WO1997017986A1 - Nima interacting proteins - Google Patents

Nima interacting proteins Download PDF

Info

Publication number
WO1997017986A1
WO1997017986A1 PCT/US1996/017334 US9617334W WO9717986A1 WO 1997017986 A1 WO1997017986 A1 WO 1997017986A1 US 9617334 W US9617334 W US 9617334W WO 9717986 A1 WO9717986 A1 WO 9717986A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
nima
pinl
pin1
cells
Prior art date
Application number
PCT/US1996/017334
Other languages
French (fr)
Inventor
Tony Hunter
Kun Ping Lu
Original Assignee
The Salk Institute For Biological Studies
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 The Salk Institute For Biological Studies filed Critical The Salk Institute For Biological Studies
Priority to EP96941949A priority Critical patent/EP0880359B1/en
Priority to CA002235225A priority patent/CA2235225C/en
Priority to DE69634671T priority patent/DE69634671T2/en
Priority to AU11158/97A priority patent/AU1115897A/en
Publication of WO1997017986A1 publication Critical patent/WO1997017986A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y502/00Cis-trans-isomerases (5.2)
    • C12Y502/01Cis-trans-Isomerases (5.2.1)
    • C12Y502/01008Peptidylprolyl isomerase (5.2.1.8), i.e. cyclophilin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/90Isomerases (5.)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates generally to the eukaryotic cell cycle and specifically to a novel class of proteins that interact with NIMA protein kinase in the NIMA mitotic pathway.
  • the CDC2 kinase associated with its cyclin partners has been shown to play an important role during G2/M progression in eukaryotic cells.
  • recent studies demonstrate that activation of the CDC2 kinase itself is not sufficient to trigger mitosis in some eukaryotic cells such as those in Saccharomyces cerevisiae
  • NIMA mitotic kinase
  • NIMA is a protein-Ser/Thr kinase, biochemically distinct from other protein kinases. and its phosphotransferase activity is regulated by Ser Thr phosphorylation
  • NIMA mitotic pathway is not restricted to Aspergillus, but also exists in vertebrate cells (Lu and Hunter, Cell, 81 :413. 1995a).
  • NIMA induces germinal vesicle breakdown without activating Mos, CDC2 or MAP kinase.
  • CDC2 In HeLa cells, NIMA induces mitotic events without activating CDC2, whereas dominant-negative NIMA mutants cause a specific G2 arrest.
  • O'Connell, et al (EMBOJ., 11:4926, 1 94) have also demonstrated that NIMA induce premature chromatin condensation in fission yeast and HeLa cells.
  • PPIases Peptidyl-prolyl cis/trans isomerases
  • proline isomerases Peptidyl-prolyl cis/trans isomerases
  • Cyclophilins and FK506-binding proteins FKBPs are two well characterized families of PPIases that share little if any amino acid similarity to each other. However, the members of each family contain a core structure that has been highly conserved from prokaryotes to eukaryotes.
  • Identification of such genes has several utilities including the identification of appropriate therapeutic targets, candidate genes for gene therapy (e.g. , gene replacement), mapping locations of disease-associated genes, and for the identification of diagnostic and prognostic indicator genes, for example.
  • the present invention provides a novel class of proteins that associate with NIMA protein kinase. Some of these proteins are characterized by inhibiting the mitosis promoting function of NIMA when overexpressed and inducing mitotic arrest and nuclear fragmentation when depleted.
  • the invention provides an exemplary NIMA associated protein called "protein interacting with NIMA" (Pinl).
  • Pinl has C- terminal peptidyl-propyl cis/trans isomerase activity and contains a conserved N- terminal tryptophan domain (WW domain) thought to mediate protein-protein interactions.
  • WW domain N- terminal tryptophan domain
  • the invention provides a method for identifying a protein that inhibits the mitosis promoting function of NIMA protein kinase.
  • the method is based on a genetic system designed to detect protein-protein interac ⁇ tions.
  • the method comprises culturing transformed cells containing the following: a nucleic acid construct comprising a DNA binding domain operatively associated with the coding sequence of NIMA, or functional fragments thereof; a nucleic acid library, wherein each member of said library comprises a transactivation domain operatively associated with a protein encoding sequence; and a nucleic acid reporter construct comprising a response element for the DNA binding domain operatively associated with a reporter gene, and monitoring for evidence of expression of reporter gene.
  • the invention provides a method for controlling the growth of a cell comprising contacting the cell with a composition which modulates Pinl activity.
  • a composition which modulates Pinl activity for example, an inhibitor of Pinl activity such as a PPlase inhibitor or an anti-Pin 1 antibody, or an inhibitor of PIN ⁇ expression such as an antisense nucleotide sequence or a ribozyme, can be used to control growth of a cell.
  • Pinl activity can be increased by an activator or PIN ⁇ expression can be increased by an enhancer, for example.
  • the invention provides a method for identifying a protein or other composition (e.g. , drug or other small molecule) that associates with and/or modulates Pinl protein activity or PIN ⁇ gene expression.
  • Figures 1 A and IB show human cDNA clones encoding proteins interacting with
  • Figure 1 A shows a ⁇ -Galactosidase Activity in the two-hybrid system.
  • Figure IB shows a comparison of NIMA and NLK1.
  • Figures 2A-2C show the cDNA and deduced amino acid sequences of human PIN ⁇ and homologies with other WW domain proteins and PPIases.
  • Figure 2 A shows the predicted Pinl amino acid sequence is indicated in one-letter code. The fusion points between GAL4 and Pinl in six different isolated clones were: clone H20 at C-9; clone H16, 24 and 38 at G+13; clones H6 and H36 at C+15. Underlined residues form a consensus bipartite nuclear localization signal. The N- and C- terminal boxes indicate the WW domain and PPlase domain. Nucleotide numbers are on the left and amino acid numbers on right.
  • Figure 2B and 2C show alignments of the WW domain (B) and PPlase Domain (C) in selected proteins. Identical residues are shown in the bottom row. Dashes indicate gaps introduced to make the alignment.
  • Cbf2 cell binding factor 2; SC, S. cerevisiae; EC, E. coli; BS, B. subtilis; CJ, C jejuni; AT, A. thaliana.
  • Figure 3 shows an analysis of PINX mRNA expression in human cell lines and human fetal liver.
  • HeLa epitheloid carcinoma cells
  • HFF human foreskin fibroblasts
  • Ramos Burkitt lymphoma cells
  • A431 epitheloid carcinoma cells
  • 293 adenovirus E1A transformed human embryonic kidney cells
  • U937 histiocytic lymphoma cells
  • Saos-2 osteosarcoma cells
  • U87-MG glioblastoma cells
  • Jurkat T leukemia cell line
  • HFL human fetal liver.
  • Position of the RNA molecular weight standards are on the left.
  • Figure 4 shows the PPlase activity of Pinl . Different concentrations of Pinl were used as indicated, with the recombinant FK506-binding protein (FKBP) PPlase being used as a positive control.
  • the control sample contained all the ingredients except the PPlase.
  • the insert shows PPlase activity during the first min of assay.
  • Figures 5A-5E show immunoprecipitations to show the interaction between Pinl and the C-terminal noncatalytic domain of NIMAs in HeLa cells.
  • Figure 5A shows the expression and purification of His-Pinl .
  • the PIN ⁇ cDNA was subcloned into pProExl and the recombinant protein was purified from bacteria using Ni 2+ -NTA agarose column, followed by analyzing the protein on a 15% SDS-containing gel, and Coomassie staining. The positions of His-Pinl and standard size markers are indicated.
  • Figure 5B shows the expression of NIMAs in HeLa cells. tTA-1 cells were transfected with different FLAG-tagged NIMA constructs and labeled with 35 S Express Label for 24 hr. followed by immunoprecipitation using the M2 mAb.
  • FIG. 5C shows a stable complex between recombinant Pinl and NIMAs.
  • Cell lysates similar to those in panel B were incubated with His-Pinl -Ni 2+ -NTA bead, as indicated in panel A and washed, followed by electrophoresis on an SDS-containing gel and autoradiography.
  • Figures 5 D and 5E show coimmunoprecipitation of K40M NIMA with Pinl .
  • tTA- 1 cells were cotransfected with FLAG-tagged NIMA and HA-/JN1 constructs for 24 hr.
  • Cell lysates were immunoprecipitated with the HA-specific 12CA5 mAb, followed by immunoblotting using the M2 mAB specific for the FLAG tag (D) and vice versa (E).
  • Figure 6 shows the colocahzation of Pinl and NIMA and its kinase-negative mutant in HeLa Cells.
  • the transfected tTA-1 cells were processed for indirect immunofluores- cence staining and examined by confocal microscopy.
  • Top panels staining pattern for NIMA or K40M NIMA obtained with FLAG tag-specific M2 mAb or SC-35 obtaining with anti-SC-35 mAb, and then FITC-conjugated IgGl -specific secondary antibodies; middle panels: staining pattern for Pinl obtaining with HA tag-specific 12CA5 mAb and Texas-Red-conjugated IgG2b-specific secondary antibodies; bottom panels: double-staining for Pinl and NIMA, Pinl and K40M NIMA, or Pinl and SC-35 displayed by superimposing the respective top and middle images, with yellow color indicating colocahzation. Arrows point to an untransfected cell, indicating very little cross reactivity among the antibodies. Bar, l ⁇ m.
  • Figures 7A-7D show overexpression of PIN ⁇ delays NIMA-induced mitotic arrest and induces a specific G2 arrest in HeLa cells.
  • Figure 7A shows the results of tTA- 1 cells cotransfected with nimA and PINX or control vector. At the times indicated, cells were fixed and doubly labeled with M2 mAb and Hoechst dye, followed by scoring for the percentage of cells with rounding and chromatin condensation in a sample of at least 250 NIMA-expressing cells. The data are the average from two independent experiments.
  • Figures 7B, C, and D show after transfection with PINX expression vector or the control vector for 48 hr, tTA-1 cells were stained with the 12CA5 mAb and then with FITC-conjugated secondary antibodies and propidium iodide, followed by
  • FIG. 8 shows depletion of Pinl /Ess 1 results in mitotic arrest and nuclear fragmentation in yeast.
  • a Pinl -dependent strain (YSH12.4) was shifted from inducing media to repressing media, harvested and fixed with 70% ethanol at the times indicated.
  • the cells were stained with DAPI or propidium iodide, followed by videomicroscopy under Nomarksi (DIC) or fluorescent (DAPI) illumination, or FACS analysis, respectively.
  • the bar is 10 ⁇ m and the inserts show a higher magnification of a representative cell.
  • a NIMA-like pathway is required for the G2/M transition in Aspergillus nidulans and human cells.
  • the present invention provides the first NIMA- interacting protein, Pinl, of mammalian origin, and methods for identification of other NIMA-interacting proteins. Overexpression of PINX and Pinl activity, induces a specific G2 arrest and delays NIMA-induced mitosis, while depletion of Pinl triggers mitotic arrest and nuclear fragmentation in budding yeast.
  • the invention provides an isolated mammalian protein characterized as associating with NIMA protein kinase, inhibiting the mitosis promoting function of NIMA when overexpressed, and inducing entry of cells into mitosis when depleted.
  • the C-terminal domain of such a protein catalyzes the cis/trans isomerization of peptidyl-propyl peptide bonds.
  • the N-terminal domain associates with NIMA and contains at least two conserved tryptophan residues (WW domain). While the exemplary polynucleotides and polypeptides of the invention are directed to Pinl , it is understood that any PIN polynucleotide or Pin protein can now be identified and characterized by the methods described herein.
  • the present invention provides a substantially pure NIMA-interacting protein (Pinl) characterized by having a molecular weight of about 18kD as determined by reducing SDS-PAGE, having peptidyl-propyl cis/trans isomerase activity, associating with NIMA protein kinase, and having essentially the amino acid sequence of SEQ ID NO:2.
  • the term "substantially pure” as used herein refers to Pinl which is substantially free of other proteins, lipids, carbohydrates or other materials with which it is naturally associated.
  • One skilled in the art can purify Pinl using standard techniques for protein purification.
  • the substantially pure polypeptide will yield a single maj or band on a non-reducing polyacrylamide gel.
  • Pinl polypeptide The purity of the Pinl polypeptide can also be determined by amino-terminal amino acid sequence analysis.
  • Pinl polypeptide includes functional fragments of the polypeptide, as long as the activity of Pinl remains. Smaller peptides containing one of the biological activies of Pinl are therefore included in the invention. Such peptides include immunologically reactive peptides capable of inducing antibody production.
  • the preferred Pinl of the invention is derived from a human cell.
  • the invention provides isolated polynucleotides encoding Pin polypeptides, including Pinl . These polynucleotides include DNA, cDNA and RNA sequences which encode Pinl . It is understood that all polynucleotides encoding all or a portion of Pinl are also included herein, as long as they encode a polypeptide with
  • Such polynucleotides include naturally occurring, synthetic, and intentionally manipulated polynucleotides.
  • PINX polynucleotide may be subjected to site-directed mutagenesis.
  • the polynucleotide sequence for PINX also includes antisense sequences.
  • the polynucleotides of the invention include sequences that are degenerate as a result of the genetic code. There are 20 natural amino acids, most of which are specified by more than one codon. Therefore, all degenerate nucleotide sequences are included in the invention as long as the amino acid sequence of Pinl polypeptide encoded by the nucleotide sequence is functionally unchanged.
  • Polynucleotides of the invention include variations thereof which encode the same amino acid sequence, but employ different codons for some of the amino acids, or splice variant nucleotide sequences thereof.
  • the sequence contains an open reading frame encoding a polypeptide 163 amino acids in length.
  • FIGURE 2A at position 25-27 is the first ATG codon.
  • PIN1 nucleotide sequence is SEQ ID NO: 1 and the deduced amino acid sequence is preferably SEQ ID NO:2 (FIGURE 2 A).
  • the polynucleotide encoding Pinl includes SEQ ID NO: 1 as well as nucleic acid sequences complementary to SEQ ID NO: 1 (FIGURE 2A).
  • a complementary sequence may include an antisense nucleotide.
  • fragments of the above-described nucleic acid sequences that are at least 15 bases in length, which is sufficient to permit the fragment to selectively hybridize to DNA that encodes the protein of SEQ ID NO:2 under physiological conditions.
  • selectively hybridize means that the fragments should hybridize to DNA encoding Pinl protein under moderate to highly stringent conditions.
  • Minor modifications of the Pinl primary amino acid sequence may result in proteins which have substantially equivalent activity as compared to the Pinl polypeptide described herein. Such proteins include those as defined by the term "having substantially the amino acid sequence of SEQ ID NO:2". Such modifications may be deliberate, as by site-directed mutagenesis, or may be spontaneous.
  • the Pinl polypeptide of the invention encoded by the polynucleotide of the invention includes the disclosed sequence (SEQ ID NO:2; FIGURE 2A) and conservative variations thereof.
  • the term "conservative variation” as used herein denotes the replacement of an amino acid residue by another, biologically similar residue. Examples of conservative variations include the substitution of one hydrophobic residue such as isoleucine, valine, leucine or methionine for another, or the substitution of one polar residue for another, such as the substitution of arginine for lysine, glutamic for aspartic acid, or glutamine for asparagine, and the like.
  • the term “conservative variation” also includes the use of a substituted amino acid in place of an unsubstituted parent amino acid provided that antibodies raised to the substituted polypeptide also immunoreact with the unsubstituted polypep ⁇ tide.
  • DNA sequences of the invention can be obtained by several methods.
  • the DNA can be isolated using hybridization techniques which are well known in the art. These include, but are not limited to: 1 ) hybridization of genomic or cDNA libraries with probes to detect homologous nucleotide sequences, 2) polymerase chain reaction (PCR) on genomic DNA or cDNA using primers capable of annealing to the DNA sequence of interest, and 3) antibody screening of expression libraries to detect cloned DNA fragments with shared structural features.
  • hybridization techniques which are well known in the art. These include, but are not limited to: 1 ) hybridization of genomic or cDNA libraries with probes to detect homologous nucleotide sequences, 2) polymerase chain reaction (PCR) on genomic DNA or cDNA using primers capable of annealing to the DNA sequence of interest, and 3) antibody screening of expression libraries to detect cloned DNA fragments with shared structural features.
  • the PIN (e.g., PIN1) polynucleotide of the invention is derived from a mammalian organism, and most preferably from human. Screening proc ⁇ edures which rely on nucleic acid hybridization make it possible to isolate any gene sequence from any organism, provided the appropriate probe is available.
  • Oligonucleotide probes which correspond to a part of the sequence encoding the protein in question, can be synthesized chemically. This requires that short, oligopeptide stretches of amino acid sequence must be known.
  • the DNA sequence encoding the protein can be deduced from the genetic code, however, the degeneracy of the code must be taken into account. It is possible to perform a mixed addition reaction when the sequence is degenerate. This includes a heterogeneous mixture of denatured double-stranded DNA. For such screening, hybridization is preferably performed on either single-stranded DNA or denatured double-stranded DNA.
  • Hybridization is particularly useful in the detection of cDNA clones derived from sources where an extremely low amount of mRNA sequences relating to the polypeptide of interest are present.
  • stringent hybridization conditions directed to avoid non-specific binding, it is possible, for example, to allow the autoradiographic visualization of a specific cDNA clone by the hybridization of the target DNA to that single probe in the mixture which is its complete complement (Wallace, et al, Nucl. Acid Res., 9:879,
  • DNA sequences encoding PIN can also be obtained by: 1) isolation of double-stranded DNA sequences from the genomic DNA; 2) chemical manufacture of a DNA sequence to provide the necessary codons for the polypeptide of interest; 3) in vitro synthesis of a double-stranded DNA sequence by reverse transcription of mRNA isolated from a eukaryotic donor cell; and PCR of genomic DNA or cDNA using primers capable of annealing to the DNA sequence of interest. In the latter case, a double-stranded DNA complement of mRNA is eventually formed which is generally referred to as cDNA.
  • genomic DNA isolates are the least common. This is especially true when it is desirable to obtain the microbial expression of mammalian polypeptides due to the possible presence of introns.
  • the synthesis of DNA sequences is frequently the method of choice when the entire sequence of amino acid residues of the desired polypeptide product is known. When the entire sequence of amino acid residues of the desired polypep ⁇ tide is not known, the direct synthesis of DNA sequences is r ⁇ A possible and the method of choice is the synthesis of cDNA sequences.
  • plasmid- or phage-carrying cDNA libraries which are derived from reverse transcription of mRNA which is abundant in donor cells that have a high level of genetic expres ⁇ sion.
  • phage-carrying cDNA libraries which are derived from reverse transcription of mRNA which is abundant in donor cells that have a high level of genetic expres ⁇ sion.
  • the production of labeled single or double-stranded DNA or RNA probe sequences duplicating a sequence putatively present in the target cDNA may be employed in DNA/DNA hybridiza ⁇ tion procedures which are carried out on cloned copies of the cDN A which have been denatured into a single-stranded form (Jay, et al., Nucl. Acid Res., 11 :2325. 1983).
  • a cDNA expression library such as lambda gtl 1
  • P1_N antibodies specific for P1_N.
  • Such antibodies can be either polyclonally or monoclonally derived and used to detect expression product indicative of the presence of PIN 1 cDNA.
  • DNA sequences encoding Pinl can be expressed in vitro by DNA transfer into a suitable host cell.
  • "Host cells” are cells in which a vector can be propagated and its DNA expressed. The term also includes any progeny of the subject host cell. It is understood that all progeny may not be identical to the parental cell since there may be mutations that occur during replication. However, such progeny are included when the term "host cell” is used. Methods of stable transfer, meaning that the foreign DNA is continuously maintained in the host, are known in the art.
  • the PIN1 polynucleotide sequences may be inserted into a recombinant expression vector.
  • recombinant expression vector refers to a plasmid, virus or other vehicle known in the art that has been manipulated by insertion or inco ⁇ oration of the PIN genetic sequences.
  • Such expression vectors contain a promoter sequence which facilitates the efficient transcription of the inserted genetic sequence of the host.
  • the expression vector typically contains an origin of replication, a promoter, as well as specific genes which allow phenotypic selection of the transformed cells.
  • Vectors suitable for use in the present invention include, but are not limited to the T7-based expression vector for expression in bacteria (Rosenberg, et al., Gene, 56: 125, 1987), the pMSXND expression vector for expression in mammalian cells (Lee and Nathans, J. Biol. Chem., 263:3521. 1988) and baculovirus-derived vectors for expression in insect cells.
  • the DNA segment can be present in the vector operably linked to regulatory elements, for example, a promoter (e.g., T7, metal lothionein I, tetracycline responsive promoter or polyhedrin promoters).
  • Polynucleotide sequences encoding Pinl can be expressed in either prokaryotes or eukaryotes.
  • Hosts can include microbial, yeast, insect and mammalian organisms. Methods of expressing DNA sequences having eukaryotic or viral sequences in prokaryotes are well known in the art.
  • Biologically functional viral and plasmid DNA vectors capable of expression and replication in a host are known in the art. Such vectors are used to incorporate DNA sequences of the invention. Transformation of a host cell with recombinant DNA may be carried out by conventional techniques as are well known to those skilled in the art. Where the host is prokaryotic, such as E. coli, competent cells which are capable of DNA uptake can be prepared from cells harvested after exponential growth phase and subsequently treated by the CaCL method using procedures well known in the art.
  • MgCI 2 or RbCl can be used. Transformation can also be performed after forming a protoplast of the host cell if desired.
  • Eukaryotic cells can also be cotransformed with DNA sequences encoding the PIN of the invention, and a second foreign DNA molecule encoding a selectable phenotype, such as the herpes simplex thymidine kinase gene.
  • Another method is to use a eukaryotic viral vector, such as simian virus 40 (SV40) or bovine papilloma virus, to transiently infect or transform eukaryotic cells and express the protein, (see for example, Eukaryotic Viral Vectors, Cold Spring Harbor Laboratory, Gluzman ed., 1982).
  • a eukaryotic viral vector such as simian virus 40 (SV40) or bovine papilloma virus
  • Isolation and purification of microbial expressed polypeptide, or fragments thereof, provided by the invention may be carried out by conventional means including preparative chromatography and immunological separations involving monoclonal or polyclonal antibodies.
  • preparative chromatography and immunological separations involving monoclonal or polyclonal antibodies For example, one of skill in the art could use (His) 6 tag affinity purification as described in the EXAMPLES herein.
  • the Pin polypeptides of the invention can also be used to produce antibodies which are immunoreactive or bind to epitopes of the Pin polypeptides.
  • Antibody which consists essentially of pooled monoclonal antibodies with different epitopic specificities, as well as distinct monoclonal antibody preparations arc provided.
  • Monoclonal antibodies are made from antigen containing fragments of the protein by methods well known in the art (Kohler, et al, Nature, 256:495. 1975; Current Protocols in Molecular Biology, Ausubel, et al., ed., 1989).
  • antibody as used in this invention includes intact molecules as well as fragments thereof, such as Fab, F(ab') 2 , and Fv which are capable of binding the epitopic determinant. These antibody fragments retain some ability to selectively bind with its antigen or receptor and are defined as follows: (1 ) Fab, the fragment which contains a monovalent antigen-binding fragment of an antibody molecule can be produced by digestion of whole antibody with the enzyme papain to yield an intact light chain and a portion of one heavy chain;
  • Fab' the fragment of an antibody molecule can be obtained by treating whole antibody with pepsin, followed by reduction, to yield an intact light chain and a portion of the heavy chain; two Fab' fragments are obtained per antibody molecule;
  • Fv defined as a genetically engineered fragment containing the variable region of the light chain and the variable region of the heavy chain expressed as two chains
  • Single chain antibody defined as a genetically engineered molecule containing the variable region of the light chain, the variable region of the heavy chain, linked by a suitable polypeptide linker as a genetically fused single chain molecule.
  • epitopic determinants means any antigenic determinant on an antigen to which the paratope of an antibody binds.
  • Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimension- al structural characteristics, as well as specific charge characteristics.
  • Antibodies which bind to the Pinl polypeptide of the invention can be prepared using an intact polypeptide or fragments containing small peptides of interest as the immunizing antigen. For example, it may be desirable to produce antibodies that specifically bind to the N- or C-terminal domains of Pinl .
  • the polypeptide or a peptide used to immunize an animal can be derived from translated cDNA or chemical synthesis which can be conjugated to a carrier protein, if desired.
  • a carrier protein e.g., a carrier protein, if desired.
  • commonly used carriers which are chemically coupled to the peptide include keyhole limpet hemocyanin (KLH), thyroglobulin, bovine serum albumin (BSA), and tetanus toxoid.
  • KLH keyhole limpet hemocyanin
  • BSA bovine serum albumin
  • the coupled peptide is then used to immunize the animal (e.g., a mouse, a rat, or a rabbit).
  • polyclonal or monoclonal antibodies can be further purified, for example, by binding to and elution from a matrix to which the polypeptide or a peptide to which the antibodies were raised is bound.
  • a matrix to which the polypeptide or a peptide to which the antibodies were raised is bound.
  • monoclonal antibodies See for example, Coligan, et ⁇ l , Unit 9, Current Protocols in Immunology, Wiley I- nterscience, 1994, incorporated by reference).
  • an anti-idiotypic monoclonal antibody made to a first monoclonal antibody will have a binding domain in the hypervariable region which is the "image" of the epitope bound by the first monoclonal antibody.
  • the invention provides a method for identifying a protein that inhibits the mitosis promoting function of NIMA protein kinase.
  • the method comprises culturing transformed cells containing the following: a nucleic acid construct comprising a DNA binding domain operably associated with the coding sequence of NIMA, or functional fragments thereof; a nucleic acid library, wherein each member of said library comprises a transactivation domain operably associated with a protein encoding sequence; and a nucleic acid reporter construct comprising a response element for the DNA binding domain operably associated with a reporter gene, and monitoring for evidence of expression of reporter gene.
  • the term “inhibits” refers to a reduction in the mitosis promoting function of NIMA protein kinase.
  • the "mitosis promoting function of NIMA” refers to the ability of NIMA to promote the progression of the G2/M transition in a cell.
  • the term “operably associated” refers to functional linkage between the promoter or regulatory sequence and the controlled nucleic acid sequence; the controlled sequence and regulatory sequence, or promoter are typically covalently joined, preferably by conventional phosphodiester bonds.
  • the method of the invention comprises culturing cells, under suitable conditions, transformed by standard methods in the art and as described above.
  • the transformed cells contain the following: a nucleic acid c 1 istruct comprising a DNA binding domain operably associated with the coding sequence of NIMA, or functional fragments thereof.
  • DNA binding domain refers to a nucleic acid sequence which contains a recognition site for a protein that binds to specific DNA sequences.
  • the coding sequence of NIMA includes nucleic acid sequence that encodes a protein having the biological activity of NIMA as described herein.
  • the method includes "functional fragments" of NIMA that are less than the full length coding sequence but which encode a protein having NIMA biological activity (e.g., Ser/Thr protein kinase).
  • the transformed cells also contain a nucleic acid library, wherein each member of the library comprises a transactivation domain operably associated with a protein encoding sequence.
  • a "transactivation domain” refers to a nucleic acid sequence which activates transcription, but typically fails to bind to DNA.
  • a nucleic acid library consists of a clone bank of genomic or complementary DNA sequences which are "protein encoding sequences”. The term refers to cDNA operably associated with the transactivation domain of a protein as exemplified in the EXAMPLES.
  • the cells also contain a nucleic acid reporter construct comprising a response element for the DNA binding domain operably associated with a reporter gene.
  • reporter refers to a gene encoding a trait or a phenotype which permits the monitoring and selection of, or the screening for, a cell containing the marker, and thus a NIMA interacting protein.
  • the nucleic acid reporter is a selectable marker such as a color indicator (e.g., lacZ). Other suitable reporter genes will be known to those of skill in the art.
  • the protein-protein interaction system exemplified in the EXAMPLES herein preferably utilize the GAL4 protein to provide the DNA binding and transactivation domains (Fields and Song, Nature, 340:245. 1989).
  • GAL4 protein to provide the DNA binding and transactivation domains
  • Other nucleic acid constructs comprising the coding sequences of proteins that have a DNA binding and transactivation domain will be known to those of skill in the art and can be utilized in the method of the invention for providing the hybrid constructs.
  • the method of the invention relies on interaction between the library encoded protein and the NIMA protein, or functional fragment thereof, in order to activate transcription of the reporter gene. Once the library encoded protein and the NIMA protein associate, they bring the DNA binding domain and the transactivation domain in close proximity, resulting in transcriptional activity. Thus, the method provides a means for identifying a protein that interacts or associates with NIMA protein kinase.
  • the transformed cell may contain the reverse constructs, e.g., a nucleic acid construct comprising a transactivation domain operably associated with the coding sequence of NIMA, or fragments thereof, and a nucleic acid library, wherein each member of said library comprises a DNA binding domain operably associated with a protein encoding sequence.
  • NIMA interacting proteins identified by the above method.
  • the invention also provides a method for controlling the growth of a cell comprising contracting the cell with a composition which modulates Pinl activity.
  • modulate envisions the suppression of expression of PIN1 or suppression of Pinl activity, when it is over-expressed, or augmentation of PIN 1 798
  • Pinl activity or protein level when it is under-expressed.
  • Growth of a cell is "controlled", for example, by inhibiting mitosis promoting function of NIMA or inducing entry of cells into mitosis.
  • An inhibitor of Pinl activity or protein level for example, would result in arrest in mitosis. Therefore, Pinl activity or protein level can be decreased, leading the cell to mitotic arrest, or alternatively, Pinl activity or protein level can be increased, arresting the cells in G2.
  • nucleic acid sequences that interfere with PINX expression at the translational level can be used.
  • This approach utilizes, for example, antisense nucleic acid, ribozymes, or triplex agents to block transcription or translation of a specific PIN mRNA, either by masking that mRNA with an antisense nucleic acid or triplex agent, or by cleaving it with a ribozyme.
  • Antisense nucleic acids are DNA or RNA molecules that are complemen ⁇ tary to at least a portion of a specific mRNA molecule (Weintraub, Scientific American, 262:40, 1990). In the cell, the antisense nucleic acids hybridize to the corresponding mRNA, forming a double-stranded molecule. The antisense nucleic acids primarily work via RNaseH-mediated degradation of the target mRNA. Antisense may also interfere with the translation of the mRNA, since the cell will not translate a mRNA that is double-stranded.
  • Antisense ohgomers of about 15 nucleotides are preferred, since they are easily synthesized and are less likely to cause problems than larger molecules when introduced into the target PIN- producing cell.
  • the use of antisense methods is well known in the art (Marcus- Sakura, Anal. iochem., 172:289, 1988).
  • triplex strategy Use of an oligonucleotide to stall transcription is known as the triplex strategy since the oligomer winds around double-helical DNA, forming a three- strand helix. Therefore, these triplex compounds can be designed to recognize a unique site on a chosen gene (Maher, et al, Antisense Res. and Dev., U3):227. 1991; Helene, C, Anticancer Drug Design, 6_(6 ⁇ :569, 1991).
  • Ribozymes are RNA molecules possessing the ability to specifically cleave other single-stranded RNA in a manner analogous to DNA restriction -21- endonucleases. Through the modification of nucleotide sequences which encode these RNAs, it is possible to engineer molecules that recognize specific nucleotide sequences in an RNA molecule and cleave it (Cech, J.Amer.Med. Assn., 260:3030. 1988). A major advantage of this approach is that, because they are sequence- specific, only mRNAs with particular sequences are inactivated.
  • Pin inhibitors include PPlase inhibitors.
  • immunosup ⁇ pressive drugs such as cyclosporin A and FK506, are PPlase inhibitors useful in the method of the invention for inhibiting some Pin proteins.
  • the Pinl or other Pin protein of the invention is useful for screening for other inhibitors as well.
  • a suspected PPlase inhibitor is incubated with Pin 1 protein under suitable conditions to allow Pinl enzymatic activity to be expressed and measured, and the level of activity in the presence and absence of the inhibitor is assayed to determine the effect on Pinl activity.
  • Pinl activity may also be increased in the presence of an activator.
  • PINX expression may be increased in the presence of an enhancer for example.
  • Stimulation of Pinl activity or overexpression of PINX arrests the cells in G2 and inhibits the mitosis promoting function of NIMA.
  • the -acting elements which control genes are called promoters, enhancers or silencers. Promoters are positioned next to the start site of transcription and function in an orientation- dependent manner, while enhancer and silencer elements, which modulate the activity of promoters, are flexible with respect to their orientation and distance to the start site of transcription.
  • One of skill in the art will know methods for stimulating expression of PINX by including an enhancer, for example, to stimulate PINX expression.
  • the effect of an enhancer or other regulatory element can be determined by standard methods in the art (e.g., Northern blot analysis, nuclear runoff assay).
  • Pinl activity can be affected by an activator.
  • An "activator” as used herein includes a protein or a small molecule, such as an organic compound, for example, which increases Pinl activity or protein level, such that a cell arrests in G2.
  • An activator can be identified by incubating Pinl and a putative activator under -21- conditions that allow interaction between the components, and measuring of the effect of the activator on Pinl . For example, one could determine whether cells were arrested in G2 (e.g. , increased Pinl activity), or whether cells continued to cycle through G2 to M (e.g. , by FACS analysis or labeled nuclei analysis).
  • the Pin 1 protein of the invention is useful in a screening method to identify compounds or compositions which affect the activity of the protein or expression of the gene.
  • the invention provides a method for identifying a composition which affects Pinl comprising incubating the compo ⁇ nents, which include the composition to be tested (e.g., a drug or a protein) and Pinl , under conditions sufficient to allow the components to interact, then subse ⁇ quently measuring the effect the composition has on Pinl activity or expression.
  • the observed effect on Pinl may be either inhibitory or stimulatory.
  • the entry of cells into mitosis or G2 arrest can be determined by nuclear content (e.g. , immunofluorescence or FACS analysis based on DNA content) or other methods known to those of skill in the art.
  • the increase or decrease of PINX transcription/translation can be measured by adding a radioactive compound to the mixture of components, such as 32 P-ATP (for nuclei) or 3 H-Uridine, or 35 S-Met, and observing radioacti e inco ⁇ oration into PINX transcripts or protein, respectively.
  • a radioactive compound such as 32 P-ATP (for nuclei) or 3 H-Uridine, or 35 S-Met
  • other labels may be used to determine the effect of a composition on PINX transcription/translation.
  • a radioisotope, a fluorescent compound, a bioluminescent compound, a chemiluminescent compound, a metal chelator or an enzyme could be used.
  • Analysis of the effect of a compound on PINX is performed by standard methods in the art, such as Northern blot analysis (to measure gene expression) or SDS-PAGE (to measure protein product), for example. Further, Pinl biological activity can also be determined, for example, by inco ⁇ oration label into nuclei.
  • the method of the invention described above also includes identifying a protein that associates with Pinl protein.
  • the method comprises incubating the protein with Pinl protein, or with a recombinant cell expressing Pinl , under conditions sufficient to allow the components to interact and determining the effect of the protein on Pinl activity or expression.
  • one of skill in the art could use the two-hybrid system described above to identify a protein that interacts or associates with Pinl protein. Once identified, such Pin E/N-associated proteins may now be used as targets for drug development.
  • the invention provides a method for treating a cell proliferative disorder.
  • the method comprises administering a subject in need of such treatment, an amount of Pinl inhibitor effective to induce entry of cells into mitosis or apoptosis.
  • the "amount of Pinl inhibitor effective to induce entry of cells into mitosis” means that the amount of polypeptide, peptide, polynucleotide, or monoclonal antibody for example, which when used, is of sufficient quantity to ameliorate the disorder.
  • cell-proliferative disorder denotes malignant as well as non-malignant cell populations which mo ⁇ hologically often appear to differ from the surrounding tissue both mo ⁇ hologically and genotypically.
  • Malignant cells develop as a result of a multistep process.
  • the PIN1 polynucleotide that is an antisense molecule is useful in treating malignancies of the various organ systems.
  • the method may be useful in treating malignancies of the various organ systems, such as, for example, lung, breast, lymphoid, gastrointestinal, and genito-urinary tract as well as adenocarcinomas which include malignancies such as most colon cancers, renal-cell carcinoma, prostate cancer, leukemia, breast cancer, non-small cell carcinoma of the lung, cancer of the small intestine, and cancer of the esophagus.
  • any disorder which is etiologically linked to altered expression of PIN] could also be considered susceptible to treatment with a PIN] suppressing/inhibiting reagent.
  • the method is also useful in treating non-malignant or immunologically- related cell-proliferative diseases such as psoriasis, pemphigus vulgaris, Bechet's syndrome, acute respiratory distress syndrome (ARDS), ischemic heart disease, post-dialysis syndrome, rheumatoid arthritis, acquired immune deficiency syndrome, vasculitis, lipid histiocytosis, septic shock and inflammation in general.
  • non-malignant or immunologically- related cell-proliferative diseases such as psoriasis, pemphigus vulgaris, Bechet's syndrome, acute respiratory distress syndrome (ARDS), ischemic heart disease, post-dialysis syndrome, rheumatoid arthritis, acquired immune deficiency syndrome, vasculitis, lipid histiocytosis, septic shock and inflammation in general.
  • any disorder which is etiologically linked to PIN would also be considered susceptible to treatment.
  • an antibody or nucleic acid probe specific for Pin l may be used to detect Pinl polypeptide (using antibody) or polynucleotide
  • the invention provides a method for detecting a cell proliferative disorder which comprises contacting an anti-Pin 1 antibody or nucleic acid probe with a cell suspected of having a Pinl associated disorder and detecting binding to the antibody or nucleic acid probe.
  • the antibody reactive with Pinl or the nucleic acid probe is preferably labeled with l o a compound which allows detection of binding to Pin 1. Any specimen containing a detectable amount of antigen or polynucleotide can be used.
  • the level of Pinl in the suspect cell can be compared with the level in a normal cell to determine whether the subject has a Pin 1 -associated cell proliferative disorder.
  • the subject is human.
  • PCR polymerase chain reaction
  • LAT ligated activated transcription
  • NASBA nucleic acid sequence-based amplification
  • the antibodies of the invention can be used in any subject in which it is desirable to administer in vitro or in vivo immunodiagnosis or immunotherapy.
  • the antibodies of the invention are suited for use, for example, in immunoassays in which they can be utilized in liquid phase or bound to a solid phase carrier.
  • the antibodies in these immunoassays can be detectably labeled in various immunoassays.
  • Examples of types of immunoassays which can utilize antibodies of the invention are competitive and non-competitive immunoassays in either a direct or indirect format. Examples of such immunoassays are the radioimmunoassay (RLA) and the sandwich (immunometric) assay. Detection of the antigens using the antibodies of the invention can be done utilizing immunoassays which are run in
  • viral vectors which can be utilized for gene therapy as taught herein include adenovirus, he ⁇ es virus, vaccinia, or, preferably, an RNA virus such
  • the retroviral vector is a derivative of a murine or avian retrovirus.
  • retroviral vectors in which a single foreign gene can be inserted include, but are not limited to: Moloney murine leukemia virus (- MoMuLV), Harvey murine sarcoma virus (HaMuSV), murine mammary tumor virus (MuMTV), and Rous Sarcoma Virus (RSV).
  • - MoMuLV Moloney murine leukemia virus
  • HaMuSV Harvey murine sarcoma virus
  • MuMTV murine mammary tumor virus
  • RSV Rous Sarcoma Virus
  • GaLV 10 gibbon ape leukemia virus
  • retroviral vectors can inco ⁇ orate multiple genes.
  • Other viral vectors include DNA vectors such as adenovirus and adeno-associated virus (AAV). All of these vectors can transfer or inco ⁇ orate a gene for a selectable marker so that transduced cells can be identified and generated.
  • Retroviral vectors can be made target specific by attaching, for example, a sugar, a glycolipid, or a protein. Preferred targeting is accomplished by using an antibody to target the retroviral vector.
  • the viral vector may contain a regulatory element such as a tetracycline responsive promoter (inducible or
  • recombinant retroviruses Since recombinant retroviruses are defective, they require assistance in order to produce infectious vector particles. This assistance can be provided, for example, by using helper cell lines that contain plasmids encoding all of the structural genes of the retrovirus under the control of regulatory sequences within
  • Helper cell lines which have deletions of the packaging signal include, but are not limited to ⁇ 2, PA317 and PA 12, for example. These cell lines produce empty virions, since no genome is packaged. If a retroviral vector is introduced into such cells in which the packaging signal is intact, but the structural genes are replaced by other genes of interest, the vector can be packaged and vector virion produced.
  • colloidal dispersion system Another targeted delivery system for PIN I polynucleotides (e.g. , antisense) is a colloidal dispersion system.
  • Colloidal dispersion systems include macromole- cule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes.
  • the preferred colloidal system of this invention is a liposome.
  • Liposomes are artificial membrane vesicles which are useful as delivery vehicles in vitro and in vivo. It has been shown that large unilamellar vesicles (LUV), which range in size from 0.2-4.0 ⁇ m can encapsulate a substantial percentage of an aqueous buffer containing large macromolecules.
  • LUV large unilamellar vesicles
  • RNA, DNA and intact virions can be encapsulated within the aqueous interior and be delivered to cells in a biologically active form (Fraley, et al, Trends Biochem. Sci., 6:11, 1981).
  • liposomes In addition to mammalian cells, liposomes have been used for delivery of polynucleotides in plant, yeast and bacterial cells.
  • a liposome In order for a liposome to be an efficient gene transfer vehicle, the following characteristics should be present: (1 ) encapsulation of the genes of interest at high efficiency while not compromising their biological activity; (2) preferential and substantial binding to a target cell in comparison to non-target cells; (3) delivery of the aqueous contents of the vesicle to the target cell cytoplasm at high efficiency; and (4) accurate and effective expression of genetic information (Mannino, et al, Biotechniques, 6:682, 1988).
  • the targeting of liposomes can be classified based on anatomical and mechanistic factors.
  • Anatomical classification is based on the level of selectivity, for example, organ-specific, cell-specific, and organelle-specific.
  • Mechanistic targeting can be distinguished based upon whether it is passive or active. Passive targeting utilizes the natural tendency of liposomes to distribute to cells of the -27- reticulo-endothelial system (RES) in organs which contain sinusoidal capillaries.
  • RES reticulo-endothelial system
  • Active targeting involves alteration of the liposome by coupling the liposome to a specific ligand such as a monoclonal antibody, sugar, glycolipid, or protein, or by changing the composition or size of the liposome in order to achieve targeting to organs and cell types other than the naturally occurring sites of localization.
  • a specific ligand such as a monoclonal antibody, sugar, glycolipid, or protein
  • the Aspergillus nidulans nimA cDNA was inserted into the pAS2 vector (from S. Elledge; HHMI, Baylor University)(Durfee et al., 1993; Ha ⁇ er, et al., 1993) as fusion to GAL4 DNA-binding domain, resulting in NIMA/PAS2.
  • a HeLa cell two-hybrid cDNA library that contained inserts fused to the transactivation domain of GAL4 (residue 768-881) (obtained from D.
  • K40M NIMA, NIMA280-699, NLK1 and a frame-shift mutant NIMA280-699 FS were also subcloned into the pAS2 vector, followed by cotransformation into Y190 ceils with the isolated cDNA clones.
  • PCR polymerase chain reaction
  • GCGCCTGCAGTATCTATAC/TATGGAATAT/CTGT/(SEQ ID NO: 3) GCGCGGATCCG/AGGTTTCAGAGGT/GTC/TG/AAAG/CAG(SEQID NO: 4) and
  • GCGCGTACCAAGT/ACCACT/CGTAC/TATTATTCC were designed specifically to the catalytic domain V, VII and VIII, respectively, which are conserved among NIMA (Osmani et al., Cell, 51:237, 1988), NPK, a budding yeast NIMA-related kinase (Schweitzer and Philippsen, Mol. Gen. Genet. , 214: 164, 1992) and human HsPK21 (Schultz and Nigg, Cell. Growth Differ ,
  • Human placenta cDNA library (from R. Evans, Salk Institute, La Jolla, CA) was used as a template. The PCR cycle was 1 min at 95°C, 2 min at 42°C and 3 min at 63°C. PCR products with the expected size were subcloned and sequenced. Three potential clones (Ping-1, -44 and -77) were consistently obtained and showed a high sequence identity to NIMA at the deduced amino acids. To obtain the full length cDNA sequence, the Ping-1 PCR product was used as a probe to screen HeLa cell cDNA library filters provided by X.D.
  • NLK1 NIMA-like kinase 1
  • NLK1 was found to be same as NEK2 reported later by Schultz et al. (Cell. Growth Differ., 5:625, 1994)
  • Biotechnology were used at 1 :50, with no significant cross-reactivity. Cells were observed and photographed with a Zeiss microscope, or an MRC-1000 laser- scanning confocal assembly (BioRad).
  • NP40 lysis buffer 50 mM Tris-HCl, pH 8.0, 0.1 % NP40, 200 mM NaCl, 20mM ⁇ -glycerophosphate, 20 mM NaF, 0.1 mM sodium orthovanadate, 50 ⁇ g/ml phenylmethylsufonyl fluoride, 10 ⁇ g/ml leupeptin, 10 ⁇ g/ml aprotinin, and 1 mM DTT or 10 mM ⁇ -mercaptoethanol (in the case of
  • Ni 2+ -NTA agarose precipitation and preclarified with boiled S.aureis or Ni 2+ -NTA agarose (Qiagen). The lysates were then incubated with M2 or 12CA5 or His-Pinl- Ni 2 ) * -NTA agarose. After washing 6 times with lysis buffer, the precipitates were subjected to SDS-polyacrylamide gel electrophoresis and immunoblotting.
  • Pinl could be phosphoryiated, recombinant Pinl protein was added to NIMA kinase reaction mixtures at concentrations up to 0.5mg/ml, with ⁇ -casein and PL1 peptide as positive controls, as described previously (Lu et al., J. Biol. Chem. ,
  • ERK1 MAPK obtained from R. Fukunaga
  • PKA Promega
  • PPlase activity of the purified recombinant proteins was assayed using the procedure of Heitman et al. (Methods, 5: 176, 1993) with the following exceptions.
  • the reaction was carried out 5°C and chymotrypsin was added just before the peptide substrate (N-Succ-Ala-Pro-Phe-p-nitroanilide, Sigma).
  • the cis to trans isomerization was monitored every 6 seconds by a change in the absorbance at 395 nm using DMS 2000 UV and Visible Spectrophotometer (Varian).
  • FKBP, cyclosporin A and FK520, a FK506 derivative were gifts of D. Schultz and C. Zuker; University of California, San Diego.
  • Plasmid pTPI-PINl was made by insertion of an -850 bp BamHI-XhoI fragment of PIN] cDNA (from H20/GADGH) into pJK305-TPI (J. Kamens, Ph.D. Thesis, Harvard University, 1991).
  • PTPI-PIN1 directs the synthesis of the native full length Pinl protein and carries the yeast 2 ⁇ replicator and LEU2 selectable marker.
  • Plasmid YepHESS carries the yeast ESS] gene, a 2 ⁇ replicator and HIS 3 selectable marker (Hanes et al., Yeast, _ ⁇ SS, 1989).
  • a heterozygous ESS I disruption strain MGG3/pSH-U (MATa/MATa ura3/ura3 leu2/ leu2 his 3/ his 3 essI::URA3/ESSI) (Hanes et al., Yeast, 5:55, 1989), was transformed with control vector pJK305-TPI, YepHESS, or pTPI-PINl .
  • Cells were induced to undergo sporulation on 1% potassium acetate plates, tetrads were dissected and haploid segregants grown for 3-4 days at 30 °C on rich medium (YEPD) plates.
  • Cells were serially passaged once per day (1/50 Dilutions) for 6 days in liquid completer synthetic medium lacking leucine; thus maintaining selection for the PIN] -expressing plasmid or control vector (2 ⁇ , LEU2), but not for the ESS1- bearing plasmid (2 ⁇ M, HIS3). Cells were plated and phenotypes of individual colonies scored by replica plating to appropriate selective media.
  • PGAL-PINl directs the synthesis of an HA1 hemagglutinin epitope-tagged version of Pinl that contains a 31 amino-terminal extension (M ASYPYDVPDYASPEF VDPPGSKNSIARGKM) (SEQ ID NO:7) where underlined residues are the HA1 epitope and the normal PIN1 initiator, respec- tively. Other residues derive from polylinker sequences.
  • I- GAL-PIN 1 An integrating version, I- GAL-PIN 1 , was made by removal of the GAL-PIN 1 cassette from pGAL-PINl with Xbal and Sad and reinsertion into the same sites in pRS305 (Sikorski and Hieter, Genetics, 122: 19, 1989).
  • I-GAL-PIN1 carries a LEU2 selectable marker.
  • the original essl ⁇ knockout strain (MGG3/pSH-U) does not grow in galactose, probably due to a gal2 mutation. Therefore, to make a strain (YSH12) that lacks ESSI function and whose growth depends on a galactose-inducible version of PIN], the following scheme was used.
  • a gal strain, FY86 (MATa his3 ⁇ 200 ura3-52 ICU2 ⁇ I) was transformed with integrating vector GAL-PIN 1 (LEU2).
  • Leu + transformants were mated with a haploid MGG3/pSH-U segregant
  • strain YSH12 was grown overnight in galactose or galactose/glucose-containing medium and inoculated
  • Figure 1A shows a ⁇ -Galactosidase activity in the two-hybrid system.
  • Yeast strain Y190 was cotransformed with vectors expressing the three different types of cDNA and different domains of NIMA or NLK1 as indicated, and grown in SC media lacking T ⁇ and Leu, followed by ⁇ -galactosidase activity filter assay as described previously (Durfee, et al. , Genes Dev. , 7:555, 1993).
  • Pin3 interacts with the catalytic domains in both ⁇ IMA and ⁇ LKls
  • Pinl and Pin2 interact with the C-terminal domain of ⁇ IMA.
  • the following examples focus on Pinl as an exemplary Pin protein.
  • the DNA and deduced amino acid sequence of the protein encoded by the longest PIN1 cDNA (l.Okb) is shown in Figure 2A. It encodes a protein of 163 amino acids with the predicted molecular weight of 18,245. When expressed in HeLa cells, Pinl migrated with an apparent size of ⁇ 18 kDa in an SDS-polyacryl- amide gel. Pinl shows a high similarity to the budding yeast Essl protein (Hanes et al., Yeast, 1:55, 1989). Based on recent RNA primer-extension studies and reexamination of the nucleotide sequence of ESS] (Hani et al., FEBS Lett.,
  • the corrected ESS] sequence encodes a 169 amino acid protein with 45% identity to Pinl .
  • Northern blot analysis of human cell line and human fetal liver RNAs showed that there is a single PIN] mRNA of - 1.Okb present in all cell lines and tissue tested ( Figure 3). Fifteen ⁇ g of the indicated total RNAs were run on each lane.
  • HeLa epitheloid carcinoma cells
  • HFF human foreskin fibroblasts
  • Ramos Burkitt lymphoma cells
  • A431 epitheloid carcinoma cells
  • 293 adenovirus El A transformed human embryonic kidney cells
  • U937 histiocytic lymphoma cells
  • Saos-2 osteosarcoma cells
  • U87-MG glioblastoma cells
  • Jurkat T leukemia cell line
  • HFL human fetal liver.
  • Position of the RNA molecular weight standards are on the left.
  • Ess 1 confirm the authenticity of the PIN] open reading frame, although there is no inframe termination condon preceding the putative initiation site.
  • Sequence analysis of six different PIN] cDNA clones identified the points of fusion with the GAL4 activation domain at Arg-(-3), Glu+(+5) and Lys-(+6), indicating that the N-terminal 5 amino acids of Pinl are not necessary for the interaction with NIMA.
  • the deduced Pinl sequence contains two identifiable domains, an N- terminal WW domain and C-terminal putative PPlase domain.
  • the WW domain contains two invariant T ⁇ residues and other residues highly conserved in the WW domains of other proteins including mammalian dystrophins and Yap, and the yeast Rspl and Essl (Sudol et al., 1995).
  • the Cys or His-rich domain flanking the WW domain found in WW domain-containing proteins including Essl is not conserved in the human Pin 1.
  • the C-termin l two-thirds of Pin 1 contains motifs that are characteristic of the recently identified third family of
  • the PPlase domain of Pinl contains three highly observed subdomains, with 45% identity to parvulin and 62% identity to a partial PPlase from A. thaliana.
  • a putative nuclear localization signal is located in the beginning of the PPlase domain, which is also conserved in Essl .
  • Figure 2 A shows the predicted Pinl amino acid sequence is indicated in one-letter code. The fusion points between GAL4 and Pinl in six different isolated clones were: clone H20 at C-9; clone H16, 24 and 38 at G+13; clones H6 and H36 at C+l 5. Underlined residues form a consensus bipartite nuclear localization signal.
  • N- and C-terminal boxes indicate the WW domain and PPlase domain. Nucleotide numbers are on the left and amino acid numbers on right.
  • Figure 2B -37- and 2C show alignments of the WW domain (B) and PPlase Domain (C) in selected proteins. Identical residues are shown in the bottom row. Dashes indicate gaps introduced to make the alignment.
  • Pin 1 was expressed in bacteria with an N-termmal (His) 6 tag and was purified from bacte ⁇ a using a Ni 2+ -NTA agarose column.
  • the insert shows PPlase activity during the first minute of the assay
  • NIMA induces mitotic arrest (Lu and Hunter, Cell, 81:413, 1995a), it is difficult to express sufficient wild type NIMA to detect complex formation, but since Pinl interacted with the kinase-negative K40M NIMA as efficiently as wild type NIMA in the yeast two-hybrid system, we used K40M NIMA and truncated derivative mutant constructs were used.
  • FIGs. 5A-E shows immunoprecipitations to show the interaction between Pinl and the C-terminal noncatalytic domain of NIMAs in HeLa cells.
  • Figure 5A shows the expression and purification of His-Pinl .
  • the PINX cDNA was subcloned into pProExl and the recombinant protein was purified from bacteria using Ni 2+ - NTA agarose column, followed by analyzing the protein on a 15% SDS-containing gel, and Coomassie staining. The positions of His-Pinl and standard size markers are indicated.
  • Figure 5B shows the expression of NIMAs in HeLa cells.
  • tTA- 1 cells were transfected with different FLAG-tagged NIMA constructs and labeled with 5 S Express Label for 24 hr, followed by immunoprecipitation using the M2 mAb.
  • the precipitated proteins were analyzed on a 10% SDS-containing gel followed by autoradiography.
  • the positions of K40M, NIMA, K40M NIMA280, NIMA280-699 and standard size markers are indicated.
  • Figure 5C shows a stable complex between recombinant Pinl and NIMAs. Cell lysates similar to those in panel B were incubated with His-Pinl -Ni 2+ -NTA bead, as indicated in panel A and washed, followed by electrophoresis on an SDS-containing gel and autoradiogra ⁇ phy.
  • Figures 5D and 5E show coimmunoprecipitation of K40M NIMA with Pinl .
  • tTA- 1 cells were cotransfected with FLAG-tagged NIMA and HA-PINX constructs -39- for 24 hr.
  • Cell lysates were immunoprecipitated with the HA-specific 12CA5 mAb, followed by immunoblotting using the M2 mAB specific for the FLAG tag (D) and vice versa (E).
  • His-Pinl was found to be most stable and easiest to purify in large quantities when expressed in bacteria
  • Figure 5A When incubated with 35 S-labeled extracts from cells transiently expressing the full length K40M NIMA, an N-terminal fragment K40M NIMA280, or a C-terminal fragment NIMA280-699 ( Figure 5B), the recombinant His-Pinl beads specifically bound K40M NIMA and NIMA280-699, but not K40M NIMA280 ( Figure 5C). Results also showed that purified GST-NIMA 280-699 directly interacts with purified Pinl in vitro. These results indicate that the recombinant Pinl interacts with C-terminal domain of NIMA in the HeLa cell extracts.
  • Figure 6 shows the colocahzation of Pinl and NIMA and its kinase- negative mutant in HeLa Cells. Twenty four hr after transfection with the vectors expressing HA-tagged Pinl only (right panels) or HA-tagged Pinl and FLAG- tagged NIMA (left panels) or its kinase-negative mutant (K40M NIMA, middle panels), the transfected tTA-1 cells were processed for indirect immunofluores ⁇ cence staining and examined by confocal microscopy.
  • Top panels staining pattern for NIMA or K40M NIMA obtained with FLAG tag-specific M2 mAb or SC-35 obtaining with anti-SC-35 mAb, and then FITC-conjugated IgG 1 -specific secondary antibodies; middle panels: staining pattern for Pinl obtaining with HA tag-specific 12CA5 mAb and Texas-Red-conjugated IgG2b-specific secondary antibodies; bottom panels: double-staining for Pinl and NIMA, Pinl and K40M -41 -
  • NIMA NIMA
  • Pinl and SC-35 displayed by superimposing the respective top and middle images, with yellow color indicating colocahzation. Arrows point to an untransfected cell, indicating very little cross reactivity among the antibodies. Bar, l ⁇ m.
  • HA- tagged PIN] and Flag-tagged nimA or its kinase-negative mutant (K40M nimA) cotransfected into tTA-1 cells and then examined the localization of expressed proteins using tag-specific 12CA5 and M2 mAbs and isotope-specific secondary antibodies.
  • Pinl nuclear substructures were examined to determine whether they were the same as those localized by SC35 or PML.
  • cells expressing Pinl were doubly stained with 12CA5 and anti- splicing factor SC35 or anti-PML antibodies, the speckles displayed by Pinl were found to be the same as those detected using anti-SC35 ( Figure 6, right panels), but not anti-PML antibodies.
  • Figures 7A-7D shows overexpression of PINX delays NIMA-induced mitotic arrest and induces a specific G2 arrest in HeLa cells.
  • Figure 7A shows the results of tTA- 1 cells cotransfected with nimA and PINX or control vector. At the times indicated, cells were fixed and doubly labeled with M2 mAb and Hoechst dye, followed by scoring for the percentage of cells with rounding and chromatin condensation in a sample of at least 250 NIMA-expressing cells. The data are the average from two independent experiments.
  • Figures 7B, C, and D show after transfection with PINX expression vector or the control vector for 48 hr, tTA-1 cells were stained with the 12CA5 mAb and then with FITC-conjugated secondary antibodies and propidium iodide, followed by
  • NIMA-expressing cells were not completely rounded up and their chromatin was not as condensed as those in cells expressing only NIMA ( Figure 5 left panels and 7A), although by 36 hr all the NIMA-expressing cells displayed the mitotic phenotype
  • plasmids that express PIN] under the control of a constitutive yeast promoter were introduced into diploid cells in which one copy of ESS] is disrupted (essl::URA3/ESS]). Cells were induced to undergo sporulation, tetrads were dissected, and viability of the resulting haploid spores was scored (Table 1A). As expected, tetrads derived from cells transformed with vector alone showed a 2:2 segregation for spore viability (viable:inviable). In contrast, -25% of the tetrads from cells transformed with a
  • PIN] -expressing plasmid showed 4:0 segregation for spore viability, indicating that PIN] complements the essl mutant to allow spore outgrowth and haploid cell viability. PIN] did not complement essl mutant to allow spore outgrowth and haploid cell viability. PIN] did not complement essl ' mutants quite as efficiently as ESS] itself, where more than half the tetrads from cells bearing an ESS1 plasmid showed a 4:0 segregation from spore viability. Growth curves revealed that PIN1- expressing ess] ' cells had doubling times only slightly longer than control cells.
  • human PIN! can functionally substitute for ESS] in haploid yeast cells.
  • the PIN] expression plasmid was introduced into diploid cells in which both chromosomal copies ofESSl are disrupted (essl : : URA3/essl: : UR ⁇ 3) but that remain viable by maintaining a plasmid-borne copy of ESS If PIN 1 functionally substitute for ESS], then it should be possible to cure cells of the ESS1 plasmid.
  • Table IB shows that cells serially passaged in media selecting only for the PIN] plasmid (LEU2) lost the ESS1 plasmid (HIS3) about 12% of the time, whereas cells containing a control vector did not lose the ESS1 plasmid.
  • human PIN] can functionally substitute for ESS1 in diploid yeast cells.
  • the heterozygous disruption strain MGGS3/pSH-U was transformed with an £551- or PINX -expression vector plasmid or control vector. Cells were induced to undergo sporulation and tetrads were dissected. The number of viable spores included only those that showed proper segregation of independent markers. As expected, almost all segregants that were ura * [i.e. carried the ess /::URA3 knockout allele) were also his * or leu * indicating the presence of the £55 or PIN1 containing plasmid, respectively.
  • B. PIN] permits loss of £55/-containing plasmids in a essl diploid knockout strain.
  • a diploid disruption strain (&s.5l _:URA3/e_j_jl ::URA3) carrying the £551 -plasmid ( //53) was transformed with the vector alone (LEU2) or a PINX -expression vector plasmid (LEU2).
  • LEU2 the vector alone
  • PINX -expression vector plasmid LEU2
  • Cells were serially passaged in leucine-deficient media that maintains selection for control or the PINX plasmid but not for the £551 plasmid. Cells were plated and phenotypes of individual colonies scored by repiica plating to appropriate selective media.
  • Loss of the £551 plasmid is detected by loss of the his * phenotype, while the presence of PINX is detected by a leu * phenotype.
  • a ura * phenotype confirms the presence of the knockout allele (essX ::URA3).
  • Pinl -expressing strains grew normally in inducing media (galactose) or noninducing media (galactose/glucose, with a basal level of PIN 1 expression), but did not grow in repressing media (glucose).
  • Figure 8 shows depletion of Pinl /Essl results in mitotic arrest and nuclear fragmentation in yeast.
  • a Pinl -dependent strain (YSH12.4) was shifted from inducing media to repressing media, harvested and fixed with 70% ethanol at the times indicated. The cells were stained with DAPI or propidium iodide, followed by videomicroscopy under Nomarksi (DIC) or fluorescent (DAPI) illumination, or
  • the bar is 10 ⁇ m and the inserts show a higher magnification of a representative cell.
  • a Pi l -dependent strain (YSH 12.4) was shifted from inducing media to repressing media, harvested and fixed with ethanol at the times indicated.
  • Bud size distribution was scored under Nomarski illumination after cells were stained with DAPI and sonicated. About 500 cells were counted for each point.
  • the present invention identifies the first NIMA- interacting protein of human origin Pinl .
  • Pinl contains an N-terminal WW domain and a C-terminal PPlase domain, and has PPlase activity in vitro.
  • Pinl is conserved from yeast to human; Pinl /Essl is the first PPlase known to be essential for life. Pinl interacts with the C-terminal noncatalytic domain of NIMA and colocalizes with NIMA in a defined nuclear substructure in HeLa cells.
  • Pinl contains two identifiable domains.
  • the C- terminal two-thirds of Pin 1 contains residues that are highly conserved in the newly delineated family of PPIases that includes parvulin, PrsA, SurA, NifM, PrtM, Cbf2 and Essl (Rudd, et al., TIBS, 20:12, 1995).
  • PrsA, SurA, NifM, PrtM have been shown to be involved in maturation and/or transport of specific proteins or protein classes.
  • Parvulin originally identified during a chromatographic purification procedure, is the prototype of this family; it contains 96 amino acids and catalyzes the cis/trans isomerization of X-Pro peptide bonds, even in the presence of immunosuppressive drugs (Rahfeld et al., FEBS Lett.. 352:180. 1994a and FEBS Lett., 343:65. 1994b).
  • the homology between Pinl and parvulin spans almost the entire parvulin molecule, strongly suggesting that Pinl is a PPlase.
  • the in vitro PPlase assay shown in the above Examples confirms that Pinl can indeed catalyze the cis/trans isomerization of peptidyl-prolyl peptide bonds.
  • AGC AGT GGT GGC AAA AAC GGG CAG GGG GAG CCT GCC AGG GTC CGC TGC 195 Ser Ser Gly Gly Lys Asn Gly Gin Gly Glu Pro Ala Arg Val Arg Cys
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic) IMMEDIATE SOURCE- (B) CLONE. nlkl(2)

Abstract

A novel class of NIMA interacting proteins (PIN), exemplified by Pin1, is provided. Pin1 induces a G2 arrest and delays NIMA-induced mitosis when overexpressed, and triggers mitotic arrest and DNA fragmentation when depleted. Methods of identifying other Pin proteins and Pin-interacting proteins and identifying compositions which affect Pin activity or expression are also provided.

Description

NIMA INTERACTING PROTEINS
Statement as to Federally Sponsored Research The present invention was made with government support from United States Public Health Services Grant No. CA 37980. The government has certain rights in this invention.
Field of the Invention
The present invention relates generally to the eukaryotic cell cycle and specifically to a novel class of proteins that interact with NIMA protein kinase in the NIMA mitotic pathway.
Background of the Invention
The CDC2 kinase associated with its cyclin partners has been shown to play an important role during G2/M progression in eukaryotic cells. However, recent studies demonstrate that activation of the CDC2 kinase itself is not sufficient to trigger mitosis in some eukaryotic cells such as those in Saccharomyces cerevisiae
(Amon, et al.. Nature, 355:368, 1992; Sorger and Murray, Nature, 355:365, 1992; Stueland et al., Mol. Cell. Bio., 13_:3744, 1993) and Aspergillus nidulans (Osmani, et al., Cell, 67:283, 1991a). Furthermore, detailed analysis of mouse oocyte maturation reveals that CDC2 histone HI kinase activity does not increase during the G2 M transition as indicated by germinal vesicle breakdown (GVBD) (Choi, et al., Development, 111:789, 1991 ; Jung, et al., Int. J. Dev. Biol., 37:595. 1993; Gavin, et al., J. Cell Sci., 107:275, 1994). These results suggest that there might be other mitotic activation pathway(s) remaining to be identified.
Recent studies have identified a novel mitotic kinase, NIMA, encoded by the Aspergillus nimA gene (Osmani, et al., Cell, 53:237, 1988). NIMA kinase activity is tightly regulated during the nuclear division cycle, peaking in late G2 and M. Overexpression of NIMA promotes entry of Aspergillus cells into M (Osmani, et al.. Cell, 51:237, 1988; Lu and Means, EMBOJ., 13:2103, 1994). Thus, NIMA is important for progression into mitosis in Aspergillus.
NIMA is a protein-Ser/Thr kinase, biochemically distinct from other protein kinases. and its phosphotransferase activity is regulated by Ser Thr phosphorylation
. It has recently been shown that the NIMA mitotic pathway is not restricted to Aspergillus, but also exists in vertebrate cells (Lu and Hunter, Cell, 81 :413. 1995a). In Xenopus oocytes, NIMA induces germinal vesicle breakdown without activating Mos, CDC2 or MAP kinase. In HeLa cells, NIMA induces mitotic events without activating CDC2, whereas dominant-negative NIMA mutants cause a specific G2 arrest. In addition, O'Connell, et al (EMBOJ., 11:4926, 1 94) have also demonstrated that NIMA induce premature chromatin condensation in fission yeast and HeLa cells. These results reveal the existence of a NIMA-like mitotic pathway in other eukaryotic cells. Peptidyl-prolyl cis/trans isomerases (PPIases, proline isomerases) are ubiquitously expressed enzymes catalyzing the cis/trans isomerization of the peptidyl-prolyl peptide bond which can be the rate-limiting step in protein folding or assembly under some circumstances. Cyclophilins and FK506-binding proteins (FKBPs) are two well characterized families of PPIases that share little if any amino acid similarity to each other. However, the members of each family contain a core structure that has been highly conserved from prokaryotes to eukaryotes. The importance of these PPIases is highlighted by the findings that cyclophilin and FK506-binding proteins are the targets of immunosuppressive drugs cyclosporin A and FK506, respectively, and play an important role in cell signaling in T cell activation, although none of these genes have been shown to be essential for life
(for review see Schreiber, Science, 211:283, 1991 ; Fruman, et al., FASEB J, 8:391 , 1994).
The recent discovery of parvulin has led to the identification of a third family of PPIases, which show little homology with either cyclophilins or FKBPs and is not sensitive to the immunosuppressive drugs (Rahfeld, et al., FEBS Lett. , 352: 180. 1994a and FEBS Lett. , 243:65, 1994b). A sequence homology search identified several other members of this family including those involved in protein maturation and/or transport and the ESS1 gene (Rudd, et al., TIBS, 20:12, 1995). ESS1 is an essential gene for the growth in budding yeast and previous results suggested that it may be required at later stages of the cell cycle (Hanes, et al..
Yeast, 5:55, 1989). ESS] was recently reisolated as PTFl in a screen for genes involved in mRNA 3' end maturation. Ptfl was shown to contain a putative PPlase domain, but PPlase activity could not be demonstrated (Hani, et al., FEBS Lett., 365: 198. 1995). So far, none of the PPIases have been shown to be specifically involved in cell cycle control.
There is a need to identify components of the mammalian NIMA mitotic pathway in order to identify genes essential for life. Identification of such genes has several utilities including the identification of appropriate therapeutic targets, candidate genes for gene therapy (e.g. , gene replacement), mapping locations of disease-associated genes, and for the identification of diagnostic and prognostic indicator genes, for example.
SUMMARY OF THE INVENTION
The present invention provides a novel class of proteins that associate with NIMA protein kinase. Some of these proteins are characterized by inhibiting the mitosis promoting function of NIMA when overexpressed and inducing mitotic arrest and nuclear fragmentation when depleted.
In a first embodiment, the invention provides an exemplary NIMA associated protein called "protein interacting with NIMA" (Pinl). Pinl has C- terminal peptidyl-propyl cis/trans isomerase activity and contains a conserved N- terminal tryptophan domain (WW domain) thought to mediate protein-protein interactions. Also included are polynucleotides encoding PIN proteins.
In another embodiment, the invention provides a method for identifying a protein that inhibits the mitosis promoting function of NIMA protein kinase. The method is based on a genetic system designed to detect protein-protein interac¬ tions. The method comprises culturing transformed cells containing the following: a nucleic acid construct comprising a DNA binding domain operatively associated with the coding sequence of NIMA, or functional fragments thereof; a nucleic acid library, wherein each member of said library comprises a transactivation domain operatively associated with a protein encoding sequence; and a nucleic acid reporter construct comprising a response element for the DNA binding domain operatively associated with a reporter gene, and monitoring for evidence of expression of reporter gene.
In yet another embodiment, the invention provides a method for controlling the growth of a cell comprising contacting the cell with a composition which modulates Pinl activity. For example, an inhibitor of Pinl activity such as a PPlase inhibitor or an anti-Pin 1 antibody, or an inhibitor of PIN\ expression such as an antisense nucleotide sequence or a ribozyme, can be used to control growth of a cell. Alternatively, Pinl activity can be increased by an activator or PIN\ expression can be increased by an enhancer, for example. Finally, the invention provides a method for identifying a protein or other composition (e.g. , drug or other small molecule) that associates with and/or modulates Pinl protein activity or PIN\ gene expression.
BRIEF DESCRIPTION OF THE DRAWINGS
Figures 1 A and IB show human cDNA clones encoding proteins interacting with
NIMA (Pins).
Figure 1 A shows a β-Galactosidase Activity in the two-hybrid system. Figure IB shows a comparison of NIMA and NLK1.
Figures 2A-2C show the cDNA and deduced amino acid sequences of human PIN\ and homologies with other WW domain proteins and PPIases. Figure 2 A shows the predicted Pinl amino acid sequence is indicated in one-letter code. The fusion points between GAL4 and Pinl in six different isolated clones were: clone H20 at C-9; clone H16, 24 and 38 at G+13; clones H6 and H36 at C+15. Underlined residues form a consensus bipartite nuclear localization signal. The N- and C- terminal boxes indicate the WW domain and PPlase domain. Nucleotide numbers are on the left and amino acid numbers on right.
Figure 2B and 2C show alignments of the WW domain (B) and PPlase Domain (C) in selected proteins. Identical residues are shown in the bottom row. Dashes indicate gaps introduced to make the alignment. Cbf2, cell binding factor 2; SC, S. cerevisiae; EC, E. coli; BS, B. subtilis; CJ, C jejuni; AT, A. thaliana.
Figure 3 shows an analysis of PINX mRNA expression in human cell lines and human fetal liver. HeLa, epitheloid carcinoma cells; HFF, human foreskin fibroblasts; Ramos, Burkitt lymphoma cells; A431 , epitheloid carcinoma cells; 293 ; adenovirus E1A transformed human embryonic kidney cells; U937, histiocytic lymphoma cells; Saos-2, osteosarcoma cells; U87-MG, glioblastoma cells; Jurkat, T leukemia cell line and HFL, human fetal liver. Position of the RNA molecular weight standards (Promega) are on the left.
Figure 4 shows the PPlase activity of Pinl . Different concentrations of Pinl were used as indicated, with the recombinant FK506-binding protein (FKBP) PPlase being used as a positive control. The control sample contained all the ingredients except the PPlase. The insert shows PPlase activity during the first min of assay.
Figures 5A-5E show immunoprecipitations to show the interaction between Pinl and the C-terminal noncatalytic domain of NIMAs in HeLa cells. Figure 5A shows the expression and purification of His-Pinl . The PIN\ cDNA was subcloned into pProExl and the recombinant protein was purified from bacteria using Ni2+-NTA agarose column, followed by analyzing the protein on a 15% SDS-containing gel, and Coomassie staining. The positions of His-Pinl and standard size markers are indicated. Figure 5B shows the expression of NIMAs in HeLa cells. tTA-1 cells were transfected with different FLAG-tagged NIMA constructs and labeled with 35S Express Label for 24 hr. followed by immunoprecipitation using the M2 mAb.
The precipitated proteins were analyzed on a 10% SDS-containing gel followed by autoradiography. The positions of K40M, NIMA, K40M NIMA280, NIMA280- 699 and standard size markers are indicated. Figure 5C shows a stable complex between recombinant Pinl and NIMAs. Cell lysates similar to those in panel B were incubated with His-Pinl -Ni2+-NTA bead, as indicated in panel A and washed, followed by electrophoresis on an SDS-containing gel and autoradiography. Figures 5 D and 5E show coimmunoprecipitation of K40M NIMA with Pinl . tTA- 1 cells were cotransfected with FLAG-tagged NIMA and HA-/JN1 constructs for 24 hr. Cell lysates were immunoprecipitated with the HA-specific 12CA5 mAb, followed by immunoblotting using the M2 mAB specific for the FLAG tag (D) and vice versa (E). Figure 6 shows the colocahzation of Pinl and NIMA and its kinase-negative mutant in HeLa Cells. Twenty four hr after transfection with the vectors expressing HA-tagged Pinl only (right panels) or HA-tagged Pinl and FLAG- tagged NIMA (left panels) or its kinase-negative mutant (K40M NIMA, middle panels), the transfected tTA-1 cells were processed for indirect immunofluores- cence staining and examined by confocal microscopy. Top panels: staining pattern for NIMA or K40M NIMA obtained with FLAG tag-specific M2 mAb or SC-35 obtaining with anti-SC-35 mAb, and then FITC-conjugated IgGl -specific secondary antibodies; middle panels: staining pattern for Pinl obtaining with HA tag-specific 12CA5 mAb and Texas-Red-conjugated IgG2b-specific secondary antibodies; bottom panels: double-staining for Pinl and NIMA, Pinl and K40M NIMA, or Pinl and SC-35 displayed by superimposing the respective top and middle images, with yellow color indicating colocahzation. Arrows point to an untransfected cell, indicating very little cross reactivity among the antibodies. Bar, l μm.
Figures 7A-7D show overexpression of PIN\ delays NIMA-induced mitotic arrest and induces a specific G2 arrest in HeLa cells. Figure 7A shows the results of tTA- 1 cells cotransfected with nimA and PINX or control vector. At the times indicated, cells were fixed and doubly labeled with M2 mAb and Hoechst dye, followed by scoring for the percentage of cells with rounding and chromatin condensation in a sample of at least 250 NIMA-expressing cells. The data are the average from two independent experiments.
Figures 7B, C, and D show after transfection with PINX expression vector or the control vector for 48 hr, tTA-1 cells were stained with the 12CA5 mAb and then with FITC-conjugated secondary antibodies and propidium iodide, followed by
FACS analysis. Based on the FITC intensity of the PINX -transfected cells, cells were divided into two populations with 12CA5-negative (B) or -positive cells (C), and cell cycle profiles were determined to compare with those in total vector- transfected cells (A). Figure 8 shows depletion of Pinl /Ess 1 results in mitotic arrest and nuclear fragmentation in yeast. A Pinl -dependent strain (YSH12.4) was shifted from inducing media to repressing media, harvested and fixed with 70% ethanol at the times indicated. The cells were stained with DAPI or propidium iodide, followed by videomicroscopy under Nomarksi (DIC) or fluorescent (DAPI) illumination, or FACS analysis, respectively. The bar is 10 μm and the inserts show a higher magnification of a representative cell.
DESCRIPTION OF THE PREFERRED EMBODIMENTS
A NIMA-like pathway is required for the G2/M transition in Aspergillus nidulans and human cells. The present invention provides the first NIMA- interacting protein, Pinl, of mammalian origin, and methods for identification of other NIMA-interacting proteins. Overexpression of PINX and Pinl activity, induces a specific G2 arrest and delays NIMA-induced mitosis, while depletion of Pinl triggers mitotic arrest and nuclear fragmentation in budding yeast.
In a first embodiment, the invention provides an isolated mammalian protein characterized as associating with NIMA protein kinase, inhibiting the mitosis promoting function of NIMA when overexpressed, and inducing entry of cells into mitosis when depleted. The C-terminal domain of such a protein catalyzes the cis/trans isomerization of peptidyl-propyl peptide bonds. The N-terminal domain associates with NIMA and contains at least two conserved tryptophan residues (WW domain). While the exemplary polynucleotides and polypeptides of the invention are directed to Pinl , it is understood that any PIN polynucleotide or Pin protein can now be identified and characterized by the methods described herein.
In a preferred embodiment, the present invention provides a substantially pure NIMA-interacting protein (Pinl) characterized by having a molecular weight of about 18kD as determined by reducing SDS-PAGE, having peptidyl-propyl cis/trans isomerase activity, associating with NIMA protein kinase, and having essentially the amino acid sequence of SEQ ID NO:2. The term "substantially pure" as used herein refers to Pinl which is substantially free of other proteins, lipids, carbohydrates or other materials with which it is naturally associated. One skilled in the art can purify Pinl using standard techniques for protein purification. The substantially pure polypeptide will yield a single maj or band on a non-reducing polyacrylamide gel. The purity of the Pinl polypeptide can also be determined by amino-terminal amino acid sequence analysis. Pinl polypeptide includes functional fragments of the polypeptide, as long as the activity of Pinl remains. Smaller peptides containing one of the biological activies of Pinl are therefore included in the invention. Such peptides include immunologically reactive peptides capable of inducing antibody production. The preferred Pinl of the invention is derived from a human cell.
The invention provides isolated polynucleotides encoding Pin polypeptides, including Pinl . These polynucleotides include DNA, cDNA and RNA sequences which encode Pinl . It is understood that all polynucleotides encoding all or a portion of Pinl are also included herein, as long as they encode a polypeptide with
Pinl activity. Such polynucleotides include naturally occurring, synthetic, and intentionally manipulated polynucleotides. For example, PINX polynucleotide may be subjected to site-directed mutagenesis. The polynucleotide sequence for PINX also includes antisense sequences. The polynucleotides of the invention include sequences that are degenerate as a result of the genetic code. There are 20 natural amino acids, most of which are specified by more than one codon. Therefore, all degenerate nucleotide sequences are included in the invention as long as the amino acid sequence of Pinl polypeptide encoded by the nucleotide sequence is functionally unchanged. Polynucleotides of the invention include variations thereof which encode the same amino acid sequence, but employ different codons for some of the amino acids, or splice variant nucleotide sequences thereof.
Specifically disclosed herein is a DNA sequence encoding the human PIN1 gene. The sequence contains an open reading frame encoding a polypeptide 163 amino acids in length. The human PIN1 initiator methionine codon shown in
FIGURE 2A at position 25-27 is the first ATG codon. Preferably, the human
PIN1 nucleotide sequence is SEQ ID NO: 1 and the deduced amino acid sequence is preferably SEQ ID NO:2 (FIGURE 2 A). The polynucleotide encoding Pinl includes SEQ ID NO: 1 as well as nucleic acid sequences complementary to SEQ ID NO: 1 (FIGURE 2A). A complementary sequence may include an antisense nucleotide. When the sequence is RNA, the deoxynucleotides A, G, C, and T of SEQ ID NO:l is replaced by ribonucleotides
A, G, C, and U, respectively. Also included in the invention are fragments of the above-described nucleic acid sequences that are at least 15 bases in length, which is sufficient to permit the fragment to selectively hybridize to DNA that encodes the protein of SEQ ID NO:2 under physiological conditions. Specifically, the term "selectively hybridize" means that the fragments should hybridize to DNA encoding Pinl protein under moderate to highly stringent conditions. Minor modifications of the Pinl primary amino acid sequence may result in proteins which have substantially equivalent activity as compared to the Pinl polypeptide described herein. Such proteins include those as defined by the term "having substantially the amino acid sequence of SEQ ID NO:2". Such modifications may be deliberate, as by site-directed mutagenesis, or may be spontaneous. All of the polypeptides produced by these modifications are included herein as long as the biological activity of PPW still exists. Further, deletion of one or more amino acids can also result in a modification of the structure of the resultant molecule without significantly altering its biological activity. This can lead to the development of a smaller active molecule which would have broader utility. For example, one can remove amino or carboxy terminal amino acids which are not required for Pinl biological activity.
The Pinl polypeptide of the invention encoded by the polynucleotide of the invention includes the disclosed sequence (SEQ ID NO:2; FIGURE 2A) and conservative variations thereof. The term "conservative variation" as used herein denotes the replacement of an amino acid residue by another, biologically similar residue. Examples of conservative variations include the substitution of one hydrophobic residue such as isoleucine, valine, leucine or methionine for another, or the substitution of one polar residue for another, such as the substitution of arginine for lysine, glutamic for aspartic acid, or glutamine for asparagine, and the like. The term "conservative variation" also includes the use of a substituted amino acid in place of an unsubstituted parent amino acid provided that antibodies raised to the substituted polypeptide also immunoreact with the unsubstituted polypep¬ tide.
DNA sequences of the invention can be obtained by several methods. For example, the DNA can be isolated using hybridization techniques which are well known in the art. These include, but are not limited to: 1 ) hybridization of genomic or cDNA libraries with probes to detect homologous nucleotide sequences, 2) polymerase chain reaction (PCR) on genomic DNA or cDNA using primers capable of annealing to the DNA sequence of interest, and 3) antibody screening of expression libraries to detect cloned DNA fragments with shared structural features.
Preferably the PIN (e.g., PIN1) polynucleotide of the invention is derived from a mammalian organism, and most preferably from human. Screening proc¬ edures which rely on nucleic acid hybridization make it possible to isolate any gene sequence from any organism, provided the appropriate probe is available.
Oligonucleotide probes, which correspond to a part of the sequence encoding the protein in question, can be synthesized chemically. This requires that short, oligopeptide stretches of amino acid sequence must be known. The DNA sequence encoding the protein can be deduced from the genetic code, however, the degeneracy of the code must be taken into account. It is possible to perform a mixed addition reaction when the sequence is degenerate. This includes a heterogeneous mixture of denatured double-stranded DNA. For such screening, hybridization is preferably performed on either single-stranded DNA or denatured double-stranded DNA. Hybridization is particularly useful in the detection of cDNA clones derived from sources where an extremely low amount of mRNA sequences relating to the polypeptide of interest are present. In other words, by using stringent hybridization conditions directed to avoid non-specific binding, it is possible, for example, to allow the autoradiographic visualization of a specific cDNA clone by the hybridization of the target DNA to that single probe in the mixture which is its complete complement (Wallace, et al, Nucl. Acid Res., 9:879,
1981 ; Maniatis, et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, N.Y. 1989).
The development of specific DNA sequences encoding PIN can also be obtained by: 1) isolation of double-stranded DNA sequences from the genomic DNA; 2) chemical manufacture of a DNA sequence to provide the necessary codons for the polypeptide of interest; 3) in vitro synthesis of a double-stranded DNA sequence by reverse transcription of mRNA isolated from a eukaryotic donor cell; and PCR of genomic DNA or cDNA using primers capable of annealing to the DNA sequence of interest. In the latter case, a double-stranded DNA complement of mRNA is eventually formed which is generally referred to as cDNA.
Of the three above-noted methods for developing specific DNA sequences for use in recombinant procedures, the isolation of genomic DNA isolates is the least common. This is especially true when it is desirable to obtain the microbial expression of mammalian polypeptides due to the possible presence of introns. The synthesis of DNA sequences is frequently the method of choice when the entire sequence of amino acid residues of the desired polypeptide product is known. When the entire sequence of amino acid residues of the desired polypep¬ tide is not known, the direct synthesis of DNA sequences is r< A possible and the method of choice is the synthesis of cDNA sequences. Among the standard procedures for isolating cDNA sequences of interest is the formation of plasmid- or phage-carrying cDNA libraries which are derived from reverse transcription of mRNA which is abundant in donor cells that have a high level of genetic expres¬ sion. When used in combination with polymerase chain reaction technology, even rare expression products can be cloned. In those cases where significant portions of the amino acid sequence of the polypeptide are known, the production of labeled single or double-stranded DNA or RNA probe sequences duplicating a sequence putatively present in the target cDNA may be employed in DNA/DNA hybridiza¬ tion procedures which are carried out on cloned copies of the cDN A which have been denatured into a single-stranded form (Jay, et al., Nucl. Acid Res., 11 :2325. 1983).
A cDNA expression library, such as lambda gtl 1 , can be screened indirectly for PIN peptides having at least one epitope, using antibodies specific for P1_N. Such antibodies can be either polyclonally or monoclonally derived and used to detect expression product indicative of the presence of PIN 1 cDNA. DNA sequences encoding Pinl can be expressed in vitro by DNA transfer into a suitable host cell. "Host cells" are cells in which a vector can be propagated and its DNA expressed. The term also includes any progeny of the subject host cell. It is understood that all progeny may not be identical to the parental cell since there may be mutations that occur during replication. However, such progeny are included when the term "host cell" is used. Methods of stable transfer, meaning that the foreign DNA is continuously maintained in the host, are known in the art.
In the present invention, the PIN1 polynucleotide sequences may be inserted into a recombinant expression vector. The term "recombinant expression vector" refers to a plasmid, virus or other vehicle known in the art that has been manipulated by insertion or incoφoration of the PIN genetic sequences. Such expression vectors contain a promoter sequence which facilitates the efficient transcription of the inserted genetic sequence of the host. The expression vector typically contains an origin of replication, a promoter, as well as specific genes which allow phenotypic selection of the transformed cells. Vectors suitable for use in the present invention include, but are not limited to the T7-based expression vector for expression in bacteria (Rosenberg, et al., Gene, 56: 125, 1987), the pMSXND expression vector for expression in mammalian cells (Lee and Nathans, J. Biol. Chem., 263:3521. 1988) and baculovirus-derived vectors for expression in insect cells. The DNA segment can be present in the vector operably linked to regulatory elements, for example, a promoter (e.g., T7, metal lothionein I, tetracycline responsive promoter or polyhedrin promoters).
Polynucleotide sequences encoding Pinl can be expressed in either prokaryotes or eukaryotes. Hosts can include microbial, yeast, insect and mammalian organisms. Methods of expressing DNA sequences having eukaryotic or viral sequences in prokaryotes are well known in the art. Biologically functional viral and plasmid DNA vectors capable of expression and replication in a host are known in the art. Such vectors are used to incorporate DNA sequences of the invention. Transformation of a host cell with recombinant DNA may be carried out by conventional techniques as are well known to those skilled in the art. Where the host is prokaryotic, such as E. coli, competent cells which are capable of DNA uptake can be prepared from cells harvested after exponential growth phase and subsequently treated by the CaCL method using procedures well known in the art.
Alternatively, MgCI 2 or RbCl can be used. Transformation can also be performed after forming a protoplast of the host cell if desired.
When the host is a eukaryote, such methods of transfection of DNA as calcium phosphate co-precipitates, conventional mechanical procedures such as microinjection, electroporation, insertion of a plasmid encased in liposomes, or virus vectors may be used. Eukaryotic cells can also be cotransformed with DNA sequences encoding the PIN of the invention, and a second foreign DNA molecule encoding a selectable phenotype, such as the herpes simplex thymidine kinase gene. Another method is to use a eukaryotic viral vector, such as simian virus 40 (SV40) or bovine papilloma virus, to transiently infect or transform eukaryotic cells and express the protein, (see for example, Eukaryotic Viral Vectors, Cold Spring Harbor Laboratory, Gluzman ed., 1982).
Isolation and purification of microbial expressed polypeptide, or fragments thereof, provided by the invention, may be carried out by conventional means including preparative chromatography and immunological separations involving monoclonal or polyclonal antibodies. For example, one of skill in the art could use (His)6 tag affinity purification as described in the EXAMPLES herein.
The Pin polypeptides of the invention can also be used to produce antibodies which are immunoreactive or bind to epitopes of the Pin polypeptides. Antibody which consists essentially of pooled monoclonal antibodies with different epitopic specificities, as well as distinct monoclonal antibody preparations arc provided. Monoclonal antibodies are made from antigen containing fragments of the protein by methods well known in the art (Kohler, et al, Nature, 256:495. 1975; Current Protocols in Molecular Biology, Ausubel, et al., ed., 1989). The term "antibody" as used in this invention includes intact molecules as well as fragments thereof, such as Fab, F(ab')2, and Fv which are capable of binding the epitopic determinant. These antibody fragments retain some ability to selectively bind with its antigen or receptor and are defined as follows: (1 ) Fab, the fragment which contains a monovalent antigen-binding fragment of an antibody molecule can be produced by digestion of whole antibody with the enzyme papain to yield an intact light chain and a portion of one heavy chain;
(2) Fab', the fragment of an antibody molecule can be obtained by treating whole antibody with pepsin, followed by reduction, to yield an intact light chain and a portion of the heavy chain; two Fab' fragments are obtained per antibody molecule;
(3) (Fab')2, the fragment of the antibody that can be obtained by treating whole antibody with the enzyme pepsin without subsequent reduction; F(ab')2 is a dimer of two Fab' fragments held together by two disulfide bonds;
(4) Fv, defined as a genetically engineered fragment containing the variable region of the light chain and the variable region of the heavy chain expressed as two chains; and
(5) Single chain antibody ("SCA"), defined as a genetically engineered molecule containing the variable region of the light chain, the variable region of the heavy chain, linked by a suitable polypeptide linker as a genetically fused single chain molecule.
Methods of making these fragments are known in the art. (See for example, Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, New York (1988), incorporated herein by reference).
As used in this invention, the term "epitope" means any antigenic determinant on an antigen to which the paratope of an antibody binds. Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimension- al structural characteristics, as well as specific charge characteristics. Antibodies which bind to the Pinl polypeptide of the invention can be prepared using an intact polypeptide or fragments containing small peptides of interest as the immunizing antigen. For example, it may be desirable to produce antibodies that specifically bind to the N- or C-terminal domains of Pinl . The polypeptide or a peptide used to immunize an animal can be derived from translated cDNA or chemical synthesis which can be conjugated to a carrier protein, if desired. Such commonly used carriers which are chemically coupled to the peptide include keyhole limpet hemocyanin (KLH), thyroglobulin, bovine serum albumin (BSA), and tetanus toxoid. The coupled peptide is then used to immunize the animal (e.g., a mouse, a rat, or a rabbit).
If desired, polyclonal or monoclonal antibodies can be further purified, for example, by binding to and elution from a matrix to which the polypeptide or a peptide to which the antibodies were raised is bound. Those of skill in the art will know of various techniques common in the immunology arts for purification and/or concentration of polyclonal antibodies, as well as monoclonal antibodies (See for example, Coligan, et αl , Unit 9, Current Protocols in Immunology, Wiley I- nterscience, 1994, incorporated by reference).
It is also possible to use the anti-idiotype technology to produce m- onoclonal antibodies which mimic an epitope. For example, an anti-idiotypic monoclonal antibody made to a first monoclonal antibody will have a binding domain in the hypervariable region which is the "image" of the epitope bound by the first monoclonal antibody.
In another embodiment, the invention provides a method for identifying a protein that inhibits the mitosis promoting function of NIMA protein kinase. The method comprises culturing transformed cells containing the following: a nucleic acid construct comprising a DNA binding domain operably associated with the coding sequence of NIMA, or functional fragments thereof; a nucleic acid library, wherein each member of said library comprises a transactivation domain operably associated with a protein encoding sequence; and a nucleic acid reporter construct comprising a response element for the DNA binding domain operably associated with a reporter gene, and monitoring for evidence of expression of reporter gene.
The term "inhibits" refers to a reduction in the mitosis promoting function of NIMA protein kinase. The "mitosis promoting function of NIMA" refers to the ability of NIMA to promote the progression of the G2/M transition in a cell. The term "operably associated" refers to functional linkage between the promoter or regulatory sequence and the controlled nucleic acid sequence; the controlled sequence and regulatory sequence, or promoter are typically covalently joined, preferably by conventional phosphodiester bonds. The method of the invention comprises culturing cells, under suitable conditions, transformed by standard methods in the art and as described above. The transformed cells contain the following: a nucleic acid c 1 istruct comprising a DNA binding domain operably associated with the coding sequence of NIMA, or functional fragments thereof. As used herein, the term "DNA binding domain" refers to a nucleic acid sequence which contains a recognition site for a protein that binds to specific DNA sequences. The coding sequence of NIMA includes nucleic acid sequence that encodes a protein having the biological activity of NIMA as described herein. The method includes "functional fragments" of NIMA that are less than the full length coding sequence but which encode a protein having NIMA biological activity (e.g., Ser/Thr protein kinase).
The transformed cells also contain a nucleic acid library, wherein each member of the library comprises a transactivation domain operably associated with a protein encoding sequence. A "transactivation domain" refers to a nucleic acid sequence which activates transcription, but typically fails to bind to DNA. A nucleic acid library consists of a clone bank of genomic or complementary DNA sequences which are "protein encoding sequences". The term refers to cDNA operably associated with the transactivation domain of a protein as exemplified in the EXAMPLES.
The cells also contain a nucleic acid reporter construct comprising a response element for the DNA binding domain operably associated with a reporter gene. As used herein, the term "reporter" refers to a gene encoding a trait or a phenotype which permits the monitoring and selection of, or the screening for, a cell containing the marker, and thus a NIMA interacting protein. The nucleic acid reporter is a selectable marker such as a color indicator (e.g., lacZ). Other suitable reporter genes will be known to those of skill in the art.
The protein-protein interaction system exemplified in the EXAMPLES herein preferably utilize the GAL4 protein to provide the DNA binding and transactivation domains (Fields and Song, Nature, 340:245. 1989). Other nucleic acid constructs comprising the coding sequences of proteins that have a DNA binding and transactivation domain will be known to those of skill in the art and can be utilized in the method of the invention for providing the hybrid constructs.
The method of the invention relies on interaction between the library encoded protein and the NIMA protein, or functional fragment thereof, in order to activate transcription of the reporter gene. Once the library encoded protein and the NIMA protein associate, they bring the DNA binding domain and the transactivation domain in close proximity, resulting in transcriptional activity. Thus, the method provides a means for identifying a protein that interacts or associates with NIMA protein kinase.
While the method of the invention as described above is preferred, it is understood that the transformed cell may contain the reverse constructs, e.g., a nucleic acid construct comprising a transactivation domain operably associated with the coding sequence of NIMA, or fragments thereof, and a nucleic acid library, wherein each member of said library comprises a DNA binding domain operably associated with a protein encoding sequence. Also included in the present invention are NIMA interacting proteins identified by the above method.
The invention also provides a method for controlling the growth of a cell comprising contracting the cell with a composition which modulates Pinl activity. The term "modulate" envisions the suppression of expression of PIN1 or suppression of Pinl activity, when it is over-expressed, or augmentation of PIN 1 798
-20- expression or Pinl activity when it is under-expressed. Growth of a cell is "controlled", for example, by inhibiting mitosis promoting function of NIMA or inducing entry of cells into mitosis. An inhibitor of Pinl activity or protein level, for example, would result in arrest in mitosis. Therefore, Pinl activity or protein level can be decreased, leading the cell to mitotic arrest, or alternatively, Pinl activity or protein level can be increased, arresting the cells in G2.
Where controlling the growth of a cell is associated with the expression of PINX polynucleotide, nucleic acid sequences that interfere with PINX expression at the translational level can be used. This approach utilizes, for example, antisense nucleic acid, ribozymes, or triplex agents to block transcription or translation of a specific PIN mRNA, either by masking that mRNA with an antisense nucleic acid or triplex agent, or by cleaving it with a ribozyme.
Antisense nucleic acids are DNA or RNA molecules that are complemen¬ tary to at least a portion of a specific mRNA molecule (Weintraub, Scientific American, 262:40, 1990). In the cell, the antisense nucleic acids hybridize to the corresponding mRNA, forming a double-stranded molecule. The antisense nucleic acids primarily work via RNaseH-mediated degradation of the target mRNA. Antisense may also interfere with the translation of the mRNA, since the cell will not translate a mRNA that is double-stranded. Antisense ohgomers of about 15 nucleotides are preferred, since they are easily synthesized and are less likely to cause problems than larger molecules when introduced into the target PIN- producing cell. The use of antisense methods is well known in the art (Marcus- Sakura, Anal. iochem., 172:289, 1988).
Use of an oligonucleotide to stall transcription is known as the triplex strategy since the oligomer winds around double-helical DNA, forming a three- strand helix. Therefore, these triplex compounds can be designed to recognize a unique site on a chosen gene (Maher, et al, Antisense Res. and Dev., U3):227. 1991; Helene, C, Anticancer Drug Design, 6_(6}:569, 1991).
Ribozymes are RNA molecules possessing the ability to specifically cleave other single-stranded RNA in a manner analogous to DNA restriction -21- endonucleases. Through the modification of nucleotide sequences which encode these RNAs, it is possible to engineer molecules that recognize specific nucleotide sequences in an RNA molecule and cleave it (Cech, J.Amer.Med. Assn., 260:3030. 1988). A major advantage of this approach is that, because they are sequence- specific, only mRNAs with particular sequences are inactivated.
Other Pin inhibitors include PPlase inhibitors. For example, immunosup¬ pressive drugs such as cyclosporin A and FK506, are PPlase inhibitors useful in the method of the invention for inhibiting some Pin proteins. The Pinl or other Pin protein of the invention is useful for screening for other inhibitors as well. For example, a suspected PPlase inhibitor is incubated with Pin 1 protein under suitable conditions to allow Pinl enzymatic activity to be expressed and measured, and the level of activity in the presence and absence of the inhibitor is assayed to determine the effect on Pinl activity.
Pinl activity may also be increased in the presence of an activator. PINX expression may be increased in the presence of an enhancer for example.
Stimulation of Pinl activity or overexpression of PINX arrests the cells in G2 and inhibits the mitosis promoting function of NIMA. The -acting elements which control genes are called promoters, enhancers or silencers. Promoters are positioned next to the start site of transcription and function in an orientation- dependent manner, while enhancer and silencer elements, which modulate the activity of promoters, are flexible with respect to their orientation and distance to the start site of transcription. One of skill in the art will know methods for stimulating expression of PINX by including an enhancer, for example, to stimulate PINX expression. The effect of an enhancer or other regulatory element can be determined by standard methods in the art (e.g., Northern blot analysis, nuclear runoff assay).
Pinl activity can be affected by an activator. An "activator" as used herein includes a protein or a small molecule, such as an organic compound, for example, which increases Pinl activity or protein level, such that a cell arrests in G2. An activator can be identified by incubating Pinl and a putative activator under -21- conditions that allow interaction between the components, and measuring of the effect of the activator on Pinl . For example, one could determine whether cells were arrested in G2 (e.g. , increased Pinl activity), or whether cells continued to cycle through G2 to M (e.g. , by FACS analysis or labeled nuclei analysis). The Pin 1 protein of the invention is useful in a screening method to identify compounds or compositions which affect the activity of the protein or expression of the gene. Thus, in another embodiment, the invention provides a method for identifying a composition which affects Pinl comprising incubating the compo¬ nents, which include the composition to be tested (e.g., a drug or a protein) and Pinl , under conditions sufficient to allow the components to interact, then subse¬ quently measuring the effect the composition has on Pinl activity or expression. The observed effect on Pinl may be either inhibitory or stimulatory. For example, the entry of cells into mitosis or G2 arrest can be determined by nuclear content (e.g. , immunofluorescence or FACS analysis based on DNA content) or other methods known to those of skill in the art.
The increase or decrease of PINX transcription/translation can be measured by adding a radioactive compound to the mixture of components, such as 32P-ATP (for nuclei) or 3H-Uridine, or 35S-Met, and observing radioacti e incoφoration into PINX transcripts or protein, respectively. Alternatively, other labels may be used to determine the effect of a composition on PINX transcription/translation. For example, a radioisotope, a fluorescent compound, a bioluminescent compound, a chemiluminescent compound, a metal chelator or an enzyme could be used. Those of ordinary skill in the art will know of other suitable labels or will be able to ascertain such, using routine experimentation. Analysis of the effect of a compound on PINX is performed by standard methods in the art, such as Northern blot analysis (to measure gene expression) or SDS-PAGE (to measure protein product), for example. Further, Pinl biological activity can also be determined, for example, by incoφoration label into nuclei.
The method of the invention described above also includes identifying a protein that associates with Pinl protein. The method comprises incubating the protein with Pinl protein, or with a recombinant cell expressing Pinl , under conditions sufficient to allow the components to interact and determining the effect of the protein on Pinl activity or expression. Alternatively, one of skill in the art could use the two-hybrid system described above to identify a protein that interacts or associates with Pinl protein. Once identified, such Pin E/N-associated proteins may now be used as targets for drug development.
In another embodiment, the invention provides a method for treating a cell proliferative disorder. The method comprises administering a subject in need of such treatment, an amount of Pinl inhibitor effective to induce entry of cells into mitosis or apoptosis. The "amount of Pinl inhibitor effective to induce entry of cells into mitosis" means that the amount of polypeptide, peptide, polynucleotide, or monoclonal antibody for example, which when used, is of sufficient quantity to ameliorate the disorder. The term "cell-proliferative disorder" denotes malignant as well as non-malignant cell populations which moφhologically often appear to differ from the surrounding tissue both moφhologically and genotypically.
Malignant cells (i.e. cancer) develop as a result of a multistep process. The PIN1 polynucleotide that is an antisense molecule is useful in treating malignancies of the various organ systems. For example, the method may be useful in treating malignancies of the various organ systems, such as, for example, lung, breast, lymphoid, gastrointestinal, and genito-urinary tract as well as adenocarcinomas which include malignancies such as most colon cancers, renal-cell carcinoma, prostate cancer, leukemia, breast cancer, non-small cell carcinoma of the lung, cancer of the small intestine, and cancer of the esophagus. Essentially, any disorder which is etiologically linked to altered expression of PIN] could also be considered susceptible to treatment with a PIN] suppressing/inhibiting reagent.
The method is also useful in treating non-malignant or immunologically- related cell-proliferative diseases such as psoriasis, pemphigus vulgaris, Bechet's syndrome, acute respiratory distress syndrome (ARDS), ischemic heart disease, post-dialysis syndrome, rheumatoid arthritis, acquired immune deficiency syndrome, vasculitis, lipid histiocytosis, septic shock and inflammation in general. Essentially, any disorder which is etiologically linked to PIN] would also be considered susceptible to treatment.
For puφoses of the invention, an antibody or nucleic acid probe specific for Pin l may be used to detect Pinl polypeptide (using antibody) or polynucleotide
5 (using nucleic acid probe) in biological fluids or tissues. The invention provides a method for detecting a cell proliferative disorder which comprises contacting an anti-Pin 1 antibody or nucleic acid probe with a cell suspected of having a Pinl associated disorder and detecting binding to the antibody or nucleic acid probe. The antibody reactive with Pinl or the nucleic acid probe is preferably labeled with l o a compound which allows detection of binding to Pin 1. Any specimen containing a detectable amount of antigen or polynucleotide can be used. The level of Pinl in the suspect cell can be compared with the level in a normal cell to determine whether the subject has a Pin 1 -associated cell proliferative disorder. Preferably the subject is human.
15 When the cell component is nucleic acid, it may be necessary to amplify the nucleic acid prior to binding with an PIN] specific probe. Preferably, polymerase chain reaction (PCR) is used, however, other nucleic acid amplification procedures such as ligase chain reaction (LCR), ligated activated transcription (LAT) and nucleic acid sequence-based amplification (NASBA) may be used.
20 The antibodies of the invention can be used in any subject in which it is desirable to administer in vitro or in vivo immunodiagnosis or immunotherapy. The antibodies of the invention are suited for use, for example, in immunoassays in which they can be utilized in liquid phase or bound to a solid phase carrier. In addition, the antibodies in these immunoassays can be detectably labeled in various
25 ways. Examples of types of immunoassays which can utilize antibodies of the invention are competitive and non-competitive immunoassays in either a direct or indirect format. Examples of such immunoassays are the radioimmunoassay (RLA) and the sandwich (immunometric) assay. Detection of the antigens using the antibodies of the invention can be done utilizing immunoassays which are run in
30 either the forward, reverse, or simultaneous modes, including immunohistochemi- -25- cal assays on physiological samples. Those of skill in the art will know, or can readily discern, other immunoassay formats without undue experimentation.
Various viral vectors which can be utilized for gene therapy as taught herein include adenovirus, heφes virus, vaccinia, or, preferably, an RNA virus such
5 as a retro virus. Preferably, the retroviral vector is a derivative of a murine or avian retrovirus. Examples of retroviral vectors in which a single foreign gene can be inserted include, but are not limited to: Moloney murine leukemia virus (- MoMuLV), Harvey murine sarcoma virus (HaMuSV), murine mammary tumor virus (MuMTV), and Rous Sarcoma Virus (RSV). Preferably, when the subject
10 is a human, a vector such as the gibbon ape leukemia virus (GaLV) is utilized. A number of additional retroviral vectors can incoφorate multiple genes. Other viral vectors include DNA vectors such as adenovirus and adeno-associated virus (AAV). All of these vectors can transfer or incoφorate a gene for a selectable marker so that transduced cells can be identified and generated. By inserting a
15 PIN] sequence of interest into the viral vector, along with another gene which encodes the ligand for a receptor on a specific target cell, for example, the vector is now target specific. Retroviral vectors can be made target specific by attaching, for example, a sugar, a glycolipid, or a protein. Preferred targeting is accomplished by using an antibody to target the retroviral vector. Those of skill in the art will
20 know of, or can readily ascertain without undue experimentation, specific polynu¬ cleotide sequences which can be inserted into the retroviral genome or attached to a viral envelope to allow target specific delivery of the retroviral vector containing the PIN] antisense polynucleotide. In addition, the viral vector may contain a regulatory element such as a tetracycline responsive promoter (inducible or
25 repressible) operably linked to a polynucleotide sequence).
Since recombinant retroviruses are defective, they require assistance in order to produce infectious vector particles. This assistance can be provided, for example, by using helper cell lines that contain plasmids encoding all of the structural genes of the retrovirus under the control of regulatory sequences within
"*0 the LTR. These plasmids are missing a nucleotide sequence which enables the packaging mechanism to recognize an RNA transcript for encapsidation. Helper cell lines which have deletions of the packaging signal include, but are not limited to Ψ2, PA317 and PA 12, for example. These cell lines produce empty virions, since no genome is packaged. If a retroviral vector is introduced into such cells in which the packaging signal is intact, but the structural genes are replaced by other genes of interest, the vector can be packaged and vector virion produced.
Another targeted delivery system for PIN I polynucleotides (e.g. , antisense) is a colloidal dispersion system. Colloidal dispersion systems include macromole- cule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes. The preferred colloidal system of this invention is a liposome. Liposomes are artificial membrane vesicles which are useful as delivery vehicles in vitro and in vivo. It has been shown that large unilamellar vesicles (LUV), which range in size from 0.2-4.0 μm can encapsulate a substantial percentage of an aqueous buffer containing large macromolecules. RNA, DNA and intact virions can be encapsulated within the aqueous interior and be delivered to cells in a biologically active form (Fraley, et al, Trends Biochem. Sci., 6:11, 1981). In addition to mammalian cells, liposomes have been used for delivery of polynucleotides in plant, yeast and bacterial cells. In order for a liposome to be an efficient gene transfer vehicle, the following characteristics should be present: (1 ) encapsulation of the genes of interest at high efficiency while not compromising their biological activity; (2) preferential and substantial binding to a target cell in comparison to non-target cells; (3) delivery of the aqueous contents of the vesicle to the target cell cytoplasm at high efficiency; and (4) accurate and effective expression of genetic information (Mannino, et al, Biotechniques, 6:682, 1988).
The targeting of liposomes can be classified based on anatomical and mechanistic factors. Anatomical classification is based on the level of selectivity, for example, organ-specific, cell-specific, and organelle-specific. Mechanistic targeting can be distinguished based upon whether it is passive or active. Passive targeting utilizes the natural tendency of liposomes to distribute to cells of the -27- reticulo-endothelial system (RES) in organs which contain sinusoidal capillaries. Active targeting, on the other hand, involves alteration of the liposome by coupling the liposome to a specific ligand such as a monoclonal antibody, sugar, glycolipid, or protein, or by changing the composition or size of the liposome in order to achieve targeting to organs and cell types other than the naturally occurring sites of localization.
The following examples are intended to illustrate but not limit the invention. While they are typical of those that might be used, other procedures known to those skilled in the art may alternatively be used.
-28-
EXAMPLE 1 MATERIALS AND METHODS
1. Yeast Two-Hybrid Screen and cDNA Isolation
For the yeast two-hybrid screen, the Aspergillus nidulans nimA cDNA was inserted into the pAS2 vector (from S. Elledge; HHMI, Baylor University)(Durfee et al., 1993; Haφer, et al., 1993) as fusion to GAL4 DNA-binding domain, resulting in NIMA/PAS2. A HeLa cell two-hybrid cDNA library that contained inserts fused to the transactivation domain of GAL4 (residue 768-881) (obtained from D. Beach; HHMI, Cold Spring Harbor Laboratory, New YorkNovember 12, 1995) (Harmon et al., 1993) was contransformed with NIMA/PAS2 into the yeast reporter strain Y190, which was then plated on yeast drop-out media lacking Leu, Tφ and His containing 35 mM 3-amino-l ,2,4 triazole and about 106 transformants were analyzed as described previously (Haφer et al., 1993). To determine the interaction properties of isolated clones, K40M NIMA, NIMA280-699, NLK1 and a frame-shift mutant NIMA280-699FS (a base pair deletion at the litigation point) were also subcloned into the pAS2 vector, followed by cotransformation into Y190 ceils with the isolated cDNA clones. To obtain additional PINX 5' untranslated sequence, a HeLa cell cDNA library constructed by R. Fukunaga; Salk Institute, La Jolla, CA) was screened with the H20 cDNA fragment. DNA sequence was determined by the dideoxynucleotide chain termination method.
2. Isolation of Human NIMA-Like Kinase 1 fNLKl
For polymerase chain reaction (PCR), three degenerate oligonucleotides GCGCCTGCAGTATCTATAC/TATGGAATAT/CTGT/(SEQ ID NO: 3) GCGCGGATCCG/AGGTTTCAGAGGT/GTC/TG/AAAG/CAG(SEQID NO: 4) and
GCGCGTACCAAGT/ACCACT/CGTAC/TATTATTCC (SEQ ID NO: 5) were designed specifically to the catalytic domain V, VII and VIII, respectively, which are conserved among NIMA (Osmani et al., Cell, 51:237, 1988), NPK, a budding yeast NIMA-related kinase (Schweitzer and Philippsen, Mol. Gen. Genet. , 214: 164, 1992) and human HsPK21 (Schultz and Nigg, Cell. Growth Differ ,
4:821 , 1993). Human placenta cDNA library (from R. Evans, Salk Institute, La Jolla, CA) was used as a template. The PCR cycle was 1 min at 95°C, 2 min at 42°C and 3 min at 63°C. PCR products with the expected size were subcloned and sequenced. Three potential clones (Ping-1, -44 and -77) were consistently obtained and showed a high sequence identity to NIMA at the deduced amino acids. To obtain the full length cDNA sequence, the Ping-1 PCR product was used as a probe to screen HeLa cell cDNA library filters provided by X.D. Fu (Gui et al., Nature, 36.9:678, 1994); over thirty positive clones were obtained and encode the same protein, referred to as NLK1 (NIMA-like kinase 1). NLK1 cDNA is 2152 bp and encodes 445 amino acids with an apparent molecular weight of 45 kDa on a
SDS-polyacrylamide gel. NLK1 was found to be same as NEK2 reported later by Schultz et al. (Cell. Growth Differ., 5:625, 1994)
3. DNA Transfection and Indirect Immunofluorescence Microscopy NIMA and its derivative mutant expression constructs were the same as described previously (Lu and Hunter, Cell, 81:413, 1995a). For expression of Pinl, an HA tag (MYDVPDYASRPQN) (SEQ ID NO:6) was added to the N-terminus of PIN] clones H20 and H6, followed by insertion into pUHD 10-3 vector as described previously (Lu and Means, EMBO J., 11:2103, 1994). Transfection of the tTA-1 cell line (Gossen and Bujard, Proc. Natl. Acad. Sci. USA, 89:5547, 1992) was described previously (Elredge et al., Meth. Enzymoi , 254:481. 1995) with the exception that cells were plated at lower density (30%), which seemed to increase the transfection efficiency and the percentage of cycling cells. Indirect immunofluorescence microscopy was performed as described (Lu and Hunter, Cell, 81:413, 1995a). The dilution of primary antibodies was: mouse M2 mAb
(Kodak/IBI, IgGl isotype), 1 :600;12CA5 mAb (IgG2b isotype, 1 : 1500; SC35 mAb (from X. Fu, IgGl isotype) (Fu and Maniatis, Nature, 141:437, 1990); straight supernatant; and rabbit anti-PML antibodies (from R. Evans) (Dyck et al.. Cell, 76:333, 1994), 1 :100. The FITC-conjugated goat anti-mouse IgGl and Texas Red-conjugated goat anti-mouse IgG2b secondary antibodies (Southern
Biotechnology) were used at 1 :50, with no significant cross-reactivity. Cells were observed and photographed with a Zeiss microscope, or an MRC-1000 laser- scanning confocal assembly (BioRad).
4. Flow Cvtometry Analysis Cells were harvested 48 hr after transfection and stained with the 12CA5 MAb, followed by flow cytometry analysis on a Becton-Dickinson FACScan machine (Lu and Hunter, Cell, &1: 13, 1995a). 12CA5-negative or -positive cells (104) were collected to determine the DNA content, and cell cycle profiles were determined using the M Cycle analysis software (Phoenix Flow Systems, San Diego, CA).
5. Metabolic Labeling. Immunoprecipitation and Immunoblot Analysis
Metabolic labeling of tTA-1 cells was as described previously (Lu and Hunter, Cell, 81:413, 1995a). Cells were lysed in a NP40 lysis buffer (50 mM Tris-HCl, pH 8.0, 0.1 % NP40, 200 mM NaCl, 20mM β-glycerophosphate, 20 mM NaF, 0.1 mM sodium orthovanadate, 50 μg/ml phenylmethylsufonyl fluoride, 10 μg/ml leupeptin, 10 μg/ml aprotinin, and 1 mM DTT or 10 mM β-mercaptoethanol (in the case of
Ni2+-NTA agarose precipitation) and preclarified with boiled S.aureis or Ni2+-NTA agarose (Qiagen). The lysates were then incubated with M2 or 12CA5 or His-Pinl- Ni2)*-NTA agarose. After washing 6 times with lysis buffer, the precipitates were subjected to SDS-polyacrylamide gel electrophoresis and immunoblotting.
6. Expression and Purification of Pinl and Kinase Assays
To express the (His)6 tag-containing Pin 1 protein, the cDNA insert from H20 clone was subcloned into Spel/Hindlll sites of pProEx l and the recombinant protein was expressed in and purified from BL21 bacterial strain using Ni:+-NTA agarose column (Qiagen) as described by the manufacturer. To examine whether
Pinl could be phosphoryiated, recombinant Pinl protein was added to NIMA kinase reaction mixtures at concentrations up to 0.5mg/ml, with β-casein and PL1 peptide as positive controls, as described previously (Lu et al., J. Biol. Chem. ,
269:6603, 1994). The effect of Pinl on NIMA was assayed using PL1 as a substrate with increasing concentrations of recombinant Pinl (Lu et al., J. Biol.
Chem. , 269:6603, 1994). Cyclin B/CDC2 (obtained from N. Watanabe; Salk
Institute) ERK1 MAPK (obtained from R. Fukunaga) and PKA (Promega) were assayed using histone HI, myelin basic protein and HI as substrates respectively.
7. PPlase Assay
PPlase activity of the purified recombinant proteins was assayed using the procedure of Heitman et al. (Methods, 5: 176, 1993) with the following exceptions. The reaction was carried out 5°C and chymotrypsin was added just before the peptide substrate (N-Succ-Ala-Pro-Phe-p-nitroanilide, Sigma). The cis to trans isomerization was monitored every 6 seconds by a change in the absorbance at 395 nm using DMS 2000 UV and Visible Spectrophotometer (Varian). FKBP, cyclosporin A and FK520, a FK506 derivative, were gifts of D. Schultz and C. Zuker; University of California, San Diego.
8. Yeast Complementation Assays
PIN] expression was driven from the strong constitutive yeast triose phosphate isomerase promoter (TPI) (Smith et al., 1985). Plasmid pTPI-PINl was made by insertion of an -850 bp BamHI-XhoI fragment of PIN] cDNA (from H20/GADGH) into pJK305-TPI (J. Kamens, Ph.D. Thesis, Harvard University, 1991). PTPI-PIN1 directs the synthesis of the native full length Pinl protein and carries the yeast 2μ replicator and LEU2 selectable marker. Plasmid YepHESS carries the yeast ESS] gene, a 2μ replicator and HIS 3 selectable marker (Hanes et al., Yeast, _{ SS, 1989).
For tetrad analysis, a heterozygous ESS I disruption strain MGG3/pSH-U (MATa/MATa ura3/ura3 leu2/ leu2 his 3/ his 3 essI::URA3/ESSI) (Hanes et al., Yeast, 5:55, 1989), was transformed with control vector pJK305-TPI, YepHESS, or pTPI-PINl . Cells were induced to undergo sporulation on 1% potassium acetate plates, tetrads were dissected and haploid segregants grown for 3-4 days at 30 °C on rich medium (YEPD) plates. Only tetrads that showed proper segregation of the URA3, and MATa and MAT alleles were included in the viable spore counts. For the curing experiment, a homozygous disruption derivative of MGG3/pSH-U (relevant genotype essl::URA3/ess]::URA3) carrying ESS] on episomai plasmid (YepHESS) was transformed with either pJK305-TPI or pTPI- PIN 1. Cells were serially passaged once per day (1/50 Dilutions) for 6 days in liquid completer synthetic medium lacking leucine; thus maintaining selection for the PIN] -expressing plasmid or control vector (2μ, LEU2), but not for the ESS1- bearing plasmid (2μM, HIS3). Cells were plated and phenotypes of individual colonies scored by replica plating to appropriate selective media.
9. PIN I -Dependent Yeast and Determination of Terminal Phenotype For Pinl depletions experiments in yeast, we used the GAL] promoter (Yocum et al., Mol. Cell. Bio., 4: 1985, 1984). Plasmid pGAL-PINl, was made by insertion of a Spel-Xohl fragment of the PIN] cDNA from pN2P 1 /GADGH into Avrll and
Xhol cut pBC103. PGAL-PINl directs the synthesis of an HA1 hemagglutinin epitope-tagged version of Pinl that contains a 31 amino-terminal extension (M ASYPYDVPDYASPEF VDPPGSKNSIARGKM) (SEQ ID NO:7) where underlined residues are the HA1 epitope and the normal PIN1 initiator, respec- tively. Other residues derive from polylinker sequences. An integrating version, I- GAL-PIN 1 , was made by removal of the GAL-PIN 1 cassette from pGAL-PINl with Xbal and Sad and reinsertion into the same sites in pRS305 (Sikorski and Hieter, Genetics, 122: 19, 1989). I-GAL-PIN1 carries a LEU2 selectable marker. The original essl~ knockout strain (MGG3/pSH-U) does not grow in galactose, probably due to a gal2 mutation. Therefore, to make a strain (YSH12) that lacks ESSI function and whose growth depends on a galactose-inducible version of PIN], the following scheme was used. A gal strain, FY86 (MATa his3Δ200 ura3-52 ICU2ΔI) was transformed with integrating vector GAL-PIN 1 (LEU2). Leu+ transformants were mated with a haploid MGG3/pSH-U segregant
(MATa ura3 his3 leu2 ess]::URA3) carrying YEpHESS. Diploids were sporulated and tetrads dissected on rich medium containing 2% galactose/ 1% raffinose. Haploid segregants that contained the ess]:: URA3 disruption (ura~) and GAL- PIN] (leu*), but lacked YEpHESS (his') were identified. Isolates that showed growth on galactose (YEPG) but not on glucose (YEPD) medium were further characterized. Cells that did not contain the ess] knockout (ura ) but did contain GAL-PIN 1 (leu*) were used as controls. Inducible expression of PIN I in several isolates of YSH12 was confirmed by immunoblot analysis.
To shut off expression of PIN 1 in ess]' cells, strain YSH12 was grown overnight in galactose or galactose/glucose-containing medium and inoculated
(~ 1/50 dilution) into glucose containing-medium. Cells were grown for approxi¬ mately 12 hours and then reinoculated into fresh glucose-containing medium for an additional 18 hours. Aliquots of cells were harvested by centrifugation, fixed by the addition of 70% ethanol and stored at 4°C. Bud size distribution was scored under DIC (Normarski) illumination after cells were resuspended in water and sonicated extensively to disperse clumped cells. For DAPI staining, the fixed cells were stained with 1 μg/ml DAPI and mounted in 75% glycerol and photographed under fluorescent illumination. FACS analysis of yeast cells was described previously (Sazer and Sherwood, J. Cell Sci., 509, 1990). EXAMPLE 2 IDENTIFICATION OF CLONES ENCODING PROTEINS INTERACT¬ ING WITH NIMA fPINS
To search for human cDNAs encoding proteins able to interact with NIMA, a yeast two-hybrid system that uses GAL4 recognition sites to regulate expression of both HIS3 and £αcZ (Durfee et al., Genes Dev., 2'555, 1993) was used. As bait, the full length coding sequence of the Aspergilius nidulans nimA was fused to the C-terminus of the GAL4 DNA-binding domain in pAS2 driven by a partially ADH promoter (Haφer, et al., Cell, 75:805, 1993). As prey, we used the GAL4 transactivation domain and a human HeLa cell cDNA fusion library driven by the highly active entire ADH promoter, resulting in high level expression (Hannon, et al., Genes Dev. , 2378, 1993). Initially, Y190 cells were transformed with the NIMA/pAS2 expression vector in an attempt to establish stable strains constitu- tively expressing NIMA, but the transformant colonies were very small and could not be propagated. The failure of the transformants to grow is probably due to the fact that NIMA induces mitotic arrest in budding yeast, as it does in the other different eukaryotic cells so far examined (Osami, et al., Cell, 11:237, 1988; Lu and Means, EMBOJ., 11:2103, 1994;O'Connell et al., EMBO J , 11:4926, 1994; Lu and Hunter, Cell, & 413, 1995a). To circumvent this problem, Y190 yeast cells were cotransformed with NIMA pAS2 and the two-hybrid screen cDNA library in the hope that expression of one or more of the cDNA library products might rescue the lethal phenotype of NIMA.
Out of 106 colonies screened, 13 were consistently positive upon repeat transformations. The specificity of the interaction was tested using full length NIMA, a C-terminal noncatalytic fragment of NIMA, NIMA280-699, and a human
NIMA-like kinase 1 (NLK1) (Figure 1A). Figure 1A shows a β-Galactosidase activity in the two-hybrid system. Yeast strain Y190 was cotransformed with vectors expressing the three different types of cDNA and different domains of NIMA or NLK1 as indicated, and grown in SC media lacking Tφ and Leu, followed by β-galactosidase activity filter assay as described previously (Durfee, et al. , Genes Dev. , 7:555, 1993). NLKl , which was isolated from a human placenta cDNA library and proved to be the same as Nek2 (Schultz et al., Cell Growth Differ., 5:625, 1994), is 47% identical to NIMA in its catalytic domain, but NIMA and NLK 1 has very little similarity in their C-terminal noncatalytic domains (Figure 1 B). Based on the sequences of their inserts and the specificities of their interaction with NIMA and NLKl , these clones fell into three different gene classes, referred to as PIN], PIN2 and PIN3 ( /N=protein interacting with ΝIMA). There were six PIN] , three PIN2 and four PIN3 clones. The analysis of the two-hybrid interactions indicate Pin3 interacts with the catalytic domains in both ΝIMA and ΝLKls, whereas Pinl and Pin2 interact with the C-terminal domain of ΝIMA. The following examples focus on Pinl as an exemplary Pin protein.
EXAMPLE 3 ANALYSIS OF THE PIN1 CDNA
The DNA and deduced amino acid sequence of the protein encoded by the longest PIN1 cDNA (l.Okb) is shown in Figure 2A. It encodes a protein of 163 amino acids with the predicted molecular weight of 18,245. When expressed in HeLa cells, Pinl migrated with an apparent size of ~ 18 kDa in an SDS-polyacryl- amide gel. Pinl shows a high similarity to the budding yeast Essl protein (Hanes et al., Yeast, 1:55, 1989). Based on recent RNA primer-extension studies and reexamination of the nucleotide sequence of ESS] (Hani et al., FEBS Lett.,
365:198. 1995), it is likely that the second ATG is used as the initiation codon, rather than the first ATG as originally proposed (Hanes et al., Yeast, 1:55, 1989); in addition, a G should be inserted at position 919, which results in a C-terminal frame shift. The corrected ESS] sequence encodes a 169 amino acid protein with 45% identity to Pinl . Northern blot analysis of human cell line and human fetal liver RNAs showed that there is a single PIN] mRNA of - 1.Okb present in all cell lines and tissue tested (Figure 3). Fifteen μg of the indicated total RNAs were run on each lane. HeLa, epitheloid carcinoma cells; HFF, human foreskin fibroblasts; Ramos, Burkitt lymphoma cells; A431 , epitheloid carcinoma cells; 293 ; adenovirus El A transformed human embryonic kidney cells; U937, histiocytic lymphoma cells; Saos-2, osteosarcoma cells; U87-MG, glioblastoma cells; Jurkat, T leukemia cell line and HFL, human fetal liver. Position of the RNA molecular weight standards (Promega) are on the left. These results, together with the sequence comparison between Pinl and
Ess 1, confirm the authenticity of the PIN] open reading frame, although there is no inframe termination condon preceding the putative initiation site. Sequence analysis of six different PIN] cDNA clones identified the points of fusion with the GAL4 activation domain at Arg-(-3), Glu+(+5) and Lys-(+6), indicating that the N-terminal 5 amino acids of Pinl are not necessary for the interaction with NIMA.
Immunoblot analysis using anti-Pinl antibodies confirmed that Pinl is an 18 kD protein in the cell.
The deduced Pinl sequence contains two identifiable domains, an N- terminal WW domain and C-terminal putative PPlase domain. The WW domain contains two invariant Tφ residues and other residues highly conserved in the WW domains of other proteins including mammalian dystrophins and Yap, and the yeast Rspl and Essl (Sudol et al., 1995). However, the Cys or His-rich domain flanking the WW domain found in WW domain-containing proteins including Essl (Sudol et al., 1995) is not conserved in the human Pin 1. The C-termin l two-thirds of Pin 1 contains motifs that are characteristic of the recently identified third family of
PPIases (Rudd et al., TIBS, 2Q:12, 1995). The PPlase domain of Pinl contains three highly observed subdomains, with 45% identity to parvulin and 62% identity to a partial PPlase from A. thaliana. A putative nuclear localization signal is located in the beginning of the PPlase domain, which is also conserved in Essl . Figure 2 A shows the predicted Pinl amino acid sequence is indicated in one-letter code. The fusion points between GAL4 and Pinl in six different isolated clones were: clone H20 at C-9; clone H16, 24 and 38 at G+13; clones H6 and H36 at C+l 5. Underlined residues form a consensus bipartite nuclear localization signal. The N- and C-terminal boxes indicate the WW domain and PPlase domain. Nucleotide numbers are on the left and amino acid numbers on right. Figure 2B -37- and 2C show alignments of the WW domain (B) and PPlase Domain (C) in selected proteins. Identical residues are shown in the bottom row. Dashes indicate gaps introduced to make the alignment. Cbf2, cell binding factor 2; SC, S cerevtsiae; EC, E coli; BS, B subtihs; CJ, C jejum; AT, A thaliana.
EXAMPLE 4
Pinl HAS PPlase ACTIVITY
Because of the strong sequence similarity between Pinl and the third PPlase family, Pin was tested to determine if Pin could catalyze the cis/trans isomerization of peptidyl-prolyl peptide bonds in vitro, a characteristic of PPlase. Pin 1 was expressed in bacteria with an N-termmal (His)6 tag and was purified from bacteπa using a Ni2+-NTA agarose column. When purified recombinant Pinl was tested for PPlase activity using a standard chymotrypsin-coupled spectrophotomet- ric assay (Heitman et al., Methods, 1: 176, 1993), PPlase activity was readily detected which was highly concentration-dependent, with a specific activity similar to that of recombinant FKBP (Figure 4). The isomerase activity was measured as described in Example 1. The cis to trans isomerization was monitored every 6 seconds by a change in the absorbance at 395 nm, with the finally stable absorbance of each sample being set at 1.0. Different concentrations of Pinl were used as indicated, with the recombinant FK506-binding protein (FKBP) PPlase being used as a positive control. The control sample contained all the ingredients except the
PPlase. The insert shows PPlase activity during the first minute of the assay
(Acontrol;opinl, 71 ug/ml; a Pinl, 14 ug/ml;" Pinl, 7 ug/ml; • FKBP 70 ug/ml).
Like parvulin, the PPlase activity of Pinl was not inhibited by either cyclosporin A or FK520, a derivative of FK506, even at 25μM. These results confirm that Pinl is a member of the third family of PPlase. EXAMPLE 5 Pinl INTERACTS WITH THE C-TERMINAL DOMAIN OF NIMA
In the two-hybrid system, Pinl interacted with NIMA, kinase-negative K40M NIMA and C-terminal NIMA280-699, but not with NLKl (Table 1), indicating that NIMA interacts specifically with the C-terminal noncatalytic domain of NIMA in yeast. To determine if there was a stable interaction between Pinl and NIMA in HeLa cell extracts, recombinant Pinl was coupled to beads to determine whether NIMA could be recovered from lysates of cells transiently expressing different NIMA mutants. Since NIMA induces mitotic arrest (Lu and Hunter, Cell, 81:413, 1995a), it is difficult to express sufficient wild type NIMA to detect complex formation, but since Pinl interacted with the kinase-negative K40M NIMA as efficiently as wild type NIMA in the yeast two-hybrid system, we used K40M NIMA and truncated derivative mutant constructs were used.
Figs. 5A-E shows immunoprecipitations to show the interaction between Pinl and the C-terminal noncatalytic domain of NIMAs in HeLa cells. Figure 5A shows the expression and purification of His-Pinl . The PINX cDNA was subcloned into pProExl and the recombinant protein was purified from bacteria using Ni2+- NTA agarose column, followed by analyzing the protein on a 15% SDS-containing gel, and Coomassie staining. The positions of His-Pinl and standard size markers are indicated. Figure 5B shows the expression of NIMAs in HeLa cells. tTA- 1 cells were transfected with different FLAG-tagged NIMA constructs and labeled with 5S Express Label for 24 hr, followed by immunoprecipitation using the M2 mAb.The precipitated proteins were analyzed on a 10% SDS-containing gel followed by autoradiography. The positions of K40M, NIMA, K40M NIMA280, NIMA280-699 and standard size markers are indicated. Figure 5C shows a stable complex between recombinant Pinl and NIMAs. Cell lysates similar to those in panel B were incubated with His-Pinl -Ni2+-NTA bead, as indicated in panel A and washed, followed by electrophoresis on an SDS-containing gel and autoradiogra¬ phy. Figures 5D and 5E show coimmunoprecipitation of K40M NIMA with Pinl . tTA- 1 cells were cotransfected with FLAG-tagged NIMA and HA-PINX constructs -39- for 24 hr. Cell lysates were immunoprecipitated with the HA-specific 12CA5 mAb, followed by immunoblotting using the M2 mAB specific for the FLAG tag (D) and vice versa (E).
Out of several different fusion proteins tested, His-Pinl was found to be most stable and easiest to purify in large quantities when expressed in bacteria
(Figure 5A). When incubated with 35S-labeled extracts from cells transiently expressing the full length K40M NIMA, an N-terminal fragment K40M NIMA280, or a C-terminal fragment NIMA280-699 (Figure 5B), the recombinant His-Pinl beads specifically bound K40M NIMA and NIMA280-699, but not K40M NIMA280 (Figure 5C). Results also showed that purified GST-NIMA 280-699 directly interacts with purified Pinl in vitro. These results indicate that the recombinant Pinl interacts with C-terminal domain of NIMA in the HeLa cell extracts.
To examine whether a Pinl and NIMA complex is formed in the cell, an HA epitope tag was inserted at the N-terminus of Pinl and coexpressed it with
FLAG-tagged NIMA constructs in HeLa cells using a tetracycline-responsive expression system, as described previously (Lu and Hunter, Cell, 8J_:413, 1995a). When cell lysates were immunoprecipitated with the HA tag-specific mAb (12CA5), followed by immunoblot analysis using the M2mAb specific for the FLAG tag and vice versa, K40M NIMA but not K40M NIMA280 was detected in Pinl immunoprecipitates (Figure 5D). Conversely, Pinl was detected only in K40MNIMA immunoprecipitates (Figure 5E). When the isolated catalytic domain of NIMA was expressed in HeLa cells, it was located in the cytosol (Lu and Hunter, Cell, 31:413, 1995a), whereas Pinl is a nuclear protein (see below); therefore it is unlikely that Pinl interacts with the NIMA catalytic domain. These results, together with the in vitro binding assay and the two-hybrid analysis, suggest that the C-terminal noncatalytic domain of NIMA is involved in the interaction with Pinl .
To examine whether Pinl affected NIMA kinase activity, purified recombinant Pinl protein was added to a NIMA kinase reaction mixture in the -40- presence of absence of the PL1 peptide substrate. Under these conditions tested, Pin l was neither phosphorylated by nor inhibited NIMA kinase. Similar results were obtained with several other protein kinases including cyclin B/CDC2, ERK1 and PKA. These results indicated that Pinl is unlikely to act as a substrate or an inhibitor of the NIMA, CDC2, ERK1 and PKA protein kinases.
EXAMPLE 6
Pinl COLOCALIZES WITH NIMA IN A DEFINED
NUCLEAR SUBSTRUCTURE
To examine whether Pinl is colocalized with Nima in human cells, the subcellular localization of Pin 1 was determined using indirect immunofluorescence staining as described previously (Lu and Hunter, Cell, & 413, 1995a). When Ha- tagged Pinl was expressed in HeLa cells and stained with the 12A5 mAB, Pinl was observed almost exclusively in the nucleus. This result indicates that Pinl is a nuclear protein, consistent with the presence of a nuclear localization signal in Pinl (Figure 2 A). Confocal microscopy further revealed that although Pinl was distributed throughout the nucleus, it was highly concentrated in certain areas with a speckled appearance (Figure 6, right panel).
Figure 6 shows the colocahzation of Pinl and NIMA and its kinase- negative mutant in HeLa Cells. Twenty four hr after transfection with the vectors expressing HA-tagged Pinl only (right panels) or HA-tagged Pinl and FLAG- tagged NIMA (left panels) or its kinase-negative mutant (K40M NIMA, middle panels), the transfected tTA-1 cells were processed for indirect immunofluores¬ cence staining and examined by confocal microscopy. Top panels: staining pattern for NIMA or K40M NIMA obtained with FLAG tag-specific M2 mAb or SC-35 obtaining with anti-SC-35 mAb, and then FITC-conjugated IgG 1 -specific secondary antibodies; middle panels: staining pattern for Pinl obtaining with HA tag-specific 12CA5 mAb and Texas-Red-conjugated IgG2b-specific secondary antibodies; bottom panels: double-staining for Pinl and NIMA, Pinl and K40M -41 -
NIMA, or Pinl and SC-35 displayed by superimposing the respective top and middle images, with yellow color indicating colocahzation. Arrows point to an untransfected cell, indicating very little cross reactivity among the antibodies. Bar, l μm. To examine whether Pinl and NIMA were colocalized in HeLa cells, HA- tagged PIN] and Flag-tagged nimA or its kinase-negative mutant (K40M nimA) cotransfected into tTA-1 cells and then examined the localization of expressed proteins using tag-specific 12CA5 and M2 mAbs and isotope-specific secondary antibodies. In cells with the NIMA-induced mitotic phenotype (Figure 6, left panels), Pinl was associated with the condensed chromatin, but was also distributed throughout the cell, as was NIMA (Lu and Hunter, 1995a). In K40M NIMA-expressing cells, Pinl was concentrated in nuclear substructures, where the mutant NIMA was also concentrated (Figure 6, middle panels). The colocahzation was extensive, but not complete (Figure 6, middle lower panel). Pinl was also colocalized with the C-terminal NIMA280-699 in the nuclear substructures. Since similar nuclear substructures have been observed by immunostaining with anti- SC35 splicing factor (Fu and Maniatis, Nature, 241:437, 1990) and anti-PML antibodies (Dyck et al., Cell, 76:333, 1994), Pinl nuclear substructures were examined to determine whether they were the same as those localized by SC35 or PML. When cells expressing Pinl were doubly stained with 12CA5 and anti- splicing factor SC35 or anti-PML antibodies, the speckles displayed by Pinl were found to be the same as those detected using anti-SC35 (Figure 6, right panels), but not anti-PML antibodies. These results indicate that Pinl and K40M NIMA are colocalized in the spliceosome nuclear substructure. Since using identical protocols, completely different localization patterns were observed for several other proteins, including the human NLKl which was colocalized with Pin3 in the nucleolus, the localization pattern of Pinl and NIMA is unlikely to be due to a nonspecific accumulation in certain areas of the nuclease resulting from over expression. Therefore, these results demonstrate that Pinl is colocalized with NIMA in the nucleus in a defined nuclear substructure. EXAMPLE 7 Pinl OVEREXPRESSION INHIBITS NIMA-INDUCED MITOTIC
ARREST AND INDUCES G2 ARREST IN HELA CELLS
In the yeast two-hybrid system, expression of Pinl rescued the lethal phenotype of NIMA, indicating that Pinl might inhibit the mitotic function of NIMA. To examine whether over expression of Pinl in HeLa cells blocked NIMA- induced mitotic arrest, cells were contransfected with expression vectors for nimA and either PIN] or an expression vector control and their phenotypes examined, as described previously (Lu and Hunter, Cell, £1:413, 1995a).
Figures 7A-7D shows overexpression of PINX delays NIMA-induced mitotic arrest and induces a specific G2 arrest in HeLa cells. Figure 7A shows the results of tTA- 1 cells cotransfected with nimA and PINX or control vector. At the times indicated, cells were fixed and doubly labeled with M2 mAb and Hoechst dye, followed by scoring for the percentage of cells with rounding and chromatin condensation in a sample of at least 250 NIMA-expressing cells. The data are the average from two independent experiments.
Figures 7B, C, and D show after transfection with PINX expression vector or the control vector for 48 hr, tTA-1 cells were stained with the 12CA5 mAb and then with FITC-conjugated secondary antibodies and propidium iodide, followed by
FACS analysis. Based on the FITC intensity of the PINX -transfected cells, cells were divided into two populations with 12CA5 -negative (B) or -positive cells (C), and cell cycle profiles were determined to compare with those in total vector- transfected cells (A). At 24 hr after transfection, -80% of NIMA-expressing cells were rounded up and contained highly condensed chromatin (Figure 7A). When cells were contransfected with nimA and PIN], NIMA was expressed at much higher levels than in cells contransfected with nimA and control vector, as detected by immunofiuorescence microscopy. However, at 24 hr only -21% of NIMA- -43- expressing cells showed the complete mitotic phenotype. The other NIMA- expressing cells were not completely rounded up and their chromatin was not as condensed as those in cells expressing only NIMA (Figure 5 left panels and 7A), although by 36 hr all the NIMA-expressing cells displayed the mitotic phenotype
5 (Figure 7A). These results indicated that Pinl can partially inhibit the mitosis- promoting function of NIMA.
To examine whether overexpression of Pinl blocked the G2/M transition, HeLa cells were transfected with either PIN] or the control vector and cycle progression was examined by immunofluorescence microscopy and FACS analysis. l o Cells expressing Pin 1 contained a large inteφhase nucleus, and it was very difficult to find mitotic cells expressing Pinl, suggesting that Pinl might induce a G2 arrest. FACS analysis indicated that the percentage of cells with 4n DNA content was significantly increased in cells expressing Pinl (12CA5-positive) (Figure 7D). About 50% of cells were in G2, with commensurately fewer cells in Gl and S.
15 Similar results were also obtained with over expression of another independent
PIN] clone (H6) encoding a 5 amino acid N-terminal deletion mutant. In contrast, the vector-transfected cells showed very little 12CA5 staining and displayed a similar cell cycle profile to that of cells not expressing Pinl (12CA5-negative) in the Pinl -transfected cell population (Figure 7B and C). These results, together
20 with the delaying effect of Pinl on NIMA-induced mitotic arrest, suggest that Pinl can inhibit the NIMA pathway which is required for the G2/M transition.
EXAMPLE 8
PIN1 IS A FUNCTIONAL HOMOLOGUE OF THE
ESSENTIAL ESS1 GENE OF S. CEREVISIAE
25 Overexpression of Pinl inhibits the G2/M transition in HeLa cells as shown above. If this inhibitory effect is due to the fact that high levels of Pinl affect a cell cycle checkpoint control, depletion of Pinl should promote the G2/M transition, resulting in a mitotic arrest. To examine this possibility, budding yeast were used where endogenous protein expression can be readily manipulated. Since Pinl shows striking similarity to the yeast Essl , the human PIN I was tested to determine whether it might functionally substitute for the yeast gene. Since ESS I knockout mutations are lethal in yeast (Hanes et al., Yeast, 5:55, 1989), the ability of PIN 1 to restore viability to ess]- mutants was asigned. First, plasmids that express PIN] under the control of a constitutive yeast promoter were introduced into diploid cells in which one copy of ESS] is disrupted (essl::URA3/ESS]). Cells were induced to undergo sporulation, tetrads were dissected, and viability of the resulting haploid spores was scored (Table 1A). As expected, tetrads derived from cells transformed with vector alone showed a 2:2 segregation for spore viability (viable:inviable). In contrast, -25% of the tetrads from cells transformed with a
PIN] -expressing plasmid showed 4:0 segregation for spore viability, indicating that PIN] complements the essl mutant to allow spore outgrowth and haploid cell viability. PIN] did not complement essl mutant to allow spore outgrowth and haploid cell viability. PIN] did not complement essl' mutants quite as efficiently as ESS] itself, where more than half the tetrads from cells bearing an ESS1 plasmid showed a 4:0 segregation from spore viability. Growth curves revealed that PIN1- expressing ess]' cells had doubling times only slightly longer than control cells.
Thus, human PIN! can functionally substitute for ESS] in haploid yeast cells.
Second, the PIN] expression plasmid was introduced into diploid cells in which both chromosomal copies ofESSl are disrupted (essl : : URA3/essl: : URΛ3) but that remain viable by maintaining a plasmid-borne copy of ESS If PIN 1 functionally substitute for ESS], then it should be possible to cure cells of the ESS1 plasmid. Table IB shows that cells serially passaged in media selecting only for the PIN] plasmid (LEU2) lost the ESS1 plasmid (HIS3) about 12% of the time, whereas cells containing a control vector did not lose the ESS1 plasmid. Thus, human PIN] can functionally substitute for ESS1 in diploid yeast cells. -45-
Table 1 PINX Complements the ESS1 Knockout Mutation in Budding Yeast
A. Tetrad Analysis
Figure imgf000047_0001
B. Curing Experiment
Plasmid His7Leu+ Ura+ Loss of JYN1 plasmid (%)
Vector 216/216 216 0
PINX 380/431 431 12
A. PINX rescues essl' lethality in haploids. The heterozygous disruption strain MGGS3/pSH-U was transformed with an £551- or PINX -expression vector plasmid or control vector. Cells were induced to undergo sporulation and tetrads were dissected. The number of viable spores included only those that showed proper segregation of independent markers. As expected, almost all segregants that were ura* [i.e. carried the ess /::URA3 knockout allele) were also his* or leu* indicating the presence of the £55 or PIN1 containing plasmid, respectively.
B. PIN] permits loss of £55/-containing plasmids in a essl diploid knockout strain. A diploid disruption strain (&s.5l _:URA3/e_j_jl ::URA3) carrying the £551 -plasmid ( //53) was transformed with the vector alone (LEU2) or a PINX -expression vector plasmid (LEU2). Cells were serially passaged in leucine-deficient media that maintains selection for control or the PINX plasmid but not for the £551 plasmid. Cells were plated and phenotypes of individual colonies scored by repiica plating to appropriate selective media. Loss of the £551 plasmid is detected by loss of the his* phenotype, while the presence of PINX is detected by a leu* phenotype. A ura* phenotype confirms the presence of the knockout allele (essX ::URA3).
EXAMPLE 9 DEPLETION OF PIN1/ESS1 RESULTS IN MITOTIC ARREST AND NUCLEAR FRAGMENTATION IN S. CEREMSIAE
To examine the effect of depleting Pinl /Essl on the cell cycle, PIN1 driven by the regulated GAL promoter was introduced into an essl' yeast strain. As expected, Pinl -expressing strains grew normally in inducing media (galactose) or noninducing media (galactose/glucose, with a basal level of PIN 1 expression), but did not grow in repressing media (glucose).
Figure 8 shows depletion of Pinl /Essl results in mitotic arrest and nuclear fragmentation in yeast. A Pinl -dependent strain (YSH12.4) was shifted from inducing media to repressing media, harvested and fixed with 70% ethanol at the times indicated. The cells were stained with DAPI or propidium iodide, followed by videomicroscopy under Nomarksi (DIC) or fluorescent (DAPI) illumination, or
FACS analysis, respectively. The bar is 10 μm and the inserts show a higher magnification of a representative cell.
Cells depleted of Pinl following the shift to repressing media displayed a striking terminal phenotype indicating mitotic arrest (Table 2, Figure 8). Following about 6 hr of normal growth, cell division was inhibited and by 12 hr, cells began to accumulate in mitosis as judged by the increasing percentage of cells containing a large bud (dumbbell shape) and by FACS analysis (Table 2, Figure 8, middle panels). As revealed by DAPI staining, a high percentage of cells containing nuclear staining material in the neck between the mother and the bud, suggesting that mitotic chromosome segregation was slowed or inhibited. Control strains that carried GALr.PINl, but were wild-type for ESSl, showed normal distribution of cells throughout the cell cycle as judged microscopically and by FACS analysis. By 24 hr., cell division stopped and most cells were arrested in mitosis. Interestingly, -47- cells depleted of Pinl for extended periods of time (18-30 hr) showed multiple nuclear fragments, which appeared randomly distributed throughout the cell (Figure 8, right panels). FACS analysis revealed that cells with a 2n DNA content accumulated over time, with most cells containing 2n DNA content by 24 hr after the shift to repressing media (Figure 8, bottom panels). When cells were shifted from noninducing media tor expressing media, this phenotypes appeared earlier. Cells were arrested at mitosis by 12 hr with a mitotic chromatin located in the neck in about 40% of cells and nuclear fragmentation in most other cells, indicating the dependence of the phenotypes on the initial level of Pinl expression. These results show that depletion of Pinl results in mitotic arrest and eventually nuclear fragmentation; phenotypes similar to those observed in Aspergillus and HeLa cells that oveφroduce NIMA (Osmain et al., Cell, 11:237, 1988; O'Connell et al., EMBOJ., 11:4926, 1994; Lu and Hunter, Cell, 81, 413, 1995a).
Table 2 Depletion of Pinl /ESS 1 Results in Mitotic Arrest in Budding Yeast
Figure imgf000049_0001
A Pi l -dependent strain (YSH 12.4) was shifted from inducing media to repressing media, harvested and fixed with ethanol at the times indicated. Bud size distribution was scored under Nomarski illumination after cells were stained with DAPI and sonicated. About 500 cells were counted for each point. SUMMARY
Using the yeast two-hybrid system, the present invention identifies the first NIMA- interacting protein of human origin Pinl . Pinl contains an N-terminal WW domain and a C-terminal PPlase domain, and has PPlase activity in vitro. PIN1 function- ally rescues a knockout mutation of the ESS! gene, which is essential for the growth of budding yeast cells. These results indicate that Pinl is conserved from yeast to human; Pinl /Essl is the first PPlase known to be essential for life. Pinl interacts with the C-terminal noncatalytic domain of NIMA and colocalizes with NIMA in a defined nuclear substructure in HeLa cells. Whereas overexpression of Pin 1 induces a specific G2 arrest, and delays NIMA-induced mitosis in HeLa cells, depletion of Pinl/Essl triggers mitotic arrest and nuclear fragmentation in budding yeast. These results indicate that Pinl regulates entry into mitosis, probably via the NIMA mitotic pathway.
The primary structure of Pinl contains two identifiable domains. The C- terminal two-thirds of Pin 1 contains residues that are highly conserved in the newly delineated family of PPIases that includes parvulin, PrsA, SurA, NifM, PrtM, Cbf2 and Essl (Rudd, et al., TIBS, 20:12, 1995). PrsA, SurA, NifM, PrtM have been shown to be involved in maturation and/or transport of specific proteins or protein classes. Parvulin, originally identified during a chromatographic purification procedure, is the prototype of this family; it contains 96 amino acids and catalyzes the cis/trans isomerization of X-Pro peptide bonds, even in the presence of immunosuppressive drugs (Rahfeld et al., FEBS Lett.. 352:180. 1994a and FEBS Lett., 343:65. 1994b). The homology between Pinl and parvulin spans almost the entire parvulin molecule, strongly suggesting that Pinl is a PPlase. The in vitro PPlase assay shown in the above Examples confirms that Pinl can indeed catalyze the cis/trans isomerization of peptidyl-prolyl peptide bonds.
Although the invention has been described with reference to the presently preferred embodiments, it should be understood that various modifications can be made without deporting from the spirit of the invention. Accordingly, the invention is limited only by the following claims. SEQUENCE LISTING (1) GENERAL INFORMATION
(l) APPLICANT The Salk Institute for Biological Studies (11) TITLE OF INVENTION NIMA INTERACTING PROTEINS (ill) NUMBER OF SEQUENCES 14
(iv) CORRESPONDENCE ADDRESS
(A) ADDRESSEE Fish __ Richardson P C
(B) STREET 4225 Executive Square Suite 1400 (C) CITY La Jolla
(D) STATE CA
(E) COUNTRY USA
(F) ZIP 92037
(v) COMPUTER READABLE FORM (A) MEDIUM TYPE Floppy disk
(B) COMPUTER IBM PC compatible
(C) OPERATING SYSTEM PC-DOS/MΞ-DOΞ
(D) SOFTWARE Patentln Release #1 0 Version #1 25
(vi) CURRENT APPLICATION DATA (A) APPLICATION NUMBER PCT/US96/
(B) FILING DATE Herewith
(C) CLASSIFICATION
(viii) ATTORNEY/AGENT INFORMATION
(A) NAME Haile Ph D Lisa A (B) REGISTRATION NUMBER 38,347
(C) REFERENCE/DOCKET NUMBER 07251/011001
(ix) TELECOMMUNICATION INFORMATION
(A) TELEPHONE (619) 678-5070
(B) TELEFAX (619) 678-5099
(2) INFORMATION FOR SEQ ID NO 1
(l) SEQUENCE CHARACTERISTICS
(A) LENGTH 1014 base pairs
(B) TYPE nucleic acid
(C) STRANDEDNESS single (D) TOPOLOGY linear
(ii) MOLECULE TYPE DNA (genomic)
(ix) FEATURE
(A) NAME/KEY CDS
(B) LOCATION 25 513
(xi) SEQUENCE DESCRIPTION SEQ ID NO 1
TGCTGGCCAG CACCTCGAGG GAAG ATG GCG GAC GAG GAG AAG CTG CCG CCC 51
Met Ala Asp Glu Glu Lys Leu Pro Pro 1 5 GGC TGG GAG AAG CGC ATG AGC CGC AGC TCA GGC CGA GTG TAC TAC TTC 99
Gly Trp Glu Lys Arg Met Ser Arg Ser Ser Gly Arg Val Tyr Tyr Phe 10 IS 20 25 AAC CAC ATC ACT AAC GCC AGC CAG TGG GAG CGG CCC AGC GGC AAC AGC 147 Asn His lie Thr Asn Ala Ser Gin Trp Glu Arg Pro Ser Gly Asn Ser 30 35 40
AGC AGT GGT GGC AAA AAC GGG CAG GGG GAG CCT GCC AGG GTC CGC TGC 195 Ser Ser Gly Gly Lys Asn Gly Gin Gly Glu Pro Ala Arg Val Arg Cys
45 50 55
TCG CAC CTG CTG GTG AAG CAC AGC CAG TCA CGG CGG CCC TCG TCC TGG 243 Ser His Leu Leu Val Lys His Ser Gin Ser Arg Arg Pro Ser Ser Trp 60 65 70 CGG CAG GAG AAG ATC ACC CGG ACC AAG GAG GAG GCC CTG GAG CTG ATC 291
Arg Gin Glu Lys lie Thr Arg Thr Lys Glu Glu Ala Leu Glu Leu lie 75 80 85
AAC GGC TAC ATC CAG AAG ATC AAG TCG GGA GAG GAG GAC TTT GAG TCT 339 Asn Gly Tyr lie Gin Lys He Lys Ser Gly Glu Glu Asp Phe Glu Ser 90 95 100 105
CTG GCC TCA CAG TTC AGC GAC TGC AGC TCA GCC AAG GCC AGG GGA GAC 387 Leu Ala Ser Gin Phe Ser Asp Cys Ser Ser Ala Lys Ala Arg Gly Asp 110 115 120
CTG GGT GCC TTC AGC AGA GGT CAG ATG CAG AAG CCA TTT GAA GAC GCC 435 Leu Gly Ala Phe Ser Arg Gly Gin Met Gin Lys Pro Phe Glu Asp Ala
125 130 135
TCG TTT GCG CTG CGG ACG GGG GAG ATG AGC GGG CCC GTG TTC ACG GAT 483
Ser Phe Ala Leu Arg Thr Gly Glu Met Ser Gly Pro Val Phe Thr Asp 140 145 150 TCC GGC ATC CAC ATC ATC CTC CGC ACT GAG TGAGGGTGGG GAGCCCAGGC 533
Ser Gly He His He He Leu Arg Thr Glu 155 160
CTGGCCTCGG GGCAGGGCAG GGCGGCTAGG CCGGCCAGCT CCCCCTTGCC CGCCAGCCAG 593
TGGCCGAACC CCCCACTCCC TGCCACCGTC ACACAGTATT TATTGTTCCC ACAATGGCTG 653 GGAGGGGGCC CTTCCAGATT GGGGGCCCTG GGGTCCCCAC TCCCTGTCCA TCCCCAGTTG 713
GGGCTGCGAC CGCCAGATTC TCCCTTAAGG AATTGACTTC AGCAGGGGTG GGAGGCTCCC 773
AGACCCAGGG CAGTGTGGTG GGAGGGGTGT TCCAAAGAGA AGGCCTGGTC AGCAGAGCCG 833
CCCCGTGTCC CCCCAGGTGC TGGAGGCAGA CTCGAGGGCC GAATTGTTTC TAGTTAGGCC 893
ACGCTCCTCT GTTCAGTCGC AAAGGTGAAC ACTCATGCGG CAGCCATGGG CCCTCTGAGC 953 AACTGTGCAG ACCCTTTCAC CCCCAATTAA ACCCAGAACC ACTAAAAAAA AAAAAAAAAA 1013
A 1014
( 2 ) INFORMATION FOR SEQ ID NO 2
( l ) SEQUENCE CHARACTERISTICS
(A) LENGTH 163 ammo acids ( B) TYPE amino acid
( D ) TOPOLOGY linear
(ii ) MOLECULE TYPE protein
(xl ) SEQUENCE DESCRIPTION SEQ ID NO 2 Met Ala Asp Glu Glu Lys Leu Pro Pro Gly Trp Glu Lys Arg Met Ser
1 5 10 15
Arg Ser Ser Gly Arg Val Tyr Tyr Phe Asn His He Thr Asn Ala Ser 20 25 30 Gin Trp Glu Arg Pro Ser Gly Asn Ser Ser Ser Gly Gly Lys Asn Gly
35 40 45
Gin Gly Glu Pro Ala Arg Val Arg Cys Ser His Leu Leu Val Lys His 50 55 60
Ser Gin Ser Arg Arg Pro Ser Ser Trp Arg Gin Glu Lys He Thr Arg 65 70 75 80
Thr Lys Glu Glu Ala Leu Glu Leu He Asn Gly Tyr He Gin Lys He 85 90 95
Lys Ser Gly Glu Glu Asp Phe Glu Ser Leu Ala Ser Gin Phe Ser Asp 100 105 110 Cys Ser Ser Ala Lys Ala Arg Gly Asp Leu Gly Ala Phe Ser Arg Gly
115 120 125
Gin Met Gin Lys Pro Phe Glu Asp Ala Ser Phe Ala Leu Arg Thr Gly 130 135 140
Glu Met Ser Gly Pro Val Phe Thr Asp Ser Gly He His He He Leu 145 ISO 155 160
Arg Thr Glu
(2) INFORMATION FOR SEQ ID NO:3-
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 33 base pairs
(B) TYPE- nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY, linear
(ii) MOLECULE TYPE: DNA (genomic)
(vn) IMMEDIATE SOURCE-
(B) CLONE: NLKl(l)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:
GCGCCTGCAG TATCTATACT ATGGAATATC TGT 33 (2) INFORMATION FOR SEQ ID NO-4.
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH- 36 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single (D) TOPOLOGY: unknown
(n) MOLECULE TYPE: DNA (genomic) IMMEDIATE SOURCE- (B) CLONE. nlkl(2)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO 4
GCGCGGATCC GAGGTTTCAG AGGTGTCTGA AAGCAG 36
(2) INFORMATION FOR SEQ ID NO:5.
(l) SEQUENCE CHARACTERISTICS
(A) LENGTH- 33 base pairs
(B) TYPE: nucleic acid (C) STRANDEDNESS: single
(D) TOPOLOGY- unknown
(n) MOLECULE TYPE. DNA (genomic)
(vn) IMMEDIATE SOURCE-
(B) CLONE: nlkl(3)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO .5 •
GCGCGTACCA AGTACCACTC GTACTATTAT TCC 33
(2) INFORMATION FOR SEQ ID NO:6:
(l) SEQUENCE CHARACTERISTICS- (A) LENGTH: 13 amino acids
(B) TYPE, ammo acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(il) MOLECULE TYPE: protein
(vn) IMMEDIATE SOURCE:
(B) CLONE: HA tag to PIN1
(XI) SEQUENCE DESCRIPTION: SEQ ID NO:6
Met Tyr Asp Val Pro Asp Tyr Ala Ser Arg Pro Gin Asn
1 5 10 (2) INFORMATION FOR SEQ ID NO:7:
(l) SEQUENCE CHARACTERISTICS.
(A) LENGTH: 32 ammo acids
(B) TYPE: amino acid
(C) STRANDEDNESS: unknown (D) TOPOLOGY: unknown
(ii) MOLECULE TYPE: protein
(vii) IMMEDIATE SOURCE:
(B) CLONE: PGAL-PINl
(IX) FEATURE: (A) NAME/KEY: B ding-site
(B) LOCATION: 6..12 -53-
(XI) SEQUENCE DESCRIPTION SEQ ID NO 7
Met Ala Ser Tyr Pro Tyr Asp Val Pro Asp Tyr Ala Ser Pro Glu Phe
1 5 10 IS
Leu Val Asp Pro Pro Gly Ser Lys Asn Ser He Ala Arg Gly Lys Met 20 25 30
(2) INFORMATION FOR SEQ ID NO 8
(l) SEQUENCE CHARACTERISTICS
(A) LENGTH 39 amino acids
(B) TYPE amino acid
(C) STRANDEDNESS unknown
(D) TOPOLOGY unknown
(n) MOLECULE TYPE protein
(vn) IMMEDIATE SOURCE
(B) CLONE PIN1/HUMAN
(xi) SEQUENCE DESCRIPTION SEQ ID NO 8
Glu Lys Leu Pro Pro Gly Trp Glu Lys Arg Met Ser Arg Ser Ser Gly 1 5 10 15
Arg Val Tyr Tyr Phe Asn His He Thr Asn Ala Ser Gin Trp Glu Arg 20 25 30
Pro Ser Gly Asn Ser Ser Ser 35
(2) INFORMATION FOR SEQ ID NO 9
(l) SEQUENCE CHARACTERISTICS
(A) LENGTH 39 ammo acids
(B) TYPE ammo acid (C) STRANDEDNESS unknown
(D) TOPOLOGY unknown
(u) MOLECULE TYPE protein
(vn) IMMEDIATE SOURCE
(B) CLONE ESS1/SC
(xi) SEQUENCE DESCRIPTION SEQ ID NO 9
Thr Gly Leu Pro Thr Pro Trp Thr Val Arg Tyr Ser Lys Ser __ys Lys 1 5 10 15 Arg Glu Tyr Phe Phe Asn Pro Glu Thr Lys His Ser Gin Trp Glu Glu 20 25 30
Pro Glu Gly Thr Asn Lys Asp 35
(2) INFORMATION FOR SEQ ID NO: 10
(1) SEQUENCE CHARACTERISTICS
(A) LENGTH 38 ammo acids
(B) TYPE amino acid (C) STRANDEDNESS. unknown
(D) TOPOLOGY unknown
(ii) MOLECULE TYPE: protein
(vil) IMMEDIATE SOURCE
(B) CLONE Yap/Human
(xi) SEQUENCE DESCRIPTION SEQ ID NO 10
Val Pro Leu Pro Ala Gly Trp Glu Met Ala Lys Thr Ser Ser Gly Gin 1 5 10 15
Arg Tyr Phe Leu Asn His He Asp Gin Thr Thr Thr Trp Gin Asp Pro 20 25 30
Arg Lys Ala Met Leu Ser 35
(2) INFORMATION FOR SEQ ID NO: 11- (l) SEQUENCE CHARACTERISTICS:
(A) LENGTH- 38 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown (n) MOLECULE TYPE, protein
(vn) IMMEDIATE SOURCE.
(B) CLONE: Nedd4/Mouse
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 11-
Ser Pro Leu Pro Pro Gly Trp Glu Glu Arg Gin Asp Val Leu Gly Arg 1 5 10 15
Thr Tyr Tyr Val Asn His Glu Ser Arg Arg Thr Gin Trp Lys Arg Pro 20 25 30
Ser Pro Asp Asp Asp Leu
35
(2) INFORMATION FOR SEQ ID NO: 12 -55-
(l) SEQUENCE CHARACTERISTICS
(A) LENGTH 38 amino acids
(B) TYPE amino acid
(C) STRANDEDNESS unknown
(D) TOPOLOGY unknown
(n) MOLECULE TYPE protein
(vn) IMMEDIATE SOURCE
(B) CLONE RSP5/SC
(xi) SEQUENCE DESCRIPTION SEQ ID NO 12
Gly Arg Leu Pro Pro Gly Trp Glu Arg Arg Thr Asp Asn Phe Gly Arg 1 5 10 15
Thr Tyr Tyr Val Asp His Asn Thr Arg Thr Thr Thr Trp Lys Arg Pro 20 25 30
Thr Leu Asp Gin Thr Glu 35
(2) INFORMATION FOR SEQ ID NO 13
(l) SEQUENCE CHARACTERISTICS (A) LENGTH 38 amino acids
(B) TYPE amino acid
(C) STRANDEDNESS unknown
(D) TOPOLOGY unknown
(ii) MOLECULE TYPE protein
( n) IMMEDIATE SOURCE
(B) CLONE Dmd/Human
(xi) SEQUENCE DESCRIPTION SEQ ID NO 13
Thr Ser Val Gin Gly Pro Trp Glu Arg Ala He Ser Pro Asn Lys Val 1 5 10 15
Pro Tyr Tyr He Asn His Glu Thr Gin Thr Thr Cys Trp Asp His Pro 20 25 30
Lys Met Thr Glu Leu Tyr 35
(2) INFORMATION FOR SEQ ID NO 14
(l) SEQUENCE CHARACTERISTICS
(A) LENGTH 37 ammo acids
(B) TYPE amino acid (C) STRANDEDNESS unknown
(D) TOPOLOGY unknown
(n) MOLECULE TYPE protein (vn) IMMEDIATE SOURCE
(B) CLONE FE65/Rat
(xi) SEQUENCE DESCRIPTION SEQ ID NO 14
Ser Asp Leu Pro Ala Gly Trp Met Arg Val Gin Asp Thr Ser Gly Thr
1 5 10 IS
Tyr Tyr Trp His He Pro Thr Gly Thr Thr Gin Trp Glu Pro Pro Gly 20 25 30
Arg Ala Ser Pro Ser 35

Claims

What is claimed is:
1. Substantially pure Pin1 protein.
2. The Pin1 of claim 1, characterized as:
having a molecular weight of about 18kD;
having peptidyl-propyl cis/trans isomerase activity;
associating with NIMA protein kinase;
inhibiting the mitosis promoting function of NIMA when overexpressed; and
inducing mitotic arrest and nuclear fragmentation when depleted.
3. The Pin1 of claim 1, wherein the Pin1 is derived from a human cell.
4. A protein according to claim 1, wherein the amino acid sequence of said protein is substantially the same as the amino acid sequence set forth in SEQ ID NO:2 (Figure 2A).
5. A protein according to claim 1 having the same amino acid sequence as set forth in SEQ ID NO:2 (Figure 2A).
6. An isolated mammalian protein characterized as associating with NIMA protein kinase.
7. An isolated nucleic acid sequence encoding a protein according to claim 1.
8. An isolated nucleic acid according to claim 7 having a nucleotide sequence substantially the same as set forth in SEQ ID NO:1 (Figure 2A), or variations thereof which encode the same amino acid sequence, but employ different codons for some of the amino acids, or splice variant nucleotide sequences thereof.
9. A recombinant expression vector comprising a nucleic acid sequence according to claim 7.
10. A host cell containing the vector of claim 9.
11. An antibody which binds to the protein of claim 1 , or fragments of said
protein.
12. A method for identifying a protein that inhibits the mitosis promoting function of NIMA protein kinase, said method comprising:
culturing transformed cells containing the following:
a nucleic acid construct comprising a DNA binding domain operably associated with the coding sequence of NIMA, or functional fragments thereof; a nucleic acid library, wherein each member of said library comprises a transactivation domain operably associated with a protein encoding sequence; and a nucleic acid reporter construct comprising a response element for the
DNA binding domain operably associated with a reporter gene; and
monitoring for evidence of expression of reporter gene.
13. A method for identifying a protein that inhibits the mitosis promoting function of NIMA protein kinase, said method comprising:
culturing cells containing the following:
a nucleic acid construct comprising a transactivation domain operably associated with the coding sequence of NIMA, or fragments thereof;
a nucleic acid library, wherein each member of said library comprises a DNA binding domain operably associated with a protein encoding sequence; and a nucleic acid reporter construct comprising a response element for the DNA binding domain operably associated with a reporter gene; and
monitoring for evidence of expression of reporter gene.
14. A protein identified by the method of claim 12 or 13.
15. A method for controlling the growth of a cell comprising contacting the cell with a composition which modulates Pin1 protein activity or PIN1 expression.
16. The method of claim 15, wherein Pin1 activity or PIN1 expression is reduced with an inhibitor.
17. The method according to claim 16, wherein said inhibitor is a PPlase inhibitor.
18. The method of claim 16, wherein said inhibitor is selected from the group consisting of an anti-Pin1 antibody, an antisense nucleotide sequence and a ribozyme.
19. The method of claim 15, wherein Pin1 activity is increased in the presence of an activator.
20. The method of claim 15, wherein PIN1 expression is increased in the presence of a regulatory element.
21. The method of claim 20, wherein the regulatory element is an enhancer.
22. A method for identifying a protein that associates with Pin1 protein, said method comprising:
incubating said protein with Pin1 protein, or with a recombinant cell expressing Pin1 under conditions sufficient to allow the components to interact; and
determining the effect of said protein on Pin1 activity or expression.
23. A method for determining whether a composition modulates Pin1 activity, said method comprising:
incubating the composition with Pin1 protein, or with a recombinant cell expressing Pin1 under conditions sufficient to allow the components to interact; and
determining the effect of the substance on Pin1 activity or expression.
24. A method for treating a cell proliferative disorder in a subject, said method comprising administering to the subject in need thereof, an amount of Pin1 or PIN1 inhibitor effective to induce mitotic arrest and nuclear fragmentation.
25. A method for treating a cell proliferative disorder in a subject, said method comprising administering to the subject in need thereof, an amount of PIN1 enhancer sufficient to block the cell in G2.
PCT/US1996/017334 1995-11-13 1996-10-28 Nima interacting proteins WO1997017986A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP96941949A EP0880359B1 (en) 1995-11-13 1996-10-28 Nima interacting proteins
CA002235225A CA2235225C (en) 1995-11-13 1996-10-28 Nima interacting proteins
DE69634671T DE69634671T2 (en) 1995-11-13 1996-10-28 Nima interacting proteins
AU11158/97A AU1115897A (en) 1995-11-13 1996-10-28 Nima interacting proteins

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/555,912 1995-11-13
US08/555,912 US5972697A (en) 1995-11-13 1995-11-13 NIMA interacting proteins

Publications (1)

Publication Number Publication Date
WO1997017986A1 true WO1997017986A1 (en) 1997-05-22

Family

ID=24219099

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/017334 WO1997017986A1 (en) 1995-11-13 1996-10-28 Nima interacting proteins

Country Status (6)

Country Link
US (7) US5972697A (en)
EP (1) EP0880359B1 (en)
AU (1) AU1115897A (en)
CA (1) CA2235225C (en)
DE (1) DE69634671T2 (en)
WO (1) WO1997017986A1 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000048621A2 (en) * 1999-02-18 2000-08-24 Beth Israel Deaconess Medical Center Methods and compositions for regulating protein-protein interactions
EP1094800A2 (en) * 1998-06-09 2001-05-02 The Salk Institute For Biological Studies PEPTIDYL-PROLYL $i(CIS)-TRANS ISOMERASE INHIBITORS AND USES THEREFOR
US6462173B1 (en) 1997-09-08 2002-10-08 Max-Planck-Gesellschaft Zur Forderung Der Inhibitors of phosphoserine and phosphothreonine-proline-specific isomerases
EP1368630A2 (en) * 2001-02-09 2003-12-10 Pintex Pharmaceuticals, Inc. Pin1 as marker for abnormal cell growth
WO2004046329A2 (en) * 2002-11-14 2004-06-03 Pintex Pharmaceuticals, Inc. Levels of pin1 in normal and cancerous tissue
EP1446044A2 (en) * 2001-02-09 2004-08-18 Pintex Pharmaceuticals Pin1 as a marker for prostate cancer
US8129131B2 (en) 1999-11-29 2012-03-06 Beth Israel Deaconess Medical Center Pin1 as a marker for abnormal cell growth
WO2013185055A1 (en) 2012-06-07 2013-12-12 Beth Israel Deaconess Medical Center, Inc. Methods and compositions for the inhibition of pin1
US9439884B2 (en) 2011-05-26 2016-09-13 Beth Israel Deaconess Medical Center, Inc. Methods for the treatment of immune disorders
US9796784B2 (en) 2009-10-27 2017-10-24 Beth Israel Deaconess Medical Center, Inc. Methods and compositions for the generation and use of conformation-specific antibodies
US9968579B2 (en) 2014-07-17 2018-05-15 Beth Isreal Deaconess Medical Center, Inc. ATRA for modulating Pin1 activity and stability
US10265288B2 (en) 2011-03-14 2019-04-23 Beth Israel Deaconess Medical Center, Inc. Methods and compositions for the treatment of proliferative disorders
US10351914B2 (en) 2014-07-17 2019-07-16 Beth Israel Deaconess Medical Center, Inc. Biomarkers for Pin1-associated disorders
US10487114B2 (en) 2011-04-27 2019-11-26 Beth Israel Deaconess Medical Center, Inc. Methods for administering peptides for the generation of effective c/s conformation-specific antibodies to a human subject in need thereof
US10548864B2 (en) 2015-03-12 2020-02-04 Beth Israel Deaconess Medical Center, Inc. Enhanced ATRA-related compounds for the treatment of proliferative diseases, autoimmune diseases, and addiction conditions

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7125677B2 (en) * 1995-11-13 2006-10-24 The Salk Institute For Biological Studies NIMA interacting proteins
US5972697A (en) * 1995-11-13 1999-10-26 The Salk Institute For Biological Studies NIMA interacting proteins
ATE400814T1 (en) * 1999-11-29 2008-07-15 Beth Israel Hospital PIN1 AS A STAGE MARKER OF AGGRESSIVITY AND/OR METASTASIZATION ABNORMAL CELL GROWTH
AU2003213698A1 (en) * 2002-03-01 2003-09-16 Pintex Pharmaceuticals, Inc. Methods for designing specific inhibitors for pin1 proline isomerase and pin1-related molecules
AU2003216501A1 (en) * 2002-03-01 2003-09-16 Pintex Pharmaceuticals, Inc. Methods of treating pin1 associated disorders
WO2004033666A2 (en) * 2002-10-11 2004-04-22 Board Of Regents, The University Of Texas System Method and compounds for inhibiting hec1 activity for the treatment of proliferative diseases
US20040176277A1 (en) * 2003-03-03 2004-09-09 Pintex Pharmaceuticals, Inc. Methods of treating Pin1 associated disorders
WO2004101745A2 (en) * 2003-05-08 2004-11-25 Beth Israel Deaconess Medical Center, Inc. NOVEL REGULATORY MECHANISMS OF NF-kappaB
US20050250742A1 (en) * 2004-03-03 2005-11-10 Pfizer Inc. Phosphate/sulfate ester compounds and pharmaceutical composition for inhibiting protein interacting NIMA (PIN1)
US20070027067A1 (en) * 2005-05-13 2007-02-01 Felicia Etzkorn Transition-state Inhibitors of Pin1, alpha-Ketoamide-containing peptidomimetics, and synthesis thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5443962A (en) * 1993-06-04 1995-08-22 Mitotix, Inc. Methods of identifying inhibitors of cdc25 phosphatase

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5972697A (en) 1995-11-13 1999-10-26 The Salk Institute For Biological Studies NIMA interacting proteins
US7125677B2 (en) 1995-11-13 2006-10-24 The Salk Institute For Biological Studies NIMA interacting proteins
US6462173B1 (en) 1997-09-08 2002-10-08 Max-Planck-Gesellschaft Zur Forderung Der Inhibitors of phosphoserine and phosphothreonine-proline-specific isomerases
US6495376B1 (en) 1999-02-18 2002-12-17 Beth Israel Deaconess Medical Center Methods and compositions for regulating protein-protein interactions
US20020025521A1 (en) 1999-11-29 2002-02-28 Lu Kun Ping Pin1 as a marker for abnormal cell growth
US6436703B1 (en) 2000-03-31 2002-08-20 Hyseq, Inc. Nucleic acids and polypeptides
US20020150898A1 (en) 2000-04-18 2002-10-17 Tang Y. Tom Novel nucleic acids and polypeptides
EP2241636A1 (en) * 2002-03-13 2010-10-20 Genomic Health, Inc. Gene expression profiling in biopsied tumor tissues
BR0312555A (en) * 2002-07-09 2007-06-19 Pfizer pin1 peptidyl prolyl isomerase polypeptides, their crystal structures and their use for drug design

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5443962A (en) * 1993-06-04 1995-08-22 Mitotix, Inc. Methods of identifying inhibitors of cdc25 phosphatase

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
CELL, 05 May 1995, Vol. 81, LU et al., "Evidence for a NIMA-Like Mitotic Pathway in Vertebrate Cells", pages 413-424. *
CELL, 22 April 1988, Vol. 53, OSMANI et al., "Mitotic Induction and Maintenance by Overexpression of a GS-Specific Gene That Encodes a Potential Protein Kinase", pages 237-244. *
EMBO JOURNAL, 1994, Vol. 13, No. 20, O'CONNELL et al., "Premature Chromatin Condensation Upon Accumulation of NIMA", pages 4926-4937. *
EMBO JOURNAL, 1994, Vol. 13, No. 9, LU et al., "Expression of the Noncatalytic Domain of the NIMA Kinase Causes a G2 Arrest in Aspergillus Nidulans", pages 2103-2113. *
See also references of EP0880359A4 *

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6462173B1 (en) 1997-09-08 2002-10-08 Max-Planck-Gesellschaft Zur Forderung Der Inhibitors of phosphoserine and phosphothreonine-proline-specific isomerases
EP1094800A4 (en) * 1998-06-09 2004-11-03 Salk Inst For Biological Studi PEPTIDYL-PROLYL $i(CIS)-TRANS ISOMERASE INHIBITORS AND USES THEREFOR
EP1094800A2 (en) * 1998-06-09 2001-05-02 The Salk Institute For Biological Studies PEPTIDYL-PROLYL $i(CIS)-TRANS ISOMERASE INHIBITORS AND USES THEREFOR
WO2000048621A3 (en) * 1999-02-18 2000-12-21 Beth Israel Hospital Methods and compositions for regulating protein-protein interactions
US6495376B1 (en) 1999-02-18 2002-12-17 Beth Israel Deaconess Medical Center Methods and compositions for regulating protein-protein interactions
EP2056111A1 (en) 1999-02-18 2009-05-06 Beth Israel Deaconess Medical Center, Inc. Methods and compositions for regulating protein-protein interactions
WO2000048621A2 (en) * 1999-02-18 2000-08-24 Beth Israel Deaconess Medical Center Methods and compositions for regulating protein-protein interactions
AU782056B2 (en) * 1999-02-18 2005-06-30 Beth Israel Deaconess Medical Center Methods and compositions for regulating protein-protein interactions
US8129131B2 (en) 1999-11-29 2012-03-06 Beth Israel Deaconess Medical Center Pin1 as a marker for abnormal cell growth
EP1368630A4 (en) * 2001-02-09 2004-06-16 Pintex Pharmaceuticals Inc Pin1 as marker for abnormal cell growth
EP1446044A2 (en) * 2001-02-09 2004-08-18 Pintex Pharmaceuticals Pin1 as a marker for prostate cancer
EP1446044A4 (en) * 2001-02-09 2007-08-08 Pintex Pharmaceuticals Inc Pin1 as a marker for prostate cancer
US7592145B2 (en) 2001-02-09 2009-09-22 Pintex Pharmaceuticals, Inc. Pin 1 as a marker for prostate cancer
EP1368630A2 (en) * 2001-02-09 2003-12-10 Pintex Pharmaceuticals, Inc. Pin1 as marker for abnormal cell growth
US7501255B2 (en) 2002-11-14 2009-03-10 Pintex Pharmaceuticals, Inc. Levels of Pin1 in normal and cancerous tissue
WO2004046329A2 (en) * 2002-11-14 2004-06-03 Pintex Pharmaceuticals, Inc. Levels of pin1 in normal and cancerous tissue
WO2004046329A3 (en) * 2002-11-14 2005-01-13 Pintex Pharmaceuticals Inc Levels of pin1 in normal and cancerous tissue
US9796784B2 (en) 2009-10-27 2017-10-24 Beth Israel Deaconess Medical Center, Inc. Methods and compositions for the generation and use of conformation-specific antibodies
US10265288B2 (en) 2011-03-14 2019-04-23 Beth Israel Deaconess Medical Center, Inc. Methods and compositions for the treatment of proliferative disorders
US10485780B2 (en) 2011-03-14 2019-11-26 Beth Israel Deaconess Medical Center, Inc. Methods and compositions for the treatment of proliferative disorders
US10487114B2 (en) 2011-04-27 2019-11-26 Beth Israel Deaconess Medical Center, Inc. Methods for administering peptides for the generation of effective c/s conformation-specific antibodies to a human subject in need thereof
US9439884B2 (en) 2011-05-26 2016-09-13 Beth Israel Deaconess Medical Center, Inc. Methods for the treatment of immune disorders
US9730941B2 (en) 2012-06-07 2017-08-15 Beth Israel Deaconess Medical Center, Inc. Methods and compositions for the inhibition of Pin1
WO2013185055A1 (en) 2012-06-07 2013-12-12 Beth Israel Deaconess Medical Center, Inc. Methods and compositions for the inhibition of pin1
US10413548B2 (en) 2012-06-07 2019-09-17 Beth Israel Deaconess Medical Center, Inc. Methods and compositions for the inhibition of Pin1
US11129835B2 (en) 2012-06-07 2021-09-28 Beth Israel Deaconess Medical Center, Inc. Methods and compositions for the inhibition of PIN1
US9968579B2 (en) 2014-07-17 2018-05-15 Beth Isreal Deaconess Medical Center, Inc. ATRA for modulating Pin1 activity and stability
US10351914B2 (en) 2014-07-17 2019-07-16 Beth Israel Deaconess Medical Center, Inc. Biomarkers for Pin1-associated disorders
US10548864B2 (en) 2015-03-12 2020-02-04 Beth Israel Deaconess Medical Center, Inc. Enhanced ATRA-related compounds for the treatment of proliferative diseases, autoimmune diseases, and addiction conditions

Also Published As

Publication number Publication date
EP0880359B1 (en) 2005-04-27
DE69634671D1 (en) 2005-06-02
EP0880359A4 (en) 1999-09-29
US5972697A (en) 1999-10-26
EP0880359A1 (en) 1998-12-02
AU1115897A (en) 1997-06-05
US20070042467A1 (en) 2007-02-22
CA2235225A1 (en) 1997-05-22
US6596848B1 (en) 2003-07-22
US20050049404A1 (en) 2005-03-03
US20050027107A1 (en) 2005-02-03
US7148003B2 (en) 2006-12-12
US20050033032A1 (en) 2005-02-10
US5952467A (en) 1999-09-14
US7164012B2 (en) 2007-01-16
US7125955B2 (en) 2006-10-24
DE69634671T2 (en) 2006-03-02
CA2235225C (en) 2008-08-26

Similar Documents

Publication Publication Date Title
EP0880359B1 (en) Nima interacting proteins
JP3745558B2 (en) Tumor protein kinase
US20060121527A1 (en) Oncoprotein protein kinase
JP2004222728A (en) Human cyclin e
JPH08509504A (en) Protein kinase
KR100536394B1 (en) Function modulators of FAS / AP01 receptors
JP2008301815A (en) STIMULUS-INDUCIBLE I(kappa)B KINASE [IKK] SIGNALSOME
SK287890B6 (en) Isolated polypeptide that binds to TRAF2 and modulates activity of NF-B, DNA sequence encoding its, pharmaceutical composition containing them and their use
US7125677B2 (en) NIMA interacting proteins
JP4633259B2 (en) Compounds and methods for modulating NF-κB activation
JP2001524813A (en) IκB kinase, subunits thereof, and methods of using them
CA2370098C (en) Human beta-trcp protein
CA2219775A1 (en) Murine and human box-dependent myc-interacting protein (bin1) and uses therefor
US6387642B2 (en) Method for identifying a reagent that modulates Myt1 activity
US6307035B1 (en) BRCA1 associated polynucleotide (BAP-1) and uses therefor
WO1998005968A9 (en) Brca1 associated protein (bap-1) and uses therefor
JPH10117788A (en) Human myt-1 kinase clone
WO2000032787A1 (en) Therapeutic agents comprising an e3 ubiquitin ligase for use in degenerative disorders
EP0812360A1 (en) Human geranylgeranyl pyrophosphate synthetase
AU696986B2 (en) Novel proteins which bind to retinoblastoma protein and their encoding DNA equences
JP2002020313A (en) Human dna topoisomerase i-alpha

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref document number: 2235225

Country of ref document: CA

Ref country code: CA

Ref document number: 2235225

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1996941949

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: JP

Ref document number: 97518874

Format of ref document f/p: F

WWP Wipo information: published in national office

Ref document number: 1996941949

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1996941949

Country of ref document: EP