US20030027313A1 - Cloned genes encoding reverse transcriptase lacking RNase H activity - Google Patents

Cloned genes encoding reverse transcriptase lacking RNase H activity Download PDF

Info

Publication number
US20030027313A1
US20030027313A1 US10/024,131 US2413101A US2003027313A1 US 20030027313 A1 US20030027313 A1 US 20030027313A1 US 2413101 A US2413101 A US 2413101A US 2003027313 A1 US2003027313 A1 US 2003027313A1
Authority
US
United States
Prior art keywords
leu
pro
ala
gln
thr
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
US10/024,131
Inventor
Michael Kotewicz
Gary Gerard
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.)
Individual
Original Assignee
Individual
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=22503881&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20030027313(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Individual filed Critical Individual
Priority to US10/024,131 priority Critical patent/US20030027313A1/en
Publication of US20030027313A1 publication Critical patent/US20030027313A1/en
Priority to US10/970,280 priority patent/US20060003341A1/en
Priority to US11/754,968 priority patent/US20080004437A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P19/00Preparation of compounds containing saccharide radicals
    • C12P19/26Preparation of nitrogen-containing carbohydrates
    • C12P19/28N-glycosides
    • C12P19/30Nucleotides
    • C12P19/34Polynucleotides, e.g. nucleic acids, oligoribonucleotides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1241Nucleotidyltransferases (2.7.7)
    • C12N9/1276RNA-directed DNA polymerase (2.7.7.49), i.e. reverse transcriptase or telomerase
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/07Nucleotidyltransferases (2.7.7)
    • C12Y207/07049RNA-directed DNA polymerase (2.7.7.49), i.e. telomerase or reverse-transcriptase
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/81Packaged device or kit
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/975Kit

Definitions

  • the invention is in the field of recombinant genetics.
  • RT reverse transcriptase
  • DNA polymerase DNA polymerase
  • RNase H ribonuclease H
  • RNA Tumor Viruses In the retrovirus life cycle, the RT DNA polymerase activity is responsible for transcribing viral RNA into double-stranded DNA.
  • Varmus, H. (1982) in Weiss, R., et al. (eds.), RNA Tumor Viruses, Cold Spring Harbor Laboratory, pp. 410-423.
  • the function of RNase H in replication is less clear, but it is thought to degrade genomic RNA during DNA synthesis to generate oligomeric RNA primers for plus-strand DNA synthesis, and to remove the RNA primers of both minus- and plus-strand DNA.
  • RT is used extensively in recombinant DNA technology to synthesize cDNA from mRNA.
  • One major problem with cDNA synthesis is that the RNase H activity of RT degrades the mRNA template during first-strand synthesis.
  • the mRNA poly(A)-oligo(dT) hybrid used as a primer for first-strand cDNA synthesis is degraded by RT RNase H.
  • a competition is established between RNase H-mediated deadenylation of mRNA and initiation of DNA synthesis, which reduces the yield of cDNA product. Berger, S. L., et al., Biochem. 22:2365-2373 (1983).
  • the RNase H causes premature termination of DNA chain growth. Unfortunately, these events eliminate the potential for repeated copying of the RNA template.
  • the invention relates to a gene which encodes reverse transcriptase having DNA polynerase activity and substantially no RNase H activity.
  • the invention also relates to a reverse transcriptase gene comprising the following DNA sequence: 1078 A ⁇ dot over (T) ⁇ G ACC CTA AA ⁇ dot over (T) ⁇ ATA GAA GAT GAG CAT CGG C ⁇ dot over (T) ⁇ A CAT GAG AC ⁇ dot over (C) ⁇ TCA AAA GAG ⁇ dot over (C) ⁇ CA GAT GTT MET Thr Leu Asn Ile Glu Asp Glu His Arg Leu His Glu Thr Ser Lys Glu Pro Asp Val 1138 TCT CTA GGG TCC ACA TGG CTG TCT GAT TTT CCT GAG GCC TGG GCG GAA ACC GGG GGC ATG Ser Leu Gly Ser Thr Trp Leu Ser Asp Phe Pro Gln Ala Trp Ala Glu Thr Gly Gly MET 1198 GGA CTG GCA GTT CGC CAA GCT CCT CTG ATC ATA CCT CTG AAA GCA
  • the invention also relates to a reverse transcriptase gene comprising the following DNA sequence: 1078 A ⁇ dot over (T) ⁇ G ACC CTA AA ⁇ dot over (T) ⁇ ATA GAA GAT GAG CAT CGG C ⁇ dot over (T) ⁇ A CAT GAG AC ⁇ dot over (C) ⁇ TCA AAA GAG ⁇ dot over (C) ⁇ CA GAT GTT MET Thr Leu Asn Ile Glu Asp Glu His Arg Leu His Glu Thr Ser Lys Glu Pro Asp Val 1138 TCT CTA GGG TCC ACA TGG CTG TCT GAT TTT CCT CAG GCC TGG GCG GAA ACC GGG GGC ATG Ser Leu Gly Ser Thr Trp Leu Ser Asp Phe Pro Gln Ala Trp Ala Glu Thr Gly Gly MET 1198 GGA CTG GCA GTT CGC CAA GCT CCT CTG ATC ATA CCT CTG AAA GCA
  • the invention also relates to the vectors containing the gene of the invention, hosts transformed with the vectors of the invention, and the reverse transcriptase expressed by the transformed hosts of the invention.
  • the invention also relates to a fusion protein comprising a polypeptide having RNA-dependent DNA polymerase activity and substantially no RNase H activity and a second peptide selected from polypeptide proteins which stabilize the fusion protein and hydrophobic leader sequences.
  • the invention also relates to a method of producing reverse transcriptase having DNA polymerase activity and substantially no RNase H activity, comprising culturing transformed hosts of the invention under conditions which produce reverse transcriptase, and isolating the reverse transcriptase so produced.
  • the invention also relates to a method of preparing cDNA from mRNA comprising contacting mRNA with a polypeptide having RNA-dependent DNA polymerase activity and substantially no RNase H activity, and isolating the cDNA so produced.
  • the invention also relates to a kit for the preparation of cDNA from mRNA comprising a carrier being compartmentalized to receive in close confinement therein one or more containers, wherein
  • a first container contains reverse transcriptase having DNA polymerase activity and substantially no RNase H activity
  • a second container contains a buffer and the nucleoside triphosphates
  • a third container contains oligo(dT)primer
  • a fourth container contains control RNA.
  • the invention is related to the discovery that portions of the RT gene can be deleted to give a deletion mutant having DNA polymerase activity but no detectable RNase H activity. This purified mutant RT lacking RNase H activity can be used to effectively synthesize cDNA from mRNA.
  • FIG. 1 This figure depicts the restriction map of plasmid pRT601.
  • the M-MLV RT gene extends from position 1,019 to 3,070.
  • FIG. 2 This figure depicts schematic representation of pRT601 and related plasmids, and the enzymatic activities and predicted structure of the M-MLV RT protein coded by each plasmid.
  • FIG. 3 This figure depicts an SDS-polyacrylamide gel of M-MLV RT.
  • pRTdEcoRV-C RT (A) and pRT601 RT (B) (3 ⁇ g of each) were run on an SDS 10% polyacrylamide gel (Laemmli, U. K., Nature 227:680-685 (1970)). The gel was stained with Coomassie blue. Lane M contained Mr standards.
  • FIG. 4 This figure depicts an autoradiogram of 32 P-labeled cDNA synthesized from 6.2 kb RNA (Materials and Methods) by pRTdEcoRV-C RT (A) or pRT601 RT (B). A 1 kb ladder was used as a standard (C). Electrophoresis was performed on an alkaline 1.4% agarose gel (McDonnel, M. W., et al., J. Mol. Biol. 110:119-146 (1977)).
  • FIG. 5 This figure depicts an autoradiogram of 32 P-labeled 2.3 kb poly(A)-tailed RNA after oligo(dT)-primed cDNA synthesis catalyzed by pRTdEcoRV-C RT or pRT601 RT. Aliquots were removed from reaction mixtures containing no enzyme ( ⁇ E) or 200 units of RT at the times indicated (in min). The minus enzyme control was incubated for 60 min. Samples were electrophoresed as described in Materials and Methods. A 1 kb ladder was used as marker (M).
  • FIG. 6 This figure depicts the DNA sequence which encodes reverse transcriptase having DNA polymerase activity and substantially no RNase H activity. Also shown is the corresponding amino acid sequence.
  • the invention relates to the production of reverse transcriptase having DNA polymerase activity and substantially no RNase H activity, using recombinant DNA techniques.
  • Recombinant plasmids constructed as described herein provide reverse transcriptase for use in recombinant DNA technology to synthesize cDNA from mRNA without the problem associated with RNase H activity which degrades mRNA template during first-strand synthesis.
  • substantially no RNase H activity is intended reverse transcriptase purified to near homogeneity and having an RNase H activity of less than 0.001 pmoles [ 3 H](A) n solubilized per ⁇ g protein with a [ 3 H](A) n .(dT) n substrate in which the [ 3 H](A) n has a specific radioactivity of 2,200 cpm/pmole. RNase H activities of this specific activity or less allows the preparation of cDNA without significant degradation of the mRNA template during first-strand synthesis.
  • the reverse transcriptase gene (or the genetic information contained therein) can be obtained from a number of different sources.
  • the gene may be obtained from eukaryotic cells which are infected with retrovirus, or from a number of plasmids which contain either a portion of or the entire retrovirus genome.
  • messenger RNA-like RNA which contains the RT gene can be obtained from retroviruses.
  • M-MLV Moloney murine leukemia virus
  • HTLV-I human T-cell leukemia virus type I
  • BLV bovine leukemia virus
  • RSV Rous Sarcoma Virus
  • HV human immunodeficiency virus
  • yeast including Saccharomyces, Neurospora, Drosophila; primates; and rodents. See, for example, Weiss et al., U.S. Pat. No. 4,663,290 (1987); Gerard, G. R., DNA 5:271-279 (1986); Kotewicz, M.
  • RT proviral DNA can be isolated using standard isolation techniques.
  • the DNA is cleaved into linear fragments, any one of which may contain the genes which encode RT.
  • Such fragmentation can be achieved using enzymes which digest or cleave DNA, such as restriction enzymes which cleave DNA as specific base sequences.
  • enzymes which digest or cleave DNA such as restriction enzymes which cleave DNA as specific base sequences.
  • Such recombinant DNA techniques may be performed as described by Maniatis, T., et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y. (1982).
  • Identification of the DNA fragment which contains the gene may be accomplished in a number of ways. For example, it is possible to sequence the DNA fragments (Maxam and Gilbert, Methods in Enzymology 64.499 (1980); Messing, J., Meth. in Enz. 101C:20 (1983)) to identify which fragment contains the reverse transcriptase gene. Alternatively, hybridization techniques (Southern, J. Mol. Biol. 98:503 (1975)) using a labeled (e.g., radioactively labeled) DNA probe may be employed.
  • a labeled e.g., radioactively labeled
  • the fractions containing the desired DNA are pooled, ligated into a suitable vector, and used to transform a host. Screening for transformed hosts containing the RT gene may be accomplished by, for example, the method disclosed by Gerard et al., Biochem. 13:1632-1641 (1974) or by Gerard et al., J. Virol. 15:785-797 (1975). Alternatively, clones containing reverse transcriptase may be identified by hybridization with complementary labeled DNA.
  • An alternative to isolating the reverse transcriptase gene from a retroviral proviral DNA is to make cDNA to the mRNA-like RNA which codes for reverse transcriptase.
  • mRNA-like RNA coding for reverse transcriptase is isolated from retrovirus. By standard techniques, the isolated mRNA is then converted into cDNA using reverse transcriptase. The cDNA can then be inserted into a plasmid vector in a conventional manner.
  • a suitable vector depends on a number of considerations known to one of ordinary skill in the art, such as the size of the fragment, nature of the host, number and position of restriction sites desired, and the selection marker and markers desired.
  • Such vectors may include replicon and control sequences from species compatible with a host cell (see Maniatis et al., supra). Expression of the RT genes may also be placed under control of other regulatory sequences homologous or heterologous to the host organism in its untransformed state.
  • lactose-dependent E. coli chromosomal DNA comprises a lactose or lac operon which mediates lactose utilization by elaborating the enzyme ⁇ -galactosidase.
  • the lac control elements may be obtained from bacteriophage lambda plac 5, which is infectious for E. coli .
  • the lac promoter-operator system can be induced by IPTG.
  • promoter-operator systems or portions thereof can be employed as well.
  • galactose alkaline phosphatase, tryptophan, xylose, tac, lambda pL, lambda pR and the like can be used.
  • the vectors may be introduced into an appropriate host.
  • Various techniques may be employed such as protoplast fusion, CaCl 2 , calcium phosphate precipitation, electroporation, or other conventional DNA transfer techniques.
  • the vectors may then be introduced into a second host by similar transfer methods, and also by cell to cell transfer methods such as conjugation.
  • This cell-to-cell transfer may be accomplished using known techniques which depend upon the nature of the transferer bacterium, the recipient bacterium, and the cloning vector used to propagate the RT DNA.
  • the transfer may require the use of a helper plasmid. See, for example, Ditta, G., et al., Proc. Natl. Acad. Sci. (USA) 77:7347-7351 (1980).
  • RT genes having DNA polymerase activity and substantially no RNase H activity may be obtained by deletion of deoxyribonucleotides at the 3′ end of the gene which encode the portion of the polypeptide having RNase H activity. Deletions of the RT gene may be accomplished by cutting the plasmid at selected restriction sites within the RT gene and discarding the excised fragment. Further deletion of consecutive deoxyribonucleotides may be accomplished by treating the fragment with an exonuclease. The DNA ends may then be joined in such a way that the translation reading frame of the gene is maintained. The plasmid thus obtained may then be used to transform hosts which may then be screened for altered RT activity.
  • RT RNase H activity may be assayed according to Gerard et al., J. Virol. 15:785-797 (1975).
  • DNA polymerase activity may be assayed according to Gerard et al., Biochem. 13:1632-1641 (1974).
  • Clones having DNA polymerase activity and substantially no RNase H activity may be used to prepare RT with altered activity.
  • the portion of the RT gene derived from M-MLV which encodes DNA polymerase was localized to about 1495 base pairs (about 1018 to about 2512) as shown in FIG. 6.
  • the protein expressed by this gene has about 503 amino acids (FIG. 6). This protein has DNA polymerase activity and substantially no RNase H activity.
  • the invention also relates to fusion proteins which comprise the reverse transcriptase of the invention.
  • fusion proteins may comprise, for example, a carrier protein which has a leader sequence of hydrophobic amino acids at the amino terminus of the reverse transcriptase.
  • This carrier protein is normally excreted through the membrane of the cell within which it is made. By cleavage of the hydrophobic leader sequence during excretion, a means is provided for producing reverse transcriptase which can be recovered either from the periplasmic space or the medium in which the bacterium is grown.
  • fusion proteins comprising the reverse transcriptase of the invention which is substituted at the amino or carboxy termini with polypeptides which stabilize or change the solubility of the reverse transcriptase.
  • a carboxy-terminal gene fusion which encodes reverse transcriptase and part of the plasmid pBR322 tet gene is taught, for example, by Xotewicz, M., et al., Gene 35:249-258 (1985) and Gerard, G., DNA 5:271-279 (1986).
  • the transformed hosts of the invention may be cultured under protein producing conditions according to any of the methods which are known to those skilled in the art.
  • the reverse transcriptase having DNA polymerase activity and substantially no RNase activity may be isolated according to conventional methods known to those skilled in the art.
  • the cells may be collected by centrifugation, washed with suitable buffers, lysed, and the reverse transcriptase isolated by column chromatography, for example, on DEAE-cellulose, phosphocellulose (see Kotewicz et al., Gene 35:249-258 (1985)) or other standard isolation and identification techniques using, for example, polyribocytidylic acid-agarose, or hydroxylapatite or by electrophoresis or immunoprecipitation.
  • the reverse transcriptase so produced may be used to prepare cDNA from RNA by, for example, hybridizing an oligo(dT) primer or other complementary primers with the mRNA.
  • the synthesis of a complete cDNA may be accomplished by adding the reverse transcriptase and all four deoxynucleoside triphosphates.
  • Using the reverse transcriptase produced by the present invention allows for the preparation of cDNA from mRNA without concomitant degradation of the mRNA which results in incomplete cDNA synthesis.
  • RNA-DNA hybrid may be treated, for example, with alkali or RNase H to selectively hydrolyze the RNA to leave cDNA which may be converted to double-stranded form in a second DNA reaction catalyzed by reverse transcriptase or other DNA polymerase.
  • alkali or RNase H to selectively hydrolyze the RNA to leave cDNA which may be converted to double-stranded form in a second DNA reaction catalyzed by reverse transcriptase or other DNA polymerase.
  • the reverse transcriptase of the invention is ideally suited for incorporation into a kit for the preparation of cDNA from RNA.
  • a kit may comprise a carrier means being compartmentalized to receive a close confinement therein, one or more container means, such as vials, tubes, and the like, each of said container means comprising one of the separate elements of the method used to prepare cDNA from RNA.
  • container means containing reverse transcriptase having DNA polymerase activity and substantially no RNase H activity, in solution.
  • Further container means may contain suitable buffers, substrates for DNA synthesis such as the deoxynucleoside triphosphate, oligo(dT) primer, and control RNA for use as a standard.
  • the reverse transcriptase may be present in the solution at a concentration of 200 units/ml to 400 units/ml.
  • the deoxynucleoside triphosphases may be present either in lyophilized form or as part of a buffer at a concentration of 0.5 mM to 2 mM.
  • a suitable buffer present at 5 times the final concentration of use, includes 250 mM Tris-HCl (pH 7.5 to 8.3), 375 mM KCl, 15 mM MgCl 2 , and 50 mM dithiothreitol.
  • the oligo (dT) may be present at a concentration of 5 ⁇ g/ml to 20 ⁇ g/ml.
  • Control RNA such as 2.3 kb control RNA, may be present at a concentration of 10 ⁇ g/ml to 20 ⁇ g/ml.
  • RT For deletion analysis of RT, a clone of M-MLV RT was constructed to overproduce stable RT in Escherichia coli , pRT601 (FIG. 1). Gerard, G. F., et al., DNA 5:271-279 (1986). It is a pBR322 replicon containing the strong lambda leftward promoter, pL, and the ribosome binding site of the lambda cII gene.
  • RT DNA polymerase activity in extracts was assayed specifically by using poly(2′-O-methylcytidylate).oligodeoxyguanylate [(Cm) n .(dG) 12-18 ] (Gerard, G. F., et al., DNA 5:271-279 (1986), eliminating interference from cellular DNA polymerases.
  • activity was assayed with (A) n .(dT) 12-18 (Houts, G. E., et al., J. Virol. 29:517-522 (1979) as described by Gerard, G. F., et al., DNA 5:271-279 (1986).
  • One unit of DNA polymerase activity is the amount of enzyme that incorporates one nmole of deoxynucleoside monophosphate into acid insoluble product at 37° C. in 10 min.
  • RNase H activity is the amount of enzyme required to solubilize one mole of [ 3 H](A) n in [ 3 H](A) n .(dT) n in 10 min at 37° C.
  • Synthetic 2.3 kb and 6.2 kb RNAs containing a 19 nucleotide poly(A) tail at the 3′ end were synthesized with T7 RNA polymerase from Xba I-cut pJD2.3 and Hind III-cut pHL3X, respectively.
  • Reaction mixtures contained 40 uM Tris-HCl (pH 8.0), 8 mM MgCl 2 , 2 mM spermidine-HCl, 5 mM dithiothreitol, 0.4 mM each of CTP, UTP, GTP, and ATP, 20 ⁇ g/ml DNA, and 2,000 units/ml T7 RNA polynerase.
  • RNA Uniformly labeled RNA was synthesized with all four [ ⁇ - 32 P]NTPs, each at 0.4 mM and 250 cpm/pmole. After 1 hr incubation at 37° C., the RNA product was phenol extracted, ethanol precipitated, and purified by oligo(dT)-cellulose chromatography to ensure the presence of a poly(A) tail.
  • reaction mixtures contained 50 mM Tris-HCl (pH 8.3), 75 mM KCl, 3 mM MgCl 2 , 10 mM dithiothreitol, 0.5 mM each dATP, dGTP, and dTTP, 0.5 mM [ 3 H]dCTP (200 cpm/pmol), 50 ⁇ g/ml (dT) 12-18 , 20 ⁇ g/ml 2.3 kb [ 32 P]labeled RNA, and 4,000 units/ml RT. The reactions were incubated at 37° C.
  • reaction mixtures (10 ⁇ l) contained the same buffer and salts, 0.5 mM each of dATP, dGTP, dTTP, and [ ⁇ - 32 P]dCTP (600 cpm/pmole), 50 ⁇ g/ml actinomycin D, 50 ⁇ g/ml (dT) 12-18 , 100 ⁇ g/ml 6.2 kb poly(A)-tailed RNA, and 20,000 units/ml RT.
  • the (NH 4 ) 2 SO 4 pellet was dissolved in 50 ml of 20 mM Tris-HCl (pH 7.5), 1 mM EDTA, 0.1 M NaCl, 5% glycerol, 1 mM, dithiothreitol, and 0.01% n-octyl- ⁇ -D-glucopyranoside, the suspension was clarified by centrifugation at 10,000 ⁇ g for 10 min, and the supernatant was desalted on a 320 ml (5 ⁇ 16 cm) Sephadex G-25 column run in buffer A (20 mM Tris-HCl, pH 7.5, 1 mM dithiothreitol, 1 mM EDTA, 5% glycerol, 0.01% NP-40) plus 0.1 M NaCl.
  • the RT peak was pooled, diluted with an equal volume of buffer A, and chromatographed on a 21-ml heparin-agarose column (1.5 ⁇ 12 cm) equilibrated in buffer A plus 0.1 M NaCl.
  • the RT peak from the heparin-agarose column was chromatographed on a Mono-S HR 5/5 column equilibrated in buffer A (Gerard, G. F., supra).
  • Deletions of the M-MLV RT gene were constructed by cutting pRT601 (FIG. 1) at selected restriction sites within the RT gene, discarding the excised fragment, and joining the DNA ends in such a way that the translation reading frame of the gene was maintained.
  • pRTdBam-Bam was constructed by deleting the Bam HI fragment between nucleotide positions 1,654 and 1,960 (FIGS. 1 and 2). Ligation of the Bam HI half sites at positions 1,654 and 1,960 maintained the translation reading frame across the site.
  • a deletion at the carboxy terminus of M-MLV RT was constructed by deleting all of the 3′ end of the gene downstream of the Eco RV site at position 2,513 (FIGS. 1 and 2).
  • pRTdEcoRV-C a Sca I (position 6,238) to Eco RV (position 2,513) fragment of pRT601 containing the 5′ portion of the RT gene was ligated to a Sca I-Eco RI fragment derived from plasmid pBRT (Gerard, G. F., supra).
  • the 3,211 base pair pBRT Sca I-Eco RI fragment contained the pBR322 origin of replication and a universal translation terminator sequence just inside the Eco RI site.
  • the Eco RI site was repaired with DNA polymerase I Klenow fragment before ligation.
  • pRT603 was constructed as described (Gerard, G. R., supra) which encodes an RT that contains 73 fewer amino acids than pRT601, all deleted from the carboxy terminus (FIG. 2).
  • pRTdEcoRV-C RT was purified and compared to RT encoded by pRT601.
  • M-MLV reverse transcriptase encoded by pRTdEcoRV-C, pRT601, and pRT603 were purified as described in Materials and Methods.
  • a summary of the purification of pRTdEcoRV-C RT is presented in Table 3.
  • Three column steps produced a nearly homogeneous mutant enzyme with the same DNA polymerase specific activity as pRT601 RT with the template-primer (Cm) n .(dG) 12-18 (Table 2). With (A) n .(dT) 12-18 , the mutant enzyme had one-fourth the DNA polymerase activity of pRT601 RT (Table 2).
  • RNase H activity of purified pRTdEcoRV-C RT was undetectable using [ 3 H](A) n .(dT) n as the substrate. Most RNase H activity in extracts was eliminated from mutant RT by precipitation of the enzyme with 40% (NH 4 ) 2 SO 4 (Table 3). Under these conditions, DNA polymerase I remains soluble (Richardson, C., et al., J. Biol. Chem. 239:222-230 (1964)), as does most of the RNase H activity in the extract.
  • pRTdEcoRV-C RT purified through the Mono-S column was greater than 90% pure and had a molecular weight of 56,000 (FIG. 3), consistent with the molecular weight (57,000) predicted by the DNA sequence.
  • FIG. 4 shows that pRTdEcoRV-C RT catalyzed the synthesis of full-length cDNA from 6.2 kb RNA more efficiently than pRT601 RT.
  • the amount of cDNA synthesized from 1 ⁇ g of RNA was 0.28 ⁇ g (34% full-length) and 0.24 ⁇ g (24% full-length) with pRTdEcoRV-C RT and pRT601 RT, respectively.
  • FIG. 5 shows that with pRT601 RT, the full-length 2.3 kb RNA template was degraded totally after 5 min of synthesis. In contrast, with pRTdEcoRV-C RT the RNA was intact even after 60 min. The amount of cDNA synthesized after 60 min from 1 ⁇ g of RNA was 0.67 and 0.76 ⁇ g with pRT601 and pRTdEcoRV-C RT, respectively. When 10 units of E.
  • coli RNase H were added to the pRTdEcoRV-C RT reaction after 60 min of incubation, all of the RNA was degraded, confirming the hybrid state of the RNA.
  • 15 ⁇ g (1,200 units) of pRTdEcoRV-C RT solubilized no radioactivity from a [ 3 H](A) n .(dT) n substrate in which the [ 3 H](A) n had a specific activity of 2,200 cpm/pmole (Materials and Methods).
  • pRT603 RT has normal levels of RNase H activity. These missing, homologous residues apparently are not required for catalysis, and might serve a nucleic acid binding or structural role. Consistent with the latter, a single amino acid change at a position 12 residues from the carboxy end of E. coli RNase H produces a 10-fold reduction in RNase H specific activity (Kanaya, S. et al., J. Bacteriol. 154:1021-1026 (1983)). This reduction appears to be the result of altered protein conformation (Kanaya, S. et al., supra).
  • RT polymerase and nuclease active sites reside on separate structural domains, it should be possible theoretically to isolate two separate protein fragments, each with a single activity.
  • a 24K to 30K proteolytic fragment of RT possessing only RNase H activity has been isolated (Lai, M. H. T. et al., J. Virol. 25:652-663 (1978); Gerard, G. F., J. Virol. 26:16-28 (1978); Gerard, G. F., J. Virol. 37:748-754 (1981)), but unfortunately, the location of this RNase H fragment in the parent RT polypeptide has not been established, and no analogous DNA polymerase containing fragment has ever been found.

Abstract

The invention relates to a gene which encodes reverse transcriptase having DNA polymerase activity and substantially no RNase H activity. The invention also relates to vectors containing the gene and hosts transformed with the vectors of the invention. The invention also relates to a method of producing reverse transcriptase having DNA polymerase activity and substantially no RNase H activity by expressing the reverse transcriptase genes of the present invention in a host. The invention also relates to a method of producing cDNA from mRNA using the reverse transcriptase of the invention. The invention also relates to a kit for the preparation of cDNA from mRNA comprising the reverse transcriptase of the invention.

Description

    FIELD OF THE INVENTION
  • The invention is in the field of recombinant genetics. [0001]
  • BACKGROUND OF THE INVENTION
  • Both viral and cloned reverse transcriptase (RT) contain at least two enzymatic activities, DNA polymerase and ribonuclease H (RNase H), that reside on a single polypeptide. Grandgenett, D. P. et al., [0002] Proc. Natl. Acad. Sci. (USA) 70:230-234 (1973); Moelling, K., Virology 62:46-59 (1974); Kotewicz, M. L., et al., Gene 35:249-258 (1985); and Roth, M. J., et al., J. Biol. Chem. 260:9326-9335 (1985). Little is known about the structure-functional relationship of these two activities, but such knowledge would be important both in understanding retroviral replication and in exploiting the enzyme as a recombinant DNA tool.
  • In the retrovirus life cycle, the RT DNA polymerase activity is responsible for transcribing viral RNA into double-stranded DNA. Varmus, H. (1982), in Weiss, R., et al. (eds.), [0003] RNA Tumor Viruses, Cold Spring Harbor Laboratory, pp. 410-423. The function of RNase H in replication is less clear, but it is thought to degrade genomic RNA during DNA synthesis to generate oligomeric RNA primers for plus-strand DNA synthesis, and to remove the RNA primers of both minus- and plus-strand DNA. Omer, C. A., et al., Cell 30:797-805 (1982); Resnick, R., et al., J. Virol. 51:813-821 (1984); Varmus, H. (1985), in Weiss, R., et al. (eds.), RNA Tumor Viruses, Cold Spring Harbor Laboratory, pp. 79-80.
  • The temporal relationship in vivo between DNA polymerization and RNA hydrolysis is not well defined. Furthermore, precisely how the two enzymatic activities are coordinated is not clear. Conditional mutations restricted to either DNA polymerase or RNase H would be invaluable in deciphering the events of retroviral |replication. Unfortunately, conditional viral mutations in the RT gene invariably affect both activities. Lai, M. H. T, et al., [0004] J. Virol. 27:823-825 (1978); Moelling, K., et al., J. Virol. 32:370-378 (1979).
  • RT is used extensively in recombinant DNA technology to synthesize cDNA from mRNA. One major problem with cDNA synthesis is that the RNase H activity of RT degrades the mRNA template during first-strand synthesis. The mRNA poly(A)-oligo(dT) hybrid used as a primer for first-strand cDNA synthesis is degraded by RT RNase H. Thus, at the outset of cDNA synthesis, a competition is established between RNase H-mediated deadenylation of mRNA and initiation of DNA synthesis, which reduces the yield of cDNA product. Berger, S. L., et al., [0005] Biochem. 22:2365-2373 (1983). Furthermore, in some cases, the RNase H causes premature termination of DNA chain growth. Unfortunately, these events eliminate the potential for repeated copying of the RNA template.
  • Efforts to selectively inactivate RT RNase H with site-specific inhibitors have been unsuccessful (for review, see Gerard, G. F. (1983), in Jacob, S. T., (ed.), [0006] Enzymes of Nucleic Acid Synthesis and Modification, Vol. I, DNA Enzymes, CRC Press, Inc., Boca Raton, Fla., pp. 1-38). Attempts to physically separate the active centers of RT polymerase and RNase H activity by proteolysis have yielded a proteolytic fragment possessing only RNase H activity (Lai, M. H. T., et al., J. Virol. 25:652-663 (1978); Gerard, G. F., J. Virol. 26:16-28 (1978); and Gerard, G. F., J. Virol. 37:748-754 (1981)), but no corresponding fragment containing only polymerase activity has been isolated.
  • Computer analysis of the amino acid sequences from the putative gene products of retroviral pol genes has revealed a 150-residue segment at the carboxyl terminus that is homologous with the ribonuclease H of [0007] E. coli and a section close to the amino terminus which can be aligned with nonretroviral polymerases. Johnson, M. S., et al., Proc. Natl. Acad. Sci. (USA) 83:7648-7652 (1986). Based on these related amino acid sequences, Johnson et al. suggest that ribonuclease H activity should be situated at the carboxyl terminus, and the DNA polymerase activity at the amino terminus.
  • There have been a number of reports concerning the cloning of genes which encode RT and their expression in hosts. Weiss et al., U.S. Pat. No. 4,663,290 (1987); Gerard, G. F., [0008] DNA 5:271-279 (1986); Kotewicz, M. L., et al., Gene 35:249-258 (1985); Tanese, N., et al., Proc. Natl. Acad. Sci. (USA) 82:4944-4948 (1985); and Roth, M. J., et al., J. Biol. Chem. 260:9326-9335 (1985).
  • There has been no direct scientific evidence that amino acid residues involved catalytically or structurally in the RNase H activity of reverse transcriptase could be altered to eliminate RNase H activity without affecting the RNA-dependant DNA polymerase activity of reverse transcriptase. Moreover, there has been no report of the cloning of RT to give a gene product without RNase activity. [0009]
  • SUMMARY OF THE INVENTION
  • The invention relates to a gene which encodes reverse transcriptase having DNA polynerase activity and substantially no RNase H activity. [0010]
  • The invention also relates to a reverse transcriptase gene comprising the following DNA sequence: [0011]
                                                                               1078
    A{dot over (T)}G ACC CTA AA{dot over (T)} ATA GAA GAT GAG CAT CGG C{dot over (T)}A CAT GAG AC{dot over (C)} TCA AAA GAG {dot over (C)}CA GAT GTT
    MET Thr Leu Asn Ile Glu Asp Glu His Arg Leu His Glu Thr Ser Lys Glu Pro Asp Val
                                                                               1138
    TCT CTA GGG TCC ACA TGG CTG TCT GAT TTT CCT GAG GCC TGG GCG GAA ACC GGG GGC ATG
    Ser Leu Gly Ser Thr Trp Leu Ser Asp Phe Pro Gln Ala Trp Ala Glu Thr Gly Gly MET
                                                                               1198
    GGA CTG GCA GTT CGC CAA GCT CCT CTG ATC ATA CCT CTG AAA GCA ACC TCT ACC CCC GTG
    Gly Leu Ala Val Arg Gln Ala Pro Leu Ile Ile Pro Leu Lys Ala Thr Ser Thr Pro Val
                                                                               1258
    TCC ATA AAA CAA TAC CCC ATG TCA CAA GAA GCC AGA CTG GGG ATC AAG CCC CAC ATA CAG
    Ser Ile Lys Gln Tyr Pro MET ser Gln Glu Ala Arg Leu Gly Ile Lys Pro His Ile Gln
                                                                               1318
    AGA CTG TTG GAC CAG GGA ATA CTG GTA CCC TGC CAG TCC CCC TGG AAC ACG CCC CTG CTA
    Arg Leu Leu Asp Gln Gly Ile Leu Val Pro Cys Gln Ser Pro Trp Asn Thr Pro Leu Leu
                                                                               1378
    CCC GTT AAG AAA CCA GGG ACT AAT GAT TAT AGG CCT GTC CAG GAT CTG AGA GAA GTC AAC
    Pro Val Lys Lys Pro Gly Thr Asn Asp Tyr Arg Pro Val Gln Asp Leu Arg Glu Val Asn
                                                                               1438
    AAG CGG GTG GAA GAC ATC CAC CCC ACC GTG CCC AAC CCT TAC AAC CTC TTG AGC GGG CTC
    Lys Arg Val Glu Asp Ile His Pro Thr Val Pro Asn Pro Tyr Asn Leu Leu Ser Gly Leu
                                                                               1498
    CCA CCG TCC CAC CAG TGG TAC ACT GTG CTT GAT TTA AAG GAT GCC TTT TTC TGC CTG AGA
    Pro Pro Ser His Gln Trp Tyr Thr Val Leu Asp Leu Lys Asp Ala Phe Phe Cys Leu Arg
                                                                               1558
    CTC CAC CCC ACC AGT CAG CCT CTC TTC GCC TTT GAG TGG AGA GAT CCA GAG ATG GGA ATC
    Leu His Pro Thr Ser Gln Pro Leu Phe Ala Phe Glu Trp Arg Asp Pro Glu MET Gly Ile
                                                                               1618
    TCA GGA CAA TTG ACC TGG ACC AGA CTC CCA CAG GGT TTC AAA AAC AGT CCC ACC CTG TTT
    Ser Gly Gln Leu Thr Trp Thr Arg Leu Pro Gln Gly Phe Lys Asn Ser Pro Thr Leu Phe
                                                                               1678
    GAT GAG GCA CTG CAC AGA GAC CTA GCA GAC TTC CGG ATC CAG CAC CCA GAC TTG ATC CTG
    Asp Glu Ala Leu His Arg Asp Leu Ala Asp Phe Arg Ile Gln His Pro Asp Leu Ile Leu
                                                                               1738
    CTA CAG TAC GTG GAT GAC TTA CTG CTG GCC GCC ACT TCT GAG CTA GAC TGC CAA CAA GGT
    Leu Gln Tyr Val Asp Asp Leu Leu Leu Ala Ala Thr Ser Glu Leu Asp Cys Gln Gln Gly
                                                                               1798
    ACT CGG GCC CTG TTA CAA ACC CTA GGG AAC CTC GGG TAT CGG GCC TCG GCC AAG AAA GCC
    Thr Arg Ala Leu Leu Gln Thr Leu Gly Asn Leu Gly Tyr Arg Ala Ser Ala Lys Lys Ala
                                                                               1858
    CAA ATT TGC CAG AAA CAG GTC AAG TAT CTG GGG TAT CTT CTA AAA GAG GGT CAG AGA TGG
    Gln Ile Cys Gln Lys Gln Val Lys Tyr Leu Gly Tyr Leu Leu Lys Glu Gly Gln Arg Trp
                                                                               1918
    CTG ACT GAG GCC AGA AAA GAG ACT GTG ATG GGG CAG CCT ACT CCG AAG ACC CCT CGA CAA
    Leu Thr Glu Ala Arg Lys Glu Thr Val MET Gly Gln Pro Thr Pro Lys Thr Pro Arg Gln
                                                                               1978
    CTA AGG GAG TTC CTA GGG ACG GCA GGC TTC TGT CGC CTC TGG ATC CCT GGG TTT GCA GAA
    Leu Arg Glu Phe Leu Gly Thr Ala Gly Phe Cys Arg Leu Trp Ile Pro Gly Phe Ala Glu
                                                                               2038
    ATG GCA GCC CCC TTG TAC CCT CTC ACC AAA ACG GGG ACT CTG TTT AAT TGG GGC CCA GAC
    MET Ala Ala Pro Leu Tyr Pro Leu Thr Lys Thr Gly Thr Leu Phe Asn Trp Gly Pro Asp
                                                                               2098
    CAA CAA AAG GCC TAT CAA GAA ATC AAG CAA GCT CTT CTA ACT GCC CCA GCC CTG GGG TTG
    Gln Gln Lys Ala Tyr Gln Glu Ile Lys Gln Ala Leu Leu Thr Ala Pro Ala Leu Gly Leu
                                                                               2158
    CCA GAT TTG ACT AAG CCC TTT GAA CTC TTT GTC GAC GAG AAG CAG GGC TAC GCC AAA GGT
    Pro Asp Leu Thr Lys Pro Phe Glu Leu Phe Val Asp Glu Lys Gln Gly Tyr Ala Lys Gly
                                                                               2218
    GTC CTA ACG CAA AAA CTG GGA CCT TGG CGT CGG CCG GTG GCC TAC CTG TCC AAA AAG CTA
    Val Leu Thr Gln Lys Leu Gly Pro Trp Arg Arg Pro Val Ala Tyr Leu Ser Lys Lys Leu
                                                                               2278
    GAC CCA GTA GCA GCT GGG TGG CCC CCT TGC CTA CGG ATG GTA GCA GCC ATT GCC GTA CTG
    Asp Pro Val Ala Ala Gly Trp Pro Pro Cys Leu Arg MET Val Ala Ala Ile Ala Val Leu
                                                                               2338
    ACA AAG GAT GCA GGC AAG CTA ACC ATG GGA CAG CCA CTA GTC ATT CIG GCC CCC CAT GCA
    Thr Lys Asp Ala Gly Lys Leu Thr MET Gly Gln Pro Leu Val Ile Leu Ala Pro His Ala
                                                                               2398
    GTA GAG GCA CTA GTC AAA CAA CCC CCC GAC CGC TGG CTT TCC AAC GCC CGG ATG ACT CAC
    Val Glu Ala Leu Val Lys Gln Pro Pro Asp Arg Trp Leu Ser Asn Ala Arg MET Thr His
                                                                               2458
    TAT CAG CGG TTG CTT TTG GAC ACG GAC CGG CTG CAG TTC GGA CCG GTG GTA GCC CTG AAC
    Tyr Gln Ala Leu Leu Leu Asp Thr Asp Arg Val Gln Phe Gly Pro Val Val Ala Leu Asn
                                                                       2512
    CCG GCT ACG CTG CTC CCA CTG CCT GAG GAA GGG CTG CAA CAC AAC TGC CTT GAT
    Pro Ala Thr Leu Leu Pro Leu Pro Glu Glu Gly Leu Gln His Asn Cys Leu Asp
  • or the degenerate variants thereof. [0012]
  • The invention also relates to a reverse transcriptase gene comprising the following DNA sequence: [0013]
                                                                               1078
    A{dot over (T)}G ACC CTA AA{dot over (T)} ATA GAA GAT GAG CAT CGG C{dot over (T)}A CAT GAG AC{dot over (C)} TCA AAA GAG {dot over (C)}CA GAT GTT
    MET Thr Leu Asn Ile Glu Asp Glu His Arg Leu His Glu Thr Ser Lys Glu Pro Asp Val
                                                                               1138
    TCT CTA GGG TCC ACA TGG CTG TCT GAT TTT CCT CAG GCC TGG GCG GAA ACC GGG GGC ATG
    Ser Leu Gly Ser Thr Trp Leu Ser Asp Phe Pro Gln Ala Trp Ala Glu Thr Gly Gly MET
                                                                               1198
    GGA CTG GCA GTT CGC CAA GCT CCT CTG ATC ATA CCT CTG AAA GCA ACC TCT ACC CCC GTG
    Gly Leu Ala Val Arg Gln Ala Pro Leu Ile Ile Pro Leu Lys Ala Thr Ser Thr Pro Val
                                                                               1258
    TCC ATA AAA CAA TAC CCC ATG TCA CAA GAA GCC AGA CTG GGG ATC AAG CCC CAC ATA CAG
    Ser Ile Lys Gln Tyr Pro MET ser Gln Glu Ala Arg Leu Gly Ile Lys Pro His Ile Gln
                                                                               1318
    AGA CTG TTG GAC CAG GGA ATA CTG GTA CCC TGC CAG TCC CCC TGG AAC ACG CCC CTG CTA
    Arg Leu Leu Asp Gln Gly Ile Leu Val Pro Cys Gln Ser Pro Trp Asn Thr Pro Leu Leu
                                                                               1378
    CCC GTT AAG AAA CCA GGG ACT AAT GAT TAT AGG CCT GTC CAG GAT CTG AGA GAA CTC AAC
    Pro Val Lys Lys Pro Gly Thr Asn Asp Tyr Arg Pro Val Gln Asp Leu Arg Glu Val Asn
                                                                               1438
    AAG CGG GTG GAA GAC ATC CAC CCC ACC GIG CCC AAC CCT TAC AAC CTC TTG AGC GGG CTC
    Lys Arg Val Glu Asp Ile His Pro Thr Val Pro Asn Pro Tyr Asn Leu Leu Ser Gly Leu
                                                                               1498
    CCA CCG TCC CAC CAG TGG TAC ACT GTG CTT GAT TTA AAG GAT GCC TTT TTC TGC CTG AGA
    Pro Pro Ser His Gln Trp Tyr Thr Val Leu Asp Leu Lys Asp Ala Phe Phe Cys Leu Arg
                                                                               1558
    CTC CAC CCC ACC AGT CAG CCT CTC TTC GCC TTT GAG TGG AGA GAT CCA GAG ATG GGA ATC
    Leu His Pro Thr Ser Gln Pro Leu Phe Ala Phe Glu Trp Arg Asp Pro Glu MET Gly Ile
                                                                               1618
    TCA GGA CAA TTG ACC TGG ACC AGA CTC CCA CAG GGT TTC AAA AAC AGT CCC ACC CTG TTT
    Ser Gly Gln Leu Thr Trp Thr Arg Leu Pro Gln Gly Phe Lys Asn Ser Pro Thr Leu Phe
                                                                               1678
    GAT GAG GCA CTG CAC AGA GAC CTA GCA GAC TTC CGG ATC CAG CAC CCA GAC TTG ATC CTG
    Asp Glu Ala Leu His Arg Asp Leu Ala Asp Phe Arg Ile Gln His Pro Asp Leu Ile Leu
                                                                               1738
    CTA CAG TAC GTG GAT GAC TTA CTG CTG GCC GCC ACT TCT GAG CTA GAC TGC CAA CAA GGT
    Leu Gln Tyr Val Asp Asp Leu Leu Leu Ala Ala Thr Ser Glu Leu Asp Cys Gln Gln Gly
                                                                               1798
    ACT CGG GCC CTG TTA CAA ACC CTA GGG AAC CTC GGG TAT CGG GCC TCG GCC AAG AAA GCC
    Thr Arg Ala Leu Leu Gln Thr Leu Gly Asn Leu Gly Tyr Arg Ala Ser Ala Lys Lys Ala
                                                                               1858
    CAA ATT TGC CAG AAA CAG GTC AAG TAT CTG GGG TAT CTT CTA AAA GAG GGT CAG AGA TGG
    Gln Ile Cys Gln Lys Gln Val Lys Tyr Leu Gly Tyr Leu Leu Lys Glu Gly Gln Arg Trp
                                                                               1918
    CTG ACT GAG GCC AGA AAA GAG ACT GTG ATG GGG CAG CCT ACT CCG AAG ACC CCT CGA CAA
    Leu Thr Glu Ala Arg Lys Glu Thr Val MET Gly Gln Pro Thr Pro Lys Thr Pro Arg Gln
                                                                               1978
    CTA AGG GAG TTC CTA GGG ACG GCA GGC TTC TGT CGC CTC TGG ATC CCT GGG TTT GCA GAA
    Leu Arg Glu Phe Leu Gly Thr Ala Gly Phe Cys Arg Leu Trp Ile Pro Gly Phe Ala Glu
                                                                               2038
    ATG GCA GCC CCC TTG TAC CCT CTC ACC AAA ACG GG ACT CTG TTT AAT TGG GGC CCA GAC
    MET Ala Ala Pro Leu Tyr Pro Leu Thr Lys Thr Gly Thr Leu Phe Asn Trp Gly Pro Asp
                                                                               
    CAA CAA AAG GCC TAT CAA GAA ATC AAG CAA GCT CTT CTA ACT GCC CCA GCC CTG GGG TTG
    Gln Gln Lys Ala Tyr Gln Glu Ile Lys Gln Ala Leu Leu Thr Ala Pro Ala Leu Gly Leu
                                                                               2158
    CCA GAT TTG ACT AAG CCC TTT GAA CTC TTT GTC GAC GAG AAG CAG GGC TAC GCC AAA GGT
    Pro Asp Leu Thr Lys Pro Phe Glu Leu Phe Val Asp Glu Lys Gln Gly Tyr Ala Lys Gly
                                                                               2218
    GTC CTA ACG CAA AAA CTG GGA CCT TGG CGT CGG CCG GTG GCC TAC CTG TCC AAA AAG CTA
    Val Leu Thr Gln Lys Leu Gly Pro Trp Arg Arg Pro Val Ala Tyr Leu Ser Lys Lys Leu
                                                                               2278
    GAC CCA GTA GCA GCT GGG TGG CCC CCT TGC CTA CGG ATG GTA GCA GCC ATT GCC GTA CTG
    Asp Pro Val Ala Ala Gly Trp Pro Pro Cys Leu Arg MET Val Ala Ala Ile Ala Val Leu
                                                                               2338
    ACA AAG GAT GCA GGC AAG CTA ACC ATG GGA CAG CCA CTA GTC ATT CTG GCC CCC CAT GCA
    Thr Lys Asp Ala Gly Lys Leu Thr MET Gly Gln Pro Leu Val Ile Leu Ala Pro His Ala
                                                                               2398
    GTA GAG GCA CTA GTC AAA CAA CCC CCC GAC CGC TGG CTT TCC AAC GCC CGG ATG ACT CAC
    Val Glu Ala Leu Al Lys Gln Pro Pro Asp Arg Trp Leu Ser Asn Ala Arg MET Thr His
                                                                               2458
    TAT CAG GCC TTG CTT TTG GAC ACG GAC CGG GTC CAG TTC GGA CCG GTG GTA GCC CTG AAC
    Tyr Gln Ala Leu Leu Leu Asp Thr Asp Arg Val Gln Phe Gly Pro Val Val Ala Leu Asn
                                                                               2518
    CCG GCT ACG CTG CTC CCA CTG CCT GAG GAA GGG CTG CAA CAC AAC TGC CTT GAT AAT TCC
    Pro Ala Thr Leu Leu Pro Leu Pro Glu Glu Gly Leu Gln His Asn Cys Leu Asp Asn Ser
               2530
    CGC TTA ATT AAT
    Arg Leu Ile Asn
  • or the degenerate variants thereof. [0014]
  • The invention also relates to the vectors containing the gene of the invention, hosts transformed with the vectors of the invention, and the reverse transcriptase expressed by the transformed hosts of the invention. [0015]
  • The invention also relates to a fusion protein comprising a polypeptide having RNA-dependent DNA polymerase activity and substantially no RNase H activity and a second peptide selected from polypeptide proteins which stabilize the fusion protein and hydrophobic leader sequences. [0016]
  • The invention also relates to a method of producing reverse transcriptase having DNA polymerase activity and substantially no RNase H activity, comprising culturing transformed hosts of the invention under conditions which produce reverse transcriptase, and isolating the reverse transcriptase so produced. [0017]
  • The invention also relates to a method of preparing cDNA from mRNA comprising contacting mRNA with a polypeptide having RNA-dependent DNA polymerase activity and substantially no RNase H activity, and isolating the cDNA so produced. [0018]
  • The invention also relates to a kit for the preparation of cDNA from mRNA comprising a carrier being compartmentalized to receive in close confinement therein one or more containers, wherein [0019]
  • (a) a first container contains reverse transcriptase having DNA polymerase activity and substantially no RNase H activity; [0020]
  • (b) a second container contains a buffer and the nucleoside triphosphates; [0021]
  • (c) a third container contains oligo(dT)primer; and [0022]
  • (d) a fourth container contains control RNA. [0023]
  • The invention is related to the discovery that portions of the RT gene can be deleted to give a deletion mutant having DNA polymerase activity but no detectable RNase H activity. This purified mutant RT lacking RNase H activity can be used to effectively synthesize cDNA from mRNA. [0024]
  • DESCRIPTION OF THE FIGURES
  • FIG. 1. This figure depicts the restriction map of plasmid pRT601. The M-MLV RT gene extends from position 1,019 to 3,070. [0025]
  • FIG. 2. This figure depicts schematic representation of pRT601 and related plasmids, and the enzymatic activities and predicted structure of the M-MLV RT protein coded by each plasmid. [0026]
  • FIG. 3. This figure depicts an SDS-polyacrylamide gel of M-MLV RT. pRTdEcoRV-C RT (A) and pRT601 RT (B) (3 μg of each) were run on an [0027] SDS 10% polyacrylamide gel (Laemmli, U. K., Nature 227:680-685 (1970)). The gel was stained with Coomassie blue. Lane M contained Mr standards.
  • FIG. 4. This figure depicts an autoradiogram of [0028] 32P-labeled cDNA synthesized from 6.2 kb RNA (Materials and Methods) by pRTdEcoRV-C RT (A) or pRT601 RT (B). A 1 kb ladder was used as a standard (C). Electrophoresis was performed on an alkaline 1.4% agarose gel (McDonnel, M. W., et al., J. Mol. Biol. 110:119-146 (1977)).
  • FIG. 5. This figure depicts an autoradiogram of [0029] 32P-labeled 2.3 kb poly(A)-tailed RNA after oligo(dT)-primed cDNA synthesis catalyzed by pRTdEcoRV-C RT or pRT601 RT. Aliquots were removed from reaction mixtures containing no enzyme (−E) or 200 units of RT at the times indicated (in min). The minus enzyme control was incubated for 60 min. Samples were electrophoresed as described in Materials and Methods. A 1 kb ladder was used as marker (M).
  • FIG. 6. This figure depicts the DNA sequence which encodes reverse transcriptase having DNA polymerase activity and substantially no RNase H activity. Also shown is the corresponding amino acid sequence. [0030]
  • DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • The invention relates to the production of reverse transcriptase having DNA polymerase activity and substantially no RNase H activity, using recombinant DNA techniques. [0031]
  • Recombinant plasmids constructed as described herein provide reverse transcriptase for use in recombinant DNA technology to synthesize cDNA from mRNA without the problem associated with RNase H activity which degrades mRNA template during first-strand synthesis. [0032]
  • By the terms “substantially no RNase H activity” is intended reverse transcriptase purified to near homogeneity and having an RNase H activity of less than 0.001 pmoles [[0033] 3H](A)n solubilized per μg protein with a [3H](A)n.(dT)n substrate in which the [3H](A)n has a specific radioactivity of 2,200 cpm/pmole. RNase H activities of this specific activity or less allows the preparation of cDNA without significant degradation of the mRNA template during first-strand synthesis.
  • By the terms “degenerate variants” is intended cloned genes having variations of DNA sequence, but which encode the same amino acid sequence. [0034]
  • The reverse transcriptase gene (or the genetic information contained therein) can be obtained from a number of different sources. For instance, the gene may be obtained from eukaryotic cells which are infected with retrovirus, or from a number of plasmids which contain either a portion of or the entire retrovirus genome. In addition, messenger RNA-like RNA which contains the RT gene can be obtained from retroviruses. Examples of sources for RT include, but are not limited to, Moloney murine leukemia virus (M-MLV); human T-cell leukemia virus type I (HTLV-I); bovine leukemia virus (BLV); Rous Sarcoma Virus (RSV); human immunodeficiency virus (HIV); yeast, including Saccharomyces, Neurospora, Drosophila; primates; and rodents. See, for example, Weiss et al., U.S. Pat. No. 4,663,290 (1987); Gerard, G. R., [0035] DNA 5:271-279 (1986); Kotewicz, M. L., et al., Gene 35:249-258 (1985); Tanese, N., et al., Proc. Natl. Acad. Sci. (USA) 82:4944-4948 (1985); Roth, M. J., et al., J. Biol. Chem. 260:9326-9335 (1985); Michel, F., et al., Nature 316:641-643 (1985); Akins, R. A., et al., Cell 47:505-516 (1986), EMBO J. 4:1267-1275 (1985); and Fawcett, D. F., Cell 47:1007-1015 (1986).
  • RT proviral DNA can be isolated using standard isolation techniques. The DNA is cleaved into linear fragments, any one of which may contain the genes which encode RT. Such fragmentation can be achieved using enzymes which digest or cleave DNA, such as restriction enzymes which cleave DNA as specific base sequences. After the linear DNA fragments are generated, they are separated according to size by standard techniques. Such recombinant DNA techniques may be performed as described by Maniatis, T., et al., [0036] Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y. (1982).
  • Identification of the DNA fragment which contains the gene may be accomplished in a number of ways. For example, it is possible to sequence the DNA fragments (Maxam and Gilbert, [0037] Methods in Enzymology 64.499 (1980); Messing, J., Meth. in Enz. 101C:20 (1983)) to identify which fragment contains the reverse transcriptase gene. Alternatively, hybridization techniques (Southern, J. Mol. Biol. 98:503 (1975)) using a labeled (e.g., radioactively labeled) DNA probe may be employed.
  • The fractions containing the desired DNA are pooled, ligated into a suitable vector, and used to transform a host. Screening for transformed hosts containing the RT gene may be accomplished by, for example, the method disclosed by Gerard et al., [0038] Biochem. 13:1632-1641 (1974) or by Gerard et al., J. Virol. 15:785-797 (1975). Alternatively, clones containing reverse transcriptase may be identified by hybridization with complementary labeled DNA.
  • An alternative to isolating the reverse transcriptase gene from a retroviral proviral DNA is to make cDNA to the mRNA-like RNA which codes for reverse transcriptase. To this end, mRNA-like RNA coding for reverse transcriptase is isolated from retrovirus. By standard techniques, the isolated mRNA is then converted into cDNA using reverse transcriptase. The cDNA can then be inserted into a plasmid vector in a conventional manner. [0039]
  • The choice of a suitable vector depends on a number of considerations known to one of ordinary skill in the art, such as the size of the fragment, nature of the host, number and position of restriction sites desired, and the selection marker and markers desired. Such vectors may include replicon and control sequences from species compatible with a host cell (see Maniatis et al., supra). Expression of the RT genes may also be placed under control of other regulatory sequences homologous or heterologous to the host organism in its untransformed state. For example, lactose-dependent [0040] E. coli chromosomal DNA comprises a lactose or lac operon which mediates lactose utilization by elaborating the enzyme β-galactosidase. The lac control elements may be obtained from bacteriophage lambda plac 5, which is infectious for E. coli. The lac promoter-operator system can be induced by IPTG.
  • Other promoter-operator systems or portions thereof can be employed as well. For example, galactose, alkaline phosphatase, tryptophan, xylose, tac, lambda pL, lambda pR and the like can be used. Once the vector or DNA sequence containing the constructs has been prepared, the vectors may be introduced into an appropriate host. Various techniques may be employed such as protoplast fusion, CaCl[0041] 2, calcium phosphate precipitation, electroporation, or other conventional DNA transfer techniques. The vectors may then be introduced into a second host by similar transfer methods, and also by cell to cell transfer methods such as conjugation. This cell-to-cell transfer may be accomplished using known techniques which depend upon the nature of the transferer bacterium, the recipient bacterium, and the cloning vector used to propagate the RT DNA. The transfer may require the use of a helper plasmid. See, for example, Ditta, G., et al., Proc. Natl. Acad. Sci. (USA) 77:7347-7351 (1980).
  • RT genes having DNA polymerase activity and substantially no RNase H activity may be obtained by deletion of deoxyribonucleotides at the 3′ end of the gene which encode the portion of the polypeptide having RNase H activity. Deletions of the RT gene may be accomplished by cutting the plasmid at selected restriction sites within the RT gene and discarding the excised fragment. Further deletion of consecutive deoxyribonucleotides may be accomplished by treating the fragment with an exonuclease. The DNA ends may then be joined in such a way that the translation reading frame of the gene is maintained. The plasmid thus obtained may then be used to transform hosts which may then be screened for altered RT activity. RT RNase H activity may be assayed according to Gerard et al., [0042] J. Virol. 15:785-797 (1975). DNA polymerase activity may be assayed according to Gerard et al., Biochem. 13:1632-1641 (1974). Clones having DNA polymerase activity and substantially no RNase H activity may be used to prepare RT with altered activity.
  • According to these methods, the portion of the RT gene derived from M-MLV which encodes DNA polymerase was localized to about 1495 base pairs (about 1018 to about 2512) as shown in FIG. 6. The protein expressed by this gene has about 503 amino acids (FIG. 6). This protein has DNA polymerase activity and substantially no RNase H activity. [0043]
  • The invention also relates to fusion proteins which comprise the reverse transcriptase of the invention. Such fusion proteins may comprise, for example, a carrier protein which has a leader sequence of hydrophobic amino acids at the amino terminus of the reverse transcriptase. This carrier protein is normally excreted through the membrane of the cell within which it is made. By cleavage of the hydrophobic leader sequence during excretion, a means is provided for producing reverse transcriptase which can be recovered either from the periplasmic space or the medium in which the bacterium is grown. The use of such a carrier protein allows isolation of reverse transcriptase without contamination by other proteins within the bacterium, and achieves production of a form of reverse transcriptase having greater stability by avoiding the enzymes within the bacterial cell which degrade foreign proteins. The DNA and amino acid sequences for such hydrophobic leader sequences, as well as methods of preparing such fusion proteins are taught, for example, by Gilbert et al., U.S. Pat. No. 4,411,994 (1983). [0044]
  • It is also possible to prepare fusion proteins comprising the reverse transcriptase of the invention which is substituted at the amino or carboxy termini with polypeptides which stabilize or change the solubility of the reverse transcriptase. An amino-terminal gene fusion which encodes reverse transcriptase, having both DNA polymerase and RNase activity, and trpE taught, for example, by Tanese, N. et al., [0045] Proc. Nat'l. Acad. Sci. 82:4944-4948 (1985). A carboxy-terminal gene fusion which encodes reverse transcriptase and part of the plasmid pBR322 tet gene is taught, for example, by Xotewicz, M., et al., Gene 35:249-258 (1985) and Gerard, G., DNA 5:271-279 (1986).
  • The transformed hosts of the invention may be cultured under protein producing conditions according to any of the methods which are known to those skilled in the art. [0046]
  • The reverse transcriptase having DNA polymerase activity and substantially no RNase activity may be isolated according to conventional methods known to those skilled in the art. For example, the cells may be collected by centrifugation, washed with suitable buffers, lysed, and the reverse transcriptase isolated by column chromatography, for example, on DEAE-cellulose, phosphocellulose (see Kotewicz et al., [0047] Gene 35:249-258 (1985)) or other standard isolation and identification techniques using, for example, polyribocytidylic acid-agarose, or hydroxylapatite or by electrophoresis or immunoprecipitation.
  • The reverse transcriptase so produced may be used to prepare cDNA from RNA by, for example, hybridizing an oligo(dT) primer or other complementary primers with the mRNA. The synthesis of a complete cDNA may be accomplished by adding the reverse transcriptase and all four deoxynucleoside triphosphates. Using the reverse transcriptase produced by the present invention allows for the preparation of cDNA from mRNA without concomitant degradation of the mRNA which results in incomplete cDNA synthesis. The resulting RNA-DNA hybrid may be treated, for example, with alkali or RNase H to selectively hydrolyze the RNA to leave cDNA which may be converted to double-stranded form in a second DNA reaction catalyzed by reverse transcriptase or other DNA polymerase. See Old, R. W., et al., [0048] Principals of Gene Manipulation, second edition, Studies in Microbiology, Vol. 2, University of California Press, p. 26 (1981).
  • The reverse transcriptase of the invention is ideally suited for incorporation into a kit for the preparation of cDNA from RNA. Such a kit may comprise a carrier means being compartmentalized to receive a close confinement therein, one or more container means, such as vials, tubes, and the like, each of said container means comprising one of the separate elements of the method used to prepare cDNA from RNA. For example, there may be provided a container means containing reverse transcriptase having DNA polymerase activity and substantially no RNase H activity, in solution. Further container means may contain suitable buffers, substrates for DNA synthesis such as the deoxynucleoside triphosphate, oligo(dT) primer, and control RNA for use as a standard. [0049]
  • The reverse transcriptase may be present in the solution at a concentration of 200 units/ml to 400 units/ml. The deoxynucleoside triphosphases may be present either in lyophilized form or as part of a buffer at a concentration of 0.5 mM to 2 mM. A suitable buffer, present at 5 times the final concentration of use, includes 250 mM Tris-HCl (pH 7.5 to 8.3), 375 mM KCl, 15 mM MgCl[0050] 2, and 50 mM dithiothreitol. The oligo (dT) may be present at a concentration of 5 μg/ml to 20 μg/ml. Control RNA, such as 2.3 kb control RNA, may be present at a concentration of 10 μg/ml to 20 μg/ml.
  • The following examples are illustrative but not limiting of the methods and compositions of the present invention. Any suitable modifications and adaptations which are obvious to one of ordinary skill in the art in recombinant DNA techniques are within the spirit and scope of the present invention. [0051]
  • EXAMPLES
  • Materials and Methods [0052]
  • Plasmids and Bacterial Strains [0053]
  • For deletion analysis of RT, a clone of M-MLV RT was constructed to overproduce stable RT in [0054] Escherichia coli, pRT601 (FIG. 1). Gerard, G. F., et al., DNA 5:271-279 (1986). It is a pBR322 replicon containing the strong lambda leftward promoter, pL, and the ribosome binding site of the lambda cII gene. (Higher copy number derivatives of pBR322, such as pUC plasmids, can also be used.) The coding sequence for the RT gene was carefully engineered into this plasmid to produce a protein with the amino terminus of the viral protein, and a carboxy terminus similar to the viral enzyme. Gerard, G. F., supra.
  • Two bacterial strains were used to propagate clones and express RT: K802 (Maniatis, T., et al., (1982), [0055] Molecular Cloning: A Laboratory Handbook, pp. 504-505, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y.), made lysogenic for lambda cIindlts857 Sam7, and N4830 (Gottesman, M. E., et al., J. Mol. Biol. 140:57-75 (1980)), which contains a deleted a cryptic lambda prophage expressing the temperature sensitive cI allele indlts857. Bacteria were grown in super broth (SB) containing 2% tryptone, 1% yeast extract, 0.1% NaCl, pH 7.5, and 50 μg/ml ampicillin.
  • Plasmid Construction [0056]
  • Standard procedures for plasmid construction were performed as described previously (Kotewicz, M. L., et al., [0057] Gene 35:249-258 (1985); Gerard, G. F., et al., DNA 5:271-279 (1986)).
  • Temperature Induction of [0058] E. coli Carrying pRT601 and Its Derivatives
  • Cultures of bacteria were grown in SB broth overnight at 32° C. and diluted 1:20 in fresh SB in the morning. The cells were grown at 32° C. until the A[0059] 590 was 0.8, and were induced by swirling in a 65° C. water bath until the temperature reached 42° C. Induction was continued for 30 minutes in a shaking water bath at 42° C., and then the cultures were incubated at 37° C. and grown an additional 30 minutes.
  • Preparation of Cell Extracts [0060]
  • Unless otherwise noted, all operations were performed at 4° C. Pelleted cells from one ml of culture were washed, lysed, and centrifuged as described previously (Kotewicz, M. L., supra). Supernatants were removed and assayed for RNase H and DNA polymerase activity. [0061]
  • Enzymes Assays [0062]
  • RT DNA polymerase activity in extracts was assayed specifically by using poly(2′-O-methylcytidylate).oligodeoxyguanylate [(Cm)[0063] n.(dG)12-18] (Gerard, G. F., et al., DNA 5:271-279 (1986), eliminating interference from cellular DNA polymerases. To establish DNA polymerase specific activities of purified RT preparations, activity was assayed with (A)n.(dT)12-18 (Houts, G. E., et al., J. Virol. 29:517-522 (1979) as described by Gerard, G. F., et al., DNA 5:271-279 (1986). One unit of DNA polymerase activity is the amount of enzyme that incorporates one nmole of deoxynucleoside monophosphate into acid insoluble product at 37° C. in 10 min.
  • RNase H activity in crude extracts and purified enzyme was assayed in reaction mixtures (50 μl) containing 50 mM Tris-HCl (pH 8.3), 2 mM MnCl[0064] 2, 1 mM dithiothreitol, and [3H](A)n.(dT)n (5 μM [3H](A)n, 35 cpm/p-mole; 20 μM (dT)n). Reactions were incubated at 37° C. for 20 min and were stopped by adding 10 μl of tRNA (1 mg/ml) and 20 μl of cold 50% TCA. After 10 minutes on ice, the mixture was centrifuged for 10 minutes in an Eppendorf centrifuge. Forty μl of the supernatant was counted in aqueous scintillant. One unit of RNase H activity is the amount of enzyme required to solubilize one mole of [3H](A)n in [3H](A)n.(dT)n in 10 min at 37° C.
  • Synthesis of Poly(A)-Tailed RNA [0065]
  • Synthetic 2.3 kb and 6.2 kb RNAs containing a 19 nucleotide poly(A) tail at the 3′ end were synthesized with T7 RNA polymerase from Xba I-cut pJD2.3 and Hind III-cut pHL3X, respectively. Reaction mixtures (0.3 ml) contained 40 uM Tris-HCl (pH 8.0), 8 mM MgCl[0066] 2, 2 mM spermidine-HCl, 5 mM dithiothreitol, 0.4 mM each of CTP, UTP, GTP, and ATP, 20 μg/ml DNA, and 2,000 units/ml T7 RNA polynerase. Uniformly labeled RNA was synthesized with all four [α-32P]NTPs, each at 0.4 mM and 250 cpm/pmole. After 1 hr incubation at 37° C., the RNA product was phenol extracted, ethanol precipitated, and purified by oligo(dT)-cellulose chromatography to ensure the presence of a poly(A) tail.
  • Conditions for cDNA Synthesis [0067]
  • When assessing the effect of cDNA synthesis upon the integrity of template RNA, reaction mixtures (50 μl) contained 50 mM Tris-HCl (pH 8.3), 75 mM KCl, 3 mM MgCl[0068] 2, 10 mM dithiothreitol, 0.5 mM each dATP, dGTP, and dTTP, 0.5 mM [3H]dCTP (200 cpm/pmol), 50 μg/ml (dT)12-18, 20 μg/ml 2.3 kb [32P]labeled RNA, and 4,000 units/ml RT. The reactions were incubated at 37° C. and duplicate 2.5 μl aliquots were removed at 0, 1, 5, 10, 30, and 60 min. One aliquot was precipitated onto glass fiber filters using TCA to determine the amount of cDNA synthesized, and the other aliquot was prepared for glyoxal gel analysis. Carmichael, G. G., et al., Method. Enzymol. 65:380-391 (1980). The glyoxalated RNA was fractionated on a 1% agarose gel, dried, and autoradiographed. In some cases, 10 units of E. coli RNase H were added to the reaction mixture after 60 min and the incubation continued for 10 more min before aliquots were taken.
  • When measuring the ability of RT to synthesize a cDNA copy of long RNA, reaction mixtures (10 μl) contained the same buffer and salts, 0.5 mM each of dATP, dGTP, dTTP, and [α-[0069] 32P]dCTP (600 cpm/pmole), 50 μg/ml actinomycin D, 50 μg/ml (dT)12-18, 100 μg/ml 6.2 kb poly(A)-tailed RNA, and 20,000 units/ml RT. After 1 hr at 37° C., the product in an aliquot (1 μl) was precipitated with TCA, counted, and the remaining DNA size fractionated on an alkaline 1.4% agarose gel according to McDonnel, M. W, et al., J. Mol. Biol. 110:119-146 (1977).
  • Purification of RT [0070]
  • Cells were grown to an A[0071] 590 of 3 in TYN and ampicillin medium (Gerard, G. F., et al., DNA 5:271-279 (1986)) at 30° C., induced at 43° C. for 45 min, and then grown at 36° C. for 3.5 hr before harvesting. RT was extracted from 100 g of cells as described (Gerard, G. F., supra) with the following exceptions. RT was precipitated by addition of solid (NH4)2SO4 to 40% saturation. The (NH4)2SO4 pellet was dissolved in 50 ml of 20 mM Tris-HCl (pH 7.5), 1 mM EDTA, 0.1 M NaCl, 5% glycerol, 1 mM, dithiothreitol, and 0.01% n-octyl-β-D-glucopyranoside, the suspension was clarified by centrifugation at 10,000×g for 10 min, and the supernatant was desalted on a 320 ml (5×16 cm) Sephadex G-25 column run in buffer A (20 mM Tris-HCl, pH 7.5, 1 mM dithiothreitol, 1 mM EDTA, 5% glycerol, 0.01% NP-40) plus 0.1 M NaCl. After phosphocellulose chromatography, the RT peak was pooled, diluted with an equal volume of buffer A, and chromatographed on a 21-ml heparin-agarose column (1.5×12 cm) equilibrated in buffer A plus 0.1 M NaCl. The RT peak from the heparin-agarose column was chromatographed on a Mono-S HR 5/5 column equilibrated in buffer A (Gerard, G. F., supra).
  • RESULTS [0072]
  • Construction of Reverse Transcriptase Gene Deletions [0073]
  • Deletions of the M-MLV RT gene were constructed by cutting pRT601 (FIG. 1) at selected restriction sites within the RT gene, discarding the excised fragment, and joining the DNA ends in such a way that the translation reading frame of the gene was maintained. pRTdBam-Bam was constructed by deleting the Bam HI fragment between nucleotide positions 1,654 and 1,960 (FIGS. 1 and 2). Ligation of the Bam HI half sites at positions 1,654 and 1,960 maintained the translation reading frame across the site. [0074]
  • A deletion at the carboxy terminus of M-MLV RT (pRTdEcoRV-C) was constructed by deleting all of the 3′ end of the gene downstream of the Eco RV site at position 2,513 (FIGS. 1 and 2). To construct pRTdEcoRV-C, a Sca I (position 6,238) to Eco RV (position 2,513) fragment of pRT601 containing the 5′ portion of the RT gene was ligated to a Sca I-Eco RI fragment derived from plasmid pBRT (Gerard, G. F., supra). The 3,211 base pair pBRT Sca I-Eco RI fragment contained the pBR322 origin of replication and a universal translation terminator sequence just inside the Eco RI site. The Eco RI site was repaired with DNA polymerase I Klenow fragment before ligation. [0075]
  • The plasmid of pRTdEcoRV-C was deposited in [0076] E. coli under the terms of the Budapest Treaty at the American Type Culture Collection (ATCC), Rockville, Md., and given accession number 67555.
  • pRT603 was constructed as described (Gerard, G. R., supra) which encodes an RT that contains 73 fewer amino acids than pRT601, all deleted from the carboxy terminus (FIG. 2). [0077]
  • DNA Polymerase and RNase H Levels in Cells Bearing Deletion Plasmids [0078]
  • Alteration of as little as 3 amino acids at the carboxy end of M-MLV RT can influence markedly the stability of the protein in [0079] E. coli (Gerard, G. F. supra). This must be taken into consideration in making correlations between cloned RT deletions and enzymatic activities in E. coli extracts. Both DNA polymerase and RNase H activity must be assayed and relative enzyme levels compared. For example, pRT603 codes for an RT with 73 fewer amino acids at the carboxy terminus than pRT601 RT (Gerard, G. F., supra; FIG. 2). The level of DNA polymerase activity in E. coli extracts of pRT603 RT is reduced 5 fold relative to pRT601 (Gerard, G. F. supra). However, the DNA polymerase and RNase H specific activities of purified pRT601 and pRT603 RT are comparable (Table 2). The reduced DNA polymerase activity in E. coli extracts of pRT603 RT is not due to a selective effect of the deletion on DNA polymerase activity, but rather to a reduction in the stability of pRT603 RT relative to pRT601 RT in cells (t½ of 7 min versus 33 min) (Gerard, G. F. supra). Therefore, deletions within 70 amino acids of the RT carboxy terminus do not affect either RNase H or DNA polymerase activity.
  • In contrast, the DNA polymerase activity of pRTdBam-Bam RT was eliminated totally without affecting RNase H activity (Table 1) by the deletion of 102 amino acid residues between [0080] amino acids 212 and 314 (FIG. 2). Introduction of a more extensive deletion of 180 amino acids at the carboxy end of RT in pRTdEcoRV-C RT (FIG. 2) yielded extracts with RT DNA polymerase levels unchanged compared to pRT601 extracts, but with RNase H levels reduced 7.5 fold (Table 1). The residual RNase H activity in pRTdEcoRV-C extracts could be due to E. coli RNase H, the 5′→3′ exonuclease of DNA polymerase I, or a small amount of residual RT-coded RNase H activity. To resolve this issue, pRTdEcoRV-C RT was purified and compared to RT encoded by pRT601.
  • Purification and Properties of pRTdEcoRV-C RT [0081]
  • M-MLV reverse transcriptase encoded by pRTdEcoRV-C, pRT601, and pRT603 were purified as described in Materials and Methods. A summary of the purification of pRTdEcoRV-C RT is presented in Table 3. Three column steps produced a nearly homogeneous mutant enzyme with the same DNA polymerase specific activity as pRT601 RT with the template-primer (Cm)[0082] n.(dG)12-18 (Table 2). With (A)n.(dT)12-18, the mutant enzyme had one-fourth the DNA polymerase activity of pRT601 RT (Table 2). RNase H activity of purified pRTdEcoRV-C RT was undetectable using [3H](A)n.(dT)n as the substrate. Most RNase H activity in extracts was eliminated from mutant RT by precipitation of the enzyme with 40% (NH4)2SO4 (Table 3). Under these conditions, DNA polymerase I remains soluble (Richardson, C., et al., J. Biol. Chem. 239:222-230 (1964)), as does most of the RNase H activity in the extract. As judged by SDS-polyacrylamide gel electrophoresis, pRTdEcoRV-C RT purified through the Mono-S column was greater than 90% pure and had a molecular weight of 56,000 (FIG. 3), consistent with the molecular weight (57,000) predicted by the DNA sequence.
  • A number of enzymatic properties of purified pRTdEcoRV-C RT and pRT601 RT were compared and were found to be similar. These included half life at 37° C., monovalent and divalent metal ion optima, fidelity of dNTP incorporation with homopolymer templates, and insensitivity to stimulation by polyanions. The abilities of the two enzymes to synthesize heteropolymeric DNA were also compared. FIG. 4 shows that pRTdEcoRV-C RT catalyzed the synthesis of full-length cDNA from 6.2 kb RNA more efficiently than pRT601 RT. The amount of cDNA synthesized from 1 μg of RNA was 0.28 μg (34% full-length) and 0.24 μg (24% full-length) with pRTdEcoRV-C RT and pRT601 RT, respectively. [0083]
  • To confirm that pRTdEcoRV-C RT completely lacked RNase H activity, the integrity of a uniformly [0084] 32P-labeled RNA template after conversion to hybrid form during RT-catalyzed DNA synthesis was examined. FIG. 5 shows that with pRT601 RT, the full-length 2.3 kb RNA template was degraded totally after 5 min of synthesis. In contrast, with pRTdEcoRV-C RT the RNA was intact even after 60 min. The amount of cDNA synthesized after 60 min from 1 μg of RNA was 0.67 and 0.76 μg with pRT601 and pRTdEcoRV-C RT, respectively. When 10 units of E. coli RNase H were added to the pRTdEcoRV-C RT reaction after 60 min of incubation, all of the RNA was degraded, confirming the hybrid state of the RNA. In addition, 15 μg (1,200 units) of pRTdEcoRV-C RT solubilized no radioactivity from a [3H](A)n.(dT)n substrate in which the [3H](A)n had a specific activity of 2,200 cpm/pmole (Materials and Methods).
  • Experiments with a frameshift mutant of MLV producing a 47K RT molecule truncated at the carboxy terminus (Levin, J. G. et al., [0085] J. Virol. 51:470-478 (1984)) and with antibodies to synthetic peptides modeled to Rous sarcoma virus pol gene sequences (Grandgenett, D. et al., J. Biol. Chem. 260:8243-8249 (1985)) suggest the RNase H activity of RT resides within the amino-terminal portion of the molecule. Conversely, the extensive homology found between the amino acids of E. coli RNase H and the 153-residue segment at the carboxy-terminal end of M-MLV RT (Johnson, M. S. et al. Proc. Natl. Acad. Sci (USA) 83:7648-7652 (1986)) suggests the RNase H activity resides within the carboxy-terminal portion of RT.
  • By deleting large segments (100 to 200 codons) of the M-MLV RT gene, the regions within the RT molecule responsible for DNA polymerase and RNase H activity have been identified. DNA polymerase was mapped to the amino half of the molecule, and RNase H to within 200 amino acids of the carboxy end, confirming the predictions based upon amino acid homology (Johnson, M. S. et al., supra). In this context, the results with one RT clone, pRT603 (FIG. 2), are of interest. The RT protein encoded by pRT603 is missing the carboxy half of the 153 amino acid segment of RT homologous to [0086] E. coli RNase H, which includes 20 of 48 homologous amino acids. Yet, pRT603 RT has normal levels of RNase H activity. These missing, homologous residues apparently are not required for catalysis, and might serve a nucleic acid binding or structural role. Consistent with the latter, a single amino acid change at a position 12 residues from the carboxy end of E. coli RNase H produces a 10-fold reduction in RNase H specific activity (Kanaya, S. et al., J. Bacteriol. 154:1021-1026 (1983)). This reduction appears to be the result of altered protein conformation (Kanaya, S. et al., supra).
  • If the RT polymerase and nuclease active sites reside on separate structural domains, it should be possible theoretically to isolate two separate protein fragments, each with a single activity. A 24K to 30K proteolytic fragment of RT possessing only RNase H activity has been isolated (Lai, M. H. T. et al., [0087] J. Virol. 25:652-663 (1978); Gerard, G. F., J. Virol. 26:16-28 (1978); Gerard, G. F., J. Virol. 37:748-754 (1981)), but unfortunately, the location of this RNase H fragment in the parent RT polypeptide has not been established, and no analogous DNA polymerase containing fragment has ever been found. The results presented here show that of the 684 amino acids in pRT601 RT, residues between amino acid 212 and 314 are required for DNA polymerase activity, and residues between amino acid 503 and 611 are required for RNase H activity. They also demonstrate for the first time that the RT DNA polymerase activity can exist independently of RNase H activity on an RT protein fragment. Purified pRTdEcoRV-C RT appeared to be totally devoid of RNase H activity, based upon two sensitive assays, and to have full DNA polymerase activity. However, these results do not rule out the possibility that the two active centers share a portion(s) of the RT molecule.
  • Demonstration of a separate structural domain for the RNase H active center was attempted by constructing two amino-terminal deletion derivatives of pRT601. The first derivative contained sequences for the Eco RV site at [0088] position 2513 to the 3′ end of the RT gene (see FIG. 2), and the second contained sequences from an Nco I site at position 2302 to the 3′ end of the RT gene. Unfortunately, neither clone produced detectable RNase H activity in E. coli crude extracts. Such negative results are difficult to interpret because the proteins might be unable to fold in an active form, or might be extremely labile.
  • Deletion of the carboxy-terminal one-fourth of the M-MLV RT molecule did not disrupt the ability of the protein to fold in an active conformation. pRTdEcoRV-C RT copied heteropolymeric RNA more efficiently than intact RT. Yields of cDNA from 1 μg of 2.3 kb and 6.2 kb RNA were 0.76 μg (50% full-length) and 0.28 μg (34% full-length), respectively. Also, the truncated and intact enzymes had the same DNA polymerase specific activity with (Cm)[0089] n.(dG)12-18. However, the truncated enzyme copied (A)n.(dT)12-18 only one fourth as efficiently as the parent RT. The origin of this difference has not yet been established.
    TABLE 1
    DNA polymerase and RNase H activity in extracts of heat
    induced E. coli K802 (lambda) bearing pRT601 or one of its
    derivatives.
    DNA polymerase
    Activitya RNase H
    (cpm incorporated/ Activityb
    Plasmid 2.5 μl extract) (cpm solubilized/2.5 μl extract)
    pRT601 10,977 2,020
    pRTdBam-Bam 179 1,564
    pRTdEcoRV-C 10,038 268
  • [0090]
    TABLE 2
    Comparison of activities of purified RT coded by pRT601,
    pRT603, and pRTdEcoRV-C
    DNA Polymerase Activity
    with
    (Cm)n.(dG)12-18 (A)n.(dT)12-18 RNase H Activity
    Enzyme (Units/mg) (Units/mg) (Units/mg)
    pRT601 21,700 350,000 2,670
    pRT603 NDa 230,000 1,100
    pRTdEcoRV-C 17,500 81,000 b
  • [0091]
    TABLE 3
    Summary of the purification of pRTdEcoRV-C RT
    DNA Polymerase Activitya RNase H Activity
    Specific Specific
    Total Total Activity Total Activity
    Proteinc (Units) (Units/mg) Yield (Units) (Units/mg) Yield
    Fraction (mg) ×103 ×103 (%) ×103 ×103 (%)
    Crude lysate 7,913 255 0.03 100 80 0.01 100
    Polymin P 2,735 323 0.12 127 157 0.06 196
    Supernatant
    (NH4)2SO4 63 168 1.38 66 6.0 0.10 7
    pellet
    Phosphocellulose 8.8 167 19.0 66 2.0 0.23 3
    pool
    Heparin-agarose 6.5 111 17.1 44 _b
    pool
    Mono S 3.1 55 17.5 22 _b
    pool

Claims (23)

What is claimed is:
1. A gene which encodes reverse transcriptase having DNA polymerase activity and substantially no RNase H activity.
2. The gene of claim 1, wherein said gene is derived from an organism selected from the group consisting of Moloney murine leukemia virus (M-MLV), human T-cell leukemia virus type I (HTLV-I), bovine leukemia virus (BLV), Rous sarcoma virus (RSV), human immunodeficiency virus (HIV), yeast, Neurospora, Drosophila, primates and rodents.
3. The gene of claim 1, wherein said microorganism is M-MLV, comprising the following DNA sequence:
                                                                           1078 A{dot over (T)}G ACC CTA AA{dot over (T)} ATA GAA GAT GAG CAT CGG C{dot over (T)}A CAT GAG AC{dot over (C)} TCA AAA GAG {dot over (C)}CA GAT GTT                                                                            1138 TCT CTA GGG TCC ACA TGG CTG TCT GAT TTT CCT CAG GCC TGG GCG GAA ACC GGG GGC ATG                                                                            1198 GGA CTG GCA GTT CGC CAA GCT CCT CTG ATC ATA CCT CTG AAA GCA ACC TCT ACC CCC GTG                                                                            1258 TCC ATA AAA CAA TAC CCC ATG TCA CAA GAA GCC AGA CTG GGG ATC AAG CCC CAC ATA CAG                                                                            1318 AGA CTG TTG GAC CAG GGA ATA CTG GTA CCC TGC CAG TCC CCC TGG AAC ACG CCC CTG CTA                                                                            1378 CCC GTT AAG AAA CCA GGG ACT AAT GAT TAT AGG CCT GTC CAG CAT CTG AGA GAA GTC AAC                                                                            1438 AAG CGG GTG GAA GAC ATC CAC CCC ACC GTG CCC AAC CCT TAC AAC CTC TTG AGC GGG CTC                                                                            1498 CCA CCG TCC CAC CAG TGG TAC ACT GTG CTT GAT TTA AAG GAT GCC TTT TTC TGC CTG AGA                                                                            1558 CTC CAC CCC ACC AGT CAG CCT CTC TTC GCC TTT GAG TGG AGA GAT CCA GAG ATG GGA ATC                                                                            1618 TCA GGA CAA TTG ACC TGG ACC AGA CTC CCA CAG GGT TTC AAA AAC AGT CCC ACC CTG TTT                                                                            1678 GAT GAG GCA CTG CAC AGA GAC CTA GCA GAC TTC CGG ATC CAG CAC CCA GAC TTG ATC CTG                                                                            1738 CTA CAG TAC GTG GAT GAC TTA CTG CTG GCC GCC ACT TCT GAG CTA GAC TGC CAA CAA GGT                                                                            1798 ACT CGG GCC CTG TTA CAA ACC CTA GGG AAC CTC GGG TAT CGG GCC TCG GCC AAG AAA GCC                                                                            1858 CAA ATT TGC CAG AAA CAG GTC AAG TAT CTG GGG TAT CTT CTA AAA GAG GGT CAG AGA TGG                                                                            1918 CTG ACT GAG GCC AGA AAA GAG ACT GTG ATG GGG CAG CCT ACT CCG AAG ACC CCT CGA CAA                                                                            1978 CTA AGG GAG TTC CTA GGG ACG GCA GGC TTC TGT CGC CTC TGG ATC CCT GGG TTT GCA GAA                                                                            2038 ATG GCA GCC CCC TTG TAC CCT CTC ACC AAA ACG GGG ACT CTG TTT AAT TGG GGC CCA GAC                                                                            2098 CAA CAA AAG GCC TAT CAA GAA ACT AAG CAA GCT CTT CTA ACT GCC CCA GCC CTG GGG TTG                                                                            2158 CCA GAT TTG ACT AAG CCC TTT GAA CTC TTT GTC GAC GAG AAG CAG GGC TAC GCC AAA GGT                                                                            2218 GTC CTA ACG CAA AAA CTG GGA CCT TGG CGT CGG CCG GTG GCC TAC CTG TCC AAA AAG CTA                                                                            2278 GAC CCA GTA GCA GCT GGG TGG CCC CCT TGC CTA CGG ATG GTA GCA GCC ATT GCC GTA CTG                                                                            2338 ACA AAG GAT GCA GGC AAG CTA ACC ATG GGA CAG CCA CTA GTC ATT CTG GCC CCC CAT GCA                                                                            2398 GTA GAG GCA CTA GTC AAA CAA CCC CCC GAC CGC TGG CTT TCC AAC GCC CGG ATG ACT CAC                                                                            2458 TAT CAG GCC TTG CTT TTG GAC ACG GAC CGG GTC CAG TTC GGA CCG GTG GTA GCC CTG AAC CCG GCT ACG CTG CTC CCA CTG CCT GAG GAA GGG CTG CAA CAC AAC TGC CTT GAT
or the degenerate variants thereof.
4. The gene of claim 1, wherein said microorganism is M-MLV, comprising the following DNA sequence:
                                                                           1078 A{dot over (T)}G ACC CTA AA{dot over (T)} ATA GAA GAT GAG CAT CGG C{dot over (T)}A CAT GAG AC{dot over (C)} TCA AAA GAG {dot over (C)}CA GAT GTT                                                                            1138 TCT CTA GGG TCC ACA TGG CTG TCT GAT TTT CCT CAG GCC TGG GCG GAA ACC GGG GGC ATG                                                                            1198 GGA CTG GCA GTT CGC CAA GCT CCT CTG ATC ATA CCT CTG AAA GCA ACC TCT ACC CCC GTG                                                                            1258 TCC ATA AAA CAA TAC CCC ATG TCA CAA GAA GCC AGA CTG GGG ATC AAG CCC CAC ATA CAG                                                                            1318 AGA CTG TTG GAC CAG GGA ATA CTG GTA CCC TGC CAG TCC CCC TGG AAC ACG CCC CTG CTA                                                                            1378 CCC GTT AAG AAA CCA GGG ACT AAT GAT TAT AGG CCT GTC CAG GAT CTG AGA GAA GTC AAC                                                                            1438 AAG CGG GTG GAA GAC ATC CAC CCC ACC GTG CCC AAC CCT TAC AAC CTC TTG AGC GGG CTC                                                                            1498 CCA CCG TCC CAC CAG TGG TAC ACT GTG CTT GAT TTA AAG GAT GCC TTT TTC TGC CTG AGA                                                                            1558 CTC CAC CCC ACC AGT CAG CCT CTC TTC GCC TTT GAG TGG AGA GAT CCA GAG ATG GGA ATC                                                                            1618 TCA GGA CAA TTG ACC TGG ACC AGA CTC CCA CAG GGT TTC AAA AAC AGT CCC ACC CTG TTT                                                                            1678 GAT GAG GCA CTG CAC AGA GAC CTA GCA GAC TTC CGG ATC CAG CAC CCA GAC TTG ATC CTG                                                                            1738 CTA CAG TAC GTG GAT GAC TTA CTG CTG GCC GCC ACT TCT GAG CTA GAC TGC CAA CAA GGT                                                                            1798 ACT CGG GCC CTG TTA CAA ACC CTA GGG AAC CTC GGG TAT CGG GCC TCG GCC AAG AAA GCC                                                                            1858 CAA ATT TGC CAG AAA CAG GTC AAG TAT CTG GGG TAT CTT CTA AAA GAG GGT CAG AGA TGG                                                                            1918 CTG ACT GAG GCC AGA AAA CAG ACT GTG ATG GGG CAG CCT ACT CCG AAG ACC CCT CGA CAA                                                                            1978 CTA AGG GAG TTC CTA GGG ACG GCA GGC TTC TGT CGC CTC TGG ATC CCT GGG TTT GCA GAA                                                                            2038 ATG GCA GCC CCC TTG TAC CCT CTC ACC AAA ACG GGG ACT CTG TTT AAT TGG GGC CCA GAC                                                                            2098 CAA CAA AAG GCC TAT CAA GAA ATC AAG CAA GCT CTT CTA ACT GCC CCA GCC CTG GGG TTG                                                                            2158 CCA GAT TTG ACT AAG CCC TTT GAA CTC TTT GTC GAC GAG AAG CAG GGC TAC GCC AAA GGT                                                                            2218 GTC CTA ACG CAA AAA CTG GGA CCT TGG CGT CGG CCG GTG GCC TAC CTG TCC AAA AAG CTA                                                                            2278 GAC CCA GTA GCA GCT GGG TGG CCC CCT TGC CTA CGG ATG GTA GCA GCC ATT GCC GTA CTG                                                                            2338 ACA AAG GAT GCA GGC AAG CTA ACC ATG GGA CAG CCA CTA GTC ATT CTG GCC CCC CAT GCA                                                                            2398 GTA GAG GCA CTA GTC AAA CAA CCC CCC GAC CGC TGG CTT TCC AAC GCC CGG ATG ACT CAC                                                                            2458 TAT CAG GCC TTG CTT TTG GAC ACG GAC CGG GTC CAG TTC GGA CCG GTG GTA GCC CTG AAC                                                                            2518 CCG GCT ACG CTG CTC CCA CTG CCT GAG GAA GGG CTG CAA CAC AAC TGC CTT GAT AAT TCC            2530 CGC TTA ATT AAT
or the degenerate variants thereof.
5. A gene which encodes a fusion protein which comprises reverse transcriptase having DNA polynerase activity and substantially no RNase H activity and a second protein comprising a hydrophobic leader peptide or a stabilizing peptide.
6. A vector containing the gene of claim 1 or 5.
7. The vector of claim 6 designated pRTdEcoRV-C which has been deposited at the American Type Culture Collection, Rockville, Md. under terms of the Budapest Treaty and given accession number 67555.
8. A host transformed with the vector of claim 6.
9. A polypeptide having an amino acid sequence encoded by the cloned gene of claim 1 or 5.
10. The polypeptide of claim 9 comprising the following amino acid sequence:
M{dot over (E)}T Thr Leu As{dot over (n)} Ile Glu Asp Glu His Arg L{dot over (e)}u His Glu Th{dot over (r)} Ser Lys Glu {dot over (P)}ro Asp Val Ser Leu Gly Ser Thr Trp Leu Ser Asp Phe Pro Gln Ala Trp Ala Glu Thr Gly Gly MET Gly Leu Ala Val Arg Gln Ala Pro Leu Ile Ile Pro Leu Lys Ala Thr Ser Thr Pro Val Ser Ile Lys Gln Tyr Pro MET ser Gln Glu Ala Arg Leu Gly Ile Lys Pro His Ile Gln Arg Leu Leu Asp Gln Gly Ile Leu Val Pro Cys Gln Ser Pro Trp Asn Thr Pro Leu Leu Pro Val Lys Lys Pro Gly Thr Asn Asp Tyr Arg Pro Val Gln Asp Leu Arg Glu Val Asn Lys Arg Val Glu Asp Ile His Pro Thr Val Pro Asn Pro Tyr Asn Leu Leu Ser Gly Leu Pro Pro Ser His Gln Trp Tyr Thr Val Leu Asp Leu Lys Asp Ala Phe Phe Cys Leu Arg Leu His Pro Thr Ser Gln Pro Leu Phe Ala Phe Glu Trp Arg Asp Pro Glu MET Gly Ile Ser Gly Gln Leu Thr Trp Thr Arg Leu Pro Gln Gly Phe Lys Asn Ser Pro Thr Leu Phe Asp Glu Ala Leu His Arg Asp Leu Ala Asp Phe Arg Ile Gln His Pro Asp Leu Ile Leu Leu Gln Tyr Val Asp Asp Leu Leu Leu Ala Ala Thr Ser Glu Leu Asp Cys Gln Gln Gly Thr Arg Ala Leu Leu Gln Thr Leu Gly Asn Leu Gly Tyr Arg Ala Ser Ala Lys Lys Ala Gln Ile Cys Gln Lys Gln Val Lys Tyr Leu Gly Tyr Leu Leu Lys Glu Gly Gln Arg Trp Leu Thr Glu Ala Arg Lys Glu Thr Val MET Gly Gln Pro Thr Pro Lys Thr Pro Arg Gln Leu Arg Glu Phe Leu Gly Thr Ala Gly Phe Cys Arg Leu Trp Ile Pro Gly Phe Ala Glu MET Ala Ala Pro Leu Tyr Pro Leu Thr Lys Thr Gly Thr Leu Phe Asn Trp Gly Pro Asp Gln Gln Lys Ala Tyr Gln Glu Ile Lys Gln Ala Leu Leu Thr Ala Pro Ala Leu Gly Leu Pro Asp Leu Thr Lys Pro Phe Glu Leu Phe Val Asp Glu Lys Gln Gly Tyr Ala Lys Gly Val Leu Thr Gln Lys Leu Gly Pro Trp Arg Arg Pro Val Ala Tyr Leu Ser Lys Lys Leu Asp Pro Val Ala Ala Gly Trp Pro Pro Cys Leu Arg MET Val Ala Ala Ile Ala Val Leu Thr Lys Asp Ala Gly Lys Leu Thr MET Gly Gln Pro Leu Val Ile Leu Ala Pro His Ala Val Glu Ala Leu Val Lys Gln Pro Pro Asp Arg Trp Leu Ser Asn Ala Arg MET Thr His Tyr Gln Ala Leu Leu Leu Asp Thr Asp Arg Val Gln Phe Gly Pro Val Val Ala Leu Asn Pro Ala Thr Leu Leu Pro Leu Pro Glu Glu Gly Leu Gln His Asn Cys Leu Asp.
11. The polypeptide of claim 9 comprising the following amino acid sequence:
M{dot over (E)}T Thr Leu As{dot over (n)} Ile Glu Asp Glu His Arg L{dot over (e)}u His Glu Th{dot over (r)} Ser Lys Glu {dot over (P)}ro Asp Val Ser Leu Gly Ser Thr Trp Leu Ser Asp Phe Pro Gln Ala Trp Ala Glu Thr Gly Gly MET Gly Leu Ala Val Arg Gln Ala Pro Leu Ile Ile Pro Leu Lys Ala Thr Ser Thr Pro Val Ser Ile Lys Gln Tyr Pro MET ser Gln Glu Ala Arg Leu Gly Ile Lys Pro His Ile Gln Arg Leu Leu Asp Gln Gly Ile Leu Val Pro Cys Gln Ser Pro Trp Asn Thr Pro Leu Leu Pro Val Lys Lys Pro Gly Thr Asn Asp Tyr Arg Pro Val Gln Asp Leu Arg Glu Val Asn Lys Arg Val Glu Asp Ile His Pro Thr Val Pro Asn Pro Tyr Asn Leu Leu Ser Gly Leu Pro Pro Ser His Gln Trp Tyr Thr Val Leu Asp Leu Lys Asp Ala Phe Phe Cys Leu Arg Leu His Pro Thr Ser Gln Pro Leu Phe Ala Phe Glu Trp Arg Asp Pro Glu MET Gly Ile Ser Gly Gln Leu Thr Trp Thr Arg Leu Pro Gln Gly Phe Lys Asn Ser Pro Thr Leu Phe Asp Glu Ala Leu His Arg Asp Leu Ala Asp Phe Arg Ile Gln His Pro Asp Leu Ile Leu Leu Gln Tyr Val Asp Asp Leu Leu Leu Ala Ala Thr Ser Glu Leu Asp Cys Gln Gln Gly Thr Arg Ala Leu Leu Gln Thr Leu Gly Asn Leu Gly Tyr Arg Ala Ser Ala Lys Lys Ala Gln Ile Cys Gln Lys Gln Val Lys Tyr Leu Gly Tyr Leu Leu Lys Glu Gly Gln Arg Trp Leu Thr Glu Ala Arg Lys Glu Thr Val MET Gly Gln Pro Thr Pro Lys Thr Pro Arg Gln Leu Arg Glu Phe Leu Gly Thr Ala Gly Phe Cys Arg Leu Trp Ile Pro Gly Phe Ala Glu MET Ala Ala Pro Leu Tyr Pro Leu Thr Lys Thr Gly Thr Leu Phe Asn Trp Gly Pro Asp Gln Gln Lys Ala Tyr Gln Glu Ile Lys Gln Ala Leu Leu Thr Ala Pro Ala Leu Gly Leu Pro Asp Leu Thr Lys Pro Phe Glu Leu Phe Val Asp Glu Lys Gln Gly Tyr Ala Lys Gly Val Leu Thr Gln Lys Leu Gly Pro Trp Arg Arg Pro Val Ala Tyr Leu Ser Lys Lys Leu Asp Pro Val Ala Ala Gly Trp Pro Pro Cys Leu Arg MET Val Ala Ala Ile Ala Val Leu Thr Lys Asp Ala Gly Lys Leu Thr MET Gly Gln Pro Leu Val Ile Leu Ala Pro His Ala Val Glu Ala Leu Val Lys Gln Pro Pro Asp Arg Trp Leu Ser Asn Ala Arg MET Thr His Tyr Gln Ala Leu Leu Leu Asp Thr Asp Arg Val Gln Phe Gly Pro Val Val Ala Leu Asn Pro Ala Thr Leu Lue Pro Leu Pro Glu Glu Gly Leu Gln His Asn Cys Leu Asp Asn Ser Arg Leu Ile Asn.
12. A method of producing reverse transcriptase having DNA polymerase activity and substantially no RNase H activity comprising culturing the transformed host of claim 8 under conditions which produce reverse transcriptase, and isolating the reverse transcriptase so produced.
13. A method of preparing cDNA from mRNA, comprising
(a) contacting mRNA with an oligo(dT) primer or other complementary primer to form a hybrid, and
(b) contacting said hybrid formed in step (a) with reverse transcriptase, having DNA polymerase and substantially no RNase activity, and the nucleoside triphosphates to give a cDNA-RNA hybrid.
14. The method of claim 13, further comprising treating the cDNA-RNA with alkali or RNase H to selectively hydrolyze said RNA to give a cDNA.
15. The method of claim 13, further comprising treating said cDNA with DNA polymerase to give second-strand cDNA.
16. A kit for the preparation of cDNA from mRNA, comprising a carrier means being compartmentalized to receive in close confinement therein, one or more containers wherein
(a) a first container contains reverse transcriptase having DNA polymerase activity and substantially no Rnase H activity;
(b) a second container contains the nucleoside triphosphates, and
(c) a third container contains oligo(dt) primer.
17. The kit of claim 16, further comprising:
(d) a fourth container containing control RNA.
18. The kit of claim 16, wherein said second container further contains a buffer.
19. The kit of claim 16, wherein said reverse transcriptase is present at a concentration of 200 μg/μl to 400 μg/μl.
20. The kit of claim 16, wherein said oligo (dT) primer is present at a concentration of 5 μg/ml to 20 μg/ml.
21. The kit of claim 18, wherein said buffer comprises Tris-HCl (pH 7.5 to 8.3), KCl, MgCl2, and dithiothreitol.
22. The kit of claim 16, wherein said nucleoside triphosphates are present at a concentration of 0.5 mM to 2 mM.
23. The kit of claim 17, wherein said control RNA is present at a concentration of 10 μg/ml to 20 μg/ml.
US10/024,131 1988-01-13 2001-12-21 Cloned genes encoding reverse transcriptase lacking RNase H activity Abandoned US20030027313A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US10/024,131 US20030027313A1 (en) 1988-01-13 2001-12-21 Cloned genes encoding reverse transcriptase lacking RNase H activity
US10/970,280 US20060003341A1 (en) 1988-01-13 2004-10-22 Cloned genes encoding reverse transcriptase lacking RNase H activity
US11/754,968 US20080004437A1 (en) 1988-01-13 2007-05-29 Cloned Genes Encoding Reverse Transcriptase lacking RNase H Activity

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US14339688A 1988-01-13 1988-01-13
US07671156 US5244797B1 (en) 1988-01-13 1991-03-18 Cloned genes encoding reverse transcriptase lacking rnase h activity
US07825260 US5405776B1 (en) 1988-01-13 1992-01-24 Cloned genes encoding reverse transcriptase lacking RNase H activity
US40490795A 1995-03-15 1995-03-15
US08/614,260 US5668005A (en) 1988-01-13 1996-03-12 Cloned genes encoding reverse transcriptase lacking RNASE H activity
US08/798,458 US6063608A (en) 1988-01-13 1997-02-10 Cloned genes encoding reverse transcriptase lacking RNase H activity
US09/220,329 US6589768B1 (en) 1988-01-13 1998-12-24 Cloned genes encoding reverse transcriptase lacking RNase H activity
US10/024,131 US20030027313A1 (en) 1988-01-13 2001-12-21 Cloned genes encoding reverse transcriptase lacking RNase H activity

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/220,329 Continuation US6589768B1 (en) 1988-01-13 1998-12-24 Cloned genes encoding reverse transcriptase lacking RNase H activity

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/970,280 Continuation US20060003341A1 (en) 1988-01-13 2004-10-22 Cloned genes encoding reverse transcriptase lacking RNase H activity

Publications (1)

Publication Number Publication Date
US20030027313A1 true US20030027313A1 (en) 2003-02-06

Family

ID=22503881

Family Applications (11)

Application Number Title Priority Date Filing Date
US07671156 Expired - Lifetime US5244797B1 (en) 1988-01-13 1991-03-18 Cloned genes encoding reverse transcriptase lacking rnase h activity
US07825260 Expired - Lifetime US5405776B1 (en) 1988-01-13 1992-01-24 Cloned genes encoding reverse transcriptase lacking RNase H activity
US08/614,260 Expired - Fee Related US5668005A (en) 1988-01-13 1996-03-12 Cloned genes encoding reverse transcriptase lacking RNASE H activity
US08/798,458 Expired - Lifetime US6063608A (en) 1988-01-13 1997-02-10 Cloned genes encoding reverse transcriptase lacking RNase H activity
US09/220,329 Expired - Fee Related US6589768B1 (en) 1988-01-13 1998-12-24 Cloned genes encoding reverse transcriptase lacking RNase H activity
US09/220,330 Expired - Fee Related US6610522B1 (en) 1988-01-13 1998-12-24 Cloned genes encoding reverse transcriptase lacking RNase H activity
US10/024,149 Abandoned US20030039988A1 (en) 1988-01-13 2001-12-21 Cloned genes encoding reverse transcriptase lacking RNase H activity
US10/024,131 Abandoned US20030027313A1 (en) 1988-01-13 2001-12-21 Cloned genes encoding reverse transcriptase lacking RNase H activity
US10/929,791 Abandoned US20050233315A1 (en) 1988-01-13 2004-08-31 Cloned genes encoding reverse transcriptase lacking RNase H activity
US10/970,280 Abandoned US20060003341A1 (en) 1988-01-13 2004-10-22 Cloned genes encoding reverse transcriptase lacking RNase H activity
US11/754,968 Abandoned US20080004437A1 (en) 1988-01-13 2007-05-29 Cloned Genes Encoding Reverse Transcriptase lacking RNase H Activity

Family Applications Before (7)

Application Number Title Priority Date Filing Date
US07671156 Expired - Lifetime US5244797B1 (en) 1988-01-13 1991-03-18 Cloned genes encoding reverse transcriptase lacking rnase h activity
US07825260 Expired - Lifetime US5405776B1 (en) 1988-01-13 1992-01-24 Cloned genes encoding reverse transcriptase lacking RNase H activity
US08/614,260 Expired - Fee Related US5668005A (en) 1988-01-13 1996-03-12 Cloned genes encoding reverse transcriptase lacking RNASE H activity
US08/798,458 Expired - Lifetime US6063608A (en) 1988-01-13 1997-02-10 Cloned genes encoding reverse transcriptase lacking RNase H activity
US09/220,329 Expired - Fee Related US6589768B1 (en) 1988-01-13 1998-12-24 Cloned genes encoding reverse transcriptase lacking RNase H activity
US09/220,330 Expired - Fee Related US6610522B1 (en) 1988-01-13 1998-12-24 Cloned genes encoding reverse transcriptase lacking RNase H activity
US10/024,149 Abandoned US20030039988A1 (en) 1988-01-13 2001-12-21 Cloned genes encoding reverse transcriptase lacking RNase H activity

Family Applications After (3)

Application Number Title Priority Date Filing Date
US10/929,791 Abandoned US20050233315A1 (en) 1988-01-13 2004-08-31 Cloned genes encoding reverse transcriptase lacking RNase H activity
US10/970,280 Abandoned US20060003341A1 (en) 1988-01-13 2004-10-22 Cloned genes encoding reverse transcriptase lacking RNase H activity
US11/754,968 Abandoned US20080004437A1 (en) 1988-01-13 2007-05-29 Cloned Genes Encoding Reverse Transcriptase lacking RNase H Activity

Country Status (1)

Country Link
US (11) US5244797B1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040152072A1 (en) * 2002-07-30 2004-08-05 Invitrogen Corporation Reverse transcription
US20070020622A1 (en) * 2001-09-14 2007-01-25 Invitrogen Corporation DNA Polymerases and mutants thereof

Families Citing this family (128)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5244797B1 (en) 1988-01-13 1998-08-25 Life Technologies Inc Cloned genes encoding reverse transcriptase lacking rnase h activity
US7150982B2 (en) 1991-09-09 2006-12-19 Third Wave Technologies, Inc. RNA detection assays
US7045289B2 (en) * 1991-09-09 2006-05-16 Third Wave Technologies, Inc. Detection of RNA Sequences
US6759226B1 (en) * 2000-05-24 2004-07-06 Third Wave Technologies, Inc. Enzymes for the detection of specific nucleic acid sequences
US6551618B2 (en) 1994-03-15 2003-04-22 University Of Birmingham Compositions and methods for delivery of agents for neuronal regeneration and survival
US6593120B1 (en) 1994-04-01 2003-07-15 Gen-Probe Incorporated Recombinant DNA encoding a reverse transcriptase derived from moloney murine leukemia virus
US5795737A (en) * 1994-09-19 1998-08-18 The General Hospital Corporation High level expression of proteins
US5786464C1 (en) * 1994-09-19 2012-04-24 Gen Hospital Corp Overexpression of mammalian and viral proteins
US5853990A (en) * 1996-07-26 1998-12-29 Edward E. Winger Real time homogeneous nucleotide assay
US6114148C1 (en) * 1996-09-20 2012-05-01 Gen Hospital Corp High level expression of proteins
US6291164B1 (en) 1996-11-22 2001-09-18 Invitrogen Corporation Methods for preventing inhibition of nucleic acid synthesis by pyrophosphate
CA2281205A1 (en) * 1997-02-12 1998-08-13 Eugene Y. Chan Methods and products for analyzing polymers
ES2355739T3 (en) 1997-04-03 2011-03-30 Life Technologies Corporation COMPOSITIONS AND METHODS FOR CHAIN REACTION OF THE INVERSE TRANSCRIPTASE POLYMERASE (RT-PCR).
ATE445630T1 (en) * 1997-04-22 2009-10-15 Life Technologies Corp METHOD FOR PRODUCING ASLV REVERSER TRANSCRIPTASE COMPOSED OF MULTIPLE SUBUNITS
US6261770B1 (en) 1997-05-13 2001-07-17 Display Systems Biotech Aps Method to clone mRNAs
EP2390258A1 (en) 1997-08-29 2011-11-30 Life Technologies Corporation High fidelity polymerases and uses thereof
US7399589B2 (en) * 1998-02-06 2008-07-15 Digene Corporation Immunological detection of RNA:DNA hybrids on microarrays
US5994079A (en) 1998-02-06 1999-11-30 Digene Corporation Direct detection of RNA mediated by reverse transcriptase lacking RNAse H function
US6787305B1 (en) 1998-03-13 2004-09-07 Invitrogen Corporation Compositions and methods for enhanced synthesis of nucleic acid molecules
AU772847B2 (en) 1998-11-12 2004-05-06 Invitrogen Corporation Transfection reagents
AU2611700A (en) * 1999-01-15 2000-08-01 Molecular Biology Resources Biologically active reverse transcriptases
CA2363924A1 (en) 1999-03-02 2000-09-08 Invitrogen Corporation Compositions and methods for use in recombinational cloning of nucleic acids
US7074556B2 (en) * 1999-03-02 2006-07-11 Invitrogen Corporation cDNA synthesis improvements
US6630333B1 (en) * 1999-03-23 2003-10-07 Invitrogen Corporation Substantially pure reverse transriptases and methods of production thereof
US7655443B1 (en) * 1999-05-07 2010-02-02 Siemens Healthcare Diagnostics, Inc. Nucleic acid sequencing with simultaneous quantitation
EP1061129A1 (en) * 1999-05-18 2000-12-20 Etablissement de Transfusion Sanguine de Lyon Infectious retroviruses from a leukemic dog cell line with extensive homologies to murine leukemia viruses
AU5968600A (en) * 1999-05-17 2000-12-05 Etablissement Francais Du Sang Infectious retroviruses from a leukemic dog cell line with extensive homologies to murine leukemia viruses
EP1185711A4 (en) * 1999-05-21 2002-09-18 Invitrogen Corp Compositions and methods for labeling of nucleic acid molecules
WO2000079009A2 (en) 1999-06-22 2000-12-28 Invitrogen Corporation Improved primers and methods for the detection and discrimination of nucleic acids
US6271004B1 (en) 1999-06-25 2001-08-07 Display Systems Biotech A/S Method for improved reverse transcription at high temperatures
US6830902B1 (en) * 1999-07-02 2004-12-14 Invitrogen Corporation Compositions and methods for enhanced sensitivity and specificity of nucleic acid synthesis
EP2210948A3 (en) 1999-12-10 2010-10-06 Life Technologies Corporation Use of multiple recombination sites with unique specificity in recombinational cloning
US7056716B2 (en) * 2000-03-15 2006-06-06 Invitrogen Corporation High fidelity reverse transcriptases and uses thereof
US7078208B2 (en) * 2000-05-26 2006-07-18 Invitrogen Corporation Thermostable reverse transcriptases and uses thereof
US9771565B2 (en) * 2000-05-26 2017-09-26 Life Technologies Corporation Thermostable reverse transcriptases and uses thereof
CN1430670A (en) * 2000-05-26 2003-07-16 茵维特罗根公司 Thermostable reverse transcriptases and uses thereof
US7198924B2 (en) 2000-12-11 2007-04-03 Invitrogen Corporation Methods and compositions for synthesis of nucleic acid molecules using multiple recognition sites
US7153672B1 (en) * 2000-08-30 2006-12-26 University Of Rochester Method of performing reverse transcription reaction using reverse transcriptase encoded by non-LTR retrotransposable element
DE10046960A1 (en) * 2000-09-22 2002-04-11 Roche Diagnostics Gmbh Process for the production of an active, heterodimeric AMW-RT in prokaryotic cells
WO2002044425A2 (en) 2000-12-01 2002-06-06 Visigen Biotechnologies, Inc. Enzymatic nucleic acid synthesis: compositions and methods for altering monomer incorporation fidelity
EP2105496B1 (en) 2000-12-08 2013-02-20 Life Technologies Corporation Methods and compositions for synthesis of nucleic acid molecules using multiple recognition sites
US20030165859A1 (en) * 2001-10-23 2003-09-04 Invitrogen Corporation Primers and methods for the detection and discrimination of nucleic acids
US20030211528A1 (en) * 2002-03-12 2003-11-13 Norman Iscove Exponential amplification of sub-picogram nucleic acid samples with retention of quantitative representation
AU2003240795A1 (en) * 2002-05-24 2003-12-12 Invitrogen Corporation Nested pcr employing degradable primers
US20040132133A1 (en) * 2002-07-08 2004-07-08 Invitrogen Corporation Methods and compositions for the production, identification and purification of fusion proteins
WO2004021986A2 (en) 2002-09-03 2004-03-18 Quanta Biosciences, Inc. Improved compositions and methods for cdna synthesis
JP2006522582A (en) * 2002-09-05 2006-10-05 インヴィトロジェン コーポレーション Compositions and methods for synthesizing nucleic acids
JP4634799B2 (en) * 2002-09-13 2011-02-16 ライフ テクノロジーズ コーポレーション Thermostable reverse transcriptase and use thereof
SE0202867D0 (en) * 2002-09-27 2002-09-27 Pyrosequencing Ab New sequencing method
US20040067492A1 (en) * 2002-10-04 2004-04-08 Allan Peng Reverse transcription on microarrays
US7560232B2 (en) * 2002-12-19 2009-07-14 Promega Corporation Methods of capturing, detecting and quantifying RNA:DNA hybrids and a modified RNase H useful therein
US7141398B2 (en) * 2002-12-30 2006-11-28 Rosetta Inpharmatics Llc Methods for recycling mRNA for linear cRNA amplification
US20040265870A1 (en) * 2003-04-09 2004-12-30 Invitrogen Corporation Methods of synthesizing and labeling nucleic acid molecules
AU2004235298A1 (en) * 2003-04-25 2004-11-11 Janssen Pharmaceutica N.V. Preservation of RNA in a biological sample
US7727752B2 (en) 2003-07-29 2010-06-01 Life Technologies Corporation Kinase and phosphatase assays
US20070009899A1 (en) * 2003-10-02 2007-01-11 Mounts William M Nucleic acid arrays for detecting gene expression in animal models of inflammatory diseases
US20050118625A1 (en) * 2003-10-02 2005-06-02 Mounts William M. Nucleic acid arrays for detecting gene expression associated with human osteoarthritis and human proteases
US20050095600A1 (en) * 2003-10-31 2005-05-05 Xiang Yu Methods of generating gene-specific probes for nucleic acid array detection
EP2287341B1 (en) 2003-12-01 2013-02-13 Life Technologies Corporation Nucleic acid molecules containing recombination sites and methods of using the same
US7595179B2 (en) 2004-04-19 2009-09-29 Applied Biosystems, Llc Recombinant reverse transcriptases
CA2575675A1 (en) 2004-07-30 2006-03-09 Adeza Biomedical Corporation Oncofetal fibronectin as a marker for disease and other conditions and methods for detection of oncofetal fibronectin
WO2006034009A2 (en) * 2004-09-20 2006-03-30 Sigma-Aldrich Co. Purification of biomolecules from contaminating intact nucleic acids
US20060105348A1 (en) * 2004-11-15 2006-05-18 Lee Jun E Compositions and methods for the detection and discrimination of nucleic acids
US20060275792A1 (en) * 2004-11-15 2006-12-07 Lee Jun E Enhancement of nucleic acid amplification using double-stranded DNA binding proteins
US9505846B2 (en) * 2005-01-28 2016-11-29 Life Technologies Corporation Multi-component inhibitors of nucleic acid polymerases
US20060292578A1 (en) 2005-06-28 2006-12-28 Weidong Zheng DNA polymerase blends and mutant DNA polymerases
US20070111219A1 (en) * 2005-11-17 2007-05-17 Minor James M Label integrity verification of chemical array data
US20070111218A1 (en) * 2005-11-17 2007-05-17 Ilsley Diane D Label integrity verification of chemical array data
US20070237716A1 (en) * 2005-11-30 2007-10-11 Mason Andrew L Compositions and methods for reverse transcriptase-polymerase chain reaction (rt-pcr) of human b-retrovirus
US20080145844A1 (en) * 2006-01-25 2008-06-19 Evrogen Joint Stock Company Methods of cDNA preparation
EP1970440A1 (en) * 2007-03-06 2008-09-17 Qiagen GmbH Polymerase stabilization by ionic detergents
GB0806562D0 (en) 2008-04-10 2008-05-14 Fermentas Uab Production of nucleic acid
EP2208785A1 (en) 2009-01-15 2010-07-21 Newbiotechnic, S.A. Methods and kits to generate miRNA- and smallRNA-expressing vectors, and its application to develop lentiviral expression libraries
WO2010085715A1 (en) * 2009-01-22 2010-07-29 Quanta Biosciences Method for enrichment of selected rna molecules
EP2392680A1 (en) 2010-06-04 2011-12-07 Labor Diagnostik GmbH Leipzig Method for specific detection of classical swine fever virus
WO2011163120A1 (en) 2010-06-21 2011-12-29 Life Technologies Corporation Compositions, methods and kits for nucleic acid synthesis and amplification
US9410194B2 (en) 2010-06-21 2016-08-09 Life Technologies Corporation Compositions, kits and methods for synthesis and/or detection of nucleic acids
US20120058481A1 (en) 2010-08-20 2012-03-08 Life Technologies Corporation Quantitative Real Time PCR Assay Using FRET Dual-Labeled Primers
EP2492279A1 (en) 2011-02-25 2012-08-29 Laboratorios Del. Dr. Esteve, S.A. Rapid immunogen selection method using lentiviral display
JP6029580B2 (en) 2011-04-18 2016-11-24 株式会社トクヤマ Photochromic composition and optical article using the composition
EP2698377A1 (en) 2012-08-17 2014-02-19 Laboratorios Del. Dr. Esteve, S.A. Enhanced rapid immunogen selection method for HIV gp120 variants
US9416405B2 (en) 2012-11-02 2016-08-16 Life Technologies Corporation Compositions, methods and kits for enhancing PCR specificity
WO2014100755A2 (en) 2012-12-20 2014-06-26 Biomatrica, Inc. Formulations and methods for stabilizing pcr reagents
CN105339505B (en) 2013-03-12 2021-08-10 生命技术公司 Universal reporter-based genotyping methods and materials
EP2805769A1 (en) 2013-05-24 2014-11-26 European Molecular Biology Laboratory Methods for nano-scale single cell analysis
US9163284B2 (en) 2013-08-09 2015-10-20 President And Fellows Of Harvard College Methods for identifying a target site of a Cas9 nuclease
US9340800B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College Extended DNA-sensing GRNAS
JP2017503521A (en) 2014-01-22 2017-02-02 ライフ テクノロジーズ コーポレーション Novel reverse transcriptase for use in high temperature nucleic acid synthesis
EP3169310A1 (en) 2014-07-15 2017-05-24 Life Technologies Corporation Compositions with lipid aggregates and methods for efficient delivery of molecules to cells
US10077453B2 (en) 2014-07-30 2018-09-18 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
ES2890776T3 (en) 2015-03-13 2022-01-24 Life Technologies Corp Methods, compositions and kits for the capture, detection and quantification of small RNA
WO2017070632A2 (en) 2015-10-23 2017-04-27 President And Fellows Of Harvard College Nucleobase editors and uses thereof
RU2757416C2 (en) 2016-04-06 2021-10-15 Лайф Текнолоджис Корпорейшн Compositions, methods and sets for synthesizing and detecting nucleic acids
US11046940B2 (en) 2016-04-12 2021-06-29 Solis Biodyne Ou Synthetic reverse transcriptases and uses thereof
CN109477150B (en) 2016-06-16 2023-03-31 生命技术公司 Compositions, methods and kits for detecting microorganisms
US11155857B2 (en) 2016-06-27 2021-10-26 Dana-Farber Cancer Institute, Inc. Methods for measuring RNA translation rates
WO2018027078A1 (en) 2016-08-03 2018-02-08 President And Fellows Of Harard College Adenosine nucleobase editors and uses thereof
CA3033327A1 (en) 2016-08-09 2018-02-15 President And Fellows Of Harvard College Programmable cas9-recombinase fusion proteins and uses thereof
WO2018039438A1 (en) 2016-08-24 2018-03-01 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
US11613777B2 (en) 2016-08-26 2023-03-28 Life Technologies Corporation Nucleic acid extraction and amplification controls and methods of use thereof
GB2573062A (en) 2016-10-14 2019-10-23 Harvard College AAV delivery of nucleobase editors
WO2018119359A1 (en) 2016-12-23 2018-06-28 President And Fellows Of Harvard College Editing of ccr5 receptor gene to protect against hiv infection
US10988762B2 (en) 2017-02-16 2021-04-27 Bio-Rad Laboratories, Inc. Reverse transcriptases and uses thereof
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
WO2018165629A1 (en) 2017-03-10 2018-09-13 President And Fellows Of Harvard College Cytosine to guanine base editor
KR20190130613A (en) 2017-03-23 2019-11-22 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Nucleobase edits comprising nucleic acid programmable DNA binding proteins
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
CN111801345A (en) 2017-07-28 2020-10-20 哈佛大学的校长及成员们 Methods and compositions using an evolved base editor for Phage Assisted Continuous Evolution (PACE)
WO2019139645A2 (en) 2017-08-30 2019-07-18 President And Fellows Of Harvard College High efficiency base editors comprising gam
WO2019079347A1 (en) 2017-10-16 2019-04-25 The Broad Institute, Inc. Uses of adenosine base editors
JP2021502825A (en) 2017-11-13 2021-02-04 ライフ テクノロジーズ コーポレイション Compositions, methods, and kits for detecting urethral microbes
CN112770776A (en) * 2018-07-30 2021-05-07 瑞德库尔有限责任公司 Method and system for sample processing or analysis
KR20210143230A (en) 2019-03-19 2021-11-26 더 브로드 인스티튜트, 인코퍼레이티드 Methods and compositions for editing nucleotide sequences
WO2021023853A1 (en) 2019-08-08 2021-02-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Rna sequencing method for the analysis of b and t cell transcriptome in phenotypically defined b and t cell subsets
WO2021072328A1 (en) 2019-10-10 2021-04-15 The Broad Institute, Inc. Methods and compositions for prime editing rna
CN116096873A (en) 2020-05-08 2023-05-09 布罗德研究所股份有限公司 Methods and compositions for editing two strands of a target double-stranded nucleotide sequence simultaneously
WO2022067130A2 (en) 2020-09-24 2022-03-31 The Broad Institute, Inc. Prime editing guide rnas, compositions thereof, and methods of using the same
CA3203876A1 (en) 2021-01-11 2022-07-14 David R. Liu Prime editor variants, constructs, and methods for enhancing prime editing efficiency and precision
WO2022265965A1 (en) 2021-06-14 2022-12-22 10X Genomics, Inc. Reverse transcriptase variants for improved performance
AU2022305653A1 (en) 2021-06-30 2024-01-18 Dana-Farber Cancer Institute, Inc. Compositions and methods for enrichment of nucleic acids using light-mediated cross-linking
WO2023015309A2 (en) 2021-08-06 2023-02-09 The Broad Institute, Inc. Improved prime editors and methods of use
WO2023076898A1 (en) 2021-10-25 2023-05-04 The Broad Institute, Inc. Methods and compositions for editing a genome with prime editing and a recombinase
WO2023102459A1 (en) 2021-12-03 2023-06-08 Medicinal Genomics Corporation Psilocybe assay
WO2023102538A1 (en) 2021-12-03 2023-06-08 The Broad Institute, Inc. Self-assembling virus-like particles for delivery of prime editors and methods of making and using same
WO2023205687A1 (en) 2022-04-20 2023-10-26 The Broad Institute, Inc. Improved prime editing methods and compositions
WO2023240137A1 (en) 2022-06-08 2023-12-14 The Board Institute, Inc. Evolved cas14a1 variants, compositions, and methods of making and using same in genome editing
WO2024059493A1 (en) 2022-09-13 2024-03-21 Medicinal Genomics Corporation Psilocybe assay
WO2024077267A1 (en) 2022-10-07 2024-04-11 The Broad Institute, Inc. Prime editing methods and compositions for treating triplet repeat disorders

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5017488A (en) * 1986-04-01 1991-05-21 University Of Medicine And Dentistry Of New Jersey Highly efficient dual T7/T3 promoter vector PJKF16 and dual SP6/T3 promoter vector PJFK15
US5405776A (en) * 1988-01-13 1995-04-11 Life Technologies, Inc. Cloned genes encoding reverse transcriptase lacking RNase H activity
US5935833A (en) * 1994-04-01 1999-08-10 Gen-Probe Incorporated Highly-purified recombinant reverse transcriptase
US20020090618A1 (en) * 2000-05-26 2002-07-11 Invitrogen Corporation Thermostable reverse transcriptases and uses thereof
US20030003452A1 (en) * 2000-03-15 2003-01-02 Invitrogen Corporation High fidelity reverse transcriptases and uses thereof
US6610552B2 (en) * 2000-03-31 2003-08-26 Seiko Epson Corporation Method of manufacturing organic EL element

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4663290A (en) * 1982-01-21 1987-05-05 Molecular Genetics, Inc. Production of reverse transcriptase
US4943531A (en) * 1985-05-06 1990-07-24 The Trustees Of Columbia University In The City Of New York Expression of enzymatically active reverse transcriptase
US5017492A (en) * 1986-02-27 1991-05-21 Life Technologies, Inc. Reverse transcriptase and method for its production
US4795699A (en) * 1987-01-14 1989-01-03 President And Fellows Of Harvard College T7 DNA polymerase
ATE445630T1 (en) * 1997-04-22 2009-10-15 Life Technologies Corp METHOD FOR PRODUCING ASLV REVERSER TRANSCRIPTASE COMPOSED OF MULTIPLE SUBUNITS
US6399334B1 (en) * 1997-09-24 2002-06-04 Invitrogen Corporation Normalized nucleic acid libraries and methods of production thereof
US6495358B1 (en) * 2000-04-19 2002-12-17 Wichita State University Sulfamide and bis-sulfamide amino acid derivatives as inhibitors of proteolytic enzymes

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5017488A (en) * 1986-04-01 1991-05-21 University Of Medicine And Dentistry Of New Jersey Highly efficient dual T7/T3 promoter vector PJKF16 and dual SP6/T3 promoter vector PJFK15
US5405776A (en) * 1988-01-13 1995-04-11 Life Technologies, Inc. Cloned genes encoding reverse transcriptase lacking RNase H activity
US5405776B1 (en) * 1988-01-13 1996-10-01 Life Technologies Inc Cloned genes encoding reverse transcriptase lacking RNase H activity
US5668005A (en) * 1988-01-13 1997-09-16 Life Technologies, Inc. Cloned genes encoding reverse transcriptase lacking RNASE H activity
US6063608A (en) * 1988-01-13 2000-05-16 Life Technologies, Inc. Cloned genes encoding reverse transcriptase lacking RNase H activity
US6589768B1 (en) * 1988-01-13 2003-07-08 Invitrogen Corporation Cloned genes encoding reverse transcriptase lacking RNase H activity
US5935833A (en) * 1994-04-01 1999-08-10 Gen-Probe Incorporated Highly-purified recombinant reverse transcriptase
US20030003452A1 (en) * 2000-03-15 2003-01-02 Invitrogen Corporation High fidelity reverse transcriptases and uses thereof
US6610552B2 (en) * 2000-03-31 2003-08-26 Seiko Epson Corporation Method of manufacturing organic EL element
US20020090618A1 (en) * 2000-05-26 2002-07-11 Invitrogen Corporation Thermostable reverse transcriptases and uses thereof

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070020622A1 (en) * 2001-09-14 2007-01-25 Invitrogen Corporation DNA Polymerases and mutants thereof
US20040152072A1 (en) * 2002-07-30 2004-08-05 Invitrogen Corporation Reverse transcription

Also Published As

Publication number Publication date
US6589768B1 (en) 2003-07-08
US5244797A (en) 1993-09-14
US20080004437A1 (en) 2008-01-03
US5405776B1 (en) 1996-10-01
US5244797B1 (en) 1998-08-25
US5405776A (en) 1995-04-11
US6063608A (en) 2000-05-16
US6610522B1 (en) 2003-08-26
US20060003341A1 (en) 2006-01-05
US20030039988A1 (en) 2003-02-27
US20050233315A1 (en) 2005-10-20
US5668005A (en) 1997-09-16

Similar Documents

Publication Publication Date Title
US6610522B1 (en) Cloned genes encoding reverse transcriptase lacking RNase H activity
Kotewicz et al. Isolation of cloned Moloney murine leukemia virus reverse transcriptase lacking ribonuclease H activity
US5939301A (en) Cloned DNA polymerases from Thermotoga neapolitana and mutants thereof
US5270179A (en) Cloning and expression of T5 DNA polymerase reduced in 3'- to-5' exonuclease activity
US6794177B2 (en) Modified DNA-polymerase from carboxydothermus hydrogenoformans and its use for coupled reverse transcription and polymerase chain reaction
US5545552A (en) Purified thermostable pyrococcus furiosus DNA polymerase I
US7045328B2 (en) Purified thermostable Pyrococcus furiosus DNA polymerase I
EP0386859B1 (en) T7 DNA polymerase
US7417133B2 (en) Methods for obtaining thermostable enzymes, DNA polymerase I variants from Thermus aquaticus having new catalytic activities, methods for obtaining the same, and applications of the same
US20050282171A1 (en) Purified thermostable Pyrococcus furiosus DNA polymerase I
US4946786A (en) T7 DNA polymerase
EP0416755A1 (en) Cloning and expression of T5 DNA polymerase reduced in 3'-to-5' exonuclease activity
US5866395A (en) Purified thermostable pyrococcus furiosus DNA polymerase I
US20120184017A1 (en) Mutant dna polymerases and uses therof
US5266466A (en) Method of using T7 DNA polymerase to label the 3' end of a DNA molecule
US9593318B2 (en) Linker-bridged gene or domain fusion reverse transcriptase enzyme
US5145776A (en) Method of using T7 DNA polymerase to mutagenize and fill-in DNA
US5514574A (en) Method of producing flavine nucleotides
EP0922765B1 (en) Modified DNA-polymerase from carboxydothermus hydrogenoformans and its use for coupled reverse transcription and polymerase chain reaction
US5256554A (en) Expression of human immunodeficiency virus (HIV) reverse transcriptase
CA1335263C (en) T7 dna polymerase
IL101061A (en) Genes encoding T7 dna polymerase

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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