EP1156819A1 - Methoden und zusammensetzungen zur regulierung der fruchtbarkeit - Google Patents

Methoden und zusammensetzungen zur regulierung der fruchtbarkeit

Info

Publication number
EP1156819A1
EP1156819A1 EP00914482A EP00914482A EP1156819A1 EP 1156819 A1 EP1156819 A1 EP 1156819A1 EP 00914482 A EP00914482 A EP 00914482A EP 00914482 A EP00914482 A EP 00914482A EP 1156819 A1 EP1156819 A1 EP 1156819A1
Authority
EP
European Patent Office
Prior art keywords
fsp95
gene
protein
sperm
amino acid
Prior art date
Legal status (The legal status 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 status listed.)
Withdrawn
Application number
EP00914482A
Other languages
English (en)
French (fr)
Other versions
EP1156819A4 (de
Inventor
John Herr
Arabinda Mandal
Michael Wolkowicz
Kenneth Klotz
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Virginia UVA
University of Virginia Patent Foundation
Original Assignee
University of Virginia UVA
University of Virginia Patent Foundation
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 University of Virginia UVA, University of Virginia Patent Foundation filed Critical University of Virginia UVA
Publication of EP1156819A1 publication Critical patent/EP1156819A1/de
Publication of EP1156819A4 publication Critical patent/EP1156819A4/de
Withdrawn legal-status Critical Current

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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/16Masculine contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to vertebrate FSP95 genes and their encoded 10 protein products, as well as derivatives and analogs thereof. Production of vertebrate
  • FSP95 proteins, derivatives and antibodies is also provided.
  • the invention further relates to therapeutic compositions and methods of diagnosis and therapy, including compositions and methods for modulating fertility (e.g., inhibiting or promoting), such as methods for contraception, and methods for promoting fertility.
  • capacitation prepares the sperm to undergo the acrosome reaction and to bind
  • Capacitated spermatozoa are also characterized by a rise in intracellular pH (Vredenburgh- Wilberg and Parrish, 1995, Mol. Reprod. Dev. 40:490-502), and loss of cholesterol from the sperm plasma membrane (Nisconti and Kopf, 1998, Biol. Reprod. 59:1-6).
  • sperm proteins are potent auto- and iso-antigens which evoke immune responses in both males and females
  • antisperm antibodies (“ASA") are capable of causing infertility in both human and animal models (Primakoff et al., 1988, Nature 335:543-546, Kutteh et al., 1996, Am. J. Reprod. Immunol. 35:429-433, Ohl and Naz, 1995,
  • ASA can be measured in serum of up to 70 percent of men after vasectomy compared with 2 to 8 percent of men without vasectomy (Raspa, 1993, Am. Fain. Physician 48:1264-1268).
  • the incidence of ASA in infertile couples varies from 9 to 36% and in infertile men is from 8 to 21% (Gubin et al., 1998, Am. J. Reprod. Immunol. 39:157-160), while the prevalence of 0 ASA in the general population ranges from 0 to 2% (Jarow and Sanzone, 1992, J. Urol. 148:1805-1807).
  • the present invention is based, in part, on the discovery of the nucleotide and amino acid sequence of a novel fibrous sheath protein of 95 kDa (FSP95), which is 5 localized to the ribs of the fibrous sheath in the principal pieoe of sperm tail, undergoes tyrosine phosphorylation during capacitation of human spermatozoa and plays a role in sperm motility.
  • FSP95 novel fibrous sheath protein of 95 kDa
  • the 853 amino acid residue protein has a calculated molecular weight of 94.6 kDa and a pi of 6.0 and contains multiple potential phosphorylation sites for protein kinase C and casein inase II as well as one tyrosine kinase phosphorylation site at amino acid position 435.
  • the present invention is directed to nucleotide sequences of vertebrate fibrous sheath protein of 95 kDa (FSP95), and amino acid sequences of their encoded proteins, as well as derivatives (e.g., fragments) and analogs thereof. Nucleic acids hybridizable to or complementary to the FSP95 nucleotide sequence are also provided.
  • the FSP95 protein is a mammalian protein, preferably a human protein.
  • the invention is directed to vertebrate FSP95 derivatives and analogs which are functionally active, i.e., capable of displaying one or more known functional activities associated with a full-length (wild-type) FSP95 protein.
  • Such functional activities include but are not hmited to antigenicity [ability to bind (or compete with FSP95 for binding) to an anti-FSP95 antibody], immunogenicity (ability to generate antibody which binds to FSP95).
  • the invention is further directed to fragments (and derivatives and analogs thereof) of a vertebrate FSP95 protein that comprise one or more domains of the FSP95 protein, including but not limited to the extracellular domain, txansmembrane domain, intracellular domain, intracellular targeting domain, RII binding site, or any combination of the foregoing.
  • fragments of FSP95 comprise amino acids 318- 335, amino acids 368-385, and/or amino acids 671-688.
  • Antibodies to a vertebrate FSP95 protein, its derivatives and analogs are additionally provided by the present invention.
  • the present invention is also directed to therapeutic and diagnostic methods and compositions based on vertebrate FSP95 proteins and nucleic acids.
  • the invention provides for treatment of sperm motility disorders by administration of a therapeutic compound of the invention.
  • therapeutic compounds include, but are not limited to, FSP95 proteins and analogs and derivatives (including fragments) thereof; antibodies thereto; nucleic acids encoding the FSP95 proteins, analogs or derivatives; and FSP95 antisense nucleic acids.
  • a therapeutic of the invention is administered to treat infertility.
  • a therapeutic of the invention is administered to decrease sperm motility.
  • a therapeutic of the invention is administered to increase sperm motility.
  • therapeutics which antagonize, or inhibit, FSP95 function are administered for therapeutic effect.
  • therapeutics which promote FSP95 function referred to as
  • Antist Therapeutics are administered for therapeutic effect. Disorders of sperm motility, involving aberrant or undesirable levels of expression or activity or localization of FSP95 protein can be diagnosed by detecting such levels, as described more fully infra.
  • a therapeutic of the invention is a fragment of an antibody to FSP95 consisting of at least the binding domain of the antibody.
  • a therapeutic of the invention is a fragment of FSP95 comprising amino acids 318-335, 368-385 and/or 671-688.
  • cAMP cyclic 3',5'-adenosine monophosphate
  • AKAP A-kinase anchor protein
  • PK-A protein kinase A
  • ASA antisperm antibodies
  • FSP95 fibrous sheath protein 95 kDa
  • OBG octyl-3-D-glucopyranoside
  • BSA bovine serum albumin
  • PBS phosphate buffer saline
  • RACE rapid amplification of cDNA ends
  • PCR polymerase chain reaction
  • IPTG isopropyl-1-thio- ⁇ -D-galactopyranoside
  • rFSP95 recombinant FSP95
  • HSA human serum albumin
  • S1Y sperm immobilization value
  • pro-mAKAP82 precursor of mouse AKAP82
  • pro-hAKAP82 precursor of human AKAP82.
  • FIG.l Identification of FSP95 as a high molecular weight major acidic tyrosine phosphorylated protein in capacitated human spermatozoa by 2-D immunoblot. Fresh swim up sperm were capacitated in absence and presence of genistein, a protein tyrosine kinase inhibitor. The location of FSP95 spot in the 2-D sperm proteome is indicated with a white half rectangular box (A) in a silver stained gel. The high molecular weight acidic proteins (box area of A) which showed phosphorylation of tyrosine residues are shown in blots probed with antiphosphotyrosine antibody before (B) and after capacitation (C).
  • Prominent high molecular weight acidic tyrosine phosphorylated proteins ( ⁇ 95 kDa, pi 5.1-5.5) show increased phosphorylation with capacitation (C). A lack of phosphorylation of the 95 kDa proteins was observed in capacitated cells in presence of genistein (D). The spot which was cored for microsequencing FSP95 is indicated with a white circle along with an arrow(A). FIG. 2. Antigenicity of FSP95 in men and women with antisperm antibodies.
  • Nucleotide and deduced amino acid sequences of the human sperm protein FSP95 are shown below the cDNA sequence.
  • the numbers on the left refer to the nucleotide sequence; numbers on the right refer to the amino aoid sequence.
  • the consensus ATG of the open reading frame and the polyadenylation signal (ATFAAA) are indicated in bold letters.
  • the termination codon (TAA) is marked with an asterisk.
  • the 5-prime and 3-prime untranslated regions are 162 bp and 218 bp respectively and are shown in italics.
  • the calculated molecular weight and pi of the predicted protein were 94.6 kDa and 6.0 respectively.
  • the 18 underlined sequences indicate the tryptic peptides obtained by microsequencing.
  • the putative tyrosine kinase phosphorylation site is indicated in bold within a box (residue number 435).
  • GenBank accession number AF087003
  • FIG.4 Homology comparison of the deduced amino acid sequences of human sperm FSP95 with those of mouse and human sperm fibrous sheath AKAPs (mouse: pro-rnAKAP82, accession # 148968; human: pro-hAKAP82, accession # AF072756). The sequences were listed in descending order of homology from the FSP95. The alignment was constructed by use of the GCG-PILEUP program and formatted with ALSCRIPT version 2.0. The shaded areas indicate the amino acid identities and similarities among the molecules (cut off 8 in ALSCRIPT). The conserved AKAP-like intracellular targeting domains are shown in boxes. The N-terminal RTI-binding domain of the mouse and human AKAP82 is highlighted with an underline. FIG.5. Analysis of human sperm FSP95 expression. A: Northern blot
  • RNA markers (kb) are indicated on the left. A single transcript of- 3.0 kb was apparent only in testis. The blot was subsequently stripped and rehybridized with actin to assess the levels of RNA in each lane (data not shown).
  • B Dot blot containing poly(A)+ RNA from 50 human tissues (obtained from Clontech) was hybridized with a radiolabelled FSP95 cDNA and signals were visualized by autoradiography. A hybridization was found only in testis following 18 h of exposure.
  • the dot blot contained normalized amounts (89-514 ng) of Poly(A)+ RNA from the following 50 tissues: Al, whole brain; A2, amygdala; A3, caudate nucleus; A4, cerebellum; AS, cerebral cortex; A6, frontal lobe; A7, hippocampus; A8, medulla oblongata; B 1, occipital lobe; B2, putamen; B3, substantia nigra; B4, temporal lobe; BS, thalamus; B6, subthalamic nucleus; B7, spinal cord; Cl, heart; C2, aorta; C3, skeletal muscle; C4, colon; CS, bladder; C6, uterus; C7, prostate; C8, stomach; Dl, testis; D2, ovary; D3, pancreas; D4, pituitary gland; DS, adrenal gland; D6, thyroid gland; D7, salivary gland; D8, mammary gland; El, kidney; E2,
  • FIG. 6 Isolation of recombinant FSP95 (rFSP95) from E. coli and immunoblotting of recombinant and sperm FSP95.
  • rFSP95 recombinant FSP95
  • a portion of the human sperm FSP95 cDNA (encoding residues 1 -779 of the protein) was expressed in E. coli using the pET2Sb plasmid, induced by addition of 1.0 mM IP TG and purified by nickel ion affinity column chromatography followed by preparative polyacrylamide gel electrophoresis.
  • A Bacterial extracts stained with Coomassie; lane 1, uninduced; lanes 2 and 3, 1.5 h and 3.0 h after induction; expressed FSP95 is indicated by the arrow.
  • B Coomassie stained purified rFSP95 (2.3 ⁇ g), used for immunization; left, molecular weight markers.
  • C Immunoblot of gel purified rFSP95 (3.5 ⁇ g) probed with rat antisera against rFSP95 at l:SOOO dilution; lane 1, preimmune serum; lane 2, immune serum.
  • D 2-D Blot of human sperm proteins probed with rat serum against gel purified rFSP95 . The 95 kDa region at pi - 5.3 originally microsequenced immunoreacts with the antibody (arrow). The pH gradient is indicated at the top. Molecular weight markers are shown to the left of all the panels.
  • FIG.7 Phase contrast (A, C) and indirect immunofluorescence staining (B, D) of FSP95 in capacitated permeabilized human spermatozoa. Immunofluoresence was noted throughout the principal piece (PP) of the flagellum (bar) with immune sera (B) while no fluorescence was observed in the head, mid piece (MP) or the end piece (EP). No irnmunostaining was observed with preimmune sera (D), or by using immune sera on live capacitated unpermeabilized sperm (data not shown).
  • PP principal piece
  • MP mid piece
  • EP end piece
  • FIG.8 Electron microscopic immunogold localization of FSP95 in a longitudinal (A) and a cross section (B) of the principal piece of ejaculated human spermatozoa.
  • Gold particles (arrows) were detected in the ribs of fibrous sheath (FS) but not within the central portion of the longitudinal columns (LC) .
  • No immunoreactivity was detected on the outer dense fibers (ODF) or on the axoneme (AXO). Only a rare gold particle was detected on control sections (C&D) treated with preimmune rat serum .
  • FIG.9 Tyrosine phosphorylation of FSP95 during in vitro capacitation of human spermatozoa. Proteins of uncapacitated sperm (A, C) were compared to 6 h capacitated sperm (B, D) after 2D SDS -PAGE, electroblotting and probing with rat antisera against rFSP95 (A, B) and antiphosphotyrosine monoclonal antibody (C, D).
  • FIG. 10 Helical wheel representation of FSP95 RII subunit binding domain alpha helix (arnino acid residues 671-688), indicating surface of hydrophobic residues, shown in boxes (o). 5
  • the present invention provides "ESRP5"(fjbrous sheath protein of 95KDa) polynucleotides, polypeptides, and derivatives and analogs thereof.
  • the invention further provides methods for the use of such sequences in contraception and modulation of fertility.
  • Production of recombinant FSP95 proteins, and peptide fragments comprising functional domains, derivatives, antibodies, and ligands are also provided.
  • the invention further provides compositions, kits, and methods for their use to target sperm and modulate fertility.
  • the invention further provides therapeutic compositions and methods for the use of FSP95 polynucleotides for targeted heterologous gene expression.
  • the invention further encompasses the use of nucleotides encoding FSP95 proteins and peptides, as well as antibodies to FSP95 (which can, for example, act as agonists or antagonists), ligands that bind to FSP95 or modulate the function, activity or expression of FSP95.
  • regulatory nucleotides and nucleotides encoding FSP95 polypeptides or one or more functional domains of FSP95 or fragments thereof, e.g., the RII binding site are effective in gene therapy, or for delivery of heterologous gene products to a cellular or subcellular locale.
  • embodiments of the invention described in the subsections below encompasses FSP95, polypeptides or peptides corresponding to functional domains of FSP95 (e.g., a ligand-binding domain, such as the RII binding site), mutated, truncated or deleted (e.g. with one or more functional domains or portions thereof deleted), FSP95 fusion proteins, nucleotide sequences encoding such products, and host cell expression systems that can produce such FSP95 products.
  • functional domains of FSP95 e.g., a ligand-binding domain, such as the RII binding site
  • mutated, truncated or deleted e.g. with one or more functional domains or portions thereof deleted
  • FSP95 fusion proteins e.g. with one or more functional domains or portions thereof deleted
  • the invention also encompasses antibodies, including anti-idiotypic antibodies, antagonists and agonists, as well as compounds or nucleotide constructs that inhibit expression of the FSP95 gene (transcription factor inhibitors, antisense and ribozyme molecules, or gene or regulatory sequence replacement constructs), or promote expression of FSP95 (e.g., expression constructs in which FSP95 coding sequences are operatively associated with expression control elements such as promoters, promoter/enhancers, etc.).
  • the invention also provides host cells and animals genetically engineered to express the o human (or mutants thereof) FSP95 regulatory or protein coding sequences, or to inhibit or "knock-out" expression of the animal's endogenous FSP95.
  • the FSP95 products and fusion protein products (i.e., fusions of the proteins or a domain of the protein, e.g., the RU binding site), antibodies and anti-idiotypic antibodies (including Fab fragments), modulators and ligands can be used as therapeutics to 5 modulate fertility.
  • the present invention provides methods of screening for agents, small molecules, or proteins that interact with FSP95.
  • the invention encompasses both in vivo and in vitro assays to screen small molecules, compounds, recombinant proteins, peptides, nucleic acids, antibodies etc. which bind to or modulate the activity of FSP95 and are thus 0 useful as therapeutics or diagnostic markers for fertility.
  • the FSP95 polynucleotides i.e., coding regions or regulatory regions of the FSP95 gene
  • fusion protein products i.e., fusions of the FSP95 proteins or a domain of the protein, e.g., the RII binding site, to another heterologous polypeptid ⁇
  • antibodies including, but without limitation, anti-idiotypic antibodies, and Fab fragments
  • modulators and ligands can be used for drug delivery or gene therapy.
  • the invention also encompasses pharmaceutical formulations and methods for contraception and treating infertility and cancer.
  • a FSP95 nucleic acid refers to: (a) a nucleic acid molecule containing the nucleotide sequence of FSP95 shown in FIG. 3
  • the invention also includes nucleic acid molecules derived from mammalian nucleic acids, preferably DNA molecules, that hybridize to, and are therefore the complements of, the nucleotide sequences (a) through (d), in the preceding paragraph.
  • Such hybridization conditions may be highly stringent or less highly stringent, as described above.
  • highly stringent conditions may refer, e.g., to washing in 6xSSC/0.05% sodium pyrophosphate at 37°C (for 14-base oligos), 48°C (for 17-base oligos), 55°C (for 20-base oligos), and 60°C (for 23-base oligos).
  • nucleic acid molecules may encode or act as FSP95 nucleic acid antisense molecules, useful, for example, in FSP95 gene regulation (for and/or as antisense primers in amplification reactions of FSP95 nucleic acid sequences).
  • FSP95 gene regulation such techniques can be used to regulate, for example, a SP9.J-regulated pathway, in order to block cell proliferation associated with cancer.
  • sequences may be used as part of ribozyme and/or triple helix sequences, also useful for FSP95 gene regulation.
  • such molecules may be used as components of diagnostic methods whereby, for example, the presence of a particular FSP95 allele responsible for causing a FSP95 related disorder, e.g., fertility or proliferative disorders such as infertility or cancer, may be detected.
  • a FSP95 related disorder e.g., fertility or proliferative disorders such as infertility or cancer
  • the nucleic acid molecules of the invention further include nucleotide sequences that encode polypeptides having at least 30%, 35%. 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or higher amino acid sequence identity to the polypeptides encoded by the FSP95 nucleotide sequences of (a)-(d) above.
  • the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence).
  • the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions arc then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position.
  • the determination of percent identity between two sequences can also be accomplished using a mathematical algorithm.
  • a preferred, non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Karlin and Altschul, 1990, Proc. Natl. Acad. Sci. USA 87:2264-2268, modified as in Karlin and Altschul, 1993, Proc. Natl. Acad. Sci. USA 90:5873-5877. Such an algorithm is incorporated into the NBLAST and XBLAST programs of Altschul, et al., 1990, J. Mol. Biol. 215:403-410.
  • Gapped BLAST can be utilized as described in Altschul et al., 1997, Nucleic Acids ⁇ es.25:3389-3402.
  • PSI-Blast can be used to perform an iterated search which detects distant relationships between molecules (Altschul et al., 1997, supra).
  • the invention further includes fragments of any of the nucleotide sequences disclosed herein.
  • the fragment of a FSP95 nucleic acid encodes a FSP95 PKA RII subunit binding domain sequence.
  • a FSP95 PKA RII subunit binding domain sequence includes: (a) any DNA sequence that encodes a peptide, e.g. , a PKA RII subunit binding domain peptide, comprising amino acids 318-335
  • the invention encompasses the FSP95 PKA RII subunit binding domain sequences, in isolated or purified form, as well as compositions containing such PKA RII subunit binding domain sequences operatively associated with a nucleic acid encoding a protein or polypeptide heterologous to FSP95.
  • FSP95 sequences of the present invention are derived from a eukaryotic genome, preferably a mammalian genome, and more preferably a human genome.
  • the nucleotides of the present invention encompass any DNA sequence derived from a mammalian genome which hybridizes under highly stringent conditions to the nucleotide sequence shown in FIG. 3.
  • the nucleotides of the present invention encompass any DNA sequence derived from a mammalian genome which hybridize under highly stringent conditions to the nucleotide sequence shown in FIG. 3 and encodes a gene product involved sperm motility, and contains a PKA RU subunit binding domain.
  • nucleic acid which encodes a given amino acid sequence
  • the nucleic acid need not only be a cDNA molecule, but can also, for example, refer to a gDNA sequence from which an mRNA species is transcribed that is processed to encode the given amino acid sequence.
  • the invention further includes regulator nucleic acids of the FSP95 gene.
  • the genomic sequence of the FSP95 gene contains regulatory sequences in the non-coding 5'- flanking region.
  • the 5 '-regulatory sequences of the FSP95 gene comprise the polynucleotide sequences located between the nucleotide in position -5000, -3000, -2000, - 1000, or -500, and the nucleotide in position +10, +100, or +300, of the nucleotide sequence of the nucleotide sequence shown in FIG. 3.
  • the invention also encompasses:
  • regulatory elements include but are not limited to inducible and non-inducible promoters, enhancers, operators and other elements known to those skilled in the art that drive and regulate expression.
  • Such regulatory elements include but are not limited to the cytomegalovirus hCMN immediate early gene, the early or late promoters of SV40 adenovirus, the lac system, the trp system, the TAC system, the TRC system, the major operator and promoter regions of phage A, the control regions of fd coat protein, the promoter for 3-phosphoglycerate kinase, the promoters of acid phosphatase, and the promoters of the yeast ⁇ -mating factors.
  • the FSP95 nucleic acid sequences of the invention are mammalian nucleic acid sequences, with human sequences being preferred.
  • the FSP95 nucleic acid sequences of the invention are nucleic acid sequences encoding FSP95 gene products containing polypeptide portions corresponding to (that is, polypeptide portions exhibiting amino acid sequence similarity to) the amino acid sequences depicted in FIG. 3 and FIG. 4, wherein the corresponding portion exhibits greater than about 50% amino acid identity with the depicted sequence, averaged across the FSP95 gene product's entire length.
  • FSP95 encoding nucleic acids comprise the cDNA sequences of the nucleotide sequences shown in FIG. 3 or the coding regions thereof, or nucleic acids encoding a FSP95 protein (e.g., a protein having the amino acid sequences depicted in FIG. 3 and FIG. 4).
  • the invention provides isolated or purified nucleic acids consisting of at least 8 nucleotides (i.e., a hybridizable portion) of a FSP95 nucleic acid
  • the nucleic acids consist of at least 25 (continuous) nucleotides, 50, 100, 150, 200, 300, 400, 500, 600, 700, 800,900, 1000, 1500, 2000, 2500, 2900, or 2941 contiguous nucleotides of a FSP95 sequence, or a full-length FSP95 coding sequence.
  • the invention provides isolated or purified nucleic acids consisting of nucleotides 1-200, 201-250, 251-300, 301-350, 351-400, 401-
  • nucleic acids are smaller than 25, 50, 75, 100, 200, 300, 400, 500, 1000, 1500, 2000, 2500, or 2941 nucleotides in length. Nucleic acids can be single or double stranded. The invention also provides nucleic acids hybridizable to or complementary to the foregoing sequences.
  • nucleic acids are provided which comprise a sequence complementary to at least 10, 25, 50, 100, 200, 300, 400, 500, 1000, 1500, 2000, 2500 nucleotides or the entire coding region of a FSP95 gene.
  • the invention also encompasses those genomic DNA sequences which give rise to the cDNA sequences of the nucleotide sequences shown in FIG. 3 described above. 5
  • FSP95 nucleic acid sequences can be identified and readily isolated, without undue experimentation, by molecular biological techniques well known in the art, used in conjunction with the FSP95 nucleic acid sequences disclosed herein. These other sequences are encompassed by the present 0 invention.
  • additional human FSP95 nucleic acid sequences at the same or at different genetic loci as those disclosed in the nucleotide sequences shown in FIG. 3 can be isolated readily. There can exist, for example, genes at other genetic or physical loci within the human genome that encode proteins that have extensive homology to one or more domains of the FSP95 gene products and that encode gene products functionally equivalent 5 to a FSP95 gene product.
  • homologous FSP95 nucleic acid sequences present in other species can be identified and isolated readily. With respect to identification and isolation of FSP95 nucleic acid sequences present at the same genetic or physical locus as those sequences disclosed in the nucleotide sequences shown in FIG. 3, such sequences can, for example, be obtained readily by utilizing standard sequencing and bacterial artificial chromosome (BAC) and PI artificial chromosome (PAC) technologies.
  • BAC bacterial artificial chromosome
  • PAC PI artificial chromosome
  • the isolated FSP95 nucleic acid sequences disclosed herein may be labeled and used to screen a cDNA library constructed from mRNA obtained from appropriate cells or tissues (e.g., testes) derived from the organism (e.g., mouse) of interest.
  • the hybridization conditions used should be of a lower stringency when the cDNA library is derived from an organism different from the type of organism from which the labeled sequence was derived.
  • the labeled fragment may be used to screen a genomic library derived from the organism of interest, again, using appropriately stringent conditions.
  • Low stringency conditions are well known to those of skill in the art, and will vary predictably depending on the specific organisms from which the library and the labeled sequences are derived. For guidance regarding such conditions see, for example, Sambrook, et al, 1 89, Molecular Cloning, A Laboratory Manual, Second Edition, Cold Spring Harbor Press, N.Y.; and Ausubel, et al, supra.
  • a FSP95 gene homologue may be isolated from, for example, human nucleic acid, by performing PCR using two degenerate oligonucleotide primer pools designed on the basis of amino acid sequences within any FSP95 gene product disclosed herein.
  • the PCR product may be subcloned and sequenced to ensure that the amplified sequences represent the sequences of a FSP95 gene nucleic acid sequence.
  • the PCR fragment may then be used to isolate a full length cDNA clone by a variety of methods.
  • the amplified fragment may be labeled and used to screen a bacteriophage cDNA library.
  • the labeled fragment may be used to isolate genomic clones via the screening of a genomic library.
  • RNA may be isolated, following standard procedures, from an appropriate cellular or tissue source (i.e., testis).
  • a reverse transcription reaction may be performed on the RNA using an oligonucleotide primer specific for the most 5' end of the amplified fragment for the prirning of first strand synthesis.
  • the resulting RNA/DNA hybrid may then be "tailed" with guanines using a standard terminal transferase reaction, the hybrid may be digested with RNAase H, and second strand synthesis may then be primed with a poly-C primer.
  • cDNA sequences upstream of the amplified fragment may easily be isolated.
  • FSP95 nucleic acid sequences may additionally be used to identify mutant FSP95 gene alleles.
  • mutant alleles may be isolated from individuals either known or proposed to have a genotype that contributes to the symptoms of a FSP95 gene disorder, such as fertility disorders, for example. Such alleles are encompassed by the present invention.
  • FSP95 alleles may be identified by single strand conformationa) polymorphism (SSCP) mutation detection techniques, Southern blot, and/or PCR amplification techniques.
  • Primers can routinely be designed to amplify overlapping regions of the whole FSP95 sequence including the promoter region.
  • primers are designed to cover the exon-intron boundaries such that, first, coding regions can be scanned for mutations.
  • Genomic DNA isolated from lymphocytes of normal and affected individuals is used as PCR template. PCR products from normal and affected individuals are compared, either by single strand conformational polymorphism (SSCP) mutation detection techniques and or by sequencing.
  • SSCP single strand conformational polymorphism
  • SSCP analysis can be performed as follows: 100 ng of genomic DNA is amplified in a 10 ⁇ l reaction, adding 10 pmols of each primer, 0.5 U of Taq DNA polymerase (Promega), 1 Ci of ⁇ -[ 3 P]dCTP (NEN; specific activity, 3000 Ci mmol), in 2.5 ⁇ U dNTPs (Pharmacia), 10 mM Tris-HCl (pH 8.8), 50 mM KC1, 1 mM MgC12, 0.01% gelatin, final concentration.
  • Autoradiography is performed by exposure to film at -70 °C with intensifying scree for different periods of time.
  • the mutations responsible for the loss or alteration of function of the mutant FSP95 gene product can then be ascertained.
  • a cDNA of a mutant FSP95 gene may be isolated, for example, using PCR.
  • the first cDNA strand may be synthesized by hybridizing an oligo-dT oligonucleotide to mRNA isolated from tissue known or suspected to be expressed in an individual putatively carrying the mutant FSP95 allele, and by extending the new strand with reverse transcriptase.
  • the second strand of the cDNA is then synthesized using an oligonucleotide that hybridizes specifically to the 5' end of the normal gene. Using these two primers, the product is then amplified via PCR, cloned into a suitable vector, and subjected to DNA sequence analysis through methods well known to those of skill in the art. By comparing the DNA sequence of the mutant FSP95 allele to that of the normal FSP95 allele, the mutation(s) responsible for the loss or alteration of function of the mutant FSP95 gene product can be ascertained.
  • a genomic library can be constructed using DNA obtained from an individual suspected of or known to carry a mutant FSP95 allele, or a cDNA library can be constructed using RNA from a tissue known, or suspected, to express a mutant FSP95 allele.
  • An unimpaired FSP95 gene or any suitable fragment thereof may then be labeled and used as a probe to identify the corresponding mutant FSP95 allele in such libraries.
  • Clones containing the mutant FSP95 nucleic acid sequences may then be purified and subjected to sequence analysis according to methods well known to those of skill in the art.
  • an expression library can be constructed utilizing cDNA synthesized from, for example, RNA isolated from a tissue known, or suspected, to express a mutant FSP95 allele in an individual suspected of or known to carry such a mutant allele.
  • gene products made by the putatively mutant tissue may be expressed and screened using standard antibody screening techniques in conjunction with antibodies raised against the normal FSP95 gene product, as described, below, in Section 5.3.
  • For screening techniques see, for example, Harlow and Lane, eds., 1988, "Antibodies: A Laboratory Manual", Cold Spring Harbor Press, Cold Spring Harbor.)
  • nucleic acids encoding derivatives and analogs of FSP95 proteins, and FSP95 antisense nucleic acids can be isolated by the methods recited above.
  • a "nucleic acid encoding a fragment or portion of a FSP95 protein” shall be construed as referring to a nucleic acid encoding only the recited fragment or portion of the FSP95 and not the other contiguous portions of the FSP95 protein as a continuous sequence.
  • FSP95 nucleic acids comprising regions conserved between (i.e., with homology to) other FSP95 nucleic acids, of the same or different species, are also provided.
  • Nucleic acids encoding one or more FSP95 domains can be isolated by the methods recited above.
  • &FSP95 mutation results in an expressed gene product with altered function (e.g., as a result of a missense or a frameshift mutation)
  • a polyclonal set of an ⁇ -FSP95 gene product antibodies are likely to cross-react with the mutant FSP95 gene product.
  • Library clones detected via their reaction with such labeled antibodies can be purified and subjected to sequence analysis according to methods well known to those of skill in the art.
  • the invention further provides ESPPJ-encoded proteins and amino acid sequences, as well as derivatives (e.g., fragments) and analogs thereof.
  • the invention provides FSP95 derivatives and analogs which are functionally active, i.e., they are capable of displaying one or more functional activities associated with a full-length (wild-type) FSP95 protein.
  • Such functional activities include, but are not limited to, binding to the RU subunit of PKA, phosphorylation, modulating sperm motility, sperm marker, antigenicity (ability to bind to an anti-FSP95 antibody or compete with FSP95 for binding), immunogenicity (ability to generate antibody which binds to FSP95).
  • the invention further provides fragments (and derivatives and analogs thereof) of FSP95 which comprise one or more domains of a FSP95 protein.
  • the FSP95 protein is a human protein.
  • the amino acid sequences depicted in FIG. 3 and FIG.4 represent FSP95 gene products.
  • the FSP95 gene product sometimes referred to herein as a "FSP95" includes those products encoded by the FSP95 nucleic acid sequences described in Section 5.1, above.
  • the nucleic acid sequences encoding the FSP95 gene products are derived from eukaryotic genomes, including mammalian genomes. In a preferred embodiment the nucleic acid sequences encoding the FSP95 products are derived from the human genome.
  • FSP95 proteins, polypeptides and peptide fragments thereof can be prepared for a variety of uses.
  • such molecules can be used for the generation of antibodies, for use in diagnostic and therapeutic assays, for the identification of other sperm gene products involved in sperm motility, or for the identification of compounds that modulate sperm motility.
  • FSP95 products of the present invention may include proteins that represent functionally equivalent (see Section 5.1 for a definition) products.
  • Functionally equivalent FSP95 products may contain deletions, including internal deletions, additions, including additions yielding fusion proteins, or substitutions of amino acid residues within and/or adjacent to the amino acid sequence encoded by the FSP95 nucleic acid sequences described, above, in Section 5.1, but that result in a "silent" change, in that the change produces a functionally equivalent FSP95 gene product.
  • Amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and or the amphipathic nature of the residues involved.
  • nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine;
  • polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine;
  • positively charged (basic) amino acids include arginine, lysine, and histidine; and negatively charged (acidic) amino acids include aspartic acid and glutamic acid.
  • FSP95 proteins encompassed by the present invention include immunoreactive proteins with the following molecular weights (MW) in kiloDaltons, and isoloelectric points (pi), obtainable from human sperm preparations: a protein
  • deletion or non- conservative alterations can be engineered to produce altered FSP95 gene products.
  • Such o alterations can, for example, alter one or more of the biological functions of the FSP95 product. Further, such alterations can be selected so as to generate FSP95 products that are better suited for expression, scale up, etc. in the host cells chosen.
  • cysteine residues can be deleted or substituted with another amino acid residue in order to eliminate disulfide bridges.
  • tyrosine residues can be deleted or substituted with another amino acid residue in order to eliminate tyrosine phosphorylation.
  • Peptides and/or proteins corresponding to one or more domains of a FSP95 protein as well as fusion proteins in which a FSP95 protein or a portion of a FSP95 protein such as a truncated FSP95 protein or peptide or a FSP95 protein domain, is fused to an unrelated protein are also within the scope of this invention.
  • Such proteins and peptides can be designed on the basis of the FSP95 nucleotide sequence disclosed in Section 5.1, above, and or on the basis of the FSP95 amino acid sequence disclosed herein.
  • Fusion proteins include, but are not limited to, IgFc fusions which stabilize the FSP95 protein or peptide and prolong half life in vivo; or fusions to any amino acid sequence that allows the fusion protein to be anchored to the cell membrane; or fusions of FSP95 protein domains to an enzyme, fluorescent protein, luminescent protein, or a flag epitope protein or peptide which provides a marker function.
  • FSP95 proteins of the invention also include FSP95 protein sequences wherein domains encoded by at least one exon of the cDNA sequence, or fragments thereof, have been deleted.
  • the FSP95 polypeptides of the invention can further comprise posttranslational modifications, including, but not limited to stearation, myristylations, palmitation, glycosylations, acetylations, and phosphorylations.
  • FSP95 polypeptides are modified to make them membrane permeable using techniques such as stearation, myristylation, palmitation, and incorporating lipophilic amino acids. If the native FSP95 protein does not have recognition motifs that allow such modifications, it would be routine for one skilled in the art to introduce into a FSP95 gene nucleotide sequences that encode motifs such as enzyme recognition signals so as to produce a modified FSP95 gene product.
  • the peptides of the invention or analogues thereof may be prepared using
  • the peptides may be prepared in linear form using conventional solution or solid phase peptide syntheses and cleaved from the resin followed by purification procedures (Creighton, 1983, Protein Structures And Molecular Principles, W.H. Freeman and Co., N.Y.). Suitable procedures for synthesizing the peptides described herein are well
  • composition of the synthetic peptides may be confirmed by amino acid analysis or sequencing (e.g., the Edman degradation procedure and mass spectroscopy).
  • analogues and derivatives of the peptides can be chemically synthesized.
  • the linkage between each amino acid of the peptides of the invention may be an amide, a substituted amide or an isostere of amide.
  • Nonclassical amino acids or 0 chemical amino acid analogues can be introduced as a substitution or addition into the sequence.
  • Non-classical amino acids include, but are not Hmited to, the D-isomers of the common amino acids, ⁇ -amino isobutyric acid, 4-aminobutyric acid, Abu, 2-amino butyric acid, ⁇ -Abu, e-Ahx, 6-amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, omithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, 5 cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, ⁇ -alanine, fiuoro-amino acids, designer amino acids such as ⁇ -methyl amino acids, C ⁇ -methyi amino acids, N ⁇ -methyl amino acids, and amino acid analogues in general.
  • the amino acid can be D (dextro
  • Cyclized peptides may be formed by the addition of Cys residues to the 0 termini of linear peptides. Formation of disulfide linkages, if desired, is generally conducted in the presence of mild oxidizing agents. Chemical oxidizing agents may be used, or the compounds may simply be exposed to atmospheric oxygen to effect these linkages. Various methods are known in the art, including those described, for example, by Tarn, J.P. et al, 1979, Synthesis 955-957; Stewart et al, 1984, Solid Phase Peptide 5 Synthesis, 2d Ed., Pierce Chemical Company Rockford, IL; Ahmed et al, 1975, J. Biol. Chem.
  • the peptide is composed entirely of gene-encoded amino acids, or a portion of it is so composed, the peptide or the relevant portion may also be synthesized using
  • a polynucleotide sequence encoding a linear form of the peptide is inserted into an appropriate expression vehicle, i.e., a vector which contains the necessary elements for the transcription and translation of the inserted coding sequence, or in the case of an RNA viral vector, the necessary elements for replication and
  • the expression vehicle is then transfected into a suitable target cell which will express the peptide.
  • the expressed peptide is then isolated by procedures well-established in the art. Methods for recombinant protein and peptide production are well known in the art (see, e.g. , Maniatis et al, 1989, Molecular Cloning A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y.; and Ausubel et al,
  • the FSP95 gene products, peptide fragments thereof and fusion proteins thereof may be produced by recombinant DNA technology using techniques well known in
  • RNA capable of encoding FSP95 gene product sequences may be chemically synthesized using, for example, synthesizers. Sec, for example, the techniques described in "Oligonucleotide Synthesis", 1984, Gait, ed., IRL Press, Oxford.
  • a variety of host-expression vector systems may be utilized to express the
  • Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells that may, when transformed or transfected with the appropriate nucleotide coding sequences, exhibit the FSP95 gene product of the invention in situ.
  • These include but are not limited to microorganisms such as bacteria (e.g., E. coli, B.
  • subtilis transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing FSP95 gene product coding sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing the FSP95 gene product coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing the FSP95 gene product coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing FSP95 gene product coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.
  • a number of expression vectors may be advantageously selected depending upon the use intended for the FSP95 gene product being expressed. For example, when a large quantity of such a protein is to be produced, for the generation of pharmaceutical compositions of FSP95 protein or for raising antibodies to FSP95 protein, for example, vectors that direct the expression of high levels of fusion protein products that are readily purified may be desirable.
  • vectors include, but are not limited, to the E.
  • coli expression vector pUR278 (Ruther et al, 1983, EMBO J.2, 1791), in which the FSP95 gene product coding sequence may be Iigated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; pIN vectors (Inouye and Inouye, 1985, Nucleic Acids Res. 13, 3101-3109; Van Heeke and Schuster, 1989, J. Biol. Chem. 264, 5503-5509); and the like.
  • pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
  • fusion proteins are soluble and can easily be purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione.
  • the pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
  • Autographa californica, nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes.
  • the virus grows in Spodoptera frugiperda cells.
  • FSP95 gene coding sequences may be cloned individually into non- essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter). Successful insertion of FSP95 gene coding sequences will result in inactivation of the polyhedrin gene and production of non-occluded recombinant virus (i.e., virus lacking the proteinaceous coat coded for by the o polyhedrin gene).
  • a number of viral-based expression systems may be utilized.
  • a FSP95 gene 5 coding sequence of interest may be Iigated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence.
  • This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e.g., region El or E3) will result in a recombinant virus that is viable and capable of expressing FSP95 gene product in infected hosts (e.g., see Logan and Shenk, 1984, Proc.
  • Specific initiation signals may also be required for efficient translation of inserted FSP95 gene product coding sequences. These signals include the ATG initiation codon and adjacent sequences. In cases where an entire FSP95 gene, including its own initiation codon and adjacent sequences, is inserted into the appropriate expression vector, no additional translational control signals may be needed. However, in cases where only a portion of the FSP95 gene coding sequence is inserted, exogenous translational control signals, including, perhaps, the ATG initiation codon, must be provided. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert.
  • exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic.
  • the efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bittner, et al, 1987, Methods in Enzymol. 153, 516-544).
  • a host cell strain may be chosen that modulates the expression of the inserted sequences, or modifies and processes the gene produot in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein.
  • Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed.
  • eukaryotic host cells that possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used.
  • mammalian host cells include but are not limited to CHO, VERO, BHK, HeLa, COS, MDCK, 293, 3T3, and WI38.
  • cell lines that stably express the FSP95 gene product may be engineered.
  • host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media.
  • the selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci that in turn can be cloned and expanded into cell lines. This method may advantageously be used to engineer cell lines that express the
  • FSP95 gene product Such engineered cell lines may be particularly useful in screening and evaluation of compounds that affect the endogenous activity of the FSP95 gene product.
  • a number of selection systems may be used, including but not limited to the herpes simplex virus thyrmdine kinase (Wigler, et al, 1977, Cell 11: 223), hypoxanthine- guanine phosphoribosyltransferase (Szybalska and Szybalski, 1962, Proc. Natl. Acad. Sci.
  • adenine phosphoribosyltransferase (Lowy, et al. , 1980, Cell 22: 817) genes can be employed in tk “ , hgprt " or aprt " cells, respectively.
  • antimetabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to mycophenolic acid (Mulligan and Berg, 1981, Proc. Natl. Acad. Sci. USA 78: 2072); neo, which confers resistance to the aminoglycoside G-418 (Colberre-Garapin, et al,
  • any fusion protein may be readily purified by utilizing an antibody specific for the fusion protein being expressed.
  • an antibody specific for the fusion protein being expressed For example, a system described by Janknecht, et al. allows for the ready purification of non-denatured fusion proteins
  • an endogenous FSP95 gene within a cell, cell line, or microorganism may be modified by inserting a heterologous DNA regulatory element into the genome of a stable cell line or cloned microorganism such that the inserted regulatory element is operatively linked with the endogenous FSP95 gene.
  • an endogenous FSP95 gene which is normally "transcriptionally silent" i.e., a FSP95 gene which is normally not expressed, or is expressed only at very low levels in a cell, cell line, or microorganism, may be activated by inserting a regulatory element which is capable of promoting the expression of a normally expressed gene product in that cell, cell line, or microorganism.
  • a transcriptionally silent, endogenous FSP95 gene may be activated by insertion of a promiscuous regulatory element that works across cell types.
  • a heterologous regulatory element may be inserted into a stable cell line or cloned microorganism, such that it is operatively linked with an endogenous FSP95 gene, using techniques, such as targeted homologous recombination, which are well known to those of skill in the art, and described e.g., in Chappel, U.S. Patent No. 5,272,071; PCT publication No. WO 91 /06667, published May 16, 1991.
  • FSP95 gene products can also be expressed in transgenic animals.
  • Animals of any species including, but not hmited to, mice, rats, rabbits, guinea pigs, pigs, micro- pigs, goats, sheep, and non-human primates, e.g., baboons, monkeys, and chimpanzees may be used to generate FSP95 transgenic animals.
  • transgenic refers to animals expressing FSP95 gene sequences from a different species (e.g.
  • mice expressing human FSP95 sequences as well as animals that have been genetically engineered to overexpress endogenous (i.e., same species) FSP95 sequences or animals that have been genetically engineered to no longer express endogenous FSP95 gene sequences (i.e., "knock-out” animals), and their progeny.
  • Any technique known in the art may be used to introduce an FSP95 gene transgene into animals to produce the founder lines of transgenic animals. Such techniques include, but are not limited to pronuclear microinjection (Hoppe and Wagner, 1989, U.S. Pat. No. 4,873,191); retrovirus mediated gene transfer into germ lines (van der Putten, et al, 1985, Proc. Natl. Acad.
  • transgenic animal clones containing an FSP95 transgene for example, nuclear transfer into enucleated oocytes of nuclei from cultured embryonic, fetal or adult cells induced to quiescence (Campbell, et al, 1996, Nature 380,: 64-66; Wil ut, et al, Nature 385,: 810-813).
  • the present invention provides for transgenic animals that carry a FSP95 transgene in all their cells, as well as animals that carry the transgene in some, but not all their cells, i.e., mosaic animals.
  • the transgene may be integrated as a single transgene or in concatamers, e.g., head-to-head tandems or head-to-tail tandems.
  • the transgene may also be selectively introduced into and activated in a particular cell type by following, for example, the teaching of Lasko et al. (Lasko, et al, 1992, Proc. Natl. Acad. Sci. USA 89, 6232-6236).
  • the regulatory sequences required for such a cell-type specific activation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art.
  • gene targeting is preferred.
  • vectors containing some nucleotide sequences homologous to the endogenous FSP95 gene are designed for the purpose of integrating, via homologous recombination with chromosomal sequences, into and disrupting the function of the nucleotide sequence of the endogenous FSP95 gene.
  • the transgene may also be selectively introduced into a particular cell type, thus inactivating the endogenous FSP95 gene in only that cell type, by following, for example, the teaching of Gu, et al. (Gu, et al, 1994, Science 265, 103-106).
  • the regulatory sequences required for such a cell-type specific inactivation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art.
  • the phenotypic expression of the recombinant FSP95 gene may be assayed utilizing standard techniques. Initial screening may be accomplished by Southern blot analysis or PCR techniques to analyze animal tissues to assay whether integration of the transgene has taken place. The level of mRNA expression of the transgene in the tissues of the transgenic animals may also be assessed using techniques that include but are not limited to Northern blot analysis of tissue samples obtained from the animal, in situ hybridization analysis, and RT-PCR (reverse transcriptase PCR). Samples of FSP95 gene-expressing tissue, may also be evaluated immunocytochemically using antibodies specific for the FSR9J transgene product.
  • FSP95 gene products, or peptide fragments thereof can be prepared for a variety of uses.
  • gene products, or peptide fragments thereof can be used for the generation of antibodies, in diagnostic assays, or for mapping and the identification of other cellular or extracellular gene products involved in the sperm motility.
  • FSP95 gene products include but are not limited to soluble derivatives such as peptides or polypeptides corresponding to one or more domains of the FSP95 gene product, particularly FSP95 gene products, that are modified such that they are deleted for one or more hydrophobic domains.
  • FSP95 gene products can be directly administered to a subject to modulate sperm motility, for example, for use as, a contraceptive or to increase fertility.
  • nucleotide constructs encoding such FSP95 gene products can be used to genetically engineer host cells to express such FSP95 gene products in vivo; these genetically engineered cells can function as "bioreactors" in the body delivering a continuous supply of FSP95 gene product, FSP95 peptides, or soluble FSP95 polypeptides.
  • the invention provides FSP95 fragments or analogs and derivatives of such fragments, that comprise, or alternatively consist of, one or more domains of a FSP95 protein, for example, a nuclear translocation domain.
  • FSP95 fragments or analogs and derivatives of such fragments that comprise, or alternatively consist of, one or more domains of a FSP95 protein, for example, a nuclear translocation domain.
  • a specific embodiment provides molecules comprising specific fragments of
  • FSP95 that are those fragments in the respective FSP95 protein most homologous to specific fragments of a human FSP95 protein.
  • a fragment comprising a domain of a FSP95 homolog can be identified by protein analysis methods as described in Sections 5.3.2 or 6.
  • the invention provides a fragment, derivative or analog of a FSP95 protein that has a functional RJJ subunit binding domain. See above Section 5.1 for nucleic acid sequences encoding a functional RII subunit binding domain.
  • the invention provides fusion proteins comprising an FSP95 RH subunit bmding domain that has been operatively linked to a heterologous protein. The fusion proteins are useful to cause the heterologous protein to be translocated to the nucleus.
  • a molecule in another specific embodiment, comprises one or more domains (or functional portion thereof) of a FSP95 protein but that also lacks one or more domains (or functional portion thereof) of a FSP95 protein.
  • FSP95 protein derivatives are provided that lack an RII subunit binding domain.
  • FSP95 5.4 FSP95 ANTIBODIES
  • FSP95 its fragments or other derivatives, or analogs thereof, maybe used as an immunogen to generate antibodies which immunospecif ⁇ cally bind such an immunogen.
  • Such antibodies include, but are not limited to, polyclonal, monoclonal, chimeric, single chain, Fab fragments, and an Fab expression library.
  • antibodies to a human FSP95 protein are produced.
  • antibodies to a domain e.g. , the RII subunit binding domain
  • FSP95 a domain (e.g. , the RII subunit binding domain) of a FSP95 are produced.
  • polyclonal antibodies to a FSP95 or derivative or analog may be obtained.
  • various host animals can be immunized by injection with the native FSP95, or a synthetic version, or derivative (e.g., fragment) thereof, including but not limited to rabbits, mice, rats, etc.
  • adjuvants may be used to increase the immunological response, depending on the host species, and including but not limited to Freund's (complete and incomplete), mineral 0 els such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful human adjuvants such as BCG bacille Calmette-Guerin) and corynebacterium parvum.
  • any technique which provides for the production of antibody molecules by continuous cell lines in culture may be used.
  • human antibodies may be used and can be obtained by using human hybridomas (Cote et al., 1983, Proc. Natl. Acad. Sci. U.S.A. 80:2026-2030) or by transforming human B cells with EB V virus in vitro (Cole et al., 1985, in Monoclonal Antibodies and Cancer Therapy. Alan R. Liss, pp. 77-96).
  • human hybridomas Cote et al., 1983, Proc. Natl. Acad. Sci. U.S.A. 80:2026-2030
  • transforming human B cells with EB V virus in vitro Cold-e et al., 1985, in Monoclonal Antibodies and Cancer Therapy. Alan R. Liss, pp. 77-96.
  • techniques developed for the production of "chimeric antibodies” (Morrison et al., 1984, Proc. Natl. Acad. Sci. U.S.A.
  • Antibody fragments which contain the idiotype of the molecule can be generated by known techniques.
  • such fragments include but are not limited to: the F(ab') 2 fragment which can be produced by pepsin digestion of the antibody
  • screening for the desired antibody can be accomplished by techniques known in the art, e.g. ELISA (enzyme-linked immunosorbent 0 assay).
  • ELISA enzyme-linked immunosorbent 0 assay
  • one may assay generated hybridomas for a product which binds to an FSP95 fragment containing such domain.
  • selection of an antibody that specifically binds a first FSP95 homolog but which does not specifically bind a different FSP95 homolog one can select on the basis of positive binding to the first FSP95 homolog and a lack of binding to the second 5 FSP95 homolog.
  • Antibodies specific to a domain of an FSP95 are also provided, such as a PKA RII subunit binding domain.
  • the foregoing antibodies can be used in methods known in the art relating to the identification of sperm containing FSP95 protein, separation of sperm, and the 0 localization and activity of the FSP95 polypeptides of the invention, e.g., for imaging these proteins, in diagnostic methods, measuring levels thereof in appropriate physiological samples etc.
  • All of these methods comprise the step of mixing an FSP95 protein or fragment with test compounds, allowing time for any binding to occur, and assaying for any bound complexes. All such methods are enabled by the present disclosure of substantially pure FSP95 proteins, substantially pure functional domain fragments, fusion proteins.
  • peptide libraries may be used to screen for agonists or antagonists of the FSP95 of the present invention
  • diversity libraries such as random or combinatorial peptide or nonpeptide libraries can be screened for molecules that specifically bind to FSP95.
  • Many libraries are known in the art that can be used, e.g., chemically
  • phage display libraries are described in Scott & Smith, 1990, Science 249:386-390; Devlin et al., 1990, Science, 249:404-406; Christian, et al., 1992, J. Mol. Biol. 227:711-718; Lenstra, 1992, J. Immunol. Meth. 152:149-157; Kay et a)., 1993, Gene 128:59-65; and PCT Publication No. WO 94/18318 dated August 18, 1994.
  • a benzodiazepine library (see e.g., Bunin et al.. 1994, Proc. Natl. Acad. Sci. USA 91 :4708-4712) can be adapted for use.
  • 5 Peptoid libraries (Simon et al., 1992, Proc. Nat). Acad. Sci. USA 89:9367-9371) can also be used.
  • Another example of a library that can be used, in which the amide functionalities in peptides have been permethylated to generate a chemically transformed combinatorial library, is described by Ostresh et al. (1 94, Proc. Natl. Acad. Sci. USA 91 : 11138-11142).
  • Screening the libraries can be accomplished by any of a variety of commonly known methods. See, e.g., the following references, which disclose screening of peptide 5 libraries: Parmley & Smith, 1989, Adv. Exp. Med. Biol.251:215-218; Scott & Smith, 1990, Science 249:386-390; Fowlkes et al., 1992; BioTechniques 13:422-427; Oldenburg et al., 1992, Proc. Natl. Acad. Sci.
  • screening can be carried out by contacting the library members with a FSP95 protein (or nucleic acid or derivative) immobilized on a solid phase and harvesting those library members that bind to the protein (or nucleic acid or 15 derivative).
  • FSP95 protein or nucleic acid or derivative
  • Examples of such screening methods termed “panning” techniques are described by way of example in Parmley & Smith, 1988, Gene 73:305-318; Fowlkes et al., 1992, BioTechniques 13:422-427; PCT Publication No. WO 94/18318; and in references cited hereinabove.
  • the two-hybrid system for selecting interacting 0 proteins or peptides in yeast can be used to identify molecules that specifically bind to an FSP95 protein or derivative.
  • Binding interactions between two or more components can be measured in a variety of ways.
  • One approach is to label one of the components with an easily detectable 5 label, place it together with the other component(s) in conditions under which they would normally interact, perform a separation step which separates bound labeled component from unbound labeled component, and then measure the amount of bound component.
  • the effect of a test agent included in the binding reaction can be determined by comparing the amount of labeled component which binds in the presence of this agent to the amount which binds 0 in its absence.
  • the separation step in this type of procedure can be accomplished in various ways.
  • (one of) the binding partner(s) for the labeled component can be immobilized on a solid phase prior to the binding reaction, and unbound labeled component can be removed after the binding reaction by washing the solid phase.
  • Attachment of the 5 binding partner to the solid phase can be accomplished in various ways known to those skilled in the art, including, but not hmited to, chemical cross-linking, non-specific adhesion to a plastic surface, interaction with an antibody attached to the solid phase, interaction between a ligand attached to the binding partner (such as biotin) and a ligand- binding protein (such as avidin or streptavidin) attached to the solid phase, and so on.
  • the separation step can be accomplished after the labeled component had been allowed to interact with its binding partner(s) in solution. If the size differences between the labeled component and its binding partner(s) permit such a separation, the separation can be achieved by passing the products of the binding reaction through an ultrafilter whose pores allow passage of unbound labeled component but not of its binding partners) or of labeled component bound to its partner(s). Separation can also o be achieved using any reagent capable of capturing a binding partner of the labeled component from solution, such as an antibody against the binding partner, a ligand-binding protein which can interact with a ligand previously attached to the binding partner, and so on.
  • FSP95 promoter sequences can be used advantageously to drive spermatid- specific expression of heterologous gene products.
  • a vector comprising the FSP95 promoter nucleotide sequences operably linked to a heterologous gene can be useful for gene therapy and contraception. In one embodiment, these sequences can be used for contraceptive or sterilization purposes.
  • FSP95 promoter sequences can be inserted into a vector operatively linked to a gene that will kill the cell in which it is expressed. Examples of such genes are known in the art, including, but not limited to, spermicides and toxins.
  • such a vector can be used to target cancer cells of a patient with a testes-specific proliferative disorder or cancer to inhibit growth or kill the cancer cell.
  • FSP95 promoter sequences can be used to drive spermatid-specific expression of drugs or toxins using gene therapy techniques in cells of a patient with a testes-specific proliferative disorder or cancer to inhibit growth or kill the cancer cell.
  • gene therapy techniques using promoter constructs to drive spermatid-specific expression of drugs or toxins can be used for sterilization or contraception in the testi s .
  • the genomic sequence of the FSP95 gene contains regulatory sequences both in the non-coding 5'-flanking gene of polynucleotide sequence of FIG. 3 can be assessed by any known method.
  • methods for FSP95 gene comprise the polynucleotide sequences located between the nucleotide in position -2000 and the nucleotide in position +100 of the nucleotide sequence of FIG. 3 or more preferably between positions -3000 and +200 of FIG. 3.
  • the expression of the reporter gene (for example, green fluorescent protein, luciferase, ⁇ -galactosidase, or chloramphenicol acetyl transferase) is detected when placed under the control of a biologically active polynucleotide fragment.
  • Genomic sequences located upstream of the first exon of the gene may be cloned into any suitable promoter reporter vector, such as the pSEAPBasic, pSEAP-Enhancer, p ⁇ gal-Basic, p ⁇ gal-Enhancer, orpEGFP-1 Promoter Reporter vectors available from Clontech, or pGL2- basic or pGL3-basic promoterl ⁇ ss luciferase reporter gene vector from Promega.
  • Each of these promoter reporter vectors include multiple cloning sites positioned upstream of a reporter gene encoding a readily assayable protein such as secreted alkaline phosphatase, green fluorescent protein, luciferase, or ⁇ -galactosidase.
  • the sequences upstream of the first FSP95 exon are inserted into the cloning sites upstream of the reporter gene in both orientations and introduced into an appropriate host cell.
  • the level of reporter protein is assayed and compared to the level obtained with a vector lacking an insert in the cloning site.
  • the presence of an elevated expression level in the vector containing the insert with respect to the control vector indicates the presence of a promoter in the insert.
  • Promoter sequences within the 5' non-coding regions of the FSP95 gene may be further defined by constructing nested 5' and/or 3' deletions using conventional techniques such as Exonuclease III or appropriate restriction endonuclease digestion.
  • the resulting deletion fragments can be inserted into the promoter reporter vector to determine whether the deletion has reduced or obliterated promoter activity, such as described, for example, by Coles et al. (Hum. Mol. Genet., 7:791-800, 1998). In this way, the boundaries of the promoters may be defined.
  • potential individual regulatory sites within the promoter may be identified using site directed mutagenesis or linker scanning to obliterate potential transcription factor binding sites within the promoter individually or in combination.
  • the effects of these mutations on transcription levels may be determined by inserting the mutations into cloning sites in promoter reporter vectors.
  • This type of assays are well known to those skilled in the art (WO 97/17359, US 5,374,544, EP 582 796, US 5,698,389, US 5,643,746, US5,502,176, and US 5,266,488).
  • the activity and the specificity of the promoter of the FSP95 gene can further be assessed by monitoring the expression level of a detectable polynucleotide operably linked to the FSP95 promoter in different types of cells and tissues.
  • the detectable polynucleotide may be either a polynucleotide that specifically hybridizes with a predefined oligonucleotide probe, or a polynucleotide encoding a detectable protein, o including a FSP95 polypeptide or a fragment or a variant thereof.
  • This type of assay is well known to those skilled in the art (US 5,502,176 and US 5,266,488).
  • Polynucleotides carrying the regulatory elements located both at the 5 ' end and at the 3' end of the FSP95 gene coding region may be advantageously used to control the transcriptional and translational activity of an heterologous polynucleotide of interest, 5 said polynucleotide being heterologous as regards to the FSP95 regulatory region.
  • the present invention also provides a purified, isolated, and recombinant nucleic acid comprising a polynucleotide sequence located between the nucleotide in position -2000 and the nucleotide in position +100 of the nucleotide sequence of FIG. 3, or a sequence complementary thereto or a functionally active fragment thereof. 0 By a "functionally active" fragment of the sequence of FIG.
  • polynucleotide 3 is intended a polynucleotide comprising or alternatively consisting of a fragment of said polynucleotide which is functional as a regulatory region for expressing a recombinant polypeptide or a recombinant polynucleotide in a recombinant cell host.
  • a nucleic acid or polynucleotide is "functionaT as a regulatory 5 region for expressing a recombinant polypeptide or a recombinant polynucleotide if said regulatory polynucleotide contains nucleotide sequences which contain transcriptional and translational regulatory information, and such sequences are "operably linked" to nucleotide sequences which encode the desired polypeptide or the desired polynucleotide.
  • the regulatory polynucleotides according to the invention may be 0 advantageously part of a recombinant expression vector that may be used to express a coding sequence in a desired host cell or host organism.
  • FSP95 gene regulatory sequences can, for example, be utilized for the treatment of proliferative disorders such as testicular cancer. Such treatment can be administered, for example, in the form of gene replacement therapy.
  • one or more copies 01 a normal FSP95 gene or a portion of the FSP95 gene that directs the production of a heterologous gene product that is toxic to the cell may be inserted into the appropriate cells within a patient, using vectors that include, but are not limited to, adenovirus, adeno- associated virus, and retrovirus vectors, in addition to other particles that introduce DNA into cells, such as liposomes.
  • gene replacement therapy techniques should be capable delivering FSP95 gene sequences to testis-specific cell types within patients.
  • techniques for delivery involve direct administration of such FSP95 gene sequences to the site of the cells in which the FSP95 nucleic acid o sequences are to be expressed.
  • cells preferably autologous cells
  • the expression of the heterologous 5 gene sequences is controlled by the appropriate FSP95 gene regulatory sequences to allow such expression in testes cells.
  • the cells to be administered are non-autologous cells, they can be adrninistered using well known techniques that prevent a host immune response against the introduced oells from developing.
  • the cells may be introduced in an encapsulated form which, while allowing for an exchange of components with the 0 immediate extracellular environment, does not allow the introduced cells to be recognized by the host immune system.
  • motif analysis identified three PKA RII subunit binding domains at amino acids 318-335 (LKKVLLKHAKEVVSDLID), 368-385 (QKATDL ⁇ AMLRKLYNNM), and 671-688 5 (EHLM ⁇ SVMKLCV ⁇ AKSC) of FIG.3.
  • the present invention encompasses the nucleic acid sequences encoding the FSP95 PKA RII subunit binding domains sequences, FSP95 PKA RII subunit binding domain polypeptides, and methods for using the PKA RII subunit binding domain peptides to inhibit sperm motility for use in contraceptive therapy.
  • the invention also encompasses isolated nucleic acid molecules comprising the D ⁇ A sequence of the PKA RII subunit binding domains described above in operative association with a nucleic acid encoding heterologous polynucleotide.
  • the invention also encompasses isolated fusion proteins comprising the FSP95 consensus PKA RII subunit binding domains operatively associated with a heterologous polypeptide.
  • the FSP95 PKA RII subunit binding domain sequences of the invention can be used, for example, in gene targeting heterologous sequences to the sperm.
  • the invention further encompasses the use of the promoter sequences described above in gene targeting heterologous sequences specifically to the sperm fibrous sheath.
  • the invention also encompasses the use of FSP95 promoter sequences either alone, respectively, in which the FSP95 promoter sequences can be used to drive spermatid-specific expression of drugs or toxins using gene therapy techniques in cells of a patient with a testes-specific proliferative disorder or cancer to inhibit growth or kill the cancer cell.
  • gene therapy techniques using promoter constructs either alone, or in combination with the nucleic acid sequences encoding the PKA RII subunit binding domain set forth in amino acids 318-335 (LKKNLLKHAKEWSDLID), 368-385 (QKATDIMDAMLRKLY ⁇ NM), and 671-688 (EHLM ⁇ SVMKLCVIIAKSC) of FIG. 3, respectively, can be used to drive spermatid- specific expression of drugs or toxins can be used for sterilization or contraception in the testis.
  • diseases and disorders for gene therapy diseases that can be treated or prevented by the methods of the present invention include, but are not limited to: diseases and disorders involving a fertility or infertility, deficiency in cell proliferation or in which cell proliferation is desired for treatment or prevention, and that can be treated or prevented by introduction of a heterologous gene in a testes-specific manner, include, but are not limited to, degenerative disorders, growth deficiencies, hypoprohferative disorders, physical trauma, lesions, and wounds; for example, to promote wound healing, or to promote regeneration in degenerated, lesioned or injured tissues, etc.
  • testicular disorders are treated.
  • Other disorders that are contemplated within the scope of the invention are fertility disorders.
  • the subject is preferably an animal, including, but not limited to, animals such as foxes, rabbits, rodents, cows, pigs, horses, chickens, cats, dogs, etc., and is preferably a mammal, and most preferably human. In a specific embodiment, a non-human mammal is the subject.
  • Formulations and methods of administration that can be employed wnen me therapeutic comprises a nucleic acid are described in Sections 5.1-5.6 above; additional appropriate formulations and routes of administration can be selected from among those described hereinbelow.
  • Various delivery systems are known and can be used to administer a therapeutic of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the therapeutic, receptor-mediated endocytosis (see, e.g., Wu and Wu, 1987, J. Biol. Chem. 262:4429-4432), construction of a therapeutic nucleic acid as part of a retroviral or other vector, etc.
  • Methods of introduction include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • the compounds may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous Unings (e.g., oral mucosa, rectal and intestinal ucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
  • compositions of the invention may be desirable to administer the pharmaceutical compositions of the invention locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion during surgery, topical application, e.g., in conjunction with a wound dressing after surgery, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • administration can be by direct injection at the site (or former site) of a malignant tumor or neoplastic or pre-neoplastic tissue.
  • the therapeutic can be delivered in a vesicle, in particular a liposome (see Langer, Science 249:1527-1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.)
  • the therapeutic can be delivered in a controlled release system.
  • a pump may be used (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng.
  • polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Florida (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, J. Macromol. Sci. Rev. Macromol. Chem. 23:61 (1983); see also Levy et al., Science 228:190 (1985); During et al., Ann. Neurol.
  • a controlled release system can be placed in proximity of the therapeutic target, i.e., the testes, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, 1984, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138).
  • the nucleic acid can be administered in vivo to promote expression of its encoded protein, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see US 4,980,286), or by direct injection, or by use of microparticle bombardment (e.g., a gene gun; Biolistic, Dupont), or coating with lipids or cell-surface receptors or transfecting agents, or by administering it in linkage to a homeobox-like peptide which is known to enter the nucleus (see e.g., Joliot et al., 1991, Proc. Natl. Acad. Sci. USA 88:1864-1868), etc.
  • a nucleic acid therapeutic can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination.
  • compositions comprise a therapeutically effective amount of a therapeutic, and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in a mals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • Water is a preferred carrier when the pharmaceutical composition is administered intravenously.
  • Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin.
  • Such compositions will contain a therapeutically effective amount of the therapeutic, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient.
  • the formulation should suit the mode of administration.
  • the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • the therapeutics of the invention can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
  • the amount of the therapeutic of the invention which will be effective in the treatment of a particular disorder or condition will depend on the nature of the disorder or condition, and can be determined by standard clinical techniques.
  • in vitro assays may optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances.
  • suitable dosage ranges for intravenous administration are generally about 20-500 micrograms of active compound per kilogram body weight.
  • Suitable dosage ranges for intranasal administration are generally about 0.01 pg/kg body weight to 1 mg/kg body weight.
  • Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • Suppositories generally contain active ingredient in the range of 0.5% to 10% by weight; oral formulations preferably contain 10% to 95% active ingredient.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • FSP95 fibrous sheath protein of 95 kDa
  • AKAPs sperm A-kinase anchor proteins
  • FSP95 is both auto- and iso-antigenic in humans as it is recognized by sera containing antisperm antibodies from infertile men and women.
  • the 853 amino acid residue protein has a calculated molecular weight of 94.6 kDa and a pi of 6.0 and contains multiple potential phosphorylation sites for protein kinase C and casein kinase II as well as one tyrosine kinase phosphorylation site at amino acid 435.
  • Northern analysis detected a single transcript of ⁇ 3.0 kb, and Northern dot blot analysis of 50 human tissues, including 7 fetal tissues, revealed FSP95 iriRNA expression only in testis.
  • FSP95 iriRNA expression revealed FSP95 iriRNA expression only in testis.
  • the protein was localized to the ribs of the fibrous sheath in the principal piece of sperm tail. The molecule showed clear evidence of generating more acidic isoforms during capacitation.
  • motile sperm were prepared by the swim-up method for 60 min in BWW medium (Irvine Scientific, CA) containing no human serum albumin (Yanagimachi et al, 1979, Fertil. Steril. 31:562-574). Motile spermatozoa were subsequently collected from the supernatant and incubated in BWW medium supplemented with 3.0% human serum albumin (HSA) (Sigma, St. Louis, MO) at 37°C in 5% C0 2 for 6 hour to induce capacitation (Luconi et al, 1996, Biol. Reprod. 55:207-216, Fenichel et al., 1996, Biol. Reprod.
  • HSA human serum albumin
  • Fresh and capacitated spermatozoa were solubilized in a lysis buffer containing 9.8 M urea, 2% octyl- ⁇ -D-glucopyranoside (OBG) (ESA Inc, MA), 2% (v:v) ampholines, 100 mM dithiothreitol (Bio Rad, CA), 5 mM iodoacetamide (Sigma, MO), 5 mM EDTA, and four protease inhibitors - 2 mM PMSF (Sigma, St. Louis, MO), 3 mg/ml TLCK (Boehringer Mannheim, IN), 1.46 mM pepstatin A (Sigma), and 2.1 mM leupeptin (Sigma).
  • OBG octyl- ⁇ -D-glucopyranoside
  • ampholines 100 mM dithiothreitol (Bio Rad, CA), 5 mM iodoacetamide (Sigma, MO), 5 mM EDTA,
  • sperm were solubilized in the lysis buffer (Naaby-Hansen, 1997, Biol. Reprod. 56:771-787).
  • the solubilized proteins were separated by 2-D SDS-PAGE and stained with silver (Hochstrasser et al., 1988, Anal. Biochem. 173:424-435).
  • the proteins were electrotransferred to nitrocellulose membranes (Towbin et al., 1979, Proc. Natl. Acad. Sci.
  • RC-20 is a well characterized monoclonal antibody with a specificity for phosphotyrosine previously documented (Ruff-Jamisson et al., 1993, Science 261:1733-1736, Burks et al., 1995, Science 269:83-86).
  • sperm proteins were extracted and separated by a 2-D SDS-PAGE system as described earlier (Naaby-Hansen et al., 1997, Biol. Reprod. 56:771- 787). Following electrophoretic transfer of the proteins, the membranes were rinsed in 0 phosphate buffer saline (PBS) (pH 7.4) and blocked with 5% dry milk in PBS-Tween (10 mM PBS with 0.05% Tween 20). The blots were then incubated with test serum diluted 1 : 1000 at 4°C overnight.
  • PBS phosphate buffer saline
  • a horseradish peroxidase conjugated goat antihuman IgG/IgM secondary antibody (Jackson ImniunoResearch Lab, PA) was then incubated with the blots for 1 h at a 1 :5000 dilution in PBS-Tween and the immunoreactive spots were visualized by enhanced chemilummescence using the manufacturer's protocol (Amersham Corp., UK).
  • a completely degenerate deoxyinosine containing sense primer (5'-A/T-C/G- I GTI TT-C/T TT-C/T AA-C/T TT-C/T A T/C-TI A/C-GI-3') was designed from one of the microsequences obtained by mass spectrometry, peptide number 6 (SVFFNFI LR), and the
  • oligonucleotide was synthesized by GIBCO BRL (Life Technologies, CA). Using this forward primer and an adapter primer, a 3' rapid amplification of cDNA ends (RACE) polymerase chain reaction (PCR) was performed using 0.25 ng of human testicular Marathon ready cDNA (Clontech, CA) in a 25 ⁇ l assay system for 40 cycles. Thermal cycling was done in a MJ Research (Watertown, MA) thermal cycler (PTC-200 DNA
  • the 3' clone contained a 786 bp open reading frame and a 218 bp untranslated region.
  • the 5' end of the cDNA was also amplified by 5' RACE PCR from the same template using an adapter primer and an antisense 3' gene specific primer (5'-AGC CTG GGG GGA GAA GAG GCC AAC GOT C-3 * ) which was 118 bp downstream from the 5' end of the 1.0 kb 3' clone.
  • a product of 2081 bp was obtained and cloned into the
  • a Northern blot containing 2 ⁇ g of poly(A)+ RNA from eight selected human tissues and a normalized RNA dot blot containing 89 to 514 ng of mRNA from 50 different human tissues were obtained from Clontech.
  • the Northern blot was probed with a
  • Probes were prepared by random oligonucleotide prime labeling (Feinberg and Vogelstein, 1983, Anal. Biochem. 132:6-13). Hybridization was performed in ExpressHyb solution (Clontech) at 68 °C for 1 h followed by three washes in 2x SSC, 0.05% SDS at room temperature and two washes in O.lx SSC.
  • FSP95 FSP95 cDNA
  • ORF human testicular Marathon ready cDNA
  • Primers were designed to create an Ndel site at the 5 * end and an Xhol site at the 3' end of the PCR product.
  • the amplified DNA was Iigated into the Ndel-Xhol sites of the pET28b expression vector (Novagen, WI).
  • the resulting construct appended 28 amino acids from the vector including six residues of histidine tag on either side of the protein.
  • the FSP95 coding sequence was preceded by the promotor for phage T7 RNA polymerase, an initiator ATG and a six consecutive His codons.
  • the recombinant vector was introduced into the Escherichia coli strain NovaBlue(DE3) cells, which contains a chromosomal copy of T7 RNA polymerase under the control of the lac promotor.
  • the expression plasmid was sequenced at the 5' and 3' end to verify the reading frame of the construct. A single positive colony was used to inoculate 10 liters of LB broth with 30 ⁇ g/ml kanamycin in New Brunswick Scientific fermentor (New Brunswick, NJ) and the culture was grown at 37 C C until the A coo was 0.6.
  • the urea dissolved supernatant obtained at 15000 x g for 15 min was loaded to an Ni 2* activated His-Binding resin column (Novagen, WI) following manufacturer's protocol, and the recombinant protein was eluted with 300 mM immidazole in lx binding buffer containing 6 M urea.
  • the affinity purified recombinant protein was further purified by preparative SDS-PAGE to remove some lower molecular weight breakdown products of the full length form.
  • ⁇ g of the recombinant protein was subjected to 10% SDS-PAGE, and the protein was electrophoretically transferred to nitrocellulose membrane (41).
  • the membrane was cut into strips (each containing - 150 ng), blocked with 5% dry fat-free milk in PBS-Tween (10 mM PBS, pH 7.4, 0.05% Tween 20) for 1 h and incubated with rat antisera at 1:5000 dilution in the blocking buffer for 1 h.
  • Immunodetection was performed with horseradish peroxidase conjugated goat anti-rat IgG (Jackson ImmunoResearch, PA) at 1 :5000 dilution in blocking buffer and visualized with a chromogenic substrate, diaminobenzidine (Sigma) in H 2 0 2 (47).
  • a chromogenic substrate diaminobenzidine (Sigma) in H 2 0 2 (47).
  • percoll washed sperm were solubilized in lysis buffer containing urea and OBG (see sperm preparation) and subjected to 2-D SDS-PAGE analysis (35). Following electrophoretic transfer of the proteins to nitrocellulose, the membrane was blocked and probed with the primary and secondary antibodies as above.
  • rat antisera against the rFSP95 were diluted 1 : 1 with swim-up medium and decomplemented at 56 °C for 30 min before use.
  • Guinea pig serum which was used as the source of complement was absorbed twice with sperm for 30 min at 4°C.
  • the micro sperm immobilization assay (Isojima and Koyama, 1989 Arch. Androl.
  • the sperm immobilization value was calculated by dividing the percentage of motile sperm in the control (inactivated complement) by that in the test sera with active complement. When the SIV is 2 or more, the test serum is judged as positive for sperm immobilizing antibodies.
  • the motile capacitated sperm were air dried onto poly-1-lysine coated slides (Polysciences, PA), permeabilized in methanol at - 20°C for 30 min, air dried and blocked in 10% normal goat serum in PBS-Tween (10 mM phosphate buffer saline with 0.01% Tween 20) for 30 min.
  • the sperm were then incubated with a 1:100 dilution of the primary antibody (rat anti rFSP95) in the blocking buffer for 2 h at 37°C followed by incubation with the fluorescence (Cyanine, Cy3) conjugated anti-rat (IgG) secondary antibody (Zymed Lab, CA) at 1: 100 dilution in the blocking buffer for 1 h at 37°C.
  • cDNA contained a 2559 bp open reading frame with untranslated regions of 162 bp at the 5' end, 218 bp at the 3' end, and a polyadenylation signal (ATTAAA) (Juretic and Theus, 1991, FEBS Lett.
  • the open reading frame encodes a protein of 853 amino acids with a predicted molecular weight of 94.6 kDa and a pi of 6.0. All ofthe I8 tryptic peptides obtained by microsequencing the 95 kDa protein spot were recovered in the predicted amino acid sequence of the molecule (Fig. 3, underlines), validating that the protein originally identified and cored from the gel had been cloned.
  • 25:217-221) demonstrated the presence of five potential N-linked glycosylation sites (amino acid 87, 117, 180, 502, 763), fifteen potential casein kinase II phosphorylation sites (amino acid 21, 34, 52, 102, 109, 120, 223, 280, 440, 448, 549, 659, 691, 713, 816), eleven possible protein kinase C phosphorylation sites (amino acid 2, 89, 102, 217, 223, 236, 303, 367, 408, 484, 597), eight myristoylation sites (amino acid 68, 116, 346, 366, 648, 722, 724, 814), and one tyrosine kinase phosphorylation site at amino acid 435.
  • BLAST Altschul et al., 1990, J. Mol. Biol. 215:403-410) and FASTA (Pearson and Lipman, 1988, Proc. Natl. Acad. Sci. USA 85:2444-2448) revealed that the human sperm FSP95 had closest amino acid identity to a mouse sperm fibrous sheath AKAP, precursor of mouse AKAP82 (pro-mAKAP82) (identity: 33.6%; similarity: 42.5%), and to a human sperm fibrous sheath AKAP, precursor of human AKAP82 (pro-hAKAP82) (identity: 32.4%; similarity: 39.4%) (Turner et al., 1998, J. Biol. Che .
  • rat antibody to rFSP95 recognized both the rFSP95 and the "native" FSP95 present in sperm extracts and stained the 95 kDa spot at a pi ⁇ 5.3 which was originally microsequenced (Fig. 6C and 6D).
  • Preimmune rat sera did not react with either rFSP95 or with sperm proteins.
  • FSP95 The intracellular distribution of FSP95 was examined at the ultrastructural level by post-embedding immunolabelling of ultrathin sections of washed ejaculated human spermatozoa.
  • Gold particles indicating the localization of FSP95 were associated with the entire thickness of the fibrous sheath in both longitudinal and cross sections. The gold particles were observed over the circumferential ribs while the central zone of the longitudinal columns (LC) remained unstained (Fig. 8A and 8B, indicated by arrows). No label was associated with the outer dense fibers or the axoneme. Only a rare gold particle was observed in sections exposed to preimmune control rat sera (Fig. 8C and 8D).
  • FSP95 is a capacitation induced tyrosine phosphorylated protein
  • FSP95 a novel human sperm antigen
  • fibrous sheath localization and calculated molecular weight of 95 kDa In order to characterize FSP95 from a 2-D SDS- PAGE protein spot derived microsequence, the 3' cDNA of the molecule was amplified using a single gene-specific inosine containing primer and an adapter primer from human testicular Marathon cDNA. The obtained cDNA revealed the presence of 4 FSP95 microsequenced peptides embedded in its open reading frame.
  • the 5' end of the cDNA was then similarly cloned by 5' RACE revealing two in frame stop codons upstream of the most 5 ' methionine identified. This indicates that the reported cDNA represents the complete coding sequence of FSP95.
  • the translation start site also conforms to the essential Kozak consensus sequence. Embedded within the deduced amino acid sequence are all 18 microsequenced peptides obtained from the FSP95 protein spot (Table 1; Fig. 3) validating that the protein spot originally cored was cloned. The success of this cloning strategy using a single microsequence derived primer is important because it was completed in only about three weeks.
  • tyrosine kinase The presence of a tyrosine kinase has previously been demonstrated in ejaculated human spermatozoa and in the mid-piece tail region of boar spermatozoa using antibodies against t y rosine kinase purified from boar male germ cells (Berruti and Borgonovo, 1996, J. Cell. Sci. 109:851-858).
  • the decrease in protein phosphorylation of tyrosine residue observed in capacitated sperm in presence of genistein below that of the uncapacitated cells (Fig. 1) is likely due to the presence of phosphatases, perhaps activity of the calmodulin-dependent protein phosphatase calcineurin (Tash et al., 1988, J. Cell Biol.
  • FSP95 as a tyrosine kinase substrate and its similarity to sperm AKAP may suggest possible interrelationship between PK-A and tyrosine kinase signaling pathways, because tyrosine phosphorylation and capacitation in other mammals has been shown to be upregulated by a cAMP/PK-A dependent pathway (Nisconti and Kopf, 1998, Biol. Reprod. 59:1-6, Galantino-Homer et al., 1997, Biol. Reprod. 56:707-719, Visconti et al., 1995, Development 121:1139-1150).
  • FSP95 is both- iso- and auto-antigenic. This finding is in concert with the remarkable testis-specificity observed for expression of the FSP95 transcript.
  • Many sperm proteins are antigenic in nature because they are tissue specific and do not appear until puberty when meiosis is initiated and sperm specific genes begin to be transcribed (Tung et al., 1985, in: The Autoimmune Diseases (Rose and MacKay, eds.) pp. 537-590, Academic Press, New York, NY). During the induction of self tolerance during the neonatal period
  • FSP95 is not a candidate contraceptive vaccinogen because both the micro sperm immobilization assay and immunofluorescence indicated that it is not on the sperm surface. Instead, 0 immunocytochemistry using antisera against rFSP95 localized FSP95 to the cytoplasm of the principal piece of the tail.
  • Immunoelectronmicroscopy identified the antigen in association with the ribs of the fibrous sheath (Fig. 8), which are believed to be involved in defining the shape of the flagellar beat (Eddy and O'Brien, 1994, in: The Physiology of Reproduction, (Knobil 5 and Neill, eds.) Vol. 1, pp. 29-77, Raven Press, New York, NY). In demembranated mouse spermatozoa, it has been demonstrated that sliding of the fibrous sheath towards the head is accompanied by extrusion of microtubules towards the distal end (Si and Oknno, 1993, Exp. Cell Res. 208:170-174).
  • Anomalies in fibrous sheath structure particularly disorganization of components and asymmetrical location of the longitudinal columns have been shown to be associated with severe sperm immobility related to sterility and flagella dyskinesia respectively (Chemes et al., 1987, Fertil. Steril. 48:664-669, Serres et al., 1986, Cell Motil. Cytoskeleton 6:68-76).
  • mutations of the genes encoding fibrous sheath proteins may be related to the disorganization of fibrous sheath structure leading to flagellar dyskinesia. Further studies on disruption of the FSP95 gene would clarify its role in fibrous sheath organization as well as aid in evaluating the importance of tyrosine phosphorylation of FSP95 in hyperactivation and capacitation.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Toxicology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Reproductive Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Endocrinology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Cell Biology (AREA)
  • Immunology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
EP00914482A 1999-02-01 2000-02-01 Methoden und zusammensetzungen zur regulierung der fruchtbarkeit Withdrawn EP1156819A4 (de)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US11803099P 1999-02-01 1999-02-01
US118030P 1999-02-01
US11899599P 1999-02-08 1999-02-08
US118995P 1999-02-08
PCT/US2000/002675 WO2000044395A1 (en) 1999-02-01 2000-02-01 Methods and compositions for modulating fertility

Publications (2)

Publication Number Publication Date
EP1156819A1 true EP1156819A1 (de) 2001-11-28
EP1156819A4 EP1156819A4 (de) 2003-01-02

Family

ID=26815894

Family Applications (1)

Application Number Title Priority Date Filing Date
EP00914482A Withdrawn EP1156819A4 (de) 1999-02-01 2000-02-01 Methoden und zusammensetzungen zur regulierung der fruchtbarkeit

Country Status (5)

Country Link
EP (1) EP1156819A4 (de)
JP (1) JP2003525021A (de)
AU (1) AU772403B2 (de)
CA (1) CA2361271A1 (de)
WO (1) WO2000044395A1 (de)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1390387A4 (de) * 2001-04-20 2004-12-08 Ludwig Inst Cancer Res Hodenkrebsantigene
CL2010000261A1 (es) 2010-03-23 2010-06-11 Laboratorios Andromaco S A 8 5% Peptidos derivados del veneno de latrodectus mirabilis, secuencias que lo codifican, procedimiento de produccion de dichos peptidos, composiciones farmaceuticas que los comprenden, y uso anticonceptivos de las mismas.

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001054477A2 (en) * 2000-01-25 2001-08-02 Hyseq, Inc. Novel nucleic acids and polypeptides
EP1130094A2 (de) * 1999-07-08 2001-09-05 Helix Research Institute Primer zur Synthese von vollständigen cDNA Klonen und ihre Verwendung

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4795634A (en) * 1986-03-14 1989-01-03 Aphton Corporation Method for contraception by immunization against the zona pellucida
US5346990A (en) * 1987-04-08 1994-09-13 Cytogam, Inc. Sex-associated membrane proteins and methods for increasing the probability that offspring will be of a desired sex
US5641487A (en) * 1989-06-12 1997-06-24 The Government Of The United States Of America As Represented By The Secretary Department Of Health And Human Services Contraceptive vaccine based on alloimmunization with zona pellucida polypeptides
US5525592A (en) * 1993-11-30 1996-06-11 Eric R. Lacy Alkaline gland fluid proteins and methods of enhancing and inhibiting sperm motility
US5637300A (en) * 1994-08-03 1997-06-10 Dunbar; Bonnie S. Contraceptive vaccine comprising a glycosylated 55 kD zona pellucida protein immunogen and method of use of the same in contraception

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1130094A2 (de) * 1999-07-08 2001-09-05 Helix Research Institute Primer zur Synthese von vollständigen cDNA Klonen und ihre Verwendung
WO2001054477A2 (en) * 2000-01-25 2001-08-02 Hyseq, Inc. Novel nucleic acids and polypeptides

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
DATABASE EMBL [Online] EMBL; 17 August 1998 (1998-08-17) "ov24d10.x1 Soares testis NHT Homo sapiens cDNA clone IMAGE:1638259" Database accession no. AI081873 XP002218762 *
DATABASE EMBL [Online] EMBL; 25 September 1998 (1998-09-25) "qe03f10.x1 Soares testis NHT homo sapiens cDNA clone IMAGE:1737931" Database accession no. AI138771 XP002218763 *
LEFEVRE A (REPRINT) ET AL: "SOB1 -g, a human gene coding for a sperm protein with potential oocyte binding activity" HUMAN REPRODUCTION, (JUN 1997) VOL. 12, SUPP. [1], PP. P24-P24. PUBLISHER: OXFORD UNIV PRESS, GREAT CLARENDON ST, OXFORD, ENGLAND OX2 6DP. ISSN: 0268-1161., XP001117984 INSERM, U355, F-92140 CLAMART, FRANCE *
LEFEVRE A ET AL: "Cloning and characterization of SOB1, a new testis-specific cDNA encoding a human sperm protein probably involved in oocyte recognition." BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 259, no. 1, 27 May 1999 (1999-05-27), pages 60-66, XP002218761 ISSN: 0006-291X *
MANDAL A ET AL: "FSP95, A TESTIS-SPECIFIC 95-KILODALTON FIBROUS SHEATH ANTIGEN THAT UNDERGOES TYROSINE PHOSPHORYLATION IN CAPACITATED HUMAN SPERMATOZOA" BIOLOGY OF REPRODUCTION, SOCIETY FOR THE STUDY OF REPRODUCTION, CHAMPAIGN, IL, US, vol. 61, no. 5, 1999, pages 1184-1197, XP001029106 ISSN: 0006-3363 -& DATABASE EMBL [Online] EMBL; 23 September 1998 (1998-09-23) MANDAL: "Homo sapiens fibrousheathin I mRNA" Database accession no. AF087003 XP002218767 *
MANDAL A ET AL: "TWO NOVEL HUMAN SPERM FIBROUS SHEATH PROTEINS WHICH UNDERGO PHOSPHORYLATION DURING CAPACITATION" MOLECULAR BIOLOGY OF THE CELL, BETHESDA, MD, US, vol. 9, no. SUPPL, November 1998 (1998-11), page 66A XP001029742 ISSN: 1059-1524 *
See also references of WO0044395A1 *
VIJAYARAGHAVAN S ET AL: "ISOLATION AND MOLECULAR CHARACTERIZATION OF AKAP110, A NOVEL, SPERM-SPECIFIC PROTEIN KINASE A-ANCHORING PROTEIN" MOLECULAR ENDOCRINOLOGY, BALTIMORE, MD, US, vol. 13, no. 5, May 1999 (1999-05), pages 705-717, XP001029110 ISSN: 0888-8809 -& DATABASE EMBL [Online] EMBL; 8 October 1998 (1998-10-08) VIJAYARAGHAVAN: "Homo sapiens protein kinase A binding protein AKAP110 mRNA" Database accession no. AF093408 XP002218764 -& DATABASE EMBL [Online] EMBL; 8 October 1998 (1998-10-08) VIJAYARAGHAVAN: "Mus musculus protein kinase A binding protein AKAP110 mRNA" Database accession no. AF093406 XP002218765 -& DATABASE EMBL [Online] EMBL; 8 October 1998 (1998-10-08) VIJAYARAGHAVAN: "Bos taurus protein kinase A binding protein AKAP110 mRNA" Database accession no. AF093407 XP002218766 *

Also Published As

Publication number Publication date
AU772403B2 (en) 2004-04-29
CA2361271A1 (en) 2000-08-03
JP2003525021A (ja) 2003-08-26
AU3586800A (en) 2000-08-18
WO2000044395A1 (en) 2000-08-03
EP1156819A4 (de) 2003-01-02
WO2000044395A9 (en) 2002-05-02

Similar Documents

Publication Publication Date Title
ES2555543T3 (es) Neuquinasa, una proteína secuencia abajo de la neuregulina
ES2284502T3 (es) 22025, una nueva nucleotido ciclico fosfodiesterasa humana.
US20010010913A1 (en) Extracellular adhesive proteins
JP2000515734A (ja) ヒトcAMP依存性プロテインキナーゼインヒビターホモログ
JP2002506625A (ja) サイトカインレセプター共通γ鎖様
CA2278596A1 (en) Novel human metallothionein
US7404952B2 (en) Protein disulfide isomerase and ABC transporter homologous proteins involved in the regulation of energy homeostasis
AU772403B2 (en) Methods and compositions for modulating fertility
US20020090693A1 (en) Novel human sodium-dependent phosphate cotransporter
US20050042653A1 (en) Human transport protein homologs
US6080848A (en) Human brain associated protein
JP2002503962A (ja) ヒトタンパク質フォスファターゼ▲ii▼−c様タンパク質
US6558950B1 (en) Methods and reagents for modulating apoptosis
WO2000011164A1 (en) Human angiopoietin-3
JP2002520055A (ja) ヒトプレセニリン関連タンパク質
US20030207393A1 (en) Cbp86, a sperm specific protein
WO2000024870A2 (en) Nucleic acids and proteins of the human span-x gene and uses thereof
US20050176659A1 (en) Endophilin homologous proteins involved in the regulation of energy homeostasis
US20040072773A1 (en) Novel functions for dp214
JP2002509718A (ja) Nek1関連プロテインキナーゼ
US20030148934A1 (en) Growth factor modulators
WO2000005368A1 (en) Human calcium regulatory proteins
AU2002308110A1 (en) Protein disulfide isomerase and ABC transporter homologous proteins involved in the regulation of energy homeostasis
EP1097206A1 (de) Menschliche kalzium-regulierende proteine
WO2001053338A2 (en) Cbp86, a sperm specific protein

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20010813

AK Designated contracting states

Kind code of ref document: A1

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

AX Request for extension of the european patent

Free format text: AL;LT;LV;MK;RO;SI

A4 Supplementary search report drawn up and despatched

Effective date: 20021115

AK Designated contracting states

Kind code of ref document: A4

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

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20050901