WO1999033985A2 - C1F150/hTAFII150 IS NECESSARY FOR CELL CYCLE PROGRESSION - Google Patents

C1F150/hTAFII150 IS NECESSARY FOR CELL CYCLE PROGRESSION Download PDF

Info

Publication number
WO1999033985A2
WO1999033985A2 PCT/US1998/027665 US9827665W WO9933985A2 WO 1999033985 A2 WO1999033985 A2 WO 1999033985A2 US 9827665 W US9827665 W US 9827665W WO 9933985 A2 WO9933985 A2 WO 9933985A2
Authority
WO
WIPO (PCT)
Prior art keywords
htaf
protein
cif150
gene
seq
Prior art date
Application number
PCT/US1998/027665
Other languages
French (fr)
Other versions
WO1999033985A3 (en
Inventor
Joerg Kaufmann
Original Assignee
Chiron Corporation
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 Chiron Corporation filed Critical Chiron Corporation
Priority to AU19483/99A priority Critical patent/AU1948399A/en
Publication of WO1999033985A2 publication Critical patent/WO1999033985A2/en
Publication of WO1999033985A3 publication Critical patent/WO1999033985A3/en

Links

Classifications

    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the invention relates to the area of cell cycle progression. More particularly, the invention relates to proteins which regulate cell cycle progression.
  • BACKGROUND OF THE INVENTION Alterations in the regulation of mitosis or cell cycle progression play an important role in diseases such as neoplasia and anemia. Manipulation of genes involved in regulating the cell cycle can be used to prevent or treat these diseases. Detections of mutations in cell-cycle regulatory genes can also be used to detect neoplastic cells and genetic predispositions to neoplasias. Thus, there is a need in the art for the identification of cell cycle regulator genes which can be used in methods of diagnosing, prognosing, and treating neoplasia and other diseases in humans and other mammals.
  • One embodiment of the invention is an isolated and purified subgenomic polynucleotide which encodes a protein comprising an amino acid sequence which is at least 85% identical to the nucleotide sequence shown in SEQ ID NO:2. Percent identity is determined using a Smith- Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 1.
  • Another embodiment of the invention is an isolated and purified subgenomic polynucleotide which comprises at least 11 contiguous nucleotides selected from the nucleotide sequence shown in SEQ ID NO:l.
  • Yet another embodiment of the invention is a construct which comprises a promoter and a polynucleotide segment encoding a human CIF150/hTAF ⁇ 150 protein as shown in SEQ ID NO:2.
  • the polynucleotide segment is located downstream from the promoter. Transcription of the polynucleotide segment initiates at the promoter.
  • Still another embodiment of the invention is a host cell comprising a construct.
  • the construct comprises a promoter and a polynucleotide segment encoding a human CIF150/hTAF ⁇ 150 protein as shown in SEQ ID NO:2.
  • Another embodiment of the invention provides a homologously recombinant cell having incorporated therein a new transcription initiation unit.
  • the new transcription initiation unit comprises an exogenous regulatory sequence, an exogenous exon, and a splice donor site.
  • the transcription initiation unit is located upstream of a coding sequence of a CIF150/hTAFj_ ⁇ l50 gene.
  • the exogenous regulatory sequence directs transcription of the coding sequence of the CIF150/hTAFjjl 50 gene.
  • Even another embodiment of the invention provides a method to aid in the diagnosis or prognosis of neoplasia in a human.
  • Expression of a first CIF150/hTAFjjl50 gene in a first tissue of a human suspected of being neoplastic is compared with expression of a second CIF150/hTAF jl50 gene in a second tissue of a human which is normal.
  • the second CIF150/hTAFjjl50 gene has the coding sequence shown in SEQ ID NO:l.
  • Increased expression of the first CIF150/hTAF/jl50 relative to the second CIF150/hTAFjjl50 gene indicates neoplasia in the first tissue.
  • Yet another embodiment of the invention provides a method to aid in the diagnosis or prognosis of neoplasia in a human.
  • a human CIF150/hTAFjjl 50 gene, mRNA, or protein in a first tissue suspected of being neoplastic is compared with a second human CIF150/hTAFjjl50 gene, mRNA, or protein in a second tissue which is normal.
  • the second CIF150/hTAFjjl 50 gene has the coding sequence shown in SEQ ID NO: 1.
  • a difference between the first and second genes, mRNAs, or proteins in the second tissue indicates neoplasia in the first tissue.
  • Still another embodiment of the invention provides a method to aid in detecting a genetic predisposition to neoplasia in a human.
  • a CIF150/hTAFj l 50 gene, mRNA, or protein in a fetal tissue of a human is compared with a wild-type human
  • CIF150/hTAFj ⁇ l50 gene mRNA, or protein.
  • the wild-type CIF150/hTAFjjl50 gene has the coding sequence shown in SEQ ID NO: 1.
  • a difference between the CIF150/hTAFj l50 gene, mRNA, or protein in the fetal tissue of the human and the wild-type human CIF150/hTAF jl50 gene, mRNA, or protein indicates a genetic predisposition to neoplasia in the human.
  • Even another embodiment of the invention provides a method of screening test compounds for the ability to interfere with the binding of a OF 130 protein to a CIF150/hTAF ⁇ 150 protein.
  • a test compound is contacted with a CIF150/hTAF ⁇ 150- binding domain of a OF 130 protein and a OF130-binding domain of a OF150/hTAF ⁇ 150 protein.
  • the CIF130 protein has the amino acid sequence shown in SEQ ID NO:4.
  • the OF150/hTAF ⁇ 150 protein has the amino acid sequence shown in SEQ ID NO:2.
  • the CIF130-binding domain binds to the OF150/hTAF ⁇ 150-binding domain in the absence of the test compound.
  • the amount of at least one of the OF 130- or OF150/hTAF ⁇ 150-binding domains which is bound or unbound is determined in the presence of the test compound.
  • a test compound which decreases the amount of bound CIF130- or OF150/hTAF ⁇ 150-binding domains or which increases the amount of unbound CIF130- and OF150/hTAF ⁇ 150-binding domains is a potential inducer of mitosis or cell cycle progression.
  • Yet another embodiment of the invention provides a method of screening test compounds for the ability to interfere with the binding of a OF 130 protein to a OF 150/hTAF ⁇ l 50 protein.
  • a cell is contacted with a test compound.
  • the cell comprises two fusion proteins.
  • a first fusion protein comprises (1) a OF150/hTAF ⁇ 150- binding domain of a OF 130 protein and (2) either a DNA binding domain or a transcriptional activating domain.
  • a second fusion protein comprises a OF130-binding domain of a OF150/hTAF ⁇ 150 protein.
  • the CIF130 protein has the amino acid sequence shown in SEQ ID NO:4.
  • the OF150/hTAF ⁇ 150 protein has the amino acid sequence shown in SEQ ID NO:2.
  • the OF130-binding domain binds to the OF150/hTAF ⁇ 150-binding domain. If the first fusion protein comprises a DNA binding domain, then the second fusion protein comprises a transcriptional activating domain. If the first fusion protein comprises a transcriptional activating domain, then the second fusion protein comprises a DNA binding domain. The interaction of the first and second fusion proteins reconstitutes a sequence-specific transcription activating factor.
  • the cell also comprises a reporter gene comprising a DNA sequence to which the DNA binding domain specifically binds. Expression of the reporter gene is measured. A test compound which decreases the expression of the reporter gene is a potential inducer of mitosis or cell cycle progression.
  • a promoter region of the gene is contacted with a CIFl 50/hTAF ⁇ l 50 protein as shown in SEQ ID NO:2.
  • the promoter region of the gene comprises a OF150/hTAF ⁇ 150 binding element.
  • the OF150/hTAF ⁇ 150 binding element comprises a nucleotide sequence 5'-Py X G A G A/C A/Py -3' (SEQ ID NO:7). Expression of the gene is thereby increased.
  • Still another embodiment of the invention is an antisense oligonucleotide as shown in SEQ ID NO:5.
  • Yet another embodiment of the invention provides an isolated and purified subgenomic polynucleotide which comprises 5'-Py X G A G A/C A Py-3' (SEQ ID NO:7).
  • the present invention thus provides the art with reagents and methods of affecting human mitosis or cell cycle progression and treating disorders associated with alterations in mitosis or cell cycle progression.
  • Figure 1 Functional knock out of OF 150/hTAF ⁇ 150 protein leads to cell cycle arrest in G 2 /M and reduced gene expression of cyclin Bl.
  • Figure 1A Analysis of the cell cycle of HeLa cells 36 hours after transfection with the OF150/hTAF ⁇ 150 specific antisense oligomer B or the oligomer Bx (reverse sequence). Concentrations of the oligomers are indicated.
  • Figure IB Cell cycle analysis of IMR 90 cells after 36 hours of oligomer treatment.
  • Figure lC Cell cycle analysis of IMR 90 cells after 36 hours of oligomer treatment.
  • RNA derived from HeLa cells treated with oligomer B or Bx analyzed by quantitative RT-PCR (lanes 1 to 6, 100, 200, 300 nM oligomer B and Bx) and Northern blot analysis (lanes 7 to 10) using OF150/hTAF ⁇ 150 and ⁇ -actin-specific primers or 32 P-labeled cDNA probes.
  • Figure ID HeLa cell extracts from different time points after antisense oligomer treatment analyzed by immunoblotting for the decrease of OF150/hTAF ⁇ 150 protein.
  • a control nuclear extract (lane 1) and a molecular size marker (lane 2) were loaded.
  • Figure IE Figure IE.
  • Figure 2 Regulation of OF 150/hTAF ⁇ l 50 expression and activity is cell cycle dependent.
  • Figure 2 A Quiescent BALB/c 3T3 cells were stimulated to undergo cell cycle traverse by serum addition. Extracts derived from cells at times indicated above were assayed for OF150/hTAF ⁇ 150, cyclin Bl, cyclin A, and cyclin E by immunoblot.
  • Figure 2B HeLa cells were assayed for OF150/hTAF ⁇ 150 and cyclin Bl expression after synchronization for 48 hours by serum deprivation.
  • Figure 2C Control of OF 150/hTAF ⁇ l 50 expression and activity is cell cycle dependent.
  • Figure 2 A Quiescent BALB/c 3T3 cells were stimulated to undergo cell cycle traverse by serum addition. Extracts derived from cells at times indicated above were assayed for OF150/hTAF ⁇ 150, cyclin Bl, cyclin A, and cyclin E by immunoblot.
  • Figure 2B HeLa cells were ass
  • Nuclear extracts of HeLa cells harvested at different time points after release from a double thymidine block were used for in vitro transcription assays using promoter with (odd numbered lanes) and without functional TdT Inr (even numbered lanes). Nuclear extracts were standardized by protein concentration.
  • OF150/hTAF ⁇ 150 is a positive regulator of cyclin Bl and cyclin A transcription.
  • Figure 3 A Purified OF150/hTAF ⁇ 150 was visualized after SDS-PAGE by silver staining.
  • Figure 3 C In vitro transcription was performed using OF150/hTAF ⁇ 150 depleted nuclear extracts (lane 1) or in combination with increasing amounts of recombinant OF150/hTAF ⁇ 150 protein (lane 2 to 4).
  • FIG. 4 OF150/hTAF ⁇ 150 has sequence-specific binding activity.
  • Figure 4A OF150/hTAF ⁇ 150 binding site selection.
  • Figure 4B Addition of anti-CIF 150/hTAF ⁇ l 50 (lane 4) but not preimmune serum (lane 3) supershifted the retarded band.
  • Figure 4C Consensus OF150/hTAF ⁇ 150 binding element (CBE).
  • Figure 4D Point mutational analysis of the OF150/hTAF ⁇ 150 binding element using gel shift experiments.
  • FIG. 1 Identification of a CIF 150/hTAF ⁇ l 50 binding element (CBE) in the cyclin B 1 core promoter. Cofransfection of increasing amounts of CIF 150/hTAF ⁇ 150 expression plasmid with cyclin B 1 wild type and cyclin B 1 promoter mutant. The values represent the average of three experiments.
  • CBE CIF 150/hTAF ⁇ l 50 binding element
  • CIF 150/hTAF ⁇ l 50 recognizes and selects TATA-less core promoters for cell cycle specific genes.
  • OF150/hTAF ⁇ 150 plays an important and selective role in establishing gene expression patterns necessary for progression through the cell cycle.
  • the present invention provides reagents and methods for affecting mitosis or cell cycle progression of human cells and for treating disorders associated with alterations in mitosis or cell cycle progression.
  • OF150/hTAF ⁇ 150 is a necessary positive transcriptional regulator of cell cycle progression through G 2 /M.
  • OF150/hTAF ⁇ 150 is an essential cofactor for TF ⁇ D- dependent transcription from promoters containing initiator elements, such as the adenovirus major late promoter.
  • OF150/hTAF ⁇ 150 directly interacts with the transcription factor hTAF ⁇ 135 and stabilizes TF ⁇ D binding to RNA polymerase II core promoters and can stimulate expression of cell cycle-specific genes such as cyclin A and B 1.
  • the promoters of such genes contain a CIF 150/hTAF ⁇ l 50 binding element which comprises a OF150/hTAF ⁇ 150 binding site, 5'-Py X G A G A/C A/Py-3' (SEQ ID NO:7).
  • OF150/hTAF ⁇ 150 binds to and is negatively regulated by a 130 kD polypeptide, CIF 130, which has sequence homology to ATP-dependent RNA helicases (DEAD-box proteins) implicated in the control of mitosis in Schizosaccharomyces pombe.
  • CIF 130 has sequence homology to ATP-dependent RNA helicases (DEAD-box proteins) implicated in the control of mitosis in Schizosaccharomyces pombe.
  • a nucleotide sequence encoding CIF130 is shown in SEQ ID NO:3.
  • the amino acid sequence of CIF130 is shown in SEQ ID NO:4.
  • Human OF150/hTAF ⁇ 150 protein has the amino acid sequence shown in SEQ ID NO:2. Protein variants of CIF150/hTAF ⁇ 150 protein can also have OF150/hTAF ⁇ 150 activity. Biologically active OF150/hTAF ⁇ 150 variants can be naturally or non-naturally occurring. Naturally occurring OF150/hTAF ⁇ 150 variants are those which are found in humans or other species and which comprise amino acid sequences which are substantially identical to the amino acid sequence shown in SEQ ID NO:2. Non-naturally occurring OF150/hTAF ⁇ 150 variants which retain substantially the same biological activities as OF150/hTAF ⁇ 150 or naturally occurring CIF 150/hTAF ⁇ l 50 variants can be constructed in the laboratory.
  • naturally or non-naturally occurring protein biologically active OF150 hTAF ⁇ 150 variants have amino acid sequences which are at least 65%, 75%, 85%, 90%, or 95% identical to the amino acid sequence shown in SEQ ID NO:2 and have similar biological properties, including the ability to bind to the OF150/hTAF ⁇ 150 binding element, to permit mitosis, to activate TATA-less core promoters, and to bind to CIF130. More preferably, the molecules are at least 98% or 99% identical.
  • Percent sequence identity between the OF150/hTAF ⁇ 150 protein shown in SEQ ID NO:2 and a biologically active OF150 hTAF ⁇ 150 variant is determined using the Smith- Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 1.
  • the Smith- Waterman homology search algorithm is taught in Smith and Waterman, Adv. Appl. Math. (1981) 2:482-489.
  • the amino acid changes in biologically active OF150/hTAF ⁇ 150 variants are conservative amino acid changes, i.e., substitutions of similarly charged or uncharged amino acids.
  • a conservative amino acid change involves substitution of one of a family of amino acids which are related in their side chains.
  • Naturally occurring amino acids are generally divided into four families: acidic (aspartate, glutamate), basic (lysine, arginine, histidine), non-polar (alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), and uncharged polar (glycine, asparagine, glutamine, cystine, serine, threonine, tyrosine) amino acids. Phenylalanine, tryptophan, and tyrosine are sometimes classified jointly as aromatic amino acids.
  • OF150/hTAF ⁇ 150 binding element Properties and functions of OF150/hTAF ⁇ 150 variants are of the same type as a OF150/hTAF ⁇ 150 protein having the amino acid sequence shown in SEQ ID NO:2, although the properties and functions may differ in degree. Whether an amino acid change results in a OF150/hTAF ⁇ 150 protein or polypeptide variant which can function as the CIF 150/hTAF ⁇ l 50 protein disclosed herein can readily be determined. For example, the ability of a OF150/hTAF ⁇ 150 variant to bind to the OF150/hTAF ⁇ 150 binding element can be tested using in vitro DNA binding assays, as taught in Kaufinann & Smale, 1994, Genes Bevel.
  • OF150 hTAF ⁇ 150 polypeptides contain less than full-length OF150/hTAF ⁇ 150.
  • CIF150/hTAF ⁇ 150 polypeptides contain at least 5, 6, 8, 10, 12, 20, 25, 50, 75, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1250, or 1300 or more amino acids of a OF150/hTAF ⁇ 150 protein or biologically active variant in the same primary order as found in a OF150/hTAF ⁇ 150 protein or variant obtained from a natural source.
  • CIF 130 variants include glycosylated forms, aggregative conjugates with other molecules, and covalent conjugates with unrelated chemical moieties.
  • Truncations or deletions of regions which do not affect the properties or functions of OF150/hTAF ⁇ 150 described above are also biologically active variants of OF150/hTAF ⁇ 150.
  • Covalent variants can be prepared by linkage of functionalities to groups which are found in the amino acid chain or at the N- or C-terminal residue, as is known in the art.
  • a subset of mutants, called muteins is a group of polypeptides in which neutral amino acids, such as serines, are substituted for cysteine residues which do not participate in disulfide bonds. These mutants may be stable over a broader temperature range than native OF150/hTAF ⁇ 150. See Mark et al, U.S. Pat. No. 4,959,314.
  • OF150/hTAF ⁇ 150 proteins or polypeptides can be purified from human cells or cell lines, such as HeLa or NIH 3T3 cells, by methods known in the art. CIF150/hTAF ⁇ 150 copurifies with CIF 130; thus, the initial purification steps for each protein are the same. OF150/hTAF ⁇ 150 can be conveniently purified from HeLa cell extracts (Dignam et al, 1983, Nucl. Acids Res. ii:1475-89) using Ni affinity chromatography. For Ni affinity purification, the 0.1 M KO flowthrough fraction of a DEAE-Sephacel column is applied to a Mono Q column and eluted with a linear KCl gradient (40 ml; 0.1 to 1 M).
  • the CIF130/OF150/hTAF ⁇ 150-containing fractions are pooled and dialyzed against buffer A (20 mM HEPES, pH 7.9, 1 mM EDTA, 3 mM dithiothreitol, 1 mM phenylmethylsulfonyl fluoride, 20% glycerol) containing 0.1 M KCl. These fractions are supplemented with imidazole (final concentration, 20 mM in buffer A) and applied to a Ni-nitrilotriacetic acid (NTA)-agarose column (Qiagen). After being washed with 10 column volumes each of 20 mM imidazole and 35 mM imidazole, bound proteins are eluted with 100 mM imidazole.
  • buffer A (20 mM HEPES, pH 7.9, 1 mM EDTA, 3 mM dithiothreitol, 1 mM phenylmethylsulfonyl fluoride
  • Protein fractions can be tested for OF150/hTAF ⁇ 150 activity as described in Kaufinann et al, 1996.
  • Purified CIF130 and CIFl 50/hTAF ⁇ 150 proteins can be visualized by sodium dodecyl sulfate-6% polyacrylamide gel electrophoresis, followed by silver staining.
  • OF150/hTAF ⁇ 150 can be separated from CIF130 by excising the OF150/hTAF ⁇ 150-containing band from the SDS gel and eluting the OF150/hTAF ⁇ 150 protein, as is known in the art.
  • a preparation of isolated and purified OF150/hTAF ⁇ 150 protein is at least 80% pure; preferably, the preparations are at least 90%, 95%, or 99% pure.
  • OF150/hTAF ⁇ 150 proteins and polypeptides can also be produced by recombinant DNA methods or by synthetic chemical methods.
  • coding sequences selected from the CIF150/hTAFjjl50 nucleotide sequence shown in SEQ ID NO: 1 can be expressed in known prokaryotic or eukaryotic expression systems (see below).
  • v/ ' tro-franslated OF150/hTAF ⁇ 150 can be purified with Ni-NTA-agarose as described above (100 mM imidazole eluate) and concentrated with a Centricon 30 concentrator (Amicon).
  • Bacterial, yeast, insect, or mammalian expression systems can be used, as is known in the art.
  • synthetic chemical methods such as solid phase peptide synthesis, can be used to synthesize OF150/hTAF ⁇ 150 protein or polypeptides.
  • OF150/hTAF ⁇ 150 biologically active variants can be similarly produced.
  • Fusion proteins comprising at least six contiguous OF150/hTAF ⁇ 150 amino acids can also be constructed. Human OF150/hTAF ⁇ 150 fusion proteins are useful for generating antibodies against OF150/hTAF ⁇ 150 amino acid sequences and for use in various assay systems.
  • OF150/hTAF ⁇ 150 fusion proteins can be used to identify proteins which interact with CIF 150/hTAF ⁇ l 50 protein and influence its ability to affect transcription.
  • Physical methods such as protein affinity chromatography, or library-based assays for protein-protein interactions, such as the yeast two-hybrid or phage display systems, can be used for this purpose. Such methods are well known in the art and can also be used as drug screens, as described below.
  • a OF150/hTAF ⁇ 150 fusion protein comprises two protein segments fused together by means of a peptide bond.
  • the first protein segment consists of at least 5, 6, 8, 10, 12, 20, 25, 50, 75, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1250, or 1300 contiguous amino acids of a OF150/hTAF ⁇ 150 protein.
  • the amino acids can be selected from the amino acid sequence shown in SEQ ID NO:2 or from a biologically active variant of that sequence.
  • the first protein segment can also be a full-length
  • the first protein segment can be N-terminal or C-terminal, as is convenient.
  • the second protein segment can be a full-length protein or a protein fragment or polypeptide.
  • Proteins commonly used in fusion protein construction include ⁇ - galactosidase, ⁇ -glucuronidase, green fluorescent protein (GFP), autofluorescent proteins, including blue fluorescent protein (BFP), glutathione-S-fransferase (GST), luciferase, horseradish peroxidase (HRP), and chloramphenicol acetyltransferase (CAT).
  • Epitope tags can be used in fusion protein constructions, including histidine (His) tags, FLAG tags, influenza hemagglutinin (HA) tags, Myc tags, NSN-G tags, and thioredoxin (Trx) tags.
  • Other fusion constructions can include maltose binding protein (MBP), S-tag, Lex A D ⁇ A binding domain (DBD) fusions, GAL4 D ⁇ A binding domain fusions, and herpes simplex virus (HSN) BP16 protein fusions.
  • OF150/hTAF ⁇ 150 fusion proteins can be made by covalently linking the first and second protein segments or by standard procedures in the art of molecular biology. Recombinant DNA methods can be used to prepare OF150/hTAF ⁇ 150 fusion proteins, for example, by making a DNA construct which comprises coding sequences selected from SEQ ID NO:l in proper reading frame with nucleotides encoding the second protein segment and expressing the DNA construct in a host cell, as is known in the art.
  • kits for constructing fusion proteins are available from companies which supply research labs with tools for experiments, including, for example, Promega Corporation (Madison, WI), Stratagene (La Jolla, CA), Clontech (Mountain View, CA), Santa Cruz Biotechnology (Santa Cruz, CA), MBL International Corporation (MIC; Watertown, MA), and Quantum Biotechnologies (Montreal, Canada; 1-888-DNA-KITS).
  • Isolated and purified CIF 150/hTAF ⁇ l 50 proteins, polypeptides, variants, or fusion proteins can be used as immunogens, to obtain a preparation of antibodies which specifically bind to a OF150/hTAF ⁇ 150 protein.
  • the antibodies can be used, inter alia, to detect wild-type OF150/hTAF ⁇ 150 proteins in human tissue and fractions thereof.
  • the antibodies can also be used to detect the presence of mutations in the CIF150/hTAFjjl50 gene which result in under- or over-expression of the
  • OF150/hTAF ⁇ 150 protein or in expression of a OF150/hTAF ⁇ 150 protein with altered size or electrophoretic mobility.
  • Antibodies which specifically bind to epitopes of OF150/hTAF ⁇ 150 proteins, polypeptides, fusion proteins, or biologically active variants can be used in immunochemical assays, including but not limited to Western blots, ELISAs, radioimmunoassays, immunohistochemical assays, immunoprecipitations, or other immunochemical assays known in the art.
  • antibodies of the invention provide a detection signal at least 5-, 10-, or 20-fold higher than a detection signal provided with other proteins when used in such immunochemical assays.
  • antibodies which specifically bind to OF150/hTAF ⁇ 150 epitopes do not detect other proteins in immunochemical assays and can immunoprecipitate CIF150/hTAF ⁇ 150 protein or polypeptides from solution.
  • CIF150/hTAF ⁇ 150-specific antibodies specifically bind to epitopes present in a OF150/hTAF ⁇ 150 protein having the amino acid sequence shown in SEQ ID NO:2 or to biologically active variants of that sequence.
  • a OF150/hTAF ⁇ 150 protein having the amino acid sequence shown in SEQ ID NO:2 or to biologically active variants of that sequence.
  • at least 6, 8, 10, or 12 contiguous amino acids are required to form an epitope.
  • epitopes which involve non-contiguous amino acids may require more, e.g., at least 15, 25, or 50 amino acids.
  • CIF150/hTAF ⁇ 150 epitopes are not present in other human proteins.
  • anti-OF 150/hTAF ⁇ l 50 antibodies are polyclonal antibodies raised against an N-terminal peptide of OF150/hTAF ⁇ 150, such as MNRKKGDKGF (amino acids 11-20 of SEQ ID NO:2) or MNRKKGDKGFESPRP (amino acids 11-25 of SEQ ID NO.2).
  • Epitopes of OF150/hTAF ⁇ 150 which are particularly antigenic can be selected, for example, by routine screening of CIF 150/hTAF ⁇ l 50 polypeptides for antigenicity or by applying a theoretical method for selecting antigenic regions of a protein to the amino acid sequence shown in SEQ ID NO:2. Such methods are taught, for example, in Hopp and Wood, Proc. Natl. Acad. Sci. U.S.A. 78, 3824-28 (1981), Hopp and Wood, Mol. Immunol. 20, 483-89 (1983), and Sutcliffe et al, Science 219, 660-66 (1983). Any type of antibody known in the art can be generated to bind specifically to
  • OF150/hTAF ⁇ 150 epitopes For example, preparations of polyclonal and monoclonal antibodies can be made using standard methods which are well known in the art. Similarly, single-chain antibodies can also be prepared. Single-chain antibodies which specifically bind to OF150/hTAF ⁇ 150 epitopes can be isolated, for example, from single-chain immunoglobulin display libraries, as is known in the art. The library is
  • Single-chain antibodies can also be constructed using a DNA amplification method, such as the polymerase chain reaction (PCR), using hybridoma cDNA as a template. Thirion et al, 1996, Eur. J. Cancer Prev. 5:507-11.
  • PCR polymerase chain reaction
  • Single-chain antibodies can be mono- or bispecific, and can be bivalent or tefravalent. Construction of tettavalent, bispecific single-chain antibodies is taught, for example, in Coloma and Morrison, 1997, Nat. Biotechnol. 75:159-63. Construction of bivalent, bispecific single-chain antibodies is taught inter alia in Mallender and Noss, 1994, J Biol. Chem. 269:199-206.
  • a nucleotide sequence encoding a single-chain antibody can be constructed using manual or automated nucleotide synthesis, cloned into an expression construct using standard recombinant D ⁇ A methods, and introduced into a cell to express the coding sequence, as described below.
  • single-chain antibodies can be produced directly using, for example, filamentous phage technology. Nerhaar et al, 1995, Int. J. Cancer 61:491-501; ⁇ icholls et al, 1993, J. Immunol. Meth. 755:81-91.
  • Monoclonal and other antibodies can also be "humanized” in order to prevent a patient from mounting an immune response against an antibody when it is used therapeutically.
  • Such antibodies may be sufficiently similar in sequence to human antibodies to be used directly in therapy or may require alteration of a few key residues. Sequence differences between, for example, rodent antibodies and human sequences can be minimized by replacing residues which differ from those in the human sequences, for example, by site directed mutagenesis of individual residues, or by grafting of entire complementarity determining regions.
  • Antibodies which specifically bind to OF150/hTAF ⁇ 150 epitopes can contain antigen binding sites which are either partially or fully humanized, as disclosed in U.S. 5,565,332.
  • Other types of antibodies can be constructed and used in methods of the invention.
  • chimeric antibodies can be constructed as disclosed, for example, in WO 93/03151.
  • Binding proteins which are derived from immunoglobulins and which are multivalent and multispecific, such as the "diabodies" described in WO 94/13804, can also be prepared.
  • Antibodies of the invention can be purified by methods well known in the art.
  • antibodies can be affinity purified by passing the antibodies over a column to which a CIF150/hTAF ⁇ 150 protein, polypeptide, biologically active variant, or fusion protein is bound. The bound antibodies can then be eluted from the column, using a buffer with a high salt concenfration.
  • OF150/hTAF ⁇ 150-specific binding polypeptides other than antibodies can also be generated.
  • OF150/hTAF ⁇ 150-specific binding polypeptides are polypeptides which bind with OF150/hTAF ⁇ 150 or its variants and which have a measurably higher binding affinity for CIF150/hTAF ⁇ 150 and polypeptide derivatives of OF150/hTAF ⁇ 150 than for other polypeptides tested for binding. Higher affinity by a factor of 10 is preferred, more preferably a factor of 100.
  • Such polypeptides can be identified, for example, using the yeast two-hybrid system.
  • the coding region of the human CIF150/hTAFjjl50 gene has the nucleotide sequence shown in SEQ ID NO: 1.
  • the complement of the nucleotide sequence shown in SEQ ID NO: 1 consists of a contiguous nucleotide sequence which forms Watson-Crick base pairs with the contiguous nucleotide sequence shown in SEQ ID NO:l.
  • CIFl 50/hTAFjjl 50 subgenomic polynucleotides can comprise at least 11, 20, 25, 30, 35, 40, 45, 50, 55, 60, 67, 70, 75, 100, 125, 150, 200, 250, 300, 400, 500, 750, 1000, 1250, 1500, 1750, 2000, 2500, 3000, 3500, or 3900 or more contiguous nucleotides selected from the nucleotide sequence shown in SEQ ID NO:l or its complement.
  • Subgenomic polynucleotides contain less than a whole chromosome and are preferably intron-free.
  • CIF150/hTAFjjl50 antisense oligonucleotides The complement of the nucleotide sequence shown in SEQ ID NO:l can be used provide CIF150/hTAFjjl50 antisense oligonucleotides.
  • CIF150/hTAFj l50 subgenomic polynucleotides also include polynucleotides which encode CIF 150/hT AF ⁇ 150-specific single-chain antibodies, ribozymes, and biologically active or altered OF150/hTAF ⁇ 150 variants.
  • nucleotide sequences encoding amino acid sequences of OF150/hTAF ⁇ 150 protein or biologically active CIF150/hTAF ⁇ 150 variants, as well as homologous nucleotide sequences which are at least 65%, 75%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to the nucleotide sequence shown in SEQ ID NO: 1, are also CIF150/hTAF jl50 subgenomic polynucleotides.
  • Percent sequence identity between the nucleotide sequence of SEQ ID NO:l and a putative homologous or degenerate CIFl 50/hTAFjjl 50 nucleotide sequence is determined using computer programs which employ the Smith- Waterman algorithm, for example as implemented in the MPSRCH program (Oxford Molecular), using an affine gap search with the following parameters: a gap open penalty of 12 and a gap extension penalty of 1.
  • Nucleotide sequences which hybridize to the coding sequence shown in SEQ ID NO: 1 or its complement with at most 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, or 35% basepair mismatches are also CIF150/hTAFjjl50 subgenomic polynucleotides of the invention.
  • homologous CIFl 50/hTAFjjl 50 sequences can be identified which contain at most about 25-30% basepair mismatches with SEQ ID NO:l or its complement. More preferably, homologous nucleic acid strands contain 15-25% basepair mismatches, even more preferably 5-15% basepair mismatches.
  • Species homologs of CIFl 50/hTAFj l 50 subgenomic polynucleotides of the invention can also be identified by making suitable probes or primers and screening cDNA expression libraries from other species, such as mice, monkeys, yeast, or bacteria, as well as human cDNA expression libraries. It is well known that the T m of a double- stranded DNA decreases by 1-1.5 °C with every 1% decrease in homology (Bonner et al, J. Mol. Biol. 81, 123 (1973).
  • Homologous CIF150/hTAFj ⁇ l50 human polynucleotides or CIFl 50/hTAFj ⁇ l 50 polynucleotides of other species can therefore be identified, for example, by hybridizing a putative homologous CIF150/hTAF jl50 polynucleotide with a polynucleotide having the nucleotide sequence of SEQ ID NO: 1 to form a test hybrids, comparing the melting temperature of the test hybrid with the melting temperature of a hybrid comprising a polynucleotide having SEQ ID NO:l and a polynucleotide which is perfectly complementary to SEQ ID NO: 1 , and calculating the number or percent of basepair mismatches within the test hybrid.
  • Nucleotide sequences which hybridize to the coding sequence shown in SEQ ID NO:l or its complement following stringent hybridization and/or wash conditions are also CIFl 50/hTAFjjl 50 subgenomic polynucleotides of the invention.
  • Stringent wash conditions are well known and understood in the art and are disclosed, for example, in Sambrook et al, MOLECULAR CLONING: A LABORATORY MANUAL, 2d ed., 1989, at pages 9.50-9.51.
  • T m of a hybrid between the CIF150/hTAFjjl50 sequence shown in SEQ ID NO:l and a polynucleotide sequence which is 65%, 75%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO:l can be calculated, for example, using the equation of Bolton and McCarthy, Proc. Natl. Acad. Sci. U.S.A. 48, 1390 (1962):
  • Stringent wash conditions include, for example, 4X SSC at 65 °C, or 50% formamide, 4X SSC at 42 °C, or 0.5X SSC, 0.1% SDS at 65 °C.
  • Highly stringent wash conditions include, for example, 0.2X SSC at 65 °C.
  • CIF150/hTAFjjl50 subgenomic polynucleotides can be isolated and purified free from other nucleotide sequences using standard nucleic acid purification techniques. For example, restriction enzymes and probes can be used to isolate polynucleotide fragments which comprise nucleotide sequences encoding a OF150/hTAF ⁇ 150 protein or variant. Isolated and purified subgenomic polynucleotides are in preparations which are free or at least 90% free of other molecules.
  • Complementary DNA (cDNA) molecules which encode CIF150/hTAF ⁇ 150 proteins are also CIFl 50/hTAFjjl 50 subgenomic polynucleotides of the invention.
  • CIF150/hTAFjjl50 cDNA molecules can be made with standard molecular biology techniques, using CIF150/hTAFjjl50 mRNA as a template.
  • CIF150/hTAFjjl50 cDNA molecules can thereafter be replicated using molecular biology techniques known in the art and disclosed in manuals such as Sambrook et al, 1989.
  • An amplification technique such as the polymerase chain reaction (PCR), can be used to obtain additional copies of subgenomic polynucleotides of the invention, using either human genomic DNA or cDNA as a template.
  • PCR polymerase chain reaction
  • CIFl 50/hTAFjjl 50 subgenomic polynucleotide molecules of the invention can be synthesized using synthetic chemistry techniques to synthesize CIFl 50/hTAFjjl 50 subgenomic polynucleotide molecules of the invention.
  • the degeneracy of the genetic code allows alternate nucleotide sequences to be synthesized which will encode a CIF150/hTAF ⁇ 150 protein having the amino acid sequence shown in SEQ ID NO:2 or a biologically active variant of that sequence. All such nucleotide sequences are within the scope of the present invention.
  • the invention also provides polynucleotide probes which can be used to detect CIF150/hTAFjjl50 sequences, for example, in hybridization protocols such as Northern or Southern blotting or in situ hybridization.
  • Polynucleotide probes of the invention comprise at least 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, or 40 or more contiguous nucleotides selected from SEQ ID NO: 1.
  • Polynucleotide probes of the invention can comprise a detectable label, such as a radioisotopic, fluorescent, enzymatic, or chemiluminescent label.
  • CIFl 50/hTAFjjl 50 subgenomic polynucleotides can be used as primers to obtain additional copies of the polynucleotides or as probes for identifying wild-type and mutant CIFl 50/hTAFjjl 50 coding sequences.
  • CIF150/hTAFjjl50 subgenomic polynucleotides can also be used to express CIF150/hTAFjjl50 mRNA, protein, polypeptides, fusion proteins and the like and to generate CIFl 50/hTAF jl 50 antisense oligonucleotides and ribozymes.
  • CIF150/hTAFj ⁇ l50 subgenomic polynucleotide comprising CIF150/hTAF ⁇ 150 coding sequences can be used in a construct, such as an RNA or DNA construct.
  • a CIF150/hTAFjjl50 construct can be an expression construct, which can be used to express all or a portion of a OF150/hTAF ⁇ 150 protein in a host cell.
  • Host cells comprising OF150/hTAF ⁇ 150 expression constructs can be prokaryotic or eukaryotic.
  • the CIFl 50/hTAF jl 50 subgenomic polynucleotide is inserted into an expression plasmid (for example, the Ecdyson system, pIND, In Vitro Gene).
  • an expression plasmid for example, the Ecdyson system, pIND, In Vitro Gene.
  • host cells are available for use in bacterial, yeast, insect, and human expression systems and can be used to express OF150/hTAF ⁇ 150 expression constructs (see below). Expression constructs can be introduced into host cells using any technique known in the art.
  • transferrin-polycation-mediated DNA transfer transfection with naked or encapsulated nucleic acids, liposome-mediated cellular fusion, intracellular transportation of DNA-coated latex beads, protoplast fusion, viral infection, electroporation, "gene gun,” and calcium phosphate-mediated transfection.
  • a OF150/hTAF ⁇ 150 expression construct comprises a promoter which is functional in the particular host cell selected. The skilled artisan can readily select an appropriate promoter from the large number of cell type-specific promoters known and used in the art.
  • the expression construct can also contain a transcription terminator which is functional in the host cell.
  • the expression construct comprises a polynucleotide segment which encodes all or a portion of a OF150/hTAF ⁇ 150 protein, biologically active variant, fusion protein, antibody, or ribozyme.
  • the polynucleotide segment is located downstream from the promoter. Transcription of the polynucleotide segment initiates at the promoter.
  • the expression construct can be linear or circular and can contain sequences, if desired, for autonomous replication.
  • CIFl 50/hTAFjjl 50 subgenomic polynucleotides can be propagated in vectors and cell lines using techniques well known in the art.
  • CIFl 50/hTAFjjl 50 subgenomic polynucleotides can be on linear or circular molecules. They can be on autonomously replicating molecules or on molecules without replication sequences. They can be regulated by their own or by other regulatory sequences, as are known in the art.
  • Expression systems in yeast include those described in Hinnen et al, Proc. Natl. Acad. Sci. USA (1978) 75: 1929; Ito et al, J. Bacteriol (1983) 755: 163; Kurtz et al, Mol. Cell. Biol. (1986) 6: 142; Kunze et al, J. Basic Microbiol. (1985) 25: 141; Gleeson et al, J. Gen. Microbiol. (1986) 132: 3459, Roggenkamp et al, Mol. Gen. Genet. (1986) 202 :302) Das et al, J. Bacteriol. (1984) 75S: 1165; De Louvencourt et al, J. Bacteriol.
  • CIFl 50/hTAFjjl 50 subgenomic polynucleotides in insects can be carried out as described in U.S. 4,745,051, Friesen et al (1986) "The Regulation of Baculovirus Gene Expression” in: THE MOLECULAR BIOLOGY OF BACULOVIRUSES (W. Doerfler, ed.), EP 127,839, EP 155,476, and Vlak et al, J. Gen. Virol. (1988) 69: 765- 776, Miller et al, Ann. Rev. Microbiol.
  • Mammalian expression of CIFl 50/hTAFjjl 50 subgenomic polynucleotides can be achieved as described in Dijkema et al, EMBO J. (1985) 4: 761, Gorman et al, Proc. Natl. Acad. Sci. USA (1982b) 79: 6777, Boshart et al, Cell (1985) 41: 521 and U.S. 4,399,216.
  • Other features of mammalian expression can be facilitated as described in Ham and Wallace, Meth. Enz. (1979) 58: 44; Barnes and Sato, Anal. Biochem. (1980) 102: 255, U.S.
  • Polynucleotides of the invention can also be used in gene delivery vehicles, for the purpose of delivering a CIF150/hTAFjjl50 mRNA or oligonucleotide (either with the sequence of native CIF150/hTAFjjl50 mRNA or its complement), full-length OF150/hTAF ⁇ 150 protein, OF150/hTAF ⁇ 150 fusion protein, CIF150/hTAF ⁇ 150 polypeptide, or CIF 150/hTAF ⁇ l 50-specific ribozyme or single-chain antibody, into a cell preferably a eukaryotic cell.
  • a gene delivery vehicle can be, for example, naked plasmid DNA, a viral expression vector comprising a CIF150/hTAFjjl50 polynucleotide, or a OF150/hTAF ⁇ 150 polynucleotide in conjunction with a liposome or a condensing agent.
  • the gene delivery vehicle comprises a promoter and a CIF150/hTAFj l50 polynucleotide.
  • Preferred promoters are tissue- specific promoters and promoters which are activated by cellular proliferation, such as the thymidine kinase and thymidylate synthase promoters.
  • promoters which are activatable by infection with a virus such as the ⁇ - and ⁇ - interferon promoters, and promoters which are activatable by a hormone, such as esfrogen.
  • promoters which can be used include the Moloney virus LTR, the CMV promoter, and the mouse albumin promoter.
  • a CIF150/hTAF l50 gene delivery vehicle can comprise viral sequences such as a viral origin of replication or packaging signal. These viral sequences can be selected from viruses such as astrovirus, coronavirus, orthomyxovirus, papovavirus, paramyxovirus, parvovirus, picornavirus, poxvirus, retrovirus, togavirus or adenovirus.
  • the CIF150/hTAF l50 gene delivery vehicle is a recombinant retroviral vector. Recombinant refroviruses and various uses thereof have been described in numerous references including, for example, Mann et al, Cell 55. 153, 1983, Cane and Mulligan, Proc. Natl Acad. Sci.
  • retroviruses are derived from retroviruses which include avian leukosis virus (ATCC Nos. VR-535 and VR-247), bovine leukemia virus (NR- 1315), murine leukemia virus (MLN), mink-cell focus-inducing virus (Koch et al, J. Vir. 49:828, 1984; and Olif et al, J. Vir. 48:542, 1983), murine sarcoma virus (ATCC ⁇ os.
  • retroviruses which include avian leukosis virus (ATCC Nos. VR-535 and VR-247), bovine leukemia virus (NR- 1315), murine leukemia virus (MLN), mink-cell focus-inducing virus (Koch et al, J. Vir. 49:828, 1984; and Olif et al, J. Vir. 48:542, 1983), murine sarcoma virus (ATCC ⁇ os.
  • NR-844, 45010 and 45016 reticuloendotheliosis virus (ATCC ⁇ os VR-994, VR-770 and 45011), Rous sarcoma virus, Mason-Pfizer monkey virus, baboon endogenous virus, endogenous feline retrovirus (e.g., RD114), and mouse or rat gL30 sequences used as a retroviral vector.
  • Particularly preferred strains of MLV from which recombinant retroviruses can be generated include 4070A and 1504A (Hartley and Rowe, J Vir. 19: 19, 1976), Abelson (ATCC No. VR-999), Friend (ATCC No. VR-245), Graffi (Ru et al, J. Vir.
  • Rous sarcoma virus A particularly preferred non-mouse retrovirus is Rous sarcoma virus.
  • Preferred Rous sarcoma viruses include Bratislava (Manly et al, J. Vir.
  • retroviral CIFl 50/hTAFjjl 50 gene delivery vehicles can be readily utilized in order to assemble or construct retroviral CIFl 50/hTAFjjl 50 gene delivery vehicles given the disclosure provided herein and standard recombinant techniques (e.g., Sambrook et al, MOLECULAR CLONING: A LABORATORY MANUAL, 2d ed., 1989, and Kunkle, Proc. Natl. Acad. Sci. U.S.A. £2:488, 1985) known in the art. Portions of retroviral CIF150/hTAFjjl50 expression vectors can be derived from different retroviruses.
  • retrovector LTRs can be derived from a murine sarcoma virus, a tRNA binding site from a Rous sarcoma virus, a packaging signal from a murine leukemia virus, and an origin of second strand synthesis from an avian leukosis virus.
  • retroviral vectors can be used to generate fransduction competent retroviral vector particles by introducing them into appropriate packaging cell lines (see Serial No. 07/800,921, filed November 29, 1991).
  • Recombinant retroviruses can be produced which direct the site-specific integration of the recombinant retroviral genome into specific regions of the host cell DNA.
  • Such site-specific integration can be mediated by a chimeric integrase incorporated into the retroviral particle (see Serial No. 08/445,466 filed May 22, 1995).
  • the recombinant viral gene delivery vehicle is a replication-defective recombinant virus.
  • Packaging cell lines suitable for use with the above-described retroviral gene delivery vehicles can be readily prepared (see Serial No. 08/240,030, filed May 9, 1994; see also WO 92/05266) and used to create producer cell lines (also termed vector cell lines or "VCLs”) for production of recombinant viral particles.
  • packaging cell lines are made from human (e.g., HT1080 cells) or mink parent cell lines, thereby allowing production of recombinant retroviral gene delivery vehicles which are capable of surviving inactivation in human serum.
  • human e.g., HT1080 cells
  • mink parent cell lines thereby allowing production of recombinant retroviral gene delivery vehicles which are capable of surviving inactivation in human serum.
  • the construction of recombinant retroviral gene delivery vehicles is described in detail in WO 91/02805.
  • These recombinant retroviral gene delivery vehicles can be used to generate fransduction competent retroviral particles by introducing them into appropriate packaging cell lines (see Serial No. 07/800,921).
  • adenovirus gene delivery vehicles can also be readily prepared and utilized given the disclosure provided herein (see also Berkner, Biotechniques 6:616-621, 1988, and Rosenfeld et al, Science 252:431-434, 1991, WO 93/07283, WO 93/06223, and WO 93/07282).
  • a CIF150/hTAFjjl50 gene delivery vehicle can also be a recombinant adenoviral gene delivery vehicle.
  • Such vehicles can be readily prepared and utilized given the disclosure provided herein (see Berkner, Biotechniques 6:616, 1988, and Rosenfeld et al, Science 252:431, 1991, WO 93/07283, WO 93/06223, and WO 93/07282).
  • Adeno-associated viral CIFl 50/hTAFj l 50 gene delivery vehicles can also be constructed and used to deliver CIF150/hTAF ⁇ 150 amino acids or nucleotides.
  • the use of adeno-associated viral gene delivery vehicles in vitro is described in Chatteriee et al, Science 258: 1485-1488 (1992), Walsh et al, Proc. Natl Acad. Sci. 89: 7257-7261 (1992), Walsh et al, J. Clin. Invest. 94: 1440-1448 (1994), Flotte et al, J. Biol. Chem. 268: 3781-3790 (1993), Ponnazhagan et al, J. Exp. Med.
  • a CIFl 50/hTAFjjl 50 gene delivery vehicle is derived from a togavirus.
  • Preferred togaviruses include alphaviruses, in particular those described in U.S. Serial No. 08/405,627, filed March 15, 1995, WO 95/07994.
  • Alpha viruses, including Sindbis and ELVS viruses can be gene delivery vehicles for CIFl 50/hTAFjjl 50 polynucleotides.
  • Alpha viruses are described in WO 94/21792, WO 92/10578 and WO 95/07994.
  • alphavirus gene delivery vehicle systems can be constructed and used to deliver CIF150/hTAFjjl50 polynucleotides to a cell according to the present invention.
  • alphavirus gene delivery vehicles for use in the present invention include those which are described in WO 95/07994, and U.S. Serial No. 08/405,627.
  • the recombinant viral vehicle is a recombinant alphavirus viral vehicle based on a Sindbis virus.
  • Sindbis constructs as well as numerous similar constructs, can be readily prepared essentially as described in U.S. Serial No. 08/198,450.
  • Sindbis viral gene delivery vehicles typically comprise a 5' sequence capable of initiating Sindbis virus transcription, a nucleotide sequence encoding Sindbis non-structural proteins, a viral junction region inactivated so as to prevent fragment transcription, and a Sindbis RNA polymerase recognition sequence.
  • the viral junction region can be modified so that polynucleotide transcription is reduced, increased, or maintained.
  • corresponding regions from other alphaviruses can be used in place of those described above.
  • the viral junction region of an alphavirus-derived gene delivery vehicle can comprise a first viral junction region which has been inactivated in order to prevent transcription of the polynucleotide and a second viral junction region which has been modified such that polynucleotide transcription is reduced.
  • An alphavirus-derived vehicle can also include a 5' promoter capable of initiating synthesis of viral RNA from cDNA and a 3' sequence which controls transcription termination.
  • recombinant togaviral gene delivery vehicles which can be utilized in the present invention include those derived from Semliki Forest virus (ATCC VR-67; ATCC VR-1247), Middleberg virus (ATCC VR-370), Ross River virus (ATCC VR-373; ATCC VR-1246), Venezuelan equine encephalitis virus (ATCC VR923; ATCC VR-1250; ATCC VR-1249; ATCC VR-532), and those described in U.S. Patents 5,091,309 and 5,217,879 and in WO 92/10578.
  • the Sindbis vehicles described above, as well as numerous similar constructs, can be readily prepared essentially as described in U.S. Serial No. 08/198,450.
  • viral gene delivery vehicles suitable for use in the present invention include, for example, those derived from poliovirus (Evans et al, Nature 559:385, 1989, and Sabin et al, J. Biol. Standardization 7:115, 1973) (ATCC VR-58); rhino virus (Arnold et al, J. Cell. Biochem. L401, 1990) (ATCC VR-1110); pox viruses, such as canary pox virus or vaccinia virus (Fisher-Hoch et al, Proc. Natl. Acad. Sci. U.S.A. 86:317, 1989; Flexne ⁇ et al, Ann. NY. Acad. Sci. 559:86, 1989; Flexner et al.
  • Vaccine 8 11, 1990; U.S. 4,603,112 and U.S. 4,769,330; WO 89/01973) (ATCC VR-111; ATCC VR-2010); SV40 (Mulligan et al, Nature 277. 108, 1979) (ATCC VR-305), (Madzak et al, J. Gen. Vir.
  • influenza virus (Luytjes et al, Cell 59:1107, 1989; McMicheal et al, The New England Journal of Medicine 309:13, 1983; and Yap et al, Nature 275:238, 1978) (ATCC VR-797); parvovirus such as adeno-associated virus (Samulski et ⁇ /., J Vir. 55:3822, 1989, and Mendelson et ⁇ /., Virology 166:154, 1988) (ATCC VR-645); herpes simplex virus (Kit et al. , Adv. Exp. Med. Biol.
  • a CIFl 50/hTAFjjl 50 polynucleotide of the invention can also be combined with a condensing agent to form a gene delivery vehicle.
  • the condensing agent is a polycation, such as polylysine, polyarginine, polyornithine, protamine, spermine, spermidine, and putrescine. Many suitable methods for making such linkages are known in the art (see, for example, Serial No. 08/366,787, filed December 30, 1994).
  • a CIFl 50/hTAFjjl 50 polynucleotide is associated with a liposome to form a gene delivery vehicle.
  • Liposomes are small, lipid vesicles comprised of an aqueous compartment enclosed by a lipid bilayer, typically spherical or slightly elongated structures several hundred Angstroms in diameter. Under appropriate conditions, a liposome can fuse with the plasma membrane of a cell or with the membrane of an endocytic vesicle within a cell which has internalized the liposome, thereby releasing its contents into the cytoplasm. Prior to interaction with the surface of a cell, however, the liposome membrane acts as a relatively impermeable barrier which sequesters and protects its contents, for example, from degradative enzymes. Additionally, because a liposome is a synthetic structure, specially designed liposomes can be produced which incorporate desirable features.
  • Liposomes can encapsulate a variety of nucleic acid molecules including DNA, RNA, plasmids, and expression constructs comprising CIFl 50/hTAFjjl 50 polynucleotides such those disclosed in the present invention.
  • Liposomal preparations for use in the present invention include cationic (positively charged), anionic (negatively charged) and dural preparations.
  • Cationic liposomes have been shown to mediate intracellular delivery of plasmid DNA (Feigner et al, Proc. Nat Acad. Sci. USA 84:1413-1416, 1987), mRNA (Malone et al, Proc. Natl. Acad. Sci. USA 86:6011-6081, 1989), and purified transcription factors (Debs et al, J. Biol. Chem. 255:10189-10192, 1990), in functional form.
  • Cationic liposomes are readily available. For example, N[l-2,3-dioleyloxy)propyl]-N,N,N-triethylammonium
  • DOTMA liposomes are available under the trademark Lipofectin, from GIBCO BRL, Grand Island, NY. See also Feigner et al, Proc. Natl. Acad. Sci. USA 91: 5148-5152.87, 1994.
  • Other commercially available liposomes include Transfectace (DDAB/DOPE) and DOTAP/DOPE (Boerhinger).
  • DDAB/DOPE Transfectace
  • DOTAP/DOPE Boerhinger
  • Other cationic liposomes can be prepared from readily available materials using techniques well known in the art. See, e.g. , Szoka et al. , Proc. Natl Acad. Sci.
  • DOTAP l,2-bis(oleoyloxy)-3-(frimethylammonio)propane liposomes.
  • anionic and neutral liposomes are readily available, such as from Avanti Polar Lipids (Birmingham, AL), or can be easily prepared using readily available materials.
  • Such materials include phosphatidyl choline, cholesterol, phosphatidyl ethanolamine, dioleoylphosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol (DOPG), dioleoylphoshatidyl ethanolamine (DOPE), among others.
  • DOPC dioleoylphosphatidyl glycerol
  • DOPE dioleoylphoshatidyl ethanolamine
  • the liposomes can comprise multilammelar vesicles (MLVs), small unilamellar vesicles (SUVs), or large unilamellar vesicles (LUVs).
  • MLVs multilammelar vesicles
  • SUVs small unilamellar vesicles
  • LUVs large unilamellar vesicles
  • the various liposome-nucleic acid complexes are prepared using methods known in the art. See, e.g., Straubinger et al, METHODS OF IMMUNOLOGY (1983), Vol. 101, pp. 512-527; Szoka et al, Proc. Natl. Acad. Sci. USA 57:3410-3414, 1990; Papahadjopoulos et al, Biochim. Biophys.
  • lipoproteins can be included with a CIFl 50/hTAFjjl 50 polynucleotide for delivery to a cell.
  • lipoproteins include chylomicrons, HDL, IDL, LDL, and NLDL. Mutants, fragments, or fusions of these proteins can also be used. Modifications of naturally occurring lipoproteins can also be used, such as acetylated LDL. These lipoproteins can target the delivery of polynucleotides to cells expressing lipoprotein receptors.
  • no other targeting ligand is included in the composition.
  • naked CIF150/hTAFj l50 polynucleotide molecules are used as gene delivery vehicles, as described in WO 90/11092 and U.S. Patent 5,580,859.
  • Such gene delivery vehicles can be either CIFl 50/hTAFj l 50 D ⁇ A or R ⁇ A and, in certain embodiments, are linked to killed adenovirus. Curiel et al, Hum. Gene. Ther. 5:147-154, 1992.
  • Other suitable vehicles include D ⁇ A-ligand (Wu et al, J. Biol. Chem. 264:16985-16981, 1989), lipid-D ⁇ A combinations (Feigner et al, Proc. Natl. Acad. Sci.
  • This approach takes advantage of the observation that latex beads, when incubated with cells in culture, are efficiently transported and concentrated in the perinuclear region of the cells. The beads will then be transported into cells when injected into muscle.
  • CIFl 50/hTAF jl 50 polynucleotide-coated latex beads will be efficiently transported into cells after endocytosis is initiated by the latex beads and thus increase gene transfer and expression efficiency.
  • This method can be improved further by freating the beads to increase their hydrophobicity, thereby facilitating the disruption of the endosome and release of CIF150/hTAFjjl50 polynucleotides into the cytoplasm.
  • OF150/hTAF ⁇ 150 protein and activity are cell cycle regulated.
  • the invention provides compositions which can be used to alter spatial or temporal patterns of division of a human cell. Depletion of OF150/hTAF ⁇ 150 activity leads to a G 2 (late S phase) arrest of the cell cycle, indicating that OF150/hTAF ⁇ 150 function is required to transcribe genes necessary for cell cycle progression. Thus, mitosis or cell cycle progression can be reduced or prevented by decreasing expression of a human CIFl 50/hTAF jl 50 gene.
  • Decreased CIFl 50/hTAFjjl 50 gene expression can be used to freat conditions characterized by high rates of mitosis, such as neoplasia, metastasis of neoplasms, benign proliferative diseases, and dysplastic and hyperplastic disorders.
  • Cells in which CIF150/hTAFjjl50 expression has been decreased can also be used to identify genes whose expression is dependent on a OF150/hTAF ⁇ 150 protein.
  • increased CIF150/hTAFjjl50 expression can be used to expand cell populations in vitro or for freating disorders such as anemia, which are characterized by lowered rates of mitosis.
  • expression of the CIF150/hTAFj l50 gene is decreased using a ribozyme, an RNA molecule with catalytic activity.
  • a ribozyme an RNA molecule with catalytic activity.
  • Ribozymes can be used to inhibit gene function by cleaving an RNA sequence, as is known in the art (e.g., Haseloff et al, U.S. 5,641,673).
  • the coding sequence of the CIF150/hTAF jl50 gene can be used to generate ribozymes which will specifically bind to mRNA franscribed from a CIF150/hTAFjjl50 gene.
  • Methods of designing and constructing ribozymes which can cleave other RNA molecules in trans in a highly sequence specific manner have been developed and described in the art (see Haseloff et al, Nature 334:585-591, 1988).
  • the cleavage activity of ribozymes can be targeted to specific CIF150/hTAF jl50 RNAs by engineering a discrete "hybridization" region into the ribozyme.
  • the hybridization region contains a sequence complementary to the target CIF150/hTAF jl50 RNA and thus specifically hybridizes with the target (see, for example, Gerlach et al, EP 321,201).
  • the nucleotide sequence shown in SEQ ID NO:l provides a source of suitable hybridization region sequences. Longer complementary sequences can be used to increase the affinity of the hybridization sequence for the target.
  • the hybridizing and cleavage regions of the CIF150/hTAF jl50 ribozyme can be integrally related; thus, upon hybridizing to the target CIFl 50/hTAFjjl 50 RNA through the complementary regions, the catalytic region of the ribozyme can cleave the target.
  • CIF150/hTAFjjl50 ribozymes can be introduced into cells, such as neoplastic cells, as part of a DNA construct, as is known in the art and described above.
  • Mechanical methods such as microinjection, liposome-mediated transfection, elecfroporation, or calcium phosphate precipitation, can be used to introduce the ribozyme-containing DNA construct into cells in which it is desired to decrease CIF150/hTAF jl50 expression.
  • the cells stably retain the DNA construct it can be supplied on a plasmid and maintained as a separate element or integrated into the genome of the cells, as is known in the art.
  • the DNA construct can include transcriptional regulatory elements, such as a promoter element, an enhancer or UAS element, and a transcriptional terminator signal, for controlling transcription of CIF150/hTAFj l50 ribozymes in the cells.
  • transcriptional regulatory elements such as a promoter element, an enhancer or UAS element, and a transcriptional terminator signal, for controlling transcription of CIF150/hTAFj l50 ribozymes in the cells.
  • CIF150/hTAFjjl50 ribozymes can be engineered so that ribozyme expression will occur in response to factors which induce expression of the CIF150/hTAF jl50 gene.
  • Ribozymes can also be engineered to provide an additional level of regulation, so that destruction of CIFl 50/hTAFjjl 50 mRNA occurs only when both a CIF150/hTAFjjl50 ribozyme and a CIF150/hTAFj ⁇ l50 gene are induced in the cells.
  • expression of the CIF150/hTAFjjl50 gene is altered using an antisense oligonucleotide sequence.
  • the antisense sequence is complementary to at least a portion of the coding sequence of a CIF150/hTAFjjl50 gene having the nucleotide sequence shown in SEQ ID NO: 1.
  • the antisense oligonucleotide sequence is at least six nucleotides in length, but can be about 8, 12, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides long. Longer sequences can also be used.
  • CIF150/hTAFjjl50 antisense oligonucleotide molecules can be provided in a DNA construct and introduced into cells as described above, to decrease CIF150/hTAFjjl50 expression.
  • Example 1 The treated cells do not enter mitosis and accumulate in G 2 phase.
  • One of skill in the art can test other oligonucleotides for antisense effect, by delivering the antisense oligonucleotide to the cell using transfection or any means known in the art and detecting CIF150/hTAF jl50 mRNA levels.
  • CIF150/hTAFjjl50 antisense oligonucleotides can be deoxyribonucleotides, ribonucleotides, or a combination of both.
  • Oligonucleotides can be synthesized manually or by an automated synthesizer, by covalently linking the 5' end of one nucleotide with the 3' end of another nucleotide with non-phosphodiester internucleotide linkages such alkylphosphonates, phosphorothioates, phosphorodithioates, alkylphosphonothioates, alkylphosphonates, phosphoramidates, phosphate esters, carbamates, acetamidate, carboxymethyl esters, carbonates, and phosphate triesters. See Brown, 1994, Meth. Mol. Biol 20:1-8; Sonveaux, 1994, Meth. Mol. Biol.
  • Antisense molecules which comprise, for example, 2, 3, 4, or 5 or more stretches of contiguous nucleotides which are precisely complementary to a CIF150/hTAF jl50 coding sequence, each separated by a stretch of contiguous nucleotides which are not complementary to adjacent CIF150/hTAFj/150 coding sequences, can provide targeting specificity for CIF150/hTAFjjl50 mRNA.
  • each stretch of contiguous nucleotides is at least 4, 5, 6, 7, or 8 or more nucleotides in length.
  • Non-complementary intervening sequences are preferably 1, 2, 3, or 4 nucleotides in length.
  • One skilled in the art can easily use the calculated melting point of an antisense-sense pair to determine the degree of mismatching which will be tolerated between a particular antisense oligonucleotide and a particular CIFl 30 coding sequence.
  • CIFl 50/hTAFjjl 50 antisense oligonucleotides can be modified without affecting their ability to hybridize to a CIF150/hTAF jl50 coding sequence. These modifications can be internal or at one or both ends of the antisense molecule.
  • intemucleoside phosphate linkages can be modified by adding cholesteryl or diamine moieties with varying numbers of carbon residues between the amino groups and terminal ribose.
  • Modified bases and/or sugars such as arabinose instead of ribose, or a 3', 5'-substituted oligonucleotide in which the 3' hydroxyl group or the 5' phosphate group are substituted, can also be employed in a modified antisense oligonucleotide.
  • modified oligonucleotides can be prepared by methods well known in the art. See, e.g., Agrawal et ⁇ /., 1992, Trends Biotechnol 70:152-158; Uhlmann et al, 1990, Chem. Rev. 90:543-584; Uhlmann et al, 1987, Tetrahedron. Lett. 275:3539-3542.
  • Antibodies which specifically bind to a OF150/hTAF ⁇ 150 protein can also be used to alter CIF150/hTAFjjl50 gene expression.
  • CIF150/hTAF ⁇ 150-specific antibodies bind to OF150/hTAF ⁇ 150 protein and prevent the protein from functioning in the cell.
  • Polynucleotides encoding antibodies which specifically bind to OF150/hTAF ⁇ 150 can be introduced into cells as described above.
  • the mechanism used to decrease expression of the CIF150/hTAFjjl50 gene whether ribozyme, antisense nucleotide sequence, or antibody, decreases expression of the CIF150/hTAF jl50 gene by 50%, 60%, 70%, or 80%.
  • expression of the CIF150/hTAFjjl50 gene is decreased by 90%, 95%, 99%, or 100%.
  • the effectiveness of the mechanism chosen to alter expression of the CIF150/hTAFjjl50 gene can be assessed using methods well known in the art, such as hybridization of nucleotide probes to CIF150/hTAFjjl50 mRNA, quantitative RT-PCR, or detection of OF150/hTAF ⁇ 150 protein using CIF150/hTAF ⁇ 150-specific antibodies.
  • compositions comprising OF150/hTAF ⁇ 150 antibodies, ribozymes, or antisense oligonucleotides can be used to treat proliferative disorders, such as neoplasias, dysplasias, and hyperplasias, and their symptoms.
  • Neoplasias which can be treated with a OF150/hTAF ⁇ 150 composition include, but are not limited to, melanomas, squamous cell carcinomas, adenocarcinomas, hepatocellular carcinomas, renal cell carcinomas, sarcomas, myosarcomas, non-small cell lung carcinomas, leukemias, lymphomas, osteosarcomas, central nervous system tumors such as gliomas, astrocytomas, oligodendrogliomas, and neuroblastomas, tumors of mixed origin, such as Wilms* tumor and teratocarcinomas, and metastatic tumors.
  • Proliferative disorders which can be treated with a OF150/hTAF ⁇ 150 composition include disorders such as anhydric hereditary ectodermal dysplasia, congenital alveolar dysplasia, epithelial dysplasia of the cervix, fibrous dysplasia of bone, and mammary dysplasia.
  • disorders such as anhydric hereditary ectodermal dysplasia, congenital alveolar dysplasia, epithelial dysplasia of the cervix, fibrous dysplasia of bone, and mammary dysplasia.
  • Hyperplasias for example, endometrial, adrenal, breast, prostate, or thyroid hyperplasias, or pseudoepitheliomatous hyperplasia of the skin can be treated with CIFl 50/hTAF ⁇ l 50 compositions.
  • decreasing OF150/hTAF ⁇ 150 function can have therapeutic application.
  • decreasing CIF150/hTAF jl50 expression or function can help to suppress tumors.
  • effecting CIF150/hTAF ⁇ l50 downregulation or decrease of OF150/hTAF ⁇ 150 activity can suppress metastases.
  • OF150/hTAF ⁇ 150 compositions of the invention can comprise a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers are well known to those in the art. Such carriers include, but are not limited to, large, slowly metabolized macromolecules, such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, and inactive virus particles.
  • Pharmaceutically acceptable salts can also be used in OF150/hTAF ⁇ 150 compositions, for example, mineral salts such as hydrochlorides, hydrobromides, phosphates, or sulfates, as well as salts of organic acids such as acetates, proprionates, malonates, or benzoates.
  • OF150/hTAF ⁇ 150 compositions can also contain liquids, such as water, saline, glycerol, and ethanol, as well as substances such as wetting agents, emulsifying agents, or pH buffering agents.
  • liquids such as water, saline, glycerol, and ethanol, as well as substances such as wetting agents, emulsifying agents, or pH buffering agents.
  • Liposomes such as those described in U.S. 5,422,120, WO 95/13796, WO 91/14445, or EP 524,968 Bl, can also be used as a carrier for a CIF 150/hTAF ⁇ l 50 composition.
  • a OF150/hTAF ⁇ 150 composition is prepared as an injectable, either as a liquid solution or suspension; however, solid forms suitable for solution or suspension in liquid vehicles prior to injection can also be prepared.
  • a CIF150/hTAF ⁇ 150 composition can also be formulated into an enteric coated tablet or gel capsule according to known methods in the art, such as those described in U.S. 4,853,230, EP 225, 189, AU 9,224,296, and AU 9,230,801.
  • OF150/hTAF ⁇ 150 compositions of the invention can include local or systemic administration, including injection, oral administration, particle gun, or catheterized administration, and topical administration.
  • Various methods can be used to administer a CIF 150/hTAF ⁇ l 50 composition directly to a specific site in the body. For example, a small metastatic lesion can be located and an appropriate OF150/hTAF ⁇ 150 composition injected several times in several different locations within the body of the lesion.
  • arteries which serve a tumor can be identified, and a CIF150/hTAF ⁇ 150 composition can be injected into such an artery, in order to deliver the composition directly into the tumor.
  • a tumor which has a necrotic center can be aspirated, and a OF150/hTAF ⁇ 150 composition can be injected directly into the now empty center of the tumor.
  • a CIFl 50/hTAF ⁇ l 50 composition can also be administered directly to the surface of a tumor, for example, by topical application of the composition.
  • X-ray imaging can be used to assist in certain of these delivery methods.
  • Combination therapeutic agents including reagents which specifically bind to a wild-type human CIFl 50/hTAFjf 150 gene or expression product, together with other therapeutic agents, can be administered simultaneously or sequentially.
  • OF150/hTAF ⁇ 150 compositions can be delivered to specific tissues using receptor-mediated targeted delivery.
  • Receptor-mediated DNA delivery techniques are described in, for example, Findeis et al. (1993), Trends in Biotechnol 11, 202-05; Chiou et al. (1994), GENE THERAPEUTICS: METHODS AND APPLICATIONS OF DIRECT GENE TRANSFER (J.A. Wolff, ed.); Wu & Wu (1988), J. Biol. Chem. 263, 621-24; Wu et al.
  • a OF150/hTAF ⁇ 150 composition can be introduced into human cells ex vivo, and the cells then replaced into the human.
  • Cells can be removed from a variety of locations including, for example, from a selected tumor or from an affected organ.
  • a OF150/hTAF ⁇ 150 composition can be inserted into non-tumorigenic cells, such as dermal fibroblasts or peripheral blood leukocytes. If desired, particular fractions of cells, such as T cell subsets or stem cells, can also be specifically removed from the blood (see, for example, PCT WO 91/16116). The removed cells can then be contacted with a OF150/hTAF ⁇ 150 composition utilizing any of the above-described techniques, followed by the return of the cells to the human.
  • mitosis or cell cycle progression can be induced, increased, or promoted by increasing expression of a CIF150/hTAFjjl50 gene.
  • Increasing CIFl 50/hTAFjjl 50 gene expression is useful, for example, for increasing the number of cells in a cell population in vitro or for freating disorders characterized by lowered rates of mitosis or cell cycle progression, such as anemia.
  • a composition comprising all or a portion of a CIF150/hTAFjjl50 gene or expression product can be introduced into a cell.
  • the entire CIFl 50/hTAF jl 50 coding sequence or protein can be introduced, as described above.
  • a portion of a CIF150/hTAF jl50 protein which promotes mitosis or cell cycle progression can be identified and that portion or a nucleotide sequence encoding it can be introduced into the cell.
  • Portions of a OF150/hTAF ⁇ 150 protein which promote mitosis or cell cycle progression can be identified by introducing expression constructs which express different portions of the protein into cells and measuring alterations in the rate of mitosis or cell cycle progression. Rates of mitosis can be measured, for example, by detecting incorporation of labeled nucleotides, as is known in the art.
  • CIF150/hTAF ⁇ 150 compositions which contain CIF150/hTAFjjl50 subgenomic polynucleotides preferably contain an expression construct comprising a promoter and a polynucleotide segment encoding at least six contiguous amino acids of a
  • OF150/hTAF ⁇ 150 protein or protein variant within the expression construct, the polynucleotide segment is located downstream from the promoter, and transcription of the polynucleotide segment initiates at the promoter.
  • gene transfer vectors especially retroviral vectors, is contained in U.S. Serial No. 08/869,309, which is incorporated herein by reference.
  • Both the dose of a particular OF150/hTAF ⁇ 150 composition and the means of administering the composition can be determined based on specific qualities of the therapeutic composition, the condition, age, and weight of the patient, the progression of the particular disease being treated, and other relevant factors. If the composition contains CIF 150/hTAF ⁇ l 50 proteins, polypeptides, or antibodies, effective dosages of the composition are in the range of about 5 ⁇ g to about 50 ⁇ g/kg of patient body weight, about 50 ⁇ g to about 5 mg/kg, about 100 ⁇ g to about 500 ⁇ g/kg of patient body weight, and about 200 to about 250 ⁇ g/kg.
  • compositions containing CIF150/hTAFjjl50 subgenomic polynucleotides can be administered in a range of about 100 ng to about 200 mg of DNA for local administration. Suitable concentrations range from about 500 ng to about 50 mg, about 1 ⁇ g to about 2 mg, about 5 ⁇ g to about 500 ⁇ g, and about 20 ⁇ g to about 100 ⁇ g of DNA.
  • Factors such as method of action and efficacy of transformation and expression are considerations which will affect the dosage required for ultimate efficacy of the OF150/hTAF ⁇ 150 composition.
  • OF150/hTAF ⁇ 150 compositions or the same amount administered successively, or several administrations to different adjacent or close tissue portions of, for example, a tumor site may be required to effect a positive therapeutic outcome.
  • routine experimentation in clinical trials will determine specific ranges for optimal therapeutic effect.
  • Expression of an endogenous CIF150/hTAFjjl50 gene in a cell can be altered by introducing in frame with the endogenous CIF150/hTAFjjl50 gene a DNA construct comprising a CIF150/hTAFjjl50 targeting sequence, a regulatory sequence, an exon, and an unpaired splice donor site by homologous recombination, such that a homologously recombinant cell comprising a new OF150/hTAF ⁇ 150 franscription unit.
  • the new transcription unit can be used to turn the CIF150/hTAF jl50 gene on or off as desired. This method of affecting endogenous gene expression is taught in U.S. Patent No. 5,641,670.
  • the targeting sequence is a segment of at least 10, 12, 15, 20, or 50 contiguous nucleotides selected from the nucleotide sequence shown in SEQ ID NO: 1.
  • the franscription unit is located upstream of a coding sequence of the endogenous CIF150/hTAFjjl50 gene.
  • the exogenous regulatory sequence directs transcription of the coding sequence of the CIF150/hTAFjjl50 gene.
  • OF150/hTAF ⁇ 150 and its binding element 5'-Py X G A G A/C A/Py -3' can be used to increase expression of a gene, either in vitro, such as in a cell culture, or in vivo.
  • the OF150/hTAF ⁇ 150 binding element can be inserted into the promoter region of a gene whose increased expression is desired.
  • the promoter region is preferably a TATA-less promoter region. Any gene of interest can be placed under control of a TATA-less promoter region comprising a OF150/hTAF ⁇ 150 binding element using standard recombinant DNA techniques.
  • the promoter region of the gene is contacted with a OF150/hTAF ⁇ 150 protein.
  • the OF150/hTAF ⁇ 150 protein can be provided to the promoter region, for example, by means of an expression construct which encodes OF150/hTAF ⁇ 150.
  • expression of the desired gene is increased.
  • the OF150/hTAF ⁇ 150 coding sequence can be placed under control of a cell-type specific or inducible promoter, as is known in the art.
  • the present invention also provides a method of diagnosing or prognosing neoplasia or of identifying neoplastic tissue of a human.
  • Expression of a human CIFl 50/hTAF jl 50 gene can be compared between a first tissue which is suspected of being neoplastic and a second tissue which is normal.
  • the first and second tissues can be obtained from the same human or from different humans.
  • the normal tissue can be any tissue of the human, including, but not limited to, spleen, thymus, prostate, testis, ovary, small intestine, mucosal lining of the colon, peripheral blood leukocytes, heart, glial cells, placenta, lung, liver, skeletal muscle, kidney, pancreas, peripheral blood leukocytes, bone marrow, and appendix.
  • the tissue suspected of being neoplastic can be derived from a different tissue type of the human, but preferably it is derived from the same tissue type, for example an intestinal polyp or other abnormal growth.
  • Overexpression of the CIF150/hTAFjjl50 gene in the suspect tissue identifies the suspect tissue as neoplastic.
  • Expression of a CIF150/hTAFjjl50 gene can be detected by measuring CIF150/hTAF jl50 mRNA.
  • PolyA + RNA can be isolated from the two tissues as is known in the art.
  • One of skill in the art can readily determine differences in the size or amount of CIFl 50/hTAF l 50 mRNA transcripts between the two tissues that are compared, using Northern blots or in situ hybridization with nucleotide probes selected from the nucleotide sequence shown in SEQ ID NO: 1.
  • CIF150/hTAF ⁇ 150 proteins can be compared between the two tissue samples. Any method for analyzing proteins can be used to compare two OF150/hTAF ⁇ 150 proteins from matched samples. The sizes of the OF150/hTAF ⁇ 150 proteins in the two tissues can be compared, for example, using antibodies to detect OF150/hTAF ⁇ 150 proteins in Western blots of protein extracts from the two tissues. Other changes, such as expression levels and subcellular localization, can also be detected immunologically. A higher OF150/hTAF ⁇ 150 protein expression level in a tissue suspected of being neoplastic compared with the CIF 150/hTAF ⁇ 150 protein expression level in a normal tissue is indicative of neoplasia.
  • CIFl 50/hTAFjjl 50 gene sequences or of CIFl 50/hTAFjf 150 gene expression products, e.g., mRNA and protein, between a tissue of a human which is suspected of being neoplastic and a normal tissue of a human can be used to diagnose or prognose neoplasia in the human.
  • the CIFl 50/hTAFjjl 50 genes in the two tissues can be compared by any means known in the art.
  • the two genes can be sequenced, and the sequence of the CIFl 50/hTAFjjl 50 gene in the tissue suspected of being neoplastic can be compared with the wild-type CIFl 50/hTAFjjl 50 sequence in the normal tissue.
  • the CIFl 50/hTAF l 50 genes or portions of the C7E750/hTAFj l 50 genes in the two tissues can be amplified, for example, using nucleotide primers selected from the nucleotide sequence shown in SEQ ID NO:l in the polymerase chain reaction (PCR) or other amplification technique.
  • the amplified genes or portions of genes can be hybridized to nucleotide probes selected from the nucleotide sequence shown in SEQ ID NO: 1.
  • the nucleotide probes can be labeled by a variety of methods, such as radiolabeling, biotinylation, or coupling to fluorescent or chemiluminescent tags, and detected by standard methods known in the art. Comparisons of CIFl 50/hTAF jl 50 genes, mRNA, or protein can be made as described above. A difference between the CIFl 50/hTAFjjl 50 genes (or a gene which regulates, for example, the expression, half-life, or degradation of CIF150/hTAFjjl50 mRNA) in the two tissues which are compared indicates neoplasia in the suspect tissue.
  • the degree of overexpression of the CIFl 50/hTAF/f 150 gene in the neoplastic tissue relative to wild-type expression of the gene in normal tissue, or differences in the amount of overexpression of the CIF150/hTAFjjl50 gene in the neoplastic tissue over time, can be used to prognose the progression of the neoplasia in that tissue or to monitor the response of the neoplastic tissue to various therapeutic regimens.
  • a genetic predisposition to neoplasia in a human can be detected by comparing a wild-type CIF150/hTAFjjl50 gene, mRNA, or protein with a CIF150/hTAFj ⁇ l50 gene, mRNA, or protein in a fetal tissue.
  • Fetal tissues which can be used for this purpose include, but are not limited to, amniotic fluid, chorionic villi, blood, and the blastomere of an in v/tro-fertilized embryo.
  • the wild-type CIF150/hTAFjjl50 gene can be obtained from any tissue.
  • the mRNA or protein can be obtained from a normal tissue of a human in which the CIF150/hTAFjjl50 gene is expressed.
  • Such tissues are disclosed above. Differences, such as alterations in the nucleotide sequence or size of the fetal CIF150/hTAFjl50 gene or mRNA, or alterations in the molecular weight, amino acid sequence, or relative abundance of fetal CIFl 50/hTAFjjl 50 protein, indicate a germline mutation in the CIF150/hTAF jl50 gene of the fetus which indicates a genetic predisposition to neoplasia.
  • Kits for use in the diagnostic methods described above are also provided.
  • CIF150/hTAFjjl50 diagnostic kits comprise reagents which specifically bind to a human CIF150/hTAF jl50 gene or expression product and which can be used in methods of the invention, such as CIF150/hTAF l50 subgenomic polynucleotide probes or antibodies.
  • Means for labeling the probes or antibodies, reagents for use in the methods, such as buffers, and instructions for using the kits can also be included.
  • the invention provides a means of identifying compounds which induce or prevent mitosis or cell cycle progression.
  • a cell is contacted with a test compound.
  • the test compound can be a pharmacologic agent already known in the art or can be a compound previously unknown to have any pharmacological activity.
  • the test compound can be naturally occurring or designed in the laboratory. It can be isolated from microorganisms, animals, or plants, and can be produced recombinantly, or synthesized by chemical methods known in the art.
  • the cell can be any primary human cell or human cell line which expresses a
  • Expression of the CIFl 50/hTAFjjl 50 gene is detected.
  • Means of detecting CIF150/hTAFjjl50 gene expression, by measuring CIF150/hTAFj l50 mRNA or CIFl 50/hTAF ⁇ 150 protein, are disclosed above. Expression can be measured in a sample of the same cell population before and after contact with the test compound. Alternatively, control cell populations can be employed.
  • a test compound which increases expression of the CIF150/hTAFjjl50 gene is identified as a potential drug for inducing mitosis or cell cycle progression.
  • a test compound which decreases expression of the CIF150/hTAFjjl50 gene is identified as a potential drug for decreasing mitosis or cell cycle progression.
  • Decreased CIF150/hTAFjjl50 expression can be achieved, for example, using ribozymes, antisense oligonucleotide sequences, or antibodies, as described above.
  • the effectiveness of the mechanism chosen to alter expression of the CIFl 50/hTAF jJ 50 gene can be assessed using methods well known in the art, such as hybridization of nucleotide probes to CIF150/hTAFjjl50 mRNA or detection of CIF150/hTAF ⁇ 150 protein using specific antibodies.
  • Differential display PCR can be carried out on the two populations of cells using methods well known in the art. See, e.g., Liang & Pardee, 1992, Science 257:961-11; Bauer et al, 1993, Nucl. Acids. Res. 27:4272-80; Bauer et al, 1994, PCR Methods Appl. 4S91-108; and Liang et al, 1995, Meth. Enz. 254:304-21. Kits for performing differential display PCR are available, for example, from Display Systems Biotech. Briefly, total RNA is isolated form the two populations of cells.
  • RNA is reverse transcribed to produce a cDNA population which represents an overlapping subset of the total expression profile of the cells in each population.
  • Each subset cDNA population is amplified using PCR with an anchored primer and a group of arbitrary primers in the presence of radiolabeled dATP. Amplified products from the two populations of cells are separated by gel electrophoresis, and patterns of separated products are detected, as is known in the art.
  • Differences in the two patterns indicates that the expression of one or more genes was altered in response to decreasing the expression of the CIF150/hTAFjjl50 gene.
  • Differentially displayed bands can be excised from the gel, reamplified, and identified by sequence analysis.
  • the sequences can be cloned before sequencing. Sequences of the differentially displayed bands can be compared with known sequences in databases to determine the identity of genes whose expression was altered in response to decreasing expression of CIFl 50/hTAFjjl 50.
  • the invention also provides means of identifying compounds which alter mitosis or cell cycle progression. A cell population is contacted with a test compound.
  • test compound can be a pharmacologic agent already known in the art or can be a compound previously unknown to have any pharmacological activity.
  • a test compound can be naturally occurring or designed in the laboratory. It can be isolated from microorganisms, animals, or plants, and can be produced recombinantly, or synthesized by chemical methods known in the art.
  • the cell population can comprise any primary human cell or human cell line which expresses a CIFl 50/hTAFjjl 50 gene, as disclosed above. Methods of establishing cultures of primary human cells or of culturing cell lines are well known in the art. Expression of the CIF150/hTAFjjl50 gene in the cell population is detected. Means of detecting CIFl 50/hTAFjjl 50 gene expression, for example by measuring CIF150/hTAF ⁇ l50 mRNA or OF150/hTAF ⁇ 150 protein, are disclosed above. Expression can be measured in a sample of the same cell population before and after contact with the test compound. Alternatively, control cell populations which have not been contacted with the test compound can be employed.
  • a test compound which increases expression of the CIF150/hTAF jl50 gene is identified as a potential compound for inducing mitosis or cell cycle progression.
  • a test compound which decreases expression of the CIF150/hTAFjjl50 gene is identified as a potential compound for inhibiting mitosis or cell cycle progression.
  • the invention also provides methods for screening test compounds for the ability to interfere with the binding of CIF 130 to OF150/hTAF ⁇ 150.
  • at least a OF150/hTAF ⁇ 150-binding domain of a CIF 130 protein as shown in SEQ ID NO:2 and at least a OF130-binding domain of a OF150/hTAF ⁇ 150 protein as shown in SEQ ID NO:4 are incubated together in the presence of a test compound.
  • a full-length CIF130 and/or a OF150/hTAF ⁇ 150 protein can also be used.
  • the OF150/hTAF ⁇ 150-binding domain binds to the OF130-binding domain.
  • the amount of bound and/or unbound proteins or binding domains is determined according to any technique known in the art, including any immunological technique.
  • one of the proteins or binding domains can be bound to a solid support, or can be labeled with a radiolabel, or other detectable label.
  • a useful agent is identified which decreases the amount of a binding domain or a protein which is bound or increases the amount of a binding domain or a protein which is unbound.
  • the binding domains or proteins can be prebound prior to the introduction of the test compound, or the test compound can be contacted with one of the two proteins or binding domains prior to incubation.
  • a two-hybrid assay is used to screen compounds which inhibit the interaction between the binding partners, CIF 130 and OF150/hTAF ⁇ 150.
  • two fusion proteins each comprising a binding domain of one of the binding partners, are used.
  • the fusion proteins can comprise full-length CIF 130 or CIF150/hTAF u 150 proteins or the portion of each protein necessary for the binding interaction.
  • One of the binding partners is fused to a DNA binding domain and the other is fused to a transcriptional activating domain. If the fusion protein comprising the CIFl 50/hTAF ⁇ l 50-binding domain comprises the DNA binding domain, then the fusion protein comprising the OF130-binding domain comprises the transcriptional activating domain.
  • the fusion protein comprising the OF130-binding domain comprises the DNA binding domain
  • the fusion protein comprising the OF150/hTAF ⁇ 150-binding domain comprises the transcriptional activating domain.
  • the two fusion proteins interact to reconstitute a sequence-specific transcriptional activating factor.
  • Many DNA binding domains and transcriptional activating domains can be used in this system, including the DNA binding domains of GAL4, LexA, and the human esfrogen receptor paired with the acidic transcriptional activating domains of GAL4 or the herpes virus simplex protein VP16.
  • the two fusion proteins are contained in a cell which also comprises a reporter gene. The reporter gene is sensitive to the activation of the reconstituted sequence- specific transcriptional activating factor.
  • Suitable reporter genes whose expression can be conveniently detected include the E. coli lacZ gene, whose expression may be measured colorimetrically, and yeast selectable genes such as H7S5 or URA3.
  • the OF150/hTAF ⁇ 150-binding domain of CIF130 and the OF130-binding domain of OF150/hTAF ⁇ 150 can be readily determined, for example, by testing various portions of each protein for the ability to bind to its partner. A variety of techniques can be used for this purpose, including but not limited to the yeast two-hybrid assay, affinity column chromatography, and polyacrylamide gel electrophoresis under non-reducing conditions.
  • the cell expresses the reporter gene.
  • a test compound is added to the cell, and the effect on expression of the reporter gene is measured.
  • CIF150/hTAF ⁇ 150 will have a negative effect on the transcriptional activation ability of the reconstituted sequence-specific transcriptional activating factor. Thus, expression of the reporter gene will be decreased. Compounds which decrease expression of the reporter gene are potential inducers of mitosis or cell cycle progression.
  • CIFl 50/hTAF jl 50 subgenomic polynucleotides can also be delivered to subjects for the purpose of screening test compounds for those which are useful for enhancing transfer of CIFl 50/hTAFjjl 50 subgenomic polynucleotides to the cell or for enhancing subsequent biological effects of the CIF150/hTAFjjl50 subgenomic polynucleotides within the cell. Such biological effects include hybridization to complementary CIF150/hTAFjjl50 mRNA and inhibition of its translation, expression of the
  • Test compounds which can be screened include any substances, whether natural products or synthetic, which can be administered to the subject. Libraries or mixtures of compounds can be tested. The compounds or substances can be those for which a pharmaceutical effect is previously known or unknown. The compounds or substances can be delivered before, after, or concomitantly with the CIF150/hTAFjjl50 subgenomic polynucleotides.
  • CIF150/hTAFjjl50 subgenomic polynucleotides can be administered separately or in admixture with the CIF150/hTAF jl50 subgenomic polynucleotides.
  • Integration of delivered CIF150/hTAFjjl50 subgenomic polynucleotides can be monitored by any means known in the art. For example, Southern blotting of the delivered CIF150/hTAFjjl50 subgenomic polynucleotides can be performed. A change in the size of the fragments of the delivered polynucleotides indicates integration.
  • Replication of the delivered polynucleotides can be monitored inter alia by detecting incorporation of labeled nucleotides combined with hybridization to a CIF150/hTAFj l50 probe.
  • CIF150/hTAFjjl50 subgenomic polynucleotide can be monitored by detecting production of CIFl 50/hTAFjjl 50 mRNA which hybridizes to the delivered polynucleotide or by detecting OF150/hTAF ⁇ 150 protein.
  • CIF 150/hTAF ⁇ l 50 protein can be detected immunologically or by activity, for example by detecting binding to CIF130.
  • delivery of CIFl 50/hTAFjjl 50 subgenomic polynucleotides according to the present invention provides an excellent system for screening test compounds for their ability to enhance delivery, integration, hybridization, expression, replication or integration in an animal, preferably a mammal, more preferably a human.
  • IMR 90 normal human lung fibroblasts
  • HeLa cells were transfected with the phosphothiorate oligomers (100 to 400 nM) according to the manufacturer's protocol (Sequiur Inc.), using lipofection and OptiMEM (GibcoBRL). The transfected cells were harvested at various times post-lipofection for FACS and for Western and Northern analysis.
  • the functional antisense oligomer designated B (5'-TGCTCATGGAAGCATAAGCAGCCAC-3'; SEQ ID NO:5) was used in combination with a control oligo Bx (5 * -CACCGACGAATACGAAGGTACTCGT-3'; SEQ ID NO:6) containing the reverse sequence (3'-5') of the oligo B to assure identical nucleotide content.
  • Density-arrested quiescent BALB/c3T3 and HeLa cells were prepared as previously described (Pledger et al, Proc. Natl Acad. Sci. U.S.A.
  • Cyclin A (-887 to+136) (28) and cyclin Bl(-893 to +110) (Oliphant et al, Mol. Cell. Biol. 9, 2944-49, 1989) promoter fragments were generated by PCR using HeLa DNA and cloned into the promoterless pGL3 luciferase reporter vector (Promega). All PCR-amplified fragments were verified by DNA sequencing. Cotransfections were performed in HeLa cells using a pENRFl (Cogswell et al, Mol. Cell. Biol.
  • CIF150/hTAFjjl50 expression vector pEVRF-OF150/hTAF ⁇ 150
  • pEVRFl-Ob expression plasmid in combination with the indicated reporter constructs. Luciferase activity was determined according to the manufacturer's protocol (Promega) 36 hr after transfection. Purification of proteins. HeLa nuclear extracts and CIF 150/hTAF ⁇ l 50 depleted nuclear extract were prepared as described previously (Zawel & Reinberg, 1995). OF150/hTAF ⁇ 150 protein was purified from SF9 cells under native conditions using the bacculovirus expression system (pBlueBacHis2, Invitrogen).
  • Plasmid DNAs containing cyclin A, cyclin Bl, CMV promoter fragments were cloned upstream of a 180 bp G-less cassette using a PCR protocol (Zawell & Reinberg, 1995; Kaufinann et al, 1996).
  • RNA samples 8 ⁇ l of the OF150/hTAF ⁇ 150 depleted nuclear extract (4 mg/ml) were preincubated for 30 min at 4 °C in the presence of DNA template with 1, 2 and 4 ⁇ l of recombinant CIF150/hTAF ⁇ 150 protein (see Fig. 3 A; 10 ⁇ l loaded), followed by addition of rNTPs to yield the following final concentrations: 500 ⁇ M ATP, 500 ⁇ M CTP, 500 ⁇ M GTP and 30 ⁇ M [ ⁇ - 32 P]UTP.
  • 32 P-labeled RNA products were digested 15 min with RNAase HI, resolved on an 8% polyacrylamide-urea gel, and visualized by autoradiography.
  • Elecfrophoretic mobility shift assays were performed using 3 and 6 ⁇ l OF150/hTAF ⁇ 150 in 40 ⁇ l GL-buffer as previously described (Zawell & Reinberg, 1995) with the exception that the reaction mixture contained 50 ng dGdC oligomer as competitor. After 30 min incubation at 40 °C the binding mixtures were loaded on a 6% TBE (0.5 x) polyacrylamide gel. Signals were quantitated by Phosphorimager analysis (BioRad, Inc.).
  • EXAMPLE 1 This example demonstrates that reduction in CIFl 50/hTAFjjl 50 mRNA or protein levels causes arrest of cells in G 2 .
  • oligomer B 5'-TGCTCATGGA AGCATAAGCA GCCAC-3'; SEQ ID NO:5
  • oligomer B 5'-TGCTCATGGA AGCATAAGCA GCCAC-3'
  • SEQ ID NO:5 5'-TGCTCATGGA AGCATAAGCA GCCAC-3'
  • EXAMPLE 3 This example demonstrates that CIFl 50/hTAF ⁇ l 50 expression and activity are cell cycle regulated and correlate with cyclin Bl and cyclin A transcription.
  • Serum-starved BALB/c 3T3 cells showed a lower level of OF150/hTAF ⁇ 150 protein and an increase in OF150/hTAF ⁇ 150 expression in G 2 /M after serum stimulation (Figure 2A).
  • OF150/hTAF ⁇ 150 upregulation began approximately 15 hr after serum addition, just before Cyclin A and Bl expression (lanes 5 and 6). Cyclin E expression did not significantly change in this cell line and serves here as a loading control.
  • OF150/hTAF ⁇ 150 is a positive regulator of cyclin Bl and A TATA-less promoters.
  • OF150/hTAF ⁇ 150 is a positive regulator of TATA-less promoters
  • OF150/hTAF ⁇ 150 protein Figure 3A
  • Cyclin Bl and Al promoter-dependent transcription was not observed in the absence of CIF150/hTAF ⁇ 150 activity.
  • the upper panel of Figure 3C shows absolute luciferase activities using the c-Fos, cyclin Bl, and cyclin A promoters in cofransfection experiments with an unrelated expression plasmid (pEVRFl-Ob) and a OF150/hTAF ⁇ 150 expression plasmid (pEVRF-OF150). Both the cyclin A and Bl promoters are preferentially stimulated by OF150/hTAF ⁇ 150 when compared to the TATA containing promoters c-Fos and CMV (see fold stimulation, lower panel of Figure 3C).
  • This example demonstrates identification of a cis-acting CIF150/hTAF ⁇ 150 responsive element.
  • Figure 4B shows that the selected protein-DNA complex is specific for OF150/hTAF ⁇ 150 protein, as demonstrated by a supershift induced by OF150/hTAF ⁇ 150-specific antiserum.
  • DNA sequences of 44 selected PCR-fragments revealed a statistically significant enrichment of fragments with the core sequence 5'GAG3' ( Figure 2C).
  • Affinity of OF150/hTAF ⁇ 150 for the core sequence was improved only about five-fold after four rounds of selection ( Figure 4A), possibly due to the presence of cryptic binding sites in the flanking sequences of the PCR primers used (see Figure 4B).
  • OF150/hTAF ⁇ 150 might be able to form stable complexes with non-specific DNA.
  • OF150/hTAF ⁇ 150 is a necessary positive transcriptional regulator of cell cycle progression through G 2 /M. Furthermore, we provide additional evidence to support the idea that one of the regulatory functions of TAF ⁇ s is to select core promoters. It is not known whether OF150/hTAF ⁇ 150-dependent transcription of the TATA-less cyclin Bl and A promoters is still TBP-dependent. An attractive hypothesis is that OF150/hTAF ⁇ 150 binding to the CBE element can compensate for the absence of TBP binding to the TATA-box.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A human protein termed CIF150/hTAFII150 recognizes and selects TATA-less core promoters for cell cycle specific genes. Thus, CIF150/hTAFII150 plays an important and selective role in establishing gene expression patterns necessary for progression through the cell cycle. The CIF150/hTAFII150 gene and its expression products can be used to alter the spatial or temporal patterns of mitosis or cell cycle progression of a human cell and to treat disorders involving alterations in the regulation of mitosis or cell cycle progression.

Description

CIF150/hTAF„150 IS NECESSARY FOR CELL CYCLE PROGRESSION
TECHNICAL AREA OF THE INVENTION
The invention relates to the area of cell cycle progression. More particularly, the invention relates to proteins which regulate cell cycle progression.
BACKGROUND OF THE INVENTION Alterations in the regulation of mitosis or cell cycle progression play an important role in diseases such as neoplasia and anemia. Manipulation of genes involved in regulating the cell cycle can be used to prevent or treat these diseases. Detections of mutations in cell-cycle regulatory genes can also be used to detect neoplastic cells and genetic predispositions to neoplasias. Thus, there is a need in the art for the identification of cell cycle regulator genes which can be used in methods of diagnosing, prognosing, and treating neoplasia and other diseases in humans and other mammals.
SUMMARY OF THE INVENTION
It is an object of the invention to provides reagents and methods for regulating mitosis or cell cycle progression in human cells and for treating disorders related to alterations in mitosis or cell cycle progression. These and other objects of the invention are provided by one or more of the embodiments described below.
One embodiment of the invention is an isolated and purified subgenomic polynucleotide which encodes a protein comprising an amino acid sequence which is at least 85% identical to the nucleotide sequence shown in SEQ ID NO:2. Percent identity is determined using a Smith- Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 1. Another embodiment of the invention is an isolated and purified subgenomic polynucleotide which comprises at least 11 contiguous nucleotides selected from the nucleotide sequence shown in SEQ ID NO:l.
Yet another embodiment of the invention is a construct which comprises a promoter and a polynucleotide segment encoding a human CIF150/hTAFπ150 protein as shown in SEQ ID NO:2. The polynucleotide segment is located downstream from the promoter. Transcription of the polynucleotide segment initiates at the promoter.
Still another embodiment of the invention is a host cell comprising a construct. The construct comprises a promoter and a polynucleotide segment encoding a human CIF150/hTAFπ150 protein as shown in SEQ ID NO:2.
Another embodiment of the invention provides a homologously recombinant cell having incorporated therein a new transcription initiation unit. The new transcription initiation unit comprises an exogenous regulatory sequence, an exogenous exon, and a splice donor site. The transcription initiation unit is located upstream of a coding sequence of a CIF150/hTAFj_τl50 gene. The exogenous regulatory sequence directs transcription of the coding sequence of the CIF150/hTAFjjl 50 gene.
Even another embodiment of the invention provides a method to aid in the diagnosis or prognosis of neoplasia in a human. Expression of a first CIF150/hTAFjjl50 gene in a first tissue of a human suspected of being neoplastic is compared with expression of a second CIF150/hTAF jl50 gene in a second tissue of a human which is normal. The second CIF150/hTAFjjl50 gene has the coding sequence shown in SEQ ID NO:l. Increased expression of the first CIF150/hTAF/jl50 relative to the second CIF150/hTAFjjl50 gene indicates neoplasia in the first tissue.
Yet another embodiment of the invention provides a method to aid in the diagnosis or prognosis of neoplasia in a human. A human CIF150/hTAFjjl 50 gene, mRNA, or protein in a first tissue suspected of being neoplastic is compared with a second human CIF150/hTAFjjl50 gene, mRNA, or protein in a second tissue which is normal. The second CIF150/hTAFjjl 50 gene has the coding sequence shown in SEQ ID NO: 1. A difference between the first and second genes, mRNAs, or proteins in the second tissue indicates neoplasia in the first tissue.
Still another embodiment of the invention provides a method to aid in detecting a genetic predisposition to neoplasia in a human. A CIF150/hTAFj l 50 gene, mRNA, or protein in a fetal tissue of a human is compared with a wild-type human
CIF150/hTAFjιl50 gene, mRNA, or protein. The wild-type CIF150/hTAFjjl50 gene has the coding sequence shown in SEQ ID NO: 1. A difference between the CIF150/hTAFj l50 gene, mRNA, or protein in the fetal tissue of the human and the wild-type human CIF150/hTAF jl50 gene, mRNA, or protein indicates a genetic predisposition to neoplasia in the human.
Even another embodiment of the invention provides a method of screening test compounds for the ability to interfere with the binding of a OF 130 protein to a CIF150/hTAFπ150 protein. A test compound is contacted with a CIF150/hTAFπ150- binding domain of a OF 130 protein and a OF130-binding domain of a OF150/hTAFπ150 protein. The CIF130 protein has the amino acid sequence shown in SEQ ID NO:4. The OF150/hTAFπ150 protein has the amino acid sequence shown in SEQ ID NO:2. The CIF130-binding domain binds to the OF150/hTAFπ150-binding domain in the absence of the test compound. The amount of at least one of the OF 130- or OF150/hTAFπ150-binding domains which is bound or unbound is determined in the presence of the test compound. A test compound which decreases the amount of bound CIF130- or OF150/hTAFπ150-binding domains or which increases the amount of unbound CIF130- and OF150/hTAFπ150-binding domains is a potential inducer of mitosis or cell cycle progression.
Yet another embodiment of the invention provides a method of screening test compounds for the ability to interfere with the binding of a OF 130 protein to a OF 150/hTAFπl 50 protein. A cell is contacted with a test compound. The cell comprises two fusion proteins. A first fusion protein comprises (1) a OF150/hTAFπ150- binding domain of a OF 130 protein and (2) either a DNA binding domain or a transcriptional activating domain. A second fusion protein comprises a OF130-binding domain of a OF150/hTAFπ150 protein. The CIF130 protein has the amino acid sequence shown in SEQ ID NO:4. The OF150/hTAFπ150 protein has the amino acid sequence shown in SEQ ID NO:2. The OF130-binding domain binds to the OF150/hTAFπ150-binding domain. If the first fusion protein comprises a DNA binding domain, then the second fusion protein comprises a transcriptional activating domain. If the first fusion protein comprises a transcriptional activating domain, then the second fusion protein comprises a DNA binding domain. The interaction of the first and second fusion proteins reconstitutes a sequence-specific transcription activating factor. The cell also comprises a reporter gene comprising a DNA sequence to which the DNA binding domain specifically binds. Expression of the reporter gene is measured. A test compound which decreases the expression of the reporter gene is a potential inducer of mitosis or cell cycle progression.
Even another embodiment of the invention provides a method of increasing expression of a gene. A promoter region of the gene is contacted with a CIFl 50/hTAFπl 50 protein as shown in SEQ ID NO:2. The promoter region of the gene comprises a OF150/hTAFπ150 binding element. The OF150/hTAFπ150 binding element comprises a nucleotide sequence 5'-Py X G A G A/C A/Py -3' (SEQ ID NO:7). Expression of the gene is thereby increased.
Still another embodiment of the invention is an antisense oligonucleotide as shown in SEQ ID NO:5.
Yet another embodiment of the invention provides an isolated and purified subgenomic polynucleotide which comprises 5'-Py X G A G A/C A Py-3' (SEQ ID NO:7).
The present invention thus provides the art with reagents and methods of affecting human mitosis or cell cycle progression and treating disorders associated with alterations in mitosis or cell cycle progression. BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1. Functional knock out of OF 150/hTAFπ 150 protein leads to cell cycle arrest in G2/M and reduced gene expression of cyclin Bl. Figure 1A. Analysis of the cell cycle of HeLa cells 36 hours after transfection with the OF150/hTAFπ150 specific antisense oligomer B or the oligomer Bx (reverse sequence). Concentrations of the oligomers are indicated. Figure IB. Cell cycle analysis of IMR 90 cells after 36 hours of oligomer treatment. Figure lC. Total RNA derived from HeLa cells treated with oligomer B or Bx analyzed by quantitative RT-PCR (lanes 1 to 6, 100, 200, 300 nM oligomer B and Bx) and Northern blot analysis (lanes 7 to 10) using OF150/hTAFπ150 and β-actin-specific primers or 32P-labeled cDNA probes. Figure ID. HeLa cell extracts from different time points after antisense oligomer treatment analyzed by immunoblotting for the decrease of OF150/hTAFπ150 protein. A control nuclear extract (lane 1) and a molecular size marker (lane 2) were loaded. Figure IE. Cyclin Bl protein expression decreases after OF150/hTAFπ150 antisense treatment of HeLa cells (left panel). Figure IF. PCR display identifies genes which are transcriptionally dependent on OF150/hTAFπ150. Total RNA was prepared 24 hours (lane 1 and 2) or 36 hours (lane 3 and 4) after oligomer transfection.
Figure 2. Regulation of OF 150/hTAFπl 50 expression and activity is cell cycle dependent. Figure 2 A. Quiescent BALB/c 3T3 cells were stimulated to undergo cell cycle traverse by serum addition. Extracts derived from cells at times indicated above were assayed for OF150/hTAFπ150, cyclin Bl, cyclin A, and cyclin E by immunoblot. Figure 2B. HeLa cells were assayed for OF150/hTAFπ150 and cyclin Bl expression after synchronization for 48 hours by serum deprivation. Figure 2C. Nuclear extracts of HeLa cells harvested at different time points after release from a double thymidine block were used for in vitro transcription assays using promoter with (odd numbered lanes) and without functional TdT Inr (even numbered lanes). Nuclear extracts were standardized by protein concentration.
Figure 3. OF150/hTAFπ150 is a positive regulator of cyclin Bl and cyclin A transcription. Figure 3 A. Purified OF150/hTAFπ150 was visualized after SDS-PAGE by silver staining. Figure 3B. OF150/hTAFπ150 is required for cyclin Bl and cyclin A transcription but not for IgH and CMN transcription. In vitro transcription was performed using OF150/hTAFπ150 depleted nuclear extracts (lane 1) or in combination with increasing amounts of recombinant OF150/hTAFπ150 protein (lane 2 to 4). Figure 3 C . Cofransfection of increasing amounts of OF 150/hTAFπ 150 expression plasmid with cyclin Bl, cyclin A, CMV, and Fos promoter driven luciferase reporter constructs. The values are represent the average of four experiments +/- standard deviation.
Figure 4. OF150/hTAFπ150 has sequence-specific binding activity. Figure 4A. OF150/hTAFπ150 binding site selection. Figure 4B. Addition of anti-CIF 150/hTAFπl 50 (lane 4) but not preimmune serum (lane 3) supershifted the retarded band. Figure 4C. Consensus OF150/hTAFπ150 binding element (CBE). Figure 4D. Point mutational analysis of the OF150/hTAFπ150 binding element using gel shift experiments.
Figure 5. Identification of a CIF 150/hTAFπl 50 binding element (CBE) in the cyclin B 1 core promoter. Cofransfection of increasing amounts of CIF 150/hTAFπ 150 expression plasmid with cyclin B 1 wild type and cyclin B 1 promoter mutant. The values represent the average of three experiments.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS It is a discovery of the present invention that a protein termed CIF 150/hTAFπl 50 recognizes and selects TATA-less core promoters for cell cycle specific genes. Thus, OF150/hTAFπ150 plays an important and selective role in establishing gene expression patterns necessary for progression through the cell cycle. Through manipulation of CIF150/hTAFjjl50 expression, the present invention provides reagents and methods for affecting mitosis or cell cycle progression of human cells and for treating disorders associated with alterations in mitosis or cell cycle progression.
OF150/hTAFπ150 is a necessary positive transcriptional regulator of cell cycle progression through G2/M. OF150/hTAFπ150 is an essential cofactor for TFπD- dependent transcription from promoters containing initiator elements, such as the adenovirus major late promoter. OF150/hTAFπ150 directly interacts with the transcription factor hTAFπ135 and stabilizes TFπD binding to RNA polymerase II core promoters and can stimulate expression of cell cycle-specific genes such as cyclin A and B 1. The promoters of such genes contain a CIF 150/hTAFπl 50 binding element which comprises a OF150/hTAFπ150 binding site, 5'-Py X G A G A/C A/Py-3' (SEQ ID NO:7).
OF150/hTAFπ150 binds to and is negatively regulated by a 130 kD polypeptide, CIF 130, which has sequence homology to ATP-dependent RNA helicases (DEAD-box proteins) implicated in the control of mitosis in Schizosaccharomyces pombe. A nucleotide sequence encoding CIF130 is shown in SEQ ID NO:3. The amino acid sequence of CIF130 is shown in SEQ ID NO:4.
Human OF150/hTAFπ150 protein has the amino acid sequence shown in SEQ ID NO:2. Protein variants of CIF150/hTAFπ150 protein can also have OF150/hTAFπ150 activity. Biologically active OF150/hTAFπ150 variants can be naturally or non-naturally occurring. Naturally occurring OF150/hTAFπ150 variants are those which are found in humans or other species and which comprise amino acid sequences which are substantially identical to the amino acid sequence shown in SEQ ID NO:2. Non-naturally occurring OF150/hTAFπ150 variants which retain substantially the same biological activities as OF150/hTAFπ150 or naturally occurring CIF 150/hTAFπl 50 variants can be constructed in the laboratory.
Preferably, naturally or non-naturally occurring protein biologically active OF150 hTAFπ150 variants have amino acid sequences which are at least 65%, 75%, 85%, 90%, or 95% identical to the amino acid sequence shown in SEQ ID NO:2 and have similar biological properties, including the ability to bind to the OF150/hTAFπ150 binding element, to permit mitosis, to activate TATA-less core promoters, and to bind to CIF130. More preferably, the molecules are at least 98% or 99% identical. Percent sequence identity between the OF150/hTAFπ150 protein shown in SEQ ID NO:2 and a biologically active OF150 hTAFπ150 variant is determined using the Smith- Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 1. The Smith- Waterman homology search algorithm is taught in Smith and Waterman, Adv. Appl. Math. (1981) 2:482-489.
Guidance in determining which amino acid residues may be substituted, inserted, or deleted without abolishing biological or immunological activity of OF150/hTAFu150 may be found using computer programs well known in the art, such as DNASTAR software. Preferably, the amino acid changes in biologically active OF150/hTAFπ150 variants are conservative amino acid changes, i.e., substitutions of similarly charged or uncharged amino acids. A conservative amino acid change involves substitution of one of a family of amino acids which are related in their side chains. Naturally occurring amino acids are generally divided into four families: acidic (aspartate, glutamate), basic (lysine, arginine, histidine), non-polar (alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), and uncharged polar (glycine, asparagine, glutamine, cystine, serine, threonine, tyrosine) amino acids. Phenylalanine, tryptophan, and tyrosine are sometimes classified jointly as aromatic amino acids. It is reasonable to expect that an isolated replacement of a leucine with an isoleucine or valine, an aspartate with a glutamate, a threonine with a serine, or a similar replacement of an amino acid with a structurally related amino acid will not have a major effect on the binding properties of the resulting OF150/hTAFπ150 variant, especially if the replacement does not involve an amino acid at the OF130-binding site of OF150/hTAFπ150 or the site at which OF150/hTAFπ150 binds to the
OF150/hTAFπ150 binding element. Properties and functions of OF150/hTAFπ150 variants are of the same type as a OF150/hTAFπ150 protein having the amino acid sequence shown in SEQ ID NO:2, although the properties and functions may differ in degree. Whether an amino acid change results in a OF150/hTAFπ150 protein or polypeptide variant which can function as the CIF 150/hTAFπl 50 protein disclosed herein can readily be determined. For example, the ability of a OF150/hTAFπ150 variant to bind to the OF150/hTAFπ150 binding element can be tested using in vitro DNA binding assays, as taught in Kaufinann & Smale, 1994, Genes Bevel. 8: 821-29 and Kaufinann et al., 1996, Genes Devel. 70:873-86. The ability of a OF150/hTAFπ150 protein or polypeptide variant to induce mitosis or cell cycle progression can be assayed as described below.
OF150 hTAFπ150 polypeptides contain less than full-length OF150/hTAFπ150. For example, CIF150/hTAFπ150 polypeptides contain at least 5, 6, 8, 10, 12, 20, 25, 50, 75, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1250, or 1300 or more amino acids of a OF150/hTAFπ150 protein or biologically active variant in the same primary order as found in a OF150/hTAFπ150 protein or variant obtained from a natural source. Polypeptide molecules having substantially the same amino acid sequence as OF150/hTAFπ150 but possessing minor amino acid substitutions which do not substantially affect the ability of the CIF 150/hTAFπl 50 polypeptide variants to interact with CIF150/hTAFπ150-specific molecules, such as CIF130 or antibodies which specifically bind to CIF150/hTAFπ150, are within the definition of CIF150/hTAFπ150. CIF 130 variants include glycosylated forms, aggregative conjugates with other molecules, and covalent conjugates with unrelated chemical moieties. Truncations or deletions of regions which do not affect the properties or functions of OF150/hTAFπ150 described above are also biologically active variants of OF150/hTAFπ150. Covalent variants can be prepared by linkage of functionalities to groups which are found in the amino acid chain or at the N- or C-terminal residue, as is known in the art. A subset of mutants, called muteins, is a group of polypeptides in which neutral amino acids, such as serines, are substituted for cysteine residues which do not participate in disulfide bonds. These mutants may be stable over a broader temperature range than native OF150/hTAFπ150. See Mark et al, U.S. Pat. No. 4,959,314.
Naturally occurring OF150/hTAFπ150 proteins or polypeptides can be purified from human cells or cell lines, such as HeLa or NIH 3T3 cells, by methods known in the art. CIF150/hTAFπ150 copurifies with CIF 130; thus, the initial purification steps for each protein are the same. OF150/hTAFπ150 can be conveniently purified from HeLa cell extracts (Dignam et al, 1983, Nucl. Acids Res. ii:1475-89) using Ni affinity chromatography. For Ni affinity purification, the 0.1 M KO flowthrough fraction of a DEAE-Sephacel column is applied to a Mono Q column and eluted with a linear KCl gradient (40 ml; 0.1 to 1 M). The CIF130/OF150/hTAFπ150-containing fractions are pooled and dialyzed against buffer A (20 mM HEPES, pH 7.9, 1 mM EDTA, 3 mM dithiothreitol, 1 mM phenylmethylsulfonyl fluoride, 20% glycerol) containing 0.1 M KCl. These fractions are supplemented with imidazole (final concentration, 20 mM in buffer A) and applied to a Ni-nitrilotriacetic acid (NTA)-agarose column (Qiagen). After being washed with 10 column volumes each of 20 mM imidazole and 35 mM imidazole, bound proteins are eluted with 100 mM imidazole.
Protein fractions can be tested for OF150/hTAFπ150 activity as described in Kaufinann et al, 1996. Purified CIF130 and CIFl 50/hTAFπ 150 proteins can be visualized by sodium dodecyl sulfate-6% polyacrylamide gel electrophoresis, followed by silver staining. OF150/hTAFπ150 can be separated from CIF130 by excising the OF150/hTAFπ150-containing band from the SDS gel and eluting the OF150/hTAFπ150 protein, as is known in the art. A preparation of isolated and purified OF150/hTAFπ150 protein is at least 80% pure; preferably, the preparations are at least 90%, 95%, or 99% pure.
OF150/hTAFπ150 proteins and polypeptides can also be produced by recombinant DNA methods or by synthetic chemical methods. For production of recombinant OF150/hTAFπ150 proteins or polypeptides, coding sequences selected from the CIF150/hTAFjjl50 nucleotide sequence shown in SEQ ID NO: 1 can be expressed in known prokaryotic or eukaryotic expression systems (see below). To avoid non-specific T7 RNA polymerase transcription in functional assays, in v/'tro-franslated OF150/hTAFπ150 can be purified with Ni-NTA-agarose as described above (100 mM imidazole eluate) and concentrated with a Centricon 30 concentrator (Amicon). Bacterial, yeast, insect, or mammalian expression systems can be used, as is known in the art. Alternatively, synthetic chemical methods, such as solid phase peptide synthesis, can be used to synthesize OF150/hTAFπ150 protein or polypeptides. OF150/hTAFπ150 biologically active variants can be similarly produced. Fusion proteins comprising at least six contiguous OF150/hTAFπ150 amino acids can also be constructed. Human OF150/hTAFπ150 fusion proteins are useful for generating antibodies against OF150/hTAFπ150 amino acid sequences and for use in various assay systems. For example, OF150/hTAFπ150 fusion proteins can be used to identify proteins which interact with CIF 150/hTAFπl 50 protein and influence its ability to affect transcription. Physical methods, such as protein affinity chromatography, or library-based assays for protein-protein interactions, such as the yeast two-hybrid or phage display systems, can be used for this purpose. Such methods are well known in the art and can also be used as drug screens, as described below. A OF150/hTAFπ150 fusion protein comprises two protein segments fused together by means of a peptide bond. The first protein segment consists of at least 5, 6, 8, 10, 12, 20, 25, 50, 75, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1250, or 1300 contiguous amino acids of a OF150/hTAFπ150 protein. The amino acids can be selected from the amino acid sequence shown in SEQ ID NO:2 or from a biologically active variant of that sequence. The first protein segment can also be a full-length
CIF150/hTAFπl 50 protein. The first protein segment can be N-terminal or C-terminal, as is convenient.
The second protein segment can be a full-length protein or a protein fragment or polypeptide. Proteins commonly used in fusion protein construction include β- galactosidase, β-glucuronidase, green fluorescent protein (GFP), autofluorescent proteins, including blue fluorescent protein (BFP), glutathione-S-fransferase (GST), luciferase, horseradish peroxidase (HRP), and chloramphenicol acetyltransferase (CAT). Epitope tags can be used in fusion protein constructions, including histidine (His) tags, FLAG tags, influenza hemagglutinin (HA) tags, Myc tags, NSN-G tags, and thioredoxin (Trx) tags. Other fusion constructions can include maltose binding protein (MBP), S-tag, Lex A DΝA binding domain (DBD) fusions, GAL4 DΝA binding domain fusions, and herpes simplex virus (HSN) BP16 protein fusions.
OF150/hTAFπ150 fusion proteins can be made by covalently linking the first and second protein segments or by standard procedures in the art of molecular biology. Recombinant DNA methods can be used to prepare OF150/hTAFπ150 fusion proteins, for example, by making a DNA construct which comprises coding sequences selected from SEQ ID NO:l in proper reading frame with nucleotides encoding the second protein segment and expressing the DNA construct in a host cell, as is known in the art. Many kits for constructing fusion proteins are available from companies which supply research labs with tools for experiments, including, for example, Promega Corporation (Madison, WI), Stratagene (La Jolla, CA), Clontech (Mountain View, CA), Santa Cruz Biotechnology (Santa Cruz, CA), MBL International Corporation (MIC; Watertown, MA), and Quantum Biotechnologies (Montreal, Canada; 1-888-DNA-KITS). Isolated and purified CIF 150/hTAFπl 50 proteins, polypeptides, variants, or fusion proteins can be used as immunogens, to obtain a preparation of antibodies which specifically bind to a OF150/hTAFπ150 protein. The antibodies can be used, inter alia, to detect wild-type OF150/hTAFπ150 proteins in human tissue and fractions thereof. The antibodies can also be used to detect the presence of mutations in the CIF150/hTAFjjl50 gene which result in under- or over-expression of the
OF150/hTAFπ150 protein or in expression of a OF150/hTAFπ150 protein with altered size or electrophoretic mobility.
Antibodies which specifically bind to epitopes of OF150/hTAFπ150 proteins, polypeptides, fusion proteins, or biologically active variants can be used in immunochemical assays, including but not limited to Western blots, ELISAs, radioimmunoassays, immunohistochemical assays, immunoprecipitations, or other immunochemical assays known in the art. Typically, antibodies of the invention provide a detection signal at least 5-, 10-, or 20-fold higher than a detection signal provided with other proteins when used in such immunochemical assays. Preferably, antibodies which specifically bind to OF150/hTAFπ150 epitopes do not detect other proteins in immunochemical assays and can immunoprecipitate CIF150/hTAFπ150 protein or polypeptides from solution.
CIF150/hTAFπ150-specific antibodies specifically bind to epitopes present in a OF150/hTAFπ150 protein having the amino acid sequence shown in SEQ ID NO:2 or to biologically active variants of that sequence. Typically, at least 6, 8, 10, or 12 contiguous amino acids are required to form an epitope. However, epitopes which involve non-contiguous amino acids may require more, e.g., at least 15, 25, or 50 amino acids. Preferably, CIF150/hTAFπ150 epitopes are not present in other human proteins. One such preparation of anti-OF 150/hTAFπl 50 antibodies are polyclonal antibodies raised against an N-terminal peptide of OF150/hTAFπ150, such as MNRKKGDKGF (amino acids 11-20 of SEQ ID NO:2) or MNRKKGDKGFESPRP (amino acids 11-25 of SEQ ID NO.2).
Epitopes of OF150/hTAFπ150 which are particularly antigenic can be selected, for example, by routine screening of CIF 150/hTAFπl 50 polypeptides for antigenicity or by applying a theoretical method for selecting antigenic regions of a protein to the amino acid sequence shown in SEQ ID NO:2. Such methods are taught, for example, in Hopp and Wood, Proc. Natl. Acad. Sci. U.S.A. 78, 3824-28 (1981), Hopp and Wood, Mol. Immunol. 20, 483-89 (1983), and Sutcliffe et al, Science 219, 660-66 (1983). Any type of antibody known in the art can be generated to bind specifically to
OF150/hTAFπ150 epitopes. For example, preparations of polyclonal and monoclonal antibodies can be made using standard methods which are well known in the art. Similarly, single-chain antibodies can also be prepared. Single-chain antibodies which specifically bind to OF150/hTAFπ150 epitopes can be isolated, for example, from single-chain immunoglobulin display libraries, as is known in the art. The library is
"panned" against OF150/hTA-Fπ150 amino acid sequences, and a number of single chain antibodies which bind with high-affinity to different epitopes of OF150/hTAFπ150 protein can be isolated. Hayashi et al, 1995, Gene 160:129-30. Single-chain antibodies can also be constructed using a DNA amplification method, such as the polymerase chain reaction (PCR), using hybridoma cDNA as a template. Thirion et al, 1996, Eur. J. Cancer Prev. 5:507-11.
Single-chain antibodies can be mono- or bispecific, and can be bivalent or tefravalent. Construction of tettavalent, bispecific single-chain antibodies is taught, for example, in Coloma and Morrison, 1997, Nat. Biotechnol. 75:159-63. Construction of bivalent, bispecific single-chain antibodies is taught inter alia in Mallender and Noss, 1994, J Biol. Chem. 269:199-206.
A nucleotide sequence encoding a single-chain antibody can be constructed using manual or automated nucleotide synthesis, cloned into an expression construct using standard recombinant DΝA methods, and introduced into a cell to express the coding sequence, as described below. Alternatively, single-chain antibodies can be produced directly using, for example, filamentous phage technology. Nerhaar et al, 1995, Int. J. Cancer 61:491-501; Νicholls et al, 1993, J. Immunol. Meth. 755:81-91.
Monoclonal and other antibodies can also be "humanized" in order to prevent a patient from mounting an immune response against an antibody when it is used therapeutically. Such antibodies may be sufficiently similar in sequence to human antibodies to be used directly in therapy or may require alteration of a few key residues. Sequence differences between, for example, rodent antibodies and human sequences can be minimized by replacing residues which differ from those in the human sequences, for example, by site directed mutagenesis of individual residues, or by grafting of entire complementarity determining regions. Alternatively, one can produce humanized antibodies using recombinant methods, as described in GB2188638B. Antibodies which specifically bind to OF150/hTAFπ150 epitopes can contain antigen binding sites which are either partially or fully humanized, as disclosed in U.S. 5,565,332. Other types of antibodies can be constructed and used in methods of the invention. For example, chimeric antibodies can be constructed as disclosed, for example, in WO 93/03151. Binding proteins which are derived from immunoglobulins and which are multivalent and multispecific, such as the "diabodies" described in WO 94/13804, can also be prepared. Antibodies of the invention can be purified by methods well known in the art.
For example, antibodies can be affinity purified by passing the antibodies over a column to which a CIF150/hTAFπ150 protein, polypeptide, biologically active variant, or fusion protein is bound. The bound antibodies can then be eluted from the column, using a buffer with a high salt concenfration. OF150/hTAFπ150-specific binding polypeptides other than antibodies can also be generated. OF150/hTAFπ150-specific binding polypeptides are polypeptides which bind with OF150/hTAFπ150 or its variants and which have a measurably higher binding affinity for CIF150/hTAFπ150 and polypeptide derivatives of OF150/hTAFπ150 than for other polypeptides tested for binding. Higher affinity by a factor of 10 is preferred, more preferably a factor of 100. Such polypeptides can be identified, for example, using the yeast two-hybrid system.
The coding region of the human CIF150/hTAFjjl50 gene has the nucleotide sequence shown in SEQ ID NO: 1. The complement of the nucleotide sequence shown in SEQ ID NO: 1 consists of a contiguous nucleotide sequence which forms Watson-Crick base pairs with the contiguous nucleotide sequence shown in SEQ ID NO:l.
CIFl 50/hTAFjjl 50 subgenomic polynucleotides can comprise at least 11, 20, 25, 30, 35, 40, 45, 50, 55, 60, 67, 70, 75, 100, 125, 150, 200, 250, 300, 400, 500, 750, 1000, 1250, 1500, 1750, 2000, 2500, 3000, 3500, or 3900 or more contiguous nucleotides selected from the nucleotide sequence shown in SEQ ID NO:l or its complement.
Subgenomic polynucleotides contain less than a whole chromosome and are preferably intron-free.
The complement of the nucleotide sequence shown in SEQ ID NO:l can be used provide CIF150/hTAFjjl50 antisense oligonucleotides. CIF150/hTAFj l50 subgenomic polynucleotides also include polynucleotides which encode CIF 150/hT AFπ 150-specific single-chain antibodies, ribozymes, and biologically active or altered OF150/hTAFπ150 variants.
Degenerate nucleotide sequences encoding amino acid sequences of OF150/hTAFπ150 protein or biologically active CIF150/hTAFπ150 variants, as well as homologous nucleotide sequences which are at least 65%, 75%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to the nucleotide sequence shown in SEQ ID NO: 1, are also CIF150/hTAF jl50 subgenomic polynucleotides. Percent sequence identity between the nucleotide sequence of SEQ ID NO:l and a putative homologous or degenerate CIFl 50/hTAFjjl 50 nucleotide sequence is determined using computer programs which employ the Smith- Waterman algorithm, for example as implemented in the MPSRCH program (Oxford Molecular), using an affine gap search with the following parameters: a gap open penalty of 12 and a gap extension penalty of 1.
Nucleotide sequences which hybridize to the coding sequence shown in SEQ ID NO: 1 or its complement with at most 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, or 35% basepair mismatches are also CIF150/hTAFjjl50 subgenomic polynucleotides of the invention. For example, using the following wash conditions~2X SSC (0.3 M sodium chloride, 0.03 M sodium citrate, pH 7.0), 0.1% SDS, room temperature twice, 30 minutes each; then 2X SSC, 0.1% SDS, 50 °C once, 30 minutes; then 2X SSC, room temperature twice, 10 minutes each— homologous CIFl 50/hTAFjjl 50 sequences can be identified which contain at most about 25-30% basepair mismatches with SEQ ID NO:l or its complement. More preferably, homologous nucleic acid strands contain 15-25% basepair mismatches, even more preferably 5-15% basepair mismatches.
Species homologs of CIFl 50/hTAFj l 50 subgenomic polynucleotides of the invention can also be identified by making suitable probes or primers and screening cDNA expression libraries from other species, such as mice, monkeys, yeast, or bacteria, as well as human cDNA expression libraries. It is well known that the Tm of a double- stranded DNA decreases by 1-1.5 °C with every 1% decrease in homology (Bonner et al, J. Mol. Biol. 81, 123 (1973). Homologous CIF150/hTAFjιl50 human polynucleotides or CIFl 50/hTAFjιl 50 polynucleotides of other species can therefore be identified, for example, by hybridizing a putative homologous CIF150/hTAF jl50 polynucleotide with a polynucleotide having the nucleotide sequence of SEQ ID NO: 1 to form a test hybrids, comparing the melting temperature of the test hybrid with the melting temperature of a hybrid comprising a polynucleotide having SEQ ID NO:l and a polynucleotide which is perfectly complementary to SEQ ID NO: 1 , and calculating the number or percent of basepair mismatches within the test hybrid.
Nucleotide sequences which hybridize to the coding sequence shown in SEQ ID NO:l or its complement following stringent hybridization and/or wash conditions are also CIFl 50/hTAFjjl 50 subgenomic polynucleotides of the invention. Stringent wash conditions are well known and understood in the art and are disclosed, for example, in Sambrook et al, MOLECULAR CLONING: A LABORATORY MANUAL, 2d ed., 1989, at pages 9.50-9.51.
Typically, for stringent hybridization conditions, a combination of temperature and salt concenfration should be chosen that is approximately 12-20 °C below the calculated Tm of the hybrid under study. The Tm of a hybrid between the CIF150/hTAFjjl50 sequence shown in SEQ ID NO:l and a polynucleotide sequence which is 65%, 75%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO:l can be calculated, for example, using the equation of Bolton and McCarthy, Proc. Natl. Acad. Sci. U.S.A. 48, 1390 (1962):
Tm = 81.5 °C - 16.6(logI0[Na+]) + 0.41(%G + C) - 0.63(%formamide) - 600//), where / = the length of the hybrid in basepairs.
Stringent wash conditions include, for example, 4X SSC at 65 °C, or 50% formamide, 4X SSC at 42 °C, or 0.5X SSC, 0.1% SDS at 65 °C. Highly stringent wash conditions include, for example, 0.2X SSC at 65 °C.
CIF150/hTAFjjl50 subgenomic polynucleotides can be isolated and purified free from other nucleotide sequences using standard nucleic acid purification techniques. For example, restriction enzymes and probes can be used to isolate polynucleotide fragments which comprise nucleotide sequences encoding a OF150/hTAFπ150 protein or variant. Isolated and purified subgenomic polynucleotides are in preparations which are free or at least 90% free of other molecules.
Complementary DNA (cDNA) molecules which encode CIF150/hTAFπ150 proteins are also CIFl 50/hTAFjjl 50 subgenomic polynucleotides of the invention. CIF150/hTAFjjl50 cDNA molecules can be made with standard molecular biology techniques, using CIF150/hTAFjjl50 mRNA as a template. CIF150/hTAFjjl50 cDNA molecules can thereafter be replicated using molecular biology techniques known in the art and disclosed in manuals such as Sambrook et al, 1989. An amplification technique, such as the polymerase chain reaction (PCR), can be used to obtain additional copies of subgenomic polynucleotides of the invention, using either human genomic DNA or cDNA as a template.
Alternatively, synthetic chemistry techniques can be used to synthesize CIFl 50/hTAFjjl 50 subgenomic polynucleotide molecules of the invention. The degeneracy of the genetic code allows alternate nucleotide sequences to be synthesized which will encode a CIF150/hTAFπ150 protein having the amino acid sequence shown in SEQ ID NO:2 or a biologically active variant of that sequence. All such nucleotide sequences are within the scope of the present invention.
The invention also provides polynucleotide probes which can be used to detect CIF150/hTAFjjl50 sequences, for example, in hybridization protocols such as Northern or Southern blotting or in situ hybridization. Polynucleotide probes of the invention comprise at least 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, or 40 or more contiguous nucleotides selected from SEQ ID NO: 1. Polynucleotide probes of the invention can comprise a detectable label, such as a radioisotopic, fluorescent, enzymatic, or chemiluminescent label.
Purified and isolated CIFl 50/hTAFjjl 50 subgenomic polynucleotides can be used as primers to obtain additional copies of the polynucleotides or as probes for identifying wild-type and mutant CIFl 50/hTAFjjl 50 coding sequences. CIF150/hTAFjjl50 subgenomic polynucleotides can also be used to express CIF150/hTAFjjl50 mRNA, protein, polypeptides, fusion proteins and the like and to generate CIFl 50/hTAF jl 50 antisense oligonucleotides and ribozymes.
CIF150/hTAFjιl50 subgenomic polynucleotide comprising CIF150/hTAFπ150 coding sequences can be used in a construct, such as an RNA or DNA construct. A CIF150/hTAFjjl50 construct can be an expression construct, which can be used to express all or a portion of a OF150/hTAFπ150 protein in a host cell. Host cells comprising OF150/hTAFπ150 expression constructs can be prokaryotic or eukaryotic. Preferably, the CIFl 50/hTAF jl 50 subgenomic polynucleotide is inserted into an expression plasmid (for example, the Ecdyson system, pIND, In Vitro Gene). A variety of host cells are available for use in bacterial, yeast, insect, and human expression systems and can be used to express OF150/hTAFπ150 expression constructs (see below). Expression constructs can be introduced into host cells using any technique known in the art. These techniques include transferrin-polycation-mediated DNA transfer, transfection with naked or encapsulated nucleic acids, liposome-mediated cellular fusion, intracellular transportation of DNA-coated latex beads, protoplast fusion, viral infection, electroporation, "gene gun," and calcium phosphate-mediated transfection.
A OF150/hTAFπ150 expression construct comprises a promoter which is functional in the particular host cell selected. The skilled artisan can readily select an appropriate promoter from the large number of cell type-specific promoters known and used in the art. The expression construct can also contain a transcription terminator which is functional in the host cell. The expression construct comprises a polynucleotide segment which encodes all or a portion of a OF150/hTAFπ150 protein, biologically active variant, fusion protein, antibody, or ribozyme. The polynucleotide segment is located downstream from the promoter. Transcription of the polynucleotide segment initiates at the promoter. The expression construct can be linear or circular and can contain sequences, if desired, for autonomous replication.
CIFl 50/hTAFjjl 50 subgenomic polynucleotides can be propagated in vectors and cell lines using techniques well known in the art. CIFl 50/hTAFjjl 50 subgenomic polynucleotides can be on linear or circular molecules. They can be on autonomously replicating molecules or on molecules without replication sequences. They can be regulated by their own or by other regulatory sequences, as are known in the art.
Expression systems in bacteria include those described in Chang et al, Nature (1978) 275: 615, Goeddel et al, Nature (1979) 281: 544, Goeddel et al, Nucleic Acids Res. (1980) 8: 4057, EP 36,776, U.S. 4,551,433, deBoer et al, Proc. Natl. Acad. Sci. USA (1983) 80: 21-25, and Siebenlist et al, Cell (1980) 20: 269.
Expression systems in yeast include those described in Hinnen et al, Proc. Natl. Acad. Sci. USA (1978) 75: 1929; Ito et al, J. Bacteriol (1983) 755: 163; Kurtz et al, Mol. Cell. Biol. (1986) 6: 142; Kunze et al, J. Basic Microbiol. (1985) 25: 141; Gleeson et al, J. Gen. Microbiol. (1986) 132: 3459, Roggenkamp et al, Mol. Gen. Genet. (1986) 202 :302) Das et al, J. Bacteriol. (1984) 75S: 1165; De Louvencourt et al, J. Bacteriol. (1983) 154: 131, Nan den Berg et al, Bio/Technology (1990) 8: 135; Kunze et al, J. Basic Microbiol. (1985) 25: 141; Cregg et al, Mol. Cell. Biol. (1985) 5: 3376, U.S. 4,837,148, US 4,929,555; Beach and Nurse, Nature (1981) 300: 706; Davidow et al, Curr. Genet. (1985) 10: 380, Gaillardin etal, Curr. Genet. (1985) 10: 49, Ballance et al, Biochem. Biophys. Res. Commun. (1983) 772: 284-289; Tilburn et al, Gene (1983) 26: 205-221, Yelton et al, Proc. Natl. Acad. Sci. USA (1984) S7: 1470-1474, Kelly and Hynes, EMBO J. (1985) 4: 475479; EP 244,234, and WO 91/00357.
Expression of CIFl 50/hTAFjjl 50 subgenomic polynucleotides in insects can be carried out as described in U.S. 4,745,051, Friesen et al (1986) "The Regulation of Baculovirus Gene Expression" in: THE MOLECULAR BIOLOGY OF BACULOVIRUSES (W. Doerfler, ed.), EP 127,839, EP 155,476, and Vlak et al, J. Gen. Virol. (1988) 69: 765- 776, Miller et al, Ann. Rev. Microbiol. (1988) 42: 111, Carbonell et al, Gene (1988) 73: 409, Maeda et al, Nature (1985) 575: 592-594, Lebacq-Nerheyden et al, Mol. Cell. Biol. (1988) 8: 3129; Smith et al, Proc. Natl. Acad. Sci. USA (1985) 82: 8404, Miyajima et al, Gene (1987) 58: 273; and Martin et al, DNA (1988) 7:99. Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts are described in Luckow et al, Bio/Technology (1988) 6: 47-55, Miller et al, in GENETIC ENGINEERING (Setlow, J.K. et al. eds.), Vol. 8 (Plenum Publishing, 1986), pp. 277-279, and Maeda et al, Nature, (1985) 575: 592-594.
Mammalian expression of CIFl 50/hTAFjjl 50 subgenomic polynucleotides can be achieved as described in Dijkema et al, EMBO J. (1985) 4: 761, Gorman et al, Proc. Natl. Acad. Sci. USA (1982b) 79: 6777, Boshart et al, Cell (1985) 41: 521 and U.S. 4,399,216. Other features of mammalian expression can be facilitated as described in Ham and Wallace, Meth. Enz. (1979) 58: 44; Barnes and Sato, Anal. Biochem. (1980) 102: 255, U.S. 4,767,704, US 4,657,866, US 4,927,762, US 4,560,655, WO 90/103430, WO 87/00195, and U.S. RE 30,985. Polynucleotides of the invention can also be used in gene delivery vehicles, for the purpose of delivering a CIF150/hTAFjjl50 mRNA or oligonucleotide (either with the sequence of native CIF150/hTAFjjl50 mRNA or its complement), full-length OF150/hTAFπ150 protein, OF150/hTAFπ150 fusion protein, CIF150/hTAFπ150 polypeptide, or CIF 150/hTAFπl 50-specific ribozyme or single-chain antibody, into a cell preferably a eukaryotic cell. According to the present invention, a gene delivery vehicle can be, for example, naked plasmid DNA, a viral expression vector comprising a CIF150/hTAFjjl50 polynucleotide, or a OF150/hTAFπ150 polynucleotide in conjunction with a liposome or a condensing agent. In one embodiment of the invention, the gene delivery vehicle comprises a promoter and a CIF150/hTAFj l50 polynucleotide. Preferred promoters are tissue- specific promoters and promoters which are activated by cellular proliferation, such as the thymidine kinase and thymidylate synthase promoters. Other preferred promoters include promoters which are activatable by infection with a virus, such as the α- and β- interferon promoters, and promoters which are activatable by a hormone, such as esfrogen. Other promoters which can be used include the Moloney virus LTR, the CMV promoter, and the mouse albumin promoter.
A CIF150/hTAF l50 gene delivery vehicle can comprise viral sequences such as a viral origin of replication or packaging signal. These viral sequences can be selected from viruses such as astrovirus, coronavirus, orthomyxovirus, papovavirus, paramyxovirus, parvovirus, picornavirus, poxvirus, retrovirus, togavirus or adenovirus. In a preferred embodiment, the CIF150/hTAF l50 gene delivery vehicle is a recombinant retroviral vector. Recombinant refroviruses and various uses thereof have been described in numerous references including, for example, Mann et al, Cell 55. 153, 1983, Cane and Mulligan, Proc. Natl Acad. Sci. USA 81 :6349, 1984, Miller et al , Human Gene Therapy 1:5-14, 1990, U.S. Patent Nos. 4,405,712, 4,861,719, and 4,980,289, and PCT Application Nos. WO 89/02,468, WO 89/05,349, and WO 90/02,806. Numerous retroviral gene delivery vehicles can be utilized in the present invention, including for example those described in EP 0,415,731; WO 90/07936; WO 94/03622; WO 93/25698; WO 93/25234; U.S. Patent No. 5,219,740; WO 9311230; WO 9310218; Vile and Hart, Cancer Res. 55:3860-3864, 1993; Vile and Hart, Cancer Res. 53:962-961, 1993; Ram et al, Cancer Res. 55:83-88, 1993; Takamiya et al., J. Neurosci. Res. 55:493-503, 1992; Babn et aL. J. Neurosurg. 79:129-135, 1993; U.S. Patent No. 4,777,127, GB 2,200,651, EP 0,345,242 and WO91/02805.
Particularly preferred retroviruses are derived from retroviruses which include avian leukosis virus (ATCC Nos. VR-535 and VR-247), bovine leukemia virus (NR- 1315), murine leukemia virus (MLN), mink-cell focus-inducing virus (Koch et al, J. Vir. 49:828, 1984; and Olif et al, J. Vir. 48:542, 1983), murine sarcoma virus (ATCC Νos. NR-844, 45010 and 45016), reticuloendotheliosis virus (ATCC Νos VR-994, VR-770 and 45011), Rous sarcoma virus, Mason-Pfizer monkey virus, baboon endogenous virus, endogenous feline retrovirus (e.g., RD114), and mouse or rat gL30 sequences used as a retroviral vector. Particularly preferred strains of MLV from which recombinant retroviruses can be generated include 4070A and 1504A (Hartley and Rowe, J Vir. 19: 19, 1976), Abelson (ATCC No. VR-999), Friend (ATCC No. VR-245), Graffi (Ru et al, J. Vir. 67:4122, 1993; and Yantchev Neoplasma 26:391, 1979), Gross (ATCC No. VR-590), Kirsten (Albino et al, J. Exp. Med. 164:1110, 1986), Harvey sarcoma virus (Manly et al, J. Vir. 52:3540, 1988; and Albino et α/., J Exp. Med. 164:1110, 1986) and Rauscher (ATCC No. VR-998), and Moloney MLV (ATCC No. VR-190). A particularly preferred non-mouse retrovirus is Rous sarcoma virus. Preferred Rous sarcoma viruses include Bratislava (Manly et al, J. Vir. 52:3540, 1988; and Albino et al, J. Exp. Med. 164:1110, 1986), Bryan high titer (e.g., ATCC Nos. VR-334, VR-657, VR-726, VR-659, and VR-728), Bryan standard (ATCC No. VR-140), Carr-Zilber (Adgighitov et al, Neoplasma 27:159, 1980), Engelbreth-Holm (Laurent et al, Biochem Biophys Acta 908:241, 1987), Harris, Prague (e.g., ATCC Nos. VR-772, and 45033), and Schmidt-Ruppin (e.g. ATCC Nos. VR-724, VR-725, VR-354) viruses.
Any of the above retroviruses can be readily utilized in order to assemble or construct retroviral CIFl 50/hTAFjjl 50 gene delivery vehicles given the disclosure provided herein and standard recombinant techniques (e.g., Sambrook et al, MOLECULAR CLONING: A LABORATORY MANUAL, 2d ed., 1989, and Kunkle, Proc. Natl. Acad. Sci. U.S.A. £2:488, 1985) known in the art. Portions of retroviral CIF150/hTAFjjl50 expression vectors can be derived from different retroviruses. For example, retrovector LTRs can be derived from a murine sarcoma virus, a tRNA binding site from a Rous sarcoma virus, a packaging signal from a murine leukemia virus, and an origin of second strand synthesis from an avian leukosis virus. These recombinant retroviral vectors can be used to generate fransduction competent retroviral vector particles by introducing them into appropriate packaging cell lines (see Serial No. 07/800,921, filed November 29, 1991). Recombinant retroviruses can be produced which direct the site-specific integration of the recombinant retroviral genome into specific regions of the host cell DNA. Such site-specific integration can be mediated by a chimeric integrase incorporated into the retroviral particle (see Serial No. 08/445,466 filed May 22, 1995). It is preferable that the recombinant viral gene delivery vehicle is a replication-defective recombinant virus. Packaging cell lines suitable for use with the above-described retroviral gene delivery vehicles can be readily prepared (see Serial No. 08/240,030, filed May 9, 1994; see also WO 92/05266) and used to create producer cell lines (also termed vector cell lines or "VCLs") for production of recombinant viral particles. In particularly preferred embodiments of the present invention, packaging cell lines are made from human (e.g., HT1080 cells) or mink parent cell lines, thereby allowing production of recombinant retroviral gene delivery vehicles which are capable of surviving inactivation in human serum. The construction of recombinant retroviral gene delivery vehicles is described in detail in WO 91/02805. These recombinant retroviral gene delivery vehicles can be used to generate fransduction competent retroviral particles by introducing them into appropriate packaging cell lines (see Serial No. 07/800,921). Similarly, adenovirus gene delivery vehicles can also be readily prepared and utilized given the disclosure provided herein (see also Berkner, Biotechniques 6:616-621, 1988, and Rosenfeld et al, Science 252:431-434, 1991, WO 93/07283, WO 93/06223, and WO 93/07282). A CIF150/hTAFjjl50 gene delivery vehicle can also be a recombinant adenoviral gene delivery vehicle. Such vehicles can be readily prepared and utilized given the disclosure provided herein (see Berkner, Biotechniques 6:616, 1988, and Rosenfeld et al, Science 252:431, 1991, WO 93/07283, WO 93/06223, and WO 93/07282). Adeno-associated viral CIFl 50/hTAFj l 50 gene delivery vehicles can also be constructed and used to deliver CIF150/hTAFπ150 amino acids or nucleotides. The use of adeno-associated viral gene delivery vehicles in vitro is described in Chatteriee et al, Science 258: 1485-1488 (1992), Walsh et al, Proc. Natl Acad. Sci. 89: 7257-7261 (1992), Walsh et al, J. Clin. Invest. 94: 1440-1448 (1994), Flotte et al, J. Biol. Chem. 268: 3781-3790 (1993), Ponnazhagan et al, J. Exp. Med. 179: 733-738 (1994), Miller et al, Proc. Natl Acad. Sci. 91: 10183-10187 (1994), Einerhand et al, Gene Ther. 2: 336- 343 (1995), Luo et al, Exp. Hematol. 23: 1261-1267 (1995), and Zhou et al, Gene Therapy 3: 223-229 (1996). In vivo use of these vehicles is described in Flotte et al, Proc. Nat'lAcad. Sci. 90: 10613-10617 (1993), and Kaplitt et al, Nature Genet. 5:148- 153 (1994).
In another embodiment of the invention, a CIFl 50/hTAFjjl 50 gene delivery vehicle is derived from a togavirus. Preferred togaviruses include alphaviruses, in particular those described in U.S. Serial No. 08/405,627, filed March 15, 1995, WO 95/07994. Alpha viruses, including Sindbis and ELVS viruses can be gene delivery vehicles for CIFl 50/hTAFjjl 50 polynucleotides. Alpha viruses are described in WO 94/21792, WO 92/10578 and WO 95/07994. Several different alphavirus gene delivery vehicle systems can be constructed and used to deliver CIF150/hTAFjjl50 polynucleotides to a cell according to the present invention. Representative examples of such systems include those described in U.S. Patents 5,091,309 and 5,217,879. Particularly preferred alphavirus gene delivery vehicles for use in the present invention include those which are described in WO 95/07994, and U.S. Serial No. 08/405,627.
Preferably, the recombinant viral vehicle is a recombinant alphavirus viral vehicle based on a Sindbis virus. Sindbis constructs, as well as numerous similar constructs, can be readily prepared essentially as described in U.S. Serial No. 08/198,450. Sindbis viral gene delivery vehicles typically comprise a 5' sequence capable of initiating Sindbis virus transcription, a nucleotide sequence encoding Sindbis non-structural proteins, a viral junction region inactivated so as to prevent fragment transcription, and a Sindbis RNA polymerase recognition sequence. Optionally, the viral junction region can be modified so that polynucleotide transcription is reduced, increased, or maintained. As will be appreciated by those in the art, corresponding regions from other alphaviruses can be used in place of those described above.
The viral junction region of an alphavirus-derived gene delivery vehicle can comprise a first viral junction region which has been inactivated in order to prevent transcription of the polynucleotide and a second viral junction region which has been modified such that polynucleotide transcription is reduced. An alphavirus-derived vehicle can also include a 5' promoter capable of initiating synthesis of viral RNA from cDNA and a 3' sequence which controls transcription termination.
Other recombinant togaviral gene delivery vehicles which can be utilized in the present invention include those derived from Semliki Forest virus (ATCC VR-67; ATCC VR-1247), Middleberg virus (ATCC VR-370), Ross River virus (ATCC VR-373; ATCC VR-1246), Venezuelan equine encephalitis virus (ATCC VR923; ATCC VR-1250; ATCC VR-1249; ATCC VR-532), and those described in U.S. Patents 5,091,309 and 5,217,879 and in WO 92/10578. The Sindbis vehicles described above, as well as numerous similar constructs, can be readily prepared essentially as described in U.S. Serial No. 08/198,450.
Other viral gene delivery vehicles suitable for use in the present invention include, for example, those derived from poliovirus (Evans et al, Nature 559:385, 1989, and Sabin et al, J. Biol. Standardization 7:115, 1973) (ATCC VR-58); rhino virus (Arnold et al, J. Cell. Biochem. L401, 1990) (ATCC VR-1110); pox viruses, such as canary pox virus or vaccinia virus (Fisher-Hoch et al, Proc. Natl. Acad. Sci. U.S.A. 86:317, 1989; Flexneτ et al, Ann. NY. Acad. Sci. 559:86, 1989; Flexner et al. , Vaccine 8:11, 1990; U.S. 4,603,112 and U.S. 4,769,330; WO 89/01973) (ATCC VR-111; ATCC VR-2010); SV40 (Mulligan et al, Nature 277. 108, 1979) (ATCC VR-305), (Madzak et al, J. Gen. Vir. 75:1533, 1992); influenza virus (Luytjes et al, Cell 59:1107, 1989; McMicheal et al, The New England Journal of Medicine 309:13, 1983; and Yap et al, Nature 275:238, 1978) (ATCC VR-797); parvovirus such as adeno-associated virus (Samulski et α/., J Vir. 55:3822, 1989, and Mendelson et α/., Virology 166:154, 1988) (ATCC VR-645); herpes simplex virus (Kit et al. , Adv. Exp. Med. Biol. 215:219, 1989) (ATCC VR-977; ATCC VR-260); Nature 277: 108, 1979); human immunodeficiency virus (EPO 386,882, Buchschacher et al, J. Vir. 55:2731, 1992); measles virus (EPO 440,219) (ATCC NR-24); A (ATCC NR-67; ATCC NR-1247), Aura (ATCC VR-368), Bebaru virus (ATCC VR-600; ATCC VR-1240), Cabassou (ATCC VR-922), Chikungunya virus (ATCC VR-64; ATCC VR-1241), Fort Morgan (ATCC VR-924), Getah virus (ATCC VR-369; ATCC VR-1243), Kyzylagach (ATCC VR-927), Mayaro (ATCC VR-66), Mucambo virus (ATCC VR-580; ATCC VR-1244), Νdumu (ATCC VR-371), Pixuna virus (ATCC VR-372; ATCC VR-1245), Tonate (ATCC VR-925), Triniti (ATCC VR-469), Una (ATCC VR-374), Whataroa (ATCC VR-926), Y-62-33 (ATCC VR-375), O*Νyong virus, Eastern encephalitis virus (ATCC VR-65; ATCC VR- 1242), Western encephalitis virus (ATCC VR-70; ATCC VR-1251; ATCC VR-622; ATCC VR-1252), and coronavirus (Hamre et al, Proc. Soc. Exp. Biol. Med. 727:190, 1966) (ATCC VR-740).
A CIFl 50/hTAFjjl 50 polynucleotide of the invention can also be combined with a condensing agent to form a gene delivery vehicle. In a preferred embodiment, the condensing agent is a polycation, such as polylysine, polyarginine, polyornithine, protamine, spermine, spermidine, and putrescine. Many suitable methods for making such linkages are known in the art (see, for example, Serial No. 08/366,787, filed December 30, 1994). In an alternative embodiment, a CIFl 50/hTAFjjl 50 polynucleotide is associated with a liposome to form a gene delivery vehicle. Liposomes are small, lipid vesicles comprised of an aqueous compartment enclosed by a lipid bilayer, typically spherical or slightly elongated structures several hundred Angstroms in diameter. Under appropriate conditions, a liposome can fuse with the plasma membrane of a cell or with the membrane of an endocytic vesicle within a cell which has internalized the liposome, thereby releasing its contents into the cytoplasm. Prior to interaction with the surface of a cell, however, the liposome membrane acts as a relatively impermeable barrier which sequesters and protects its contents, for example, from degradative enzymes. Additionally, because a liposome is a synthetic structure, specially designed liposomes can be produced which incorporate desirable features. See Stryer, Biochemistry, pp. 236- 240, 1975 (W.H. Freeman, San Francisco, CA); Szoka et al, Biochim. Biophys. Acta 600:1, 1980; Bayer et al, Biochim. Biophys. Acta. 550:464, 1979; Rivnay et al, Meth. Enzymol 149:119, 1987; Wang etal, Proc. Natl. Acad. Sci. U.S.A. 84: 7851, 1987, Plant et al, Anal Biochem. 775:420, 1989, and U.S. Patent 4,762,915. Liposomes can encapsulate a variety of nucleic acid molecules including DNA, RNA, plasmids, and expression constructs comprising CIFl 50/hTAFjjl 50 polynucleotides such those disclosed in the present invention.
Liposomal preparations for use in the present invention include cationic (positively charged), anionic (negatively charged) and neufral preparations. Cationic liposomes have been shown to mediate intracellular delivery of plasmid DNA (Feigner et al, Proc. Nat Acad. Sci. USA 84:1413-1416, 1987), mRNA (Malone et al, Proc. Natl. Acad. Sci. USA 86:6011-6081, 1989), and purified transcription factors (Debs et al, J. Biol. Chem. 255:10189-10192, 1990), in functional form. Cationic liposomes are readily available. For example, N[l-2,3-dioleyloxy)propyl]-N,N,N-triethylammonium
(DOTMA) liposomes are available under the trademark Lipofectin, from GIBCO BRL, Grand Island, NY. See also Feigner et al, Proc. Natl. Acad. Sci. USA 91: 5148-5152.87, 1994. Other commercially available liposomes include Transfectace (DDAB/DOPE) and DOTAP/DOPE (Boerhinger). Other cationic liposomes can be prepared from readily available materials using techniques well known in the art. See, e.g. , Szoka et al. , Proc. Natl Acad. Sci. USA 75:4194-4198, 1978; and WO 90/11092 for descriptions of the synthesis of DOTAP (l,2-bis(oleoyloxy)-3-(frimethylammonio)propane) liposomes. Similarly, anionic and neutral liposomes are readily available, such as from Avanti Polar Lipids (Birmingham, AL), or can be easily prepared using readily available materials. Such materials include phosphatidyl choline, cholesterol, phosphatidyl ethanolamine, dioleoylphosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol (DOPG), dioleoylphoshatidyl ethanolamine (DOPE), among others. These materials can also be mixed with the DOTMA and DOTAP starting materials in appropriate ratios. Methods for making liposomes using these materials are well known in the art.
The liposomes can comprise multilammelar vesicles (MLVs), small unilamellar vesicles (SUVs), or large unilamellar vesicles (LUVs). The various liposome-nucleic acid complexes are prepared using methods known in the art. See, e.g., Straubinger et al, METHODS OF IMMUNOLOGY (1983), Vol. 101, pp. 512-527; Szoka et al, Proc. Natl. Acad. Sci. USA 57:3410-3414, 1990; Papahadjopoulos et al, Biochim. Biophys. Acta 394:483, 1975; Wilson et al, Cell 17:11, 1979; Deamer and Bangham, Biochim. Biophys. Acta 443:629, 1976; Osfro et al, Biochem. Biophys. Res. Commun. 75:836, 1977; Fraley et al, Proc. Natl. Acad. Sci. USA 75:3348, 1979; Enoch and Strittaiatter, Proc. Natl. Acad. Sci. USA 76:145, 1979; Fraley et al, J. Biol. Chem. 255:10431, 1980; Szoka and Papahadjopoulos, Proc. Natl. Acad. Sci. USA 75:145, 1979; and Schaefer- Ridder et al, Science 275:166, 1982.
In addition, lipoproteins can be included with a CIFl 50/hTAFjjl 50 polynucleotide for delivery to a cell. Examples of such lipoproteins include chylomicrons, HDL, IDL, LDL, and NLDL. Mutants, fragments, or fusions of these proteins can also be used. Modifications of naturally occurring lipoproteins can also be used, such as acetylated LDL. These lipoproteins can target the delivery of polynucleotides to cells expressing lipoprotein receptors. Preferably, if lipoproteins are included with a polynucleotide, no other targeting ligand is included in the composition.
In another embodiment, naked CIF150/hTAFj l50 polynucleotide molecules are used as gene delivery vehicles, as described in WO 90/11092 and U.S. Patent 5,580,859. Such gene delivery vehicles can be either CIFl 50/hTAFj l 50 DΝA or RΝA and, in certain embodiments, are linked to killed adenovirus. Curiel et al, Hum. Gene. Ther. 5:147-154, 1992. Other suitable vehicles include DΝA-ligand (Wu et al, J. Biol. Chem. 264:16985-16981, 1989), lipid-DΝA combinations (Feigner et al, Proc. Natl. Acad. Sci. USA 84:1413 7417, 1989), liposomes (Wang et al, Proc. Natl. Acad. Sci. 84:1851-1855, 1987) and microprojectiles (Williams et al, Proc. Natl. Acad. Sci. 88:2126-2130, 1991).
One can increase the efficiency of naked CIF150/hTAFjjl50 polynucleotide uptake into cells by coating the polynucleotides onto biodegradable latex beads. This approach takes advantage of the observation that latex beads, when incubated with cells in culture, are efficiently transported and concentrated in the perinuclear region of the cells. The beads will then be transported into cells when injected into muscle. CIFl 50/hTAF jl 50 polynucleotide-coated latex beads will be efficiently transported into cells after endocytosis is initiated by the latex beads and thus increase gene transfer and expression efficiency. This method can be improved further by freating the beads to increase their hydrophobicity, thereby facilitating the disruption of the endosome and release of CIF150/hTAFjjl50 polynucleotides into the cytoplasm.
Expression of OF150/hTAFπ150 protein and activity are cell cycle regulated. Thus, the invention provides compositions which can be used to alter spatial or temporal patterns of division of a human cell. Depletion of OF150/hTAFπ150 activity leads to a G2 (late S phase) arrest of the cell cycle, indicating that OF150/hTAFπ150 function is required to transcribe genes necessary for cell cycle progression. Thus, mitosis or cell cycle progression can be reduced or prevented by decreasing expression of a human CIFl 50/hTAF jl 50 gene. Decreased CIFl 50/hTAFjjl 50 gene expression can be used to freat conditions characterized by high rates of mitosis, such as neoplasia, metastasis of neoplasms, benign proliferative diseases, and dysplastic and hyperplastic disorders. Cells in which CIF150/hTAFjjl50 expression has been decreased can also be used to identify genes whose expression is dependent on a OF150/hTAFπ150 protein. Conversely, increased CIF150/hTAFjjl50 expression can be used to expand cell populations in vitro or for freating disorders such as anemia, which are characterized by lowered rates of mitosis.
In one embodiment of the invention, expression of the CIF150/hTAFj l50 gene is decreased using a ribozyme, an RNA molecule with catalytic activity. See, e.g., Cech, 1987, Science 236: 1532-1539; Cech, 1990, Ann. Rev. Biochem. 59:543-568; Cech, 1992, Curr. Opin. Struct. Biol. 2: 605-609; Couture and Stinchcomb, 1996, Trends Genet. 12: 510-515. Ribozymes can be used to inhibit gene function by cleaving an RNA sequence, as is known in the art (e.g., Haseloff et al, U.S. 5,641,673).
The coding sequence of the CIF150/hTAF jl50 gene can be used to generate ribozymes which will specifically bind to mRNA franscribed from a CIF150/hTAFjjl50 gene. Methods of designing and constructing ribozymes which can cleave other RNA molecules in trans in a highly sequence specific manner have been developed and described in the art (see Haseloff et al, Nature 334:585-591, 1988). For example, the cleavage activity of ribozymes can be targeted to specific CIF150/hTAF jl50 RNAs by engineering a discrete "hybridization" region into the ribozyme. The hybridization region contains a sequence complementary to the target CIF150/hTAF jl50 RNA and thus specifically hybridizes with the target (see, for example, Gerlach et al, EP 321,201). The nucleotide sequence shown in SEQ ID NO:l provides a source of suitable hybridization region sequences. Longer complementary sequences can be used to increase the affinity of the hybridization sequence for the target. The hybridizing and cleavage regions of the CIF150/hTAF jl50 ribozyme can be integrally related; thus, upon hybridizing to the target CIFl 50/hTAFjjl 50 RNA through the complementary regions, the catalytic region of the ribozyme can cleave the target.
CIF150/hTAFjjl50 ribozymes can be introduced into cells, such as neoplastic cells, as part of a DNA construct, as is known in the art and described above. Mechanical methods, such as microinjection, liposome-mediated transfection, elecfroporation, or calcium phosphate precipitation, can be used to introduce the ribozyme-containing DNA construct into cells in which it is desired to decrease CIF150/hTAF jl50 expression. Alternatively, if it is desired that the cells stably retain the DNA construct, it can be supplied on a plasmid and maintained as a separate element or integrated into the genome of the cells, as is known in the art. The DNA construct can include transcriptional regulatory elements, such as a promoter element, an enhancer or UAS element, and a transcriptional terminator signal, for controlling transcription of CIF150/hTAFj l50 ribozymes in the cells. As taught in Haseloff et al, U.S. 5,641,673, CIF150/hTAFjjl50 ribozymes can be engineered so that ribozyme expression will occur in response to factors which induce expression of the CIF150/hTAF jl50 gene. Ribozymes can also be engineered to provide an additional level of regulation, so that destruction of CIFl 50/hTAFjjl 50 mRNA occurs only when both a CIF150/hTAFjjl50 ribozyme and a CIF150/hTAFjιl50 gene are induced in the cells.
In another embodiment of the invention, expression of the CIF150/hTAFjjl50 gene is altered using an antisense oligonucleotide sequence. The antisense sequence is complementary to at least a portion of the coding sequence of a CIF150/hTAFjjl50 gene having the nucleotide sequence shown in SEQ ID NO: 1. Preferably, the antisense oligonucleotide sequence is at least six nucleotides in length, but can be about 8, 12, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides long. Longer sequences can also be used. CIF150/hTAFjjl50 antisense oligonucleotide molecules can be provided in a DNA construct and introduced into cells as described above, to decrease CIF150/hTAFjjl50 expression.
Cells treated with an antisense oligonucleotide in order to interfere with expression of CIF150/hTAFjjl50 arrest in the cell cycle in the S/G2 phase. This result indicates that CIF150/hTAFjjl50 function is important for cells to enter mitosis. The antisense oligonucleotide 5' TGCTCATGGA AGCATAAGCA GCCAC 3* (SEQ ID NO:5) reduces CIF150/hTAFjjl50 mRNA levels in HeLa and IMR 90 cells (see
Example 1). The treated cells do not enter mitosis and accumulate in G2 phase. One of skill in the art can test other oligonucleotides for antisense effect, by delivering the antisense oligonucleotide to the cell using transfection or any means known in the art and detecting CIF150/hTAF jl50 mRNA levels. CIF150/hTAFjjl50 antisense oligonucleotides can be deoxyribonucleotides, ribonucleotides, or a combination of both. Oligonucleotides can be synthesized manually or by an automated synthesizer, by covalently linking the 5' end of one nucleotide with the 3' end of another nucleotide with non-phosphodiester internucleotide linkages such alkylphosphonates, phosphorothioates, phosphorodithioates, alkylphosphonothioates, alkylphosphonates, phosphoramidates, phosphate esters, carbamates, acetamidate, carboxymethyl esters, carbonates, and phosphate triesters. See Brown, 1994, Meth. Mol. Biol 20:1-8; Sonveaux, 1994, Meth. Mol. Biol. 25:1-72; Uhlmann et al, 1990, Chem. Rev. 90:543-583. Precise complementarity is not required for successful duplex formation between an antisense molecule and the complementary coding sequence of a CIF150/hTAF jl50 gene. Antisense molecules which comprise, for example, 2, 3, 4, or 5 or more stretches of contiguous nucleotides which are precisely complementary to a CIF150/hTAF jl50 coding sequence, each separated by a stretch of contiguous nucleotides which are not complementary to adjacent CIF150/hTAFj/150 coding sequences, can provide targeting specificity for CIF150/hTAFjjl50 mRNA. Preferably, each stretch of contiguous nucleotides is at least 4, 5, 6, 7, or 8 or more nucleotides in length. Non-complementary intervening sequences are preferably 1, 2, 3, or 4 nucleotides in length. One skilled in the art can easily use the calculated melting point of an antisense-sense pair to determine the degree of mismatching which will be tolerated between a particular antisense oligonucleotide and a particular CIFl 30 coding sequence.
CIFl 50/hTAFjjl 50 antisense oligonucleotides can be modified without affecting their ability to hybridize to a CIF150/hTAF jl50 coding sequence. These modifications can be internal or at one or both ends of the antisense molecule. For example, intemucleoside phosphate linkages can be modified by adding cholesteryl or diamine moieties with varying numbers of carbon residues between the amino groups and terminal ribose. Modified bases and/or sugars, such as arabinose instead of ribose, or a 3', 5'-substituted oligonucleotide in which the 3' hydroxyl group or the 5' phosphate group are substituted, can also be employed in a modified antisense oligonucleotide. These modified oligonucleotides can be prepared by methods well known in the art. See, e.g., Agrawal et α/., 1992, Trends Biotechnol 70:152-158; Uhlmann et al, 1990, Chem. Rev. 90:543-584; Uhlmann et al, 1987, Tetrahedron. Lett. 275:3539-3542.
Antibodies which specifically bind to a OF150/hTAFπ150 protein, particularly single-chain antibodies, can also be used to alter CIF150/hTAFjjl50 gene expression. CIF150/hTAFπ150-specific antibodies bind to OF150/hTAFπ150 protein and prevent the protein from functioning in the cell. Polynucleotides encoding antibodies which specifically bind to OF150/hTAFπ150 can be introduced into cells as described above. Preferably, the mechanism used to decrease expression of the CIF150/hTAFjjl50 gene, whether ribozyme, antisense nucleotide sequence, or antibody, decreases expression of the CIF150/hTAF jl50 gene by 50%, 60%, 70%, or 80%. Most preferably, expression of the CIF150/hTAFjjl50 gene is decreased by 90%, 95%, 99%, or 100%. The effectiveness of the mechanism chosen to alter expression of the CIF150/hTAFjjl50 gene can be assessed using methods well known in the art, such as hybridization of nucleotide probes to CIF150/hTAFjjl50 mRNA, quantitative RT-PCR, or detection of OF150/hTAFπ150 protein using CIF150/hTAFπ150-specific antibodies. Compositions comprising OF150/hTAFπ150 antibodies, ribozymes, or antisense oligonucleotides can be used to treat proliferative disorders, such as neoplasias, dysplasias, and hyperplasias, and their symptoms. Neoplasias which can be treated with a OF150/hTAFπ150 composition include, but are not limited to, melanomas, squamous cell carcinomas, adenocarcinomas, hepatocellular carcinomas, renal cell carcinomas, sarcomas, myosarcomas, non-small cell lung carcinomas, leukemias, lymphomas, osteosarcomas, central nervous system tumors such as gliomas, astrocytomas, oligodendrogliomas, and neuroblastomas, tumors of mixed origin, such as Wilms* tumor and teratocarcinomas, and metastatic tumors. Proliferative disorders which can be treated with a OF150/hTAFπ150 composition include disorders such as anhydric hereditary ectodermal dysplasia, congenital alveolar dysplasia, epithelial dysplasia of the cervix, fibrous dysplasia of bone, and mammary dysplasia. Hyperplasias, for example, endometrial, adrenal, breast, prostate, or thyroid hyperplasias, or pseudoepitheliomatous hyperplasia of the skin can be treated with CIFl 50/hTAFπl 50 compositions.
Even in disorders in which CIF150/hTAFjjl50 mutations are not implicated, decreasing OF150/hTAFπ150 function can have therapeutic application. In these disorders, decreasing CIF150/hTAF jl50 expression or function can help to suppress tumors. Similarly, in tumors in which CIF150/hTAFjjl50 expression is not aberrant, effecting CIF150/hTAF ιl50 downregulation or decrease of OF150/hTAFπ150 activity can suppress metastases.
OF150/hTAFπ150 compositions of the invention can comprise a pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers are well known to those in the art. Such carriers include, but are not limited to, large, slowly metabolized macromolecules, such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, and inactive virus particles. Pharmaceutically acceptable salts can also be used in OF150/hTAFπ150 compositions, for example, mineral salts such as hydrochlorides, hydrobromides, phosphates, or sulfates, as well as salts of organic acids such as acetates, proprionates, malonates, or benzoates. OF150/hTAFπ150 compositions can also contain liquids, such as water, saline, glycerol, and ethanol, as well as substances such as wetting agents, emulsifying agents, or pH buffering agents. Liposomes, such as those described in U.S. 5,422,120, WO 95/13796, WO 91/14445, or EP 524,968 Bl, can also be used as a carrier for a CIF 150/hTAFπl 50 composition.
Typically, a OF150/hTAFπ150 composition is prepared as an injectable, either as a liquid solution or suspension; however, solid forms suitable for solution or suspension in liquid vehicles prior to injection can also be prepared. A CIF150/hTAFπ150 composition can also be formulated into an enteric coated tablet or gel capsule according to known methods in the art, such as those described in U.S. 4,853,230, EP 225, 189, AU 9,224,296, and AU 9,230,801.
Administration of OF150/hTAFπ150 compositions of the invention can include local or systemic administration, including injection, oral administration, particle gun, or catheterized administration, and topical administration. Various methods can be used to administer a CIF 150/hTAFπl 50 composition directly to a specific site in the body. For example, a small metastatic lesion can be located and an appropriate OF150/hTAFπ150 composition injected several times in several different locations within the body of the lesion. Alternatively, arteries which serve a tumor can be identified, and a CIF150/hTAFπ150 composition can be injected into such an artery, in order to deliver the composition directly into the tumor.
A tumor which has a necrotic center can be aspirated, and a OF150/hTAFπ150 composition can be injected directly into the now empty center of the tumor. A CIFl 50/hTAFπl 50 composition can also be administered directly to the surface of a tumor, for example, by topical application of the composition. X-ray imaging can be used to assist in certain of these delivery methods. Combination therapeutic agents, including reagents which specifically bind to a wild-type human CIFl 50/hTAFjf 150 gene or expression product, together with other therapeutic agents, can be administered simultaneously or sequentially.
OF150/hTAFπ150 compositions can be delivered to specific tissues using receptor-mediated targeted delivery. Receptor-mediated DNA delivery techniques are described in, for example, Findeis et al. (1993), Trends in Biotechnol 11, 202-05; Chiou et al. (1994), GENE THERAPEUTICS: METHODS AND APPLICATIONS OF DIRECT GENE TRANSFER (J.A. Wolff, ed.); Wu & Wu (1988), J. Biol. Chem. 263, 621-24; Wu et al.
(1994), J. Biol. Chem. 269, 542-46; Zenke et al. (1990), Proc. Natl Acad. Sci. U.S.A. 87, 3655-59; Wu et al (1991), J. Biol. Chem. 266, 338-42.
Alternatively, a OF150/hTAFπ150 composition can be introduced into human cells ex vivo, and the cells then replaced into the human. Cells can be removed from a variety of locations including, for example, from a selected tumor or from an affected organ. A OF150/hTAFπ150 composition can be inserted into non-tumorigenic cells, such as dermal fibroblasts or peripheral blood leukocytes. If desired, particular fractions of cells, such as T cell subsets or stem cells, can also be specifically removed from the blood (see, for example, PCT WO 91/16116). The removed cells can then be contacted with a OF150/hTAFπ150 composition utilizing any of the above-described techniques, followed by the return of the cells to the human.
In another embodiment of the invention, mitosis or cell cycle progression can be induced, increased, or promoted by increasing expression of a CIF150/hTAFjjl50 gene. Increasing CIFl 50/hTAFjjl 50 gene expression is useful, for example, for increasing the number of cells in a cell population in vitro or for freating disorders characterized by lowered rates of mitosis or cell cycle progression, such as anemia.
A composition comprising all or a portion of a CIF150/hTAFjjl50 gene or expression product can be introduced into a cell. The entire CIFl 50/hTAF jl 50 coding sequence or protein can be introduced, as described above. Alternatively, a portion of a CIF150/hTAF jl50 protein which promotes mitosis or cell cycle progression can be identified and that portion or a nucleotide sequence encoding it can be introduced into the cell. Portions of a OF150/hTAFπ150 protein which promote mitosis or cell cycle progression can be identified by introducing expression constructs which express different portions of the protein into cells and measuring alterations in the rate of mitosis or cell cycle progression. Rates of mitosis can be measured, for example, by detecting incorporation of labeled nucleotides, as is known in the art.
CIF150/hTAFπ150 compositions which contain CIF150/hTAFjjl50 subgenomic polynucleotides preferably contain an expression construct comprising a promoter and a polynucleotide segment encoding at least six contiguous amino acids of a
OF150/hTAFπ150 protein or protein variant. Within the expression construct, the polynucleotide segment is located downstream from the promoter, and transcription of the polynucleotide segment initiates at the promoter. A more complete description of gene transfer vectors, especially retroviral vectors, is contained in U.S. Serial No. 08/869,309, which is incorporated herein by reference.
Both the dose of a particular OF150/hTAFπ150 composition and the means of administering the composition can be determined based on specific qualities of the therapeutic composition, the condition, age, and weight of the patient, the progression of the particular disease being treated, and other relevant factors. If the composition contains CIF 150/hTAFπl 50 proteins, polypeptides, or antibodies, effective dosages of the composition are in the range of about 5 μg to about 50 μg/kg of patient body weight, about 50 μg to about 5 mg/kg, about 100 μg to about 500 μg/kg of patient body weight, and about 200 to about 250 μg/kg.
Compositions containing CIF150/hTAFjjl50 subgenomic polynucleotides, including antisense oligonucleotides and ribozyme- or antibody-encoding sequences, can be administered in a range of about 100 ng to about 200 mg of DNA for local administration. Suitable concentrations range from about 500 ng to about 50 mg, about 1 μg to about 2 mg, about 5 μg to about 500 μg, and about 20 μg to about 100 μg of DNA. Factors such as method of action and efficacy of transformation and expression are considerations which will affect the dosage required for ultimate efficacy of the OF150/hTAFπ150 composition. If greater expression is desired over a larger area of tissue, larger amounts of OF150/hTAFπ150 compositions or the same amount administered successively, or several administrations to different adjacent or close tissue portions of, for example, a tumor site, may be required to effect a positive therapeutic outcome. In all cases, routine experimentation in clinical trials will determine specific ranges for optimal therapeutic effect.
Expression of an endogenous CIF150/hTAFjjl50 gene in a cell can be altered by introducing in frame with the endogenous CIF150/hTAFjjl50 gene a DNA construct comprising a CIF150/hTAFjjl50 targeting sequence, a regulatory sequence, an exon, and an unpaired splice donor site by homologous recombination, such that a homologously recombinant cell comprising a new OF150/hTAFπ150 franscription unit. The new transcription unit can be used to turn the CIF150/hTAF jl50 gene on or off as desired. This method of affecting endogenous gene expression is taught in U.S. Patent No. 5,641,670.
The targeting sequence is a segment of at least 10, 12, 15, 20, or 50 contiguous nucleotides selected from the nucleotide sequence shown in SEQ ID NO: 1. The franscription unit is located upstream of a coding sequence of the endogenous CIF150/hTAFjjl50 gene. The exogenous regulatory sequence directs transcription of the coding sequence of the CIF150/hTAFjjl50 gene.
OF150/hTAFπ150 and its binding element 5'-Py X G A G A/C A/Py -3' (SEQ ID NO: 7) can be used to increase expression of a gene, either in vitro, such as in a cell culture, or in vivo. The OF150/hTAFπ150 binding element can be inserted into the promoter region of a gene whose increased expression is desired. The promoter region is preferably a TATA-less promoter region. Any gene of interest can be placed under control of a TATA-less promoter region comprising a OF150/hTAFπ150 binding element using standard recombinant DNA techniques.
The promoter region of the gene is contacted with a OF150/hTAFπ150 protein. The OF150/hTAFπ150 protein can be provided to the promoter region, for example, by means of an expression construct which encodes OF150/hTAFπ150. Upon binding of OF150/hTAFπ150 to the OF150/hTAFπ150 binding element of the promoter region, expression of the desired gene is increased. The OF150/hTAFπ150 coding sequence can be placed under control of a cell-type specific or inducible promoter, as is known in the art.
The present invention also provides a method of diagnosing or prognosing neoplasia or of identifying neoplastic tissue of a human. Expression of a human CIFl 50/hTAF jl 50 gene can be compared between a first tissue which is suspected of being neoplastic and a second tissue which is normal. The first and second tissues can be obtained from the same human or from different humans. The normal tissue can be any tissue of the human, including, but not limited to, spleen, thymus, prostate, testis, ovary, small intestine, mucosal lining of the colon, peripheral blood leukocytes, heart, glial cells, placenta, lung, liver, skeletal muscle, kidney, pancreas, peripheral blood leukocytes, bone marrow, and appendix. The tissue suspected of being neoplastic can be derived from a different tissue type of the human, but preferably it is derived from the same tissue type, for example an intestinal polyp or other abnormal growth.
Overexpression of the CIF150/hTAFjjl50 gene in the suspect tissue identifies the suspect tissue as neoplastic. Expression of a CIF150/hTAFjjl50 gene can be detected by measuring CIF150/hTAF jl50 mRNA. PolyA+ RNA can be isolated from the two tissues as is known in the art. One of skill in the art can readily determine differences in the size or amount of CIFl 50/hTAF l 50 mRNA transcripts between the two tissues that are compared, using Northern blots or in situ hybridization with nucleotide probes selected from the nucleotide sequence shown in SEQ ID NO: 1. Overexpression of CIF150/hTAFjjl50 mRNA in a tissue sample suspected of being neoplastic compared with the expression of CIFl 50/hTAFjjl 50 mRNA in a normal tissue is indicative of neoplasia.
Alternatively, CIF150/hTAFπ150 proteins can be compared between the two tissue samples. Any method for analyzing proteins can be used to compare two OF150/hTAFπ150 proteins from matched samples. The sizes of the OF150/hTAFπ150 proteins in the two tissues can be compared, for example, using antibodies to detect OF150/hTAFπ150 proteins in Western blots of protein extracts from the two tissues. Other changes, such as expression levels and subcellular localization, can also be detected immunologically. A higher OF150/hTAFπ150 protein expression level in a tissue suspected of being neoplastic compared with the CIF 150/hTAFπ 150 protein expression level in a normal tissue is indicative of neoplasia.
Similarly, comparison of CIFl 50/hTAFjjl 50 gene sequences or of CIFl 50/hTAFjf 150 gene expression products, e.g., mRNA and protein, between a tissue of a human which is suspected of being neoplastic and a normal tissue of a human can be used to diagnose or prognose neoplasia in the human. The CIFl 50/hTAFjjl 50 genes in the two tissues can be compared by any means known in the art. For example, the two genes can be sequenced, and the sequence of the CIFl 50/hTAFjjl 50 gene in the tissue suspected of being neoplastic can be compared with the wild-type CIFl 50/hTAFjjl 50 sequence in the normal tissue. The CIFl 50/hTAF l 50 genes or portions of the C7E750/hTAFj l 50 genes in the two tissues can be amplified, for example, using nucleotide primers selected from the nucleotide sequence shown in SEQ ID NO:l in the polymerase chain reaction (PCR) or other amplification technique. The amplified genes or portions of genes can be hybridized to nucleotide probes selected from the nucleotide sequence shown in SEQ ID NO: 1. The nucleotide probes can be labeled by a variety of methods, such as radiolabeling, biotinylation, or coupling to fluorescent or chemiluminescent tags, and detected by standard methods known in the art. Comparisons of CIFl 50/hTAF jl 50 genes, mRNA, or protein can be made as described above. A difference between the CIFl 50/hTAFjjl 50 genes (or a gene which regulates, for example, the expression, half-life, or degradation of CIF150/hTAFjjl50 mRNA) in the two tissues which are compared indicates neoplasia in the suspect tissue. The degree of overexpression of the CIFl 50/hTAF/f 150 gene in the neoplastic tissue relative to wild-type expression of the gene in normal tissue, or differences in the amount of overexpression of the CIF150/hTAFjjl50 gene in the neoplastic tissue over time, can be used to prognose the progression of the neoplasia in that tissue or to monitor the response of the neoplastic tissue to various therapeutic regimens.
A genetic predisposition to neoplasia in a human can be detected by comparing a wild-type CIF150/hTAFjjl50 gene, mRNA, or protein with a CIF150/hTAFjιl50 gene, mRNA, or protein in a fetal tissue. Fetal tissues which can be used for this purpose include, but are not limited to, amniotic fluid, chorionic villi, blood, and the blastomere of an in v/tro-fertilized embryo. The wild-type CIF150/hTAFjjl50 gene can be obtained from any tissue. The mRNA or protein can be obtained from a normal tissue of a human in which the CIF150/hTAFjjl50 gene is expressed. Such tissues are disclosed above. Differences, such as alterations in the nucleotide sequence or size of the fetal CIF150/hTAFjl50 gene or mRNA, or alterations in the molecular weight, amino acid sequence, or relative abundance of fetal CIFl 50/hTAFjjl 50 protein, indicate a germline mutation in the CIF150/hTAF jl50 gene of the fetus which indicates a genetic predisposition to neoplasia.
Kits for use in the diagnostic methods described above are also provided. CIF150/hTAFjjl50 diagnostic kits comprise reagents which specifically bind to a human CIF150/hTAF jl50 gene or expression product and which can be used in methods of the invention, such as CIF150/hTAF l50 subgenomic polynucleotide probes or antibodies. Means for labeling the probes or antibodies, reagents for use in the methods, such as buffers, and instructions for using the kits can also be included.
The invention provides a means of identifying compounds which induce or prevent mitosis or cell cycle progression. A cell is contacted with a test compound. The test compound can be a pharmacologic agent already known in the art or can be a compound previously unknown to have any pharmacological activity. The test compound can be naturally occurring or designed in the laboratory. It can be isolated from microorganisms, animals, or plants, and can be produced recombinantly, or synthesized by chemical methods known in the art. The cell can be any primary human cell or human cell line which expresses a
CIF150/hTAFjjl50 gene, as disclosed above. Methods of establishing cultures of primary human cells or of culturing cell lines are well known in the art.
Expression of the CIFl 50/hTAFjjl 50 gene is detected. Means of detecting CIF150/hTAFjjl50 gene expression, by measuring CIF150/hTAFj l50 mRNA or CIFl 50/hTAFπ 150 protein, are disclosed above. Expression can be measured in a sample of the same cell population before and after contact with the test compound. Alternatively, control cell populations can be employed. A test compound which increases expression of the CIF150/hTAFjjl50 gene is identified as a potential drug for inducing mitosis or cell cycle progression. A test compound which decreases expression of the CIF150/hTAFjjl50 gene is identified as a potential drug for decreasing mitosis or cell cycle progression.
The function of OF150/hTAFπ150 as a transcription factor can be exploited to identify genes whose transcription is dependent on the presence of OF150/hTAFπ150. In one population of a cell type, such as HeLa or NIH 3T3 cells, expression of a CIF150/hTAF jl50 gene is unaltered; in another population of the cell type, expression of a CIFl 50/hTAFjjl 50 gene is decreased. RNA can be isolated from the two populations by methods well known in the art. Isolated RNA from the two populations can be compared to identify genes which are differentially transcribed in the two populations. Decreased CIF150/hTAFjjl50 expression can be achieved, for example, using ribozymes, antisense oligonucleotide sequences, or antibodies, as described above. The effectiveness of the mechanism chosen to alter expression of the CIFl 50/hTAF jJ 50 gene can be assessed using methods well known in the art, such as hybridization of nucleotide probes to CIF150/hTAFjjl50 mRNA or detection of CIF150/hTAFπ150 protein using specific antibodies.
Genes which are differentially transcribed in the two populations can be compared, for example, using differential display PCR. Differential display PCR can be carried out on the two populations of cells using methods well known in the art. See, e.g., Liang & Pardee, 1992, Science 257:961-11; Bauer et al, 1993, Nucl. Acids. Res. 27:4272-80; Bauer et al, 1994, PCR Methods Appl. 4S91-108; and Liang et al, 1995, Meth. Enz. 254:304-21. Kits for performing differential display PCR are available, for example, from Display Systems Biotech. Briefly, total RNA is isolated form the two populations of cells. The RNA is reverse transcribed to produce a cDNA population which represents an overlapping subset of the total expression profile of the cells in each population. Each subset cDNA population is amplified using PCR with an anchored primer and a group of arbitrary primers in the presence of radiolabeled dATP. Amplified products from the two populations of cells are separated by gel electrophoresis, and patterns of separated products are detected, as is known in the art.
Differences in the two patterns, such as the presence, absence, altered position within the gel, or amount of one or more cDNA species, indicates that the expression of one or more genes was altered in response to decreasing the expression of the CIF150/hTAFjjl50 gene. Differentially displayed bands can be excised from the gel, reamplified, and identified by sequence analysis. Optionally, the sequences can be cloned before sequencing. Sequences of the differentially displayed bands can be compared with known sequences in databases to determine the identity of genes whose expression was altered in response to decreasing expression of CIFl 50/hTAFjjl 50. The invention also provides means of identifying compounds which alter mitosis or cell cycle progression. A cell population is contacted with a test compound. A test compound can be a pharmacologic agent already known in the art or can be a compound previously unknown to have any pharmacological activity. A test compound can be naturally occurring or designed in the laboratory. It can be isolated from microorganisms, animals, or plants, and can be produced recombinantly, or synthesized by chemical methods known in the art.
The cell population can comprise any primary human cell or human cell line which expresses a CIFl 50/hTAFjjl 50 gene, as disclosed above. Methods of establishing cultures of primary human cells or of culturing cell lines are well known in the art. Expression of the CIF150/hTAFjjl50 gene in the cell population is detected. Means of detecting CIFl 50/hTAFjjl 50 gene expression, for example by measuring CIF150/hTAFπl50 mRNA or OF150/hTAFπ150 protein, are disclosed above. Expression can be measured in a sample of the same cell population before and after contact with the test compound. Alternatively, control cell populations which have not been contacted with the test compound can be employed. A test compound which increases expression of the CIF150/hTAF jl50 gene is identified as a potential compound for inducing mitosis or cell cycle progression. A test compound which decreases expression of the CIF150/hTAFjjl50 gene is identified as a potential compound for inhibiting mitosis or cell cycle progression.
The invention also provides methods for screening test compounds for the ability to interfere with the binding of CIF 130 to OF150/hTAFπ150. According to one method, at least a OF150/hTAFπ150-binding domain of a CIF 130 protein as shown in SEQ ID NO:2 and at least a OF130-binding domain of a OF150/hTAFπ150 protein as shown in SEQ ID NO:4 are incubated together in the presence of a test compound. A full-length CIF130 and/or a OF150/hTAFπ150 protein can also be used. In the absence of the test compound, the OF150/hTAFπ150-binding domain binds to the OF130-binding domain. The amount of bound and/or unbound proteins or binding domains is determined according to any technique known in the art, including any immunological technique. In order to facilitate the assay, one of the proteins or binding domains can be bound to a solid support, or can be labeled with a radiolabel, or other detectable label. A useful agent is identified which decreases the amount of a binding domain or a protein which is bound or increases the amount of a binding domain or a protein which is unbound. The binding domains or proteins can be prebound prior to the introduction of the test compound, or the test compound can be contacted with one of the two proteins or binding domains prior to incubation.
In another embodiment, a two-hybrid assay is used to screen compounds which inhibit the interaction between the binding partners, CIF 130 and OF150/hTAFπ150. According to such an assay, two fusion proteins, each comprising a binding domain of one of the binding partners, are used. The fusion proteins can comprise full-length CIF 130 or CIF150/hTAFu150 proteins or the portion of each protein necessary for the binding interaction. One of the binding partners is fused to a DNA binding domain and the other is fused to a transcriptional activating domain. If the fusion protein comprising the CIFl 50/hTAFπl 50-binding domain comprises the DNA binding domain, then the fusion protein comprising the OF130-binding domain comprises the transcriptional activating domain. If the fusion protein comprising the OF130-binding domain comprises the DNA binding domain, then the fusion protein comprising the OF150/hTAFπ150-binding domain comprises the transcriptional activating domain. The two fusion proteins interact to reconstitute a sequence-specific transcriptional activating factor. Many DNA binding domains and transcriptional activating domains can be used in this system, including the DNA binding domains of GAL4, LexA, and the human esfrogen receptor paired with the acidic transcriptional activating domains of GAL4 or the herpes virus simplex protein VP16. The two fusion proteins are contained in a cell which also comprises a reporter gene. The reporter gene is sensitive to the activation of the reconstituted sequence- specific transcriptional activating factor. Suitable reporter genes whose expression can be conveniently detected include the E. coli lacZ gene, whose expression may be measured colorimetrically, and yeast selectable genes such as H7S5 or URA3. The OF150/hTAFπ150-binding domain of CIF130 and the OF130-binding domain of OF150/hTAFπ150 can be readily determined, for example, by testing various portions of each protein for the ability to bind to its partner. A variety of techniques can be used for this purpose, including but not limited to the yeast two-hybrid assay, affinity column chromatography, and polyacrylamide gel electrophoresis under non-reducing conditions.
In the absence of the test compound, the cell expresses the reporter gene. A test compound is added to the cell, and the effect on expression of the reporter gene is measured. A test compound which disrupts the binding of the CIF 130 and
CIF150/hTAFπ150 will have a negative effect on the transcriptional activation ability of the reconstituted sequence-specific transcriptional activating factor. Thus, expression of the reporter gene will be decreased. Compounds which decrease expression of the reporter gene are potential inducers of mitosis or cell cycle progression. CIFl 50/hTAF jl 50 subgenomic polynucleotides can also be delivered to subjects for the purpose of screening test compounds for those which are useful for enhancing transfer of CIFl 50/hTAFjjl 50 subgenomic polynucleotides to the cell or for enhancing subsequent biological effects of the CIF150/hTAFjjl50 subgenomic polynucleotides within the cell. Such biological effects include hybridization to complementary CIF150/hTAFjjl50 mRNA and inhibition of its translation, expression of the
CIF150/hTAFjjl50 subgenomic polynucleotide to form CIF150/hTAFjjl50 mRNA and/or OF150/hTAFπ150 protein, and replication and integration of the CIF150/hTAFjjl50 subgenomic polynucleotide. Test compounds which can be screened include any substances, whether natural products or synthetic, which can be administered to the subject. Libraries or mixtures of compounds can be tested. The compounds or substances can be those for which a pharmaceutical effect is previously known or unknown. The compounds or substances can be delivered before, after, or concomitantly with the CIF150/hTAFjjl50 subgenomic polynucleotides. They can be administered separately or in admixture with the CIF150/hTAF jl50 subgenomic polynucleotides. Integration of delivered CIF150/hTAFjjl50 subgenomic polynucleotides can be monitored by any means known in the art. For example, Southern blotting of the delivered CIF150/hTAFjjl50 subgenomic polynucleotides can be performed. A change in the size of the fragments of the delivered polynucleotides indicates integration. Replication of the delivered polynucleotides can be monitored inter alia by detecting incorporation of labeled nucleotides combined with hybridization to a CIF150/hTAFj l50 probe. Expression of a CIF150/hTAFjjl50 subgenomic polynucleotide can be monitored by detecting production of CIFl 50/hTAFjjl 50 mRNA which hybridizes to the delivered polynucleotide or by detecting OF150/hTAFπ150 protein. CIF 150/hTAFπl 50 protein can be detected immunologically or by activity, for example by detecting binding to CIF130. Thus, delivery of CIFl 50/hTAFjjl 50 subgenomic polynucleotides according to the present invention provides an excellent system for screening test compounds for their ability to enhance delivery, integration, hybridization, expression, replication or integration in an animal, preferably a mammal, more preferably a human.
The complete consents of all references cited in this disclosure are incorporated herein by reference. The following are provided for exemplification purposes only and are not intended to limit the scope of the invention which has been described in broad terms above.
EXPERIMENTAL METHODS
Cell culture and cell-cycle analysis. IMR 90 (normal human lung fibroblasts) and HeLa cells were transfected with the phosphothiorate oligomers (100 to 400 nM) according to the manufacturer's protocol (Sequiur Inc.), using lipofection and OptiMEM (GibcoBRL). The transfected cells were harvested at various times post-lipofection for FACS and for Western and Northern analysis.
Initially, four single-strand antisense oligomers were designed to target distinct regions of CIFl 50/hTAFjjl 50 mRNA. The functional antisense oligomer designated B (5'-TGCTCATGGAAGCATAAGCAGCCAC-3'; SEQ ID NO:5) was used in combination with a control oligo Bx (5*-CACCGACGAATACGAAGGTACTCGT-3'; SEQ ID NO:6) containing the reverse sequence (3'-5') of the oligo B to assure identical nucleotide content. Density-arrested quiescent BALB/c3T3 and HeLa cells were prepared as previously described (Pledger et al, Proc. Natl Acad. Sci. U.S.A. 75, 2839- 43, 1978). To monitor cell synchrony, 106 cells from each sample were fixed in ethanol, treated with RNAase A (0.5 mg/ml, 1 hour at 37 °C), and stained with propidium iodide (40μg/ml ) 2 hr at 4 °C, than analyzed by flow cytometry on a Becton Dickinson FACScan. Immunoblot analysis were performed as described (Zawel & Reinberg, Ann. Rev.
Biochem. 64, 533-61, 1995) using CIF 150/hTAFπl 50-specific polyclonal antiserum generated against the N-terminal peptide MNRKKGDKGFESPRP (amino acids 11-25 of SEQ ID NO:2) or monoclonal antibodies specific for cyclin A , Bl, and E (Santa Cruz biotechnology, Inc). Differential display was performed as described by the manufacturer (Genomyx, Corp.).
Transfection and Reporter Assays. Cyclin A (-887 to+136) (28) and cyclin Bl(-893 to +110) (Oliphant et al, Mol. Cell. Biol. 9, 2944-49, 1989) promoter fragments were generated by PCR using HeLa DNA and cloned into the promoterless pGL3 luciferase reporter vector (Promega). All PCR-amplified fragments were verified by DNA sequencing. Cotransfections were performed in HeLa cells using a pENRFl (Cogswell et al, Mol. Cell. Biol. 15, 2782-90, 1995) based CIF150/hTAFjjl50 expression vector (pEVRF-OF150/hTAFπ150) or a pEVRFl-Ob expression plasmid in combination with the indicated reporter constructs. Luciferase activity was determined according to the manufacturer's protocol (Promega) 36 hr after transfection. Purification of proteins. HeLa nuclear extracts and CIF 150/hTAFπl 50 depleted nuclear extract were prepared as described previously (Zawel & Reinberg, 1995). OF150/hTAFπ150 protein was purified from SF9 cells under native conditions using the bacculovirus expression system (pBlueBacHis2, Invitrogen). Whole cell extract from Bacculo-infected SF9 cells were prepared by sonication and applied to TALOΝ™ metal affinity resin (Oontech) according to the manufacturer's protocol applying immidazole step elution. Recombinant OF150/hTAFπ150 protein was tested for CIF150/hTAFπ150 activity as described previously (Zawell & Reinberg, 1995; Kaufinann et al, 1996) and analyzed by SDS-PAGE (8%), followed by silver staining. In vitro franscription and in vitro DNA binding assays. In vitro transcription reactions were performed using the templates containing the G-less cassette as described before (Zawell & Reinberg, 1995 and references therein). Plasmid DNAs containing cyclin A, cyclin Bl, CMV promoter fragments were cloned upstream of a 180 bp G-less cassette using a PCR protocol (Zawell & Reinberg, 1995; Kaufinann et al, 1996).
For the complementation assay, 8 μl of the OF150/hTAFπ150 depleted nuclear extract (4 mg/ml) were preincubated for 30 min at 4 °C in the presence of DNA template with 1, 2 and 4 μl of recombinant CIF150/hTAFπ150 protein (see Fig. 3 A; 10 μl loaded), followed by addition of rNTPs to yield the following final concentrations: 500 μM ATP, 500 μM CTP, 500 μM GTP and 30 μM [α-32P]UTP. 32P-labeled RNA products were digested 15 min with RNAase HI, resolved on an 8% polyacrylamide-urea gel, and visualized by autoradiography. Elecfrophoretic mobility shift assays were performed using 3 and 6 μl OF150/hTAFπ150 in 40 μl GL-buffer as previously described (Zawell & Reinberg, 1995) with the exception that the reaction mixture contained 50 ng dGdC oligomer as competitor. After 30 min incubation at 40 °C the binding mixtures were loaded on a 6% TBE (0.5 x) polyacrylamide gel. Signals were quantitated by Phosphorimager analysis (BioRad, Inc.).
EXAMPLE 1 This example demonstrates that reduction in CIFl 50/hTAFjjl 50 mRNA or protein levels causes arrest of cells in G2.
Four different OF150/hTAFπ150 specific antisense phosphorothioate oligonucleotides were tested for their ability to modulate OF150/hTAFπ150 function.
One antisense oligonucleotide (oligomer B, 5'-TGCTCATGGA AGCATAAGCA GCCAC-3'; SEQ ID NO:5) led to a concentration-dependent increase in G2/M cells when transfected into HeLa cells (Figure 1A), whereas the other three oligomers had no effect on cell cycle progression.
Because HeLa cells are highly transformed and bear defects in cell cycle checkpoints (p53" and RB"), we confirmed the antisense effect using the primary lung fibroblast cell line IMR90 (Figure IB). A control oligo Bx (reverse sequence of B) did not affect the cell cycle progression in either cell line (Figures 1 A and IB). In Figure 1C, we analyzed the RNA derived from HeLa cells treated with the antisense oligo B and the control oligo Bx. Quantitative RT-PCR (lanes 1 to 6) as well as Northern hybridization (lanes 7 to 10) revealed dramatically reduced OF150/hTAFπ150 mRNA levels 24 h after antisense oligonucleotide treatment.
To demonstrate the antisense effect on OF150/hTAFπ150 protein levels we performed an immunoblot analysis using OF150/hTAFπ150-specific antiserum on cell ly sates from different time points after antisense treatment (Figures ID and IE). CIF 150/hT AFπ 150 protein decreased 36 h after antisense oligo treatment but remained unchanged after 24 h or in control oligonucleotide-freated cells (Figure ID). Since the cells were transiently transfected with the antisense oligomer, OF150/hTAFπ150 protein level increased again after 48 h (lane 3). In order to confirm our FACS analysis, we analyzed cyclin B 1 expression using extracts from antisense and control oligomer treated HeLa cells. Again, the level of CIF 150/hTAFπl 50 protein was reduced in a concentration-dependent manner using antisense oligo B (Figure IE, left panel). The level of cyclin Bl expression was similarly affected, indicating a cell cycle arrest and confirming our FACS analysis (Figure IE).
EXAMPLE 2
This example demonstrates that OF150/hTAFπ150 mediates selective transcription of a specific set of genes.
To confirm our FACS analysis, we analyzed cyclin Bl expression using extracts from antisense and control oligomer treated HeLa cells. Again, OF150/hTAFπ150 protein levels were concenfration dependently reduced by using the antisense oligo B
(Figure IE). The level of cyclin Bl expression seemed to be affected in the same way by the antisense treatment indicating a cell cycle arrest and confirming our FACS analysis (Figure IE).
To address the question whether the decrease of cyclin B 1 was a result of a more general effect on franscription in the absence of CIF 150, we determined the expression levels of other mRNAs. We analyzed RNA derived from cells treated with the antisense or control oligonucleotides by differential PCR-display, in which each band is the PCR product of a specific mRNA. Only a minority of transcripts were affected by the loss of CIF150/hTAFπ150 function (Figure IF), suggesting that OF150/hTAFπ150 mediates selective franscription of a specific set of class II genes. Similar results have been reported for a ts-mutant of mammalian TAFπ250 (Sekiguchi et al, Gene Cells 1, 687- 705, 1996; Suzuki- Yagawa et al, Mol. Cell. Biol. 17, 3284-94, 1997; Ware et al, Genes Dev. 11, 2658-69 1977) and after functional knock-out experiments using yeast TAFπ90 (Apone et al, Genes Dev. 10, 2368-80, 1996) and TAFπ145 (Wieczorek et al, Nature
393, 187-91, 1998; Walker et al, Cell 90, 607-14, 1997; Sheu & Green, Cell 90, 615-24, 1997).
EXAMPLE 3 This example demonstrates that CIFl 50/hTAFπl 50 expression and activity are cell cycle regulated and correlate with cyclin Bl and cyclin A transcription.
Serum-starved BALB/c 3T3 cells showed a lower level of OF150/hTAFπ150 protein and an increase in OF150/hTAFπ150 expression in G2/M after serum stimulation (Figure 2A). OF150/hTAFπ150 upregulation began approximately 15 hr after serum addition, just before Cyclin A and Bl expression (lanes 5 and 6). Cyclin E expression did not significantly change in this cell line and serves here as a loading control. We repeated the same experiments using HeLa cells and observed the same change in CIF150/hTAFπ150 and cyclin Bl expression (Figure 2B).
Taken together, these results indicate that regulation of OF150/hTAFπ150 protein levels is cell cycle dependent and suggests that OF150/hTAFπ150 is a positive regulator of cyclin Bl and A expression. EXAMPLE 4
This example demonstrates that OF150/hTAFπ150 activity changes during cell cycle progression.
In vitro transcription assays were performed using HeLa nuclear exfracts derived from different cell cycle stages after synchronization at the G,/S boundary by a double thymidine block (Shen & Green, 1997; Rao & Johnson, Nature 225, 159-64, 1970; Lew et al, Cell 66, 1197-1206, 1991). CIF150/hTAFπ150 activity was measured using synthetic Inr (TdT initiator)-containing and Inr-lacking promoters as described previously (Kaufinann et al, Mol. Cell. Biol. 18, 233-39, 1998). We observed an Inr-dependent increase in transcriptional activity in nuclear exfracts derived from cells 8 to 12 hr after release from the double thymidine block (Figure 2B; lanes 11, 13, and 15). This result clearly indicates a cell-cycle dependent change in OF150/hTAFπ150 activity.
We then used the same nuclear exfracts in combination with the TATA-less cyclin Bl and A promoter fragments fused to a G-less cassette in in vitro franscription assays (Zawell & Reinberg, 1995; Kaufinann et al, 1996). Both native promoters seemed to be more transcriptionally active only in the 8 to 12 hr nuclear exfracts where OF150/hTAFπ150 activity was strongest (Figure 2C). In contrast, the activity of the TATA-box containing control promoters IgH and CMV did not correlate with OF150/hTAFπ150 activity.
EXAMPLE 5
This example demonstrates that OF150/hTAFπ150 is a positive regulator of cyclin Bl and A TATA-less promoters.
To test whether OF150/hTAFπ150 is a positive regulator of TATA-less promoters, we analyzed the effect of OF150/hTAFπ150 in in vitro transcription and cofransfection assays. For in vitro franscription, we used highly purified recombinant OF150/hTAFπ150 protein (Figure 3A) in combination with nuclear exfracts depleted for OF150/hTAFπ150 activity, as described previously (Zawell & Reinberg, 1995). Cyclin Bl and Al promoter-dependent transcription was not observed in the absence of CIF150/hTAFπ150 activity. Titration of CIF150/hTAFπ150 protein stimulated the TATA-less cyclin A and Bl promoters, indicating that OFlSO/hTAFjjlSO is required for their transcription (Figure 3B, compare lane 1 with lanes 2, 3, and 4). The TATA-containing control promoters (IgH and CMV) were not affected by the absence of OF150/hTAFπ150.
To demonstrate the stimulation affect of OF150/hTAFπ150 in vivo, we performed cofransfection experiments of OF150/hTAFπ150 expression vectors in combination with a lucifierase reporter fused to cyclin Bl and A promoter fragments. Overexpression of OFlSO/hTAFjjlSO did have only a minor effect on the TATA- containing CMV and the minimal Fos promoter activity, but was able to stimulate cyclin Bl and cyclin A transcription in HeLa cells (Figure 3C).
The upper panel of Figure 3C shows absolute luciferase activities using the c-Fos, cyclin Bl, and cyclin A promoters in cofransfection experiments with an unrelated expression plasmid (pEVRFl-Ob) and a OF150/hTAFπ150 expression plasmid (pEVRF-OF150). Both the cyclin A and Bl promoters are preferentially stimulated by OF150/hTAFπ150 when compared to the TATA containing promoters c-Fos and CMV (see fold stimulation, lower panel of Figure 3C).
These findings support our in vitro data suggesting that OF150/hTAFπ150 is required for cyclin Bl and A franscription but is dispensable for transcription of TATA-containing promoters including the CMV, IgH, and the minimal Fos promoter.
EXAMPLE 6
This example demonstrates identification of a cis-acting CIF150/hTAFπ150 responsive element.
Because dTAFπ150 has been reported to recognize specific core promoter elements (Verrijzer et al, Science 264, 933-41, 1994) we have started to identify a cis- acting OF150/hTAFπ150-responsive element. As a first step we performed binding-site selection (Oliphant et al, 1989) using highly purified recombinant CIF150/hTAFπ150 (Figure 3 A). A pool of DNA fragments with 7 bp randomized nucleotide pairs were 32P-labeled and used in three successive rounds of gel shift experiments (Figure 4A).
Figure 4B shows that the selected protein-DNA complex is specific for OF150/hTAFπ150 protein, as demonstrated by a supershift induced by OF150/hTAFπ150-specific antiserum. DNA sequences of 44 selected PCR-fragments revealed a statistically significant enrichment of fragments with the core sequence 5'GAG3' (Figure 2C). Affinity of OF150/hTAFπ150 for the core sequence was improved only about five-fold after four rounds of selection (Figure 4A), possibly due to the presence of cryptic binding sites in the flanking sequences of the PCR primers used (see Figure 4B). Alternatively, OF150/hTAFπ150 might be able to form stable complexes with non-specific DNA.
To validate further the putative sequence element, we performed elecfrophoretic mobility shift experiments using DNA fragments with defined base pair substitutions (Fig 4D). Most substitutions reduced binding efficiency of OF150/hTAFπ150 by 50%, with the exception of mutations which contain an intact GAG core sequence (Figure 4D, compare oligo a and d with oligos b, c, e, f and g). These experiments clearly demonstrate that OF150/hTAFπ150 binds the 5'GAG3' core sequence with higher affinity than randomized DNA (Figure 4D; compare lane 11 with lanes 14, 16, and 18), indicating a potential role of OF150/hTAFπ150 in promoter selection.
To test this idea we created point mutations in putative OF150/hTAFπ150 binding elements (CBE) in the cyclin Bl core promoter and tested these constructs in cofransfection experiments (Figure 5 A). The substitution of the putative OF150/hTAFπ150 binding site 5'-GAGGCTA-3' (SEQ ID NO:8) just 4 bp downstream of a non-canonical TATA box abolished the stimulation of reporter activity by CIF150/hTAFπ150 (Figure 5).
These data suggest that OF150/hTAFπ150 is a necessary positive transcriptional regulator of cell cycle progression through G2/M. Furthermore, we provide additional evidence to support the idea that one of the regulatory functions of TAFπs is to select core promoters. It is not known whether OF150/hTAFπ150-dependent transcription of the TATA-less cyclin Bl and A promoters is still TBP-dependent. An attractive hypothesis is that OF150/hTAFπ150 binding to the CBE element can compensate for the absence of TBP binding to the TATA-box.

Claims

1. An isolated and purified subgenomic polynucleotide which encodes a protein comprising an amino acid sequence which is at least 85% identical to the nucleotide sequence shown in SEQ ID NO:2, wherein percent identity is determined using a Smith- Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 1.
2. The isolated and purified subgenomic polynucleotide of claim 1 wherein the protein has the amino acid sequence shown in SEQ ID NO:2.
3. The isolated and purified subgenomic polynucleotide of claim 2 which comprises the nucleotide sequence shown in SEQ ID NO:l.
4. An isolated and purified subgenomic polynucleotide which comprises at least 11 contiguous nucleotides selected from the nucleotide sequence shown in SEQ ID NO:l.
5. A construct which comprises: a promoter; and a polynucleotide segment encoding a human OF150/hTAFπ150 protein as shown in SEQ ID NO:2, wherein the polynucleotide segment is located downstream from the promoter, and wherein franscription of the polynucleotide segment initiates at the promoter.
6. A host cell comprising a construct, wherein the construct comprises a promoter and a polynucleotide segment encoding a human OF150/hTAFπ150 protein as shown in SEQ ID NO:2.
7. A homologously recombinant cell having incorporated therein a new transcription initiation unit, wherein the new transcription initiation unit comprises:
(a) an exogenous regulatory sequence;
(b) an exogenous exon; and
(c) a splice donor site, wherein the franscription initiation unit is located upstream of a coding sequence of a CIFl 50/hTAFjjl 50 gene as shown in SEQ ID NO:l, wherein the exogenous regulatory sequence directs transcription of the coding sequence of the CIF150/hTAFπ150 gene.
8. A method to aid in the diagnosis or prognosis of neoplasia in a human, comprising the step of: comparing expression of a first CIFl 50/hTAFjjl 50 gene in a first tissue of a human suspected of being neoplastic with expression of a second CIF150/hTAFjjl50 gene in a second tissue of a human which is normal, wherein the second CIFl 50/hTAFjjl 50 gene has the coding sequence shown in SEQ ID NO:l, wherein increased expression of the first CIFl 50/hTAFjjl 50 indicates neoplasia in the first tissue.
9. The method of claim 8 wherein CIF150/hTAFjjl50 mRNAs in the first and second tissues are compared.
10. The method of claim 9 wherein CIF 150/hTAFπ 150 proteins in the first and second tissues are compared.
11. A method to aid in the diagnosis or prognosis of neoplasia in a human, comprising the step of: comparing a first human CIFl 50/hTAF/f 150 gene, mRNA, or protein in a first tissue suspected of being neoplastic with a second human CIFl 50/hTAFjjl 50 gene, mRNA, or protein in a second tissue which is normal, wherein the second human CIFl 50/hTAFjjl 50 gene has the coding sequence shown in SEQ ID NO:l, wherein a difference between the first and second genes, mRNAs, or proteins indicates neoplasia in the first tissue.
12. The method of claim 11 wherein a CIFl 50/hTAFjjl 50 gene is compared.
13. The method of claim 11 wherein a CIF150/hTAFjjl50 mRNA is compared.
14. The method of claim 11 wherein a CIF 150/hTAFπl 50 protein is compared.
15. The method of claim 11 wherein the first and second tissues are obtained from the same human.
16. A method to aid in detecting a genetic predisposition to neoplasia in a human, comprising: comparing a CIF150/hTAFj l50 gene, mRNA, or protein in a fetal tissue of a human with a wild-type human CIF150/hTAFjjl50 gene, mRNA, or protein, wherein the wild-type human CIF150/hTAFjjl50 gene has the coding sequence shown in SEQ ID NO:l, wherein a difference between the CIF150/hTAF jl50 gene, mRNA, or protein in the fetal tissue of the human and the wild-type human CIF150/hTAFjjl50 gene, mRNA, or protein indicates a genetic predisposition to neoplasia in the human.
17. The method of claim 16 wherein a CIF150/hTAFjjl50 mRNA is compared.
18. The method of claim 16 wherein a CIFl 50/hTAFjjl 50 gene is compared.
19. The method of claim 16 wherein a CIF 150/hTAFπ 150 protein is compared.
20. A method of screening test compounds for the ability to interfere with the binding of a CIFl 30 protein to a OF150/hTAFπ150 protein, comprising the steps of:
(a) contacting a test compound with a OF150/hTAFπ150-binding domain of a CIF130 protein as shown in SEQ ID NO:4 and a OF130-binding domain of a OF150/hTAFπ150 protein as shown in SEQ ID NO:2, wherein the OF150/hTAFπ150- binding domain binds to the OF130-binding domain in the absence of the test compound; and
(b) determining the amount of OF150/hTAFπ150-binding domains which are bound or unbound to OF130-binding domains or determining the amount of OF130-binding domains which are bound or unbound to OF150/hTAFπ150-binding domains in the presence of the test compound, wherein a test compound which decreases the amount of bound CIF 130- or OF150/hTAFπ150-binding domains or which increases the amount of unbound CIF130- or OF150/hTAFπ150-binding domains is a potential inducer of mitosis or cell cycle progression.
21. The method of claim 20 wherein the CIF130- and the CIF150/hTAFπ150- binding domains are prebound prior to the step of contacting.
22. The method of claim 20 wherein the test compound is contacted with either of the CIF130- or CIF150/hTAFΓÇ₧150-binding domains prior to the step of contacting.
23. A method of screening test compounds for the ability to interfere with the binding of a CIF130 protein to a OF150/hTAFπ150 protein, comprising the steps of:
(a) contacting a cell with a test compound, wherein the cell comprises: i) a first fusion protein comprising (1) a OF150/hTAFπ150- binding domain of a CIF130 protein as shown in SEQ ID NO:4 and (2) either a DNA binding domain or a transcriptional activating domain; ii) a second fusion protein comprising a OF130-binding domain of a CIF150/hTAFπ150 protein as shown in SEQ ID NO:2, wherein the CIF130-binding domain binds to the OF150/hTAFπ150-binding domain, wherein if the first fusion protein comprises a DNA binding domain, then the second fusion protein comprises a transcriptional activating domain, wherein if the first fusion protein comprises a transcriptional activating domain, then the second fusion protein comprises a DNA binding domain, wherein the interaction of the first and second fusion proteins reconstitutes a sequence-specific transcription activating factor; and iii) a reporter gene comprising a DNA sequence to which the DNA binding domain specifically binds; and
(b) measuring the expression of the reporter gene, wherein a test compound which decreases the expression of the reporter gene is a potential inducer of mitosis or cell cycle progression.
24. A method of increasing expression of a gene, comprising the step of: contacting a promoter region of a gene with a OF150/hTAFπ150 protein as shown in SEQ ID NO:2, wherein the promoter region comprises a OF150/hTAFπ150 binding element comprising a nucleotide sequence 5'-Py X G A G A/C A/Py -3' (SEQ ID NO: 7), whereby expression of the gene is increased.
25. An antisense CIF150/hTAFjjl50 oligonucleotide as shown in SEQ ID NO:5.
26. An isolated and purified subgenomic polynucleotide which comprises 5'- Py X G A G A/c A/Py - 3' (SEQ ID NO:7).
PCT/US1998/027665 1997-12-30 1998-12-28 C1F150/hTAFII150 IS NECESSARY FOR CELL CYCLE PROGRESSION WO1999033985A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU19483/99A AU1948399A (en) 1997-12-30 1998-12-28 C1f150/h TAF II 150 is necessary for cell cycle progression

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US6895697P 1997-12-30 1997-12-30
US60/068,956 1997-12-30

Publications (2)

Publication Number Publication Date
WO1999033985A2 true WO1999033985A2 (en) 1999-07-08
WO1999033985A3 WO1999033985A3 (en) 1999-08-19

Family

ID=22085795

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/027665 WO1999033985A2 (en) 1997-12-30 1998-12-28 C1F150/hTAFII150 IS NECESSARY FOR CELL CYCLE PROGRESSION

Country Status (2)

Country Link
AU (1) AU1948399A (en)
WO (1) WO1999033985A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999064450A2 (en) * 1998-06-12 1999-12-16 Chiron Corporation Cif130 inhibits cell cycle progression

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994017087A1 (en) * 1993-01-28 1994-08-04 The Regents Of The University Of California Tata-binding protein associated factors, nucleic acids encoding tafs, and methods of use

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994017087A1 (en) * 1993-01-28 1994-08-04 The Regents Of The University Of California Tata-binding protein associated factors, nucleic acids encoding tafs, and methods of use

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
DATABASE EMEST 19 E.M.B.L. Databases Accession Number: AA103510, 30 October 1996 MARRA M ET AL: "Mus musculus cDNA clone 554617 5' similar to:G541665 DTAFII-150 mRNA" XP002106718 *
DATABASE EMROD E.M.B.L. Databases Accession Number: K01334, 1 March 1991 WODNAR-FILIPOWICZ A ET AL: "Cloning and sequence analysis of cDNA for rat spleen thymosin beta 4" XP002106719 *
KAUFMANN J ET AL: "CIF, an essential cofactor for TFIID-dependent initiator function" GENES AND DEVELOPMENT, vol. 10, no. 7, 1 April 1996, pages 873-886, XP002106713 cited in the application *
KAUFMANN J ET AL: "CIF150, a human cofactor for transcription factor IID-dependent initiator function" MOL. CELL. BIOL., vol. 18, no. 1, January 1998, pages 233-239, XP002106714 cited in the application *
MARTINEZ E ET AL: "Novel cofactors and TFIIA mediate functional core promoter selectivity by the human TAFII150-containing TFIID complex" MOL. CELL. BIOL., vol. 18, no. 11, November 1998, pages 6571-6583, XP002106715 *
VERRIJZER C ET AL: "Drosophila TAFII150: similarity to yeast gene TSM-1 and specific binding to core promoter DNA" SCIENCE, vol. 264, no. 5161, 13 May 1994, pages 933-941, XP002106716 cited in the application *
WALKER S ET AL: "Transcription activation in cells lacking TAFIIs" NATURE, vol. 383, 12 September 1996, pages 185-188, XP002106747 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999064450A2 (en) * 1998-06-12 1999-12-16 Chiron Corporation Cif130 inhibits cell cycle progression
WO1999064450A3 (en) * 1998-06-12 2000-02-03 Chiron Corp Cif130 inhibits cell cycle progression
US6423822B1 (en) 1998-06-12 2002-07-23 Chiron Corporation Human CIF130 polypeptides
US6589745B2 (en) 1998-06-12 2003-07-08 Chiron Corporation CIF130 inhibits cell cycle progression

Also Published As

Publication number Publication date
WO1999033985A3 (en) 1999-08-19
AU1948399A (en) 1999-07-19

Similar Documents

Publication Publication Date Title
US6635748B2 (en) Metastatic breast and colon cancer regulated genes
US7566559B2 (en) Human cyclin-dependent kinase (hPNQALRE)
US6030834A (en) Human IKK-beta DNA constructs and cells
US6492112B1 (en) Mitogen-activated protein kinase kinase 7 (MKK7)
US6432668B1 (en) Polynucleotides encoding human cyclin-dependent kinase (hPFTAIRE)
US6489137B2 (en) Detection of loss of the wild-type huBUB1 gene
US6410312B1 (en) huBUB3 gene involved in human cancers
US6174679B1 (en) CIF150/hTAFII150 is necessary for cell cycle progression
WO1999033985A2 (en) C1F150/hTAFII150 IS NECESSARY FOR CELL CYCLE PROGRESSION
WO1999033990A1 (en) HUMAN CXC CHEMOKINE (Tim-1)
WO1999033986A1 (en) MAMMALIAN ENHANCER OF POLYCOMB (epc) ACTS AS A TUMOR SUPPRESSOR
WO1999029850A1 (en) HUMAN KISMET PROTEIN (hkis) ACTS AS AN ONCOGENE
WO1999000499A1 (en) Human fadd-interacting protein (fip)
WO1999029851A1 (en) Human 'kismet' protein hkis acts as a tumor suppressor
WO1998057985A2 (en) Human rip-interacting factor (rif)
EP1693457A2 (en) Metastatic breast and colon cancer regulated genes

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A2

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

AK Designated states

Kind code of ref document: A3

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A3

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
122 Ep: pct application non-entry in european phase