WO2005075508A2 - Hyperactive stat molecules and their use in assays employing gene activation - Google Patents

Hyperactive stat molecules and their use in assays employing gene activation Download PDF

Info

Publication number
WO2005075508A2
WO2005075508A2 PCT/EP2005/001462 EP2005001462W WO2005075508A2 WO 2005075508 A2 WO2005075508 A2 WO 2005075508A2 EP 2005001462 W EP2005001462 W EP 2005001462W WO 2005075508 A2 WO2005075508 A2 WO 2005075508A2
Authority
WO
WIPO (PCT)
Prior art keywords
stat
statl
cell
cells
nuclear
Prior art date
Application number
PCT/EP2005/001462
Other languages
French (fr)
Other versions
WO2005075508A3 (en
WO2005075508A8 (en
Inventor
Thomas Meyer
Uwe Vinkemeyer
Original Assignee
Forschungsverbund Berlin E.V.
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
Priority claimed from EP04077629A external-priority patent/EP1637540A1/en
Application filed by Forschungsverbund Berlin E.V. filed Critical Forschungsverbund Berlin E.V.
Publication of WO2005075508A2 publication Critical patent/WO2005075508A2/en
Publication of WO2005075508A8 publication Critical patent/WO2005075508A8/en
Publication of WO2005075508A3 publication Critical patent/WO2005075508A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4705Regulators; Modulating activity stimulating, promoting or activating activity

Definitions

  • the invention relates to Stat molecules which are mutated and their use in the activation of gene expression, in the screening of inhibitors or activators of transcription factors, the identification of binding proteins, the production of a drug for diagnosis and/or treatment of diseases associated with the activation of gene expression as well as in the targeting of active substances.
  • DNA replication and RNA biogenesis take place in the cell nucleus, whereas protein synthesis essentially proceeds in the cytoplasm.
  • the close integration of these activities depends on a selective transport of the proteins and ribonucleoprotein particles between nucleus and cytoplasm requiring a coupled import and export of compounds at many molecular processes.
  • This translocation of proteins, DNA and/or RNA proceeds via nuclear pores which are located in the membrane enclosing the cell nucleus. Since the translocation is mediated, inter alia, via distinct signals, the process is termed signal transduction.
  • the Stat (signal transducers and activators of transcription) proteins constitute an ancient and evolutionary conserved family of transcription factors involved in the regulated expression of numerous target genes in response to cytokines and growth factors (Darnell, J . E., Jr., Science 277, 1630- 1635 (1997)).
  • Polypeptide growth factors and cytokines elicit their effects by activating specific cell-surface receptors, thereby initiating signaling cascades.
  • Canonical signaling through the Janus tyrosine kinase (Jak) / Stat pathway begins at the cell membrane within the cytoplasm.
  • the binding of the mentioned ligands to their cognate receptors initiates a number of tyrosine phosphorylations catalyzed by Jak.
  • the bound Stat monomers detach from their receptor docking sites after phosphorylation of a single tyrosine residue at their C-terminus finally forming reciprocal homo- or heterodimers.
  • This sequence of events is commonly referred to as “Stat activation” triggering the accumulation of Stat dimers in the nucleus within a certain period.
  • Stat activation triggering the accumulation of Stat dimers in the nucleus within a certain period.
  • GAS palindromic DNA recognition sites
  • NPC nuclear pore complex
  • the transport substrates are distinguished in their amino acid sequence by the presence of cis-acting nuclear localization signals (NLS) and/or nuclear export signals (NES). Recently, such transport signals were also identified in the Stats.
  • dsNLS dimer-specific NLS
  • the DNA binding domain of the Statl molecule harbors a dimer-specific NLS (dsNLS) that constitutes the binding surface for the adaptor protein NPI-1 of the importin- ⁇ family, which tethers the Statl protein to the nuclear import factor p97 in a Ran-dependent manner. Destruction of the dsNLS precludes nuclear translocation of phosphorylated Statl and thus results in the loss of cytokine-inducible transcriptional responses.
  • dsNLS dimer-specific NLS
  • interface II the alternate surface - termed interface II - was proposed to mediate Stat dimerization as requirement for the formation of higher order Stat complexes on DNA (Chen et al, Protein Science 12, 361-365 (2003)). These studies analyzed point mutations which were introduced in the Statl N-domain at several sites of each interface. The dimerization properties of these mutated proteins were determined by analytical ultracentrifugation and gel filtration. Among others, the mutant Statl F77A characterized by a phenylalanine-to-alanine substitution on interface II was investigated and shown to be a monomer. Beside the fact that interface II is more extensive than interface I, it also involves interactions between hydrophobic residues probably affected by the replacement of phenylalanine.
  • the new results reflect a nonspecific domain destabilization rather than a specific defect in dimerization.
  • Stat proteins regulate many of the genes controlling cellular functions like cellular growth, differentiation and survival. Given the importance of Stats in the control of normal physiological processes, it is self-evident that inappropriate activation of these proteins is found in various disease patterns. Usually, cytokine-induced transcription is a transitory process which can take from a few minutes to hours. Cells with a deviantly high and prolonged Jak or Stat activation are susceptible to transformations and associated with abnormal developments (Frank, D. A., Mol. Med. 5, 432-456 (1999)).
  • Interferences in the signaling pathways are the crucial factor for the appearance of pathogenic phenotypes such as human malignancies, leukemia, disorders in cell growth, defects of the immune system or in the metabolic performance of liver, kidneys and mammary gland, septic shock, arthritis, numerous cardiovascular diseases and other things.
  • the technical problem forming the basis of the present invention is to provide agents which modify the transcription activity of target genes and to provide agents and methods for the screening and targeting of active substances which modify the transcription activity, so that they are suitable in the diagnosis and/or treatment of diseases associated with the abnormal Stat induction of target genes.
  • the present invention solves this problem by providing a Stat molecule which is characterized in that at least one conserved hydrophobic amino acid on the surface of the N-domain of the Stat wild type is mutated.
  • Stat proteins consist of a large core domain including the Src homology domain 2 (SH2) responsible for dimer formation and the DNA binding module which denotes an amino acid sequence capable of binding to a specific DNA sequence.
  • the core is connected through a proteolytically cleavable peptide to the smaller N-domain which engages in a multitude of protein-protein interactions. This N-domain mediates cooperative DNA binding of Stat dimers.
  • Stat6 are able to form higher order complexes (dimer-dimer or higher) on promoters containing two or more neighboring Stat binding sites.
  • Stat tetramers are also mandatory for full transcription activity at many promoters that contain only a single (not optimal) canonical GAS site.
  • promoter refers to a DNA sequence, mostly situated upstream (5') of the coding sequence of a structural gene which controls the expression of the coding region by providing a recognition sequence for an RNA polymerase and/or other factors required to start transcription at the appropriate position. Promoter sequences are necessary, but not sufficient in any case, to control expression of the gene.
  • the mutation of a conserved residue located in the alternate surface does not compromise protein stability, but causes a severe decrease in tetramer formation of several Stat molecules without overtly affecting the DNA binding of the dimer.
  • the novel penotype of the mutants makes insights possible into the role that DNA binding plays in nuclear accumulation of Stat proteins. It was previously hypothesized that the access to a nuclear export signal (NES) within the coiled-coil domain is regulated by DNA binding. According to this model, the Stat association to DNA would mask the NES and thus preclude nuclear export. Consequently, the loss of DNA binding would result in the loss of nuclear accumulation.
  • the novel phenotype of the mutants actually reveals that it is dimerization only (and not the subsequent DNA binding) that masks the region required for the nuclear export of Stat proteins. Thus, DNA binding is not necessary, but sufficient for nuclear retention of Stat molecules.
  • the mutated Stats are still functional transcription factors that retain cytokine responsiveness.
  • the interface centered around the critical hydrophobic residue, mainly represented by phenylalanine, fulfills distinct functions that are subjected to control by the tyrosine phosphorylation and DNA binding status of the Stat molecules. Due to the mutation, the gene induction depends on promoter strength whereby either an increase or a decrease of the transcription activity could be recognized.
  • the wild type of Stat2, Stat3, Stat ⁇ or Stat6 is mutated.
  • the Stat molecule is mutated in at least one conserved hydrophobic amino acid on the surface of the N-domain and characterized in that the Stat molecule is derived from the wild type of Stat2, Stat3, Stat ⁇ or State.
  • the Stat family of transcription factors comprise seven mammalian proteins with known homologues in all multicellular animal phyla. Stat proteins are activated by a wide range of different cytokines such as interleukins and prolactin, as well as growth factors. Accordingly, gene ablation experiments uncovered mammary gland defects, growth abnormalities and an absent interleukin response underlining the significance of regulating the Stat activity as tool for gene induction. In particular, disturbances in the signaling pathways were figured out as cancer inducing parameter.
  • the amino acids of the N- domain in position 81 and/or position 82 of Stat ⁇ or in equivalent positions of the homologous proteins are mutated.
  • Stat ⁇ a and Stat ⁇ b are two highly related proteins with a sequence identity of -96%. Possible mutations include the substitution, deletion or modification of the amino acid. They may occur in position 81 and/or position 82 of Stat ⁇ or in equivalent positions of homologous proteins.
  • Homologous proteins are members of the aforementioned Stat family, preferably Stat2, Stat3, Stat ⁇ or Stat6. Although they display a great sequence similarity, minor shifts of distinct residues within the primary structure are present. With regard to higher structure levels (secondary and/or tertiary structure), the shifted positions are referred to as equivalent.
  • the positions 77 and 78 in Stat 3 and Stat6 as well as the positions 81 and 82 in Stat2 and Stat ⁇ are considered as equivalent.
  • the altered residues in position 81 and/or position 82 of Stat ⁇ or equivalent in homologous proteins cannot maintain N-domain interactions in the full-length Stat molecule resulting in the linked phenomena of tetramerization- and dephosphorylation-deficiency, and their consequences on gene induction in a promoter-specific manner.
  • the amino acid in position 81 of Stat ⁇ or in the equivalent position of the homologous proteins is substituted by alanine. Due to its neutral properties originated from the short side chain of a single methyl group, alanine is especially suited for mutagenesis studies.
  • different residues in an equivalent position are present. These differences are preferably of equivalent matter by changing a hydrophobic amino acid by another hydrophobic one, for example.
  • the hydrophobic residues in the equivalent positions 77 and 81 comprise the following amino acids within those Stat molecules preferred in the meaning of the invention: leucine in position 81 of Stat2, valine in position 77 of Stat3, phenylalanine in position 81 of Stat ⁇ and isoleucine in position 77 of Stat6.
  • Human Stat5aF81A (1-710): magwiqaqqql qgdalrq qv lygqhfpiev rhylaq ies qpwdaidldn pqdraqatql leglvqelq kaehqvgedg allkiklghy atqlqktydr cplelvrcir hilyneqrlv reanncsspa gilvdamsqk hlqinqtfee lr vtqdten elkklqqtqe yfiiqyqesl riqaqfaqla qlspqerlsr etalqqkqvs lea lqreaq tlqqyrvela ek qktlqll rkqqtiildd eliq krrqq lagnggppeg sldvlqs
  • Human Stat5bF81A (1-715): mavwiqaqqql qgealhqmqa lygqhfpiev rhylsqwies qa dsvdldn pqenikatql leglvqelqk kaehqvgedg allkiklghy atqlqntydr cpmelvrcir hilyneqrlv reanngsspa gsladamsqk hlqinqtfee lrlvtqdten elkklqqtqe yfiiqyqesl riqaqfgpla qlspqerlsr etalqqkqvs leawlqreaq tlqqyrvela ekhqktlqll rkqqtiildd eliqwkrrqq lagnggppeg sldv
  • This invention is based on a mutation which is bold.
  • the amino acid in position 82 of Stat ⁇ or in the equivalent position of homologous proteins is substituted by serine.
  • the amino acid in position 82 of Stat ⁇ or in the equivalent position of homologous proteins is leucine.
  • the mutants Stat ⁇ aF81A and Stat ⁇ bF81A of this invention display a couple of novel features which are contrary to the wild type phenotype.
  • the mutants of this invention are tetramerization-deficient causing a promoter- dependent decrease in the strength of DNA binding. Their ability to bind tande ly linked GAS motifs in a cooperative manner is completely lost in comparison with the wild type. Contrary, the binding to a single optimal GAS site is similar to the wild type indicating a DNA recognition as dimer only. In consequence of the usage of natural promoter fragments, the reduced DNA binding is accompanied by a reduced transcription activity as well. Since natural promoters often harbor a weak single canonical GAS site, Stat oligomerization is required for high transcriptional efficiency. This ability to form tetramers is tested by EMSAs (electrophoretic mobility shift assays).
  • the nuclear accumulation of the Statl mutants is uncoupled from the DNA binding capacity.
  • the mutated phenotype displays a Stat accumulation which is 200 to 300%, prolonged compared to the wild type. Furthermore, due to the tetramerization defect the mutants form oligomers less frequently and thus maintain a high intranuclear mobility which is equivalent to the wild type.
  • Stat ⁇ aF81A and Stat ⁇ bF81A show prolonged tyrosine phosphorylation and insensitivity towards staurosporine, which is indicative of a reduced dephosphorylation rate.
  • the kinetics of dephosphorylation of wild type Stat ⁇ proteins is longer than that observed in lymphoid cells, where it occurs within 0.3 to 2 h.
  • mutation of residue 81 also prolongs tyrosine phosphorylation even further, as the phosphotyrosine signal now remains detectable for more than 10 h.
  • the N-domain point mutation has an impact on the gene transcription in a promoter-specific manner.
  • Reporter genes containing optimal GAS sites are transactivated as good as or even better up to 300%, preferably up to 200% and more preferably up to 100% by the Stat mutants than by the respective wild type counterpart.
  • the effective binding to such sites is less dependent upon cooperativity since the multimerization of optimal sites mimics oligomerization.
  • transactivation is lower up to 100%, preferably up to 7 ⁇ % and more preferably up to 50% with the mutants in comparison to the wild type Stat molecule.
  • Stat oligomerization acts as requirement of the subsequent DNA binding ability and an adequate gene induction.
  • the mutated Stat molecules are dimerized.
  • Stat dimers are the basic scaffold for subsequent DNA binding. Based on the dimerization, Statl and Stat3 to Stat6 are able to form higher order complexes (dimer-dimer or higher) on any DNA promoter which does not display an optimal consensus sequence. However, this inherent feature of higher oligomerization is abrogated in the described mutants.
  • the present invention also relates to a fusion protein comprising the mutated
  • Stat molecule and at least one marker structure.
  • a “fusion protein” is a protein which consists of amino acid sequences derived from at least two different sources. In context with fusion proteins, a
  • heterologous amino acid sequence is a sequence derived from a source other than the source of the other components of the fusion protein.
  • heterologous "marker structures” are understood to be proteins which are coupled to the Stat mutants, enabling the detection of them by means of various biological, chemical and/or physical methods.
  • a reporter gene encodes such a detectable marker protein.
  • the reporter gene according to the invention either can be the overall DNA construct of the protein to be detected. However, it may also be not more than that DNA portion of the protein which has detectable activity. Consequently, any DNA structure encoding a region detectable by means of various detection methods may serve as reporter gene. Preferred in this context are expression constructs encoding proteins which can be detected easily and with high sensitivity.
  • the inventive marker structure in the fusion protein can be chloramphenicol acetyl transferase (CAT).
  • CAT catalyzes the transfer of an acetyl residue from acetyl-CoA to chloramphenicol.
  • CAT has the advantage of being stable in a cell, with a half-life of about 50 h. This is particularly advantageous where e. g. yields of reporter gene as high as possible are to be achieved.
  • CAT has a good signal/background ratio.
  • Luciferase advantageously has a high sensitivity and, owing to a relatively short half-life of about three hours, it is well-suited for induction studies.
  • a marker structure including two luciferases with distinct properties, one luciferase, e. g. from Phottinus pyralis, detecting the activity of the own promoter, while the co-transfected luciferase, e. g. from Renilla reniformis, is under control of the constitutive promoter and thus can be used advantageously for standardization purposes.
  • marker structures in the meaning of the invention are ⁇ >-gaiactosidase, human growth hormone and secreted alkaline phosphatase.
  • the marker structures of the invention can also be used as reporter genes in qualitative detection. In qualitative detection, it is possible e. g. to determine whether a selected cell has been transfected or not, or, it is possible to determine in the entire organism, in which type of tissue a promoter is active.
  • reporter genes for qualitative detection are the genes for luciferase and ⁇ -galactosidase.
  • the marker structure is green fluorescent protein (GFP), glutathione S-transferase (GST) and/or a fragment thereof.
  • the green fluorescent protein advantageously has low cytotoxic activity or none at all.
  • the absorption and emission maxima of green fluorescent protein are similar to that of fluorescein, so that e. g. a GFP-positive cell can be detected using the same methods as with cells stained with fluorescein, i. e., under UV light, under a fluorescence microscope, or in a cell sorter (FACS), for example.
  • preferred marker proteins which can be fused with the Stat mutants are GST as well as other fluorescent proteins such as the red or the yellow protein, as well as the maltose-binding protein (MBP), and others.
  • the fusion protein contains the marker structure at the N-terminus and/or the C-terminus.
  • the arrangement will depend on the maintenance of the three-dimensional structure and the functional activity of both Stat mutant and marker structure.
  • the integration within the Stat mutant may disrupt its intermolecular order, however it should not be excluded.
  • the invention also relates to a nucleic acid sequence which encodes the Stat mutant or the fusion protein.
  • nucleic acid sequence refers to a natural or synthetic polymer of single- or double-stranded DNA or RNA alternatively including synthetic, non- natural or modified nucleotides which could be incorporated in DNA or RNA polymers.
  • Each nucleotide consists of a sugar moiety, a phosphate moiety, and either a purine or pyrimidine residue.
  • nucleic acid sequence which encodes refers to that part of a gene which enciphers a protein, a polypeptide or a part thereof.
  • the regulatory sequences and/or elements controlling the initiation or termination of transcription are excluded.
  • the coding sequence and/or the regulatory element can normally be found in cells, in which case it is referred to as “autologous” or “endogenic”, or cannot be situated in cells, in which case it is referred to as “heterologous”.
  • Gene denotes a DNA sequence encoding a specific protein, and regulatory elements controlling the expression of said DNA sequence.
  • a heterologous gene may also be composed of autologous elements arranged in an order and/or orientation which is normally not found in that cell the gene is transferred into.
  • a heterologous gene can be derived completely or partially from any source known in the art, including a bacterial or viral genome or episome, eukaryotic nuclear or plasmid DNA, cDNA, or chemically synthesized DNA.
  • the structural gene may form a continuous coding region, or may comprise one or more introns bordered by suitable splice junctions.
  • the structural gene can consist of segments derived from various naturally occurring or synthetic sources.
  • the present invention also relates to a vector comprising the above-described nucleic acid sequence, particularly to a bacterial vector such as a plasmid, or a virus.
  • vector denotes a recombinant DNA construct which can be a plasmid, a virus, or an autonomously replicating sequence, a phage, or a nucleotide sequence, which is linear or circular, consists of single- or double-stranded DNA or RNA, wherein a number of nucleotide sequences are linked or recombined to form a unique construction, and which is capable of introducing a promoter fragment and a DNA sequence of a selected gene product in sense or antisense orientation into a cell, together with suitable non-translated 3' sequences.
  • Plasmids are genetic elements which are stable inherited without being part of the chromosome of their host cell. They may comprise DNA or RNA, and they can be both linear and circular. Plasmids encode molecules ensuring their replication and stable inheritance during cell replication, and they are capable of encoding products of considerable importance for medicine, agriculture and environment. For example, they encode toxins which massively increase the virulence of pathogenic bacteria. They are also capable of encoding genes imparting resistance to antibiotics. In molecular biology, plasmids are generally used as vectors to clone and express recombinant genes.
  • plasmids are generally described using the small letter p preceded or followed by capital letters and/or numerals.
  • the starting plasmids disclosed in the present specification either are commercially available, accessible to the public, or can be constructed from available plasmids by routine use of well-known, published methods.
  • Many plasmids and other cloning and expression vectors which can be used according to the invention are well-known and easily available to the skilled artisan.
  • a person skilled in the art can easily construct any number of other plasmids suitable for the use in this invention.
  • the properties, construction and use of such plasmids, as well as other vectors, are readily accessible to those skilled in the art from the present disclosure.
  • the present invention relates to host cells transfected with a vector of the invention, particularly to prokaryotic or eukaryotic cells.
  • the present invention also relates to cell cultures, tissues, organs and the like, which comprise cells including an above-described plasmid or vector.
  • the term "host cell” relates to a cell which has been genetically modified by the transfer of a chimeric, heterologous or autologous nucleic acid sequence or derivatives thereof still including said sequence. These cells are also referred to as "transgenic cells". Where an autologous nucleic acid sequence is transferred, the number of copies of this sequence in the host cell is higher than that of the naturally occurring sequences.
  • Object of the present invention is also the use of the Stat mutants of the invention, their dimers, the fusion protein of the invention, the nucleic acid sequence of the invention, the vector of the invention or the host cell of the invention for expression of a target protein, for investigating effects caused by different gene induction levels, for screening of active substances which modify the transcription activity of target genes, for identifying binding partners or for the production of a drug for the diagnosis and/or treatment of diseases associated with abnormal Stat induction of target genes.
  • Binding assays are usually performed to detect protein-protein interactions.
  • a well-established system represents the two-hybrid system, preferably a yeast two-hybrid system.
  • the latter exploits the modular nature of many transcription factors which are typically composed of a DNA-binding domain (DBD) and an transactivation domain (TAD).
  • DBD DNA-binding domain
  • TAD transactivation domain
  • the original transcription factor is split and two types of hybrids are created: the first one is a DBD-bait fusion, in which the bait represents a protein of interest or a fragment thereof, and the second hybrid " comprises an TAD-prey fusion, in which the prey represents a protein library to be investigated.
  • Hyperactive STAT proteins may be exploited for interaction trap assays such as the yeast two-hybrid system.
  • a probe or "bait" protein fused to a heterologous DNA-binding protein domain reacts with components of a conditionally expressed library comprising fusion proteins with a portable transcriptional activation domain.
  • the function of STAT proteins as constitutive shuttling molecules may facilitate the search for interactions between the bait and the fusion proteins harboring a transactivation domain.
  • any of the Stat mutants could act as bait which is fused to the LexA DBD, for example, whereas the protein library could be merged with the Gal4 TAD domain.
  • Stat mutant-LexA DBD fusion proteins encoded in a first vector and Iibrary-Gal4 TAD fusion proteins encoded in a second vector are transformed into two yeast strains which are cultured under appropriate conditions and over an appropriate period. Subsequently, the yeast strains are mated, grown on plates and examined for LacZ expression.
  • the identified binding partners may elucidate the control of signaling pathways and/or different gene induction levels, for instance by cooperative binding. In addition, they could be useful as markers for pathogenic phenotypes and/or as activators or inhibitors of Stat molecules in order to modify the transcription activity of target genes.
  • the present invention also relates to a method for the expression of a target protein including the addition of the mutated Stat monomer of the invention, their dimers or the fusion protein of the invention to an in-vitro system containing genes under Stat control.
  • the in-vitro system preferably represents a cell and more preferably a cell-free system.
  • the method for the expression of a target protein includes the nucleic acid sequence of the invention preferably inserted into the vector of the invention which is preferably transformed or transfected into the host cell of the invention.
  • the Stat mutants exert an influence on gene activation in a promoter-specific manner. While the transcription yield is strongly enhanced if activated on optimal GAS sites, the opposite phenomenon appears on natural promoter sequences. Thus, either an overexpression or an underexpression of a target protein can be realized compared to the matching gene induction by the Stat wild type.
  • a suitable vector containing a promoter of choice followed by the DNA sequence of the Stat mutant is transformed into an appropriate cell culture.
  • a phosphorylation system could be included for Stat activation.
  • a construct of promoter harboring at least a single optimal GAS sequence and a target gene of choice is preferably inserted into a second vector. High protein amounts can be advantageously obtained in this way.
  • the cell-free system In addition to the cellular expression, the cell-free system is applicable.
  • Cell-free systems involve coupled transcription and translation reactions in-vitro which are based on a cell lysate (generally called S30). The lysate is pre-produced by means of cell disruption and subsequent centrifugation at 30.000 g.
  • the advantages of the cell-free protein biosynthesis include, inter alia, the solely dependence of protein expression on the template, high yields as well as the possibilities to produce cell-toxic proteins and to incorporate unusual amino acids into the protein.
  • Two vectors which are generated to harbor the Stat insert and the target gene as described in the cellular in-vitro section are introduced to the cell-free system. The encoded proteins are expressed and the tyrosine phosphorylation of the Stat mutant is performed with EGF receptor or supplemented Jak.
  • the expression used in the present specification is intended to describe the transcription and translation of the sequence of the gene product.
  • the gene product is a protein
  • the expression involves initial transcription of a DNA chain encoding the sequence of a gene product, to form a complementary RNA, frequently a mRNA, followed by the translation of the mRNA, to form the above- mentioned gene product.
  • expression also comprises transcription of a DNA inserted in antisense direction with respect to its regulatory elements.
  • An expression which proceeds in a constitutive fashion and is possibly further enhanced by an externally controlled promoter fragment - to produce multiple mRNA copies and large amounts of the selected gene product - may also involve overproduction of a gene product.
  • the wild type Stat cDNA sequence preferably cloned into a baculovirus transfer vector is mutated by site-directed mutagenesis at the appropriate residues which is confirmed by DNA sequence analysis.
  • Tyrosine phosphorylation is checked by Western blot analysis using phosphotyrosine-specific antibodies.
  • the present invention also relates to a method for investigating effects caused by different gene induction levels including the addition of the mutated Stat monomer of the invention, their dimers or the fusion protein of the invention to an in-vitro system containing genes under Stat control.
  • effects are understood to be any influence on transcription activity, resulting in an increase or drop in activity, which can be determined by means of biological, chemical or physical measuring methods.
  • the in-vitro system preferably represents a cell and more preferably a cell-free system.
  • the method for investigating effects caused by different gene induction levels includes the nucleic acid sequence of the invention preferably inserted into the vector of the invention which is preferably transformed or transfected into the host cell of the invention.
  • the Stat mutation of the tetramerization interface affects the transcription in a promoter-specific manner.
  • biochemical effects on the living cell will appear. They may concern the entire range of biochemical life processes, like metabolic rate, cell growth, proliferation, aging etc.
  • the up- or down-regulation of genes is tightly controlled. The resulting observations are very helpful to reveal unknown functions of genes.
  • the general experimental procedure is identical to that already described for the method for expression performed in cells. The monitoring of the effects depends on the target gene to be investigated. A couple of methods to detect a gene product is mentioned within the description of the next embodiment concerning the screening of active substances.
  • a “detectable gene product” is a nucleotide sequence or amino acid sequence which can be detected by means of an appropriate test depending on the gene product itself.
  • the expression of a detectable gene product provides the cell with a feature allowing easy selection of that cell among other cells not expressing the detectable gene product.
  • the present invention also relates to a method for the screening of active substances which modify the transcription activity of target genes including the addition of the mutated Stat monomer of the invention, their dimers or the fusion protein of the invention to an in-vitro system containing genes under Stat control.
  • active substances which modify the transcription activity of target genes including the addition of the mutated Stat monomer of the invention, their dimers or the fusion protein of the invention to an in-vitro system containing genes under Stat control.
  • such modifying is understood to be any influence on transcription activity, resulting in an increase or drop in activity, which can be determined by means of biological, chemical or physical measuring methods.
  • the in-vitro system preferably represents a cell and more preferably a. cell-free system.
  • the screening method includes the nucleic acid sequence of the invention preferably inserted into the vector of the invention which is preferably transformed or transfected into the host cell of the invention.
  • a suitable expression vector containing a promoter of choice followed by the DNA sequence of the Stat mutant is transformed into an appropriate cell culture firstly.
  • a target gene which is cell-inherent can be chosen thereby exclusively investigating the effect of an active substance on the interaction between Stat mutant and natural promoters.
  • the construct of promoter and reporter gene of choice is preferably inserted into a second vector choosing a weak natural promoter (e. g. PRRIII or ICAM-1 ) for the screening of transcription activators or a strong synthetic promoter (e. g. the PRRIII derivative M10 or Ly6E) for the opposite selection of transcription inhibitors.
  • a weak natural promoter e. g. PRRIII or ICAM-1
  • a strong synthetic promoter e. g. the PRRIII derivative M10 or Ly6E
  • the treatment of human malignancies requires agents that reduce Stat levels and/or Stat activity, along with the promoter-binding capability.
  • a fusion protein construct can be used to recognize Stat relocations. Those skilled in the art
  • RNA can be detected via DNA by semiquantitative RT-PCR.
  • Cells stable expressing any mutant are cultivated and spread on a microtiter plate containing a substance library.
  • Cells are treated with cytokines, like the ⁇ nterferons IFN ⁇ and IFN ⁇ or interleukins, to induce corresponding responsive genes. They are kept interacting for a certain time before the RNA is extracted and reverse transcribed followed by PCR.
  • the PCR products can be analyzed either directly or by gel electrophoresis and quantified by scoring the intensities of ethidium bromide staining. The results are finally compared to the untreated cell sample thus revealing the mode and direction of changes in mRNA yield.
  • the cell-free system already described can be treated with a library of substances followed by the detection of the target mRNA.
  • the protein can be translated either in cells or in the cell-free system.
  • eukaryotic cells are transfected with the plasmid constructs as described above. After a certain period of adaptation the cells are spread on a microtiter plate containing a library of substances to be screened.
  • the Stat transport to the nucleus is induced by administering the appropriate cytokines leading to nuclear accumulation firstly.
  • changes in the expression level of target genes may occur due to association of library substances with Stat mutants.
  • association - without being limited thereto - relates to any type of interaction between the Stat molecule and the target, particularly covalent or non-covalent binding or association, such as a covalent bond, hydrophobic/hydrophilic interactions, van der Waals forces, ion pairs, ligand-receptor interactions, interactions between epitope and antibody binding site, nucleotide base pairing, and the like.
  • association may also encompass the presence of other molecules such as peptides, proteins or other nucleotide sequences.
  • the changed yield of a reporter gene product as marker for transcription activity is monitored via firefly luminescence in the luciferase assay system.
  • an immunoassay For the detection of cell-inherent gene products, an immunoassay is applicable. ..Immunoassay" denotes a test wherein an antibody is used for specific binding an analyte. The immunoassay is characterized in that specific binding properties of a particular antibody are utilized in the isolation, purposeful testing and/or quantitative determination of the analyte. Those skilled in the art can easily produce antibodies directed against the target protein to be monitored and fragments thereof.
  • ..Antibody denotes a polypeptide essentially encoded by an immunoglobulin gene or fragments thereof specifically binding and recognizing an analyte (antigen).
  • Known immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon, and ' mu genes for the constant region, as well as the innumerable genes for the variable immunoglobulin region.
  • antibodies are present as intact immunoglobulins or as a number of well- characterized fragments produced by cleavage using various peptidases.
  • An antibody binds specifically to a protein, e. g.
  • the antibodies mentioned preferably bind to a specific protein, whereas no significant binding to other proteins present in the sample takes place. Specific binding to a protein under such conditions requires an antibody which has been selected according to its specificity for a particular protein.
  • Various variants of immunoassays can be used to select antibodies having specific immunoreactivity with a particular protein. A description of immunoassay variants and conditions which can be used to determine a specific immunoreactivity is presented in Harlow & Lane, Antibodies; A Laboratory Manual (1988), Cold Spring Harbor Publications, New York.
  • the application of the cell-free system is imaginable.
  • Two vectors which are generated to harbor the Stat insert and the target gene as described in the cellular in-vitro section are introduced to the cell-free system which is subsequently spread on a microtiter plate containing a library of substances.
  • the encoded proteins are expressed and the tyrosine phosphorylation of the
  • Stat mutant is performed with EGF receptor or supplemented Jak. Instead of the simultaneous expression of Stat mutant and target protein in the cell-free system, the addition of a mutated Stat monomer, a dimerized Stat or a fusion protein separately expressed in advance is possible as already described above.
  • the present invention also relates to a method for the production of a drug for the diagnosis and/or treatment of diseases associated with abnormal Stat induction of target genes including the detection of the mutated Stat monomer of the invention, their dimers or the fusion protein of the invention in a cell.
  • the method for the production of a drug includes the nucleic acid sequence of the invention preferably inserted into the vector of the invention which is preferably transformed or transfected into the host cell of the invention.
  • a "disease associated with abnormal Stat induction of target genes” is arthritis, carcinogenesis, nephritis, proteinuria, dermatitis, diabetes/obesity, acute and chronic rejection of allogeneic organ transplants, neuron degeneration, Parkinson's disease, septic shock, endotoxemia, hypersensitivity, uveitis, wound closure, and others.
  • the abnormal Stat induction of target genes also plays an important role in the regulation of the immune system, in inflammation and effector mechanisms of cellular and humoral immunity, septic shock, inflammation of sensory organs, like the eyes, in the regulation of transcription, cell cycles and cell growth, acute respiratory insufficiency, physiological stress, numerous cardiovascular diseases and other disorders.
  • the term “treatment” refers to the prophylactic and/or therapeutic effect of a drug.
  • Dysfunctions of signaling pathways are e. g. reflected by inappropriate activation and accumulation of Stat molecules. Therefore, agents that inhibit Stat functions by a decrease of Stat levels and/or binding specifity to target DNA sequences would represent a promising approach which may have the potential to be more selective and less toxic than current anti-cancer treatments.
  • the phenotype of the Stat mutants this invention is based on corresponds with the mentioned requirements. The interactions of the alternate N-domain interface of the mutants which do not mediate oligomerization and block dephosphorylation are probably accompanied by a three-dimensional structural rearrangement. Hence, the application of high affinity molecules which are able to discriminate between the wild type structure and the mutated one is suggested for diagnosis.
  • RNA aptamers These high affinity molecules involve antibodies already described or RNA aptamers. The latter are identified by an in-vitro selection procedure - the so-called SELEX process (systematic evolution of ligands by exponential enrichment). Since RNA is very susceptible to nucleolytic degradation in biological solutions, aptamers are chemically modified using phosphorothioates, locked nucleic acids or SpiegelmersTM.. Because of their high affinity for a broad spectrum of structural targets, aptamers act very similar to antibodies. Both, antibodies and aptamers carry functional coumpounds which are detectable to determine their binding quantity to the Stat target. As example, the fluorescence labelling is mentioned.
  • the wild type Stat molecule should be replaced by the Stat mutant.
  • This transfer may happen by means of a gene therapy.
  • the most common method involves the introduction of genetic material into a gene shuttle, for instance retro viruses.
  • the replacement of the Stat gene in certain cell types can also be performed in-vitro and the modified cells are subsequently re-injected into the organism.
  • the recombination of the vehicle genome or the cell genome, respectively, with the nucleic acid sequence encoding the Stat mutant follows standard techniques, whereas targeting and the stable incorporation of the new gene represent current research topics.
  • the present invention also relates to a method for the importing and targeting of substances into the nucleus which includes the vector encoding the proteins of the invention.
  • the vector is preferably transformed or transfected into the host 5 cell of the invention.
  • These substances preferably represent peptides or proteins which can be attached to any terminus of the Stat molecule and simultaneously expressed as fusion construct. Firstly, the peptides or proteins should be able to interact specifically with physiologically interesting endogenous compounds of the0 nucleus or exogenous compounds expressed by co-transfection. Secondly, these imported substances have to display a higher target affinity in comparison to the carrier Stat molecule. The latter fact is essential for their preferred successful association to the target. Inhibitors and activators of transcription which bind to DNA or enzymes are suited molecules to be transferred into the5 nucleus.
  • Eukaryotic cells are transfected in-vitro with the vector encoding the Stat mutant in combination with an active peptide structure.
  • the vector encoding the Stat mutant in combination with an active peptide structure.
  • the fusion protein containing a marker structure can be used to pursuit Stat relocations during import and within the nucleus.
  • the transport to the nucleus is induced by administering interferon or interleukin, respectively, leading to nuclear accumulation.
  • the effects of the5 imported substance on the cell metabolism are detected by an appropriate method depending on the gene product.
  • the construct of Stat mutant and active substance may be introduced in-vivo by using a vehicle like a virus. In this case, the procedure is to be o considered as gene therapy.
  • Stat molecule derived from wild type Statl which is mutated in at least one conserved hydrophobic amino acid on the surface of the N-domain,5 preferably in the amino acids in position 77 and/or position 78.
  • the mutation in position 77 may involve a substitution by alanine
  • the mutation in position 78 may involve a substitution by serine.
  • the mutants of the following sequences are termed Statl ⁇ F77A and Statl ⁇ F77A.
  • the mutated amino acid is bold.
  • All statements concerning the methods for several purposes also include the addition or detection of the dimerized Statl mutant, which is mutated as described above, or of a fusion protein including the Statl mutant and at least one marker structure. Furthermore, these methods include the nucleic acid sequence which encodes the Statl mutant or the fusion protein preferably inserted into a vector which is preferably transformed or transfected into the host cell.
  • the present invention also relates to a kit comprising the amino acid sequence or the nucleic acid sequence of the Stat mutants of the invention, their dimers or fusion proteins, and/or synthetic analogues, modifications and pharmacologically active fragments thereof and an information about the using of the kit:
  • kits to be built-up are either based on the genotype or the phenotype of the Stat mutant. They can also include parts of the amino acid sequence or the nucleic acid sequence originated from the Stat mutant, a vector harboring the nucleic acid sequence or a host cell containing the vector. The kits should be preferably applied for any of the methods described above.
  • Example 1 The plasmid expressing full-length human STAT1 (amino acids 1-746) fused to the N-terminus of GFP (pSTAT1-GFP) was described previously (Begirt et al, Cell Biol. 97, 10418-10423 (2000)).
  • Stat expression constructs were generated by cloning the Statl full-length coding region in the SamHI and Not ⁇ sites of the vector pcDNA3 (Invitrogen). Those skilled in the art can easily produce these constructs by well-known recombination and cloning techniques.
  • RT-PCR analysis of endogenous IFN ⁇ - and IFN ⁇ -responsive genes was performed with U3A cells stably expressing Statl wild type or Statl F77A mutant. Cells were grown for 1 ⁇ h in Dulbecco's modified Eagle's medium with reduced serum (1 %), followed by the addition of serum to 10% and treatment with or without IFN ⁇ or IFN ⁇ for 4 h. RNA was extracted and reverse transcribed followed by PCR.
  • IFN ⁇ which signals via a Statl homodimer
  • stimulation of cells with IFN ⁇ induced the additional formation of a ternary complex called ISGF-3, consisting 5 of a Stat1/Stat2 heterodimer and IRF9, which binds to an unrelated recognition site termed interferon-stimulated response element (ISRE).
  • ISGF-3 a ternary complex
  • IRF9 an unrelated recognition site termed interferon-stimulated response element
  • Example 2 The plasmids pCIStat ⁇ a and pCIStat ⁇ b were described (John et al, Mol. Cell.5 Biol. 19, 1910-1918 (1999)). Site specific mutagenesis of these plasmids according to manufactures' instructions (QuikChangeTM, Stratagene; MORPH site-specific plasmid mutagenesis kit, ⁇ '-3' Inc.) generated the respective N- domain mutants Stat ⁇ aF81A and Stat ⁇ bF81A.
  • 293T cells were co-transfected with the 0 following DNAs: 2 ⁇ g IL-2 receptor ⁇ , O. ⁇ ⁇ g ⁇ c , 0.2 ⁇ ⁇ g Jak3, 10 ng of each of the wild type Stat ⁇ (control), Stat ⁇ aF81A or Stat5bF81A mutant expression plasmids and 1 ⁇ g of PRRIII-E1b or M10-E1b-luciferase reporter construct as described (John et al, Mol. Cell. Biol. 19, 1910-1918 (1999)). Either the weak natural promoter PRRIII (Soldani et al, Mol. Cell. Biol.
  • Unphosphorylated Stats preferably Statl
  • NPC nuclear pore complex
  • Mr > 87 kD the signal-independent nuclear import of unphosphorylated Stats
  • Stats derived form any Stat wild type or fragments thereof shuttle between cytosol and nucleoplasm, preferably Statl , Stat3 and Stat ⁇ , more preferably Statl and most preferably Statltc (representing a truncated Statl protein lacking the NH 2 - and the COOH-terminal domain).
  • the direct binding of Stats to nucleoporins indicated that unphosphorylated Stats migrate into the nucleus via specific molecular interactions with components of the NPC.
  • these components of the NPC comprise nucleoporins and fragments thereof, preferably Nup1 ⁇ 3 and Nup214.
  • the invention also relates to a Stat mutant which could preclude the aforementioned nuclear import.
  • a Stat molecule derived form any Stat wild type or fragments thereof is mutated, preferably Statl , Stat3 and Stat ⁇ , more preferably Statl and most preferably
  • At least one amino acid is mutated within the linker domain of the Stat molecule, preferably cysteines, more preferably cysteine in position 643 of Statl or in the equivalent position of homologous proteins.
  • Possible mutations comprise the deletion, insertion, substitution and/or modification of at least one amino acid, preferably a single residue. Possible modifications are alkylation, arylation and acetylation, preferably the alkylation.
  • the amino acid Cys ⁇ 43 in Statl or in the equivalent position of homologous proteins is alkylated.
  • the phenotype of shuttle loss can be effectively used as negative control in bioassays requiring shuttling.
  • the phenotype represents a test within gene induction assays, like the two-hybrid system.
  • mouse Stat3 (aa 136-716), or sheep Stat ⁇ (aa 138-704) were amplified by PCR and cloned into the EcoRI and BamHl sites of pASKlBA3 (IBA, G ⁇ ttingen).
  • the cDNA encoding aa 4 ⁇ -462 of p97 was PCR-amplified and cloned into pGEX-5X-2 (Pharmacia).
  • MBP fusion proteins of the FG repeat regions of human Nup62 (aa 1-308, a kind gift of E. Hurt) and human Nup153 (aa 333-618, a kind gift of B. K.
  • Felber were prepared after PCR-amplification of the respective cDNAs and cloned into the BamHI site of pMal-2CX (NEB). PCR reactions were performed with Vent-proofreading polymerase (NEB). Site- directed mutagenesis was performed with the Quick-change kit (Stratagene). Baculovirus transfer vectors (pFastBac) encoding human Statl wt (aa 1-746) or the Tyr701Phe mutant and vectors for bacterial expression of GST-NLS-GFP and GST-NES-GFP have been described. Expression constructs for His-tagged human p97 (aa 1-878) and human Nup214 (aa 1549-2090) were kind gifts of U. Kutay and M.
  • Protein expression was at 18° C for 15 h (Strep-tagged proteins), or at 30° C for 5 h or 3 h with GST- or MBP-tagged proteins, respectively.
  • Cells were lysed by sonication in PBS containing 1% Triton X-100, 2 mM EDTA, 2 mM EGTA, ⁇ mM DTT.
  • Proteins were single-step purified by virtue of their Strep- or GST-tag, respectively, as recommended by the manufacturers (IBA; Pharmacia). Where indicated, purified proteins were phosphorylated alkylated with NEM (20 mM) as described.
  • Proteins were concentrated by ultrafiltration (Centriprep, Millipore) in injection buffer containing 5 mM DTT; protein concentrations were determined by UV spectroscopy (extinction coefficient 1.25 for Statlwt, 1.27 for Statltc, 1.24 for Stat3tc, and 1 .43 for Stat ⁇ tc), or by Coomassie dye-binding (Biorad) with a BSA standard. Proteins were quick-frozen on dry ice and stored at -80° C. Fluorescence labeling of Statlwt and Statltc was done with succinimidyl esters of Alexa Fluor 594 or Oregon Green 488 as described by the manufacturer (Molecular Probes) at a molar protein:dye ratio of 1 :5. Efficiency of labeling was close to 100% as judged by mobility shift in SDS-PAGE ( Figure 1). His-tagged p97 was expressed and purified as described.
  • Microinjections of antibodies (at 0.1 ⁇ mg/ml in PBS) and concentrated recombinant proteins (at 0. ⁇ -1 mg/ml) were as described. Injection site markers were used at a concentration of 0. ⁇ -1 mg/ml, WGA (Sigma) was co- microinjected at a concentration of O. ⁇ mg/ml. Immunocytochemistry was with monoclonal Statl antibody C-136 (0.2 ⁇ g/ml; Santa Cruz) or anti-Strep-tag II rabbit polyclonal antibody (0.2 ⁇ g/ml; IBA).
  • HeLa-S3 cells were grown on coverslips and permeabilized with 40 ⁇ g/ml d ⁇ gitonin (Roche) in transport buffer (TB, 20 mM Hepes, pH 7.3, 110 mM KOAC, 2 mM Mg(OAC) 2l 1 mM EGTA, 2 mM DTT, Complete protease inhibitors (Roche)) for 6 min on ice. Subsequently, the cells were washed in TB twice. Incubation with 20 ⁇ l import mix was performed at RT for 60 min or as indicated.
  • transport buffer TB, 20 mM Hepes, pH 7.3, 110 mM KOAC, 2 mM Mg(OAC) 2l 1 mM EGTA, 2 mM DTT, Complete protease inhibitors (Roche)
  • the cytosol-free import mix contained TB supplemented with 10 mg/ml BSA and one or several of the following purified proteins as indicated: 1 ⁇ M Statltc, 1 ⁇ M Stat3tc, 1 ⁇ M Stat ⁇ tc, 1 ⁇ M Oregon Green 488- or Alexa Fluor 594-labeled Statltc, 15 ⁇ M Statlwt, 0.8 ⁇ M GST-NLS-GFP, or 0.8 ⁇ M full- length His-tagged p97.
  • the complete import mix contained 75% (v/v) rabbit reticulocyte lysate (Promega), 25% IM, and an energy regenerating system (0,5 mM ATP, 0,5 mM GTP, 10 mM creatine phosphate, 30 U/ml creatine phosphokinase).
  • CIM complete import mix
  • digitonin-permeabilized cells were incubated for 10 min on ice in IM containing 250 ⁇ g/ml WGA or as indicated, before washing and addition of the import mix.
  • IM or CIM were preincubated for 15 min on ice in the presence of 0.8 U/ml apyrase (Sigma) before addition to the permeabilized cells.
  • In vitro binding assay was perfomed as follows: Bacterial lysates (containing approx. 6 ⁇ g NPC proteins or MBP) and 6 ⁇ g purified His-tagged control protein (tandem SH3 domains from human FYB; a kind gift of C. Freund) were resolved by 10% SDS-PAGE and transferred to a nitrocellulose membrane. The membrane was rinsed in transport buffer (20 mM HEPES, pH 7.4, 110 mM KOAc, 2 mM Mg(OAc) 2) 0.5 mM EGTA, 2 mM DTT, 0.5% Tween-20, Complete protease inhibitors), and then blocked in this buffer plus 5% (w/v) non-fat milk for 1 h at 4°C.
  • transport buffer (20 mM HEPES, pH 7.4, 110 mM KOAc, 2 mM Mg(OAc) 2 0.5 mM EGTA, 2 mM DTT, 0.5% Tween-20, Complete protease inhibitors
  • Statl and Statltc were labeled on lysine residues with fluorescent dyes (either Oregon Green 488 or Alexa Fluor 594) to directly observe their nuclear translocation. As is shown in Figure 21 for Oregon Green 488-labeled Statltc, conjugation with fluorescent dyes did not interfere with nuclear import (identical result were obtained with Alexa Fluor 594 and for full length Statl).
  • WGA wheat germ agglutinin
  • fusion proteins of glutathione S-transferase (GST) and green fluorescent protein (GFP) that included at their domain junction either a triple SV40 NLS or a Statl -derived NES (termed GST-NLS- GFP or GST-NES-GFP, respectively) were co-microinjected with recombinant Statl.
  • GST glutathione S-transferase
  • GFP green fluorescent protein
  • the effective nucleocytoplasmic translocation of the reporter constructs was confirmed in cells growing in full-medium, whereas pre-exposure of cells to azide and 2-deoxyglucose for 2 h prevented the carrier-dependent nuclear translocation of the reporter protein ( Figures 4 and ⁇ E, F).
  • Endogenous Statl is constitutively shuttling across the nuclear envelope independently of metabolic energy.
  • a different experimental approach was used to investigate the nucleocytoplasmic flux rates also of endogenous Statl .
  • To this end we performed microinjections of Statl antibodies, which has been shown to immobilize the shuttling target antigen and hence cause its accumulation in the microinjected compartment. The results are assembled in Figure 5 and Table I. As is shown in panel A of Figure 5, maximal nuclear accumulation of unphosphorylated Statl was seen already 15 min after antibody microinjection into the nucleus. Next, cytoplasmic antibody microinjections were used to estimate the nuclear export rate of the endogenous Statl protein.
  • Statl, Stat3, and Stat ⁇ have the ability to migrate into the nucleus by themselves in the absence of metabolic energy or added transport factors. They share binding partners in the NPC among each other and with the import receptor p97.
  • Statl binds to nucleoporin Nup1 ⁇ 3 and Nup214, but not Nup62.
  • the above data are not compatible with a carrier-mediated nuclear translocation process. Rather, they are characteristic for a carrier-free mechanism that occurs through direct interactions between the Stats and constituents of the nuclear pore.
  • a distinctive feature of numerous NPC proteins is the presence of Phenylalanine/Glycine (FG)-rich repeat motifs, which provide interaction sites for transport factors. Since Stats and p97 appeared to share binding sites on nuclear pore proteins, we examined whether Statl could also interact with constituents of the nuclear pore.
  • FG Phenylalanine/Glycine
  • FG repeat-containing parts of His-tagged Nup214 (residues 1649-2090), Nup62 (residues 1-308), and Nup153 (residues 333-618; the latter two were fusion proteins with maltose-binding-protein, MBP) were expressed in bacteria and detected by immunoblotting with the monoclonal antibody 414 (Supplemental Figure 2).
  • the bacterial lysates were resolved by denaturing SDS-PAGE ( Figure 7, lower panel) and subsequently blotted on nitrocellulose. After extensive incubation in renaturing buffer, the immobilized proteins were incubated with wild type Statl, the binding of which was detected by Western blotting ( Figure 7, upper panel).
  • the linker domain of Statl plays a role in cytokine-independent nuclear translocation.
  • Statl contains highly reactive cysteine residues which can cause aggregation of recombinant purified protein. This problem can be overcome by blocking reactive SH-groups with N-ethylmaleimide (NEM), which has been shown to leave in vitro DNA binding undisturbed, and this treatment also did not influence nuclear import of full length Statl .
  • NEM N-ethylmaleimide
  • Carrier-dependent and -independent pathways cooperatively determine the subcellular distribution of Statl in resting cells. It is currently unknown how the Stats achieve their predominantly cytoplasmic localization in unstimulated cells. As is shown above, before stimulation with cytokines Statl enters the nucleus independent of metabolic energy and transport factors. Export of Statl from the nucleus, however, can occur via two pathways, only one of which functions without metabolic energy, whereas the second pathway requires an NES, the exportin CRM1 and metabolic energy. Therefore we used energy-depletion and employed the specific CRM1 inhibitor LMB to suppress the NES-dependent active nuclear export of Statl .
  • FIG. 1 Recombinant Stat proteins.
  • A SDS-PAGE (7%) analysis of the Stat protein preparations used in this work (1 ⁇ g of each). Molecular size markers are indicated to the left. OG488, Oregon Green 488.
  • B Schematic representation of the respective Stat proteins. The truncated mutants were expressed with a C-terminal Strep-tag of the sequence SAWSHPQFEK (single letter code). The Stat proteins were of human (Statl), mouse (Stat3), or sheep (Stat ⁇ ) origin. N, N domain; CC, coiled coil domain; DNA, DNA binding domain; L, linker domain; SH2, SH2 domain; TAD, transactivation domain.
  • FIG. 1 Unphosphorylated recombinant Stat proteins translocate into the nucleus of unstimulated cells.
  • HeLa-S3 cells except for panels B and C were used. The injection site is indicated by the location of co-microinjected FITC- labeled or Tetramethyl-rhodamine (TMR)-labeled BSA. Nuclei were stained with Hoechst dye. Arrows indicate injected cells.
  • A-C Cytoplasmic microinjection of purified Statlwt into Hela cells (A), COS7 cells (B), or 2fTGH cells (C). After 15 min the cells were fixed and Statl was detected by immunocytochemistry with a specific antibody.
  • FIG. 3 Unphosphorylated recombinant Stat proteins exit the nucleus of unstimulated cells. The injection sites of the HeLa cells are indicated by co- microinjected FITC-labeled BSA.
  • A, B Recombinant Statlwt was injected into the nucleus. After 1 h (A) or 3 h (B) immunocytochemistry with a Statl -specific antibody was performed on fixed cells.
  • C, D Microinjection of Statlwt into cells pretreated for 1 h with LMB. After nuclear (filled arrow) or cytoplasmic (open arrow) microinjection the cells were further incubated with LMB for 1 h (C) or 4 h (D) before fixation and immunocytochemistry with a Statl -specific antibody.
  • FIG. 4 The nucleocytoplasmic translocation of recombinant Stat proteins continues in energy-depleted cells. HeLa cells were kept in energy depletion medium for 2 h before microinjection (EDM).
  • EDM microinjection
  • A-E Cytoplasmic microinjection of tyrosine-phosphorylated Statltc (A), or unphosphorylated Statltc (B), Statlwt (C), Stat3tc (D), or Stat ⁇ tc (E). Injection site and energy depletion are indicated by the location of co-microinjected GST-NLS-GFP. Tine cells were fixed at the indicated times after injection and Stats were detected by immunocytochemistry (see Figure 2).
  • F Nuclear co-microinjection of recombinant Statlwt and GST- NES-GFP, followed by 3 h of incubation in EDM. Then , cells were processed for immunocytochemistry as before. Bar, 20 ⁇ m.
  • FIG. ⁇ Nucleocytoplasmic translocation of endogenous Statl in normal and energy-depleted HeLa cells.
  • Statl antibodies were microinjected followed by immunocytochemical detection of Statl .
  • A Microinjaction of Statl antibodies into the cell nucleus with fixation after 15 min. The injection site is marked by co-microinjected FITC-BSA.
  • B Identical with (A), except that the antibodies were microinjected into the cytoplasm.
  • C Identical wit-h (B) except that the cells were incubated with 5 ng/ml LMB starting 1 h before microinjection.
  • D Identical with (B), except that the cells were kept at 4°C for 60 min both before and after cytoplasmic microinjection.
  • FIG. 6 The nuclear import of Stat proteins continues in cytosol-depleted digitonin-permeabilized cells.
  • HeLa-S3 cells were treated with 40 ⁇ g/ml digitonin, washed, and then incubated with 20 ⁇ l of import mix (IM; protein-free transport buffer supplemented with 10 mg/ml BSA) at RT unless noted otherwise. The nuclei are stained with Hoechst dye. Unlabeled Stat proteins were detected immunocytochemically in fixed cells.
  • IM protein-free transport buffer supplemented with 10 mg/ml BSA
  • Unlabeled Stat proteins were detected immunocytochemically in fixed cells.
  • Permeabilized HeLa cells were incubated for 60 min with 1 ⁇ M unphosphorylated Statltc in IM. Additionally, the import reaction was performed on ice (4°C), or after preincubation with 250 ⁇ g/ml WGA (+WGA).
  • FIG. 7 Statl interacts with FG repeat-containing nucleoporins.
  • (Upper panel) Bacterial lysates containing the indicated fusion proteins (lanes 1 -4) or purified His-tagged tandem SH3 domains from FYB (lane 5) were resolved by SDS- PAGE, transferred to a nitrocellulose membrane, and probed with wild type unphosphorylated Statl The association of Statl with the immobilized proteins was detected by immunoblotting using a Statl specific antibody. The lower panel shows the respective Coomassie-blue stained SDS-gel before transfer to a nitrocellulose membrane (input).
  • FIG. 8 Alkylation of cysteine 643 in the linker domain precludes nuclear import of Statl
  • A-C Purified Statltc or the indicated mutants were alkylated by NEM in vitro and injected in the cytosol of HeLa cells. After 1 h the cells were fixed and the intracellular distribution of the microinjected proteins was determined by immunocytochemistry. Bar, 20 ⁇ m.
  • D Summary of the microinjection data and mass spectrometric analyses of alkylated Statl . Purified bacterially expressed Statltc was treated without or with NEM and subsequently injected into the cytoplasm of HeLa cells. The ability to enter the nucleus during an 1 h incubation period is stated.
  • cysteine residues in position 155, 174, 247, 25 ⁇ , 324, 440, 492, 543, 677 of human Statl are numbered 1 to 9 in the diagram to the left.
  • Mutated cysteines (Cys to Ala) are blackened. Open circles denote absence of alkylation, filled circles denote alkylated cysteines as determined by mass spectrometry. For position 7 both alkylated and non-alkylated peptides were found. Peptides covering Cys- positions 3, 4, and 6 were not detected
  • FIG. 9 The inhibition of active nuclear export prevents cytoplasmic accumulation of Statl .
  • 3T3 cells were left untreated (DMEM) or treated with energy depletion medium for 2 h (EDM) followed by growth medium for 5 h (EDM+DMEM). Alternatively, cells were treated with the CRM1 inhibitor leptomycin B (LMB) for 2 h or 5 h as indicated. Shown is the distribution of the endogenous Statl as revealed by immunocytochemistry with a specific antibody and conventional microscopy.
  • B Corresponding bar diagram with a quantitative analysis of the ratio of the mean cytoplasmic and nuclear immunofluorescence densities. The median slice (x/y image) of a confocal microscopical image was used to quantify the signals in 13 randomly chosen cells. Statistically significant differences between cells incubating in control (DMEM) or experimental medium are indicated by (*). Bar, 20 nm.
  • FIG. 10 A model of Statl nucleocytoplasmic shuttling.
  • Unphosphorylated "latent” Statl constitutively shuttles between cytosol and nucleoplasm via direct interactions with the nucleoporins Nup153 and Nup214. The surface of the linker domain of Statl is likely to provide the contact surface.
  • B Additionally, NES-mediated transport of unphosphorylated Statl via CRM1 enhances the export rate and achieves cytoplasmic accumulation.
  • C After cytokine-induced receptor activation, Statl is tyrosine-phosphorylated and dimerizes, which precludes further carrier-free nucleocytoplasmic cycling.
  • IL-2-reconstituted 293T cells are co-transfected with Stat ⁇ wild type or mutant Stat ⁇ aF81A or Stat5bF81A and a luciferase reporter gene. Stimulation without (gray) or with IL-2 is overnight (black). Luciferase expression is activated from a natural PRRIII promoter or an M10 element. The diagrams represent the ground expression level which is not affected by the substance library.
  • the cells were incubated for 60 min at RT with GST-NLS-GFP control protein (0.8 ⁇ M) dissolved in 20 ⁇ l IM or complete import mix (CIM: IM supplemented with reticulocyte lysate (76% of volume) and an energy regeneration system (O. ⁇ mM ATP, O. ⁇ mM GTP, 10 mM creatine phosphate, 30 U/ml creatine phosphokinase). Where indicated, the reaction (with CIM) was performed at 4°C (4°C).
  • CIM was supplemented with O. ⁇ mg/ml WGA (+WGA); or with 16 ⁇ M p97 (aa 45-462 of p97) (+p97tc), or with 0.8 U/ml apyrase (+apyrase).
  • B the cytosol-independent nuclear import of p97 is demonstrated. Import reaction without or with 0.8 ⁇ M His-tagged importin- ⁇ (p97) dissolved in IM. Where indicated, IM was supplemented with WGA at a concentration of 0.1 mg/ml (+WGA/100), or 0.25 mg/ml (+WGA/2 ⁇ O). An anti- His antibody was used to detect p97. Bar, 30 ⁇ m.
  • Protein loading was 0.2% of the input used in Figure 7.
  • Table I Nucleocytoplasmic translocation of unphosphorylated Stat proteins as determined by microinjection. a) Microinjection of recombinant Stat proteins. Listed is the amount of time required to reach equilibrium (min or h). b) Microinjection of Statl antibodies. Listed are the times required to observe maximal precipitation of endogenous Statl in the injected compartment. Each entry represents an averaged value observed by immunocytochemistry in 15-20 microinjected cells after fixation.
  • GM growth medium with 10% serum
  • EDM serum- and glucose-free energy depletion medium with azide and deoxyglucose
  • n.a. not applicable
  • n.d. not done
  • AF Alexa Fluor 594
  • OG Oregon Green 488.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The invention relates to Stat molecules which are mutated and their use in the activation of gene expression, in the screening of inhibitors or activators of transcription factors, the identification of binding proteins, the production of a drug for diagnosis and/or treatment of diseases associated with the activation of gene expression as well as in the targeting of active substances. The Stat molecules occurring in the nucleus of a cutaneous T cell lymphoma cell line are characterized in that at least one conserved hydrophobic amino acid on the surface of the N-domain of the Stat wild type is mutated.

Description

Hyperactive Stat molecules and their use in assays employing gene activation
The invention relates to Stat molecules which are mutated and their use in the activation of gene expression, in the screening of inhibitors or activators of transcription factors, the identification of binding proteins, the production of a drug for diagnosis and/or treatment of diseases associated with the activation of gene expression as well as in the targeting of active substances.
In eukaryotic organisms, DNA replication and RNA biogenesis take place in the cell nucleus, whereas protein synthesis essentially proceeds in the cytoplasm. The close integration of these activities depends on a selective transport of the proteins and ribonucleoprotein particles between nucleus and cytoplasm requiring a coupled import and export of compounds at many molecular processes. This translocation of proteins, DNA and/or RNA proceeds via nuclear pores which are located in the membrane enclosing the cell nucleus. Since the translocation is mediated, inter alia, via distinct signals, the process is termed signal transduction.
For example, the Stat (signal transducers and activators of transcription) proteins constitute an ancient and evolutionary conserved family of transcription factors involved in the regulated expression of numerous target genes in response to cytokines and growth factors (Darnell, J . E., Jr., Science 277, 1630- 1635 (1997)). Polypeptide growth factors and cytokines elicit their effects by activating specific cell-surface receptors, thereby initiating signaling cascades. Canonical signaling through the Janus tyrosine kinase (Jak) / Stat pathway begins at the cell membrane within the cytoplasm. The binding of the mentioned ligands to their cognate receptors initiates a number of tyrosine phosphorylations catalyzed by Jak. The bound Stat monomers detach from their receptor docking sites after phosphorylation of a single tyrosine residue at their C-terminus finally forming reciprocal homo- or heterodimers. This sequence of events is commonly referred to as "Stat activation" triggering the accumulation of Stat dimers in the nucleus within a certain period. Eventually, they induce transcription by binding to palindromic DNA recognition sites named γ-activated sequence (GAS).
The mechanisms that control the predominantly cytoplasmic localization in unstimulated cells have not been resolved.
The translocation of protein substrates across the nuclear envelope commonly involves transport factors that share homology with the transport factor p97 (also called importin-β). These proteins mediate passage of protein cargoes through the nuclear pore complex (NPC). Based on the direction of cargo transport they have been classified as importins or exportins. The transport process requires metabolic energy and it is propelled by a concentration gradient across the nuclear membrane of the GTP-bound form of the G-protein Ran, which is found predominantly in the nucleus. The transport substrates are distinguished in their amino acid sequence by the presence of cis-acting nuclear localization signals (NLS) and/or nuclear export signals (NES). Recently, such transport signals were also identified in the Stats. These proteins contain canonical leucine-rich export signals which are required for binding to the exportin CRM1. Since pharmacological or mutational inactivation of the NES/CRM1 pathway does not preclude nuclear export, further transport mechanisms for the removal of Statl from the nucleus remain to be characterized. In addition, the DNA binding domain of the Statl molecule harbors a dimer-specific NLS (dsNLS) that constitutes the binding surface for the adaptor protein NPI-1 of the importin-α family, which tethers the Statl protein to the nuclear import factor p97 in a Ran-dependent manner. Destruction of the dsNLS precludes nuclear translocation of phosphorylated Statl and thus results in the loss of cytokine-inducible transcriptional responses.
It was shown that Stat proteins can leave the nucleus only after tyrosine dephosphorylation, and DNA binding was identified as the critical regulator of the conversion (Meyer et al, Genes Dev. 17, 1992-2005 (2003)). However, at present the physiological interfaces that modulate cooperative DNA binding and tyrosine dephosphorylation of Stat proteins are poorly defined. Packing in the Stat4 crystal suggested two interfaces (I and II) that are potentially relevant for dimer formation. Previous biochemical investigations concerning mutations of an invariant tryptophane residue in position 37 of the N-domain led to the loss of cooperative Stat binding to tandem sites on DNA. Consequently, the crystal structure was interpreted in terms of the putative dimer interface I including the amino acid tryptophane.
Due to further experimental studies, the alternate surface - termed interface II - was proposed to mediate Stat dimerization as requirement for the formation of higher order Stat complexes on DNA (Chen et al, Protein Science 12, 361-365 (2003)). These studies analyzed point mutations which were introduced in the Statl N-domain at several sites of each interface. The dimerization properties of these mutated proteins were determined by analytical ultracentrifugation and gel filtration. Among others, the mutant Statl F77A characterized by a phenylalanine-to-alanine substitution on interface II was investigated and shown to be a monomer. Beside the fact that interface II is more extensive than interface I, it also involves interactions between hydrophobic residues probably affected by the replacement of phenylalanine.
Referring to the mentioned tryptohane mutant, the new results reflect a nonspecific domain destabilization rather than a specific defect in dimerization.
In particular, Stat proteins regulate many of the genes controlling cellular functions like cellular growth, differentiation and survival. Given the importance of Stats in the control of normal physiological processes, it is self-evident that inappropriate activation of these proteins is found in various disease patterns. Usually, cytokine-induced transcription is a transitory process which can take from a few minutes to hours. Cells with a deviantly high and prolonged Jak or Stat activation are susceptible to transformations and associated with abnormal developments (Frank, D. A., Mol. Med. 5, 432-456 (1999)). Interferences in the signaling pathways are the crucial factor for the appearance of pathogenic phenotypes such as human malignancies, leukemia, disorders in cell growth, defects of the immune system or in the metabolic performance of liver, kidneys and mammary gland, septic shock, arthritis, numerous cardiovascular diseases and other things.
The lack of more detailed information on the pathogenic mechanisms of signal transduction is particularly disadvantageous since current therapeutic strategies, for example to treat cancer, are both less selective and fairly toxic. Therefore, the technical problem forming the basis of the present invention is to provide agents which modify the transcription activity of target genes and to provide agents and methods for the screening and targeting of active substances which modify the transcription activity, so that they are suitable in the diagnosis and/or treatment of diseases associated with the abnormal Stat induction of target genes. The present invention solves this problem by providing a Stat molecule which is characterized in that at least one conserved hydrophobic amino acid on the surface of the N-domain of the Stat wild type is mutated.
The formation of Stat tetramers and higher order oligomers on DNA results from cooperative DNA binding of Stat dimers that appears to be conserved throughout the Stat family. Generally, Stat proteins consist of a large core domain including the Src homology domain 2 (SH2) responsible for dimer formation and the DNA binding module which denotes an amino acid sequence capable of binding to a specific DNA sequence. The core is connected through a proteolytically cleavable peptide to the smaller N-domain which engages in a multitude of protein-protein interactions. This N-domain mediates cooperative DNA binding of Stat dimers. Several Stat molecules (Statl , Stat3, Stat4, Statδ,
Stat6) are able to form higher order complexes (dimer-dimer or higher) on promoters containing two or more neighboring Stat binding sites. Likely, Stat tetramers are also mandatory for full transcription activity at many promoters that contain only a single (not optimal) canonical GAS site.
The term "promoter" refers to a DNA sequence, mostly situated upstream (5') of the coding sequence of a structural gene which controls the expression of the coding region by providing a recognition sequence for an RNA polymerase and/or other factors required to start transcription at the appropriate position. Promoter sequences are necessary, but not sufficient in any case, to control expression of the gene.
The mutation of a conserved residue located in the alternate surface does not compromise protein stability, but causes a severe decrease in tetramer formation of several Stat molecules without overtly affecting the DNA binding of the dimer.
Importingly, the novel penotype of the mutants makes insights possible into the role that DNA binding plays in nuclear accumulation of Stat proteins. It was previously hypothesized that the access to a nuclear export signal (NES) within the coiled-coil domain is regulated by DNA binding. According to this model, the Stat association to DNA would mask the NES and thus preclude nuclear export. Consequently, the loss of DNA binding would result in the loss of nuclear accumulation. The novel phenotype of the mutants actually reveals that it is dimerization only (and not the subsequent DNA binding) that masks the region required for the nuclear export of Stat proteins. Thus, DNA binding is not necessary, but sufficient for nuclear retention of Stat molecules.
Most striking is the complete loss of nucleocytoplasmic shuttling during nuclear accumulation proposing that the mutants bear a dephosphorylation defect in- vivo. Tyrosine dephosphorylation is localized within the N-domain representing the crucial step in the control of nuclear export. Both, the dephosphorylation defect as well as the tetramerization deficiency affect the nuclear accumulation of the mutants concluding that the interfaces for dimer-phosphatase and dimer- dimer interactions are overlapping.
In summary, the mutated Stats are still functional transcription factors that retain cytokine responsiveness. The interface centered around the critical hydrophobic residue, mainly represented by phenylalanine, fulfills distinct functions that are subjected to control by the tyrosine phosphorylation and DNA binding status of the Stat molecules. Due to the mutation, the gene induction depends on promoter strength whereby either an increase or a decrease of the transcription activity could be recognized.
In another embodiment of the invention, the wild type of Stat2, Stat3, Statδ or Stat6 is mutated. The Stat molecule is mutated in at least one conserved hydrophobic amino acid on the surface of the N-domain and characterized in that the Stat molecule is derived from the wild type of Stat2, Stat3, Statδ or State. The Stat family of transcription factors comprise seven mammalian proteins with known homologues in all multicellular animal phyla. Stat proteins are activated by a wide range of different cytokines such as interleukins and prolactin, as well as growth factors. Accordingly, gene ablation experiments uncovered mammary gland defects, growth abnormalities and an absent interleukin response underlining the significance of regulating the Stat activity as tool for gene induction. In particular, disturbances in the signaling pathways were figured out as cancer inducing parameter.
In another preferred embodiment of the invention, the amino acids of the N- domain in position 81 and/or position 82 of Statδ or in equivalent positions of the homologous proteins are mutated. Statδa and Statδb are two highly related proteins with a sequence identity of -96%. Possible mutations include the substitution, deletion or modification of the amino acid. They may occur in position 81 and/or position 82 of Statδ or in equivalent positions of homologous proteins. Homologous proteins are members of the aforementioned Stat family, preferably Stat2, Stat3, Statδ or Stat6. Although they display a great sequence similarity, minor shifts of distinct residues within the primary structure are present. With regard to higher structure levels (secondary and/or tertiary structure), the shifted positions are referred to as equivalent. For example, the positions 77 and 78 in Stat 3 and Stat6 as well as the positions 81 and 82 in Stat2 and Statδ are considered as equivalent. In consequence of their mutation, the altered residues in position 81 and/or position 82 of Statδ or equivalent in homologous proteins cannot maintain N-domain interactions in the full-length Stat molecule resulting in the linked phenomena of tetramerization- and dephosphorylation-deficiency, and their consequences on gene induction in a promoter-specific manner.
In another preferred embodiment of the invention, the amino acid in position 81 of Statδ or in the equivalent position of the homologous proteins is substituted by alanine. Due to its neutral properties originated from the short side chain of a single methyl group, alanine is especially suited for mutagenesis studies. Within the Stat family, different residues in an equivalent position are present. These differences are preferably of equivalent matter by changing a hydrophobic amino acid by another hydrophobic one, for example. The hydrophobic residues in the equivalent positions 77 and 81 comprise the following amino acids within those Stat molecules preferred in the meaning of the invention: leucine in position 81 of Stat2, valine in position 77 of Stat3, phenylalanine in position 81 of Statδ and isoleucine in position 77 of Stat6.
The amino acid sequence of the mutants termed StatδaF81A or StatδbF81A, respectively, are given below:
Human Stat5aF81A (1-710): magwiqaqql qgdalrq qv lygqhfpiev rhylaq ies qpwdaidldn pqdraqatql leglvqelq kaehqvgedg allkiklghy atqlqktydr cplelvrcir hilyneqrlv reanncsspa gilvdamsqk hlqinqtfee lr vtqdten elkklqqtqe yfiiqyqesl riqaqfaqla qlspqerlsr etalqqkqvs lea lqreaq tlqqyrvela ek qktlqll rkqqtiildd eliq krrqq lagnggppeg sldvlqswce klaeii qnr qqirraehlc qqlpipgpve emlaevnati tdiisalvts tfiiekqppq vlktqtkfaa tvrllvggkl nvhrnnppqvk atiiseqqak sllknentrn ecsgeilnnc cvmeyhqatg tlsa frnms Ikrikradrr gaesvteekf tvlfesqfsv gsnelvfqvk tlslpwviv gsqdlinata tvlwdnafae pgrvpfavpd kvlv qlcea lnmkfkaevq snrgltkenl vflaqklfnn ssshledysg lsvswsqfnr enlpgwnytf wqwfdgvmev lkkhhkphwn dgailgfvnk qqahdllink pdgtfllrfs dseiggitia kfdspernl wnlkpfttrd fsirsladrl gdlsyliyvf pdrpkdevfs kyytpvlaka vdgyvkpqik
Human Stat5bF81A (1-715): mavwiqaqql qgealhqmqa lygqhfpiev rhylsqwies qa dsvdldn pqenikatql leglvqelqk kaehqvgedg allkiklghy atqlqntydr cpmelvrcir hilyneqrlv reanngsspa gsladamsqk hlqinqtfee lrlvtqdten elkklqqtqe yfiiqyqesl riqaqfgpla qlspqerlsr etalqqkqvs leawlqreaq tlqqyrvela ekhqktlqll rkqqtiildd eliqwkrrqq lagnggppeg sldvlqswce klaeiiwqnr qqirraehlc qqlpipgpve emlaevnati tdiisalvts tfiiekqppq vlktqtkfaa tvrllvggkl nvhrnnppqvk atiiseqqak sllknentrn dysgeilnnc cvmeyhqatg tlsahfrnms lkrikrsdrr gaesvteekf tilfesqfsv ggnelvfqvk tlslpwviv hgsqdnnata tvlwdnafae pgrvpfavpd kvlwpqlcea Inmkfkaevq snrgltkenl vflaqklfnn ssshledysg lsvswsqfnr enlpgrnytf wqwfdgvmev lkkhlkphwn dgailgfvnk qqahdllink pdgtfllrfs dseiggitia wkfdsqer f wnlmpfttrd fsirsladrl gdlnyliyvf pdrpkdevys kyytpvpces atakavdgyv kpqikqwpe fvnas
This invention is based on a mutation which is bold.
In another preferred embodiment of the invention, the amino acid in position 82 of Statδ or in the equivalent position of homologous proteins is substituted by serine. Preferably, the amino acid in position 82 of Statδ or in the equivalent position of homologous proteins is leucine.
The mutants StatδaF81A and StatδbF81A of this invention display a couple of novel features which are contrary to the wild type phenotype.
The mutants of this invention are tetramerization-deficient causing a promoter- dependent decrease in the strength of DNA binding. Their ability to bind tande ly linked GAS motifs in a cooperative manner is completely lost in comparison with the wild type. Contrary, the binding to a single optimal GAS site is similar to the wild type indicating a DNA recognition as dimer only. In consequence of the usage of natural promoter fragments, the reduced DNA binding is accompanied by a reduced transcription activity as well. Since natural promoters often harbor a weak single canonical GAS site, Stat oligomerization is required for high transcriptional efficiency. This ability to form tetramers is tested by EMSAs (electrophoretic mobility shift assays).
Whereas any impaired DNA binding normally results in shortened nuclear retention, the nuclear accumulation of the Statl mutants is uncoupled from the DNA binding capacity. The mutated phenotype displays a Stat accumulation which is 200 to 300%, prolonged compared to the wild type. Furthermore, due to the tetramerization defect the mutants form oligomers less frequently and thus maintain a high intranuclear mobility which is equivalent to the wild type.
StatδaF81A and StatδbF81A show prolonged tyrosine phosphorylation and insensitivity towards staurosporine, which is indicative of a reduced dephosphorylation rate. Interestingly, in the IL-2 receptor-reconstituted 293T system the kinetics of dephosphorylation of wild type Statδ proteins is longer than that observed in lymphoid cells, where it occurs within 0.3 to 2 h. Nevertheless, mutation of residue 81 also prolongs tyrosine phosphorylation even further, as the phosphotyrosine signal now remains detectable for more than 10 h.
Finally, the N-domain point mutation has an impact on the gene transcription in a promoter-specific manner. Reporter genes containing optimal GAS sites are transactivated as good as or even better up to 300%, preferably up to 200% and more preferably up to 100% by the Stat mutants than by the respective wild type counterpart. The effective binding to such sites is less dependent upon cooperativity since the multimerization of optimal sites mimics oligomerization. In contrast, when natural promoter fragments are used, transactivation is lower up to 100%, preferably up to 7δ% and more preferably up to 50% with the mutants in comparison to the wild type Stat molecule. Here, Stat oligomerization acts as requirement of the subsequent DNA binding ability and an adequate gene induction.
In another preferred embodiment of this invention, the mutated Stat molecules are dimerized. Stat dimers are the basic scaffold for subsequent DNA binding. Based on the dimerization, Statl and Stat3 to Stat6 are able to form higher order complexes (dimer-dimer or higher) on any DNA promoter which does not display an optimal consensus sequence. However, this inherent feature of higher oligomerization is abrogated in the described mutants.
The present invention also relates to a fusion protein comprising the mutated
Stat molecule and at least one marker structure.
A "fusion protein" is a protein which consists of amino acid sequences derived from at least two different sources. In context with fusion proteins, a
"heterologous" amino acid sequence is a sequence derived from a source other than the source of the other components of the fusion protein.
In the meaning of the invention, heterologous "marker structures" are understood to be proteins which are coupled to the Stat mutants, enabling the detection of them by means of various biological, chemical and/or physical methods. Advantageously, it is possible to localize proteins under investigation in a living cell, especially in a host cell. Furthermore, it is possible e. g. to monitor the migration of proteins in real-time investigations.
A reporter gene encodes such a detectable marker protein. The reporter gene according to the invention either can be the overall DNA construct of the protein to be detected. However, it may also be not more than that DNA portion of the protein which has detectable activity. Consequently, any DNA structure encoding a region detectable by means of various detection methods may serve as reporter gene. Preferred in this context are expression constructs encoding proteins which can be detected easily and with high sensitivity.
For example, the inventive marker structure in the fusion protein can be chloramphenicol acetyl transferase (CAT). In particular, CAT catalyzes the transfer of an acetyl residue from acetyl-CoA to chloramphenicol. Among other things, CAT has the advantage of being stable in a cell, with a half-life of about 50 h. This is particularly advantageous where e. g. yields of reporter gene as high as possible are to be achieved. Advantageously, CAT has a good signal/background ratio.
Another advantageous marker structure in the meaning of the invention is luciferase. Luciferase advantageously has a high sensitivity and, owing to a relatively short half-life of about three hours, it is well-suited for induction studies. Particularly advantageous is a marker structure including two luciferases with distinct properties, one luciferase, e. g. from Phottinus pyralis, detecting the activity of the own promoter, while the co-transfected luciferase, e. g. from Renilla reniformis, is under control of the constitutive promoter and thus can be used advantageously for standardization purposes.
Further examples of marker structures in the meaning of the invention are β>-gaiactosidase, human growth hormone and secreted alkaline phosphatase. Advantageously, the marker structures of the invention can also be used as reporter genes in qualitative detection. In qualitative detection, it is possible e. g. to determine whether a selected cell has been transfected or not, or, it is possible to determine in the entire organism, in which type of tissue a promoter is active. For example, reporter genes for qualitative detection are the genes for luciferase and β-galactosidase.
In another preferred embodiment of the invention, the marker structure is green fluorescent protein (GFP), glutathione S-transferase (GST) and/or a fragment thereof. The green fluorescent protein advantageously has low cytotoxic activity or none at all. Advantageously, the absorption and emission maxima of green fluorescent protein are similar to that of fluorescein, so that e. g. a GFP-positive cell can be detected using the same methods as with cells stained with fluorescein, i. e., under UV light, under a fluorescence microscope, or in a cell sorter (FACS), for example.
Furthermore, preferred marker proteins which can be fused with the Stat mutants are GST as well as other fluorescent proteins such as the red or the yellow protein, as well as the maltose-binding protein (MBP), and others.
In another advantageous embodiment, the fusion protein contains the marker structure at the N-terminus and/or the C-terminus. The arrangement will depend on the maintenance of the three-dimensional structure and the functional activity of both Stat mutant and marker structure. The integration within the Stat mutant may disrupt its intermolecular order, however it should not be excluded.
The invention also relates to a nucleic acid sequence which encodes the Stat mutant or the fusion protein.
The term "nucleic acid sequence" refers to a natural or synthetic polymer of single- or double-stranded DNA or RNA alternatively including synthetic, non- natural or modified nucleotides which could be incorporated in DNA or RNA polymers. Each nucleotide consists of a sugar moiety, a phosphate moiety, and either a purine or pyrimidine residue.
The term "nucleic acid sequence which encodes" refers to that part of a gene which enciphers a protein, a polypeptide or a part thereof. The regulatory sequences and/or elements controlling the initiation or termination of transcription are excluded. The coding sequence and/or the regulatory element can normally be found in cells, in which case it is referred to as "autologous" or "endogenic", or cannot be situated in cells, in which case it is referred to as "heterologous".
"Gene" denotes a DNA sequence encoding a specific protein, and regulatory elements controlling the expression of said DNA sequence. A heterologous gene may also be composed of autologous elements arranged in an order and/or orientation which is normally not found in that cell the gene is transferred into. A heterologous gene can be derived completely or partially from any source known in the art, including a bacterial or viral genome or episome, eukaryotic nuclear or plasmid DNA, cDNA, or chemically synthesized DNA. The structural gene may form a continuous coding region, or may comprise one or more introns bordered by suitable splice junctions. The structural gene can consist of segments derived from various naturally occurring or synthetic sources.
The present invention also relates to a vector comprising the above-described nucleic acid sequence, particularly to a bacterial vector such as a plasmid, or a virus. The term "vector" denotes a recombinant DNA construct which can be a plasmid, a virus, or an autonomously replicating sequence, a phage, or a nucleotide sequence, which is linear or circular, consists of single- or double-stranded DNA or RNA, wherein a number of nucleotide sequences are linked or recombined to form a unique construction, and which is capable of introducing a promoter fragment and a DNA sequence of a selected gene product in sense or antisense orientation into a cell, together with suitable non-translated 3' sequences.
"Plasmids" are genetic elements which are stable inherited without being part of the chromosome of their host cell. They may comprise DNA or RNA, and they can be both linear and circular. Plasmids encode molecules ensuring their replication and stable inheritance during cell replication, and they are capable of encoding products of considerable importance for medicine, agriculture and environment. For example, they encode toxins which massively increase the virulence of pathogenic bacteria. They are also capable of encoding genes imparting resistance to antibiotics. In molecular biology, plasmids are generally used as vectors to clone and express recombinant genes. According to the standard nomenclatural rules familiar to those skilled in the art, plasmids are generally described using the small letter p preceded or followed by capital letters and/or numerals. The starting plasmids disclosed in the present specification either are commercially available, accessible to the public, or can be constructed from available plasmids by routine use of well-known, published methods. Many plasmids and other cloning and expression vectors which can be used according to the invention are well-known and easily available to the skilled artisan. Furthermore, a person skilled in the art can easily construct any number of other plasmids suitable for the use in this invention. The properties, construction and use of such plasmids, as well as other vectors, are readily accessible to those skilled in the art from the present disclosure.
Moreover, the present invention relates to host cells transfected with a vector of the invention, particularly to prokaryotic or eukaryotic cells. The present invention also relates to cell cultures, tissues, organs and the like, which comprise cells including an above-described plasmid or vector. The term "host cell" relates to a cell which has been genetically modified by the transfer of a chimeric, heterologous or autologous nucleic acid sequence or derivatives thereof still including said sequence. These cells are also referred to as "transgenic cells". Where an autologous nucleic acid sequence is transferred, the number of copies of this sequence in the host cell is higher than that of the naturally occurring sequences.
Object of the present invention is also the use of the Stat mutants of the invention, their dimers, the fusion protein of the invention, the nucleic acid sequence of the invention, the vector of the invention or the host cell of the invention for expression of a target protein, for investigating effects caused by different gene induction levels, for screening of active substances which modify the transcription activity of target genes, for identifying binding partners or for the production of a drug for the diagnosis and/or treatment of diseases associated with abnormal Stat induction of target genes. Binding assays are usually performed to detect protein-protein interactions. A well-established system represents the two-hybrid system, preferably a yeast two-hybrid system. The latter exploits the modular nature of many transcription factors which are typically composed of a DNA-binding domain (DBD) and an transactivation domain (TAD). The original transcription factor is split and two types of hybrids are created: the first one is a DBD-bait fusion, in which the bait represents a protein of interest or a fragment thereof, and the second hybrid" comprises an TAD-prey fusion, in which the prey represents a protein library to be investigated. Hyperactive STAT proteins may be exploited for interaction trap assays such as the yeast two-hybrid system. In these assays a probe or "bait" protein fused to a heterologous DNA-binding protein domain reacts with components of a conditionally expressed library comprising fusion proteins with a portable transcriptional activation domain. The function of STAT proteins as constitutive shuttling molecules may facilitate the search for interactions between the bait and the fusion proteins harboring a transactivation domain.
Furthermore, in the meaning of the invention, any of the Stat mutants could act as bait which is fused to the LexA DBD, for example, whereas the protein library could be merged with the Gal4 TAD domain. Stat mutant-LexA DBD fusion proteins encoded in a first vector and Iibrary-Gal4 TAD fusion proteins encoded in a second vector are transformed into two yeast strains which are cultured under appropriate conditions and over an appropriate period. Subsequently, the yeast strains are mated, grown on plates and examined for LacZ expression. The identified binding partners may elucidate the control of signaling pathways and/or different gene induction levels, for instance by cooperative binding. In addition, they could be useful as markers for pathogenic phenotypes and/or as activators or inhibitors of Stat molecules in order to modify the transcription activity of target genes.
The present invention also relates to a method for the expression of a target protein including the addition of the mutated Stat monomer of the invention, their dimers or the fusion protein of the invention to an in-vitro system containing genes under Stat control. The in-vitro system preferably represents a cell and more preferably a cell-free system. Furthermore, the method for the expression of a target protein includes the nucleic acid sequence of the invention preferably inserted into the vector of the invention which is preferably transformed or transfected into the host cell of the invention.
The Stat mutants exert an influence on gene activation in a promoter-specific manner. While the transcription yield is strongly enhanced if activated on optimal GAS sites, the opposite phenomenon appears on natural promoter sequences. Thus, either an overexpression or an underexpression of a target protein can be realized compared to the matching gene induction by the Stat wild type.
A suitable vector containing a promoter of choice followed by the DNA sequence of the Stat mutant is transformed into an appropriate cell culture. Depending on the cell culture, a phosphorylation system could be included for Stat activation. Alternatively, a construct of promoter harboring at least a single optimal GAS sequence and a target gene of choice is preferably inserted into a second vector. High protein amounts can be advantageously obtained in this way.
In addition to the cellular expression, the cell-free system is applicable. Cell-free systems involve coupled transcription and translation reactions in-vitro which are based on a cell lysate (generally called S30). The lysate is pre-produced by means of cell disruption and subsequent centrifugation at 30.000 g. The advantages of the cell-free protein biosynthesis include, inter alia, the solely dependence of protein expression on the template, high yields as well as the possibilities to produce cell-toxic proteins and to incorporate unusual amino acids into the protein. Two vectors which are generated to harbor the Stat insert and the target gene as described in the cellular in-vitro section are introduced to the cell-free system. The encoded proteins are expressed and the tyrosine phosphorylation of the Stat mutant is performed with EGF receptor or supplemented Jak.
Instead of the simultaneous expression of Stat mutant and target protein in the cell-free system, the addition of a mutated Stat monomer, a dimerized Stat molecule or a fusion protein separately expressed in advance is imaginable. The term "expression" used in the present specification is intended to describe the transcription and translation of the sequence of the gene product. Where the gene product is a protein, the expression involves initial transcription of a DNA chain encoding the sequence of a gene product, to form a complementary RNA, frequently a mRNA, followed by the translation of the mRNA, to form the above- mentioned gene product. However, expression also comprises transcription of a DNA inserted in antisense direction with respect to its regulatory elements. An expression which proceeds in a constitutive fashion and is possibly further enhanced by an externally controlled promoter fragment - to produce multiple mRNA copies and large amounts of the selected gene product - may also involve overproduction of a gene product.
For expression of recombinant Stat proteins the wild type Stat cDNA sequence preferably cloned into a baculovirus transfer vector is mutated by site-directed mutagenesis at the appropriate residues which is confirmed by DNA sequence analysis. Tyrosine phosphorylation is checked by Western blot analysis using phosphotyrosine-specific antibodies.
The present invention also relates to a method for investigating effects caused by different gene induction levels including the addition of the mutated Stat monomer of the invention, their dimers or the fusion protein of the invention to an in-vitro system containing genes under Stat control. In the meaning of the invention, such effects are understood to be any influence on transcription activity, resulting in an increase or drop in activity, which can be determined by means of biological, chemical or physical measuring methods. The in-vitro system preferably represents a cell and more preferably a cell-free system. Furthermore, the method for investigating effects caused by different gene induction levels includes the nucleic acid sequence of the invention preferably inserted into the vector of the invention which is preferably transformed or transfected into the host cell of the invention.
The Stat mutation of the tetramerization interface affects the transcription in a promoter-specific manner. Depending on the overexpression or the underexpression of selected target proteins, biochemical effects on the living cell will appear. They may concern the entire range of biochemical life processes, like metabolic rate, cell growth, proliferation, aging etc. The up- or down-regulation of genes is tightly controlled. The resulting observations are very helpful to reveal unknown functions of genes. The general experimental procedure is identical to that already described for the method for expression performed in cells. The monitoring of the effects depends on the target gene to be investigated. A couple of methods to detect a gene product is mentioned within the description of the next embodiment concerning the screening of active substances.
A "detectable gene product" is a nucleotide sequence or amino acid sequence which can be detected by means of an appropriate test depending on the gene product itself. Preferably, the expression of a detectable gene product provides the cell with a feature allowing easy selection of that cell among other cells not expressing the detectable gene product.
The present invention also relates to a method for the screening of active substances which modify the transcription activity of target genes including the addition of the mutated Stat monomer of the invention, their dimers or the fusion protein of the invention to an in-vitro system containing genes under Stat control. In the meaning of the invention, such modifying is understood to be any influence on transcription activity, resulting in an increase or drop in activity, which can be determined by means of biological, chemical or physical measuring methods. The in-vitro system preferably represents a cell and more preferably a. cell-free system. Furthermore, the screening method includes the nucleic acid sequence of the invention preferably inserted into the vector of the invention which is preferably transformed or transfected into the host cell of the invention.
Generally, a suitable expression vector containing a promoter of choice followed by the DNA sequence of the Stat mutant is transformed into an appropriate cell culture firstly. A target gene which is cell-inherent can be chosen thereby exclusively investigating the effect of an active substance on the interaction between Stat mutant and natural promoters. Alternatively, the construct of promoter and reporter gene of choice is preferably inserted into a second vector choosing a weak natural promoter (e. g. PRRIII or ICAM-1 ) for the screening of transcription activators or a strong synthetic promoter (e. g. the PRRIII derivative M10 or Ly6E) for the opposite selection of transcription inhibitors. For example, the treatment of human malignancies requires agents that reduce Stat levels and/or Stat activity, along with the promoter-binding capability. Besides, a fusion protein construct can be used to recognize Stat relocations. Those skilled in the art can easily produce these constructs by well-known recombination and cloning techniques.
For screening, a couple of methods are to be distinguished by the quantitative determination either of the product of transcription or that of translation. On the level of transcription, the synthesized RNA can be detected via DNA by semiquantitative RT-PCR. Cells stable expressing any mutant are cultivated and spread on a microtiter plate containing a substance library. Cells are treated with cytokines, like the ϊnterferons IFNα and IFNγ or interleukins, to induce corresponding responsive genes. They are kept interacting for a certain time before the RNA is extracted and reverse transcribed followed by PCR. The PCR products can be analyzed either directly or by gel electrophoresis and quantified by scoring the intensities of ethidium bromide staining. The results are finally compared to the untreated cell sample thus revealing the mode and direction of changes in mRNA yield. Likewise, the cell-free system already described can be treated with a library of substances followed by the detection of the target mRNA.
The protein can be translated either in cells or in the cell-free system. In the first one, eukaryotic cells are transfected with the plasmid constructs as described above. After a certain period of adaptation the cells are spread on a microtiter plate containing a library of substances to be screened. The Stat transport to the nucleus is induced by administering the appropriate cytokines leading to nuclear accumulation firstly. Secondly, changes in the expression level of target genes may occur due to association of library substances with Stat mutants.
In the context of the present invention, the term "association" - without being limited thereto - relates to any type of interaction between the Stat molecule and the target, particularly covalent or non-covalent binding or association, such as a covalent bond, hydrophobic/hydrophilic interactions, van der Waals forces, ion pairs, ligand-receptor interactions, interactions between epitope and antibody binding site, nucleotide base pairing, and the like. Such association may also encompass the presence of other molecules such as peptides, proteins or other nucleotide sequences.
As an example in the context of the invention, the changed yield of a reporter gene product as marker for transcription activity is monitored via firefly luminescence in the luciferase assay system.
For the detection of cell-inherent gene products, an immunoassay is applicable. ..Immunoassay" denotes a test wherein an antibody is used for specific binding an analyte. The immunoassay is characterized in that specific binding properties of a particular antibody are utilized in the isolation, purposeful testing and/or quantitative determination of the analyte. Those skilled in the art can easily produce antibodies directed against the target protein to be monitored and fragments thereof.
..Antibody" denotes a polypeptide essentially encoded by an immunoglobulin gene or fragments thereof specifically binding and recognizing an analyte (antigen). Known immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon, and' mu genes for the constant region, as well as the innumerable genes for the variable immunoglobulin region. For example, antibodies are present as intact immunoglobulins or as a number of well- characterized fragments produced by cleavage using various peptidases. An antibody binds specifically to a protein, e. g. another biological structure, or exhibits specific immunoreactivity in this way, if the antibody assumes its function in a binding reaction in the presence of a heterogeneous population of proteins and other biological substances, which reaction allows a decision to be made whether the protein or another biological structure is present. Under the established conditions of an immunoassay, the antibodies mentioned preferably bind to a specific protein, whereas no significant binding to other proteins present in the sample takes place. Specific binding to a protein under such conditions requires an antibody which has been selected according to its specificity for a particular protein. Various variants of immunoassays can be used to select antibodies having specific immunoreactivity with a particular protein. A description of immunoassay variants and conditions which can be used to determine a specific immunoreactivity is presented in Harlow & Lane, Antibodies; A Laboratory Manual (1988), Cold Spring Harbor Publications, New York.
In addition to the cellular gene expression and substance screening, the application of the cell-free system is imaginable. Two vectors which are generated to harbor the Stat insert and the target gene as described in the cellular in-vitro section are introduced to the cell-free system which is subsequently spread on a microtiter plate containing a library of substances. The encoded proteins are expressed and the tyrosine phosphorylation of the
Stat mutant is performed with EGF receptor or supplemented Jak. Instead of the simultaneous expression of Stat mutant and target protein in the cell-free system, the addition of a mutated Stat monomer, a dimerized Stat or a fusion protein separately expressed in advance is possible as already described above.
The present invention also relates to a method for the production of a drug for the diagnosis and/or treatment of diseases associated with abnormal Stat induction of target genes including the detection of the mutated Stat monomer of the invention, their dimers or the fusion protein of the invention in a cell.
Furthermore, the method for the production of a drug includes the nucleic acid sequence of the invention preferably inserted into the vector of the invention which is preferably transformed or transfected into the host cell of the invention.
According to the invention, a "disease associated with abnormal Stat induction of target genes" is arthritis, carcinogenesis, nephritis, proteinuria, dermatitis, diabetes/obesity, acute and chronic rejection of allogeneic organ transplants, neuron degeneration, Parkinson's disease, septic shock, endotoxemia, hypersensitivity, uveitis, wound closure, and others. The abnormal Stat induction of target genes also plays an important role in the regulation of the immune system, in inflammation and effector mechanisms of cellular and humoral immunity, septic shock, inflammation of sensory organs, like the eyes, in the regulation of transcription, cell cycles and cell growth, acute respiratory insufficiency, physiological stress, numerous cardiovascular diseases and other disorders. In the context with the present invention, the term "treatment" refers to the prophylactic and/or therapeutic effect of a drug.
Dysfunctions of signaling pathways are e. g. reflected by inappropriate activation and accumulation of Stat molecules. Therefore, agents that inhibit Stat functions by a decrease of Stat levels and/or binding specifity to target DNA sequences would represent a promising approach which may have the potential to be more selective and less toxic than current anti-cancer treatments. The phenotype of the Stat mutants this invention is based on corresponds with the mentioned requirements. The interactions of the alternate N-domain interface of the mutants which do not mediate oligomerization and block dephosphorylation are probably accompanied by a three-dimensional structural rearrangement. Hence, the application of high affinity molecules which are able to discriminate between the wild type structure and the mutated one is suggested for diagnosis. These high affinity molecules involve antibodies already described or RNA aptamers. The latter are identified by an in-vitro selection procedure - the so-called SELEX process (systematic evolution of ligands by exponential enrichment). Since RNA is very susceptible to nucleolytic degradation in biological solutions, aptamers are chemically modified using phosphorothioates, locked nucleic acids or Spiegelmers™.. Because of their high affinity for a broad spectrum of structural targets, aptamers act very similar to antibodies. Both, antibodies and aptamers carry functional coumpounds which are detectable to determine their binding quantity to the Stat target. As example, the fluorescence labelling is mentioned.
Referring to a desired reduction of Stat activity for the treatment of diseases caused by a hyperactivation, the wild type Stat molecule should be replaced by the Stat mutant. This transfer may happen by means of a gene therapy. The most common method involves the introduction of genetic material into a gene shuttle, for instance retro viruses. The replacement of the Stat gene in certain cell types can also be performed in-vitro and the modified cells are subsequently re-injected into the organism. The recombination of the vehicle genome or the cell genome, respectively, with the nucleic acid sequence encoding the Stat mutant follows standard techniques, whereas targeting and the stable incorporation of the new gene represent current research topics. The present invention also relates to a method for the importing and targeting of substances into the nucleus which includes the vector encoding the proteins of the invention. The vector is preferably transformed or transfected into the host 5 cell of the invention. These substances preferably represent peptides or proteins which can be attached to any terminus of the Stat molecule and simultaneously expressed as fusion construct. Firstly, the peptides or proteins should be able to interact specifically with physiologically interesting endogenous compounds of the0 nucleus or exogenous compounds expressed by co-transfection. Secondly, these imported substances have to display a higher target affinity in comparison to the carrier Stat molecule. The latter fact is essential for their preferred successful association to the target. Inhibitors and activators of transcription which bind to DNA or enzymes are suited molecules to be transferred into the5 nucleus.
Eukaryotic cells are transfected in-vitro with the vector encoding the Stat mutant in combination with an active peptide structure. Those skilled in the art can easily produce these constructs by well-known recombination and cloning o techniques. Alternatively to the single Stat mutant, the fusion protein containing a marker structure can be used to pursuit Stat relocations during import and within the nucleus. The transport to the nucleus is induced by administering interferon or interleukin, respectively, leading to nuclear accumulation. The effects of the5 imported substance on the cell metabolism are detected by an appropriate method depending on the gene product. Furthermore, the construct of Stat mutant and active substance may be introduced in-vivo by using a vehicle like a virus. In this case, the procedure is to be o considered as gene therapy.
All statements concerning the use and the methods for several purposes also include a Stat molecule derived from wild type Statl , which is mutated in at least one conserved hydrophobic amino acid on the surface of the N-domain,5 preferably in the amino acids in position 77 and/or position 78. The mutation in position 77 may involve a substitution by alanine, and the mutation in position 78 may involve a substitution by serine. There are two slicing variants alpha and beta differing in the terminal amino acid sequence from 713 to 7δ0 which is absent in the beta isoform. The mutants of the following sequences are termed StatlαF77A and StatlβF77A.
Human StatlαF77A (1-750): msqwyelqql dskfleqvhq lyddsfpmei rqylaqwlek qdwehaandv sfatirfhdl lsqlddqysr fslennallq hnirkskrnl qdnfqedpiq msmiiysclk eerkilenaq rfnqaqsgni qstvmldkqk eldskvrnvk dkvmciehei ksledlqdey dfkcktlqnr ehetngvaks dqkqeqlllk kmylmldnkr kewhkiiel lnvteltqna lindelvewk rrqqsacigg ppnacldqlq nwftivaesl qqvrqqlkkl eeleqkytye hdpitknkqv lwdrtfslfq qliqssfwe rqpcmpthpq rplvlktgvq ftvklrllvk lqelnynlkv kvlfdkdvne rntvkgfrkf nilgthtkvm nmeestngsl aaefrhlqlk eqknagtrtn egplivteel hslsfetqlc qpglvidlet tslpvwisn vsqlpsgwas ilwynmlvae prnlsffltp pcarwaqlse vlswqfssvt krglnvdqln mlgekllgpn aspdglipwt rfckenindk nfpfwlwies ilelikkhll plwndgcimg fiskereral lkdqqpgtfl lrfsessreg aitftwvers qnggepdfha vepytkkels avtfpdiirn ykvmaaenip enplkylypn idkdhafgky ysrpkeapep eldgpkgtg yiktelisvs evhpsrlqtt dnllpmspee fdevsrivgs vefdsmmntv
Human Statl βF77A (1-712): msqwyelqql dskfleqvhq lyddsfpmei rqylaqwlek qdwehaandv sfatirfhdl lsqlddqysr fslennallq hnirkskrnl qdnfqedpiq msmiiysclk eerkilenaq rfnqaqsgni qstvmldkqk eldskvrnvk dkvmciehei ksledlqdey dfkcktlqnr ehetngvaks dqkqeqlllk kmylmldnkr kewhkiiel lnvteltqna lindelvewk rrqqsacigg ppnacldqlq nwftivaesl qqvrqqlkkl eeleqkytye hdpitknkqv lwdrtfslfq qliqssfwe rqpcmpthpq rplvlktgvq ftvklrllvk lqelnynlkv kvlfdkdvne rntvkgfrkf nilgthtkvm nmeestngsl aaefrhlqlk eqknagtrtn egplivteel hslsfetqlc qpglvidlet tslpvwisn vsqlpsgwas ilwynmlvae prnlsffltp pcarwaqlse vlswqfssvt krglnvdqln mlgekllgpn aspdglipwt rfckenindk nfpfwlwies ilelikkhll plwndgcimg fiskereral lkdqqpgtfl lrfsessreg aitftwvers qnggepdfha vepytkkels avtfpdiirn ykvmaaenip enplkylypn idkdhafgky ysrpkeapep meldgpkgtg yiktelisvs ev
The mutated amino acid is bold.
All statements concerning the methods for several purposes also include the addition or detection of the dimerized Statl mutant, which is mutated as described above, or of a fusion protein including the Statl mutant and at least one marker structure. Furthermore, these methods include the nucleic acid sequence which encodes the Statl mutant or the fusion protein preferably inserted into a vector which is preferably transformed or transfected into the host cell.
Their integration into any special method mentioned proceeds in accordance to the previous description. The methods are performed identically to those already described for other Stat derivatives.
The present invention also relates to a kit comprising the amino acid sequence or the nucleic acid sequence of the Stat mutants of the invention, their dimers or fusion proteins, and/or synthetic analogues, modifications and pharmacologically active fragments thereof and an information about the using of the kit:
The kits to be built-up are either based on the genotype or the phenotype of the Stat mutant. They can also include parts of the amino acid sequence or the nucleic acid sequence originated from the Stat mutant, a vector harboring the nucleic acid sequence or a host cell containing the vector. The kits should be preferably applied for any of the methods described above.
The following examples are provided by way of illustration and not by way of limitation. Within the examples, standard reagents and buffers that are free from contaminating activities (whenever practical) are used. It is preferred to exercise care to avoid ribonucleases and PCR product contamination.
1. Example The plasmid expressing full-length human STAT1 (amino acids 1-746) fused to the N-terminus of GFP (pSTAT1-GFP) was described previously (Begirt et al, Cell Biol. 97, 10418-10423 (2000)). Stat expression constructs were generated by cloning the Statl full-length coding region in the SamHI and Not\ sites of the vector pcDNA3 (Invitrogen). Those skilled in the art can easily produce these constructs by well-known recombination and cloning techniques.
RT-PCR analysis of endogenous IFNα- and IFNγ-responsive genes was performed with U3A cells stably expressing Statl wild type or Statl F77A mutant. Cells were grown for 1δ h in Dulbecco's modified Eagle's medium with reduced serum (1 %), followed by the addition of serum to 10% and treatment with or without IFNα or IFNγ for 4 h. RNA was extracted and reverse transcribed followed by PCR. The following primer pairs were used: IRF-1 (GenBank™ accession number NM_002198) δ'-ATGAGACCCTGGCTAGAG-3' and δ'-AAGCATCCGGTACACTCG-3'; LMP- 2 (GenBank™ accession number U0102δ) δ'-GG-GGGCGTTGTGATGGGTTCTGATTC-3' and δ'-TCGGGAGACATGCCTGGCTTATAT-3'; ISG-15 (GenBank™ accession number NM_00δ101) δ'-GGCGGGCAACGAATTCCAGGTGTC-3' and δ'-CCCCGCAGGCGCAGATTCATGA-3'; ISG-δ6k (GenBank™ accession number M24594) δ'-AGGGCAGAACAGAGAAAAGCTAGACAA-3' and δ'-CCGCTCATAGTACTCCAGGGCTTCATTC-3'. The ethidium bromide-stained PCR products were quantified. Absolute intensities were scored in arbitrary units, and the ratio between induced and uninduced signal was depicted for each gene.
Of the seven genes tested, only LMP-2 expression was unaffected by the N- domain mutation of Statl , whereas the others showed a strongly diminished IFNγ-responsiveness in cells expressing Statl F77A. Strong effects were to be seen with the MIG-1 , IRF-1 , IRF-9, and GBP-1 genes, which had lost their IFNγ- responsiveness in cells expressing the Statl mutant protein. The importance of tetramer formation for gene induction in a natural setting was further evaluated by scoring gene induction in response to IFNα. Contrary to IFNγ, which signals via a Statl homodimer, stimulation of cells with IFNα induced the additional formation of a ternary complex called ISGF-3, consisting 5 of a Stat1/Stat2 heterodimer and IRF9, which binds to an unrelated recognition site termed interferon-stimulated response element (ISRE). Expression of the ISG-1δ gene was highly induced by IFNα, and mutation of F77 reduced its IFNα response by one half, whereas the weak induction of ISG-δ6 was not adversely affected by the N-domain mutation (Figure 11).0 Due to the association of added library substances with Stat mutants, changes in the transcription yield of target genes may occur and can be easily detected.
2. Example The plasmids pCIStatδa and pCIStatδb were described (John et al, Mol. Cell.5 Biol. 19, 1910-1918 (1999)). Site specific mutagenesis of these plasmids according to manufactures' instructions (QuikChange™, Stratagene; MORPH site-specific plasmid mutagenesis kit, δ'-3' Inc.) generated the respective N- domain mutants StatδaF81A and StatδbF81A. For reporter gene assay, 4*106 293T cells were co-transfected with the 0 following DNAs: 2 μg IL-2 receptor β, O.δ μg γc, 0.2δ μg Jak3, 10 ng of each of the wild type Statδ (control), StatδaF81A or Stat5bF81A mutant expression plasmids and 1 μg of PRRIII-E1b or M10-E1b-luciferase reporter construct as described (John et al, Mol. Cell. Biol. 19, 1910-1918 (1999)). Either the weak natural promoter PRRIII (Soldani et al, Mol. Cell. Biol. 20, 389-401 (2000)) for 5 the possible screening of transcription activators or the strong synthetic promoter, the PRRIII derivative M10 (Vinkemeier et al, EMBO J. 1δ, δ616-δ626 (1996)), for the possible screening of transcription inhibitors was applied to drive the luciferase gene. After 48 hours cells were spread on a microtiter plate containing a library of substances to be screened. The Statδ transport to the o nucleus was induced by administering 2 nM human interieukin 2 (IL-2) overnight firstly leading to nuclear accumulation and secondly to reporter gene expression (Figure 12). Due to the association of added library substances with Stat mutants, changes in the expression level of the luciferase gene may occur and are monitored via5 luminescence according to manufacturer's protocols (Promega). 3. Example
In this control study we examined the behavior of various unphosphorylated Stat proteins in the absence of cytokine stimulation. Unphosphorylated Stats, preferably Statl , can enter the nucleus independently of p97 by an unknown mechanism. As generally only molecules of less than 30 kD can freely cross the nuclear pore complex (NPC) by passive diffusion, the signal-independent nuclear import of unphosphorylated Stats (Mr > 87 kD) requires specific interactions with either the NPC or another intermediary carrier. Here, it was demonstrated that Stats can undergo rapid translocation through the nuclear pore in a cytosol-unassisted and carrier-independent manner that does not require metabolic energy. In the meaning of theinvention, Stats derived form any Stat wild type or fragments thereof shuttle between cytosol and nucleoplasm, preferably Statl , Stat3 and Statδ, more preferably Statl and most preferably Statltc (representing a truncated Statl protein lacking the NH2- and the COOH-terminal domain). The direct binding of Stats to nucleoporins (Nups) indicated that unphosphorylated Stats migrate into the nucleus via specific molecular interactions with components of the NPC. In the meaning of the invention, these components of the NPC comprise nucleoporins and fragments thereof, preferably Nup1δ3 and Nup214. The nucleoporins Nup1δ3 and Nup214
(the number refers to the molecular weight) were preferred chosen because of the exposed arrangement within the nuclear pore making heterologous intermolecular interactions possible, their detailed characterization and high expression yields for subsequent in-vitro investigations. In another embodiment of the present invention, molecules which are structurally and functionally homologous to nucleoporins interact with Stats.
The invention also relates to a Stat mutant which could preclude the aforementioned nuclear import. In the meaning of the present invention, a Stat molecule derived form any Stat wild type or fragments thereof is mutated, preferably Statl , Stat3 and Statδ, more preferably Statl and most preferably
Statltc. Furthermore, at least one amino acid is mutated within the linker domain of the Stat molecule, preferably cysteines, more preferably cysteine in position 643 of Statl or in the equivalent position of homologous proteins. Possible mutations comprise the deletion, insertion, substitution and/or modification of at least one amino acid, preferably a single residue. Possible modifications are alkylation, arylation and acetylation, preferably the alkylation. In a specific embodiment of this invention, the amino acid Cysδ43 in Statl or in the equivalent position of homologous proteins is alkylated. The phenotype of shuttle loss can be effectively used as negative control in bioassays requiring shuttling. Preferably, the phenotype represents a test within gene induction assays, like the two-hybrid system.
Cells were grown in complete growth medium. LMB (5 ng/ml, Sigma) was added to the cells 60 min before microinjection. For energy depletion studies, HeLa-S3 or NIH-3T3 cells were washed with PBS, and incubated in serum- and glucose-free DMEM (GIBCO). This medium was supplemented with 10 mM sodium azide and 10 mM 2-deoxy-D-glucose (energy depletion-medium, EDM) and left on the washed cells for 2 h. Subsequently, the cells were either fixed in methanol for 6 min at -20° C,. or microinjected with antibodies or recombinant protein and the incubation in EDM was continued for the indicated times. Alternatively, after 2 h in EDM the cells were kept in complete growth medium for 5 h before fixation. Statl was detected by immunocytochemistry with antibody E-23 (0.2 μg/ml; Santa Cruz).
For expression in bacteria, the cDNAs encoding truncated human Statl (aa
130-712), mouse Stat3 (aa 136-716), or sheep Statδ (aa 138-704) were amplified by PCR and cloned into the EcoRI and BamHl sites of pASKlBA3 (IBA, Gδttingen). The cDNA encoding aa 4δ-462 of p97 was PCR-amplified and cloned into pGEX-5X-2 (Pharmacia). MBP fusion proteins of the FG repeat regions of human Nup62 (aa 1-308, a kind gift of E. Hurt) and human Nup153 (aa 333-618, a kind gift of B. K. Felber) were prepared after PCR-amplification of the respective cDNAs and cloned into the BamHI site of pMal-2CX (NEB). PCR reactions were performed with Vent-proofreading polymerase (NEB). Site- directed mutagenesis was performed with the Quick-change kit (Stratagene). Baculovirus transfer vectors (pFastBac) encoding human Statl wt (aa 1-746) or the Tyr701Phe mutant and vectors for bacterial expression of GST-NLS-GFP and GST-NES-GFP have been described. Expression constructs for His-tagged human p97 (aa 1-878) and human Nup214 (aa 1549-2090) were kind gifts of U. Kutay and M. Fomerod, respectively. Expression, purification, alkylation, and fluorescent labeling of recombinant proteins were performed as follows. Expression of Statl wt and Statl Y701F in baculovirus-infected Sf9 insect cells, the purification and tyrosine phosphorylation were done as previously described. Protein expression in BL21p(Lys)S bacteria was induced at an OD6oo of 0.8 with 1 mM anhydrotetracycline (Acros, pASKIBA3 vector), or IPTG (1 M for pGEX and O.3 mM for pMal vectors, respectively). Protein expression was at 18° C for 15 h (Strep-tagged proteins), or at 30° C for 5 h or 3 h with GST- or MBP-tagged proteins, respectively. Cells were lysed by sonication in PBS containing 1% Triton X-100, 2 mM EDTA, 2 mM EGTA, δ mM DTT. Proteins were single-step purified by virtue of their Strep- or GST-tag, respectively, as recommended by the manufacturers (IBA; Pharmacia). Where indicated, purified proteins were phosphorylated alkylated with NEM (20 mM) as described. Proteins were concentrated by ultrafiltration (Centriprep, Millipore) in injection buffer containing 5 mM DTT; protein concentrations were determined by UV spectroscopy (extinction coefficient 1.25 for Statlwt, 1.27 for Statltc, 1.24 for Stat3tc, and 1 .43 for Statδtc), or by Coomassie dye-binding (Biorad) with a BSA standard. Proteins were quick-frozen on dry ice and stored at -80° C. Fluorescence labeling of Statlwt and Statltc was done with succinimidyl esters of Alexa Fluor 594 or Oregon Green 488 as described by the manufacturer (Molecular Probes) at a molar protein:dye ratio of 1 :5. Efficiency of labeling was close to 100% as judged by mobility shift in SDS-PAGE (Figure 1). His-tagged p97 was expressed and purified as described.
Microinjections of antibodies (at 0.1δ mg/ml in PBS) and concentrated recombinant proteins (at 0.δ-1 mg/ml) were as described. Injection site markers were used at a concentration of 0.δ-1 mg/ml, WGA (Sigma) was co- microinjected at a concentration of O.δ mg/ml. Immunocytochemistry was with monoclonal Statl antibody C-136 (0.2 μg/ml; Santa Cruz) or anti-Strep-tag II rabbit polyclonal antibody (0.2 μg/ml; IBA).
Import assays with permeabilized cells were performed as follows: Adherent
HeLa-S3 cells were grown on coverslips and permeabilized with 40 μg/ml dϊgitonin (Roche) in transport buffer (TB, 20 mM Hepes, pH 7.3, 110 mM KOAC, 2 mM Mg(OAC)2l 1 mM EGTA, 2 mM DTT, Complete protease inhibitors (Roche)) for 6 min on ice. Subsequently, the cells were washed in TB twice. Incubation with 20 μl import mix was performed at RT for 60 min or as indicated. The cytosol-free import mix (IM) contained TB supplemented with 10 mg/ml BSA and one or several of the following purified proteins as indicated: 1 μM Statltc, 1 μM Stat3tc, 1 μM Statδtc, 1 μM Oregon Green 488- or Alexa Fluor 594-labeled Statltc, 15 μM Statlwt, 0.8 μM GST-NLS-GFP, or 0.8 μM full- length His-tagged p97. The complete import mix (CIM) contained 75% (v/v) rabbit reticulocyte lysate (Promega), 25% IM, and an energy regenerating system (0,5 mM ATP, 0,5 mM GTP, 10 mM creatine phosphate, 30 U/ml creatine phosphokinase). For WGA treatment, digitonin-permeabilized cells were incubated for 10 min on ice in IM containing 250 μg/ml WGA or as indicated, before washing and addition of the import mix. For ATP depletion, IM or CIM were preincubated for 15 min on ice in the presence of 0.8 U/ml apyrase (Sigma) before addition to the permeabilized cells. For competition assays, 16 μM truncated p97 (aa 45-462) or 20 μM full length Statl was added to IM and left on the cells for 45 min. After the import reaction the cells were washed with ice-cold TB and fixed for 10 min at RT with 3.7% para-formaldehyde in PBS, before permeabilization with 0.2% Triton X-100 in PBS (2 min at RT). Truncated Stats were detected with Strep-tag II antibody (0.17 μg/ml); for Statlwt antibody C-136 was used (0.07 μg/ml); and His-tagged p97 was detected with anti-His antibody (0.25 μg/ml; Abeam). Fixed cells were mounted in TB containing 50% glycerol, 1% (w/v) NaN , 1% (w/v) Diazobicyclo (2,2,2)-odone.
In vitro binding assay was perfomed as follows: Bacterial lysates (containing approx. 6 μg NPC proteins or MBP) and 6 μg purified His-tagged control protein (tandem SH3 domains from human FYB; a kind gift of C. Freund) were resolved by 10% SDS-PAGE and transferred to a nitrocellulose membrane. The membrane was rinsed in transport buffer (20 mM HEPES, pH 7.4, 110 mM KOAc, 2 mM Mg(OAc)2) 0.5 mM EGTA, 2 mM DTT, 0.5% Tween-20, Complete protease inhibitors), and then blocked in this buffer plus 5% (w/v) non-fat milk for 1 h at 4°C. Recombinant full length Statl was added at 3 μg/ml, and the incubation continued for 16 h at 4°C. Subsequently, the blot was rinsed three times with transport buffer and subjected to Western analysis using anti-Statl antibody C-136 (0.2 μg/ml). Fluorescence microscopy and fluorescence quantification were performed as follows: Conventional and confocal fluorescence analysis and quantification of immunofluorescence intensities were performed as described. The data were analysed by ANOVA followed by Tukey multiple comparison tests. Differences were considered statistically significant at p < O.Oδ.
Various recombinant Stat proteins were prepared to gain insight into the cytokine-independent nucleocytoplasmic translocation of these transcription factors (Figure 1). Full length Statl was isolated from baculovirus infected insect cells. A stable and well-characterized truncated variant, Statltc, which lacks both the N-domain of 129 residues and the C-terminal transactivation domain of 38 residues was expressed in bacteria, as were analogous mutants of Stat3 and Statδ (Mr > 6δ kD). The truncated Stat proteins were purified by virtue of a small C-terminal Strep-tag, which was also useful for indirect immunocytochemical detection. The recombinant proteins were microinjected into unstimulated cell lines or used for import assays with permeabilized cells. In addition, endogenous Statl was targeted by antibody microϊnjection to reveal the flux rates of the native protein.
Microinjected Statl , Stat3, and Statδ rapidly migrate into the nucleus of unstimulated living cells. Several unstimulated cell lines were microinjected into the cytosol with full length unphosphorylated Statl . In HeLa-S3 cells (Figure 2A), COS7 cells (Figure 2B), and 2fTGH cells (Figure 2C) nuclear import occurred with identical velocity, as it took only 1δ min to reach pancellular distribution. Thereafter, no further nuclear accumulation could be observed and the nucleocytoplasmic distribution remained stable (Figure 2D). We then compared wild type Statl with a tyrosine phosphorylation-defective mutant (Tyr701 Phe) and with truncated Statltc. As is shown in Figure 2E and F, nuclear translocation of the two Statl variant proteins was indiscriminable from wild type and also resulted in a pancellular distribution at equilibrium, which indicated that tyrosine phosphorylation as well as N- and C-termini were dispensable for cytokine-independent nuclear import. Next we tested the import abilities of truncated Stat3 and Statδ, and found that both proteins enter the nucleus with kinetics similar to Statl (Figure 2G and H). Notably, the injected Stat3 repeatedly showed a higher steady state concentration in the nucleus than the two other Stat proteins. Additionally, Statl and Statltc were labeled on lysine residues with fluorescent dyes (either Oregon Green 488 or Alexa Fluor 594) to directly observe their nuclear translocation. As is shown in Figure 21 for Oregon Green 488-labeled Statltc, conjugation with fluorescent dyes did not interfere with nuclear import (identical result were obtained with Alexa Fluor 594 and for full length Statl). To demonstrate exclusive passage of the microinjected material through the nuclear pore, we co-injected wheat germ agglutinin (WGA), a lectin known to inhibit nuclear pore-mediated translocation of macromolecules. Expectedly, passage across the nuclear envelope of both full length and truncated Statl was effectively blocked (Figure 2J, K).
The requirements for the constitutive nuclear import and export of recombinant Statl are biochemically distinguishable. Next, recombinant Statlwt was injected into the nucleus to investigate export kinetics. The microinjected protein quickly left the nucleus and spread evenly across the cell. However, nuclear export appeared to occur slower than nuclear import. It usually took about 30 to 60 min to reach a pancellular distribution (Figure 3A), which then remained stable (observation for another 2 h) (Figure 3B). Nuclear export of Statl after cytokine- induced nuclear accumulation is achieved in part via the transport receptor CRMi . It is unclear, however, whether CRM1 -dependent nuclear export also functions in the absence of cytokine stimulation. Therefore, cells were treated with the CRM1 inhibitor leptomycin B (LMB) starting 60 min before nuclear microinjection. This treatment attenuated nuclear export, since the recombinant protein was still predominantly nuclear after 1 h (Figure 3C), and a pancellular distribution was achieved not even after 4 h (Figure 3D, filled arrow). Notably, preincubation with LMB followed by cytoplasmic microinjection of Statl did not cause its nuclear accumulation (Figure 3D, open arrow). These results show that LMB reduces the constitutive nuclear export of Statl in unstimulated cells. We have shown in Figure 2F that removal of N- and C-domains did not influence the nuclear import of Statl . Contrary, nuclear export of the truncated mutant Statltc was strongly diminished, since it took about four hours to achieve an even nucleocytoplasmic distribution (Figure 3E, F), and treatment with LMB reduced the export rate even further (Table I). Similar results were obtained also for truncated Stat3 and Statδ (Table I). Moreover, fluorescently labeled full length Statl was not exported from the nucleus even after 2 h (Figure 3G).
The constitutive nuclear import of recombinant Statl continues in energy- depleted cells. Another set of microinjection experiments was performed in living cells that were depleted of ATP by the addition of sodium azide and 2- deoxyglucose. Such treatment was shown to reversibly inhibit classical Ran- dependent nuclear transport by limiting the pool of GTP-bound Ran. In order to demonstrate the energy-depleted status of the microinjected cells, we used different injection markers. Here, fusion proteins of glutathione S-transferase (GST) and green fluorescent protein (GFP) that included at their domain junction either a triple SV40 NLS or a Statl -derived NES (termed GST-NLS- GFP or GST-NES-GFP, respectively) were co-microinjected with recombinant Statl. The effective nucleocytoplasmic translocation of the reporter constructs was confirmed in cells growing in full-medium, whereas pre-exposure of cells to azide and 2-deoxyglucose for 2 h prevented the carrier-dependent nuclear translocation of the reporter protein (Figures 4 and δE, F). Expectedly, the nuclear import of microinjected tyrosine-phosphorylated Statlwt and Statltc was also prevented in energy-depleted cells (shown for Statltc in Figure 4A). However, nuclear import of unphosphorylated Statltc (Figure 4B) and wild type (Figure 4C) continued under this condition, albeit at a reduced pace, as it now took about 30 min to reach a pancellular distribution after injection into the cytoplasm. Similarly, nuclear import of recombinant Stat3tc and Statδtc also continued in energy-depleted cells (Figure 4D, E). Notably, the predominantly nuclear localization of Stat3tc seen in cells growing in normal medium was not maintained in ATP-depleted cells (compare Figures 4D and 2G). Next, the nuclear export of recombinant wild type Statl and Statltc in energy- depleted cells was examined, and it was found that energy depletion blocked the export from the nucleus of both proteins during a 3 h observation period (shown in Figure 4F for Statlwt; compare with Figure 3A, B).
Endogenous Statl is constitutively shuttling across the nuclear envelope independently of metabolic energy. A different experimental approach was used to investigate the nucleocytoplasmic flux rates also of endogenous Statl . To this end we performed microinjections of Statl antibodies, which has been shown to immobilize the shuttling target antigen and hence cause its accumulation in the microinjected compartment. The results are assembled in Figure 5 and Table I. As is shown in panel A of Figure 5, maximal nuclear accumulation of unphosphorylated Statl was seen already 15 min after antibody microinjection into the nucleus. Next, cytoplasmic antibody microinjections were used to estimate the nuclear export rate of the endogenous Statl protein. Depletion of nuclear Statl was observable as early as δ min post injection, and maximal depletion was achieved with this technique already after 15 min (Figure δB). This assay was also used to determine the inhibitory influence of LMB on the nuclear export of endogenous Statl. However, the antibody microinjection assay did not allow us to record differences, as the nuclear export rate was not measurably diminished in the presence of LMB (Figure δC). Incubation of the cells at 4PC, however, precluded nuclear export of endogenous Statl (Figure δD). Nuclear import of the endogenous Statl continued also during ATP-depletion, as revealed after nuclear injection of a Statl antibody. Already 1δ min past antibody microinjection nuclear accumulation was observable, which was maximal after 30 min (Figure δE). We then explored the nuclear export of endogenous Statl under these conditions. This was achieved by cytoplasmic co-microinjection of a Statl antibody and the GST-NLS-GFP reporter (Figure δF). Clearly, nuclear export of endogenous Statl continued in azide/2- deoxyglucose-treated cells, as the nuclear compartment was depleted of Statl immunoreactivity after cytoplasmic antibody injection. Energy depletion reduced the transport rate by about δ0%, as the time required to clear the nucleus of Statl had doubled to 30 min. Together, these results indicated that nucleocytoplasmic shuttling of Statl continued in the absence of a physiological Ran-GTP pool.
Exclusively unphosphorylated Stats enter the nucleus of digitonin-permeabilized cells in the absence of cytosol. The behavior of Stat proteins was investigated further with a permeabilized cell transport assay. HeLa cells were incubated with a concentration of digitonin (40 μg/ml) that selectively permeabilizes the plasma membrane, thereby releasing cytoplasmic proteins. An extensive validation of this assay was performed with the carrier-dependent import substrate GFP-NLS-GST. As expected, nuclear import required the addition of cytosol and metabolic energy (see Supplemental Figure 1). A similar analysis was also performed for unphosphorylated Statl . As is shown in Figure 6A, Statltc entered the nucleus in the presence of cytosol and metabolic energy (+CIM, complete import mix). However, nuclear import was even enhanced in the absence of added exogenous cytosol (Figure 6A, +IM, import mix). Pre- treatment of digitonin-permeabilized cells with apyrase to further reduce residual ATP in the absence of added cytosol was without effect on the import of unphosphorylated Statltc (Figure 6A, ÷apyrase), in contrast to tyrosine- phosphorylated Statltc, which was incapable to enter the nucleus in the absence of added cytosol (Figure 6B).
Several conditions were found to prevent nuclear import of unphosphorylated Statltc in the absence of added cytosol. First, the addition of WGA strongly reduced Statl nuclear uptake (Figure 6A; +WGA), as did incubation of the permeabilized cells at 4° C (Figure 6A; 4° C). To further characterize cytosol- independent nuclear translocation, we examined the saturability of the import process. For this experiment, an N- and C-terminally truncated fragment of the import receptor p97 was added to the cytosol-free import reaction. This mutant binds irreversibly to proteins of the nuclear pore and thus obstructs multiple import pathways in a dominant fashion. As is shown in Figure 6A (+p97tc), the inclusion of a 16-fold molar excess of the inhibitory p97 fragment (residues 45- 462) completely abrogated nuclear import of Statltc. The same result was obtained after adding a 20-fold molar excess of wild type Statl to the import reaction (Figure 6A; +1wt). Cytosol-free import assays with digitonin-permeabilized HeLa cells were succesfully performed also with Stat3tc (Figure 6C). Since both Stat proteins could enter the nucleus by a carrier-free mechanism, we tested whether they also compete for the same NPC binding sites. This was done by adding a 20- fold molar excess of wild type Statl to the Stat3tc import reaction, which resulted in a strong suppression of Stat3tc nuclear import (Figure 6C; +1wt). Similarly, addition of a 16-fold molar excess of truncated p97 also reduced nuclear import of Stat3tc, as did pre-incubation with WGA (Figure 6C; labeled +p97tc and +WGA, respectively).
To estimate the nuclear entry rates of Statlwt and truncated Statl and Stat3 in digitonin-permeabilized cells, a time-course experiment was performed. As is shown in Figure 6D-F, all three proteins entered the nucleus with similar kinetics. Already after 5 min the Stat proteins were detectable at the nuclear rim and inside some nuclei, and after 10 min the Stats had accumulated in most of the nuclei. The accumulation phase continued and reached a plateau between 30 and 60 min. Interestingly, we noted that the import rate of Statl and Stat3 differed among individual nuclei (Figure 6D-F). Incorrect cell permeabilization leading to rupturing of the nuclear membrane is an unlikely explanation for this, since staining of digitonin-permeabilized cells with labeled concanavalin-A was limited to the cell membrane. Moreover, the cytosol-free nuclear import of p97 did not differ among individual nuclei of permeabilized cells (see Supplemental Figure 1B). Of note, fluorescent labeling increased both the transport rate and the homogeneity of Statl nuclear import (shown for Statltc in Figure 6G). We also attempted to establish cytosol-free nuclear import for Statδtc. While about 10% of the nuclei showed nuclear import of Statδtc, the remaining cells displayed strong labeling in the extranuclear space of what appeared to constitute precipitated Statδtc protein.
These results indicate that Statl, Stat3, and Statδ have the ability to migrate into the nucleus by themselves in the absence of metabolic energy or added transport factors. They share binding partners in the NPC among each other and with the import receptor p97.
Statl binds to nucleoporin Nup1δ3 and Nup214, but not Nup62. The above data are not compatible with a carrier-mediated nuclear translocation process. Rather, they are characteristic for a carrier-free mechanism that occurs through direct interactions between the Stats and constituents of the nuclear pore. A distinctive feature of numerous NPC proteins is the presence of Phenylalanine/Glycine (FG)-rich repeat motifs, which provide interaction sites for transport factors. Since Stats and p97 appeared to share binding sites on nuclear pore proteins, we examined whether Statl could also interact with constituents of the nuclear pore. FG repeat-containing parts of His-tagged Nup214 (residues 1649-2090), Nup62 (residues 1-308), and Nup153 (residues 333-618; the latter two were fusion proteins with maltose-binding-protein, MBP) were expressed in bacteria and detected by immunoblotting with the monoclonal antibody 414 (Supplemental Figure 2). The bacterial lysates were resolved by denaturing SDS-PAGE (Figure 7, lower panel) and subsequently blotted on nitrocellulose. After extensive incubation in renaturing buffer, the immobilized proteins were incubated with wild type Statl, the binding of which was detected by Western blotting (Figure 7, upper panel). No binding of Statl was seen with MBP and a His-tagged control protein, but Statl bound strongly to both Nup214 and Nup153. Contrary, no binding was detected to the FG repeat region of Nup62. These results suggested that Statl is able to bind directly to the FG repeat region of Nup1δ3 and Nup214, supporting a model in which unphosphorylated Statl engages in carrier-free nucleocytoplasmic shuttling through direct interactions with components of the nuclear pore.
The linker domain of Statl plays a role in cytokine-independent nuclear translocation. Statl contains highly reactive cysteine residues which can cause aggregation of recombinant purified protein. This problem can be overcome by blocking reactive SH-groups with N-ethylmaleimide (NEM), which has been shown to leave in vitro DNA binding undisturbed, and this treatment also did not influence nuclear import of full length Statl . However, when truncated Statl was alkylated and microinjected into cells, we could no longer detect nucleocytoplasmic shuttling, as both import and export were strongly diminished (Figure 8A). Mutation of eight out of the nine cysteine residues of human Statltc prevented NEM from inhibiting nuclear import (see summary in Figure 8D), indicating that cysteine alkylation caused the inhibitory effect. Further experiments unambigously demonstrated that mutation of Cysδ43, which is located in the linker domain, was responsible for rescuing Statl from the shuttling defects caused by NEM (Figure 8B). Mutation of other cysteine residues did not prevent NEM from inhibiting nuclear translocation of Statl (shown for Cys492 in Figure 8C). Mass spectrometry confirmed alkylation of recombinant Statltc in position 8 (Cysδ43), and also of positions 5 (Cys324) and 9 (Cys577) (see Figure 8D). Interestingly, mutation to alanine or arginine of residue 543 was without adverse effects on nuclear import.
Carrier-dependent and -independent pathways cooperatively determine the subcellular distribution of Statl in resting cells. It is currently unknown how the Stats achieve their predominantly cytoplasmic localization in unstimulated cells. As is shown above, before stimulation with cytokines Statl enters the nucleus independent of metabolic energy and transport factors. Export of Statl from the nucleus, however, can occur via two pathways, only one of which functions without metabolic energy, whereas the second pathway requires an NES, the exportin CRM1 and metabolic energy. Therefore we used energy-depletion and employed the specific CRM1 inhibitor LMB to suppress the NES-dependent active nuclear export of Statl . As is shown in Figures 9A, exposure of 3T3 cells to energy depletion medium for 2 h reduced the cytoplasmic accumulation of Statl , and a statistically significant relocation to the nucleus resulting in a pancellular distribution was observed (Figure 9B). Expectedly, the nuclear relocation of Statl was reversible, since the subsequent incubation in full growth medium for 5 h restored the cytoplasmic accumulation of Statl (Figure 9A). Treatment of cells with the CRM1 inhibitor LMB also caused relocalization
• and pancellular distribution of Statl . Importantly, prolonged exposure to LMB for 5 h had the same effects and did not induce nuclear accumulation (Figure 9A, B). In the present study we have investigated both in vitro and in vivo the nucleocytoplasmic translocation of Stats before their activation. The results shown here reveal that unphosphorylated Statl, Stat3 and Statδ are shuttling proteins that rapidly traverse the nuclear envelope. Several lines of evidence indicated that this is a process distinct from the conventional import mechanism described for tyrosine-phosphorylated Statl (Figure 10). 1) All transport processes known to date that depend upon transport factors rely on metabolic energy in vivo. Nuclear translocation of unphosphorylated Statl , Stat3, and Statδ, however, continued in energy-depleted cells, whereas the translocation of tyrosine phosphorylated Statl or of a transport factor-dependent reporter construct did not (Figure 4, δE, F). 2) Nuclear import of un hosphorylated Statl and Stat3 was readily observable in digitonin-permeabilized cells in the absence of added cytosol (Figure 6A, C). Contrary, tyrosine-phosphorylated Statl or a p97-dependent reporter construct could not enter the nucleus in this system (Figure 6B and Supplemental Figure 1A). 3) Unphosphorylated Statl was demonstrated to directly interact with the FG repeat region of Nup1δ3 and Nup214, but not Nup62 (Figure 7). These results are compatible with a model describing nucleocytoplasmic translocation of unphosphorylated Stats as a carrier-independent process that relies on direct interactions between Stat proteins and the nuclear pore (Figure 10). The carrier-free nuclear import of the Stats is rapid and saturable. Nuclear import in digitonin-permeabilized cells or after cytoplasmic microinjection was detectable already after 5 min, and trapping of endogenous Statl by antibody microinjection in the nucleus of resting HeLa cells resulted in nuclear accumulation after about 15 min. Contrary to the results presented here for wild type Statl , examination of GFP-tagged Statl with FLIP (Fluorescence Loss in Photobleaching) indicates that the nuclear and cytoplasmic pools do not exchange rapidly. However, in unstimulated cells GFP-tagged Statl traverses the nuclear membrane much less efficiently in comparison to the wild type (manuscript in preparation). Active nuclear export has been demonstrated to participate in the termination of the interferon-induced nuclear accumulation of Statl . Here, we extend these data and show that both active, CRM1 -dependent and carrier-free nuclear export occur simultaneously regardless of cytokine stimulation, as indicated by the protracted export of nuclear-microinjected Statl during LMB-treatment (Figure 3C, D). Several observations suggested that nuclear import of unphosphorylated Statl, on the other hand, is a carrier-free process. Namely, the cytoplasmic microinjection of fluorescently labeled Statl that cannot leave the nucleus did not result in nuclear accumulation, but import ceased when a pancellular distribution was achieved (Figure 21). Similarly, the specific inactivation of CRM1 -mediated export did not cause nuclear accumulation either (Figure 3D). Both results argue against constitutive active nuclear import. In addition, no evidence for a functional NLS was found in unphosphorylated Statl . We therefore propose a model in which diffusion-controlled import and energy-consuming export contribute to the observed accumulation of Statl in the cytoplasm of unstimulated cells. In line with this model we found that the cytoplasmic accumulation of Statl was significantly diminished upon ATP- depletion or exposure of resting cells to the CRM1 inhibitor LMB (Figure 9). Similar observations were previously made also for Stat2, Stat3 and Statδ. We thus conclude that the influx rate of Statl into the nuclei of unstimulated cells is influenced by its concentration gradient between cytosol and nucleoplasm. Given the active CRM1 -dependent nuclear export, the steady-state distribution of microinjected Statl should be predominantly cytoplasmic. However, we observed a pancellular distribution at equilibrium. This could be explained if CRM1 played only a minor role in comparison to the transport capacity of carrier-free translocation. The following observations support such an assumption. First, the cytoplasmic accumulation of Statl is not very pronounced in most cell lines. Moreover, nuclear export of endogenous Statl both before (Figure 5C) and during interferon treatment of cells was not measurably decreased by LMB, which certainly reflects also the limitations of our microinjection assay. And third, LMB prolonged nuclear accumulation of Statl only modestly. However, the Stat proteins are likely to differ in the extent to which these pathways determine their overall transport rate. Contrary to Statl , a constitutive carrier-dependent import signal was described for Stat3. Accordingly, the cytoplasmic microinjection of Stat3tc resulted in its nuclear accumulation (Figure 2G), but an even nucleocytoplasmic distribution resulted if the cells were cultured in energy-depleted medium (Figure 4D). The structural requirements that enable the nucleocytoplasmic shuttling of unphosphorylated Stats are likely to be complex. Similar to observations that
• were previously made with importin p97, where alkylation with NEM precluded binding to the nuclear pore, also the translocation of Statl was sensitive to NEM. Alkylation of a single variant cysteine residue in the linker domain precluded nuclear translocation and thus implicated this rather non-descript region of Statl in nuclear transport. Further work is also necessary to explore the role of the Stat N- and C-domains, which were dispensable for nuclear import, but the presence of which accelerated nuclear export. These observations as well as the competition data, which indicated that the Stat proteins share NPC-binding sjtes among themselves and with p97 (Figure 6A, C) raise the issue of, specificity in carrier-free protein translocation. Besides the truncation of Stats several other conditions affected transport in a direction- specific manner. Energy depletion and fluorescent labeling specifically blocked the nuclear export of Statl Also, for unknown reasons the import rates of both Statl and -3 differed among individual nuclei of digitonin permeabilized cells. Addition of ATP, which remedied a similar problem in carrier-free nuclear translocation of importin-α, or using recombinant protein from insect cells instead of bacteria had no effect on nuclear import. However, labeling of Statl with fluorescent dyes, while preventing export (Figure 3G), made nuclear import more homogenous (Figure 6G). Of note, several experiments indicated that the shuttling rate of endogenous Statl exceeded that of the microinjected recombinant protein (Table I). Therefore, it is conceivable that posttranslational modifications influence the translocation through the nuclear pore, possibly even in a direction-specific manner. Such mechanisms could also account for cell-type or Stat-specific differences in the subcellular distribution of members of this protein family.
In summary, our results indicate that a dynamic equilibrium of constitutive nuclear export and import maintains the steady-state distribution of Stats both before and after cytokine stimulation of cells. Thus, in light of the constant cycling that characterizes the Stat transcription factors, the cytokine signal transduction is best described as a "continous signaling cycle" rather than a linear pathway that starts at the cell membrane and ends in the nucleus.
Figure 1. Recombinant Stat proteins. (A) SDS-PAGE (7%) analysis of the Stat protein preparations used in this work (1 μg of each). Molecular size markers are indicated to the left. OG488, Oregon Green 488. (B) Schematic representation of the respective Stat proteins. The truncated mutants were expressed with a C-terminal Strep-tag of the sequence SAWSHPQFEK (single letter code).The Stat proteins were of human (Statl), mouse (Stat3), or sheep (Statδ) origin. N, N domain; CC, coiled coil domain; DNA, DNA binding domain; L, linker domain; SH2, SH2 domain; TAD, transactivation domain.
Figure 2. Unphosphorylated recombinant Stat proteins translocate into the nucleus of unstimulated cells. HeLa-S3 cells (except for panels B and C) were used. The injection site is indicated by the location of co-microinjected FITC- labeled or Tetramethyl-rhodamine (TMR)-labeled BSA. Nuclei were stained with Hoechst dye. Arrows indicate injected cells. (A-C) Cytoplasmic microinjection of purified Statlwt into Hela cells (A), COS7 cells (B), or 2fTGH cells (C). After 15 min the cells were fixed and Statl was detected by immunocytochemistry with a specific antibody. (D) Identical experimental setup as in (A), but microinjected cells were fixed after 60 min (E-H). Cytoplasmic microinjection of full length tyrosine mutant Y701F (E), or truncated Statltc (F), Stat3tc (G), or Statδtc (H). Fixation was 15 min post injection, truncated Stats were detected with Strep-tag antibody. (I) Cytoplasmic microinjection of Oregon Green 488-labeled Statltc. Cells were fixed after 15 min followed by direct fluorescence microscopy. (J, K) Cytoplasmic co-micrdinjection of WGA and Statlwt (J) or Statltc (K). After 1 h the cells were fixed and Statl variant proteins were detected by immunocytochemistry as described. Bar, 20 μm.
Figure 3. Unphosphorylated recombinant Stat proteins exit the nucleus of unstimulated cells. The injection sites of the HeLa cells are indicated by co- microinjected FITC-labeled BSA. (A, B) Recombinant Statlwt was injected into the nucleus. After 1 h (A) or 3 h (B) immunocytochemistry with a Statl -specific antibody was performed on fixed cells. (C, D) Microinjection of Statlwt into cells pretreated for 1 h with LMB. After nuclear (filled arrow) or cytoplasmic (open arrow) microinjection the cells were further incubated with LMB for 1 h (C) or 4 h (D) before fixation and immunocytochemistry with a Statl -specific antibody. (E, F) Statltc was injected into the nucleus and the cells were subsequently incubated for 1 h (E) or 4 h (F), before fixation and immunocytochemistry with Strep-tag antibody. (G) Nuclear microinjection of Alexa Fluor 594-labeled Statlwt, followed by incubation for 2 h, fixation, and direct fluorescence microscopy. Bar, 20 μm.
Figure 4. The nucleocytoplasmic translocation of recombinant Stat proteins continues in energy-depleted cells. HeLa cells were kept in energy depletion medium for 2 h before microinjection (EDM). (A-E) Cytoplasmic microinjection of tyrosine-phosphorylated Statltc (A), or unphosphorylated Statltc (B), Statlwt (C), Stat3tc (D), or Statδtc (E). Injection site and energy depletion are indicated by the location of co-microinjected GST-NLS-GFP. Tine cells were fixed at the indicated times after injection and Stats were detected by immunocytochemistry (see Figure 2). (F) Nuclear co-microinjection of recombinant Statlwt and GST- NES-GFP, followed by 3 h of incubation in EDM. Then , cells were processed for immunocytochemistry as before. Bar, 20 μm.
Figure δ. Nucleocytoplasmic translocation of endogenous Statl in normal and energy-depleted HeLa cells. Statl antibodies were microinjected followed by immunocytochemical detection of Statl . (A) Microinjaction of Statl antibodies into the cell nucleus with fixation after 15 min. The injection site is marked by co-microinjected FITC-BSA. (B) Identical with (A), except that the antibodies were microinjected into the cytoplasm. (C) Identical wit-h (B) except that the cells were incubated with 5 ng/ml LMB starting 1 h before microinjection. (D) Identical with (B), except that the cells were kept at 4°C for 60 min both before and after cytoplasmic microinjection. (E) Statl antibodies were microinjected into the nucleus of cells after a 2 h preincubation in EDM. Injection site and energy depletion are indicated by co-microinjected GST-NES-GFP. The cells were fixed 30 min after microinjection. (F) Identical with (E), except that the Statl antibody was microinjected into the cytoplasm. Injection site and energy depletion are indicated by co-microinjected GST-NLS-GFP. Bar, 20 μm.
Figure 6. The nuclear import of Stat proteins continues in cytosol-depleted digitonin-permeabilized cells. HeLa-S3 cells were treated with 40 μg/ml digitonin, washed, and then incubated with 20 μl of import mix (IM; protein-free transport buffer supplemented with 10 mg/ml BSA) at RT unless noted otherwise. The nuclei are stained with Hoechst dye. Unlabeled Stat proteins were detected immunocytochemically in fixed cells. (A) Permeabilized HeLa cells were incubated for 60 min with 1 μM unphosphorylated Statltc in IM. Additionally, the import reaction was performed on ice (4°C), or after preincubation with 250 μg/ml WGA (+WGA). Where indicated, the following additions were made to IM. Rabbit reticulocyte lysate (75%) and an energy regeneration system (+CIM); 0.8 U/ml apyrase (+apyrase); 16 μM truncated p97 (residues 45-462) fused to GST (+p97tc); 20 μM Statlwt (+1wt). (B) Import reaction with tyrosine-phosphorylated Statltc in IM. (C) Identical experimental setup as in (A), but with Stat3tc replacing Statltc. (D-G) Time-course of carrier- free nuclear import of Statlwt (D), Statltc (E), Stat3tc (F), or Oregon Green 488-labeled Statltc (G). The import mix contained 1μM of the respective Stat proteins in IM. In (G) Statltc was detected by direct fluorescence microscopy. Note the different time scale. Bar, 30 μm.
Figure 7. Statl interacts with FG repeat-containing nucleoporins. (Upper panel) Bacterial lysates containing the indicated fusion proteins (lanes 1 -4) or purified His-tagged tandem SH3 domains from FYB (lane 5) were resolved by SDS- PAGE, transferred to a nitrocellulose membrane, and probed with wild type unphosphorylated Statl The association of Statl with the immobilized proteins was detected by immunoblotting using a Statl specific antibody. The lower panel shows the respective Coomassie-blue stained SDS-gel before transfer to a nitrocellulose membrane (input).
Figure 8. Alkylation of cysteine 643 in the linker domain precludes nuclear import of Statl (A-C) Purified Statltc or the indicated mutants were alkylated by NEM in vitro and injected in the cytosol of HeLa cells. After 1 h the cells were fixed and the intracellular distribution of the microinjected proteins was determined by immunocytochemistry. Bar, 20 μm. (D) Summary of the microinjection data and mass spectrometric analyses of alkylated Statl . Purified bacterially expressed Statltc was treated without or with NEM and subsequently injected into the cytoplasm of HeLa cells. The ability to enter the nucleus during an 1 h incubation period is stated. The cysteine residues in position 155, 174, 247, 25δ, 324, 440, 492, 543, 677 of human Statl are numbered 1 to 9 in the diagram to the left. Mutated cysteines (Cys to Ala) are blackened. Open circles denote absence of alkylation, filled circles denote alkylated cysteines as determined by mass spectrometry. For position 7 both alkylated and non-alkylated peptides were found. Peptides covering Cys- positions 3, 4, and 6 were not detected
Figure 9. The inhibition of active nuclear export prevents cytoplasmic accumulation of Statl . (A) 3T3 cells were left untreated (DMEM) or treated with energy depletion medium for 2 h (EDM) followed by growth medium for 5 h (EDM+DMEM). Alternatively, cells were treated with the CRM1 inhibitor leptomycin B (LMB) for 2 h or 5 h as indicated. Shown is the distribution of the endogenous Statl as revealed by immunocytochemistry with a specific antibody and conventional microscopy. (B) Corresponding bar diagram with a quantitative analysis of the ratio of the mean cytoplasmic and nuclear immunofluorescence densities. The median slice (x/y image) of a confocal microscopical image was used to quantify the signals in 13 randomly chosen cells. Statistically significant differences between cells incubating in control (DMEM) or experimental medium are indicated by (*). Bar, 20 nm.
Figure 10. A model of Statl nucleocytoplasmic shuttling. (A) Unphosphorylated "latent" Statl constitutively shuttles between cytosol and nucleoplasm via direct interactions with the nucleoporins Nup153 and Nup214. The surface of the linker domain of Statl is likely to provide the contact surface. (B) Additionally, NES-mediated transport of unphosphorylated Statl via CRM1 enhances the export rate and achieves cytoplasmic accumulation. (C) After cytokine-induced receptor activation, Statl is tyrosine-phosphorylated and dimerizes, which precludes further carrier-free nucleocytoplasmic cycling. However, a dimer- specific NLS in the DNA binding domain is exposed, and nuclear import occurs in complex with NPI-1 and p97. (D) Until its dephosphorylation, which is inhibited by DNA-binding, Statl is retained in the nucleus, thus allowing for the signal-induced nuclear accumulation. GAS, Statl binding site.
Figure 11. RT-PCR analysis of endogenous INFα- and IFNγ-responsive genes.
Figure 12. IL-2-reconstituted 293T cells are co-transfected with Statδ wild type or mutant StatδaF81A or Stat5bF81A and a luciferase reporter gene. Stimulation without (gray) or with IL-2 is overnight (black). Luciferase expression is activated from a natural PRRIII promoter or an M10 element. The diagrams represent the ground expression level which is not affected by the substance library.
Online Supplemental Figure SI Control experiments in digitonin-permeabilized HeLa cells with a carrier-dependent control protein or p97. (A) shows that a carrier-dependent control protein requires cytosol for nuclear import into digitonin-permeabilized HeLa cells. Cells were incubated with digitonin (40 μg/ml) in transport buffer (TB) for 6 min on ice. After washing with TB the cells were incubated for 60 min at RT with GST-NLS-GFP control protein (0.8 μM) dissolved in 20 μl IM or complete import mix (CIM: IM supplemented with reticulocyte lysate (76% of volume) and an energy regeneration system (O.δ mM ATP, O.δ mM GTP, 10 mM creatine phosphate, 30 U/ml creatine phosphokinase). Where indicated, the reaction (with CIM) was performed at 4°C (4°C). Additionally, CIM was supplemented with O.δ mg/ml WGA (+WGA); or with 16 μM p97 (aa 45-462 of p97) (+p97tc), or with 0.8 U/ml apyrase (+apyrase). In (B) the cytosol-independent nuclear import of p97 is demonstrated. Import reaction without or with 0.8 μM His-tagged importin-β (p97) dissolved in IM. Where indicated, IM was supplemented with WGA at a concentration of 0.1 mg/ml (+WGA/100), or 0.25 mg/ml (+WGA/2δO). An anti- His antibody was used to detect p97. Bar, 30 μm.
Online Supplemental Figure S2. Immunoblotting results with the FG repeat- specific antibody 414 and bacterial lysates containing recombinant nucleoporins. Identification of bacterially expressed nucleoporin fusion proteins. Bacterial lysates containing maltose binding protein (MBP, lane 1) or FG repeat regions of Nup214 (residues 1649-2090, His-tagged), Nup62 (residues 1-308 fused to MBP), Nup1δ3 (residues 333-618 fused to MBP) (lanes 2-4), or purified His-tagged tandem SH3 domains from FYB (lane δ) were resolved by 10% SDS-PAGE, transferred to a nitrocellulose membrane, and probed with FG repeat-specific monoclonal antibody 414 (Covance). Protein loading was 0.2% of the input used in Figure 7. Table I. Nucleocytoplasmic translocation of unphosphorylated Stat proteins as determined by microinjection. a) Microinjection of recombinant Stat proteins. Listed is the amount of time required to reach equilibrium (min or h). b) Microinjection of Statl antibodies. Listed are the times required to observe maximal precipitation of endogenous Statl in the injected compartment. Each entry represents an averaged value observed by immunocytochemistry in 15-20 microinjected cells after fixation. GM, growth medium with 10% serum; EDM, serum- and glucose-free energy depletion medium with azide and deoxyglucose; n.a., not applicable; n.d., not done; AF, Alexa Fluor 594; OG, Oregon Green 488.

Claims

Claims:
1. Stat molecule occurring in the nucleus of a cutaneous T cell lymphoma cell line characterized in that at least one conserved hydrophobic amino acid on the surface of the N-domain of the Stat wild type is mutated.
2. Stat molecule according to claim 1 , characterized in that the wild type of Stat2, Stat3, Statδ or Stat6 is mutated.
3. Stat molecule according to claim 1 or 2, characterized in that the amino acids of the N-domain in position 81 and/or position 82 of Statδ or in equivalent positions of homologous Stat proteins are mutated.
4. Stat molecule according to claim 3, characterized in that the arhino acid in position 81 is substituted by alanine and/or the amino acid in position 82 is substituted by serine.
δ. Stat molecules according to one of claims 1 to 4 which are dimerized.
6. A fusion protein which includes the Stat molecule according to one of claims 1 to 4 in combination with at least one marker structure.
7. A nucleic acid sequence which encodes the Stat molecule according to one of claims 1 to 4, or the fusion protein according to claim 6.
8. A vector, including the nucleic acid sequence according to claim 7.
9. A host cell, including the vector according to claim 8.
10. Use of the Stat molecule according to one of claims 1 to δ, the fusion protein according to claim 6, the nucleic acid sequence according to claim 7, the vector according to claim 8 or the host cell according to claim 9 - for expression of a target protein; or - for investigating effects caused by different gene induction levels; or - for screening of active substances which modify the transcription activity of target genes; or - for the identification of binding partners; or - for the production of a drug for the diagnosis and/or treatment of 5 diseases associated with abnormal Stat induction of target genes.
11. Use of a Stat molecule derived from wild type Statl , which is mutated in at least one conserved hydrophobic amino acid on the surface of the N- domain,0 - for expression of a target protein; or - for investigating effects caused by different gene induction levels; or - for screening of active substances which modify the transcription activity of target genes; or - for the identification of binding partners; or5 - for the production of a drug for the diagnosis and/or treatment of diseases associated with abnormal Stat induction of target genes.
12. Method for overexpression of a target protein comprising the following steps:0 - introduction of a vector according to claim 8 into a host cell or a cell-free system; - introduction of a second vector harboring at least a single optimal GAS sequence followed by a target gene into a host cell or a cell-free system;5 - induction of gene expressions; and - maintenance of gene expressions over an appropriate period.
13. Method for investigating effects caused by different gene induction levels comprising the following steps: o - introduction of a vector according to claim 8 into a host cell; - introduction of a second vector harboring a promoter sequence followed by a target gene into a host cell; - induction of gene expressions; - maintenance of gene expressions over an appropriate period; and - monitoring effects on the host cell according to claim 9 by an appropriate detection procedure.
14. Method for screening of active substances which modify the transcription activity of target genes comprising the following steps: - introduction of a vector according to claim 8 into a host cell or a cell-free system; - introduction of a second vector harboring a promoter sequence followed by a target gene into a host cell or a cell-free system; - induction of gene expressions; - addition of a substance library; - maintenance of the gene expressions over an appropriate period; and - monitoring effects on the host cell according to claim 9 or the cell-free system by an appropriate detection procedure.
16. Method for the production of a drug for the diagnosis and/or treatment of diseases associated with abnormal Stat induction of target genes comprising the following steps: - withdrawal of a tissue sample; - cell disruption; - separation of the intracellular protein extract; - probing the protein extract with a high affinity molecule selectively recognizing the Stat mutant; - gene therapy.
16. A kit comprising a Stat molecule according to one of claims 1 to 6, and/or synthetic analogues, modifications and pharmacologically active fragments thereof and an information about the using of the kit.
17. A kit comprising a nucleic acid sequence according to claim 7, and/or synthetic analogues, modifications and pharmacologically active fragments thereof and an information about the using of the kit.
PCT/EP2005/001462 2004-02-10 2005-02-10 Hyperactive stat molecules and their use in assays employing gene activation WO2005075508A2 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
EP04090039.1 2004-02-10
EP04090039 2004-02-10
EP04090242.1 2004-06-18
EP04090242 2004-06-18
EP04077629A EP1637540A1 (en) 2004-09-17 2004-09-17 Hyperactive Stat molecules and their use in assays employing gene activation
EP04077629.6 2004-09-17

Publications (3)

Publication Number Publication Date
WO2005075508A2 true WO2005075508A2 (en) 2005-08-18
WO2005075508A8 WO2005075508A8 (en) 2005-10-27
WO2005075508A3 WO2005075508A3 (en) 2006-07-20

Family

ID=34841403

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2005/001462 WO2005075508A2 (en) 2004-02-10 2005-02-10 Hyperactive stat molecules and their use in assays employing gene activation

Country Status (1)

Country Link
WO (1) WO2005075508A2 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001096560A1 (en) * 2000-05-09 2001-12-20 Shanghai Biowindow Gene Development Inc. A novel polypeptide, a human protein stat2 and the polynucleotide encoding the polypeptide
US20040009571A1 (en) * 1998-01-23 2004-01-15 John Kuriyan Dimerization interface of signal transducer and activator of transcription (STAT) proteins
WO2004048524A2 (en) * 2002-11-23 2004-06-10 Isis Pharmaceuticals, Inc. Modulation of stat2 expression

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040009571A1 (en) * 1998-01-23 2004-01-15 John Kuriyan Dimerization interface of signal transducer and activator of transcription (STAT) proteins
WO2001096560A1 (en) * 2000-05-09 2001-12-20 Shanghai Biowindow Gene Development Inc. A novel polypeptide, a human protein stat2 and the polynucleotide encoding the polypeptide
WO2004048524A2 (en) * 2002-11-23 2004-06-10 Isis Pharmaceuticals, Inc. Modulation of stat2 expression

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
CHEN W ET AL: "Turning off signal transducer and activator of transcription (STAT): The negative regulation of STAT signaling" JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY, MOSBY - YEARLY BOOK, INC, US, vol. 114, no. 3, September 2004 (2004-09), pages 476-489, XP004660844 ISSN: 0091-6749 *
CHEN XIAOMIN ET AL: "A reinterpretation of the dimerization interface of the N-terminal domains of STATs." PROTEIN SCIENCE : A PUBLICATION OF THE PROTEIN SOCIETY. FEB 2003, vol. 12, no. 2, February 2003 (2003-02), pages 361-365, XP002351562 ISSN: 0961-8368 *
HORVATH C M ET AL: "A STAT PROTEIN DOMAIN THAT DETERMINES DNA SEQUENCE RECOGNITION SUGGESTS A NOVEL DNA-BINDING DOMAIN" GENES AND DEVELOPMENT, COLD SPRING HARBOR, NY, US, vol. 9, no. 8, 15 April 1995 (1995-04-15), pages 984-994, XP000617031 ISSN: 0890-9369 *
MARG ANDREAS ET AL: "Nucleocytoplasmic shuttling by nucleoporins Nup153 and Nup214 and CRM1-dependent nuclear export control the subcellular distribution of latent Stat1." THE JOURNAL OF CELL BIOLOGY. 21 JUN 2004, vol. 165, no. 6, 21 June 2004 (2004-06-21), pages 823-833, XP002319835 ISSN: 0021-9525 *
MEYER THOMAS ET AL: "A single residue modulates tyrosine dephosphorylation, oligomerization, and nuclear accumulation of stat transcription factors." THE JOURNAL OF BIOLOGICAL CHEMISTRY. 30 APR 2004, vol. 279, no. 18, 30 April 2004 (2004-04-30), pages 18998-19007, XP002319833 ISSN: 0021-9258 *
MEYER THOMAS ET AL: "Nucleocytoplasmic shuttling of STAT transcription factors." EUROPEAN JOURNAL OF BIOCHEMISTRY / FEBS. DEC 2004, vol. 271, no. 23-24, December 2004 (2004-12), pages 4606-4612, XP002319838 ISSN: 0014-2956 *
OTA NARUHISA ET AL: "N-domain-dependent nonphosphorylated STAT4 dimers required for cytokine-driven activation." NATURE IMMUNOLOGY. FEB 2004, vol. 5, no. 2, February 2004 (2004-02), pages 208-215, XP002319834 ISSN: 1529-2908 *
STREHLOW I ET AL: "Amino-terminal signal transducer and activator of transcription (STAT) domains regulate nuclear translocation and STAT deactivation." THE JOURNAL OF BIOLOGICAL CHEMISTRY. 23 OCT 1998, vol. 273, no. 43, 23 October 1998 (1998-10-23), pages 28049-28056, XP002319836 ISSN: 0021-9258 *
ZHANG J J ET AL: "Two contact regions between Stat1 and CBP/p300 in interferon gamma signaling." PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA. 24 DEC 1996, vol. 93, no. 26, 24 December 1996 (1996-12-24), pages 15092-15096, XP002319837 ISSN: 0027-8424 *

Also Published As

Publication number Publication date
WO2005075508A3 (en) 2006-07-20
WO2005075508A8 (en) 2005-10-27

Similar Documents

Publication Publication Date Title
Geley et al. Anaphase-promoting complex/cyclosome–dependent proteolysis of human cyclin A starts at the beginning of mitosis and is not subject to the spindle assembly checkpoint
Huang et al. SR splicing factors serve as adapter proteins for TAP-dependent mRNA export
Garcia-Bustos et al. Nuclear protein localization
Morris et al. Myosin VI binds to and localises with Dab2, potentially linking receptor‐mediated endocytosis and the actin cytoskeleton
US6207452B1 (en) Antibody of the anti-proliferation domain of human Bcl-2
Singh et al. DNA binding domains and nuclear localization signal of LEDGF: contribution of two helix-turn-helix (HTH)-like domains and a stretch of 58 amino acids of the N-terminal to the trans-activation potential of LEDGF
US20060141529A1 (en) Compositions, kits and assays containing reagents directed to cortactin and an ARG/ABL protein kinase
Ohtakara et al. Densin‐180, a synaptic protein, links to PSD‐95 through its direct interaction with MAGUIN‐1
JP5143552B2 (en) Cell cycle phase marker
Lindsay et al. Myosin Vb localises to nucleoli and associates with the RNA polymerase I transcription complex
EP1990413B1 (en) Screening method for candidate drugs
Mattock et al. Inhibition of cell proliferation by the PCNA-binding region of p21 expressed as a GFP miniprotein
KR100648761B1 (en) Hypoxia-inducible factor 1? hif-1? variants and methods for identifying hif-1? modulators
US6436654B1 (en) Methods for identifying compounds that modulate HIF-1α
Xu et al. Regulation of thyroid hormone receptor α2 RNA binding and subcellular localization by phosphorylation
EP1637540A1 (en) Hyperactive Stat molecules and their use in assays employing gene activation
JP2006515159A (en) MK2 interacting protein
Boettner et al. Ras and rap 1 interaction with AF-6 effector target
WO2005075508A2 (en) Hyperactive stat molecules and their use in assays employing gene activation
US20040067497A1 (en) Transcription-based assay for identification of post-translational modification and its application in proteomics
US20030211523A1 (en) Two-hybrid double screening system and method
WO2005049868A1 (en) Methods and agents for screening for compounds capable of modulating her2 expression
Jaradat Investigating the role of the P2X7 intracellular domains in coupling to downstream signalling
Monzo et al. CD2AP, Rabip4, and Rabip4′: Analysis of Interaction with Rab4a and Regulation of Endosomes Morphology
EP3003352B1 (en) Protein involved in dna replication, and modulation of its activity

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i

Free format text: IN PCT GAZETTE 33/2005 UNDER (81) ADD "SM"

NENP Non-entry into the national phase in:

Ref country code: DE

WWW Wipo information: withdrawn in national office

Country of ref document: DE

122 Ep: pct application non-entry in european phase