EP0672185A1 - Sondes d'acide nucleique et leur utilisation dans la detection d'acides nucleiques bicatenaires - Google Patents

Sondes d'acide nucleique et leur utilisation dans la detection d'acides nucleiques bicatenaires

Info

Publication number
EP0672185A1
EP0672185A1 EP93917236A EP93917236A EP0672185A1 EP 0672185 A1 EP0672185 A1 EP 0672185A1 EP 93917236 A EP93917236 A EP 93917236A EP 93917236 A EP93917236 A EP 93917236A EP 0672185 A1 EP0672185 A1 EP 0672185A1
Authority
EP
European Patent Office
Prior art keywords
probe
target
complementary
seq
nucleic acid
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.)
Withdrawn
Application number
EP93917236A
Other languages
German (de)
English (en)
Other versions
EP0672185A4 (fr
Inventor
Nagindra Prashad
Mark Blick
William Dugald Weber
Michael Lee Cubbage
Shyh Chen Ju
Morteza Asgari
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.)
Aprogenex Inc
Original Assignee
Aprogenex Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Aprogenex Inc filed Critical Aprogenex Inc
Publication of EP0672185A1 publication Critical patent/EP0672185A1/fr
Publication of EP0672185A4 publication Critical patent/EP0672185A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • 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
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers

Definitions

  • This inventions described herein concern the detection of nucleic acids through the use of nucleic acid probes.
  • RNA or DNA probes By the use of specific nucleic acid (RNA or DNA) probes, nucleic acid molecule that signify infection and other disease states may be detected. Certain genetic diseases are characterized by the presence of genes which are not present in normal tissue. Oth diseased conditions are characterized by the expression of RNAs or RNA translation products (i.e. peptides or proteins) which are not expressed in normal cells. Some disease states are characterized by the absence of certain genes or gene portions, or the absence or alteration of expression of gene products or proteins. When the probe target is DNA, the target is generally a two-stranded target: an RNA translation products (i.e. peptides or proteins) which are not expressed in normal cells. Some disease states are characterized by the absence of certain genes or gene portions, or the absence or alteration of expression of gene products or proteins. When the probe target is DNA, the target is generally a two-stranded target: an
  • anti-sense strand from which RNA such as mRNA is transcribed and a “sense strand” that is complementary in base sequence to the anti-sense strand. This suggests that, for given amount of cellular DNA target, one can double the signal generated by target- bound probes by including both probes against the sense strand and the anti-sense stran of the target.
  • a nucleic acid probe population is created so that although some of the probe molecules are capable of hybridizing to one strand of a double-stranded target and other probe molecules are capable of hybridizing to the othe strand of that target, the probe molecules cannot hybridize to each other.
  • the probe molecules cannot hybridize to each other.
  • Fig. 1 Nucleotides 101 to 400 of Gen Bank sequence HUMREPA84 are marked and are shown such that nucleotide 101 is at the 5' end of the sequence and nucleotide 400 is at the 3' end. Nucleotides in the sequence complementary to that between nucleotides 101 and 400 are shown below those latter nucleotides. The nucleotide sequences of five probes, H18-100L (SEQ ID NO:16), H18-100R (SEQ ID NO:17), H18 110R (SEQ ID NO:18), H18-10 (SEQ ID NO:14), and H18-11 (SEQ ID NO:18), are indicated.
  • FIG. 3b Photomicrograph of results obtained with probe HYR-7-12 (SEQ ID NO:21) i Example 4.
  • any pair of single-stranded molecules if one has within itself a nucleotide sequence that is complementary to a nucleotide sequence in the second molecule, and both of those sequences are N nucleotides long (the total length of either molecule can be greater than N) then the molecules will form a hybrid only if N is large than some critical value.
  • the critical value will depending partly on the hybridization conditions (temperature, choice of solvent, etc.) and partly on the nucleotide compositio of the complementary sequences.
  • the critical value in the experiments exemplified in the Examples below will be seen to be between 12 an 23.
  • the length of the probe molecule be about 24 nucleotides, and by not letting N exceed 12 for any pair of probe molecules, one has an effective probe population for hybridizing to a two-stranded target.
  • the population is effective because one obtains about twice as much as signal as one would obtain with probes to just one strand.
  • the invention is a process for detecting a two-stranded nucleic acid target, which process comprises the steps of:
  • step (2) co-incubating the sufficiently separated strands of the target with a nucleic acid probe population that comprises molecules complementary in nucleotide sequence to o target strand and molecules complementary in nucleotide sequence to the other target strand; and (3) detecting the nucleic acid probe molecules that are hybridized to target molecules; such that step (2) is performed under conditions that allow each strand of the target to form a hybrid with a nucleic acid probe molecule complementary in nucleotide sequence to that strand; such that, as to the nucleotide sequence of each nucleic acid probe molecule, there a totally complementary sequence in the target; such that each nucleic acid probe molecule is partially complementary in nucleotide sequence to at least one other nucleic acid probe molecule; such that no two nucleic acid probe molecules are completely complementary in nucleotide sequence to each other; such that, where a portion of one nucleic acid probe molecule is complementary in nucleotide sequence to another nucleic acid probe molecule, that portion has a length which is
  • nucleotide sequence is intended to cover a sequence where there is som atom (e.g., sulfur) other than phosphorus at some of the positions where internucleosid phosphorus normally occur. In such a situation, one could alternatively two molecules complementary as to nucleotide sequence as being complementary as to nucleoside sequence or complementary as to nucleotide sequence.
  • the probe molecule will normally be labelled with a detectable label, e.g., radioactively (e.g. with 3 P), a dye molecule such as fluorescein, or a moiety that can enter into a chemiluminescence reaction.
  • the two target strands are located in a biological entity that is either a cell or a virus.
  • the cell or virus may be suspended in solution and not immobilized on a solid support.
  • the cell or virus may be immobilized on a solid support.
  • the cell or virus may be part of a tissue section (histologic section).
  • the cells containing the target nucleic acid molecules may be eukaryotic cells (e.g., human cells), prokaryotic cells (e.g., bacteria), plant cells, or any other type of cell. They can be simple eukaryotes such as yeast or derived from complex eukaryotes such as humans.
  • the target strands of nucleic acid may be in a non-enveloped virus or an enveloped virus (having a non-enveloped membrane such as a lipid protein membrane).
  • a plurality of molecules in the probe population are each covalently attached to a fluorescent dye molecule either directly or via a cross- linker molecule.
  • the two target strands may be purified nucleic acids. They may have been extracted from a virus, cell or multi-cellular organism.
  • the two target strands may be immobilized on a solid support (such as on nitrocellulose paper or a nylon sheet) during Step (2) of the process. Alternatively, they may be in solution and not immobilized on a solid support.
  • the target strands may be DNA.
  • the target strands may be RNA, as in the case of a virus (e.g., human immunodeficiency virus) where complementary RNA strands can exist simultaneously in a single cell.
  • a viral nucleic acid target can be part of a virus, in which case the virus may or may not be inside a cell. Alternatively, a viral nucleic acid target may not be part of a virus, but may be inside a cell.
  • the probe molecules have nucleotide sequences such that, if one strand of the target strand is saturated with probe molecules, then there will be no unhybridized target strand sequences forming gaps between the probe molecules.
  • each probe molecule is complementary to a sequence, present in at least one other probe molecule, not less than about 12 nucleotides but not more than about 100 nucleotides in length. More preferably, each probe molecule is complementary to a sequence, present in a least one other probe molecule, not less than about 12 nucleotides but not more than about 20 nucleotides in length.
  • each probe is between about 15 nucleotides and 10 nucleotides. It is more preferred that the length of each probe is between about 15 nucleotides and 40 nucleotides.
  • the portion of a probe molecule that is complementary to another probe molecule is not less than about 12 nucleotides but not more than about 100 nucleotides in length. It is more preferred that the portion of a probe molecule that is complementary to another probe molecule is not less than about 12 nucleotides but not more than about 20 nucleotides in length. In one highly preferre embodiment of the process, the portion of a probe molecule that is complementary to another probe molecule is about 12 nucleotides in length.
  • the two-stranded target has a firs target strand and a second target strand and wherein the probe molecules that are complementary in nucleotide sequence to the first target strand have a detectable label with a structure different from the detectable label on the probe molecules that complementary to the second target strand.
  • the detectable label on the probe molecules that are complementary in nucleotide sequence to the first target stran may be a fluorescent dye and the detectable label on the probe molecules that are complementary to the second strand may also be a fluorescent dye.
  • the probe molecules tha are complementary in nucleotide sequence to the first target strand are also complementary in nucleotide sequence to cellular RNA molecules.
  • RNA molecules complementary in nucleotide sequence to cellular RNA molecules.
  • An example of wher the latter particular embodiment is useful is where there may be a double-stranded DN viral genome (or the reverse transcriptase DNA copy of an RNA viral genome) in the target cell of interest and, if indeed there is such a genome present, then there may or may not be RNA transcribed from such a genome.
  • it of interest from a clinical point of view, to know whether the DNA genome is present, it is of clinical interest to know whether that genome is being expressed into mRNA or other RNA copies of the genome.
  • the amount of nucleic acid detected by the probe against the anti-sense strand will equal the amount of nucleic acid detected by the probe against the sense strand. If there is also viral mRNA present, then the amount of nucleic acid detected by the probe against the sense strand of DNA will exceed the amount of nucleic acid detected by the probe against the anti-sense strand of DNA. The excess will be due to the mRNA present.
  • the two-stranded target may be cellular DNA, cellular RNA, viral DNA, or viral RNA.
  • nucleic acid probe populations including all specific and preferred embodiments, disclosed here for use in those processes.
  • Related inventions are probe populations used in the above-noted process of the invention.
  • An example is a nucleic acid probe population wherein
  • each probe molecule is between about 15 nucleotides and about 100 nucleotides, 2) no probe molecule is totally complementary in nucleotide sequence to another probe molecule, and
  • each probe molecule is at least partially complementary in nucleotide sequence to at least one other probe molecule.
  • the nucleic acid probe may be DNA, RNA, or oligonucleotides or polynucleotides comprised of DNA or RNA.
  • the DNA or RNA may be composed of the bases adenosine, uridine, thymidine, guanine, cytosine, or any natural or artificial chemical derivatives thereof.
  • the probe is capable of binding to a complementary or mirror image target cellular genetic sequence through one or more types of chemical bonds, usually through hydrogen bond formation.
  • Nucleic acid probes may be detectably labeled prior to addition to the hybridization solution.
  • a detectable label may be selected which binds to the hybridization product.
  • Probes may be labeled with any detectable group for use in practicing the invention.
  • Such detectable group can be any " material having a detectable physical or chemical property.
  • detectable labels have been well-developed in the field of immunoassays and in general most any label useful in such methods can be applied to the present invention. Particularly useful are enzymatically active groups, suc as enzymes (see Clin. Chem.. 22:1243 (1976)), enzyme substrates (see British Pat. Spec. 1,548,741), coenzymes (see U.S.. Patents Nos.
  • nucleic acid probe is considered to include nucleic acids that have been labeled in any manner, including the foregoing manners.
  • Biotin labeled nucleotides can be incorporated into DNA or RNA by nick translatio enzymatic, or chemical means. The biotinylated probes are detected after hybridization using avidin/strepavidin, fluorescent, enzymatic or colloidal gold conjugates. Nucleic acid may also be labeled with other fluorescent compounds, with immunodetectable fluorescent derivatives or with biotin analogues. Nucleic acids may also be labeled by means of attaching a protein. Nucleic acids cross-linked to radioactive or fluorescent histone HI, enzymes (alkaline phosphatase and peroxidases), or single-stranded binding (ssB) protein may also be used. To increase the sensitivity of detecting the colloidal gol or peroxidase products, a number of enhancement or amplification procedures using silver solutions may be used.
  • PhotobiotinTM labeling of probes is preferable to biotin labeling.
  • Nucleic acid probes can be used against a variety of nucleic acid targets, viral, prokaryotic, and eukaryotic.
  • the target for probe populations of this invention will usually be a DNA target such as a gene (e.g., oncogene), control element (e.g., promote repressor, or enhancer), or sequence coding for ribosomal RNA, transfer RNA, or RNase P.
  • the target may be any nucleic acid target, either RNA or DNA that comprises one of the two complementary target nucleotide sequences; that will be the situation, for example, where the desire is to detect any DNA or mRNA molecule with a specific sequence or its complement.
  • the target may be RNA where, as in the case of some viruses, a viral RNA sequence and its RNA complement may be present in the same cell.
  • probes of any desired sequence can be made.
  • the target is a purified nucleic acid
  • a purified nucleic acid is considered here to be one that has been extracted from a cell or has been synthesized in vitro in a cell-free system. Many procedures have been published for hybridizing probes to such purified nucleic acids. Generally, if the target is a DNA molecule, its strands are separated by heat or other means before the hybridization step takes place. The hybridization can take place with the target immobilized on a solid support (e.g., nitrocellulose paper for DNA, nylon for RNA) by well-established procedures.
  • the probes may be labeled in the same way as probes are labeled for in situ experiments as described below; or they may be labeled in other detectable ways. The manner of labeling is not critical for implementation of this experiment. If a labeling procedure is known to work for probes against purified nucleic acid targets, it would be expected to work for probe populations where both strands are targeted.
  • the hybridization assay can be done for targets in biological entities in liquid suspension, in cells on slides or other solid supports, in tissue culture cells, and in tissue sections.
  • the biological entity can come from solid tissue (e.g., nerves, muscle, heart, skin, lungs, kidneys, pancreas, spleen, lymph nodes, testes, cervix, and brain) or cells present in membranes lining various tracts, conduits and cavities (such as the gastrointestinal tract, urinary tract, vas deferens, uterine cavity, uterine tube, vagina, respiratory tract, nasal cavity, oral cavity, pharynx, larynx, trachea, bronchi and lungs) or cells in an organism's fluids (e.g., urine, stomach fluid, sputum, blood and lymph fluid) o stool.
  • solid tissue e.g., nerves, muscle, heart, skin, lungs, kidneys, pancreas, spleen, lymph nodes, testes, cervix
  • a chaotropic agent such as 50% formamide
  • a buffer such as 0.1M sodium phosphate (pH 7.4)
  • about 100 micrograms (ug)/milliliter (ml) low molecular weight DNA to diminish non-specific binding 0.1% Trit
  • a probe population to hybridize with the target nucleic acids. If the cells are to be ultimately viewed on glass slides (or other soli supports), the cells as either single cell suspensions or as tissue slices are deposited on the slides. The cells are fixed by choosing a fixative which provides the best spatial resolution of the cells and the optimal hybridization efficiency. After fixation, the support bound cells may be dehydrated and stored at room temperature or the hybridization procedure may be carried out immediately. The hybridization solution containing the probe is added in an amount sufficient to cover the cells. The cells are then incubated at an appropriate temperature.
  • the hybridization solution may include a chaotropic denaturing agent, a buffer, a pore forming agent, a hybrid stabilizing agent, and the target-specific probe molecule.
  • the chaotropic denaturing agents include formamide, urea, thiocyanate, guanidine, trichloroacetate, tetramethylamine, perchlorate, and sodium iodide. Any buffer which maintains pH at least between 7.0 and 8.0 is preferred.
  • the pore forming agent is for instance, a detergent such as Brij 35, Brij 58, sodium dodecyl sulfate, CHAPSTM Triton X-100.
  • the pore-forming agent is chosen to facilitate probe entry through plasma, or nuclear membranes or cellular compartmental structures. For instance, 0.05% Brij 35 or 0.1% Triton X-100 will permit probe entry through the plasma membrane but not the nuclear membrane. Alternatively, sodium desoxycholate will allow probes to traverse the nuclear membrane. Thus, in order to restrict hybridization to the cytoplasmic biopolyme targets, nuclear membrane pore-forming agents are avoided.
  • Such selective subcellular localization contributes to the specificity and sensitivity of the assay by eliminating probe hybridization to complementary nuclear sequences when the target biopolymer is located in the cytoplasm.
  • Agents other than detergents such as fixatives may serve this function.
  • a biopolymer probe may also be selected such that its size is sufficiently small to traverse the plasma membrane of a cell but is too large to pass through the nuclear membrane.
  • Hybrid stabilizing agents such as salts of mono- and di-valent cations are included in the hybridization solution to promote formation of hydrogen bonds between complementary nucleotide sequences of the probe and its target biopolymer.
  • sodium chloride at a concentration from .15M to IM is used.
  • nucleic acids unrelated to the target biopolymers are added to the hybridization solution at a concentration of about 100 fold the concentration of the probe.
  • Specimens are removed after each of the above steps and analyzed by observation of cellular morphology as compared to fresh, untreated cells using a phase contrast microscope. The condition determined to maintain the cellular morphology and the spatial resolution of the various subcellular structures as close as possible to the fresh untreated cells is chosen as optimal for each step.
  • the cells Prior to nucleic acid hybridization, the cells may be reacted with antibodies in phosphate buffered saline. After hybridization one may analyze the cells for both bound antibodies and bound hybridization probes.
  • Supports which may be utilized include, but are not limited to, glass, Scotch tape (3M), nylon, Gene Screen Plus (New England Nuclear) and nitrocellulose. Most preferably glass microscope slides are used. The use of these supports and the procedures for depositing specimens thereon will be obvious to those of skill in the art. The choice of support material will depend upon the procedure for visualization of cells and the quantitation procedure used. Some filter materials are not uniformly thick and, thus, shrinking and swelling during in situ hybridization procedures is not uniform. In addition, some supports which autofluoresce will interfere with the determination of low level fluorescence. Glass microscope slides are most preferable as a solid support since they have high signal-to-noise ratios and can be treated to better retain tissue.
  • a fixative may be selected from the group consisting of any precipitating agent or cross-linking agent used alone or in combination, and may be aqueous or non-aqueous.
  • the fixative may be selected from the group consisting of formaldehyde solutions, alcohols, salt solutions, mercuric chloride sodium chloride, sodium sulfate, potassium dichromate, potassium phosphate, ammonium bromide, calcium chloride, sodium acetate, lithium chloride, cesium acetate, calcium or magnesium acetate, potassium nitrate, potassium dichromate, sodium chromate, potassium iodide, sodium iodate, sodium thiosulfate, picric acid, acetic acid, paraformaldehyde, sodium hydroxide, acetones, chloroform, glycerin, thymol, etc.
  • the fixative will comprise an agent which fixes the cellular constituents through a precipitating action and has the following characteristics: the effect is reversible, the cellular (or viral) morphology is maintained, the antigenicity of desired cellular constituents is maintained, the nucleic acids are retained in the appropriate location in the cell, the nucleic acids are not modified in such a way that they become unable to form double or triple stranded hybrids, and cellular constituents are not affected in such a way so as to inhibit the process of nucleic acid hybridization to all resident target sequences.
  • Choice of fixatives and fixation procedures can affect cellular constituents and cellular morphology; such effects can be tissue specific.
  • fixatives for use in the invention are selected from the group consisting of ethanol, ethanol-acetic acid, methanol, and methanol-acetone which fixatives afford the highest hybridization efficiency with good preservation of cellular morphology.
  • Fixatives for practicing the present invention include 95% ethanol/5% acetic acid for HL-60 and normal bone marrow cells, 75% ethanol/20% acetic acid for K562 and normal peripheral blood cells, 50% methanol/50% acetone for fibroblast cells and normal bone marrow cells, and 10% formaldehyde/90% methanol for cardiac muscle tissue. These fixatives provide good preservation of cellular morphology and preservation and accessibility of antigens, and high hybridization efficiency.
  • the fixative may contain a compound which fixes the cellular components by cross-linking these materials together, for example, glutaraldehyde or formaldehyde. While this cross-linking agent must meet all of the requirements above for the precipitating agent, it is generally more "sticky" and causes the cells and membrane components to be secured or sealed, thus, maintaining the characteristics described above.
  • the cross linking agents when used are preferably less than 10% (v/v).
  • Cross-linking agents while preserving ultrastructure, often reduce hybridization efficiency; they form networks trapping nucleic acids and antigens and rendering them inaccessible to probes and antibodies. Some also covalently modify nucleic acids preventing later hybrid formation. Storage of Biological Entities/Tissues
  • microscope slides containing cells may be stored air dried at room temperature for up to three weeks, in cold (4°C) 70% ethanol in water for 6-12 months, or in paraplast for up to two years. If specimens are handled under RNase free conditions, they can be dehydrated in graded alcohols and stored for at least 5 months at room temperature.
  • Reagents can be purchased from any of a variety of sources including Aldrich Chemical Co., Milwaukee, Wisconsin, Sigma Chemical Co., St. Louis, Missouri, Molecula Probes, Inc., Eugene, Oregon, Clontech, Palo Alto, California, Kodak, Rochester, NY, and SPectrum Chemical Manufacturing Corp., Gardenea, California.
  • cells either as single cell suspensions or as tissue slices may be deposited on solid supports such as glass slides.
  • cells are placed into a single cell suspension of about 10 5 -10 6 cells per ml. The cells are fixed by choosing a fixative which provides the best spatial resolution of the cells and the optimal hybridization efficiency.
  • the hybridization is then carried out in the same solution which effects fixation.
  • This solution contains both a fixative and a chaotropic agent such as formamide.
  • a hybrid stabilizing agent such as concentrated lithium chlorid or ammonium acetate solution, a buffer, low molecular weight DNA and/or ribosomal RNA (sized to 50 bases) to diminish non-specific binding, and a pore forming agent to facilitate probe entry into the cells.
  • Nuclease inhibitors such as vanadyl ribonucleoside complexes may also be included.
  • a probe or probes
  • the one-step procedure is a means of carrying out the fixation, prehybridization, hybridization and detection steps normally associated with in situ hybridization procedures all in one step.
  • a convenient temperature may be used to carry out the hybridization reaction.
  • this provides a hybridization assay which can be accomplished with viable or non-viable cells in solution. In either case, the assay is rapid and sensitive.
  • the hybridization procedure is carried out utilizing a single hybridization solution which also fixes the cells. This fixation is accomplished in the same solution and along with the hybridization reaction.
  • the fixative may be selected from the group consisting of any precipitating agent or cross-linking agent used alone or in combination, and may be aqueous or non-aqueous.
  • Tissue samples are broken apart by physical, chemical or enzymatic means into single cell suspension.
  • Cells are placed into a PBS solution (maintained to cellular osmolality with bovine serum albumin (BSA) at a concentration of 10 5 to 10° cells per ml.
  • BSA bovine serum albumin
  • Cells in suspension may be fixed and processed at a later time, fixed and processed immediately, or not fixed and processed in the in situ hybridization system of the present invention.
  • a single solution is added to the cells/tissues (hereafter referred to as the specimen).
  • This solution contains the following: a mild fixative, a chaotrope, a nucleic acid probe (RNA or DNA probe which is prelabeled) and/or antibody probe, salts, detergents, buffers, and blocking agents.
  • the incubation in this solution can be carried out at 55 °C for 20 minutes as well as other conditions such as those in the Examples below.
  • the fixative is one which has been found to be optimal for the particular cell type being assayed (eg., there is one optimal fixative for bone marrow and peripheral blood even though this "tissue" contains numerous distinct cell types).
  • the fixative is usually a combination of precipitating fixatives (such as alcohols) and cross-linking fixatives (such as aldehydes), with the concentration of the cross-linking fixatives kept very low (less than 10%). Frequently, the solution contains 10-40% ethanol, and 5% formalin.
  • concentration and type of precipitating agent and crosslinking agent may be varied depending upon the probe and the stringency requirements of the probe, as well as the desired temperature of hybridization. Typical useful precipitating and cross-linking agents are specified in PCT applications WO 90/02173 and WO 90/02204.
  • the hybridization cocktail contains a denaturing agent, usually formamide at about 30% (v/v), but other chaotropic agents such as Nal, urea, etc. may also be used. Furthermore, several precipitating and/or cross-linking fixatives also have mild denaturing properties; these properties can be used in conjunction with the primary denaturant in either an additive or synergistic fashion.
  • the hybridization cocktail may be constructed to preferentially allow only the formation of RNA-RNA or RNA-DNA hybrids. This is accomplished by adjusting the concentration of the denaturing agents along with the concentration of salts (primarily monovalent cations of the Group I series of metals along with the ammonium ion) and along with the temperature of hybridization which is used.
  • the present invention may be provided in the form of a kit adapted for a one-step process.
  • kits for detecting a nucleic acid molecule in a biological entity comprising a probe population described herein and one more reagents for use in a solution for reacting said probe population with said biological entity so that a hybrid molecule can form between a molecule of the probe population and a nucleic acid molecule in the biological entity.
  • the biological entity is a cell and the one or more reagents comprise a reagent selected from the group, a fixative and a chaotropic agent.
  • kits could include a solution containing a .fixation hybridization cocktail and one or more labeled probes.
  • This solution could, for example, contain 15-40% ethanol, 25-40% formamide, 0-10% formaldehyde, 0.1-1.5 M LiCl, 0.05-0.5 M Tris-acetate (pH 7-8), 0.05%-0.15% Triton X-100, 20 ug/ml-200 ug/ml of a non-specific nucleic acid which does not react with the probe(s), and 0.1 ug/ml to 10 ug/ml of single stranded probes directly labeled with a reporter molecule.
  • this solution could contain 30% ethanol, 30% formamide, 5% formaldehyde, 0.8M LiCl, 0.1M Tris-acetate (pH 7.4), 0.1% Triton X-100, 50 ug/ml of the non-specific nucleic acid, and 2.5 ug/ml of each single stranded probes directly labeled with a fluorescent reporter molecule.
  • kits may also include:
  • a second detectable reporter system which would react with the probe or the probe-target hybrid.
  • Any mechanical components which may be necessary or useful to practice the present invention such as a solid support (e.g. a microscope slide), an apparatus to affix cells to said support, or a device to assist with any incubations or washings of the specimens. 4. A photographic film or emulsion with which to record results of assays carried out with the present invention.
  • the H9 cell line was used in the following experiment. Cultured cells were washed with nuclease-free Phosphate Buffered Saline (PBS) and placed in a single cell suspension at a concentration that resulted in clearly separated cells. The cells were spun down to a pellet and the supernatant, drained off. The cells were resuspend in 40% ethanol, 50% PBS, and 10% glacial acetic acid and left for 12-16 hours at 4°C. After fixation, the cells were spun to remove the fixative and then washed once in IX PBS and resuspend in 2X SSC. The cells should be used immediately.
  • PBS nuclease-free Phosphate Buffered Saline
  • a conserved segment of the eukaryotic 28S rRNA was designed and utilized; it was designated 28S-25-AL (SEQ ID NO:l) and it served as a positive probe for the experiment described herein.
  • the negative probe designated NR- 25-AL (SEQ ID NO:9), was derived from the nitrogen reductase gene found in bacteria and was known to not hybridize to nucleic acid within eukaryotic cells.
  • the DNA sequences for these two probes used are shown in Table 1 below. Twelve base, ten base, eight base, and six base oligomers, derived from these 25-base oligomers were also prepared with the sequences shown in the Table 1 below. All sequences displayed in the Examples have the 5' end as the left end of the sequence.
  • NR 25-AL TACGCTCGATCCAGCTATCAGCCGT (SEQ ID NO:9)
  • NR 12-AL TACGCTCGATCC (SEQ ID NO:10)
  • NR 10-AL TACGCTCGAT (SEQ ID NO:11)
  • NR8-AL TACGCTCG (SEQ ID NO:12)
  • NR6-AL TACGCT (SEQ ID NO:13)
  • the ohgodeoxynucleotides were synthesized (Applied Biosystems DNA Synthesizer Model 380 B using the recommended A.B.I. reagents), and in the last stage an aminohexyl linker was attached to the 5' end phosphate.
  • the 5'-aminohexyl ohgodeoxynucleotides were purified and coupled to a rhodamine derivative from Molecular Probes and purified by Waters HPLC using a baseline 810 chromatography work station.
  • hybridization procedure to pelleted cells was added 50 ⁇ l of an hybridization cocktail consisting of 30% formamide, 5X SSC, 0.16M sodium phosphate buffer, pH 7.4, 1 ⁇ g/ ⁇ l sheared DNA, 3% (v/v) Triton X-100 (alcohol derivative of polyoxylene ether, se Aldrich Chemical Co. catalogue for 1990-91), 5% PEG 4000 (polyethylene glycol), 25 mM DTT (dithiothreitol), 0.4 M guanidinium isothiocyanate, 15X Ficoll/PVP, and the probe added at a concentration of 2.5 ⁇ g/ml. Hybridizations were carried out at 42°C fo 30 minutes.
  • 500X Ficoll/PVP is 5g of Ficoll type 400 (polysucrose 400,000 mol wt) plus 5 g of PVP (polyvinylpyrollidone) dissolved in water to a total volume of 100 ml; 15X FIcoll/PVP is 500X Ficoll/PVP diluted with water by a factor of 15/500.
  • the cells were analyzed on a Profile IITM made by Coulter Instruments.
  • the Instrument uses a 488nm argon laser, a 525nm band pass filter for FL1 and a 635nm band pass filter for the counterstain.
  • the sample containing the negative probe was analyzed first and the quad-stats were set so that less than 0.01% of the cells fell in the upper-right quadrant.
  • the sample analyzed with the positive probe was analyzed under the exact same parameters as the sample analyzed with the negative probe. Since the quad-stats were set correctly and the two samples had been handled identically, any number of cells (above 0.01%) that were recorded in the upper right quadrant were scored as positive.
  • H9 cells Approximately 500,000 H9 cells were equally divided into two tubes and fixed a described above. For one of these sample aliquots was added a hybridization solution containing a positive probe (28S) and to the other a negative probe (NR), corresponding to the same size as the positive probe as in the list in Table 1 above. Following hybridization and washing, flow cytometry was performed.
  • 28S positive probe
  • NR negative probe
  • nucleotide sequence in the top strand starting at position 100 and endin at position 212 is SEQ ID NO:25 for a segment of double-stranded DNA.
  • nucleotide sequence in the top strand starting at position 288 and endin at position 403 is SEQ ID NO:27 for a segment of double-stranded DNA.
  • sequences H18-10, H18-11, H18-100L, H18-100R, and H18-110R ar SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, and SEQ ID NO: 18 respectively.
  • the ohgodeoxynucleotides were synthesized (Applied Biosystems DNA Synthesize Model 380 B using the recommended A.B.I. reagents), and in the last stage an aminohexy linker was attached to the 5' end phosphate.
  • the 5'-aminohexyl ohgodeoxynucleotides were then coupled to a rhodamine dye from Clontech and purified by Waters HPLC using baseline 810 chromatography work station.
  • hybridization procedure to the cells deposited onto the slides was added 20 to 25 ⁇ l of a hybridization cocktail consisting of 30% formamide, 5X SSC, O.l M sodiu phosphate buffer, pH 7.4, 100 ⁇ g/ml low molecular weight, denatured, salmon or herrin sperm DNA, 5% (v/v) Triton X-100, 15X Ficoll/ PVP, 0.4 M guanidinium isothiocaynate, 1 mM DTT, and 0.025 M EDTA and the probe, added at a concentration of 2.5 ⁇ g/ml. Denaturation and hybridization was carried out simultaneously by placing the slides in an incubator for 15 minutes at 85°C.
  • HTB 31 "C-33A” is a human cervical carcinoma derived cell line from cervical cancer biopsies (J. National Cancer Institute 32:135-148, 1964) and contains no human papilloma virus was used as the negative control.
  • Culture Media Eagles MEM with non-essential Amino Acids, sodium pyruvate, 10% fetal bovine serum.
  • CCL 1550 "CAski” is a human cervical carcinoma cell line containing 400-500 copies of HPV16 integrated into its genome.(Science 196:1456-1458, 1977), and was used as the positive control.
  • Cells from both cell lines were grown to confluence in 5% C02, in 100 ml culture flasks. They were rinsed 1 time in IX PBS. To the cells was added 2 ml of 0.25% Trypsin, in 0.02 EDTA. These were incubated for 5 minutes at 37 °C, gently tapped to dislodge cells. To these cells were add 10 ml. of their respective media. 5 x 10 3 cells were then cytospun for 7 minutes at 700 rpm's onto clean glass slides, and left to air dry. To these cells was added 20 ul of ethano methanol (3:1). They were then allowed to air dry.
  • the ohgodeoxynucleotides were synthesized (Applied Biosystems DNA Synthesizer Model 380 B using the recommended A.B.I. reagents), and in the last stage an aminohexyl linker was attached to the 5' end phosphate.
  • the 5'-aminohexyl ohgodeoxynucleotides were then coupled to a rhodamine dye from Clontech and purified by Waters HPLC using a baseline 810 chromatography work station.
  • hybridization procedure 20 ⁇ l of an hybridization cocktail consisting of PEG 21%, formamide 25%, 5X SSC, salmon sperm DNA 1 mg/ml, Ficoll/PVP 15X, 0.4 M guanidinium isothiocyanate, 50 mM DTT, 5% Triton X-100, 50 mM EDTA, 50 mM Na 2 P0 4 and probe at a concentration of 0.06 ug/ul is added to the slide. A coverslip was applied and the slide was heated to 95°C for 5 minutes, allowed to cool to 42°C and incubated for 25 minutes at that temperature.
  • Fluorescence Detection Photomicrographs were taken on an Olympus BH10 microscope with fluorescence capabilities, using Kodak Ektachrome EES-135 (PS 800/1600) film, exposed, and pus processed at 1600 ASA. A 20-second exposure time was consistently used, so that direc comparisons could be made between all photomicrographs taken.
  • the cell lines C-33A and Caski were used to determine the intensity difference between the signal obtained using probes directed at one strand of the DNA vs probes directed at both strands ("staggered overlap" probes).
  • the sequences for the 25-base synthetic oligonucleotide probes listed below and designated HYR 7 were obtained from the published sequences for the alpha centromeric repetitive DNA sequence on the Y chromosome. Twelve base, ten base, eight base, and six base oligomers, derived from these 25-base oligomers were also prepared as shown in the Table 5 below.
  • the ohgodeoxynucleotides were synthesized (Applied Biosystems DNA Synthesizer Model 380 B using the recommended A.B.I. reagents), and in the last stage an aminohexyl linker was attached to the 5' end phosphate.
  • the 5'-aminohexyl ohgodeoxynucleotides were then coupled to a rhodamine dye from Clontech and purified by Waters HPLC using a baseline 810 chromatography work station.
  • hybridization procedure to the cells deposited onto the slides was added 20 to 25 ⁇ l of a hybridization cocktail consisting of 30% formamide, 5X SSC, O.IM sodium phosphate buffer, pH 7.4, 100 ⁇ g/ml low molecular weight, denatured, salmon or herring sperm DNA, 5% (v/v) Triton X-100, 15X Ficoll/ PVP, 0.4 M guanidinium isothiocaynate, 10 mM DTT, and 0.025 M EDTA and the probe, added at a concentration of 2.5 ⁇ g/ml. Denaturation and hybridization was carried out simultaneously by placing the slides in an incubator for 15 minutes at 85°C.
  • the cell line (GM 02504G, Coriell Inst. of Med. Research, Camden NJ), grown as a monolayer and were trypinsized.
  • DNA isolated essentially by the method of Maniatis et al (Molecular Cloning. T. Maniatis, E.F. Fritsch and J. Sambrook, eds., Cold Spring Harbor Laboratory, NY, 1982) and digested to completion using restriction enzymes Bam HI and EcoRl under conditions described by Maniatiset al. Then an aliquot of 10 ug of each digested DNA was electrophoresed from a 2 mm-wide slot through a 1.25 percent agarose gel. The electrophoretically fractionated DNA was then immobilized on nitrocellulose filter paper using the procedure of Southern (see Maniatis et al). Preparation of Probes
  • the sequences for the 25-base synthetic oligonucleotide probes listed below and designated HYR 7 were obtained from the published sequences for the alpha centromeric repetitive DNA sequence on the Y chromosome. Twelve base, ten base, eight base, and six base oligomers, derived from these 25-base oligomers were also prepared as shown in the Table 6 below.
  • the probes were then end labeled with digoxigenin at the 3' end using an end labeling kit from Boehringer Mannheim Biochemicals (BMB) and using the BMB recommended procedure.
  • BMB Boehringer Mannheim Biochemicals
  • the filters were cut to a size of about 10 cm x 2 cm and were incubated for 3 hrs at 65° C in a pre-hybridization solution followed by incubation at 56° C overnight in a hybridization solution containing end-labeled oligonucleotide probes.
  • the hybridization cocktail consisted of 30% formamide, 5X SSC, 0.1M sodium phosphate buffer, pH 7.4, 100 ug/ml low molecular weight denatured salmon or herring sperm DNA, 5% (v/v) Triton X-100, 15X Ficoll/PVP, 0.4 M guanidinium isothiocyanate, 10 mM DTT, and 0.025 M EDTA and the probe, added at a concentration of 2.4 ug/ml (micrograms/ml).
  • the filters were washed, blocked, equilibrated and reacted with anti- anti-digoxigenin/alkaline phosphatase conjugate according to BMB protocol and soaked in the substrate (lumipos 530, BMB). The filters were then exposed to x-ray film and the films were developed.
  • This Example demonstrates that oligomers prepared to both strands of a DNA targe and that the results can be monitored by flow cytometry. It also demonstrates the ability t hybridize to both DNA strands allows one to quantitate simultaneously the amount of DN and RNA within individual cells.
  • the H9 cell line is used in the following experiment. Cultured cells are washed wit nuclease-free Phosphate Buffered Saline (PBS) and placed in a single cell suspension at concentration that results in clearly separated cells. The cells are spun down to a pellet an the supernatent drained off. The cells are resuspended in 40% ethanol, 50% PBS, and 10 glacial acetic acid and left for 12-16 hours at 4°C. After fixation, the cells are spun t remove the fixative and then washed once in IX PBS and resuspended in 2X SSC. The cell should be used immediately.
  • PBS nuclease-free Phosphate Buffered Saline
  • HIV sequences used as probes are accessed via GenBank, version 69.0, prepare as probe by cutting them into 30-mers as described in figure 2, for HPV sequences. Thi design results in an "overlap" region of 15 bases. Probe Designation
  • sequences are cut into 30-base oligonucleotides and synthesized as phosphorothioate oligonucleotides using DNA synthesizers (Applied Biosystem DNA Synthesizer, Model 380B) and using the recommended ABI ' reagents.
  • the polysulfurized oligonucleotides are then coupled to a fluorescent dye and purified by column chromatography and HPLC.
  • 30-base NR oligonucleotides (30-mers) serve as the negative control probes.
  • Probes are made as phosphorothioate oligonucleotides, each 30-mer having four sulfur atoms, using an Applied Biosystem (ABI) DNA Synthesizer, Model 380B and the recommended ABI reagents.
  • the sulfur atoms are located as follows: one is at the extreme 5' end of the probe, a second is between the 7th and 8th nucleosides (counting from the 5' end), the third is between the 22nd and 23rd nucleosides, and the fourth is between the 29th and 30th nucleosides.
  • the sulfur atoms of the polysulfurized oligonucleotides are then coupled to a fluorescent dye, iodoacetamido-fluorescein, as follows (smaller amounts can be synthesized by adjusting the volumes): 200 ⁇ g of dried oligonucleotide is dissolved in 100 ⁇ l of 250 mM Tris buffer, pH 7.4 to form a first solution. Then one mg of iodoacetamido- fluorescein is combined with 100 ⁇ l of dry dimethylformamide (i.e., 100 percent DMF) in a second solution. The two solutions are mixed together and shaken overnight.
  • a fluorescent dye iodoacetamido-fluorescein
  • the labeled oligonucleotide is precipitated with ethanol and 3M sodiu acetate. This crude material is then loaded on to a PD-10 column to remove free dye. Th desired fractions are then collected. The liquid phase is then removed under vacuum. Th crude material is then purified with HPLC (high performance liquid chromatography).
  • Hybridizations are carried out at 42°C for 30 minutes.
  • the cells are placed in a 15 ml conica tube to which is added 10 ml of a wash solution, consisting of .IX SSC, .4M guanidiniu isothiocyanate, and .1% Triton at a temperature of 42°C.
  • a wash solution consisting of .IX SSC, .4M guanidiniu isothiocyanate, and .1% Triton at a temperature of 42°C.
  • the solution is agitated until th cells are a single cell suspension and then spun at 250 X g for 5 minutes.
  • the supernatan is removed and to the pellet is added 10 ml of a wash solution, consisting of .IX SSC, .1 Triton at a temperature of 42°C.
  • the solution is agitated until the cells are a single cel suspension.
  • the cells are spun at 250 X g for 5 minutes.
  • the supernatant is removed an the cell pellet resuspended in 0.2 ml counterstain solution consisting of .0025% Evans Blu in IX PBS.
  • the cells are analyzed on a FACSTARTM made by Beckon Dickinson.
  • the Instrumen uses a 5 watt argon laser coupled to a dye head, a 525nm band pass filter for FLl and a 584nm band pass filter for the Rhodamine.
  • the sample containing the negative probe is analyzed first and the quad-stats are set so that less than 0.01% of the cells fall in the upper-right quadrant or lower-right quandant.
  • the sample analyzed with the HIV probes is analyzed under the exact same parameters as the sample analyzed with the negative probe.
  • any number of cells (about 0.01%) that are recorded in the upper right quadrant are scored as positive for both strands and/or mRNA. Any number of cells (above 0.01%) that are recorded in the lower right quadrant are scored positive for DNA only.
  • the Histogram is constructed so that FL-3 is the Y axis and FL-1 is the X axis.
  • Example 4or 6 can be followed with one or more of the following changes: 1) the hybridization cocktail additionally contains 10% DMSO (v/v) and 5% (v/v) vitamin
  • MOLECULE TYPE cDNA to rRNA
  • HYPOTHETICAL N
  • MOLECULE TYPE cDNA to rRNA
  • HYPOTHETICAL N
  • MOLECULE TYPE cDNA to rRNA
  • HYPOTHETICAL N
  • MOLECULE TYPE cDNA to rRNA
  • HYPOTHETICAL N
  • MOLECULE TYPE cDNA to rRNA
  • HYPOTHETICAL N
  • MOLECULE TYPE cDNA to rRNA
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:9: TACGCTCGAT CCAGCTATCA GCCGT 25
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:11: TACGCTCGAT 10
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:12: TACGCTCG 8
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:13: TACGCT 6
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:14: ACTCTACACA CATGAGTGTG ATTCT 25
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:17: ACTCACACTA AGAGAATTGT TCCAC 25
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:20: GAGTCGATTT TATTG 15
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:28: GTTTCAAAAC TG 12
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:31: TCTCGCCCAG TGCCACGCCT AGGAT 25
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:33: TAAAGTTGTA GACCCTGCTT TTGTA 25
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:34: ACCACTCCCA CTAAACTTAT TACAT 25
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:39: TATAGTTGCT TTACATAGGC CAGCA 25
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:40: TTAACCTCTA GGCGTACTGG CATTA 25
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:41: GGTACAGTAG AATTGGTAAT AAACA 25
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:43: GTGCTACTAG TTACTGTGTT 20
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:47: AGTGGGAGTG GTTACAAAAG CAGGG 25
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:51 GATCTGGAGC TATATTAATA CTATT 25
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • SEQUENCE DESCRIPTION SEQ ID NO:54 TTCTACTGTA CCTAATGCCA GTACG 25

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Engineering & Computer Science (AREA)
  • Analytical Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Pathology (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

L'invention concerne des procédés et des populations de sondes qui permettent une hybridation efficace de sondes au niveau des deux brins d'un acide nucléique cible bicaténaire sans qu'il y ait une auto-hybridation importante à l'intérieur de la population de sondes.
EP93917236A 1992-07-17 1993-07-16 Sondes d'acide nucleique et leur utilisation dans la detection d'acides nucleiques bicatenaires. Withdrawn EP0672185A4 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US91590092A 1992-07-17 1992-07-17
US915900 1992-07-17
PCT/US1993/006715 WO1994002643A1 (fr) 1992-07-17 1993-07-16 Sondes d'acide nucleique et leur utilisation dans la detection d'acides nucleiques bicatenaires

Publications (2)

Publication Number Publication Date
EP0672185A1 true EP0672185A1 (fr) 1995-09-20
EP0672185A4 EP0672185A4 (fr) 1997-04-23

Family

ID=25436402

Family Applications (1)

Application Number Title Priority Date Filing Date
EP93917236A Withdrawn EP0672185A4 (fr) 1992-07-17 1993-07-16 Sondes d'acide nucleique et leur utilisation dans la detection d'acides nucleiques bicatenaires.

Country Status (5)

Country Link
EP (1) EP0672185A4 (fr)
CN (1) CN1088260A (fr)
AU (2) AU4681693A (fr)
IL (1) IL106379A0 (fr)
WO (2) WO1994002500A1 (fr)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996000234A1 (fr) * 1994-06-23 1996-01-04 Aprogenex, Inc. Sondes d'hybridation centromeres
DE19610255B4 (de) * 1996-03-15 2004-11-04 Universität Heidelberg Verfahren zur Herstellung von Nukleinsäuresequenzen und Verfahren zum Nachweis von Translokationen zwischen Chromosomen
FR2770539B1 (fr) * 1997-10-30 2001-07-27 Jean Gabert Methode de diagnostic in vitro de pathololgies associees a des remaniements geniques et trousses de diagnostic
GB0821457D0 (en) * 2008-11-24 2008-12-31 Trillion Genomics Ltd Oligonucleotides
AU2016281718B2 (en) * 2015-06-24 2022-03-31 Dana-Farber Cancer Institute, Inc. Selective degradation of wild-type DNA and enrichment of mutant alleles using nuclease

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0318245A2 (fr) * 1987-11-24 1989-05-31 Gen-Probe Incorporated Moyen et méthode pour augmenter l'hybridation d'acide nucléique
EP0385410A2 (fr) * 1989-02-28 1990-09-05 Canon Kabushiki Kaisha Oligonucléotide à double brin partiel et son procédé de formation

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4868105A (en) * 1985-12-11 1989-09-19 Chiron Corporation Solution phase nucleic acid sandwich assay
US4925785A (en) * 1986-03-07 1990-05-15 Biotechnica Diagnostics, Inc. Nucleic acid hybridization assays
US5024934A (en) * 1988-03-14 1991-06-18 The Board Of Regents, The University Of Texas System Detection of minimal numbers of neoplastic cells carrying DNA translocations by DNA sequence amplification
US4999290A (en) * 1988-03-31 1991-03-12 The Board Of Regents, The University Of Texas System Detection of genomic abnormalities with unique aberrant gene transcripts
US5198338A (en) * 1989-05-31 1993-03-30 Temple University Molecular probing for human t-cell leukemia and lymphoma

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0318245A2 (fr) * 1987-11-24 1989-05-31 Gen-Probe Incorporated Moyen et méthode pour augmenter l'hybridation d'acide nucléique
EP0385410A2 (fr) * 1989-02-28 1990-09-05 Canon Kabushiki Kaisha Oligonucléotide à double brin partiel et son procédé de formation

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of WO9402643A1 *

Also Published As

Publication number Publication date
IL106379A0 (en) 1993-11-15
CN1088260A (zh) 1994-06-22
WO1994002643A1 (fr) 1994-02-03
EP0672185A4 (fr) 1997-04-23
WO1994002500A1 (fr) 1994-02-03
AU4774293A (en) 1994-02-14
AU4681693A (en) 1994-02-14

Similar Documents

Publication Publication Date Title
US5521061A (en) Enhancement of probe signal in nucleic acid-mediated in-situ hybridization studies
EP1287165B1 (fr) Méthode pour la détection et la localisation de gènes in situ à l'aide d'hybridation d'ADN branché
RU2618868C2 (ru) Композиции и способы определения хромосомных аберраций с новыми буферами для гибридизации
US7368245B2 (en) Method and probes for the detection of chromosome aberrations
Bauman et al. Cytochemical hybridization with fluorochrome-labeled RNA. II. Applications.
EP0662151A1 (fr) Composes reducteurs de fond pour essais fluorimetriques in-situ a mediation par sonde
JP2012065660A (ja) イン・サイチュー解析のためのオリゴヌクレオチドプローブおよびタンパク質を標識するためのオリゴヌクレオチド
WO1994002645A1 (fr) Detection rapide de biopolymeres dans des echantillons traites par un colorant
EP0357436A2 (fr) Essai d'hybridation "in situ" en une seule étape
EP0662153A1 (fr) Piegeurs de radicaux libres servant a reduire l'autofluorescence dans des cellules fixes
JP2002538838A (ja) 黒色腫の検出
EP0862650B1 (fr) Hybridation in situ pour detecter des sequences d'acides nucleiques specifiques dans des echantillons eucaryotes
WO1994002643A1 (fr) Sondes d'acide nucleique et leur utilisation dans la detection d'acides nucleiques bicatenaires
WO1996000234A1 (fr) Sondes d'hybridation centromeres
JP2005503753A5 (fr)
WO1995019449A1 (fr) Populations de sondes d'hybridation non adjacentes
WO1994002644A9 (fr) Detection in situ d'acides nucleiques utilisant l'amplification 3sr
JP5476989B2 (ja) ゲノムdna断片の増幅または欠失の検出方法
WO1994002644A1 (fr) Detection in situ d'acides nucleiques utilisant l'amplification 3sr
WO1994002640A1 (fr) Sondes d'acide nucleique marquees par plusieurs reporters
WO1994002641A1 (fr) Analogues de groupes reporters utilises comme reducteurs de fond dans des essais d'hybridation
WO1995019450A1 (fr) Composes attenuant la fluorescence du fond dans des dosages fluorimetriques effectues in situ a l'aide de sondes

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19950421

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LI LU MC NL PT SE

A4 Supplementary search report drawn up and despatched

Effective date: 19970306

AK Designated contracting states

Kind code of ref document: A4

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LI LU MC NL PT SE

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Withdrawal date: 19970603