WO2002064776A2 - Signal transduction proteins 15b3, 15b3-1 and 15b3-2, and related dna sequences - Google Patents

Signal transduction proteins 15b3, 15b3-1 and 15b3-2, and related dna sequences Download PDF

Info

Publication number
WO2002064776A2
WO2002064776A2 PCT/EP2002/001562 EP0201562W WO02064776A2 WO 2002064776 A2 WO2002064776 A2 WO 2002064776A2 EP 0201562 W EP0201562 W EP 0201562W WO 02064776 A2 WO02064776 A2 WO 02064776A2
Authority
WO
WIPO (PCT)
Prior art keywords
protein
sequences
cell
gene
sequence
Prior art date
Application number
PCT/EP2002/001562
Other languages
German (de)
French (fr)
Other versions
WO2002064776A3 (en
Inventor
Armin Schneider
Achim Fischer
Bettina Klausner
Moritz Rossner
Birgitta Kammandel
Rebecca Widenmeyer
Bernhard Götz
Gisela Eisenhardt
Stephanie Jomana Naim
Dieter Newrzella
Original Assignee
Axaron Bioscience Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Axaron Bioscience Ag filed Critical Axaron Bioscience Ag
Priority to AU2002250943A priority Critical patent/AU2002250943A1/en
Publication of WO2002064776A2 publication Critical patent/WO2002064776A2/en
Publication of WO2002064776A3 publication Critical patent/WO2002064776A3/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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Definitions

  • the present invention relates to (i) DNA sequences, (ii) expression vectors which contain DNA sequences according to the invention, (iv) host cells which have expression vectors according to the invention, (v) gene products which are encoded by sequences according to the invention, (vi) transgenic animals modified with respect to sequences according to the invention, (vii) antibodies directed against gene products according to the invention, (viii) methods for the expression or isolation of gene products according to the invention, (ix) the use of DNA sequences or gene products according to the invention as medicaments, (x) pharmaceutically active Compounds and processes for their preparation and uses of such compounds according to the invention and (xi) non-therapeutic uses of DNA sequences or gene products according to the invention.
  • Ras / Erk signal transduction pathway has a great influence on NMDA receptor / NOS activation and the neuronal ischemic preconditioning.
  • the proteins of the Ras / Erk cascade represent pharmacological targets because specific inhibitors can be found based on defined biochemical functions.
  • Stroke is the third leading cause of death and the main cause of disabilities in western industrialized nations. Every year around 250,000 people in Germany suffer from a stroke, a third die within the first month, 60% retain hindrances back. So far, there is no effective therapy for the majority of these patients. The majority of all strokes in Western countries are due to cerebral blood flow disorders caused by thrombosis or embolism. For this reason, many of the previous therapy concepts have been based on thrombolysis (for example by means of streptokinase, rtPA, Ancrod). From the prior art, however, Newer Ar is now also known that a spontaneous reperfusion occurs in the majority of "strokes. BEITEN in rodents also suggest that the damage is rather larger by reperfusion in a certain time window.
  • the object of the present invention is to identify further proteins and their underlying nucleotide sequences which have a protective effect, in particular impart a certain resistance to the stroke, as well as those which cause an expansion of the ischemic damage.
  • Another object of the present invention is to provide methods on the basis of identified proteins which make it possible to develop pharmaceuticals which can intervene therapeutically in the corresponding pathophysiology. The provision of corresponding pharmaceuticals is therefore also an object of the present invention.
  • nucleic acid sequences in particular DNA sequences, which contain a sequence region which is suitable for a polypeptide with an amino acid sequence of AS 312 to AS 711 (15B3), AS 180 to AS 579 (15B3-1) or AS 306 to AS 705 (15B3-2) (numbering according to FIG. 4), including all functionally homologous derivatives, fragments or alleles.
  • all nucleic acid sequences are included, those with the invention Hybridize sequences, including the complementary sequences in the double strand (claim 1).
  • DNA sequences are disclosed whose gene product codes for a polypeptide, as shown in one of FIGS. 9, 11 or 13 for the sequences 15B3, 15B3-1 or 15B3-2, including all functionally homologous derivatives, alleles or Fragments of such a DNA sequence and also infunctional derivatives, alleles, analogs or fragments (for example DN variants) which can inhibit the physiological function, for example apoptotic signal cascade (claim 2).
  • DNA sequences hybridizing with these DNA sequences according to the invention are also disclosed. Derivatives, analogs, fragments or alleles of this type are prepared by standard methods (Sambrook et al.
  • the subject matter of the present application also includes partial DNA sequences of the sequences shown in FIGS. 8, 10 or 12. These partial sequences typically contain at least 60, more preferably at least 150 and even more preferably at least 250 nucleotides comprising fragments of the nucleotide sequences shown in FIGS. 8, 10 or 12.
  • preferred partial sequences for polypeptides encode that of AS 312 (15B3), 180 (15B3-1) or 306 (15B3-2) run at least 20 AS in the direction of the N-terminus and possibly extend to the N-terminus (numbering according to Figure 4).
  • the partial sequence coding for at least 20 AS can also begin at a codon lying further proximally or distally from the aforementioned points.
  • Partial sequences which code for at least one LRR are very particularly preferred, preferably at least 2 and more preferably at least 5 (LRRs are indicated in FIG. 4 by arrows). Those partial nucleotide sequences which code for at least one of the LRRs marked in FIG. 4 or nucleotide sequences which contain at least one of the LRRs indicated in FIG. 4 are particularly preferred. All derivatives, analogs or alleles of the aforementioned partial sequences disclosed are also disclosed. The AS sequences resulting from these partial DNA sequences according to the invention are also disclosed as such or as a constituent in larger recombinant proteins. All native splice variants of the sequences according to the invention also belong to the scope of the present invention.
  • nucleic acid sequences in particular DNA sequences, which code for a protein which has at least 60% sequence identity, preferably at least 80% and even more preferably at least 95%, with the sequences shown in FIGS. 9, 11 or 13.
  • the nucleotide sequences according to the invention shown in FIGS. 8, 10 or 12 or their functional or non-functional equivalents, such as, for example, allele variants or isoforms are obtainable.
  • allele variants are understood to mean variants which have 60 to 100% homology at the amino acid level, preferably 70 to 100%, very particularly preferably 90 to 100%.
  • Allelic variants include in particular those functional or infunctional len variants which can be obtained by deleting, inserting or substituting nucleotides from the sequences shown in FIGS. 8, 10 or 12, at least one of the essential biological properties being retained.
  • Homologous or sequence-related DNA sequences can be isolated from all mammalian species, including humans, by conventional methods by homology screening by hybridization with a sample of the nucleic acid sequences according to the invention or parts thereof.
  • Functional equivalents are also to be understood as homologs of the sequences shown in FIGS. 8, 10 or 12, for example their homologues from other mammals, shortened sequences, single-stranded DNA or RNA of the coding and non-coding DNA sequence.
  • Such functional equivalents can be isolated from other vertebrates such as Mammalia, starting from the DNA sequences shown in FIGS. 8, 10 or 12 or parts of these sequences, for example using conventional hybridization methods or the PCR technique. According to the invention, all sequences hybridizing with the sequences according to FIGS.
  • DNA sequences hybridize under standard conditions with the sequences according to the invention. Short oligonucleotides of the conserved regions, which can be determined in a manner known to the person skilled in the art, are advantageously used for the hybridization. However, longer fragments of the nucleic acids according to the invention or the complete sequences can also be used for the hybridization.
  • RNA hybrids are approx. 10 ° C lower than those of DNA: RNA hybrids of the same length.
  • temperatures between 42 and 58 ° C in an aqueous buffer solution with a concentration between 0.1 to 5 x SSC (1 X SSC 0.15 M NaCl, 15 mM sodium citrate, pH 7.2 ) or additionally in the presence of 50% formamide, such as 42 ° C in 5 x SSC, 50% formamide.
  • the hybridization conditions for DNA are advantageously 0.1 ⁇ SSC and temperatures between approximately 20 ° C. to 45 ° C., preferably between approximately 30 ° C. to 45 ° C.
  • DNA RNA hybrids
  • the hybridization conditions are advantageously 0.1 ⁇ SSC and temperatures between approximately 30 ° C. to 55 ° C., preferably between approximately 45 ° C. to 55 ° C.
  • These specified temperatures for the hybridization are, for example, calculated melting temperature values for a nucleic acid with a length of approx. 100 nucleotides and a G + C content of 50% in the absence of formamide.
  • the experimental conditions for DNA hybridization are in relevant textbooks of genetics, such as in Sambrook et al.
  • nucleic acid sequences according to the invention also include derivatives of the sequences shown in FIGS. 8, 10 or 12, such as promoter variants.
  • the promoters which are upstream of the specified nucleotide sequences together or individually can be changed by one or more nucleotide changes, by insertion (s) and / or deletion (s), functionality or effectiveness of the promoters being retained or, depending can be changed as required.
  • the effectiveness of the promoters can be increased by changing their sequence, or they can be completely replaced by more effective promoters, including organisms of other species.
  • derivatives are also to be understood to mean variants whose nucleotide sequence in the range -1 to -1000 before the start codon have been changed such that the gene expression and / or the protein expression is changed, preferably increased.
  • derivatives are also to be understood as variants which were preferably changed at the 3 'end.
  • tags in the literature, for. B. Hexa-histidine anchor known or epitopes that can be recognized as antigens of various antibodies (Studier et al., Meth. Enzymol., 185, 1990: 60 - 89 and Ausubel et al. (Eds.) 1998, Current Protocols in Molecular Biology, John Wiley & Sons, New York). and / or at least one signal sequence for transporting the translated protein, for example into a specific cell organelle or into the extracellular space.
  • nucleic acid construct according to the invention or a nucleic acid according to the invention for example according to FIGS. 8, 10 or 12 or their derivatives, variants, homologs or, in particular, fragments can also be expressed in a therapeutically or diagnostically suitable form.
  • vector systems or 0-ligonucleotides can be used which extend the nucleic acids or the nucleic acid construct by certain nucleotide sequences and thus code for modified polypeptides which, for example, serve for easier purification.
  • DNA sequences which contain one of the (c) DNA sequences indicated in FIG. 8, 10 or 12 are further preferred (claim 4).
  • all DNA sequences which code for a protein which essentially corresponds to the amino acid sequence of the proteins 15B3, 15B3-1 or 15B3-2 according to FIGS. 9, 11 or 13 are preferably disclosed. These DNA sequences receive only a small number of changes compared to the sequences given in the aforementioned figures, for example they can be isoforms. The number of sequence changes will typically not be greater than 10.
  • Such DNA sequences which essentially correspond to the DNA sequences coding for the proteins 15B3, 15B3-1 or 15B3-2 and which likewise code for a biologically active protein, can be obtained by generally known mutagenesis methods and the biological activity of the proteins encoded by the mutants can be identified by screening methods, for example binding studies or the ability to express the biological function, for example in connection with apoptosis.
  • the corresponding mutagenesis methods include “site-directed” mutagenesis, which provides for the automatically carried out synthesis of a primer with at least one base change. After the polymerization reaction, the heteroduplex vector is converted into a suitable netes cell system transferred (eg E. coli) and isolated transformed clones isolated.
  • PCR Innis et al. PCR Protocols: A Guide to Methods and Applications
  • Appropriate PCR primers can be used, for example, to introduce new functions into a DNA sequence according to the invention, such as Restriction interfaces, termination codons. In this way, sequences according to the invention can be designed accordingly for transfer into cloning vectors.
  • the present invention further relates to expression vectors or a recombinant nucleic acid construct which, as described above, contains a nucleic acid sequence according to the invention, typically a DNA sequence (claim 5).
  • the nucleic acid sequences according to the invention are functionally linked to at least one genetic regulatory element, such as, for example, transcription and translation signals. Depending on the desired application, this linkage can lead to a native expression rate or also to an increase or decrease in the native gene expression.
  • host organisms or host cells can then be transformed, e.g. Cell cultures from mammalian cells.
  • the expression vector according to the invention will typically use the native regulatory element (s), ie the promoter and / or enhancer Region of the gene for the protein 15B3, 15B3-1 or 15B3-2, for example from mammals, in particular corresponding human regulatory sequences. Possibly. these native 15B3, 15B3-1 or 15B3-2 regulatory sequences can also be genetically modified in order to produce a changed expression intensity.
  • native regulatory element ie the promoter and / or enhancer Region of the gene for the protein 15B3, 15B3-1 or 15B3-2
  • these native 15B3, 15B3-1 or 15B3-2 regulatory sequences can also be genetically modified in order to produce a changed expression intensity.
  • native regulatory elements according to the invention for other genes can be connected upstream and / or downstream (5 ' - or 3 'regulatory sequences) and, if necessary, also have been genetically modified so that the natural regulation is switched off under the control of the 15B3, 15B3-1 or 15B3-2 regulatory sequences and the expression of the genes - as desired - this can be increased or decreased.
  • Advantageous regulatory sequences for the method according to the invention are, for example, in promoters such as cos, tac, trp, tet, trp-tet, lpp, lac, lpp-lac, laclq, T7, T5, T3 contain, gal, trc, ara, SP6, 1-PR or in the 1-PL promoter, which are advantageously used in gram-negative bacteria.
  • promoters such as cos, tac, trp, tet, trp-tet, lpp, lac, lpp-lac, laclq, T7, T5, T3 contain, gal, trc, ara, SP6, 1-PR or in the 1-PL promoter, which are advantageously used in gram-negative bacteria.
  • promoters such as amy and SP02
  • yeast promoters such as ADC1, MFa, AC, P-60, CYC1, GAPDH
  • mammalian promoters such as CaM-Kinasell, CMV, Nestin, L7, BDNF, NF , MBP, NSE, beta-globin, GFAP, GAP43, tyrosine hydroxylase, kainate receptor subunit 1, glutamate receptor subunit B included.
  • all natural promoters with their regulatory sequences, such as those mentioned above, can be used for an expression vector according to the invention.
  • synthetic promoters can also be used advantageously.
  • regulatory sequences are intended to enable the targeted expression of the nucleic acid sequences according to the invention. Depending on the host organism, this can mean, for example, that the gene is only expressed or overexpressed after induction, or that it is expressed and / or overexpressed immediately.
  • the regulatory sequences or factors can preferably have a positive influence on the expression and thereby increase it.
  • the regulatory elements can advantageously be strengthened at the transcription level by using strong transcription signals such as promoters and / or "enhancers".
  • an increase in translation is also possible, for example, by improving the stability of the mRNA.
  • Regulatory sequences are all elements familiar to the person skilled in the art which can influence the expression of the sequences according to the invention at the transcription and / or translation level.
  • so-called “enhancer” sequences are to be emphasized, which can bring about increased expression via an improved interaction between RNA polymerase and DNA.
  • locus control regions so-called “silencers” or respective partial sequences thereof may be mentioned as further regulatory sequences.
  • These sequences can be used advantageously for tissue-specific expression.
  • terminator sequences will also advantageously be present in an expression vector according to the invention and, according to the invention, are subsumed under the term “regulatory sequence”.
  • a preferred embodiment of the present invention is the linkage of the nucleic acid sequence according to the invention with a promoter, the promoter typically being located 5 '"upstream" from a DNA sequence according to the invention.
  • Further regulation signals such as, for example, 3 '-terminal, polyadenylation signals or enhancers, can be functionally contained in the expression vector.
  • nucleic acid sequences according to the invention in particular for the sequences according to FIGS. 8, 10 or 12 or for the corresponding proteins, can be contained in one or more copies in a gene construct according to this invention, or, if appropriate, can also be localized on separate gene constructs.
  • expression vector includes both recombinant nucleic acid constructs or gene constructs, as described above, and complete vector constructs, which typically contain further elements in addition to the DNA sequences according to the invention and any regulatory sequences. These vector constructs or vectors are used for expression in a suitable host organism.
  • at least one DNA sequence according to the invention for example the 15B3 gene or a partial sequence of the 15B3 gene, is inserted into a host-specific vector which enables optimal expression of the genes in the selected host.
  • Vectors are well known to those skilled in the art and can be found, for example, in "Cloning Vectors" (Eds. Pouwels PH et al.
  • vectors are also understood to mean all other vectors known to the person skilled in the art, such as phages, viruses such as SV40, CMV, baculovirus, adenovirus, Sindbis virus, transposons, IS elements, phasmids, phagemids, cosmids, linear or circular DNA. These vectors can be replicated autonomously in the host organism or replicated chromosomally. Linear DNA is typically used for integration into Mammalia.
  • nucleic acid sequences according to the invention can advantageously be increased by increasing the number of gene copies and / or by strengthening regulatory factors which have a positive effect on gene expression.
  • regulatory elements can preferably be amplified at the transcription level by using stronger transcription signals, such as promoters and enhancers.
  • an increase in translation is also possible, for example, by improving the stability of the mRNA or increasing the reading efficiency of this mRNA on the ribosomes.
  • the nucleic acid sequences or the homologous genes can be incorporated, for example, into a nucleic acid fragment or into a vector which preferably contains the regulatory gene sequences assigned to the respective genes or promoter activity having an analogous effect. In particular, those regulatory sequences are used which increase gene expression.
  • Nucleic acid sequences according to the invention can be cloned together with the sequences coding for interacting or for potentially interacting proteins into a single vector and then expressed in vitro in a host cell or in vivo in a host organism.
  • each of the potentially interacting nucleic acid sequences and the sequences coding for 15B3, 15B3-1 or 15B3-2 can each be brought into a single vector and these separated into the respective organism using customary methods, such as, for example, transformation, transfection, transduction, Electroporation or particle gun can be spent.
  • At least one marker gene (for example antibiotic resistance genes and / or genes which code for a fluorescent protein, in particular GFP), can be contained in an expression vector according to the invention, in particular a complete vector construct.
  • the present invention further relates to host cells which have been transformed with a DNA sequence according to the invention and / or an expression vector according to the invention, in particular a vector construct (claim 6).
  • all cells are suitable as host cells that allow expression of DNA sequences according to the invention alone or in combination with other sequences, in particular regulatory sequences.
  • All cells of a pro- or eukaryotic nature can be considered as host cells, for example bacteria, fungi, yeasts, plant or animal cells.
  • Preferred host cells are bacteria such as Escherichia coli, Streptomyces, Bacillus or Pseudomonas, eukaryotic microorganisms such as Aspergillus or Saccharomyces cerevisiae or the usual baker's yeast (Stinchcomb et al., Nature, 282: 39, (1997)).
  • cells from multicellular organisms are selected for the expression of DNA sequences according to the invention. This also happens against the background of a possibly required glycosylation (N- and / or O-coupled) of the encoded proteins.
  • This function can be carried out in a suitable manner in higher eukaryotic cells - in comparison to prokaryotic cells.
  • any higher eukaryotic cell culture is available as a host cell, although cells from mammals, for example monkeys, rats, hamsters or humans, are very particularly preferred.
  • a large number of established cell lines are known to the person skilled in the art. In a by no means exhaustive list named the following cell lines: 293T (embryonic kidney cell line), (Graham et al., J. Gen.
  • Human transformed cells according to the invention in particular autologous cells of the patient, are suitable after (above all ex vivo) transformation with DNA sequences according to the invention or expression vectors according to the invention, very particularly as a medicament for, for example, gene therapy purposes, that is to say after cell removal, possibly ex vivo Expansion, transformation, selection and final retransplantation.
  • a host cell and an expression vector according to the invention which matches the host cells, such as plasmids, viruses or phages, such as plasmids with the RNA polymerase / promoter system, the phages 1, Mu or other tempered phages or transposons, and / or further advantageous regulatory sequences form a host cell according to the invention which can serve as an expression system.
  • Preferred expression systems according to the invention based on host cells according to the invention are, for example, the combination of mammalian cells, such as, for example, CHO cells, and vectors, such as, for example, pcDNA3neo vector, or, for example, HEK293 cells and CMV vector, which are particularly suitable for mammalian cells.
  • gene products are understood to mean both primary transcripts, that is to say RNA, preferably mRNA, and proteins or polypeptides, in particular in purified form (claim 9).
  • these proteins have at least one LRR sequence, preferably at least 5, more preferably at least 8 and most preferably 10 or even more than
  • LRR sequences and regulate or transport in particular apoptotic or necrotic, possibly also inflammatory signals or signals which concern cell growth or cell plasticity. Typically, they are directly or indirectly involved in signal transduction via the Ras cascade.
  • a purified gene product is preferred if it contains at least one of the amino acid sequences given in FIG. 4 (and also contained in FIGS. 9, 11 and 13 (for an LRR domain), or a function-homologous or function-inhibiting (infunctional) allele "Contains a fragment, analog or derivative of this sequence or typically consists of such an amino acid sequence. Functional homology is defined in the sense of the present invention in such a way that at least one of the essential functional properties of those according to FIG.
  • Proteins 15B3, 15B3-1 or 15B3-2 shown in FIG. 11 or 13 are retained.
  • functionally homologous proteins according to the invention will in particular have a characteristic, for example at least 60%, preferably at least 80% sequence identity in the sequence sections designated as leucine-rich regions (LRRs), which represent protein interaction domains and typically 20-28 amino acids with a specific core consensus. Sequence of leucines and asparagines (LXXLXLXXN) have.
  • a functionally homologous allele, derivative or analog of the amino acid sequences disclosed in FIGS. 9, 11 or 13 is typically at least four, preferably at least at least 6 and even more preferably have at least 8 LRRs with the aforementioned consensus sequences.
  • a derivative is understood to mean, in particular, those AS sequences which have been modified by modifications to their side chains. For example. by conjugation of an antibody, enzyme or receptor to an AS sequence according to the invention.
  • Derivatives can also be the coupling of a sugar or fatty (acid) residue or a phosphate group or any modification of a side chain, in particular a free OH group or NH2 group or at the N or C terminus.
  • the term "derivative” also includes fusion proteins in which an amino acid sequence according to the invention is coupled to any oligo- or polypeptides.
  • sequences that are characterized by at least one AS change compared to the native sequence are referred to as "analogs".
  • analogs such conservative substitutions are preferred in which the physicochemical character (space filling, basicity, hydrophobicity, etc.) of the exchanged AS is retained (polar AS, long aliphatic chain, short aliphatic chain, negatively or positively charged AS, AS with aromatic group).
  • the substitutions can be biologically functional, some. Functional or bio-functional sequences result. For example, arginine residues can be exchanged for lysine residues, valine residues for isoleucine residues or aspartic acid residues for glutamic acid residues.
  • the proteins modified in this way compared to the ones in FIG. 9, 11 or 13 typically have at least 60%, preferably at least 70% and particularly preferably at least 90% sequence identity to the sequences in the aforementioned figures, calculated according to the algorithm of Altschul et al. (J. Mol. Biol., 215, 403-410, 1990).
  • the isolated protein and its functional variants can advantageously be isolated from the brain of mammals such as Homo sapiens, Rattus norvegicus or Mus musculus. Homologs from other mammals are also to be understood as functional variants.
  • Analogs are preferred according to the invention if the secondary structure as occurs in the native sequence is also retained in them.
  • less conservative AS variations can also be introduced into the native sequence according to the invention. They typically retain their biological function, in particular as a transducer of an apoptotic or necrotic, cell proliferative, growth-indicating or regenerative signal. The effect of a substitution or deletion can easily be checked by appropriate investigations, for example binding assays or cytotoxic tests.
  • sequences are also included which produce a so-called dominant-negative effect, ie, due to their changed primary sequence, still have binding activity to a sequence upstream in the cascade, but cannot pass the signal downstream.
  • Analogs of this type therefore function as inhibitors of biological function, in particular as inhibitors of apoptosis, cell growth, cell proliferation, cell plasticity or cell regeneration.
  • Such analogs are produced by genetic engineering measures, typically by the so-called “site directed "mutagenesis of a DNA sequence which codes for a protein according to the invention (typically 15B3, 15B3-1 or 15B3-2).
  • fragments of a native AS sequence according to the invention also form part of the subject matter of the present invention. Fragments are characterized by deletions (N- or C-terminal or also intrasequential). They can have a dominant-negative or dominant-positive effect.
  • the gene products (proteins) according to the invention also include all those gene products (proteins) which, according to the invention, are derived from DNA derivatives, DNA fragments or DNA alleles of the DNA sequences shown in FIGS. 8, 10 or 12 after transcription and translation ,
  • proteins according to the invention can be chemically modified.
  • Glycosyl groups can be added to hydroxyl or amino groups, lipids can be covalently linked to the protein according to the invention, as can phosphates or acetyl groups and the like.
  • Any chemical substances, compounds or groups can also be bound to the protein according to the invention by any synthetic route.
  • Additional amino acids for example in the form of individual amino acids or in the form of peptides or in the form of protein domains and the like, can also be used with the N- and / or C-terminus of a protein according to the invention, but in particular also at the N- or the C-terminus of a leucine-rich domain may be fused, but may also be integrated into the sequence of the leucine-rich domain according to the invention.
  • signal or "leader” sequences at the N-terminus of the amino acid sequence of a protein according to the invention are preferred, which lead the peptide cotranslationally or post-translationally into a specific cell organelle or into the extracellular space (or the culture medium). Ren.
  • Amino acid sequences can also be present at the N- or at the C-terminus, which allow the binding of the amino acid sequence according to the invention to antibodies as antigen.
  • the flag peptide the sequence of which in the single-letter code of the amino acids is: DYKDDDDK. Or a His tag with at least 3, preferably at least 6, histidine residues. These sequences have strong antigenic properties and thus allow the recombinant protein to be checked and purified quickly.
  • Monoclonal antibodies that bind the flag peptide are available from Eastman Kodak Co., Scientific Imaging Systems Division, New Haven, Connecticut.
  • the present invention furthermore relates to at least 20, more preferably at least 30 and even more preferably at least 50, amino acid segments of the sequences disclosed in FIGS. 9, 11 or 13.
  • Such partial sequences can, for example, be chemically synthesized by methods familiar to the person skilled in the art and can preferably be used as antigens for the production of antibodies.
  • These subsections are preferably regions of the sequences disclosed in FIGS. 9, 11 or 13 that at least partially make up the protein surface in the spatial model of the proteins.
  • Transgenic animals are a further subject of the present invention (claim 11).
  • the transgenic animals according to the invention are animals which have been genetically modified in such a way that they express or contain an amount of a gene product according to the invention which is modified in comparison to the normal animal in at least one tissue.
  • those animals are also included that either tw. or completely no longer have, or (b) indeed have sequences according to the invention at the genetic level, but cannot transcribe and / or translate them and therefore no longer contain the gene product.
  • the native 15B3 sequence (s), 15B3-1 or 15B3-2 sequence (s) can be supplemented by at least one DNA sequence according to the invention at least one DNA sequence according to the invention may be substituted.
  • the substituted and / or supplemented sequence (s) can be sequences according to the invention which are non-native in nature.
  • transgenic and “knock-out” animals in particular mice, rats, pigs, fruit flies or zebra fish, with respect to the sequences according to the invention, is carried out in a manner familiar to the person skilled in the art.
  • a 15B3, 15B3-1 or 15B3-2 cDNA sequence or native or non-native variant expressed in transgenic mice eg under an NSE promoter in neurons, under an MBP promoter in oligodendrocytes etc.
  • the genetically modified animals can then be examined in different disease models (eg experimentally induced stroke, MCAO )
  • MCAO experimentally induced stroke
  • all multicellular organisms can be transgenic, in particular mammals, for example mice, rats, sheep, cattle or pigs.
  • transgenic plants are also conceivable.
  • the transgenic organisms can also be so-called "knock-out" animals.
  • the transgenic animals can contain a functional or non-functional nucleic acid sequence according to the invention or a functional or non-functional nucleic acid construct alone or in combination with a functional or non-functional sequence which codes for 15B3 proteins.
  • transgenic animals described above are transgenic animals, in their germ cells or all or part of the somatic cells or in their germ cells or all or part of the somatic cells the native 15B3 nucleotide sequence (s) have been modified by genetic engineering or interrupted by the insertion of DNA elements.
  • a further possibility of using a nucleotide sequence according to the invention or parts thereof is the generation of transgenic or knock-out or conditional or region-specific knock-out animals or specific mutations in genetically modified animals (Ausubel et al. (Eds.) 1998, Current Protocols in Molecular Biology, John Wiley & Sons, New York and Torres et al., (Eds.) 1997, Laboratory protocols for conditional gene targeting, Oxford University Press, Oxford).
  • animal models Via transgenic overexpression or genetic mutation (zero mutation or specific deletions, insertions or changes) by homologous recombination in embryonic stem cells, animal models can be generated which provide valuable further information about the (patho-) physiology of the sequences according to the invention. Animal models produced in this way can represent essential test systems for evaluating novel therapeutic agents which influence the biological function of proteins according to FIGS. 9, 11 or 13 for neural, vascular or other processes.
  • the present invention furthermore relates to an antibody which recognizes an epitope on a gene product according to the invention, in particular a protein according to the invention according to FIGS. 9, 11 or 13 or derivatives, fragments or isoforms or alleles (claim 12), but also against, for example , mRNA according to the invention can be directed.
  • antibody encompasses both polyclonal antibodies and monoclonal antibodies (claim 13), chimeric antibodies, anti-idiotypic antibodies (directed against antibodies according to the invention), all of which are present in bound or soluble form and, if appropriate, by "Label” can be marked, as well as fragments of the aforementioned antibodies.
  • antibodies according to the invention can also occur in recombinant form as fusion proteins with other (protein) components. Fragments as such or fragments of antibodies according to the invention as components of fusion proteins are typically produced by the methods of enzymatic cleavage, protein synthesis or the recombination methods familiar to the person skilled in the art. As antibodies according to the present invention thus both polyclonal, monoclonal, human or humanized or recombinant antibodies or fragments thereof, single chain antibodies or synthetic antibodies.
  • the polyclonal antibodies are heterogeneous mixtures of antibody molecules which are produced from sera from animals which have been immunized with an antigen.
  • the subject of the invention also includes polyclonal monospecific antibodies which are obtained after the antibodies have been purified (for example via a column which is loaded with peptides of a specific epitope).
  • a monoclonal antibody contains an essentially homogeneous population of antibodies which are specifically directed against antigens, the antibodies having essentially the same epitope binding sites.
  • Monoclonal antibodies can be obtained by methods known in the art (e.g., Koehler and Milstein, Nature, 256, 495-397, (1975); U.S.
  • Patent 4,376,110 Ausübel et al., Harlow and Lane “Antibodies” : Laboratory Manual, Cold Spring, Harbor Laboratory (1988); Ausubel et al., (Eds), 1998, Current Protocols in Molecular Biology, John Wiley & Sons, New York).).
  • the description contained in the abovementioned references is included as part of the present invention in the disclosure of the present invention.
  • Antibodies according to the invention which have been genetically engineered can also be produced by methods as described in the aforementioned publications.
  • antibody-producing cells are attracted to this and the mRNA, provided the optical density of the cells is sufficient, is lysed with guanidinium thiocyanate, acidified with sodium acetate, extracted with phenol, chloroform / isoamyl alcohol, precipitated with isopropyl alcohol. panol and washing with ethanol isolated from the cells in a known manner. Then the reverse transcriptase is used to synthesize cDNA from the mRNA.
  • the synthesized cDNA can be inserted directly or after genetic manipulation, for example by "site directed mutagenesis", introduction of insertions, inversions, deletions or base exchanges into suitable animal, fungal, bacterial or viral vectors and expressed in the corresponding host organisms.
  • Bacterial or yeast vectors such as pBR322, pUC18 / 19, pACYC184, lambda or yeast mu vectors for cloning the genes and expression in bacteria such as E. coli or in yeast such as Saccharomyces cerevisiae are preferred.
  • Specific antibodies against the proteins according to the invention can be suitable both as diagnostic reagents and as therapeutic agents for diseases in which 15B3 is of pathophysiological importance.
  • Antibodies according to the invention can belong to one of the following immunoglobulin classes: IgG, IgM, IgE, IgA, GILD and possibly a subclass of the abovementioned classes, such as the subclasses of the IgG or mixtures thereof.
  • IgG and its subclasses such as IgGl, IgG2, IgG2a, IgG2b, IgG3 or IgGM are preferred.
  • the IgG subtypes IgGl / k or IgG2b / k are particularly preferred.
  • a hybridoma cell clone that produces monoclonal antibodies according to the invention can be cultivated in vitro, in situ or in vivo. Large titers of monoclonal antibodies are preferably produced in vivo or in situ.
  • the chimeric antibodies according to the invention are molecules which contain different constituents, which are derived from different animal species (e.g.
  • Chimeric antibodies are preferably used, on the one hand, to reduce the immunogenicity in use and, on the other hand, to increase the yields in production, for example murine monoclonal antibodies give higher yields from hybrid cell lines, but also lead to higher immunogenicity in humans , so that human / murine chimeric antibodies are preferably used.
  • Chimeric antibodies and methods for their preparation are known from the prior art (Cabilly et al., Proc. Natl. Sei. USA 81: 3273-3277 (1984); Morrison et al. Proc. Natl. Acad.
  • Such an antibody according to the invention against a leucine-rich sequence section on a protein according to FIGS. 9, 11 or 13 is very particularly preferably directed as an epitope (claim 14).
  • An anti-idiotypic antibody of the present invention is an antibody which is a determinant generally related to that
  • An anti-idiotypic antibody can be produced by immunizing an animal of the same type and the same genetic type (for example a mouse strain) as the starting point for a monoclonal antibody against which an anti-idiotypic antibody according to the invention is directed. The immunized animal will recognize the idiotypical determinants of the immunizing antibody by the production of an antibody which is directed against the idiotypic determinants (namely an anti-idiotypisher antibody according to the invention) (US Pat. No. 4,699,880).
  • An anti-idiotypic antibody according to the invention can also be used as an immunogen in order to elicit an immune response in another animal and to produce an anti-idiotypic antibody there.
  • the epitope construction of the anti-anti-idiotypic antibody may, but need not, be identical to the original monoclonal antibody that caused the anti-idiotypic reaction. In this way, by using antibodies directed against idiotypic determinants of a monoclonal antibody, other clones which express antibodies of identical specificity can be identified.
  • Monoclonal antibodies which are directed against proteins, analogs, fragments or derivatives of these proteins according to the invention, can be used to bind anti-idiotypic antibodies in corresponding animals, such as, for. B. the BALB / c mouse. Spleen cells from such an immunized mouse can be used to produce anti-idiotypic hybridoma cell lines that secrete anti-idiotypic monoclonal antibodies.
  • Anti-idiotypic monoclonal antibodies can also be coupled to a carrier (KLH, "keyhole lim- pet hemocyanin ”) and can then be used to immunize further BALB / c mice.
  • the sera of these mice then contain anti-anti-idiotypic antibodies which have the binding properties of the original monoclonal antibodies and are specific for an epitope of the protein according to the invention or one
  • the anti-idiotypic monoclonal antibodies thus have their own idiotypic epitopes or "idiotopes" which are structurally similar to the epitope under investigation.
  • antibody is intended to include both intact molecules and fragments thereof. Fragments are all shortened or modified antibody fragments with one or two binding sites complementary to the antigen, such as antibody parts with a binding site formed by the light and heavy chain corresponding to the antibody, such as Fv, Fab or F (ab ') 2 fragments or single-strand fragments, called. Shortened double-strand fragments such as Fv, Fab or F (ab ') 2 are preferred. Fab and F (ab ') 2 fragments lack an Fc fragment, such as present in an intact antibody, so that they can be transported faster in the bloodstream and have comparatively less non-specific tissue binding than intact antibodies.
  • Fab and F (ab ') 2 fragments of antibodies according to the invention can be used in the detection and quantification of proteins according to the invention. Such fragments are typically made by proteolytic cleavage using enzymes such as. B. papain (for the production of Fab fragments) or pepsin (for the production of F (ab ') 2 , fragments) can be used, or can be obtained by chemical oxidation or by genetic engineering manipulation of the antibody genes.
  • the present invention also relates to mixtures of antibodies for the purposes of the present invention. In addition to the antibodies, mixtures of antibodies can also be used for all of the methods or uses described according to the present invention.
  • Both purified fractions of monoclonal antibodies, polyclonal antibodies or mixtures of monoclonal antibodies are used as medicaments and are used in the production of medicaments for the treatment of cerebral ischemia (eg stroke), degenerative diseases, in particular newly-regenerative diseases, and neurological diseases, such as, for example Epilepsy, as well as tumor diseases.
  • cerebral ischemia eg stroke
  • degenerative diseases in particular newly-regenerative diseases
  • neurological diseases such as, for example Epilepsy
  • Antibodies according to the invention can be used in a sample for the quantitative or qualitative detection of a gene product according to the invention, in particular proteins according to FIGS. 9, 11 or 13 or their fragments or derivatives, or also for detection of cells that express and, if necessary, secrete proteins according to the invention.
  • the use of antibodies according to the invention as diagnostic agents is disclosed.
  • the amount of gene product according to the invention and possibly. the activity (e.g. specific phosphorylation) of the proteins is determined according to FIGS. 9, 11 or 13.
  • the detection can be achieved with the aid of immunofluorescence methods, the fluorescence-labeled antibodies in combination with light microscopy, flow cytometry or fluorometric detection.
  • Antibodies according to the invention in the sense of the invention are suitable for histological examinations.
  • gene such as in the context of immunofluorescence or immunoelectromicroscopy, for the in situ detection of a protein according to the invention.
  • the in situ detection can take place in that a histological sample is taken from a patient and labeled antibodies according to the invention are added to such a sample.
  • the antibody (or a fragment of this antibody) is applied to the biological sample in labeled form. In this way it is not only possible to determine the presence of protein according to the invention in the sample, but also the distribution of the protein according to the invention in the tissue examined.
  • the biological sample can be a biological fluid, a tissue extract, harvested cells, such as.
  • the labeled antibody can be detected by methods known in the art (eg by fluorescence methods).
  • the biological sample can also on a solid phase support, such as. B. nitrocellulose or another carrier material, so that the cells, cell parts or soluble proteins are immobilized.
  • the carrier can then be washed one or more times with a suitable buffer, with subsequent treatment with a detectably labeled antibody according to the present invention.
  • the solid phase support can then be washed with the buffer a second time to remove unbound antibody.
  • the amount of bound label on the solid phase support can then be determined using a conventional method.
  • the carrier can be of either partially soluble or insoluble character to meet the conditions of the present invention.
  • the carrier material can take any shape, e.g. B. in the form of beads, or cylindrical or spherical, with polystyrene beads are preferred as a carrier.
  • Detectable antibody labeling can be done in different ways.
  • the antibody can be bound to an enzyme, the enzyme finally being used in an immunoassay (EIA).
  • EIA immunoassay
  • the enzyme can then react later with a corresponding substrate, so that a chemical compound is formed which can be detected in a manner familiar to the person skilled in the art and, if necessary, quantified, e.g. B. by spectrophotometry, fluorometry or the other optical method.
  • the enzyme can be malate dehydrogenase, staphylococcal nuclease, delta-5 steroid isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate dehydrogenase, triosephosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparticase, glucose oxydase, -Galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase or acetylcholinesterase.
  • the detection is then enabled via a chromogenic substrate that is specific for the enzyme used for the labeling and can finally be e.g. by visual comparison of the substrate converted by the enzyme reaction compared to control standards.
  • the detection can be ensured by other immunoassays, for example by radioactive labeling of the antibodies or antibody fragments (that is to say by means of a radioimmunoassay (RIA; Laboratory Techniques and Biochemistry in Molecular Biology, Work, T. et al. North Holland Publishing Company, New York (1978).
  • RIA radioimmunoassay
  • the radioactive isotope can be detected and quantified using scintillation counters or auto-radigraphy.
  • Fluorescent compounds can also be used for labeling, for example compounds such as fluororescin isothiocyanate, rhodamine, phyoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde and fluorescamine. Also fluorescent-emitting metals, such as. B. 152 E or other metals from the lanthanide group can be used. These metals are attached to the antibody via chelate groups, such as. B. Diethylenetriaminepentaacetic acid (ETPA) or EDTA coupled. Furthermore, the antibody according to the invention can be coupled via a compound which acts with the aid of chemiluminescence.
  • fluorescent-emitting metals such as. B. 152 E or other metals from the lanthanide group can be used. These metals are attached to the antibody via chelate groups, such as. B. Diethylenetriaminepentaacetic acid (ETPA) or EDTA coupled.
  • the antibody according to the invention can be coupled via
  • the presence of the chemiluminescence-labeled antibody is then detected via the luminescence which arises in the course of a chemical reaction.
  • luminol examples of such compounds are luminol, isoluminol, acridinium ester, imidazole, acridinium salt or oxalate ester.
  • bioluminescent compounds can also be used. Bioluminescence is a subspecies of chemiluminescence found in biological systems, where a catalytic protein increases the efficiency of the chemiluminescent reaction. The detection of the bioluminescent protein is again carried out via luminescence, with luciferin, luciferase or “aequorin” being considered as the bioluminescent compound.
  • An antibody according to the invention can be used for use in an immunometric assay, also known as a "two-site” or “sandwich” assay.
  • Typical immunometric assay systems include so-called “forward” assays, which are distinguished by the fact that body are bound to a solid phase system and that the antibody is brought into contact with the sample being examined in this way.
  • the antigen is isolated from the sample by the formation of a binary solid phase-antibody-antigen complex from the sample.
  • the solid support is washed to remove the remainder of the liquid sample, including any unbound antigen, and then contacted with a solution containing an unknown amount of the labeled detection antibody.
  • the labeled antibody serves as a so-called reporter molecule.
  • the solid phase support is washed again to remove unreacted labeled antibodies.
  • a so-called "sandwich” assay can also be used.
  • a single incubation step can be sufficient if the antibody bound to the solid phase and the labeled antibody are both applied to the sample to be tested at the same time.
  • the solid phase support is washed to remove residues of the liquid sample and the non-associated labeled antibodies.
  • the presence of labeled antibody on the solid phase support is determined in the same way as in the conventional "forward" sandwich assay.
  • a solution of the labeled antibody is first gradually added to the liquid sample, followed by the addition of unlabeled antibody, bound to a solid phase support, after a suitable incubation time has elapsed. After a second incubation step, the solid phase support becomes conventional washed to remove sample residues and labeled antibody that has not responded. The determination of the labeled antibody which has reacted with the solid phase support is then carried out as described above.
  • the use of the host cells transformed with the aid of expression vectors as a medicament or for the production of a medicament in particular for the purpose of treating diseases, for example tumor diseases, neurological diseases, neurodegenerative diseases (for example multiple sclerosis, Parkinson's disease), cerebral ischemia (e.g. stroke).
  • diseases for example tumor diseases, neurological diseases, neurodegenerative diseases (for example multiple sclerosis, Parkinson's disease), cerebral ischemia (e.g. stroke).
  • host cells according to the invention are made available in the case of diseases which are based on incorrect regulation of apoptosis, necrosis, cell proliferation, cell growth or cell plasticity.
  • the autologous or allogeneic host cells transformed in this way ex vivo according to the invention can then be transplanted to patients.
  • the gene product according to the invention of the DNA sequence according to the invention can, depending on the expression system, be isolated from a culture medium or from cell extracts.
  • the respective isolation methods and the method for the purification of the recombinant protein encoded by a DNA according to the invention strongly depend on the type of the host cell or also on the fact whether the protein is secreted into the medium.
  • expression systems can be used which lead to the secretion of the recombinant protein from the host cell.
  • the culture medium must be concentrated using commercially available protein concentration filters, for example Amicon or Millipore Pelicon.
  • a purification step can be carried out, for example a gel filtration step or a purification using column chromatography methods.
  • an anion exchanger can be used which has a matrix with DEAE.
  • HPLC steps can then be used to further purify a polypeptide encoded by a DNA according to the invention. It can be one or more Act steps. In particular, the "reversed phase" method is used. These steps serve to obtain an essentially homogeneous recombinant protein of a DNA sequence according to the invention.
  • transformed yeast cells can also be used.
  • the translated protein can be secreted, so that protein purification is simplified.
  • Secreted recombinant protein from a yeast host cell can be obtained by methods as described in Urdal et al. (J. Chromato. 296: 171 (1994)) and form part of the disclosure of the present invention.
  • Nucleic acid sequences according to the invention in particular DNA sequences according to the invention, and / or gene products according to the invention can be used as medicaments or for the production of a medicament (claim 17). These can be administered as such (for example buccally, intravenously, orally, parenterally, nasally, subcutaneously) or in combination with other active ingredients, auxiliaries or additives typical of drugs.
  • Nucleic acid according to the invention can be injected as naked nucleic acid, in particular intravenously, or can be administered to the patient using vectors. These vectors can be plasmids as such, but also viral vectors, in particular retroviral or adenoviral vectors, or also liposomes which can have naked DNA according to the invention or a plasmid which contains DNA according to the invention.
  • sequences according to the invention in particular the nucleotide or amino acid sequences of 15B3, 15B3-1 or 15B3-2 or their variants, and protein proteins according to the invention
  • Heteromeric and reagents according to the invention derived therefrom are thus suitable for the production of a medicament for therapeutic purposes, ie for the treatment of diseases.
  • the therapeutic use for the treatment or for the manufacture of a medicament for the treatment of diseases or pathophysiological conditions which are based, for example, on incorrectly controlled regulation of the homeostasis of cell death and proliferation events, is very particularly preferred (claim 18).
  • cell death processes for example cascades which lead to apoptosis or processes which lead to necrosis
  • cell death processes can be expressed in all cell types which express 15B3, 15B3-1 and / or 15B3-2 and or a native variant thereof, in particular in neural ones
  • Cells are influenced, for example by modulating cell-cell interactions.
  • the native proteins 15B3, 15B3-1 and 15B3-2 according to the invention are, according to the invention, part of the ras-MAPK signal transduction pathway, typically as a modulator thereof, the faulty control of which is the cause of a large number of diseases.
  • the aforementioned proteins according to the invention are found in particular as components in the following cellular processes and have cellular functions in: signal transduction, cell differentiation, growth, plasticity, regeneration. Accordingly, by Infunktion gleich of a protein according to the invention, for example. 1563 ⁇ or by infunktionelle expression or overexpression thereof a pathophysiological state are triggered which, for example with a dysregulation. Of cell differentiation, cell growth, cell plasticity or cell regeneration associated.
  • the administration of functional protein according to the invention or at least a higher expression of the same or an inhibition of the cellularly overexpressed or the expressed infunctional protein may be desired.
  • the use of 15B3, 15B3-1 and / or 15B3-2 is very particularly preferred in connection with their function in neuronal cell death, excitation and neurogenesis.
  • the pathophysiology of cerebral ischemia is associated with an increased transcription of 15B3 according to the invention, which is why its use for the treatment of this pathophysiology is very particularly preferred.
  • sequences according to the invention for the production of a medicament for the treatment of tumor diseases and neurological diseases, in particular ischemic conditions (stroke), results.
  • multiple sclerosis neurodegenerative diseases, such as, for example, Parkinson's disease, amyotrophic lateral sclerosis, heredodegenerative ataxias, neuropathies, Huntington's disease, epilepsy and Alzheimer's disease, viral infectious diseases, for example hepatitis infections (claim 19).
  • Another object of the present invention is related to the therapeutic use of sequences according to the invention, namely the use of a nucleic acid sequence or protein sequence according to the invention, in particular the nucleotide sequence or amino acid sequence of protein 15B3, 15B3-1 or 15B3-2 or a variant - as defined above - Of these, in particular a fragment, for gene therapy in mammals, for example in humans, or also such gene therapy processes.
  • Gene therapy encompasses all forms of therapy which either introduce sequences according to the invention according to one of claims 1 to 4 into the body or parts thereof, for example individual tissues, or the expression of invented affect sequences according to the invention.
  • oligonucleotides for example antisense or hybrid RNA-DNA oligonucleotides, with any modifications which contain sequences according to the invention.
  • Viral constructs containing a sequence according to the invention can also be used.
  • Corresponding naked DNA sequences according to the invention are also considered in the context of gene therapy. Nucleic acid pieces with enzymatic activity (eg ribozymes) can also be used for gene therapy purposes.
  • nucleic acids or polypeptides according to the invention protein heteromers according to the invention and reagents according to the invention (oligonucleotides, antibodies, peptides) derived therefrom are also possible, for example for the diagnosis of human diseases or of genetic predispositions, for example. also in the context of pregnancy examinations. These diseases or predispositions are, in particular, those mentioned above in connection with therapeutic applications.
  • diagnostic methods can be designed as in vivo, but typically ex vivo methods. Ex vivo will serve a typical application of a diagnostic method according to the invention for the qualitative and / or quantitative detection of a nucleic acid according to the invention in a biological sample.
  • Such a method preferably comprises the following steps: (a) incubation of a biological sample with a known amount of nucleic acid according to the invention or a known amount of oligonucleotides which are suitable as primers for an amplification of the nucleic acid according to the invention, (b) after the nucleic acid according to the invention by specific hybridization or PCR amplification, (c) comparison of the amount of hybridizing nucleic acid or of nucleic acid obtained by PCR amplification with a quantity standard.
  • the invention also relates to a method for the qualitative and / or quantitative detection of a protein heteromer or a protein according to the invention in a biological sample, which comprises the following steps: (a) Incubation of a biological sample with an antibody which is specific to the protein heteromer or to the invention Protein / polypeptide is directed, (b) detection of the antibody / antigen complex, (c) comparison of the amounts of the antibody / antigen complex with a quantity standard.
  • a biological sample is usually taken from a healthy organism.
  • the property of the 15B3, 15B3-1 and / or 15B3-2 gene can be used here, that after characteristic pathophysiological stimuli, such as e.g. cerebral ischemia, the mRNA amount of 15B3, 15B3-1 and / or 15B3-2 in
  • Proteins of associated diseases such as stroke
  • the assessment of therapeutic success the grading of a disease.
  • the treatment according to the invention can be used to monitor the treatment of the diseases mentioned above.
  • Sequences according to the invention can be used in methods for determining polymorphisms thereof, for example in humans.
  • the polymorphisms of sequences according to the invention which are determined in this way are not only subject to the disclosure of the present gene invention, but can also be used as a prognostic marker for the diagnosis or for the diagnosis of a predisposition to diseases which are associated with a sequence by means of infunctional expression according to the invention, by expression of sequence sequences according to the invention and / or their overexpression.
  • sequences according to the invention permit research into human hereditary diseases, both monogenic and polygenic diseases.
  • nucleic acids or polypeptides according to the invention are also suitable for scientific use.
  • sequences according to the invention allow, in a manner known to the person skilled in the art, for example to identify sequences related to single or multi-cell organisms via cDNA libraries or to localize sequences related to the human genome.
  • nucleotide sequences according to the invention in particular the sequences of 15B3, 15B3-1 and / or 15B3-2, can thus be used to generate genes for mRNAs which code for these nucleic acids or their functional equivalents, homologs or derivatives, for example in murine and in to isolate, map and correlate the human genome with common methods using homology screening and markers for human hereditary diseases. This enables the gene to be identified as the cause of certain hereditary diseases, which considerably simplifies their diagnosis and enables new therapeutic approaches.
  • hereditary diseases can be diagnosed, which is why they are used as markers.
  • markers For example. there will be a correlation of the chromosomal locus of protein 15B3 in humans (chromosome 9) result in certain hereditary diseases, from which a diagnostic method according to the invention for hereditary diseases arises. The same applies to the proteins according to the invention.
  • test system for scientific use, which is based on amino acid and / or nucleotide sequences according to the invention.
  • the cDNA, the genomic DNA, the regulatory elements of the nucleic acid sequences according to the invention, and also the polypeptide, as well as partial fragments thereof can be used in recombinant or non-recombinant form to develop a test system.
  • Such a test system according to the invention is particularly suitable for measuring the activity of the promoter or of the protein in the presence of the test substance.
  • test systems are preferably simple measurement methods (colorimetric, luminometric, based on fluorescence or radioactive) which allow the rapid measurement of a large number of test substances (Böhm, Klebe, Kubinyi, 1996, active ingredient design, Spektrum-Verlag, Heidelberg).
  • the test systems described allow chemical libraries to be searched for substances which have inhibitory or activating effects on proteins according to the invention.
  • the identification of such substances is the first step on the way to the identification of new drugs that specifically affect 15B3, 15B3-1 or 15B3-2-associated signal transduction.
  • simple test systems are conceivable that use the activity of the LLR domain from 15B3, 15B3-1 or 15B3-2.
  • Connection with apoptosis as is desired, for example, in the case of stroke, septic shock, GvHD, degenerative diseases, in particular neurodegenerative diseases, acute hepatitis or other indications disclosed herein, can also be achieved by the fact that oligonucleotides which code for an antisense strand of the native sequences of the proteins 15B3, 15B3-1 or 15B3-2 or another native protein with a domain rich in leucine, to be introduced into the affected cells.
  • the method described above can be used with the aid of recombinant viruses.
  • ribozymes are used which can cut a target mRNA.
  • ribozymes are therefore disclosed and represent an object of the present invention which can cleave native 15B3 (or 15B3-1 or -15B3-2) mRNA or the mRNA of proteins containing domains rich in leucine.
  • Ribozymes according to the invention must be able to interact with the target mRNA according to the invention, for example via base pairing, and then cleave the mRNA in order to block the translation of 15B3, for example.
  • the ribozymes according to the invention are introduced into the target cells via suitable vectors (in particular plasmids, modified animal viruses, in particular retroviruses), the vectors, in addition to possibly other sequences, having a cDNA sequence for a ribozyme according to the invention).
  • a chemical compound according to the invention will in particular modulate the intracellular function of the proteins 15B3, 15B3-1 and / or 15B3-2, typically inhibit (claim 20) or influence the biological function at the level of the underlying DNA sequences according to the invention.
  • Compounds according to the invention will typically bind specifically to a protein with an amino acid sequence according to FIGS. 9, 11 or 13 or to a nucleic acid sequence according to FIGS. 8, 10 or 12 and thereby a pharmacological, in particular neuroprotective or immunomodulating, anti-apoptotic or anti-proliferative Effect.
  • Chemical compounds are therefore disclosed according to the invention, preferably an organic chemical compound with a molecular weight of ⁇ 5000, in particular ⁇ 3000, in particular ⁇ 1500, which is typically well tolerated physiologically and can preferably pass the blood-brain barrier 21). Possibly. it will be part of a composition with at least one further active ingredient and preferably auxiliaries and / or additives and can be used as a medicament.
  • the organic molecule will be particularly preferred if the binding constant for the Binding to a protein according to the invention, in particular to the leucine-rich domain of a protein according to the invention, is at least 10 7 mol -1 .
  • the compound according to the invention will preferably be such that it can pass through the cell membrane, be it by diffusion or via (intra) membranous transport proteins (claim 22), if appropriate after appropriate modification, for example with a coupled AS sequence.
  • such compounds occupy positions on the surface of proteins according to the invention or cause a local conformational change in the proteins according to the invention, so that the binding of a native binding partner to a protein according to the invention is prevented.
  • Structural analyzes of the protein according to the invention can be used to specifically find compounds according to the invention which have a specific binding affinity (rational drug design (Böhm, Klebe, Kubinyi, 1996, active ingredient design, Spektrum Verlag, Heidelberg)).
  • a specific binding affinity rational drug design (Böhm, Klebe, Kubinyi, 1996, active ingredient design, Spektrum Verlag, Heidelberg)
  • the structure or a partial structure, derivative, allele, isoform or part of one of one of the proteins shown in FIGS. 9, 11 or 13 is determined by means of NMR or X-ray crystallography methods or, if such a high-resolution structure is not available, with the help of structure prediction algorithms creates a structural model of a protein according to the invention, and uses this (s) to identify compounds with the support of molecular modeling programs for which a high affinity for the protein according to the invention can be predicted. These substances are then synthesized and tested in suitable test procedures for their binding capacity and their therapeutic usability.
  • the compound according to the invention is an antibody, preferably an antibody directed against a protein according to the invention, for example 15B3, 15B3-1 or 15B3-2, or also against the underlying mRNA, which retransplanted ex vivo Host cells or by gene therapeutic in vivo methods is introduced into host cells and is not secreted there as "intrabody”, but can exert its effect intracellularly.
  • introduction preferably an antibody directed against a protein according to the invention, for example 15B3, 15B3-1 or 15B3-2, or also against the underlying mRNA, which retransplanted ex vivo Host cells or by gene therapeutic in vivo methods is introduced into host cells and is not secreted there as "intrabody”, but can exert its effect intracellularly.
  • Such a procedure will typically be considered for cells of those tissues which show apoptotic behavior that is exaggerated in the patient's physiology, that is to say, for example, in cells of the pancreas, keratinocytes, connective tissue cells, immune cells, neurons or muscle cells.
  • cells modified in this way with "intrabodies" according to the invention are also part of the present invention.
  • a compound according to the invention with the function of blocking the biological function of native 15B3, 15B3-1 or 15B3-2 according to FIGS. 9, 11 or 13 or corresponding native alleles or native splice variants, for example the apoptotic function, can be used as a medicament .
  • All the aforementioned variants are included as compounds here, for example, organic chemical compounds, antibodies, anti-sense oligonucleotides, ribozymes.
  • a compound according to the invention for producing a nes drug for the treatment of diseases for which at least tw. a pathological hyperapoptotic or hyper necrotic or inflammatory reaction is causal or symptomatic.
  • An inhibitor according to the invention for example an antibody with an inhibitory effect (in particular an intrabody), a ribozyme, anti-sense RNA, dominant-negative mutants or one of the aforementioned inhibitory organic chemical compounds
  • an inhibitor according to the invention can inhibit the cellular function of a native protein 15B3, 15B3 according to the invention -1 or 15B3-2 or its native variants, for example the a-poptotic, necrotic or proliferative reaction, are used as medicaments and very particularly in the treatment of the following diseases or in the manufacture of a medicament for the treatment of the following diseases: tumor diseases , Autoimmune diseases, especially diabetes, psoriasis, multiple sclerosis, rheumatoid arthritis or asthma, viral infectious diseases (e.g.
  • apoptosis-inhibiting substances according to the invention can also be part of a pharmaceutical composition which can contain further pharmaceutical excipients and / or additives in order to stabilize such compositions for therapeutic administration, for example to stabilize the biological availability and / or to improve pharmacodynamics.
  • the present invention furthermore relates to methods (“screening methods”) for identifying pharmaceutically active compounds, in particular with inhibitory properties with regard to triggering or Transmission of signals which are related to apoptotic reactions triggered by physiologically occurring sequences according to the invention (claim 23).
  • Methods according to the invention provide that (a) cells with an expression vector according to claim 5, in particular an expression vector which codes for the polypeptide 15B3, 15B3-1 or 15B3-2, and optionally at least one expression vector which is for at least one reporter Gene are encoded, transfected, and (b) a parameter suitable for observing the 15B3-mediated function, for example apoptosis, in particular the activation of caspase-3 after addition of a test compound compared to the control without addition of a test compound for the (a) the host cell system obtained is measured.
  • “high-throughput screening assays” for identifying inhibitors of 15B3 are disclosed.
  • Systems with the “Scintillation Proximity Assay” (SPA) are preferably available (Amersham Life Science, MAP kinase SPA (see McDonald et al., 1999, Anal Biochem, 268, 318-29)) describes an assay set-up for the Raf / MEK / ERK cascade that can identify inhibitors of the entire cascade. tion of the present invention.
  • the MAP cascade is reconstituted in vitro, with the individual components as GST fusion proteins (E. coli expressed) or, in the case of cRAF1, with the baculovirus system (claim 25).
  • the first element of the cascade (MAP-KKK) must be constantly activated in order to be able to reliably test inhibitors. Typically this is achieved by coexpression of src in the baculovirus system. This ensures a ras-analog activation of cRaf.
  • a modulation of the cascade is brought about, which is used in order to be able to measure an influence on this modulation in an HTS by adding substances to be tested.
  • the binding sites of the identified and pharmacologically active substances to the gene products according to the invention in particular 15B3, 15B3-1 or 15B3-2, can be determined with the aid of the two-hybrid system or other assays, ie that those responsible for the interaction Amino acids are limited (claim 24). In this way, substances can also be found with which the interaction between polypeptides according to the invention and any native intracellular interaction partners thereof can be influenced, in particular inhibited.
  • a target for a pharmaceutically active compound is therefore preferably the regulation of transcription, for example by specific binding of the substances to a regulator region (for example promoter or enhancer sequences) of a gene product according to the invention, binding to one or more transcription factors of a gene product according to the invention (with the result of an activation or inhibition of the transcription factor) or a regulation of the expression (transcription or translation) of such a transcription factor itself.
  • a pharmaceutically active compound according to the invention can also intervene in other control processes of the cell which, for example, affect the expression rate of a protein according to the invention.
  • can flow eg translation, splicing processes, native derivatization of gene product according to the invention, for example phosphorylation, or regulation of the degradation of gene product according to the invention.
  • the present invention further relates to methods for identifying cellular interaction partners of polypeptides according to the invention, in particular the proteins 15B3, 15B3-1 or 15B3-2 or their natively occurring variants (isoforms, alleles, splice forms, fragments) (claim 27) ,
  • proteins can be identified as interaction partners which have specific binding affinities for the protein according to the invention, or for the identification of nucleic acids which code for proteins which have specific binding affinities for the protein according to the invention.
  • Such a method according to the invention or the use of polypeptides according to the invention, nucleic acid sequences according to the invention and / or nucleic acid constructs according to the invention for carrying out such methods is preferably carried out with the aid of a "yeast-two-hybrid" screening (y2h "screens") alone or in combination with other biochemical Procedures performed (Fields and Song, 1989, Nature, 340, 245-6).
  • y2h "screens” screening y2h "screens” screening
  • Such screens can also be found in Van Aelst et al. (1993, Proc Natl Acad Sei U S A, 90, 6213-7) and Vojtek et al. (1993, Cell, 74, 205-14).
  • mammal systems can also be used to carry out a method according to the invention instead of yeast systems, for example as in Luo et al. (1997, Biotechniques, 22, 350-2).
  • the open reading frame of 15B3, 15B3-1, 15B3-2 or a native variant is cloaked, for example, in a so-called bait vector "in-frame" with the GAL4 binding domain.
  • a so-called bait vector "in-frame” with the GAL4 binding domain e.g. pGBTIO, from Clontech.
  • a so-called “prey library” in a yeast strain can thus preferably be searched for interacting proteins according to the customary protocol.
  • Kinase-negative mutants are preferably used for this, since these often interact more stably with any phosphorylation targets.
  • Serine / threonine kinases in a metabolic pathway can be caused by Adapter molecules are brought into close proximity in order to be able to carry out specific phosphorylations better (Chang et al., 1998, Science, 281, 1860-3), (Yasuda et al., 1999, Mol Cell Biol, 19, 7245-54, ( Whitmarsh and Davis, 1998, Trends Biochem Sei, 23, 481-5), Whitmarsh et al., 1998, Science, 281, 1671-4) It is therefore also possible according to the invention, via two steps in the yeast-two-hybrid system to encounter phosphorylation targets by first cloning an adapter protein and using this to determine the specific target protein (“target”) as “bait”. In addition, so-called mapping experts can also be used with y2h systems imente to identify specific interaction domains.
  • interaction partners can also share
  • Immunoprecipitations from cells transfected with 15B3-, 15B3-1- or 15B3-2- are used to purify proteins that bind to them, and subsequently using protein sequencing methods (eg MALDI-TOF, ESI-tandem-MALDI) identify the associated genes.
  • protein sequencing methods eg MALDI-TOF, ESI-tandem-MALDI
  • Another object of the present invention is therefore the use of the two-hybrid system or other biochemical methods for the identification of interaction domains of 15B3, 15B3-1 or 15B3-2 or natively occurring variants thereof and the use of these interaction domains (Fragments of the native sequences) for pharmacotherapeutic intervention.
  • polypeptides according to the invention in particular natively occurring forms or also non-native, artificially generated variants whose biological function is to be investigated, can be used according to the invention in an apoptosis assay or in a method for examining the function and / or effectiveness of polypeptides according to the invention are used for induction, transduction or inhibition of cell death signals.
  • the involvement of 15B3, 15B3-1, 15B3-2 or the aforementioned variants in apoptotic cascades can be examined by transfecting expression constructs with 15B3, 15B3-1 or 15B3-2 or variants in eukaryotic cells, and then examining the induction of apoptosis can.
  • apoptosis may be cell type-specific, which is why several cell lines and primary cells are preferably investigated according to the invention.
  • the induction of apoptosis can also be stimulus-specific. Therefore, in a method according to the invention, preferably several stress situations are taken as a basis, for example heat shock, hypoxia conditions, cytokine treatments (for example 11-1, 11-6, TNF-alpha, H 2 O 2 treatment).
  • Typical cell types for such a method according to the invention are customary cell lines, for example Cos cells, HEK cells, PC12 cells, THP-1 cells, or primary cells, such as neurons, astrocytes, as well as others in - Mortalized and primary cell lines as needed.
  • the present invention further relates to the use of nucleic acids according to the invention, nucleic acid constructs or gene products according to the invention for carrying out a proliferation assay. For example.
  • 15B3, 15B3-1, 15B3-2 and native or non-native variants thereof in the growth of cells, in the passage of the cell cycle or in tumorigenic transformation can be examined by using expression constructs with 15B3, 15B3-1 or 15B3-2 or equivalent Variants are transfected into eukaryotic cells and then, for example, the induction of tumorigenicity is examined, for example with the aid of a soft agar test (Housey, et al., 1988, Adv Exp Med Biol, 234, 127-40).
  • Common cell lines such as Cos cells, HEK cells, PC12 cells, THP-1 cells, and primary cells such as neurons, astrocytes, as well as other immortalized and primary cell lines as required, are considered as preferred cell types.
  • the function of gene products according to the invention on the ras signal transmission path and the interaction of gene products according to the invention with other components of the ras signal transmission path can be investigated, in particular with regard to proliferative or apoptotic processes.
  • Another object of the present invention is the use of a DNA sequence according to one of claims 1 to 4 or a gene product according to one of claims 8 to 10 as a suicide gene / protein for the in vivo or ex vivo transformation of host cells (claim 29).
  • a DNA sequence according to one of claims 1 to 4 or a gene product according to one of claims 8 to 10 as a suicide gene / protein for the in vivo or ex vivo transformation of host cells (claim 29).
  • cell death can be triggered in a targeted manner in host cells.
  • a DNA sequence according to the invention or a protein according to the invention be designed such that the suicide gene is operably linked to a promoter, the transcription being repressed and being activated only when necessary (claim 30).
  • the transfected cell can be specifically switched off after transplantation of patient cells ex vivo or in vivo as part of a gene therapy.
  • FIG. 1 shows an RMDD scheme (restriction-mediated differential display). A description is given in embodiment 1.
  • the result of a 15B3 LightCycler verification is shown in FIG.
  • the upregulation of 15B3 was measured 2 hours after an experimental stroke (MCAO, 90 min occlusion), specifically in the contralateral and in the ipsolateral area.
  • the expression (on the y axis) is normalized against the expression of the standard gene product cyclophilin, ie plotted as a relative expression.
  • FIG. 3 shows the 15B3 tissue expression in various mouse tissues (heart, spleen, brain, lung, liver, skeletal muscle, kidney, testis) or after various phases of mouse embryonic development. The result was determined using a quantitative PCR with 15B3 primers on a cDNA set from Fa. Clontech obtained, ie the gene presented, 15B3, is ubiquitously expressed in humans and mice. The expression rate against the individual tissues is plotted.
  • FIG. 4 shows a homology comparison at the protein level of the proteins 15B3, 15B3-1 and 15B3-2 with the aid of the Clustal program (Megalign, Lasergene, Madison, WI).
  • the amino acids which are identical in each case are shaded with a dark background.
  • the lines drawn above or below the sequence represent those areas (a total of 4, approximately between (1) AS 234 and 254, (2) 45 and 63, (3) 24 and 43 and (4) 184 and 203 ( Numbering according to the sequence of 15B3-2), which are referred to as transmembrane domains, lines with arrows (10 in total) mark regions which are leucine-rich regions ("leucine-rich repeats" (LRR)).
  • Interaction modules are known in proteins with various functions, for example the human and murine toll-like receptor 6 (TLR6) contains an extracellular LRR domain in addition to the transmembrane domains (Takeuchi et al., 1999).
  • FIG. 5 shows a phylogenetic family tree of the proteins with "leucine-rich-repeat” domains.
  • the Megalign program was used to determine the family tree.
  • the 15B3 family represents a new subset within the proteins containing LRR domains.
  • FIG. 6 shows a protein structure prediction for protein 15B3 according to the invention with the aid of the Protean program (DNAStar, Lasergene, Madison, WI). The 4 putative transmembrane domains are clearly visible.
  • Figure 7 Ras-MAPK cascade. Putative interactions of 15B3 within the Ras signal transduction path are shown. 15B3 has modulatory functions here.
  • Figure 8 represents the human 15B3 nucleic acid sequence (SEQ ID 1).
  • Figure 9 depicts the amino acid sequence of human 15B3 protein (SEQ ID 2) running from the N to the C terminus.
  • Figure 10 represents the human nucleotide sequence of human 15B3-1 (Seq ID 3).
  • FIG. 11 shows the amino acid sequence of the human protein 15B3-1 (Seq ID 4), namely continuously from the N- to the C-terminus.
  • Figure 12 depicts the nucleotide sequence of protein 15B3-2 (Seq ID 5).
  • Figure 13 shows the amino acid sequence of protein 15B3-2 (Seq ID 6), running from the N to the C terminus.
  • RNA products of the 15B3 gene were increasingly expressed and could be cloned using a differential cloning system (restriction-mediated differential display, RMDD).
  • the thread model (middle cerebral artery occlusion, MCAO) was used to induce focal cerebral ischemia, a valid model for stroke disease in humans.
  • This experimental system was used according to the invention to identify genes which, in addition to other functions, are also of pathophysiological importance for events after cerebral ischemia.
  • the experimental procedure was as follows, and molecular targets for new drugs in neurodegenerative diseases can be represented.
  • PCR samples from the ischemic and contralateral hemisphere were applied to the gel (24 h MCAO right and left and 90 min MCAO right and left). Bands of different intensities in the right or left hemisphere were cut out and the corresponding PCR product was reamplified. Amplified products obtained were in the TOPO TA vector pcDNA 2.1 (Fa. Invitrogen) cloned and sequenced with T7 and M13rev primers (ABI 3700 capillary electrophoresis sequencer). Sequences obtained are compared with the EMBL database.
  • This sequence showed strong homology to the SUR-8 etc. genes.
  • the isolated 3 'PCR fragment was used to hybridize a mouse brain bank. Several clones were found that contained parts of the sequence from this sequence (Seql). These were compared with ESTs from the EMBL database, and the sequences Seql assembled. A mouse sample from the 5 'region was used to search a human fetal brain bank in lambdaZapII (Stratagene) ("screening"). An illustration of the preparation of the human cDNA library used is shown in FIG. 1. The human clones obtained were sequenced and compared with human ESTs from the EMBL database and the human mRNA of the KIAA1437 protein AB037858.
  • nucleotide and protein sequences of the 15B3-related clones 15B3-.1 and 15B3-2 yielded a database search of the EMBL, nrdb and swissprot databases (with the BLAST 2 program, described by Altschul et al. (1997, Nucleic Acids Res, 25, 3389-402) This publication is fully part of the present disclosure, and further human ESTs were determined using the EMBL database, so that for 15B3-2 the ORF could be averaged. These sequences are shown as sequences Seq ID 5, 6 in FIGS. 10 to 13.
  • the ligation was transformed by electroporation into E. coli (DH10B, Gibco) and amplified on LB ampicillin agar plates.
  • the plasmid DNA was isolated by means of alkaline lysis and ion exchange chromatography (QIAfilter kit, from Qiagen).
  • the complexity of single clones was 4 million for the fetal human brain cDNA library. From each cDNA library 24 individual clones were randomly analyzed for insert sizes, which showed a size distribution from 800 bp up to 4.5 kb, the average length of the cDNA inserts for the human Bajik was approximately 1.2 kb.
  • Embodiment 2 Carrying out a reporter gene assay
  • the sequence obtained from 15B3 was used to obtain information about the classification of the protein in its function as E- element of signal transduction cascades, in particular the ras / MapK cascade.
  • the open reading frame of the gene was cloned into a common expression vector (eg pCMV-tag, from Stratagene).
  • This construct was transfected together with other constructs into eukaryotic cells (eg using the calcium phosphate method, see Ausubel et al., Current Protocols in Molecular Biology, New York, 1997).
  • Extracts from the cells were then subjected to a measurement in a luminometer (for example from Bertold).
  • a luminometer for example from Bertold.
  • An increase in the luciferase value was assessed as influencing the signal transduction pathway, which results in the activation of a specific transcription factor (ELK).
  • ELK specific transcription factor
  • reporter assays have also been carried out with other systems, e.g. lacZ or chloramphenicol transferase (CAT assays), the assay being based on the same principle as described above.
  • lacZ lacZ
  • CAT assays chloramphenicol transferase
  • the quantitative analysis (FIG. 3) shows a relatively ubiquitous tissue distribution with a focus on the heart, lungs, liver and brain. 15B3 expression is also detectable in the testis, muscle, kidney and spleen as well as during embryonic development.
  • a homology search with the human 15B3 sequence identified human ESTs (expressed sequence tags) from the 3 'untranslated region of the gene. There are clones from aorta, bone, brain, CNS, colon, eye, genital organs, heart, kidney, lungs, lymphatic system, pancreas, prostate, stomach, testis, uterus, embryo, breast tissue, muscle, nervous tumor, ovary, Uterus, which suggest a broad distribution of tissue. This coincides with the quantitative PCR data obtained.
  • the computer program T pred was used to determine transmembrane domains. It predicts with high probability 4 transmembrane domains for the N-terminus of protein 15B3 (see FIG. 6).
  • a "score" above 500 indicates the significance of potential transmembrane segments, for the protein 15B3 the "score” for all four potential domains is very high (1361, 1965, 757, 2592).
  • the nucleotide sequence of clone BAA92675 shows a nucleotide exchange in position 2748bp (C instead of T) and in position 3891 bp (A instead of G).
  • the errors in the nucleotide sequence do not result in an amino acid exchange at the protein level.
  • the annotated protein sequence of 476 AA from clone Q92627 did not show a complete ORF.
  • the start codon was determined and the ORF on 804 AS was completed by translating the 5 'genomic sequences found. There is also a certain homology to 15B3 in the N-terminal sequences.
  • the protein sequence also has four potential conserved transmembrane segments, 10 leucine-rich repeats and a motif similar to "ER membrane retention signal".
  • the protein sequence of clone BAA9131 starts with a start codon, however, based on the sequence comparisons to 15B3 and Q92627, it is assumed that the ORF in the N-terminus is incomplete.
  • the protein sequence shows strong conservation to the potential transmembrane segments (3 and 4, Fig. 6) and also has 10 leucine-rich repeats. Due to the conservation at the protein level, especially within the potential transmembrane domains and the LRR domains, it can be assumed that 15B3 with BAA9131 and Q92627 belong to a common gene family and define a new subgroup (FIG. 5). For this reason, BAA9131 was renamed to 15B3-1 and Q92627 to 15B3-2.
  • H. sapiens PID: gl504042 - similar to yeast adenylate cyclase (69% / 148 aa)
  • M. muscle PID: g3252981 - Ras-binding protein SUR-8 (35% / 141 aa)
  • E. coli PID: gl788752 - cell division protein involved in FtsZ ring (34% / 123 aa)
  • Soc-2 a protein with an 18 tandem LRR domain, suppresses the activated form of the EGL-15 FGF receptor (fibroblast growth factor).
  • the FGF signal cascade is also said to have an activating effect on the Ras cascade.
  • the human form Shoc-2 is localized to chromosome 10q25 in humans, a region that is associated with certain bones and cancer.
  • the other subgroups of the LLR proteins are the LAP proteins, which have both an LLR domain in the N-terminus and a PDZ domain in the carboxy terminus.
  • the LAP proteins include the C. elgans protein Let-413, the Sophosila protein Scribble, the vertebrate proteins Densin-180, Erbin and the human Scribble. Interestingly, the LAP proteins have a common expression pattern along the cell membrane.
  • Genomic structure of 15B3 in humans A database search based on the knowledge according to the invention with the BLAST 2 program (Altschul, et al., 1997, Nucleic Acids Res, 25, 3389-402)) in the EMBL database yielded additional human genomic sequence information.
  • the clone AL136108 consisting of 36 composite contigs, contains large areas of the 15B3 cDNA. Sequence comparisons between the genomic nucleotide sequence indicate that the human cDNA clone is encoded by at least two exons. The 3 'area is completely preserved. 15B3 is located on chromosome 9 in humans (position: 9q22.32-31.3; http: // www.sanger.ac.uk/cgi-bin/humace/searcher. Cgi).
  • the Ras associated LRR domain of S. Cerevisiae adenylyl cyclase belongs to a small subgroup that has a specific 23-AS consensus unit (PXXaXXLXXLXXLXLNXLXXa, a is an aliphatic, X any amino acid).
  • Other members of this subgroup, such as RSU1 and SUR-8, are two putative scaffold proteins that have a modulating effect on the Ras-dependent signal cascade. Both have in common the formation of a ternary complex with Ras and Raf and the resulting activation of Ras-MAP kinase signal transduction.
  • Ras and the Raf / Erk cascade also has an essential role for the oxygen-glucose deficiency tolerance in neurons.
  • a signal cascade is started, which is activated by the vation of the NMDA receptors is triggered by Ca2 + inflow and production of NO.
  • NO is a mediator of Ras activation using NMDA receptor stimulation.
  • the ischemic tolerance is achieved by activating the NMDA glutamate receptors.
  • the proteins according to the invention contain a tandem leucine repeat-rich domain (LRR) in the C-terminus.
  • LRRs in the protein according to the invention are protein-protein interaction domains consisting of 20-28 amino acids with a core consensus of leucines and asparagines (LXXLXLXXN). In the 15B3 protein, this domain typically consists of 10 leucine rich repeats (FIG. 4).
  • the experimental data according to the present invention show that the identified proteins play a crucial role in the regulation of focal cerebral ischemia, a stroke model.
  • Proteins 15B3, 15B3-1 and 15B3-2 were used to identify drug target sequences using a method for cloning differentially regulated genes (RMDD) in the ischemic hemisphere of mice after focal cerebral ischemia.
  • RMDD differentially regulated genes
  • the regulation of gene expression plays a crucial role in the course and extent of neuronal damage in cerebral ischemia (Koistinaho and Hokfelt, 1997, Neuroreport, 8, i-viii; Schneider et al., 1999, Nat. Med., 5, 554-9).
  • So-called “immediate early genes” in particular play a role here (Atkins et al., 1996, Stroke, 27, 1682-1687), such as cox-2, (Nogawa, et al., 1997, J.
  • the Animal model of focal cerebral ischemia represents a valid model for human ischemic stroke.
  • the so-called thread model was used, in which a coated nylon thread was advanced through the internal carotid artery to the outlet of the cerebral artery and induces an ischemic stroke (Clark et al., 1997, Neurol. Res., 19, 641-648).
  • a quantitative PCR was carried out using the LightCycler TM system (Röche Diagnostics, Mannheim).
  • cDNA content of the samples was normalized for the expression of cyclophilin and S20 (Schneider, et al., 1995, Proc Natl Acad Sei U S A, 92, 4447-51).
  • Primers used for the amplification of cyclophilin were: cyc5 ACCCCACCGTGTTCTTCGAC acyc300 CATTTGCCATGGACAAGATG and for the amplification of mouse 15B3: seq_15B3.1 GAGCTGCCAAGAAAGGGGGAGACT (in Exon 2) seq_15B3 2AT CAGGAAConAg (in Exon 2) a 'End of mouse cDNA is).
  • the result shows a clear upregulation by a factor of 2-3 of 15B3-RNA on the ischemic (left) hemisphere 2h after the ischemic event (middle cerebral artery occlusion of 90 min and 2 h reperfusion, i.e. in one Acute neurodegeneration model (Fig. 2); the error bars show standard deviations, these result from measurements with 3-fold serial diluted cDNA samples and thus reflect the reliability of the measurement results). After 24 hours (in a permanent model), however, no difference could be determined. Proteins according to the invention, for example 15B3, play an important role in intracellular signal transduction processes, which is why they are also involved in pathological processes (for example stroke).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Neurosurgery (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurology (AREA)
  • Hospice & Palliative Care (AREA)
  • Genetics & Genomics (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Psychiatry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to DNA sequences, expression vectors containing the inventive DNA sequences, host cells, gene products of the cited DNA sequences coding for signal transduction proteins, and other derivatives of the cited objects of the invention. Said DNA sequences and gene products either have a protective action, i.e. impart a certain resistance to apoplexy, or they cause an increase in ischaemic damage. The invention also relates to the use of the inventive DNA sequences or the signal transduction proteins expressed by said DNA sequences as pharmaceuticals, and to the non-therapeutic uses of the inventive DNA sequences or gene products.

Description

Signaltransduktionsproteine 15B3, 15B3-1 und 15B3-2 und zugrundeliegende DNA-Sequenzen Signal transduction proteins 15B3, 15B3-1 and 15B3-2 and underlying DNA sequences
Die vorliegende Erfindung betrifft (i) DNA-Sequenzen, (ii) Expressionsvektoren, die erfindungsgemäße DNA-Sequenzen enthalten, (iv) Wirtszellen, die erfindungsgemäße Expressionsvektoren aufweisen, (v) Genprodukte, die durch erfindungsge- mäße Sequenzen codiert werden, (vi) hinsichtlich erfindungsgemäßer Sequenzen veränderte transgene Tiere, (vii) gegen erfindungsgemäße Genprodukte gerichtete Antikörper, (viii) Verfahren zur Expression bzw. Isolierung von erfindungsgemäßen Genprodukten, (ix) die Verwendung von erfindungsgemäßen DNA- Sequenzen bzw. Genprodukten als Arzneimittel, (x) pharmazeutisch wirksame Verbindungen und Verfahren zu deren Herstellung sowie Verwendungen derartiger erfindungsgemäßer Verbindungen und (xi) nicht-therapeutische Verwendungen erfindungsgemäßer DNA-Sequenzen bzw. Genprodukte.The present invention relates to (i) DNA sequences, (ii) expression vectors which contain DNA sequences according to the invention, (iv) host cells which have expression vectors according to the invention, (v) gene products which are encoded by sequences according to the invention, (vi) transgenic animals modified with respect to sequences according to the invention, (vii) antibodies directed against gene products according to the invention, (viii) methods for the expression or isolation of gene products according to the invention, (ix) the use of DNA sequences or gene products according to the invention as medicaments, (x) pharmaceutically active Compounds and processes for their preparation and uses of such compounds according to the invention and (xi) non-therapeutic uses of DNA sequences or gene products according to the invention.
Es ist aus dem Stand der Technik bekannt, daß die Pathophysi- ologie des Schlaganfalls, daß der Ras/Erk Signaltransdukti- onsweg einen großen Einfluß auf NMDA Rezeptor/NOS Aktivierung und die neuronale ischämische Prekonditionierung hat. Die Proteine der Ras/Erk-Kaskade stellen pharmakologische targets dar, da aufgrund definierter biochemischer Funktionen spezifische Inhibitoren gefunden werden können.It is known from the prior art that the pathophysiology of the stroke, that the Ras / Erk signal transduction pathway has a great influence on NMDA receptor / NOS activation and the neuronal ischemic preconditioning. The proteins of the Ras / Erk cascade represent pharmacological targets because specific inhibitors can be found based on defined biochemical functions.
Der Schlaganfall stellt die dritthäufigste Todesursache und die Hauptursache für Behinderungen in den westlichen Industrienationen dar. Jedes Jahr erleiden in Deutschland ungefähr 250 000 Menschen einen Schlaganfall, ein Drittel stirbt innerhalb des ersten Monats daran, 60% behalten Be- hinderungen zurück. Bislang gibt es keine wirksame Therapie für die Mehrzahl dieser Patienten. Die Mehrzahl aller Schlaganfälle in den westlichen Ländern beruht auf Störungen des cerebralen Blutflusses durch Thrombosen oder Embo- lien. Aus diesem Grund beruhen viele der bisherigen Therapiekonzepte auf der Thrombolyse (bspw. mittels Strep- tokinase, rtPA, Ancrod) . Aus dem Stand der Technik ist allerdings nunmehr auch bekannt, daß bei der Mehrzahl der "Schlaganfälle eine spontane Reperfusion auftritt. Neuere Ar- beiten bei Nagetieren lassen auch vermuten, dass der Schaden durch die Reperfusion in einem gewissen Zeitfenster eher größer wird.Stroke is the third leading cause of death and the main cause of disabilities in western industrialized nations. Every year around 250,000 people in Germany suffer from a stroke, a third die within the first month, 60% retain hindrances back. So far, there is no effective therapy for the majority of these patients. The majority of all strokes in Western countries are due to cerebral blood flow disorders caused by thrombosis or embolism. For this reason, many of the previous therapy concepts have been based on thrombolysis (for example by means of streptokinase, rtPA, Ancrod). From the prior art, however, Newer Ar is now also known that a spontaneous reperfusion occurs in the majority of "strokes. BEITEN in rodents also suggest that the damage is rather larger by reperfusion in a certain time window.
Die klinische Wirksamkeit ist für die Thrombolyse teilweise erbracht, hat jedoch nicht zu einer Therapie geführt, die bei der Mehrzahl der Schlaganfälle eingesetzt werden kann. Neuere Forschungsentwicklungen stellen daher auf die Entwicklung neuer Medikamente ab, die direkt das Zelltodprogramm in Neuronen beeinflussen.Clinical efficacy has been partially demonstrated for thrombolysis, but has not led to therapy that can be used in the majority of strokes. Recent research developments are therefore based on the development of new drugs that directly influence the cell death program in neurons.
Um neuartige Konzepte für die Behandlung der cerebralen Ischämie, deren ausgeprägteste Form der Schlaganfall darstellt, zu entwickeln, werden in neueren Modellsystemen der cerebralen Ischämie Veränderungen der Genexpressi- on registriert, die in funktionellem Zusammenhang mit dem Ausmaß der Hirnschädigung stehen. Das beste verfügbare Tierraodell ist das sog. Fadenmodell in Nagetieren ("fila- ment model") bei dem ein beschichteter Nylonfaden die Abzweigung der A. cerebri media verschließt. Durch Zurückzie- hen des Fadens wird eine Wiederdurchblutung des Infarktgebietes möglich. Aus der Analyse des Transkriptoms zu unterschiedlichen postischämischen Zeitpunkten können neue therapeutische Targets identifiziert werden, die neue Behand- lungswege für die Akuttherapie erschliessen. Einzelne Genprodukte, die einer Regulation während des Präkonditionierens unterliegen, konnten bereits identifiziert werden (z.B. IL- lra, TIMP-1, hsp-72) . Bislang ist jedoch eine protektive Wir- kung der identifizierten Proteine nicht eindeutig nachgewiesen worden.In order to develop novel concepts for the treatment of cerebral ischemia, the most pronounced form of which is stroke, changes in gene expression are registered in newer model systems of cerebral ischemia, which are functionally related to the extent of brain damage. The best available animal model is the so-called filament model, in which a coated nylon thread closes the branch of the cerebral artery. By pulling back the thread, blood circulation to the infarct area is possible. New therapeutic targets can be identified from the analysis of the transcriptome at different post-ischemic times. open up pathways for acute therapy. Individual gene products that are subject to regulation during preconditioning have already been identified (eg IL-lra, TIMP-1, hsp-72). So far, however, a protective effect of the identified proteins has not been clearly demonstrated.
Die Aufgabe der vorliegenden Erfindung ist es, weitere Proteine und deren zugrundeliegenden Nukleotidsequenzen zu identi- fizieren, die protektive Wirkung haben, insbesondere eine gewisse Resistenz gegenüber dem Schlaganfall vermitteln, wie auch solche, die eine Ausweitung des ischämischen Schadens bedingen. Eine weitere Aufgabe der vorliegenden Erfindung ist es, auf der Basis identifizierter Proteine Ver- fahren zur Verfügung zu stellen, die es erlauben, Pharmaka zu entwickeln, die in die entsprechende Pathophysiologie therapeutisch eingreifen können. Damit ist auch das Bereitstellen von entsprechenden Pharmaka eine Aufgabe der vorliegenden Erfindung.The object of the present invention is to identify further proteins and their underlying nucleotide sequences which have a protective effect, in particular impart a certain resistance to the stroke, as well as those which cause an expansion of the ischemic damage. Another object of the present invention is to provide methods on the basis of identified proteins which make it possible to develop pharmaceuticals which can intervene therapeutically in the corresponding pathophysiology. The provision of corresponding pharmaceuticals is therefore also an object of the present invention.
Die vorliegende Erfindung löst diese Aufgabe durch die Gegenstände der Ansprüche 1, 5, 6, 8, 11, 12, 15, 16, 17, 20, 23, 26 und 27. Vorteilhafte Ausführungsformen sind in den jeweiligen Unteransprüchen beschrieben.The present invention solves this problem by the subject matter of claims 1, 5, 6, 8, 11, 12, 15, 16, 17, 20, 23, 26 and 27. Advantageous embodiments are described in the respective subclaims.
Ein Gegenstand der vorliegenden Erfindung betrifft Nuklein- säure-Sequenzen, insbesondere DNA-Sequenzen, die einen Sequenzbereich enthalten, der für ein Polypeptid mit einer Aminosäuresequenz von AS 312 bis AS 711 (15B3) , AS 180 bis AS 579 (15B3-1) oder AS 306 bis AS 705 (15B3-2) (Numerierung gemäß Figur 4) codiert, einschließlich aller funktionshomologen Derivate, Fragmente oder Allele. Insbesondere sind alle Nuk- leinsäure-Sequenzen mitumfaßt, die mit den erfindungsgemäßen Sequenzen hybridisieren, einschließlich der jeweils im Doppelstrang komplementären Sequenzen (Anspruch 1) .One object of the present invention relates to nucleic acid sequences, in particular DNA sequences, which contain a sequence region which is suitable for a polypeptide with an amino acid sequence of AS 312 to AS 711 (15B3), AS 180 to AS 579 (15B3-1) or AS 306 to AS 705 (15B3-2) (numbering according to FIG. 4), including all functionally homologous derivatives, fragments or alleles. In particular, all nucleic acid sequences are included, those with the invention Hybridize sequences, including the complementary sequences in the double strand (claim 1).
In einer weiteren bevorzugten Ausführungsform werden DNA- Sequenzen offenbart, deren Genprodukt für ein Polypeptid codiert, wie in einer der Figuren 9, 11 oder 13 für die Sequenzen 15B3, 15B3-1 bzw. 15B3-2 wiedergegeben, einschließlich aller funktionshomologen Derivate, Allele oder Fragmente einer solchen DNA-Sequenz und auch infunktioneller Derivate, Allele, Analoga oder Fragmente (bspw. DN-Varianten), die die physiologische Funktion, bspw. apoptotische Signalkaskade, inhibieren können (Anspruch 2) . Auch mit diesen erfindungsgemäßen DNA-Sequenzen hybridisierende DNA-Sequenzen (einschließlich der Sequenzen des komplementären DNA-Stranges) sind mitoffenbart. Die Herstellung derartiger Derivate, Analoga, Fragmente oder Allele geschieht durch Standardverfahren (Sambrook et al. 1989, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, NY) . Hierbei werden bei den DNA- Sequenzen gemäß Figuren 8, 10 oder 12 ein oder mehrere Co- don(s) insertiert, deletiert oder substituiert, um nach Transkription und Translation ein Polypeptid zu erhalten, das einen Unterschied in bezug auf mindestens eine Aminosäure gegenüber den nativen in den Figuren 9, 11 oder 13 dargestellten Sequenzen aufweist.In a further preferred embodiment, DNA sequences are disclosed whose gene product codes for a polypeptide, as shown in one of FIGS. 9, 11 or 13 for the sequences 15B3, 15B3-1 or 15B3-2, including all functionally homologous derivatives, alleles or Fragments of such a DNA sequence and also infunctional derivatives, alleles, analogs or fragments (for example DN variants) which can inhibit the physiological function, for example apoptotic signal cascade (claim 2). DNA sequences hybridizing with these DNA sequences according to the invention (including the sequences of the complementary DNA strand) are also disclosed. Derivatives, analogs, fragments or alleles of this type are prepared by standard methods (Sambrook et al. 1989, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, NY). 8, 10 or 12, one or more codon (s) are inserted, deleted or substituted in order to obtain, after transcription and translation, a polypeptide which has a difference in at least one amino acid compared to the native sequences shown in Figures 9, 11 or 13.
Zum Erfindungsgegenstand der vorliegenden Anmeldung gehören auch .DNA-Teilsequenzen der in den Figuren 8, 10 oder 12 dargestellten Sequenzen. Diese Teilsequenzen enthalten typischerweise mindestens 60, stärker bevorzugt mindestens 150 und noch stärker bevorzugt mindestens 250 Nukleotide umfassende Fragmente der in den Figuren 8, 10 oder 12 dargestellten Nukleotidsequenzen. Insbesondere codieren bevorzugte Teilsequenzen für Polypeptide, die von AS 312 (15B3), 180 (15B3-1) bzw. 306 (15B3-2) mindestens 20 AS in Richtung N- Terminus verlaufen und ggf. sich bis zum N-Terminus erstrecken können (Numerierung gemäß Figur 4) . Weiterhin kann die für mindestens 20 AS codierende Teilsequenz aber auch an ei- nem von den vorgenannten Punkten weiter proximal oder distal liegenden Codons beginnen. Ganz besonders bevorzugt sind Teilsequenzen, die für mindestens eine LRR codieren, vorzugsweise mindestens 2 und stärker bevorzugt mindestens 5 (LRRs sind in Figur 4 durch unterlegte Pfeile gekennzeichnet) . Ins- besondere bevorzugt sind solche Nukleotidteilsequenzen, die für mindestens eine der in Figur 4 markierten LRRs codieren bzw. Nukleotidsequenzen, die mindestens eine der in Figur 4 indizierten LRRs enthalten. Mitoffenbart sind alle Derivate, Analoga oder Allele der vorgenannten offenbarten Teilsequen- zen. Auch die sich aus diesen erfindungsgemäßen DNA- Teilsequenzen ergebenden AS-Sequenzen sind als solche oder als Bestandteil in größeren rekombinanten Proteinen mitoffenbart. Zum Rahmen der vorliegenden Erfindung gehören auch alle nativen Spleißvarianten der erfindungsgemäßen Sequenzen.The subject matter of the present application also includes partial DNA sequences of the sequences shown in FIGS. 8, 10 or 12. These partial sequences typically contain at least 60, more preferably at least 150 and even more preferably at least 250 nucleotides comprising fragments of the nucleotide sequences shown in FIGS. 8, 10 or 12. In particular, preferred partial sequences for polypeptides encode that of AS 312 (15B3), 180 (15B3-1) or 306 (15B3-2) run at least 20 AS in the direction of the N-terminus and possibly extend to the N-terminus (numbering according to Figure 4). Furthermore, the partial sequence coding for at least 20 AS can also begin at a codon lying further proximally or distally from the aforementioned points. Partial sequences which code for at least one LRR are very particularly preferred, preferably at least 2 and more preferably at least 5 (LRRs are indicated in FIG. 4 by arrows). Those partial nucleotide sequences which code for at least one of the LRRs marked in FIG. 4 or nucleotide sequences which contain at least one of the LRRs indicated in FIG. 4 are particularly preferred. All derivatives, analogs or alleles of the aforementioned partial sequences disclosed are also disclosed. The AS sequences resulting from these partial DNA sequences according to the invention are also disclosed as such or as a constituent in larger recombinant proteins. All native splice variants of the sequences according to the invention also belong to the scope of the present invention.
Weiterhin bevorzugt sind Nukleinsäure-Sequenzen, insbesondere DNA-Sequenzen, die für ein Protein codieren, das mindestens 60% Sequenzidentität, vorzugsweise mindestens 80% und noch stärker bevorzugt mindestens 95%, mit den in den Figuren 9, 11 oder 13 dargestellten Sequenzen hat. Nach Isolierung und Sequenzierung sind die erfindungsgemäßen Nukleotidsequenzen gemäß Figuren 8, 10 oder 12 oder deren funktioneile oder in- funktionelle Äquivalente, wie z.B. Allelvariantenn oder Isoformen, erhältlich. Unter Allelvarianten werden im Sinne der vorliegenden Erfindung Varianten verstanden, die 60 bis 100 % Homologie auf Aminosäureebene, bevorzugt 70 bis 100 %, ganz besonders bevorzugt 90 bis 100 % aufweisen. Allelvarianten umfassen insbesondere solche funktioneilen oder infunktionel- len Varianten, die durch Deletion, Insertion oder Substitution von Nukleotiden aus der in den Figuren 8, 10 oder 12 dargestellten Sequenzen erhältlich sind, wobei wenigstens noch eine der wesentlichen biologischen Eigenschaften erhalten bleibt.Also preferred are nucleic acid sequences, in particular DNA sequences, which code for a protein which has at least 60% sequence identity, preferably at least 80% and even more preferably at least 95%, with the sequences shown in FIGS. 9, 11 or 13. After isolation and sequencing, the nucleotide sequences according to the invention shown in FIGS. 8, 10 or 12 or their functional or non-functional equivalents, such as, for example, allele variants or isoforms, are obtainable. For the purposes of the present invention, allele variants are understood to mean variants which have 60 to 100% homology at the amino acid level, preferably 70 to 100%, very particularly preferably 90 to 100%. Allelic variants include in particular those functional or infunctional len variants which can be obtained by deleting, inserting or substituting nucleotides from the sequences shown in FIGS. 8, 10 or 12, at least one of the essential biological properties being retained.
Homologe oder sequenzverwandte DNA-Sequenzen können aus allen Säugerspezies, einschließlich Mensch, nach gängigen Verfahren durch Homologie-Screening durch Hybridisierung mit einer Probe der erfindungsgemäßen Nukleinsäuresequenzen oder Teilen davon isoliert werden. Unter funktionellen Äquivalenten sind auch Homologe der in den Figuren 8, 10 oder 12 dargestellten Sequenzen, beispielsweise ihre Homologen aus anderen Mamma- lia, verkürzte Sequenzen, Einzelstrang-DNA oder RNA der codierenden und nicht-codierenden DNA-Sequenz zu verstehen. Solche funktionellen Äquivalente lassen sich ausgehend von den in den Figuren 8, 10 oder 12 dargestellten DNA-Sequenzen oder Teilen dieser Sequenzen, beispielsweise mit üblichen Hybridisierungsverfahren oder der PCR-Technik aus anderen Vertebraten wie Mammalia, isolieren. Damit sind erfindungsge- maß alle mit den Sequenzen gemäß Figuren 8, 10 oder 12 hybridisierenden Sequenzen mitumfaßt. Diese DNA-Sequenzen hybridisieren unter Standardbedingungen mit den erfindungsgemäßen Sequenzen. Zur Hybrisierung werden vorteilhaft kurze Oligo- nukleotide der konservierten Bereiche, die auf dem Fachmann bekannte Weise ermittelt werden können, verwendet. Es können aber auch längere Fragmente der erfindungsgemäßen Nukleinsäuren oder die vollständigen Sequenzen für die Hybridisierung verwendet werden.Homologous or sequence-related DNA sequences can be isolated from all mammalian species, including humans, by conventional methods by homology screening by hybridization with a sample of the nucleic acid sequences according to the invention or parts thereof. Functional equivalents are also to be understood as homologs of the sequences shown in FIGS. 8, 10 or 12, for example their homologues from other mammals, shortened sequences, single-stranded DNA or RNA of the coding and non-coding DNA sequence. Such functional equivalents can be isolated from other vertebrates such as Mammalia, starting from the DNA sequences shown in FIGS. 8, 10 or 12 or parts of these sequences, for example using conventional hybridization methods or the PCR technique. According to the invention, all sequences hybridizing with the sequences according to FIGS. 8, 10 or 12 are also included. These DNA sequences hybridize under standard conditions with the sequences according to the invention. Short oligonucleotides of the conserved regions, which can be determined in a manner known to the person skilled in the art, are advantageously used for the hybridization. However, longer fragments of the nucleic acids according to the invention or the complete sequences can also be used for the hybridization.
Je nach der verwendeten Nukleinsäure-Sequenz (Oligonukleotid, längeres Fragment oder vollständige Sequenz) bzw. je nachdem, welche Nukleinsäureart (DNA oder RNA) für die Hybridisierung verwendet werden, variieren diese Standardbedingungen. So liegen beispielsweise die Schmelztemperaturen für DNA: DNA- Hybride ca. 10 °C niedriger als die von DNA:RNA-Hybriden gleicher Länge. Unter Standardbedingungen sind beispielsweise, je nach Nukleinsäure, Temperaturen zwischen 42 und 58 °C in einer wäßrigen Pufferlösung mit einer Konzentration zwischen 0,1 bis 5 x SSC (1 X SSC = 0,15 M NaCl, 15 mM Natriumeitrat, pH 7,2) oder zusätzlich in Gegenwart von 50% Forma- mid, wie beispielsweise 42 °C in 5 x SSC, 50% Formamid, zu verstehen. Vorteilhafterweise liegen die Hybridisierungsbe- dingungen für DNA: DNA-Hybride bei 0,1 x SSC und Temperaturen zwischen etwa 20 °C bis 45 °C, bevorzugt zwischen etwa 30 °C bis 45 °C. Für DNA:RNA-Hybride liegen die Hybridisierungsbe- dingungen vorteilhaft bei 0,1 x SSC und Temperaturen zwischen etwa 30 °C bis 55 °C, bevorzugt zwischen etwa 45 °C bis 55 °C. Diese angegebenen Temperaturen für die Hybridisierung sind beispielhaft kalkulierte Schmelztemperaturwerte für eine Nukleinsäure mit einer Länge von ca. 100 Nukleotiden und einem G + C-Gehalt von 50 % in Abwesenheit von Formamid. Die experimentellen Bedingungen für die DNA-Hybridisierung sind in einschlägigen Lehrbüchern der Genetik, wie beispielsweise bei Sambrook et al. ("Molecular Cloning", Cold Spring Harbor Laboratory, 1989), beschrieben und lassen sich nach dem Fachmann bekannten Formeln, beispielsweise abhängig von der Länge der Nukleinsäuren, der Art der Hybride oder dem G + C-Gehalt berechnen. Weitere Informationen zur Hybridisierung kann der Fachmann folgenden Lehrbüchern entnehmen: Ausübel et al. (eds) , 1985, Current Protocols in Molecular Biology, John Wiley & Sons, New York; Harnes and Higgins (eds) , 1985, Nu- cleic Acids Hybridization: A Practical Approach, IRL Press at Oxford University Press, Oxford; Brown (ed) , 1991, Essential Molecular Biology: A Practical Approach, IRL Press at Oxford University Press, Oxford. Zu Äquivalenten von erfindungsgemäßen Nukleinsäure-Sequenzen gehören auch Derivate der in den Figuren 8, 10 oder 12 dargestellten Sequenzen, wie beispielsweise Promotorvarianten. Die Promotoren, die den angegebenen Nukleotidsequenzen gemeinsam oder einzeln vorgeschaltet sind, können durch ein oder mehrere Nukleotidaustausche, durch Insertion (en) und/oder Deleti- on(en) verändert sein, wobei Funktionalität bzw. Wirksamkeit der Promotoren erhalten bleiben kann oder, je nach Bedarf, verändert werden kann. So können die Promotoren durch Verän- derung ihrer Sequenz in ihrer Wirksamkeit erhöht oder komplett durch wirksamere Promotoren auch artfremder Organismen ausgetauscht werden. Unter Derivaten sind erfindungsgemäß auch Varianten zu verstehen, deren Nukleotidsequenz im Bereich -1 bis -1000 vor dem Startkodon so verändert wurden, daß die Genexpression und/oder die Proteinexpression verändert, bevorzugt erhöht, wird.These standard conditions vary depending on the nucleic acid sequence used (oligonucleotide, longer fragment or complete sequence) or on the type of nucleic acid (DNA or RNA) used for the hybridization. So For example, the melting temperatures for DNA: DNA hybrids are approx. 10 ° C lower than those of DNA: RNA hybrids of the same length. Under standard conditions, for example, depending on the nucleic acid, temperatures between 42 and 58 ° C in an aqueous buffer solution with a concentration between 0.1 to 5 x SSC (1 X SSC = 0.15 M NaCl, 15 mM sodium citrate, pH 7.2 ) or additionally in the presence of 50% formamide, such as 42 ° C in 5 x SSC, 50% formamide. The hybridization conditions for DNA: DNA hybrids are advantageously 0.1 × SSC and temperatures between approximately 20 ° C. to 45 ° C., preferably between approximately 30 ° C. to 45 ° C. For DNA: RNA hybrids, the hybridization conditions are advantageously 0.1 × SSC and temperatures between approximately 30 ° C. to 55 ° C., preferably between approximately 45 ° C. to 55 ° C. These specified temperatures for the hybridization are, for example, calculated melting temperature values for a nucleic acid with a length of approx. 100 nucleotides and a G + C content of 50% in the absence of formamide. The experimental conditions for DNA hybridization are in relevant textbooks of genetics, such as in Sambrook et al. ("Molecular Cloning", Cold Spring Harbor Laboratory, 1989), and can be calculated according to formulas known to the person skilled in the art, for example depending on the length of the nucleic acids, the type of hybrid or the G + C content. The person skilled in the art can obtain further information on hybridization from the following textbooks: Ausübel et al. (eds), 1985, Current Protocols in Molecular Biology, John Wiley & Sons, New York; Harnes and Higgins (eds), 1985, Nuclear Acids Hybridization: A Practical Approach, IRL Press at Oxford University Press, Oxford; Brown (ed), 1991, Essential Molecular Biology: A Practical Approach, IRL Press at Oxford University Press, Oxford. Equivalents of nucleic acid sequences according to the invention also include derivatives of the sequences shown in FIGS. 8, 10 or 12, such as promoter variants. The promoters which are upstream of the specified nucleotide sequences together or individually can be changed by one or more nucleotide changes, by insertion (s) and / or deletion (s), functionality or effectiveness of the promoters being retained or, depending can be changed as required. For example, the effectiveness of the promoters can be increased by changing their sequence, or they can be completely replaced by more effective promoters, including organisms of other species. According to the invention, derivatives are also to be understood to mean variants whose nucleotide sequence in the range -1 to -1000 before the start codon have been changed such that the gene expression and / or the protein expression is changed, preferably increased.
Weiterhin sind unter Derivaten auch Varianten zu verstehen, die vorzugsweise am 3' -Ende verändert wurden. Als solche "Tags" sind in der Literatur z. B. Hexa-Histidin-Anker be- kannt oder Epitope, die als Antigene verschiedener Antikörper erkannt werden können (Studier et al., Meth. Enzymol., 185, 1990: 60 - 89 und Ausubel et al. (eds.) 1998, Current Proto- cols in Molecular Biology, John Wiley & Sons, New York) . und/oder mindestens eine Signalsequenz zum Transport des translatierten Proteins, bspw. in eine bestimmte Zellorganelle oder in den extrazellulären Raum.Furthermore, derivatives are also to be understood as variants which were preferably changed at the 3 'end. As such "tags" in the literature, for. B. Hexa-histidine anchor known or epitopes that can be recognized as antigens of various antibodies (Studier et al., Meth. Enzymol., 185, 1990: 60 - 89 and Ausubel et al. (Eds.) 1998, Current Protocols in Molecular Biology, John Wiley & Sons, New York). and / or at least one signal sequence for transporting the translated protein, for example into a specific cell organelle or into the extracellular space.
Darüber hinaus kann ein erfindungsgemäßes Nukleinsäurekon- strukt oder eine erfindungsgemäße Nukleinsäure, bspw. gemäß Figuren 8, 10 oder 12 bzw. deren Derivate, Varianten, Homologe oder insbesondere Fragmente auch in therapeutisch oder diagnostisch geeigneter Form exprimiert werden. Zur Generie- rung des rekombinanten Proteins können Vektorsysteme oder 0- ligonukleotide verwendet werden, die die Nukleinsäuren oder das Nukleinsäurekonstrukt um bestimmte Nukleotidsequenzen verlängern und damit für veränderte Polypeptide kodieren, die bspw. einer einfacheren Reinigung dienen.In addition, a nucleic acid construct according to the invention or a nucleic acid according to the invention, for example according to FIGS. 8, 10 or 12 or their derivatives, variants, homologs or, in particular, fragments can also be expressed in a therapeutically or diagnostically suitable form. To the genius For the recombinant protein, vector systems or 0-ligonucleotides can be used which extend the nucleic acids or the nucleic acid construct by certain nucleotide sequences and thus code for modified polypeptides which, for example, serve for easier purification.
Bevorzugt sind weiterhin DNA-Sequenzen, die eine der in Figur 8, 10 oder 12 angegebenen (c) DNA-Sequenzen enthalten (An- spruch 4) .DNA sequences which contain one of the (c) DNA sequences indicated in FIG. 8, 10 or 12 are further preferred (claim 4).
Weiterhin werden erfindungsgemäß vorzugsweise alle DNA- Sequenzen mitoffenbart, die für ein Protein codieren, das im wesentlichen der Aminosäuresequenz der Proteine 15B3, 15B3-1 oder 15B3-2 gemäß Figuren 9, 11 oder 13 entspricht. Diese DNA-Sequenzen erhalten nur eine geringe Zahl an Veränderungen gegenüber der in den vorgenannten Figuren angegebenen Sequenzen, bspw. kann es sich um Isoformen handeln. Die Zahl der Sequenzveränderungen wird typischerweise nicht größer als 10 sein. Derartige im wesentlichen mit den für die Proteine 15B3, 15B3-1 oder 15B3-2 codierenden DNA-Sequenzen entsprechenden DNA-Sequenzen, die gleichfalls für ein biologisch aktives Protein codieren, können durch allgemein bekannte Muta- genese-Verfahren erhalten und die biologische Aktivität der durch die Mutanten codierten Proteine durch Screening- Verfahren, bspw. Bindungsstudien oder die Fähigkeit zur Ausprägung der biologischen Funktion, bspw. im Zusammenhang mit der Apoptose, identifiziert werden. Zu den entsprechenden Mu- tagenese-Verfahren gehören die „site-directed"-Mutagenese, die die automatisch durchgeführte Synthese eines Primers mit mindestens einer Basenveränderung vorsieht. Nach der Poly er- sierungsreaktion wird der Heteroduplex-Vektor in einen geeig- netes Zellsystem transferiert (z.B. E.coli) und entsprechend transformierte Klone isoliert.Furthermore, according to the invention, all DNA sequences which code for a protein which essentially corresponds to the amino acid sequence of the proteins 15B3, 15B3-1 or 15B3-2 according to FIGS. 9, 11 or 13 are preferably disclosed. These DNA sequences receive only a small number of changes compared to the sequences given in the aforementioned figures, for example they can be isoforms. The number of sequence changes will typically not be greater than 10. Such DNA sequences, which essentially correspond to the DNA sequences coding for the proteins 15B3, 15B3-1 or 15B3-2 and which likewise code for a biologically active protein, can be obtained by generally known mutagenesis methods and the biological activity of the proteins encoded by the mutants can be identified by screening methods, for example binding studies or the ability to express the biological function, for example in connection with apoptosis. The corresponding mutagenesis methods include “site-directed” mutagenesis, which provides for the automatically carried out synthesis of a primer with at least one base change. After the polymerization reaction, the heteroduplex vector is converted into a suitable netes cell system transferred (eg E. coli) and isolated transformed clones isolated.
Darüber hinaus kommen alle dem Fachmann geläufigen Methoden für die Herstellung, Modifikation und/oder Detektion von erfindungsgemäßen DNA-Sequenzen, die in vivo, in situ oder in vitro ausgeführt werden können in Betracht (PCR (Innis et al. PCR Protocols: A Guide to Methods and Applications) oder chemische Synthese) . Durch entsprechende PCR-Primer können bspw. neue Funktionen in eine erfindungsgemäße DNA-Sequenz eingeführt werden, wie z.B. Restriktionsschnittstellen, Ter- minationscodons. Hierdurch können erfindungsgemäße Sequenzen für den Transfer in Klonierungsvektoren entsprechend entworfen werden.In addition, all methods familiar to the person skilled in the art for the production, modification and / or detection of DNA sequences according to the invention, which can be carried out in vivo, in situ or in vitro, are suitable (PCR (Innis et al. PCR Protocols: A Guide to Methods and Applications) or chemical synthesis). Appropriate PCR primers can be used, for example, to introduce new functions into a DNA sequence according to the invention, such as Restriction interfaces, termination codons. In this way, sequences according to the invention can be designed accordingly for transfer into cloning vectors.
Ein weiterer Gegenstand der vorliegenden Erfindung betrifft Expressionsvektoren oder ein rekombinantes Nukleinsäurekon- strukt, das eine, wie oben beschrieben, erfindungsgemäße Nuk- leinsäuresequenz, typischerweise eine DNA-Sequenz enthält (Anspruch 5) . Vorteilhafterweise werden hierbei die erfindungsgemäßen Nukleinsäuresequenzen mit mindestens einem genetischen Regulationselement, wie bspw. Transkriptions- und Translationssignalen, funktioneil verknüpft. Diese Verknüpfung kann je nach gewünschter Anwendung zu einer nativen Ex- pressionsrate oder auch zu einer Erhöhung bzw. Erniedrigung der nativen Genexpression führen. Mit den solchermaßen hergestellten Expressionsvektoren können anschließend Wirtsorganismen bzw. Wirtszellen transformiert werden, z.B. Zellkulturen aus Säugetierzellen.The present invention further relates to expression vectors or a recombinant nucleic acid construct which, as described above, contains a nucleic acid sequence according to the invention, typically a DNA sequence (claim 5). Advantageously, the nucleic acid sequences according to the invention are functionally linked to at least one genetic regulatory element, such as, for example, transcription and translation signals. Depending on the desired application, this linkage can lead to a native expression rate or also to an increase or decrease in the native gene expression. With the expression vectors produced in this way, host organisms or host cells can then be transformed, e.g. Cell cultures from mammalian cells.
Bei dem erfindungsgemäßen Expressionsvektor wird (werden) typischerweise das (die) native(n) Regulationselement (e) eingesetzt werden, d.h. die bspw. Promotor und/oder Enhancer- Region des Gens für das Protein 15B3, 15B3-1 oder 15B3-2, bspw. aus Säugetieren, insbesondere entsprechende humane Regulationssequenzen. Ggf. können diese nativen 15B3-, 15B3-1 oder 15B3-2-Regulationssequenzen auch genetisch verändert sein, um eine veränderte Expressionsintensität hervorzurufen. Zusätzlich zu diesen nativen 15B3, 15B3-1 oder 15B3-2- Regulationssequenzen oder anstelle dieser nativen 15B3-, 15B3-1 oder 15B3-2-Regulationssequenzen können für andere Gene native Regulationselemente erfindungsgemäßen DNA-Sequenzen vor- und/oder nachgeschaltet (5'- oder 3'- Regulationssequenzen) sein und gegebenenfalls auch genetisch verändert worden sein, so daß die natürliche Regulation unter der Kontrolle der 15B3-, 15B3-1- oder 15B3-2- Regulationssequenzen ausgeschaltet ist und die Expression der Gene - je nach Wunsch - hierdurch erhöht oder erniedrigt werden kann.The expression vector according to the invention will typically use the native regulatory element (s), ie the promoter and / or enhancer Region of the gene for the protein 15B3, 15B3-1 or 15B3-2, for example from mammals, in particular corresponding human regulatory sequences. Possibly. these native 15B3, 15B3-1 or 15B3-2 regulatory sequences can also be genetically modified in order to produce a changed expression intensity. In addition to these native 15B3, 15B3-1 or 15B3-2 regulatory sequences or instead of these native 15B3, 15B3-1 or 15B3-2 regulatory sequences, native regulatory elements according to the invention for other genes can be connected upstream and / or downstream (5 ' - or 3 'regulatory sequences) and, if necessary, also have been genetically modified so that the natural regulation is switched off under the control of the 15B3, 15B3-1 or 15B3-2 regulatory sequences and the expression of the genes - as desired - this can be increased or decreased.
Vorteilhafte Regulationssequenzen für das erfindungsgemäße Verfahren sind beispielsweise in Promotoren wie cos-, tac-, trp-, tet-, trp-tet-, lpp-, lac-, lpp-lac-, laclq-, T7-, T5- , T3-, gal-, trc-, ara-, SP6-, 1-PR- oder im 1-PL-Promotor enthalten, die vorteilhafterweise in gram-negativen Bakterien Anwendung finden. Weitere vorteilhafte Regulationssequenzen sind beispielsweise in den gram-positiven Promotoren wie amy und SP02, in den Hefepromotoren wie ADC1, MFa , AC, P-60, CYC1, GAPDH oder in Mammaliapromotoren wie CaM-Kinasell, CMV, Nestin, L7, BDNF, NF, MBP, NSE, beta-Globin, GFAP, GAP43, Ty- rosin Hydroxylase, Kainat-Rezeptor-Untereinheit 1, Glutamat- Rezeptor-Untereinheit B enthalten. Prinzipiell können alle natürlichen Promotoren mit ihren Regulationssequenzen wie die bspw. oben genannten für einen erfindungsgemäße Expressionsvektor verwendet werden. Darüber hinaus können auch synthetische Promotoren vorteilhaft verwendet werden. Diese regulatorischen Sequenzen sollen die gezielte Expression der erfindungsgemäßen Nukleinsäurese- quenzen ermöglichen. Dies kann beispielsweise je nach Wirts- Organismus bedeuten, daß das Gen erst nach Induktion expri- miert oder überexprimiert wird, oder daß es sofort exprimiert und/oder überexprimiert wird. Die regulatorischen Sequenzen bzw. Faktoren können dabei vorzugsweise die Expression positiv beeinflussen und dadurch erhöhen. So kann eine Verstär- kung der regulatorischen Elemente vorteilhafterweise auf der Transkriptionsebene erfolgen, indem starke Transkriptionssignale wie Promotoren und/oder "Enhancer" verwendet werden. Daneben ist aber auch eine Verstärkung der Translation möglich, indem beispielsweise die Stabilität der mRNA verbessert wird.Advantageous regulatory sequences for the method according to the invention are, for example, in promoters such as cos, tac, trp, tet, trp-tet, lpp, lac, lpp-lac, laclq, T7, T5, T3 contain, gal, trc, ara, SP6, 1-PR or in the 1-PL promoter, which are advantageously used in gram-negative bacteria. Further advantageous regulatory sequences are, for example, in the gram-positive promoters such as amy and SP02, in the yeast promoters such as ADC1, MFa, AC, P-60, CYC1, GAPDH or in mammalian promoters such as CaM-Kinasell, CMV, Nestin, L7, BDNF, NF , MBP, NSE, beta-globin, GFAP, GAP43, tyrosine hydroxylase, kainate receptor subunit 1, glutamate receptor subunit B included. In principle, all natural promoters with their regulatory sequences, such as those mentioned above, can be used for an expression vector according to the invention. In addition, synthetic promoters can also be used advantageously. These regulatory sequences are intended to enable the targeted expression of the nucleic acid sequences according to the invention. Depending on the host organism, this can mean, for example, that the gene is only expressed or overexpressed after induction, or that it is expressed and / or overexpressed immediately. The regulatory sequences or factors can preferably have a positive influence on the expression and thereby increase it. Thus, the regulatory elements can advantageously be strengthened at the transcription level by using strong transcription signals such as promoters and / or "enhancers". In addition, an increase in translation is also possible, for example, by improving the stability of the mRNA.
Als Regulationssequenzen werden alle dem Fachmann geläufigen Elemente bezeichnet, die auf der Transkriptions- und/oder Translationsebene die Expression der erfindungsgemäßen Se- quenzen beeinflussen können. Insbesondere sind dabei neben Promotorsequenzen sog. "Enhancer"-Sequenzen hervorzuheben, die über eine verbesserte Wechselwirkung zwischen RNA- Polymerase und DNA eine erhöhte Expression bewirken können. Als weitere Regulationssequenzen seien beispielhaft die sog. "Locus Control Regions", "Silencer" oder jeweilige Teilsequenzen davon genannt. Diese Sequenzen können vorteilhaft für eine gewebespezifische Expression verwendet werden. Auch sog. Terminatorsequenzen werden vorteilhafterweise in einem erfindungsgemäßen Expressionsvektor vorhanden sein und erfindungs- gemäß unter den Terminus "Regulationssequenz" subsumiert.Regulatory sequences are all elements familiar to the person skilled in the art which can influence the expression of the sequences according to the invention at the transcription and / or translation level. In particular, in addition to promoter sequences, so-called "enhancer" sequences are to be emphasized, which can bring about increased expression via an improved interaction between RNA polymerase and DNA. The so-called "locus control regions", "silencers" or respective partial sequences thereof may be mentioned as further regulatory sequences. These sequences can be used advantageously for tissue-specific expression. So-called terminator sequences will also advantageously be present in an expression vector according to the invention and, according to the invention, are subsumed under the term "regulatory sequence".
Eine bevorzugte Ausführungsform der vorliegenden Erfindung ist die Verknüpfung der erfindungsgemäßen Nukleinsäuresequenz mit einem Promotor, wobei der Promotor typisch 5' "upstream" von einer erfindungsgemäßen DNA-Sequenz zu liegen kommt. Weitere Regulationssignale, wie bspw. 3' -gelegene Terminatoren, Polyadenylierungssignale oder Enhancer, können funktioneil in dem Expressionsvektor enthalten sein. Darüber hinaus können erfindungsgemäße Nukleinsäuresequenzen, insbesondere für die Sequenzen gemäß Figuren 8, 10 oder 12 bzw. für die entsprechenden Proteine, in einer oder mehreren Kopien in einem Gen- konstrukt nach dieser Erfindung enthalten sein, oder ggf. auch auf getrennten Genkonstrukten lokalisiert sein.A preferred embodiment of the present invention is the linkage of the nucleic acid sequence according to the invention with a promoter, the promoter typically being located 5 '"upstream" from a DNA sequence according to the invention. Further regulation signals, such as, for example, 3 '-terminal, polyadenylation signals or enhancers, can be functionally contained in the expression vector. In addition, nucleic acid sequences according to the invention, in particular for the sequences according to FIGS. 8, 10 or 12 or for the corresponding proteins, can be contained in one or more copies in a gene construct according to this invention, or, if appropriate, can also be localized on separate gene constructs.
Unter den Begriff "Expressionsvektor" fallen sowohl rekombi- nante Nukleinsäurekonstrukte bzw. Genkonstrukte, wie zuvor beschrieben, als auch komplette Vektorkonstrukte, die neben erfindungsgemäßen DNA-Sequenzen und etwaigen Regulationssequenzen typischerweise auch weitere Elemente enthalten. Diese Vektorkonstrukte oder Vektoren werden zur Expression in einem geeigneten Wirtsorganismus verwendet. Vorteilhafterweise wird mindestens eine erfindungsgemäße DNA-Sequenz, bspw. das 15B3- Gen oder eine Teilsequenz des 15B3-Gens, in einen wirtsspezifischen Vektor insertiert, der eine optimale Expression der Gene im ausgesuchten Wirt ermöglicht. Vektoren sind dem Fachmann wohl bekannt und können beispielsweise aus "Cloning Vectors" (Eds. Pouwels P. H. et al. Elsevier, Amsterdam-New Y- ork-Oxford, 1985, ISBN 0 444 904018) entnommen werden. Unter Vektoren sind außer Plasmiden auch alle anderen dem Fachmann bekannten Vektoren wie beispielsweise Phagen, Viren wie SV40, CMV, Baculovirus, Adenovirus, Sindbisvirus, Transposons, IS- Elemente, Phasmide, Phagemide, Cosmide, lineare oder zirkulä- re DNA zu verstehen. Diese Vektoren können autonom im Wirtsorganismus repliziert oder chromosomal repliziert werden. Für die Integration in Mammalia wird typischerweise lineare DNA verwendet. Die Expression erfindungsgemäßer Nukleinsäuresequenzen kann vorteilhaft durch Erhöhen der Genkopienzahl und/oder durch Verstärkung regulatorischer Faktoren, die die Genexpression positiv beeinflussen, erhöht werden. So kann eine Verstärkung regulatorischer Elemente vorzugsweise auf der Transkriptionsebene erfolgen, indem stärkere Transkriptionssignale, wie Promotoren und Enhancer, verwendet werden. Daneben ist aber auch eine Verstärkung der Translation möglich, indem bei- spielsweise die Stabilität der mRNA verbessert oder die Ableseeffizienz dieser mRNA an den Ribosomen erhöht wird. Zur Erhöhung der Genkopienzahl können die Nukleinsäuresequenzen o- der homologe Gene, beispielsweise in ein Nukleinsäurefragment bzw. in einen Vektor eingebaut werden, der vorzugsweise die den jeweiligen Genen zugeordnete, regulatorische Gensequenzen oder analog wirkende Promotoraktivität enthält. Insbesondere werden solche regulatorische Sequenzen verwendet, die die Genexpression verstärken.The term “expression vector” includes both recombinant nucleic acid constructs or gene constructs, as described above, and complete vector constructs, which typically contain further elements in addition to the DNA sequences according to the invention and any regulatory sequences. These vector constructs or vectors are used for expression in a suitable host organism. Advantageously, at least one DNA sequence according to the invention, for example the 15B3 gene or a partial sequence of the 15B3 gene, is inserted into a host-specific vector which enables optimal expression of the genes in the selected host. Vectors are well known to those skilled in the art and can be found, for example, in "Cloning Vectors" (Eds. Pouwels PH et al. Elsevier, Amsterdam-New York-Oxford, 1985, ISBN 0 444 904018). In addition to plasmids, vectors are also understood to mean all other vectors known to the person skilled in the art, such as phages, viruses such as SV40, CMV, baculovirus, adenovirus, Sindbis virus, transposons, IS elements, phasmids, phagemids, cosmids, linear or circular DNA. These vectors can be replicated autonomously in the host organism or replicated chromosomally. Linear DNA is typically used for integration into Mammalia. The expression of nucleic acid sequences according to the invention can advantageously be increased by increasing the number of gene copies and / or by strengthening regulatory factors which have a positive effect on gene expression. For example, regulatory elements can preferably be amplified at the transcription level by using stronger transcription signals, such as promoters and enhancers. In addition, an increase in translation is also possible, for example, by improving the stability of the mRNA or increasing the reading efficiency of this mRNA on the ribosomes. To increase the number of gene copies, the nucleic acid sequences or the homologous genes can be incorporated, for example, into a nucleic acid fragment or into a vector which preferably contains the regulatory gene sequences assigned to the respective genes or promoter activity having an analogous effect. In particular, those regulatory sequences are used which increase gene expression.
Erfindungsgemäße Nukleinsäuresequenzen können zusammen mit den für interagierende oder für potentiell interagierende Proteine kodierenden Sequenzen in einen einzelnen Vektor klo- niert werden und anschließend in vitro in einer Wirtszelle oder in vivo in einem Wirtsorganismus exprimiert werden. Al- ternativ kann auch jede der potentiell interagierenden Nukleinsäuresequenzen und die für 15B3, 15B3-1 oder 15B3-2 kodierenden Sequenzen in je einen einzelnen Vektor gebracht und diese getrennt in den jeweiligen Organismus über übliche Methoden, wie bspw. Transformation, Transfektion, Transduktion, Elektroporation oder Partikel-Gun verbracht werden.Nucleic acid sequences according to the invention can be cloned together with the sequences coding for interacting or for potentially interacting proteins into a single vector and then expressed in vitro in a host cell or in vivo in a host organism. Alternatively, each of the potentially interacting nucleic acid sequences and the sequences coding for 15B3, 15B3-1 or 15B3-2 can each be brought into a single vector and these separated into the respective organism using customary methods, such as, for example, transformation, transfection, transduction, Electroporation or particle gun can be spent.
In einer weiteren vorteilhaften Ausführungsform kann mindestens ein Marker-Gen (bspw. Antibiotika-Resistenz-Gene und/oder Gene, die für ein fluoreszierendes Protein kodieren, insbesondere GFP) in einem erfindungsgemäßen Expressionsvektor, insbesondere einem kompletten Vektorkonstrukt, enthalten sein.In a further advantageous embodiment, at least one marker gene (for example antibiotic resistance genes and / or genes which code for a fluorescent protein, in particular GFP), can be contained in an expression vector according to the invention, in particular a complete vector construct.
Ein weiterer Gegenstand der vorliegenden Erfindung betrifft Wirtszellen, die mit einer erfindungsgemäßen DNA-Sequenz und/oder einem erfindungsgemäßen Expressionsvektor, insbesondere einem Vektorkonstrukt transformiert sind (Anspruch 6) . Als Wirtszellen sind prinzipiell alle Zellen geeignet, die eine Expression erfindungsgemäßer DNA-Sequenzen allein oder im Verbund mit weiteren Sequenzen, insbesondere Regulationssequenzen, gestatten. Als Wirtszellen kommen alle Zellen pro- oder eukaryontischer Natur in Betracht, beispielsweise Bakte- rien, Pilze, Hefen, pflanzliche oder tierische Zellen. Bevorzugte Wirtszellen sind Bakterien, wie Escherichia coli, Streptomyces, Bacillus oder Pseudomonas, eukaryotische Mikroorganismen, wie Aspergillus oder Saccharomyces cerevisiae o- derr die gewöhnliche Bäckerhefe (Stinchcomb et al., Nature, 282:39, (1997)).The present invention further relates to host cells which have been transformed with a DNA sequence according to the invention and / or an expression vector according to the invention, in particular a vector construct (claim 6). In principle, all cells are suitable as host cells that allow expression of DNA sequences according to the invention alone or in combination with other sequences, in particular regulatory sequences. All cells of a pro- or eukaryotic nature can be considered as host cells, for example bacteria, fungi, yeasts, plant or animal cells. Preferred host cells are bacteria such as Escherichia coli, Streptomyces, Bacillus or Pseudomonas, eukaryotic microorganisms such as Aspergillus or Saccharomyces cerevisiae or the usual baker's yeast (Stinchcomb et al., Nature, 282: 39, (1997)).
In einer bevorzugten Ausführungsform werden jedoch zur Expression von erfindungsgemäßen DNA-Sequenzen Zellen aus multizellulären Organismen gewählt. Dies geschieht auch vor dem Hin- tergrund einer möglicherweise erforderlichen Glykosylierung (N- und/oder O-gekoppelt) der codierten Proteine. Diese Funktion kann in höheren Eukaryotenzellen - im Vergleich zu Proka- ryotenzellen - in geeigneter Weise ausgeführt werden. Im Prinzip ist jede höhere eukaryotische Zellkultur als Wirtszelle verfügbar, wenn auch Zellen von Säugern, beispielsweise Affen, Ratten, Hamstern oder Menschen, ganz besonders bevorzugt sind. Dem Fachmann ist eine Vielzahl von etablierten Zellinien bekannt. In einer keineswegs abschließenden Aufzählung werden die folgenden Zellinien genannt: 293T (Embryonennierenzelli- nie), (Graham et al., J. Gen. Virol., 36:59 (1997)), BHK (Ba- byhamsternierenzellen) , CHO (Zellen aus den Hamsterovarien) , (Urlaub und Chasin, P. N. A. S. (USA) 77:4216, (1980)), HeLa (humane Cervixkarzinomzellen) und weitere - insbesondere für den Laboreinsatz etablierte - Zellinien, wie bspw. HEK293-, Sf9- oder COS-Zellen. Ganz besonders bevorzugt sind humane Zellen, insbesondere Zellen des Immunsystems oder adulte Stammzellen, bspw. Stammzellen des Blut bildenden Systems (aus dem Knochenmark) (Anspruch 7) . Humane erfindungsgemäße transformierte Zellen, insbesondere autologe Zellen des Patienten, eignen sich nach (vor allem ex vivo) Transformation mit erfindungsgemäßen DNA-Sequenzen oder erfindungsgemäßen Expressionsvektoren, ganz besonders als Arzneimittel für bspw. gentherapeutische Zwecke, also nach Durchführung einer Zellentnahme, ggf. ex vivo Expansion, Transformation, Selektion und abschließender Retransplantation.In a preferred embodiment, however, cells from multicellular organisms are selected for the expression of DNA sequences according to the invention. This also happens against the background of a possibly required glycosylation (N- and / or O-coupled) of the encoded proteins. This function can be carried out in a suitable manner in higher eukaryotic cells - in comparison to prokaryotic cells. In principle, any higher eukaryotic cell culture is available as a host cell, although cells from mammals, for example monkeys, rats, hamsters or humans, are very particularly preferred. A large number of established cell lines are known to the person skilled in the art. In a by no means exhaustive list named the following cell lines: 293T (embryonic kidney cell line), (Graham et al., J. Gen. Virol., 36:59 (1997)), BHK (baby hamster kidney cells), CHO (cells from the hamster ovaries), (Urlaub and Chasin, PNAS (USA) 77: 4216, (1980)), HeLa (human cervical carcinoma cells) and further cell lines, such as HEK293, Sf9 or COS cells, which have been established especially for laboratory use. Human cells, in particular cells of the immune system or adult stem cells, for example stem cells of the blood-forming system (from the bone marrow) are very particularly preferred (claim 7). Human transformed cells according to the invention, in particular autologous cells of the patient, are suitable after (above all ex vivo) transformation with DNA sequences according to the invention or expression vectors according to the invention, very particularly as a medicament for, for example, gene therapy purposes, that is to say after cell removal, possibly ex vivo Expansion, transformation, selection and final retransplantation.
Die Kombination aus einer Wirtszelle und einem zu den Wirts- zellen passenden erfindungsgemäßen Expressionsvektor, wie Plasmide, Viren oder Phagen, wie beispielsweise Plasmide mit dem RNA-Polymerase/Promoter System, die Phagen 1, Mu oder andere temperänte Phagen oder Transposons, und/oder weiteren vorteilhaften regulatorischen Sequenzen, bilden eine erfin- dungsgemäße Wirtszelle, die als Expressionssystem dienen kann. Bevorzugte erfindungsgemäße Expressionssysteme auf der Basis erfindungsgemäßer Wirtszellen sind beispielsweise die Kombination aus Säugetierzellen, wie bspw. CHO-Zellen, und Vektoren, wie bspw. pcDNA3neo-Vektor, oder bspw. HEK293- Zellen und CMV-Vektor, die für Säugetierzellen besonders geeignet sind.The combination of a host cell and an expression vector according to the invention which matches the host cells, such as plasmids, viruses or phages, such as plasmids with the RNA polymerase / promoter system, the phages 1, Mu or other tempered phages or transposons, and / or further advantageous regulatory sequences form a host cell according to the invention which can serve as an expression system. Preferred expression systems according to the invention based on host cells according to the invention are, for example, the combination of mammalian cells, such as, for example, CHO cells, and vectors, such as, for example, pcDNA3neo vector, or, for example, HEK293 cells and CMV vector, which are particularly suitable for mammalian cells.
Ein weiterer Aspekt der vorliegenden Erfindung sind die Genprodukte der erfindungsgemäßen DNA-Sequenzen (Anspruch 8) . Un- ter Genprodukten versteht man im Sinne dieser Erfindung sowohl Primärtranskripte, also RNA, vorzugsweise mRNA, als auch Proteine bzw. Polypeptide, insbesondere in aufgereinigter Form (Anspruch 9) . Diese Proteine weisen erfindungsgemäß mindestens eine LRR-Sequenz, vorzugsweise mindestens 5, stärker bevorzugt mindestens 8 und am stärksten bevorzugt 10 oder sogar mehr alsAnother aspect of the present invention are the gene products of the DNA sequences according to the invention (claim 8). U.N- For the purposes of this invention, gene products are understood to mean both primary transcripts, that is to say RNA, preferably mRNA, and proteins or polypeptides, in particular in purified form (claim 9). According to the invention, these proteins have at least one LRR sequence, preferably at least 5, more preferably at least 8 and most preferably 10 or even more than
10 LRR-Sequenzen auf und regulieren oder transportieren insbesondere apoptotische oder nekrotische, ggf. auch infla matori- sche Signale oder Signale, die das Zellwachstum oder die Zell- Plastizität betreffen. Typischerweise sind sie an der Signaltransduktion über die Ras-Kaskade unmittelbar oder mittelbar beteiligt. Bevorzugt ist ein aufgereinigtes Genprodukt dann, wenn es mindestens eine der in Figur 4 angegebenen (und auch in den Figuren 9, 11 und 13 enthaltenen Aminosäurese- quenzen (für eine LRR-Domäne) , oder ein funktionshomologes oder die Funktion inhibierendes (infunktionelles) Allel, Fragment, Analoges oder Derivat dieser Sequenz enthält (Anspruch 10) oder typischerweise aus einer solchen Aminosäuresequenz besteht. Funktionshomologie wird im Sinne der vorlie- genden Erfindung so definiert, daß mindestens noch eine der wesentlichen funktionellen Eigenschaften der gemäß Figuren 9,10 LRR sequences and regulate or transport in particular apoptotic or necrotic, possibly also inflammatory signals or signals which concern cell growth or cell plasticity. Typically, they are directly or indirectly involved in signal transduction via the Ras cascade. A purified gene product is preferred if it contains at least one of the amino acid sequences given in FIG. 4 (and also contained in FIGS. 9, 11 and 13 (for an LRR domain), or a function-homologous or function-inhibiting (infunctional) allele "Contains a fragment, analog or derivative of this sequence or typically consists of such an amino acid sequence. Functional homology is defined in the sense of the present invention in such a way that at least one of the essential functional properties of those according to FIG.
11 oder 13 dargestellten Proteine 15B3, 15B3-1 oder 15B3-2 erhalten bleibt. Typischerweise werden funktionshomologe erfindungsgemäße Proteine insbesondere eine charakteristische, bspw. mindestens 60%ige, vorzugsweise mindestens 80%ige Sequenzidentität in den als Leucin reichen Regionen (LRRs) bezeichneten Sequenzabschnitten, die Protein- Interaktionsdomänen darstellen und typischerweise 20-28 Aminosäuren mit einer spezifischen Kernkonsensus-Sequenz von Leucinen und Asparaginen (LXXLXLXXN) aufweisen. Typischerweise wird ein erfindungsgemäßes funktionshomologes Allel, Derivat oder Analogon der in den Figuren 9, 11 oder 13 offenbarten Aminosäure-Sequenzen mindestens vier, vorzugsweise min- destens 6 und noch stärker bevorzugt mindestens 8 LRRs mit den vorgenannten Konsensus-Sequenzen aufweisen.Proteins 15B3, 15B3-1 or 15B3-2 shown in FIG. 11 or 13 are retained. Typically, functionally homologous proteins according to the invention will in particular have a characteristic, for example at least 60%, preferably at least 80% sequence identity in the sequence sections designated as leucine-rich regions (LRRs), which represent protein interaction domains and typically 20-28 amino acids with a specific core consensus. Sequence of leucines and asparagines (LXXLXLXXN) have. A functionally homologous allele, derivative or analog of the amino acid sequences disclosed in FIGS. 9, 11 or 13 is typically at least four, preferably at least at least 6 and even more preferably have at least 8 LRRs with the aforementioned consensus sequences.
Unter einem Derivat werden dabei insbesondere solche AS- Sequenzen verstanden, die durch Modifikationen ihrer Seitenketten verändert sind. Bspw. durch Konjugation eines Antikörpers, Enzyms oder Rezeptors an eine erfindungsgemäße AS- Sequenz. Derivate können aber auch die Kopplung eines Zuckers oder Fett (säure) -restes oder einer Phosphatgruppe oder jeder beliebigen Modifikation einer Seitenkette, insbesondere einer freien OH-Gruppe oder NH2-Gruppe oder am N- oder C-Terminus. Darüber hinaus schließt der Begriff "Derivat" auch Fusionsproteine ein, bei denen also eine erfindungsgemäße Aminosäuresequenz an beliebige Oligo- oder Polypeptide gekoppelt ist.A derivative is understood to mean, in particular, those AS sequences which have been modified by modifications to their side chains. For example. by conjugation of an antibody, enzyme or receptor to an AS sequence according to the invention. Derivatives can also be the coupling of a sugar or fatty (acid) residue or a phosphate group or any modification of a side chain, in particular a free OH group or NH2 group or at the N or C terminus. In addition, the term "derivative" also includes fusion proteins in which an amino acid sequence according to the invention is coupled to any oligo- or polypeptides.
Als "Analoge" werden Sequenzen bezeichnet, die sich durch mindestens eine AS-Veränderung gegenüber der nativen Sequenz auszeichnen (Insertion, Substitution) . Im Rahmen der vorliegenden Erfindung sind solche konservativen Substitutionen be- vorzugt, bei denen der physikochemische Charakter (Raumerfüllung, Basizität, Hydrophobizität etc.) der ausgetauschten AS erhalten bleibt (polare AS, lange aliphatische Kette, kurze aliphatische Kette, negativ oder positiv geladene AS, AS mit aromatischer Gruppe) . Die Substitutionen können biologisch funktioneile, tw. Funktionelle oder biologisch infunktionelle Sequenzen ergeben. Beispielsweise können Argininreste gegen Lysinreste, Valinreste gegen Isoleucinreste oder Asparagin- säurereste gegen Glutaminsäurereste ausgetauscht werden. Es können aber auch ein oder mehrere Aminosäuren in ihrer Rei- henfolge vertauscht, hinzugefügt oder entfernt werden, oder es können mehrere dieser Maßnahmen miteinander kombiniert werden. Die solchermaßen gegenüber den in Fig. 9, 11 oder 13 veränderten Proteine besitzen typischerweise wenigstens 60%, bevorzugt wenigstens 70% und besonders bevorzugt wenigstens 90% Sequenzidentität zu den Sequenzen in den vorgenannten Figuren, berechnet nach dem Algorithmus von Altschul et al. (J. Mol. Biol., 215, 403-410, 1990). Das isolierte Protein und seine funktionellen Varianten lassen sich vorteilhafterweise aus dem Gehirn von Mammalia wie Homo sapiens, Rattus norvegicus oder Mus musculus isolieren. Auch Homologe aus anderen Mammalia sind unter funktionellen Varianten zu verstehen.Sequences that are characterized by at least one AS change compared to the native sequence (insertion, substitution) are referred to as "analogs". In the context of the present invention, such conservative substitutions are preferred in which the physicochemical character (space filling, basicity, hydrophobicity, etc.) of the exchanged AS is retained (polar AS, long aliphatic chain, short aliphatic chain, negatively or positively charged AS, AS with aromatic group). The substitutions can be biologically functional, some. Functional or bio-functional sequences result. For example, arginine residues can be exchanged for lysine residues, valine residues for isoleucine residues or aspartic acid residues for glutamic acid residues. However, one or more amino acids can also be interchanged, added or removed in their order, or several of these measures can be combined with one another. The proteins modified in this way compared to the ones in FIG. 9, 11 or 13 typically have at least 60%, preferably at least 70% and particularly preferably at least 90% sequence identity to the sequences in the aforementioned figures, calculated according to the algorithm of Altschul et al. (J. Mol. Biol., 215, 403-410, 1990). The isolated protein and its functional variants can advantageously be isolated from the brain of mammals such as Homo sapiens, Rattus norvegicus or Mus musculus. Homologs from other mammals are also to be understood as functional variants.
Bevorzugt sind erfindungsgemäß Analoge dann, wenn die Sekundärstruktur, wie sie in der nativen Sequenz auftritt, auch bei ihnen erhalten bleibt. Neben konservative Substitutionen können auch weniger konservative AS-Variationen erfindungsge- maß in die native Sequenz eingeführt werden. Dabei behalten sie typischerweise ihre biologische Funktion, insbesondere als Transduktor eines apoptotischen oder nekrotischen, zellproliferativen, wachstumsindizierenden oder regenerativen Signals, bei. Der Effekt einer Substitution oder Deletion kann ohne weiteres durch entsprechende Untersuchungen, bspw. Bindungsassays oder zytotoxische Tests, überprüft werden.Analogs are preferred according to the invention if the secondary structure as occurs in the native sequence is also retained in them. In addition to conservative substitutions, less conservative AS variations can also be introduced into the native sequence according to the invention. They typically retain their biological function, in particular as a transducer of an apoptotic or necrotic, cell proliferative, growth-indicating or regenerative signal. The effect of a substitution or deletion can easily be checked by appropriate investigations, for example binding assays or cytotoxic tests.
Gleichwohl werden erfindungsgemäß aber auch Sequenzen einbezogen, die einen sogenannten dominant-negativen Effekt her- vorrufen, d.h. auf Grund ihre veränderten Primärsequenz zwar noch Bindungsaktivität an eine in der Kaskade stromaufwärts gelegene Sequenz aufweisen, das Signal aber nicht stromabwärts weitergeben können. Derartige Analoge fungieren daher als Inhibitoren der biologischen Funktion, insbesondere als Inhibitoren der Apoptose, des Zellwachstums, der Zellprolife- ration, der Zellplastizität oder der Zellregeneration. Derartige Analoge werden durch gentechnische Maßnahmen hergestellt, und zwar typischerweise durch die sog. "site- directed"-Mutagenese einer DNA-Sequenz, die für ein erfin- dunsgemäßes Protein (typischerweise 15B3, 15B3-1 oder 15B3- 2), codiert. Hierdurch wird die dem Analogen zugrundliegende DNA-Sequenz hergestellt, die schließlich das Protein in einer rekombinanten Zellkultur exprimieren kann (Sambrook et al., 1989, s.o.). Auch alle Derivate der vorbeschriebenen Analoge werden mitoffenbart, genauso wie die den vorbeschriebenen AS- Sequenzen zugrundeliegenden DNA-Sequenzen.Nevertheless, according to the invention, sequences are also included which produce a so-called dominant-negative effect, ie, due to their changed primary sequence, still have binding activity to a sequence upstream in the cascade, but cannot pass the signal downstream. Analogs of this type therefore function as inhibitors of biological function, in particular as inhibitors of apoptosis, cell growth, cell proliferation, cell plasticity or cell regeneration. Such analogs are produced by genetic engineering measures, typically by the so-called "site directed "mutagenesis of a DNA sequence which codes for a protein according to the invention (typically 15B3, 15B3-1 or 15B3-2). This produces the DNA sequence on which the analogue is based, which ultimately contains the protein in a recombinant cell culture (Sambrook et al., 1989, see above). All derivatives of the above-described analogs are also disclosed, as is the DNA sequence on which the above-described AS sequences are based.
Weiterhin gehören auch Fragmente einer nativen erfindungsgemäßen AS-Sequenz zum Gegenstand der vorliegenden Erfindung. Fragmente zeichnen sich durch Deletionen aus (N- oder C- terminal oder auch intrasequentiell) . Sie können einen dominant-negativen oder dominant-positiven Effekt haben.Furthermore, fragments of a native AS sequence according to the invention also form part of the subject matter of the present invention. Fragments are characterized by deletions (N- or C-terminal or also intrasequential). They can have a dominant-negative or dominant-positive effect.
Zu den erfindungsgemäßen Genprodukten (Proteinen) gehören aber auch all jene Genprodukte (Proteine) , die sich erfindungsgemäß von DNA-Derivaten, DNA-Fragmenten oder DNA-Allelen der in den Figuren 8, 10 oder 12 angegebenen DNA-Sequenzen nach Transkription und Translation ableiten.The gene products (proteins) according to the invention also include all those gene products (proteins) which, according to the invention, are derived from DNA derivatives, DNA fragments or DNA alleles of the DNA sequences shown in FIGS. 8, 10 or 12 after transcription and translation ,
Darüber hinaus können die erfindungsgemäßen Proteine chemisch modifiziert sein. So etwa kann eine Schutzgruppe am N-Terminus vorliegen. Es können Glykosylgruppen an Hydroxyl- oder Ami- nogruppen angefügt sein, Lipide können kovalent mit dem erfindungsgemäßen Protein verbunden sein, ebenso Phosphate oder A- cetylgruppen und ähnliches. Auch beliebige chemische Substanzen, Verbindungen oder Gruppen können auf einem beliebigen Syntheseweg an das erfindungsgemäße Protein gebunden sein. Auch zusätzliche Aminosäuren, z.B. in Form einzelner Aminosäuren oder in Form von Peptiden oder in Form von Proteindomänen und ähnliches, können mit dem N- und/oder C-Terminus eines erfindungsgemäßen Proteins, insbesondere aber auch an den N- o- der C-Terminus einer Leucin reichen Domäne, fusioniert sein, ggf. aber auch in die Sequenz der erfindungsgemäßen Leucin reichen Domäne integriert sein.In addition, the proteins according to the invention can be chemically modified. For example, there may be a protecting group at the N-terminus. Glycosyl groups can be added to hydroxyl or amino groups, lipids can be covalently linked to the protein according to the invention, as can phosphates or acetyl groups and the like. Any chemical substances, compounds or groups can also be bound to the protein according to the invention by any synthetic route. Additional amino acids, for example in the form of individual amino acids or in the form of peptides or in the form of protein domains and the like, can also be used with the N- and / or C-terminus of a protein according to the invention, but in particular also at the N- or the C-terminus of a leucine-rich domain may be fused, but may also be integrated into the sequence of the leucine-rich domain according to the invention.
Insbesondere sind hier sogenannte Signal- oder "Leader"- Sequenzen am N-Terminus der Aminosäuresequenz eines erfindungsgemäßen Proteins bevorzugt, die das Peptid cotranslatio- nal oder posttranslational in eine bestimmte Zellorganelle o- der in den extrazellulären Raum (bzw. das Kulturmedium) füh- ren. Am N- oder am C-Terminus können auch Aminosäuresequenzen vorliegen, die als Antigen die Bindung der erfindungsgemäßen Aminosäuresequenz an Antikörper erlauben. Zu nennen ist hier insbesondere das Flag-Peptid, dessen Sequenz im Einbuchstabencode der Aminosäuren lautet: DYKDDDDK. Oder auch ein His-Tag mit mindestens 3, vorzugsweise mindestens 6 Histidin-Resten. Diese Sequenzen haben stark antigene Eigenschaften und erlaubt somit eine schnelle Überprüfung und leichte Reinigung des rekombinanten Proteins. Monoklonale Antikörper, die das Flag- Peptid binden, sind von der Firma Eastman Kodak Co., Scienti- fic Imaging Systems Division, New Haven, Connecticut erhältlich.In particular, so-called signal or "leader" sequences at the N-terminus of the amino acid sequence of a protein according to the invention are preferred, which lead the peptide cotranslationally or post-translationally into a specific cell organelle or into the extracellular space (or the culture medium). Ren. Amino acid sequences can also be present at the N- or at the C-terminus, which allow the binding of the amino acid sequence according to the invention to antibodies as antigen. Particularly noteworthy here is the flag peptide, the sequence of which in the single-letter code of the amino acids is: DYKDDDDK. Or a His tag with at least 3, preferably at least 6, histidine residues. These sequences have strong antigenic properties and thus allow the recombinant protein to be checked and purified quickly. Monoclonal antibodies that bind the flag peptide are available from Eastman Kodak Co., Scientific Imaging Systems Division, New Haven, Connecticut.
Gegenstand der vorliegenden Erfindung sind ferner mindestens 20, stärker bevorzugt mindestens 30 und noch stärker bevor- zugt mindestens 50 Aminosäuren umfassende Teilabschnitte der in den Figuren 9, 11 oder 13 offenbarten Sequenzen. Derartige Teilsequenzen können nach dem Fachmann geläufigen Verfahren bspw. chemisch synthetisiert werden und können vorzugsweise als Antigene für die Produktion von Antikörpern eingesetzt werden. Vorzugsweise wird es sich bei diesen Teilabschnitten um solche Regionen der in den Figuren 9, 11 oder 13 offenbarten Sequenzen handeln, die im räumlichen Modell der Proteine zumindest teilweise die Proteinoberfläche ausmachen. Transgene Tiere stellen einen weiteren Gegenstand der vorliegenden Erfindung dar (Anspruch 11) . Bei erfindungsgemäßen transgenen Tieren handelt es sich um Tiere, die genetisch dahingehend verändert sind, daß sie eine im Vergleich zum Normaltier veränderte Menge eines erfindungsgemäßen Genprodukts in mindestens einem Gewebe exprimieren bzw. enthalten. Hierbei sind erfindungsgemäß auch solche Tiere eingeschlossen, die die nativ vorhandene erfindungsgemäße DNA-Sequenz (a) auf der genetischen Ebene entweder tw. oder vollständig nicht mehr aufweisen oder (b) zwar auf der genetischen Ebene erfindungsgemäße Sequenzen aufweisen, diese jedoch nicht transkribieren und/oder translatieren können und daher das Genprodukt nicht mehr enthalten. Darüber hinaus kann (können) bei einem transgenen Tier die native 15B3-Sequenz (en) , 15B3-1- oder 15B3-2-Sequenz (en) (ob vorhanden oder nicht vorhanden) um mindestens eine erfindungsgemäße DNA-Sequenz ergänzt bzw. durch mindestens eine erfindungsgemäße DNA-Sequenz substitu- iert sein. Insbesondere kann es sich bei der (den) substituierten und/oder ergänzten Sequenz (en) um erfindungsgemäße Sequenzen handeln, die nicht-nativer Natur sind.The present invention furthermore relates to at least 20, more preferably at least 30 and even more preferably at least 50, amino acid segments of the sequences disclosed in FIGS. 9, 11 or 13. Such partial sequences can, for example, be chemically synthesized by methods familiar to the person skilled in the art and can preferably be used as antigens for the production of antibodies. These subsections are preferably regions of the sequences disclosed in FIGS. 9, 11 or 13 that at least partially make up the protein surface in the spatial model of the proteins. Transgenic animals are a further subject of the present invention (claim 11). The transgenic animals according to the invention are animals which have been genetically modified in such a way that they express or contain an amount of a gene product according to the invention which is modified in comparison to the normal animal in at least one tissue. Here, according to the invention, those animals are also included that either tw. or completely no longer have, or (b) indeed have sequences according to the invention at the genetic level, but cannot transcribe and / or translate them and therefore no longer contain the gene product. In addition, in a transgenic animal, the native 15B3 sequence (s), 15B3-1 or 15B3-2 sequence (s) (whether present or not present) can be supplemented by at least one DNA sequence according to the invention at least one DNA sequence according to the invention may be substituted. In particular, the substituted and / or supplemented sequence (s) can be sequences according to the invention which are non-native in nature.
Die Herstellung von in bezug auf erfindungsgemäße Sequenzen transgenen und „knock-out" Tieren, insbesondere Mäusen, Ratten Schweinen, Fruchtfliegen oder Zebrafischen, erfolgt auf dem Fachmann geläufige Weise. Hierzu wird z.B. eine 15B3, 15B3-1 oder 15B3-2 cDNA Sequenz oder native oder nicht-native Variante in transgenen Mäusen exprimiert, z.B. unter einem NSE-Promotor in Neuronen, unter einem MBP-Promotor in Oligo- dendrozyten etc.. Die genetisch veränderten Tiere können danach in unterschiedlichen Krankheitsmodellen untersucht werden (z.B. experimentell herbeigeführtem Schlaganfall, MCAO) . Die Herstellung von „knock-out" Tieren kann zudem Hinweise auf die Auswirkungen von Inhibitoren auf den Gesamtorganismen liefern, da ein „knock out Modell" insoweit der Inhibition erfindungsgemäßer nativer Sequenzen entspricht.The production of transgenic and “knock-out” animals, in particular mice, rats, pigs, fruit flies or zebra fish, with respect to the sequences according to the invention, is carried out in a manner familiar to the person skilled in the art. For this purpose, for example, a 15B3, 15B3-1 or 15B3-2 cDNA sequence or native or non-native variant expressed in transgenic mice, eg under an NSE promoter in neurons, under an MBP promoter in oligodendrocytes etc. The genetically modified animals can then be examined in different disease models (eg experimentally induced stroke, MCAO ) The production of "knock-out" animals can also provide clues on the effects of inhibitors on the whole organism, since a "knock out model" corresponds in this respect to the inhibition of native sequences according to the invention.
Sämtliche multizellulären Organismen können erfindungsgemäß transgen ausgestaltet sein, insbesondere Säugetiere, bspw. Mäuse, Ratten, Schafe, Rinder oder Schweine. Auch transgene Pflanzen sind im Prinzip denkbar. Bei den transgenen Organis- men kann es sich auch um sogenannte "Knock-Out"-Tiere handeln. Dabei können die transgenen Tiere eine funktioneile o- der nicht funktionelle erfindungsgemäße Nukleinsäuresequenz oder ein funktionelles oder nicht funktionelles Nukleinsäure- konstrukt allein oder in Kombination mit einer funktionellen oder nicht funktionellen Sequenz, die für 15B3 Proteine kodiert, enthalten.According to the invention, all multicellular organisms can be transgenic, in particular mammals, for example mice, rats, sheep, cattle or pigs. In principle, transgenic plants are also conceivable. The transgenic organisms can also be so-called "knock-out" animals. The transgenic animals can contain a functional or non-functional nucleic acid sequence according to the invention or a functional or non-functional nucleic acid construct alone or in combination with a functional or non-functional sequence which codes for 15B3 proteins.
Eine weitere erfindungsgemäße Ausgestaltung der oben beschriebenen transgenen Tiere sind transgene Tiere, in deren Keimzellen oder der Gesamtheit oder einem Teil der somati- schen Zellen oder in deren Keimzellen oder der Gesamtheit o- der einem Teil der somatischen Zellen die native (n) 15B3- Nukleotidsequenz (en) durch gentechnische Verfahren verändert oder durch Einfügen von DNA-Elementen unterbrochen wurden. Eine weitere Möglichkeit des Einsatzes einer erfindungsgemäßen Nukleotidsequenz oder Teilen davon ist die Erzeugung transgener oder knock-out- oder konditioneller oder regionenspezifischer knock-out Tiere oder spezifischer Mutationen bei gentechnisch veränderten Tieren (Ausubel et al. (eds.) 1998, Current Protocols in Molecular Biology, John Wiley & Sons, New York und Torres et al., (eds.) 1997, Laboratory protocols for conditional gene targeting, Oxford University Press, Oxford) . Über transgene Überexpression oder genetische Mutation (Nullmutation oder spezifische Deletionen, Insertionen oder Veränderungen) durch homologe Rekombination in embryonalen Stamm- zellen kann man Tiermodelle erzeugen, die wertvolle weitere Informationen über die (Patho-) Physiologie der erfindungsgemäßen Sequenzen liefern. Solchermaßen hergestellte Tiermodelle können essentielle Testsysteme zur Evaluierung neuartiger Therapeutika darstellen, die die biologische Funktion von Proteinen gemäß Figuren 9, 11 oder 13 für neurale, vaskuläre oder andere Prozesse beeinflussen.A further embodiment of the transgenic animals described above are transgenic animals, in their germ cells or all or part of the somatic cells or in their germ cells or all or part of the somatic cells the native 15B3 nucleotide sequence (s) have been modified by genetic engineering or interrupted by the insertion of DNA elements. A further possibility of using a nucleotide sequence according to the invention or parts thereof is the generation of transgenic or knock-out or conditional or region-specific knock-out animals or specific mutations in genetically modified animals (Ausubel et al. (Eds.) 1998, Current Protocols in Molecular Biology, John Wiley & Sons, New York and Torres et al., (Eds.) 1997, Laboratory protocols for conditional gene targeting, Oxford University Press, Oxford). Via transgenic overexpression or genetic mutation (zero mutation or specific deletions, insertions or changes) by homologous recombination in embryonic stem cells, animal models can be generated which provide valuable further information about the (patho-) physiology of the sequences according to the invention. Animal models produced in this way can represent essential test systems for evaluating novel therapeutic agents which influence the biological function of proteins according to FIGS. 9, 11 or 13 for neural, vascular or other processes.
Ein weiterer Gegenstand der vorliegenden Erfindung ist ein Antikörper, der ein Epitop auf einem erfindungsgemäßen Gen- produkt, insbesondere einem erfindungsgemäßen Protein gemäß Figuren 9, 11 oder 13 oder Derivaten, Fragmenten oder Isoformen oder Allelen, erkennt (Anspruch 12), aber auch gegen z.B. , erfindungsgemäße mRNA gerichtet sein kann. Der Begriff "Antikörper" umfaßt i.S. der vorliegenden Erfindung sowohl po- lyklonale Antikörper als auch monoklonale Antikörper (Anspruch 13), chimärische Antikörper, anti-idiotypische Antikörper (gerichtet gegen erfindungsgemäße Antikörper) , die alle in gebundener oder löslicher Form vorliegen und ggf. durch "Label" markiert sein können, sowie auch Fragmente der vorge- nannten Antikörper. Neben den Fragmenten von erfindungsgemäßen Antikörpern in Alleinstellung können erfindungsgemäße Antikörper auch in rekombinanter Form als Fusionsproteine mit anderen (Protein) -Bestandteilen auftreten. Fragmente als solche oder Fragmente von erfindungsgemäßen Antikörpern als Be- standteile von Fusionsproteinen werden typischerweise durch die Methoden enzymatischer Spaltung, der Protein-Synthese o- der die dem Fachmann geläufigen Rekombinationsmethoden hergestellt. Als Antikörper werden nach der vorliegenden Erfindung also sowohl polyklonale, monoklonale, humane oder humanisierte oder rekombinante Antikörper oder Fragmente davon, single chain Antikörper oder auch synthetische Antikörper bezeichnet.The present invention furthermore relates to an antibody which recognizes an epitope on a gene product according to the invention, in particular a protein according to the invention according to FIGS. 9, 11 or 13 or derivatives, fragments or isoforms or alleles (claim 12), but also against, for example , mRNA according to the invention can be directed. In the sense of the present invention, the term “antibody” encompasses both polyclonal antibodies and monoclonal antibodies (claim 13), chimeric antibodies, anti-idiotypic antibodies (directed against antibodies according to the invention), all of which are present in bound or soluble form and, if appropriate, by "Label" can be marked, as well as fragments of the aforementioned antibodies. In addition to the fragments of antibodies according to the invention in isolation, antibodies according to the invention can also occur in recombinant form as fusion proteins with other (protein) components. Fragments as such or fragments of antibodies according to the invention as components of fusion proteins are typically produced by the methods of enzymatic cleavage, protein synthesis or the recombination methods familiar to the person skilled in the art. As antibodies according to the present invention thus both polyclonal, monoclonal, human or humanized or recombinant antibodies or fragments thereof, single chain antibodies or synthetic antibodies.
Bei den polyklonalen Antikörpern handelt es sich um heterogene Mischungen von Antikörpermolekülen, die aus Seren von Tieren hergestellt werden, die mit einem Antigen immunisiert worden sind. Zum Gegenstand der Erfindung gehören aber auch polyklonale monospezifische Antikörper, die nach Aufreinigung der Antikörper (bspw. über eine Säule, die mit Peptiden eines spezifischen Epitops beladen sind) erhalten werden. Ein mo- noklonaler Antikörper enthält eine im wesentlichen homogene Population von Antikörpern, die spezifisch gegen Antigene gerichtet sind, wobei die Antikörper im wesentlichen gleiche Epitop-Bindungsstellen aufweisen. Monoklonale Antikörper können durch die im Stand der Technik bekannten Verfahren erhalten werden (z. B. Köhler und Milstein, Nature, 256, 495-397, (1975); US-Patent 4,376,110; Ausübel et al . , Harlow und Lane "Antikörper": Laboratory Manual, Cold Spring, Harbor Labora- tory (1988); Ausubel et al., (eds), 1998, Current Protocols in Molecular Biology, John Wiley & Sons, New York).). Die in den vorgenannten Literaturstellen enthaltene Beschreibung wird als Bestandteil der vorliegenden Erfindung in die Offenbarung der vorliegenden Erfindung einbezogen.The polyclonal antibodies are heterogeneous mixtures of antibody molecules which are produced from sera from animals which have been immunized with an antigen. However, the subject of the invention also includes polyclonal monospecific antibodies which are obtained after the antibodies have been purified (for example via a column which is loaded with peptides of a specific epitope). A monoclonal antibody contains an essentially homogeneous population of antibodies which are specifically directed against antigens, the antibodies having essentially the same epitope binding sites. Monoclonal antibodies can be obtained by methods known in the art (e.g., Koehler and Milstein, Nature, 256, 495-397, (1975); U.S. Patent 4,376,110; Ausübel et al., Harlow and Lane "Antibodies" : Laboratory Manual, Cold Spring, Harbor Laboratory (1988); Ausubel et al., (Eds), 1998, Current Protocols in Molecular Biology, John Wiley & Sons, New York).). The description contained in the abovementioned references is included as part of the present invention in the disclosure of the present invention.
Auch lassen sich gentechnisch manipulierte erfindungsgemäße Antikörper nach Verfahren, wie in den vorgenannten Druckschriften beschrieben herstellen. Kurz gesagt, werden dazu Antikörper-produzierende Zellen angezogen und die mRNA bei ausreichender optischer Dichte der Zellen über Zellyse mit Guanidiniumthiocyanat, Ansäuern mit Natriumacetat, Extraktion mit Phenol, Chloroform/Isoamylalkohol, Fällungen mit Isopro- panol und Waschen mit Ethanol aus den Zellen in bekannter Weise isoliert. Anschließend wird mit Hilfe der Reversen Transcriptase cDNA aus der mRNA synthetisiert. Die synthetisierte cDNA kann direkt oder nach genetischer Manipulation beispielsweise durch "site directed mutagenesis", Einführung von Insertionen, Inversionen, Deletionen oder Basenaustausche in geeignete tierische, pilzliche, bakterielle oder virale Vektoren inseriert und in den entsprechenden Wirtsorganismen exprimiert werden. Bevorzugt werden bakterielle oder Hefe Vektoren wie pBR322, pUC18/19, pACYC184, Lambda oder Hefe-mu- Vektoren zur Klonierung der Gene und die Expression in Bakterien wie E. coli bzw. in der Hefe wie Saccharomyces cerevisi- ae. Spezifische Antikörper gegen die erfindungsgemäßen Proteine können sich sowohl als diagnostische Reagenzien als auch als Therapeutika bei Erkrankungen eignen, bei denen 15B3 von pathophysiologischer Bedeutung ist.Antibodies according to the invention which have been genetically engineered can also be produced by methods as described in the aforementioned publications. In short, antibody-producing cells are attracted to this and the mRNA, provided the optical density of the cells is sufficient, is lysed with guanidinium thiocyanate, acidified with sodium acetate, extracted with phenol, chloroform / isoamyl alcohol, precipitated with isopropyl alcohol. panol and washing with ethanol isolated from the cells in a known manner. Then the reverse transcriptase is used to synthesize cDNA from the mRNA. The synthesized cDNA can be inserted directly or after genetic manipulation, for example by "site directed mutagenesis", introduction of insertions, inversions, deletions or base exchanges into suitable animal, fungal, bacterial or viral vectors and expressed in the corresponding host organisms. Bacterial or yeast vectors such as pBR322, pUC18 / 19, pACYC184, lambda or yeast mu vectors for cloning the genes and expression in bacteria such as E. coli or in yeast such as Saccharomyces cerevisiae are preferred. Specific antibodies against the proteins according to the invention can be suitable both as diagnostic reagents and as therapeutic agents for diseases in which 15B3 is of pathophysiological importance.
Erfindungsgemäße Antikörper können einer der folgenden Im- munglobulinklassen angehören: IgG, IgM, IgE, IgA, GILD und ggf. einer Unterklasse der vorgenannten Klassen, wie die Subklassen des IgG oder deren Mischungen zu verstehen. Bevorzugt sind IgG und seine Subklassen wie beispielsweise IgGl, IgG2, IgG2a, IgG2b, IgG3 oder IgGM. Besonders bevorzugt sind die IgG Subtypen IgGl/k oder IgG2b/k. Ein Hybridom-Zellklon, der erfindungsgemäße monoklonale Antikörper produziert, kann in vitro, in situ oder in vivo kultiviert werden. Die Herstellung von großen Titern an monoklonalen Antikörpern erfolgt vorzugsweise in vivo oder in situ.Antibodies according to the invention can belong to one of the following immunoglobulin classes: IgG, IgM, IgE, IgA, GILD and possibly a subclass of the abovementioned classes, such as the subclasses of the IgG or mixtures thereof. IgG and its subclasses such as IgGl, IgG2, IgG2a, IgG2b, IgG3 or IgGM are preferred. The IgG subtypes IgGl / k or IgG2b / k are particularly preferred. A hybridoma cell clone that produces monoclonal antibodies according to the invention can be cultivated in vitro, in situ or in vivo. Large titers of monoclonal antibodies are preferably produced in vivo or in situ.
Bei den erfindungsgemäßen chimärische Antikörpern handelt es sich um Moleküle, die verschiedene Bestandteile enthalten, wobei diese sich aus verschiedenen Tierarten ableiten (z. B.The chimeric antibodies according to the invention are molecules which contain different constituents, which are derived from different animal species (e.g.
Antikörper, die eine variable Region, die aus einem Mäuse- monoklonalen Antikörper abgeleitet ist, und eine konstante Region eines humanen Immunglobulins aufweisen) . Chimärische Antikörper werden vorzugsweise eingesetzt, um einerseits die Immunogenizität bei der Anwendung zu reduzieren und anderer- seits die Ausbeuten bei der Produktion zu erhöhen, z.B. ergeben murine monoklonale Antikörper höhere Ausbeuten aus Hybri- dom-Zellinien, führen aber auch zu einer höheren Immunogenizität beim Menschen, so daß human/murine chimärische Antikörper vorzugsweise eingesetzt werden. Chimärische Antikörper und Verfahren zu ihrer Herstellung sind aus dem Stand der Technik bekannt (Cabilly et al., Proc. Natl. Sei. USA 81: 3273-3277 (1984); Morrison et al. Proc. Natl. Acad. Sei USA 81:6851-6855 (1984); Boulianne et al . Nature 312 643-646 (1984); Cabilly et al., EP-A-125023; Neuberger et al., Nature 314: 268-270 (1985); Taniguchi et al., EP-A-171496; Morrion et al., EP-A-173494; Neuberger et al., WO 86/01533; Kudo et al., EP-A-184187; Sahagan et al., J. Immuno1. 137: 1066-1074Antibodies that have a variable region derived from a mouse monoclonal antibody is derived, and have a constant region of a human immunoglobulin). Chimeric antibodies are preferably used, on the one hand, to reduce the immunogenicity in use and, on the other hand, to increase the yields in production, for example murine monoclonal antibodies give higher yields from hybrid cell lines, but also lead to higher immunogenicity in humans , so that human / murine chimeric antibodies are preferably used. Chimeric antibodies and methods for their preparation are known from the prior art (Cabilly et al., Proc. Natl. Sei. USA 81: 3273-3277 (1984); Morrison et al. Proc. Natl. Acad. Sei USA 81: 6851-6855 (1984); Boulianne et al. Nature 312 643-646 (1984); Cabilly et al., EP-A-125023; Neuberger et al., Nature 314: 268-270 (1985); Taniguchi et al. , EP-A-171496; Morrion et al., EP-A-173494; Neuberger et al., WO 86/01533; Kudo et al., EP-A-184187; Sahagan et al., J. Immuno1. 137: 1066-1074
(1986); Robinson et al., WO 87/02671; Liu et al., Proc. Natl.(1986); Robinson et al., WO 87/02671; Liu et al., Proc. Natl.
Acad. Sei USA 84:3439-3443 (1987); Sun et al., Proc. Natl. Acad. Sei USA 84:214218 (1987); Better et al., Science 240: 1041-1043 (1988) und Harlow und Lane, Antikörper: A Laborato- ry Manual, wie oben zitiert. Diese Zitatstellen werden als zur Offenbarung gehörig in die vorliegende Erfindung einbezogen.Acad. Sci USA 84: 3439-3443 (1987); Sun et al., Proc. Natl. Acad. Be USA 84: 214218 (1987); Better et al., Science 240: 1041-1043 (1988) and Harlow and Lane, Antibodies: A Laboratory Manual, as cited above. These citations are included as part of the disclosure in the present invention.
Ganz besonders bevorzugt wird ein solcher erfindungsgemäßer Antikörper gegen einen Leucin reichen Sequenzabschnitt auf einem Protein gemäß Figuren 9, 11 oder 13 als Epitop gerichtet sein (Anspruch 14) .Such an antibody according to the invention against a leucine-rich sequence section on a protein according to FIGS. 9, 11 or 13 is very particularly preferably directed as an epitope (claim 14).
Ein erfindungsgemäßer anti-idiotypischer Antikörper ist ein Antikörper, der eine Determinante, die im allgemeinen mit derAn anti-idiotypic antibody of the present invention is an antibody which is a determinant generally related to that
Antigenbindungsstelle eines erfindungsgemäßen Antikörpers as- soziiert ist, erkennt. Ein anti-idiotypiseher Antikörper kann durch die Immunisierung eines Tieres der gleichen Art und des gleichen genetischen Typs (z.B. eines Mäusestamms) als Ausgangspunkt für einen monoklonalen Antikörper, gegen welchen ein erfindungsgemäßer anti-idiotypiseher Antikörper gerichtet ist, hergestellt werden. Das immunisierte Tier wird die idio- typischen Determinanten des immunisierenden Antikörpers durch die Produktion eines Antikörpers, der gegen die idiotypischen Determinanten gerichtet ist (nämlich ein erfindungsgemäßer anti-idiotypiseher Antikörper), erkennen (U.S. 4,699,880). Ein erfindungsgemäßer anti-idiotypiseher Antikörper kann auch als Immunogen eingesetzt werden, um eine Immunantwort in einem weiteren Tier hervorzurufen und um dort zur Produktion eines sog. anti-anti-idiotypischen Antikörpers zu führen. Der anti-anti-idiotypische Antikörper kann, muß aber nicht, bezüglich seiner Epitop-Konstruktion identisch mit dem originären monoklonalen Antikörper sein, der die anti-idiotypische Reaktion hervorgerufen hat. Auf diese Weise können durch die Verwendung von gegen idiotypische Determinanten eines mo- noklonalen Antikörpers gerichtete Antikörper andere Klone, die Antikörper von identischer Spezifität exprimieren, identifiziert werden.Antigen binding site of an antibody according to the invention as is sociated, recognizes. An anti-idiotypic antibody can be produced by immunizing an animal of the same type and the same genetic type (for example a mouse strain) as the starting point for a monoclonal antibody against which an anti-idiotypic antibody according to the invention is directed. The immunized animal will recognize the idiotypical determinants of the immunizing antibody by the production of an antibody which is directed against the idiotypic determinants (namely an anti-idiotypisher antibody according to the invention) (US Pat. No. 4,699,880). An anti-idiotypic antibody according to the invention can also be used as an immunogen in order to elicit an immune response in another animal and to produce an anti-idiotypic antibody there. The epitope construction of the anti-anti-idiotypic antibody may, but need not, be identical to the original monoclonal antibody that caused the anti-idiotypic reaction. In this way, by using antibodies directed against idiotypic determinants of a monoclonal antibody, other clones which express antibodies of identical specificity can be identified.
Monoklonale Antikörper, die gegen erfindungsgemäße Proteine, Analoge, Fragmente oder Derivate dieser erfindungsgemäßen Proteine gerichtet sind, können eingesetzt werden, um die Bindung von anti-idiotypischen Antikörpern in entsprechenden Tieren, wie z. B. der BALB/c Maus, zu induzieren. Zellen aus der Milz einer solchen immunisierten Maus können verwendet werden, um anti-idiotypische Hybridom-Zellinien, die anti- idiotypische monoklonale Antikörper sekretieren, zu produzieren. Weiterhin können anti-idiotypische monoklonale Antikörper auch an einen Träger gekoppelt werden (KLH, "keyhole lim- pet hemocyanin") und dann verwendet werden, um weitere BALB/c-Mäuse zu immunisieren. Die Sera dieser Mäuse enthalten dann anti-anti-idiotypische Antikörper, die die Bindungseigenschaften der originären monoklonalen Antikörper haben und spezifisch für ein Epitop des erfindungsgemäßen Proteins oder eines Fragments oder Derivats von demselben sind. Die anti- idiotypischen monoklonalen Antikörper haben auf diese Weise ihre eigenen idiotypischen Epitope oder "Idiotope", die strukturell mit dem zu untersuchenden Epitop ähnlich sind.Monoclonal antibodies, which are directed against proteins, analogs, fragments or derivatives of these proteins according to the invention, can be used to bind anti-idiotypic antibodies in corresponding animals, such as, for. B. the BALB / c mouse. Spleen cells from such an immunized mouse can be used to produce anti-idiotypic hybridoma cell lines that secrete anti-idiotypic monoclonal antibodies. Anti-idiotypic monoclonal antibodies can also be coupled to a carrier (KLH, "keyhole lim- pet hemocyanin ") and can then be used to immunize further BALB / c mice. The sera of these mice then contain anti-anti-idiotypic antibodies which have the binding properties of the original monoclonal antibodies and are specific for an epitope of the protein according to the invention or one The anti-idiotypic monoclonal antibodies thus have their own idiotypic epitopes or "idiotopes" which are structurally similar to the epitope under investigation.
Die Bezeichnung "Antikörper" soll sowohl intakte Moleküle als auch Fragmente derselben einschließen. Als Fragmente seien alle verkürzten oder veränderten Antikörperfragmente mit einer oder zwei dem Antigen-komplementären Bindungsstellen, wie Antikörperteile mit einer den Antikörper entsprechenden von leichter und schwerer Kette gebildeten Bindungsstelle wie Fv- , Fab- oder F(ab' ) 2-Fragmente oder Einzelstrangfragmente, genannt. Bevorzugt sind verkürzte Doppelstrangfragmente wie Fv- , Fab- oder F(ab')2. Fab und F(ab' ) 2-Fragmente entbehren eines Fc-Fragments, wie etwa in einem intakten Antikörper vorhanden, so daß sie im Blutkreislauf schneller transportiert werden können und vergleichsweise weniger nicht-spezifische Gewebsbindung als intakte Antikörper aufweisen. Hierbei wird hervorgehoben, daß Fab und F(ab')2 Fragmente von erfindungsgemäßen Antikörpern bei der Detektion und Quantifizierung von erfindungsgemäßen Proteinen eingesetzt werden können. Solche Fragmente werden typischerweise durch proteolytische Spaltung hergestellt, indem Enzyme, wie z. B. Papain (zur Herstellung von Fab- Fragmenten) oder Pepsin (zur Herstellung von F(ab' ) 2, -Frag- menten) verwendet werden, oder durch chemische Oxidation oder durch gentechnische Manipulation der Antikörpergene erhalten werden. Gegenstand der vorliegenden Erfindung sind auch Mischungen von Antikörpern im Sinne der vorliegenden Erfindung. Neben den Antikörpern können auch Mischungen von Antikörpern für alle gemäß der vorliegenden Erfindung beschriebenen Verfahren oder Verwendungen eingesetzt werden. Sowohl gereinigte Fraktionen monoklonaler Antikörper, polyklonale Antikörper oder Mischungen monoklonaler Antikörper kommen als Arzneimittel zum Einsatz und finden Verwendung bei der Herstellung von Arzneimitteln zur Behandlung von cerebralen Ischämien (z.B. Schlaganfall) , degenerativen Erkrankungen, insbesondere neu- rodegenerativen Erkrankungen, und neurologischen Erkrankungen, wie z.B. Epilepsie, wie auch Tumorerkrankungen.The term "antibody" is intended to include both intact molecules and fragments thereof. Fragments are all shortened or modified antibody fragments with one or two binding sites complementary to the antigen, such as antibody parts with a binding site formed by the light and heavy chain corresponding to the antibody, such as Fv, Fab or F (ab ') 2 fragments or single-strand fragments, called. Shortened double-strand fragments such as Fv, Fab or F (ab ') 2 are preferred. Fab and F (ab ') 2 fragments lack an Fc fragment, such as present in an intact antibody, so that they can be transported faster in the bloodstream and have comparatively less non-specific tissue binding than intact antibodies. It is emphasized here that Fab and F (ab ') 2 fragments of antibodies according to the invention can be used in the detection and quantification of proteins according to the invention. Such fragments are typically made by proteolytic cleavage using enzymes such as. B. papain (for the production of Fab fragments) or pepsin (for the production of F (ab ') 2 , fragments) can be used, or can be obtained by chemical oxidation or by genetic engineering manipulation of the antibody genes. The present invention also relates to mixtures of antibodies for the purposes of the present invention. In addition to the antibodies, mixtures of antibodies can also be used for all of the methods or uses described according to the present invention. Both purified fractions of monoclonal antibodies, polyclonal antibodies or mixtures of monoclonal antibodies are used as medicaments and are used in the production of medicaments for the treatment of cerebral ischemia (eg stroke), degenerative diseases, in particular newly-regenerative diseases, and neurological diseases, such as, for example Epilepsy, as well as tumor diseases.
Erfindungsgemäße Antikörper, einschließlich der Fragmente von diesen Antikörpern, bzw. deren Mischungen können zur quanti- tativen oder qualitativen Detektion von erfindungsgemäßem Genprodukt, insbesondere Proteinen gemäß Figuren 9, 11 oder 13 oder deren Fragmente oder Derivate, in einer Probe eingesetzt werden oder auch zur Detektion von Zellen, die erfindungsgemäße Proteine exprimieren und ggf. sekretieren. Inso- weit wird die Verwendung erfindungsgemäßer Antikörper als Di- agnostika offenbart. So kann etwa über erfindungsgemäße Antikörper die Menge an erfindungsgemäßem Genprodukt und u.U. deren Aktivität (z.B. spezifische Phosphorylierungen) der Proteine gemäß Figuren 9, 11 oder 13 bestimmt werden. Die Detek- tion kann mit Hilfe von Immunofluoreszenz-Verfahren erreicht werden, die Fluoreszenz-markierte Antikörper in Kombination mit Lichtmikroskopie, Flußzytometrie oder fluorometrischer Detektion durchgeführt werden.Antibodies according to the invention, including the fragments of these antibodies, or mixtures thereof, can be used in a sample for the quantitative or qualitative detection of a gene product according to the invention, in particular proteins according to FIGS. 9, 11 or 13 or their fragments or derivatives, or also for detection of cells that express and, if necessary, secrete proteins according to the invention. In this respect, the use of antibodies according to the invention as diagnostic agents is disclosed. For example, the amount of gene product according to the invention and possibly. the activity (e.g. specific phosphorylation) of the proteins is determined according to FIGS. 9, 11 or 13. The detection can be achieved with the aid of immunofluorescence methods, the fluorescence-labeled antibodies in combination with light microscopy, flow cytometry or fluorometric detection.
Erfindungsgemäße Antikörper im Sinne der Erfindung (dies schließt Fragmente dieser Antikörper ein oder auch Mischungen von Antikörpern) eignen sich für histologische Untersuchun- gen, wie z.B. im Rahmen der Immunofluoreszenz oder Immunoe- lektromikroskopie, für die in situ Detektion eines erfindungsgemäßen Proteins. Die in situ Detektion kann dadurch erfolgen, daß eine histologische Probe von einem Patienten ge- nommen wird und markierte erfindungsgemäße Antikörper zu einer solchen Probe hinzugegeben werden. Der Antikörper (oder ein Fragment dieses Antikörpers) wird in markierter Form auf die biologische Probe aufgetragen. Auf diese Weise ist es nicht nur möglich, die Anwesenheit von erfindungsgemäßem Pro- tein in der Probe zu bestimmen, sondern auch die Verteilung des erfindungsgemäßen Proteins in dem untersuchten Gewebe. Bei der biologischen Probe kann es sich um eine biologische Flüssigkeit, ein Gewebeextrakt, geerntete Zellen, wie z. B. Immunzellen oder Herzmuskel- oder Leberzellen, oder allgemein um Zellen, die in einer Gewebekultur inkubiert worden sind, handeln. Die Detektion des markierten Antikörpers kann je nach Art der Markierung durch im Stand der Technik bekannte Verfahren (z. B. durch Fluoreszenzverfahren) erfolgen. Die biologische Probe kann aber auch auf einem Festphasenträger, wie z. B. Nitrocellulose oder ein anderes Trägermaterial, aufgetragen werden, so daß die Zellen, Zellteile oder löslichen Proteine immobilisiert werden. Der Träger kann dann mit einem geeigneten Puffer ein- oder mehrfach gewaschen werden, wobei nachfolgend mit einem detektierbar markierten Antikör- per nach der vorliegenden Erfindung behandelt wird. Der Festphasenträger kann dann mit dem Puffer ein zweites Mal gewaschen werden, um nicht-gebundenen Antikörper zu beseitigen. Die Menge an gebundener Markierung auf dem Festphasenträger kann dann mit einem herkömmlichen Verfahren bestimmt werden.Antibodies according to the invention in the sense of the invention (this includes fragments of these antibodies or mixtures of antibodies) are suitable for histological examinations. gene, such as in the context of immunofluorescence or immunoelectromicroscopy, for the in situ detection of a protein according to the invention. The in situ detection can take place in that a histological sample is taken from a patient and labeled antibodies according to the invention are added to such a sample. The antibody (or a fragment of this antibody) is applied to the biological sample in labeled form. In this way it is not only possible to determine the presence of protein according to the invention in the sample, but also the distribution of the protein according to the invention in the tissue examined. The biological sample can be a biological fluid, a tissue extract, harvested cells, such as. B. immune cells or cardiac muscle or liver cells, or generally cells that have been incubated in a tissue culture. Depending on the type of labeling, the labeled antibody can be detected by methods known in the art (eg by fluorescence methods). The biological sample can also on a solid phase support, such as. B. nitrocellulose or another carrier material, so that the cells, cell parts or soluble proteins are immobilized. The carrier can then be washed one or more times with a suitable buffer, with subsequent treatment with a detectably labeled antibody according to the present invention. The solid phase support can then be washed with the buffer a second time to remove unbound antibody. The amount of bound label on the solid phase support can then be determined using a conventional method.
Als Träger eignen sich insbesondere Glas, Polystyrol, Polypropylen, Polyethylen, Dextran, Nylon-Amylasen, natürliche oder modifizierte Zellulosen, Polyacrylamide und Magnetit. Der Träger kann entweder bedingt löslichen oder unlöslichen Charakters sein, um die Bedingungen nach Maßgabe der vorliegenden Erfindung zu erfüllen. Das Trägermaterial kann beliebige Formen einnehmen, z. B. in Form von Kügelchen ("beads"), oder zylindrisch oder sphärisch sein, wobei Polystyrol- Kügelchen als Träger bevorzugt sind.Glass, polystyrene, polypropylene, polyethylene, dextran, nylon amylases, natural or modified celluloses, polyacrylamides and magnetite are particularly suitable carriers. The carrier can be of either partially soluble or insoluble character to meet the conditions of the present invention. The carrier material can take any shape, e.g. B. in the form of beads, or cylindrical or spherical, with polystyrene beads are preferred as a carrier.
Eine detektierbare Antikörpermarkierung kann auf verschiedene Weise erfolgen. Beispielsweise kann der Antikörper an ein Enzym gebunden werden, wobei das Enzym schließlich in einem Im- munoassay (EIA) eingesetzt werden kann. Das Enzym kann dann später mit einem entsprechenden Substrat reagieren, so daß eine chemische Verbindung entsteht, die auf eine dem Fachmann geläufige Art und Weise detektiert und ggf. quantifiziert werden kann, z. B. durch Spektrophotometrie, Fluorometrie o- der andere optische Verfahren. Bei dem Enzym kann es sich um Malat-Dehydrogenase, Staphylokokken-Nuklease, delta-5-Steroid Isomerase, Hefe-Alkohol-Dehydrogenase, alpha-Glycerophosphat- dehydrogenase, Triosephosphatisomerase, Meerrettich- Peroxidase, alkalische Phosphatase, Aspariginase, Glucoseoxi- dase, beta-Galactosidase, Ribonuklease, Urease, Katalase, Glucose-6-phosphat-Dehydrogenase, Glucoamylase oder Acetyl- cholinesterase handeln. Die Detektion wird dann über ein chromogenes Substrat, das spezifisch für das für die Markierung eingesetzte Enzym ist, ermöglicht und kann schließlich z.B. über Sichtvergleich des durch die Enzymreaktion umgesetzten Substrats im Vergleich zu Kontrollstandards erfolgen.Detectable antibody labeling can be done in different ways. For example, the antibody can be bound to an enzyme, the enzyme finally being used in an immunoassay (EIA). The enzyme can then react later with a corresponding substrate, so that a chemical compound is formed which can be detected in a manner familiar to the person skilled in the art and, if necessary, quantified, e.g. B. by spectrophotometry, fluorometry or the other optical method. The enzyme can be malate dehydrogenase, staphylococcal nuclease, delta-5 steroid isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate dehydrogenase, triosephosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparticase, glucose oxydase, -Galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase or acetylcholinesterase. The detection is then enabled via a chromogenic substrate that is specific for the enzyme used for the labeling and can finally be e.g. by visual comparison of the substrate converted by the enzyme reaction compared to control standards.
Weiterhin kann die Detektion durch andere Immunoassays sichergestellt werden, z.B. durch radioaktive Markierung der Antikörper oder Antikörperfragmente (also durch einen Radi- oimmunoassay (RIA; Laboratory Techniques and Biochemistry in Molecular Biology, Work, T. et al. North Holland Publishing Company, New York (1978) . Das radioaktive Isotop kann dabei durch die Verwendung von Szintillationszählern oder durch Au- toradigraphie detektiert und quantifiziert werden.Furthermore, the detection can be ensured by other immunoassays, for example by radioactive labeling of the antibodies or antibody fragments (that is to say by means of a radioimmunoassay (RIA; Laboratory Techniques and Biochemistry in Molecular Biology, Work, T. et al. North Holland Publishing Company, New York (1978). The radioactive isotope can be detected and quantified using scintillation counters or auto-radigraphy.
Fluoreszierende Verbindungen können gleichfalls zur Markie- rung eingesetzt werden, beispielsweise Verbindungen wie Fluo- rescinisothiocyanat, Rhodamin, Phyoerythrin, Phycocyanin, Al- lophycocyanin, o-Phthaldehyd und Fluorescamin. Auch fluores- zensemittierende Metalle, wie z. B. 152E oder andere Metalle aus der Lanthanid-Gruppe, können eingesetzt werden. Diese Me- talle werden an den Antikörper über Chelatgruppen, wie z. B. Diethylentriaminpentaessigsäure (ETPA) oder EDTA angekoppelt. Weiterhin kann der erfindungsgemäße Antikörper über eine mit Hilfe von Chemilumineszenz wirkende Verbindung angekoppelt werden. Die Gegenwart des Chemilumineszenz-markierten Anti- körpers wird dann über die Lumineszenz, die im Verlauf einer chemischen Reaktion entsteht, detektiert. Beispiele für der- artige Verbindungen sind Luminol, Isoluminol, Acridiniu- mester, Imidazol, Acridiniumsalz oder Oxalatester. Gleichermaßen können auch biolumineszente Verbindungen zum Einsatz kommen. Biolumineszenz ist eine Unterart der Chemilumineszenz, die bei biologischen Systemen vorgefunden wird, wobei ein katalytisches Protein die Effizienz der chemilumineszen- ten Reaktion verstärkt. Die Detektion des biolumineszenten Proteins erfolgt wiederum über die Lumineszenz, wobei als bi- olumineszente Verbindung beispielsweise Luciferin, Luciferase oder „Aequorin in Betracht kommen.Fluorescent compounds can also be used for labeling, for example compounds such as fluororescin isothiocyanate, rhodamine, phyoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde and fluorescamine. Also fluorescent-emitting metals, such as. B. 152 E or other metals from the lanthanide group can be used. These metals are attached to the antibody via chelate groups, such as. B. Diethylenetriaminepentaacetic acid (ETPA) or EDTA coupled. Furthermore, the antibody according to the invention can be coupled via a compound which acts with the aid of chemiluminescence. The presence of the chemiluminescence-labeled antibody is then detected via the luminescence which arises in the course of a chemical reaction. Examples of such compounds are luminol, isoluminol, acridinium ester, imidazole, acridinium salt or oxalate ester. Similarly, bioluminescent compounds can also be used. Bioluminescence is a subspecies of chemiluminescence found in biological systems, where a catalytic protein increases the efficiency of the chemiluminescent reaction. The detection of the bioluminescent protein is again carried out via luminescence, with luciferin, luciferase or “aequorin” being considered as the bioluminescent compound.
Ein erfindungsgemäßer Antikörper kann für die Verwendung in einem immunometrisehen Assay, auch bekannt als "two-site" o- der "sandwich" Assay, zur Anwendung gelangen. Typische immu- nometrische Assay-Systeme schließen sog. "Vorwärts"-Assays ein, die sich dadurch auszeichnen, daß erfindungsgemäße Anti- körper an ein Festphasensystem gebunden sind und daß der Antikörper mit der Probe, die untersucht wird, auf diese Weise in Kontakt gebracht wird. Derart wird das Antigen aus der Probe durch die Bildung eines binären Festphasen-Antikörper- Antigen-Komplexes aus der Probe isoliert. Nach einer geeigneten Inkubationszeit wird der feste Träger gewaschen, um den verbleibenden Rest der flüssigen Probe zu beseitigen, einschließlich des ggf. nicht gebundenen Antigens, und daraufhin mit einer Lösung in Kontakt gebracht, die eine unbekannte Menge an markiertem Detektionsantikörper enthält. Der markierte Antikörper dient hierbei als sog. Reporter-Molekül. Nach einer zweiten Inkubationszeit, die es den markierten Antikörper erlaubt, mit dem an die Festphase gebundenen Antigen zu assoziieren, wird der Festphasenträger erneut gewaschen, um markierte Antikörper, die nicht reagiert haben, zu beseitigen.An antibody according to the invention can be used for use in an immunometric assay, also known as a "two-site" or "sandwich" assay. Typical immunometric assay systems include so-called "forward" assays, which are distinguished by the fact that body are bound to a solid phase system and that the antibody is brought into contact with the sample being examined in this way. In this way, the antigen is isolated from the sample by the formation of a binary solid phase-antibody-antigen complex from the sample. After an appropriate incubation period, the solid support is washed to remove the remainder of the liquid sample, including any unbound antigen, and then contacted with a solution containing an unknown amount of the labeled detection antibody. The labeled antibody serves as a so-called reporter molecule. After a second incubation period that allows the labeled antibody to associate with the antigen bound to the solid phase, the solid phase support is washed again to remove unreacted labeled antibodies.
In einer alternativen Assay-Form kann auch ein sog. "sand- wich"-Assay zum Einsatz kommen. Hierbei kann ein einziger Inkubationsschritt ausreichen, wenn der an die Festphase gebun- dene Antikörper und der markierte Antikörper beide gleichzeitig auf die zu testende Probe aufgebracht werden. Nach Abschluß der Inkubation wird der Festphasenträger gewaschen, um Rückstände der flüssigen Probe und der nicht-assoziierten markierten Antikörper zu beseitigen. Die Anwesenheit von mar- kiertem Antikörper auf dem Festphasenträger wird genau so bestimmt, wie bei den konventionellen "Vorwärts"-Sandwich- Assay. Bei dem sog. reversen Assay wird schrittweise zunächst eine Lösung des markierten Antikörpers zur Flüssigprobe hinzugefügt, gefolgt von der Beimischung von nicht-markiertem Antikörper, gebunden an einen Festphasenträger, nach Ablauf einer geeigneten Inkubationszeit. Nach einem zweiten Inkubationsschritt wird der Festphasenträger in herkömmlicher Weise gewaschen, um ihn von Probenüberresten und von markiertem Antikörper, der nicht reagiert hat, zu befreien. Die Bestimmung des markierten Antikörpers, der mit dem Festphasenträger reagiert hat, wird dann, so wie oben beschrieben, durchgeführt.In an alternative form of assay, a so-called "sandwich" assay can also be used. A single incubation step can be sufficient if the antibody bound to the solid phase and the labeled antibody are both applied to the sample to be tested at the same time. After the incubation is complete, the solid phase support is washed to remove residues of the liquid sample and the non-associated labeled antibodies. The presence of labeled antibody on the solid phase support is determined in the same way as in the conventional "forward" sandwich assay. In the so-called reverse assay, a solution of the labeled antibody is first gradually added to the liquid sample, followed by the addition of unlabeled antibody, bound to a solid phase support, after a suitable incubation time has elapsed. After a second incubation step, the solid phase support becomes conventional washed to remove sample residues and labeled antibody that has not responded. The determination of the labeled antibody which has reacted with the solid phase support is then carried out as described above.
Nach der vorliegenden Erfindung werden weiterhin Verfahren zur Expression von erfindungsgemäßen Genprodukten, insbesondere also von Polypeptiden gemäß Figuren 9, 11 oder 13, einschließlich aller Derivate, Analoge und Fragmente, offenbart, wobei hierfür Wirtszellen mit einem erfindungsgemäßen Expressionsvektor transformiert werden (Anspruch 15) . Dieses Verfahren zur Expression von Genprodukten, die auf einer erfindungsgemäßen DNA-Sequenz beruhen, dient nicht dazu, das entsprechende Genprodukt zu konzentrieren und aufzureinigen, sondern viel- mehr dazu, den Zellstoffwechsel durch das Einführen der erfindungsgemäßen DNA-Sequenzen über die Expression des dazugehörigen Genprodukts zu beeinflussen. Hier ist insbesondere an die Verwendung der mit Hilfe von Expressionsvektoren transformierten Wirtszellen als Arzneimittel bzw. zur Herstellung eines Arzneimittels, insbesondere zum Zwecke der Behandlung von Erkrankungen, bspw. von Tumorerkrankungen, neurologischen Erkrankungen, neurodegenerativen Erkrankungen (bspw. Multipler Sklerose, Morbus Parkinson), cerebralen Ischämien (z.B. Schlaganfall) . Allgemein werden erfindungsgemäße Wirtszellen bei Erkrankungen zur Verfügung gestellt, denen eine Fehlregulation der Apoptose, der Nekrose, der Zeilproliferation, des Zellwachstums oder der Zellplastizität zugrunde liegt. Die derart erfindungsgemäß ex vivo transformierten autologen oder allogenen Wirtszellen können dann Patienten transplantiert werden.According to the present invention, methods for the expression of gene products according to the invention, in particular thus of polypeptides according to FIGS. 9, 11 or 13, including all derivatives, analogs and fragments, are further disclosed, for which host cells are transformed with an expression vector according to the invention (claim 15). This method for the expression of gene products which are based on a DNA sequence according to the invention does not serve to concentrate and purify the corresponding gene product, but rather to improve the cell metabolism by introducing the DNA sequences according to the invention via the expression of the associated gene product to influence. Here, in particular, the use of the host cells transformed with the aid of expression vectors as a medicament or for the production of a medicament, in particular for the purpose of treating diseases, for example tumor diseases, neurological diseases, neurodegenerative diseases (for example multiple sclerosis, Parkinson's disease), cerebral ischemia (e.g. stroke). In general, host cells according to the invention are made available in the case of diseases which are based on incorrect regulation of apoptosis, necrosis, cell proliferation, cell growth or cell plasticity. The autologous or allogeneic host cells transformed in this way ex vivo according to the invention can then be transplanted to patients.
Gemäß einem weiteren Aspekt der vorliegenden Erfindung wird ein Verfahren zur Isolierung von Genprodukten mit mindestens einer mit der erfindungsgemäßen Aminosäuresequenz von 15B3, 15B3-1 oder 15B3-2 zumindest über eine Teilsequenz von mindestens 20, vorzugsweise mindestens 30 AS homologen Teilsequenz, wobei die Wirtszellen mit einem erfindungsgemäßen Expressions- vektor transformiert und dann unter geeigneten, die Expression fördernden Bedingungen kultiviert werden, so daß das Genprodukt schließlich aus der Kultur aufgereinigt werden kann (Anspruch 16) . Das erfindungsgemäße Genprodukt der erfindungsgemäßen DNA-Sequenz kann dabei, abhängig von dem Expressionssys- tem, aus einem Kulturmedium oder aus Zellextrakten isoliert werden. Der Fachmann kann ohne weiteres erkennen, daß die jeweiligen Isolierungsmethoden und das Verfahren bei der Aufreinigung des von einer erfindungsgemäßen DNA kodierten, rekombinanten Proteins stark vom Typ der Wirtszelle oder auch von dem Umstand, ob das Protein in das Medium sekretiert wird, abhängt. Zum Beispiel können Expressionssysteme eingesetzt werden, die zur Sekretion des rekombinanten Proteins aus der Wirtszelle führen. Das Kulturmedium muß in diesem Fall durch kommerziell erhältliche Proteinkonzentrationsfilter, z.B. Ami- con oder Millipore Pelicon, aufkonzentriert werden. Nach dem Konzentrationsschritt kann ein Reinigungsschritt erfolgen, z.B. ein Gelfiltrationsschritt oder eine Reinigung mit Hilfe von säulenchromatographische Methoden. Alternativ kann aber auch ein Anionenaustauscher eingesetzt werden, der eine Matrix mit DEAE aufweist.According to a further aspect of the present invention, a method for isolating gene products with at least a partial sequence which is homologous with the amino acid sequence of 15B3, 15B3-1 or 15B3-2 according to the invention, at least over a partial sequence of at least 20, preferably at least 30 AS, the host cells being transformed with an expression vector according to the invention and then cultivated under suitable conditions which promote expression are so that the gene product can finally be purified from the culture (claim 16). The gene product according to the invention of the DNA sequence according to the invention can, depending on the expression system, be isolated from a culture medium or from cell extracts. The person skilled in the art can readily recognize that the respective isolation methods and the method for the purification of the recombinant protein encoded by a DNA according to the invention strongly depend on the type of the host cell or also on the fact whether the protein is secreted into the medium. For example, expression systems can be used which lead to the secretion of the recombinant protein from the host cell. In this case, the culture medium must be concentrated using commercially available protein concentration filters, for example Amicon or Millipore Pelicon. After the concentration step, a purification step can be carried out, for example a gel filtration step or a purification using column chromatography methods. Alternatively, however, an anion exchanger can be used which has a matrix with DEAE.
Als .Matrix dienen dabei alle aus der Proteinreinigung bekannten Materialien, z.B. Acrylamid oder Agarose oder Dextran oder ähnliches. Es kann aber auch ein Kationenaustauscher eingesetzt werden, der dann typischerweise Carboxymethyl-Gruppen enthält. Zur weiteren Reinigung eines durch eine erfindungsgemäße DNA codierten Polypeptids können dann HPLC-Schritte dienen. Es kann sich um einen oder mehrere Schritte handeln. Insbesondere wird die "Reversed- Phase"- Methode eingesetzt. Diese Schritte dienen zum Erhalt eines im wesentlichen homogenen rekombinanten Proteins einer erfindungsgemäßen DNA-Sequenz.All materials known from protein purification, such as acrylamide or agarose or dextran or the like, serve as the matrix. However, a cation exchanger can also be used, which then typically contains carboxymethyl groups. HPLC steps can then be used to further purify a polypeptide encoded by a DNA according to the invention. It can be one or more Act steps. In particular, the "reversed phase" method is used. These steps serve to obtain an essentially homogeneous recombinant protein of a DNA sequence according to the invention.
Neben bakteriellen Zellkulturen zur Isolierung des Genprodukts können auch transformierte Hefezellen eingesetzt werden. In diesem Fall kann das translatierte Protein sekretiert werden, so daß die Proteinreinigung vereinfacht wird. Sekretiertes re- kombinantes Protein aus einer Hefewirtszelle kann durch Methoden erhalten werden, wie sie bei Urdal et al. (J. Chromato. 296:171 (1994)) offenbart sind und Bestandteil der Offenbarung der vorliegenden Erfindung sind.In addition to bacterial cell cultures for isolating the gene product, transformed yeast cells can also be used. In this case, the translated protein can be secreted, so that protein purification is simplified. Secreted recombinant protein from a yeast host cell can be obtained by methods as described in Urdal et al. (J. Chromato. 296: 171 (1994)) and form part of the disclosure of the present invention.
Erfindungsgemäße Nukleinsäuresequenzen, insbesondere erfindungsgemäße DNA-Sequenzen, und/oder erfindungsgemäße Genprodukte können als Arzneimittel bzw. zur Herstellung eines Arzneimittels Verwendung finden (Anspruch 17) . Diese können als solche verabreicht werden (bspw. bukkal, intravenös, oral, parenteral, nasal, subkutan) oder in Kombination mit weiteren Wirk-, Hilfs- oder arzneimitteltypischen Zusatzstoffen. Erfindungsgemäße Nukleinsäure kann als nackte Nukleinsäure, insbesondere intravenös, injiziert werden oder aber mit Hilfe von Vektoren dem Patienten verabreicht werden. Bei diesen Vektoren kann es sich um Plasmide als solche handeln, aber auch um virale Vektoren, insbesondere retrovirale oder adeno- virale Vektoren, oder auch um Liposomen, die nackte erfindungsgemäße DNA oder ein Plasmid, das erfindungsgemäße DNA enthält, aufweisen können.Nucleic acid sequences according to the invention, in particular DNA sequences according to the invention, and / or gene products according to the invention can be used as medicaments or for the production of a medicament (claim 17). These can be administered as such (for example buccally, intravenously, orally, parenterally, nasally, subcutaneously) or in combination with other active ingredients, auxiliaries or additives typical of drugs. Nucleic acid according to the invention can be injected as naked nucleic acid, in particular intravenously, or can be administered to the patient using vectors. These vectors can be plasmids as such, but also viral vectors, in particular retroviral or adenoviral vectors, or also liposomes which can have naked DNA according to the invention or a plasmid which contains DNA according to the invention.
Die Verwendung von erfindungsgemäßen Sequenzen, insbesondere der Nukleotid- oder Aminosäuresequenzen von 15B3, 15B3-1 oder 15B3-2 bzw. deren Varianten, sowie erfindungsgemäßer Protein- heteromere sowie davon abgeleiteter erfindungsgemäßer Reagenzien (Oligonukleotide, Antikörper, Peptide) kommt somit für die Herstellung eines Arzneimittels zu therapeutischen Zwecken, d.h. zur Behandlung von Erkrankungen, in Betracht. Ganz besonders bevorzugt ist dabei der therapeutische Einsatz zur Behandlung bzw. zur Herstellung eines Arzneimittels zur Behandlung von Erkrankungen oder pathophysiologischen Zuständen, die bspw. auf fehlgesteuerter Regulation der Homöostase von Zelltod- und Proliferationsereignissen beruhen (Anspruch 18) . Durch die erfindungsgemäße Verwendung können Zelltodprozesse, z.B. Kaskaden, die zur Apoptose führen, oder Prozesse, die zur Nekrose führen, in allen Zelltypen, die 15B3, 15B3-1 und/oder 15B3-2 und oder eine native Variante hiervon exprimieren, insbesondere in neuralen Zellen, beeinflusst werden, bspw. durch Modulation von Zell-Zell-Interaktionen.The use of sequences according to the invention, in particular the nucleotide or amino acid sequences of 15B3, 15B3-1 or 15B3-2 or their variants, and protein proteins according to the invention Heteromeric and reagents according to the invention derived therefrom (oligonucleotides, antibodies, peptides) are thus suitable for the production of a medicament for therapeutic purposes, ie for the treatment of diseases. The therapeutic use for the treatment or for the manufacture of a medicament for the treatment of diseases or pathophysiological conditions, which are based, for example, on incorrectly controlled regulation of the homeostasis of cell death and proliferation events, is very particularly preferred (claim 18). Through the use according to the invention, cell death processes, for example cascades which lead to apoptosis or processes which lead to necrosis, can be expressed in all cell types which express 15B3, 15B3-1 and / or 15B3-2 and or a native variant thereof, in particular in neural ones Cells are influenced, for example by modulating cell-cell interactions.
Die nativen erfindungsgemäßen Proteine 15B3, 15B3-1 und 15B3- 2 sind erfindungsgemäß unter anderem Bestandteil des ras- MAPK-Signaltransduktionswegs, typischerweise als Modulator desselben, dessen Fehlsteuerung für eine Vielzahl von Erkrankungen ursächlich ist. Insofern finden sich die vorgenannten erfindungsgemäßen Proteine insbesondere als Komponenten bei den folgenden zellulären Prozessen wieder und hat zelluläre Funktionen bei: Signaltransduktion, Zelldifferenzierung, Wachstum, Plastizität, Regeneration. Entsprechend kann durch Infunktionalität eines erfindungsgemäßen Proteins, bspw. 1563,^ oder durch infunktionelle Expression oder durch Überexpression desselben ein pathophysiologischer Zustand ausgelöst werden, der mit einer Fehlsteuerung bspw. der Zelldifferen- zierung, des Zellwachstums, der Zellplastizität oder der Zellregeneration einhergeht. Je nach molekularem Mechanismus der pathophysiologischen Störung kann zu therapeutischen Zwecken die Gabe funktionellen erfindungsgemäßen Proteins oder zumindest eine höhere Expression desselben oder aber eine Inhibition des zellulär überexprimierten oder des exprimierten infunktionellen Proteins erwünscht sein. Ganz besonders bevorzugt ist die Verwendung von 15B3, 15B3-1 und/oder 15B3-2 im Zusammenhang mit deren Funktion beim neuronalen Zelltod, Excitation und Neurogenese. Insbesondere geht die Pathophysi- ologie der cerebralen Ischämie mit einer erhöhten Transkription von erfindungsgemäßem 15B3 einher, weswegen dessen Verwendung zur Behandlung dieser Pathophysiologie ganz besonders bevorzugt ist. Aus diesen erfindungsgemäßen Erkenntnissen ergibt sich die Verwendung erfindungsgemäßer Sequenzen (Nukleo- tid- und Aminosäuresequenzen) sowie entsprechender Derivate (bspw. Peptide, Oligos oder Antikörper) zur Herstellung eines Arzneimittels zur Behandlung von Tumorerkrankungen und neuro- logischen Erkrankungen, insbesondere ischämische Zustände (Schlaganfall), multipler Sklerose, neurodegenerative Erkrankungen, wie bspw. Morbus Parkinson, Amyotrophe Lateralsklerose, heredodegenerative Ataxien, Neuropathien, Morbus Huntington, Epilepsien und Morbus Alzheimer, viralen Infektionser- krankungen, bspw. Hepatitis-Infektionen (Anspruch 19) .The native proteins 15B3, 15B3-1 and 15B3-2 according to the invention are, according to the invention, part of the ras-MAPK signal transduction pathway, typically as a modulator thereof, the faulty control of which is the cause of a large number of diseases. In this respect, the aforementioned proteins according to the invention are found in particular as components in the following cellular processes and have cellular functions in: signal transduction, cell differentiation, growth, plasticity, regeneration. Accordingly, by Infunktionalität of a protein according to the invention, for example. 1563 ^ or by infunktionelle expression or overexpression thereof a pathophysiological state are triggered which, for example with a dysregulation. Of cell differentiation, cell growth, cell plasticity or cell regeneration associated. Depending on the molecular mechanism of the pathophysiological disorder, the administration of functional protein according to the invention or at least a higher expression of the same or an inhibition of the cellularly overexpressed or the expressed infunctional protein may be desired. The use of 15B3, 15B3-1 and / or 15B3-2 is very particularly preferred in connection with their function in neuronal cell death, excitation and neurogenesis. In particular, the pathophysiology of cerebral ischemia is associated with an increased transcription of 15B3 according to the invention, which is why its use for the treatment of this pathophysiology is very particularly preferred. From these findings according to the invention, the use of sequences according to the invention (nucleotide and amino acid sequences) and corresponding derivatives (for example peptides, oligos or antibodies) for the production of a medicament for the treatment of tumor diseases and neurological diseases, in particular ischemic conditions (stroke), results. , multiple sclerosis, neurodegenerative diseases, such as, for example, Parkinson's disease, amyotrophic lateral sclerosis, heredodegenerative ataxias, neuropathies, Huntington's disease, epilepsy and Alzheimer's disease, viral infectious diseases, for example hepatitis infections (claim 19).
Im Zusammenhang mit der therapeutischen Anwendung erfindungsgemäßer Sequenzen steht ein weiterer Gegenstand der vorliegenden Erfindung, nämlich die Verwendung einer erfindungsge- mäßen Nukleinsäuresequenz oder Proteinsequenz, insbesondere der Nukleotidsequenz oder Aminosäuresequenz von Protein 15B3, 15B3-1 oder 15B3-2 oder einer Variante - wie oben definiert - hiervon, insbesondere eines Fragments, zur Gentherapie bei Säugetieren, z.B. beim Menschen bzw. auch derartige genthera- peutische Verfahren. Gentherapie umfaßt dabei alle Therapieformen, die entweder erfindungsgemäße Sequenzen nach einem der Ansprüche 1 bis 4 in den Körper oder Teile davon, bspw. einzelne Gewebe, einbringen, oder die Expression von erfin- dungsgemäßer Sequenzen beeinflussen. Dazu können alle dem Fachmann geläufigen Modifikationen im Rahmen der Gentherapie verwendet werden, bspw. Oligonukleotide, z.B. antisense- oder Hybrid-RNA-DNA Oligonukleotide, mit beliebigen Modifikatio- nen, die erfindungsgemäße Sequenzen enthalten, benutzt werden. Ebenfalls können virale Konstrukte, enthaltend eine erfindungsgemäße Sequenzen (dies schließt alle Varianten, wie Fragmente, Isoformen, Allele, Derivate ein) benutzt werden. Auch entsprechende erfindungsgemäße nackte DNA-Sequenzen, kommen im Rahmen der Gentherapie in betracht. Ebenso können Nukleinsäurestücke mit enzymatischer Aktivität (z.B. Ribozy- me) für gentherapeutische Zwecke benutzt werden.Another object of the present invention is related to the therapeutic use of sequences according to the invention, namely the use of a nucleic acid sequence or protein sequence according to the invention, in particular the nucleotide sequence or amino acid sequence of protein 15B3, 15B3-1 or 15B3-2 or a variant - as defined above - Of these, in particular a fragment, for gene therapy in mammals, for example in humans, or also such gene therapy processes. Gene therapy encompasses all forms of therapy which either introduce sequences according to the invention according to one of claims 1 to 4 into the body or parts thereof, for example individual tissues, or the expression of invented affect sequences according to the invention. For this purpose, all modifications familiar to the person skilled in the art can be used in the context of gene therapy, for example oligonucleotides, for example antisense or hybrid RNA-DNA oligonucleotides, with any modifications which contain sequences according to the invention. Viral constructs containing a sequence according to the invention (this includes all variants, such as fragments, isoforms, alleles, derivatives) can also be used. Corresponding naked DNA sequences according to the invention are also considered in the context of gene therapy. Nucleic acid pieces with enzymatic activity (eg ribozymes) can also be used for gene therapy purposes.
Neben den therapeutischen Anwendungen kommen auch diagnosti- sehe Verwendungen erfindungsgemäßer Nukleinsäuren oder Poly- peptide, erfindungsgemäßer Proteinheteromere sowie davon abgeleiteter erfindungsgemäßer Reagenzien (Oligonukleotide, Antikörper, Peptide) in Betracht, bspw. zur Diagnose von menschlichen Erkrankungen oder von genetischen Prädispositio- nen, bspw. auch im Rahmen von Schwangerschaftsuntersuchungen. Bei diesen Erkrankungen oder Prädispositionen handelt es sich insbesondere um die oben im Zusammenhang mit therapeutischen Anwendungen genannten. Diese diagnostischen Verfahren können als in vivo, typischerweise jedoch ex vivo Verfahren ausges- taltet sein. Ex vivo wir eine typische Anwendung eines diagnostischen erfindungsgemäßen Verfahrens zum qualitativen und/αder quantitativen Nachweis einer erfindungsgemäßen Nukleinsäure in einer biologischen Probe dienen. Ein solches Verfahren umfasst vorzugsweise die folgenden Schritte: (a) Inkubation einer biologischen Probe mit einer bekannten Menge an erfindungsgemäßer Nukleinsäure oder einer bekannten Menge an Oligonukleotiden, die als Primer für eine Amplifikation der erfindungsgemäßen Nukleinsäure geeignet sind, (b) Nach- weis der erfindungsgemäßen Nukleinsäure durch spezifische Hybridisierung oder PCR-Amplifikation, (c) Vergleich der Menge an hybridisierender Nukleinsäure oder an durch PCR Ampli- fikation gewonnener Nukleinsäure mit einem Mengenstandard.In addition to therapeutic applications, diagnostic uses of nucleic acids or polypeptides according to the invention, protein heteromers according to the invention and reagents according to the invention (oligonucleotides, antibodies, peptides) derived therefrom are also possible, for example for the diagnosis of human diseases or of genetic predispositions, for example. also in the context of pregnancy examinations. These diseases or predispositions are, in particular, those mentioned above in connection with therapeutic applications. These diagnostic methods can be designed as in vivo, but typically ex vivo methods. Ex vivo will serve a typical application of a diagnostic method according to the invention for the qualitative and / or quantitative detection of a nucleic acid according to the invention in a biological sample. Such a method preferably comprises the following steps: (a) incubation of a biological sample with a known amount of nucleic acid according to the invention or a known amount of oligonucleotides which are suitable as primers for an amplification of the nucleic acid according to the invention, (b) after the nucleic acid according to the invention by specific hybridization or PCR amplification, (c) comparison of the amount of hybridizing nucleic acid or of nucleic acid obtained by PCR amplification with a quantity standard.
Außerdem betrifft die Erfindung ein Verfahren zum qualitativen und/oder quantitativen Nachweis eines erfindungsgemäßen Proteinheteromers oder eines erfindungsgemäßen Proteins in einer biologischen Probe, das folgende Schritte umfaßt: (a) Inkubation einer biologischen Probe mit einem Antikörper, der spezifisch gegen das Proteinheteromer oder gegen das erfindungsgemäße Protein/Polypeptid gerichtet ist, (b) Nachweis des Antikörper / Antigenkomplexes, (c) Vergleich der Mengen des Antikörper/Antigenkomplexes mit einem Mengenstandard. Als Standard wird üblicherweise eine biologische Probe aus einem gesunden Organismus entnommen. Insbesondere zu diagnostischen Zwecken kann hierbei die Eigenschaft des 15B3-, 15B3-1- und/oder 15B3-2-Gens benutzt werden, daß nach charakteristischen pathophysiologischen Stimuli, wie z.B. cerebralen I- schämien, die mRNA-Menge von 15B3, 15B3-1 und/oder 15B3-2 inThe invention also relates to a method for the qualitative and / or quantitative detection of a protein heteromer or a protein according to the invention in a biological sample, which comprises the following steps: (a) Incubation of a biological sample with an antibody which is specific to the protein heteromer or to the invention Protein / polypeptide is directed, (b) detection of the antibody / antigen complex, (c) comparison of the amounts of the antibody / antigen complex with a quantity standard. As a standard, a biological sample is usually taken from a healthy organism. In particular for diagnostic purposes, the property of the 15B3, 15B3-1 and / or 15B3-2 gene can be used here, that after characteristic pathophysiological stimuli, such as e.g. cerebral ischemia, the mRNA amount of 15B3, 15B3-1 and / or 15B3-2 in
Zellen sich verändert, z.B. sich erhöht. Auf diese Art kann erfindungsgemäß z.B. eine Verlaufsbeurteilung (Prognose) von mit Veränderungen von Expressionsraten erfindungsgemäßerCells change, e.g. increases. In this way, according to the invention, e.g. a course assessment (prognosis) of changes in expression rates according to the invention
Proteine einhergehender Krankheiten (wie z.B. des Schlagan- falls), die Beurteilung von Therapieerfolgen, die Graduierung einer Krankheit vorgenommen werden. Schließlich kann mit erfindungsgemäßen Verfahren ein Monitoring der Behandlung oben angegebener Erkrankungen durchgeführt werden. Proteins of associated diseases (such as stroke), the assessment of therapeutic success, the grading of a disease. Finally, the treatment according to the invention can be used to monitor the treatment of the diseases mentioned above.
Erfindungsgemäße Sequenzen können in Verfahren zur Bestimmung von Polymorphismen derselben z.B. bei Menschen verwendet werden. Auf diese ermittelte Polymorphismen erfindungsgemäßer Sequenzen unterfallen nicht nur der Offenbarung der vorlie- genden Erfindung, sondern können auch prognostische Marker für die Diagnose bzw. für die Diagnose einer Prädisposition von Erkrankungen, die im Zusammenhang mit einer durch infunktionale Expression erfindungsgemäßer Sequenzen, durch Expression infunktionaler erfindungsgemäßer Sequenzen und/oder oder deren Überexpression dienen. Darüber hinaus ist erlauben er- findunsgemäße Sequenzen die Erforschung humaner Erbkrankheiten, und zwar sowohl monogener als auch polygener Erkrankungen.Sequences according to the invention can be used in methods for determining polymorphisms thereof, for example in humans. The polymorphisms of sequences according to the invention which are determined in this way are not only subject to the disclosure of the present gene invention, but can also be used as a prognostic marker for the diagnosis or for the diagnosis of a predisposition to diseases which are associated with a sequence by means of infunctional expression according to the invention, by expression of sequence sequences according to the invention and / or their overexpression. In addition, sequences according to the invention permit research into human hereditary diseases, both monogenic and polygenic diseases.
Neben therapeutischen und/oder diagnostischen Verwendungszwecken im Bereich der Human- und/oder Tiermedizin kommt auch die Verwendung erfindungsgemäßer Nukleinsäuren oder Polypep- tide zum wissenschaftlichen Einsatz in Betracht. Insbesondere erlauben die erfindungsgemäßen Sequenzen auf dem Fachmann bekannte Weise, bspw. über cDNA-Bibliotheken verwandte Sequenzen bei ein- oder mehrzelligen Organismen zu identifizieren oder aber im humanen Genom verwandte Sequenzen zu lokalisieren. Die erfindungsgemäßen Nukleotidsequenzen, insbesondere die Sequenzen von 15B3, 15B3-1 und/oder 15B3-2, können also dazu verwendet werden, Gene für mRNAs, die für diese Nukleinsäuren oder deren funktionellen Äquivalente, Homologe oder Derivate kodieren, im bspw. murinen und im menschlichen Genom mit gängigen Methoden durch Homologiescreening zu isolieren, zu kartieren und mit Markern für humane Erbkrankheiten zu korrelieren. Dies ermöglicht die Identifizierung des Gens als Ursache für bestimmte Erbkrankheiten, was ihre Diagnose erheblich vereinfacht und neue Therapieansätze ermöglicht.In addition to therapeutic and / or diagnostic uses in the field of human and / or veterinary medicine, the use of nucleic acids or polypeptides according to the invention is also suitable for scientific use. In particular, the sequences according to the invention allow, in a manner known to the person skilled in the art, for example to identify sequences related to single or multi-cell organisms via cDNA libraries or to localize sequences related to the human genome. The nucleotide sequences according to the invention, in particular the sequences of 15B3, 15B3-1 and / or 15B3-2, can thus be used to generate genes for mRNAs which code for these nucleic acids or their functional equivalents, homologs or derivatives, for example in murine and in to isolate, map and correlate the human genome with common methods using homology screening and markers for human hereditary diseases. This enables the gene to be identified as the cause of certain hereditary diseases, which considerably simplifies their diagnosis and enables new therapeutic approaches.
Mit Hilfe erfindungsgemäßer Nukleinsäuren lassen sich damit Erbkrankheiten diagnostizieren, weswegen sie als Marker Verwendung finden. Bspw. wird sich eine Korrelation des chromo- somalen Locus des Protein 15B3 beim Menschen (Chromosom 9) zu bestimmten erblichen Erkrankungen ergeben, wobei hieraus ein erfindungsgemäßes Diagnoseverfahren für erbliche Erkrankungen erwachsen wird. Gleiches gilt für die erfindungsgemäßen Proteine.With the help of nucleic acids according to the invention, hereditary diseases can be diagnosed, which is why they are used as markers. For example. there will be a correlation of the chromosomal locus of protein 15B3 in humans (chromosome 9) result in certain hereditary diseases, from which a diagnostic method according to the invention for hereditary diseases arises. The same applies to the proteins according to the invention.
Insbesondere wird erfindungsgemäß für die wissenschaftliche Anwendung ein TestSystem offenbart, das auf erfindungsgemäßen Aminosäure- und/oder Nukleotid-Sequenzen beruht. In diesem Zusammenhang können die cDNA, die genomische DNA, die regula- torischen Elemente der erfindungsgemäßen Nukleinsäuresequenzen, als auch das Polypeptid, sowie Teilfragmente davon in rekombinanter oder nicht-rekombinanter Form zur Ausarbeitung eines Testsystems verwendet werden. Ein solches erfindungsgemäßes Testsystem ist insbesondere geeignet, die Aktivität des Promotors oder des Proteins in Anwesenheit der Testsubstanz zu messen. Bevorzugt handelt es sich hierbei um einfache Meßmethoden (kolorimetrische, luminometrische, auf Fluoreszenz beruhende oder radioaktive) , die die schnelle Messung einer Vielzahl von Testsubstanzen erlauben (Böhm, Klebe, Kubinyi, 1996, Wirkstoffdesign, Spektrum-Verlag, Heidelberg) . Die beschriebenen Testsysteme erlauben das Durchsuchen von chemischen Bibliotheken nach Substanzen, die hemmende oder aktivierende Wirkungen auf erfindungsgemäße Proteine haben. Die Identifizierung solcher Substanzen stellt den ersten Schritt auf dem Weg zur Identifizierung neuartiger Medikamente dar, die spezifisch auf die 15B3, 15B3-1 oder 15B3-2-assoziierte Signaltransduktion wirken. Insbesondere sind einfache Testsysteme denkbar, die die Aktivität der LLR-domäne vom 15B3, 15B3-1 oder 15B3-2 benutzen.In particular, a test system is disclosed according to the invention for scientific use, which is based on amino acid and / or nucleotide sequences according to the invention. In this context, the cDNA, the genomic DNA, the regulatory elements of the nucleic acid sequences according to the invention, and also the polypeptide, as well as partial fragments thereof, can be used in recombinant or non-recombinant form to develop a test system. Such a test system according to the invention is particularly suitable for measuring the activity of the promoter or of the protein in the presence of the test substance. These are preferably simple measurement methods (colorimetric, luminometric, based on fluorescence or radioactive) which allow the rapid measurement of a large number of test substances (Böhm, Klebe, Kubinyi, 1996, active ingredient design, Spektrum-Verlag, Heidelberg). The test systems described allow chemical libraries to be searched for substances which have inhibitory or activating effects on proteins according to the invention. The identification of such substances is the first step on the way to the identification of new drugs that specifically affect 15B3, 15B3-1 or 15B3-2-associated signal transduction. In particular, simple test systems are conceivable that use the activity of the LLR domain from 15B3, 15B3-1 or 15B3-2.
Eine Inhibition der biologischen Aktivität von erfindungsgemäßem Protein, insbesondere von Proteinen gemäß Figuren 9, 11 oder 13, typischerweise der biologischen Aktivität im Zusam- menhang mit der Apoptose, wie sie bspw. beim Schlaganfall, dem septischen Schock, GvHD, degenerativen Erkrankungen, insbesondere neurodegenerativen Erkrankungen, akuter Hepatitis oder anderen vorliegend offenbarten Indikationen erwünscht ist, kann auch dadurch erreicht werden, daß durch dem Fachmann geläufige Verfahren Oligonukleotide, die für einen Anti- sense-Strang der nativen Sequenzen der Proteine 15B3, 15B3-1 oder 15B3-2 oder eines anderen nativen Proteins mit einer Leucin reichen Domäne codieren, in die betroffenen Zellen einzuschleusen. Hierdurch wird die in den entsprechend transformierten Zellen die Translation der nativen mRNA von bspw. 15B3 oder 15B3-1 bzw. 15B3-2 blockiert, was im Ergebnis vorzugsweise die Überlebensfähigkeit der transfizierten Zelle steigert oder modulierend auf Zellwachstum, Zellplastizität, Zeilproliferation wirkt. Auch in diesem Fall kann das oben beschriebene Verfahren mit Hilfe rekombinanter Viren eingesetzt werden.An inhibition of the biological activity of protein according to the invention, in particular of proteins according to FIGS. 9, 11 or 13, typically the biological activity in combination. Connection with apoptosis, as is desired, for example, in the case of stroke, septic shock, GvHD, degenerative diseases, in particular neurodegenerative diseases, acute hepatitis or other indications disclosed herein, can also be achieved by the fact that oligonucleotides which code for an antisense strand of the native sequences of the proteins 15B3, 15B3-1 or 15B3-2 or another native protein with a domain rich in leucine, to be introduced into the affected cells. This blocks the translation of the native mRNA of, for example, 15B3 or 15B3-1 or 15B3-2 in the appropriately transformed cells, which, as a result, preferably increases the survivability of the transfected cell or has a modulating effect on cell growth, cell plasticity, cell proliferation. In this case too, the method described above can be used with the aid of recombinant viruses.
Die ggf. pathologisch gesteigerte Zellapoptose oder Zellpro- liferation bei Erkrankungen, die auf einer entsprechenden Fehlsteuerung erfindungsgemäßer Sequenzen beruhen (bspw. bei den vorgenannten Indikationen) , kann auch durch Ribozym- Methoden therapiert werden. Hierzu werden Ribozyme verwendet, die eine Ziel-mRNA schneiden können. Im vorliegenden Fall werden daher Ribozyme offenbart und stellen einen Gegenstand der vorliegenden Erfindung dar, die native 15B3 (oder 15B3-1 oder -15B3-2) -mRNA oder die mRNA anderer Leucin reiche Domänen enthaltender Proteine spalten können. Erfindungsgemäße Ribozyme müssen dabei mit der erfindungsgemäßen Ziel-mRNA inter- agieren können, bspw. über Basenpaarung, und anschließend die mRNA spalten, um die Translation von bspw. 15B3 zu blockieren. Die erfindungsgemäßen Ribozyme werden über geeignete Vektoren in die Zielzellen geschleust (insbesondere Plasmide, modifizierte Tierviren, insbesondere Retroviren) , wobei die Vektoren neben ggf. anderen Sequenzen eine cDNA -Sequenz für ein erfindungsgemäßes Ribozym aufweist) .The possibly pathologically increased cell apoptosis or cell proliferation in the case of diseases which are based on a corresponding incorrect control of sequences according to the invention (for example in the aforementioned indications) can also be treated by ribozyme methods. For this purpose ribozymes are used which can cut a target mRNA. In the present case, ribozymes are therefore disclosed and represent an object of the present invention which can cleave native 15B3 (or 15B3-1 or -15B3-2) mRNA or the mRNA of proteins containing domains rich in leucine. Ribozymes according to the invention must be able to interact with the target mRNA according to the invention, for example via base pairing, and then cleave the mRNA in order to block the translation of 15B3, for example. The ribozymes according to the invention are introduced into the target cells via suitable vectors (in particular plasmids, modified animal viruses, in particular retroviruses), the vectors, in addition to possibly other sequences, having a cDNA sequence for a ribozyme according to the invention).
Neben den vorgenannten Möglichkeiten zur Modulation der biologischen Funktion erfindungsgemäßer Genprodukte, insbesondere der Genprodukte gemäß Figuren 9, 11 oder 13, typischerweise der Modulation der Funktion erfindungsgemäßer Genprodukte bei der apoptotischen oder nekrotischen oder proliferativen Signaltransduktion ist dies auch mit Hilfe eines weiteren Gegenstands der vorliegenden Erfindung möglich. Eine erfindungsgemäße chemische Verbindung wird insbesondere die intrazelluläre Funktion der Proteine 15B3, 15B3-1 und/oder 15B3-2 modulieren, typischerweise inhibieren (Anspruch 20) oder auf der Ebene der zugrundeliegenden erfindungsgemäßen DNA- Sequenzen die biologische Funktion beeinflussen. Erfindungsgemäße Verbindungen werden typischerweise spezifisch an ein Protein mit einer Aminosäuresequenz gemäß Figuren 9, 11 oder 13 bzw. an eine Nukleinsäuresequenz gemäß Figuren 8, 10 oder 12 binden und dadurch eine pharmakologische, insbesondere neuroprotektive oder immunmodulierende, anti-apoptotische o- der anti-proliferative Wirkung, hervorrufen.In addition to the abovementioned possibilities for modulating the biological function of gene products according to the invention, in particular the gene products according to FIGS. 9, 11 or 13, typically modulating the function of gene products according to the invention in apoptotic or necrotic or proliferative signal transduction, this is also possible with the aid of a further object of the present invention , A chemical compound according to the invention will in particular modulate the intracellular function of the proteins 15B3, 15B3-1 and / or 15B3-2, typically inhibit (claim 20) or influence the biological function at the level of the underlying DNA sequences according to the invention. Compounds according to the invention will typically bind specifically to a protein with an amino acid sequence according to FIGS. 9, 11 or 13 or to a nucleic acid sequence according to FIGS. 8, 10 or 12 and thereby a pharmacological, in particular neuroprotective or immunomodulating, anti-apoptotic or anti-proliferative Effect.
Daher werden erfindungsgemäß chemische Verbindungen offen- bart, vorzugsweise eine organisch-chemische Verbindung, mit einem Molekulargewicht von <5000, insbesondere <3000, vor allem <1500, die typischerweise physiologisch gut verträglich ist und vorzugsweise die Blut-Hirn-Schranke passieren kann (Anspruch 21) . Ggf. wird sie Bestandteil einer Zusammenset- zung mit mindestens einem weiteren Wirkstoff sowie vorzugsweise Hilfs- und/oder Zusatzstoffen sein und als Arzneimittel eingesetzt werden können. Besonders bevorzugt wird das organische Molekül dann sein, wenn die Bindungskonstante für die Bindung an ein erfindungsgemäßes Protein, insbesondere an die Leucin reiche Domäne eines erfindungsgemäßen Proteins, mindestens 107 mol-1 beträgt. Die erfindungsgemäße Verbindung wird vorzugsweise so beschaffen sein, daß sie die Zellmembran passieren kann, sei es durch Diffusion oder über (intra)membranöse Transportproteine (Anspruch 22), ggf. nach entsprechender Modifikation, bspw. mit einer angekoppelten AS-Sequenz. Weiterhin bevorzugt sind jene Verbindungen, die die Interaktion von 15B3 oder 15B3-1 oder 15B3-2 mit Bin- dungspartnern, insbesondere für die Transduktion eines apop- totischen oder nekrotischen Signals, inhibiert oder verstärkt. Insbesondere besetzen derartige Verbindungen Positionen auf der Oberfläche erfindungsgemäßer Proteine oder rufen bei den erfindungsgemäßen Proteinen einen lokalen Konformati- onswechsel hervor, so daß die Bindung eines nativen Bindungspartners an ein erfindungsgemäßes Protein verhindert wird.Chemical compounds are therefore disclosed according to the invention, preferably an organic chemical compound with a molecular weight of <5000, in particular <3000, in particular <1500, which is typically well tolerated physiologically and can preferably pass the blood-brain barrier 21). Possibly. it will be part of a composition with at least one further active ingredient and preferably auxiliaries and / or additives and can be used as a medicament. The organic molecule will be particularly preferred if the binding constant for the Binding to a protein according to the invention, in particular to the leucine-rich domain of a protein according to the invention, is at least 10 7 mol -1 . The compound according to the invention will preferably be such that it can pass through the cell membrane, be it by diffusion or via (intra) membranous transport proteins (claim 22), if appropriate after appropriate modification, for example with a coupled AS sequence. Also preferred are those compounds which inhibit or enhance the interaction of 15B3 or 15B3-1 or 15B3-2 with binding partners, in particular for the transduction of an apoptotic or necrotic signal. In particular, such compounds occupy positions on the surface of proteins according to the invention or cause a local conformational change in the proteins according to the invention, so that the binding of a native binding partner to a protein according to the invention is prevented.
Über Strukturanalysen des erfindungsgemäßen Proteins lassen sich gezielt erfindungsgemäße Verbindungen finden, die eine spezifische Bindungsaffinität aufweisen (Rationales Drug Design (Böhm, Klebe, Kubinyi, 1996, Wirkstoffdesign, Spektrum- Verlag, Heidelberg) ) . Hier wird die Struktur oder eine Teilstruktur, Derivat, Allel, Isoform oder ein Teil einer solchen von einem der gemäß Figuren 9, 11 oder 13 dargestellten Pro- teinen über NMR- oder Röntgenkristallographie-Verfahren ermittelt oder, sofern eine solche hochaufgelöste Struktur nicht vorliegt, mit Hilfe von Strukturvorhersage-Algorithmen ein Strukturmodell eines erfindungsgemäßen Proteins erstellt, und diese (s) benutzt, um mit Unterstützung von Molecular Mo- delling Programmen Verbindungen zu identifizieren, für die sich eine hohe Affinität zum erfindungsgemäßen Protein vorhersagen läßt. Diese Substanzen werden dann synthetisiert und in geeigneten Testverfahren auf ihr Bindungsvermögen und ihre therapeutische Nutzbarkeit getestet.Structural analyzes of the protein according to the invention can be used to specifically find compounds according to the invention which have a specific binding affinity (rational drug design (Böhm, Klebe, Kubinyi, 1996, active ingredient design, Spektrum Verlag, Heidelberg)). Here, the structure or a partial structure, derivative, allele, isoform or part of one of one of the proteins shown in FIGS. 9, 11 or 13 is determined by means of NMR or X-ray crystallography methods or, if such a high-resolution structure is not available, with the help of structure prediction algorithms creates a structural model of a protein according to the invention, and uses this (s) to identify compounds with the support of molecular modeling programs for which a high affinity for the protein according to the invention can be predicted. These substances are then synthesized and tested in suitable test procedures for their binding capacity and their therapeutic usability.
In einer anderen bevorzugten Ausführungsform der vorliegenden Erfindung ist die erfindungsgemäße Verbindung ein Antikörper, vorzugsweise ein gegen ein erfindungsgemäßes Protein, bspw. 15B3, 15B3-1 oder 15B3-2, gerichteter Antikörper oder auch gegen die zugrundeliegenden mRNA gerichteter Antikörper, der ex vivo in retransplantierte Wirtszellen oder durch genthera- peutische in vivo Verfahren in Wirtszellen eingeschleust wird und dort als "intrabody" nicht sekretiert wird, sondern intrazellulär seine Wirkung entfalten kann. Durch derartige erfindungsgemäße "Intrabodies" können die Zellen vor einer fehlgeleiteten apoptotischen Reaktion, bspw. durch Überex- pression eines erfindungsgemäßen Proteins, geschützt werden. Eine derartige Vorgehensweise wird typischerweise für Zellen jener Gewebe in Betracht kommen, die beim Patienten ein pa- thophysiologisch übersteigertes apoptotisches Verhalten zeigen, also bspw. bei Zellen der Bauchspeicheldrüse, Keratino- zyten, Bindegewebszellen, Immunzellen, Neuronen oder Muskelzellen. Neben den Antikörper oder Intrabodies sind auch derartig mit erfindungsgemäßen "Intrabodies" gentherapeutisch modifizierte Zellen Bestandteil der vorliegenden Erfindung.In another preferred embodiment of the present invention, the compound according to the invention is an antibody, preferably an antibody directed against a protein according to the invention, for example 15B3, 15B3-1 or 15B3-2, or also against the underlying mRNA, which retransplanted ex vivo Host cells or by gene therapeutic in vivo methods is introduced into host cells and is not secreted there as "intrabody", but can exert its effect intracellularly. By means of such “intrabodies” according to the invention, the cells can be protected against a misdirected apoptotic reaction, for example by overexpression of a protein according to the invention. Such a procedure will typically be considered for cells of those tissues which show apoptotic behavior that is exaggerated in the patient's physiology, that is to say, for example, in cells of the pancreas, keratinocytes, connective tissue cells, immune cells, neurons or muscle cells. In addition to the antibodies or intrabodies, cells modified in this way with "intrabodies" according to the invention are also part of the present invention.
Eine erfindungsgemäße Verbindung mit der Funktion der Blockade der biologischen Funktion von nativem 15B3, 15B3-1 oder 15B3-2 gemäß Figuren 9, 11 oder 13 oder entsprechender nati- ver Allele oder nativer Spleißvarianten, bspw. der apoptotischen Funktion, kann als Arzneimittel Verwendung finden. Hierbei sind als Verbindungen alle vorgenannten Varianten eingeschlossen, also bspw. organisch-chemische Verbindungen, Antikörper, anti-sense Oligonukleotide, Ribozyme. Insbesondere ist eine erfindungsgemäße Verbindung (zur Herstellung ei- nes Arzneimittels) zur Behandlung von Erkrankungen, für die zumindest tw. eine pathologische hyperapoptotische oder hy- pernekrotische oder inflammatorische Reaktion kausal oder symptomatisch ist, geeignet. Damit kann ein erfindungsgemäßer Inhibitor (bspw. ein inhibitorisch wirkender Antikörper (insbesondere ein Intrabody) , ein Ribozym, anti-sense RNA, dominant-negative Mutanten oder eine der vorgenannten inhibitorischen organisch-chemischen Verbindungen) der zellulären Funktion eines erfindungsgemäßen nativen Proteins 15B3, 15B3-1 oder 15B3-2 oder seiner nativen Varianten, also bspw. der a- poptotischen, nekrotischen oder proliferativen Reaktion, als Arzneimittel und ganz besonders bei der Behandlung der folgenden Erkrankungen bzw. zur Herstellung eines Arzneimittels zur Behandlung der folgenden Erkrankungen eingesetzt werden: Tumorerkrankungen, Autoimmunerkrankungen, insbesondere Diabetes, Psoriasis, multiple Sklerose, rheumatoide Arthritis oder Asthma, virale Infektionserkrankungen (bspw. HIV) , degenerativen Erkrankungen, insbesondere neurodegenerativen Erkrankungen, bspw. Morbus Alzheimer oder Morbus Parkinson, Epilep- sie oder Muskeldystrophien, GvHD (bspw. Leber, Niere oder Herz), ischämischer Infarkt oder akuter Hepatitis. Die vorgenannten bspw. apoptoseinhibitorischen erfindungsgemäßen Substanzen können auch Bestandteil einer pharmazeutischen Zusammensetzung sein, die weitere pharmazeutische Träger- Hilfs- und/oder Zusatzstoffe enthalten kann, um derartige Zusammensetzungen für die therapeutische Verabreichung bspw. zu sta- bilis-ieren, die biologische Verfügbarkeit und/oder die Phar- makodynamik zu verbessern.A compound according to the invention with the function of blocking the biological function of native 15B3, 15B3-1 or 15B3-2 according to FIGS. 9, 11 or 13 or corresponding native alleles or native splice variants, for example the apoptotic function, can be used as a medicament , All the aforementioned variants are included as compounds here, for example, organic chemical compounds, antibodies, anti-sense oligonucleotides, ribozymes. In particular, a compound according to the invention (for producing a nes drug) for the treatment of diseases for which at least tw. a pathological hyperapoptotic or hyper necrotic or inflammatory reaction is causal or symptomatic. An inhibitor according to the invention (for example an antibody with an inhibitory effect (in particular an intrabody), a ribozyme, anti-sense RNA, dominant-negative mutants or one of the aforementioned inhibitory organic chemical compounds) can inhibit the cellular function of a native protein 15B3, 15B3 according to the invention -1 or 15B3-2 or its native variants, for example the a-poptotic, necrotic or proliferative reaction, are used as medicaments and very particularly in the treatment of the following diseases or in the manufacture of a medicament for the treatment of the following diseases: tumor diseases , Autoimmune diseases, especially diabetes, psoriasis, multiple sclerosis, rheumatoid arthritis or asthma, viral infectious diseases (e.g. HIV), degenerative diseases, especially neurodegenerative diseases, e.g. Alzheimer's or Parkinson's disease, epilepsy or muscular dystrophies, GvHD (e.g., liver disease) , Kidney or Heart), ischemic infarction or acute hepatitis. The above-mentioned, for example, apoptosis-inhibiting substances according to the invention can also be part of a pharmaceutical composition which can contain further pharmaceutical excipients and / or additives in order to stabilize such compositions for therapeutic administration, for example to stabilize the biological availability and / or to improve pharmacodynamics.
Ein weiterer Gegenstand der vorliegenden Erfindung sind Verfahren ("Screening-Methoden") zur Identifizierung von pharmazeutisch wirksamen Verbindungen, insbesondere mit inhibitorischen Eigenschaften in Hinblick auf die Auslösung oder Wei- terleitung von Signalen, die mit durch physiologisch auftretende, erfindungsgemäße Sequenzen ausgelöste apoptotischen Reaktionen, in Zusammenhang stehen (Anspruch 23) . Erfindungsgemäße Verfahren sehen vor, daß (a) Zellen mit einem Expres- sionsvektor nach Anspruch 5, insbesondere einem Expressionsvektor, der für das Polypeptid 15B3, 15B3-1 oder 15B3-2 codiert, und ggf. mindestens einem Expressionsvektor, der für mindestens ein Reporter-Gen codiert, transfiziert werden, und (b) ein zur Beobachtung der 15B3 vermittelten Funktion, bspw. der Apoptose, geeigneter Parameter, insbesondere die Aktivierung der Caspase-3, nach Zugabe einer Testverbindung im Vergleich zur Kontrolle ohne Zugabe einer Testverbindung für das gemäß (a) erhaltene Wirtszellsystem gemessen wird. Hierzu werden nach dem erfindungsgemäßen Verfahren vorzugsweise meh- rere parallele Versuche mit ansteigenden Konzentrationen der Testsubstanz angesetzt, um im Falle einer pharmazeutischen Wirksamkeit, bspw. die Apoptose inhibierenden Wirkung, der Testsubstanz deren ID50-Wert bestimmen zu können.The present invention furthermore relates to methods (“screening methods”) for identifying pharmaceutically active compounds, in particular with inhibitory properties with regard to triggering or Transmission of signals which are related to apoptotic reactions triggered by physiologically occurring sequences according to the invention (claim 23). Methods according to the invention provide that (a) cells with an expression vector according to claim 5, in particular an expression vector which codes for the polypeptide 15B3, 15B3-1 or 15B3-2, and optionally at least one expression vector which is for at least one reporter Gene are encoded, transfected, and (b) a parameter suitable for observing the 15B3-mediated function, for example apoptosis, in particular the activation of caspase-3 after addition of a test compound compared to the control without addition of a test compound for the (a) the host cell system obtained is measured. For this purpose, according to the method according to the invention, several parallel tests with increasing concentrations of the test substance are preferably carried out in order to be able to determine the ID 50 value of the test substance in the case of pharmaceutical activity, for example the apoptosis-inhibiting effect.
Insbesondere werden in einer bevorzugten Ausführungsform der vorliegenden Erfindung „Hochdurchsatz-Suchverfahren" („High- throughput screening assays", HTS) zur Identifikation von Inhibitoren von 15B3 offenbart. Ganz besonders bevorzugt ist dabei der Einsatz von (allen bekannten) Komponenten des ras- Signaltransduktionswegs im Rahmen des erfindungsgemäßen Verfahrens zur Identifikation von Inhibitoren, insbesondere zur Identifikation kleiner organischer Verbindungen an. Vorzugsweise bieten sich Systeme mit dem „Scintillation Proximity Assay" (SPA) an (Fa. Amersham Life Science, MAP kinase SPA (s. McDonald et al., 1999, Anal Biochem, 268, 318-29)). Vorgenannte Druckschrift, die einen Assayaufbau für die Raf/MEK/ERK-Kaskade, die Inhibitoren der ganzen Kaskade identifizieren kann, beschreibt, gehört vollinhaltlich zur Offen- barung der vorliegenden Erfindung. Die MAP-Kaskade wird hierbei in vitro rekonstituiert, mit den einzelnen Bestandteilen als GST-Fusionsproteinen (E. coli exprimiert) oder im Falle von cRAFl mit dem Baculovirussystem hergestellt (Anspruch 25) . Das erste Element der Kaskade (MAP-KKK) muss hierbei ständig gleichmäßig aktiviert sein, um verläßlich Inhibitoren testen zu können. Typischerweise wird dies wird durch Coexpression von src im Baculovirussystem erreicht. Dadurch wird eine ras-analoge Aktivierung von cRaf sichergestellt. Nach Transfektion von 15B3, 15B3-1, 15B3-2 oder anderen erfindungsgemäßen Varianten wird eine Modulierung der Kaskade hervorgerufen, die herangezogen wird, um in einem HTS durch Zugabe von zu testenden Substanzen eine Beeinflussung dieser Modulation messen zu können.In particular, in a preferred embodiment of the present invention, “high-throughput screening assays” (HTS) for identifying inhibitors of 15B3 are disclosed. The use of (all known) components of the ras signal transduction path in the context of the method according to the invention for identifying inhibitors, in particular for identifying small organic compounds, is very particularly preferred. Systems with the “Scintillation Proximity Assay” (SPA) are preferably available (Amersham Life Science, MAP kinase SPA (see McDonald et al., 1999, Anal Biochem, 268, 318-29)) describes an assay set-up for the Raf / MEK / ERK cascade that can identify inhibitors of the entire cascade. tion of the present invention. The MAP cascade is reconstituted in vitro, with the individual components as GST fusion proteins (E. coli expressed) or, in the case of cRAF1, with the baculovirus system (claim 25). The first element of the cascade (MAP-KKK) must be constantly activated in order to be able to reliably test inhibitors. Typically this is achieved by coexpression of src in the baculovirus system. This ensures a ras-analog activation of cRaf. After transfection of 15B3, 15B3-1, 15B3-2 or other variants according to the invention, a modulation of the cascade is brought about, which is used in order to be able to measure an influence on this modulation in an HTS by adding substances to be tested.
Nach Identifizierung hochaffiner, selektiver Substanzen nach vorgenanntem erfindungsgemäßen Verfahren werden diese auf ihre Verwendung als Medikamente gegen Epilepsie, Schlaganfall und andere neurologische, immunologische oder proliferative Erkrankungen (Tumorerkrankungen) getestet. Darüber hinaus können die Bindungsplätze der identifizierten und pharmakolo- gisch wirksamen Substanzen an die erfindungsgemäßen Genprodukte, insbesondere 15B3, 15B3-1 oder 15B3-2, mit Hilfe des two-hybrid Systems oder anderen Assays ermittelt werden, d.h., dass die für die Interaktion verantwortlichen Aminosäuren eingegrenzt werden (Anspruch 24) . Auf diese Weise können auch .Substanzen aufgefunden werden, mit denen die Interaktion zwischen erfindungsgemäßen Polypeptiden und etwaigen nativen intrazellulären Interaktionspartnern derselben beeinflusst, insbesondere inhibiert, werden können. Hierdurch wird erfindungsgemäß ein Verfahren zum Auffinden von Substanzen mit spezifischer Bindungsaffinität zum erfindungsgemäßen Protein offenbart. Aufgrund der pharmakologischen Bedeutung von 15B3, 15B3-1 und 15B3-2 bzw. deren nativen Varianten für zahlreiche Erkrankungen, bspw. in neurodegenerativen oder proliferativen Erkran- kungen, haben gemäß erfindungsgemäßem Verfahren identifizierte pharmazeutisch wirksame Substanzen ein weites Anwendungsspektrum. Neben der Inhibierung einer Interaktion mit einem oder mehreren anderen Molekülen, z.B. im Signaltransduktions- weg downstream befindlichen Proteinkinasen, oder Adaptoren kann insbesondere auch eine Beeinflussung der Transkription oder der Transkriptmenge von erfindungsgemäßen Proteinen in der Zelle Ursache pharmazeutischer Wirksamkeit sein. Beispielhaft wäre die Suppression der schnellen Hochregulation von Transkripten erfindungsgemäßer DNA-Sequenzen nach patho- logischen Prozessen durch erfindungsgemäße Verbindungen zu nennen, da 15B3, 15B3-1 bzw. 15B3-2 einer sehr raschen Regulation durch transkriptionelle Aktivierung unterliegen. Vorzugsweise ist ein Angriffsziel für eine pharmazeutisch wirksame Verbindung daher die Regulation der Transkription, bspw. durch spezifische Bindung der Substanzen an eine Regulatorregion (bspw. Promotor- oder Enhancer-Sequenzen) eines erfindungsgemäßen Genprodukts, Bindung an einen oder mehrere Transkriptionsfaktoren eines erfindungsgemäßen Genprodukts (mit dem Resultat einer Aktivierung oder Inhibierung des Transkriptionsfaktors) oder eine Regulation der Expression (Transkription oder Translation) eines solchen Transkriptionsfaktors selbst.After identification of high-affinity, selective substances by the aforementioned method according to the invention, these are tested for their use as medicaments for epilepsy, stroke and other neurological, immunological or proliferative diseases (tumor diseases). In addition, the binding sites of the identified and pharmacologically active substances to the gene products according to the invention, in particular 15B3, 15B3-1 or 15B3-2, can be determined with the aid of the two-hybrid system or other assays, ie that those responsible for the interaction Amino acids are limited (claim 24). In this way, substances can also be found with which the interaction between polypeptides according to the invention and any native intracellular interaction partners thereof can be influenced, in particular inhibited. In this way, a method for finding substances with specific binding affinity for the protein according to the invention is disclosed according to the invention. Due to the pharmacological importance of 15B3, 15B3-1 and 15B3-2 or their native variants for numerous diseases, for example in neurodegenerative or proliferative diseases, pharmaceutically active substances identified according to the method according to the invention have a wide range of uses. In addition to the inhibition of an interaction with one or more other molecules, for example protein kinases located downstream in the signal transduction pathway, or adapters, in particular an influence on the transcription or the transcript amount of proteins according to the invention in the cell can also be the cause of pharmaceutical effectiveness. The suppression of the rapid up-regulation of transcripts of DNA sequences according to the invention after pathological processes by compounds according to the invention should be mentioned as examples, since 15B3, 15B3-1 and 15B3-2 are subject to very rapid regulation through transcriptional activation. A target for a pharmaceutically active compound is therefore preferably the regulation of transcription, for example by specific binding of the substances to a regulator region (for example promoter or enhancer sequences) of a gene product according to the invention, binding to one or more transcription factors of a gene product according to the invention (with the result of an activation or inhibition of the transcription factor) or a regulation of the expression (transcription or translation) of such a transcription factor itself.
Neben der transkriptionellen Regulation, d.h. Regulation der mRNA-Menge eines erfindungsgemäßen Gens in der Zelle, kann eine erfindungsgemäße pharmazeutisch wirksame Verbindung auch in andere Kontrollprozesse der Zelle eingreifen, die bspw. die Expressionsrate eines erfindungsgemäßem Proteins beein- flussen können (z.B. Translation, Spleißvorgänge, native De- rivatisierung von erfindungsgemäßem Genprodukt, bspw. Phosphorylierungen, oder Regulation der Degradation von erfindungsgemäßem Genprodukt.In addition to the transcriptional regulation, ie regulation of the amount of mRNA of a gene according to the invention in the cell, a pharmaceutically active compound according to the invention can also intervene in other control processes of the cell which, for example, affect the expression rate of a protein according to the invention. can flow (eg translation, splicing processes, native derivatization of gene product according to the invention, for example phosphorylation, or regulation of the degradation of gene product according to the invention.
Ein weiterer Gegenstand der vorliegenden Erfindung betrifft Verfahren zur Identifizierung von zellulären Interaktionspartnern von erfindungsgemäßen Polypeptiden, insbesondere der Proteine 15B3, 15B3-1 oder 15B3-2 bzw. deren nativ auftreten- den Varianten (Isoformen, Allele, Spleißformen, Fragmente) (Anspruch 27) . Auf diese Weise können als Interaktionspartner Proteine identifiziert werden, die zum erfindungsgemäßen Protein spezifische Bindungsaffinitäten aufweisen, oder zur I- dentifizierung von Nukleinsäuren, die für Proteine kodieren, die zum erfindungsgemäßen Protein spezifische Bindungsaffinitäten aufweisen. Ein derartiges erfindungsgemäßes Verfahren bzw. die Verwendung erfindungsgemäßer Polypeptide, erfindungsgemäßer Nukleinsäuresequenzen und/oder erfindungsgemäßer Nukleinsäurekonstrukte zur Durchführung derartiger Verfahren wird vorzugsweise mit Hilfe einer „Yeast-two-hybrid"- Durchmusterung (y2h-„Screens") allein oder in Kombination mit anderen biochemischen Verfahren durchgeführt (Fields and Song, 1989, Nature, 340, 245-6) . Derartige Screens finden sich auch bei Van Aelst et al. (1993, Proc Natl Acad Sei U S A, 90, 6213-7) und Vojtek et al. (1993, Cell, 74, 205-14) beschrieben. Typischerweise können anstelle von Hefesystemen auch -Säugetiersysteme zur Durchführung eines erfindungsgemäßen Verfahrens verwendet werden, bspw. wie bei Luo et al. (1997, Biotechniques, 22, 350-2) beschrieben.The present invention further relates to methods for identifying cellular interaction partners of polypeptides according to the invention, in particular the proteins 15B3, 15B3-1 or 15B3-2 or their natively occurring variants (isoforms, alleles, splice forms, fragments) (claim 27) , In this way, proteins can be identified as interaction partners which have specific binding affinities for the protein according to the invention, or for the identification of nucleic acids which code for proteins which have specific binding affinities for the protein according to the invention. Such a method according to the invention or the use of polypeptides according to the invention, nucleic acid sequences according to the invention and / or nucleic acid constructs according to the invention for carrying out such methods is preferably carried out with the aid of a "yeast-two-hybrid" screening (y2h "screens") alone or in combination with other biochemical Procedures performed (Fields and Song, 1989, Nature, 340, 245-6). Such screens can also be found in Van Aelst et al. (1993, Proc Natl Acad Sei U S A, 90, 6213-7) and Vojtek et al. (1993, Cell, 74, 205-14). Typically, mammal systems can also be used to carry out a method according to the invention instead of yeast systems, for example as in Luo et al. (1997, Biotechniques, 22, 350-2).
Für einen y2h-Screen wird das offene Leseraster von 15B3, 15B3-1, 15B3-2 oder einer nativen Variante bspw. in einen sog. bait-Vektor „in-frame" mit der GAL4-Bindungsdomäne klo- niert (z.B. pGBTIO, Fa. Clontech) . Damit kann vorzugsweise eine sog. „prey-library" in einem Hefestamm nach gängigem Protokoll auf interagierende Proteine durchsucht werden. Vorzugsweise werden hierfür Kinase-negative Mutanten eingesetzt, da diese oft stabiler mit etwaigen Phosphorylierungstargets interagieren. Serin-/Threoninkinasen in einem Stoffwechselweg können durch Adaptermoleküle in räumliche Nähe gebracht werden, um spezifische Phosphorylierungen besser durchführen zu können (Chang et al., 1998, Science, 281 , 1860-3), (Yasuda et al., 1999, Mol Cell Biol, 19, 7245-54, (Whitmarsh and Davis, 1998, Trends Biochem Sei, 23, 481-5), Whitmarsh et al., 1998, Science, 281, 1671-4) . Es ist deshalb erfindungsgemäß auch möglich, über zwei Schritte im yeast-two-hybrid System auf Phosphorylierungstargets zu stoßen, indem zunächst ein Adapterprotein kloniert wird und mit diesem als „bait" das spezifische Zielprotein („target") ermittelt wird. Darüber hinaus können mit y2h-Systemen auch sog. Mapping-Experimente durchgeführt werden, um spezifische Interaktionsdomänen zu identifizieren.For a y2h screen, the open reading frame of 15B3, 15B3-1, 15B3-2 or a native variant is cloaked, for example, in a so-called bait vector "in-frame" with the GAL4 binding domain. (e.g. pGBTIO, from Clontech). A so-called “prey library” in a yeast strain can thus preferably be searched for interacting proteins according to the customary protocol. Kinase-negative mutants are preferably used for this, since these often interact more stably with any phosphorylation targets. Serine / threonine kinases in a metabolic pathway can be caused by Adapter molecules are brought into close proximity in order to be able to carry out specific phosphorylations better (Chang et al., 1998, Science, 281, 1860-3), (Yasuda et al., 1999, Mol Cell Biol, 19, 7245-54, ( Whitmarsh and Davis, 1998, Trends Biochem Sei, 23, 481-5), Whitmarsh et al., 1998, Science, 281, 1671-4) It is therefore also possible according to the invention, via two steps in the yeast-two-hybrid system to encounter phosphorylation targets by first cloning an adapter protein and using this to determine the specific target protein (“target”) as “bait”. In addition, so-called mapping experts can also be used with y2h systems imente to identify specific interaction domains.
Erfindungsgemäß können Interaktionspartner auch über Co-According to the invention, interaction partners can also
Immunpräzipitationen aus mit 15B3-, 15B3-1- oder 15B3-2-(bzw. deren native Varianten) Expressionsvektoren transfizierten Zellen zu benutzen, um daran bindende Proteine aufzureinigen, und über Proteinsequenzierungsmethoden (z.B. MALDI-TOF, ESI- tandem-MALDI) nachfolgend die dazugehörigen Gene zu identifizieren.Immunoprecipitations from cells transfected with 15B3-, 15B3-1- or 15B3-2- (or their native variants) are used to purify proteins that bind to them, and subsequently using protein sequencing methods (eg MALDI-TOF, ESI-tandem-MALDI) identify the associated genes.
Daher ist ein weiterer Gegenstand der vorliegenden Erfindung die Verwendung des two-hybrid Systems oder anderer biochemischer Verfahren zur Identifizierung von Interaktionsdomänen von 15B3, 15B3-1 oder 15B3-2 bzw. nativ auftretender Varianten derselben und die Verwendung dieser Interaktionsdomänen (Fragmente der nativen Sequenzen) zur pharmakotherapeutischen Intervention.Another object of the present invention is therefore the use of the two-hybrid system or other biochemical methods for the identification of interaction domains of 15B3, 15B3-1 or 15B3-2 or natively occurring variants thereof and the use of these interaction domains (Fragments of the native sequences) for pharmacotherapeutic intervention.
Die erfindungsgemäßen Polypeptide, insbesondere nativ auftretende Formen oder aber auch nicht-native, artifiziell erzeugte Varianten, deren biologische Funktion untersucht werden soll, können erfindungsgemäß in einem Apoptoseassay verwendet werden bzw. in einem Verfahren zur Untersuchung der Funk- tion und/oder Wirksamkeit erfindungsgemäßer Polypeptide bei Induktion, Transduktion oder Inhibition von Zelltodsignalen zum Einsatz kommen. Die Beteiligung von 15B3, 15B3-1, 15B3-2 oder vorgenannten Varianten an apoptotischen Kaskaden kann untersucht werden, indem Expressionskonstrukte mit 15B3, 15B3-1 oder 15B3-2 oder Varianten in eukaryontische Zellen transfiziert werden, und danach die Induktion von Apoptose untersucht werden kann. Dies kann z.B. durch Anfärbung mit Annexin geschehen (Fa. Röche Diagnostics) , durch Antikörper, die die aktive Form von Caspase-3 erkennen (Fa. New England Biolabs) , oder durch ELISAs, die DNA-Histon-Bruchstücke erkennen (cell-death elisa, Röche Diagnostics) . Diese Induktion von Apoptose ist ggf zelltyp-spezifisch, weswegen erfindungsgemäß vorzugsweise mehrere Zelllinien und primäre Zellen untersucht werden. Die Induktion von Apoptose kann ggf. auch Stimulus-spezifisch sein. Daher werden in einem erfindungsgemäßen Verfahren vorzugsweise mehrere Streß-Situationen zugrundegelegt, z.B. Hitzeschock, Hypoxiebedingungen, Cytokinbe- handlungen (z.B. 11-1, 11-6, TNF-alpha, H202-Behandlung) . Als typische Zelltypen für ein derartiges erfindungsgemäßes Ver- fahren kommen gebräuchliche Zellinien, z.B. Cos-Zellen, HEK- Zellen, PC12-Zellen, THP-1-Zellen, oder primäre Zellen, wie z.B. Neurone, Astrozyten, in Betracht, ebenso wie andere im- mortalisierte und primäre Zellinien nach Bedarf. Ein weiterer Gegenstand der vorliegenden Erfindung betrifft die Verwendung erfindungsgemäßer Nukleinsäuren, Nukleinsäure- konstrukte oder erfindungsgemäßer Genprodukte zur Durchfüh- rung eines Proliferationsassays. Bspw. kann die Beteiligung von 15B3, 15B3-1, 15B3-2 sowie nativer oder nicht nativer Varianten derselben beim Wachstum von Zellen, beim Durchlauf des Zellzyklus oder bei tumorigener Transformation untersucht werden, indem Expressionskonstrukte mit 15B3, 15B3-1 oder 15B3-2 oder entsprechender Varianten in eukaryontische Zellen transfiziert werden und danach bspw. die Induktion von Tumo- rigenizität untersucht wird, z.B. mit Hilfe eines Soft-Agar- Tests (Housey, et al., 1988, Adv Exp Med Biol, 234, 127-40). Als bevorzugte Zelltypen kommen gebräuchliche Linien, z.B. Cos-Zellen, HEK-Zellen, PC12-Zellen, THP-1-Zellen, und primäre Zellen, wie z.B. Neurone, Astrozyten, in Betracht, ebenso wie andere immortalisierte und primäre Zellinien nach Bedarf. Insbesondere kann mit Hilfe eines solchen erfindungsgemäßen Verfahrens die Funktion erfindungsgemäßer Genprodukte am ras- Signalübertragungsweg und die Wechselwirkung erfindungsgemäßer Genprodukte mit anderen Komponenten des ras- Signalübertragungswegs, insbesondere in Hinblick auf prolife- rative oder apoptotische Prozesse, untersucht werden.The polypeptides according to the invention, in particular natively occurring forms or also non-native, artificially generated variants whose biological function is to be investigated, can be used according to the invention in an apoptosis assay or in a method for examining the function and / or effectiveness of polypeptides according to the invention are used for induction, transduction or inhibition of cell death signals. The involvement of 15B3, 15B3-1, 15B3-2 or the aforementioned variants in apoptotic cascades can be examined by transfecting expression constructs with 15B3, 15B3-1 or 15B3-2 or variants in eukaryotic cells, and then examining the induction of apoptosis can. This can be done, for example, by staining with annexin (from Röche Diagnostics), through antibodies that recognize the active form of caspase-3 (from New England Biolabs), or by ELISAs that recognize DNA histone fragments (cell death elisa, Röche Diagnostics). This induction of apoptosis may be cell type-specific, which is why several cell lines and primary cells are preferably investigated according to the invention. The induction of apoptosis can also be stimulus-specific. Therefore, in a method according to the invention, preferably several stress situations are taken as a basis, for example heat shock, hypoxia conditions, cytokine treatments (for example 11-1, 11-6, TNF-alpha, H 2 O 2 treatment). Typical cell types for such a method according to the invention are customary cell lines, for example Cos cells, HEK cells, PC12 cells, THP-1 cells, or primary cells, such as neurons, astrocytes, as well as others in - Mortalized and primary cell lines as needed. The present invention further relates to the use of nucleic acids according to the invention, nucleic acid constructs or gene products according to the invention for carrying out a proliferation assay. For example. The involvement of 15B3, 15B3-1, 15B3-2 and native or non-native variants thereof in the growth of cells, in the passage of the cell cycle or in tumorigenic transformation can be examined by using expression constructs with 15B3, 15B3-1 or 15B3-2 or equivalent Variants are transfected into eukaryotic cells and then, for example, the induction of tumorigenicity is examined, for example with the aid of a soft agar test (Housey, et al., 1988, Adv Exp Med Biol, 234, 127-40). Common cell lines such as Cos cells, HEK cells, PC12 cells, THP-1 cells, and primary cells such as neurons, astrocytes, as well as other immortalized and primary cell lines as required, are considered as preferred cell types. In particular, with the aid of such a method according to the invention, the function of gene products according to the invention on the ras signal transmission path and the interaction of gene products according to the invention with other components of the ras signal transmission path can be investigated, in particular with regard to proliferative or apoptotic processes.
Ein weiterer Gegenstand der vorliegenden Erfindung ist die Verwendung einer DNA-Sequenz nach einem der Ansprüche 1 bis 4 oder eines Genprodukts nach einem der Ansprüche 8 bis 10 als Suizidgen/-protein für die in vivo oder ex vivo Transformation von Wirtszellen (Anspruch 29) . Insbesondere in Hinblick auf die biologische Funktion von erfindungsgemäßem Protein bei der Signaltransduktion von apoptotischen und/oder nekro- tischen Signalen kann auf diese Weise in Wirtszellen der Zelltod gezielt ausgelöst werden. Vorzugsweise wird hierbei die Verwendung einer erfindungsgemäßen DNA-Sequenz bzw. eines erfindungsgemäßen Proteins so ausgestaltet sein, daß das Suizidgen operabel mit einem Promotor verbunden ist, wobei die Transkription reprimiert ist und nur im Bedarfsfall aktiviert wird (Anspruch 30) . Insbesondere kann nach Transplantation von Patienten-Zellen ex vivo oder in vivo im Rahmen einer Gentherapie gezielt die transfizierte Zelle ausgeschaltet werden.Another object of the present invention is the use of a DNA sequence according to one of claims 1 to 4 or a gene product according to one of claims 8 to 10 as a suicide gene / protein for the in vivo or ex vivo transformation of host cells (claim 29). In particular with regard to the biological function of protein according to the invention in the signal transduction of apoptotic and / or necrotic signals, cell death can be triggered in a targeted manner in host cells. This is preferred the use of a DNA sequence according to the invention or a protein according to the invention be designed such that the suicide gene is operably linked to a promoter, the transcription being repressed and being activated only when necessary (claim 30). In particular, the transfected cell can be specifically switched off after transplantation of patient cells ex vivo or in vivo as part of a gene therapy.
Die vorliegende Erfindung wird durch die nachfolgenden Figuren näher erläutert.The present invention is explained in more detail by the following figures.
Figur 1 zeigt ein RMDD-Schema (restriction-mediated differen- tial display) . Eine Beschreibung ist in Ausführungsbeispiel 1 dargelegt.FIG. 1 shows an RMDD scheme (restriction-mediated differential display). A description is given in embodiment 1.
In Figur 2 ist das Ergebnis einer 15B3-LightCycler- Verifizierung dargestellt. Wie in Ausführungsbeispiel 1 erläutert, wurde 2 Stunden nach einem experimentellen Schlaganfall (MCAO, 90 min Okklusion) die Hochregulation von 15B3 gemessen, und zwar im kontralateralen und im ipsolateralen Be- reich. Die Expression (auf der y-Achse) ist normalisiert gegen die Expression des Standardgenprodukts Cyclophilin, also als relative Expression aufgetragen.The result of a 15B3 LightCycler verification is shown in FIG. As explained in exemplary embodiment 1, the upregulation of 15B3 was measured 2 hours after an experimental stroke (MCAO, 90 min occlusion), specifically in the contralateral and in the ipsolateral area. The expression (on the y axis) is normalized against the expression of the standard gene product cyclophilin, ie plotted as a relative expression.
Figur 3 gibt die 15B3-Gewebeexpression in verschiedenen Gewe- ben der Maus (Herz, Milz, Hirn, Lunge, Leber, Skelettmuskel, Niere, Hoden) oder nach verschiedenen Phasen der Embryonalentwicklung der Maus wieder. Das Ergebnis wurde mittels einer quantitativen PCR mit 15B3-Primern auf ein cDNA-Set der Fa. Clontech gewonnen, d.h. das vorgestellte Gen, 15B3, wird bei Mensch und Maus ubiqitär exprimiert. Aufgetragen ist die Expressionsrate gegen die einzelnen Gewebe.FIG. 3 shows the 15B3 tissue expression in various mouse tissues (heart, spleen, brain, lung, liver, skeletal muscle, kidney, testis) or after various phases of mouse embryonic development. The result was determined using a quantitative PCR with 15B3 primers on a cDNA set from Fa. Clontech obtained, ie the gene presented, 15B3, is ubiquitously expressed in humans and mice. The expression rate against the individual tissues is plotted.
Figur 4 stellt einen Homologie-Vergleich auf Proteinebene der Proteine 15B3, 15B3-1 und 15B3-2 mit Hilfe des Programms Clustal (Megalign, Lasergene, Madison, WI) dar. Dunkel unterlegt sind die jeweils identischen Aminosäuren. Insgesamt fällt der hohe Grad der Konservierung der Aminosäuren bei den drei Genprodukten auf. Die ober- oder unterhalb der Sequenz eingezeichneten Linien stellen jene Bereiche (insgesamt 4, und zwar ca. zwischen (1) AS 234 und 254, (2) 45 und 63, (3) 24 und 43 und (4) 184 und 203 (Numerierung nach Sequenz von 15B3-2) dar, die als Transmembrandomänen bezeichnet werden. Linien mit Pfeilen (insgesamt 10) markieren Bereiche, die Leucin-reiche Bereiche sind ("leucin-rich-repeats" (LRR) ) . Derartige LRR-Protein-Interaktionmodule sind in Proteinen mit diversen Funktionen bekannt. So enthält z.B. der humane und murine Toll-like Receptor 6 (TLR6) neben den Transmembrando- mänen eine extrazelluläre LRR-Domäne (Takeuchi et al., 1999).FIG. 4 shows a homology comparison at the protein level of the proteins 15B3, 15B3-1 and 15B3-2 with the aid of the Clustal program (Megalign, Lasergene, Madison, WI). The amino acids which are identical in each case are shaded with a dark background. Overall, the high degree of amino acid conservation in the three gene products is striking. The lines drawn above or below the sequence represent those areas (a total of 4, approximately between (1) AS 234 and 254, (2) 45 and 63, (3) 24 and 43 and (4) 184 and 203 ( Numbering according to the sequence of 15B3-2), which are referred to as transmembrane domains, lines with arrows (10 in total) mark regions which are leucine-rich regions ("leucine-rich repeats" (LRR)). Interaction modules are known in proteins with various functions, for example the human and murine toll-like receptor 6 (TLR6) contains an extracellular LRR domain in addition to the transmembrane domains (Takeuchi et al., 1999).
In Figur 5 ist ein phylogenetischer Stammbaum der Proteine mit "leucin-rich-repeat"-Domänen dargestellt. Zur Bestimmung des Stammbaums wurde das Programm Megalign eingesetzt. Die 15B3-Familie stellt dabei eine neue Untergruppe innerhalb der LRR-Domänen enthaltenden Proteine dar.FIG. 5 shows a phylogenetic family tree of the proteins with "leucine-rich-repeat" domains. The Megalign program was used to determine the family tree. The 15B3 family represents a new subset within the proteins containing LRR domains.
In Figur 6 ist eine Proteinstruktur-Vorhersage für erfindungsgemäßes Protein 15B3 mit Hilfe des Programms Protean (DNAStar, Lasergene, Madison, WI) abgebildet. Deutlich sichtbar sind die 4 putativen Transmembrandomänen. Figur 7: Ras-MAPK-Kaskade. Gezeigt sind putative Interaktionen von 15B3 innerhalb des Räs-Signaltransduktionsweges. 15B3 hat hier modulatorische Funktionen.FIG. 6 shows a protein structure prediction for protein 15B3 according to the invention with the aid of the Protean program (DNAStar, Lasergene, Madison, WI). The 4 putative transmembrane domains are clearly visible. Figure 7: Ras-MAPK cascade. Putative interactions of 15B3 within the Ras signal transduction path are shown. 15B3 has modulatory functions here.
Fig. 8 stellt die humane 15B3-Nukleinsäuresequenz (SEQ ID 1) dar.Figure 8 represents the human 15B3 nucleic acid sequence (SEQ ID 1).
Fig. 9 stellt die Aminosäuresequenz von humanem 15B3-Protein dar (SEQ ID 2) , und zwar durchlaufend vom N- zum C-Terminus.Figure 9 depicts the amino acid sequence of human 15B3 protein (SEQ ID 2) running from the N to the C terminus.
Fig. 10 stellt die humane Nukleotidsequenz von humanem 15B3-1 dar (Seq ID 3) .Figure 10 represents the human nucleotide sequence of human 15B3-1 (Seq ID 3).
Fig. 11 stelt die Aminosäuresequenz vom humanen Protein 15B3- 1 dar (Seq ID 4), und zwar durchlaufend vom N- zum C- Terminus.FIG. 11 shows the amino acid sequence of the human protein 15B3-1 (Seq ID 4), namely continuously from the N- to the C-terminus.
Fig. 12 stellt die Nukleotidsequenz von Protein 15B3-2 dar (Seq ID 5) .Figure 12 depicts the nucleotide sequence of protein 15B3-2 (Seq ID 5).
Fig. 13 stellt die Aminosäuresequenz von Protein 15B3-2 dar (Seq ID 6), und zwar durchlaufend vom N- zum C-Terminus.Figure 13 shows the amino acid sequence of protein 15B3-2 (Seq ID 6), running from the N to the C terminus.
Die vorliegende Erfindung wird durch die nachfolgenden Ausführungsbeispiele näher erläutert:The present invention is explained in more detail by the following exemplary embodiments:
Ausführungsbeispieleembodiments
Aus ührungsbeispiel 1 :From example 1:
Der Gegenstand der vorliegenden Erfindung, ein bislang nicht in der Vollänge definiertes und entsprechend bislang in sei- ner Funktion nicht charakterisiertes Protein, einschließlich der zugrundeliegenden DNA-Sequenz, wurde aufgrund einer dif- ferentiellen Regulation im zentralen Nervensystem kloniert. Die RNA-Produkte des Gens 15B3 wurden nach fokaler cerebraler Ischämie im Gehirn von Mäusen in der ischämischen Hemisphäre verstärkt exprimiert, und konnten durch ein differentielles Klonierungssystem (restriction-mediated differential display, RMDD) kloniert werden. Zur Herbeiführung von fokalen cerebralen Ischämien wurde hierbei das Fadenmodell benutzt (middle cerebral artery occlusion, MCAO) , ein valides Modell für die Schlaganfallerkrankung beim Menschen. Dieses experimentelle System wurde erfindungsgemäß eingesetzt, um Gene zu identifizieren, die neben anderen Funktionen auch von pathophysiolo- gischer Bedeutung für das Ereignisse nach einer cerebralen Ischämie sind. Im einzelnen wurde wie folgt experimentell verfahren, und molekulare targets für neue Medikamente bei neurodegenerativen Erkrankungen darstellen können.The subject matter of the present invention, a hitherto not defined in full length and accordingly hitherto in its A protein that was not characterized, including the underlying DNA sequence, was cloned due to differential regulation in the central nervous system. After focal cerebral ischemia in the brain of mice in the ischemic hemisphere, the RNA products of the 15B3 gene were increasingly expressed and could be cloned using a differential cloning system (restriction-mediated differential display, RMDD). The thread model (middle cerebral artery occlusion, MCAO) was used to induce focal cerebral ischemia, a valid model for stroke disease in humans. This experimental system was used according to the invention to identify genes which, in addition to other functions, are also of pathophysiological importance for events after cerebral ischemia. In detail, the experimental procedure was as follows, and molecular targets for new drugs in neurodegenerative diseases can be represented.
(a) Herbeiführen des Fadenmodells in Mäusen Zum Herbeiführen einer fokalen cerebralen Ischämie in c57/bl6 Mäusen wurden 3 Monate alte Mäuse benutzt. Nach Herbeiführen einer Inhalationsnarkose (70% N20, 30% 02, 0,8 - 1 % Ha- lothan) wurde ein 5-0 Prolenefaden (Fa. Ethicon) , der mit 0.1% Poly-L-Lysin beschichtet war, über die A. carotis exter- na in die A. carotis interna bis zum Abgang der A. cerebri media. vorgeschoben. Die richtige Position des Fadens wird durch einen Abfall des Laser-Doppler-Signals (Fa. Perimed) auf 10 - 20% des Ausgangssignals angezeigt. Nach Durchführung dieser Operation und gegebenenfalls Bestimmung zusätzlicher physiologischer Parameter (Blutdruck, Puls, Blutgase, Blutglukose) erwachen die Mäuse aus der Narkose. Nach bestimmten Okklusionszeiten werden die Mäuse wieder einer Narkose unter- zogen, und der Faden zurückgezogen. Dadurch findet eine Reperfusion des Gewebes statt. Nach bestimmten Reperfusionszei- ten werden die Mäuse durch Genickbruch getötet, und das Gehirn sofort präpariert und auf Trockeneis weggefroren.(a) Establishing the suture model in mice 3-month-old mice were used to induce focal cerebral ischemia in c57 / bl6 mice. After induction of inhalation (70% N20, 30% 02, 0.8-1% halothane), a 5-0 Prolenefaden (Ethicon), which was coated with 0.1% poly-L-lysine, was coated over the A External carotid artery into the internal carotid artery up to the exit of the cerebral media. advanced. The correct position of the thread is indicated by a drop in the laser Doppler signal (from Perimed) to 10-20% of the output signal. After performing this operation and, if necessary, determining additional physiological parameters (blood pressure, pulse, blood gases, blood glucose), the mice awake from anesthesia. After certain occlusion times, the mice are again anesthetized. pulled, and the thread pulled back. This causes tissue reperfusion. After certain reperfusion times, the mice are killed by a broken neck, and the brain is immediately prepared and frozen on dry ice.
Im vorliegenden Fall wurden keine Reperfusionen durchgeführt, sondern nur eine Okklusion von 90 min bzw. 24 h.In the present case, no reperfusions were carried out, but only an occlusion of 90 min or 24 h.
(b) Präparation von mRNA aus den Hirnen(b) Preparation of brain mRNA
Dazu wurde das mRNA-Präparationskit von der Fa. InvitrogenFor this purpose, the mRNA preparation kit from Invitrogen
(Fasttrack) verwendet. Es wird auf die beschriebene Standard- präparationsmethode verwiesen, die Bestandteil der Offenbarung der vorliegenden Patentanmeldung ist.(Fasttrack) used. Reference is made to the standard preparation method described, which is part of the disclosure of the present patent application.
(c) Durchführung des RMDD-Protokolls (siehe auch Abb. 1)(c) Implementation of the RMDD protocol (see also Fig. 1)
Die Durchführung erfolgte nach den in den Druckschriften EP 0 743 367 A2 und US 5,876,932 beschriebenen Verfahren, wobei abweichend hiervon 2 ug polyA-RNA für die Erststrangsynthese eingesetzt wurden. Nach Durchführung von Erststrang-, Zweit- Strangsynthese, Mbol-Restriktion wurde eine Ligation mit A- daptoren durchgeführt.' Zwei aufeinanderfolgende PCR- Reaktionen mit Subsets von Primerkombinationen schlössen sich an. Die PCR-Reaktionen wurden danach auf ein denaturierendes Gel geladen und auf eine Nylonmembran geblottet (Fa. GATC) . Die biotin-markierten Banden wurden mit Hilfe einer gebräuchlichen Streptavidin-Peroxidase-Reaktion visualisiert . Auf das Gel wurden jeweils PCR-Proben von der ischämischen und kontralateralen Hemisphäre zusammen aufgetragen (24 h MCAO rechts und links und 90 min MCAO rechts und links) . Banden unterschiedlicher Intensität in der rechten oder linken Hemisphäre wurden ausgeschnitten, und eine Reamplifikation des entsprechenden PCR-Produktes durchgeführt. Erhaltene amplifi- zierte Produkte wurden in den TOPO TA Vektor pcDNA 2.1 (Fa. Invitrogen) kloniert und mit T7 und M13rev-Primern sequenziert (ABI 3700 Kapillarelektrophoresesequencer) . Erhaltene Sequenzen werden mit der EMBL-Datenbank verglichen.The procedure was carried out according to the methods described in the publications EP 0 743 367 A2 and US 5,876,932, with the exception that 2 μg of polyA-RNA were used for the first strand synthesis. After first-strand, second-strand synthesis and Mbol restriction had been carried out, ligation with adapters was carried out. ' Two successive PCR reactions with subsets of primer combinations followed. The PCR reactions were then loaded onto a denaturing gel and blotted onto a nylon membrane (GATC). The biotin-labeled bands were visualized using a common streptavidin-peroxidase reaction. PCR samples from the ischemic and contralateral hemisphere were applied to the gel (24 h MCAO right and left and 90 min MCAO right and left). Bands of different intensities in the right or left hemisphere were cut out and the corresponding PCR product was reamplified. Amplified products obtained were in the TOPO TA vector pcDNA 2.1 (Fa. Invitrogen) cloned and sequenced with T7 and M13rev primers (ABI 3700 capillary electrophoresis sequencer). Sequences obtained are compared with the EMBL database.
(d) Klonierung von 15B3(d) Cloning 15B3
Nach Ausführung der vorbeschriebenen Verfahrensschritte wurde eine Sequenz identifiziert, die nach 90 min auf der ischämischen Seite hochreguliert schien. Eine "Lightcycler"-Analyse bestätigte eine schnelle Regulation nach MCAO (90 min MCAO und 2 h Reperfusion) (s. auch Fig. 2) .After carrying out the above-described process steps, a sequence was identified which appeared to be upregulated on the ischemic side after 90 min. A "light cycler" analysis confirmed rapid regulation after MCAO (90 min MCAO and 2 h reperfusion) (see also FIG. 2).
Diese Sequenz wies starke Homologie zu den Genen SUR-8 etc. auf. Das isolierte 3' -gelegene PCR-Fragment wurde benutzt, um eine Maushirnbank zu hybridisieren. Dabei fanden sich mehrere Klone, die Sequenzteile von dieser Sequenz (Seql) enthielten. Diese wurden mit ESTs aus der EMBL-Datenbank verglichen, und die Sequenzen Seql assembliert. Eine Maus Probe aus dem 5'- Bereich wurde benutzt, um eine humane fötale Hirnbank in lambdaZapII (Stratagene) zu durchsuchen ("screening") Eine Darstellung der Herstellung der benutzten humanen cDNA- Bibliothek ist in Fig. 1 dargestellt. Die erhaltenen humanen Klone wurden sequenziert und mit humanen ESTs aus der EMBL- Datenbank und der humanen mRNA des KIAA1437-Proteins AB037858 verglichen.This sequence showed strong homology to the SUR-8 etc. genes. The isolated 3 'PCR fragment was used to hybridize a mouse brain bank. Several clones were found that contained parts of the sequence from this sequence (Seql). These were compared with ESTs from the EMBL database, and the sequences Seql assembled. A mouse sample from the 5 'region was used to search a human fetal brain bank in lambdaZapII (Stratagene) ("screening"). An illustration of the preparation of the human cDNA library used is shown in FIG. 1. The human clones obtained were sequenced and compared with human ESTs from the EMBL database and the human mRNA of the KIAA1437 protein AB037858.
Die Nukleotid- und Proteinsequenzen der 15B3 verwandten Klone 15B3-.1 und 15B3-2 erbrachte eine Datenbanksuche der EMBL-, nrdb- und swissprot-Datenbanken (mit dem Programm BLAST 2, beschrieben bei Altschul et al. (1997, Nucleic Acids Res, 25, 3389-402) . Diese Druckschrift ist vollinhaltlich Bestandteil der vorliegenden Offenbarung. Mittels EMBL-Datenbank wurden weitere humane ESTs ermittelt, so dass für 15B3-2 der ORF er- mittelt werden konnte. Diese Sequenzen sind als Sequenzen Seq ID 5, 6 in den Figuren 10 bis 13 dargestellt.The nucleotide and protein sequences of the 15B3-related clones 15B3-.1 and 15B3-2 yielded a database search of the EMBL, nrdb and swissprot databases (with the BLAST 2 program, described by Altschul et al. (1997, Nucleic Acids Res, 25, 3389-402) This publication is fully part of the present disclosure, and further human ESTs were determined using the EMBL database, so that for 15B3-2 the ORF could be averaged. These sequences are shown as sequences Seq ID 5, 6 in FIGS. 10 to 13.
(e) Herstellung der humanen cDNA-Bibliothek Mit dem cDNA-Synthese Kit der Fa. Stratagene wurde ausgehend von 2 μg humaner fötaler Gehirn-mRNA (Fa. Clontech) und von 5 μg mRNA aus adultem Mausgehirn entsprechende cDNA- Bibliotheken hergestellt. Dabei wurde im wesentlichen entsprechend der Angaben des Herstellers verfahren. Zur Synthese der Erststrang cDNA wurde nach den Herstellerangaben ein oli- godT-Primer verwendet. Die klonierungs-kompatiblen (Eco- Rl/Xhol) doppel-strängigen cDNA-Fragmente wurden größenselek- tioniert (nach Herstellerangaben/ Fa. Stratagene) und in den Plasmidvektor pBluescript SKII (Stratagene) ligiert. Die Li- gation wurde durch Elektroporation in E.coli (DH10B, Gibco) transformiert und auf LB-Ampizillin Agar-Platten amplifi- ziert. Die Plasmid-DNA wurde mittels alkalischer Lyse und Ionenaustauscher-Chromatographie isoliert (QIAfilter-Kit, Fa. Qiagen) .(e) Production of the human cDNA library With the cDNA synthesis kit from Stratagene, corresponding cDNA libraries were produced starting from 2 μg human fetal brain mRNA (Clontech) and 5 μg mRNA from adult mouse brain. The procedure was essentially according to the manufacturer's instructions. According to the manufacturer's instructions, an oligotT primer was used to synthesize the first strand cDNA. The cloning-compatible (Eco-Rl / Xhol) double-stranded cDNA fragments were size-selected (according to the manufacturer's instructions / Stratagene) and ligated into the plasmid vector pBluescript SKII (Stratagene). The ligation was transformed by electroporation into E. coli (DH10B, Gibco) and amplified on LB ampicillin agar plates. The plasmid DNA was isolated by means of alkaline lysis and ion exchange chromatography (QIAfilter kit, from Qiagen).
Die Komplexität an Einzelklonen betrug für die fötale humane Gehirn-cDNA-Bank 4 Millionen. Von jeder cDNA-Bank wurden zufallsmäßig 24 Einzelklone nach Insertgrößen analysiert, die eine Größenverteilung von 800 bp bis zu 4,5 kb zeigten, die durchschnittliche Länge der cDNA-Inserts betrug für die humane Bajik ca. 1,2 kb.The complexity of single clones was 4 million for the fetal human brain cDNA library. From each cDNA library 24 individual clones were randomly analyzed for insert sizes, which showed a size distribution from 800 bp up to 4.5 kb, the average length of the cDNA inserts for the human Bajik was approximately 1.2 kb.
Ausführungsbeispiel 2 Durchführung eines Reportergen-AssaysEmbodiment 2 Carrying out a reporter gene assay
Die erhaltene Sequenz von 15B3 wurde dazu benutzt, Aufschlüsse über die Einordnung des Proteins in deren Funktion als E- lement von Signaltransduktionskaskaden, insbesondere der ras/MapK- Kaskade, zu gewinnen. Dazu wurde das offene Leseraster des Gens in einen gängigen Expressionsvektor (z.B. pCMV-tag, Fa. Stratagene) kloniert. Dieses Konstrukt wurde mit anderen Konstrukten zusammen in eukaryontische Zellen transfiziert werden (z.B. mit der Calciumphosphatmethode, siehe Ausubel et al., Current Protocols in Molecular Biology, New York, 1997) . Hierbei handelte es sich typischerweise um Reporterkonstrukte, z.B. ein Luciferase-Gen, unter Kontrolle eines Minimalpromotors mit mehreren SRE-Elementen, der Bindungssequenz für den ELK-Transkriptionsfaktor. Extrakte aus den Zellen wurden dann einer Messung im Luminometer (z.B. Fa. Bertold) unterzogen. Eine Erhöhung des Luciferase-Wertes wurde als Beeinflussung des Signaltransduktionsweges gewertet, der in der Aktivierung eines bestimmten Transkriptionsfaktors (ELK) mündet. Kombinationen mit anderen Expressionskonstruk- ten (bspw. für andere Gene, z.B. MAP-Kinasen) konnten Aufschlüsse über die genaue Position von 15B3 in Signalkaskaden geben.The sequence obtained from 15B3 was used to obtain information about the classification of the protein in its function as E- element of signal transduction cascades, in particular the ras / MapK cascade. For this purpose, the open reading frame of the gene was cloned into a common expression vector (eg pCMV-tag, from Stratagene). This construct was transfected together with other constructs into eukaryotic cells (eg using the calcium phosphate method, see Ausubel et al., Current Protocols in Molecular Biology, New York, 1997). These were typically reporter constructs, for example a luciferase gene, under the control of a minimal promoter with several SRE elements, the binding sequence for the ELK transcription factor. Extracts from the cells were then subjected to a measurement in a luminometer (for example from Bertold). An increase in the luciferase value was assessed as influencing the signal transduction pathway, which results in the activation of a specific transcription factor (ELK). Combinations with other expression constructs (eg for other genes, eg MAP kinases) could provide information about the exact position of 15B3 in signal cascades.
Ggf. wurden diese Reporter-Assays auch mit anderen Systemen durchgeführt, z.B. lacZ oder Chloramphenicol-Transferase (CAT-Assays) , wobei dem Assay das gleiche Prinzip, wie oben beschrieben, zugrundelag.Possibly. these reporter assays have also been carried out with other systems, e.g. lacZ or chloramphenicol transferase (CAT assays), the assay being based on the same principle as described above.
Ausführungsbeispiel 3 :Example 3:
Untersuchungen zur Gewebeexpression von 15B3Tissue expression studies of 15B3
Zur Untersuchung der Gewebeexpression von 15B3 wurde eine quantitative PCR mit Hilfe des LightCyclers (Röche Diagnostics, Mannheim) durchgeführt. Es wurden cDNA-Proben von 8 murinen Geweben verwendet, die bereits mengenstandardisiert auf 4 verschiedene housekeeping Gene sind (Fa. Clontech) . Als Kontrolle verwendet wurde Plasmid xxl5B3, das ein amplifi- ziertes Fragment der murinen 15B3-CDNA enthielt. Als PCR- Programm wurde ein Standard-Protokoll (annealling-temp 60 °C) gewählt. Es fand in allen Gewebeproben Amplifikation eines Produktes mit dem Schmelzpunkt 89°C statt, dieser deckte sich mit der Kontroll-PCR. Die Amplifikation wird im LightCycler ab Zyklus 25 sichtbar. Folgende Primer wurden verwendet: seq_15B3.1 GAGCTGCCAAGAAAGGGGGAGACT (in Exon 2) seq_15B3.2 CAGAAAACACAAATGGACGGAGAG (in Exon 2)To examine the tissue expression of 15B3, a quantitative PCR was carried out using the LightCycler (Röche Diagnostics, Mannheim). CDNA samples from 8 murine tissues were used, which were already standardized in terms of quantity on 4 different housekeeping genes (from Clontech). Plasmid xxl5B3, which contained an amplified fragment of the murine 15B3 CDNA, was used as a control. A standard protocol (annealling temp 60 ° C) was chosen as the PCR program. In all tissue samples there was amplification of a product with a melting point of 89 ° C, which coincided with the control PCR. The amplification is visible in the LightCycler from cycle 25. The following primers were used: seq_15B3.1 GAGCTGCCAAGAAAGGGGGAGACT (in exon 2) seq_15B3.2 CAGAAAACACAAATGGACGGAGAG (in exon 2)
Die quantitative Analyse (Fig. 3) zeigt eine relativ ubiqui- täre Gewebeverteilung mit Schwerpunkten in Herz, Lunge, Leber und Gehirn. 15B3 Expression ist ebenfalls in Testis, Muskel, Niere und Milz sowie während der Embryonalentwicklung nachweisbar.The quantitative analysis (FIG. 3) shows a relatively ubiquitous tissue distribution with a focus on the heart, lungs, liver and brain. 15B3 expression is also detectable in the testis, muscle, kidney and spleen as well as during embryonic development.
Durch eine Homologiesuche (BLAST) mit der humanen 15B3 Sequenz konnten humane ESTs (expressed sequence tags) aus dem 3' untranslatierten Bereich des Gens identifiziert werden. Dabei finden sich Klone aus Aorta, Knochen, Gehirn, CNS, Co- lon, Auge, Geschlechtsorganen, Herz, Niere, Lunge, Lymphsystem, Pankreas, Prostata, Magen, Testis, Uterus, Embryo, Brustgewebe, Muskel, nervous Tumor, Ovar, Uterus, die auf ei- ne breite Gewebeverteilung schließen lassen. Dies deckt sich mit den erhaltenen quantitativen PCR-Daten.A homology search (BLAST) with the human 15B3 sequence identified human ESTs (expressed sequence tags) from the 3 'untranslated region of the gene. There are clones from aorta, bone, brain, CNS, colon, eye, genital organs, heart, kidney, lungs, lymphatic system, pancreas, prostate, stomach, testis, uterus, embryo, breast tissue, muscle, nervous tumor, ovary, Uterus, which suggest a broad distribution of tissue. This coincides with the quantitative PCR data obtained.
Ausführungsbeispiel :Design example:
Untersuchungen zur und Charakterisierung der Proteinsequenz von 15B3 Für die humane Proteinsequenz ergibt sich ein offenes Leseraster, das für ein Protein mit 811 Aminosäuren kodiert (94.2 kD Molekulargewicht; isoelektr. Punkt bei pH 7.9).Studies on and characterization of the protein sequence of 15B3 There is an open reading frame for the human protein sequence, which codes for a protein with 811 amino acids (94.2 kD molecular weight; isoelectric point at pH 7.9).
(a) Bestimmung von Protein-Domänen(a) Determination of protein domains
Zur Ermittlung von Transmembran-Domänen wurde das Computerprogramm T pred verwendet Es sagt mit hohen Wahrscheinlichkeiten 4 Transmembran-Domänen für den N-terminus des Proteins 15B3 voraus (s. Fig. 6) . Ein "Score" über 500 weist auf die Signifikanz potentieller Transmembransegmente hin, für das Protein 15B3 ist der "Score" für alle vier potentiellen Domänen sehr hoch (1361, 1965, 757, 2592) .The computer program T pred was used to determine transmembrane domains. It predicts with high probability 4 transmembrane domains for the N-terminus of protein 15B3 (see FIG. 6). A "score" above 500 indicates the significance of potential transmembrane segments, for the protein 15B3 the "score" for all four potential domains is very high (1361, 1965, 757, 2592).
4 strong transmembrane helices, total score : 6675 from to length score orientation 1 26 45 (20) 1361 i-o4 strong transmembrane helices, total score: 6675 from to length score orientation 1 26 45 (20) 1361 i-o
2 124 144 (21) 1965 o-i2 124 144 (21) 1965 o-i
3 267 287 (21) 757 i-o3,267,287 (21) 757 OK
4 320 341 (22) 2592 o-i4,320,341 (22) 2,592 o-i
Weiterhin wurde Kyte-Doolittle Hydrophobizitätsplot erstellt. Das Ergebnis dieser Auftragung indiziert im N-Terminus vier hydrophobe Bereiche an (Index > -3), die mit der oben angegebenen Sequenz übereinstimmen (AS: 26-45, 124-144, 267-287, 320-341) , dies unterstützt die Hypothese, daß es sich hierbei um Transmembranregionen handelt.Furthermore, Kyte-Doolittle hydrophobicity plot was created. The result of this plot indicates four hydrophobic areas (index> -3) in the N-terminus, which correspond to the sequence given above (AS: 26-45, 124-144, 267-287, 320-341), this supports the Hypothesis that these are transmembrane regions.
Mit Hilfe des Kyte-Doolittle Hydrophobizitätsplots wurde erfindungsgemäß festgestellt, daß eine hydrophile Region für AS 350-435 vorliegt, während der anschließende C-Terminus einen grösseren hydrophoben Bereich aufweist. Unter Verwendung des Programm HMMPFAM wurde weiterhin erfindungsgemäß bestimmt, daß diese Region (AS 461-803) einen charakteristischen Leu- cin-Rich-Repeats-Bereich (LRR) darstellt. Desweiteren wurde das Programm PSORTII zur näheren Charakterisierung der erfindungsgemäßen Sequenz eingesetzt. Derart wurden in der Proteinsequenz zwei Signalsequenzen identifi- ziert. Die letzten 4 Aminosäuren des 15B3-C-Terminus, DKEQ, weisen signifikante Ähnlichkeit zu einem "ER Membrane Retention Signal" ähnlichen Motif, KKXX, auf. Eine weitere Sequenz an Position 695 as des Proteins 15B3 wurde ggf. als peroxiso- males Targeting-Signal (KIEKIPTQL) identifiziert. Die Ergeb- nisse der vorgenannten Experimente erlauben den Schluß, daß ein erfindungsgemäßes Protein im Endoplasmatischen Retikulum lokalisiert ist.Using the Kyte-Doolittle hydrophobicity plot, it was found according to the invention that there is a hydrophilic region for AS 350-435, while the subsequent C-terminus has a larger hydrophobic region. Using the program HMMPFAM, it was further determined according to the invention that this region (AS 461-803) represents a characteristic leucine-rich repeats area (LRR). Furthermore, the PSORTII program was used to characterize the sequence according to the invention in more detail. In this way, two signal sequences were identified in the protein sequence. The last 4 amino acids of the 15B3-C terminus, DKEQ, show significant similarity to a Motif, KKXX, similar to "ER Membrane Retention Signal". Another sequence at position 695 as of protein 15B3 was identified as a peroxisomal targeting signal (KIEKIPTQL). The results of the aforementioned experiments allow the conclusion that a protein according to the invention is localized in the endoplasmic reticulum.
(b) Sequenzvergleiche des 15B3 Proteins(b) Sequence comparisons of the 15B3 protein
Es wurden Sequenzvergleiche der 15B3 Sequenz mit EMBL, nrdb und swissprot-Datenbanken durchgeführt. Hierbei ergaben sich Übereinstimmungen mit folgenden humanen Sequenzen: (i) BAA92675 und (ii) BAA91549.Sequence comparisons of the 15B3 sequence were carried out with EMBL, nrdb and swissprot databases. This resulted in agreement with the following human sequences: (i) BAA92675 and (ii) BAA91549.
Der Protein-Sequenzvergleich zwischen dem erfindungsgemäßen Protein 15B3 und BAA92675 ergab eine 100% Identität auf Proteinebene. Die Autoren geben für den Klon BAA92675 an, dass das Startcodon nicht identifiziert wurde. Durch die Ermitt- lung des offenen Leserahmens der 4,7 kb langen Nukleotidsequenz von 15B3 konnten wir jedoch feststellen, das es sich bei der zweiten AS der annotierten BAA92675-Proteinsequenz sehr wahrscheinlich um das Startcodon von 15B3 handelt. Es konnten keine längeren ORF identifiziert werden, die mit dem translatierten Protein übereinstimmen. Auch konnten keine weiteren ORFs mit anderem Leseraster aufgezeigt werden. Die Nukleotidsequenz des Klons BAA92675 zeigt einen Nukleoti- daustausch in Position 2748bp (C statt T) und in Position 3891 bp (A statt G) . Die Fehler in der Nukleotidsequenz bewirken jedoch keinen Aminosäureaustausch auf Proteinebene.The protein sequence comparison between the protein 15B3 and BAA92675 according to the invention showed 100% identity at the protein level. For the clone BAA92675, the authors state that the start codon was not identified. However, by determining the open reading frame of the 4.7 kb nucleotide sequence of 15B3, we were able to determine that the second AS of the annotated BAA92675 protein sequence is very likely the start codon of 15B3. No longer ORF could be identified that matched the translated protein. Also, no other ORFs with a different reading frame could be shown. The nucleotide sequence of clone BAA92675 shows a nucleotide exchange in position 2748bp (C instead of T) and in position 3891 bp (A instead of G). However, the errors in the nucleotide sequence do not result in an amino acid exchange at the protein level.
(c) Klassifizierung von 15B3, homologe Proteine(c) Classification of 15B3, homologous proteins
Eine Datenbanksuche (BLAST 2 (Altschul, et al. , 1997,Nucleic Acids Res, 25, 3389-402) ) der EMBL, nrdb und swissprot- Datenbanken erbrachte Verwandte dieses Proteins.A database search (BLAST 2 (Altschul, et al., 1997, Nucleic Acids Res, 25, 3389-402)) of the EMBL, nrdb and swissprot databases revealed relatives of this protein.
Die stärksten Homologien zeigt 15B3 zu zwei annotierten Proteinsequenzen:15B3 shows the strongest homologies to two annotated protein sequences:
-58.8% Identität auf 670 AS zu BAA9131 -58.8% Identität auf 803 AS zu Q92627. Konserviert ist hierbei die LRR-Bindungsdomäne im C-Terminus sowie die vier potentiellen Transmembrandomänen im N-Terminus (Fig. 4).-58.8% identity on 670 AS to BAA9131 -58.8% identity on 803 AS to Q92627. The LRR binding domain in the C-terminus and the four potential transmembrane domains in the N-terminus are preserved (FIG. 4).
Die annotierte Proteinsequenz von 476 AS des Klons Q92627 zeigte keinen vollständigen ORF. Durch Translation der ermit- telten 5 'liegenden genomischen Sequenzen konnte das Startcodon bestimmt sowie der ORF auf 804 AS vervollständigt werden. Auch in den N-terminalen Sequenzen besteht eine gewisse Ho- molgie zu 15B3. Die Proteinsequenz weist ebenfalls vier potentielle konservierte Transmembran-segmente, 10 Leucin- reiche repeats sowie ein "ER Membrane Retention Signal" ähnlichen Motif auf.The annotated protein sequence of 476 AA from clone Q92627 did not show a complete ORF. The start codon was determined and the ORF on 804 AS was completed by translating the 5 'genomic sequences found. There is also a certain homology to 15B3 in the N-terminal sequences. The protein sequence also has four potential conserved transmembrane segments, 10 leucine-rich repeats and a motif similar to "ER membrane retention signal".
Die Proteinsequenz des Klon BAA9131 beginnt zwar mit einem Startcodon, jedoch besteht aufgrund der Sequenzvergleiche zu 15B3 und Q92627 die Annahme, daß der ORF im N-Terminus unvollständig ist. Die Proteinsequenz zeigt starke Konservierung zu den potentiellen Transmembran-segmenten (3 und 4, Fig. 6) und weist ebenfalls 10 Leucin-reiche repeats auf. Aufgrund der Konservierung auf Proteinebene, besonders innerhalb der potentiellen Transmembrandomänen und der LRR-Domänen kann davon ausgegangen werden, dass 15B3 mit BAA9131 und Q92627 einer gemeinsamen Genfamilie angehört und eine neue Subgruppe definiert (Figur 5) . Aus diesem Grund wurde BAA9131 in 15B3-1 und Q92627 in 15B3-2 umbenannt.The protein sequence of clone BAA9131 starts with a start codon, however, based on the sequence comparisons to 15B3 and Q92627, it is assumed that the ORF in the N-terminus is incomplete. The protein sequence shows strong conservation to the potential transmembrane segments (3 and 4, Fig. 6) and also has 10 leucine-rich repeats. Due to the conservation at the protein level, especially within the potential transmembrane domains and the LRR domains, it can be assumed that 15B3 with BAA9131 and Q92627 belong to a common gene family and define a new subgroup (FIG. 5). For this reason, BAA9131 was renamed to 15B3-1 and Q92627 to 15B3-2.
Mit Hilfe einer Datenbanksuche konnten außerdem Sequenzähnlichkeiten zu Proteinen aus anderen Spezies oder Organismen wie Mensch, Maus, Ratte, Drosophila melanogaster, C.elegans, S. cerevisiae, E. coli: nachgewiesen werden:With the help of a database search, sequence similarities to proteins from other species or organisms such as humans, mice, rats, Drosophila melanogaster, C.elegans, S. cerevisiae, E. coli:
(1) H. sapiens: PID:gl504042 - similar to yeast adenylate cy- clase (69 % / 148 aa)(1) H. sapiens: PID: gl504042 - similar to yeast adenylate cyclase (69% / 148 aa)
(2) M. musculus: PID:g3252981 - Ras-binding protein SUR-8 (35 % / 141 aa)(2) M. muscle: PID: g3252981 - Ras-binding protein SUR-8 (35% / 141 aa)
(3) R. norvegicus: PID:gl657758 - densin-180 (30 % / 112 aa)(3) R. norvegicus: PID: gl657758 - densin-180 (30% / 112 aa)
(4) D. melanogaster: PIR:S60461 - S60461 gene flightless-I protein - fruit fly (34 % / 141 )(4) D. melanogaster: PIR: S60461 - S60461 gene flightless-I protein - fruit fly (34% / 141)
(5) C. elegans: PID:g3168891 - contains similarity to re- peated leucine-rich (29 % / 145 aa)(5) C. elegans: PID: g3168891 - contains similarity to repeated leucine-rich (29% / 145 aa)
(6) S. cerevisiae: PID:gl006714 - ORF YJL005w (34 % / 114 aa)(6) S. cerevisiae: PID: gl006714 - ORF YJL005w (34% / 114 aa)
(7) E. coli: PID:gl788752 - cell division protein involved in FtsZ ring (34 % / 123 aa)(7) E. coli: PID: gl788752 - cell division protein involved in FtsZ ring (34% / 123 aa)
Weitere Homolgien zu bekannten Proteinen des Menschen und der Maus hat erfindungsgemäßes 15B3 zu Sur-8 (Li W., et al., Genes Dev. 2000 Apr 15; 14 (8) : 895-900) , Soc-2 (Proc. Natl.15B3 of Sur-8 (Li W., et al., Genes Dev. 2000 Apr 15; 14 (8): 895-900), Soc-2 (Proc. Natl ,
Acad. SciUSA Vol. 95, pp. 6903-6908, June 1998), Rsu-1 (Tsuda et al., Genomics. 1993 Nov; 18(2): 461-2), Densin-180 (Apper- son et al., J Neurosci. 1996 Nov 1,16(21) : 6839-52), ScribbleAcad. SciUSA Vol. 95, pp. 6903-6908, June 1998), Rsu-1 (Tsuda et al., Genomics. 1993 Nov; 18 (2): 461-2), Densin-180 (Apperson et al., J Neurosci. 1996 Nov 1, 16 (21): 6839-52), Scribble
(Nature 403 (6770) : 676-680 (2000) und der Adenylate Cyclase aus Hefe (X) . Dabei liegen verschiedene Gruppen von homologen Sequenzen vor, die verschiedene Untergruppen der LLR-Proteine bilden, wie in Fig. 5 in einem phylogenetischen Baum dargestellt. Die Proteine Adenylyl Cyclase aus Saccharomyces cerevisiae sowie Sur-8, egl-15 und Rsu-1 haben eine wichtige Funktion in der Ras/Erk Signalkaskade. Die in C.elegans identifizierten Ξoc- Gene (egl-15, sem-5, soc-1 und soc-2), Suppressoren der Rezeptor Tyrosin Phophatase clr-1, beeinflussen die FGF- Signalkaskade. Soc-2, ein Protein mit einer 18 Tandem-LRR- Domäne, unterdrückt die aktivierte Form des EGL-15 FGF- Rezeptors (Fibroblast Wachstumstumsfactor) . Die FGF- Signalkaskade soll ebenfalls einen aktivierenden Effekt auf die Ras-Kaskade bewirken. Die humane Form Shoc-2, ist beim Menschen auf Chromosom 10q25 lokalisiert, einer Region, die mit bestimmten Knochen und -Krebserkrankungen assoziiert wird.(Nature 403 (6770): 676-680 (2000) and the adenylate cyclase from yeast (X). There are different groups of homologous sequences that form different subsets of the LLR proteins, as shown in FIG. 5 in a phylogenetic tree. The proteins adenylyl cyclase from Saccharomyces cerevisiae and Sur-8, egl-15 and Rsu-1 have an important function in the Ras / Erk signal cascade. The Ξoc genes identified in C.elegans (egl-15, sem-5, soc-1 and soc-2), suppressors of the receptor tyrosine phosphatase clr-1, influence the FGF signal cascade. Soc-2, a protein with an 18 tandem LRR domain, suppresses the activated form of the EGL-15 FGF receptor (fibroblast growth factor). The FGF signal cascade is also said to have an activating effect on the Ras cascade. The human form Shoc-2, is localized to chromosome 10q25 in humans, a region that is associated with certain bones and cancer.
Die andere Untergruppen der LLR-Proteine sind die LAP- Proteine, die sowohl eine LLR-Domäne im N-Terminus und als auch eine PDZ-Domäne im Carboxy-Terminus besitzen. Zu den LAP-Proteinen gehört das C. elgans Protein Let-413, das Dro- sophila-Protein Scribble, die Vertebraten-Proteine Densin- 180, Erbin und das humane Scribble. Interessanterweise haben die LAP-Proteine ein gemeinsames Expressionsmuster entlang der Zellmembran.The other subgroups of the LLR proteins are the LAP proteins, which have both an LLR domain in the N-terminus and a PDZ domain in the carboxy terminus. The LAP proteins include the C. elgans protein Let-413, the Sophosila protein Scribble, the vertebrate proteins Densin-180, Erbin and the human Scribble. Interestingly, the LAP proteins have a common expression pattern along the cell membrane.
Aufgrund der Ergebinsse des hieraus resultierenden phylogenetischen Baum (Fig. 5) kann angenommen werden, dass 15B3 nicht zu den bisher bekannten LLR-Subgruppen zugeordnet werden kann.Based on the yield rate of the resulting phylogenetic tree (FIG. 5), it can be assumed that 15B3 cannot be assigned to the previously known LLR subgroups.
(d) Genomische Struktur von 15B3 beim Menschen Eine auf den erfindungsgemäßen Erkenntnissen beruhende Datenbanksuche mit dem Programm BLAST 2 (Altschul, et al., 1997, Nucleic Acids Res, 25, 3389-402)) in der EMBL-Datenbank er- brachte zusätzliche humane genomische Sequenzinformation. Der Klon AL136108, bestehend aus 36 Zusammengesetzen contigs, enthält große Bereiche der 15B3-cDNA. Sequenzvergleiche zwischen der genomischen Nucleotidsequenz deuten darauf hin, daß der humane cDNA Klon mindestens von zwei Exons kodiert wird. Der 3 '-Bereich ist dabei komplett konserviert. 15B3 befindet sich beim Menschen auf Chromosom 9 (Position: 9q22.32-31.3; http: //www. sanger.ac.uk/cgi-bin/humace/searcher. cgi) .(d) Genomic structure of 15B3 in humans A database search based on the knowledge according to the invention with the BLAST 2 program (Altschul, et al., 1997, Nucleic Acids Res, 25, 3389-402)) in the EMBL database yielded additional human genomic sequence information. The clone AL136108, consisting of 36 composite contigs, contains large areas of the 15B3 cDNA. Sequence comparisons between the genomic nucleotide sequence indicate that the human cDNA clone is encoded by at least two exons. The 3 'area is completely preserved. 15B3 is located on chromosome 9 in humans (position: 9q22.32-31.3; http: // www.sanger.ac.uk/cgi-bin/humace/searcher. Cgi).
(e) LRR-Domäne der erfindungsgemäßen Proteine(e) LRR domain of the proteins according to the invention
Aufgrund der Konservierung der LLR-Domäne bei 15B3 weist eine LLR-Domäne auf, wie sie für Proteine, die an einer Interaktion mit Ras beteiligt sind, typisch ist. So gehört die mit Ras assoziierende LRR-Domäne von S. Cerevisiae Adenylyl Cyclase einer kleine Untergruppe an, die einen bestimmten 23-AS Con- sensus-Unit besitzt (PXXaXXLXXLXXLXLNXLXXa, a ist hierbei eine aliphatische, X jede beliebige Aminosäure) . Andere Mitglieder dieser Untergruppe wie z.B. RSU1 und SUR-8 sind zwei mutmaßliche Gerüstproteine, die einen modulierenden Effekt auf die Ras-abhängige Signalkaskade haben. Beiden gemeinsam ist die Bildung eines ternären Komplexes mit Ras und Raf und der »daraus resultierenden Aktivierung der Ras-MAP-Kinase Signaltransduktion. Aus dem Stand der Technik ist bspw. ferner bekannt, daß die Aktivierung von Ras und der Raf/Erk- Kaskade (Raf, Mek; Erk2) auch eine wesentliche Rolle für die Sauerstoff-Glucosemangel Toleranz in Neuronen besitzt. Während der Sauerstoff-Glucosemangel-Preconditionierung in Neuronen wird eine Signalkasade gestartet, die durch die Akti- vierung der NMDA-Rezeptoren mittels Ca2+ Einstrom und Produktion von NO ausgelöst wird. NO ist ein Mediator der Ras Aktivierung mittels NMDA Rezeptor Stimulation. Die ischämische Toleranz erfolgt durch eine Aktvierung der NMDA-Glutamat Re- zeptoren.Because of the conservation of the LLR domain at 15B3, it has an LLR domain that is typical of proteins involved in interacting with Ras. The Ras associated LRR domain of S. Cerevisiae adenylyl cyclase belongs to a small subgroup that has a specific 23-AS consensus unit (PXXaXXLXXLXXLXLNXLXXa, a is an aliphatic, X any amino acid). Other members of this subgroup, such as RSU1 and SUR-8, are two putative scaffold proteins that have a modulating effect on the Ras-dependent signal cascade. Both have in common the formation of a ternary complex with Ras and Raf and the resulting activation of Ras-MAP kinase signal transduction. It is also known, for example, from the prior art that the activation of Ras and the Raf / Erk cascade (Raf, Mek; Erk2) also has an essential role for the oxygen-glucose deficiency tolerance in neurons. During the oxygen-glucose deficiency preconditioning in neurons, a signal cascade is started, which is activated by the vation of the NMDA receptors is triggered by Ca2 + inflow and production of NO. NO is a mediator of Ras activation using NMDA receptor stimulation. The ischemic tolerance is achieved by activating the NMDA glutamate receptors.
Die erfindungsgemäßen Proteine enthalten im C-terminus eine Tandem-Leucin-Repeat-reichen Domäne (LRR) . Diese LRRs im er- findungemäßen Protein sind Protein-Protein- Interaktionsdomänen bestehend aus 20-28 Aminosäuren mit einem Core-Consensus von Leucinen und Asparaginen (LXXLXLXXN) . Bei dem Protein 15B3 besteht diese Domäne typischerweise aus 10 Leucin-Rich Repeats (Fig. 4) .The proteins according to the invention contain a tandem leucine repeat-rich domain (LRR) in the C-terminus. These LRRs in the protein according to the invention are protein-protein interaction domains consisting of 20-28 amino acids with a core consensus of leucines and asparagines (LXXLXLXXN). In the 15B3 protein, this domain typically consists of 10 leucine rich repeats (FIG. 4).
Zusammenfassung der experimentellen DatenSummary of experimental data
Die experimentellen Daten nach der vorliegenden Erfindung zeigen, daß die identifizierten Proteine eine entscheidende Rolle bei der Regulation der fokalen cerebralen Ischämie, ei- nem Schlaganfallmodell, spielen.The experimental data according to the present invention show that the identified proteins play a crucial role in the regulation of focal cerebral ischemia, a stroke model.
Mit den Proteinen 15B3, 15B3-1 bzw. 15B3-2 wurden Zielsequenzen für Pharmaka („drug targets") mittels eines Verfahrens zur Klonierung differentiell regulierter Gene (RMDD) in der ischämischen Hemisphäre von Mäusen nach fokaler cerebraler Ischämie identifiziert. Die Regulation der Genexpression spielt bei der cerebralen Ischämie eine entscheidende Rolle für den Ablauf und das Ausmaß des Neuronenschadens (Koistinaho and Hokfelt, 1997, Neuroreport, 8, i-viii; Schneider et al., 1999, Nat. Med., 5, 554-9). Insbesondere sog. „immediate early genes" spielen hier eine Rolle (Atkins et al., 1996, Stroke, 27, 1682-1687), wie z.B. cox-2, (Nogawa, et al., 1997, J. Neurosci., 17, 2746-2755). Das Tiermodell der fokalen cerebralen Ischämie stellt dabei ein valides Modell für den humanen ischämischen Schlaganfall dar. Um eine fokale cerebrale Ischmämie herbeizuführen, wurde das sog. Fadenmodell, bei dem ein beschichteter Nylonfaden durch die A. carotis interna an den Abgang der A. cerebri media vorgeschoben wird und einen ischämischen Schlaganfall induziert, benutzt (Clark et al., 1997, Neurol. Res., 19, 641- 648) .Proteins 15B3, 15B3-1 and 15B3-2 were used to identify drug target sequences using a method for cloning differentially regulated genes (RMDD) in the ischemic hemisphere of mice after focal cerebral ischemia. The regulation of gene expression plays a crucial role in the course and extent of neuronal damage in cerebral ischemia (Koistinaho and Hokfelt, 1997, Neuroreport, 8, i-viii; Schneider et al., 1999, Nat. Med., 5, 554-9). So-called “immediate early genes” in particular play a role here (Atkins et al., 1996, Stroke, 27, 1682-1687), such as cox-2, (Nogawa, et al., 1997, J. Neurosci., 17 , 2746-2755). The Animal model of focal cerebral ischemia represents a valid model for human ischemic stroke. In order to induce focal cerebral ischemia, the so-called thread model was used, in which a coated nylon thread was advanced through the internal carotid artery to the outlet of the cerebral artery and induces an ischemic stroke (Clark et al., 1997, Neurol. Res., 19, 641-648).
Die 15B3-Expression wurde nach einer fokalen cerebralen I- schämie in drei Zeitverläufen beobachtet: Zum einen in einer transienten Ischämie nach zwei Reperfusionszeiten (90 min I- schämie, 2 h und 6 h Reperfusion) , und zum anderen in einer permanenten Ischämie von 24 h (Fig. 2) . RNA wurde aus den beiden Hemisphärenhälften von 3-4 Gehirnen ohne Hirnstamm und Kleinhirn gewonnen (Fasttrack kit, Invitrogen) . Mit Hilfe des LightCycler™ Systems (Röche Diagnostics, Mannheim) wurde eine quantitative PCR durchgeführt. Der cDNA Gehalt der Proben wurde auf die Expression von Cyclophilin und S20 normiert (Schneider, et al., 1995, Proc Natl Acad Sei U S A, 92, 4447- 51) . Benutzte Primer zur Amplifikation von Cyclophilin waren: cyc5 ACCCCACCGTGTTCTTCGAC acyc300 CATTTGCCATGGACAAGATG und zur Amplifikation von Maus 15B3: seq_15B3.1 GAGCTGCCAAGAAAGGGGGAGACT (in Exon 2) seq_15B3.2 CAGAAAACACAAATGGACGGAGAG (in Exon 2) (Diese Primer amplifizieren ein Amplimer von 501 bp, das am 3' Ende der Maus-cDNA liegt) .15B3 expression was observed in three time courses after focal cerebral ischemia: on the one hand in transient ischemia after two reperfusion times (90 min ischemia, 2 h and 6 h reperfusion), and on the other hand in permanent ischemia of 24 h (Fig. 2). RNA was obtained from the two hemispheres of 3-4 brains without a brain stem and cerebellum (Fasttrack kit, Invitrogen). A quantitative PCR was carried out using the LightCycler ™ system (Röche Diagnostics, Mannheim). The cDNA content of the samples was normalized for the expression of cyclophilin and S20 (Schneider, et al., 1995, Proc Natl Acad Sei U S A, 92, 4447-51). Primers used for the amplification of cyclophilin were: cyc5 ACCCCACCGTGTTCTTCGAC acyc300 CATTTGCCATGGACAAGATG and for the amplification of mouse 15B3: seq_15B3.1 GAGCTGCCAAGAAAGGGGGAGACT (in Exon 2) seq_15B3 2AT CAGGAAConAg (in Exon 2) a 'End of mouse cDNA is).
Im Ergebnis zeigt sich eine deutliche Hochregulation um den Faktor 2-3 von 15B3-RNA auf der ischämischen (linken) Hirnhälfte 2h nach dem ischämischen Ereignis (middle cerebral artery occlusion von 90 min und 2 h Reperfusion, also in einem Modell akuter Neurodegeneration (Fig. 2) ; die Fehlerbalken zeigen Standardabweichungen, diese ergeben sich aus Messungen mit 3-fach seriell verdünnten cDNA-Proben und spiegelt damit die Reliabilität der Messergebnisse dar) . Nach 24 h (in einem permanenten Modell) hingegen konnte kein Unterschied mehr festgestellt werden. Erfindungsgemäße Proteine, also bspw. 15B3 nimmt bei intrazellulären Signaltransduktionsprozessen eine wichtige Funktion ein, weswegen sie auch bei pathologischen Prozessen (z.B. dem Schlaganfall) beteiligt sind. The result shows a clear upregulation by a factor of 2-3 of 15B3-RNA on the ischemic (left) hemisphere 2h after the ischemic event (middle cerebral artery occlusion of 90 min and 2 h reperfusion, i.e. in one Acute neurodegeneration model (Fig. 2); the error bars show standard deviations, these result from measurements with 3-fold serial diluted cDNA samples and thus reflect the reliability of the measurement results). After 24 hours (in a permanent model), however, no difference could be determined. Proteins according to the invention, for example 15B3, play an important role in intracellular signal transduction processes, which is why they are also involved in pathological processes (for example stroke).

Claims

Patentansprüche claims
1. DNA-Sequenz, dadurch gekennzeichnet, daß sie einen Sequenzbereich enthält, der für ein Polypeptid mit einer Aminosäuresequenz von AS 312 bis AS 711 (15B3) , AS 180 bis AS 579 (15B3-1) oder AS 306 bis AS 705 (15B3-2) (Nu- merierung gemäß Figur 4) codiert, einschließlich aller funktionshomologen Derivate, Fragmente oder Allele, oder hiermit hybridisierenden DNA-Sequenzen.1. DNA sequence, characterized in that it contains a sequence region for a polypeptide with an amino acid sequence from AS 312 to AS 711 (15B3), AS 180 to AS 579 (15B3-1) or AS 306 to AS 705 (15B3 -2) (numbering according to FIG. 4), including all functionally homologous derivatives, fragments or alleles, or DNA sequences hybridizing therewith.
2. DNA-Sequenz nach Anspruch 1, dadurch gekennzeichnet, daß sie einen Sequenzbereich enthält, der für ein Polypeptid mit einer Aminosäuresequenz gemäß Figur 9, 11 oder 13 codiert, einschließlich aller funktionshomologen Derivate, Fragmente oder Allele, oder hiermit hybridisierenden DNA-Sequenzen.2. DNA sequence according to claim 1, characterized in that it contains a sequence region which codes for a polypeptide with an amino acid sequence according to Figure 9, 11 or 13, including all functionally homologous derivatives, fragments or alleles, or DNA sequences hybridizing therewith.
3. DNA-Sequenz nach Anspruch 1 oder 2, dadurch gekennzeichnet, daß sie für ein Polypeptid gemäß Figur 9, 11 oder 13 codiert, einschließlich aller funktions-homologen Derivate, Fragmente oder Allele, oder hiermit hybridisie- renden DNA-Sequenzen.3. DNA sequence according to claim 1 or 2, characterized in that it codes for a polypeptide according to Figure 9, 11 or 13, including all function-homologous derivatives, fragments or alleles, or with this hybridizing DNA sequences.
4. DNA-Sequenz nach einem der vorgenannten Ansprüche, dadurch gekennzeichnet, daß sie die in Figur 8, 10 oder 12 angegebene (c) DNA-Sequenz enthält.4. DNA sequence according to one of the preceding claims, characterized in that it contains the (c) DNA sequence indicated in Figure 8, 10 or 12.
Expressionsvektor, dadurch gekennzeichnet, daß er eine DNA-Sequenz nach einem der Ansprüche 1 bis 4 enthält. Expression vector, characterized in that it contains a DNA sequence according to one of Claims 1 to 4.
6. Wirtszelle, dadurch gekennzeichnet, daß sie mit einem Expressionsvektor nach Anspruch 5 transformiert ist.6. Host cell, characterized in that it is transformed with an expression vector according to claim 5.
7. Wirtszelle nach Anspruch 6, dadurch gekennzeichnet, daß sie eine Säugetierzelle, insbesondere eine humane Zelle, ist.7. Host cell according to claim 6, characterized in that it is a mammalian cell, in particular a human cell.
8. Aufgereinigtes Genprodukt, dadurch gekennzeichnet, daß es durch eine DNA-Sequenz nach einem der Ansprüche 1 bis 4 codiert wird.8. Purified gene product, characterized in that it is encoded by a DNA sequence according to one of claims 1 to 4.
9. Aufgereinigtes Genprodukt nach Anspruch 8, dadurch gekennzeichnet, daß es ein Polypeptid ist.9. Purified gene product according to claim 8, characterized in that it is a polypeptide.
10. Aufgereinigtes Genprodukt nach Anspruch 8 oder 9, dadurch gekennzeichnet, daß es die in Figur 9, 11 oder 13 angegebene Aminosäuresequenz enthält, einschließlich aller funktionshomologen Allele, Fragmente oder Derivate.10. Purified gene product according to claim 8 or 9, characterized in that it contains the amino acid sequence shown in Figure 9, 11 or 13, including all functional homologous alleles, fragments or derivatives.
11. Transgene Tiere, dadurch gekennzeichnet, daß sie genetisch dahingehend verändert, daß sie eine im Vergleich zum Normaltier (spezifisch) veränderte Menge mindestens einer Aminosäuresequenz nach Anspruch 9 oder 10 oder eine spezifisch gegenüber einer nativen Sequenz (gemäß Fi- guren 9, 11 oder 13) veränderte Aminosäuresequenz gemäß Anspruch 9 oder 10 enthalten oder daß ihnen mindestens eine Aminosäuresequenz gemäß Anspruch 9 oder Teile davon fehlt oder verändert vorliegt.11. Transgenic animals, characterized in that they are genetically modified in such a way that they change (specifically) a quantity of at least one amino acid sequence according to claim 9 or 10 or a specific one compared to a native sequence (according to FIGS. 9, 11 or 13) contain modified amino acid sequence according to claim 9 or 10 or that at least one amino acid sequence according to claim 9 or parts thereof is missing or changed.
12. Antikörper, dadurch gekennzeichnet, daß er ein Epitop auf einem Genprodukt nach einem der Ansprüche 8 bis 10 erkennt. 12. Antibody, characterized in that it recognizes an epitope on a gene product according to one of claims 8 to 10.
13. Antikörper nach Anspruch 12, dadurch gekennzeichnet, daß er monoklonal ist.13. Antibody according to claim 12, characterized in that it is monoclonal.
14. Antikörper nach einem der Ansprüche 12 oder 13, dadurch gekennzeichnet, daß er gegen einen Sequenzabschnitt auf der cytoplasmatischen Domäne als Epitop gerichtet ist.14. Antibody according to one of claims 12 or 13, characterized in that it is directed against a sequence section on the cytoplasmic domain as an epitope.
15. Verfahren zur Expression von Genprodukten nach einem der Ansprüche 8 bis 10, dadurch gekennzeichnet, daß Wirts- zellen mit einem Expressionsvektor nach Anspruch 5 transformiert werden.15. A method for the expression of gene products according to one of claims 8 to 10, characterized in that host cells are transformed with an expression vector according to claim 5.
16. Verfahren zur Isolierung von Genprodukten nach einem der Ansprüche 8 bis 10, dadurch gekennzeichnet, daß Wirts- zellen nach Anspruch 6 oder 7 unter geeigneten, die Expression fördernden Bedingungen kultiviert werden und das Genprodukt anschließend aus der Kultur aufgereinigt wird.16. A method for isolating gene products according to one of claims 8 to 10, characterized in that host cells according to claim 6 or 7 are cultivated under suitable conditions which promote expression and the gene product is subsequently purified from the culture.
17. DNA-Sequenz nach einem der Ansprüche 1 bis 4 oder eines Genprodukts nach einem der Ansprüche 8 bis 10 als Arzneimittel.17. DNA sequence according to one of claims 1 to 4 or a gene product according to one of claims 8 to 10 as a medicament.
18. Verwendung einer DNA-Sequenz nach einem der Ansprüche 1 bis 4 oder eines Genprodukts nach einem der Ansprüche 8 bis 10 zur Behandlung (bzw. zur Herstellung eines Arzneimittels zur Behandlung) von Erkrankungen, die auf fehlgesteuerter Regulation von Zelltod- und/oder Zellproliferationsereignissen beruhen.18. Use of a DNA sequence according to one of claims 1 to 4 or of a gene product according to one of claims 8 to 10 for the treatment (or for the manufacture of a medicament for the treatment) of diseases which are based on incorrectly controlled cell death and / or cell proliferation events based.
19. Verwendung einer DNA-Sequenz nach einem der Ansprüche 1 bis 4 oder eines Genprodukts nach einem der Ansprüche 8 bis 10 zur Behandlung (bzw. zur Herstellung eines Arzneimittels zur Behandlung) von Tumorerkrankungen und neurologischen Erkrankungen, insbesondere Schlaganfall, Multipler Sklerose, Morbus Parkinson, Amyotrophe Late- ralsklerose, Heredodegenerative Ataxien, Morbus Huntington, Neuropathien und Epilepsien.19. Use of a DNA sequence according to one of claims 1 to 4 or a gene product according to one of claims 8 to 10 for the treatment (or for the manufacture of a medicament for the treatment) of tumor diseases and neurological diseases, in particular stroke, multiple sclerosis, Parkinson's disease, amyotrophic latex sclerosis, heredodegenerative ataxias, Huntington's disease, neuropathies and epilepsy.
20. Verbindung, dadurch gekennzeichnet, daß sie die intrazelluläre Funktion des Proteins 15B3, 15B3-1 und/oder 15B3-2 moduliert, insbesondere inhibiert.20. Compound, characterized in that it modulates, in particular inhibits, the intracellular function of the 15B3, 15B3-1 and / or 15B3-2 protein.
21. Verbindung nach Anspruch 20 oder 21, dadurch gekennzeichnet, daß sie eine organisch-chemische Verbindung mit einem Molekulargewicht von vorzugsweise < 3000 ist.21. A compound according to claim 20 or 21, characterized in that it is an organic chemical compound with a molecular weight of preferably <3000.
22. Verbindung nach einem der Ansprüche 19 bis 21, dadurch gekennzeichnet, daß sie die Zellmembran durch Diffusion oder über membranöse Transportproteine passiert.22. A compound according to any one of claims 19 to 21, characterized in that it passes through the cell membrane by diffusion or via membrane transport proteins.
23. Verfahren zur Identifizierung von pharmazeutisch wirksamen Verbindungen nach einem der Ansprüche 19 bis 22, dadurch gekennzeichnet, daß23. A method for identifying pharmaceutically active compounds according to any one of claims 19 to 22, characterized in that
(a) ein geeignetes Wirtszellsystem mit einem Expressionsvektor nach Anspruch 5, insbesondere einem Expressi- onsvektor, der für das Protein 15B3, 15B3-1 oder(a) a suitable host cell system with an expression vector according to claim 5, in particular an expression vector which is suitable for the protein 15B3, 15B3-1 or
15B3-2 codiert, und ggf. mindestens einem Expressionsvektor, der für mindestens ein Reporter-Gen codiert, transfiziert wird, und15B3-2, and if appropriate at least one expression vector which codes for at least one reporter gene, is transfected, and
(b) ein zur Beobachtung der 15B3, 15B3-1 und/oder 15B3-2 vermittelten Funktion, insbesondere ein zur Beobachtung der Apoptose, des Zellwachstums, der Zeilproliferation und/oder der Zellplastizität geeigneter Parameter nach Zugabe einer Testverbindung im Vergleich zur Kontrolle ohne Zugabe einer Testverbindung für das gemäß (a) erhaltene Wirtszellsystem in einem geeigneten Testsystem gemessen wird.(b) a function mediated for the observation of the 15B3, 15B3-1 and / or 15B3-2, in particular a parameter suitable for the observation of apoptosis, cell growth, cell proliferation and / or cell plasticity after adding a test compound in comparison as a control without adding a test compound for the host cell system obtained in (a) is measured in a suitable test system.
24. Verfahren nach Anspruch 23, dadurch gekennzeichnet, daß (c) der Bindungsplatz der pharmazeutisch wirksamen Verbindung auf einem erfindungsgemäßen Protein durch ein geeignetes biochemisches oder strukturbiologisches Verfahren ermit- telt wird.24. The method according to claim 23, characterized in that (c) the binding site of the pharmaceutically active compound on a protein according to the invention is determined by a suitable biochemical or structural biological method.
25. Verfahren nach Anspruch 23 oder 24, dadurch gekennzeichnet, daß ein Parameter gemäß (b) innerhalb eines Assay- aufbaus für die Raf/MEK/ERK-Kaskade gemessen wird.25. The method according to claim 23 or 24, characterized in that a parameter according to (b) is measured within an assay setup for the Raf / MEK / ERK cascade.
26. Verwendung einer Verbindung nach Anspruch 19 bis 22 zur Behandlung von (bzw. zur Herstellung eines Arzneimittels zur Behandlung von) neurodegenerativen Erkrankungen, insbesondere Alzheimer-Demenz und Morbus Parkinson, Mus- keldystrophie, viralen Infektionserkrankungen, Tumorerkrankungen und Autoimmunerkrankungen oder cerebralen I- schämien.26. Use of a compound according to claims 19 to 22 for the treatment of (or for the manufacture of a medicament for the treatment of) neurodegenerative diseases, in particular Alzheimer's disease and Parkinson's disease, muscular dystrophy, viral infectious diseases, tumor diseases and autoimmune diseases or cerebral ischemia ,
27. Verfahren zur Identifizierung eines zellulären Interak- tionspartner des Proteins 15B3, 15B3-1 und/oder 15B3-2 oder eines nativen Alieis, Fragments oder Derivats, wobei ein sog. "yeast-two-hybrid"-System eingesetzt wird.27. A method for identifying a cellular interaction partner of the 15B3, 15B3-1 and / or 15B3-2 protein or a native alien, fragment or derivative, using a so-called "yeast-two-hybrid" system.
28. Verwendung einer DNA-Sequenz nach einem der Ansprüche 1 bis 4 oder eines Genprodukts nach einem der Ansprüche 8 bis 10 zur Identifizierung von weiteren an der durch das Protein 15B3, 15B3-1 und/oder 15B3-2 vermittelten Signaltransduktion beteiligten Proteinen. 28. Use of a DNA sequence according to one of claims 1 to 4 or of a gene product according to one of claims 8 to 10 for the identification of further proteins involved in the signal transduction mediated by the protein 15B3, 15B3-1 and / or 15B3-2.
29. Verwendung einer DNA-Sequenz nach einem der Ansprüche 1 bis 4 oder eines Genprodukts nach einem der Ansprüche 8 bis 10 als Suizidgen/-protein für die in vivo oder ex vivo Transformation von Wirtszellen.29. Use of a DNA sequence according to one of claims 1 to 4 or a gene product according to one of claims 8 to 10 as a suicide gene / protein for the in vivo or ex vivo transformation of host cells.
30. Verwendung einer DNA-Sequenz nach Anspruch 29, wobei das Suizidgen operabel mit einem Promotor verbunden ist, wobei die Transkription reprimiert ist und nur im Bedarfs- fall aktiviert wird. 30. Use of a DNA sequence according to claim 29, wherein the suicide gene is operably linked to a promoter, wherein the transcription is repressed and is only activated when necessary.
PCT/EP2002/001562 2001-02-14 2002-02-14 Signal transduction proteins 15b3, 15b3-1 and 15b3-2, and related dna sequences WO2002064776A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002250943A AU2002250943A1 (en) 2001-02-14 2002-02-14 Signal transduction proteins 15b3, 15b3-1 and 15b3-2, and related dna sequences

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
DE10106835.2 2001-02-14
DE10106835A DE10106835A1 (en) 2001-02-14 2001-02-14 Signal transduction proteins 15B3, 15B3-1 and 15B3-2 and underlying DNA sequences

Publications (2)

Publication Number Publication Date
WO2002064776A2 true WO2002064776A2 (en) 2002-08-22
WO2002064776A3 WO2002064776A3 (en) 2003-08-21

Family

ID=7674010

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2002/001562 WO2002064776A2 (en) 2001-02-14 2002-02-14 Signal transduction proteins 15b3, 15b3-1 and 15b3-2, and related dna sequences

Country Status (3)

Country Link
AU (1) AU2002250943A1 (en)
DE (1) DE10106835A1 (en)
WO (1) WO2002064776A2 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998037210A1 (en) * 1997-02-21 1998-08-27 KANTON ZÜRICH vertreten durch DIE ERZIEHUNGSDIREKTION Immunological detection of prions
WO1999014328A2 (en) * 1997-09-17 1999-03-25 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2883900A (en) * 1999-07-07 2001-01-30 Genentech Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
EP1396543A3 (en) * 1999-07-08 2004-03-31 Research Association for Biotechnology Primers for synthesizing full length cDNA clones and their use
EP1074617A3 (en) * 1999-07-29 2004-04-21 Research Association for Biotechnology Primers for synthesising full-length cDNA and their use
AU7721500A (en) * 1999-09-29 2001-04-30 Human Genome Sciences, Inc. Colon and colon cancer associated polynucleotides and polypeptides
WO2001055326A2 (en) * 2000-01-31 2001-08-02 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
AU6802801A (en) * 2000-03-01 2001-09-24 Genentech Inc Secreted and transmembrane polypeptides and nucleic acids encoding the same
AU2001274888A1 (en) * 2000-05-19 2001-12-03 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
AU6531101A (en) * 2000-06-02 2001-12-17 Genentech Inc Secreted and transmembrane polypeptides and nucleic acids encoding the same
CA2412211A1 (en) * 2000-06-23 2002-01-03 Genetech, Inc. Compositions and methods for the diagnosis and treatment of disorders involving angiogenesis
CA2416456A1 (en) * 2000-07-20 2002-01-31 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998037210A1 (en) * 1997-02-21 1998-08-27 KANTON ZÜRICH vertreten durch DIE ERZIEHUNGSDIREKTION Immunological detection of prions
WO1999014328A2 (en) * 1997-09-17 1999-03-25 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same

Non-Patent Citations (11)

* Cited by examiner, † Cited by third party
Title
DATABASE EPOP [Online] 1. März 2002 (2002-03-01) BAKER K.P. ET AL.: "Secreted and transmembrane polypeptides and nucleic acids encoding the same" Database accession no. AX376345 XP002214593 -& WO 01 68848 A (GENETECH INC.) 20. September 2001 (2001-09-20) *
DATABASE EPOP [Online] 13. Februar 2002 (2002-02-13) BAKER K.P. ET AL.: "Secreted and transmembrane polypeptides and nucleic acids encoding the same" Database accession no. AX358871 XP002214597 -& WO 01 93983 A (GENETECH INC) 13. Dezember 2001 (2001-12-13) *
DATABASE EPOP [Online] 15. Februar 2002 (2002-02-15) BAKER K.P. ET AL.: "Secreted and transmembrane polypeptides and nucleic acids encoding the same" Database accession no. AX362364 XP002214596 -& WO 02 08288 A (GENETECH INC.) 31. Januar 2002 (2002-01-31) *
DATABASE GSP [Online] 16. Mai 2002 (2002-05-16) BAKER K.P.: "Human PRO247 protein sequence SEQ ID NO:40" Database accession no. ABB84836 XP002214595 -& WO 02 00690 A (GENETECH INC) 3. Januar 2002 (2002-01-03) *
DATABASE GSP [Online] 24. April 2001 (2001-04-24) ASHKENAZI A.J. ET AL.: "Human PRO247 protein" Database accession no. AAB80254 XP002214589 -& WO 01 04311 A (GENETECH INC) 18. Januar 2001 (2001-01-18) *
DATABASE GSP [Online] 24. Mai 2002 (2002-05-24) BIRSE C.E. ET AL.: "Human polypeptide SEQ ID NO:2312" Database accession no. ABB89936 XP002214598 -& WO 01 90304 A (HUMAN GENOME SCI INC.) 29. November 2001 (2001-11-29) *
DATABASE GSP [Online] 26. Juni 2001 (2001-06-26) OTA T. ET AL.: "Human protein sequence SEQ ID NO:14536" Database accession no. AAB94199 XP002214592 -& EP 1 074 617 A (HELIX RES INST.) 7. Februar 2001 (2001-02-07) *
DATABASE GSP [Online] 3. September 2001 (2001-09-03) RUBEN S.M. ET AL.: "Human colon cancer antigen protein SEQ ID NO:6177" Database accession no. AAG75413 XP002214590 -& WO 01 22920 A (HUMAN GENOME SCI INC) 5. April 2001 (2001-04-05) *
DATABASE GSP [Online] 4. Dezember 2001 (2001-12-04) ROSEN C.A. ET AL.: "Human secreted protein SEQ ID NO:418" Database accession no. AAU20426 XP002214594 -& WO 01 55326 A (HUMAN GENOME SCI INC) 2. August 2001 (2001-08-02) *
DATABASE GSP [Online] 6. November 2001 (2001-11-06) OTA T. ET AL.: "Human polypeptide SEQ ID NO:3764" Database accession no. AAM93765 XP002214591 -& EP 1 130 094 A (HELIX RES. INST.) 5. September 2001 (2001-09-05) *
NAGASE T ET AL: "PREDICTION OF THE CODING SEQUENCES OF UNIDENTIFIED HUMAN GENES. XVI THE COMPLETE SEQUENCES OF 150 NEW CDNA CLONES FROM BRAIN WHICH CODE FOR LARGE PROTEINS IN VITRO" DNA RESEARCH, Bd. 7, 2000, Seiten 65-73, XP000949814 ISSN: 1340-2838 *

Also Published As

Publication number Publication date
AU2002250943A1 (en) 2002-08-28
WO2002064776A3 (en) 2003-08-21
DE10106835A1 (en) 2002-09-05

Similar Documents

Publication Publication Date Title
DE69820419T2 (en) CLONING AND USE OF THE LSR RECEPTOR
DE69819345T2 (en) NUCLEIC ACID SEQUENCES ENCODING THE CAPSAICIN RECEPTOR AND POLYPEPTIDES SIMILAR TO THE CAPSAICIN RECEPTOR AND THEIR USE
DE69534402T2 (en) APOPTOSIS PROTEASES 3 AND 4, SIMILAR TO THE CONVERSION CYCLE FOR INTERLEUKIN-1BETA
DE69838905T2 (en) TRANSCRIPTION FACTOR ISLET-BRAIN 1 (IB1)
DE60034450T2 (en) 22025, A NEW HUMAN CYCLIC NUCLEOTIDE PHOSPHODIESTERASE
DE69926764T2 (en) METHODS FOR DETERMINING COMPONENTS FOR MODULATING THE BODY WEIGHT
DE69932153T2 (en) PHOSPHODIESTERASE 10
DE10294485T5 (en) Proteins homologous with Mnk kinase, which are involved in the regulation of energy homeostasis and organelle metabolism
WO2002040668A2 (en) Proteins and dna sequences underlying these proteins used for treating inflammations
DE69635349T2 (en) NUCLEOTIDE SEQUENCES, PROTEINS, MEDICAMENTS AND DIAGNOSIS AGENTS FOR THE USE OF CANCER TREATMENT
WO2006000213A9 (en) Peptides for inhibiting the interaction of protein kinase a and protein kinase a anchor proteins
DE60031241T2 (en) KALIUM CHANNELS AND GENES CODING THESE KALIUM CHANNELS
EP0805204B1 (en) Epididymis-specific receptor protein and its use
DE69931345T2 (en) GEN, WHICH CODES FOR NEW TRANSMEMBRANE PROTEIN
DE60036548T2 (en) NEW KALIUM CHANNELS, AND GENES THAT ARE CODED FOR
DE60117788T2 (en) HUMAN WINGLESS-SIMILAR GENES
DE69434090T2 (en) EPSILON SUB-UNIT OF THE GABA A-RECEPTOR
WO2002064776A2 (en) Signal transduction proteins 15b3, 15b3-1 and 15b3-2, and related dna sequences
DE60124380T2 (en) ANGIOGENESIS ASSOCIATED PROTEINS AND NUCLEIC ACID CODING THEREFOR
EP1436327B1 (en) Ee3-protein family and corresponding dna sequences
WO2002064777A2 (en) Proteins 7b6 and 11b4 and dna sequences encoding the same
DE10163467A1 (en) New DNA sequence encoding protein 24B2, useful for treatment and diagnosis or cancer and degenerative diseases, also related polypeptides and modulators
DE69839150T2 (en) INTRA-CELLULAR GLUCOCORTICOID-INDUCED LEUCIN-ZIPPER MODULATORS OF MECHANISMS OF APOPTOTIC CELL-DATE
DE60217711T2 (en) PTP10D NUCLEIC ACIDS AND PEPTIDES IN THE REGULATION OF ENERGY HOMEOSTASIS
DE60024862T2 (en) &#34;INSULIN-DEPENDENT SEQUENCE DNA BINDING PROTEIN-1&#34; (IRSDBP-1), A GENERIC ENCODER AND ITS USES

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 BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE 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 NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE 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
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP