Replica amplification of nucleic acid arrays
JP2001519538A (en) 1997-10-10 2001-10-23 プレジデント・アンド・フェローズ・オブ・ハーバード・カレッジ Replica amplification of nucleic acid arrays
US6511803B1 (en) 1997-10-10 2003-01-28 President And Fellows Of Harvard College Replica amplification of nucleic acid arrays
CA2306501C (en) 1997-10-14 2011-03-29 Luminex Corporation Precision fluorescently dyed particles and methods of making and using same
US6660149B1 (en) 1997-10-24 2003-12-09 Beckman Coulter, Inc. Multichannel microscale system for high throughput preparative separation with comprehensive collection and analysis
CA2313047A1 (en) 1997-12-04 1999-12-16 Nicholas Thomas Multiple assay method
WO1999033963A1 (en) 1997-12-31 1999-07-08 Chiron Corporation Metastatic cancer regulated gene
AU765378B2 (en) 1998-02-19 2003-09-18 President And Fellows Of Harvard College Monovalent, multivalent, and multimeric MHC binding domain fusion proteins and conjugates, and uses therefor
AU3196099A (en) 1998-03-27 1999-10-18 President And Fellows Of Harvard College Systematic identification of essential genes by (in vitro) transposon mutagenesis
US6022716A (en) 1998-04-10 2000-02-08 Genset Sa High throughput DNA sequencing vector
AU3555599A (en) 1998-04-13 1999-11-01 Luminex Corporation Liquid labeling with fluorescent microparticles
US6780591B2 (en) 1998-05-01 2004-08-24 Arizona Board Of Regents Method of determining the nucleotide sequence of oligonucleotides and DNA molecules
US5997636A (en) 1998-05-01 1999-12-07 Instrumentation Technology Associates, Inc. Method and apparatus for growing crystals
US6123798A (en) 1998-05-06 2000-09-26 Caliper Technologies Corp. Methods of fabricating polymeric structures incorporating microscale fluidic elements
US6534262B1 (en) 1998-05-14 2003-03-18 Whitehead Institute For Biomedical Research Solid phase technique for selectively isolating nucleic acids
US6306590B1 (en) 1998-06-08 2001-10-23 Caliper Technologies Corp. Microfluidic matrix localization apparatus and methods
JP3662850B2 (en) 1998-06-24 2005-06-22 イルミナ インコーポレイテッド Decoding array sensors with microspheres
WO2000008212A1 (en) 1998-08-07 2000-02-17 Cellay, Llc Gel microdrops in genetic analysis
US6159736A (en) 1998-09-23 2000-12-12 Wisconsin Alumni Research Foundation Method for making insertional mutations using a Tn5 synaptic complex
AR021833A1 (en) 1998-09-30 2002-08-07 Applied Research Systems METHODS OF AMPLIFICATION AND SEQUENCING OF NUCLEIC ACID
WO2000022436A1 (en) 1998-10-13 2000-04-20 Biomicro Systems, Inc. Fluid circuit components based upon passive fluid dynamics
US6489096B1 (en) 1998-10-15 2002-12-03 Princeton University Quantitative analysis of hybridization patterns and intensities in oligonucleotide arrays
SE9803614L (en) 1998-10-19 2000-04-20 Muhammed Mamoun Method and apparatus for producing nanoparticles
WO2000026412A1 (en) 1998-11-02 2000-05-11 Kenneth Loren Beattie Nucleic acid analysis using sequence-targeted tandem hybridization
US6569631B1 (en) 1998-11-12 2003-05-27 3-Dimensional Pharmaceuticals, Inc. Microplate thermal shift assay for ligand development using 5-(4″dimethylaminophenyl)-2-(4′-phenyl)oxazole derivative fluorescent dyes
US5942609A (en) 1998-11-12 1999-08-24 The Porkin-Elmer Corporation Ligation assembly and detection of polynucleotides on solid-support
US6465193B2 (en) 1998-12-11 2002-10-15 The Regents Of The University Of California Targeted molecular bar codes and methods for using the same
NO986133D0 (en) 1998-12-23 1998-12-23 Preben Lexow Method of DNA Sequencing
GB9900298D0 (en) 1999-01-07 1999-02-24 Medical Res Council Optical sorting method
US6416642B1 (en) 1999-01-21 2002-07-09 Caliper Technologies Corp. Method and apparatus for continuous liquid flow in microscale channels using pressure injection, wicking, and electrokinetic injection
US6635419B1 (en) 1999-02-16 2003-10-21 Applera Corporation Polynucleotide sequencing method
US20030027214A1 (en) 1999-02-17 2003-02-06 Kamb Carl Alexander Methods for substrate-ligand interaction screening
EP1163052B1 (en) 1999-02-23 2010-06-02 Caliper Life Sciences, Inc. Manipulation of microparticles in microfluidic systems
US7615373B2 (en) 1999-02-25 2009-11-10 Virginia Commonwealth University Intellectual Property Foundation Electroprocessed collagen and tissue engineering
US6171850B1 (en) 1999-03-08 2001-01-09 Caliper Technologies Corp. Integrated devices and systems for performing temperature controlled reactions and analyses
US6303343B1 (en) 1999-04-06 2001-10-16 Caliper Technologies Corp. Inefficient fast PCR
US6908737B2 (en) 1999-04-15 2005-06-21 Vitra Bioscience, Inc. Systems and methods of conducting multiplexed experiments
US20060275782A1 (en) 1999-04-20 2006-12-07 Illumina, Inc. Detection of nucleic acid reactions on bead arrays
EP1196630B2 (en) 1999-04-20 2018-10-17 Illumina, Inc. Detection of nucleic acid reactions on bead arrays
WO2000065042A1 (en) 1999-04-28 2000-11-02 The Board Of Trustees Of The Leland Stanford Junior University P element derived vector and methods for its use
JP3815969B2 (en) 1999-05-12 2006-08-30 アクララ バイオサイエンシーズ, インコーポレイテッド Multiplex fluorescence detection in microfluidic devices
WO2000070095A2 (en) 1999-05-17 2000-11-23 Dade Behring Inc. Homogeneous isothermal amplification and detection of nucleic acids using a template switch oligonucleotide
US20020051971A1 (en) 1999-05-21 2002-05-02 John R. Stuelpnagel Use of microfluidic systems in the detection of target analytes using microsphere arrays
US6846622B1 (en) 1999-05-26 2005-01-25 Oregon Health & Science University Tagged epitope protein transposable element
US20030124509A1 (en) 1999-06-03 2003-07-03 Kenis Paul J.A. Laminar flow patterning and articles made thereby
US6372813B1 (en) 1999-06-25 2002-04-16 Motorola Methods and compositions for attachment of biomolecules to solid supports, hydrogels, and hydrogel arrays
AU6068300A (en) 1999-07-06 2001-01-22 Caliper Technologies Corporation Microfluidic systems and methods for determining modulator kinetics
US6977145B2 (en) 1999-07-28 2005-12-20 Serono Genetics Institute S.A. Method for carrying out a biochemical protocol in continuous flow in a microreactor
US6524456B1 (en) 1999-08-12 2003-02-25 Ut-Battelle, Llc Microfluidic devices for the controlled manipulation of small volumes
EP1210358A4 (en) 1999-08-13 2005-01-05 Univ Brandeis Detection of nucleic acids
AU6788100A (en) 1999-08-20 2001-03-19 Luminex Corporation Liquid array technology
US6492118B1 (en) 1999-08-27 2002-12-10 Matrix Technologies Corporation Methods of immobilizing ligands on solid supports
US6982146B1 (en) 1999-08-30 2006-01-03 The United States Of America As Represented By The Department Of Health And Human Services High speed parallel molecular nucleic acid sequencing
CA2386193A1 (en) 1999-10-13 2001-04-19 Signature Bioscience, Inc. System and method for detecting and identifying molecular events in a test sample
US6958225B2 (en) 1999-10-27 2005-10-25 Affymetrix, Inc. Complexity management of genomic DNA
AU1100201A (en) 1999-10-28 2001-05-08 Board Of Trustees Of The Leland Stanford Junior University Methods of in vivo gene transfer using a sleeping beauty transposon system
JP4721603B2 (en) 1999-11-08 2011-07-13 栄研化学株式会社 Mutation and / or polymorphism detection method
US6432290B1 (en) 1999-11-26 2002-08-13 The Governors Of The University Of Alberta Apparatus and method for trapping bead based reagents within microfluidic analysis systems
US20010051348A1 (en) 2000-01-28 2001-12-13 Lee Chee Wee Novel ligands and methods for preparing same
KR20020089357A (en) 2000-02-23 2002-11-29 자이오믹스, 인코포레이티드 Chips having elevated sample surfaces
AU2001249071B2 (en) 2000-02-23 2005-09-08 Caliper Life Sciences, Inc. Multi-reservoir pressure control system
IL134830A0 (en) 2000-03-01 2001-05-20 Chay 13 Medical Res Group N V Peptides and immunostimulatory and anti-bacterial pharmaceutical compositions containing them
CN1487836A (en) 2000-03-14 2004-04-07 埃米林药品公司 Effects of glucagon-like peptide-1 (7-36) on antro-pyloro-duodenal motility
JP4927287B2 (en) 2000-03-31 2012-05-09 マイクロニックス、インコーポレーテッド Microfluidic device for protein crystallization
US20020001856A1 (en) 2000-04-06 2002-01-03 Chow Andrea W. Methods and devices for achieving long incubation times in high-throughput systems
DK2206791T3 (en) 2000-04-10 2016-10-24 Taxon Biosciences Inc Methods of study and genetic analysis of populations
US6481453B1 (en) 2000-04-14 2002-11-19 Nanostream, Inc. Microfluidic branch metering systems and methods
US6800298B1 (en) 2000-05-11 2004-10-05 Clemson University Biological lubricant composition and method of applying lubricant composition
US20060008799A1 (en) 2000-05-22 2006-01-12 Hong Cai Rapid haplotyping by single molecule detection
CA2408574A1 (en) 2000-05-24 2001-11-29 Micronics, Inc. Microfluidic concentration gradient loop
US6645432B1 (en) 2000-05-25 2003-11-11 President & Fellows Of Harvard College Microfluidic systems including three-dimensionally arrayed channel networks
US20060263888A1 (en) 2000-06-02 2006-11-23 Honeywell International Inc. Differential white blood count on a disposable card
US6632606B1 (en) 2000-06-12 2003-10-14 Aclara Biosciences, Inc. Methods for single nucleotide polymorphism detection
DE60117556T2 (en) 2000-06-21 2006-11-02 Bioarray Solutions Ltd. MULTI-ANALYTIC MOLECULAR ANALYSIS THROUGH THE USE OF APPLICATION SPECIFIC RAPID PARTICLE ARRAYS
WO2002011888A2 (en) 2000-08-07 2002-02-14 Nanostream, Inc. Fluidic mixer in microfluidic system
US6773566B2 (en) 2000-08-31 2004-08-10 Nanolytics, Inc. Electrostatic actuators for microfluidics and methods for using same
US6610499B1 (en) 2000-08-31 2003-08-26 The Regents Of The University Of California Capillary array and related methods
DE60140553D1 (en) 2000-09-14 2009-12-31 Caliper Life Sciences Inc MICROFLUIDIC DEVICES AND METHODS FOR CARRYING OUT TEMPERATURE-MEDIATED REACTIONS
EP1334347A1 (en) 2000-09-15 2003-08-13 California Institute Of Technology Microfabricated crossflow devices and methods
AU2002211389A1 (en) 2000-10-03 2002-04-15 California Institute Of Technology Microfluidic devices and methods of use
JP2005501217A (en) 2000-10-10 2005-01-13 ディベルサ コーポレーション High-throughput or capillary-based screening for bioactivity or biomolecules
JP2002155305A (en) 2000-11-14 2002-05-31 Akira Kawasaki Equipment and method for manufacturing monodispersed particle, and monodispersed particle manufactured by the manufacturing method
US6492154B2 (en) 2001-01-31 2002-12-10 Applera Corporation Isolated human kinase proteins, nucleic acid molecules encoding human kinase proteins, and uses thereof
CA2332186A1 (en) 2001-02-08 2002-08-08 Her Majesty In Right Of Canada As Represented By The Minister Of Agricul Ture And Agri-Food Canada Replicative in vivo gene targeting
US7670559B2 (en) 2001-02-15 2010-03-02 Caliper Life Sciences, Inc. Microfluidic systems with enhanced detection systems
WO2002068383A2 (en) 2001-02-22 2002-09-06 Anika Therapeutics, Inc. Thiol-modified hyaluronan
EP1447127B1 (en) 2001-02-23 2007-07-04 Japan Science and Technology Agency Apparatus for producing emulsion
US20150329617A1 (en) 2001-03-14 2015-11-19 Dynal Biotech Asa Novel MHC molecule constructs, and methods of employing these constructs for diagnosis and therapy, and uses of MHC molecules
US7211654B2 (en) 2001-03-14 2007-05-01 Regents Of The University Of Michigan Linkers and co-coupling agents for optimization of oligonucleotide synthesis and purification on solid supports
US20020160518A1 (en) 2001-04-03 2002-10-31 Hayenga Jon W. Microfluidic sedimentation
US7138267B1 (en) 2001-04-04 2006-11-21 Epicentre Technologies Corporation Methods and compositions for amplifying DNA clone copy number
US20030027221A1 (en) 2001-04-06 2003-02-06 Scott Melissa E. High-throughput screening assays by encapsulation
US7572642B2 (en) 2001-04-18 2009-08-11 Ambrigen, Llc Assay based on particles, which specifically bind with targets in spatially distributed characteristic patterns
DE60229246D1 (en) 2001-05-26 2008-11-20 One Cell Systems Inc
US6880576B2 (en) 2001-06-07 2005-04-19 Nanostream, Inc. Microfluidic devices for methods development
US7179423B2 (en) 2001-06-20 2007-02-20 Cytonome, Inc. Microfluidic system including a virtual wall fluid interface port for interfacing fluids with the microfluidic system
US7262063B2 (en) 2001-06-21 2007-08-28 Bio Array Solutions, Ltd. Directed assembly of functional heterostructures
US6613523B2 (en) 2001-06-29 2003-09-02 Agilent Technologies, Inc. Method of DNA sequencing using cleavable tags
US7682353B2 (en) 2001-06-29 2010-03-23 Coloplast A/S Catheter device
US7077152B2 (en) 2001-07-07 2006-07-18 Nanostream, Inc. Microfluidic metering systems and methods
US7799552B2 (en) 2001-07-20 2010-09-21 California Institute Of Technology Protein and nucleic acid expression systems
US6767731B2 (en) 2001-08-27 2004-07-27 Intel Corporation Electron induced fluorescent method for nucleic acid sequencing
WO2003031591A2 (en) 2001-10-10 2003-04-17 Superarray Bioscience Corporation Detecting targets by unique identifier nucleotide tags
US7297485B2 (en) 2001-10-15 2007-11-20 Qiagen Gmbh Method for nucleic acid amplification that results in low amplification bias
US6783647B2 (en) 2001-10-19 2004-08-31 Ut-Battelle, Llc Microfluidic systems and methods of transport and lysis of cells and analysis of cell lysate
EP1442131A4 (en) 2001-10-19 2006-06-07 Univ West Virginia Microfluidic system for proteome analysis
US20030149307A1 (en) 2001-10-24 2003-08-07 Baxter International Inc. Process for the preparation of polyethylene glycol bis amine
JP2005514224A (en) 2001-10-26 2005-05-19 アクララ バイオサイエンシーズ, インコーポレイテッド Systems and methods for injection micro-replication of microfluidic substrates
CA2466164A1 (en) 2001-10-30 2003-05-08 Nanomics Biosystems Pty, Ltd. Device and methods for directed synthesis of chemical libraries
US7262056B2 (en) 2001-11-08 2007-08-28 Mirus Bio Corporation Enhancing intermolecular integration of nucleic acids using integrator complexes
GB0127564D0 (en) 2001-11-16 2002-01-09 Medical Res Council Emulsion compositions
DE60235491D1 (en) 2001-11-28 2010-04-08 Bio Rad Laboratories PARALLEL SCORING OF POLYMORPHISMS WITH AMPLIFICATION AND ERROR CORRECTION
US7335153B2 (en) 2001-12-28 2008-02-26 Bio Array Solutions Ltd. Arrays of microparticles and methods of preparation thereof
AU2003210438A1 (en) 2002-01-04 2003-07-24 Board Of Regents, The University Of Texas System Droplet-based microfluidic oligonucleotide synthesis engine
WO2003062462A2 (en) 2002-01-16 2003-07-31 Dynal Biotech Asa Method for isolating nucleic acids and protein from a single sample
KR100459870B1 (en) 2002-02-22 2004-12-04 한국과학기술원 CONSTRUCTION OF NOVEL STRAINS CONTAINING MINIMIZING GENOME BY Tn5-COUPLED Cre/loxP EXCISION SYSTEM
ATE397096T1 (en) 2002-03-20 2008-06-15 Innovativebio Biz CONTROLLED PERMEABILITY MICROCapsules CONTAINING A NUCLEIC ACID AMPLIFICATION REACTION MIXTURE AND THEIR USE AS A REACTION VESSEL FOR PARALLEL REACTIONS
EP2283918B1 (en) 2002-05-09 2022-10-05 The University of Chicago Device and method for pressure-driven plug transport and reaction
US7901939B2 (en) 2002-05-09 2011-03-08 University Of Chicago Method for performing crystallization and reactions in pressure-driven fluid plugs
US7527966B2 (en) 2002-06-26 2009-05-05 Transgenrx, Inc. Gene regulation in transgenic animals using a transposon-based vector
JP2006507921A (en) 2002-06-28 2006-03-09 プレジデント・アンド・フェロウズ・オブ・ハーバード・カレッジ Method and apparatus for fluid dispersion
CA2493808A1 (en) 2002-07-24 2004-01-29 Ptc Therapeutics, Inc. Methods for identifying small molecules that modulate premature translation termination and nonsense mediated mrna decay
IL151660A0 (en) 2002-09-09 2003-04-10 Univ Ben Gurion Method for isolating and culturing unculturable microorganisms
WO2004031408A1 (en) 2002-09-30 2004-04-15 F.Hoffmann-La Roche Ag Oligonucleotides for genotyping thymidylate synthase gene
US20040081962A1 (en) 2002-10-23 2004-04-29 Caifu Chen Methods for synthesizing complementary DNA
US6979713B2 (en) 2002-11-25 2005-12-27 3M Innovative Properties Company Curable compositions and abrasive articles therefrom
US20050266582A1 (en) 2002-12-16 2005-12-01 Modlin Douglas N Microfluidic system with integrated permeable membrane
US20040248299A1 (en) 2002-12-27 2004-12-09 Sumedha Jayasena RNA interference
US7385995B2 (en) 2003-01-13 2008-06-10 Brooktree Broadband Holding, Inc. System and method for dynamic bandwidth allocation on PONs
US7700325B2 (en) 2003-01-17 2010-04-20 Trustees Of Boston University Haplotype analysis
EP2261372B1 (en) 2003-01-29 2012-08-22 454 Life Sciences Corporation Methods of amplifying and sequencing nucleic acids
EP1594973B1 (en) 2003-02-10 2011-12-07 Max-Delbrück-Centrum für Molekulare Medizin (MDC) Transposon-based targeting system
US7041481B2 (en) 2003-03-14 2006-05-09 The Regents Of The University Of California Chemical amplification based on fluid partitioning
US7316903B2 (en) 2003-03-28 2008-01-08 United States Of America As Represented By The Department Of Health And Human Services Detection of nucleic acid sequence variations using phase Mu transposase
GB0307403D0 (en) 2003-03-31 2003-05-07 Medical Res Council Selection by compartmentalised screening
GB0307428D0 (en) 2003-03-31 2003-05-07 Medical Res Council Compartmentalised combinatorial chemistry
US20060078893A1 (en) 2004-10-12 2006-04-13 Medical Research Council Compartmentalised combinatorial chemistry by microfluidic control
NZ542576A (en) 2003-04-04 2008-11-28 Pfizer Prod Inc Submicron oil-in-water emulsions composed of a non-metabolizable oil, at least one surfactant, and an aqueous component, where the oil is dispersed in the aqueous component with al droplet size in the submicron range
US20100035254A1 (en) 2003-04-08 2010-02-11 Pacific Biosciences Of California, Inc. Composition and method for nucleic acid sequencing
AU2004229440B2 (en) 2003-04-10 2010-08-12 President And Fellows Of Harvard College Formation and control of fluidic species
US20060275915A1 (en) 2003-05-16 2006-12-07 Global Technologies (Nz) Ltd. Method and apparatus for mixing sample and reagent in a suspension fluid
WO2004103565A2 (en) 2003-05-19 2004-12-02 Hans-Knöll-Institut für Naturstoff-Forschung e.V. Device and method for structuring liquids and for dosing reaction liquids into liquid compartments immersed in a separation medium
WO2004105734A1 (en) 2003-05-28 2004-12-09 Valorisation Recherche, Societe En Commandite Method of preparing microcapsules
GB0313170D0 (en) 2003-06-09 2003-07-16 Qinetiq Ltd Method and apparatus for spore disruption and/or detection
EP1636564A1 (en) 2003-06-13 2006-03-22 The General Hospital Corporation Microfluidic systems for size based removal of red blood cells and platelets from blood
GB2403475B (en) 2003-07-01 2008-02-06 Oxitec Ltd Stable integrands
GB0315438D0 (en) 2003-07-02 2003-08-06 Univ Manchester Analysis of mixed cell populations
WO2005010145A2 (en) 2003-07-05 2005-02-03 The Johns Hopkins University Method and compositions for detection and enumeration of genetic variations
EP1658133A1 (en) 2003-08-27 2006-05-24 President And Fellows Of Harvard College Electronic control of fluidic species
CA2542512A1 (en) 2003-09-04 2005-03-17 Nathan Ravi Hydrogel nanocompsites for ophthalmic applications
WO2005023427A1 (en) 2003-09-05 2005-03-17 Stokes Bio Limited A microfluidic analysis system
US7354706B2 (en) 2003-09-09 2008-04-08 The Regents Of The University Of Colorado, A Body Corporate Use of photopolymerization for amplification and detection of a molecular recognition event
JP4353945B2 (en) 2003-09-22 2009-10-28 独立行政法人理化学研究所 Efficient DNA inverted repeat structure preparation method
CN102928584B (en) 2003-09-25 2017-06-13 富山县政府 Microwell array chip and its manufacture method
JP2007512811A (en) 2003-11-10 2007-05-24 インベスチゲン, インコーポレイテッド Methods for preparing nucleic acids for detection
EP1691792A4 (en) 2003-11-24 2008-05-28 Yeda Res & Dev Compositions and methods for in vitro sorting of molecular and cellular libraries
US20050136417A1 (en) 2003-12-19 2005-06-23 Affymetrix, Inc. Amplification of nucleic acids
WO2005062881A2 (en) 2003-12-24 2005-07-14 Transgenrx, Inc. Gene therapy using transposon-based vectors
ES2432040T3 (en) 2004-01-28 2013-11-29 454 Life Sciences Corporation Nucleic acid amplification with continuous flow emulsion
US20050181379A1 (en) 2004-02-18 2005-08-18 Intel Corporation Method and device for isolating and positioning single nucleic acid molecules
US20100216153A1 (en) 2004-02-27 2010-08-26 Helicos Biosciences Corporation Methods for detecting fetal nucleic acids and diagnosing fetal abnormalities
JP2007526772A (en) 2004-02-27 2007-09-20 プレジデント・アンド・フェロウズ・オブ・ハーバード・カレッジ Polony fluorescent beads for in situ sequencing
KR100552706B1 (en) 2004-03-12 2006-02-20 삼성전자주식회사 Method and apparatus for nucleic acid amplification
JP4777238B2 (en) 2004-03-23 2011-09-21 独立行政法人科学技術振興機構 Method and apparatus for generating microdroplets
US20050221339A1 (en) 2004-03-31 2005-10-06 Medical Research Council Harvard University Compartmentalised screening by microfluidic control
US20060020371A1 (en) 2004-04-13 2006-01-26 President And Fellows Of Harvard College Methods and apparatus for manipulation and/or detection of biological samples and other objects
US20050250147A1 (en) 2004-05-10 2005-11-10 Macevicz Stephen C Digital profiling of polynucleotide populations
WO2006047183A2 (en) 2004-10-21 2006-05-04 New England Biolabs, Inc. Recombinant dna nicking endonuclease and uses thereof
US7799553B2 (en) 2004-06-01 2010-09-21 The Regents Of The University Of California Microfabricated integrated DNA analysis system
US7700281B2 (en) 2004-06-30 2010-04-20 Usb Corporation Hot start nucleic acid amplification
US7968085B2 (en) 2004-07-05 2011-06-28 Ascendis Pharma A/S Hydrogel formulations
CN1648671B (en) 2005-02-06 2012-09-26 成都夸常医学工业有限公司 Detecting method for multiple reactor analytic chip and analytic chip and detector
US7608434B2 (en) 2004-08-04 2009-10-27 Wisconsin Alumni Research Foundation Mutated Tn5 transposase proteins and the use thereof
WO2006030993A1 (en) 2004-09-14 2006-03-23 Jin-Ho Choy Information code system using dna sequences
US7892731B2 (en) 2004-10-01 2011-02-22 Radix Biosolutions, Ltd. System and method for inhibiting the decryption of a nucleic acid probe sequence used for the detection of a specific nucleic acid
US7968287B2 (en) 2004-10-08 2011-06-28 Medical Research Council Harvard University In vitro evolution in microfluidic systems
US9492400B2 (en) 2004-11-04 2016-11-15 Massachusetts Institute Of Technology Coated controlled release polymer particles as efficient oral delivery vehicles for biopharmaceuticals
WO2006051552A2 (en) 2004-11-15 2006-05-18 Yeda Research And Development Co. Ltd. At The Weizmann Institute Of Science Directed evolution and selection using in vitro compartmentalization
US7329493B2 (en) 2004-12-22 2008-02-12 Asiagen Corporation One-tube nested PCR for detecting Mycobacterium tuberculosis
JP5165383B2 (en) 2004-12-23 2013-03-21 アイ−スタツト・コーポレイシヨン Molecular diagnostic system and method
WO2006078841A1 (en) 2005-01-21 2006-07-27 President And Fellows Of Harvard College Systems and methods for forming fluidic droplets encapsulated in particles such as colloidal particles
US7579153B2 (en) 2005-01-25 2009-08-25 Population Genetics Technologies, Ltd. Isothermal DNA amplification
US7393665B2 (en) 2005-02-10 2008-07-01 Population Genetics Technologies Ltd Methods and compositions for tagging and identifying polynucleotides
US7407757B2 (en) 2005-02-10 2008-08-05 Population Genetics Technologies Genetic analysis by sequence-specific sorting
EP1871903B1 (en) 2005-02-18 2011-12-21 Canon U.S. Life Sciences, Inc. Devices and methods for identifying genomic dna of organisms
JP4649621B2 (en) 2005-02-21 2011-03-16 国立大学法人 鹿児島大学 Purification method of biodiesel fuel
US9040237B2 (en) 2005-03-04 2015-05-26 Intel Corporation Sensor arrays and nucleic acid sequencing applications
US20070054119A1 (en) 2005-03-04 2007-03-08 Piotr Garstecki Systems and methods of forming particles
JP2008535644A (en) 2005-03-04 2008-09-04 プレジデント・アンド・フエローズ・オブ・ハーバード・カレツジ Method and apparatus for the formation of multiple emulsions
JP2006289250A (en) 2005-04-08 2006-10-26 Kao Corp Micro mixer and fluid mixing method using the same
US8407013B2 (en) 2005-06-07 2013-03-26 Peter K. Rogan AB initio generation of single copy genomic probes
CA2611743C (en) 2005-06-15 2019-12-31 Callida Genomics, Inc. Nucleic acid analysis by forming and tracking aliquoted fragments of a target polynucleotide
JP2006349060A (en) 2005-06-16 2006-12-28 Ntn Corp Ball screw
WO2007002490A2 (en) 2005-06-22 2007-01-04 The Research Foundation Of State University Of New York Massively parallel 2-dimensional capillary electrophoresis
US20070154903A1 (en) 2005-06-23 2007-07-05 Nanosphere, Inc. Selective isolation and concentration of nucleic acids from complex samples
US9175295B2 (en) 2005-07-05 2015-11-03 The Chemo-Sero-Therapeutic Research Institute Modified transposon vector and its use
JP5051490B2 (en) 2005-07-08 2012-10-17 独立行政法人産業技術総合研究所 Inorganic microcapsule encapsulating macro-biomaterial and method for producing the same
US20070020640A1 (en) 2005-07-21 2007-01-25 Mccloskey Megan L Molecular encoding of nucleic acid templates for PCR and other forms of sequence analysis
FR2888912B1 (en) 2005-07-25 2007-08-24 Commissariat Energie Atomique METHOD FOR CONTROLLING COMMUNICATION BETWEEN TWO ZONES BY ELECTROWRINKING, DEVICE COMPRISING ISOLABLE ZONES AND OTHERS AND METHOD FOR PRODUCING SUCH DEVICE
ATE510930T1 (en) 2005-08-02 2011-06-15 Rubicon Genomics Inc COMPOSITIONS AND METHODS FOR EDITING AND AMPLIFICATION OF DNA USING MULTIPLE ENZYMES IN A SINGLE REACTION
DE102005037401B4 (en) 2005-08-08 2007-09-27 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Formation of an emulsion in a fluidic microsystem
WO2007024840A2 (en) 2005-08-22 2007-03-01 Critical Therapeutics, Inc. Method of quantitating nucleic acids by flow cytometry microparticle-based array
US7556776B2 (en) 2005-09-08 2009-07-07 President And Fellows Of Harvard College Microfluidic manipulation of fluids and reactions
JP2007074967A (en) 2005-09-13 2007-03-29 Canon Inc Identifier probe and method for amplifying nucleic acid by using the same
CN101523156A (en) 2005-09-16 2009-09-02 加利福尼亚大学董事会 A colorimetric bio-barcode amplification assay for analyte detection
US7960104B2 (en) 2005-10-07 2011-06-14 Callida Genomics, Inc. Self-assembled single molecule arrays and uses thereof
AU2007249635B2 (en) 2005-10-07 2012-05-31 Complete Genomics, Inc. High throughput genome sequencing on DNA arrays
US20070111241A1 (en) 2005-10-14 2007-05-17 Nezih Cereb System and method for accessing, tracking, and editing sequence analysis and software to accomplish the same
US7709544B2 (en) 2005-10-25 2010-05-04 Massachusetts Institute Of Technology Microstructure synthesis by flow lithography and polymerization
EP1954838B1 (en) 2005-11-14 2014-02-26 Life Technologies Corporation Coded molecules for detecting target analytes
US20070134277A1 (en) 2005-12-09 2007-06-14 Children's Medical Center Corporation Pharmaceutical formulation for sulfur-containing drugs in liquid dosage forms
US20070141584A1 (en) 2005-12-20 2007-06-21 Roberts Douglas N Methods for assessment of native chromatin on microarrays
US7932037B2 (en) 2007-12-05 2011-04-26 Perkinelmer Health Sciences, Inc. DNA assays using amplicon probes on encoded particles
WO2007081385A2 (en) 2006-01-11 2007-07-19 Raindance Technologies, Inc. Microfluidic devices and methods of use in the formation and control of nanoreactors
US7537897B2 (en) 2006-01-23 2009-05-26 Population Genetics Technologies, Ltd. Molecular counting
EP1987162A4 (en) 2006-01-23 2009-11-25 Population Genetics Technologi Nucleic acid analysis using sequence tokens
CA2640024A1 (en) 2006-01-27 2007-08-09 President And Fellows Of Harvard College Fluidic droplet coalescence
PT2385143T (en) 2006-02-02 2016-10-18 Univ Leland Stanford Junior Non-invasive fetal genetic screening by digital analysis
WO2007092538A2 (en) 2006-02-07 2007-08-16 President And Fellows Of Harvard College Methods for making nucleotide probes for sequencing and synthesis
GB0603251D0 (en) 2006-02-17 2006-03-29 Isis Innovation DNA conformation
SG170028A1 (en) 2006-02-24 2011-04-29 Callida Genomics Inc High throughput genome sequencing on dna arrays
WO2007111937A1 (en) 2006-03-23 2007-10-04 Applera Corporation Directed enrichment of genomic dna for high-throughput sequencing
JP4921829B2 (en) 2006-03-30 2012-04-25 株式会社東芝 Fine particle production apparatus, emulsifier holding part, fine particle production method, and molecular film production method
WO2007114794A1 (en) 2006-03-31 2007-10-11 Nam Trung Nguyen Active control for droplet-based microfluidics
JP2009538123A (en) 2006-04-19 2009-11-05 アプライド バイオシステムズ, エルエルシー Reagents, methods and libraries for gel-free bead-based sequencing
US7811603B2 (en) 2006-05-09 2010-10-12 The Regents Of The University Of California Microfluidic device for forming monodisperse lipoplexes
EP2530168B1 (en) 2006-05-11 2015-09-16 Raindance Technologies, Inc. Microfluidic Devices
US9074242B2 (en) 2010-02-12 2015-07-07 Raindance Technologies, Inc. Digital analyte analysis
EP2030011B1 (en) 2006-05-22 2017-02-01 Nanostring Technologies, Inc. Systems and methods for analyzing nanoreporters
RU2321638C2 (en) 2006-05-23 2008-04-10 Закрытое акционерное общество "Молекулярно-медицинские технологии" Method for preparing multifunctional multichip, multichip for successive or parallel screening biopolymers, method for analysis of biopolymers and set for realization of method
CA2653321A1 (en) 2006-05-26 2007-12-06 Althea Technologies, Inc. Biochemical analysis of partitioned cells
FR2901717A1 (en) 2006-05-30 2007-12-07 Centre Nat Rech Scient METHOD FOR TREATING DROPS IN A MICROFLUIDIC CIRCUIT
EP4108780A1 (en) 2006-06-14 2022-12-28 Verinata Health, Inc. Rare cell analysis using sample splitting and dna tags
CA2656022C (en) 2006-06-19 2017-10-17 The Johns Hopkins University Single-molecule pcr on microparticles in water-in-oil emulsions
US20080076909A1 (en) 2006-06-30 2008-03-27 Applera Corporation Emulsion pcr and amplicon capture
EP1878501A1 (en) 2006-07-14 2008-01-16 Roche Diagnostics GmbH Instrument for heating and cooling
US8394590B2 (en) 2006-08-02 2013-03-12 California Institute Of Technology Capture agents and related methods and systems for detecting and/or sorting targets
US9012390B2 (en) 2006-08-07 2015-04-21 Raindance Technologies, Inc. Fluorocarbon emulsion stabilizing surfactants
WO2008030433A2 (en) 2006-09-06 2008-03-13 Canon U.S. Life Sciences, Inc. Chip and cartridge design configuration for performing micro-fluidic assays
AU2007302586B2 (en) 2006-09-25 2013-06-27 Archer-Daniels-Midland Company Superabsorbent surface-treated carboxyalkylated polysaccharides and process for producing same
US7935518B2 (en) 2006-09-27 2011-05-03 Alessandra Luchini Smart hydrogel particles for biomarker harvesting
US20080166720A1 (en) 2006-10-06 2008-07-10 The Regents Of The University Of California Method and apparatus for rapid nucleic acid analysis
US8841116B2 (en) 2006-10-25 2014-09-23 The Regents Of The University Of California Inline-injection microdevice and microfabricated integrated DNA analysis system using same
WO2008070352A2 (en) 2006-10-27 2008-06-12 Complete Genomics, Inc. Efficient arrays of amplified polynucleotides
WO2008061165A2 (en) 2006-11-14 2008-05-22 Handylab, Inc. Microfluidic cartridge and method of making same
DK2518162T3 (en) 2006-11-15 2018-06-18 Biospherex Llc Multi-tag sequencing and ecogenomic analysis
CN101583879A (en) 2006-11-20 2009-11-18 纳提维斯公司 Apparatus and method for transducing an in vitro or mammalian system with a low-frequency signal
US20080242560A1 (en) 2006-11-21 2008-10-02 Gunderson Kevin L Methods for generating amplified nucleic acid arrays
JP5320546B2 (en) 2006-12-13 2013-10-23 国立大学法人名古屋大学 Tol1 element transposase and DNA introduction system using the same
CN101669026B (en) 2006-12-14 2014-05-07 生命技术公司 Methods and apparatus for measuring analytes using large scale FET arrays
US8338166B2 (en) 2007-01-04 2012-12-25 Lawrence Livermore National Security, Llc Sorting, amplification, detection, and identification of nucleic acid subsequences in a complex mixture
JP2008167722A (en) 2007-01-15 2008-07-24 Konica Minolta Medical & Graphic Inc Nucleic acid isolation method by heating on magnetic support
US7844658B2 (en) 2007-01-22 2010-11-30 Comcast Cable Holdings, Llc System and method for providing an application to a device
US20080176768A1 (en) 2007-01-23 2008-07-24 Honeywell Honeywell International Hydrogel microarray with embedded metal nanoparticles
EP2121983A2 (en) 2007-02-02 2009-11-25 Illumina Cambridge Limited Methods for indexing samples and sequencing multiple nucleotide templates
US8003312B2 (en) 2007-02-16 2011-08-23 The Board Of Trustees Of The Leland Stanford Junior University Multiplex cellular assays using detectable cell barcodes
FI20075124A0 (en) 2007-02-21 2007-02-21 Valtion Teknillinen Method and test kit for detection of nucleotide variations
WO2008109176A2 (en) 2007-03-07 2008-09-12 President And Fellows Of Harvard College Assays and other reactions involving droplets
US20080228268A1 (en) 2007-03-15 2008-09-18 Uluru, Inc. Method of Formation of Viscous, Shape Conforming Gels and Their Uses as Medical Prosthesis
US7943330B2 (en) 2007-03-23 2011-05-17 Academia Sinica Tailored glycoproteomic methods for the sequencing, mapping and identification of cellular glycoproteins
WO2008121342A2 (en) 2007-03-28 2008-10-09 President And Fellows Of Harvard College Emulsions and techniques for formation
US20100130369A1 (en) 2007-04-23 2010-05-27 Advanced Liquid Logic, Inc. Bead-Based Multiplexed Analytical Methods and Instrumentation
CN101293191B (en) 2007-04-25 2011-11-09 中国科学院过程工程研究所 Agarose gelatin microsphere preparation method
WO2008135512A2 (en) 2007-05-02 2008-11-13 Jerzy Paszkowski Dna amplification method
US20080295909A1 (en) 2007-05-24 2008-12-04 Locascio Laurie E Microfluidic Device for Passive Sorting and Storage of Liquid Plugs Using Capillary Force
CN101720359A (en) 2007-06-01 2010-06-02 454生命科学公司 System and meth0d for identification of individual samples from a multiplex mixture
US8476382B2 (en) 2007-06-05 2013-07-02 Eugenia Kumacheva Multiple continuous microfluidic reactors for the scaled up synthesis of gel or polymer particles
WO2009005680A1 (en) 2007-06-29 2009-01-08 President And Fellows Of Harvard College Methods and apparatus for manipulation of fluidic species
WO2009011808A1 (en) 2007-07-13 2009-01-22 President And Fellows Of Harvard College Droplet-based selection
WO2009015296A1 (en) 2007-07-24 2009-01-29 The Regents Of The University Of California Microfabricated dropley generator
US20130084243A1 (en) 2010-01-27 2013-04-04 Liliane Goetsch Igf-1r specific antibodies useful in the detection and diagnosis of cellular proliferative disorders
WO2009023821A1 (en) 2007-08-15 2009-02-19 Opgen, Inc. Method, system and software arrangement for comparative analysis and phylogeny with whole-genome optical maps
US8563527B2 (en) 2007-08-20 2013-10-22 Pharmain Corporation Oligonucleotide core carrier compositions for delivery of nucleic acid-containing therapeutic agents, methods of making and using the same
US8268564B2 (en) 2007-09-26 2012-09-18 President And Fellows Of Harvard College Methods and applications for stitched DNA barcodes
WO2009048532A2 (en) 2007-10-05 2009-04-16 President And Fellows Of Harvard College Formation of particles for ultrasound application, drug release, and other uses, and microfluidic methods of preparation
US20090099040A1 (en) 2007-10-15 2009-04-16 Sigma Aldrich Company Degenerate oligonucleotides and their uses
US20100086914A1 (en) 2008-10-03 2010-04-08 Roche Molecular Systems, Inc. High resolution, high throughput hla genotyping by clonal sequencing
WO2009061372A1 (en) 2007-11-02 2009-05-14 President And Fellows Of Harvard College Systems and methods for creating multi-phase entities, including particles and/or fluids
US8334013B2 (en) 2007-11-02 2012-12-18 Stc.Unm Mesoporous metal oxide microspheres and method for forming same
US8592150B2 (en) 2007-12-05 2013-11-26 Complete Genomics, Inc. Methods and compositions for long fragment read sequencing
WO2009076485A2 (en) 2007-12-10 2009-06-18 Xiaolian Gao Sequencing of nucleic acids
US7771944B2 (en) 2007-12-14 2010-08-10 The Board Of Trustees Of The University Of Illinois Methods for determining genetic haplotypes and DNA mapping
CN101946010B (en) 2007-12-21 2014-08-20 哈佛大学 Systems and methods for nucleic acid sequencing
EP3360972B1 (en) 2008-01-17 2019-12-11 Sequenom, Inc. Single molecule nucleic acid sequence analysis processes
KR20090081260A (en) 2008-01-23 2009-07-28 삼성전자주식회사 Assay method of microarray hybridization
WO2009100382A1 (en) 2008-02-07 2009-08-13 Pacific Biosciences Of California, Inc. Cis reactive oxygen quenchers integrated into linkers
JP5468271B2 (en) 2008-02-08 2014-04-09 花王株式会社 Method for producing fine particle dispersion
US20090203531A1 (en) 2008-02-12 2009-08-13 Nurith Kurn Method for Archiving and Clonal Expansion
AU2009224170B2 (en) 2008-03-11 2012-03-29 National Cancer Center Method for measuring chromosome, gene or specific nucleotide sequence copy numbers using SNP array
US9011777B2 (en) 2008-03-21 2015-04-21 Lawrence Livermore National Security, Llc Monodisperse microdroplet generation and stopping without coalescence
US8961902B2 (en) 2008-04-23 2015-02-24 Bioscale, Inc. Method and apparatus for analyte processing
US9068181B2 (en) 2008-05-23 2015-06-30 The General Hospital Corporation Microfluidic droplet encapsulation
DE102008025656B4 (en) 2008-05-28 2016-07-28 Genxpro Gmbh Method for the quantitative analysis of nucleic acids, markers therefor and their use
GB0810051D0 (en) 2008-06-02 2008-07-09 Oxford Biodynamics Ltd Method of diagnosis
WO2009148598A1 (en) 2008-06-05 2009-12-10 President And Fellows Of Harvard College Polymersomes, colloidosomes, liposomes, and other species associated with fluidic droplets
WO2010003132A1 (en) 2008-07-02 2010-01-07 Illumina Cambridge Ltd. Using populations of beads for the fabrication of arrays on surfaces
WO2010004018A2 (en) 2008-07-11 2010-01-14 Eth Zurich Degradable microcapsules
EP4047367A1 (en) 2008-07-18 2022-08-24 Bio-Rad Laboratories, Inc. Method for detecting target analytes with droplet libraries
WO2010009735A2 (en) 2008-07-23 2010-01-28 Dako Denmark A/S Combinatorial analysis and repair
US20100062494A1 (en) 2008-08-08 2010-03-11 President And Fellows Of Harvard College Enzymatic oligonucleotide pre-adenylation
US8383345B2 (en) 2008-09-12 2013-02-26 University Of Washington Sequence tag directed subassembly of short sequencing reads into long sequencing reads
WO2010033200A2 (en) 2008-09-19 2010-03-25 President And Fellows Of Harvard College Creation of libraries of droplets and related species
US9764322B2 (en) 2008-09-23 2017-09-19 Bio-Rad Laboratories, Inc. System for generating droplets with pressure monitoring
US20110159499A1 (en) 2009-11-25 2011-06-30 Quantalife, Inc. Methods and compositions for detecting genetic material
US9156010B2 (en) 2008-09-23 2015-10-13 Bio-Rad Laboratories, Inc. Droplet-based assay system
US20120252015A1 (en) 2011-02-18 2012-10-04 Bio-Rad Laboratories Methods and compositions for detecting genetic material
US8709762B2 (en) 2010-03-02 2014-04-29 Bio-Rad Laboratories, Inc. System for hot-start amplification via a multiple emulsion
US9417190B2 (en) 2008-09-23 2016-08-16 Bio-Rad Laboratories, Inc. Calibrations and controls for droplet-based assays
CN104224703A (en) 2008-09-25 2014-12-24 赛福伦公司 Liquid Formulations Of Bendamustine
US8361299B2 (en) 2008-10-08 2013-01-29 Sage Science, Inc. Multichannel preparative electrophoresis system
US9080211B2 (en) 2008-10-24 2015-07-14 Epicentre Technologies Corporation Transposon end compositions and methods for modifying nucleic acids
EP3272879B1 (en) 2008-10-24 2019-08-07 Epicentre Technologies Corporation Transposon end compositions and methods for modifying nucleic acids
US20100113296A1 (en) 2008-11-05 2010-05-06 Joel Myerson Methods And Kits For Nucleic Acid Analysis
US8748103B2 (en) 2008-11-07 2014-06-10 Sequenta, Inc. Monitoring health and disease status using clonotype profiles
US20100203647A1 (en) 2008-11-21 2010-08-12 The Rockefeller University Chemical Reporters of Protein Acylation
EP2352852A4 (en) 2008-12-02 2012-10-24 Bio Rad Laboratories Chromatin structure detection
WO2010080134A1 (en) 2008-12-19 2010-07-15 President And Fellows Of Harvard College Particle-assisted nucleic acid sequencing
WO2010075570A2 (en) 2008-12-24 2010-07-01 New York University Methods, computer-accessible medium, and systems for score-driven whole-genome shotgun sequence assemble
KR101065807B1 (en) 2009-01-23 2011-09-19 충남대학교산학협력단 Preparation method for micro-capsule using a microfluidic chip system
US9347092B2 (en) 2009-02-25 2016-05-24 Roche Molecular System, Inc. Solid support for high-throughput nucleic acid analysis
JP5457222B2 (en) 2009-02-25 2014-04-02 エフ.ホフマン−ラ ロシュ アーゲー Miniaturized high-throughput nucleic acid analysis
KR101793744B1 (en) 2009-03-13 2017-11-03 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Scale-up of flow-focusing microfluidic devices
WO2010104604A1 (en) 2009-03-13 2010-09-16 President And Fellows Of Harvard College Method for the controlled creation of emulsions, including multiple emulsions
EP2230312A1 (en) 2009-03-19 2010-09-22 Helmholtz-Zentrum für Infektionsforschung GmbH Probe compound for detecting and isolating enzymes and means and methods using the same
EP3415235A1 (en) 2009-03-23 2018-12-19 Raindance Technologies Inc. Manipulation of microfluidic droplets
DK3495498T3 (en) 2009-03-30 2022-01-17 Illumina Inc GENE EXPRESSION ANALYSIS IN SINGLE CELLS
CN102439177B (en) 2009-04-02 2014-10-01 弗卢伊蒂格姆公司 Multi-primer amplification method for barcoding of target nucleic acids
WO2010127186A1 (en) 2009-04-30 2010-11-04 Prognosys Biosciences, Inc. Nucleic acid constructs and methods of use
EP2427572B1 (en) 2009-05-01 2013-08-28 Illumina, Inc. Sequencing methods
US9334531B2 (en) 2010-12-17 2016-05-10 Life Technologies Corporation Nucleic acid amplification
FR2945545B1 (en) 2009-05-14 2011-08-05 Univ Aix Marseille Ii METHOD FOR DETECTION OF PROCARYOTE DNA EXTRACTED FROM A SAMPLE SAMPLE
FR2945819B1 (en) 2009-05-19 2011-06-17 Commissariat Energie Atomique DEVICE AND METHOD FOR ISOLATING BIOLOGICAL OR CHEMICAL TARGETS
US8574835B2 (en) 2009-05-29 2013-11-05 Life Technologies Corporation Scaffolded nucleic acid polymer particles and methods of making and using
EP2443236B1 (en) 2009-06-15 2015-05-13 Complete Genomics, Inc. Methods and compositions for long fragment read sequencing
US9524369B2 (en) 2009-06-15 2016-12-20 Complete Genomics, Inc. Processing and analysis of complex nucleic acid sequence data
WO2010151776A2 (en) 2009-06-26 2010-12-29 President And Fellows Of Harvard College Fluid injection
US20110028412A1 (en) 2009-08-03 2011-02-03 Cappellos, Inc. Herbal enhanced analgesic formulations
US20110033548A1 (en) 2009-08-05 2011-02-10 E.I. Du Pont De Nemours And Company Degradable crosslinked aminated dextran microspheres and methods of use
WO2011021102A2 (en) 2009-08-20 2011-02-24 Population Genetics Technologies Ltd Compositions and methods for intramolecular nucleic acid rearrangement
JP2013503605A (en) 2009-09-01 2013-02-04 コーニンクレッカ フィリップス エレクトロニクス エヌ ヴィ Devices and methods for microarray selection
EP2473618B1 (en) 2009-09-02 2015-03-04 Bio-Rad Laboratories, Inc. System for mixing fluids by coalescence of multiple emulsions
BR112012004719A2 (en) 2009-09-02 2016-04-05 Harvard College multiple emulsions created by blasting and other techniques
US9625454B2 (en) 2009-09-04 2017-04-18 The Research Foundation For The State University Of New York Rapid and continuous analyte processing in droplet microfluidic devices
GB0918564D0 (en) 2009-10-22 2009-12-09 Plasticell Ltd Nested cell encapsulation
AU2010315580B2 (en) 2009-10-27 2014-11-06 President And Fellows Of Harvard College Droplet creation techniques
WO2011056872A2 (en) 2009-11-03 2011-05-12 Gen9, Inc. Methods and microfluidic devices for the manipulation of droplets in high fidelity polynucleotide assembly
EP2496700B1 (en) 2009-11-04 2017-03-01 The University Of British Columbia Nucleic acid-containing lipid particles and related methods
CN102985552B (en) 2009-11-25 2016-02-17 伯乐生命医学产品有限公司 For detecting the method and composition of genetic material
US9023769B2 (en) 2009-11-30 2015-05-05 Complete Genomics, Inc. cDNA library for nucleic acid sequencing
US8835358B2 (en) 2009-12-15 2014-09-16 Cellular Research, Inc. Digital counting of individual molecules by stochastic attachment of diverse labels
US8932812B2 (en) 2009-12-17 2015-01-13 Keygene N.V. Restriction enzyme based whole genome sequencing
EP2517025B1 (en) 2009-12-23 2019-11-27 Bio-Rad Laboratories, Inc. Methods for reducing the exchange of molecules between droplets
JP5901046B2 (en) 2010-02-19 2016-04-06 国立大学法人 千葉大学 Novel alternative splicing variant of OATP1B3 mRNA
US20110257889A1 (en) 2010-02-24 2011-10-20 Pacific Biosciences Of California, Inc. Sequence assembly and consensus sequence determination
US20130203606A1 (en) 2010-02-25 2013-08-08 Advanced Liquid Logic Inc Method of Preparing a Nucleic Acid Library
US8236574B2 (en) 2010-03-01 2012-08-07 Quanterix Corporation Ultra-sensitive detection of molecules or particles using beads or other capture objects
WO2011119881A1 (en) 2010-03-24 2011-09-29 Northeastern University Multi-compartmental macrophage delivery
CA2767182C (en) 2010-03-25 2020-03-24 Bio-Rad Laboratories, Inc. Droplet generation for droplet-based assays
FR2958186A1 (en) 2010-03-30 2011-10-07 Ecole Polytech DEVICE FOR FORMING DROPS IN A MICROFLUID CIRCUIT.
CA2794522C (en) 2010-04-05 2019-11-26 Prognosys Biosciences, Inc. Spatially encoded biological assays
US9255291B2 (en) 2010-05-06 2016-02-09 Bioo Scientific Corporation Oligonucleotide ligation methods for improving data quality and throughput using massively parallel sequencing
US20110287947A1 (en) 2010-05-18 2011-11-24 University Of Southern California Tethered Conformation Capture
CN107029223A (en) 2010-05-26 2017-08-11 西莱克塔生物科技公司 Synthesize nano-carrier combined vaccine
US20120000777A1 (en) 2010-06-04 2012-01-05 The Regents Of The University Of California Devices and methods for forming double emulsion droplet compositions and polymer particles
EP2580378A4 (en) 2010-06-08 2014-01-01 Nugen Technologies Inc Methods and composition for multiplex sequencing
US8703493B2 (en) 2010-06-15 2014-04-22 Src, Inc. Location analysis using fire retardant-protected nucleic acid-labeled tags
US20120003657A1 (en) 2010-07-02 2012-01-05 Samuel Myllykangas Targeted sequencing library preparation by genomic dna circularization
WO2012012037A1 (en) 2010-07-19 2012-01-26 New England Biolabs, Inc. Oligonucleotide adaptors: compositions and methods of use
US20120034603A1 (en) 2010-08-06 2012-02-09 Tandem Diagnostics, Inc. Ligation-based detection of genetic variants
CN103202812B (en) 2010-08-09 2015-10-28 南京大学 A kind of method of protein nano grain for the preparation of sending pharmacological active substance in body
WO2012019765A1 (en) 2010-08-10 2012-02-16 European Molecular Biology Laboratory (Embl) Methods and systems for tracking samples and sample combinations
WO2012037358A1 (en) 2010-09-16 2012-03-22 The University Of North Carolina At Chapel Hill Asymmetric bifunctional silyl monomers and particles thereof as prodrugs and delivery vehicles for pharmaceutical, chemical and biological agents
CN102409048B (en) 2010-09-21 2013-10-23 深圳华大基因科技服务有限公司 DNA index library building method based on high throughput sequencing
CN110878345A (en) 2010-09-21 2020-03-13 安捷伦科技有限公司 Increasing confidence in allele calls by molecular counting
EP2625526B1 (en) 2010-10-04 2017-03-15 Genapsys Inc. Systems and methods for automated reusable parallel biological reactions
US9999886B2 (en) 2010-10-07 2018-06-19 The Regents Of The University Of California Methods and systems for on demand droplet generation and impedance based detection
US9902950B2 (en) 2010-10-08 2018-02-27 President And Fellows Of Harvard College High-throughput single cell barcoding
EP3561073A1 (en) 2010-10-08 2019-10-30 President and Fellows of Harvard College High-throughput immune sequencing
CN102050953B (en) 2010-10-18 2012-11-07 武汉理工大学 Method for preparing reducible and degradable supermolecule hydrogel
EP2633069B1 (en) 2010-10-26 2015-07-01 Illumina, Inc. Sequencing methods
US20130225623A1 (en) 2010-10-27 2013-08-29 Mount Sinai School Of Medicine Methods of Treating Psychiatric or Neurological Disorders with MGLUR Antagonists
DE202011110979U1 (en) 2010-11-01 2017-12-04 Bio-Rad Laboratories, Inc. System for forming emulsions
CA2821299C (en) 2010-11-05 2019-02-12 Frank J. Steemers Linking sequence reads using paired code tags
US8829171B2 (en) 2011-02-10 2014-09-09 Illumina, Inc. Linking sequence reads using paired code tags
US9074251B2 (en) 2011-02-10 2015-07-07 Illumina, Inc. Linking sequence reads using paired code tags
US20140057799A1 (en) 2010-12-16 2014-02-27 Gigagen System and Methods for Massively Parallel Analysis of Nucleic Acids in Single Cells
US9163281B2 (en) 2010-12-23 2015-10-20 Good Start Genetics, Inc. Methods for maintaining the integrity and identification of a nucleic acid template in a multiplex sequencing reaction
CA2822439A1 (en) 2010-12-23 2012-06-28 Sequenom, Inc. Fetal genetic variation detection
WO2012100216A2 (en) 2011-01-20 2012-07-26 Knome, Inc. Methods and apparatus for assigning a meaningful numeric value to genomic variants, and searching and assessing same
US8765455B2 (en) 2011-01-27 2014-07-01 Lawrence Livermore National Security, Llc Chip-based droplet sorting
GB201101429D0 (en) 2011-01-27 2011-03-16 Biocompatibles Uk Ltd Drug delivery system
ES2762866T3 (en) 2011-01-28 2020-05-26 Illumina Inc Nucleotide replacement by doubly-labeled and directional libraries
US10144950B2 (en) 2011-01-31 2018-12-04 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
WO2012106546A2 (en) 2011-02-02 2012-08-09 University Of Washington Through Its Center For Commercialization Massively parallel continguity mapping
CA2826748C (en) 2011-02-09 2020-08-04 Bio-Rad Laboratories, Inc. Method of detecting variations in copy number of a target nucleic acid
EP3412778A1 (en) 2011-02-11 2018-12-12 Raindance Technologies, Inc. Methods for forming mixed droplets
US9266104B2 (en) 2011-02-11 2016-02-23 Raindance Technologies, Inc. Thermocycling device for nucleic acid amplification and methods of use
WO2012112804A1 (en) 2011-02-18 2012-08-23 Raindance Technoligies, Inc. Compositions and methods for molecular labeling
WO2012116250A1 (en) 2011-02-25 2012-08-30 University Of Massachusetts Medical School Monomers and polymers for functional polycarbonates and poly (ester-carbonates) and peg-co-polycarbonate hydrogels
WO2012116331A2 (en) 2011-02-25 2012-08-30 Illumina, Inc. Methods and systems for haplotype determination
EP2681196B1 (en) 2011-03-04 2015-09-09 Life Technologies Corporation Compounds and methods for conjugation of biomolecules
US8982879B2 (en) 2011-03-09 2015-03-17 Annai Systems Inc. Biological data networks and methods therefor
CA2830443C (en) 2011-03-18 2021-11-16 Bio-Rad Laboratories, Inc. Multiplexed digital assays with combinatorial use of signals
US9260753B2 (en) 2011-03-24 2016-02-16 President And Fellows Of Harvard College Single cell nucleic acid detection and analysis
GB2489714B (en) 2011-04-05 2013-11-06 Tracesa Ltd Fluid Identification Method
GB201106254D0 (en) 2011-04-13 2011-05-25 Frisen Jonas Method and product
CN107368705B (en) 2011-04-14 2021-07-13 完整基因有限公司 Method and computer system for analyzing genomic DNA of organism
CA2834291A1 (en) 2011-04-25 2012-11-01 Biorad Laboratories, Inc. Methods and compositions for nucleic acid analysis
CA3078215A1 (en) 2011-04-28 2012-11-01 Department Of Veterans Affairs Identification of polynucleotides associated with a sample
CN106912197B (en) 2011-04-28 2022-01-25 生命技术公司 Methods and compositions for multiplex PCR
EP2705156B1 (en) 2011-05-05 2015-08-26 Institut National de la Santé et de la Recherche Médicale (INSERM) Linear dna amplification
WO2012156919A1 (en) 2011-05-16 2012-11-22 Koninklijke Philips Electronics N.V. Bio-orthogonal drug activation
US20140227706A1 (en) 2011-05-16 2014-08-14 Dna Chip Research Inc. Method for assessing progression of clinical state of malignant neoplasm by quantitative detection of DNA in blood
BR112013029729A2 (en) 2011-05-23 2017-01-24 Basf Se emulsion control including multiple emulsions
US9005935B2 (en) 2011-05-23 2015-04-14 Agilent Technologies, Inc. Methods and compositions for DNA fragmentation and tagging by transposases
CN103890191B (en) 2011-05-27 2018-12-04 哈佛大学校长及研究员协会 Single cell whole genome amplification method
US8841071B2 (en) 2011-06-02 2014-09-23 Raindance Technologies, Inc. Sample multiplexing
WO2012167142A2 (en) 2011-06-02 2012-12-06 Raindance Technolgies, Inc. Enzyme quantification
US9150916B2 (en) 2011-06-24 2015-10-06 Beat Christen Compositions and methods for identifying the essential genome of an organism
US8927218B2 (en) 2011-06-27 2015-01-06 Flir Systems, Inc. Methods and compositions for segregating target nucleic acid from mixed nucleic acid samples
WO2013006824A2 (en) 2011-07-07 2013-01-10 Life Technologies Corporation Polymer particles, nucleic acid polymer particles and methods of making and using the same
US20130017978A1 (en) 2011-07-11 2013-01-17 Finnzymes Oy Methods and transposon nucleic acids for generating a dna library
US8658430B2 (en) 2011-07-20 2014-02-25 Raindance Technologies, Inc. Manipulating droplet size
US9605304B2 (en) 2011-07-20 2017-03-28 The Hong Kong Polytechnic University Ultra-stable oligonucleotide-gold and-silver nanoparticle conjugates and method of their preparation
US20130189700A1 (en) 2011-07-25 2013-07-25 Bio-Rad Laboratories, Inc. Breakage of an emulsion containing nucleic acid
EP2737089B1 (en) 2011-07-29 2017-09-06 Bio-rad Laboratories, Inc. Library characterization by digital assay
CA2844056A1 (en) 2011-08-04 2013-02-07 Sage Science, Inc. Systems and methods for processing fluids
WO2013022961A1 (en) 2011-08-08 2013-02-14 3The Broad Institute Compositions and methods for co-amplifying subsequences of a nucleic acid fragment sequence
US8846883B2 (en) 2011-08-16 2014-09-30 University Of Southhampton Oligonucleotide ligation
WO2013035114A1 (en) 2011-09-08 2013-03-14 Decode Genetics Ehf Tp53 genetic variants predictive of cancer
US9249460B2 (en) 2011-09-09 2016-02-02 The Board Of Trustees Of The Leland Stanford Junior University Methods for obtaining a sequence
CN107058059B (en) 2011-09-26 2020-08-07 基因技术股份公司 Efficient small volume nucleic acid synthesis
SG11201401810QA (en) 2011-09-28 2014-10-30 Harvard College Systems and methods for droplet production and/or fluidic manipulation
US9514272B2 (en) 2011-10-12 2016-12-06 Complete Genomics, Inc. Identification of DNA fragments and structural variations
US9469874B2 (en) 2011-10-18 2016-10-18 The Regents Of The University Of California Long-range barcode labeling-sequencing
US20130109576A1 (en) 2011-10-28 2013-05-02 Anthony P. Shuber Methods for detecting mutations
WO2013066416A1 (en) 2011-11-04 2013-05-10 Intel Corporation Signaling for configuration of downlink coordinated multipoint communications
CN103946548B (en) 2011-11-16 2016-10-05 国际商业机器公司 There is the microfluidic device of deformable valve
US9938524B2 (en) 2011-11-22 2018-04-10 Active Motif, Inc. Multiplex isolation of protein-associated nucleic acids
US10689643B2 (en) 2011-11-22 2020-06-23 Active Motif, Inc. Targeted transposition for use in epigenetic studies
SG11201402558QA (en) 2011-12-03 2014-06-27 Emd Millipore Corp Micro-incubation systems for microfluidic cell culture and methods
WO2013096851A1 (en) 2011-12-22 2013-06-27 President And Fellows Of Harvard College Compositions and methods for analyte detection
WO2013096643A1 (en) 2011-12-23 2013-06-27 Gigagen Methods and apparatuses for droplet mixing
KR102019297B1 (en) 2012-02-09 2019-09-06 라이프 테크놀로지스 코포레이션 Hydrophilic polymeric particles and methods for making same
JP2015507928A (en) 2012-02-14 2015-03-16 ザ・ジョンズ・ホプキンス・ユニバーシティ MIRNA analysis method
US9090662B2 (en) 2012-02-15 2015-07-28 Wisconsin Alumni Research Foundation Dithioamine reducing agents
WO2013123125A1 (en) 2012-02-17 2013-08-22 President And Fellows Of Harvard College Assembly of nucleic acid sequences in emulsions
DK3363901T3 (en) 2012-02-17 2021-02-22 Hutchinson Fred Cancer Res Compositions and methods for accurately identifying mutations
EP2817418B1 (en) 2012-02-24 2017-10-11 Raindance Technologies, Inc. Labeling and sample preparation for sequencing
LT3305918T (en) 2012-03-05 2020-09-25 President And Fellows Of Harvard College Methods for epigenetic sequencing
NO2694769T3 (en) 2012-03-06 2018-03-03
US9552458B2 (en) 2012-03-16 2017-01-24 The Research Institute At Nationwide Children's Hospital Comprehensive analysis pipeline for discovery of human genetic variation
WO2013148189A1 (en) 2012-03-30 2013-10-03 Massachusetts Institute Of Technology Probe incorporation mediated by enzymes
EP2647426A1 (en) 2012-04-03 2013-10-09 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Replication of distributed nucleic acid molecules with preservation of their relative distribution through hybridization-based binding
US8209130B1 (en) 2012-04-04 2012-06-26 Good Start Genetics, Inc. Sequence assembly
MX356508B (en) 2012-04-16 2018-05-30 Biological dynamics inc Nucleic acid sample preparation.
US20130296173A1 (en) 2012-04-23 2013-11-07 Complete Genomics, Inc. Pre-anchor wash
WO2013177220A1 (en) 2012-05-21 2013-11-28 The Scripps Research Institute Methods of sample preparation
WO2013177046A1 (en) 2012-05-21 2013-11-28 Solulink, Inc. Methods and/or use of oligonucleotide conjugates for suppressing background due to cross-hybridization
CA2875695C (en) 2012-06-15 2022-11-15 The Board Of Regents Of The University Of Texas System High throughput sequencing of multiple transcripts of a single cell
JP2015519909A (en) 2012-06-15 2015-07-16 アダプティブ バイオテクノロジーズ コーポレイション Uniquely tagged rearranged adaptive immune receptor genes in a complex gene set
AU2013293240A1 (en) 2012-07-24 2015-03-05 Adaptive Biotechnologies Corp. Single cell analysis using sequence tags
JP6525872B2 (en) 2012-08-08 2019-06-05 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Increasing dynamic range to identify multiple epitopes in cells
EP2882872B1 (en) 2012-08-13 2021-10-06 The Regents of The University of California Methods and systems for detecting biological components
US20150005199A1 (en) 2012-08-14 2015-01-01 10X Technologies, Inc. Compositions and methods for sample processing
US20140378349A1 (en) 2012-08-14 2014-12-25 10X Technologies, Inc. Compositions and methods for sample processing
CA2881685C (en) 2012-08-14 2023-12-05 10X Genomics, Inc. Microcapsule compositions and methods
US9567631B2 (en) 2012-12-14 2017-02-14 10X Genomics, Inc. Methods and systems for processing polynucleotides
US20140378345A1 (en) 2012-08-14 2014-12-25 10X Technologies, Inc. Compositions and methods for sample processing
US10752949B2 (en) 2012-08-14 2020-08-25 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9951386B2 (en) 2014-06-26 2018-04-24 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10323279B2 (en) 2012-08-14 2019-06-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9701998B2 (en) 2012-12-14 2017-07-11 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9388465B2 (en) 2013-02-08 2016-07-12 10X Genomics, Inc. Polynucleotide barcode generation
US10584381B2 (en) 2012-08-14 2020-03-10 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10273541B2 (en) 2012-08-14 2019-04-30 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11591637B2 (en) 2012-08-14 2023-02-28 10X Genomics, Inc. Compositions and methods for sample processing
US20150005200A1 (en) 2012-08-14 2015-01-01 10X Technologies, Inc. Compositions and methods for sample processing
US20140378322A1 (en) 2012-08-14 2014-12-25 10X Technologies, Inc. Compositions and methods for sample processing
US10221442B2 (en) 2012-08-14 2019-03-05 10X Genomics, Inc. Compositions and methods for sample processing
WO2014047561A1 (en) 2012-09-21 2014-03-27 The Broad Institute Inc. Compositions and methods for labeling of agents
US9644199B2 (en) 2012-10-01 2017-05-09 Agilent Technologies, Inc. Immobilized transposase complexes for DNA fragmentation and tagging
GB201217772D0 (en) 2012-10-04 2012-11-14 Base4 Innovation Ltd Sequencing method
FR2996544B1 (en) 2012-10-08 2015-03-13 Ecole Polytech MICROFLUIDIC CIRCUIT FOR COMBINING DROPS OF MULTIPLE FLUIDS AND CORRESPONDING MICROFLUIDIC PROCESS.
FR2996545B1 (en) 2012-10-08 2016-03-25 Ecole Polytech MICROFLUIDIC METHOD FOR PROCESSING AND ANALYZING A SOLUTION CONTAINING BIOLOGICAL MATERIAL, AND CORRESPONDING MICROFLUIDIC CIRCUIT
US9133510B2 (en) 2012-10-15 2015-09-15 Life Technologies Corporation Compositions, methods, systems and kits for target nucleic acid enrichment
EP3901280A1 (en) 2012-10-17 2021-10-27 Spatial Transcriptomics AB Methods and product for optimising localised or spatial detection of gene expression in a tissue sample
WO2014071361A1 (en) 2012-11-05 2014-05-08 Rubicon Genomics Barcoding nucleic acids
US9995728B2 (en) 2012-11-06 2018-06-12 Oxford Nanopore Technologies Ltd. Quadruplex method
EP2917368A1 (en) 2012-11-07 2015-09-16 Good Start Genetics, Inc. Methods and systems for identifying contamination in samples
WO2014109845A1 (en) 2012-12-03 2014-07-17 Yilin Zhang Single-stranded polynucleotide amplification methods
WO2014093701A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Functional genomics using crispr-cas systems, compositions, methods, knock out libraries and applications thereof
PT2896697E (en) 2012-12-12 2015-12-31 Massachusetts Inst Technology Engineering of systems, methods and optimized guide compositions for sequence manipulation
US10533221B2 (en) 2012-12-14 2020-01-14 10X Genomics, Inc. Methods and systems for processing polynucleotides
EP2749653A1 (en) 2012-12-28 2014-07-02 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Molecular coding for analysis of composition of macromolecules and molecular complexes
EP2752664A1 (en) 2013-01-07 2014-07-09 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Label-free method for the detection of analytes
US9683230B2 (en) 2013-01-09 2017-06-20 Illumina Cambridge Limited Sample preparation on a solid support
GB202020510D0 (en) 2013-01-17 2021-02-03 Edico Genome Corp Bioinformatics systems, apparatuses, and methods executed on an integrated circuit processing platform
KR101984699B1 (en) 2013-01-24 2019-05-31 삼성전자주식회사 Micro-fluidic system for analysis of nucleic acid
EP3473905B1 (en) 2013-01-25 2020-07-29 Bio-rad Laboratories, Inc. System and method for performing droplet inflation
US10381106B2 (en) 2013-01-28 2019-08-13 Hasso-Plattner-Institut Fuer Softwaresystemtechnik Gmbh Efficient genomic read alignment in an in-memory database
WO2014121241A1 (en) 2013-02-01 2014-08-07 Bio-Rad Laboratories, Inc. System for detection of spaced droplets
WO2014138154A1 (en) 2013-03-06 2014-09-12 President And Fellows Of Harvard College Devices and methods for forming relatively monodisperse droplets
EP3418398B1 (en) 2013-03-08 2020-05-13 Bio-Rad Laboratories, Inc. Compositions for polymerase chain reaction assays
ES2887177T3 (en) 2013-03-13 2021-12-22 Illumina Inc Nucleic Acid Sequencing Library Preparation Method
US9273349B2 (en) 2013-03-14 2016-03-01 Affymetrix, Inc. Detection of nucleic acids
US10612088B2 (en) 2013-03-14 2020-04-07 The Broad Institute, Inc. Massively multiplexed RNA sequencing
US20140272996A1 (en) 2013-03-15 2014-09-18 Bio-Rad Laboratories, Inc. Droplet generator with collection tube
CN105283558B (en) 2013-03-15 2019-05-10 西格尼斯生物技术有限责任公司 The method for expanding and being sequenced using heat-staple TthPrimPol
US20140274729A1 (en) 2013-03-15 2014-09-18 Nugen Technologies, Inc. Methods, compositions and kits for generation of stranded rna or dna libraries
US9328382B2 (en) 2013-03-15 2016-05-03 Complete Genomics, Inc. Multiple tagging of individual long DNA fragments
CN105358984B (en) 2013-03-15 2020-02-18 普罗格诺西斯生物科学公司 Methods for detecting peptide/MHC/TCR binding
US10119134B2 (en) 2013-03-15 2018-11-06 Abvitro Llc Single cell bar-coding for antibody discovery
CA3156663A1 (en) 2013-03-15 2014-09-18 Verinata Health, Inc. Generating cell-free dna libraries directly from blood
EP2981349A4 (en) 2013-04-02 2016-11-16 Raindance Technologies Inc Systems and methods for handling microfluidic droplets
WO2014182835A1 (en) 2013-05-09 2014-11-13 Bio-Rad Laboratories, Inc. Magnetic immuno digital pcr assay
AU2014268710B2 (en) 2013-05-23 2018-10-18 The Board Of Trustees Of The Leland Stanford Junior University Transposition into native chromatin for personal epigenomics
US20160122753A1 (en) 2013-06-12 2016-05-05 Tarjei Mikkelsen High-throughput rna-seq
WO2014200767A1 (en) 2013-06-12 2014-12-18 The General Hospital Corporation Methods, kits, and systems for multiplexed detection of target molecules and uses thereof
RU2716420C2 (en) 2013-06-17 2020-03-11 Те Брод Инститьют Инк. Delivery and use of systems of crispr-cas, vectors and compositions for targeted action and therapy in liver
US20160208323A1 (en) 2013-06-21 2016-07-21 The Broad Institute, Inc. Methods for Shearing and Tagging DNA for Chromatin Immunoprecipitation and Sequencing
US9879313B2 (en) 2013-06-25 2018-01-30 Prognosys Biosciences, Inc. Methods and systems for determining spatial patterns of biological targets in a sample
CN117568449A (en) 2013-06-27 2024-02-20 10X基因组学有限公司 Compositions and methods for sample processing
WO2015006700A1 (en) 2013-07-12 2015-01-15 University Of South Alabama Minimal piggybac vectors for genome integration
CN103394410B (en) 2013-07-25 2016-04-20 博奥生物集团有限公司 A kind of intelligent magnetic frame of position-adjustable
GB2516684A (en) 2013-07-30 2015-02-04 Sphere Fluidics Ltd Microfluidic devices and systems
KR102536833B1 (en) 2013-08-28 2023-05-26 벡톤 디킨슨 앤드 컴퍼니 Massively parallel single cell analysis
US10395758B2 (en) 2013-08-30 2019-08-27 10X Genomics, Inc. Sequencing methods
US20160231324A1 (en) 2013-09-24 2016-08-11 The Regents Of The University Of California Encapsulated sensors and sensing systems for bioassays and diagnostics and methods for making and using them
GB201317301D0 (en) 2013-09-30 2013-11-13 Linnarsson Sten Method for capturing and encoding nucleic acid from a plurality of single cells
CN106062209B (en) 2013-10-09 2021-08-06 Stc.Unm 公司 Synthetic long read DNA sequencing
US9937495B2 (en) 2013-10-28 2018-04-10 Massachusetts Institute Of Technology Hydrogel microstructures with immiscible fluid isolation for small reaction volumes
US9824068B2 (en) 2013-12-16 2017-11-21 10X Genomics, Inc. Methods and apparatus for sorting data
US20140315755A1 (en) 2013-12-26 2014-10-23 Tao Chen Genome-wide Antisense Oligonucleotide and RNAi
JP6608368B2 (en) 2013-12-30 2019-11-20 アトレカ インコーポレイテッド Method for analyzing nucleic acids associated with single cells using nucleic acid barcodes
KR101464100B1 (en) 2014-01-29 2014-11-21 성균관대학교산학협력단 Fusion nano liposome-fluorescence labeled nucleic acid for in vivo application, uses thereof and preparation method thereof
CN106103713B (en) 2014-02-03 2021-05-28 赛默飞世尔科技波罗的海封闭股份公司 Method for controlled DNA fragmentation
US9822396B2 (en) 2014-02-13 2017-11-21 Bio-Rad Laboratories, Inc. Chromosome conformation capture in partitions
JP6996844B2 (en) 2014-02-27 2022-02-03 ジャンプコード ゲノミクス,インク. Methods for the analysis of somatic mobile elements, and their use
AU2015243445B2 (en) 2014-04-10 2020-05-28 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same
CN106507677B (en) 2014-04-15 2021-03-12 伊鲁米那股份有限公司 Modified transposases for improved insert sequence bias and increased DNA import tolerance
EP3456846B1 (en) 2014-04-21 2022-06-22 President and Fellows of Harvard College Systems and methods for barcoding nucleic acid
US20150298091A1 (en) 2014-04-21 2015-10-22 President And Fellows Of Harvard College Systems and methods for barcoding nucleic acids
US10975371B2 (en) 2014-04-29 2021-04-13 Illumina, Inc. Nucleic acid sequence analysis from single cells
WO2015179706A1 (en) 2014-05-23 2015-11-26 Fluidigm Corporation Haploidome determination by digitized transposons
SG10202005892SA (en) 2014-06-06 2020-07-29 Herlev Hospital Determining antigen recognition through barcoding of mhc multimers
DK3155119T3 (en) 2014-06-11 2018-12-17 Samplix Aps NUCLEOTIDE SEQUENCE EXCLUSION ENHANCEMENT BY DROP SORTING (NEEDLS)
WO2015191877A1 (en) 2014-06-11 2015-12-17 Life Technologies Corporation Systems and methods for substrate enrichment
WO2015188839A2 (en) 2014-06-13 2015-12-17 Immudex Aps General detection and isolation of specific cells by binding of labeled molecules
US10480021B2 (en) 2014-06-23 2019-11-19 Yale University Methods for closed chromatin mapping and DNA methylation analysis for single cells
WO2015200541A1 (en) 2014-06-24 2015-12-30 Bio-Rad Laboratories, Inc. Digital pcr barcoding
JP2017522866A (en) 2014-06-26 2017-08-17 10エックス ジェノミクス, インコーポレイテッド Nucleic acid sequence analysis
EP4053292A1 (en) 2014-06-26 2022-09-07 10X Genomics, Inc. Methods of analyzing nucleic acids from individual cells or cell populations
US10017759B2 (en) 2014-06-26 2018-07-10 Illumina, Inc. Library preparation of tagged nucleic acid
CA2953473A1 (en) 2014-06-26 2015-12-30 10X Genomics, Inc. Methods and compositions for sample analysis
KR20170023979A (en) 2014-06-26 2017-03-06 10엑스 제노믹스, 인크. Processes and systems for nucleic acid sequence assembly
EP3656875B1 (en) 2014-07-18 2021-09-22 Illumina, Inc. Non-invasive prenatal diagnosis
US20160024558A1 (en) 2014-07-23 2016-01-28 10X Genomics, Inc. Nucleic acid binding proteins and uses thereof
CN105335116B (en) 2014-07-30 2018-11-09 联想(北京)有限公司 A kind of display control method and electronic equipment
EP3186418A2 (en) 2014-08-26 2017-07-05 Nugen Technologies, Inc. Compositions and methods for targeted nucleic acid sequence enrichment and high efficiency library generation
JP2017532024A (en) 2014-09-09 2017-11-02 ザ・ブロード・インスティテュート・インコーポレイテッド Droplet-based methods and instruments for composite single cell nucleic acid analysis
KR102541849B1 (en) 2014-09-15 2023-06-09 에이비비트로, 엘엘씨 High-throughput nucleotide library sequencing
WO2016061517A2 (en) 2014-10-17 2016-04-21 Illumina Cambridge Limited Contiguity preserving transposition
WO2016069939A1 (en) 2014-10-29 2016-05-06 10X Genomics, Inc. Methods and compositions for targeted nucleic acid sequencing
US9975122B2 (en) 2014-11-05 2018-05-22 10X Genomics, Inc. Instrument systems for integrated sample processing
EP3234602B1 (en) 2014-12-19 2021-08-25 F. Hoffmann-La Roche AG Methods for identifying multiple epitopes in selected sub-populations of cells
CA2972969A1 (en) 2015-01-12 2016-07-21 10X Genomics, Inc. Processes and systems for preparing nucleic acid sequencing libraries and libraries prepared using same
EP3245605B1 (en) 2015-01-13 2022-04-20 10X Genomics, Inc. Systems and methods for visualizing structural variation and phasing information
US10669304B2 (en) 2015-02-04 2020-06-02 Twist Bioscience Corporation Methods and devices for de novo oligonucleic acid assembly
EP3253479B1 (en) 2015-02-04 2022-09-21 The Regents of The University of California Sequencing of nucleic acids via barcoding in discrete entities
WO2016130578A1 (en) 2015-02-09 2016-08-18 10X Genomics, Inc. Systems and methods for determining structural variation and phasing using variant call data
KR20210135626A (en) 2015-02-10 2021-11-15 일루미나, 인코포레이티드 The method and the composition for analyzing the cellular constituent
WO2016138148A1 (en) 2015-02-24 2016-09-01 10X Genomics, Inc. Methods for targeted nucleic acid sequence coverage
EP3262407B1 (en) 2015-02-24 2023-08-30 10X Genomics, Inc. Partition processing methods and systems
EP3262192B1 (en) 2015-02-27 2020-09-16 Becton, Dickinson and Company Spatially addressable molecular barcoding
EP3268462B1 (en) 2015-03-11 2021-08-11 The Broad Institute, Inc. Genotype and phenotype coupling
EP3271713B1 (en) 2015-03-18 2021-05-05 The Broad Institute, Inc. Massively parallel on-chip coalescence of microemulsions
WO2016160908A1 (en) 2015-03-30 2016-10-06 Verily Life Sciences Llc Methods for combining single cell profiling with combinatorial nanoparticle conjugate library screening
EP4321627A3 (en) 2015-04-10 2024-04-17 10x Genomics Sweden AB Spatially distinguished, multiplex nucleic acid analysis of biological specimens
AU2016248995B2 (en) 2015-04-17 2022-04-28 President And Fellows Of Harvard College Barcoding systems and methods for gene sequencing and other applications
WO2016169431A1 (en) 2015-04-20 2016-10-27 深圳华大基因研究院 Method for constructing long fragment dna library
US10632465B2 (en) 2015-04-22 2020-04-28 Stilla Technologies Contact-less priming method for loading a solution in a microfluidic device and associated system
US20160314242A1 (en) 2015-04-23 2016-10-27 10X Genomics, Inc. Sample indexing methods and compositions for sequencing applications
EP3298168A4 (en) 2015-05-18 2019-02-20 10X Genomics, Inc. Stabilized reducing agents and methods using same
CN107580627A (en) 2015-05-18 2018-01-12 10X基因组学有限公司 For the flowing solid compositions in biochemical reaction and analysis
EP3304383B1 (en) 2015-05-26 2021-07-07 Pacific Biosciences of California, Inc. De novo diploid genome assembly and haplotype sequence reconstruction
WO2016187717A1 (en) 2015-05-26 2016-12-01 Exerkine Corporation Exosomes useful for genome editing
US20180087050A1 (en) 2015-05-27 2018-03-29 Jianbiao Zheng Methods of inserting molecular barcodes
WO2016207647A1 (en) 2015-06-24 2016-12-29 Oxford Biodynamics Limited Epigenetic chromosome interactions
AU2016297510B2 (en) 2015-07-17 2021-09-09 President And Fellows Of Harvard College Methods of amplifying nucleic acid sequences
US10934636B2 (en) 2015-08-12 2021-03-02 CeMM—FORSCHUNGSZENTRUM FÜR MOLEKULARE MEDIZIN GmbH Methods for studying nucleic acids
US11479805B2 (en) 2015-08-21 2022-10-25 The General Hospital Corporation Combinatorial single molecule analysis of chromatin
CA2999888C (en) 2015-09-24 2024-04-09 Abvitro Llc Affinity-oligonucleotide conjugates and uses thereof
EP3366818B1 (en) 2015-10-19 2021-04-14 Zhejiang Annoroad Bio-Technology Co., Ltd. Method for constructing high-resolution single cell hi-c library with a lot of information
EP3365454A1 (en) 2015-10-20 2018-08-29 10X Genomics, Inc. Methods and systems for high throughput single cell genetic manipulation
WO2017075294A1 (en) 2015-10-28 2017-05-04 The Board Institute Inc. Assays for massively combinatorial perturbation profiling and cellular circuit reconstruction
US11092607B2 (en) 2015-10-28 2021-08-17 The Board Institute, Inc. Multiplex analysis of single cell constituents
EP3371309B1 (en) 2015-11-04 2023-07-05 Atreca, Inc. Combinatorial sets of nucleic acid barcodes for analysis of nucleic acids associated with single cells
SG11201803983UA (en) 2015-11-19 2018-06-28 10X Genomics Inc Transformable tagging compositions, methods, and processes incorporating same
CN115369161A (en) 2015-12-04 2022-11-22 10X 基因组学有限公司 Methods and compositions for nucleic acid analysis
WO2017117358A1 (en) 2015-12-30 2017-07-06 Bio-Rad Laboratories, Inc. Digital protein quantification
WO2017139690A1 (en) 2016-02-11 2017-08-17 10X Genomics, Inc. Cell population analysis using single nucleotide polymorphisms from single cell transcriptomes
JP6735348B2 (en) 2016-02-11 2020-08-05 10エックス ジェノミクス, インコーポレイテッド Systems, methods and media for de novo assembly of whole genome sequence data
WO2017145476A1 (en) 2016-02-26 2017-08-31 富士フイルム株式会社 Method for producing lithographic printing plate
US20190078150A1 (en) 2016-03-01 2019-03-14 Universal Sequencing Technology Corporation Methods and Kits for Tracking Nucleic Acid Target Origin for Nucleic Acid Sequencing
CN109072206A (en) 2016-03-10 2018-12-21 斯坦福大学托管董事会 The imaging to accessible genome that transposase mediates
WO2017180420A1 (en) 2016-04-11 2017-10-19 Board Of Regents, The University Of Texas System Methods and compositions for detecting single t cell receptor affinity and sequence
WO2017197343A2 (en) 2016-05-12 2017-11-16 10X Genomics, Inc. Microfluidic on-chip filters
WO2017197338A1 (en) 2016-05-13 2017-11-16 10X Genomics, Inc. Microfluidic systems and methods of use
WO2018031631A1 (en) 2016-08-10 2018-02-15 President And Fellows Of Harvard College Methods of de novo assembly of barcoded genomic dna fragments
WO2018039338A1 (en) 2016-08-23 2018-03-01 10X Genomics, Inc. Microfluidic surface-mediated emulsion stability control
WO2018044831A1 (en) 2016-08-30 2018-03-08 Integrated Dna Technologies, Inc. Cleavable hairpin primers
MX2019002376A (en) 2016-08-31 2019-06-20 Harvard College Methods of whole genome digital amplification.
GB2569252A (en) 2016-08-31 2019-06-12 Harvard College Methods of combining the detection of biomolecules into a single assay using fluorescent in situ sequencing
US20180080021A1 (en) 2016-09-17 2018-03-22 The Board Of Trustees Of The Leland Stanford Junior University Simultaneous sequencing of rna and dna from the same sample
SG11201901733PA (en) 2016-09-26 2019-04-29 Cellular Res Inc Measurement of protein expression using reagents with barcoded oligonucleotide sequences
CN109983126A (en) 2016-10-19 2019-07-05 10X基因组学有限公司 For bar coded individual cells or the method and system of the nucleic acid molecules of cell mass
GB201619458D0 (en) 2016-11-17 2017-01-04 Spatial Transcriptomics Ab Method for spatial tagging and analysing nucleic acids in a biological specimen
DK3553180T3 (en) 2016-12-07 2022-07-25 Mgi Tech Co Ltd PROCEDURE FOR SINGLE CELL SEQUENCE LIBRARY CONSTRUCTION AND USE THEREOF
CN110139932A (en) 2016-12-19 2019-08-16 生物辐射实验室股份有限公司 The labeling DNA that the adjacency of drop mark-on retains
EP3571308A4 (en) 2016-12-21 2020-08-19 The Regents of The University of California Single cell genomic sequencing using hydrogel based droplets
US10815525B2 (en) 2016-12-22 2020-10-27 10X Genomics, Inc. Methods and systems for processing polynucleotides
US20190177800A1 (en) 2017-12-08 2019-06-13 10X Genomics, Inc. Methods and compositions for labeling cells
US10011872B1 (en) 2016-12-22 2018-07-03 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10550429B2 (en) 2016-12-22 2020-02-04 10X Genomics, Inc. Methods and systems for processing polynucleotides
CA3048863A1 (en) 2016-12-29 2018-07-05 Illumina, Inc. Analysis system for orthogonal access to and tagging of biomolecules in cellular compartments
EP4095263A1 (en) 2017-01-06 2022-11-30 Editas Medicine, Inc. Methods of assessing nuclease cleavage
EP3568508A4 (en) 2017-01-12 2020-12-16 Massachusetts Institute Of Technology Methods for analyzing t cell receptors and b cell receptors
EP4029939B1 (en) 2017-01-30 2023-06-28 10X Genomics, Inc. Methods and systems for droplet-based single cell barcoding
US10995333B2 (en) 2017-02-06 2021-05-04 10X Genomics, Inc. Systems and methods for nucleic acid preparation
US10347365B2 (en) 2017-02-08 2019-07-09 10X Genomics, Inc. Systems and methods for visualizing a pattern in a dataset
GB201704402D0 (en) 2017-03-20 2017-05-03 Blacktrace Holdings Ltd Single cell DNA sequencing
CA3054901A1 (en) 2017-03-24 2018-09-27 National University Of Singapore Methods for multiplex detection of molecules
CA3059370C (en) 2017-04-12 2022-05-10 Karius, Inc. Methods for concurrent analysis of dna and rna in mixed samples
US20210139941A1 (en) 2017-04-14 2021-05-13 The Broad Institute, Inc. High-throughput screens for exploring biological functions of microscale biological systems
US20180312822A1 (en) 2017-04-26 2018-11-01 10X Genomics, Inc. Mmlv reverse transcriptase variants
CN117143960A (en) 2017-05-18 2023-12-01 10X基因组学有限公司 Method and system for sorting droplets and beads
US10544413B2 (en) 2017-05-18 2020-01-28 10X Genomics, Inc. Methods and systems for sorting droplets and beads
EP3625715A4 (en) 2017-05-19 2021-03-17 10X Genomics, Inc. Systems and methods for analyzing datasets
US10914729B2 (en) 2017-05-22 2021-02-09 The Trustees Of Princeton University Methods for detecting protein binding sequences and tagging nucleic acids
US20180340169A1 (en) 2017-05-26 2018-11-29 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
WO2018226546A1 (en) 2017-06-05 2018-12-13 10X Genomics, Inc. Gaskets for the distribution of pressures in a microfluidic system
AU2018281745B2 (en) 2017-06-05 2022-05-19 Becton, Dickinson And Company Sample indexing for single cells
WO2018236615A1 (en) 2017-06-20 2018-12-27 10X Genomics, Inc. Methods and systems for improved droplet stabilization
US20200131506A1 (en) 2017-06-21 2020-04-30 Bluedot Llc Systems and methods for identification of nucleic acids in a sample
CN108336542B (en) 2017-06-23 2020-02-21 番禺得意精密电子工业有限公司 Electrical connector
SG11201911871TA (en) 2017-08-01 2020-01-30 Illumina Inc Hydrogel beads for nucleotide sequencing
US9946577B1 (en) 2017-08-14 2018-04-17 10X Genomics, Inc. Systems and methods for distributed resource management
US10549279B2 (en) 2017-08-22 2020-02-04 10X Genomics, Inc. Devices having a plurality of droplet formation regions
WO2019071039A1 (en) 2017-10-04 2019-04-11 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
US10590244B2 (en) 2017-10-04 2020-03-17 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
WO2019084328A1 (en) 2017-10-26 2019-05-02 10X Genomics, Inc. Methods for preparing nucleic acid molecules
WO2019083852A1 (en) 2017-10-26 2019-05-02 10X Genomics, Inc. Microfluidic channel networks for partitioning
US20190127731A1 (en) 2017-10-26 2019-05-02 10X Genomics, Inc. Methods for preparing nucleic acid molecules
WO2019084043A1 (en) 2017-10-26 2019-05-02 10X Genomics, Inc. Methods and systems for nuclecic acid preparation and chromatin analysis
CN114525273A (en) 2017-10-27 2022-05-24 10X基因组学有限公司 Methods and systems for sample preparation and analysis
SG11201913654QA (en) 2017-11-15 2020-01-30 10X Genomics Inc Functionalized gel beads
US10829815B2 (en) 2017-11-17 2020-11-10 10X Genomics, Inc. Methods and systems for associating physical and genetic properties of biological particles
WO2019108851A1 (en) 2017-11-30 2019-06-06 10X Genomics, Inc. Systems and methods for nucleic acid preparation and analysis
WO2019113235A1 (en) 2017-12-06 2019-06-13 10X Genomics, Inc. Methods and systems for processing nucleic acid molecules
EP3724658A1 (en) 2017-12-12 2020-10-21 10X Genomics, Inc. Systems and methods for single cell processing
WO2019126789A1 (en) 2017-12-22 2019-06-27 10X Genomics, Inc. Systems and methods for processing nucleic acid molecules from one or more cells
CN108173158B (en) 2018-01-05 2023-09-08 浙江正泰电器股份有限公司 Drawer type circuit breaker
WO2019148042A1 (en) 2018-01-26 2019-08-01 10X Genomics, Inc. Compositions and methods for sample processing
EP4299755A3 (en) 2018-02-05 2024-04-10 The Board of Trustees of the Leland Stanford Junior University Systems and methods for multiplexed measurements in single and ensemble cells
WO2019157529A1 (en) 2018-02-12 2019-08-15 10X Genomics, Inc. Methods characterizing multiple analytes from individual cells or cell populations
CN112074610A (en) 2018-02-22 2020-12-11 10X基因组学有限公司 Conjugation-mediated nucleic acid analysis
WO2019169028A1 (en) 2018-02-28 2019-09-06 10X Genomics, Inc. Transcriptome sequencing through random ligation
WO2019169347A1 (en) 2018-03-02 2019-09-06 10X Genomics, Inc. Systems and apparatus for holding plates
WO2019191321A1 (en) 2018-03-28 2019-10-03 10X Genomics, Inc. Nucleic acid enrichment within partitions
EP3775271A1 (en) 2018-04-06 2021-02-17 10X Genomics, Inc. Systems and methods for quality control in single cell processing
US20190345636A1 (en) 2018-05-10 2019-11-14 10X Genomics, Inc. Methods and systems for molecular library generation
WO2019217758A1 (en) 2018-05-10 2019-11-14 10X Genomics, Inc. Methods and systems for molecular library generation
US20190352717A1 (en) 2018-05-18 2019-11-21 10X Genomics, Inc. Targeted non-invasive prenatal testing
US11932899B2 (en) 2018-06-07 2024-03-19 10X Genomics, Inc. Methods and systems for characterizing nucleic acid molecules
US11703427B2 (en) 2018-06-25 2023-07-18 10X Genomics, Inc. Methods and systems for cell and bead processing
US11574706B2 (en) 2018-06-28 2023-02-07 10X Genomics, Inc. Systems and methods for visualization of single-cell resolution characteristics
US20200033366A1 (en) 2018-07-27 2020-01-30 10X Genomics, Inc. Systems and methods for metabolome analysis
US20200032335A1 (en) 2018-07-27 2020-01-30 10X Genomics, Inc. Systems and methods for metabolome analysis
US20200263232A1 (en) 2018-08-03 2020-08-20 10X Genomics, Inc. Methods and systems to minimize barcode exchange
WO2020041148A1 (en) 2018-08-20 2020-02-27 10X Genomics, Inc. Methods and systems for detection of protein-dna interactions using proximity ligation
US20200056223A1 (en) 2018-08-20 2020-02-20 10X Genomics, Inc. Compositions and methods for cellular processing
US20200105373A1 (en) 2018-09-28 2020-04-02 10X Genomics, Inc. Systems and methods for cellular analysis using nucleic acid sequencing
SG11202107418SA (en) 2019-01-06 2021-08-30 10X Genomics Inc Methods and systems for enrichment of barcodes
SG11202108788TA (en) 2019-02-12 2021-09-29 10X Genomics Inc Methods for processing nucleic acid molecules
WO2020167866A1 (en) 2019-02-12 2020-08-20 10X Genomics, Inc. Systems and methods for transposon loading
WO2020167862A1 (en) 2019-02-12 2020-08-20 10X Genomics, Inc. Systems and methods for transfer of reagents between droplets
CN113811619A (en) 2019-03-27 2021-12-17 10X基因组学有限公司 Systems and methods for processing RNA from cells
CN114729392A (en) 2019-09-06 2022-07-08 10X基因组学有限公司 Systems and methods for barcoding cells and cell beads
WO2021222301A1 (en) 2020-04-27 2021-11-04 10X Genomics, Inc. Methods and systems for analysis and identification of barcode multiplets
WO2021222302A1 (en) 2020-04-27 2021-11-04 10X Genomics, Inc. Methods and systems for increasing cell recovery efficiency

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120022049A1 (en) * 2002-03-25 2012-01-26 Universite Laval Nicotinic receptor agonists for the treatment of inflammatory diseases
US20150001143A1 (en) * 2013-06-27 2015-01-01 Desmond Bell Oil Filter Conversion Kit

Cited By (125)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10436700B1 (en) 2011-08-01 2019-10-08 Celsee Diagnostics, Inc. Cell capture system and method of use
US10746648B2 (en) 2011-08-01 2020-08-18 Bio-Rad Laboratories, Inc. Cell capture and method of use
US11946855B2 (en) 2011-08-01 2024-04-02 Bio-Rad Laboratories, Inc. Cell capture system and method of use
US11635365B2 (en) 2011-08-01 2023-04-25 Bio-Rad Laboratories, Inc. Cell capture system and method of use
US10782226B1 (en) 2011-08-01 2020-09-22 Bio-Rad Laboratories, Inc. Cell capture system and method of use
US10794817B1 (en) 2011-08-01 2020-10-06 Bio-Rad Laboratories, Inc. Cell capture system and method of use
US10914672B2 (en) 2011-08-01 2021-02-09 Bio-Rad Laboratories, Inc. System and method for retrieving and analyzing particles
US10641700B2 (en) 2011-08-01 2020-05-05 Celsee Diagnostics, Inc. Cell capture system and method of use
US10921237B2 (en) 2011-08-01 2021-02-16 Bio-Rad Laboratories, Inc. Cell capture system and method of use
US10591404B1 (en) 2011-08-01 2020-03-17 Celsee Diagnostics, Inc. Cell capture system and method of use
US10564090B2 (en) 2011-08-01 2020-02-18 Celsee Diagnostics, Inc. System and method for retrieving and analyzing particles
US10345219B2 (en) 2011-08-01 2019-07-09 Celsee Diagnostics, Inc. Cell capture system and method of use
US10533936B1 (en) 2011-08-01 2020-01-14 Celsee Diagnostics, Inc. Cell capture system and method of use
US11073468B2 (en) 2011-08-01 2021-07-27 Bio-Rad Laboratories, Inc. Cell capture system and method of use
US10481077B1 (en) 2011-08-01 2019-11-19 Celsee Diagnostics, Inc. Cell capture system and method of use
US10466160B2 (en) 2011-08-01 2019-11-05 Celsee Diagnostics, Inc. System and method for retrieving and analyzing particles
US11300496B2 (en) 2011-08-01 2022-04-12 Bio-Rad Laboratories, Inc. Cell capture system and method of use
US11231355B2 (en) 2011-08-01 2022-01-25 Bio-Rad Laboratories, Inc. Cell capture system and method of use
US11275015B2 (en) 2011-08-01 2022-03-15 Bio-Rad Laboratories, Inc. System and method for retrieving and analyzing particles
US10401277B2 (en) 2011-08-01 2019-09-03 Celsee Diagnostics, Inc. Cell capture system and method of use
US10408737B1 (en) 2011-08-01 2019-09-10 Celsee Diagnostics, Inc. Cell capture system and method of use
US10408736B1 (en) 2011-08-01 2019-09-10 Celsee Diagnostics, Inc. Cell capture system and method of use
US10416070B1 (en) 2011-08-01 2019-09-17 Celsee Diagnostics, Inc. Cell capture system and method of use
US11237096B2 (en) 2011-08-01 2022-02-01 Bio-Rad Laboratories, Inc. Cell capture system and method of use
US11359239B2 (en) 2012-08-14 2022-06-14 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10669583B2 (en) 2012-08-14 2020-06-02 10X Genomics, Inc. Method and systems for processing polynucleotides
US10450607B2 (en) 2012-08-14 2019-10-22 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10752950B2 (en) 2012-08-14 2020-08-25 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10221442B2 (en) 2012-08-14 2019-03-05 10X Genomics, Inc. Compositions and methods for sample processing
US11078522B2 (en) 2012-08-14 2021-08-03 10X Genomics, Inc. Capsule array devices and methods of use
US10752949B2 (en) 2012-08-14 2020-08-25 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10400280B2 (en) 2012-08-14 2019-09-03 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10273541B2 (en) 2012-08-14 2019-04-30 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10323279B2 (en) 2012-08-14 2019-06-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11441179B2 (en) 2012-08-14 2022-09-13 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11035002B2 (en) 2012-08-14 2021-06-15 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11021749B2 (en) 2012-08-14 2021-06-01 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10626458B2 (en) 2012-08-14 2020-04-21 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10597718B2 (en) 2012-08-14 2020-03-24 10X Genomics, Inc. Methods and systems for sample processing polynucleotides
US10584381B2 (en) 2012-08-14 2020-03-10 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11591637B2 (en) 2012-08-14 2023-02-28 10X Genomics, Inc. Compositions and methods for sample processing
US10676789B2 (en) 2012-12-14 2020-06-09 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10612090B2 (en) 2012-12-14 2020-04-07 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10533221B2 (en) 2012-12-14 2020-01-14 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11421274B2 (en) 2012-12-14 2022-08-23 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11473138B2 (en) 2012-12-14 2022-10-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10253364B2 (en) 2012-12-14 2019-04-09 10X Genomics, Inc. Method and systems for processing polynucleotides
US10227648B2 (en) 2012-12-14 2019-03-12 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10975422B2 (en) 2013-01-26 2021-04-13 Bio-Rad Laboratories, Inc. System and method for capturing and analyzing cells
US10718007B2 (en) 2013-01-26 2020-07-21 Bio-Rad Laboratories, Inc. System and method for capturing and analyzing cells
US11345951B2 (en) 2013-01-26 2022-05-31 Bio-Rad Laboratories, Inc. System and method for capturing and analyzing cells
US10150964B2 (en) 2013-02-08 2018-12-11 10X Genomics, Inc. Partitioning and processing of analytes and other species
US10150963B2 (en) 2013-02-08 2018-12-11 10X Genomics, Inc. Partitioning and processing of analytes and other species
US11193121B2 (en) 2013-02-08 2021-12-07 10X Genomics, Inc. Partitioning and processing of analytes and other species
US10509022B2 (en) 2013-03-13 2019-12-17 Celsee Diagnostics, Inc. System for imaging captured cells
US11199532B2 (en) 2013-03-13 2021-12-14 Bio-Rad Laboratories, Inc. System for imaging captured cells
US10350601B2 (en) 2013-03-13 2019-07-16 Celsee Diagnostics, Inc. System and method for capturing and analyzing cells
US10690650B2 (en) 2013-03-13 2020-06-23 Bio-Rad Laboratories, Inc. System for imaging captured cells
US10391490B2 (en) 2013-05-31 2019-08-27 Celsee Diagnostics, Inc. System and method for isolating and analyzing cells
US11358147B2 (en) 2013-05-31 2022-06-14 Bio-Rad Laboratories, Inc. System and method for isolating and analyzing cells
US10512914B2 (en) 2013-05-31 2019-12-24 Celsee Diagnostics, Inc. System for isolating and analyzing cells in a single-cell format
US11052396B2 (en) 2013-05-31 2021-07-06 Bio-Rad Laboratories, Inc. System and method for isolating and analyzing cells
US10533229B2 (en) 2013-05-31 2020-01-14 Celsee Diagnostics, Inc. System and method for isolating and analyzing cells
US10449543B2 (en) 2013-05-31 2019-10-22 Celsee Diagnostics, Inc. System and method for isolating and analyzing cells
US10851426B2 (en) 2013-05-31 2020-12-01 Bio-Rad Laboratories, Inc. System and method for isolating and analyzing cells
US10343166B2 (en) 2014-04-10 2019-07-09 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same
US11629344B2 (en) 2014-06-26 2023-04-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11713457B2 (en) 2014-06-26 2023-08-01 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10457986B2 (en) 2014-06-26 2019-10-29 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10208343B2 (en) 2014-06-26 2019-02-19 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10344329B2 (en) 2014-06-26 2019-07-09 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10337061B2 (en) 2014-06-26 2019-07-02 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10480028B2 (en) 2014-06-26 2019-11-19 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10760124B2 (en) 2014-06-26 2020-09-01 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11135584B2 (en) 2014-11-05 2021-10-05 10X Genomics, Inc. Instrument systems for integrated sample processing
US10221436B2 (en) 2015-01-12 2019-03-05 10X Genomics, Inc. Processes and systems for preparation of nucleic acid sequencing libraries and libraries prepared using same
US10557158B2 (en) 2015-01-12 2020-02-11 10X Genomics, Inc. Processes and systems for preparation of nucleic acid sequencing libraries and libraries prepared using same
US11414688B2 (en) 2015-01-12 2022-08-16 10X Genomics, Inc. Processes and systems for preparation of nucleic acid sequencing libraries and libraries prepared using same
US10697000B2 (en) 2015-02-24 2020-06-30 10X Genomics, Inc. Partition processing methods and systems
US11603554B2 (en) 2015-02-24 2023-03-14 10X Genomics, Inc. Partition processing methods and systems
US11274343B2 (en) 2015-02-24 2022-03-15 10X Genomics, Inc. Methods and compositions for targeted nucleic acid sequence coverage
US11873528B2 (en) 2015-12-04 2024-01-16 10X Genomics, Inc. Methods and compositions for nucleic acid analysis
US11624085B2 (en) 2015-12-04 2023-04-11 10X Genomics, Inc. Methods and compositions for nucleic acid analysis
US11473125B2 (en) 2015-12-04 2022-10-18 10X Genomics, Inc. Methods and compositions for nucleic acid analysis
US10774370B2 (en) 2015-12-04 2020-09-15 10X Genomics, Inc. Methods and compositions for nucleic acid analysis
US11084036B2 (en) 2016-05-13 2021-08-10 10X Genomics, Inc. Microfluidic systems and methods of use
US10550429B2 (en) 2016-12-22 2020-02-04 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10815525B2 (en) 2016-12-22 2020-10-27 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10793905B2 (en) 2016-12-22 2020-10-06 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10858702B2 (en) 2016-12-22 2020-12-08 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11180805B2 (en) 2016-12-22 2021-11-23 10X Genomics, Inc Methods and systems for processing polynucleotides
US11193122B2 (en) 2017-01-30 2021-12-07 10X Genomics, Inc. Methods and systems for droplet-based single cell barcoding
US10428326B2 (en) 2017-01-30 2019-10-01 10X Genomics, Inc. Methods and systems for droplet-based single cell barcoding
US10400235B2 (en) 2017-05-26 2019-09-03 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US11773389B2 (en) 2017-05-26 2023-10-03 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US11198866B2 (en) 2017-05-26 2021-12-14 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US10927370B2 (en) 2017-05-26 2021-02-23 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US10844372B2 (en) 2017-05-26 2020-11-24 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US11155810B2 (en) 2017-05-26 2021-10-26 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US11358146B2 (en) 2017-08-29 2022-06-14 Bio-Rad Laboratories, Inc. System and method for isolating and analyzing cells
US10391492B2 (en) 2017-08-29 2019-08-27 Celsee Diagnostics, Inc. System and method for isolating and analyzing cells
US11865542B2 (en) 2017-08-29 2024-01-09 Bio-Rad Laboratories, Inc. System and method for isolating and analyzing cells
US10821440B2 (en) 2017-08-29 2020-11-03 Bio-Rad Laboratories, Inc. System and method for isolating and analyzing cells
US11504714B2 (en) 2017-08-29 2022-11-22 Bio-Rad Laboratories, Inc. System and method for isolating and analyzing cells
US10391493B2 (en) 2017-08-29 2019-08-27 Celsee Diagnostics, Inc. System and method for isolating and analyzing cells
US11884962B2 (en) 2017-11-15 2024-01-30 10X Genomics, Inc. Functionalized gel beads
US10876147B2 (en) 2017-11-15 2020-12-29 10X Genomics, Inc. Functionalized gel beads
US10745742B2 (en) 2017-11-15 2020-08-18 10X Genomics, Inc. Functionalized gel beads
US10829815B2 (en) 2017-11-17 2020-11-10 10X Genomics, Inc. Methods and systems for associating physical and genetic properties of biological particles
US11155881B2 (en) 2018-04-06 2021-10-26 10X Genomics, Inc. Systems and methods for quality control in single cell processing
US11814671B2 (en) 2019-04-16 2023-11-14 Bio-Rad Laboratories, Inc. System and method for leakage control in a particle capture system
US11866766B2 (en) 2019-04-16 2024-01-09 Bio-Rad Laboratories, Inc. System and method for leakage control in a particle capture system
US10947581B2 (en) 2019-04-16 2021-03-16 Bio-Rad Laboratories, Inc. System and method for leakage control in a particle capture system
US10633693B1 (en) 2019-04-16 2020-04-28 Celsee Diagnostics, Inc. System and method for leakage control in a particle capture system
US10900032B2 (en) 2019-05-07 2021-01-26 Bio-Rad Laboratories, Inc. System and method for automated single cell processing
US11833507B2 (en) 2019-05-07 2023-12-05 Bio-Rad Laboratories, Inc. System and method for target material retrieval from microwells
US11578322B2 (en) 2019-05-07 2023-02-14 Bio-Rad Laboratories, Inc. System and method for automated single cell processing
US11273439B2 (en) 2019-05-07 2022-03-15 Bio-Rad Laboratories, Inc. System and method for target material retrieval from microwells
US11724256B2 (en) 2019-06-14 2023-08-15 Bio-Rad Laboratories, Inc. System and method for automated single cell processing and analyses
US11504719B2 (en) 2020-03-12 2022-11-22 Bio-Rad Laboratories, Inc. System and method for receiving and delivering a fluid for sample processing
WO2022066992A1 (en) * 2020-09-24 2022-03-31 University Of Cincinnati Continuous sensing with adapters and aptamers
EP4217722A4 (en) * 2020-09-24 2024-02-14 Univ Cincinnati Continuous sensing with adapters and aptamers
EP4217495A4 (en) * 2020-09-24 2024-03-27 Univ Cincinnati Continuous optical aptamer sensors
EP4217492A4 (en) * 2020-09-24 2024-03-27 Univ Cincinnati Solute-phase electrochemical aptamer sensors for improved longevity and sensitivity
EP4217493A4 (en) * 2020-09-24 2024-04-10 Univ Cincinnati Solute-phase electrochemical aptamer sensors with rapid time-to-measurement

Also Published As

Publication number Publication date
US10676789B2 (en) 2020-06-09
US11421274B2 (en) 2022-08-23
US20180274027A1 (en) 2018-09-27
US20210079463A1 (en) 2021-03-18
US20170114390A1 (en) 2017-04-27
US9856530B2 (en) 2018-01-02
US20170247757A1 (en) 2017-08-31
US9701998B2 (en) 2017-07-11

Similar Documents

Publication Publication Date Title
US11421274B2 (en) Methods and systems for processing polynucleotides
US10612090B2 (en) Methods and systems for processing polynucleotides
US11473138B2 (en) Methods and systems for processing polynucleotides
AU2014214682B2 (en) Polynucleotide barcode generation
US20180282796A1 (en) Typing and Assembling Discontinuous Genomic Elements
US20100035249A1 (en) Rna sequencing and analysis using solid support
EP3553175A1 (en) Methods and compositions for nucleic acid sequencing
AU2021297880A1 (en) Methods and compositions for analyzing nucleic acid

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION