WO2000066728A1 - Homologues de star - Google Patents

Homologues de star Download PDF

Info

Publication number
WO2000066728A1
WO2000066728A1 PCT/IL2000/000252 IL0000252W WO0066728A1 WO 2000066728 A1 WO2000066728 A1 WO 2000066728A1 IL 0000252 W IL0000252 W IL 0000252W WO 0066728 A1 WO0066728 A1 WO 0066728A1
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
star
acid sequence
sequence
product
Prior art date
Application number
PCT/IL2000/000252
Other languages
English (en)
Inventor
Kinneret Savitzky
Liat Mintz
Original Assignee
Compugen Ltd.
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 Compugen Ltd. filed Critical Compugen Ltd.
Priority to AU43104/00A priority Critical patent/AU4310400A/en
Priority to EP00922827A priority patent/EP1175492A1/fr
Publication of WO2000066728A1 publication Critical patent/WO2000066728A1/fr

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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention concerns novel nucleic acid sequences, vectors and host cells containing them, amino acid sequences encoded by said sequences, and antibodies reactive with said amino acid sequences, as well as pharmaceutical compositions comprising any of the above.
  • the present invention further concerns methods for screening for candidate activator or deactivators utilizing said amino acid sequences.
  • Steroid hormones produced in the placenta, adrenals, ovaries, and gonads are important mediators of tissue differentiation, development, and homeostasis.
  • the synthesis of these important mediators of both genomic and non-genomic effects is tightly regulated in a temporal and tissue-specific manner.
  • the acute stimulation of steroidogenesis in the adrenals and gonads is triggered by trophic hormone-induced generation of cAMP with subsequent activation of phosphorylation and gene transcription by the cAMP-dependent protein kinase A (PKA).
  • PKA cAMP-dependent protein kinase A
  • the rate-limiting step in steroid hormone synthesis is the side-chain cleavage of cholesterol to form pregnenolone, a step catalyzed by the side-chain cleavage enzyme system (P450scc), which is located on the matrix side of the inner mitochondrial membrane.
  • P450scc side-chain cleavage enzyme system
  • the translocation of cholesterol from the cholesterol-rich outer mitochondrial membrane to the cholesterol-poor inner membrane is the true rate-limiting step in the acute control of steroidogenesis. Consistent with this proposition is the finding that freely diffusible cholesterol analogs (hydroxysterols) that are able to reach the inner mitochondrial membrane promote maximal levels of steroidogenesis in the absence of trophic stimulation. It has also been shown that the acute steroidogenic response is blocked within minutes of treatment with inhibitors of protein synthesis (i.e. cyclo-heximide or puromycin).
  • the protein responsible for this transport is termed “steroidogenic acute regulatory protein” (StAR).
  • lipoid CAH congenital lipoid adrenal hyperplasia
  • the human StAR cDNA encodes a 285 amino acid protein with 37 kDa mobility on SDS polyacrylamide gels (SDS-PAGE).
  • SDS-PAGE SDS polyacrylamide gels
  • the StAR protein sequence is highly conserved (80-88% identity and 90% similarity) across all of the species thus far compared, including: the mouse, human, hamster, rat, cow, sheep and pig.
  • the protein contains putative phosphorylation sites for several protein kinases including the protein kinase A (PKA), calmodulin- dependent kinase II (CAM kinase II), and protein kinase C (PKC).
  • PKA protein kinase A
  • CAM kinase II calmodulin- dependent kinase II
  • PKC protein kinase C
  • the amino terminal 33 amino acids of the predicted polypeptide possesses a high net positive charge and the potential to form an amphipathic helical structure typical of mitochondrial signal sequences. Both murine and human StAR proteins are efficiently imported and processed by mitochondria of steroidogenic and non-steroidogenic cells.
  • PCOS Polycystic ovary syndrome
  • Hyperandrogenaemia is seen both in women with PCOS and men with premature male pattern baldness suggesting an underlying disorder of androgen biosynthesis or metabolism. Androgens are synthesized by the adrenals, the theca cell layer of the developing ovarian follicle and the testicular Ley dig cells. Both scalp hair loss and hirsutism are known to be mediated by androgens. The sensitivity of the hair follicle to androgens is dependent on a number of factors, such as serum concentrations of bioavailable androgens and the presence and number of androgen receptors.
  • StepAR-B Steroidogenic acute regulatory protein-homolog (StAR-B) nucleic acid sequence
  • SEQ ID NO: 1 sequence shown in SEQ ID NO: 1, sequences having at least 70% identity to said sequence and fragments of the above sequences being 20 b.p. long.
  • This sequence is a sequence coding for a novel homolog of the known StAR protein.
  • the two sequences share homology at the C-terminus which is the region responsible for physiological and catalytic activity of StAR.
  • StAR-B does not necessarily signify that StAR-B protein coded by the above sequence has the same or even similar physiological effects as known StAR, merely that it shows sequence homology with the known StAR .
  • Steproidogenic acute protein (StAR-B product) also referred at times as the "StAR-B protein" or “StAR-B polypeptide” - is an amino acid shown in SEQ ID NO: 2.
  • the amino acid sequence may be a peptide, a protein, as well as peptides or proteins having chemically modified amino acids (see below) such as a glycopeptide or glycoprotein.
  • An example of an StAR-B product is shown in SEQ ID NO: 2.
  • the term also includes analogues of said sequences in which one or more amino acids has been added, deleted, substituted (see below) or chemically modified (see below) as well as fragments of this sequence having at least 10 amino acids.
  • Nucleic acid sequence a sequence composed of DNA nucleotides, RNA nucleotides or a combination of both types and may includes natural nucleotides, chemically modified nucleotides and synthetic nucleotides.
  • amino acid sequence - a sequence composed of any one of the 20 naturally appearing amino acids, amino acids which have been chemically modified (see below), or composed of synthetic amino acids.
  • “Fragment of StAR-B product” a polypeptide which has an amino acid sequence which is the same as part of but not all of the amino acid sequence of the StAR-B product.
  • “Fragments of StAR-B nucleic acid sequence” a continuous portion, preferably of about 20 nucleic acid sequences of the StAR-B nucleic acid sequence.
  • Constant substitution refers to the substitution of an amino acid in one class by an amino acid of the same class, where a class is defined by common physicochemical amino acid side chain properties and high substitution frequencies in homologous proteins found in nature, as determined, for example, by a standard Dayhoff frequency exchange matrix or BLOSUM matrix.
  • Class I Cys
  • Class II Ser, Thr, Pro, Ala, Gly
  • Class III Asn, Asp, Gin, Glu
  • Class IV His, Arg, Lys
  • Class V He, Leu, Val, Met
  • Class VI Phe, Tyr, Trp
  • substitution of an Asp for another class III residue such as Asn, Gin, or Glu is a conservative substitution.
  • “Non-conservative substitution" refers to the substitution of an amino acid in one class with an amino acid from another class; for example, substitution of an Ala, a class II residue, with a class III residue such as Asp, Asn, Glu, or Gin.
  • “Chemically modified” - when referring to the product of the invention, means a product (protein) where at least one of its amino acid resides is modified either by natural processes, such as processing or other post-translational modifications, or by chemical modification techniques which are well known in the art.
  • modifications typical, but not exclusive examples include: acetylation, acylation, amidation, ADP-ribosylation, glycosylation, GPI anchor formation, covalent attachment of a lipid or lipid derivative, methylation, myristlyation, pegylation, prenylation, phosphorylation, ubiqutination, or any similar process.
  • Bioly active refers to the StAR-B product which have, regulatory or biochemical functions on the same target sites which naturally occurring StAR-B influence, for example an effect in regulation of steroid synthesis, in particular androgen synthesis. This term may refer to the same biological activity as manifested in different cells in different tissues which are responsible for hyperandrogonism, such as Theca cells, Leydig cells and cells of the adrenal cortex.
  • Immunologically active defines the capability of a natural, recombinant or synthetic StAR-B product, or any fragment thereof, to induce a specific immune response in appropriate animals or cells and to bind with specific antibodies.
  • a biologically active fragment of StAR-B product denotes a fragment which retains some or all of the immunological properties of the StAR-B product, e.g can bind specific anti-StAR-B product antibodies or which can elicit an immune response which will generate such antibodies or cause proliferation of specific immune cells which produce StAR-B.
  • Optimal alignment is defined as an alignment giving the highest percent identity score. Such alignment can be performed using a variety of commercially available sequence analysis programs, such as the local alignment program LALIGN using a ktup of 1, default parameters and the default PAM. A preferred alignment is the one performed using the CLUSTAL-W program from MacVector (TM), operated with an open gap penalty of 10.0, an extended gap penalty of 0.1, and a BLOSUM similarity matrix. If a gap needs to be inserted into a first sequence to optimally align it with a second sequence, the percent identity is calculated using only the residues that are paired with a corresponding amino acid residue (i.e., the calculation does not consider residues in the second sequences that are in the "gap" of the first sequence).
  • TM MacVector
  • Having at least X% identity refers to the percentage of residues that are identical in the two sequences when the sequences are optimally aligned.
  • 70% amino acid sequence identity means that 70% of the amino acids in two or more optimally aligned polypeptide sequences are identical.
  • isolated nucleic acid molecule having an StAR-B nucleic acid sequence is a nucleic acid molecule that includes the coding StAR-B nucleic acid sequence.
  • Said isolated nucleic acid molecule may include the StAR-B nucleic acid sequence as an independent insert; may include the StAR-B nucleic acid sequence fused to an additional coding sequences, encoding together a fusion protein in which the StAR-B coding sequence is the dominant coding sequence (for example, the additional coding sequence may code for a signal peptide); the StAR-B nucleic acid sequence may be in combination with non-coding sequences, e.g., introns or control elements, such as promoter and terminator elements or 5' and/or 3' untranslated regions, effective for expression of the coding sequence in a suitable host; or may be a vector in which the StAR-B protein coding sequence is a heterologous.
  • Expression vector refers to vectors that have the ability to incorporate and express heterologous DNA fragments in a foreign cell. Many prokaryotic and eukaryotic expression vectors are known and/or commercially available. Selection of appropriate expression vectors is within the knowledge of those having skill in the art.
  • “Deletion " - is a change in either nucleotide or amino acid sequence in which one or more nucleotides or amino acid residues, respectively, are absent.
  • “Insertion” or “addition” - is that change in a nucleotide or amino acid sequence which has resulted in the addition of one or more nucleotides or amino acid residues, respectively, as compared to the naturally occurring sequence.
  • substitution - replacement of one or more nucleotides or amino acids by different nucleotides or amino acids, respectively. As regards amino acid sequences the substitution may be conservative or non- conservative.
  • Antibody refers to IgG, IgM, IgD, IgA, and IgG antibody.
  • the definition includes polyclonal antibodies or monoclonal antibodies. This term refers to whole antibodies or fragments of the antibodies comprising the antigen-binding domain of the anti-StAR-B product antibodies, e.g. antibodies without the Fc portion. single chain antibodies, fragments consisting of essentially only the variable, antigen-binding domain of the antibody, etc.
  • Activator refers to a molecule which mimics the effect of the natural StAR-B product or at times even increases or prolongs the duration of the biological activity of said product, as compared to that induced by the natural product.
  • the mechanism may be by binding to the inner mitochondria membrane thus increasing the activity of StAR-B, by prolonging the lifetime of the StAR-B, by increasing the activity of the StAR-B on its target (transport of cholesterol), by increasing the affinity of StAR-B to moieties which it binds (such as cholesterol) etc.
  • Activators may be polypeptides, nucleic acids, carbohydrates, lipids, or derivatives thereof, or any other molecules which can bind to and activate the StAR-B product.
  • Deactivator or (“Inhibitor”) - refers to a molecule which modulates the activity of the StAR-B product in an opposite manner to that of the activator, by decreasing or shortening the duration of the biological activity of the StAR-B product. This may be done by blocking the binding of the StAR-B to cholesterol (competitive or non-competitive inhibition), by causing rapid degradation of the StAR-B, etc. by inhibiting association of the StAR-B with the inner membrane of the mitochondria, etc.
  • Deactivators may be polypeptides, nucleic acids, carbohydrates, lipids, or derivatives thereof, or any other molecules which bind to and modulate the activity of said product.
  • Treating a disease refers to administering a therapeutic substance effective to ameliorate symptoms associated with a disease, to lessen the severity or cure the disease, or to prevent the disease from occurring.
  • Detection ⁇ - refers to a method of detection of a disease. This term may refer to detection of a predisposition to a disease.
  • Probe the StAR-B nucleic acid sequence, or a sequence complementary 5 therewith, when used to detect presence of other similar sequences in a sample. The detection is carried out by identification of hybridization complexes between the probe and the assayed sequence.
  • the probe may be attached to a solid support or to a detectable label.
  • the present invention is based on the surprising finding that there exist in humans a novel homolog of the StAR having a significant homology to the C-terminus of native StAR, which is the physiologically active region of that protein.
  • the novel homolog was termed "StAR-B".
  • StAR-B was found to be localized on chromosome 15q23-q24. On this locus are also localized cytochrome
  • PCOS Stein Levental Syndrome
  • nucleic acid molecule comprising or consisting of the sequence SEQ ID NO: 1, fragments of said sequence having at least 20 nucleic acids, or a molecule comprising a sequence having at least 70%, preferably 80%, and most preferably 90% identity to SEQ ID NO:l
  • present invention further provides a protein or polypeptide comprising or consisting of an amino acid sequence encoded by any of
  • StAR-B product for example, an amino acid sequence having the sequence as depicted in SEQ ID NO: 2, fragments of the above amino acid sequence having a length of at least 10 amino acids, as well as homologs of the amino acid sequences SEQ ID NO.:2 in which one or more of the amino acid residues has been substituted (by conservative or non-conservative substitution) added, deleted, or chemically modified.
  • the present invention further provides nucleic acid molecule comprising or consisting of a sequence which encodes the above amino acid sequences, (including the fragments and analogs of the amino acid sequences). Due to the degenerative nature of the genetic code, a plurality of alternative nucleic acid sequences, beyond SEQ ID NO:l, can code for the amino acid sequence of the invention. Those alternative nucleic acid sequences which code for the same amino acid sequences codes by the sequence SEQ ID NO: 1 are also an aspect of the of the present invention.
  • the present invention further provides expression vectors and cloning vectors comprising any of the above nucleic acid sequences, as well as host cells transfected by said vectors.
  • the present invention still further provides pharmaceutical compositions comprising, as an active ingredient, said nucleic acid molecules, said expression vectors, or said protein or polypeptide.
  • compositions are suitable for the treatment of diseases and pathological conditions, which can be ameliorated or cured by raising the level of the StAR-B product.
  • diseases which are manifested by non-normal levels of steroid hormones (which can be higher or lower than normal levels).
  • the compositions are intended to restore the levels to normal levels.
  • the pharmaceutical compositions may serve as alternative regions for hormonal administration.
  • the present invention provides a nucleic acid molecule comprising or consisting of a non-coding sequence which is complementary to that of SEQ ID NO:l, or complementary to a sequence having at least 70% identity to said sequence or a fragment of said two sequences.
  • the complementary sequence may be a DNA sequence which hybridizes with the SEQ of ID NO:l or hybridizes to a portion of that sequence having a length sufficient to inhibit the transcription of the complementary sequence.
  • the complementary sequence may be a DNA sequence which can be transcribed into an mRNA being an antisense to the mRNA transcribed from SEQ ID NO:l or into an mRNA which is an antisense to a fragment of the mRNA transcribed from SEQ ID NO.:l which has a length sufficient to hybridize with the mRNA transcribed from SEQ ID NO:. 1, so as to inhibit its translation.
  • the complementary sequence may also be the mRNA or the fragment of the mRNA itself.
  • the nucleic acids of the second aspect of the invention may be used for therapeutic or diagnostic applications for example for detection of the expression of StAR-B in various tissues such as to ovary, adrenal, placenta, etc.
  • the present invention also provides expression vectors comprising any one of the above defined complementary nucleic acid sequences and host cells transfected with said nucleic acid sequences or vectors, being complementary to those specified in the first aspect of the invention.
  • the invention also provides anti-StAR-B product antibodies, namely antibodies directed against the StAR-B product which specifically bind to said StAR-B product. Said antibodies are useful both for diagnostic and therapeutic purposes.
  • said antibody may be as an active ingredient in a pharmaceutical composition as will be explained below.
  • the present invention also provides pharmaceutical compositions comprising, as an active ingredient, the nucleic acid molecules which comprise or consist of said complementary sequences, or of a vector comprising said complementary sequences.
  • the pharmaceutical composition thus provides pharmaceutical compositions comprising, as an active ingredient, said anti-StAR-B product antibodies.
  • compositions comprising said anti-StAR-B product antibodies or the nucleic acid molecule comprising said complementary sequence, are suitable for the treatment of diseases and pathological conditions where a therapeutically beneficial effect may be achieved by neutralizing the StAR-B or decreasing the amount of the StAR-B product or blocking its binding to its effector (cholesterol), for example, by the neutralizing effect of the antibodies, or by the decrease of the effect of the antisense mRNA in decreasing expression level of the StAR-B product.
  • cholesterol cholesterol
  • these diseases are manifested by non-normal level of steroid hormones in the diseased persons which may be regulated to produce normal levels by utilizing the pharmaceutical compositions of the invention.
  • the compositions of the invention may serve as alternative treatment regimes for ho ⁇ onal administration.
  • the phamiaceutical compositions are for the treatment of conditions involving PCOS and MPB.
  • the therapeuticals may be utilized for the treatment of prostate cancer.
  • the present invention provides methods for detecting the level of the transcript (mRNA) of said StAR-B product in a body fluid sample, or in a specific tissue sample (notably the ovary, placenta, adrenal), for example by use of probes comprising or consisting of said coding sequences; as well as methods for detecting levels of expression of said product in tissue, e.g. by the use of antibodies capable of specifically reacting with the above amino acid sequences.
  • Detection of the level of the expression of the StAR-B variant of the invention may be indicative of a plurality of physiological or pathological conditions.
  • the method, according to this latter aspect, for detection of a nucleic acid sequence which encodes the StAR-B product in a biological sample comprises the steps of: (a) providing a probe comprising at least one of the nucleic acid sequence defined above;
  • the probe is part of a nucleic acid chip used for detection purposes, i.e. the probe is a part of an array of probes each present in a known location on a solid support.
  • the nucleic acid sequence used in the above method may be a DNA sequence an RNA sequence, etc; it may be a coding or a sequence or a sequence complementary thereto (for respective detection of RNA transcripts or coding-DNA sequences).
  • RNA transcripts for respective detection of RNA transcripts or coding-DNA sequences.
  • Methods for detecting mutations in the region coding for the StAR-B product are also provided, which may be methods carried-out in a binary fashion, namely merely detecting whether there is any mismatches between the normal
  • StAR-B nucleic acid sequence and the one present in the sample, or carried-out by specifically detecting the nature and location of the mutation.
  • the present invention also concerns a method for detecting StAR-B product both for determining its presence ,as well as its level or alterations in its levelin a biological sample, comprising the steps of:
  • Both detection of StAR-B product and transcript, for example in urine samples, may be used to discriminate between various stages of prostate cancer. Changes in levels as compared to normal controls may be indicative of a pathological state.
  • the invention also provides a method for identifying candidate compounds capable of binding to the StAR-B product and modulating its activity (being either activators or deactivators). The method includes:
  • the activity of the amino acid which should be changed by the modulator may be for example the binding of the StAR-B product to cholesterol, effect on the transport rate of cholesterol to the inner mitochondrial membrane, effect or steroid synthesis, etc.
  • Any modulator 15 which changes such an activity has an intersecting potential, as serving as an actuator or deactivator.
  • the present invention also concerns compounds identified by the above methods described above, which compound may either be an activator of the StAR-B product or a deactivator thereof.
  • FIG. 1 is the nucleic acid sequence of the invention SEQ ID NO: 1 ;
  • Fig. 2 is the amino acid sequence of the invention SEQ ID NO: 2;
  • Fig. 3 is a comparison of the known StAR sequence and the StAR-B sequence of the invention.
  • the nucleic acid sequences of the invention include nucleic acid sequences which encode StAR-B product and fragments and analogs thereof.
  • the nucleic acid sequences may alternatively be sequences complementary to the above coding sequence, or to a region of said coding sequence. The length of the complementary sequence is sufficient to avoid the expression of the coding sequence.
  • the nucleic acid sequences may be in the form of RNA or in the form of DNA, and include messenger RNA, synthetic RNA and DNA, cDNA, and genomic DNA.
  • the DNA may be double-stranded or single-stranded, and if single-stranded may be the coding strand or the non-coding (anti-sense, complementary) strand.
  • the nucleic acid sequences may also both include dNTPs, rNTPs as well as non naturally occurring sequences.
  • the sequence may also be a part of a hybrid between an amino acid sequence and a nucleic acid sequence.
  • the nucleic acid sequence has at least 70%, preferably 80% or 90% sequence identity with the sequence identified as SEQ ID NO:l.
  • the nucleic acid sequences may include the coding sequence by itself.
  • the coding region may be in combination with additional coding sequences, such as those coding for fusion protein or signal peptides, in combination with non-coding sequences, such as introns and control elements, promoter and terminator elements or 5' and/or 3' untranslated regions, effective for expression of the coding sequence in a suitable host, and/or in a vector or host environment in which the StAR-B nucleic acid sequence is introduced as a heterologous sequence.
  • the nucleic acid sequences of the present invention may also have the product coding sequence fused in-frame to a marker sequence which allows for purification of the StAR-B product.
  • the marker sequence may be, for example, a hexahistidine tag to provide for purification of the mature polypeptide fused to the marker in the case of a bacterial host, or, the marker sequence may be a hemagglutinin (HA) tag when a mammalian host, e.g. COS-7 cells, is used.
  • the HA tag corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson, I, et al. Cell 37:767 (1984)).
  • fragments also referred to herein as oligonucleotides, typically having at least 20 bases, preferably 20-30 bases corresponding to a region of the coding-sequence nucleic acid sequence.
  • the fragments may be used as probes, primers, and when complementary also as antisense agents, and the like, according to known methods.
  • the nucleic acid sequence may be substantially a depicted in SEQ ID NO:l or fragments thereof or sequences having at least 70%, preferably 70-80%), most preferably 90% identity to the above sequence.
  • the sequence may be a sequence coding the amino acid sequence of SEQ ID NO:2, or fragments or analogs of said amino acid sequence.
  • nucleic acid sequences may be obtained by screening cDNA libraries using oligonucleotide probes which can hybridize to or PCR-amplify nucleic acid sequences which encode the StAR-B products disclosed above.
  • cDNA libraries prepared from a variety of tissues are commercially available and procedures for screening and isolating cDNA clones are well-known to those of skill in the art. Such techniques are described in, for example, Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual (2nd Edition), Cold Spring Harbor Press, Plainview, N.Y. and Ausubel FM et al. (1989) Current Protocols in Molecular Biology, John Wiley & Sons, New York, N.Y.
  • the nucleic acid sequences may be extended to obtain upstream and downstream sequences such as promoters, regulatory elements, and 5' and 3' untranslated regions (UTRs). Extension of the available transcript sequence may be performed by numerous methods known to those of skill in the art, such as PCR or primer extension (Sambrook et al, supra), or by the RACE method using, for example, the Marathon RACE kit (Clontech, Cat. # Kl 802-1).
  • genomic DNA is amplified in the presence of primer to a linker sequence and a primer specific to the known region.
  • the amplified sequences are subjected to a second round of PCR with the same linker primer and another specific primer internal to the first one.
  • Products of each round of PCR are transcribed with an appropriate RNA polymerase and sequenced using reverse transcriptase.
  • Inverse PCR can be used to amplify or extend sequences using divergent primers based on a known region (Triglia, T. et al, Nucleic Acids Res. 16:8186, (1988)).
  • the primers may be designed using OLIGO(R) 4.06 Primer Analysis Software (1992; National Biosciences Inc, Madison, Minn.), or another appropriate program, to be 22-30 nucleotides in length, to have a GC content of
  • the method uses several restriction enzymes to generate a suitable fragment in the known region of a gene.
  • the fragment is then circularized by intramolecular ligation and used as a PCR template.
  • Capture PCR (Lagerstrom, M. et al, PCR Methods Applic. 1:111-19, (1991)) is a method for PCR amplification of DNA fragments adjacent to a known sequence in human and yeast artificial chromosome DNA. Capture PCR also requires multiple restriction enzyme digestions and ligations to place an engineered double-stranded sequence into a flanking part of the DNA molecule before PCR.
  • flanking sequences Another method which may be used to retrieve flanking sequences is that of Parker, J.D., et al, Nucleic Acids Res., 19:3055-60, (1991)). Additionally, one can use PCR, nested primers and PromoterFinderTM libraries to "walk in" genomic DNA (PromoterFinderTM; Clontech, Palo Alto, CA). This process avoids the need to screen libraries and is useful in finding intron/exon junctions. Preferred libraries for screening for full length cDNAs are ones that have been size-selected to include larger cDNAs. Also, random primed libraries are preferred in that they will contain more sequences which contain the 5' and upstream regions of genes.
  • a randomly primed library may be particularly useful if an oligo d(T) library does not yield a full-length cDNA.
  • Genomic libraries are useful for extension into the 5' nontranslated regulatory region.
  • the nucleic acid sequences and oligonucleotides of the invention can also be prepared by solid-phase methods, according to known synthetic methods. Typically, fragments of up to about 100 bases are individually synthesized, then joined to form continuous sequences up to several hundred bases.
  • nucleic acid sequences specified above may be used as recombinant DNA molecules that direct the expression of StAR-B products.
  • Codons preferred by a particular prokaryotic or eukaryotic host can be selected, for example, to increase the rate of StAR-B product expression or to produce recombinant RNA transcripts having desirable properties, such as a longer half-life, than transcripts produced from naturally occurring sequence.
  • nucleic acid sequences of the present invention can be engineered in order to alter a StAR-B product coding sequence for a variety of reasons, including but not limited to, alterations which modify the cloning, processing and/or expression of the product.
  • alterations may be introduced using techniques which are well known in the art, e.g., site-directed mutagenesis, to insert new restriction sites, to alter glycosylation patterns, to change codon preference, to produce splice variants, etc.
  • the present invention also includes recombinant constructs comprising one or more of the sequences as broadly described above.
  • the constructs comprise a vector, such as a plasmid or viral vector, into which a nucleic acid sequence of the invention has been inserted, in a forward or reverse orientation.
  • the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence.
  • suitable vectors and promoters are known to those of skill in the art, and are commercially available. Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are also described in Sambrook, et al, (supra).
  • the present invention also relates to host cells which are genetically engineered with vectors of the invention, and the production of the product of the invention by recombinant techniques.
  • Host cells are genetically engineered (i.e., transduced, transformed or transfected) with the vectors of this invention which may be, for example, a cloning vector or an expression vector.
  • the vector may be, for example, in the form of a plasmid, a viral particle, a phage, etc.
  • the engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants or amplifying the expression of the StAR-B nucleic acid sequence.
  • the culture conditions are those previously used with the host cell selected for expression, and will be apparent to those skilled in the art.
  • the nucleic acid sequences of the present invention may be included in any one of a variety of expression vectors for expressing a product.
  • Such vectors include chromosomal, nonchromosomal and synthetic DNA sequences, e.g., derivatives of SV40; bacterial plasmids; phage DNA; baculovirus; yeast plasmids; vectors derived from combinations of plasmids and phage DNA, viral DNA such as vaccinia, adenovirus, fowl pox virus, and pseudorabies.
  • any other vector may be used as long as it is replicable and viable in the host.
  • the appropriate DNA sequence may be inserted into the vector by a variety of procedures.
  • the DNA sequence is inserted into an appropriate restriction endonuclease site(s) by procedures known in the art. Such procedures and related sub-cloning procedures are deemed to be within the scope of those skilled in the art.
  • the DNA sequence in the expression vector is operatively linked to an appropriate transcription control sequence (promoter) to direct mRNA synthesis.
  • promoters include: LTR or SV40 promoter, the E.coli lac or trp promoter, the phage lambda PL promoter, and other promoters known to control expression of genes in prokaryotic or eukaryotic cells or their viruses.
  • the expression vector also contains a ribosome binding site for translation initiation, and a transcription terminator.
  • the vector may also include appropriate sequences for amplifying expression.
  • the expression vectors preferably contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampicillin resistance in E.coli.
  • the vector containing the appropriate DNA sequence as described above, as well as an appropriate promoter or control sequence, may be employed to transform an appropriate host to permit the host to express the protein.
  • Examples of appropriate expression hosts include: bacterial cells, such as E.coli, Streptomyces, Salmonella typhimurium; fungal cells, such as yeast; insect cells such as Drosophila and Spodoptera Sf9; animal cells such as CHO, COS, HEK 293 or Bowes melanoma; adenoviruses; plant cells, etc.
  • bacterial cells such as E.coli, Streptomyces, Salmonella typhimurium
  • fungal cells such as yeast
  • insect cells such as Drosophila and Spodoptera Sf9
  • animal cells such as CHO, COS, HEK 293 or Bowes melanoma
  • adenoviruses adenoviruses
  • plant cells etc.
  • the selection of an appropriate host is deemed to be within the scope of those skilled in the art from the teachings herein.
  • the invention is not limited by the host cells employed. In bacterial systems, a number of expression vectors may be selected depending upon the use intended
  • vectors which direct high level expression of fusion proteins that are readily purified may be desirable.
  • Such vectors include, but are not limited to, multifunctional E.coli cloning and expression vectors such as Bluescript( ) (Stratagene), in which the StAR-B polypeptide coding sequence may be ligated into the vector in-frame with sequences for the amino-terminal Met and the subsequent 7 residues of beta-galactosidase so that a hybrid protein is produced; pIN vectors (Van Heeke & Schuster J. Biol Chem. 264:5503-5509, (1989)); pET vectors (Novagen, Madison WI); and the like.
  • Bluescript( ) Stratagene
  • pIN vectors Vector Heeke & Schuster J. Biol Chem. 264:5503-5509, (1989)
  • pET vectors Novagen, Madison WI
  • yeast Saccharomyces cerevisiae a number of vectors containing constitutive or inducible promoters such as alpha factor, alcohol oxidase and PGH may be used.
  • constitutive or inducible promoters such as alpha factor, alcohol oxidase and PGH.
  • StAR-B product may also be expressed in an insect system.
  • Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes in Spodoptera frugiperda cells or in Trichoplusia larvae.
  • the StAR-B product coding sequence may be cloned into a nonessential region of the virus, such as the polyhedrin gene, and placed under control of the polyhedrin promoter. Successful insertion of StAR-B coding sequence will render the polyhedrin gene inactive and produce recombinant virus lacking coat protein coat. The recombinant viruses are then used to infect S.
  • a number of viral-based expression systems may be utilized.
  • a StAR-B product coding sequence may be ligated into an adenovirus transcription/translation complex consisting of the late promoter and tripartite leader sequence. Insertion in a nonessential El or E3 region of the viral genome will result in a viable virus capable of expressing StAR-B protein in infected host cells (Logan and Shenk. Proc. Natl Acad. Sci. 81:3655-59, (1984).
  • transcription enhancers such as the Rous sarcoma virus (RSV) enhancer, may be used to increase expression in mammalian host cells.
  • RSV Rous sarcoma virus
  • Specific initiation signals may also be required for efficient translation of a StAR-B protein coding sequence. These signals include the ATG initiation codon and adjacent sequences. In cases where StAR-B product coding sequence, its initiation codon and upstream sequences are inserted into the appropriate expression vector, no additional translational control signals may be needed. However, in cases where only coding sequence, or a portion thereof, is inserted, exogenous transcriptional control signals including the ATG initiation codon must be provided. Furthermore, the initiation codon must be in the correct reading frame to ensure transcription of the entire insert. Exogenous transcriptional elements and initiation codons can be of various origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of enhancers appropriate to the cell system in use (Scharf, D. et al, (1994) Results Probl Cell Differ., 20:125-62, (1994); Bittner et al., Methods in Enzvmol 153:516-544, (1987)).
  • the present invention relates to host cells containing the above-described constructs.
  • the host cell can be a higher eukaryotic cell, such as a mammalian cell, or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell.
  • Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DEAE-Dextran mediated transfection, or electroporation (Davis, L., Dibner, M., and Battey, I. (1986) Basic Methods in Molecular Biology).
  • Cell-free translation systems can also be employed to produce polypeptides using RNAs derived from the DNA constructs of the present invention.
  • a host cell strain may be chosen for its ability to modulate the expression of the inserted sequences or to process the expressed protein in the desired fashion.
  • modifications of the protein include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation and
  • Post-translational processing which cleaves a "pre-pro" form of the protein may also be important for correct insertion, folding and/or function.
  • Different host cells such as CHO, HeLa, MDCK, 293, WI38, etc. have specific cellular machinery and characteristic mechanisms for such post-translational activities and may be chosen to ensure the correct modification and processing of i o the introduced, foreign protein.
  • cell lines which stably express StAR-B product may be transformed using expression vectors which contain viral origins of replication or endogenous expression elements and a selectable marker gene.
  • cells may be allowed to grow for 1-2 days in an enriched media before they are switched to selective media.
  • the purpose of the selectable marker is to confer resistance to selection, and its presence allows growth and recovery of cells which successfully express the introduced sequences. Resistant clumps of stably transformed cells can be
  • Any number of selection systems may be used to recover transformed cell lines. These include, but are not limited to, the herpes simplex virus thymidine kinase (Wigler M., et al, Cell 11:223-32, (1977)) and adenine phosphoribosyltransferase (Lowy I., et al., Cell 22:817-23, (1980)) genes which are not limited to recover transformed cell lines. These include, but are not limited to, the herpes simplex virus thymidine kinase (Wigler M., et al, Cell 11:223-32, (1977)) and adenine phosphoribosyltransferase (Lowy I., et al., Cell 22:817-23, (1980)) genes which
  • 25 can be employed in tk- or aprt- cells, respectively.
  • antimetabolite, antibiotic or herbicide resistance can be used as the basis for selection; for example, dhfr which confers resistance to methotrexate (Wigler M., et al, Proc. Natl Acad. Sci. TT3567-70, (1980)); npt, which confers resistance to the aminoglycosides neomycin and G-418 (Colbere-Garapin, F. et al, J.
  • als ox pat which confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively (Murry, supra). Additional selectable genes have been described, for example, trpB, which allows cells to utilize indole in place of tryptophan, or hisD, which allows cells to utilize histinol in place of histidine (Hartman S.C. and R.C. Mulligan, Proc. Natl Acad. Sci. 85:8047-51, (1988)).
  • Host cells transformed with a nucleotide sequence encoding StAR-B product may be cultured under conditions suitable for the expression and recovery of the encoded protein from cell culture.
  • the product produced by a recombinant cell may be secreted or contained intracellularly depending on the sequence and/or the vector used.
  • expression vectors containing nucleic acid sequences encoding StAR-B product can be designed with signal sequences which direct secretion of StAR-B product through a prokaryotic or eukaryotic cell membrane.
  • StAR-B product may also be expressed as a recombinant protein with one or more additional polypeptide domains added to facilitate protein purification.
  • purification facilitating domains include, but are not limited to, metal chelating peptides such as histidine-tryptophan modules that allow purification on immobilized metals, protein A domains that allow purification on immobilized immuno globulin, and the domain utilized in the FLAGS extension/affinity purification system (Immunex Corp, Seattle, Wash.).
  • the inclusion of a protease-cleavable polypeptide linker sequence between the purification domain and StAR-B protein is useful to facilitate purification.
  • One such expression vector provides for expression of a fusion protein compromising a StAR-B polypeptide fused to a polyhistidine region separated by an enterokinase cleavage site.
  • the histidine residues facilitate purification on IMIAC (immobilized metal ion affinity chromatography, as described in Porath, et al., Protein Expression and Purification, 3:263-281, (1992)) while the enterokinase cleavage site provides a means for isolating StAR-B polypeptide from the fusion protein.
  • pGEX vectors Promega, Madison, Wis.
  • GST glutathione S-transferase
  • fusion proteins are soluble and can easily be purified from lysed cells by adsorption to ligand-agarose beads (e.g., glutathione-agarose in the case of GST- fusions) followed by elution in the presence of free ligand.
  • ligand-agarose beads e.g., glutathione-agarose in the case of GST- fusions
  • the selected promoter is induced by appropriate means (e.g., temperature shift or chemical induction) and cells are cultured for an additional period.
  • Cells are typically harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification.
  • Microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents, or other methods, which are well know to those skilled in the art.
  • the StAR-B products can be recovered and purified from recombinant cell cultures by any of a number of methods well known in the art, including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography, and lectin chromatography. Protein refolding steps can be used, as necessary, in completing configuration of the mature protein. Finally, high performance liquid chromatography (HPLC) can be employed for final purification steps.
  • HPLC high performance liquid chromatography
  • nucleic acid sequences of the present invention may be used for a variety of diagnostic purposes.
  • the nucleic acid sequences may be used to detect and quantitate expression of StAR-B in patient's cells, e.g. biopsied tissues, by detecting the presence of mRNA coding for StAR-B product.
  • the assay may be used to detect soluble StAR-B in the serum or bloodThis assay typically involves obtaining total mRNA from the tissue or serum and contacting the mRNA with a nucleic acid probe.
  • the probe is a nucleic acid molecule of at least 20 nucleotides, preferably 20-30 nucleotides, capable of specifically hybridizing with a sequence included within the sequence of a nucleic acid molecule encoding StAR-B under hybridizing conditions, detecting the presence of mRNA hybridized to the probe, and thereby detecting the expression of StAR-B.
  • This assay can be used to distinguish between absence, presence, and excess expression of StAR-B product and to monitor levels of StAR-B expression during therapeutic intervention.
  • the invention also contemplates the use of the nucleic acid sequences as a diagnostic for diseases resulting from inherited defective StAR-B sequences, or diseases in which the purpose of the amount of the known StAR to the novel StAR variant of the invention is altered. These sequences can be detected by comparing the sequences of the defective (i.e., mutant) StAR-B coding region with that of a normal coding region. Association of the sequence coding for mutant StAR-B product with abnormal StAR-B product activity may be verified.
  • sequences encoding mutant StAR-B products can be inserted into a suitable vector for expression in a functional assay system (e.g., colorimetric assay, complementation experiments in a StAR-B protein deficient strain of HEK293 cells) as yet another means to verify or identify mutations. Once mutant genes have been identified, one can then screen populations of interest for carriers of the mutant gene.
  • a functional assay system e.g., colorimetric assay, complementation experiments in a StAR-B protein deficient strain of HEK293 cells
  • Nucleic acids used for diagnosis may be obtained from a patient's cells, including but not limited to such as from blood, urine, saliva, placenta, tissue biopsy and autopsy material. Genomic DNA may be used directly for detection or may be amplified enzymatically by using PCR (Saiki, et al, Nature 324:163-166. (1986)) prior to analysis. RNA or cDNA may also be used for the same purpose. As an example, PCR primers complementary to the nucleic acid of the present invention can be used to identify and analyze mutations in the gene of the present invention.
  • Deletions and insertions can be detected by a change in size of the amplified product in comparison to the normal genotype.
  • Point mutations can be identified by hybridizing amplified DNA to radiolabeled RNA of the invention or alternatively, radiolabeled antisense DNA sequences of the invention. Sequence changes at specific locations may also be revealed by nuclease protection assays, such RNase and SI protection or the chemical cleavage method (e.g. Cotton, et alProc. Natl. Acad. Sci. USA, 85:4397-4401, (1985)), or by differences in melting temperatures. "Molecular beacons" (Kostrikis L.G.
  • hairpin-shaped, single-stranded synthetic oligo- nucleotides containing probe sequences which are complementary to the nucleic acid of the present invention may also be used to detect point mutations or other sequence changes as well as monitor expression levels of StAR-B product. Such diagnostics would be particularly useful for prenatal testing.
  • Another method for detecting mutations uses two DNA probes which are designed to hybridize to adjacent regions of a target, with abutting bases, where the region of known or suspected mutation(s) is at or near the abutting bases.
  • the two probes may be joined at the abutting bases, e.g., in the presence of a ligase enzyme, but only if both probes are correctly base paired in the region of probe junction.
  • the presence or absence of mutations is then detectable by the presence or absence of ligated probe.
  • oligonucleotide array methods based on sequencing by hybridization (SBH), as described, for example, in U.S. Patent No. 5,547,839.
  • SBH sequencing by hybridization
  • the DNA target analyte is hybridized with an array of oligonucleotides formed on a microchip.
  • the sequence of the target can then be "read" from the pattern of target binding to the array.
  • the nucleic acid sequences of the present invention are also valuable for chromosome identification.
  • the sequence is specifically targeted to and can hybridize with a particular location on an individual human chromosome.
  • Few chromosome marking reagents based on actual sequence data (repeat polymorphisms) are presently available for marking chromosomal location.
  • the mapping of DNAs to chromosomes according to the present invention is an important first step in correlating those sequences with genes associated with disease.
  • sequences can be mapped to chromosomes by preparing PCR primers (preferably 20-30 bp) from the StAR-B cDNA. Computer analysis of the 3' untranslated region is used to rapidly select primers that do not span more than one exon in the genomic DNA, which would complicate the amplification process. These primers are then used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene corresponding to the primer will yield an amplified fragment.
  • mapping of somatic cell hybrids or using instead radiation hybrids are rapid procedures for assigning a particular DNA to a particular chromosome.
  • sublocalization can be achieved with panels of fragments from specific chromosomes or pools of large genomic clones in an analogous manner.
  • Other mapping strategies that can similarly be used to map to its chromosome include in situ hybridization, prescreening with labeled flow-sorted chromosomes and preselection by hybridization to construct chromosome specific-cDNA libraries.
  • Fluorescence in situ hybridization of a cDNA clone to a metaphase chromosomal spread can be used to provide a precise chromosomal location in one step.
  • This technique can be used with cDNA as short as 50 or 60 bases.
  • Verma et al Human Chromosomes: a Manual of Basic Techniques, (1988) Pergamon Press, New York.
  • a sequence Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data.
  • genetic map data are found, for example, in the OMIM database (Center for Medical Genetics, Johns Hopkins University, Baltimore, MD and National Center for Biotechnology Information, National Library of Medicine, Bethesda, MD).
  • the OMIM gene map presents the cytogenetic map location of disease genes and other expressed genes.
  • the OMIM database provides information on diseases associated with the chromosomal location. Such associations include the results of linkage analysis mapped to this interval, and the correlation of translocations and other chromosomal aberrations in this area with the advent of polygenic diseases, such as cancer, in general and prostate cancer in particular.
  • Therapeutic applications of nucleic acid sequences include the results of linkage analysis mapped to this interval, and the correlation of translocations and other chromosomal aberrations in this area with the advent of polygenic diseases, such as cancer, in general
  • Nucleic acid sequences of the invention may also be used for therapeutic purposes.
  • expression of StAR-B product may be modulated through antisense technology, which controls gene expression through hybridization of complementary nucleic acid sequences, i.e. antisense DNA or RNA, to the control, 5' or regulatory regions of the gene encoding StAR-B product.
  • the 5' coding portion of the nucleic acid sequence sequence which codes for the product of the present invention is used to design an antisense oligonucleotide of from about 10 to 40 base pairs in length. Oligonucleotides derived from the transcription Start site, e.g.
  • An antisense DNA oligonucleotide is designed to be complementary to a region of the nucleic acid sequence involved in transcription (Lee et al, Nucl Acids, Res., 6:3073, (1979); Cooney et al., Science 241:456. (1988); and Dervan et al, Science 251:1360, (1991)), thereby preventing transcription and the production of the StAR-B products.
  • An antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into the StAR-B products (Okano J. Neurochem. 56:560, (1991)).
  • the antisense constructs can be delivered to cells by procedures known in the art such that the antisense RNA or DNA may be expressed in vivo.
  • the antisense may be antisense mRNA or DNA sequence capable of coding such antisense mRNA.
  • the antisense mRNA or the DNA coding thereof can be complementary to the full sequence of nucleic acid sequences coding to the StAR-B protein or to a fragment of such a sequence which is sufficient to inhibit production of a protein product.
  • expression of StAR-B product may be increased by providing coding sequences for coding for said product under the control of suitable control elements ending its expression in the desired host.
  • compositions comprise a therapeutically effective amount of the compound, and a pharmaceutically acceptable carrier or excipient.
  • a carrier includes but is not limited to saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof.
  • the formulation should suit the mode of administration.
  • Cells from a patient may be engineered with a nucleic acid sequence (DNA or RNA) encoding a polypeptide ex vivo, with the engineered cells then being provided to a patient to be treated with the polypeptide.
  • DNA or RNA nucleic acid sequence
  • Such methods are well-known in the art.
  • cells may be engineered by procedures known in the art by use of a retroviral particle containing RNA encoding a polypeptide of the present invention.
  • cells may be engineered in vivo for expression of a polypeptide in vivo by procedures known in the art.
  • a producer cell for producing a retroviral particle containing RNA encoding the polypeptide of the present invention may be administered to a patient for engineering cells in vivo and expression of the polypeptide in vivo.
  • the expression vehicle for engineering cells may be other than a retrovirus, for example, an adenovirus which may be used to engineer cells in vivo after combination with a suitable delivery vehicle.
  • Retroviruses from which the retroviral plasmid vectors mentioned above may be derived include, but are not limited to, Moloney Murine Leukemia Virus, spleen necrosis virus, retroviruses ' such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, gibbon ape leukemia virus, human immunodeficiency virus, adenovirus, Myeloproliferative Sarcoma Virus, and mammary tumor virus.
  • the retroviral plasmid vector is employed to transduce packaging cell lines to form producer cell lines.
  • packaging cells which may be transfected include, but are not limited to, the PE501, PA317, psi-2, psi-AM, PA12, T19-14X, VT-19-17-H2, psi-CRE, psi-CRIP, GP+E-86, GP+envAml2, and DAN cell lines as described in Miller (Human Gene Therapy, Vol. 1, pg. 5-14, (1990)).
  • the vector may transduce the packaging cells through any means known in the art. Such means include, but are not limited to, electroporation, the use of liposomes, and CaP0 4 precipitation.
  • the retroviral plasmid vector may be encapsulated into a liposome, or coupled to a lipid, and then administered to a host.
  • the producer cell line generates infectious retroviral vector particles which include the nucleic acid sequence(s) encoding the polypeptides.
  • retroviral vector particles then may be employed, to transduce eukaryotic cells, either in vitro or in vivo.
  • the transduced eukaryotic cells will express the nucleic acid sequence(s) encoding the polypeptide.
  • Eukaryotic cells which may be transduced include, but are not limited to, embryonic stem cells, embryonic carcinoma cells, as well as hematopoietic stem cells, hepatocytes, fibroblasts, myoblasts, keratinocytes, endothelial cells, and bronchial epithelial cells.
  • the genes introduced into cells may be placed under the control of inducible promoters, such as the radiation-inducible Egr-1 promoter, (Maceri, H.J., et al, Cancer Res., 56(19):431 1 (1996)), to stimulate StAR-B production or antisense inhibition in response to radiation, eg., radiation therapy for treating tumors.
  • inducible promoters such as the radiation-inducible Egr-1 promoter, (Maceri, H.J., et al, Cancer Res., 56(19):431 1 (1996)
  • the substantially purified StAR-B product of the invention has been defined above as the product coded from the nucleic acid sequence of the 5 invention.
  • the amino acid sequence is an amino acid sequence having at least 70%, preferably at least 80% or 90% identity to the sequence identified as SEQ ID NO:2.
  • the protein or polypeptide may be in mature and/or modified form, also as defined above. Also contemplated are protein fragments having at least 10 contiguous amino acid residues, preferably at least 10-20 residues, l o derived from the StAR-B product.
  • sequence variations are preferably those that are considered conserved substitutions, as defined above.
  • a protein with a sequence having at least 80%> sequence identity with the protein identified as SEQ ID NO:2, preferably by utilizing conserved substitutions as defined above is also part
  • the protein has or contains the sequence identified SEQ ID NO:2.
  • the StAR-B product may be (i) one in which one or more of the amino acid residues in a sequence listed above are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue), or (ii) one in which one or more of the amino acid residues
  • the StAR-B product includes a substituent group, or (iii) one in which the StAR-B product is fused with another compound, such as a compound to increase the half-life of the protein (for example, polyethylene glycol (PEG)), or a moiety which serves as targeting means to direct the protein to its target tissue or target cell population (such as an antibody), or (iv) one in which additional amino acids are fused to the
  • a compound to increase the half-life of the protein for example, polyethylene glycol (PEG)
  • PEG polyethylene glycol
  • a moiety which serves as targeting means to direct the protein to its target tissue or target cell population such as an antibody
  • fragments and portions of StAR-B product may be produced by direct peptide synthesis using solid-phase techniques 5 (cf. Stewart et al., (1969) Solid-Phase Peptide Synthesis, WH Freeman Co, San Francisco; Merrifield J., J. Am. Chem. Soc, 85:2149-2154, (1963)).
  • In vitro peptide synthesis may be performed using manual techniques or by automation. Automated synthesis may be achieved, for example, using Applied Biosystems 43 1 A Peptide Synthesizer (Perkin Elmer, Foster City, Calif.) in accordance with l o the instructions provided by the manufacturer.
  • Fragments of StAR-B product may be chemically synthesized separately and combined using chemical methods to produce the full length molecule.
  • the StAR-B product of the invention is generally useful in treating diseases and disorders which are characterized by a lower than normal level of StAR-B expression, and or diseases which can be cured or ameliorated by raising the level of the StAR-B product, even if the level is normal.
  • StAR-B products or fragments may be administered by any of a number of
  • the product-containing compositions may be administered alone or in combination with other agents, such as stabilizing compounds, and/or in combination with other pharmaceutical agents such as drugs or hormones.
  • StAR-B product-containing compositions may be administered by a number of routes including, but not limited to oral, intravenous, intramuscular, transdermal, subcutaneous, topical, sublingual, or rectal means as well as by nasal application. StAR-B product-containing compositions may also be administered via liposomes. Such administration routes and appropriate formulations are generally known to those of skill in the art.
  • the product can be given via intravenous or intraperitoneal injection. Similarly, the product may be injected to other localized regions of the body. The product may also be administered via nasal insufflation. Enteral administration is also possible. For such administration, the product should be formulated into an appropriate capsule or elixir for oral administration, or into a suppository for rectal administration.
  • a therapeutic composition for use in the treatment method can include the product in a sterile injectable solution, the polypeptide in an oral delivery vehicle, the product in an aerosol suitable for nasal administration, or the product in a nebulized form, all prepared according to well known methods.
  • Such compositions comprise a therapeutically effective amount of the compound, and a phamiaceutically acceptable carrier or excipient.
  • a carrier includes but is not limited to saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof.
  • the product of the invention may also be used to modulate endothelial differentiation and proliferation as well as to modulate apoptosis either ex vivo or in vitro, for example, in cell cultures.
  • Example III Screening methods for activators and deactivators (inhibitors)
  • the present invention also includes an assay for identifying molecules, such as synthetic drugs, antibodies, peptides, or other molecules, which have a modulating effect on the activity of the StAR-B product, e.g. activators or deactivators of the StAR-B product of the present invention.
  • an assay comprises the steps of providing an StAR-B product encoded by the nucleic acid sequences of the present invention, contacting the StAR-B protein with one or more candidate molecules to determine the candidate molecules modulating effect on the activity of the StAR-B product, and selecting from the molecules a candidate's molecule capable of modulating StAR-B product physiological activity.
  • StAR-B product its catalytic or immunogenic fragments or oligopeptides thereof, can be used for screening therapeutic compounds in any of a variety of drug screening techniques.
  • the fragment employed in such a test may be free in solution, affixed to a solid support, bome on a cell membrane or located intracellularly.
  • the formation of binding complexes, between StAR-B product and the agent being tested, may be measured.
  • the activator or deactivator may work by serving as agonist or antagonist, respectively, of the StAR-B receptor and their effect may be determined in connection with the receptor.
  • Antibodies to the StAR-B product may also be used in screening assays according to methods well known in the art.
  • a "sandwich" assay may be perfo ⁇ ned, in which an anti-StAR-B antibody is affixed to a solid surface such as a microtiter plate and StAR-B product is added.
  • Such an assay can be used to capture compounds which bind to the StAR-B product.
  • such an assay may be used to measure the ability of compounds to influence with the binding of StAR-B product to the o StAR-B receptor [ I and then select those compounds which effect the binding.
  • the purified StAR-B product is 5 used to produce anti-StAR-B antibodies which have diagnostic and therapeutic uses related to the activity, distribution, and expression of the StAR-B product, in particular therapeutic applications in inhibiting the effect of the StAR-B cholesterol in the transport.
  • Antibodies to StAR-B product may be generated by methods well known 0 in the art. Such antibodies may include, but are not limited to, polyclonal, monoclonal, chimeric, humanized, single chain, Fab fragments and fragments produced by an Fab expression library. Antibodies, i.e., those which inhibit dimer formation, are especially preferred for therapeutic use.
  • a fragment StAR-B product for antibody induction does not require 5 biological activity but have to feature immunological activity; however, the protein fragment or oligopeptide must be antigenic.
  • Peptides used to induce specific antibodies may have an amino acid sequence consisting of at least five amino acids, preferably at least 10 amino acids of the sequences specified in SEQ ID NO: 2. Preferably they should mimic a portion of the amino acid sequence of the natural protein and may contain the entire amino acid sequence of a small, naturally occurring molecule. Short stretches of StAR-B protein amino acids may be fused with those of another protein such as keyhole limpet hemocyanin and antibody produced against the chimeric molecule. Procedures well known in the art can be used for the production of antibodies to StAR-B product.
  • various hosts including goats, rabbits, rats, mice, etc may be immunized by injection with StAR-B product or any portion, fragment or oligopeptide which retains immunogenic properties.
  • various adjuvants may be used to increase immunological response.
  • adjuvants include but are not limited to Freund's, mineral gels such as aluminum hydroxide, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol.
  • BCG Bacilli Calmette-Guerin
  • Corynebacterium parvum are potentially useful human adjuvants.
  • Monoclonal antibodies to StAR-B protein may be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include but are not limited to the hybridoma technique originally described by Koehler and Milstein (Nature 256:495-497, (1975)), the human B-cell hybridoma technique (Kosbor et al, Immunol. Today 4:72, (1983); Cote et al, Proc. Natl Acad. Sci. 80:2026-2030, (1983)) and the EBV-hybridoma technique (Cole, et al, Mol Cell Biol. 62:109-120, (1984)).
  • Antibodies may also be produced by inducing in vivo production in the lymphocyte population or by screening recombinant immunoglobulin libraries or 5 panels of highly specific binding reagents as disclosed in Orlandi et al. (Proc.
  • Antibody fragments which contain specific binding sites for StAR-B protein may also be generated.
  • fragments include, but are not 10 limited to, the F(ab') 2 fragments which can be produced by pepsin digestion of the antibody molecule and the Fab fragments which can be generated by reducing the disulfide bridges of the F(ab') 2 fragments.
  • Fab expression libraries may be constructed to allow rapid and easy identification of monoclonal
  • Antibodies which specifically bind StAR-B product are useful for the diagnosis of conditions or diseases characterized by over or under expression of StAR-B. Alternatively, such antibodies may be used in assays to monitor patients being treated with StAR-B product, its activators, or its deactivators. Diagnostic assays for StAR-B protein include methods utilizing the antibody and a label to detect StAR-B product in human body fluids or extracts of cells or tissues.
  • the products and antibodies of the present invention may be used with or without modification. Frequently, the proteins and antibodies will be labeled by joining them, either covalently or noncovalently, with a reporter molecule. A wide variety of reporter molecules are known in the art.
  • a variety of protocols for measuring StAR-B product, using either polyclonal or monoclonal antibodies specific for the respective protein are known in the art. Examples include enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), and fluorescent activated cell sorting (FACS). As noted above, a two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering epitopes on StAR-B product is preferred, but a competitive binding assay may be employed. These assays are described, among other places, in Maddox, et al. (supra). Such protocols provide a basis for diagnosing altered or abnormal levels of StAR-B product expression.
  • ELISA enzyme-linked immunosorbent assay
  • RIA radioimmunoassay
  • FACS fluorescent activated cell sorting
  • Normal or standard values for StAR-B product expression are established by combining body or cell extracts taken from normal subjects, preferably human, with antibody to StAR-B product under conditions suitable for complex formation which are well known in the art.
  • the amount of standard complex formation may be quantified by various methods, preferably by photometric methods.
  • standard values obtained from normal samples may be compared with values obtained from samples from subjects potentially affected by disease. Deviation between standard and subject values establishes the presence of disease state.
  • the antibody assays are useful to determine the level of StAR-B present in a body fluid sample, in order to determine whether it is being overexpressed or underexpressed in the tissue, or as an indication of how StAR-B levels are responding to drug treatment.
  • the antibodies may have a therapeutical utility in blocking or decreasing the activity of the StAR-B product in pathological conditions where beneficial effect can be achieved by such a decrease.
  • the antibody employed is preferably a humanized monoclonal antibody, or a human Mab produced by known globulin-gene library methods.
  • the antibody is administered typically as a sterile solution by IV injection, although other parenteral routes may be suitable.
  • the antibody is administered in an amount between about 1-15 mg/kg body weight of the subject. Treatment is continued, e.g., with dosing every 1-7 days, until a therapeutic improvement is seen.

Landscapes

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

Abstract

L'invention concerne de nouvelles séquences d'acides nucléiques et d'acides aminés qui constituent des homologues de la protéine régulatrice de réponse stéroïdogène aiguë. L'invention concerne en outre des vecteurs d'expression contenant ces séquences et des cellules hôtes transfectées à l'aide des vecteurs. L'invention concerne également des compositions pharmaceutiques.
PCT/IL2000/000252 1999-05-03 2000-05-03 Homologues de star WO2000066728A1 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU43104/00A AU4310400A (en) 1999-05-03 2000-05-03 Star homologues
EP00922827A EP1175492A1 (fr) 1999-05-03 2000-05-03 Homologues de star

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IL129734 1999-05-03
IL12973499A IL129734A0 (en) 1999-05-03 1999-05-03 Novel nucleic acid and amino acid sequences

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US09959656 A-371-Of-International 2002-04-30
US10/147,170 Continuation-In-Part US20030105049A1 (en) 1999-05-03 2002-05-15 StAR homologues

Publications (1)

Publication Number Publication Date
WO2000066728A1 true WO2000066728A1 (fr) 2000-11-09

Family

ID=11072759

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2000/000252 WO2000066728A1 (fr) 1999-05-03 2000-05-03 Homologues de star

Country Status (4)

Country Link
EP (1) EP1175492A1 (fr)
AU (1) AU4310400A (fr)
IL (1) IL129734A0 (fr)
WO (1) WO2000066728A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001031342A2 (fr) * 1999-10-26 2001-05-03 Tularik Inc. DIAGNOSTIC ET TRAITEMENT DU CANCER UTILISANT DES POLYPEPTIDES ET DES POLYNUCLEOTIDES StAR
WO2003104811A2 (fr) * 2002-06-10 2003-12-18 Evotec Neurosciences Gmbh Utilisation diagnostique et therapeutique de la proteine regulatrice de reponse steroidogene aigue pour traiter des maladies neurodegeneratives
US8889398B2 (en) 2006-05-19 2014-11-18 Viroblock Sa Composition for inactivating an enveloped virus

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113025700A (zh) * 2021-03-05 2021-06-25 中国农业科学院北京畜牧兽医研究所 Star及其调控基因的用途

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996029338A1 (fr) * 1995-03-23 1996-09-26 The Regents Of The University Of California Identification de mutations de genes associees a l'hyperplasie surrenale lipoide congenitale
WO1998020165A2 (fr) * 1996-11-06 1998-05-14 Whitehead Institute For Biomedical Research Marqueurs bialleliques
US5872230A (en) * 1995-11-04 1999-02-16 Texas Tech University Health Sciences Center Compositions and methods for regulation of steroidogenesis

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996029338A1 (fr) * 1995-03-23 1996-09-26 The Regents Of The University Of California Identification de mutations de genes associees a l'hyperplasie surrenale lipoide congenitale
US5872230A (en) * 1995-11-04 1999-02-16 Texas Tech University Health Sciences Center Compositions and methods for regulation of steroidogenesis
WO1998020165A2 (fr) * 1996-11-06 1998-05-14 Whitehead Institute For Biomedical Research Marqueurs bialleliques

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
DATABASE STRAND GenBank; 15 January 2000 (2000-01-15), KOEHRER K ET AL.: "Homo sapiens mRNA; cDNA DKFZp434G2416; partial cds", XP002145552 *
DATABASE STRAND GenBank; 21 August 1997 (1997-08-21), STRAUSBERG R: "nc49d1.r1 NCI_CGAP_Pr3 Homo sapiens cDNA clone IMAGE:1011479, mRNA sequence", XP002145551 *
DATABASE STRAND GenBank; 3 May 1996 (1996-05-03), HILLIER L ET AL.: "zb40d12.r1 Soares_parathyroid_tumor_NbHPA Homo sapiens cDNA clone IMAGE:306071 5', mRNA sequence", XP002145550 *
DATABASE STRAND GenBank; 9 May 1997 (1997-05-09), HILLIER L ET AL.: "zk02a01.r1 Soares_pregnant_uterus_NbHPU Homo sapiens cDNA clone IMAGE:469320 5', mRNA sequence", XP002145549 *
KALLEN C B ET AL.: "Unveiling the mechanism of action and regulation of the steroidogenic acute regulatory protein", MOLECULAR AND CELLULAR ENDOCRINOLOGY, vol. 145, no. 1-2, 25 October 1998 (1998-10-25), pages 39 - 45, XP000900751 *
STOCCO D M: "Intramitochondrial cholesterol transfer", BIOCHIMICA ET BIOPHYSICA ACTA, vol. 1486, no. 1, 26 June 2000 (2000-06-26), pages 184 - 197, XP000937443 *
SUGAWARA T ET AL.: "Human steroidogenic acute regulatory protein: functional activity in COS-1 cells, tissue-specific expression, and mapping of the structural gene to 8p11.2 and a pseudogene to chromosome 13.", PROC. NAT'L.ACAD. SCI. USA, vol. 92, no. 11, 23 May 1995 (1995-05-23), pages 4778 - 4782, XP002145548 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001031342A2 (fr) * 1999-10-26 2001-05-03 Tularik Inc. DIAGNOSTIC ET TRAITEMENT DU CANCER UTILISANT DES POLYPEPTIDES ET DES POLYNUCLEOTIDES StAR
WO2001031342A3 (fr) * 1999-10-26 2002-01-24 Tularik Inc DIAGNOSTIC ET TRAITEMENT DU CANCER UTILISANT DES POLYPEPTIDES ET DES POLYNUCLEOTIDES StAR
WO2003104811A2 (fr) * 2002-06-10 2003-12-18 Evotec Neurosciences Gmbh Utilisation diagnostique et therapeutique de la proteine regulatrice de reponse steroidogene aigue pour traiter des maladies neurodegeneratives
WO2003104811A3 (fr) * 2002-06-10 2004-10-14 Evotec Neurosciences Gmbh Utilisation diagnostique et therapeutique de la proteine regulatrice de reponse steroidogene aigue pour traiter des maladies neurodegeneratives
US8889398B2 (en) 2006-05-19 2014-11-18 Viroblock Sa Composition for inactivating an enveloped virus

Also Published As

Publication number Publication date
IL129734A0 (en) 2000-02-29
EP1175492A1 (fr) 2002-01-30
AU4310400A (en) 2000-11-17

Similar Documents

Publication Publication Date Title
US20070086997A1 (en) Compositions, reagents and kits for and methods of diagnosing, monitoring and treating obesity and/or diabetes
WO1999067382A2 (fr) Sequences d'un facteur de croissance apparente a l'angiopoietine
WO2000049158A2 (fr) Nouvel acide nucleique et sequences d'acides amines
US20020061525A1 (en) Sequences of trail variants
US20040170975A1 (en) Variant of TNF-receptor
US20050244877A1 (en) Splice variants of CD40-receptor
US6783954B2 (en) VEGF nucleic acid and amino acid sequences
WO2002006315A2 (fr) Sequences d'acides nucleiques et d'acides amines
EP1175492A1 (fr) Homologues de star
US6720182B1 (en) Alternative splice variants of CD40
US6506884B1 (en) Variant of vascular endothelial growth factor
US20030105049A1 (en) StAR homologues
US20050281810A1 (en) Variants of alternative splicing
US20020081655A1 (en) Splice variant of mGluR
EP1097166A2 (fr) Compositions de polypeptides et de polynucleotides de canaux k
WO2001034796A1 (fr) Homologues de type chordine
WO2000043506A1 (fr) Nouvelles sequences d'acides nucleiques et d'acides amines
US20030125288A1 (en) PI3K - regulatory subunit homology
WO2000044784A1 (fr) Sequences d'acide nucleique et d'acide amine
WO2002102848A2 (fr) Nouvelles sequences d'acides nucleiques et d'acides amines
CA2339406A1 (fr) Nouveau polypeptide esk du canal potassium et compositions polynucleotidiques
WO2001029215A2 (fr) Homologues de molecules d'adherence cellulaire neuronale
WO1999060121A1 (fr) PROTEINE DU TYPE RECEPTEUR DE GLUTAMATE METABOTROPIQUE ET ADNc DE CODAGE

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM DZ 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 NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG 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 BF BJ CF CG CI CM GA GN 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)
WWE Wipo information: entry into national phase

Ref document number: 2000922827

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2000922827

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWE Wipo information: entry into national phase

Ref document number: 09959656

Country of ref document: US

WWW Wipo information: withdrawn in national office

Ref document number: 2000922827

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: JP