US20030166281A1 - Methods of discovering chemicals capable of functioning as gene expression modulators - Google Patents

Methods of discovering chemicals capable of functioning as gene expression modulators Download PDF

Info

Publication number
US20030166281A1
US20030166281A1 US10/124,113 US12411302A US2003166281A1 US 20030166281 A1 US20030166281 A1 US 20030166281A1 US 12411302 A US12411302 A US 12411302A US 2003166281 A1 US2003166281 A1 US 2003166281A1
Authority
US
United States
Prior art keywords
cell
csf
expression
gene
molecule
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
US10/124,113
Inventor
J. Foulkes
Franz Leichtfried
Christian Pieler
John Stephenson
Robert Franco
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.)
OSI Pharmaceuticals LLC
Original Assignee
OSI Pharmaceuticals LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by OSI Pharmaceuticals LLC filed Critical OSI Pharmaceuticals LLC
Priority to US10/124,113 priority Critical patent/US20030166281A1/en
Publication of US20030166281A1 publication Critical patent/US20030166281A1/en
Priority to US11/704,094 priority patent/US20080009060A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/775Apolipopeptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/67General methods for enhancing the expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6897Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids involving reporter genes operably linked to promoters
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds
    • 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
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the expression of a specific gene can be regulated at any step in the process of producing an active protein. Modulation of total protein activity may occur via transcriptional, transcript-processing, translational or post-translational mechanisms. Transcription may be modulated by altering the rate of transcriptional initiation or the progression of RNA polymerase (28). Transcript-processing may be influenced by circumstances such as the pattern of RNA splicing, the rate of mRNA transport to the cytoplasm or mRNA stability.
  • This invention concerns the use of molecules which act by modulating the in vivo concentration of their target proteins via regulating gene transcription. The functional properties of these chemicals are distinct from previously described molecules which also affect gene transcription.
  • the eucaryotic transcriptional unit is much more complex than its procaryotic counterpart and consists of additional elements which are not found in bacteria.
  • Eucaryotic transcriptional units include enhancers and other cis-acting DNA sequences (30, 19). Procaryotic transcription factors most commonly exhibit a “helix-turn-helix” motif in the DNA binding domain of the protein (29, 37). Eucaryotic transcriptional factors frequently contain a “zinc finger” (37, 12) or a “leucine zipper” (24) in addition to sometimes possessing the “helix-turn-helix” motif (26). Furthermore, several critical mechanisms at the post-transcriptional level such as RNA splicing and polyadenylation are not found in procaryotic systems (21, 35).
  • modulation of gene transcription in response to extracellular factors can be regulated in both a temporal and tissue specific manner (22).
  • extracellular factors can exert their effects by directly or indirectly activating or inhibiting transcription factors (22, 28).
  • Modulators of transcription factors involved in direct regulation of gene expression have been described, and include those extracellular chemicals entering the cell passively and binding with high affinity to their receptor-transcription factors.
  • This class of direct transcriptional modulators include steroid hormones and their analogs, thyroid hormones, retinoic acid, vitamin D 3 and its derivatives, and dioxins, a chemical family of polycyclic aromatic hydrocarbons (12, 38, 9).
  • Dioxins are molecules generally known to modulate transcription, however, dioxins bind to naturally-occurring receptors which respond normally to xenobiotic agents via transcriptionally activating the expression of cytochrome P450, part of an enzyme involved in detoxification. Similarly, plants also have naturally occurring receptors to xenobiotics to induce defense pathways. For example, the fungal pathogen Phytophthora megasperma induces an anti-fungal compound in soybeans. Such molecules which bind to the ligand binding domains of such naturally occurring receptors are not included on the scope of this invention.
  • Indirect transcriptional regulation involves one or more signal transduction mechanisms.
  • the regulation typically involves interaction with a receptor, the receptor being part of a multistep intracellular signaling pathway, the pathway ultimately modulating the activity of nuclear transcription factors.
  • This class of indirect transcriptional modulators include polypeptide growth factors such as platelet-derived growth factor, epidermal growth factor, cyclic nucleotide analogs, and mitogenic tumor promoters (18, 1, 2).
  • nucleotide analogs in methods to modulate transcription. The mechanism involves incorporating nucleotide analogs into nascent mRNA or non-specifically blocking mRNA synthesis. Similarly, researchers have used alkylating agents, e.g. cyclophosphamide, or intercalating agents, e.g. doxorubicin, to non-specifically inhibit transcription.
  • alkylating agents e.g. cyclophosphamide
  • intercalating agents e.g. doxorubicin
  • Signal effector type molecules such as cyclic AMP, diacylglycerol, and their analogs are known to non-specifically regulate transcription by acting as part of a multistep protein kinase cascade reaction. These signal effector type molecules bind to domains on proteins which are thus subject to normal physiological regulation by low molecular weight ligands (10, 39).
  • PCT/US88/10095 The specific use of sterol regulatory elements from the LDL receptor gene to control expression of a reporter gene has recently been documented in PCT/US88/10095.
  • One aspect of PCT/US88/10095 deals with the use of specific sterol regulatory elements coupled to a reporter as a means to screen for drugs capable of stimulating cells to synthesize the LDL receptor.
  • PCT/US88/10095 describes neither the concept of simultaneously screening large numbers of chemicals against multiple target genes nor the existence of transcriptional modulators which (a) do not naturally occur in the cell, (b) specifically transcriptionally modulate expression of the gene-of-interest, and (c) bind to DNA or RNA or bind to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with the defined physiological effect.
  • the main focus of PCT/US88/10095 is the use of the sterol regulatory elements from the LDL receptor as a means to inhibit expression of toxic recombinant biologicals.
  • reporter gene to analyze nucleotide sequences which regulate transcription of a gene-of-interest.
  • the demonstrated utility of a reporter gene is in its ability to define domains of transcriptional regulatory elements of a gene-of-interest.
  • Reporter genes which express proteins, e.g. luciferase are widely utilized in such studies. Luciferases expressed by the North American firefly, Photinus pyralis and the bacterium, Vibrio fischeri were first described as transcriptional reporters in 1985 (8, 11)
  • a method to define domains of transcriptional regulating elements of a gene-of-interest typically has also involved use of phorbol esters, cyclic nucleotide analogs, concanavalin A, or steroids, molecules which are commonly known as transcriptional modulators.
  • phorbol esters cyclic nucleotide analogs
  • concanavalin A or steroids
  • transcriptional modulators molecules which are commonly known as transcriptional modulators.
  • available literature shows that researchers have not considered using a transcription screen to identify specific transcriptional modulators. Apparently, success would be unlikely in doing so, however, we have demonstrated herein that this is not the case.
  • molecules specifically regulating the activity of one member of a group of closely related proteins are difficult to produce.
  • Molecules, structurally related at the protein level may possess distinct regulatory elements at the DNA level which control their expression.
  • molecules such as the chemical transcriptional modulators defined herein can provide a greater opportunity for specifically modulating the activity of structurally related proteins.
  • One example is the ras oncogene family, where the H-, N- and K-ras proteins are highly related but wherein the three genes have distinct structures.
  • the molecules described herein may also serve to mimic normal physiological response mechanisms, typically involving the coordinated expression of one or more groups of functionally related genes. Therefore, determining whether a molecule can specifically transcriptionally modulate the expression of a gene-of-interest and the ultimate clinical use of the molecule provides a therapeutic advantage over the use of single recombinant biologicals, or drugs which bind directly to the final target protein encoded by the gene-of-interest.
  • the present invention provides a method of transcriptionally modulating the expression of a gene-of-interest, the expression of which is associated with a defined physiological or pathological effect within a multicellular organism.
  • the method comprises contacting a cell which is capable of expressing the gene with an amount of a molecule effective to transcriptionally modulate expression of the gene and thereby affect the level of the protein encoded by the gene which is expressed by the cell.
  • Molecules useful in the practice of the invention are characterized as follows (a) do not naturally occur in the cell, (b) specifically transcriptionally modulate expression of the gene-of-interest, and (c) bind to DNA or RNA or bind to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with the defined physiological or pathological effect.
  • this invention provides a method of determining whether a molecule, not previously known to be a modulator of protein biosynthesis, is capable of transcriptionally modulating the expression of a gene-of-interest.
  • the method comprises contacting a sample which contains a predefined number of cells with a predetermined amount of a molecule to be tested.
  • Each such cell comprises DNA consisting essentially of (i) a modulatable transcriptional regulatory sequence of the gene-of-interest, (ii) a promoter of the gene-of-interest, and (iii) a reporter gene which expresses a polypeptide capable of producing a detectable signal, coupled to, and under the control of, the promoter, and the contacting is carried out under conditions such that the molecule, if capable of acting as a transcriptional modulator of the gene-of-interest, causes a measurable detectable signal to be produced by the polypeptide expressed by the reporter gene and the amount of the signal produced may be quantitatively determined.
  • the amount of produced signal so determined is compared with the amount of produced signal detected in the absence of any molecule being tested or upon contacting the sample with any other molecule so as to thereby identify the molecule as one which causes a change in the detectable signal produced by the polypeptide expressed by the reporter gene and thus identify the molecule as a molecule capable of transcriptionally modulating the expression of the gene-of-interest.
  • the present invention still further provides a method for transcriptionally modulating in a multicellular organism the expression of a gene-of-interest, the expression of which is associated with a defined physiological or pathological effect in the organism.
  • the method comprises administering to the organism an amount of a molecule effective to transcriptionally modulate expression of the gene and thus affect the defined physiological or pathological effect.
  • a molecule useful in the method (a) does not naturally occur in the organism, (b) specifically transcriptionally modulates expression of the gene-of-interest, and (c) binds to DNA or RNA or binds to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the organism, the binding of a ligand to which ligand binding domain is normally associated with the defined physiological or pathological effect.
  • This invention provides a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • this invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising administering to the human being an amount of a molecule having the structure:
  • the invention also provides a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising contacting the cell with an amount of a molecule having the structure:
  • Another provision of the present invention concerns a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone.
  • the method comprises administering to the human being an amount of a molecule having the structure:
  • This invention also concerns a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising contacting the cell with an amount of a molecule having the structure:
  • this invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising administering to the human being an amount of a molecule having the structure:
  • this invention concerns a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising contacting the cell with an amount of a molecule having the structure:
  • the invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone.
  • the method comprises administering to the human being an amount of a molecule having the structure:
  • the invention provides another method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising administering to the human being an amount of a molecule having the structure:
  • this invention provides a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • this invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone.
  • the method comprises administering to the human being an amount of a molecule having the structure:
  • the present invention provides a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the present invention also provides a method for increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises administering to the human being an amount of a molecule having the structure:
  • the present invention also provides a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the invention provides a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises administering to the human being an amount of a molecule having the structure:
  • the invention provides another method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the present invention provides another method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises administering to the human being an amount of a molecule having the structure:
  • the invention also concerns a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF comprising contacting the cell with an amount of a molecule having the structure:
  • the invention concerns a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • This invention further concerns a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the invention provides a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • the present invention concerns a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
  • the invention further concerns a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF wherein the method comprises administering to the human being an amount of a molecule having the structure:
  • the invention provides a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the invention additionally concerns a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, wherein the method comprises administering to the human being an amount of a molecule having the structure:
  • Another provision of the present invention is a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the invention provides a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF.
  • the method comprises administering to the human being an amount of a molecule having the structure:
  • the present invention provides another method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the present invention concerns a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF.
  • the method comprises administering to the human being an amount of a molecule having the structure:
  • This invention also concerns a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • this invention provides a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • the invention also concerns a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • this invention provides a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • the invention further provides a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • this invention provides a method of decreasing the formation of neutrophils and effecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • the present invention a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • this invention provides a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • the present invention further provides a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the invention provides a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • the present invention also concerns a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the present invention concerns a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • the present invention also concerns a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the invention concerns a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • the invention provides a method of decreasing the expression of a mammary tumor virus by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, a mammary tumor virus.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the invention also provides a method of suppressing the proliferation of a mammary tumor virus in a subject who (i) comprises DNA encoding, and (ii) is capable of expressing, a mammary tumor virus, comprising administering to the subject an amount of a molecule having the structure:
  • FIG. 1 is a partial restriction enzyme cleavage map of the plasmid pD0432 which contains the luciferase gene from the firefly, Photinus pyralis.
  • FIG. 2 is a partial restriction enzyme cleavage map of the plasmid pSVLuci which contains the luciferase gene from the firefly, Photinus pyralis.
  • FIG. 3 is a partial restriction enzyme cleavage map of the plasmid pMLuci which contains the luciferase gene of the firefly, Photinus pyralis and the mouse mammary tumor virus long terminal repeat.
  • FIG. 4 is a partial restriction enzyme cleavage map of the plasmid pUXLuci which contains the luciferase gene from the firefly, Photinus pyralis.
  • FIG. 5 is a partial restriction enzyme cleavage map of the plasmid phGH:CAT which contains the CAT gene and human growth hormone promoter sequences.
  • FIG. 6 is a partial restriction enzyme cleavage map of the plasmid phGH-Luci which contains the luciferase gene from the firefly, Photinus pyralis and human growth hormone promoter sequences.
  • FIG. 7 is a partial restriction enzyme cleavage map of the plasmid pJM710 which contains G-CSF upstream sequences.
  • FIG. 8 is a partial restriction enzyme cleavage map of the plasmid pGEM5-Luci which contains the luciferase gene from the firefly, Photinus pyralis.
  • FIG. 9 is a partial restriction enzyme cleavage map of the plasmid PG-Luc 1 which contains both the luciferase gene from the firefly, Photinus pyralis, and G-CSF upstream sequences.
  • FIG. 10 is a view of the mammalian expression shuttle vector pUV102 with its features.
  • the mammalian expression shuttle vector was designed to allow the construction of the promoter-reporter gene fusions and the insertion of a neomycin resistance gene coupled to the herpes simplex virus thymidine kinase promoter (TK-NEO).
  • TK-NEO herpes simplex virus thymidine kinase promoter
  • FIG. 11 a provides the nucleotide sequences of six oligonucleotides, pUV-1 through pUV-6 (SEQ ID NOS: 25-30), which were annealed, ligated, and inserted into the SalI/EcoR1 sites of the plasmid pTZ18R.
  • FIG. 11 b is a diagrammatic representation of the construction of the plasmid pUV001 from the plasmids pTZ18R and pBluescript KS(+).
  • FIG. 12 is a diagrammatic representation of the construction of the plasmid pUV100 from the plasmid pUV001 and two DNA fragments, the XbaI/XmaI fragment from pMLuci and the XmaI/BamHI fragment from pMSG.
  • FIG. 13 is a diagrammatic representation of the construction of the plasmid pUV100-3 from the plasmid pUV100 and a 476 b fragment containing a dimeric SV40 polyadenylation site.
  • FIG. 14 is a diagrammatic representation of the construction of the plasmids pUV102 and pUV103 from the plasmid pUV100-3 and D-link oligonucleotides and the plasmid pUV100-3 and R-link oligonucleotides, respectively.
  • FIG. 15 provides the nucleotide sequences of oligos (SEQ ID NOS: 31-34) used for the construction of a synthetic HSV-Thymidine Kinase promoter and provides a diagrammatic representation of the HSV-TK promoter.
  • FIG. 16 is a diagrammatic representation of the construction of the plasmid pTKL100 which contains the luciferase gene from the firefly, Photinus pyralis and the HSV-TK promoter sequence.
  • FIG. 17 is a diagrammatic representation of the construction of the plasmid pTKNEO which contains the neo gene, from about 3.5 kb NheI/XmaI fragment from pTKL100, and the about 0.9 kb BstBI/BglII fragment containing the neo coding region from pRSVNEO.
  • FIG. 18 is a diagrammatic representation of the construction of the plasmid pTKNEO2 from the plasmid pTKNEO and the oligonucleotides Neo 1 and 2 (SEQ ID NOS: 35 and 36).
  • FIG. 19 is a diagrammatic representation of the construction of the plasmid pTKNEO3 from the plasmid PTKNEO2 and about 0.9 kb EcoR1/SalI fragment from pMC1NEO.
  • FIG. 20 is a partial restriction enzyme cleavage map of a human genomic clone which contains the entire GM-CSF coding region.
  • FIG. 21 provides the nucleotide sequence of oligonucleotides 1 through 4 (SEQ ID NOS: 37-40) and provides a diagrammatic representation of GM-CSF upstream sequences fused with the ATG of the coding region of the luciferase gene from the firefly, Phoxinus pyralis.
  • FIG. 22 is a diagrammatic representation of the construction of the plasmids pGMLS102 and pGMLS103 from plasmid pUV 102 and a 0.7 kb fragment from pGM-2 and from pUV 103 and a 0.7 kb fragment from pGM-2, respectively.
  • FIG. 23 is a diagrammatic representation of the construction of plasmids pGMLL102 and pGMLL103 from the plasmid pGMLS102 and the GM-CSF clone and the plasmid pGMLS103 and the GM-CSF clone, respectively.
  • FIG. 24 is a diagrammatic representation of the construction of the plasmid CSF1-pTZ18 from the plasmid pTZ18R and a gene fragment comprising the first exon and 5′ flanking region of M-CSF.
  • FIG. 25 is a diagrammatic representation of the construction of the plasmid p100-RH from the plasmid pUV100 and the gene fragment comprising the first exon and 5′ flanking region of M-CSF from the plasmid CSF1-pTZ18.
  • FIG. 26 is a diagrammatic representation of the construction of the plasmid p100-2 by insertion of a PSTI/PVUII fragment from CSFI-pTZ18 and synthetic oligonucleotides into the plasmid pUV100.
  • FIG. 27 is a diagrammatic representation of the construction of p100-RHC by insertion of short M-CSF 5′ upstream sequences fused to the luciferase coding sequence into the plasmid p100-RH containing a longer M-CSF 5′ upstream fragment.
  • FIG. 28 is a diagrammatic representation of the construction of pCSF1-102 from a 5 kb NotI/Xba I fragment isolated from p100-RHC and inserted into pUV102.
  • FIG. 29 is a diagrammatic representation of the construction of pGUC84 from oligonucleotides containing the G-CSF leader sequence from +15 to the ATG cloned into the plasmid pUC19.
  • FIG. 30 is a diagrammatic representation of the construction of pGUV100 from the NcoI/ScaI fragment from pGUC84 containing G-CSF leader sequences cloned into the plasmid pUV100.
  • FIG. 31 is a diagrammatic representation of the construction of pGUV1 from the Pst I fragment of the G-CSF promoter from the plasmid pJM710 inserted into the Pst I site in PGUV 100.
  • FIG. 32 is a diagrammatic representation of the construction of the plasmid pGUV-2 by insertion of more G-CSF upstream sequences from pJM710 into pGUV1.
  • FIG. 33 is a diagrammatic representation of the construction of the plasmid pGUV140 from the XbaI fragment from pGUV2 containing the G-CSF-luciferase fusion and the plasmid pUV103.
  • FIG. 34 is a diagrammatic representation of the construction of the plasmid pGUV150 from the ScaI/XbaI fragment from pGUV140 which contains the G-CSF-luciferase fusion and the plasmid pTKNEO-103.
  • FIG. 35 is a diagrammatic representation of the construction of the plasmid pEP-7.5B from a 7.5 kb BamH1 fragment consisting of 6.2 kb of the EPO promoter region and the first three EPO exons and the plasmid pBluescript KS(+).
  • FIG. 36 is a diagrammatic representation of the construction of the plasmid pRE from oligonucleotides EPO 9-12 and the plasmid pUV100.
  • FIG. 37 is a diagrammatic representation of the construction of the plasmid pUV2-EP6 from a 6 kb fragment of EPO upstream sequences and the plasmid pUV 102.
  • FIG. 38 is a diagrammatic representation of the construction of the plasmid IL-3-pTZ18R from IL-3 upstream sequences and the plasmid pTZ18R.
  • FIG. 39 is a diagrammatic representation of the construction of the plasmid pTZ-IL-3-Luci from oligonucleotides IL-lu-1 to IL-lu-4, pUV-3 and pUV-6 and the plasmid IL3/PstI-pTZ18R.
  • FIG. 40 is a diagrammatic representation of the construction of the plasmid pTZ-IL3-C from a 500 kb NcoI/EcoR1 fragment from pTZ-IL-3-Luci and the plasmid IL-3-pTZ18R.
  • FIG. 41 is a diagrammatic representation of the construction of the plasmid pUV-IL-3 from a 6.4 kb Hind III/XbaI fragment from pTZ-IL3-C and the plasmid pUV102.
  • FIG. 42 is a bar graph illustrating induction of luciferase expression in reporter cell lines containing MMTV (M10), human growth hormone (532), and human G-CSF (G21) promoter sequences, in response to known transcriptional inducers.
  • FIG. 43 is a bar graph illustrating the effect of steroids on luciferase expression in the MMTV reporter cell line M10.
  • FIG. 44 is a bar graph illustrating specific induction of luciferase expression in reporter cell lines for MMTV (M10), human growth hormone (532) and human G-CSF (G21) promoters in response to chemicals identified in a high throughput screen and known transcriptional inducers.
  • FIG. 45 is a bar graph illustrating specific inhibition of luciferase expression in reportor cell lines for MMTV (M10), human growth hormone (532), and human G-CSF (G21) in response to chemicals identified in a high throughput screen.
  • FIG. 46 is an autoradiograph of a Northern blot illustrating increased G-CSF mRNA production by the human epithelial cell line U5637 in response to chemicals #670 and #1255 and IFN-gamma as compared to the solvent DMSO. Reprobing with beta-actin was used to normalize for the amount of mRNA that had been loaded onto the gel.
  • FIG. 47 illustrates a dose response analysis of chemicals #80, #670, and #1780 using the G-CSF reporter cell line G21. The amount of luciferase expression is indicated in arbitrary units.
  • FIG. 48 illustrates the linear dependence of the bioluminescent signal (in arbitrary luminometer units) from the luciferase reporter concentration (1,280 fg luciferase correspond to 50 relative reporter concentration units).
  • FIG. 49 illustrates the time course of bioluminescent signal decay after addition of Actinomycin D to the cell clones G 1002, GM 1074 and CM 1. Time in hours is plotted against the logarithm of the ratio of the bioluminescent signal generated by Actinomycin D-treated cells over the signal of untreated control cells.
  • FIG. 50 is an autoradiograph of a Southern blot illustrating the correct integration of luciferase fusion constructs containing the G-CSF, M-CSF and GM-CSF promoters, respectively, into the genomes of 5637 cells (G- and GM-CSF) and HL 60 cells (M-CSF).
  • Lanes designated 5637 and HL 60 had been loaded with DNA preparations from the parental cell lines not containing the luciferase constructs (negative controls).
  • Lanes labeled G/5637, GM/5637 and M/HL 60 had been loaded with the same DNA preparations with the addition of the purified plasmid preparations, which had been used for the original transfections (positive controls).
  • the two low-molecular-weight bands appearing in almost all lanes except for the negative control lanes are derived from non-specific cross-hybridizing sequences contained in the probe.
  • FIG. 51 illustrates a dose response analysis of DMSO, methanol and ethanol using the reporter cell lines M 10, G 2005 and GM 1105. The level of luciferase expression of solvent-treated cells expressed in percent of that of untreated control cells is plotted against solvent concentration.
  • FIG. 52 is a summary of the number of lead samples obtained in a luciferase expression screen of Actinomyces fermentation broth samples for modulation of the G-CSF, GM-CSF and MMTV promoters.
  • TIR stands for “Transcriptional Induction/Inhibition Ratio” and represents the ratio of luciferase expression generated by a test sample treated cells over that of control cells treated by solvent alone.
  • FIG. 53 is a summary of those lead samples listed in FIG. 52, which specifically modulate one promoter only according to the TIR criteria indicated.
  • FIG. 54 is an autoradiograph of a polyacrylamide gel illustrating an S1 nuclease protection analysis of increased mRNA production by the human bladder carcinoma cell line 5637 in response to lead chemicals #542, #1255, #1793 and #1904.
  • RNA indicates the sources of the RNA preparations used in individual lanes.
  • Probe indicates the mRNA-specificities of probes used in individual lanes.
  • Compound lists the compounds with which the 5637 cells were treated prior to RNA extraction and loading on individual gel lanes (“Cyclo” means cycloheximide).
  • G, GM and A indicate the correct sizes of G-CSF-, GM-CSF- and Actin-specific nuclease-protected mRNA/Probe hybrids.
  • FIG. 55 is a bar graph illustrating increased G-CSF secretion by 5637 cells treated for 48 hours in serum-containing media with the samples indicated on the abscissa.
  • TNF-alpha was used at 5 ng/ml.
  • Chemicals #542 and #1780 were used at 50 uM or 1 uM and 0.2 uM final concentration, respectively. Both chemicals were used in DMSO at a final concentration of 0.5%.
  • the ordinate indicates the concentration of G-CSF secreted into 5 ml of serum-containing media by 25 square cm of confluent 5637 cells.
  • FIG. 56 illustrates a dose response analysis of chemical #542 using the G-CSF reporter cell line G 21 (solid line) and the MTT respiratory inhibition cytotoxicity assay (dotted line). Respiratory inhibition in percent of untreated control cells (Ordinate, left scale) and luciferase expression of #542-treated over solvent-treated cells (ordinate, right scale) are plotted against #542 concentration (abscissa).
  • the present invention provides a method of transcriptionally modulating the expression of a homologous gene-of-interest, the expression of which is associated with a defined physiological or pathological effect within a multicellular organism.
  • the method comprises contacting a cell which is capable of expressing the gene with an amount of a molecule effective to transcriptionally modulate the expression of the gene. Modulating the expression of the gene affects the level of the protein encoded by the gene which is expressed by the cell.
  • a homologous gene-of-interest means a gene which is naturally associated with an organism or virus encoding and expressing the gene. Therefore, this definition would not include a synthetic or recombinant gene constructed by genetic engineering methods so as to position the gene-of-interest under the control of a heterologous promoter.
  • physiological effect as used herein is defined as an effect characteristic of, or appropriate to, an organism's healthy or normal functioning.
  • pathological effect as used herein is defined as an effect altered or caused by disease.
  • Molecules useful in the practice of the invention are characterized as follows (a) do not naturally occur in the cell, (b) specifically transcriptionally modulate expression of the gene-of-interest, and (c) bind to DNA or RNA or bind to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with the defined physiological or pathological effect.
  • the molecule would not occur naturally in any cell of a higher eucaryotic organism. In another example of this invention the molecule would not occur naturally in any cell, e.g. minerals. In yet another example, the molecule would not occur naturally, e.g. synthetic molecules.
  • the phrase “specifically transcriptionally modulate expression of the gene-of-interest” as used herein means modulating the expression of the gene-of-interest without modulating the expression of other genes in the cell in a way which would cause an adverse effect on (a) an organism containing the cell in the case where the cell is within the organism or (b) the growth or the culturing of the cell, in the case where the cell is being grown or cultured to make a product where the amount of product produced is associated with expression of the gene-of-interest.
  • drug application is intended to cause an adverse effect on the cells of the parasite (which may contain the gene-of-interest), but not on the cells of the host organism.
  • a gene-of-interest may constitute a single gene or a limited number of genes whose expression can be functionally coordinated.
  • TGF- ⁇ family of polypeptides termed the physiological growth modulators termed the transforming growth factor- ⁇ family of polypeptides (TGF- ⁇ s) (90).
  • TGF- ⁇ s control the extracellular matrix (ECM) by; (1) increasing expression of the genes encoding ECM polypeptides, e.g. collagen, fibronectin and osteopontin; (2) increasing expression of receptors for the ECM, e.g. the integrins; and (3) increasing expression of protease inhibitors (e.g. TIMP and PAI-I) while decreasing expression of secreted proteases (e.g.
  • TGF- ⁇ s collagenase and stromelysin.
  • This coordinate regulation may allow TGF- ⁇ s to be useful in the repair of surface wounds, cartilage and bone.
  • transcriptionally modulate expression of a gene-of-interest means the effect upon transcription of the gene resulting from either (a) direct binding of the molecule to DNA or RNA, a DNA- or RNA-binding protein, and/or a DNA- or RNA-binding protein complex, or (b) direct binding of the molecule to a protein which directly chemically modifies a DNA- or RNA-binding protein or protein complex.
  • DNA- or RNA-binding protein or protein complex means to modify the protein or protein complex through a chemical reaction, including but not limited to, phosphorylation, glycosylation, methylation, acetylation, adenoribosylation, acylation, myristylation, reduction, oxidation, covalent oligomerization or polymerization or proteolytic cleavage.
  • the invention provides a cell capable of expressing the gene-of-interest, the expression of which is associated with a defined physiological or pathological effect within a multicellular organism.
  • the cell may be a human cell, an animal cell, a plant cell or any eucaryotic cell or procaryotic cell from whatever source.
  • the gene-of-interest whose expression is associated with a defined physiological or pathological effect within a multicellular organism may be a human gene.
  • the gene-of-interest may encode a hematopoietic protein.
  • Hematopoietic proteins may include, but are not limited to, colony stimulating factors and erythropoietin (EPO).
  • colony stimulating factors useful in the practice of this invention include, but are not limited to, granulocyte-macrophage colony stimulating factor (GM-CSF), granulocyte colony stimulating factor (G-CSF), and macrophage colony stimulating factor (M-CSF).
  • GM-CSF granulocyte-macrophage colony stimulating factor
  • G-CSF granulocyte colony stimulating factor
  • M-CSF macrophage colony stimulating factor
  • the gene-of-interest of the invention may encode an interleukin (IL) or a cytokine, or a growth modulating factor.
  • IL interleukin
  • a growth modulating factor would be a member of the transforming growth factor- ⁇ (TGF- ⁇ ) family i.e. TGF- ⁇ 1 or TGF- ⁇ 2 or TGF- ⁇ 3.
  • TGF- ⁇ transforming growth factor- ⁇
  • a gene-of-interest may also encode a receptor for a steroid hormone, such as the testosterone receptor or the estrogen receptor or for a TGF- ⁇ .
  • the gene-of-interest may also encode a growth hormone.
  • growth hormones include, but are not limited to, human, bovine, porcine, avian, ovine, piscine, and equine growth hormones.
  • the gene-of-interest may also encode polypeptide analogs of the above-identified growth hormones.
  • the gene-of-interest could encode a growth hormone releasing factor.
  • the present invention also provides a viral gene as the gene-of-interest.
  • the viral gene may be a retroviral gene.
  • Retroviral genes of the invention may be from the HIV, HTLV-1, or HTLV-2 virus.
  • the viral gene may be a gene from a hepatitis virus, a herpes virus, a papilloma virus, a cytomegalovirus, or an animal virus.
  • Animal viruses of the invention may include, but are not limited to, pseudorabies, Marek's, Newcastle's Disease, and IBR viruses.
  • the gene-of-interest whose expression is associated with a defined physiological or pathological effect within a multicellular organism, may also be a plant gene.
  • the plant gene may encode an agronomically important trait. Examples of agronomically important traits may include, but are not limited to, germination, sprouting, flowering, fruit ripening, salt tolerance, herbicide resistance, pesticide resistance, fungicide resistance, temperature resistance, and growth.
  • the gene-of-interest may be a protozoan gene.
  • protozoans may include, but are not limited to, a selection from the group consisting of Trypanosoma, Plasmodium, Leishmania, Giardia, Entamoeba, Toxoplasma, Babesia, and Cryptosporidiosis.
  • the gene-of-interest whose expression is associated with a defined physiological or pathological effect within a multicellular organism may be a helminth gene.
  • the gene-of-interest may also be an oncogene.
  • oncogenes may include, but are not limited to, the phl-abl oncogene, the neu oncogene, or the src oncogene. Additionally, the oncogene may be selected from the group consisting of H-ras, N-ras, and K-ras oncogenes.
  • the present invention additionally provides that the gene-of-interest, whose expression is associated with a defined physiological or pathological effect within a multicellular organism, may encode a naturally occurring receptor.
  • the naturally occurring receptor may be the human low density lipoprotein (LDL) receptor.
  • the receptor may be the receptor for a hemopoietic protein. Examples of hematopoietic proteins may include, but are not limited to, a selection from the group consisting of M-CSF, G-CSF, GM-CSF, and EPO.
  • the naturally occurring receptor encoded by the gene-of-interest may also be the receptor for an interleukin (IL).
  • IL interleukin
  • Examples of an IL may include, but are not limited to, a selection from the group consisting of IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7 and IL-8.
  • the naturally occurring receptor may be a cell surface protein which mediates infection of the cell by a virus.
  • viruses may include, but are not limited to, HIV, HTLV-1, HTLV-2, a hepatitis virus, a herpes virus, a papilloma virus, a cytomegalovirus and a rhinovirus.
  • the receptor which naturally occurs in the cell is a testosterone receptor. In another example of the invention, the receptor which naturally occurs in the cell is an estrogen receptor.
  • a ligand in the context of this invention, is a molecule with a molecular weight of less than 5,000 daltons, more typically less than 2,000 daltons.
  • This invention also provides a method of determining whether a molecule not previously known to be a modulator of protein biosynthesis is capable of transcriptionally modulating the expression of a gene-of-interest.
  • the method comprises contacting a sample which contains a predefined number of cells with a predetermined amount of a molecule to be tested.
  • Each cell comprises DNA consisting essentially of (i) a modulatable transcriptional regulatory sequence of the gene-of-interest, (ii) a promoter of the gene-of-interest, and (iii) a reporter gene which expresses a polypeptide capable of producing a detectable signal, coupled to, and under the control of, the promoter.
  • the polypeptide expressed by the reporter gene produces the detectable signal under conditions such that the molecule, if capable of acting as a transcriptional modulator of the gene-of-interest, causes a measurable detectable signal to be produced by the polypeptide expressed by the reporter gene.
  • Quantitatively determining the amount of the signal produced requires comparing the amount of signal produced compared to the amount of produced signal detected in the absence of any molecule being tested or upon contacting the sample with any other molecule. The comparison permits the identification of the molecule as one which causes a change in the detectable signal produced by the polypeptide expressed by the reporter gene and thus identifying the molecule as a molecule capable of transcriptionally modulating the expression of the gene-of-interest.
  • a modulatable transcriptional regulatory sequence of a gene-of-interest concerns a DNA sequence capable of regulating the initiation of transcription from the promoter of the gene-of-interest.
  • Molecules useful in the practice of this invention have the following characteristics.
  • the molecule does not naturally occur in the cell.
  • the molecule specifically transcriptionally modulates expression of the gene-of-interest.
  • the molecule binds to DNA or RNA or binds to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell.
  • the binding of a ligand to the ligand binding domain is normally associated with the defined physiological or pathological effect.
  • promoter of a gene-of-interest is defined herein as a minimal DNA sequence necessary for specific initiation of transcription.
  • the sample may comprise cells in monolayers or cells in suspension.
  • the cells of the invention may comprise human, animal, or plant cells.
  • the cells are bacterial cells.
  • the cells are fungal cells.
  • the invention provides that the predefined number of cells contained in the sample may be from about 2 ⁇ 10 2 to about 5 ⁇ 10 5 cells, typically from about 10 3 to about 5 ⁇ 10 4 cells.
  • the ideal method might use only one cell.
  • the method may include at least 10 cells per samples. More practically the method may include at least 100 cells per sample.
  • the predetermined amount of the molecule to be tested may be based on the volume of the sample. Further, the predetermined amount of the molecule to be tested may be from about 1 pM to about 500 ⁇ M. Typically, in a primary high-throughput screen the predetermined amount would be from about 10 nM to about 500 ⁇ M. Typically in a secondary screen to evaluate initial lead compounds, the predetermined amount would be from about 1 pM to about 20 ⁇ M.
  • contacting a sample which contains a predefined number of cells with a predetermined amount of a molecule to be tested may be effected from about 1 to about 24 hours, typically from about 2 to about 12 hours. Moreover, contacting a sample which contains a predefined number of cells with a predetermined amount of a molecule to be tested may be effected with more than one predetermined amount of the molecule to be tested.
  • the molecule to be tested may be, but is not limited to, a purified molecule. As used herein, the molecule may also be a mixture of different molecules, e.g. in a fermentation broth or in a natural product extract.
  • a fermentation broth would be produced by fermentation of a suitable sample under standard conditions known to those skilled in the art.
  • natural product extracts would include, but are not restricted to, plant extracts, seaweed extracts, insect extracts, spider venoms, etc.
  • a minimal modulatable transcriptional regulatory sequence may comprise a few nucleotides in length, such as the so-called CCAAT box motif, the octamer binding motif or the heat shock element (91).
  • a gene-of-interest may typically possess multiple motifs with different combinations of modulatable transcriptional regulatory sequences. It has been shown that enhancer elements or other regulatory motifs can be present at many kilobases upstream or downstream from the transcription start site of a gene-of-interest.
  • a modulatable transcriptional regulatory sequence may comprise a few nucleotides or may be naturally or synthetically constructed having multiple elements spanning several hundred kilobases in length. Transcription can also be influenced by the particular nuclear location or local chromatin configuration.
  • the reporter gene may be situated anywhere in a chromosome.
  • the modulatable transcriptional regulatory sequence comprises a cloned genomic regulatory sequence.
  • the DNA consists essentially of more than one modulatable transcriptional regulatory sequence.
  • TK Herpes Simplex thymidine kinase
  • the invention provides that the reporter gene in the DNA contained in the cell sample, which expresses a polypeptide capable of producing a detectable signal coupled to, and under control of, the promoter, may be inserted downstream of the endogenous promoter of the gene-of-interest by homologous recombination.
  • the following provides a method of determining whether a molecule not previously known to be a modulator of protein biosynthesis is capable of transcriptionally modulating the expression of a gene-of-interest which comprises contacting a sample which contains a predefined number of cells with a predetermined amount of a molecule to be tested, each such cell comprising DNA consisting essentially of (i) a modulatable transcriptional regulatory sequence of the gene-of-interest, (ii) a promoter of the gene-of-interest, and (iii) a DNA sequence transcribable into mRNA coupled to and under the control of, the promoter, under conditions such that the molecule, if capable of acting as a transcriptional modulator of the gene-of-interest, causes a measurable difference in the amount of mRNA transcribed from the DNA sequence, quantitatively determining the amount of the mRNA produced, comparing the amount so determined with the amount of mRNA detected in the absence of any molecule being tested or upon contacting the sample with any other molecule, and thereby identifying
  • the molecule (a) does not naturally occur in the cell, (b) specifically transcriptionally modulates expression of the gene-of-interest, and (c) binds to DNA or RNA or binds to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with a defined physiological or pathological effect.
  • Modulatable transcriptional regulatory sequences may also occur within introns of a gene-of-interest. Typically, the exact location of all such regulatory sequences of a gene-of-interest will be unknown.
  • the reporter gene will be a reporter gene which can be transcribed into mRNA which can be quantified as a detectable signal. This approach would (i) allow the modulatable transcriptional regulatory sequences of the gene to be in their native environment and arrangement relative to the gene-of-interest including any and all transcriptional regulatory sequences within introns, (ii) obviate the necessity of using permanent cell lines to construct reporter cell lines and (iii) not require knowledge of promoter and modulatable transcriptional regulatory sequences.
  • mRNA is detected by quantitative polymerase chain reaction.
  • Methods for direct detection of mRNA in a rapid and quantitative manner that fulfill the needs of a high throughput screening method described in this invention are not available.
  • a suitable assay would have to meet the following criteria: (i) specific detection of fewer than 50,000 mRNA molecules, (ii) short assay time of a few hours, (iii) simple chemistry, that is amenable to automation.
  • a “single tube” assay would be used which can be carried out within the same container in which the cells to be tested have been cultured, by eliminating transfer steps which may be time consuming and contribute to assay variation.
  • DNA may be detected by a method using an amplification oligonucleotide with a branched structure that can specifically bind to the target molecule and at the same time to many molecules carrying label resulting in an up to 500-fold amplification of specific signal. This method can potentially be carried out in a single tube. Its reported sensitivity is 60,000 target DNA molecules with an assay duration of 4 hours (48).
  • the invention also provides the use of a reporter gene whose product is easily detectable.
  • the reporter gene may encode a luciferase, chloramphenicol acetyltransferase, ⁇ glucuronidase, ⁇ galactosidase, neomycin phosphotransferase, or guanine xanthine phosphoribosyltransferase.
  • the present invention also provides a screening method for determining whether a molecule not previously known to be a modulator of protein biosynthesis is capable of transcriptionally modulating the expression of a gene-of-interest which comprises separately contacting each of a plurality of substantially identical samples, e.g. more than about 10 4 samples, preferably more than about 10 5 samples, each sample containing a predefined number of cells under conditions such that contacting is effected with a predetermined amount of each different molecule to be tested.
  • a screening method for determining whether a molecule not previously known to be a modulator of protein biosynthesis is capable of transcriptionally modulating the expression of a gene-of-interest which comprises separately contacting each of a plurality of substantially identical samples, e.g. more than about 10 4 samples, preferably more than about 10 5 samples, each sample containing a predefined number of cells under conditions such that contacting is effected with a predetermined amount of each different molecule to be tested.
  • the ideal screening method provides contacting one sample with a predetermined amount of each molecule to be tested.
  • the method may include contacting at least 3 substantially identical samples, with a predetermined amount of each molecule to be tested; more practically, contacting about 9 substantially identical samples with a predetermined amount of each molecule to be tested.
  • the invention provides a method of essentially simultaneously screening molecules to determine whether the molecules are capable of transcriptionally modulating one or more genes of interest in a panel of such genes.
  • the method comprises essentially simultaneously screening the molecules against each of the genes of interest by separately contacting each of a plurality of substantially identical samples, each of which contains a predefined number of cells, with a predetermined amount of a different molecule to be tested.
  • the screening method also provides a quantitative testing range wherein at least about 10 3 samples per week are contacted with different molecules against either one or a panel of such genes of interest.
  • a method for determining whether a molecule not previously known to be a modulator of protein biosynthesis is capable of transcriptionally modulating the expression of a gene-of-interest is provided.
  • This method works in human and in animal cells. Data is presented so that one skilled in the art could make the method work in fungal cells. In addition, one skilled in the art would realize that the method may be effected in plant cells or in bacterial cells. In the case wherein luciferase is used as a reporter, this enzyme has been shown by others to be active in plant cells (89).
  • the invention also provides a method for transcriptionally modulating in a multicellular organism, the expression of a gene-of-interest, the expression of which is associated with a defined physiological or pathological effect in the organism.
  • the method comprises administering, e.g. oral administration, administration as a suppository, topical contact, intravenous, intramuscular or subcutaneous administration, to the organism an amount of a molecule effective to transcriptionally modulate expression of the gene thereby affecting the defined physiological or pathological effect.
  • the molecule (a) does not naturally occur in the organism, (b) specifically transcriptionally modulates expression of the gene-of-interest, and (c) binds to DNA or RNA or binds to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the organism. Moreover, the binding of a ligand to the ligand binding domain is normally associated with the defined physiological or pathological effect.
  • examples of a multicellular organism include, but are not limited to, a human, an animal, or a plant.
  • the defined pathological effect may be associated with a disorder and the modulated expression of the gene-of-interest may be associated with amelioration of the disorder.
  • disorders include but are not limited to, a selection from the group consisting of cancer, a hematopoietic dysfunction, diabetes, tissue inflammation, atherosclerosis, dysfunctions of memory or learning, dysfunctions in a cholesterol or other metabolic pathway; viral, fungal or parasitic infections.
  • the gene-of-interest is not necessarily part of the normal genetic make-up of the multicellular organism but rather can be introduced via infection by a pathogen.
  • the defined physiological effect is growth and the organism is an animal such as a man, cow, a pig, a bird, a fish, a sheep or a horse.
  • the gene-of-interest associated with growth could be the gene encoding growth hormone or growth hormone releasing factor.
  • the defined physiological or pathological effect is an agronomically important trait.
  • administration comprises topical contact.
  • administration comprises oral, transdermal, intravenous, intramuscular or subcutaneous administration.
  • the gene-of-interest encodes a naturally occurring receptor.
  • the receptor is a testosterone receptor.
  • the receptor is an estrogen receptor.
  • the receptor which naturally occurs in the cell is a testosterone receptor.
  • the receptor which naturally occurs in the cell is an estrogen receptor.
  • the gene-of-interest encodes a TGF- ⁇ receptor.
  • the gene-of-interest encodes a TGF- ⁇ .
  • the TGF- ⁇ is TGF- ⁇ 1.
  • the TGF- ⁇ is TGF- ⁇ 2.
  • the TGF- ⁇ is TGF- ⁇ 3.
  • the gene-of-interest encodes an oncogene.
  • the oncogene is the neu oncogene.
  • the oncogene is selected from the group consisting of H-, N-, and K-ras oncogenes.
  • This invention further provides a method for modulating the expression of a polypeptide by a cell which i) comprises DNA encoding and ii) is capable of expressing such a polypeptide, which comprises contacting the cell with an amount of a molecule effective so to transcriptionally modulate expression of the polypeptide by the cell which molecule (a) does not naturally occur in the cell and (b) binds to DNA or RNA, or binds to a protein through a domain of such protein which is not a ligand-binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with modulating transcriptional expression of a polypeptide.
  • the polypeptide is a homologous polypeptide.
  • the molecule specifically transcriptionally modulates expression of the polypeptide.
  • the polypeptide is a desired product.
  • the desired product is a monoclonal antibody.
  • the DNA is recombinant DNA.
  • the cell is a animal cell.
  • the cell is a plant cell.
  • the cell is a bacterial cell.
  • the cell is a fungal cell.
  • the polypeptide is associated with production of a desired product.
  • the desired product is an antibiotic.
  • the desired product is citric acid.
  • the following provides a biological method for recovering a substance from a mixture containing the substance which involves contacting the mixture with cells so as to separately recover the substance, which cells (i) comprise DNA encoding, and (ii) are capable of expressing a gene product, which gene product facilitates separating the substance from the mixture so as to recover the substance from the mixture, the improvement comprising (1) treating the cells with a molecule which (a) does not naturally occur in the cell and (b) binds to DNA or RNA or binds to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with increased production of the gene product.
  • the substance is a metal.
  • the following provides a biological method for treating a substance with cells so as to effect a biochemical transformation by contacting the substance with cells which (i) comprises DNA encoding, and (ii) is capable of expressing a gene product which permits biochemical transformation, the improvement comprising contacting the cells with a molecule which (a) does not naturally occur in the cell and (b) binds to DNA or RNA or binds to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with enhanced production of the gene product.
  • the biochemical transformation is associated with the production of a steroid. Further, in another example of the invention, the biochemical transformation is associated with the production of an alcohol. Further, in another example of the invention, the biochemical transformation is associated with the degradation of petroleum products.
  • this invention would have commercial applications both in the case where the polypeptide itself is commercially important, and in the case where expression of the polypeptide mediates the production of a molecule which is commercially important. Examples include, but are not limited to:
  • Vaccines can be used against viral, bacterial or parasitic infections. Such viral vaccines include poliomyelitis, measles, mumps and rubella. In addition, foot and mouth disease vaccine and rabies vaccine are of major commercial importance. Viable, disease-associated viruses can be subsequently inactivated, live viruses can be attenuated to lose their pathogenicity or genetically engineered. The expression of a viral antigen could be under the control of a heterologous promoter (80);
  • hybridoma cells ie., cell lines resulting from the fusion of a B-lymphocytes with a myeloma cell lines.
  • Monoclonal antibodies could be produced by growing the hybridoma in tissue culture or in vivo (80);
  • heterologous polypeptide increases the expression of a heterologous polypeptide by a cell i.e. a polypeptide introduced into the cell by genetic engineering and, typically, under the control of a strong promoter.
  • promoters include the promoter of the SV40 virus, the immediate early promoter of the cytomegalovirus or the baculovirus promoter.
  • heterologous polypeptides would include tissue plasminogen activator, human or animal growth hormones, blood clotting factors, erythropoietin, interleukins, interferons, the colony stimulating factors G-, GM- and M-CSF, and transforming growth factors- ⁇ 1, - ⁇ 2 and - ⁇ 3.
  • the polypeptide of interest could be expressed by the cell without genetic engineering.
  • One example would be the production of interferon alpha by the lymphoblastoid cell line ‘Namalva’ (80);
  • biodegradation is effected by the conversion of petroleum products to emulsified fatty acids.
  • Bacteria useful in this invention include, but are not restricted to, Archromobacter, Arthrobacter, Flavobacterium, Nocardia, Pseudomonas (e.g. Pseudomonas oleovorans ) and Cytophaga.
  • Yeast useful in this invention include, but are not restricted to, Candida (e.g., Candida tropicalis ), Rhodotorula, and Trichosporon (84);
  • steroids and sterols e.g., by Rhizopus sp., Saccharomyces (70) Corynebacterium sp.
  • aromatic and heterocyclic structures including phthalic acid esters, lignosulfonates, surfactants and dyes (75)
  • [0276] 28 increasing the expression of a polypeptide which is associated with the rate limiting step in the production of extracellular polysaccharides, e.g. by Corynebacterium sp., Pseudomonas sp., or Erwinia tahitica.
  • Other examples include the production of scleroglycan from the fungus Sclerotium sr., pullulan from Aureobasidium pullulans, curdlan from Alcaligens faecalis, and dextrans from Streptobacterium sp. or Streptocucus sp.
  • Other examples include anionic polysacharides from Arthrobacter viscosus, bacterial alginates from Azotobacter vinelandii, xanthan from Xanthomonas campestris.
  • the invention would include the following steps (i) identification of the protein responsible for controlling the rate limiting step and (ii) screening for molecules capable of increasing the production of that protein using the methods described herein as will be clearly and readily understood by one skilled in the art.
  • the invention provides both for the method of screening for such molecules and for the use of such molecules to regulate expression of a rate limiting polypeptide as described herein.
  • the method for transcriptionally modulating in a multicellular organism the expression of a gene-of-interest provides that growth may be the defined physiological effect and the organism is an animal such as a cow, a pig, a bird, a fish, a sheep, or a horse.
  • the method for transcriptionally modulating in a multicellular organism the expression of a gene-of-interest provides that the agronomically important trait may be the defined physiological or pathological effect.
  • This invention provides a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • this invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising administering to the human being an amount of a molecule having the structure:
  • the invention also provides a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising contacting the cell with an amount of a molecule having the structure:
  • Another provision of the present invention concerns a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone.
  • the method comprises administering to the human being an amount of a molecule having the structure:
  • This invention also concerns a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising contacting the cell with an amount of a molecule having the structure:
  • this invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising administering to the human being an amount of a molecule having the structure:
  • this invention concerns a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising contacting the cell with an amount of a molecule having the structure:
  • the invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone.
  • the method comprises administering to the human being an amount of a molecule having the structure:
  • the invention provides another method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising administering to the human being an amount of a molecule having the structure:
  • this invention provides a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • this invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone.
  • the method comprises administering to the human being an amount of a molecule having the structure:
  • the present invention provides a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the present invention also provides a method for increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises administering to the human being an amount of a molecule having the structure:
  • the present invention also provides a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the invention provides a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises administering to the human being an amount of a molecule having the structure:
  • the invention provides another method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the present invention provides another method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises administering to the human being an amount of a molecule having the structure:
  • the invention also concerns a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF comprising contacting the cell with an amount of a molecule having the structure:
  • the invention concerns a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • This invention further concerns a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the invention provides a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • the present invention concerns a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
  • the invention further concerns a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF wherein the method comprises administering to the human being an amount of a molecule having the structure:
  • the invention provides a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the invention additionally concerns a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, wherein the method comprises administering to the human being an amount of a molecule having the structure:
  • Another provision of the present invention is a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the invention provides a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF.
  • the method comprises administering to the human being an amount of a molecule having the structure:
  • the present invention provides another method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the present invention concerns a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF.
  • the method comprises administering to the human being an amount of a molecule having the structure:
  • This invention also concerns a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • this invention provides a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • the invention also concerns a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • this invention provides a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • the invention further provides a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • this invention provides a method of decreasing the formation of neutrophils and effecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • the present invention a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • this invention provides a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • the present invention further provides a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the invention provides a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • the present invention also concerns a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the present invention concerns a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • the present invention also concerns a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the invention concerns a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
  • the invention provides a method of decreasing the expression of a mammary tumor virus by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, a mammary tumor virus.
  • the method comprises contacting the cell with an amount of a molecule having the structure:
  • the invention also provides a method of suppressing the proliferation of a mammary tumor virus in a subject who (i) comprises DNA encoding, and (ii) is capable of expressing, a mammary tumor virus, comprising administering to the subject an amount of a molecule having the structure:
  • FCS Fetal calf serum
  • NIH/3T3 fibroblast cells (ATCC number CRL 1658) were used for transfection of plasmids containing the mouse mammary tumor virus (MMTV) promoter linked to firefly luciferase coding sequences (see below). Cells were propagated in Dulbecco's modified Eagle's medium (DMEM) obtained from Gibco, Grand Island, N.Y. and supplemented with 10% FCS. For high-throughput (HTP) screening, transfected NIH/3T3 clones were transferred to serum free defined medium consisting of Iscove's modified Eagle's medium (IMEM) and Ham's F12 medium (1:1) supplemented with growth factors, hormones and nutrients as described previously (43).
  • DMEM Dulbecco's modified Eagle's medium
  • IMEM Iscove's modified Eagle's medium
  • Ham's F12 medium 1:1 supplemented with growth factors, hormones and nutrients as described previously (43).
  • a rat pituitary cell line, designated GC, (4, 25) was used for transfection of plasmids containing the human growth hormone promoter (see below) and was maintained in DMEM and Ham's F12 medium (1:1), supplemented with 12.5% FCS.
  • DMEM and Ham's F12 medium (1:1), supplemented with 12.5% FCS.
  • transfected GC clones were transferred to serum free defined medium consisting of DMEM and Ham's F12 medium (1:1) supplemented with growth factors, hormones and nutrients as described previously (17, 5).
  • a human bladder carcinoma cell line (U5637, ATCC number HTB 9) was used for transfection of plasmids containing the human Granulocyte-Colony Stimulating Factor (G-CSF) promoter (see below) and was maintained in RPMI medium supplemented with 10% FCS.
  • G-CSF Granulocyte-Colony Stimulating Factor
  • transfected 5637 clones were transferred to a serum free defined medium identical to that used for the NIH/3T3 clones.
  • G418 (Geneticin, Gibco) at 0.2 mg/ml was routinely added to both serum and serum free defined media for selection and maintenance of cell lines transfected with the neomycin resistance gene.
  • This section describes (a) the molecular cloning of the human G-CSF promoter and adjacent 5′ transcriptionally modulatable regulatory sequences and (b) the making of constructs where these regulatory sequences or those of the human growth hormone (hGH) gene or those of the mouse mammary tumor virus (MMTV) long terminal repeat (LTR) control the expression of the firefly luciferase gene.
  • hGH human growth hormone
  • MMTV mouse mammary tumor virus
  • LTR long terminal repeat
  • the firefly luciferase gene was removed from the plant expression plasmid pDO432 (33) (FIG. 1) as a 1.9 kb BamHI fragment and cloned into the BamHI site of pSVL (Pharmacia, Piscataway, N.J.), a mammalian expression vector containing the SV40 promoter.
  • the resulting plasmid (pSVLuci; FIG. 2) was digested with XhoI and SalI to produce a 2.4 kb fragment containing the luciferase coding sequences and the SV40 late polyadenylation site.
  • MMTV promoter-luciferase fusion plasmid (pMLuci; FIG. 3) was used to transfect NIH/3T3 cells as described below (section E1). Similar constructs can be made using luciferase vectors from Clontech (Palo Alto, Calif.).
  • sequences of the oligonucleotide probes were: (SEQ ID NO:1) 5′ GCTTTTTGTTCCAACCCCCCTGCATT 3′ (OL-1); (SEQ ID NO:2) 5′ CCCTGCATTGTCTTGGACACCAAAT 3′ (OL-2); (SEQ ID NO:3) 5′ GCGCTCCAGGAGAAGCTGGTGAGT 3′ (OL-3); (SEQ ID NO:4) 5′ AAGCTGATGGGTGAGTGTCTTGGC 3′ (OL-4); and (SEQ ID NO:5) 5′ ATCAGCGGCTCAGCCTTCTT 3′ (OL-5).
  • OL-1, OL-2 and OL-5 recognize the G-CSF promoter region
  • OL-4 recognizes the first intron/exon junction
  • OL-3 recognizes sequences within the second exon (32).
  • One of the clones isolated from the leukocyte library using these oligonucleotide probes contains a 3.5 kb SalI-BamHI fragment of G-CSF genomic sequence consisting of 3.3 kb of promoter sequence and two hundred base pairs of the coding region. This fragment was inserted into the vector pGEM-7-Zf (Promega, Madison, Wis.) which had previously been digested with SalI/BamHI, resulting in the vector pJM710 (FIG. 7).
  • pJM710 was then digested with PstI, and the resulting 1.6 kb fragment containing G-CSF upstream sequences and the first 15 bases of the G-CSF leader sequence was inserted into the PstI site of pGEM5-Luci (FIG. 8) to generate pG-Luc1 (FIG. 9).
  • This construct was then used for transfections of 5637 human bladder carcinoma cells as described below in section E3.
  • pGEM5-Luci (FIG. 8) had previously been constructed by inserting the XbaI/SalI fragment from pSVLuci (FIG. 2) containing the luciferase coding sequence and the SV40 late polyadenylation signal into PGEM 5-Zf (Promega, Madison Wis.) digested with XhoI/SalI.
  • a mammalian expression shuttle vector was designed to allow the construction of the promoter-reporter gene fusions to be used in high-throughput screens to identify transcriptionally modulating chemicals. Features of the plasmid are shown in FIG. 10. The shuttle vector was constructed in several steps. Initially six oligonucleotides (pUV-1 through pUV-6) were synthesized (see FIG. 11 a for sequence). The sequences of pUV-1, pUV-2 and pUV-3 correspond to a multicloning site, the b-globin leader sequence and the first 53 bases of the firefly luciferase coding region.
  • the sequences of pUV-4, pUV-5 and pUV-6 are complementary to the first three oligonucleotides.
  • the pUV oligonucleotides were annealed, ligated and inserted into the SalI/EcoRI sites of pTZ18R (Pharmacia, Piscataway N.J.) (FIG. 11( b )).
  • the resulting vector was then digested with SmaI/PvuII and the oligonucleotide containing fragment was cloned into the bluescript KS(+) plasmid (Stratagene, La Jolla, Calif.), previously digested with PvuII, to yield pUV001 (FIG. 11( b )).
  • a 476 b fragment containing a dimeric SV40 polyadenylation site was then cloned into the BclI site of pUV100 (FIG. 13).
  • a 238 bp BclI/BamHI fragment was obtained from SV40 genomic DNA (BRL), ligated, digested with BclI/BamHI, gel isolated, and inserted into pUV100, resulting in the vector pUV100-3 (FIG. 13).
  • Linkers containing one SfiI and one NotI restriction site were then cloned into the PvuII/BamHI sites of pUV100-3.
  • oligonucleotides D-link1 and D-link2 and oligonucleotides R-link1 and R-link2 Two sets of linkers were synthesized containing the SfiI site in opposite orientations (oligonucleotides D-link1 and D-link2 and oligonucleotides R-link1 and R-link2).
  • the sequences of the oligonucleotides were: (SEQ ID NO:6) 5′ GATCGGCCCCTAGGGCCGCGGCCGCAT 3′ (D-link1); (SEQ ID NO:7) 5′ ATGCGGCCGCGGCCCTAGGGGCC 3′ (D-link2); and (SEQ ID NO:8) 5′ GATCGGCCCTAGGGGCGGCCGCAT 3′ (R-link1). (SEQ ID NO:9) 5′ ATGCGGCCGCGGCCCCCTAGGGCC 3′ (R-link2)
  • pUV102 The plasmid that contains D-link oligonucleotides was named pUV102 and the plasmid that contains R-link oligonucleotides was named pUV103 (FIG. 14).
  • neomycin resistance gene was then placed under control of the Herpes Simplex Virus thymidine kinase (HSV-TK) promoter to generate a resistance cassette which is free of known enhancer sequences.
  • HSV-TK Herpes Simplex Virus thymidine kinase
  • the HSV-TK promoter was synthesized using four oligonucleotides (FIG. 15) designed according to published sequence information (31), and including an SfiI restriction site 5′ of the HSV-TK sequences. These oligonucleotides were phosphorylated, annealed, ligated and inserted into pUV100 digested previously with HindIII/NheI, generating the vector pTKL 100 (FIG. 16).
  • the about 3.5 kb NheI/SmaI fragment was isolated from pTKL100, and the about 0.9 kb BstBI/BglII fragment containing the neo coding region was isolated from pRSVNEO (14). These two fragments were filled in with Klenow polymerase and ligated to form pTKNEO (FIG. 17).
  • This section describes: (a) the molecular cloning of transcriptionally modulatable regulatory sequences of several genes of interest (in this case members of the family of hematopoietic growth factors) and (b) the making of constructs where these regulatory sequences now control the expression of the luciferase gene.
  • genes of interest in this case members of the family of hematopoietic growth factors
  • constructs where these regulatory sequences now control the expression of the luciferase gene.
  • GM-CSF Human Granulocyte-Macrophage Colony Stimulating Factor
  • GM-CSF promoter sequences were performed by using oligonucleotide probes based on the GM-CSF genomic sequence (20). Two DNA oligonucleotide probes were synthesized, one corresponding to GM-CSF sequences 5′ of the coding region (5′ GGTGACCACAAAATGCCAGGGAGGCGGG 3′) (SEQ ID NO: 10) and the other to sequences in the first exon (5′GCAGGCCACAGTGCCCAAGAGACAGCAGCAGGCT 3′) (SEQ ID NO: 11).
  • the oligonucleotide probes were used to screen a human leukocyte cell gnomic DNA library (Clontech, Palo Alto) Calif.) following the manufacturer's instructions. One clone was obtained which contains the entire GM-CSF coding region along with 2 kb of upstream sequences (see FIG. 20).
  • oligonucleotides (FIG. 21) were synthesized, phosphorylated, annealed, ligated, and inserted into pUC19 previously digested with Eco RI/XbaI, generating pGM-1 (FIG. 22). pGM-1 was then sequenced (Sequenase Kit, US Biochemicals, Cleveland, Ohio) using the M13 forward (U.S. Biochem.) and reverse primers (Pharmacia, Piscataway, N.J.) to ensure that there were no mutations in the synthesized oligonucleotides.
  • pGM-2 The about 1.8 kb BstEII/ScaI fragment from pGM-1 was then isolated and ligated to about 1.5 kb BstEII/ScaI fragment from PGMLUCI to generate pGM-2 (FIG. 22).
  • pGM-2 was then digested with Hind III/XbaI and about 0.7 kb fragment was cloned into pUV 102 and pUV 103 previously digested with HindIII/XbaI. This generated pGMLS102 and pGMLS103 (FIG.
  • the sequence of this probe corresponds to sequences within the second exon of the M-CSF gene.
  • One of the clones isolated from the leukocyte library contains a 5 kb EcoRI/HindIII fragment which includes the first exon and 5′ flanking region of M-CSF. This fragment was inserted into the pTZ18R vector (Pharmacia, Piscataway N.J.) which had been previously digested with EcoRI/HindIII resulting in the vector CSF1-pTZ18 (FIG. 24).
  • the M-CSF untranslated leader sequence (23) was then fused to the first codon of the luciferase coding region as follows: (a) a 740 bp PstI/PvuII fragment was isolated from CSF1-pTZ18 containing 570 bp of the M-CSF promoter and 170 bp of the untranslated leader sequence; (b) oligonucleotides containing sequences from the 3′ end of the M-CSF leader sequence and the 5′ end of the luciferase coding region were synthesized: 5′ CTGCCCGTATGGA 3′ (CSF-luci5) (SEQ ID NO:13) 5′ ACGGGCAG 3′ (CSF-luci6);
  • a 2 kb XmaI fragment was isolated from the plasmid p100-2. This fragment contains the 3′ end of the M-CSF leader sequence fused to the luciferase start codon. The 2 kb XmaI fragment was inserted in p100-RH previously digested with XmaI, to yield p100-RHC (FIG. 27).
  • the fused 5 kb M-CSF promoter-luciferase construct was then inserted into pUV102 as follows: a 5 kb NotI/XbaI fragment (blunt ended at the NotI end) was isolated from p100-RHC and inserted into pUV102, previously digested with SnaBI/XbaI, to generate CSF1-102 (FIG. 28). This construct was then used for transfections of 5637 human bladder carcinoma cells.
  • G-CSF Human Granulocyte Colony Stimulating Factor
  • the G-CSF-oligonucleotide-containing NcoI/ScaI fragment from pGUC84 was then isolated and ligated to the luciferase-containing NcoI/ScaI fragment from pUV100 to create pGUV100 (FIG. 30).
  • the PstI fragment of the G-CSF promoter was isolated from pJM710 (FIG. 7) and inserted into the PstI site in pGUV100 generating pGUV1 (FIG. 31).
  • the rest of the G-CSF promoter clone was added by ligating the G-CSF-luciferase containing SfiI/ScaI fragment from pGUV1 to the appropriate SfiI/ScaI fragment from pJM710, creating the plasmid pGUV2 (FIG. 32).
  • the XbaI fragment from pGUV2 containing the G-CSF-luciferase fusion was then cloned into pUV103 previously digested with XbaI/SpeI, generating pGUV140 (FIG. 33).
  • TK-Neo cassette (Section C) was included by ligating the ScaI/XbaI fragment from pGUV140 which contains the G-CSF-luciferase fusion into pTKNEO103 previously digested with ScaI/XbaI, yielding the final vector pGUV150 (FIG. 34).
  • EPO6 and EPO8 oligonucleotide probes
  • human leukocyte genomic DNA library Clontech, Palo Alto, Calif.
  • the sequences of the oligonucleotide probes were: 5′AATGAGAATATCACTGTCCCAGACACCAAAGTTAATTTCTATGCCTGGAA 3′ (EPO08); and (SEQ ID NO:14) 5′TTCCAGGCATAGAAATTAAC 3′ (EPO6).
  • SEQ ID NO:15 5′ATGAGAATATCACTGTCCCAGACACCAAAGTTAATTTCTATGCCTGGAA 3′
  • EPO8 is complementary to sequences within the third exon of the EPO gene (27).
  • EPO6 is complementary to the 3′ end of EPO8 and was used as a primer for filling in the complementary strand of EPO8 with labelled nucleotides, thereby generating a probe for cloning.
  • One of the clones isolated from the leukocyte genomic DNA library contained a 7.5 kb BamHI fragment consisting of 6.2 kb of the EPO promoter region and the first three EPO exons. This fragment was inserted into the plasmid Bluescript KS(+) (Stratagene, La Jolla, Calif.), previously digested with BamHI, resulting in the vector pEP-7.5B (FIG. 35).
  • Epo leader sequence was fused to the start codon of the luciferase gene by using four synthetic oligonucleotides (EPO9 to EPO12).
  • the sequences of the oligonucleotides were: 5′CCCGGTGTGGTCACCCGGCGCCCCAGGTCGCTGAGGGACCCCGGCCAGGCGCGGA 3′ (EP09); (SEQ ID NO:16) 5′CATCTCCGCGCCTGGCCGGGGTCCCTCAGCGACCTGGGGCGCCGGGTGACCACACCGGGGGGCC 3′ (EP010); (SEQ ID NO:17) 5′GATGGAAGACGCCAAAAACATCAAGAAAGGCCCGGCGCCATTCTATCCT 3′ (EP011); and (SEQ ID NO:18) 5′CTAGAGGATAGAATGGCGCCGGGCCTTTCTTGATGTTTTTGGCGTCTTC 3′ (EP012). (SEQ ID NO:19)
  • EPO9 and EPO11 consist of 63 bases upstream of the EPO translational start site fused to the first 53 bases of the luciferase coding region.
  • EPO10 and EPO12 oligonucleotides are complementary to EPO9 and EPO11, respectively. These oligonucleotides were inserted into the plasmid pUV100 previously digested with ApaI/XbaI to generate the vector pRE (FIG. 36).
  • IL-3 II oligonucleotide probe
  • a 900 bp PstI fragment was isolated from IL-3-pTZ18R (Eco-Hind). This fragment contains 700 bp of the IL-3 promoter along with exon 1 of IL-3 (FIG. 39), and was inserted into PTZ18R (Pharmacia, Piscataway, N.J.) previously digested with PstI, resulting in the vector IL3/PstI-pTZ18R (FIG. 39).
  • oligonucleotides (IL-lu-1 to IL-lu-4) were synthesized, with the following sequences: 5′CCGGGGTTGTGGGCACCTTGCTGCTGCACATATAAGGCGGGAGGTTGTTGCCAACTCTTC3′ (IL-lu-1); (SEQ ID NO:21) 5′AGTTGGCAACAACCTCCCGCCTTATATGTGCAGCAGCAAGGTGCCCACAACC 3′ (IL-lu-2); (SEQ ID NO:22) 5′AGAGCCCCACGAAGGACCAGAACAAGACAGAGTGCCTCCTGCCGATCCAAACATGGA 3′ (IL-lu-3); and (SEQ ID NO:23) 5′GTTTGGATCGGCAGGAGGCACTCTGTCTTGTTCTGGTCCTTCGTGGGGCTCTGAAG 3′ (IL-lu-4). (SEQ ID NO:24)
  • IL-lu-1 and IL-lu-3 correspond to 112 bases of the 3′ end of the IL-3 promoter fused to the first 5 bases of the luciferase coding region.
  • IL-lu-2 and IL-lu-4 oligonucleotides are complementary to IL-lu-I and IL-lu-3, respectively.
  • Oligonucleotides IL-lu-1 to IL-lu-4 along with oligonucleotides pUV-3 and pUV-6 were annealed, ligated and inserted into IL3/PstI-pTZ18R previously digested with XmaI/EcoRI, to generate pTZ-IL-3-Luci (XmaI-XbaI) (FIG. 39).
  • a 6 kb IL-3 promoter fragment was cloned into pUV102 as follows: a 500 kb NcoI/EcoRI fragment was isolated from pTZ-IL-3-Luci (XmaI/XbaI) and inserted into IL-3-pTZ18R (EcoRI/HindIII) previously digested with NcoI/EcoRI to yield pTZ-IL3-C (FIG. 40). A 6.4 kb HindIII/XbaI fragment was then obtained from pTZIL3-C and inserted into pUV102 previously digested with HindIII/XbaI, resulting in the vector pUV-IL-3 (FIG. 41). Finally, the 1.8 kb SfiI fragment from pTKNEO3 (FIG. 19) was inserted into pUV-IL-3 previously digested with SfiI, generating the vector pIL3-102-TKNEO.
  • pMluci FGF (FIG. 3) and pSV2Neo, an antibiotic resistance plasmid (34), were co-transfected into NIH/3T3 mouse fibroblast cells using the calcium phosphate precipitation method (15) with a commercially available kit (Pharmacia, Piscataway N.J.). Two days later, cells were transferred to media containing 0.4 mg/ml G418 and were grown for an additional 10-14 days. G418-resistant clones were isolated by standard methods. Once sufficient cell numbers were obtained, clones were analyzed based on several criteria: constitutive luciferase production, induction of luciferase expression by dexamethasone (1 ⁇ m, Sigma, St.
  • phGH-LUCI (FIG. 6) and pRSVNeo, an antibiotic resistance plasmid (14), were co-transfected into GC rat pituitary cells as described above. Selection of G418-resistant cell clones was described above except for using a concentration of 0.2 mg/ml G418. Analysis of the cell clones was performed as above, except that known inducers of hGH expression (10-100 nM rat growth hormone releasing factor (RGRF, Bachem, Torrance, Calif.) and 10 ⁇ m forskolin (Sigma, St. Louis, Mo.) were used in place of dexamethasone. One clone, 532, was selected for further use in the high throuhgput screen.
  • inducers of hGH expression (10-100 nM rat growth hormone releasing factor (RGRF, Bachem, Torrance, Calif.) and 10 ⁇ m forskolin (Sigma, St. Louis, Mo.) were used in place of dexamet
  • pG-LUC1 (FIG. 9) and pRSVNeo were co-transfected into 5637 human bladder carcinoma cells as described above. Selection of G418 resistant clones was as described above except for using a concentration of 0.1 mg/ml G418. Analysis of cell clones was performed as above except that a known inducer of G-CSF expression (1-5 lg/ml lipopolysaccharide (LPS), E. coli serotype 055:b5, Difco, Detroit, Mich. or Sigma, St. Louis, Mo.) was used in place of dexamethasone. One clone, G21, was selected for use.
  • LPS lipopolysaccharide
  • Clones G 1002, G 2005, G 2071, G 2085, G 3014, G3031 included Southern blotting, reaction to known inducers, satisfactory attachment to microtiter plates and acceptable standard deviation in multiple luciferase expression assays.
  • Clones G 2005 and G 2085 were derived from 5637 cells lipofected with 100 ug/ml of linearized pGVU150 (FIG. 34), G 3031 descends from cells electroporated with 100 ug/ml of linearized pGVU150, whereas 200 ug/ml of the same plasmid were used to generate G 3014.
  • Cotransfection of circular pGVU140 (FIG. 33) with circular pTKNeo3 FIG.
  • G 1002 resulted from co-electroporation of 50 and 5 ug/ml of the two plasmids
  • G 2071 descends from cells lipofected with 100 and 10 ug.
  • M10, 532, and G21 cells maintained in their respective serum media were washed with PBS, harvested by trypsinization, and counted using a hemocytometer and the Trypan Blue exclusion method according to protocols provided by Sigma, St. Louis, Mo. Chemical Company.
  • Cells were then diluted into serum free defined media (with 0.2 mg/ml G418), and 0.2 ml of cell suspension per well was plated onto Dynatech treated plates (532 and G21) or hFN-treated plates (M10) using a Cetus Pro/Pette (Cetus, Emeryville Calif.). Plates were incubated overnight at 37C in a humidified 5% CO 2 atmosphere.
  • Bioluminescence Assay After incubation with OSI-file chemicals, cell plates were washed 3 times with PBS using an Ultrawash 2 Microplate Washer (Dynatech Labs) and 75 ll of Lysis Buffer 2 were added to each well (Lysis Buffer 2 is the same as Lysis buffer 1 exept that the ATP and DTT concentrations were changed to 2.67 mM and 133 mM, respectively). Bioluminescence was initiated by the addition of 25 ll 0.4 ⁇ m Luciferin in Buffer B to each well, and was measured in a Dynatech ML 1000 luminometer following a 1 minute incubation at room temperature. Data were captured using Lotus-Measure (Lotus) software and processed by custom-designed macros written in Lotus.
  • the cell lysis buffer was modified to also contain the luciferin. Therefore, lysis of cells and the bioluminescence reaction begin simultaneously and the production of bioluminescent light reaches a maximum at about 5 min. The level of light output declines by about 20% within further 30 min.
  • bovine serum albumin has been omitted. This improved lysis buffer has been shown to remain fully functional for at least 12 hours, when kept on ice and protected from direct light.
  • Total cellular RNA was isolated from the G21 cell clone or from untransfected 5637 cells following incubation for 6 hours with various transcriptionally modulating chemicals identified in the high-throughput screen. Cells were grown in serum free medium as described above. Total cellular RNA was isolated using the RNAZol method (CINNA/BIOTECX, Friendswood, Tex., Laboratories International, Inc.). Cells were resuspended and lysed with RNAZol solution (1.5 ml/9 cm petri dish) and the RNA was solubilized by passing the lysate a few times through a pipette.
  • RNAZol method CINNA/BIOTECX, Friendswood, Tex., Laboratories International, Inc.
  • Chloroform was added to the homogenate (0.1 ml/1 ml), and samples were shaken for 15 seconds followed by a 5 minute incubation on ice. After centrifuging for 10 minutes, the upper phase was collected and an equal volume of isopropanol was added. Samples were incubated for 45 minutes at ⁇ 20C, and the RNA was pelleted for 15 minutes at 12,000 ⁇ g at 4C. The RNA pellet was then washed with 70% ethanol and dried briefly under vacuum.
  • the G-CSF probe was a 0.6 kb AflII to XhoI fragment which contained most of exon 5 of the human G-CSF gene.
  • the B-actin probe was used as a control probe to normalize for the total amount of RNA.
  • the probes were labeled with alpha- 32 P dCTP using a random primed DNA labeling kit (Amersham, Arlington, Ill.). Following hybridization, filters were first probed with, G-CSF-and Fusion reprobed with B-Actin Probe were washed three times at room temperature with 1 ⁇ SSC, 0.13% SDS and three times at 65C with 0.2 ⁇ SSC, 0.1% SDS. Filters were first probed with G-CSF-and then reprobed with B-actin-probe. Exposure to x-ray fi ⁇ ms was performed overnight. Bands were excised and counted in a liquid scintillation counter (LKB, Gaithersburg, Md.), and counts obtained with the G-CSF specific probe were normalized relative to the counts obtained with the B-Actin specific probe.
  • LLB liquid scintillation counter
  • stably transfected cell clones were subjected to Southern blot analysis (92). Genomic DNA was prepared of each clone to be tested and restriction-cut with Dra I. After electrophoresis, transfer to nylon filters and immobilization by UV irradiation using a Stratalinker UV device (Stratagene, La Jolla, Calif.), integrated promoter/luciferase fusion constructs were visualized by probing with radioactively labelled XbaI-EcoRI fragments of the luciferase coding region. Probes were labelled using the random primer method (93).
  • Dra I cuts in the SV 40 polyadenylation sites located in the OSI mammalian expression shuttle vector just upstream the inserted promoter sequences as well as downstream of the luciferase coding region, but not in any of the 3 promoter sequences used for generating stably transfected cell clones, a single fragment should be visualized by the probe used. The size of that fragment should be characteristic for each of the three promoter sequences analyzed.
  • Plasmid pHZ18 (97, 98) contains 2 ⁇ DNA for propagation in S. cerevisiae, the yeast promoter cyc 1, which is compatible with expression in yeast cells, and the URA 3 gene for selection.
  • the plasmid was linearized with BamH1, the ends were filled-in using deoxynucleotides and E. coli DNA polymerase Klenow fragment, and then the plasmid was digested with Aat II.
  • a 4.1 kb fragment containing the cyc1 promoter, URA3 and 2 ⁇ genes was separated by agarose gel electrophoresis and subsequently purified by electroelution onto ion-exchange paper (Whatman, DE81).
  • Plasmid pBR322 was treated with endonucleases Aat II and Pvu II and a 2.2 kb fragment containing the plasmid's origin of replication and the amp R gene was isolated by agarose gel electrophoresis and eluted onto DE81 paper.
  • S. cerevisiae DB745 were made competent according to published methods (99). One and 4 ⁇ g of either pHZluci24 or pHZ18 (transfection control) were added to the competent cells and incubated at 30° C. for 30 minutes. Lithium acetate-PEG was mixed gently with the cells and allowed to incubate for another 45 minutes at which time the cells were shifted to 42° C. for 5 minutes. The cells were spread onto uracil( ⁇ ) plates and incubated at 30° C. for several days. Cell colonies were picked, grown to saturation in YPD media and analyzed for luciferase activity. Stock cultures were made from positive clones, and each was subsequently analyzed for suitability for the 96-well plate high-throughput assay.
  • 1-2 ⁇ 10 4 yeast cells were seeded into 96-well plates which have been custome designed to allow filtration of the media while retaining the cells, and which are opaque to permit analysis using a luminometer (Millipore). After the media was removed from the cells by suction, 100 ⁇ l of lysis buffer (50 mM Tris/acetate pH 7.9, 1 mM EGTA, 10 mM Mg-acetate, 0.5% Brij 58, 100 mM DTT and 4 mM ATP, 0.2 mM Luciferin, 800 U/ml lyticase) was added and the plates were incubated at room temperature for 10 minutes. Bioluminescence was monitored in a Dynatech ML1000 luminometer.
  • lysis buffer 50 mM Tris/acetate pH 7.9, 1 mM EGTA, 10 mM Mg-acetate, 0.5% Brij 58, 100 mM DTT and 4 mM ATP, 0.2 mM Luciferin, 800 U/ml
  • Reporter concentration 50 corresponds to 1280 femtograms luciferase).
  • the assay was highly accurate and reproducible as shown by the small standard deviations obtained from a total of 2,304 individual measurements obtained in a randomized fashion. As few as 200,000 molecules of luciferase can be detected in one well of a 96-well microtiter plate. When 10,000 cells expressing luciferase are plated into one well, as few as 20 molecules of luciferase per cell can thus be detected in a simple, high-throughput, fully automatable format.
  • the novel lysis buffer described in Materials and Methods, Sections F and G allows unique sensitivity and stability, accuracy and reproducibility of the luciferase-catalysed bioluminescent reaction and facilitates easy automation of the luciferase transcriptional reporter system.
  • the luciferase assay system using the lysis and reaction buffer described above not only surpasses other available luciferase assays but all other available transcriptional reporter technologies.
  • the half-life of the reporter molecule becomes a crucial parameter in determining the minimal incubation time that would be necessary to allow enough decay of reporter molecules so that the inhibition of their synthesis became visible.
  • the cell lines G1002, GM1074 and CM1 containing luciferase reporter constructs for the G-CSF, GM-CSF or the immediate early promoter of cytomegalovirus were therefore tested for the time dependency of luciferase activity after treatment of the cells with Actinomycin D, an inhibitor of transcription. This experiment measures the combined half-life of luciferase mRNA and of the luciferase protein.
  • Cells derived from clones G 1002, GM 1074 and CM 1 were seeded into 96-well microtiter plates (20,000 cells G 1002 or GM 1074/well and 5000 cells CM 1/well) and incubated overnight in cell culture conditions. At time 0 Actinomycin D (25 microg/ml) was added. At the times indicated in FIG. 49 cells were washed with PBS and luciferase activity of Actinomycin-treated cells determined as described in Materials and Methods, Section G. was compared to the luciferase activity of untreated controls. The logarithm of the treated/untreated ratio is plotted versus time.
  • transfected promoter-luciferase fusion plasmids were reacting to transcriptional inducers in a manner as predicted based on the published literature.
  • all three transfected cell clones chosen responded to inducers which have been reported to stimulate the endogenous genes; the MMTV-luciferase containing clone M10 was stimulated 11.6 fold by 1 ⁇ m dexamethasone, the hGH-luciferase containing clone 532, was stimulated 2.2 fold by 100 nM rat growth hormone releasing factor (GRF), and the hG-CSF containing clone G21 was stimulated 5.7 fold by 5 lg/ml lipopolysaccharide (LPS).
  • GRF rat growth hormone releasing factor
  • Dra I cuts in the SV 40 polyadenylation sites located in the OSI mammalian expression shuttle vector just upstream the inserted promoter sequences as well as downstream of the luciferase coding region, but not in any of the 3 promoter sequences used for generating stably transfected cell clones, a single fragment should be visualized by the probe used. The size of that fragment should be characteristic for each of the three promoter sequences analyzed.
  • G-CSF clones G 1002, G 3014 and G 3031, GM-CSF clones GM 1073, GM 1088 and GM 1105 and all 3 M-CSF clones tested contained only a single fragment with the correct molecular size (uppermost fragments in plasmid controls G/5637, M/HL 60 and GM/5637 generated by loading mixtures of the purified plasmids and extracts of the parental cell lines 5637 or HL 60 on the gel).
  • the 2 smaller fragments are non-specific, cross-hybridizing probe impurities.
  • the other clones all contained additional rearranged fragments of various molecular sizes.
  • G-CSF clones with correctly integrated promoter/reporter constructs were derived from electroporation, whereas the other G-CSF cell clones analysed were obtained either by lipofection (G 2005, G 2071 and G 2085) or calcium phosphate precipitation (G 21; see Materials and Methods).
  • G 21 cells contain multiple copies of the promoter/luciferase construct, the majority of which migrate at approximately correct molecular weights. The data suggest that electroporation under the optimized conditions described is the transfection method of choice to obtain cell clones with correctly integrated, complete promoter/luciferase reporter constructs.
  • TNF-alpha- and PMA-induction levels of clone G 1002 were influenced by the presence or absence of epidermal growth factor (EGF).
  • EGF epidermal growth factor
  • 7-Hour incubations in serum-free defined media with 20 ng/ml EGF resulted in a 3fold G-CSF promoter induction by TNF-alpha versus 4fold in the absence of EGF.
  • the 9.3fold induction by PMA in the absence of EGF was reduced to 5.6fold by including EGF in the serum-free incubation mixture. Similar differences were observed, when EGF was substituted by 10% fetal calf serum.
  • GM-CSF reporter cell lines GM 1073, GM 1088 and GM 1105 were all shown to contain correctly inserted constructs (see above). 10.5 hours incubation of GM 1073 cells with 20 ng/ml PMA in serum-containing media resulted in a 3.4 fold induction of the GM-CSF promoter, which was increased to 7.5 fold in serum-free media. Luciferase expression of clones 1088 and 1105 was induced by PMA 2.8 fold and 2 fold, respectively, while TNF-alpha induced both clones 2 fold.
  • Clones GM 1088, GM 1073 and G 1002 consistently produced bioluminescence signals varying by less than 10% between wells, when multiple 96-well microtiter plates containing these cells were assayed.
  • Table 1 shows a summary of the results of a one-week, high-throughput screen of 2,000 chemicals to identify those chemicals specifically stimulating or inhibiting transcription from the G-CSF, hGH or MMTV promoters. This screen concurrently tested chemicals at three concentrations on quadruplicate samples of the M10, 532 and G21 cell lines.
  • a chemical to be considered a selective activator A minimum inhibition of 3 fold of one promoter and less than 20% inhibition of the other two promoters was required for a chemical to be considered a selective inhibitor.
  • Table 2 gives the names and induction or inhibition ratios of the lead chemicals identified for each promoter.
  • FIG. 44 illustrates the transcriptional stimulation and FIG. 45 the transcriptional inhibition observed with some of the lead chemicals.
  • Some of the chemicals activating G-CSF transcription fell into conspicuous groups of analogs (Table 2; Group A and B). Although not specifically indicated in Table 2, groups of homologs and analogs can also be found for G-CSF-inhibiting as well as hGH-activating chemicals.
  • G-CSF lead chemicals #1780, #58, #1783, #1374 were subjected to 48 independent luciferase assays performed on the same day.
  • Compounds #58, #1780 and #1374 scored as positives in every single one of these assays inducing luciferase expression between 2 and 28fold (#58), 20 and 80fold (#1780) and 5 and 40fold (#1374).
  • Compound #1783 scored as positive only in half of the 48 repeat assays.
  • the ED50 for induction of luciferase acitvity differed from the ED50 for inhibition of respiration by a factor of almost 10, which might indicate that compound #542 exerts a specific effect on G- and GM-CSF transcription.
  • yeast expression plasmid carrying the luciferase gene under control of a yeast promoter was constructed and transfected into appropriate yeast cells. Using these cells as a model system, a format for a 96-well luciferase expression assay in yeast cells was developed as described in more detail in Materials and Methods.
  • Phorbol ester-inducible genes contain a common cis element recognized by a TPA-modulated trans-acting factor, Cell, 49:729.
  • Cyclic AMP-responsive DNA-binding protein Structure based on a cloned placental cDNA, Science, 242:1430.

Abstract

The present invention provides a method of transcriptionally modulating the expression of a gene-of-interest. The method comprises contacting a cell which is capable of expressing the gene with an amount of a molecule effective to transcriptionally modulate expression of the gene and thereby affect the level of the protein encoded by the gene which is expressed by the cell. Molecules useful in the practice of the invention are characterized as follows (a) do not naturally occur in the cell, (b) bind to DNA or RNA or bind to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell. Additionally, this invention provides a method for determining whether a molecule known to be a modulator of protein biosynthesis is capable of transcriptionally modulating expression of a gene-of-interest.

Description

  • This application is a continuation-in-part of U.S. Ser. No. 382,712, filed Jul. 18, 1989 the contents of which are hereby incorporated by reference into the present application.[0001]
  • BACKGROUND OF THE INVENTION
  • Throughout this application, various publications are referenced by Arabic numerals within parentheses. Full citations for these publications may be found at the end of the specification immediately preceding the claims. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art as known to those skilled therein as of the date of the invention described and claimed herein. [0002]
  • The expression of a specific gene can be regulated at any step in the process of producing an active protein. Modulation of total protein activity may occur via transcriptional, transcript-processing, translational or post-translational mechanisms. Transcription may be modulated by altering the rate of transcriptional initiation or the progression of RNA polymerase (28). Transcript-processing may be influenced by circumstances such as the pattern of RNA splicing, the rate of mRNA transport to the cytoplasm or mRNA stability. This invention concerns the use of molecules which act by modulating the in vivo concentration of their target proteins via regulating gene transcription. The functional properties of these chemicals are distinct from previously described molecules which also affect gene transcription. [0003]
  • Researchers have documented the regulation of transcription in bacteria by low molecular weight chemicals (42, 36). Extracellular xenobiotics, amino acids and sugars have been reported to interact directly with an intracellular proteinaceous transcriptional activator or repressor to affect the transcription of specific genes. [0004]
  • Transcriptional regulation is sufficiently different between procaryotic and eucaryotic organisms so that a direct comparison cannot readily be made. Procaryotic cells lack a distinct membrane bound nuclear compartment. The structure and organization of procaryotic DNA elements responsible for initiation of transcription differ markedly from those of eucaryotic cells. [0005]
  • The eucaryotic transcriptional unit is much more complex than its procaryotic counterpart and consists of additional elements which are not found in bacteria. Eucaryotic transcriptional units include enhancers and other cis-acting DNA sequences (30, 19). Procaryotic transcription factors most commonly exhibit a “helix-turn-helix” motif in the DNA binding domain of the protein (29, 37). Eucaryotic transcriptional factors frequently contain a “zinc finger” (37, 12) or a “leucine zipper” (24) in addition to sometimes possessing the “helix-turn-helix” motif (26). Furthermore, several critical mechanisms at the post-transcriptional level such as RNA splicing and polyadenylation are not found in procaryotic systems (21, 35). [0006]
  • In higher eucaryotes, modulation of gene transcription in response to extracellular factors can be regulated in both a temporal and tissue specific manner (22). For example, extracellular factors can exert their effects by directly or indirectly activating or inhibiting transcription factors (22, 28). [0007]
  • Modulators of transcription factors involved in direct regulation of gene expression have been described, and include those extracellular chemicals entering the cell passively and binding with high affinity to their receptor-transcription factors. This class of direct transcriptional modulators include steroid hormones and their analogs, thyroid hormones, retinoic acid, vitamin D[0008] 3 and its derivatives, and dioxins, a chemical family of polycyclic aromatic hydrocarbons (12, 38, 9).
  • Dioxins are molecules generally known to modulate transcription, however, dioxins bind to naturally-occurring receptors which respond normally to xenobiotic agents via transcriptionally activating the expression of cytochrome P450, part of an enzyme involved in detoxification. Similarly, plants also have naturally occurring receptors to xenobiotics to induce defense pathways. For example, the fungal pathogen [0009] Phytophthora megasperma induces an anti-fungal compound in soybeans. Such molecules which bind to the ligand binding domains of such naturally occurring receptors are not included on the scope of this invention.
  • The clinical use of steroid hormones, thyroid hormones, vitamin D[0010] 3 and their analogs demonstrates that agents which modulate gene transcription can be used for beneficial effects, although these agents can exhibit significant adverse side effects. Obviously, analogs of these agents could have similar clinical utility as their naturally occurring counterparts by binding to the same ligand binding domain of such receptors.
  • Indirect transcriptional regulation involves one or more signal transduction mechanisms. The regulation typically involves interaction with a receptor, the receptor being part of a multistep intracellular signaling pathway, the pathway ultimately modulating the activity of nuclear transcription factors. This class of indirect transcriptional modulators include polypeptide growth factors such as platelet-derived growth factor, epidermal growth factor, cyclic nucleotide analogs, and mitogenic tumor promoters (18, 1, 2). [0011]
  • It is well documented that a large number of chemicals, both organic and inorganic, e.g. metal ions, can non-specifically modulate transcription. [0012]
  • Researchers have used nucleotide analogs in methods to modulate transcription. The mechanism involves incorporating nucleotide analogs into nascent mRNA or non-specifically blocking mRNA synthesis. Similarly, researchers have used alkylating agents, e.g. cyclophosphamide, or intercalating agents, e.g. doxorubicin, to non-specifically inhibit transcription. [0013]
  • Moreover, chemical inhibitors of hydroxymethyl-glutaryl CoA reductase, e.g. lovastatin, are known to modulate transcription by indirectly increasing expression of hepatic low density lipoprotein receptors as a consequence of lowered cholesterol levels. [0014]
  • Signal effector type molecules such as cyclic AMP, diacylglycerol, and their analogs are known to non-specifically regulate transcription by acting as part of a multistep protein kinase cascade reaction. These signal effector type molecules bind to domains on proteins which are thus subject to normal physiological regulation by low molecular weight ligands (10, 39). [0015]
  • The specific use of sterol regulatory elements from the LDL receptor gene to control expression of a reporter gene has recently been documented in PCT/US88/10095. One aspect of PCT/US88/10095 deals with the use of specific sterol regulatory elements coupled to a reporter as a means to screen for drugs capable of stimulating cells to synthesize the LDL receptor. PCT/US88/10095 describes neither the concept of simultaneously screening large numbers of chemicals against multiple target genes nor the existence of transcriptional modulators which (a) do not naturally occur in the cell, (b) specifically transcriptionally modulate expression of the gene-of-interest, and (c) bind to DNA or RNA or bind to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with the defined physiological effect. The main focus of PCT/US88/10095 is the use of the sterol regulatory elements from the LDL receptor as a means to inhibit expression of toxic recombinant biologicals. [0016]
  • The use of molecules to specifically modulate transcription of a gene-of-interest as described herein has not previously been reported and its use will bring surprise since available literature does not propose the use of a molecule, as described, in a method to specifically modulate transcription. Instead, the available literature has reported methods which define domains of transcriptional regulating elements of a gene-of-interest. [0017]
  • Further, the practice of using a reporter gene to analyze nucleotide sequences which regulate transcription of a gene-of-interest is well documented. The demonstrated utility of a reporter gene is in its ability to define domains of transcriptional regulatory elements of a gene-of-interest. Reporter genes which express proteins, e.g. luciferase, are widely utilized in such studies. Luciferases expressed by the North American firefly, [0018] Photinus pyralis and the bacterium, Vibrio fischeri were first described as transcriptional reporters in 1985 (8, 11)
  • A method to define domains of transcriptional regulating elements of a gene-of-interest typically has also involved use of phorbol esters, cyclic nucleotide analogs, concanavalin A, or steroids, molecules which are commonly known as transcriptional modulators. However, available literature shows that researchers have not considered using a transcription screen to identify specific transcriptional modulators. Apparently, success would be unlikely in doing so, however, we have demonstrated herein that this is not the case. [0019]
  • There is utility in developing the method of transcriptional modulation of a gene-of-interest by using such molecule as described herein. This method will allow the development of novel pharmaceuticals and circumvent many of the problems associated with the therapeutic use of recombinant biological factors. [0020]
  • Problems associated with the therapeutic use of recombinant biological factors include the technical difficulties of large scale protein purification, the high costs of protein production, the limited shelf-life of most proteins and in some cases a short biological half-life of the administered protein in the organism. Additionally, therapeutic delivery of proteins normally requires injection and frequently induces an immune reaction in situations where chronic administration is required. The method described herein provides a means of up-regulating the expression of proteins, e.g. membrane receptors, which are not readily amenable to administration as injectable biologicals. [0021]
  • Furthermore, chemical molecules specifically regulating the activity of one member of a group of closely related proteins are difficult to produce. Molecules, structurally related at the protein level, may possess distinct regulatory elements at the DNA level which control their expression. Thus, molecules such as the chemical transcriptional modulators defined herein can provide a greater opportunity for specifically modulating the activity of structurally related proteins. One example is the ras oncogene family, where the H-, N- and K-ras proteins are highly related but wherein the three genes have distinct structures. [0022]
  • Finally, the molecules described herein may also serve to mimic normal physiological response mechanisms, typically involving the coordinated expression of one or more groups of functionally related genes. Therefore, determining whether a molecule can specifically transcriptionally modulate the expression of a gene-of-interest and the ultimate clinical use of the molecule provides a therapeutic advantage over the use of single recombinant biologicals, or drugs which bind directly to the final target protein encoded by the gene-of-interest. [0023]
  • SUMMARY OF THE INVENTION
  • The present invention provides a method of transcriptionally modulating the expression of a gene-of-interest, the expression of which is associated with a defined physiological or pathological effect within a multicellular organism. The method comprises contacting a cell which is capable of expressing the gene with an amount of a molecule effective to transcriptionally modulate expression of the gene and thereby affect the level of the protein encoded by the gene which is expressed by the cell. [0024]
  • Molecules useful in the practice of the invention are characterized as follows (a) do not naturally occur in the cell, (b) specifically transcriptionally modulate expression of the gene-of-interest, and (c) bind to DNA or RNA or bind to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with the defined physiological or pathological effect. [0025]
  • Additionally, this invention provides a method of determining whether a molecule, not previously known to be a modulator of protein biosynthesis, is capable of transcriptionally modulating the expression of a gene-of-interest. The method comprises contacting a sample which contains a predefined number of cells with a predetermined amount of a molecule to be tested. Each such cell comprises DNA consisting essentially of (i) a modulatable transcriptional regulatory sequence of the gene-of-interest, (ii) a promoter of the gene-of-interest, and (iii) a reporter gene which expresses a polypeptide capable of producing a detectable signal, coupled to, and under the control of, the promoter, and the contacting is carried out under conditions such that the molecule, if capable of acting as a transcriptional modulator of the gene-of-interest, causes a measurable detectable signal to be produced by the polypeptide expressed by the reporter gene and the amount of the signal produced may be quantitatively determined. The amount of produced signal so determined is compared with the amount of produced signal detected in the absence of any molecule being tested or upon contacting the sample with any other molecule so as to thereby identify the molecule as one which causes a change in the detectable signal produced by the polypeptide expressed by the reporter gene and thus identify the molecule as a molecule capable of transcriptionally modulating the expression of the gene-of-interest. [0026]
  • The present invention still further provides a method for transcriptionally modulating in a multicellular organism the expression of a gene-of-interest, the expression of which is associated with a defined physiological or pathological effect in the organism. The method comprises administering to the organism an amount of a molecule effective to transcriptionally modulate expression of the gene and thus affect the defined physiological or pathological effect. A molecule useful in the method: (a) does not naturally occur in the organism, (b) specifically transcriptionally modulates expression of the gene-of-interest, and (c) binds to DNA or RNA or binds to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the organism, the binding of a ligand to which ligand binding domain is normally associated with the defined physiological or pathological effect. [0027]
  • This invention provides a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone. The method comprises contacting the cell with an amount of a molecule having the structure: [0028]
    Figure US20030166281A1-20030904-C00001
  • effective to enhance the expression of human growth hormone by the cell. [0029]
  • Additionally, this invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising administering to the human being an amount of a molecule having the structure: [0030]
    Figure US20030166281A1-20030904-C00002
  • effective to enhance expression of human growth hormone by, and thus growth of, the human being. [0031]
  • The invention also provides a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising contacting the cell with an amount of a molecule having the structure: [0032]
    Figure US20030166281A1-20030904-C00003
  • effective to enhance the expression of human growth hormone by the cell. [0033]
  • Another provision of the present invention concerns a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone. The method comprises administering to the human being an amount of a molecule having the structure: [0034]
    Figure US20030166281A1-20030904-C00004
  • effective to enhance expression of human growth hormone by, and thus growth of, the human being. [0035]
  • This invention also concerns a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising contacting the cell with an amount of a molecule having the structure: [0036]
    Figure US20030166281A1-20030904-C00005
  • effective to enhance the expression of human growth hormone by the cell. [0037]
  • Further, this invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising administering to the human being an amount of a molecule having the structure: [0038]
    Figure US20030166281A1-20030904-C00006
  • effective to enhance expression of human growth hormone by, and thus growth of, the human being. [0039]
  • Additionally, this invention concerns a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising contacting the cell with an amount of a molecule having the structure: [0040]
    Figure US20030166281A1-20030904-C00007
  • effective to enhance the expression of human growth hormone by the cell. [0041]
  • Additionally, the invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone. The method comprises administering to the human being an amount of a molecule having the structure: [0042]
    Figure US20030166281A1-20030904-C00008
  • effective to enhance expression of human growth hormone by, and thus growth of, the human being. [0043]
  • Further, the invention provides another method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone. The method comprises contacting the cell with an amount of a molecule having the structure: [0044]
    Figure US20030166281A1-20030904-C00009
  • wherein R is [0045]
    Figure US20030166281A1-20030904-C00010
  • effective to enhance the expression of human growth hormone by the cell. [0046]
  • Additionally, the invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising administering to the human being an amount of a molecule having the structure: [0047]
    Figure US20030166281A1-20030904-C00011
  • wherein R is [0048]
    Figure US20030166281A1-20030904-C00012
  • effective to enhance expression of human growth hormone by, and thus growth of, the human being. [0049]
  • Further, this invention provides a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone. The method comprises contacting the cell with an amount of a molecule having the structure: [0050]
    Figure US20030166281A1-20030904-C00013
  • effective to enhance the expression of human growth hormone by the cell. [0051]
  • Furthermore, this invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone. The method comprises administering to the human being an amount of a molecule having the structure: [0052]
    Figure US20030166281A1-20030904-C00014
  • effective to enhance expression of human growth hormone by, and thus growth of, the human being. [0053]
  • The present invention provides a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0054]
    Figure US20030166281A1-20030904-C00015
  • effective to enhance the expression of G-CSF by the cell. [0055]
  • The present invention also provides a method for increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises administering to the human being an amount of a molecule having the structure: [0056]
    Figure US20030166281A1-20030904-C00016
  • effective to enhance the expression of GCSF by, and thus increase the formation of neutrophils in, the human being. [0057]
  • The present invention also provides a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0058]
    Figure US20030166281A1-20030904-C00017
  • effective to enhance the expression of G-CSF by the cell. [0059]
  • Additionally, the invention provides a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises administering to the human being an amount of a molecule having the structure: [0060]
    Figure US20030166281A1-20030904-C00018
  • effective to enhance expression of G-CSF by, and thus increase the formation of neutrophils in, the human being. [0061]
  • Further, the invention provides another method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0062]
    Figure US20030166281A1-20030904-C00019
  • effective to enhance the expression of G-CSF by the cell. [0063]
  • The present invention provides another method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises administering to the human being an amount of a molecule having the structure: [0064]
    Figure US20030166281A1-20030904-C00020
  • effective to enhance expression of G-CSF by, and thus increase the formation of neutrophils in, the human being. [0065]
  • The invention also concerns a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF comprising contacting the cell with an amount of a molecule having the structure: [0066]
    Figure US20030166281A1-20030904-C00021
  • effective to enhance the expression of G-CSF by the cell. [0067]
  • Additionally, the invention concerns a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0068]
    Figure US20030166281A1-20030904-C00022
  • effective to enhance expression of G-CSF by, and thus increase the formation of neutrophils in, the human being. [0069]
  • This invention further concerns a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0070]
    Figure US20030166281A1-20030904-C00023
  • effective to enhance the expression of G-CSF by the cell. [0071]
  • Further the invention provides a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0072]
    Figure US20030166281A1-20030904-C00024
  • effective to enhance expression of G-CSF by, and thus increase the formation of neutrophils in, the human being. [0073]
  • Additionally, the present invention concerns a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure: [0074]
    Figure US20030166281A1-20030904-C00025
  • effective to enhance the expression of G-CSF by the cell. [0075]
  • The invention further concerns a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF wherein the method comprises administering to the human being an amount of a molecule having the structure: [0076]
    Figure US20030166281A1-20030904-C00026
  • effective to enhance expression of G-CSF by, and thus increase the formation of neutrophils in, the human being. [0077]
  • Also, the invention provides a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0078]
    Figure US20030166281A1-20030904-C00027
  • effective to enhance the expression of G-CSF by the cell. [0079]
  • The invention additionally concerns a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, wherein the method comprises administering to the human being an amount of a molecule having the structure: [0080]
    Figure US20030166281A1-20030904-C00028
  • effective to enhance expression of G-CSF by, and thus increase the formation of neutrophils in, the human being. [0081]
  • Another provision of the present invention is a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0082]
    Figure US20030166281A1-20030904-C00029
  • effective to enhance the expression of G-CSF by the cell. [0083]
  • Additionally, the invention provides a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF. The method comprises administering to the human being an amount of a molecule having the structure: [0084]
    Figure US20030166281A1-20030904-C00030
  • effective to enhance expression of G-CSF by, and thus increase the formation of neutrophils in the human being. [0085]
  • The present invention provides another method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0086]
    Figure US20030166281A1-20030904-C00031
  • effective to enhance the expression of G-CSF by the cell. [0087]
  • Further, the present invention concerns a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF. The method comprises administering to the human being an amount of a molecule having the structure: [0088]
    Figure US20030166281A1-20030904-C00032
  • effective to enhance expression of G-CSF by, and thus increase the formation of neutrophils in, the human being. [0089]
  • This invention also concerns a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0090]
    Figure US20030166281A1-20030904-C00033
  • effective to decrease the expression of G-CSF by the cell. [0091]
  • Additionally, this invention provides a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0092]
    Figure US20030166281A1-20030904-C00034
  • effective to decrease expression of G-CSF by, and thus decrease the formation and affect the metabolic functions of neutrophils in, the human being. [0093]
  • Further, the invention also concerns a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0094]
    Figure US20030166281A1-20030904-C00035
  • effective to decrease the expression of G-CSF by the cell. [0095]
  • Additionally, this invention provides a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0096]
    Figure US20030166281A1-20030904-C00036
  • effective to decrease expression of G-CSF by, and thus decrease the formation and affect the metabolic functions of neutrophils in, the human being. [0097]
  • The invention further provides a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0098]
    Figure US20030166281A1-20030904-C00037
  • effective to decrease the expression of G-CSF by the cell. [0099]
  • Additionally, this invention provides a method of decreasing the formation of neutrophils and effecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0100]
    Figure US20030166281A1-20030904-C00038
  • effective to decrease expression of G-CSF by, and thus decrease the formation and affect the metabolic functions of neutrophils in, the human being. [0101]
  • Further, the present invention a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0102]
    Figure US20030166281A1-20030904-C00039
  • effective to decrease the expression of G-CSF by the cell. [0103]
  • Additionally, this invention provides a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0104]
    Figure US20030166281A1-20030904-C00040
  • effective to decrease expression of G-CSF by, and thus decrease the formation and affect the metabolic functions of neutrophils in, the human being. [0105]
  • The present invention further provides a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0106]
    Figure US20030166281A1-20030904-C00041
  • effective to decrease the expression of G-CSF by the cell. [0107]
  • Further still the invention provides a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0108]
    Figure US20030166281A1-20030904-C00042
  • effective to decrease expression of G-CSF by, and thus decrease the formation and affect the metabolic functions of neutrophils in, the human being. [0109]
  • The present invention also concerns a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0110]
    Figure US20030166281A1-20030904-C00043
  • effective to decrease the expression of G-CSF by the cell. [0111]
  • Additionally, the present invention concerns a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0112]
    Figure US20030166281A1-20030904-C00044
  • effective to decrease expression of G-CSF by, and thus decrease the formation and affect the metabolic functions of neutrophils in, the human being. [0113]
  • The present invention also concerns a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0114]
    Figure US20030166281A1-20030904-C00045
  • effective to decrease the expression of G-CSF by the cell. [0115]
  • Further, the invention concerns a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0116]
    Figure US20030166281A1-20030904-C00046
  • effective to decrease expression of G-CSF by, and thus decrease the formation and affect the metabolic functions of neutrophils in, the human being. [0117]
  • Moreover, the invention provides a method of decreasing the expression of a mammary tumor virus by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, a mammary tumor virus. The method comprises contacting the cell with an amount of a molecule having the structure: [0118]
    Figure US20030166281A1-20030904-C00047
  • effective to decrease the expression of a mammary tumor virus by the cell. [0119]
  • Finally, the invention also provides a method of suppressing the proliferation of a mammary tumor virus in a subject who (i) comprises DNA encoding, and (ii) is capable of expressing, a mammary tumor virus, comprising administering to the subject an amount of a molecule having the structure: [0120]
    Figure US20030166281A1-20030904-C00048
  • effective to suppress the proliferation of a mammary tumor virus in the subject. [0121]
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 is a partial restriction enzyme cleavage map of the plasmid pD0432 which contains the luciferase gene from the firefly, [0122] Photinus pyralis.
  • FIG. 2 is a partial restriction enzyme cleavage map of the plasmid pSVLuci which contains the luciferase gene from the firefly, [0123] Photinus pyralis.
  • FIG. 3 is a partial restriction enzyme cleavage map of the plasmid pMLuci which contains the luciferase gene of the firefly, [0124] Photinus pyralis and the mouse mammary tumor virus long terminal repeat.
  • FIG. 4 is a partial restriction enzyme cleavage map of the plasmid pUXLuci which contains the luciferase gene from the firefly, [0125] Photinus pyralis.
  • FIG. 5 is a partial restriction enzyme cleavage map of the plasmid phGH:CAT which contains the CAT gene and human growth hormone promoter sequences. [0126]
  • FIG. 6 is a partial restriction enzyme cleavage map of the plasmid phGH-Luci which contains the luciferase gene from the firefly, [0127] Photinus pyralis and human growth hormone promoter sequences.
  • FIG. 7 is a partial restriction enzyme cleavage map of the plasmid pJM710 which contains G-CSF upstream sequences. [0128]
  • FIG. 8 is a partial restriction enzyme cleavage map of the plasmid pGEM5-Luci which contains the luciferase gene from the firefly, [0129] Photinus pyralis.
  • FIG. 9 is a partial restriction enzyme cleavage map of the plasmid PG-[0130] Luc 1 which contains both the luciferase gene from the firefly, Photinus pyralis, and G-CSF upstream sequences.
  • FIG. 10 is a view of the mammalian expression shuttle vector pUV102 with its features. The mammalian expression shuttle vector was designed to allow the construction of the promoter-reporter gene fusions and the insertion of a neomycin resistance gene coupled to the herpes simplex virus thymidine kinase promoter (TK-NEO). [0131]
  • FIG. 11[0132] a provides the nucleotide sequences of six oligonucleotides, pUV-1 through pUV-6 (SEQ ID NOS: 25-30), which were annealed, ligated, and inserted into the SalI/EcoR1 sites of the plasmid pTZ18R.
  • FIG. 11[0133] b is a diagrammatic representation of the construction of the plasmid pUV001 from the plasmids pTZ18R and pBluescript KS(+).
  • FIG. 12 is a diagrammatic representation of the construction of the plasmid pUV100 from the plasmid pUV001 and two DNA fragments, the XbaI/XmaI fragment from pMLuci and the XmaI/BamHI fragment from pMSG. [0134]
  • FIG. 13 is a diagrammatic representation of the construction of the plasmid pUV100-3 from the plasmid pUV100 and a 476 b fragment containing a dimeric SV40 polyadenylation site. [0135]
  • FIG. 14 is a diagrammatic representation of the construction of the plasmids pUV102 and pUV103 from the plasmid pUV100-3 and D-link oligonucleotides and the plasmid pUV100-3 and R-link oligonucleotides, respectively. [0136]
  • FIG. 15 provides the nucleotide sequences of oligos (SEQ ID NOS: 31-34) used for the construction of a synthetic HSV-Thymidine Kinase promoter and provides a diagrammatic representation of the HSV-TK promoter. [0137]
  • FIG. 16 is a diagrammatic representation of the construction of the plasmid pTKL100 which contains the luciferase gene from the firefly, [0138] Photinus pyralis and the HSV-TK promoter sequence.
  • FIG. 17 is a diagrammatic representation of the construction of the plasmid pTKNEO which contains the neo gene, from about 3.5 kb NheI/XmaI fragment from pTKL100, and the about 0.9 kb BstBI/BglII fragment containing the neo coding region from pRSVNEO. [0139]
  • FIG. 18 is a diagrammatic representation of the construction of the plasmid pTKNEO2 from the plasmid pTKNEO and the [0140] oligonucleotides Neo 1 and 2 (SEQ ID NOS: 35 and 36).
  • FIG. 19 is a diagrammatic representation of the construction of the plasmid pTKNEO3 from the plasmid PTKNEO2 and about 0.9 kb EcoR1/SalI fragment from pMC1NEO. [0141]
  • FIG. 20 is a partial restriction enzyme cleavage map of a human genomic clone which contains the entire GM-CSF coding region. [0142]
  • FIG. 21 provides the nucleotide sequence of [0143] oligonucleotides 1 through 4 (SEQ ID NOS: 37-40) and provides a diagrammatic representation of GM-CSF upstream sequences fused with the ATG of the coding region of the luciferase gene from the firefly, Phoxinus pyralis.
  • FIG. 22 is a diagrammatic representation of the construction of the plasmids pGMLS102 and pGMLS103 from [0144] plasmid pUV 102 and a 0.7 kb fragment from pGM-2 and from pUV 103 and a 0.7 kb fragment from pGM-2, respectively.
  • FIG. 23 is a diagrammatic representation of the construction of plasmids pGMLL102 and pGMLL103 from the plasmid pGMLS102 and the GM-CSF clone and the plasmid pGMLS103 and the GM-CSF clone, respectively. [0145]
  • FIG. 24 is a diagrammatic representation of the construction of the plasmid CSF1-pTZ18 from the plasmid pTZ18R and a gene fragment comprising the first exon and 5′ flanking region of M-CSF. [0146]
  • FIG. 25 is a diagrammatic representation of the construction of the plasmid p100-RH from the plasmid pUV100 and the gene fragment comprising the first exon and 5′ flanking region of M-CSF from the plasmid CSF1-pTZ18. [0147]
  • FIG. 26 is a diagrammatic representation of the construction of the plasmid p100-2 by insertion of a PSTI/PVUII fragment from CSFI-pTZ18 and synthetic oligonucleotides into the plasmid pUV100. [0148]
  • FIG. 27 is a diagrammatic representation of the construction of p100-RHC by insertion of short M-[0149] CSF 5′ upstream sequences fused to the luciferase coding sequence into the plasmid p100-RH containing a longer M-CSF 5′ upstream fragment.
  • FIG. 28 is a diagrammatic representation of the construction of pCSF1-102 from a 5 kb NotI/Xba I fragment isolated from p100-RHC and inserted into pUV102. [0150]
  • FIG. 29 is a diagrammatic representation of the construction of pGUC84 from oligonucleotides containing the G-CSF leader sequence from +15 to the ATG cloned into the plasmid pUC19. [0151]
  • FIG. 30 is a diagrammatic representation of the construction of pGUV100 from the NcoI/ScaI fragment from pGUC84 containing G-CSF leader sequences cloned into the plasmid pUV100. [0152]
  • FIG. 31 is a diagrammatic representation of the construction of pGUV1 from the Pst I fragment of the G-CSF promoter from the plasmid pJM710 inserted into the Pst I site in [0153] PGUV 100.
  • FIG. 32 is a diagrammatic representation of the construction of the plasmid pGUV-2 by insertion of more G-CSF upstream sequences from pJM710 into pGUV1. [0154]
  • FIG. 33 is a diagrammatic representation of the construction of the plasmid pGUV140 from the XbaI fragment from pGUV2 containing the G-CSF-luciferase fusion and the plasmid pUV103. [0155]
  • FIG. 34 is a diagrammatic representation of the construction of the plasmid pGUV150 from the ScaI/XbaI fragment from pGUV140 which contains the G-CSF-luciferase fusion and the plasmid pTKNEO-103. [0156]
  • FIG. 35 is a diagrammatic representation of the construction of the plasmid pEP-7.5B from a 7.5 kb BamH1 fragment consisting of 6.2 kb of the EPO promoter region and the first three EPO exons and the plasmid pBluescript KS(+). [0157]
  • FIG. 36 is a diagrammatic representation of the construction of the plasmid pRE from oligonucleotides EPO 9-12 and the plasmid pUV100. [0158]
  • FIG. 37 is a diagrammatic representation of the construction of the plasmid pUV2-EP6 from a 6 kb fragment of EPO upstream sequences and the [0159] plasmid pUV 102.
  • FIG. 38 is a diagrammatic representation of the construction of the plasmid IL-3-pTZ18R from IL-3 upstream sequences and the plasmid pTZ18R. [0160]
  • FIG. 39 is a diagrammatic representation of the construction of the plasmid pTZ-IL-3-Luci from oligonucleotides IL-lu-1 to IL-lu-4, pUV-3 and pUV-6 and the plasmid IL3/PstI-pTZ18R. [0161]
  • FIG. 40 is a diagrammatic representation of the construction of the plasmid pTZ-IL3-C from a 500 kb NcoI/EcoR1 fragment from pTZ-IL-3-Luci and the plasmid IL-3-pTZ18R. [0162]
  • FIG. 41 is a diagrammatic representation of the construction of the plasmid pUV-IL-3 from a 6.4 kb Hind III/XbaI fragment from pTZ-IL3-C and the plasmid pUV102. [0163]
  • FIG. 42 is a bar graph illustrating induction of luciferase expression in reporter cell lines containing MMTV (M10), human growth hormone (532), and human G-CSF (G21) promoter sequences, in response to known transcriptional inducers. [0164]
  • FIG. 43 is a bar graph illustrating the effect of steroids on luciferase expression in the MMTV reporter cell line M10. [0165]
  • FIG. 44 is a bar graph illustrating specific induction of luciferase expression in reporter cell lines for MMTV (M10), human growth hormone (532) and human G-CSF (G21) promoters in response to chemicals identified in a high throughput screen and known transcriptional inducers. [0166]
  • FIG. 45 is a bar graph illustrating specific inhibition of luciferase expression in reportor cell lines for MMTV (M10), human growth hormone (532), and human G-CSF (G21) in response to chemicals identified in a high throughput screen. [0167]
  • FIG. 46 is an autoradiograph of a Northern blot illustrating increased G-CSF mRNA production by the human epithelial cell line U5637 in response to chemicals #670 and #1255 and IFN-gamma as compared to the solvent DMSO. Reprobing with beta-actin was used to normalize for the amount of mRNA that had been loaded onto the gel. [0168]
  • FIG. 47 illustrates a dose response analysis of [0169] chemicals #80, #670, and #1780 using the G-CSF reporter cell line G21. The amount of luciferase expression is indicated in arbitrary units.
  • FIG. 48 illustrates the linear dependence of the bioluminescent signal (in arbitrary luminometer units) from the luciferase reporter concentration (1,280 fg luciferase correspond to 50 relative reporter concentration units). [0170]
  • FIG. 49 illustrates the time course of bioluminescent signal decay after addition of Actinomycin D to the [0171] cell clones G 1002, GM 1074 and CM 1. Time in hours is plotted against the logarithm of the ratio of the bioluminescent signal generated by Actinomycin D-treated cells over the signal of untreated control cells.
  • FIG. 50 is an autoradiograph of a Southern blot illustrating the correct integration of luciferase fusion constructs containing the G-CSF, M-CSF and GM-CSF promoters, respectively, into the genomes of 5637 cells (G- and GM-CSF) and [0172] HL 60 cells (M-CSF). Lanes designated 5637 and HL 60 had been loaded with DNA preparations from the parental cell lines not containing the luciferase constructs (negative controls). Lanes labeled G/5637, GM/5637 and M/HL 60 had been loaded with the same DNA preparations with the addition of the purified plasmid preparations, which had been used for the original transfections (positive controls). The two low-molecular-weight bands appearing in almost all lanes except for the negative control lanes are derived from non-specific cross-hybridizing sequences contained in the probe.
  • FIG. 51 illustrates a dose response analysis of DMSO, methanol and ethanol using the reporter [0173] cell lines M 10, G 2005 and GM 1105. The level of luciferase expression of solvent-treated cells expressed in percent of that of untreated control cells is plotted against solvent concentration.
  • FIG. 52 is a summary of the number of lead samples obtained in a luciferase expression screen of Actinomyces fermentation broth samples for modulation of the G-CSF, GM-CSF and MMTV promoters. “TIR” stands for “Transcriptional Induction/Inhibition Ratio” and represents the ratio of luciferase expression generated by a test sample treated cells over that of control cells treated by solvent alone. [0174]
  • FIG. 53 is a summary of those lead samples listed in FIG. 52, which specifically modulate one promoter only according to the TIR criteria indicated. [0175]
  • FIG. 54 is an autoradiograph of a polyacrylamide gel illustrating an S1 nuclease protection analysis of increased mRNA production by the human bladder [0176] carcinoma cell line 5637 in response to lead chemicals #542, #1255, #1793 and #1904. “RNA” indicates the sources of the RNA preparations used in individual lanes. “Probe” indicates the mRNA-specificities of probes used in individual lanes. “Compound” lists the compounds with which the 5637 cells were treated prior to RNA extraction and loading on individual gel lanes (“Cyclo” means cycloheximide). “Conc” indicates three different compound concentrations used in the experiment (L=low, M=medium, H=high). G, GM and A indicate the correct sizes of G-CSF-, GM-CSF- and Actin-specific nuclease-protected mRNA/Probe hybrids.
  • FIG. 55 is a bar graph illustrating increased G-CSF secretion by 5637 cells treated for 48 hours in serum-containing media with the samples indicated on the abscissa. TNF-alpha was used at 5 ng/ml. [0177] Chemicals #542 and #1780 were used at 50 uM or 1 uM and 0.2 uM final concentration, respectively. Both chemicals were used in DMSO at a final concentration of 0.5%. The ordinate indicates the concentration of G-CSF secreted into 5 ml of serum-containing media by 25 square cm of confluent 5637 cells.
  • FIG. 56 illustrates a dose response analysis of [0178] chemical #542 using the G-CSF reporter cell line G 21 (solid line) and the MTT respiratory inhibition cytotoxicity assay (dotted line). Respiratory inhibition in percent of untreated control cells (Ordinate, left scale) and luciferase expression of #542-treated over solvent-treated cells (ordinate, right scale) are plotted against #542 concentration (abscissa).
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides a method of transcriptionally modulating the expression of a homologous gene-of-interest, the expression of which is associated with a defined physiological or pathological effect within a multicellular organism. The method comprises contacting a cell which is capable of expressing the gene with an amount of a molecule effective to transcriptionally modulate the expression of the gene. Modulating the expression of the gene affects the level of the protein encoded by the gene which is expressed by the cell. As used herein a homologous gene-of-interest means a gene which is naturally associated with an organism or virus encoding and expressing the gene. Therefore, this definition would not include a synthetic or recombinant gene constructed by genetic engineering methods so as to position the gene-of-interest under the control of a heterologous promoter. [0179]
  • The term “physiological effect” as used herein is defined as an effect characteristic of, or appropriate to, an organism's healthy or normal functioning. Further, the term “pathological effect” as used herein is defined as an effect altered or caused by disease. [0180]
  • Molecules useful in the practice of the invention are characterized as follows (a) do not naturally occur in the cell, (b) specifically transcriptionally modulate expression of the gene-of-interest, and (c) bind to DNA or RNA or bind to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with the defined physiological or pathological effect. [0181]
  • In one example of this invention, the molecule would not occur naturally in any cell of a higher eucaryotic organism. In another example of this invention the molecule would not occur naturally in any cell, e.g. minerals. In yet another example, the molecule would not occur naturally, e.g. synthetic molecules. [0182]
  • The phrase “specifically transcriptionally modulate expression of the gene-of-interest” as used herein means modulating the expression of the gene-of-interest without modulating the expression of other genes in the cell in a way which would cause an adverse effect on (a) an organism containing the cell in the case where the cell is within the organism or (b) the growth or the culturing of the cell, in the case where the cell is being grown or cultured to make a product where the amount of product produced is associated with expression of the gene-of-interest. However, within this definition where the drug is used to treat, for example, parasitic infection, drug application is intended to cause an adverse effect on the cells of the parasite (which may contain the gene-of-interest), but not on the cells of the host organism. In this context, a gene-of-interest may constitute a single gene or a limited number of genes whose expression can be functionally coordinated. One example of coordinate gene regulation is exhibited by the physiological growth modulators termed the transforming growth factor-β family of polypeptides (TGF-βs) (90). TGF-βs control the extracellular matrix (ECM) by; (1) increasing expression of the genes encoding ECM polypeptides, e.g. collagen, fibronectin and osteopontin; (2) increasing expression of receptors for the ECM, e.g. the integrins; and (3) increasing expression of protease inhibitors (e.g. TIMP and PAI-I) while decreasing expression of secreted proteases (e.g. collagenase and stromelysin). This coordinate regulation may allow TGF-βs to be useful in the repair of surface wounds, cartilage and bone. Molecules with properties as described in this invention, with lower molecular weights than TGF-β, and which can be either chemically synthesized or readily isolated from natural sources, yet mimic the coordinate regulation of extracellular matrix as induced by the TGF-βs, have significant advantages as therapeutic agents over the use of such complex polypeptides. [0183]
  • Moreover, the phrase “transcriptionally modulate the gene-of-interest” infers a notion of directness. Thus, as used herein, “transcriptionally modulate expression of a gene-of-interest” by a molecule means the effect upon transcription of the gene resulting from either (a) direct binding of the molecule to DNA or RNA, a DNA- or RNA-binding protein, and/or a DNA- or RNA-binding protein complex, or (b) direct binding of the molecule to a protein which directly chemically modifies a DNA- or RNA-binding protein or protein complex. [0184]
  • As used herein “chemically modifies” a DNA- or RNA-binding protein or protein complex means to modify the protein or protein complex through a chemical reaction, including but not limited to, phosphorylation, glycosylation, methylation, acetylation, adenoribosylation, acylation, myristylation, reduction, oxidation, covalent oligomerization or polymerization or proteolytic cleavage. [0185]
  • The invention provides a cell capable of expressing the gene-of-interest, the expression of which is associated with a defined physiological or pathological effect within a multicellular organism. The cell may be a human cell, an animal cell, a plant cell or any eucaryotic cell or procaryotic cell from whatever source. [0186]
  • Further, in the practice of the invention, the gene-of-interest whose expression is associated with a defined physiological or pathological effect within a multicellular organism, may be a human gene. [0187]
  • Moreover, the gene-of-interest may encode a hematopoietic protein. Hematopoietic proteins may include, but are not limited to, colony stimulating factors and erythropoietin (EPO). [0188]
  • Examples of colony stimulating factors useful in the practice of this invention include, but are not limited to, granulocyte-macrophage colony stimulating factor (GM-CSF), granulocyte colony stimulating factor (G-CSF), and macrophage colony stimulating factor (M-CSF). [0189]
  • Further, the gene-of-interest of the invention may encode an interleukin (IL) or a cytokine, or a growth modulating factor. One example of such a growth modulating factor would be a member of the transforming growth factor-β (TGF-β) family i.e. TGF-β1 or TGF-β2 or TGF-β3. A gene-of-interest may also encode a receptor for a steroid hormone, such as the testosterone receptor or the estrogen receptor or for a TGF-β. [0190]
  • The gene-of-interest may also encode a growth hormone. Examples of growth hormones include, but are not limited to, human, bovine, porcine, avian, ovine, piscine, and equine growth hormones. Additionally, the gene-of-interest may also encode polypeptide analogs of the above-identified growth hormones. Additionally, the gene-of-interest could encode a growth hormone releasing factor. [0191]
  • The present invention also provides a viral gene as the gene-of-interest. The viral gene may be a retroviral gene. Retroviral genes of the invention may be from the HIV, HTLV-1, or HTLV-2 virus. [0192]
  • In the practice of the invention the viral gene may be a gene from a hepatitis virus, a herpes virus, a papilloma virus, a cytomegalovirus, or an animal virus. [0193]
  • Animal viruses of the invention may include, but are not limited to, pseudorabies, Marek's, Newcastle's Disease, and IBR viruses. [0194]
  • The gene-of-interest, whose expression is associated with a defined physiological or pathological effect within a multicellular organism, may also be a plant gene. The plant gene may encode an agronomically important trait. Examples of agronomically important traits may include, but are not limited to, germination, sprouting, flowering, fruit ripening, salt tolerance, herbicide resistance, pesticide resistance, fungicide resistance, temperature resistance, and growth. [0195]
  • Additionally, in the practice of the invention the gene-of-interest may be a protozoan gene. Examples of protozoans may include, but are not limited to, a selection from the group consisting of Trypanosoma, Plasmodium, Leishmania, Giardia, Entamoeba, Toxoplasma, Babesia, and Cryptosporidiosis. [0196]
  • Moreover, the gene-of-interest whose expression is associated with a defined physiological or pathological effect within a multicellular organism, may be a helminth gene. [0197]
  • Further, the gene-of-interest may also be an oncogene. Examples of oncogenes may include, but are not limited to, the phl-abl oncogene, the neu oncogene, or the src oncogene. Additionally, the oncogene may be selected from the group consisting of H-ras, N-ras, and K-ras oncogenes. [0198]
  • The present invention additionally provides that the gene-of-interest, whose expression is associated with a defined physiological or pathological effect within a multicellular organism, may encode a naturally occurring receptor. The naturally occurring receptor may be the human low density lipoprotein (LDL) receptor. Further, the receptor may be the receptor for a hemopoietic protein. Examples of hematopoietic proteins may include, but are not limited to, a selection from the group consisting of M-CSF, G-CSF, GM-CSF, and EPO. [0199]
  • The naturally occurring receptor encoded by the gene-of-interest may also be the receptor for an interleukin (IL). Examples of an IL may include, but are not limited to, a selection from the group consisting of IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7 and IL-8. [0200]
  • Additionally, in the practice of the invention, the naturally occurring receptor may be a cell surface protein which mediates infection of the cell by a virus. Examples of viruses may include, but are not limited to, HIV, HTLV-1, HTLV-2, a hepatitis virus, a herpes virus, a papilloma virus, a cytomegalovirus and a rhinovirus. [0201]
  • In one example of the invention, the receptor which naturally occurs in the cell is a testosterone receptor. In another example of the invention, the receptor which naturally occurs in the cell is an estrogen receptor. [0202]
  • Typically, a ligand, in the context of this invention, is a molecule with a molecular weight of less than 5,000 daltons, more typically less than 2,000 daltons. [0203]
  • This invention also provides a method of determining whether a molecule not previously known to be a modulator of protein biosynthesis is capable of transcriptionally modulating the expression of a gene-of-interest. The method comprises contacting a sample which contains a predefined number of cells with a predetermined amount of a molecule to be tested. Each cell comprises DNA consisting essentially of (i) a modulatable transcriptional regulatory sequence of the gene-of-interest, (ii) a promoter of the gene-of-interest, and (iii) a reporter gene which expresses a polypeptide capable of producing a detectable signal, coupled to, and under the control of, the promoter. The polypeptide expressed by the reporter gene produces the detectable signal under conditions such that the molecule, if capable of acting as a transcriptional modulator of the gene-of-interest, causes a measurable detectable signal to be produced by the polypeptide expressed by the reporter gene. [0204]
  • Quantitatively determining the amount of the signal produced requires comparing the amount of signal produced compared to the amount of produced signal detected in the absence of any molecule being tested or upon contacting the sample with any other molecule. The comparison permits the identification of the molecule as one which causes a change in the detectable signal produced by the polypeptide expressed by the reporter gene and thus identifying the molecule as a molecule capable of transcriptionally modulating the expression of the gene-of-interest. [0205]
  • The phrase “a modulatable transcriptional regulatory sequence of a gene-of-interest” as used herein concerns a DNA sequence capable of regulating the initiation of transcription from the promoter of the gene-of-interest. [0206]
  • Molecules useful in the practice of this invention have the following characteristics. The molecule does not naturally occur in the cell. The molecule specifically transcriptionally modulates expression of the gene-of-interest. Further, the molecule binds to DNA or RNA or binds to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell. The binding of a ligand to the ligand binding domain is normally associated with the defined physiological or pathological effect. [0207]
  • The term “promoter” of a gene-of-interest is defined herein as a minimal DNA sequence necessary for specific initiation of transcription. [0208]
  • In the practice of the invention the sample may comprise cells in monolayers or cells in suspension. The cells of the invention may comprise human, animal, or plant cells. In one example of the invention, the cells are bacterial cells. In another example of the invention, the cells are fungal cells. [0209]
  • Further, the invention provides that the predefined number of cells contained in the sample may be from about 2×10[0210] 2 to about 5×105 cells, typically from about 103 to about 5×104 cells. In theory, the ideal method might use only one cell. In practice, the method may include at least 10 cells per samples. More practically the method may include at least 100 cells per sample.
  • The invention also provides that the predetermined amount of the molecule to be tested may be based on the volume of the sample. Further, the predetermined amount of the molecule to be tested may be from about 1 pM to about 500 μM. Typically, in a primary high-throughput screen the predetermined amount would be from about 10 nM to about 500 μM. Typically in a secondary screen to evaluate initial lead compounds, the predetermined amount would be from about 1 pM to about 20 μM. [0211]
  • Further, the invention provides that contacting a sample which contains a predefined number of cells with a predetermined amount of a molecule to be tested may be effected from about 1 to about 24 hours, typically from about 2 to about 12 hours. Moreover, contacting a sample which contains a predefined number of cells with a predetermined amount of a molecule to be tested may be effected with more than one predetermined amount of the molecule to be tested. The molecule to be tested may be, but is not limited to, a purified molecule. As used herein, the molecule may also be a mixture of different molecules, e.g. in a fermentation broth or in a natural product extract. Typically, a fermentation broth would be produced by fermentation of a suitable sample under standard conditions known to those skilled in the art. Examples of natural product extracts would include, but are not restricted to, plant extracts, seaweed extracts, insect extracts, spider venoms, etc. [0212]
  • A minimal modulatable transcriptional regulatory sequence may comprise a few nucleotides in length, such as the so-called CCAAT box motif, the octamer binding motif or the heat shock element (91). A gene-of-interest may typically possess multiple motifs with different combinations of modulatable transcriptional regulatory sequences. It has been shown that enhancer elements or other regulatory motifs can be present at many kilobases upstream or downstream from the transcription start site of a gene-of-interest. Thus, as used herein, a modulatable transcriptional regulatory sequence may comprise a few nucleotides or may be naturally or synthetically constructed having multiple elements spanning several hundred kilobases in length. Transcription can also be influenced by the particular nuclear location or local chromatin configuration. In order to optimize a screen for substances that modulate regulatory elements, it may be preferable to situate a reporter gene at the exact chromosomal region where the gene-of-interest resides normally. However, it would be clear to one skilled in the art that the reporter gene may be situated anywhere in a chromosome. In one example in the invention, the modulatable transcriptional regulatory sequence comprises a cloned genomic regulatory sequence. In another example of the invention, the DNA consists essentially of more than one modulatable transcriptional regulatory sequence. [0213]
  • A series of experiments described by K. R. Thomas and M. R. Capecchi (44, 45, 46) have shown that it is possible to direct transfected or microinjected DNA, using homologous DNA sequences, to a specific chromosomal location in mammalian cells. Using homologous recombination, it is possible to replace a gene-of-interest with other sequences at a high frequency. The materials required include a reporter gene (such as luciferase), a selectable marker (such as the neomycin phosphotransferase II gene from Tn5), and sequences derived from the gene-of-interest. These materials would be used to construct a vector which would include, in one embodiment of this invention, a gene-of-interest (free of its normal upstream promoter sequences) and reporter gene, where the reporter gene product was still active. [0214]
  • Two types of selectable markers, one positive and one negative, such as the Herpes Simplex thymidine kinase (TK), greatly enhance the likelihood of achieving homologous recombination. Use of one copy of the TK gene located outside the homologous regions at either end allows for selection of “correct” homologous recombinants. Neomycin resistant cells which are recombined randomly will usually insert via the ends of the linearized, transfected DNA and thus include the TK gene(s). This will allow for selection against these cells in the presence of gancyclovir which is converted to a toxic product. This is the method specifically described by Capecchi. However, this method has not been used to produce cell lines to screen for compounds which modulate the expression of a target gene-of-interest as described herein. [0215]
  • The invention provides that the reporter gene in the DNA contained in the cell sample, which expresses a polypeptide capable of producing a detectable signal coupled to, and under control of, the promoter, may be inserted downstream of the endogenous promoter of the gene-of-interest by homologous recombination. The following provides a method of determining whether a molecule not previously known to be a modulator of protein biosynthesis is capable of transcriptionally modulating the expression of a gene-of-interest which comprises contacting a sample which contains a predefined number of cells with a predetermined amount of a molecule to be tested, each such cell comprising DNA consisting essentially of (i) a modulatable transcriptional regulatory sequence of the gene-of-interest, (ii) a promoter of the gene-of-interest, and (iii) a DNA sequence transcribable into mRNA coupled to and under the control of, the promoter, under conditions such that the molecule, if capable of acting as a transcriptional modulator of the gene-of-interest, causes a measurable difference in the amount of mRNA transcribed from the DNA sequence, quantitatively determining the amount of the mRNA produced, comparing the amount so determined with the amount of mRNA detected in the absence of any molecule being tested or upon contacting the sample with any other molecule, and thereby identifying the molecule as one which causes a change in the detectable mRNA amount of, and thus identifying the molecule as a molecule capable of transcriptionally modulating the expression of the gene-of-interest. In one example of the above-described method, the molecule (a) does not naturally occur in the cell, (b) specifically transcriptionally modulates expression of the gene-of-interest, and (c) binds to DNA or RNA or binds to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with a defined physiological or pathological effect. [0216]
  • Modulatable transcriptional regulatory sequences may also occur within introns of a gene-of-interest. Typically, the exact location of all such regulatory sequences of a gene-of-interest will be unknown. In one example of this invention, the reporter gene will be a reporter gene which can be transcribed into mRNA which can be quantified as a detectable signal. This approach would (i) allow the modulatable transcriptional regulatory sequences of the gene to be in their native environment and arrangement relative to the gene-of-interest including any and all transcriptional regulatory sequences within introns, (ii) obviate the necessity of using permanent cell lines to construct reporter cell lines and (iii) not require knowledge of promoter and modulatable transcriptional regulatory sequences. [0217]
  • In one example of the invention, mRNA is detected by quantitative polymerase chain reaction. Methods for direct detection of mRNA in a rapid and quantitative manner that fulfill the needs of a high throughput screening method described in this invention are not available. A suitable assay would have to meet the following criteria: (i) specific detection of fewer than 50,000 mRNA molecules, (ii) short assay time of a few hours, (iii) simple chemistry, that is amenable to automation. Ideally a “single tube” assay would be used which can be carried out within the same container in which the cells to be tested have been cultured, by eliminating transfer steps which may be time consuming and contribute to assay variation. [0218]
  • Several techniques have been described by others, which may be modified to be useful in the context of this invention. All assays are based on the principle of using a single stranded nucleotide probe containing sequences complementary to the target mRNA transcribed from the gene-of-interest which probe is attached to a solid support to rapidly concentrate (capture) the mRNA out of a crude cell lysate. Cell lysates can be prepared such that ribonucleases are inhibited while hybridization of nucleic acids remains possible, for example in high concentrations of guanidinium isothiocyanate (47). Target mRNA bound to the solid phase can be freed of contaminants by intensive washing and subsequently detected with a second probe which is complementary to a different region of the target mRNA than the capture probe. The second probe would carry an appropriate detectable label. [0219]
  • To obtain optimal sensitivity i.e. obtain a maximal number of labels being specifically bound to a target molecule in the presence of a minimal number of labels being unspecifically retained by other components of the assay mixture, various techniques can be applied to increase signal to noise ratios. A procedure termed reversible target capture has been designed to reduce the background noise by capturing the target molecule-label complexes onto capture probes linked to solid phase, release of the complexes and transfer of the complexes onto new capture probes linked to solid phase and so forth, cycling between steps. Background is reduced by this procedure, because the release step is designed not to release non-specifically bound label that will not therefore be transferred together with the target molecules and will eventually be removed from the mixture (48). This method has a reported sensitivity of 15,000 target mRNA molecules, it has, however the disadvantage of involving multiple transfer steps into new containers. [0220]
  • Beside reducing background the signal to noise ratio can be improved by binding more than one label to a target molecule or by creating many copies of an initial target molecule (amplification). For example, DNA may be detected by a method using an amplification oligonucleotide with a branched structure that can specifically bind to the target molecule and at the same time to many molecules carrying label resulting in an up to 500-fold amplification of specific signal. This method can potentially be carried out in a single tube. Its reported sensitivity is 60,000 target DNA molecules with an assay duration of 4 hours (48). Further methods of amplification include the hybridization dependant replication of RNA via the Q-β bacteriophage system (49) or polymerase chain reaction based replication of a cDNA copy of the target mRNA molecule (50). The methods described herein above, have not been used by others in the context of this invention. However, in light of the subject invention, it would be clear to those skilled in the art that use of these methods would facilitate the practice of this invention. [0221]
  • The invention also provides the use of a reporter gene whose product is easily detectable. The reporter gene may encode a luciferase, chloramphenicol acetyltransferase, β glucuronidase, β galactosidase, neomycin phosphotransferase, or guanine xanthine phosphoribosyltransferase. [0222]
  • The present invention also provides a screening method for determining whether a molecule not previously known to be a modulator of protein biosynthesis is capable of transcriptionally modulating the expression of a gene-of-interest which comprises separately contacting each of a plurality of substantially identical samples, e.g. more than about 10[0223] 4 samples, preferably more than about 105 samples, each sample containing a predefined number of cells under conditions such that contacting is effected with a predetermined amount of each different molecule to be tested.
  • In theory, the ideal screening method provides contacting one sample with a predetermined amount of each molecule to be tested. In practice, the method may include contacting at least 3 substantially identical samples, with a predetermined amount of each molecule to be tested; more practically, contacting about 9 substantially identical samples with a predetermined amount of each molecule to be tested. [0224]
  • Moreover, the invention provides a method of essentially simultaneously screening molecules to determine whether the molecules are capable of transcriptionally modulating one or more genes of interest in a panel of such genes. The method comprises essentially simultaneously screening the molecules against each of the genes of interest by separately contacting each of a plurality of substantially identical samples, each of which contains a predefined number of cells, with a predetermined amount of a different molecule to be tested. [0225]
  • The screening method also provides a quantitative testing range wherein at least about 10[0226] 3 samples per week are contacted with different molecules against either one or a panel of such genes of interest.
  • Further, in the practice of this invention a method for determining whether a molecule not previously known to be a modulator of protein biosynthesis is capable of transcriptionally modulating the expression of a gene-of-interest is provided. This method works in human and in animal cells. Data is presented so that one skilled in the art could make the method work in fungal cells. In addition, one skilled in the art would realize that the method may be effected in plant cells or in bacterial cells. In the case wherein luciferase is used as a reporter, this enzyme has been shown by others to be active in plant cells (89). [0227]
  • The invention also provides a method for transcriptionally modulating in a multicellular organism, the expression of a gene-of-interest, the expression of which is associated with a defined physiological or pathological effect in the organism. The method comprises administering, e.g. oral administration, administration as a suppository, topical contact, intravenous, intramuscular or subcutaneous administration, to the organism an amount of a molecule effective to transcriptionally modulate expression of the gene thereby affecting the defined physiological or pathological effect. The molecule (a) does not naturally occur in the organism, (b) specifically transcriptionally modulates expression of the gene-of-interest, and (c) binds to DNA or RNA or binds to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the organism. Moreover, the binding of a ligand to the ligand binding domain is normally associated with the defined physiological or pathological effect. [0228]
  • In the practice of the invention, examples of a multicellular organism include, but are not limited to, a human, an animal, or a plant. [0229]
  • The defined pathological effect may be associated with a disorder and the modulated expression of the gene-of-interest may be associated with amelioration of the disorder. Further, examples of disorders include but are not limited to, a selection from the group consisting of cancer, a hematopoietic dysfunction, diabetes, tissue inflammation, atherosclerosis, dysfunctions of memory or learning, dysfunctions in a cholesterol or other metabolic pathway; viral, fungal or parasitic infections. Thus, the gene-of-interest is not necessarily part of the normal genetic make-up of the multicellular organism but rather can be introduced via infection by a pathogen. In one example of the invention, the defined physiological effect is growth and the organism is an animal such as a man, cow, a pig, a bird, a fish, a sheep or a horse. The gene-of-interest associated with growth could be the gene encoding growth hormone or growth hormone releasing factor. In another example of the invention the defined physiological or pathological effect is an agronomically important trait. In a further example of the invention administration comprises topical contact. Further in another example of the invention, administration comprises oral, transdermal, intravenous, intramuscular or subcutaneous administration. Further in one example of the invention, provides the gene-of-interest encodes a naturally occurring receptor. Further, in one example of the invention, the receptor is a testosterone receptor. Further, in another example of the invention the receptor is an estrogen receptor. Further, in another example of the invention, the receptor which naturally occurs in the cell is a testosterone receptor. Further, in another example of the invention the receptor which naturally occurs in the cell is an estrogen receptor. Further, in another example of the invention the gene-of-interest encodes a TGF-β receptor. Moreover, in another example of this invention, the gene-of-interest encodes a TGF-β. In one example of the invention the TGF-β is TGF-β1. Further, in another example of the invention the TGF-β is TGF-β2. Further, in another example of the invention the TGF-β is TGF-β3. Further, in another example of the invention the gene-of-interest encodes an oncogene. Further, in another example of the invention the oncogene is the neu oncogene. Further, in another example of the invention the oncogene is selected from the group consisting of H-, N-, and K-ras oncogenes. [0230]
  • This invention further provides a method for modulating the expression of a polypeptide by a cell which i) comprises DNA encoding and ii) is capable of expressing such a polypeptide, which comprises contacting the cell with an amount of a molecule effective so to transcriptionally modulate expression of the polypeptide by the cell which molecule (a) does not naturally occur in the cell and (b) binds to DNA or RNA, or binds to a protein through a domain of such protein which is not a ligand-binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with modulating transcriptional expression of a polypeptide. In one example of the invention, wherein the polypeptide is a homologous polypeptide. In another example of the invention, the molecule specifically transcriptionally modulates expression of the polypeptide. Further, in another example of the invention the polypeptide is a desired product. Further, in another example of the invention, the desired product is a monoclonal antibody. Moreover, in a further example of the invention, the DNA is recombinant DNA. Further, in another example of the invention, the cell is a animal cell. Further, in another example of the invention, the cell is a plant cell. Further, in another example of the invention, the cell is a bacterial cell. Further, in another example of the invention, the cell is a fungal cell. Further, in another example of the invention, the polypeptide is associated with production of a desired product. Further, in another example of the invention, the desired product is an antibiotic. Further, in another example of the invention, the desired product is citric acid. The following provides a biological method for recovering a substance from a mixture containing the substance which involves contacting the mixture with cells so as to separately recover the substance, which cells (i) comprise DNA encoding, and (ii) are capable of expressing a gene product, which gene product facilitates separating the substance from the mixture so as to recover the substance from the mixture, the improvement comprising (1) treating the cells with a molecule which (a) does not naturally occur in the cell and (b) binds to DNA or RNA or binds to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with increased production of the gene product. Further, in another example of the invention, the substance is a metal. The following provides a biological method for treating a substance with cells so as to effect a biochemical transformation by contacting the substance with cells which (i) comprises DNA encoding, and (ii) is capable of expressing a gene product which permits biochemical transformation, the improvement comprising contacting the cells with a molecule which (a) does not naturally occur in the cell and (b) binds to DNA or RNA or binds to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with enhanced production of the gene product. Further, in another example of the invention, the biochemical transformation is associated with the production of a steroid. Further, in another example of the invention, the biochemical transformation is associated with the production of an alcohol. Further, in another example of the invention, the biochemical transformation is associated with the degradation of petroleum products. [0231]
  • Clearly, this invention would have commercial applications both in the case where the polypeptide itself is commercially important, and in the case where expression of the polypeptide mediates the production of a molecule which is commercially important. Examples include, but are not limited to: [0232]
  • 1. increasing expression of a polypeptide which is associated with the rate limiting step in antigen production wherein the antigen is produced for a vaccine. Vaccines can be used against viral, bacterial or parasitic infections. Such viral vaccines include poliomyelitis, measles, mumps and rubella. In addition, foot and mouth disease vaccine and rabies vaccine are of major commercial importance. Viable, disease-associated viruses can be subsequently inactivated, live viruses can be attenuated to lose their pathogenicity or genetically engineered. The expression of a viral antigen could be under the control of a heterologous promoter (80); [0233]
  • 2. increasing expression of monoclonal antibodies by hybridoma cells, ie., cell lines resulting from the fusion of a B-lymphocytes with a myeloma cell lines. Monoclonal antibodies could be produced by growing the hybridoma in tissue culture or in vivo (80); [0234]
  • 3. increasing the expression of a heterologous polypeptide by a cell i.e. a polypeptide introduced into the cell by genetic engineering and, typically, under the control of a strong promoter. Examples of such promoters include the promoter of the SV40 virus, the immediate early promoter of the cytomegalovirus or the baculovirus promoter. Examples of heterologous polypeptides would include tissue plasminogen activator, human or animal growth hormones, blood clotting factors, erythropoietin, interleukins, interferons, the colony stimulating factors G-, GM- and M-CSF, and transforming growth factors-β1, -β2 and -β3. In a variation of this invention, the polypeptide of interest could be expressed by the cell without genetic engineering. One example would be the production of interferon alpha by the lymphoblastoid cell line ‘Namalva’ (80); [0235]
  • 4. increasing plant derived products as used in fragrances and perfumes, flavoring compounds or sweeteners e.g. the basic proteins from [0236] Thaumatococcus danielli, insecticides, anti-fungal compounds or pesticides (79);
  • 5. increasing the expression of a polypeptide which is associated with the rate limiting step in bioleaching of metals such as uranium, copper, silver, manganese etc. For example, as carried out by the organism Thiobacillus sp., algae or fungi (81); [0237]
  • 6. increasing the expression of a polypeptide which is associated with the rate limiting step in removal of nitrogen or phosphate or toxic waste minerals from water, e.g. as carried out by Nitrobacter sp. or Acinetobacter sp. (82); [0238]
  • 7. increasing the expression of a polypeptide which is associated with the rate limiting step in stimulating methane production from biological waste, typically from the methanogenic micro-organisms archaebacteria (83); [0239]
  • 8. increasing the expression of a polypeptide which is associated with the rate limiting step in the biodegradation of marine oil spills (e.g., aliphatic hydrocarbons, halogenated aliphatics, halogenated aromatics). In one instance, biodegradation is effected by the conversion of petroleum products to emulsified fatty acids. Bacteria useful in this invention include, but are not restricted to, Archromobacter, Arthrobacter, Flavobacterium, Nocardia, Pseudomonas (e.g. [0240] Pseudomonas oleovorans) and Cytophaga. Yeast useful in this invention include, but are not restricted to, Candida (e.g., Candida tropicalis), Rhodotorula, and Trichosporon (84);
  • 9. increasing the expression of a polypeptide which is associated with the rate limiting step in biodegradation of lignin (84); [0241]
  • 10. increasing the expression of a polypeptide which is associated with the rate limiting step in the biotransformation of: [0242]
  • steroids and sterols (71), e.g., by Rhizopus sp., Saccharomyces (70) Corynebacterium sp. [0243]
  • D sorbitol to L sorbose by [0244] Acetobacter suboxydans (78)
  • racemic mixtures (77) [0245]
  • prochiral substrates [0246]
  • terpenoids (72) [0247]
  • alicyclic and heteroalicylic compounds (73) [0248]
  • antibiotics (74) [0249]
  • aromatic and heterocyclic structures including phthalic acid esters, lignosulfonates, surfactants and dyes (75) [0250]
  • naphthyridines by Penicillium sp (75) [0251]
  • polynuclear aromatic hydrocarbons (75) [0252]
  • aliphatic hydrocarbons (76) [0253]
  • amino acid and peptides (77) [0254]
  • glucose to fructose (78) [0255]
  • glucose to gluconic acid (78) [0256]
  • raffinose to sucrose and galactose (79) [0257]
  • lactose [0258]
  • sucrose [0259]
  • 12. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of commercially important enzymes from micro-organisms, e.g. lactase from [0260] Aspergillus oryzae, Escherichia coli, Bacillus stearothermophilus;
  • 13. increasing the expression of a polypeptide which is associated with the rate limiting step in the growth of Saccharomyces on molasses (51); [0261]
  • 14. increasing the expression of a polypeptide which is associated with the rate limiting step in the growth of Candida on spent sulphite liquor (51); [0262]
  • 15. increasing the expression of a polypeptide which is associated with the rate limiting step in the growth of yeast on higher n-alkanes (52); [0263]
  • 16. increasing the expression of a polypeptide which is associated with the rate limiting step in the growth of bacteria on higher n-alkanes (52); [0264]
  • 17. increasing the expression of a polypeptide which is associated with the rate limiting step in the growth of bacteria or yeast on methane or methanol (53); [0265]
  • 18. increasing the expression of a polypeptide which is associated with the rate limiting step in the assimilation of atmospheric nitrogen by e.g., Azotobacteria sp., Rhizobium sp., or Cyanobacteria; [0266]
  • 19. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of insectides from Bacillus sp., e.g. [0267] Bacillus thuringiensis (54);
  • 20. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of insectides from entomogenous fungi such as Deuteromycetes, e.g. [0268] Verticillium lecanii and Hirsutella thompsonii (55);
  • 21. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of ethanol from cellulosic materials, starch crops, sugar cane, fodder beats, molasses by, for example, [0269] Saccharomyces cerevisiae, S. uvarum, Schizosaccharomyces pombe or Kluyveromyces sp. (55, 59);
  • 22. increasing the expression of a polypeptide which is associated with the rate limiting step in acetic acid production from ethanol by Acetobacter sp. or Gluconobacter sp. (55); [0270]
  • 23. increasing the expression of a polypeptide which is associated with the rate limiting step in the lactic acid production by the family of Lactobacillaceae (56); [0271]
  • 24. increasing the expression of a polypeptide which is associated with the rate limiting step in the citric acid production by Candida sp., [0272] Aspergillus niger using e.g., molasses or starch (56);
  • 25. increasing the expression of a polypeptide which is associated with the rate limiting step in gluconic acid production by e.g. Pseudomonas sp., Gluconobacter sp., and Acetobacter sp. (57); [0273]
  • 26. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of amino acids by bacteria or fungi (58); [0274]
  • 27. increasing the expression of a enzyme in a cell, which enzyme catalyzes the resolution of racemic mixtures of amino acids (58); [0275]
  • 28. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of extracellular polysaccharides, e.g. by Corynebacterium sp., Pseudomonas sp., or [0276] Erwinia tahitica. Other examples include the production of scleroglycan from the fungus Sclerotium sr., pullulan from Aureobasidium pullulans, curdlan from Alcaligens faecalis, and dextrans from Streptobacterium sp. or Streptocucus sp. Other examples include anionic polysacharides from Arthrobacter viscosus, bacterial alginates from Azotobacter vinelandii, xanthan from Xanthomonas campestris.
  • 29. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of an antifungal compound (e.g. Griseofulvin) and penicillins from Penicillium sps. [0277]
  • 30. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of antibiotics by fungi, e.g. polyether antibiotics (66), chloramphenicol (66), ansamycines (65), tetracyclines (64), macrolides (63), aminoglycosides (62), clavans, cephalosporins, cephamycins (61) from Streptomyces sp.; [0278]
  • 31. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of antitumor substances, e.g., actinomycin D, anthracyclines, and bleomycin from Streptomyces sp.; [0279]
  • 32. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of nucleic acids, nucleotides and related compounds, e.g., 5′ inosinate (IMP), 5′ guanylate (GMP), cAMP by e.g. [0280] Brevibacterium ammoniagenes (60);
  • 33. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of vitamins, e.g. vitamin B12 by [0281] Pseudomonas denitrificans, Propionibacterium shermanii, or Rhodopseudomonas protamicus;
  • 34. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of riboflavin by [0282] Ashbya gossypii or Bacillus subtilis;
  • 35. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of ergosterol by yeast, e.g. [0283] Saccharomyces cerevisiae;
  • 36. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of ergot alkaloids by Claviceps sp (67); [0284]
  • 37. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of secondary metabolites useful for selected therapeutic uses in human medicine, e.g. cyclosporin from [0285] Trichoderma polysporum (68);
  • 38. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of secondary products from plant cell cultures, e.g. cinnamic acid derivatives in [0286] Coleus blume; shikonins from Lithospermum erythrophizon (69);
  • 39. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of wine and beer by Sacchromyces sp. (85, 86); [0287]
  • 40. increasing the expression of a polypeptide which is associated with the rate limiting step in the production of yogurt or cheese by Staphylococcus sp., Lactobacillus sp. and Propionibacterium sp. (87, 88); [0288]
  • 41. increasing the expression of a polypeptide which is associated with the rate limiting step in the fermentation of cocoa from [0289] Theobroma cacao by fungi and bacteria;
  • 42. increasing the expression of a polypeptide which is associated with the rate limiting step in the fermentation of coffee beans from Coffea sp. by fungi and bacteria. [0290]
  • In each case the invention would include the following steps (i) identification of the protein responsible for controlling the rate limiting step and (ii) screening for molecules capable of increasing the production of that protein using the methods described herein as will be clearly and readily understood by one skilled in the art. The invention provides both for the method of screening for such molecules and for the use of such molecules to regulate expression of a rate limiting polypeptide as described herein. [0291]
  • Additionally, the method for transcriptionally modulating in a multicellular organism the expression of a gene-of-interest provides that growth may be the defined physiological effect and the organism is an animal such as a cow, a pig, a bird, a fish, a sheep, or a horse. [0292]
  • Further, the method for transcriptionally modulating in a multicellular organism the expression of a gene-of-interest provides that the agronomically important trait may be the defined physiological or pathological effect. [0293]
  • This invention provides a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone. The method comprises contacting the cell with an amount of a molecule having the structure: [0294]
    Figure US20030166281A1-20030904-C00049
  • effective to enhance the expression of human growth hormone by the cell. [0295]
  • Additionally, this invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising administering to the human being an amount of a molecule having the structure: [0296]
    Figure US20030166281A1-20030904-C00050
  • effective to enhance expression of human growth hormone by, and thus growth of, the human being. [0297]
  • The invention also provides a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising contacting the cell with an amount of a molecule having the structure: [0298]
    Figure US20030166281A1-20030904-C00051
  • effective to enhance the expression of human growth hormone by the cell. [0299]
  • Another provision of the present invention concerns a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone. The method comprises administering to the human being an amount of a molecule having the structure: [0300]
    Figure US20030166281A1-20030904-C00052
  • effective to enhance expression of human growth hormone by, and thus growth of, the human being. [0301]
  • This invention also concerns a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising contacting the cell with an amount of a molecule having the structure: [0302]
    Figure US20030166281A1-20030904-C00053
  • effective to enhance the expression of human growth hormone by the cell. [0303]
  • Further, this invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising administering to the human being an amount of a molecule having the structure: [0304]
    Figure US20030166281A1-20030904-C00054
  • effective to enhance expression of human growth hormone by, and thus growth of, the human being. [0305]
  • Additionally, this invention concerns a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising contacting the cell with an amount of a molecule having the structure: [0306]
    Figure US20030166281A1-20030904-C00055
  • effective to enhance the expression of human growth hormone by the cell. [0307]
  • Additionally, the invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone. The method comprises administering to the human being an amount of a molecule having the structure: [0308]
    Figure US20030166281A1-20030904-C00056
  • effective to enhance expression of human growth hormone by, and thus growth of, the human being. [0309]
  • Further, the invention provides another method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone. The method comprises contacting the cell with an amount of a molecule having the structure: [0310]
    Figure US20030166281A1-20030904-C00057
  • wherein R is [0311]
    Figure US20030166281A1-20030904-C00058
  • effective to enhance the expression of human growth hormone by the cell. [0312]
  • Additionally, the invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising administering to the human being an amount of a molecule having the structure: [0313]
    Figure US20030166281A1-20030904-C00059
  • wherein R is [0314]
    Figure US20030166281A1-20030904-C00060
  • effective to enhance expression of human growth hormone by, and thus growth of, the human being. [0315]
  • Further, this invention provides a method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone. The method comprises contacting the cell with an amount of a molecule having the structure: [0316]
    Figure US20030166281A1-20030904-C00061
  • effective to enhance the expression of human growth hormone by the cell. [0317]
  • Furthermore, this invention provides a method of increasing the growth of a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone. The method comprises administering to the human being an amount of a molecule having the structure: [0318]
    Figure US20030166281A1-20030904-C00062
  • effective to enhance expression of human growth hormone by, and thus growth of, the human being. [0319]
  • The present invention provides a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0320]
    Figure US20030166281A1-20030904-C00063
  • effective to enhance the expression of G-CSF by the cell. [0321]
  • The present invention also provides a method for increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises administering to the human being an amount of a molecule having the structure: [0322]
    Figure US20030166281A1-20030904-C00064
  • effective to enhance the expression of GCSF by, and thus increase the formation of neutrophils in the human being. [0323]
  • The present invention also provides a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0324]
    Figure US20030166281A1-20030904-C00065
  • effective to enhance the expression of G-CSF by the cell. [0325]
  • Additionally, the invention provides a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises administering to the human being an amount of a molecule having the structure: [0326]
    Figure US20030166281A1-20030904-C00066
  • effective to enhance expression of G-CSF by, and thus increase the formation of neutrophils in, the human being. [0327]
  • Further, the invention provides another method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0328]
    Figure US20030166281A1-20030904-C00067
  • effective to enhance the expression of G-CSF by the cell. [0329]
  • The present invention provides another method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises administering to the human being an amount of a molecule having the structure: [0330]
    Figure US20030166281A1-20030904-C00068
  • effective to enhance expression of G-CSF by, and thus increase the formation of neutrophils in, the human being. [0331]
  • The invention also concerns a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF comprising contacting the cell with an amount of a molecule having the structure: [0332]
    Figure US20030166281A1-20030904-C00069
  • effective to enhance the expression of G-CSF by the cell. [0333]
  • Additionally, the invention concerns a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0334]
    Figure US20030166281A1-20030904-C00070
  • effective to enhance expression of G-CSF by, and thus increase the formation of neutrophils in, the human being. [0335]
  • This invention further concerns a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0336]
    Figure US20030166281A1-20030904-C00071
  • effective to enhance the expression of G-CSF by the cell. [0337]
  • Further the invention provides a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0338]
    Figure US20030166281A1-20030904-C00072
  • effective to enhance expression of G-CSF by, and thus increase the formation of neutrophils in, the human being. [0339]
  • Additionally, the present invention concerns a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure: [0340]
    Figure US20030166281A1-20030904-C00073
  • effective to enhance the expression of G-CSF by the cell. [0341]
  • The invention further concerns a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF wherein the method comprises administering to the human being an amount of a molecule having the structure: [0342]
    Figure US20030166281A1-20030904-C00074
  • effective to enhance expression of G-CSF by, and thus increase the formation of neutrophils in, the human being. [0343]
  • Also, the invention provides a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0344]
    Figure US20030166281A1-20030904-C00075
  • effective to enhance the expression of G-CSF by the cell. [0345]
  • The invention additionally concerns a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, wherein the method comprises administering to the human being an amount of a molecule having the structure: [0346]
    Figure US20030166281A1-20030904-C00076
  • effective to enhance expression of G-CSF by, and thus increase the formation of neutrophils in, the human being. [0347]
  • Another provision of the present invention is a method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0348]
    Figure US20030166281A1-20030904-C00077
  • effective to enhance the expression of G-CSF by the cell. [0349]
  • Additionally, the invention provides a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF. The method comprises administering to the human being an amount of a molecule having the structure: [0350]
    Figure US20030166281A1-20030904-C00078
  • effective to enhance expression of G-CSF by, and thus increase the formation of neutrophils in the human being. [0351]
  • The present invention provides another method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0352]
    Figure US20030166281A1-20030904-C00079
  • effective to enhance the expression of G-CSF by the cell. [0353]
  • Further, the present invention concerns a method of increasing the formation of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing G-CSF. The method comprises administering to the human being an amount of a molecule having the structure: [0354]
    Figure US20030166281A1-20030904-C00080
  • effective to enhance expression of G-CSF by, and thus increase the formation of neutrophils in, the human being. [0355]
  • This invention also concerns a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0356]
    Figure US20030166281A1-20030904-C00081
  • effective to decrease the expression of G-CSF by the cell. [0357]
  • Additionally, this invention provides a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0358]
    Figure US20030166281A1-20030904-C00082
  • effective to decrease expression of G-CSF by, and thus decrease the formation and affect the metabolic functions of neutrophils in, the human being. [0359]
  • Further, the invention also concerns a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0360]
    Figure US20030166281A1-20030904-C00083
  • effective to decrease the expression of G-CSF by the cell. [0361]
  • Additionally, this invention provides a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0362]
    Figure US20030166281A1-20030904-C00084
  • effective to decrease expression of G-CSF by, and thus decrease the formation and affect the metabolic functions of neutrophils in, the human being. [0363]
  • The invention further provides a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0364]
    Figure US20030166281A1-20030904-C00085
  • effective to decrease the expression of G-CSF by the cell. [0365]
  • Additionally, this invention provides a method of decreasing the formation of neutrophils and effecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0366]
    Figure US20030166281A1-20030904-C00086
  • effective to decrease expression of G-CSF by, and thus decrease the formation and affect the metabolic functions of neutrophils in, the human being. [0367]
  • Further, the present invention a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0368]
    Figure US20030166281A1-20030904-C00087
  • effective to decrease the expression of G-CSF by the cell. [0369]
  • Additionally, this invention provides a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0370]
    Figure US20030166281A1-20030904-C00088
  • effective to decrease expression of G-CSF by, and thus decrease the formation and affect the metabolic functions of neutrophils in, the human being. [0371]
  • The present invention further provides a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0372]
    Figure US20030166281A1-20030904-C00089
  • effective to decrease the expression of G-CSF by the cell. [0373]
  • Further still the invention provides a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0374]
    Figure US20030166281A1-20030904-C00090
  • effective to decrease expression of G-CSF by, and thus decrease the formation and affect the metabolic functions of neutrophils in, the human being. [0375]
  • The present invention also concerns a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0376]
    Figure US20030166281A1-20030904-C00091
  • effective to decrease the expression of G-CSF by the cell. [0377]
  • Additionally, the present invention concerns a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0378]
    Figure US20030166281A1-20030904-C00092
  • effective to decrease expression of G-CSF by, and thus decrease the formation and affect the metabolic functions of neutrophils in, the human being. [0379]
  • The present invention also concerns a method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF. The method comprises contacting the cell with an amount of a molecule having the structure: [0380]
    Figure US20030166281A1-20030904-C00093
  • effective to decrease the expression of G-CSF by the cell. [0381]
  • Further, the invention concerns a method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils, which has implications for the treatment of inflammatory and autoimmune disorders in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure: [0382]
    Figure US20030166281A1-20030904-C00094
  • effective to decrease expression of G-CSF by, and thus decrease the formation and affect the metabolic functions of neutrophils in, the human being. [0383]
  • Moreover, the invention provides a method of decreasing the expression of a mammary tumor virus by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, a mammary tumor virus. The method comprises contacting the cell with an amount of a molecule having the structure: [0384]
    Figure US20030166281A1-20030904-C00095
  • effective to decrease the expression of a mammary tumor virus by the cell. [0385]
  • Finally, the invention also provides a method of suppressing the proliferation of a mammary tumor virus in a subject who (i) comprises DNA encoding, and (ii) is capable of expressing, a mammary tumor virus, comprising administering to the subject an amount of a molecule having the structure: [0386]
    Figure US20030166281A1-20030904-C00096
  • effective to suppress the proliferation of a mammary tumor virus in the subject. [0387]
  • EXPERIMENTAL DETAILS Materials and Methods A. Cell Culture
  • All media and reagents used for routine cell culture were purchased from Gibco (Grand Island, N.Y.), Hazelton (Lenexa, Kans.), or Whittaker M.A. Biologicals (Walkersville, Md.). Fetal calf serum (FCS) was from Hyclone (Logan, Utah), and nutrients used for serum-free defined media were purchased from Sigma (St. Louis, Mo.), Boehringer Mannheim (Indianapolis, Ind.), Bachem (Torrance, Calif.) and Collaborative Research (Bedford, Mass.). [0388]
  • NIH/3T3 fibroblast cells (ATCC number CRL 1658) were used for transfection of plasmids containing the mouse mammary tumor virus (MMTV) promoter linked to firefly luciferase coding sequences (see below). Cells were propagated in Dulbecco's modified Eagle's medium (DMEM) obtained from Gibco, Grand Island, N.Y. and supplemented with 10% FCS. For high-throughput (HTP) screening, transfected NIH/3T3 clones were transferred to serum free defined medium consisting of Iscove's modified Eagle's medium (IMEM) and Ham's F12 medium (1:1) supplemented with growth factors, hormones and nutrients as described previously (43). [0389]
  • A rat pituitary cell line, designated GC, (4, 25) was used for transfection of plasmids containing the human growth hormone promoter (see below) and was maintained in DMEM and Ham's F12 medium (1:1), supplemented with 12.5% FCS. For HTP screening, transfected GC clones were transferred to serum free defined medium consisting of DMEM and Ham's F12 medium (1:1) supplemented with growth factors, hormones and nutrients as described previously (17, 5). [0390]
  • A human bladder carcinoma cell line (U5637, ATCC number HTB 9) was used for transfection of plasmids containing the human Granulocyte-Colony Stimulating Factor (G-CSF) promoter (see below) and was maintained in RPMI medium supplemented with 10% FCS. For HTP screening, transfected 5637 clones were transferred to a serum free defined medium identical to that used for the NIH/3T3 clones. [0391]
  • G418 (Geneticin, Gibco) at 0.2 mg/ml was routinely added to both serum and serum free defined media for selection and maintenance of cell lines transfected with the neomycin resistance gene. [0392]
  • B. Plasmid Construction and Molecular Cloning of Promoter-Reporter Fusion Constructs to Be Transfected into Cells Used for a 2,000-Chemical Transcription Screen
  • This section describes (a) the molecular cloning of the human G-CSF promoter and adjacent 5′ transcriptionally modulatable regulatory sequences and (b) the making of constructs where these regulatory sequences or those of the human growth hormone (hGH) gene or those of the mouse mammary tumor virus (MMTV) long terminal repeat (LTR) control the expression of the firefly luciferase gene. These constructs were transfected into cells as described in Section E and used for a high-throughput pilot screen of 2,000 chemicals to identify chemicals acting as specific transcriptional modulators (see Section G and “Results”). [0393]
  • Unless otherwise indicated cloning procedures were performed essentially according to Maniatis et al. (1982) (28). Oligonucleotides were synthesized by the beta-cyanoethyl phosphoramidite method according to protocols provided by the manufacturer of the DNA-synthesizer (Model 380A, Applied Biosystems (Foster City, Calif.). [0394]
  • 1. Construction of the MMTV Promoter-Luciferase fusion plasmid (pMluci) [0395]
  • The firefly luciferase gene was removed from the plant expression plasmid pDO432 (33) (FIG. 1) as a 1.9 kb BamHI fragment and cloned into the BamHI site of pSVL (Pharmacia, Piscataway, N.J.), a mammalian expression vector containing the SV40 promoter. The resulting plasmid (pSVLuci; FIG. 2) was digested with XhoI and SalI to produce a 2.4 kb fragment containing the luciferase coding sequences and the SV40 late polyadenylation site. This fragment was inserted into the XhoI site of pMSG (Pharmacia, Piscataway, N.J.), a eukaryotic expression vector containing the MMTV promoter. The resulting MMTV promoter-luciferase fusion plasmid (pMLuci; FIG. 3) was used to transfect NIH/3T3 cells as described below (section E1). Similar constructs can be made using luciferase vectors from Clontech (Palo Alto, Calif.). [0396]
  • 2. Construction of the human growth hormone (hGH) promoter-luciferase fusion plasmid [0397]
  • The SalI-XhoI fragment of pSVLuci (FIG. 2) containing the luciferase coding sequences and the SV40 late polyadenylation site was inserted into pUC 8 (Biorad, Richmond, Calif.), which had been linearized by a SmaI/HinCII digestion and ligated to XhoI linkers (New England Biolabs, Beverly, Mass.). The new plasmid thus generated (pUXLuci; FIG. 4) was linearized by XhoI digestion followed by incubation with the Klenow fragment of [0398] E. coli DNA polymerase and the four deoxyribonucleotides to fill in the single-stranded ends of the vector. This linear (5.1 KB) form of pUXLuci was then ligated to the filled-in 550 bP HindIII-XbaI fragment of the plasmid phGH:CAT (FIG. 5) (25). Human growth hormone promoter sequences located on the HindIII-XbaI fragment were thus fused to the luciferase coding sequences located on pUXLuci generating the plasmid phGH-Luci (FIG. 6), which was used in transfections of GC cells as described below (Section E2).
  • 3. Construction of the human Granulocyte-Colony stimulating factor (hG-CSF) Promoter-Luciferase fusion plasmid (pG-Luc1) [0399]
  • Information on the G-CSF upstream and coding sequences was published by Nagata et al. (1986) and was used to synthesize 5 oligonucleotide probes (OL-1 to OL-5) to screen a human leukocyte genomic DNA library (Clontech, Palo Alto, Calif.) according to the supplier's instructions. The sequences of the oligonucleotide probes were: [0400]
    (SEQ ID NO:1)
    5′ GCTTTTTGTTCCAACCCCCCTGCATT 3′ (OL-1);
    (SEQ ID NO:2)
    5′ CCCTGCATTGTCTTGGACACCAAAT 3′ (OL-2);
    (SEQ ID NO:3)
    5′ GCGCTCCAGGAGAAGCTGGTGAGT 3′ (OL-3);
    (SEQ ID NO:4)
    5′ AAGCTGATGGGTGAGTGTCTTGGC 3′ (OL-4); and
    (SEQ ID NO:5)
    5′ ATCAGCGGCTCAGCCTTCTT 3′ (OL-5).
  • The sequences of OL-1, OL-2 and OL-5 recognize the G-CSF promoter region, OL-4 recognizes the first intron/exon junction and OL-3 recognizes sequences within the second exon (32). One of the clones isolated from the leukocyte library using these oligonucleotide probes contains a 3.5 kb SalI-BamHI fragment of G-CSF genomic sequence consisting of 3.3 kb of promoter sequence and two hundred base pairs of the coding region. This fragment was inserted into the vector pGEM-7-Zf (Promega, Madison, Wis.) which had previously been digested with SalI/BamHI, resulting in the vector pJM710 (FIG. 7). pJM710 was then digested with PstI, and the resulting 1.6 kb fragment containing G-CSF upstream sequences and the first 15 bases of the G-CSF leader sequence was inserted into the PstI site of pGEM5-Luci (FIG. 8) to generate pG-Luc1 (FIG. 9). This construct was then used for transfections of 5637 human bladder carcinoma cells as described below in section E3. pGEM5-Luci (FIG. 8) had previously been constructed by inserting the XbaI/SalI fragment from pSVLuci (FIG. 2) containing the luciferase coding sequence and the SV40 late polyadenylation signal into PGEM 5-Zf (Promega, Madison Wis.) digested with XhoI/SalI. [0401]
  • C. Construction of the OSI Mammalian Expression Shuttle Vector
  • A mammalian expression shuttle vector was designed to allow the construction of the promoter-reporter gene fusions to be used in high-throughput screens to identify transcriptionally modulating chemicals. Features of the plasmid are shown in FIG. 10. The shuttle vector was constructed in several steps. Initially six oligonucleotides (pUV-1 through pUV-6) were synthesized (see FIG. 11[0402] a for sequence). The sequences of pUV-1, pUV-2 and pUV-3 correspond to a multicloning site, the b-globin leader sequence and the first 53 bases of the firefly luciferase coding region. The sequences of pUV-4, pUV-5 and pUV-6 are complementary to the first three oligonucleotides. The pUV oligonucleotides were annealed, ligated and inserted into the SalI/EcoRI sites of pTZ18R (Pharmacia, Piscataway N.J.) (FIG. 11(b)). The resulting vector was then digested with SmaI/PvuII and the oligonucleotide containing fragment was cloned into the bluescript KS(+) plasmid (Stratagene, La Jolla, Calif.), previously digested with PvuII, to yield pUV001 (FIG. 11(b)). Several fragments were ligated into pUV001 to create pUV100. The luciferase coding sequences (except first 53 bases) and polyadenylation site were obtained as a 1.8 kilobase XbaI/XmaI fragment from pMLuci (section B-1, FIG. 3). The SV40 early splice site and the SV40 late polyadenylation site were obtained as an 871 bp XmaI/BamHI fragment from pMSG (Pharmacia, Piscataway N.J., FIG. 12). Both DNA fragments were cloned into pUV001, previously digested with XbaI/BamHI to yield pUV100 (FIG. 12).
  • A 476 b fragment containing a dimeric SV40 polyadenylation site was then cloned into the BclI site of pUV100 (FIG. 13). To do this, a 238 bp BclI/BamHI fragment was obtained from SV40 genomic DNA (BRL), ligated, digested with BclI/BamHI, gel isolated, and inserted into pUV100, resulting in the vector pUV100-3 (FIG. 13). Linkers containing one SfiI and one NotI restriction site were then cloned into the PvuII/BamHI sites of pUV100-3. Two sets of linkers were synthesized containing the SfiI site in opposite orientations (oligonucleotides D-link1 and D-link2 and oligonucleotides R-link1 and R-link2). The sequences of the oligonucleotides were: [0403]
    (SEQ ID NO:6)
    5′ GATCGGCCCCTAGGGCCGCGGCCGCAT 3′ (D-link1);
    (SEQ ID NO:7)
    5′ ATGCGGCCGCGGCCCTAGGGGCC 3′ (D-link2); and
    (SEQ ID NO:8)
    5′ GATCGGCCCTAGGGGCGGCCGCAT 3′ (R-link1).
    (SEQ ID NO:9)
    5′ ATGCGGCCGCGGCCCCCTAGGGCC 3′ (R-link2)
  • The plasmid that contains D-link oligonucleotides was named pUV102 and the plasmid that contains R-link oligonucleotides was named pUV103 (FIG. 14). [0404]
  • The neomycin resistance gene (neo) was then placed under control of the Herpes Simplex Virus thymidine kinase (HSV-TK) promoter to generate a resistance cassette which is free of known enhancer sequences. To do this the HSV-TK promoter was synthesized using four oligonucleotides (FIG. 15) designed according to published sequence information (31), and including an [0405] SfiI restriction site 5′ of the HSV-TK sequences. These oligonucleotides were phosphorylated, annealed, ligated and inserted into pUV100 digested previously with HindIII/NheI, generating the vector pTKL 100 (FIG. 16). After verifying the HSV TK sequence, the about 3.5 kb NheI/SmaI fragment was isolated from pTKL100, and the about 0.9 kb BstBI/BglII fragment containing the neo coding region was isolated from pRSVNEO (14). These two fragments were filled in with Klenow polymerase and ligated to form pTKNEO (FIG. 17). An additional SfiI site was then inserted 3′ of the neo gene by isolating the about 1.8 kb SfiI/BamHI and about 2.6 kb SfiI/PVUII fragments of pTKNEO and conducting a three way ligation along with a synthesized SfiI oligonucleotide generating pTKNEO2 (FIG. 18). The HSV-TK/NEO vector containing an optimized Kozac sequence was also utilized (Stratagene, La Jolla, Calif., pMC1NEO). An additional vector was constructed by replacing the about 0.9 kb EcoRI/SalI fragment of pTKNEO2 with the about 0.9 kb EcoR1/SalI fragment from pMC1NEO. This vector was termed pTKNEO3. (FIG. 19).
  • D. Molecular Cloning of Hematopoietic Promoters and Insertion into the OSI Mammalian Expression Shuttle Vector
  • 1. Strategy [0406]
  • This section describes: (a) the molecular cloning of transcriptionally modulatable regulatory sequences of several genes of interest (in this case members of the family of hematopoietic growth factors) and (b) the making of constructs where these regulatory sequences now control the expression of the luciferase gene. To make such constructs, several kilobases of sequence upstream of the transcription start site, along with 5′ untranslated sequences up to the translation start site (ATG), of a gene of interest were inserted 5′ of the luciferase coding region. In this way constructs can be made where all sequences upstream of their translation start site are from the gene of interest, and all coding sequences are from the luciferase gene. How this was accomplished for the hematopoietic growth factor genes is described in sections D2-6. [0407]
  • 2. Human Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF) [0408]
  • Cloning of GM-CSF promoter sequences was performed by using oligonucleotide probes based on the GM-CSF genomic sequence (20). Two DNA oligonucleotide probes were synthesized, one corresponding to GM-[0409] CSF sequences 5′ of the coding region (5′ GGTGACCACAAAATGCCAGGGAGGCGGG 3′) (SEQ ID NO: 10) and the other to sequences in the first exon (5′GCAGGCCACAGTGCCCAAGAGACAGCAGCAGGCT 3′) (SEQ ID NO: 11). The oligonucleotide probes were used to screen a human leukocyte cell gnomic DNA library (Clontech, Palo Alto) Calif.) following the manufacturer's instructions. One clone was obtained which contains the entire GM-CSF coding region along with 2 kb of upstream sequences (see FIG. 20).
  • All routine subcloning procedures were performed as described by Maniatis et al. (1982), (28) unless otherwise noted. The about 0.7 kb HindIII/RsaI fragment of the GM-CSF clone (FIG. 20) was inserted into pUC 18 previously digested with HindIII/HincII. The about 0.7 kb HindIII/SmaI fragment was then isolated from the resulting vector and cloned into [0410] pUV 100 digested previously with HindIII/SnaBI, thereby generating pGMLUCI (FIG. 22.) In order to correctly fuse the GM-CSF ATG with the coding region of luciferase, four oligonucleotides (FIG. 21) were synthesized, phosphorylated, annealed, ligated, and inserted into pUC19 previously digested with Eco RI/XbaI, generating pGM-1 (FIG. 22). pGM-1 was then sequenced (Sequenase Kit, US Biochemicals, Cleveland, Ohio) using the M13 forward (U.S. Biochem.) and reverse primers (Pharmacia, Piscataway, N.J.) to ensure that there were no mutations in the synthesized oligonucleotides. The about 1.8 kb BstEII/ScaI fragment from pGM-1 was then isolated and ligated to about 1.5 kb BstEII/ScaI fragment from PGMLUCI to generate pGM-2 (FIG. 22). pGM-2 was then digested with Hind III/XbaI and about 0.7 kb fragment was cloned into pUV 102 and pUV 103 previously digested with HindIII/XbaI. This generated pGMLS102 and pGMLS103 (FIG. 22), which contain 663 bp of GM-CSF sequence 5′ of the ATG fused directly to the second (correct) ATG of luciferase and the rest of the luciferase coding region. An additional about 1.4 kb of upstream sequences were cloned into this construct by isolating the about 1.4 kb SalI/HindIII fragment from the GM-CSF clone (FIG. 20), blunting the SalI end by filling in with Klenow polymerase, and inserting the fragment into pGMLS102 and pGMSL103 previously digested with HindIII/SnaBI. This step generated pGMLL102 and PGMLL103, respectively (FIG. 23). Finally, the pTKNEO2 and PTKNEO3 about 1.8 kb SfiI fragments were cloned directly into the SfiI site of pGMLL103 to generate pGMLL103 NEO2 and pGMLL103 NEO3.
  • 3. Human Macrophage Colony Stimulating Factor (M-CSF or CSF-1) [0411]
  • Sequence information on the M-CSF gene (23) was used to synthesize an oligonucleotide probe (CSF1-a) to screen a human leukocyte genomic DNA library (Clontech, Palo Alto, Calif.) according to the supplier's instructions. The sequence of the oligonucleotide probe was: [0412]
  • 5′ CCGGCGCGGTCATACGGGCAGCTGG 3′ (CSF1-a) (SEQ ID NO: 12)
  • The sequence of this probe corresponds to sequences within the second exon of the M-CSF gene. One of the clones isolated from the leukocyte library contains a 5 kb EcoRI/HindIII fragment which includes the first exon and 5′ flanking region of M-CSF. This fragment was inserted into the pTZ18R vector (Pharmacia, Piscataway N.J.) which had been previously digested with EcoRI/HindIII resulting in the vector CSF1-pTZ18 (FIG. 24). The same fragment was isolated from CSF1-pTZ18, blunt ended at the EcoRI end, and inserted into the pUV100 vector which had been previously digested with SnaBI/HindIII, resulting in the vector p100-RH (FIG. 25). The M-CSF untranslated leader sequence (23) was then fused to the first codon of the luciferase coding region as follows: (a) a 740 bp PstI/PvuII fragment was isolated from CSF1-pTZ18 containing 570 bp of the M-CSF promoter and 170 bp of the untranslated leader sequence; (b) oligonucleotides containing sequences from the 3′ end of the M-CSF leader sequence and the 5′ end of the luciferase coding region were synthesized: [0413]
    5′ CTGCCCGTATGGA 3′ (CSF-luci5) (SEQ ID NO:13)
    5′ ACGGGCAG 3′ (CSF-luci6);
  • (c) oligonucleotides pUV3 and pUV6 (previously used to construct pUV001, FIG. 11([0414] b)) were annealed, and digested with XbaI to release a 48 bp fragment which contains 48 bases of the luciferase coding region; (d) DNA fragments and oligonucleotides (from a, b and c) were ligated and inserted into pUV100 previously digested with PstI/XbaI to yield p100-2 (FIG. 26). A construct containing a larger M-CSF promoter fragment (5 kb) was also made. A 2 kb XmaI fragment was isolated from the plasmid p100-2. This fragment contains the 3′ end of the M-CSF leader sequence fused to the luciferase start codon. The 2 kb XmaI fragment was inserted in p100-RH previously digested with XmaI, to yield p100-RHC (FIG. 27). The fused 5 kb M-CSF promoter-luciferase construct was then inserted into pUV102 as follows: a 5 kb NotI/XbaI fragment (blunt ended at the NotI end) was isolated from p100-RHC and inserted into pUV102, previously digested with SnaBI/XbaI, to generate CSF1-102 (FIG. 28). This construct was then used for transfections of 5637 human bladder carcinoma cells.
  • 4. Human Granulocyte Colony Stimulating Factor (G-CSF) [0415]
  • Cloning of G-CSF sequences is described in Experimental Details, section B-3. In order to correctly fuse the G-CSF upstream sequences to the luciferase start codon, oligonucleotides were synthesized which contain the G-CSF leader sequence from +15 to the ATG (32), and were cloned into pUC19 to create pGUC84 (FIG. 29). The sequence of the inserted fragment was determined and was found to be as expected. The G-CSF-oligonucleotide-containing NcoI/ScaI fragment from pGUC84 was then isolated and ligated to the luciferase-containing NcoI/ScaI fragment from pUV100 to create pGUV100 (FIG. 30). Following this, the PstI fragment of the G-CSF promoter was isolated from pJM710 (FIG. 7) and inserted into the PstI site in pGUV100 generating pGUV1 (FIG. 31). The rest of the G-CSF promoter clone was added by ligating the G-CSF-luciferase containing SfiI/ScaI fragment from pGUV1 to the appropriate SfiI/ScaI fragment from pJM710, creating the plasmid pGUV2 (FIG. 32). The XbaI fragment from pGUV2 containing the G-CSF-luciferase fusion was then cloned into pUV103 previously digested with XbaI/SpeI, generating pGUV140 (FIG. 33). Finally, a TK-Neo cassette (Section C) was included by ligating the ScaI/XbaI fragment from pGUV140 which contains the G-CSF-luciferase fusion into pTKNEO103 previously digested with ScaI/XbaI, yielding the final vector pGUV150 (FIG. 34). [0416]
  • 5. Erythropoietin (EPO) [0417]
  • Information on the EPO upstream and coding sequences has been published (27) and was used to synthesize two oligonucleotide probes (EPO6 and EPO8) to screen a human leukocyte genomic DNA library (Clontech, Palo Alto, Calif.) according to the supplier's instructions. The sequences of the oligonucleotide probes were: [0418]
    5′AATGAGAATATCACTGTCCCAGACACCAAAGTTAATTTCTATGCCTGGAA 3′ (EPO08); and (SEQ ID NO:14)
    5′TTCCAGGCATAGAAATTAAC 3′ (EPO6). (SEQ ID NO:15)
  • EPO8 is complementary to sequences within the third exon of the EPO gene (27). EPO6 is complementary to the 3′ end of EPO8 and was used as a primer for filling in the complementary strand of EPO8 with labelled nucleotides, thereby generating a probe for cloning. One of the clones isolated from the leukocyte genomic DNA library contained a 7.5 kb BamHI fragment consisting of 6.2 kb of the EPO promoter region and the first three EPO exons. This fragment was inserted into the plasmid Bluescript KS(+) (Stratagene, La Jolla, Calif.), previously digested with BamHI, resulting in the vector pEP-7.5B (FIG. 35). The Epo leader sequence was fused to the start codon of the luciferase gene by using four synthetic oligonucleotides (EPO9 to EPO12). The sequences of the oligonucleotides were: [0419]
    5′CCCGGTGTGGTCACCCGGCGCGCCCCAGGTCGCTGAGGGACCCCGGCCAGGCGCGGA 3′ (EP09); (SEQ ID NO:16)
    5′CATCTCCGCGCCTGGCCGGGGTCCCTCAGCGACCTGGGGCGCGCCGGGTGACCACACCGGGGGGCC 3′ (EP010); (SEQ ID NO:17)
    5′GATGGAAGACGCCAAAAACATCAAGAAAGGCCCGGCGCCATTCTATCCT 3′ (EP011); and (SEQ ID NO:18)
    5′CTAGAGGATAGAATGGCGCCGGGCCTTTCTTGATGTTTTTGGCGTCTTC 3′ (EP012). (SEQ ID NO:19)
  • The sequences of EPO9 and EPO11 consist of 63 bases upstream of the EPO translational start site fused to the first 53 bases of the luciferase coding region. EPO10 and EPO12 oligonucleotides are complementary to EPO9 and EPO11, respectively. These oligonucleotides were inserted into the plasmid pUV100 previously digested with ApaI/XbaI to generate the vector pRE (FIG. 36). 6 kb of EPO upstream sequence was cloned into pUV102 by inserting a 6.2 kb BamHI/partial ApaI fragment from pEP-7.5 B and a 1.7 kb ApaI/XhoI fragment from pRE into pUV102 previously digested with BglI/XhoI, yielding pUV2-EP6 (FIG. 37). The 1.8 kb SfiI fragment from pTKNEO3 (FIG. 19) was then inserted into pUV2-EP6 previously digested with SfiI, generating pEP6.0-102-TKNEO. [0420]
  • 6. Interleukin-3 or Multi-CSF (IL-3) [0421]
  • Information on the IL-3 promoter and coding sequences (40-41) was used to synthesize an oligonucleotide probe (IL-3 II) to screen a human leukocyte genomic DNA library (Clontech, Palo Alto, Calif.) according to the supplier's instructions. The sequence of the oligonucleotide probe was: [0422]
  • 5′ TAAGTGTGTTATAATTTCATCGATCATGTT 3′ (IL-3 II) (SEQ ID NO: 20)
  • which corresponds to sequences within the first exon of IL-3. One of the clones isolated from the leukocyte library using the IL-3 II probe contained an 8 kb HindII/EcoRI fragment of IL-3 sequence consisting of 6.4 kb of upstream sequences and 2 kb of the coding region. This fragment was inserted into the vector pTZ18R (Pharmacia, Piscataway N.J.) previously digested with HindIII/EcoRI, resulting in the vector IL-3-pTZ18R (Eco-Hind) (FIG. 38). The IL-3 leader sequence was fused to the first codon of the luciferase gene as follows. A 900 bp PstI fragment was isolated from IL-3-pTZ18R (Eco-Hind). This fragment contains 700 bp of the IL-3 promoter along with [0423] exon 1 of IL-3 (FIG. 39), and was inserted into PTZ18R (Pharmacia, Piscataway, N.J.) previously digested with PstI, resulting in the vector IL3/PstI-pTZ18R (FIG. 39). Four oligonucleotides (IL-lu-1 to IL-lu-4) were synthesized, with the following sequences:
    5′CCGGGGTTGTGGGCACCTTGCTGCTGCACATATAAGGCGGGAGGTTGTTGCCAACTCTTC3′ (IL-lu-1); (SEQ ID NO:21)
    5′AGTTGGCAACAACCTCCCGCCTTATATGTGCAGCAGCAAGGTGCCCACAACC 3′ (IL-lu-2); (SEQ ID NO:22)
    5′AGAGCCCCACGAAGGACCAGAACAAGACAGAGTGCCTCCTGCCGATCCAAACATGGA 3′ (IL-lu-3); and (SEQ ID NO:23)
    5′GTTTGGATCGGCAGGAGGCACTCTGTCTTGTTCTGGTCCTTCGTGGGGCTCTGAAG 3′ (IL-lu-4). (SEQ ID NO:24)
  • The sequences of IL-lu-1 and IL-lu-3 correspond to 112 bases of the 3′ end of the IL-3 promoter fused to the first 5 bases of the luciferase coding region. IL-lu-2 and IL-lu-4 oligonucleotides are complementary to IL-lu-I and IL-lu-3, respectively. Oligonucleotides IL-lu-1 to IL-lu-4 along with oligonucleotides pUV-3 and pUV-6 (see Section C) were annealed, ligated and inserted into IL3/PstI-pTZ18R previously digested with XmaI/EcoRI, to generate pTZ-IL-3-Luci (XmaI-XbaI) (FIG. 39). A 6 kb IL-3 promoter fragment was cloned into pUV102 as follows: a 500 kb NcoI/EcoRI fragment was isolated from pTZ-IL-3-Luci (XmaI/XbaI) and inserted into IL-3-pTZ18R (EcoRI/HindIII) previously digested with NcoI/EcoRI to yield pTZ-IL3-C (FIG. 40). A 6.4 kb HindIII/XbaI fragment was then obtained from pTZIL3-C and inserted into pUV102 previously digested with HindIII/XbaI, resulting in the vector pUV-IL-3 (FIG. 41). Finally, the 1.8 kb SfiI fragment from pTKNEO3 (FIG. 19) was inserted into pUV-IL-3 previously digested with SfiI, generating the vector pIL3-102-TKNEO. [0424]
  • E. Construction of Single Cell Clones Containing Various Promoter-Luciferase Fusion Constructs
  • 1. pMluci [0425]
  • pMluci (FIG. 3) and pSV2Neo, an antibiotic resistance plasmid (34), were co-transfected into NIH/3T3 mouse fibroblast cells using the calcium phosphate precipitation method (15) with a commercially available kit (Pharmacia, Piscataway N.J.). Two days later, cells were transferred to media containing 0.4 mg/ml G418 and were grown for an additional 10-14 days. G418-resistant clones were isolated by standard methods. Once sufficient cell numbers were obtained, clones were analyzed based on several criteria: constitutive luciferase production, induction of luciferase expression by dexamethasone (1 μm, Sigma, St. Louis, Mo.), satisfactory attachment to microtiter plates used in the high-throughput screen (see section G) and acceptable standard deviation in multiple luciferase expression assays (see below for assay protocol). This analysis was carried out using the luciferase assay conditions described in sections F and G. Of the clones which satisfied the above criteria for the high throuphput screen, one clone, M10, was selected for use. [0426]
  • 2. phGH-LUCI [0427]
  • phGH-LUCI (FIG. 6) and pRSVNeo, an antibiotic resistance plasmid (14), were co-transfected into GC rat pituitary cells as described above. Selection of G418-resistant cell clones was described above except for using a concentration of 0.2 mg/ml G418. Analysis of the cell clones was performed as above, except that known inducers of hGH expression (10-100 nM rat growth hormone releasing factor (RGRF, Bachem, Torrance, Calif.) and 10 μm forskolin (Sigma, St. Louis, Mo.) were used in place of dexamethasone. One clone, 532, was selected for further use in the high throuhgput screen. [0428]
  • 3. pG-LUC1 [0429]
  • pG-LUC1 (FIG. 9) and pRSVNeo were co-transfected into 5637 human bladder carcinoma cells as described above. Selection of G418 resistant clones was as described above except for using a concentration of 0.1 mg/ml G418. Analysis of cell clones was performed as above except that a known inducer of G-CSF expression (1-5 lg/ml lipopolysaccharide (LPS), [0430] E. coli serotype 055:b5, Difco, Detroit, Mich. or Sigma, St. Louis, Mo.) was used in place of dexamethasone. One clone, G21, was selected for use.
  • 4. pGVU150 and pGVU140/pTKNEO3 [0431]
  • All the following constructs were transfected into 5637 bladder carcinoma cells either by electroporation using a BRL (Gaithersburg, Md.) Cellporator electroporation device or by lipofection using BRL lipofectin and following the manufacturer's protocol. For electroporation, cells were trypsinized, treated with Soybean trypsin inhibitor (1 mg/ml), washed three times in Dulbecco's modified Eagle's medium (DMEM) without pH indicator, and 1 ml of cell suspension in DMEM was electroporated at room temperature and at a cell density of 5 million cells per ml at a voltage of 250 V and a capacitance of 1180 microFarad with the electroporation device set at low resistance. In previous experiments these parameters had been determined to result in optimal electroporation efficiency. About 15 min after electroporation cells were recovered from the disposable electroporation containers (BRL, Gaithersburg, Md.) and plated in RPMI containing 10% fetal calf serum. 2 days after electroporation 0.4 mg/ml G 418 were added and clonal cell colonies were picked 14-20 days after transfection. Clones were first analysed for luciferase expression using [0432] Lysis Buffer 1 without bovine serum albumin (see Section F) and those testing positive expanded further and frozen in liquid nitrogen. Further analysis of 6 clones (G 1002, G 2005, G 2071, G 2085, G 3014, G3031) included Southern blotting, reaction to known inducers, satisfactory attachment to microtiter plates and acceptable standard deviation in multiple luciferase expression assays. Clones G 2005 and G 2085 were derived from 5637 cells lipofected with 100 ug/ml of linearized pGVU150 (FIG. 34), G 3031 descends from cells electroporated with 100 ug/ml of linearized pGVU150, whereas 200 ug/ml of the same plasmid were used to generate G 3014. Cotransfection of circular pGVU140 (FIG. 33) with circular pTKNeo3 (FIG. 19) was used to generate clones G 1002 and G 2071. While G 1002 resulted from co-electroporation of 50 and 5 ug/ml of the two plasmids, G 2071 descends from cells lipofected with 100 and 10 ug.
  • 5. pCSF1-102/pTKNeo3 [0433]
  • 3 clones generated by co-electroporation with linearized pCSF1-102 (FIG. 28) and linearized pTKNeo3 (FIG. 19) were subjected to further analysis as outlined above: M 2071 (6 ug pCSF1-102; 0.5 ug pTKNeo3), M 2085 and M 2086 (both: 10 ug pCSF1-102 and 1 ug pTKNeo3). [0434]
  • 6. pGMLL103 NEO3 [0435]
  • 6 clones generated by electroporation with pGMLL103 NEO3 (FIG. 23; Materials and Methods, section D.2) were subjected to further analysis as described above: GM 1073 (10 ug; circular); [0436] GM 1081, 1088 and 1090 (5 ug; linear); and GM 1098 and 1105 (10 ug; linear).
  • F. Liquid Scintillation Counter Bioluminescence Assay
  • To assay for luciferase expression in transient expression assays in the various transfected clones, cells were incubated with various transcriptional inducers in serum free defined media, washed 3 times with Dulbecco's phosphate-buffered saline (D-PBS, Gibco) and lysed in Lysis Buffer 1 (50 mM Tris acetate pH 7.9, 1 mM EGTA, 10 mM magnesium acetate, 1 mg/ml bovine serum albumin [BSA], 0.5[0437] % Brij 58, 2 mM ATP, 100 mM dithiothreitol [DTT]). All reagents were obtained from Sigma except for DTT which was from Boehringer Mannheim. After lysis, cell debris was sedimented by brief centrifugation, and 950 ll of supernatant extract were added to a glass scintillation vial. Samples were counted individually in an LKB (Gaithersburg, Md.) scintillation counter on a setting which allows measurement of individual photons by switching off the coincidence circuit. The reaction was started by addition of 5011 of 2 mM luciferin (Sigma, St. Louis, Mo. or Boehringer Mannheim, Indianapolis Ind.) in Buffer B (Buffer B-Lysis Buffer 1 without Brij 58, ATP and DTT) to the 950 ll of lysate. Measurement was started 20 seconds after luciferin addition and continued for 1 minute. Results were normalized to protein concentration using the Bradford protein assay (BioRad, Richmond Calif.) or to cell numbers using Trypan Blue (Sigma) exclusion counting in a hemocytometer (see section G).
  • G. High-Throughput (HTP) Screening
  • Cell Plating: Dynatech Microlite 96 well plates were custom pretreated for cell attachment by Dynatech Laboratories, Inc. (Chantilly, Va.). Alternatively, the 96 well plates were treated with 50 ll per well of human fibronectin (hFN, 15 lg/ml in PBS, Collaborative Research, Bedford, Mass.) overnight at 37C. hFN-treated plates were washed with PBS using an [0438] Ultrawash 2 Microplate Washer (Dynatech Labs), to remove excess hFN prior to cell plating. M10, 532, and G21 cells maintained in their respective serum media (with 0.2 mg/ml G418) were washed with PBS, harvested by trypsinization, and counted using a hemocytometer and the Trypan Blue exclusion method according to protocols provided by Sigma, St. Louis, Mo. Chemical Company. Cells were then diluted into serum free defined media (with 0.2 mg/ml G418), and 0.2 ml of cell suspension per well was plated onto Dynatech treated plates (532 and G21) or hFN-treated plates (M10) using a Cetus Pro/Pette (Cetus, Emeryville Calif.). Plates were incubated overnight at 37C in a humidified 5% CO2 atmosphere.
  • Addition of Chemicals to Cells: Chemicals from the Oncogene Science file were dissolved in DMSO at concentrations of 3-30 mg/ml. A liquid handling laboratory work station (RSP 5052, Tecan U.S. Chapel Hill, N.C.) was used to dilute the chemicals (three dilutions; 5 fold, 110 fold, and 726 fold). 10 ll of each dilution were added to each of quadruplicate samples of cells contained in the wells of 96-well Dynatech Microlite Plates. Cell plates were then shaken on a microplate shaker (Dynatech, medium setting, 30 sec.) and incubated for 6 hours at 37C, 5% CO[0439] 2.
  • Bioluminescence Assay: After incubation with OSI-file chemicals, cell plates were washed 3 times with PBS using an [0440] Ultrawash 2 Microplate Washer (Dynatech Labs) and 75 ll of Lysis Buffer 2 were added to each well (Lysis Buffer 2 is the same as Lysis buffer 1 exept that the ATP and DTT concentrations were changed to 2.67 mM and 133 mM, respectively). Bioluminescence was initiated by the addition of 25 ll 0.4 μm Luciferin in Buffer B to each well, and was measured in a Dynatech ML 1000 luminometer following a 1 minute incubation at room temperature. Data were captured using Lotus-Measure (Lotus) software and processed by custom-designed macros written in Lotus.
  • More recently the cell lysis buffer was modified to also contain the luciferin. Therefore, lysis of cells and the bioluminescence reaction begin simultaneously and the production of bioluminescent light reaches a maximum at about 5 min. The level of light output declines by about 20% within further 30 min. For better lysis buffer stability bovine serum albumin has been omitted. This improved lysis buffer has been shown to remain fully functional for at least 12 hours, when kept on ice and protected from direct light. [0441]
  • Also, more recently, a fully automated device as described in U.S. patent application Ser. No. 382,483 was used to incubate luciferase reporter cells in 96-well microtiter plates, transfer chemicals and known transcriptional modulators to the cells, incubate cells with the chemicals, remove the chemicals by washing with PBS, add lysis buffer to the cells and measure the bioluminescence produced. [0442]
  • H. Isolation of Total Cellular RNA
  • Total cellular RNA was isolated from the G21 cell clone or from untransfected 5637 cells following incubation for 6 hours with various transcriptionally modulating chemicals identified in the high-throughput screen. Cells were grown in serum free medium as described above. Total cellular RNA was isolated using the RNAZol method (CINNA/BIOTECX, Friendswood, Tex., Laboratories International, Inc.). Cells were resuspended and lysed with RNAZol solution (1.5 ml/9 cm petri dish) and the RNA was solubilized by passing the lysate a few times through a pipette. Chloroform was added to the homogenate (0.1 ml/1 ml), and samples were shaken for 15 seconds followed by a 5 minute incubation on ice. After centrifuging for 10 minutes, the upper phase was collected and an equal volume of isopropanol was added. Samples were incubated for 45 minutes at −20C, and the RNA was pelleted for 15 minutes at 12,000×g at 4C. The RNA pellet was then washed with 70% ethanol and dried briefly under vacuum. [0443]
  • I. Northern Blotting
  • Total cellular RNA was isolated from 5637 cells following incubation with chemicals as described above and electrophoresed in a 1% Agarose-μm Formaldehyde gel. The RNA was transferred to Duralon-UV nylon filters (Stratagene, La Jolla, Calif.) using the manufacturer's recommended protocol. The filters were prehybridized for 4 hours (prehybridizing solution=5×SSC, 50 mM sodium pyrophosphate, 10×Denhardt's solution, 10% dextran sulfate, 7% SDS and 250 lg/ml denatured ssDNA) and then hybridized in the same solution for 16 hours at 65C in the presence of G-CSF or Beta-Actin (Oncor, Gaithersburg, Md.) specific probes. The G-CSF probe was a 0.6 kb AflII to XhoI fragment which contained most of [0444] exon 5 of the human G-CSF gene. The B-actin probe was used as a control probe to normalize for the total amount of RNA. The probes were labeled with alpha-32P dCTP using a random primed DNA labeling kit (Amersham, Arlington, Ill.). Following hybridization, filters were first probed with, G-CSF-and Fusion reprobed with B-Actin Probe were washed three times at room temperature with 1×SSC, 0.13% SDS and three times at 65C with 0.2×SSC, 0.1% SDS. Filters were first probed with G-CSF-and then reprobed with B-actin-probe. Exposure to x-ray fiμms was performed overnight. Bands were excised and counted in a liquid scintillation counter (LKB, Gaithersburg, Md.), and counts obtained with the G-CSF specific probe were normalized relative to the counts obtained with the B-Actin specific probe.
  • J. S1 Nuclease Protection
  • S1 Nuclease protection assays were carried out as described in reference 94. [0445]
  • K. Southern Blotting
  • To monitor correct and complete stable integration of transfected promoter/reporter constructs, stably transfected cell clones were subjected to Southern blot analysis (92). Genomic DNA was prepared of each clone to be tested and restriction-cut with Dra I. After electrophoresis, transfer to nylon filters and immobilization by UV irradiation using a Stratalinker UV device (Stratagene, La Jolla, Calif.), integrated promoter/luciferase fusion constructs were visualized by probing with radioactively labelled XbaI-EcoRI fragments of the luciferase coding region. Probes were labelled using the random primer method (93). Since Dra I cuts in the [0446] SV 40 polyadenylation sites located in the OSI mammalian expression shuttle vector just upstream the inserted promoter sequences as well as downstream of the luciferase coding region, but not in any of the 3 promoter sequences used for generating stably transfected cell clones, a single fragment should be visualized by the probe used. The size of that fragment should be characteristic for each of the three promoter sequences analyzed.
  • L. MTT Cell Toxicity Assay
  • To determine cytotoxic concentrations of chemicals registering as positives in the High-Throughput luciferase assay the MTT cytotoxicity assay was employed (95). In this assay, a tetrazolium salt [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide, MTT] is reduced to a colored formazan product by reducing enzymes present only in living metabolically active cells. [0447]
  • M. Two-Antibody Sandwich Immunoassay
  • Supernatants from 5637 bladder carcinoma cells incubated with chemicals registering as positives in the G-CSF promoter/luciferase High-Throughput assay were assayed for secreted G-CSF protein using the two-antibody sandwich immunoassay (96). The G-CSF Assay kit manufactured by Oncogene Science, Inc. was used and the manufacturer's instructions were followed. [0448]
  • N. Construction of a Yeast Expression Vector
  • Plasmid pHZ18 (97, 98) contains 2μ DNA for propagation in [0449] S. cerevisiae, the yeast promoter cyc 1, which is compatible with expression in yeast cells, and the URA 3 gene for selection. The plasmid was linearized with BamH1, the ends were filled-in using deoxynucleotides and E. coli DNA polymerase Klenow fragment, and then the plasmid was digested with Aat II. A 4.1 kb fragment containing the cyc1 promoter, URA3 and 2μ genes was separated by agarose gel electrophoresis and subsequently purified by electroelution onto ion-exchange paper (Whatman, DE81). Plasmid pBR322 was treated with endonucleases Aat II and Pvu II and a 2.2 kb fragment containing the plasmid's origin of replication and the ampR gene was isolated by agarose gel electrophoresis and eluted onto DE81 paper.
  • The 2.2 kb pBR322 and 4.1 kb pHZ18 fragments were ligated using T4 DNA ligase according to standard procedures (94). The resulting 6.3 kb vector pHZBR was digested with BamH1 for subsequent insertion of the luciferase coding sequence downstream of the cyc1 promoter. [0450]
  • An Nco I-Sal I fragment of pUV102 containing the luciferase gene starting at the second ATG, was made blunt-ended by filling in and ligated into the filled-in BamH1 site of pHZBR. Clones of the correct orientation were identified via restriction mapping to yield plasmid pHZluci24. This plasmid was used to transform [0451] S. cerevisiae strain DB745.
  • O. Transformation of Yeast Cells
  • [0452] S. cerevisiae DB745 were made competent according to published methods (99). One and 4 μg of either pHZluci24 or pHZ18 (transfection control) were added to the competent cells and incubated at 30° C. for 30 minutes. Lithium acetate-PEG was mixed gently with the cells and allowed to incubate for another 45 minutes at which time the cells were shifted to 42° C. for 5 minutes. The cells were spread onto uracil(−) plates and incubated at 30° C. for several days. Cell colonies were picked, grown to saturation in YPD media and analyzed for luciferase activity. Stock cultures were made from positive clones, and each was subsequently analyzed for suitability for the 96-well plate high-throughput assay.
  • P. Luciferase Bioluminescence Assay in Microtiter Plates
  • Expression of the firefly luciferase gene was determined by measuring luminescence in the presence of substrates essentially as described above. [0453]
  • Formatting the assay to a 96-well plate required optimization of cell lysis conditions, substrate concentrations and the reaction measurement time. Initial experiments were conducted using purified luciferase and substrates. Bioluminescence was measured either by scintillation counting or in a Dynatech ML1000 luminometer and the reaction conditions were optimized to provide the highest signal-to-noise ratio. Cell lysis conditions were optimized to result in complete lysis of the cells yet not interfere with the luciferase reaction. The detergent Brij 58 fulfilled these requirements. In the current format the 96-well assay was carried out as follows: [0454]
  • 1-2×10[0455] 4 yeast cells were seeded into 96-well plates which have been custome designed to allow filtration of the media while retaining the cells, and which are opaque to permit analysis using a luminometer (Millipore). After the media was removed from the cells by suction, 100 μl of lysis buffer (50 mM Tris/acetate pH 7.9, 1 mM EGTA, 10 mM Mg-acetate, 0.5% Brij 58, 100 mM DTT and 4 mM ATP, 0.2 mM Luciferin, 800 U/ml lyticase) was added and the plates were incubated at room temperature for 10 minutes. Bioluminescence was monitored in a Dynatech ML1000 luminometer.
  • RESULTS A. Validation of Luciferase Reporter Technology
  • Linearity of the luciferase reporter system [0456]
  • Various_amounts of luciferase (0, 20, 40, 80, 160, 320, 640, 1280 femtograms; 20 femtograms correspond to 200,000 molecules) in [0457] Lysis buffer 1 were added to 24 96-well microtiter plates (0 and 1280 femtograms 576 replicate wells, all other concentrations 192 replicate wells). Bioluminescence reactions were started and light output measured as described in Materials and Methods, Section G. The 24 replicate plates were processed in a random order determined by a random number generator. Mean bioluminecent signals and standard deviations obtained from 576 or 192 replicate measurements (see above) were plotted against luciferase concentration (FIG. 48; Reporter concentration 50 corresponds to 1280 femtograms luciferase). The signal was shown to be proportional to luciferase concentration over an at least 50fold range with a linear correlation coefficient of r=0.99. The assay was highly accurate and reproducible as shown by the small standard deviations obtained from a total of 2,304 individual measurements obtained in a randomized fashion. As few as 200,000 molecules of luciferase can be detected in one well of a 96-well microtiter plate. When 10,000 cells expressing luciferase are plated into one well, as few as 20 molecules of luciferase per cell can thus be detected in a simple, high-throughput, fully automatable format. Thus the novel lysis buffer described in Materials and Methods, Sections F and G, allows unique sensitivity and stability, accuracy and reproducibility of the luciferase-catalysed bioluminescent reaction and facilitates easy automation of the luciferase transcriptional reporter system. In all these qualities, the luciferase assay system using the lysis and reaction buffer described above not only surpasses other available luciferase assays but all other available transcriptional reporter technologies.
  • In vivo signal half-life of the luciferase reporter system [0458]
  • When screening for inhibitors rather than inducers of transcription, the half-life of the reporter molecule becomes a crucial parameter in determining the minimal incubation time that would be necessary to allow enough decay of reporter molecules so that the inhibition of their synthesis became visible. The cell lines G1002, GM1074 and CM1 containing luciferase reporter constructs for the G-CSF, GM-CSF or the immediate early promoter of cytomegalovirus were therefore tested for the time dependency of luciferase activity after treatment of the cells with Actinomycin D, an inhibitor of transcription. This experiment measures the combined half-life of luciferase mRNA and of the luciferase protein. Cells derived from [0459] clones G 1002, GM 1074 and CM 1 were seeded into 96-well microtiter plates (20,000 cells G 1002 or GM 1074/well and 5000 cells CM 1/well) and incubated overnight in cell culture conditions. At time 0 Actinomycin D (25 microg/ml) was added. At the times indicated in FIG. 49 cells were washed with PBS and luciferase activity of Actinomycin-treated cells determined as described in Materials and Methods, Section G. was compared to the luciferase activity of untreated controls. The logarithm of the treated/untreated ratio is plotted versus time.
  • As demonstrated in FIG. 49, apparent half-lifes found in the three cell lines tested ranged from about 2.5 to 6 hours. [0460]
  • A 24 hour incubation with a 100% efficient inhibitor of transcription would therefore be sufficient to reduce luciferase levels to maximal 6% of the control in the tested cell lines. [0461]
  • B. Validation of Cell Lines
  • Prior to initiation of drug screening, it was demonstrated that the transfected promoter-luciferase fusion plasmids were reacting to transcriptional inducers in a manner as predicted based on the published literature. As shown in FIG. 42, all three transfected cell clones chosen responded to inducers which have been reported to stimulate the endogenous genes; the MMTV-luciferase containing clone M10 was stimulated 11.6 fold by 1 μm dexamethasone, the hGH-[0462] luciferase containing clone 532, was stimulated 2.2 fold by 100 nM rat growth hormone releasing factor (GRF), and the hG-CSF containing clone G21 was stimulated 5.7 fold by 5 lg/ml lipopolysaccharide (LPS).
  • It was also demonstrated that certain steroidal chemicals other than dexamethasone modulated luciferase expression in the cell clone M10, which harbours the MMTV promoter-luciferase fusion construct. As shown in FIG. 43, dexamethasone stimulated the MMTV promoter in cell clone M10 (mouse fibroblast origin), while progesterone did not. It has been shown that a rat fibroblast cell line which contains high levels of glucocorticoid receptor but low levels of progesterone receptor, shows stimulation of the MMTV promoter by the glucocorticoid dexamethasone but not by progesterone (7). In additon, FIG. 43 shows that the mineralocorticoid aldosterone stimulates clone M10, as is expected based on previously published work which indicates that aldosterone can act through the glucocorticoid receptor to stimulate the MMTV promoter (6). [0463]
  • Cell clones transfected with the OSI mammalian expression shuttle vector fused to the G-CSF, GM-CSF and M-CSF promoters (Materials and Methods, Sections E.4, E.5 and E.6) were analysed for correct and complete integration of the promoter/luciferase constructs by Southern blotting. Genomic DNA was prepared of each clone to be tested and restriction-cut with Dra I. Blots were probed with XbaI-EcoRI fragments of the luciferase coding region. Since Dra I cuts in the [0464] SV 40 polyadenylation sites located in the OSI mammalian expression shuttle vector just upstream the inserted promoter sequences as well as downstream of the luciferase coding region, but not in any of the 3 promoter sequences used for generating stably transfected cell clones, a single fragment should be visualized by the probe used. The size of that fragment should be characteristic for each of the three promoter sequences analyzed.
  • As shown in FIG. 50, G-[0465] CSF clones G 1002, G 3014 and G 3031, GM-CSF clones GM 1073, GM 1088 and GM 1105 and all 3 M-CSF clones tested contained only a single fragment with the correct molecular size (uppermost fragments in plasmid controls G/5637, M/HL 60 and GM/5637 generated by loading mixtures of the purified plasmids and extracts of the parental cell lines 5637 or HL 60 on the gel). The 2 smaller fragments are non-specific, cross-hybridizing probe impurities. The other clones all contained additional rearranged fragments of various molecular sizes. Conspicously, all 3 G-CSF clones with correctly integrated promoter/reporter constructs were derived from electroporation, whereas the other G-CSF cell clones analysed were obtained either by lipofection (G 2005, G 2071 and G 2085) or calcium phosphate precipitation (G 21; see Materials and Methods). Interestingly, G 21 cells contain multiple copies of the promoter/luciferase construct, the majority of which migrate at approximately correct molecular weights. The data suggest that electroporation under the optimized conditions described is the transfection method of choice to obtain cell clones with correctly integrated, complete promoter/luciferase reporter constructs.
  • Cell clones with correctly integrated promoter/reporter constructs were analysed for correct reaction to known transcriptional inducers. Of the G-CSF clones tested, G1002 showed the most consistent levels of induction after 10.5 hours of incubation in serum-containing media with 8.3 ng/ml tumor necrosis factor-alpha (TNF-alpha; 2fold), 20 ng/ml phorbol-myristate-acetate (PMA; 4.4fold), 0.5 ng/ml Interleukin-1 beta (2.6fold), a mixture of 4.2 ng/ml TNF-alpha and 0.3 ng/ml Interleukin-1 beta (3.8fold) and a mixture of 4.2 ng/ml TNF-alpha and 10 ng/ml PMA (7.6fold). Both TNF-alpha- and PMA-induction levels of [0466] clone G 1002 were influenced by the presence or absence of epidermal growth factor (EGF). 7-Hour incubations in serum-free defined media with 20 ng/ml EGF resulted in a 3fold G-CSF promoter induction by TNF-alpha versus 4fold in the absence of EGF. The 9.3fold induction by PMA in the absence of EGF was reduced to 5.6fold by including EGF in the serum-free incubation mixture. Similar differences were observed, when EGF was substituted by 10% fetal calf serum.
  • A 7-hour incubation of [0467] G 21 cells with PMA in serum-free media increased luciferase expression directed by the G-CSF promoter by 34.6 fold in the absence and by 24.6 fold in the presence of EGF. TNF-Alpha induction did not significantly change on EGF addition (2.8 fold with and 2.1 fold without EGF).
  • Promoter induction experiments were also conducted with the GM-CSF reporter cell lines GM 1073, [0468] GM 1088 and GM 1105, which were all shown to contain correctly inserted constructs (see above). 10.5 hours incubation of GM 1073 cells with 20 ng/ml PMA in serum-containing media resulted in a 3.4 fold induction of the GM-CSF promoter, which was increased to 7.5 fold in serum-free media. Luciferase expression of clones 1088 and 1105 was induced by PMA 2.8 fold and 2 fold, respectively, while TNF-alpha induced both clones 2 fold.
  • All three M-CSF clones responded to a 16-hour incubation with 2,000 units/ml Interferon-gamma by a 20-fold increase of luciferase expression from the M-CSF promoter. [0469]
  • All 3 GM-CSF clones described above attached to the well surfaces of microtiter plates after overnight incubation. Levels of luciferase expression from [0470] clone 1105 were not appreciably affected by the omission of the fibronectin-coating step before cell plating. Luciferase expression levels were strongly increased by fibronectin coating, however, when clones G 1002 or G21 were used (about 8 fold or 3 fold, respectively).
  • [0471] Clones GM 1088, GM 1073 and G 1002 consistently produced bioluminescence signals varying by less than 10% between wells, when multiple 96-well microtiter plates containing these cells were assayed.
  • To be able to screen large random collections of compounds it may be necessary to use a variety of solvents to account for different solubilities of the compounds of interest. The effect of three organic solvents, DMSO, methanol and ethanol, which are frequently used to dissolve screening samples, was therefore determined on three reporter cell lines. Cells of the [0472] lines G 2005, GM 1105 and M10 containing luciferase reporter constructs for the G-CSF, GM-CSF or the promoter of the mouse mammary tumor virus were seeded into 96 well microtiter plates (10,000 cells/well) and cultured overnight. Various amounts of DMSO, methanol or ethanol were added to cultures. Luciferase activity in the cells was determined 8 hours after addition of the solvents. The relative amount of luciferase activity compared to untreated controls is plotted versus solvent concentration (FIG. 51).
  • G2005 and GM1105, which were constructed using the same parental cell line show very similar behaviour. It seems that a final concentration of 1% can be used in each case. [0473]
  • C. High-Throughput Drug Screen
  • Table 1 shows a summary of the results of a one-week, high-throughput screen of 2,000 chemicals to identify those chemicals specifically stimulating or inhibiting transcription from the G-CSF, hGH or MMTV promoters. This screen concurrently tested chemicals at three concentrations on quadruplicate samples of the M10, 532 and G21 cell lines. A minimum stimulation of one promoter, to the degree indicated, and less than 50% activation of the other two promoters was required for [0474]
    TABLE 1
    SUMMARY OF HIGH-THROUGHPUT SCREEN
    Number (%) of Chemicals Which Activate Expression:
    2-3 3-5 5-7 7-10 >10
    Promoter Fold Fold Fold Fold Fold Total
    G-CSF NA 23 10 3 2 38
    (1.1%) (0.5%) (0.15%) (0.10%) (1.9%)
    hGH NA NA 12 5 6 23
    (0.6%) (0.03%) (0.03%) (1.14%)
    MMTV 15 1 0 1 1 18
    (0.7%) (0.05%) (0%) (0.05%) (0.05%) (0.9%)
    Promoter Number (%) of Chemicals Which Inhibit Expression >3 Fold
    G-CSF 7 (0.35%)
    hGH 42 (2.1%) 
    MMTV 1 (0.05%)
  • [0475]
    TABLE 2
    FOLD
    INDUCTION RELATIVE
    TO SOLVENT CONTROL
    Chemical# Chemical Name GCSF hGH MMTV
    A) TRANSCRIPTIONAL ACTIVATORS
    G-CSF:
    40 3-Acetyl-2-6-Bis (tertiary 5.62 0.62 0.27
    butyl amino) -4-methyl-
    pyridine
    58 1-Acetylimidazole 6.03 0.17 0.42
    237 N-Carbethoxy-phthalimide 4.77 0.06 0.62
    254 1-(2-Chloroethyl) piperidine 4.09 0.90 0.98
    364 Melamine 3.67 1.18 1.07
    473 1,3,5,-Triazine >3 0.50 0.87
    542 5-Bromo-2′-deoxycytidine 6.28 1.08 1.26
    543 5-Bromo-2′-deoxyuridine 7.17 0.72 0.98
    878 Blueberry leaf extract 3.84 1.17 0.78
    1025 Culvers Root extract 4.09 0.98 1.24
    1234 4-Aminocinnamic Acid 4.97 0.51 1.03
    hydrochloride
    1255 1-Bromo-3,5-dichlorobenzene 6.74 0.43 1.09
    1374 4′-Amino-N-methylacetanilide 11.03 0.05 1.05
    1375 4′- (aminomethyl) benzene 8.94 0.04 1.37
    sulfonamide hydrochloride
    1376 2-Amino-5-Methyl benzene 6.37 0.04 1.32
    sulfonic acid
    1397 5-Amino-3-methylisothiazole 3.63 0.57 1.13
    hydrochloride
    1482 2-Aminophenyl disulfide 3.99 0.54 1.07
    1483 4-Aminophenyl disulfide 4.64 0.38 1.09
    FOLD INDUCTION
    Chemical# Chemical Name GCSF hGH MMTV
    1521 2-Amino-6-purinethiol 3.59 0.73 0.92
    1583 8-Bromoadenosine 5.82 0.12 0.88
    1592 Bis(2,2,3,3,4,4,5,5,6,6,7,7,) 3.20 0.74 1.34
    dodecafluoroheptyl- (+) -
    camphorate
    1783 Cupferron 6.55 0.32 0.89
    1793 Cyanomethyl-N,N-dimethyl 9.50 0.52 1.21
    dithiocarbamate
    1994 3-Bromobiphenyl 3.29 0.34 0.63
    2001 1-Bromo-4-tertiary butyl 3.11 0.74 1.12
    benzene
    2030 4-Bromo-2-fluoro-6- 5.53 0.67 0.87
    nitroanizol
    2096 (+) -1-Bromo-3-Chloro-2methyl 3.27 0.61 0.89
    propane
    2097 1-Bromo-5-Chloro pentane 5.09 0.88 1.22
    2129 4-Chlorobenzyl Chloride 3.23 0.75 0.95
    Group A:
    378 7-Oxo-7H-benzo [e] perimidine 4.12 0.26 0.59
    4-carboxylic acid
    423 Quinacrine dihydrochloride 2.39 0.56 0.64
    hydrate
    427 Resazurin 3.14 0.43 0.71
    836 Thionin 3.20 0.23 0.58
    1776 Cresyl Violet Acetate 3.50 0.15 1.36
    1904 9-Aminoacridine hydrochloride 4.12 0.54 0.82
    Group B:
    670 Methyl Green >3 0.52 0.79
    1780 Crystal Violet 20.39 0.38 1.15
    GROUP A AND B:
    80 Acridine Orange 5.87 0.66 0.83
    hGH:
    70 2-Acetylpyrrole 0.43 9.26 0.85
    299 10,11- 0.53 5.46 0.47
    Dihydrocarbamazepine
    322 1-ethyl-2- 0.60 11.18 1.12
    benzimidazolinone
    325 Fisetin 0.14 5.42 1.0
    552 3- (4-chlorophenyl) -1- 0.81 5.31 0.86
    methoxy-1-methyl urea
    790 Rivanol 0.01 5.94 0.58
    792 Rose Bengal 0.94 5.31 1.21
    856 Tripaumitin 0.28 6.49 0.42
    1004 Arnica 4X 0.85 6.48 1.22
    1160 Rochester # 6180 0.38 5.79 0.80
    1251 Bromocresol Green 0.14 15.19 0.33
    1337 4-Amino-5-hydroxy-1- 0.07 15.87 0.23
    naphthalene sulfonic acid
    1499 2-Amino-4-phenylthiazole 0.24 5.55 0.61
    hydrobromide monohydrate
    1550 2-Aminothiazole 0.04 5.44 0.87
    1552 2-amino-2-thiazoline 1.23 7.26 0.52
    1561 4-Amino-3,5,6- 0.23 8.05 0.48
    trichloropicolinic acid
    1598 N,N′-Bis- [3- (4,5-dihydro- 0.72 5.32 1.27
    1H-imidizol-2-yl) phenyl]
    urea dipropanoate
    1678 4,8-Bis (hydroxymethyl)- 0.36 7.08 0.89
    tricyclo [5,2,1,02.6]
    decane
    1740 5-carbethoxy-2-thiouracil 0.74 17.77 0.87
    1747 N6-carbobenzyloxy-L-lysine 0.78 6.16 0.86
    1804 Cyclobutane carboxylic 1.05 9.41 0.49
    acid
    1876 Alec Blue 0.87 11.91 0.40
    1881 Alizarin Blue Black B 0.21 18.87 0.69
    MMTV:
    189 Bathocuproinedisulfonic 1.06 1.47 2.80
    Acid disodium salt hydrate
    453 2,2′:6′,2″-Terpyridine 0.79 0.58 13.30
    519 b-Apo-8′-carotenal 1.15 0.68 2.76
    562 Copaiva Balsam 1.10 0.15 2.34
    629 Homoveratric acid 0.85 1.05 2.48
    633 5-Iodorotic acid 1.02 0.86 2.46
    765 Prednisolone-21-Acetate 0.96 1.30 2.66
    828 2,4,5,4′- 1.47 1.34 2.20
    Tetrachlorodiphenylsulfide
    848 Triamcinolone acetonide 0.75 1.28 2.43
    944 Peanut 1.15 0.91 2.10
    1269 5-Amino-4,6- 0.72 0.91 2.18
    dichloropyrimidine
    1316 2-Aminofluorene 0.74 1.39 2.33
    1318 2-Amino-9-fluorenone 1.13 0.85 2.41
    1384 2-Amino-4′- 1.33 0.50 2.43
    methylbenzophenone
    1573 5-Bromoacenapthene 1.49 0.34 4.30
    2064 4- (Bromomethyl) -6,7- 0.82 1.10 2.53
    dimethoxy-coumarin
    2148 2-chlorocyclohexanone 0.45 0.92 2.82
    2191 Chloramphenicol 0.37 0.35 7.32
    FOLD
    INHIBITION RELATIVE
    TO SOLVENT CONTROL
    Chemical# Chemical Name GCSF hGH MMTV
    B) TRANSCRIPTIONAL INHIBITORS
    G-CSF:
    209 4-Benzoylpyridine 6.66 1.08 0.81
    371 Morin hydrate 11.11 0.41 0.89
    660 Maclurin 10.0 0.34 1.04
    798 Salicylamide 4.76 0.90 0.68
    2009 4-Bromo-3,5- 3.70 0.57 0.64
    dimethylpyrazole
    2082 4-Bromo-3-Methylpyrazole 5.26 0.65 1.23
    2121 3-Chlorobenzyl alcohol 4.76 0.40 1.14
    hGH:
    183 Auramine O 0.72 4.00 0.70
    240 Carminic acid 0.63 5.26 0.80
    443 Sulfamethazine 0.60 4.76 0.79
    512 Amaranth 0.81 5.26 0.68
    FOLD INDUCTION
    Chemical# Chemical Name GCSF hGH MMTV
    541 5-Bromo-4-Chloro-3-indoxyl- 0.90 6.25 0.86
    phoshate K-salt
    556 Chromazurol S 0.73 33.33 0.87
    561 Clove Oil 0.62 5.00 0.05
    577 Na-Ne-Diacetyl-L-lysine 0.64 4.00 0.68
    578 Dibenzoyl-D-tartaric acid 0.65 4.00 0.91
    630 Hydantoin-5-acetic acid 0.70 3.57 0.74
    640 Kernechtrot 0.64 5.00 0.59
    759 Piperidine 0.64 5.88 0.95
    764 Prednisolone 0.82 4.54 0.59
    875 Black Walnut extract 0.69 6.25 0.80
    892 Colts Foot Leaves extract 0.68 11.11 0.87
    893 Comfrey Leaf extract 0.74 11.11 0.90
    920 Horehound Herb extract 0.56 3.84 0.84
    921 Horsetail Grass extract 0.72 3.44 0.86
    942 Pau D'Arco extract 0.80 6.25 0.63
    970 Thyme extract 0.57 4.34 1.07
    1591 1,2-Bis(di-p- 0.56 5.55 0.96
    tolylphosphino) -ethane
    1604 2,4-Bis [5,6-bis(4- 0.77 5.00 0.97
    sulfophenyl) -1,2,4-
    Thiazine-3-yl) -pyridine,
    tetrasodium salt hydrate
    1635 [(15) -endo]- (-) -Borneol 0.71 9.09 0.99
    1640 1,2-Bis (2-pyridyl) -ethylene 0.79 5.00 0.59
    1641 2,3-Bis (2-pyridyl) -pyrazine 0.83 5.55 0.60
    1648 2- [5,6-Bis (4-sulfophenyl) - 0.86 7.69 1.00
    1,2,4-triazine-3-yl] -4- (4-
    sulfophenyl) -pyridine,
    trisodium salt
    1651 Bis(2,2,2- 0.69 3.57 0.70
    trifluoroethyl) (methocarb-
    onyl -methyl) -phosphonate
    1655 2,5-Bis (trifluoro-methyl) 0.54 4.76 0.81
    benzoic acid
    1703 3-Bromobenzonitrile 0.76 10.00 0.90
    1704 4-Bromobenzonitrile 0.77 4.16 0.94
    1705 4-Bromobenzophenone 0.54 14.28 0.62
    1712 Calcein Blue 0.74 8.33 0.94
    1720 (15) - (-) -Camphor 0.65 4.76 0.66
    1764 7 - (Carboxymethoxy) -4- 0.55 7.14 0.82
    Methylcoumarin
    1770 Carminic acid 0.54 10.00 0.57
    1771 L-Carnosine 0.71 10.00 0.72
    1773 O-Cresolphthalein Complexone 0.62 10.00 0.67
    1890 Alloxazine 0.80 5.26 0.58
    2035 5-Bromofuroic acid 0.57 7.14 0.89
    2036 8-Bromoguanosine 0.58 4.34 0.81
    2037 1-Bromohexadecane 0.51 4.00 0.50
    MMTV: 2-Bromo-4,6-dinitroaniline 0.80 0.63 3.57
    2010
  • a chemical to be considered a selective activator. A minimum inhibition of 3 fold of one promoter and less than 20% inhibition of the other two promoters was required for a chemical to be considered a selective inhibitor. Table 2 gives the names and induction or inhibition ratios of the lead chemicals identified for each promoter. FIG. 44 illustrates the transcriptional stimulation and FIG. 45 the transcriptional inhibition observed with some of the lead chemicals. Some of the chemicals activating G-CSF transcription fell into conspicuous groups of analogs (Table 2; Group A and B). Although not specifically indicated in Table 2, groups of homologs and analogs can also be found for G-CSF-inhibiting as well as hGH-activating chemicals. [0476]
  • To determine the number of lead chemicals, which reproducibly score as positives in repeated luciferase assays, two types of experiments were conducted: [0477]
  • 1) G-CSF lead chemicals #1780, #58, #1783, #1374 were subjected to 48 independent luciferase assays performed on the same day. Compounds #58, #1780 and #1374 scored as positives in every single one of these assays inducing luciferase expression between 2 and 28fold (#58), 20 and 80fold (#1780) and 5 and 40fold (#1374). Probably due to its relatively low induction of luciferase expression (1.5 to 8fold), Compound #1783 scored as positive only in half of the 48 repeat assays. [0478]
  • 2) All of the 18 lead chemicals inducing luciferase expression from the MMTV promoter were again subjected to luciferase assays: 10 chemicals (#453, #519, #562, #765, #828, #848, #1269, #1316, #1384 and #2148) again induced luciferase expression between 2.1 and 2.8fold. Probably due to the relatively low induction level close to the background of the assay, the other eight lead chemicals did not repeat on that particular day. The most prominent lead chemical, #453 (13.3fold induction in the original high-throughput assay), was repeated in a total of 3 independent assays and consistently induced luciferase expression from the MMTV promoter between 10 and 35fold. Replacing DMSO by methanol to dissolve the chemical did not affect its ability to activate the MMTV promoter. [0479]
  • Aqueous clarified supernatants derived from individual Actinomyces colonies prepared by standard methods known to those skilled in the art as well as corresponding methanol extracts were subjected at 1:10 initial dilution to a fully automated, robotic High-Throughput luciferase assay using the system described in U.S. patent application Ser. No. 382,483. Out of 356 samples tested for modulation of the G-CSF, GM-CSF and MMTV promoters, 25 samples scored as positives, 7 of which were promoter-specific. A summary of the obtained results is contained in FIGS. 52 and 53. Thus high-throughput screening of fermentation broth samples using a luciferase expression assay consistently leads to the discovery of lead samples with the potential to be developed into novel pharmaceuticals. [0480]
  • D. Effects of Lead Chemicals on Endogenous G-CSF mRNA Levels
  • Northern blot analysis was used to demonstrate the stimulatory effects of lead chemicals #670 and #1255 on endogenous G-CSF mRNA levels. As shown in FIG. 46, both [0481] OSI #670 and #1255 stimulated production of G-CSF mRNA, as shown by a G-CSF-specific probe, but not of actin mRNA, as shown by a b-actin-specific probe. Also shown are the effects of the solvent, DMSO, used to dissolve the chemicals and a proteinaceous positive regulator, interferon-gamma. From these data it is concluded that chemicals, which induce luciferase expression from specific promoters, in plasmids stably integrated into cells, are also capable of stimulating mRNA production from the corresponding endogenous promoters without using a reporter system.
  • To further confirm, that compounds that had been identified in a luciferase expression assay using a G-CSF specific reporter cell line would be active in inducing transcription of the endogenous G-CSF gene, cells from the [0482] parental cell line 5637 used to construct the reporter cell line were incubated with cycloheximide (25 microg/ml), DMSO (0.5%, solvent control) and low, medium and high concentrations of the compounds 542 (10, 50, 250 microM), 1255 (20, 100, 500 microM), 1793 (0.25, 1.2, 6.25 microM) and 1904 (20, 100 microM) for 18 hours. RNA was extracted and the concentration of G-CSF, GM-CSF and gamma-actin mRNA was determined by the S1 protection method as described in Materials and Methods. The positions of G-CSF GM-CSF and gamma-actin specific protected fragments are indicated (G, GM, A) at the left side of the gel (FIG. 54).
  • Interestingly, all four compounds tested increased the amount of G-CSF mRNA and at least two of them, namely #542 and #1793, also increased the amount of GM-CSF mRNA. Compound #543, a structural analog of #542 showed similar activity. [0483]
  • E. Dose Response Analysis of Structurally Related Lead Chemicals
  • Among the chemicals which specifically activated the G-CSF promoter were groups of structural homologs. Three such homologs, #80, #670, and #1780, belong to groups listed in Table 2. These three structurally related chemicals all specifically activated the G-CSF promoter. Dose response graphs obtained with [0484] chemicals #80, #670, and #1780 are shown in FIG. 47. Although these chemicals all demonstrate large maximal stimulations, it is clear that their potencies, as measured by their ED50's (concentration of chemical resulting in 50% maximal stimulation), show wide variabilty (5-70 μm)).
  • F. Effects of Lead Chemicals on Target Protein Secretion
  • Two of the most promising lead chemicals (#542 and #1780), which were shown to stimulate levels of endogenous G-CSF mRNA as well as luciferase expression from the G-CSF promoter/luciferase fusion constructs, were further investigated for their ability to increase G-CSF secretion into the media of 5637 bladder carcinoma cells incubated with the chemicals for 48 hours. The levels of G-CSF in the cell supernatants were determined by a sandwich-antibody assay as described in Materials and Methods (FIG. 55). [0485]
  • G. Cytotoxicity of Lead Chemical #542
  • To address the question whether the induction of G-CSF, and GM-CSF transcription by the [0486] compound #542 was a specific effect or rather a phenomenon linked to a potential sensitivity of these promoters to stress exerted by toxic compounds, the concentration dependency of induction of luciferase activity in the reporter cell line G21 was compared to the concentration dependency of inhibition of respiration in FRE cells.
  • Cells were seeded into 96 well microtiter plates (20000 cells/well) and cultured overnight. [0487] Compound #542 was added at various concentrations and the cells were incubated for 6 hours. Luciferase activity was determined as described in Materials and Methods. The MTT-colorimetric assay was carried out on identically treated samples of FRE cells. Induction of luciferase reporter signal (plain line) and on inhibition of respiration (dashed line) are plotted versus the concentrations of compound (FIG. 56). The ED50 for induction of luciferase acitvity differed from the ED50 for inhibition of respiration by a factor of almost 10, which might indicate that compound #542 exerts a specific effect on G- and GM-CSF transcription.
  • H. Luciferase Expression Assay in Yeast
  • To determine, wheter the luciferase expression assay would also be useful to study other than mammalian cells, a yeast expression plasmid carrying the luciferase gene under control of a yeast promoter was constructed and transfected into appropriate yeast cells. Using these cells as a model system, a format for a 96-well luciferase expression assay in yeast cells was developed as described in more detail in Materials and Methods. [0488]
  • REFERENCES
  • 1. Angel, P., Baumann, I., Stein, B., Delius, H., Rahmsdorf, H. J. and Herrlich, P. (1987) 12-0-tetradecanoyl-phorbol-13-acetate (TPA) induction of the human collagenase gene is mediated by an inducible enhancer element located in the 5′-flanking region. Mol. Cell. Biol., 7:2256. [0489]
  • 2. Angel, P., Imagawa, M., Chiu, R. Stein, B., Imbra, R. J., Rahmsdorf, H. J., Jonat, C., Herrlich, P. and Karin, M. (1987) Phorbol ester-inducible genes contain a common cis element recognized by a TPA-modulated trans-acting factor, Cell, 49:729. [0490]
  • 3. Arnheim, N. (1979) Characterization of mouse ribsomal gene fragments purified by molecular cloning, Gene, 7:83. [0491]
  • 4. Bancroft, F. C., Wu, G. J., and Zubay, G. (1973) Proc. Natl. Acad. Sci. USA, 73:29. [0492]
  • 5. Bottenstein, J., Hayashi, I., Hutchings, S., Masui, H., Mather, J., McClure, D. B., Ohasa, S., Rizzino, A., Sato G., Serrero, G., Wolfe, R., and Wu, R. (1979) The growth of cells in serum-free hormone-supplemented media, Methods in Enzymology, 58:94. [0493]
  • 6. Cato, A. C. B. and Weinmann, J. (1988) Mineralcorticoid regulation of transcription of transfected mouse mammary tumor virus DNA in cultured kidney cells, J. Cell Biol., 106(6):2119. [0494]
  • 7. Cato, A. C. B., Miksicek, R., Schutz, G., Arnemann, J., and Beato, M. (1986) The hormone regulatory element of mouse mammary tumor virus mediates progesterone induction, EMBO J., 5(9):2237. [0495]
  • 8. De Wet, J. R., Wood, K. V., Helinski, D. R., and DeLuca, M. (1985) Cloning of firefly luciferase cDNA and the expression of active luciferase in [0496] Escherichia coli, Proc. Natl. Acad. Sci. USA, 82:7870.
  • 9. Denison, M. S., Fisher, Springfield, N.J., J. M., and Whitlock, Jr., J. P. (1988) Inducible, receptor-dependent protein-DNA interactions at a dioxin-responsive transcriptional enhancer, Proc. Natl. Acad. Sci. USA, 85:2528. [0497]
  • 10. Edeμman, A. M., Blumenthal, D. R. and Krebs, E. G. (1987) Protein Serine/Threonine Kinases Ann. Rev. 56:567-613. [0498]
  • 11. Engebrecht, J. M., Simon, M., and Silverman, M. (1985) Measuring gene expression with light. Science, 227:1345. [0499]
  • 12. Evans, R. M. and Hollenberg, S. M. (1988) Zinc fingers: Gilt by assocation, Cell, 52:1. [0500]
  • 13. Evans, R. M. (1988) The steroid and thyroid hormone receptor superfamily, Science, 240:889. [0501]
  • 14. Gorman, C. (1985) Vectors used in mammalian cell expression. In DNA Cloning, Vol. II (D. M. Glover, ed). IRL Press, Washington, D.C. [0502]
  • 15. Graham, F. L. and Van der Ed, A. J. (1973) A new technique for the assay of [0503] human adenovirus 5 DNA, Virology, 52:456.
  • 16. Hatzopoulos, A. K., Schlokat, U., and Gruss, P. (1988) Enhancers and other cis-acting regulatory sequences. In Transcription and Splicing. (Hames, B. D. and Glover, D. M., eds.) IRL Press, Washington, D.C., Vol. 1, p. 43. [0504]
  • 17. Hayashi, I., Larner, J., and Sato, G. (1978) Hormonal growth control of cells in culture., In Vitro, 14:23. [0505]
  • 18. Hoeffler, J. P., Meyer, T. E., Yun, Y., Jameson, J. L., and Habener, J. F. (1988) Cyclic AMP-responsive DNA-binding protein: Structure based on a cloned placental cDNA, Science, 242:1430. [0506]
  • 19. Hoopes, B. C. and McClure, W. R. Strategies in Regulation of Transcription Initiation. In [0507] Escherichia coli and Saμmonella Typhimurium: Cellular and Molecular Biology (F. C. Neidhardt, J. L. Ingraham, K. Brooks Low, B. Magasanik, M. Schaechter, eds.) Vol. 2, p. 1231.
  • 20. Kaushansky, K., O'Hara, P. J., Berkner, K., Segal, G. M., Hagen, F. S., and Adamson, J. W. (1986) Genomic cloning, characterization, and multilineage growth-promoting activity of human granulocyte-macrophage colony-stimulating factor, Proc. Natl. Acad. Sci. USA, 83:3101. [0508]
  • 21. Krainer, A. R. and Maniatis, T. (1988) RNA splicing. In Transcription and Splicing (Hames, B. D. and Glover, D. M., eds.) IRL Press, Washington, D.C., Vol. 1. [0509]
  • 22. La Thangue, N. B. and Rigby, P. W. J. (1988) Transacting protein factors and the regulation of eukaryotic transcription. In Transcription and Splicing (Hames, B. D. and Glover, D. M., eds) IRL Press, Washington, D.C., Vol. 1. [0510]
  • 23. Ladner, M. B., Martin, G. A., Noble, J. A., Nikoloff, D. N., Tal, R., Kawaski, E. S., and White, T. J. (1987) Human CSF-1: gene structure and alternative splicing of mRNA precursors, EMBO J., 6:2693. [0511]
  • 24. Landschulz, W. H., Johnson, P. F., and McKnight, S. L. (1988). The leucine zipper: A hypothetical structure common to a new class of DNA binding proteins, Science, 240:1759. [0512]
  • 25. Lefevre, C., Imagawa, M., Dana, S., Grindlay, J., Bodner, M., and Karin, M. (1987) Tissue-specific expression of the human growth hormone gene is conferred in part by the binding of a specific trans-acting factor, EMBO J., 6:971. [0513]
  • 26. Levine, M. and Hoey, T. (1988) Homeobox proteins as sequence-specific transcription factors, Cell, 55:537. [0514]
  • 27. Lin, F. K., Suggs, S., Lin, C. H, Browne, J. K., Smalling, R. Egrie, J. C., Chen, K. K., Fox, G. M., Martin, F., Stabinsky, Z., Badrawi, S. M., Lai, P. H., and Goldwasser, E. (1985) Cloning and expression of the human erythropoietin gene, Pro. Natl. Acad. Sci. USA, 82:7580. [0515]
  • 28. Maniatis, T., Goodbourn, S. and Fischer, J. A. (1987) Regulation of inducible and tissue-specific gene expression, Science, 236:1237. [0516]
  • 29. Matthews, B. W. (1987) Cro repressor structure and its interaction with DNA. In DNA: Protein Interactions and Gene Regulation (E. B. Thompson and J. Papaconstantinou, eds.) University of Texas Press, Austin. [0517]
  • 30. McClure (1985) Ann. Rev. Biochem., 54:171. [0518]
  • 31. McKnight, S. L. (1982) Functional relationships between transcriptional control signals of the thymidine kinase gene of herpes simplex virus, Cell, 31:355. [0519]
  • 32. Nagata, S., Tsuchiya, M., Asano, S., Yamamoto, O., Hirata, Y., Kubota, N., Oheda, M., Nomura, H. and Yamazaki, T. (1986) The chromosomal gene structure and two mRNAs for human granulocyte colony-stimulating factor, EMBO J., 5:575. [0520]
  • 33. Ow, D. W., Wood, K. U, Deluca, M, Dewet, J. R., Melinski, D., and Howell, S. H. Science 234:856-859 [0521]
  • 34. Pouwels, Ph. H., Enger-Valk, B. E., and Brammar, W. J. (1985) Cloning Vectors. Elsevier Science Publishers, B.V., Amsterdam. [0522]
  • 35. Proudfoot, N. J. and Whitelaw, E. (1988) Termination and 3′ end processing of eukaryotic RNA. In Transcription and Splicing (Hames, B. D. and Glover, D. M., eds.) ERL Press, Washington, D.C., Vol. 1, p. 97. [0523]
  • 36. Schlief, R. The L-Arabinose Dperon. In [0524] Escherichia coli and Salmonella typhimurium: Cellular and Molecular Biology (F. C. Neidhardt, J. L. Ingraham, K. Brooks Low, B. Maga Sanik, M. Schaecter, eds.) Vol. 2, p. 1473.
  • 37. Schlief, R. (1988) DNA binding by proteins, Science, 241:1182. [0525]
  • 38. Yamamoto, K. R. (1985) Steroid receptor regulated transcription of specific genes and gene networks, Ann. Rev., Genet., 19:209. [0526]
  • 39. Yamamoto, K. K., Gonzalez, G. A., Biggs III, W. H., and Montminy, M. R. (1988) Phosphorylation-induced binding and transcriptional efficacy of nuclear factor CREB, Nature, 334:494. [0527]
  • 40. Yang, Y. C., Ciarletta, A. B., Temple, P. A., Chung, M. P., Kovacic, S., Witek-Gianotti, J. S., Learyn, A. C., Kriz, R., Donahue, R. E., Wong, G. G., and Clark, S. C. (1986) Human IL-3 (multi-CSF): Identification by expression cloning of a novel hematopoietic growth factor related to murine IL-3, Cell, 47:3. [0528]
  • 41. Yang, T. C. and Clark, S. C. (1988) Molecular cloning of a primate cDNA and the human gene for [0529] interleukin 3, Lymphokines, 15:375.
  • 42. Yanofsky, C. and Crawford, I. P. The Tryptophan Operon. In [0530] Escherichia coli and Salmonella typhimurium: Cellular and Molecular Biology (F. C. Neidhardt, J. L. Ingraham, K. Brooks Low, B. Magasanik, M. Schaechter, eds.) Vol. 2, p. 1453.
  • 43. Zhan, A., Culpepper, A., Reddy, M., Loveless, J., and Goldfarb, M. (1987) Human oncogenes detected by a defined medium culture assay, Oncogene, 1:369. [0531]
  • 44. Thomas, K. R. and Capecchi, M. R., Cell, 1987, 51:503-512 [0532]
  • 45. Mansour, S. L., Thomas, K. R. and Capecchi, M. R., Nature, 1988, 366:348-352 [0533]
  • 46. M. R. Capecchi, 1989, Science, 244:1288-1292 [0534]
  • 47. J. Thompson and D. Gillespie (1987) Molecular hybridization with RNA probes in concentrated solutions of guanidine thiocyanate, Anal. Biochem. 163:281-291. [0535]
  • 48. J. Thompson, R. Solomon, M. Pellegrino, K. Sakai, M. Lewin, M. Feild, M. Castrovinci, L. Sacramone and D. Gillespie (1989) A noise-free molecular hybridization procedure for measuring RNA in cell lysates, Anal. Biochem. 181:371-378. [0536]
  • 49. M. S. Urdea, J. A. Running, T. Horn, J. Clyne, L. Ku and B. D. Warner (1987) A novel method for the rapid detection of specific nucleotide sequences in crude biological samples without blotting or radioactivity; application to the analysis of hepatitis B virus in human serum, GENE 61:253-264. [0537]
  • 50. B. Chu and L. Orgel (1989) Target nucleic acid amplification/detection systems, WO 90/03445, published Apr. 5, 1990. Applicant: The Salk Institute for Biological Studies. [0538]
  • 51. E. Oura (1983) “Biomass from Carbohydrates” In Biotechnology, [0539] Vol 3, Chpt. 1a (ed H. Dellweg) VCH Press, Germany.
  • 52. A. Einsele (1983) “Biomass from Higher n-Alkanes” In Biotechnology, [0540] Vol 3, Chpt. 1b (ed H. Dellweg) VCH Press, Germany.
  • 53. U. Faust and P. Prave (1983) “Biomass from Methane and Methanol” In Biotechnology, [0541] Vol 3, Chpt. 1c (ed H. Dellweg) VCH Press, Germany.
  • 54. R. J. Quinlan and S. G. Lisansky (1983) “Microbial Insectidides” In Biotechnology, [0542] Vol 3, Chpt. 2e (ed H. Dellweg) VCH Press, Germany.
  • 55. N. Kosaric, A. Wieczorek, G. P. Cosentino, R. J. Magee and J. E. Prenosil (1983) “Ethanol Fermentation” In Biotechnology, [0543] Vol 3, Chpt. 3a (ed H. Dellweg) VCH Press, Germany.
  • 56. M. Rohr, C. P. Kubicek and J. Kominek (1983) “Citric Acid” In Biotechnology, [0544] Vol 3, Chpt. 3d (ed H. Dellweg) VCH Press, Germany.
  • 57. M. Rohr, C. P. Kubicek and J. Kominek (1983) “Gluconic Acid” In Biotechnology, [0545] Vol 3, Chpt. 3e (ed H. Dellweg) VCH Press, Germany.
  • 58. K. Soda, H. Tanaka and N. Esaki (1983) “Amino Acids” In Biotechnology, [0546] Vol 3, Chpt. 3g (ed H. Dellweg) VCH Press, Germany.
  • 59. G. W. Barnard and D. O. Hall (1983) “Energy from Renewable Resources” In Biotechnology, [0547] Vol 3, Chpt. 4 (ed H. Dellweg) VCH Press, Germany.
  • 60. A. Kuninaka (1986) “Nucleic Acids, Nucleotides, and Related Compounds” In Biotechnology, [0548] Vol 4, Chpt. 4 (eds H. Pape and H.-J. Rehm) VCH Press, Germany.
  • 61. H. Kleinkauf and H. van Dohren (1986) “Peptide Antibiotics” In Biotechnology, [0549] Vol 4, Chpt. 10 (eds H. Pape and H.-J. Rehm) VCH Press, Germany.
  • 62. S. Umezawa, S. Kondo and Y. Ito (1986) “Aminoglycoside Antibiotics” In Biotechnology, [0550] Vol 4, Chpt. 11 (eds H. Pape and H.-J. Rehm) VCH Press, Germany.
  • 63. S. Omura and Y. Tanaka (1986) “Macrolide Antibiotics” In Biotechnology, [0551] Vol 4, Chpt. 12 (eds H. Pape and H.-J. Rehm) VCH Press, Germany.
  • 64. Z. Hostalek and Z. Vanek (1986) “Tetracyclines” In Biotechnology, [0552] Vol 4, Chpt. 13 (eds H. Pape and H.-J. Rehm) VCH Press, Germany.
  • 65. G. Lancini (1986) “Ansamycins” In Biotechnology, [0553] Vol 4, Chpt. 14 (eds H. Pape and H.-J. Rehm) VCH Press, Germany.
  • 66. J. Berdy (1986) “Further Antibiotics with Practical Application” In Biotechnology, [0554] Vol 4, Chpt. 15 (eds H. Pape and H.-J. Rehm) VCH Press, Germany.
  • 67. H. Kobel and J.-J. Sanglier (1986) “Ergot Alkaloids” In Biotechnology, [0555] Vol 4, Chpt. 18 (eds H. Pape and H.-J. Rehm) VCH Press, Germany.
  • 68. T. Anke (1986) “Further Secondary Products of Biotechnological Interest” In Biotechnology, [0556] Vol 4, Chpt. 19 (eds H. Pape and H.-J. Rehm) VCH Press, Germany.
  • 69. J. Berlin (1986) “Secondary Products from Plant Cell Cultures” In Biotechnology, [0557] Vol 4, Chpt. 20 (eds H. Pape and H.-J. Rehm) VCH Press, Germany.
  • 70. L. L. Smith (1984) “Steroids” In Biotechnology, Vol 6a, Chpt. 2 (ed K. Kieslich) VCH Press, Germany. [0558]
  • 71. C. K. A. Martin (1984) “Sterols” In Biotechnology, Vol 6a, Chpt. 3 (ed K. Kieslich) VCH Press, Germany. [0559]
  • 72. V. A. Krasnobajew (1984) “Terpenoids” In Biotechnology, Vol 6a, Chpt. 4 (ed K. Kieslich) VCH Press, Germany. [0560]
  • 73. A. Kergomard (1984) “Alicyclic and Heteroalicyclic Compounds” In Biotechnology, Vol 6a, Chpt. 5 (ed K. Kieslich) VCH Press, Germany. [0561]
  • 74. O. K. Sebek (1984) “Antibiotics” In Biotechnology, Vol 6a, Chpt. 7 (ed K. Kieslich) VCH Press, Germany. [0562]
  • 75. P. R. Wallnofer and G. Engelhardt (1984) “Aromatic and Heterocyclic Structures” In Biotechnology, Vol 6a, Chpt. 8 (ed K. Kieslich) VCH Press, Germany. [0563]
  • 76. M. Buhler and J. Schindler (1984) “Aliphatic Hydrocarbons” In Biotechnology, Vol 6a, Chpt. 9 (ed K. Kieslich) VCH Press, Germany. [0564]
  • 77. G. Schmidt-Kastner and P. Egerer (1984) “Amino Acids and Peptides” In Biotechnology, Vol 6a, Chpt. 10 (ed K. Kieslich) VCH Press, Germany. [0565]
  • 78. A. Crueger and W. Crueger (1984) “Carbohydrates” In Biotechnology, Vol 6a, Chpt. 11 (ed K. Kieslich) VCH Press, Germany. [0566]
  • 79. P. F. Heinstein and A. Emery (1988) “Processes with Plant Cell Cultures” In Biotechnology, Vol 6b, Chpt. 9 (ed H.-J. Rehm) VCH Press, Germany. [0567]
  • 80. M. Butler (1988) “Processes with Animal Cell and Tissue Cultures” In Biotechnology, Vol 6b, Chpt 10 (ed H.-J. Rehm) VCH press, Germany. [0568]
  • 81. A. E. Torma (1988) “Biosensors and ‘Bioelectronics’” In Biotechnology, Vol 6b, Chpt 12 (ed H.-J. Rehm) VCH press, Germany. [0569]
  • 82. S. Kunst and K. Mudrack (1988) “Microbial Elimination of Nitrogen and Phosphorus” In Biotechnology, Vol 6b, Chpt 14 (ed H.-J. Rehm) VCH press, Germany. [0570]
  • 83. E.-J. Nyns (1986) “Biomethanation Processes” In Biotechnology, [0571] Vol 8, Chpt 5 (ed W. Schonborn) VCH press, Germany.
  • 84. T. Leisinger and W. Brunner (1986) “Poorly Degradable Substances” In Biotechnology, [0572] Vol 8, Chpt 14 (ed W. Schonborn) VCH press, Germany.
  • 85. S. Lafon-Lafourcade (1983) “Wine and Brandy” In Biotechnology, [0573] Vol 5, Chpt 2 (ed G. Reed) VCH press, Germany.
  • 86. W. A. Hardwick (1983) “Beer” In Biotechnology, [0574] Vol 5, Chpt 3 (ed G. Reed) VCH press, Germany.
  • 87. E. R. Vedamuthu and C. Washam (1983) “Cheese” In Biotechnology, [0575] Vol 5, Chpt 4 (ed G. Reed) VCH press, Germany.
  • 88. V. Bottazzi (1983) “Other Fermented Dairy Products” In Biotechnology, [0576] Vol 5, Chpt 5 (ed G. Reed) VCH press, Germany.
  • 89. D. W. Ow, K. V. Wood, M. DeLuca, J. R. DeWet, D. R. Helinski, S. H. Howell (1986) Transient and stable expression of the firefly luciferase gene in plant cells and transgenic plants. Science 234:836-839. [0577]
  • 90. D. R. Edwards and J. K. Heath (1990) “Regulation of transcription by transforming growth factor-β” In Molecular Aspects of Cellular Regulation, [0578] Vol 6, Elsevier Biomedical Press, in press.
  • 91. Transcription and Splicing (1988) In the series Frontiers in Molecular Biology, ed. B. D. Hames and D. M. Glover, IRL Press, UK. [0579]
  • 92. Southern, E. (1980) Methods in Enzymology, 69: 152. [0580]
  • 93. Feinberg, A. and Vogelstein (1984) Anal. Biochem. 137:266. [0581]
  • 94. Sambrook, J., Fritsch, E. F. and Maniatis, T. (1989) Molecular Cloning. Cold Spring Harbor Laboratory Press, 1:7.58. [0582]
  • 95. Cole, S. P. C. (1986) Rapid chemosensitivity testing of human lung tumor cells using the MTT assay. Cancer Chemother. Pharmacol., 17:259. [0583]
  • 96. Harlow, E. and Lane, D. (1988). Antibodies—A Laboratory Manual. Cold Spring Harbor Laboratory Press, page 578. [0584]
  • 97. Teem, J. L. and Rosbash, M. (1983) Proc. Natl. Acad. Sci. USA 80:4403-4407. [0585]
  • 98. Pikielny, C. W., Teem, J. L., and Rosbash, M. (1983) Cell 34:395-403. [0586]
  • 99. Sherman, F., Fink, G. R., Hicks, J. B. (1986) Laboratory Course Manual for Methods in Yeast Genetics, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y. [0587]

Claims (160)

What is claimed is:
1. A method of transcriptionally modulating the expression of a homologous gene-of-interest, the expression of which is associated with a defined physiological or pathological effect within a multicellular organism, which comprises contacting a cell, which is capable of expressing the gene, with an amount of a molecule effective to transcriptionally modulate expression of the gene and thereby affect the level of the protein encoded by the gene which is expressed by the cell, which molecule (a) does not naturally occur in the cell, (b) specifically transcriptionally modulates expression of the gene-of-interest, and (c) binds to DNA or RNA, or binds to a protein through a domain of such protein which is not a ligand-binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand-binding domain is normally associated with the defined physiological or pathological effect.
2. A method of claim 1, wherein the molecule does not naturally occur in any cell of a higher eucaryotic organism.
3. A method of claim 1, wherein the molecule does not naturally occur in any cell.
4. A method of claim 1, wherein the molecule is not a naturally occurring molecule.
5. A method of claim 1, wherein the cell is a cell of the multicellular organism.
6. A method of claim 1, wherein the cell is an animal cell.
7. A method of claim 6, wherein the animal cell is a human cell.
8. A method of claim 1, wherein the cell is a plant cell.
9. A method of claim 1, wherein the cell is a fungal cell.
10. A method of claim 1, wherein the cell is a protozoan cell.
11. A method of claim 1, wherein the cell is a bacterial cell.
12. A method of claim 1, wherein the gene-of-interest is a human gene.
13. A method of claim 1, wherein the gene-of-interest encodes a hematopoietic protein.
14. A method of claim 13, wherein the hematopoietic protein is a colony stimulating factor.
15. A method of claim 14, wherein the colony stimulating factor is granulocyte-macrophage colony stimulating factor (GM-CSF).
16. A method of claim 14, wherein the colony stimulating factor is granulocyte colony stimulating factor (G-CSF).
17. A method of claim 14, wherein the colony stimulating factor is macrophage colony stimulating factor (M-CSF).
18. A method of claim 13, wherein the hematopoietic protein is erythropoietin (EPO).
19. A method of claim 1, wherein the gene-of-interest encodes an interleukin (IL).
20. A method of claim 1, wherein the gene-of-interest encodes a growth hormone selected from the group consisting of human, bovine, porcine, avian, ovine, piscine, and equine growth hormone, and polypeptide analogs thereof having the biological activity of the corresponding naturally occurring growth hormone.
21. A method of claim 1, wherein the gene-of-interest encodes a growth hormone releasing factor.
22. A method of claim 1, wherein the gene-of-interest is a viral gene.
23. A method of claim 22, wherein the viral gene is a retroviral gene.
24. A method of claim 23, wherein the retroviral gene is a gene from the HIV, HTLV-1, or HTLV-2 virus.
25. A method of claim 22, wherein the viral gene is a gene from a hepatitis virus.
26. A method of claim 22, wherein the viral gene is a gene from a herpes virus.
27. A method of claim 22, wherein the viral gene is a gene from an animal virus.
28. A method of claim 22, wherein the viral gene is a gene from a papilloma virus.
29. A method of claim 22, wherein the viral gene is a gene from a cytomegalovirus.
30. A method of claim 27, wherein the animal virus is a pseudorabies, Marek's, Newcastle's Disease, or IBR virus.
31. A method of claim 1, wherein the gene-of-interest is a plant gene.
32. A method of claim 31, wherein the plant gene encodes an agronomically important trait.
33. A method of claim 32, wherein the agronomically important trait is selected from the group consisting of germination, sprouting, flowering, fruit ripening, salt tolerance, herbicide resistance, pesticide resistance, fungicide resistance, temperature resistance, and growth.
34. A method of claim 1, wherein the gene-of-interest is a protozoan gene.
35. A method of claim 34, wherein the protozoan is selected from the group consisting of Trypanosoma, Plasmodium, Leishmania, Giardia, Entamoeba, Toxoplasma, Babesia, and Cryptosporidiosis.
36. A method of claim 1, wherein the gene-of-interest is a helminth gene.
37. A method of claim 1, wherein the gene-of-interest is an oncogene.
38. A method of claim 37, wherein the oncogene is the phl-abl oncogene.
39. A method of claim 37, wherein the oncogene is selected from the group consisting of H-, N-, and K-ras oncogenes.
40. A method of claim 37, wherein the oncogene is the neu oncogene.
41. A method of claim 37, wherein the oncogene is the src oncogene.
42. A method of claim 1, wherein the gene-of-interest encodes TGF-β1.
43. A method of claim 1, wherein the gene-of-interest encodes TGF-β2.
44. A method of claim 1, wherein the gene-of-interest encodes TGF-β3.
45. A method of claim 1, wherein the gene-of-interest encodes a naturally occurring receptor.
46. A method of claim 45, wherein the receptor is a TGF-β receptor.
47. A method of claim 45, wherein the receptor is a testosterone receptor.
48. A method of claim 45, wherein the receptor is an estrogen receptor.
49. A method of claim 45, wherein the receptor is the human low density lipoprotein (LDL) receptor.
50. A method of claim 45, wherein the receptor is the receptor for a hematopoietic protein selected from the group consisting of M-CSF, G-CSF, GM-CSF, and EPO.
51. A method of claim 45, wherein the receptor is the receptor for an interleukin (IL) selected from the group consisting of IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7and IL-8.
52. A method of claim 45, wherein the receptor is a cell surface protein which mediates infection of the cell by a virus.
53. A method of claim 52, wherein the virus is selected from the group consisting of HIV, HTLV-1, HTLV-2, a hepatitis virus, a herpes virus, an animal virus, a papilloma virus, a cytomegalovirus, and a rhinovirus.
54. A method of claim 1, wherein the receptor which naturally occurs in the cell is a testosterone receptor.
55. A method of claim 1, wherein the receptor which naturally occurs in the cell is an estrogen receptor.
56. A method of claim 1, wherein in (c) the protein is not the protein encoded by the gene-of-interest.
57. A method of determining whether a molecule not previously known to be a modulator of protein biosynthesis is capable of transcriptionally modulating the expression of a gene-of-interest which comprises contacting a sample which contains a predefined number of cells with a predetermined amount of a molecule to be tested, each such cell comprising DNA consisting essentially of (i) a modulatable transcriptional regulatory sequence of the gene-of-interest, (ii) a promoter of the gene-of-interest, and (iii) a reporter gene, which expresses a polypeptide capable of producing a detectable signal, coupled to, and under the control of, the promoter, under conditions such that the molecule, if capable of acting as a transcriptional modulator of the gene-of-interest, causes a measureable detectable signal to be produced by the polypeptide expressed by the reporter gene, quantitatively determining the amount of the signal produced, comparing the amount so determined with the amount of produced signal detected in the absence of any molecule being tested or upon contacting the sample with any other molecule, and thereby identifying the molecule as one which causes a change in the detectable signal produced by the polypeptide expressed by the reporter gene, and thus identifying the molecule as a molecule capable of transcriptionally modulating the expression of the gene-of-interest.
58. A method of determining whether a molecule not previously known to be a modulator of protein biosynthesis is capable of transcriptionally modulating the expression of a gene-of-interest which comprises contacting a sample which contains a predefined number of cells with a predetermined amount of a molecule to be tested, each such cell comprising DNA consisting essentially of (i) a modulatable transcriptional regulatory sequence of the gene-of-interest, (ii) a promoter of the gene-of-interest, and (iii) a DNA sequence transcribable into mRNA coupled to and under the control of, the promoter, under conditions such that the molecule, if capable of acting as a transcriptional modulator of the gene-of-interest, causes a measureable difference in the amount of mRNA transcribed from the DNA sequence, quantitatively determining the amount of the mRNA produced, comparing the amount so determined with the amount of mRNA detected in the absence of any molecule being tested or upon contacting the sample with any other molecule, and thereby identifying the molecule as one which causes a change in the detectable mRNA amount of, and thus identifying the molecule as a molecule capable of transcriptionally modulating the expression of the gene-of-interest.
59. A method of any of claims 57 or 58, wherein the molecule (a) does not naturally occur in the cell, (b) specifically transcriptionally modulates expression of the gene-of-interest, and (c) binds to DNA or RNA or binds to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with a defined physiological or pathological effect.
60. A method of any of claims 57 or 58, wherein the sample comprises cells in monolayers.
61. A method of any of claims 57 or 58, wherein the sample comprises cells in suspension.
62. A method of any of claims 57 or 58, wherein the cells comprise human, animal, or plant cells.
63. A method of any of claims 57 or 58, wherein the cells are bacterial cells.
64. A method of any of claims 57 or 58, wherein the cells are fungal cells.
65. A method of any of claims 57 or 58, wherein the predefined number of cells is from about 1 to about 5×105 cells.
66. A method of claim 65, wherein the predefined number of cells is from about 2×102 to about 5×104 cells.
67. A method of any of claims 57 or 58, wherein the predetermined amount of the molecule to be tested is based upon the volume of the sample.
68. A method of any of claims 57 or 58, wherein the predetermined amount is from about 1.0 pM to about 20 μM.
69. A method of any of claims 57 or 58, wherein the predetermined amount is from about 10 nM to about 500 μM.
70. A method of any of claims 57 or 58, wherein the contacting is effected from about 1 hour to about 24 hours.
71. A method of claim 70, wherein the contacting is effected from about 2 to about 12 hours.
72. A method of any of claims 57 or 58, wherein the contacting is effected with more than one predetermined amount of the molecule to be tested.
73. A method of any of claims 57 or 58, wherein the molecule to be tested is a purified molecule.
74. A method of any of claims 57 or 58, wherein the modulatable transcriptional regulatory sequence comprises a cloned genomic regulatory sequence.
75. A method of any of claims 57 or 58, wherein the DNA consists essentially of more than one modulatable transcriptional regulatory sequence.
76. A method of claim 57, wherein the reporter gene is inserted downstream of the promoter of the gene-of-interest by homologous recombination.
77. A method of claim 57, wherein the reporter gene encodes a luciferase.
78. A method of claim 57, wherein the reporter gene encodes chloramphenicol acetyltransferase.
79. A method of claim 57, wherein the reporter gene encodes β glucuronidase.
80. A method of claim 57, wherein the reporter gene encodes β galactosidase.
81. A method of claim 57, wherein the reporter gene encodes neomycin phosphotransferase.
82. A method of claim 57, wherein the reporter gene encodes guanine xanthine phosphoribosyltransferase.
83. A method of claim 58, wherein mRNA is detected by quantitative polymerase chain reaction.
84. A screening method according to any of claims 57 or 58 which comprises separately contacting each of a plurality of substantially identical samples, each sample containing a predefined number of cells under conditions such that contacting is effected with a predetermined amount of each different molecule to be tested.
85. A screening method of claim 84, wherein the plurality of samples comprises more that about 104 samples.
86. A screening method of claim 84, wherein the plurality of samples comprises more than about 5×104 samples.
87. A method of essentially simultaneously screening molecules to determine whether the molecules are capable of transcriptionally modulating one or more genes of interest in a panel of such genes which comprises essentially simultaneously screening the molecules against each of the genes of interest according to the method of claim 84.
88. A screening method of any of claims 86 or 87, wherein more that about 103 samples per week are contacted with different molecules.
89. A method for transcriptionally modulating in a multicellular organism the expression of a gene-of-interest, the expression of which is associated with a defined physiological or pathological effect in the organism, which comprises administering to the organism an amount of a molecule effective to transcriptionally modulate expression of the gene and thus affect the defined physiological or pathological effect, which molecule (a) does not naturally occur in the organism, (b) specifically transcriptionally modulates expression of the gene-of-interest, and (c) binds to DNA or RNA or binds to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the organism, the binding of a ligand to which ligand binding domain is normally associated with the defined physiological or pathological effect.
90. A method of claim 89, wherein the multicellular organism is a human being.
91. A method of claim 89, wherein the multicellular organism is an animal.
92. A method of claim 89, wherein the multicellular organism is a plant.
93. A method of claim 89, wherein the defined pathological effect is a disorder and modulated expression of the gene-of-interest is associated with amelioration of the disorder.
94. A method of claim 90, wherein the defined pathological effect is a disorder selected from the group consisting of cancer, a hematopoietic dysfunction, diabetes, tissue inflammation, atherosclerosis, viral infections, dysfunctions of memory or learning, and dysfunctions in a cholesterol or other metabolic pathway.
95. A method of claim 91, wherein the defined physiological effect is growth and the organism is an animal such as a man, cow, a pig, a bird, a fish, a sheep or a horse.
96. A method of claim 92, wherein the defined physiological or pathological effect is an agronomically important trait.
97. A method of claim 90 or 91, wherein the administering comprises topical contact.
98. A method of claim 90 or 91, wherein the administering comprises oral, transdermal, intravenous, intramuscular or subcutaneous administration.
99. A method of any of claims 57, 58, or 89, wherein the gene-of-interest encodes a naturally occurring receptor.
100. A method of claim 99, wherein the receptor is a testosterone receptor.
101. A method of claim 99, wherein the receptor is an estrogen receptor.
102. A method of any of claims 57, 58, or 89, wherein the receptor which naturally occurs in the cell is a testosterone receptor.
103. A method of any of claims 57, 58, or 89, wherein the receptor which naturally occurs in the cell is an estrogen receptor.
104. A method of any of claims 57, 58, or 89, wherein the gene-of-interest encodes a TGF-β receptor.
105. A method of any of claims 57, 58, or 89, wherein the gene-of-interest encodes TGF-β1.
106. A method of any of claims 57, 58, or 89, wherein the gene-of-interest encodes TGF-β2.
107. A method of any of claims 57, 58, or 89, wherein the gene-of-interest encodes TGF-β3.
108. A method of any of claims 57, 58, or 89, wherein the gene-of-interest encodes an oncogene.
109. A method of claim 108, wherein the oncogene is the neu oncogene.
110. A method of claim 108, wherein the oncogene is selected from the group consisting of H-, N-, and K-ras oncogenes.
111. A method for modulating the expression of a polypeptide by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing such polypeptide which comprises contacting the cell with an amount of a molecule effective so to transcriptionally modulate expression of the polypeptide by the cell, which molecule (a) does not naturally occur in the cell and (b) binds to DNA or RNA, or binds to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with modulating transcriptional expression of a polypeptide.
112. A method of claim 111, wherein the polypeptide is a homologous polypeptide.
113. A method of claim 111, wherein the molecule specifically transcriptionally modulates expression of the polypeptide.
114. A method of claim 111, wherein the polypeptide is a desired product.
115. A method of claim 114, wherein the desired product is a monoclonal antibody.
116. A method of claim 111, wherein the DNA is recombinant DNA.
117. A method of claim 111, wherein the cell is a animal cell.
118. A method of claim 111, wherein the cell is a plant cell.
119. A method of claim 111, wherein the cell is a bacterial cell.
120. A method of claim 111, wherein the cell is a fungal cell.
121. A method of claim 111, wherein expression of the polypeptide is associated with production of a desired product.
122. A method of claim 121, wherein the desired product is an antibiotic.
123. A method of claim 121, wherein the desired product is citric acid.
124. A biological method for recovering a substance from a mixture containing the substance which involves contacting the mixture with cells so as to separately recover the substance, which cells (i) comprise DNA encoding, and (ii) are capable of expressing a gene product, which gene product facilitates separating the substance from the mixture so as to recover the substance from the mixture, the improvement comprising (1) treating the cells with a molecule which (a) does not naturally occur in the cell and (b) binds to DNA or RNA or binds to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with increased production of the gene product.
125. A method of claim 124, wherein the substance is a metal.
126. A biological method for treating a substance with cells so as to effect a biochemical transformation by contacting the substance with cells which (i) comprises DNA encoding, and (ii) is capable of expressing a gene product which permits biochemical transformation, the improvement comprising contacting the cells with a molecule which (a) does not naturally occur in the cell and (b) binds to DNA or RNA or binds to a protein through a domain of such protein which is not a ligand binding domain of a receptor which naturally occurs in the cell, the binding of a ligand to which ligand binding domain is normally associated with enhanced production of the gene product.
127. A method of claim 126, wherein the biochemical transformation is associated with the production of a steroid.
128. A method of claim 126, wherein the biochemical transformation is associated with the production of an alcohol.
129. A method of claim 126, wherein the biochemical transformation is associated with the degradation of petroleum products.
130. A method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00097
effective to enhance the expression of human growth hormone by the cell.
131. A method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00098
effective to enhance the expression of human growth hormone by the cell.
132. A method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00099
effective to enhance the expression of human growth hormone by the cell.
133. A method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00100
effective to enhance the expression of human growth hormone by the cell.
134. A method for enhancing the expression of human growth hormone by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, human growth hormone, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00101
wherein R is
Figure US20030166281A1-20030904-C00102
effective to enhance the expression of human growth hormone by the cell.
135. A method for enhancing the expression of G-CSF by a cell which (i) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00103
effective to enhance the expression of G-CSF by the cell.
136. A method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of molecule having the structure:
Figure US20030166281A1-20030904-C00104
effective to enhance the expression of G-CSF by the cell.
137. A method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00105
effective to enhance the expression of G-CSF by the cell.
138. A method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00106
effective to enhance the expression of G-CSF by the cell.
139. A method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00107
wherein R1 is hydrogen or a lower alkyl group,
wherein R2 is hydrogen or a lower alkyl group,
wherein X is oxygen or —NH2,
wherein Y is bromine, fluorine, chlorine, or iodine, and
which molecule is effective to enhance the expression of G-CSF by the cell.
140. A method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00108
effective to enhance the expression of G-CSF by the cell.
141. A method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00109
effective to enhance the expression of G-CSF by the cell.
142. A method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00110
effective to enhance the expression of G-CSF by the cell.
143. A method for enhancing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00111
effective to enhance the expression of G-CSF by the cell.
144. A method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00112
effective to decrease the expression of G-CSF by the human being.
145. A method of suppressing the proliferation of a mammary tumor virus in a subject who (i) comprises DNA encoding, and (ii) is capable of expressing, a mammary tumor virus, comprising administering to the subject an amount of a molecule having the structure:
115
effective to suppress the proliferation of a mammary tumor virus in the subject.
146. A method for decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00113
effective to decrease the expression of G-CSF by the human being.
147. A method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00114
effective to decrease expression of G-CSF by, and thus decrease the formation and effect the metabolic functions of neutrophils in, the human being.
148. A method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00115
effective to decrease the expression of G-CSF by the human being.
149. A method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00116
effective to decrease expression of G-CSF by, and thus decrease the formation and effect the metabolic functions of neutrophils in, the human being.
150. A method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00117
effective to decrease the expression of G-CSF by the human being.
151. A method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00118
effective to decrease expression of G-CSF by, and thus decrease the formation and effect the metabolic functions of neutrophils in, the human being.
152. A method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00119
effective to decrease the expression of G-CSF by the human being.
153. A method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00120
effective to decrease expression of G-CSF by, and thus decrease the formation and effect the metabolic functions of neutrophils in, the human being.
154. A method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00121
effective to decrease the expression of G-CSF by the human being.
155. A method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00122
effective to decrease expression of G-CSF by, and thus decrease the formation and effect the metabolic functions of neutrophils in, the human being.
156. A method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00123
effective to decrease the expression of G-CSF by the human being.
157. A method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00124
effective to decrease expression of G-CSF by, and thus decrease the formation and effect the metabolic functions of neutrophils in, the human being.
158. A method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00125
effective to decrease the expression of G-CSF by the human being.
159. A method of decreasing the formation of neutrophils and affecting the metabolic functions of neutrophils in a human being who (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising administering to the human being an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00126
effective to decrease expression of G-CSF by, and thus decrease the formation and effect the metabolic functions of neutrophils in, the human being.
160. A method of decreasing the expression of G-CSF by a cell which (i) comprises DNA encoding, and (ii) is capable of expressing, G-CSF, comprising contacting the cell with an amount of a molecule having the structure:
Figure US20030166281A1-20030904-C00127
effective to decrease the expression of G-CSF by the human being.
US10/124,113 1989-07-18 2002-04-16 Methods of discovering chemicals capable of functioning as gene expression modulators Abandoned US20030166281A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/124,113 US20030166281A1 (en) 1989-07-18 2002-04-16 Methods of discovering chemicals capable of functioning as gene expression modulators
US11/704,094 US20080009060A1 (en) 1989-07-18 2007-02-08 Methods of discovering chemicals capable of functioning as gene expression modulators

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US38271289A 1989-07-18 1989-07-18
US55519690A 1990-07-18 1990-07-18
US1334393A 1993-02-04 1993-02-04
US13421593A 1993-10-08 1993-10-08
US08/267,834 US5665543A (en) 1989-07-18 1994-06-28 Method of discovering chemicals capable of functioning as gene expression modulators
US08/683,455 US5976793A (en) 1989-07-18 1996-07-18 Methods of transcriptionally modulating gene expression and of discovering chemicals capable as gene expression modulators
US09/123,728 US6376175B1 (en) 1989-07-18 1998-07-28 Methods of discovering chemicals capable of functioning as gene expression modulators
US10/124,113 US20030166281A1 (en) 1989-07-18 2002-04-16 Methods of discovering chemicals capable of functioning as gene expression modulators

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/123,728 Continuation US6376175B1 (en) 1989-07-18 1998-07-28 Methods of discovering chemicals capable of functioning as gene expression modulators

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/704,094 Continuation US20080009060A1 (en) 1989-07-18 2007-02-08 Methods of discovering chemicals capable of functioning as gene expression modulators

Publications (1)

Publication Number Publication Date
US20030166281A1 true US20030166281A1 (en) 2003-09-04

Family

ID=27486286

Family Applications (6)

Application Number Title Priority Date Filing Date
US08/267,834 Expired - Lifetime US5665543A (en) 1989-07-18 1994-06-28 Method of discovering chemicals capable of functioning as gene expression modulators
US08/683,455 Expired - Lifetime US5976793A (en) 1989-07-18 1996-07-18 Methods of transcriptionally modulating gene expression and of discovering chemicals capable as gene expression modulators
US08/778,754 Expired - Fee Related US6136779A (en) 1989-07-18 1997-01-06 Methods of specifically transcriptionally modulating the expression of gene of interest
US09/123,728 Expired - Fee Related US6376175B1 (en) 1989-07-18 1998-07-28 Methods of discovering chemicals capable of functioning as gene expression modulators
US10/124,113 Abandoned US20030166281A1 (en) 1989-07-18 2002-04-16 Methods of discovering chemicals capable of functioning as gene expression modulators
US11/704,094 Abandoned US20080009060A1 (en) 1989-07-18 2007-02-08 Methods of discovering chemicals capable of functioning as gene expression modulators

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US08/267,834 Expired - Lifetime US5665543A (en) 1989-07-18 1994-06-28 Method of discovering chemicals capable of functioning as gene expression modulators
US08/683,455 Expired - Lifetime US5976793A (en) 1989-07-18 1996-07-18 Methods of transcriptionally modulating gene expression and of discovering chemicals capable as gene expression modulators
US08/778,754 Expired - Fee Related US6136779A (en) 1989-07-18 1997-01-06 Methods of specifically transcriptionally modulating the expression of gene of interest
US09/123,728 Expired - Fee Related US6376175B1 (en) 1989-07-18 1998-07-28 Methods of discovering chemicals capable of functioning as gene expression modulators

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/704,094 Abandoned US20080009060A1 (en) 1989-07-18 2007-02-08 Methods of discovering chemicals capable of functioning as gene expression modulators

Country Status (1)

Country Link
US (6) US5665543A (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050080021A1 (en) * 2002-08-15 2005-04-14 Joseph Tucker Nitric oxide donating derivatives of stilbenes, polyphenols and flavonoids for the treatment of cardiovascular disorders
US20050080024A1 (en) * 2002-08-15 2005-04-14 Joseph Tucker Nitric oxide donating derivatives for the treatment of cardiovascular disorders
US20060147904A1 (en) * 2002-08-15 2006-07-06 Resverlogix, Inc. Use of resveratrol to regulate expression of apolipoprotein A1
US8076159B2 (en) 2006-08-04 2011-12-13 The Board Of Trustees Of The Leland Stanford Junior University Ligand-regulable transactivation systems, methods of use thereof, methods of detecting estrogen receptor ligands, and methods of differentiating estrogen receptor ligand agonists and antagonists
WO2012092645A1 (en) * 2011-01-04 2012-07-12 Co-Operative Research Centre For Asthma And Airways Novel antisense oligonucleotides

Families Citing this family (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6060592A (en) * 1990-01-11 2000-05-09 Isis Pharmaceuticals, Inc. Pyrimidine nucleoside compounds and oligonucleoside compounds containing same
US5610289A (en) 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
DE69132546T2 (en) 1990-09-13 2001-10-04 Univ Durham EXPRESSION OF RECEPTORS COUPLED TO G-PROTEIN IN YEAST
ES2111289T5 (en) * 1993-01-21 2005-07-16 President And Fellows Of Harvard College METHODS AND DIAGNOSIS KITS THAT USE STRESS PROMOTERS IN MAMMALS TO DETERMINE THE TOXICITY OF A COMPOUND.
GB9415167D0 (en) * 1994-07-27 1994-09-14 Springer Caroline J Improvements relating to cancer therapy
US6090544A (en) * 1995-07-26 2000-07-18 Creative Biomolecules, Inc. Methods and compositions for identifying morphogen analogs
US5834188A (en) * 1995-07-26 1998-11-10 Creative Biomolecule, Inc. Methods and compositions for identifying morphogen analogs
US7306903B1 (en) 1995-07-26 2007-12-11 Curis, Inc. Methods and compositions for identifying morphogen analogs
FR2749857B1 (en) * 1996-06-12 1998-08-14 Centre Nat Rech Scient GENERATION OF IN VIVO REPLICATIVE MOLECULES
US6004808A (en) * 1996-06-21 1999-12-21 Aurora Biosciences Corporation Promiscuous G-protein compositions and their use
US6358737B1 (en) * 1996-07-31 2002-03-19 Board Of Regents, The University Of Texas System Osteocyte cell lines
JP2000517185A (en) 1996-08-29 2000-12-26 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Novel metalloprotease family KUZ
US6566089B1 (en) * 1996-09-04 2003-05-20 Tularik Inc. Cell-based drug screens for regulators of gene expression
US5928888A (en) * 1996-09-26 1999-07-27 Aurora Biosciences Corporation Methods and compositions for sensitive and rapid, functional identification of genomic polynucleotides and secondary screening capabilities
US5985214A (en) * 1997-05-16 1999-11-16 Aurora Biosciences Corporation Systems and methods for rapidly identifying useful chemicals in liquid samples
US6586661B1 (en) 1997-06-12 2003-07-01 North Carolina State University Regulation of quinolate phosphoribosyl transferase expression by transformation with a tobacco quinolate phosphoribosyl transferase nucleic acid
US20030220234A1 (en) * 1998-11-02 2003-11-27 Selvaraj Naicker Deuterated cyclosporine analogs and their use as immunodulating agents
US5849581A (en) * 1997-10-09 1998-12-15 Tularik Inc. Regulators of UCP3 gene expression
US6303302B1 (en) 1997-11-19 2001-10-16 The Whitehead Institute For Biomedical Research Regulation of fungal gene expression
AU2325099A (en) * 1998-01-16 1999-08-02 University Of Virginia Patent Foundation Identification of a smooth muscle cell (smc) specific smooth muscle myosin heavychain (sm-mhc) promoter/enhancer
US6780610B1 (en) 1998-01-16 2004-08-24 Setagon, Inc. Identification of a smooth muscle cell (SMC) specific smooth muscle heavy chain (SM-MHC) promoter/enhancer
US6388788B1 (en) 1998-03-16 2002-05-14 Praelux, Inc. Method and apparatus for screening chemical compounds
US20030036855A1 (en) * 1998-03-16 2003-02-20 Praelux Incorporated, A Corporation Of New Jersey Method and apparatus for screening chemical compounds
WO2000004051A2 (en) * 1998-07-15 2000-01-27 Metamorphix, Inc. Growth differentiation factor promoter and uses therefor
US20030219427A1 (en) * 1998-08-18 2003-11-27 Allen Hamish J. TPL-2/COT kinase and methods of use
GB9819666D0 (en) * 1998-09-09 1998-11-04 Univ Aberdeen Biosensor
US8124630B2 (en) 1999-01-13 2012-02-28 Bayer Healthcare Llc ω-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
ATE556713T1 (en) 1999-01-13 2012-05-15 Bayer Healthcare Llc OMEGA-CARBOXYARYL SUBSTITUTED DIPHENYL UREAS AS P38 KINASE INHIBITORS
WO2000060122A1 (en) * 1999-04-05 2000-10-12 Gene Logic, Inc. IDENTIFICATION OF A cDNA ASSOCIATED WITH ISCHEMIA IN HUMAN HEART TISSUE
DE19926216A1 (en) * 1999-06-09 2001-02-22 Metallgesellschaft Ag Process for producing barium sulfate, barium sulfate and use of barium sulfate
US6673554B1 (en) * 1999-06-14 2004-01-06 Trellie Bioinformatics, Inc. Protein localization assays for toxicity and antidotes thereto
US6479241B1 (en) * 1999-09-10 2002-11-12 Southern Research Institute High throughput screening of the effects of anti-cancer agents on expression of cancer related genes in various cell lines
US6949356B1 (en) 1999-10-20 2005-09-27 Microbia, Inc. Methods for improving secondary metabolite production in fungi
CA2301158A1 (en) * 2000-03-24 2001-09-24 Stephen J. Allen Screening methods for compounds useful for modulating lipid metabolism in disease
CA2405440A1 (en) * 2000-04-12 2001-10-25 Puracyp Compositions and methods for induction of proteins involved in xenobiotic metabolism
US20040077080A1 (en) * 2000-04-12 2004-04-22 Judy Raucy Compositions and methods for induction of proteins involved in xenobiotic metabolism
US20040115627A1 (en) * 2000-04-12 2004-06-17 Judy Raucy Compositions and methods for induction of proteins involved in xenobiotic metabolism
US7071323B2 (en) * 2000-05-02 2006-07-04 Wisconsin Alumni Research Foundation Promoter sequences for corticotropin releasing-factor receptor CRF2α and method of identifying agents that alter the activity of the promoter sequences
JP2002034575A (en) * 2000-07-28 2002-02-05 Shiseido Co Ltd Human type ii 5 alpha-reductase promoter gene and its use
DE60121603T2 (en) * 2000-08-30 2007-06-21 North Carolina State University TRANSGENIC PLANTS CONTAINING MOLECULAR DECOYS THAT CHANGE THE PROTEIN CONTENT
US20040185094A1 (en) * 2000-09-01 2004-09-23 Maw Graham Nigel Pharmaceutical
EP1356061A2 (en) * 2000-10-13 2003-10-29 Crosslink Genetics Corporation Artificial transcriptional factors and methods of use
CA2427472A1 (en) 2000-10-27 2002-06-06 Tae Kook Kim Double and triple readout assay systems
KR20020035393A (en) * 2000-11-06 2002-05-11 신호진 Reporter Vectors for Evaluating the Activity of Transcription Factors or Promoters and Cell Lines Transformed with the same
WO2002038588A2 (en) * 2000-11-07 2002-05-16 North Carolina State University Putrescine-n-methyltransferase promoter
AU2002259160A1 (en) * 2001-05-07 2002-11-18 Thomas Jefferson University Oligonucleotide inhibitors of cancer cell proliferation
US20060157072A1 (en) * 2001-06-08 2006-07-20 Anthony Albino Method of reducing the harmful effects of orally or transdermally delivered nicotine
WO2003000730A1 (en) * 2001-06-21 2003-01-03 The General Hospital Corporation Compounds that modulate estrogen actions
MXPA04003625A (en) 2001-10-19 2004-12-02 Sotechnika Inc Synthesis of cyclosporin analogs.
WO2003068228A1 (en) 2002-02-11 2003-08-21 Bayer Pharmaceuticals Corporation Aryl ureas with angiogenesis inhibiting activity
US20050282814A1 (en) * 2002-10-03 2005-12-22 Targegen, Inc. Vasculostatic agents and methods of use thereof
EP1549614A4 (en) * 2002-10-03 2008-04-16 Targegen Inc Vasculostatic agents and methods of use thereof
EP1570061A2 (en) * 2002-12-05 2005-09-07 Imperial College Innovations Limited Control of apoptosis using a complex of an oligonucleotide and a regulatory peptide
UY28213A1 (en) 2003-02-28 2004-09-30 Bayer Pharmaceuticals Corp NEW CYANOPIRIDINE DERIVATIVES USEFUL IN THE TREATMENT OF CANCER AND OTHER DISORDERS.
PT1626714E (en) 2003-05-20 2007-08-24 Bayer Pharmaceuticals Corp Diaryl ureas for diseases mediated by pdgfr
CN1856469B (en) 2003-07-23 2013-03-06 拜耳医药保健有限责任公司 Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
JP2007521806A (en) * 2004-01-16 2007-08-09 セレス,インコーポレーテッド Plant cell with receptor polypeptide
EP1809614B1 (en) 2004-04-08 2014-05-07 TargeGen, Inc. Benzotriazine inhibitors of kinases
KR101347830B1 (en) 2004-05-23 2014-01-07 에이치엠아이, 인코포레이티드 Theramutein modulators
NZ588896A (en) 2004-08-25 2012-05-25 Targegen Inc Heterocyclic compounds and methods of use
US8431110B2 (en) 2005-05-23 2013-04-30 Hmi Medical Innovations, Llc. Compounds and method of identifying, synthesizing, optimizing and profiling protein modulators
BRPI0606172A2 (en) * 2005-06-08 2009-06-02 Targegen Inc methods and compositions for treating eye disorders
RU2448959C2 (en) * 2005-11-01 2012-04-27 Таргеджен, Инк. Bi-aryl-metha-pyrimidine kinase inhibitors
US8604042B2 (en) * 2005-11-01 2013-12-10 Targegen, Inc. Bi-aryl meta-pyrimidine inhibitors of kinases
US8133900B2 (en) * 2005-11-01 2012-03-13 Targegen, Inc. Use of bi-aryl meta-pyrimidine inhibitors of kinases
JP2009523408A (en) * 2005-11-23 2009-06-25 ジェラルド・エム・ハウシー Group of compounds and methods for identifying, synthesizing, optimizing and analyzing protein modulators
WO2007127366A2 (en) * 2006-04-25 2007-11-08 Targegen, Inc. Kinase inhibitors and methods of use thereof
US8029980B2 (en) * 2006-09-29 2011-10-04 The Board Of Trustees Of The University Of Illinois Identification and use of agents that modulate oncogenic transcription agent activity
US20090011516A1 (en) * 2007-07-03 2009-01-08 Pioneer Hi-Bred International, Inc. Methods and Assays for the Detection of Nitrogen Uptake by a Plant and Uses Thereof
AU2010363329A1 (en) 2010-11-07 2013-05-09 Targegen, Inc. Compositions and methods for treating myelofibrosis
WO2012083363A1 (en) 2010-12-22 2012-06-28 Murdoch Childrens Research Institute A method of treatment

Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4535058A (en) * 1982-10-01 1985-08-13 Massachusetts Institute Of Technology Characterization of oncogenes and assays based thereon
US4601978A (en) * 1982-11-24 1986-07-22 The Regents Of The University Of California Mammalian metallothionein promoter system
US4699877A (en) * 1982-11-04 1987-10-13 The Regents Of The University Of California Methods and compositions for detecting human tumors
US4736866B1 (en) * 1984-06-22 1988-04-12 Transgenic non-human mammals
US4738922A (en) * 1984-05-25 1988-04-19 Dana Farber Cancer Institute Trans-acting transcriptional factors
US4740461A (en) * 1983-12-27 1988-04-26 Genetics Institute, Inc. Vectors and methods for transformation of eucaryotic cells
US4740463A (en) * 1984-04-13 1988-04-26 Massachusetts Institute Of Technology Methods and artificial genes for antagonizing the function of an oncogene
US4761367A (en) * 1984-11-07 1988-08-02 The University Of North Carolina At Chapel Hill Vectors suitable for detection of eukaryotic DNA regulatory sequences
US4761371A (en) * 1985-02-12 1988-08-02 Genentech, Inc. Insulin receptor
US4806463A (en) * 1986-05-23 1989-02-21 Worcester Foundation For Experimental Biology Inhibition of HTLV-III by exogenous oligonucleotides
US4827079A (en) * 1983-08-22 1989-05-02 Dna Plant Technology Corporation Generation of somaclonal non-mendelian variants
US4861709A (en) * 1985-05-31 1989-08-29 Technicon Research A.G. Detection and/or identification of microorganisms in a test sample using bioluminescence or other exogenous genetically-introduced marker
US4885238A (en) * 1987-10-30 1989-12-05 The United States Of America As Represented By The Department Of Health And Human Services Immortalized human bronchial epitherial mesothelial cell lines
US4935363A (en) * 1987-03-30 1990-06-19 Board Of Regents, The University Of Texas System Sterol regulatory elements
US4981784A (en) * 1987-12-02 1991-01-01 The Salk Institute For Biological Studies Retinoic acid receptor method
US4981783A (en) * 1986-04-16 1991-01-01 Montefiore Medical Center Method for detecting pathological conditions
US5070012A (en) * 1988-03-30 1991-12-03 The Board Of Trustees Of The Leland Stanford Junior University Monitoring of cells and trans-activating transcription elements
US5071773A (en) * 1986-10-24 1991-12-10 The Salk Institute For Biological Studies Hormone receptor-related bioassays
US5075229A (en) * 1987-06-16 1991-12-24 Ohio University Edison Animal Biotechnology Center Dietary and hormonal regulation of expression of exogenous genes in transgenic animals under control of the promoter of the gene for phosphoenolpyruvate carboxykinase
US5262300A (en) * 1988-11-30 1993-11-16 The Salk Institute For Biological Studies Receptors: their identification, characterization, preparation and use
US5578483A (en) * 1988-12-23 1996-11-26 The Salk Institute For Biological Studies Receptor transcription-repression activity compositions and methods
US5602009A (en) * 1988-12-23 1997-02-11 The Salk Institute For Biological Studies Dominant negative chimeras of the steroid/thyroid superfamily of receptors

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5804374A (en) 1980-12-05 1998-09-08 Massachusetts Insti. Technology Nuclear factors associates with transcriptional regulation
US4713339A (en) * 1983-01-19 1987-12-15 Genentech, Inc. Polycistronic expression vector construction
US5087438A (en) * 1984-07-03 1992-02-11 Gordon Robert T Method for affecting intracellular and extracellular electric and magnetic dipoles
US4699978A (en) * 1985-01-18 1987-10-13 The Trustees Of Columbia University In The City Of New York Site-specific chiral ruthenium (II) and cobalt (III) antitumor agents
US4721669A (en) * 1985-01-18 1988-01-26 The Trustees Of Columbia University In The City Of New York Chemical probes for left-handed DNA and chiral metal complexes as Z-specific anti-tumor agents
US4810643A (en) * 1985-08-23 1989-03-07 Kirin- Amgen Inc. Production of pluripotent granulocyte colony-stimulating factor
WO1988005083A1 (en) 1986-12-24 1988-07-14 Whitehead Institute For Biomedical Research Method of inducible gene expression
WO1989002472A1 (en) * 1987-09-21 1989-03-23 Amrad Corporation Limited Regulation of expression of gm-csf gene
WO1989007614A1 (en) 1988-02-12 1989-08-24 Massachusetts Institute Of Technology Nuclear factors associated with transcriptional regulation
IL89495A (en) * 1988-03-08 1995-08-31 Ciba Geigy Ag Chemically regulatable DNA sequence its use for regulating transcription of an associated DNA sequence in plants or plant tissue and its production
US5540931A (en) * 1989-03-03 1996-07-30 Charles W. Hewitt Methods for inducing site-specific immunosuppression and compositions of site specific immunosuppressants
US5776502A (en) * 1989-07-18 1998-07-07 Oncogene Science, Inc. Methods of transcriptionally modulating gene expression
US5580722A (en) * 1989-07-18 1996-12-03 Oncogene Science, Inc. Methods of determining chemicals that modulate transcriptionally expression of genes associated with cardiovascular disease

Patent Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4535058A (en) * 1982-10-01 1985-08-13 Massachusetts Institute Of Technology Characterization of oncogenes and assays based thereon
US4699877A (en) * 1982-11-04 1987-10-13 The Regents Of The University Of California Methods and compositions for detecting human tumors
US4601978A (en) * 1982-11-24 1986-07-22 The Regents Of The University Of California Mammalian metallothionein promoter system
US4827079A (en) * 1983-08-22 1989-05-02 Dna Plant Technology Corporation Generation of somaclonal non-mendelian variants
US4740461A (en) * 1983-12-27 1988-04-26 Genetics Institute, Inc. Vectors and methods for transformation of eucaryotic cells
US4740463A (en) * 1984-04-13 1988-04-26 Massachusetts Institute Of Technology Methods and artificial genes for antagonizing the function of an oncogene
US4738922A (en) * 1984-05-25 1988-04-19 Dana Farber Cancer Institute Trans-acting transcriptional factors
US4981790A (en) * 1984-05-25 1991-01-01 Dana Farber Cancer Institute Stable TatIII cell lines, TatIII gene products, and assay methods
US4736866B1 (en) * 1984-06-22 1988-04-12 Transgenic non-human mammals
US4736866A (en) * 1984-06-22 1988-04-12 President And Fellows Of Harvard College Transgenic non-human mammals
US4761367A (en) * 1984-11-07 1988-08-02 The University Of North Carolina At Chapel Hill Vectors suitable for detection of eukaryotic DNA regulatory sequences
US4761371A (en) * 1985-02-12 1988-08-02 Genentech, Inc. Insulin receptor
US4861709A (en) * 1985-05-31 1989-08-29 Technicon Research A.G. Detection and/or identification of microorganisms in a test sample using bioluminescence or other exogenous genetically-introduced marker
US4981783A (en) * 1986-04-16 1991-01-01 Montefiore Medical Center Method for detecting pathological conditions
US4806463A (en) * 1986-05-23 1989-02-21 Worcester Foundation For Experimental Biology Inhibition of HTLV-III by exogenous oligonucleotides
US5071773A (en) * 1986-10-24 1991-12-10 The Salk Institute For Biological Studies Hormone receptor-related bioassays
US4935363A (en) * 1987-03-30 1990-06-19 Board Of Regents, The University Of Texas System Sterol regulatory elements
US5075229A (en) * 1987-06-16 1991-12-24 Ohio University Edison Animal Biotechnology Center Dietary and hormonal regulation of expression of exogenous genes in transgenic animals under control of the promoter of the gene for phosphoenolpyruvate carboxykinase
US4885238A (en) * 1987-10-30 1989-12-05 The United States Of America As Represented By The Department Of Health And Human Services Immortalized human bronchial epitherial mesothelial cell lines
US4981784A (en) * 1987-12-02 1991-01-01 The Salk Institute For Biological Studies Retinoic acid receptor method
US5070012A (en) * 1988-03-30 1991-12-03 The Board Of Trustees Of The Leland Stanford Junior University Monitoring of cells and trans-activating transcription elements
US5262300A (en) * 1988-11-30 1993-11-16 The Salk Institute For Biological Studies Receptors: their identification, characterization, preparation and use
US5578483A (en) * 1988-12-23 1996-11-26 The Salk Institute For Biological Studies Receptor transcription-repression activity compositions and methods
US5602009A (en) * 1988-12-23 1997-02-11 The Salk Institute For Biological Studies Dominant negative chimeras of the steroid/thyroid superfamily of receptors

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050080021A1 (en) * 2002-08-15 2005-04-14 Joseph Tucker Nitric oxide donating derivatives of stilbenes, polyphenols and flavonoids for the treatment of cardiovascular disorders
US20050080024A1 (en) * 2002-08-15 2005-04-14 Joseph Tucker Nitric oxide donating derivatives for the treatment of cardiovascular disorders
US20060147904A1 (en) * 2002-08-15 2006-07-06 Resverlogix, Inc. Use of resveratrol to regulate expression of apolipoprotein A1
US8076159B2 (en) 2006-08-04 2011-12-13 The Board Of Trustees Of The Leland Stanford Junior University Ligand-regulable transactivation systems, methods of use thereof, methods of detecting estrogen receptor ligands, and methods of differentiating estrogen receptor ligand agonists and antagonists
WO2012092645A1 (en) * 2011-01-04 2012-07-12 Co-Operative Research Centre For Asthma And Airways Novel antisense oligonucleotides

Also Published As

Publication number Publication date
US20080009060A1 (en) 2008-01-10
US6136779A (en) 2000-10-24
US5665543A (en) 1997-09-09
US6376175B1 (en) 2002-04-23
US5976793A (en) 1999-11-02

Similar Documents

Publication Publication Date Title
US6376175B1 (en) Methods of discovering chemicals capable of functioning as gene expression modulators
US5863733A (en) Methods of transcriptionally modulating gene expression and of discovering chemicals capable of functioning as gene expression modulators
US6165712A (en) Methods of transcriptionally modulating expression of viral genes and genes useful for production of proteins
US6589733B1 (en) Methods of preparing compositions comprising chemicals capable of transcriptional modulators
Südhof et al. 42 bp element from LDL receptor gene confers end-product repression by sterols when inserted into viral TK promoter
US5580722A (en) Methods of determining chemicals that modulate transcriptionally expression of genes associated with cardiovascular disease
Deiss et al. A genetic tool used to identify thioredoxin as a mediator of a growth inhibitory signal
Cunningham et al. Expression of the DAL80 gene, whose product is homologous to the GATA factors and is a negative regulator of multiple nitrogen catabolic genes in Saccharomyces cerevisiae, is sensitive to nitrogen catabolite repression
Johansen et al. Regulated expression at high copy number allows production of a growth-inhibitory oncogene product in Drosophila Schneider cells.
JP2580036B2 (en) Expression of human serum albumin in methylotrophic yeast
Suzuki-Yagawa et al. Housekeeping Na, K-ATPase α1 subunit gene promoter is composed of multiple cis elements to which common and cell type-specific factors bind
Lagosky et al. Molecular characterization of the Saccharomyces cerevisiae dihydrofolate reductase gene (DFRl)
Inoue et al. The 5′‐flanking region of the human P‐450 (SCC) gene shows responsiveness to CAMP‐dependent regulation in a transient gene‐expression system of Y‐1 adrenal tumor cells
WO1992013063A1 (en) Methods of transcriptionally modulating expression of growth factor genes and growth factor receptor genes
WO1992012635A1 (en) Methods of transcriptionally modulating gene expression of viral genes and other genes
US5759806A (en) Coding, promoter and regulator sequences of IRF-1
JPH025862A (en) Secretive gene manifestation indicator gene product
FI106720B (en) Preparation of fusion proteins in vitro
WO1992013092A1 (en) Methods of transcriptionally modulating expression of hematopoietic growth factor genes
Yamaguchi et al. Cell‐type‐specific expression of mouse DNA polymerase β‐gene is regulated by silencer elements
Leshkowitz et al. Insulin-producing cells contain a cell-specific repressor activity that functions through multiple E-box sequences
Gelb Genomic structure of and a cardiac promoter for the mousecarnitine palmitoyltransferase II gene
Detke Identification of a transcription factor like protein at the TOR locus in Leishmania mexicana amazonensis
JPH06503718A (en) Production of human lysothyme and its efficient secretion in methylotrophic yeast cells
Wurm et al. Use of transfected and amplified Drosophila heat shock promoter construction for inducible production of toxic mouse c-myc proteins in CHO cells

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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