WO2000010594A1 - The use of glial cell line-derived neurotrophic factor family-related compounds for regulating spermatogenesis and for preparing male contraceptives - Google Patents

The use of glial cell line-derived neurotrophic factor family-related compounds for regulating spermatogenesis and for preparing male contraceptives Download PDF

Info

Publication number
WO2000010594A1
WO2000010594A1 PCT/FI1999/000687 FI9900687W WO0010594A1 WO 2000010594 A1 WO2000010594 A1 WO 2000010594A1 FI 9900687 W FI9900687 W FI 9900687W WO 0010594 A1 WO0010594 A1 WO 0010594A1
Authority
WO
WIPO (PCT)
Prior art keywords
gdnf
family
compounds
related compounds
receptor
Prior art date
Application number
PCT/FI1999/000687
Other languages
French (fr)
Inventor
Hannu Sariola
Meng Xiaojuan
Mervi Salo
Maria Lindahl
Mart Saarma
Original Assignee
Hannu Sariola
Meng Xiaojuan
Mervi Salo
Maria Lindahl
Mart Saarma
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from FI981793A external-priority patent/FI981793A0/en
Priority claimed from FI990415A external-priority patent/FI990415A0/en
Priority to HU0103419A priority Critical patent/HUP0103419A3/en
Priority to AU53752/99A priority patent/AU763678B2/en
Priority to CA002338644A priority patent/CA2338644A1/en
Priority to IL14103799A priority patent/IL141037A0/en
Priority to EP99939468A priority patent/EP1105151A1/en
Priority to JP2000565914A priority patent/JP2002523374A/en
Application filed by Hannu Sariola, Meng Xiaojuan, Mervi Salo, Maria Lindahl, Mart Saarma filed Critical Hannu Sariola
Priority to PL99346298A priority patent/PL346298A1/en
Priority to SK247-2001A priority patent/SK2472001A3/en
Priority to KR1020017002234A priority patent/KR20010088793A/en
Priority to EEP200100103A priority patent/EE200100103A/en
Priority to NZ509592A priority patent/NZ509592A/en
Publication of WO2000010594A1 publication Critical patent/WO2000010594A1/en
Priority to NO20010859A priority patent/NO20010859L/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/185Nerve growth factor [NGF]; Brain derived neurotrophic factor [BDNF]; Ciliary neurotrophic factor [CNTF]; Glial derived neurotrophic factor [GDNF]; Neurotrophins, e.g. NT-3
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention is related to the use of glial cell line-derived neurotrophic factor (GDNF) family-related compounds, such as glial cell line-derived neurotrophic factor (GDNF) , GDNF-like factors and/or other compounds which act like GDNF on its receptor and/or co-receptors, for regulating and studying spermatogenesis, for inhibiting the differentiation of sperm cells, for developing male contraceptives or as a male contraceptive as well as their use for manufacturing male contraceptive compositions.
  • GDNF glial cell line-derived neurotrophic factor family-related compounds, such as glial cell line-derived neurotrophic factor (GDNF) , GDNF-like factors and/or other compounds which act like GDNF on its receptor and/or co-receptors, for regulating and studying spermatogenesis, for inhibiting the differentiation of sperm cells, for developing male contraceptives or as a male contraceptive as well as their use for manufacturing male contraceptive compositions.
  • GDNF
  • Spermatogenesis is a complex sequence of events involving both hormonal regulators and local interactions between Sertoli and germ cells (Bellve, A. R. & Zhang, W. J., Reprod. Fertil. 85, 771-793 (1989); Skinner, M. K. , Endocr. Rev. 12, 45-72 (1991); Pescovitz, 0. H., et al . , Trends Endocrinol . Metab. 5, 126-131 (1994). Parvinen, M., et al . , J. Cell Biol . 117, 629-41 (1992), Djakiew, D., et al . , Biol. Reprod. 51, 214-21 (1994), Cancilla, B.
  • the differentiation of sperm cells or spermatogenesis can be divided in three phases. In the first phase of the spermatogenesis undifferentiated stem cells from the germ line, the spermato- gonia, are divided mitotically and differentiate into sper- matocytes .
  • Spermatogonia are very resistant against different types of damages and after damage spermatogenesis starts from said cells. At present, it is not known by which mechanism the spermatogonia are maintained and secondly by which signals they are differentiated into spermatocytes .
  • Spermatogonia are located in the peripheral parts of the seminiferous tubules and are in contact with the Sertoli cells, which represent the regulating cells of the seminiferous tubules and are classified as somatic cells of the organism.
  • the spermatogonia are at first differentiated into primary and thereafter into secondary spermatocytes, whereby their chromosomes become haploid by two meiotic cell divisions after which they are called spermatides.
  • the spermatides mature into spermazoa or mature sperm cells.
  • the luteinizing hormone, the follicle stimulating hormone and testosterone are the most important hormonal regulators of spermatogenesis.
  • the cells of the germ line in the seminiferous tubules of testis are not direct targets of the hormonal regulation but the hormonal regulation is transmitted by the Sertoli cells. They modulate the hormonal signals into paracrine signals which are believed to be signal molecules and growth factors, such as stem cell factor (c-kit ligand) , insulin like growth factor- 1 and three members of the transforming growth factor- ⁇ family (transforming growth factor- ⁇ , activin and inhibin) (Bellve A. R. & Zhang . J., Reprod. Fertil. 85, 771-793 (1989); Skinner, . K. , Endocr. Rev.
  • the spermatogenetic cells can also send the signals back to the Sertoli cells (Bellve, A. R. & Zhang W. J., Reprod. Fertil. 85, 771-793 (1989); Skinner, M. K. , Endocr. Rev. 12, 45-72 (1991); Pescovitz, 0. H., et al . , Trends Endocrinol . Metab. 5, 126-131 (1994)).
  • This is indicated for example by the fact that nerve growth factor and fibroblast growth factor- 1 are expressed in the cells of the germ line and the p75 nerve growth factor receptor and fibroblast growth factor receptor are located in the Sertoli cells. Hints of auto- crine regulation of spermatogenesis also exist.
  • Bone morphoge- netic protein 8a and 8b and their receptors are expressed in the same spermatogenetic cells (Zhao, G. Q., et al . , Gene Dev . 10, 1657-1669 (1996); Zhao, G. Q., et al . , Dev. 125, 1103-1112 (1998)) .
  • the functional evidence for the autocrine regulation is still lacking.
  • GDNF glial cell line-derived neurotrophic factor
  • the glial cell line-derived neurotrophic factor (GDNF) is a member of the transforming growth factor- ⁇ superfamily (Lin, L. F., et al., Science 260, 1130-1132 (1993)).
  • GDNF maintains the dopaminergic, noradrenergic, cholinergic and motor neurons in the nervous system and also protects peripheral parasympa- thetic ciliary and sensory nerve cells (Lin, L.F., Neural Notes. 2, 3-7, (1996).
  • GDNF is expressed also in other tissues, such as embryonic kidney and testis (Suvanto, P., et al .
  • mice European Journal of Neuro- science 8, 816-822, (1996); Trupp M, et al . , Journal of Cell Biology 130, 137-148, (1996)).
  • the role of GDNF as a regulator of the differentiation of the kidney has been shown with gene knockout or gene deletion techniques in mice (Pichel, J.G., et al., Nature 382, 73-75 (1996). Because said mice die as newborn, it has not been possible to study with said deletion gene model - or other experimental models - the function of GDNF in spermatogenesis, which is initiated much later, only after birth. Studies with different model systems indicate that there is a multitude of ways for interfering with spermatogenesis and for developing male contraceptives .
  • the problem yet to be solved is how to select from the multitude of hormonal growth regulators one which can sufficiently specifically be targeted to the testis and at the same time provides a system for regulating spermatogenesis without disturbing side-effects and which system does not interfere negatively with the male mating activity.
  • mice with human GDNF were capable of mating but not capable of fertilizing the female.
  • the solution to the problem is to use glial cell line-derived neurotrophic factor family-related compounds as defined in the claims of the present invention for regulating spermatogenesis and inhibiting or preventing spermatogenesis followed by infertility and as active ingredients for manufacturing compositions useful as male contraceptives, which are both effective and without severe side-effects .
  • the first objective of the present invention is to provide an effective male contraceptive without severe, undesired side-effects by using GDNF-family-related compounds as male contraceptives.
  • Another objective of the present invention is to use the GDNF-family-related compounds as active ingredients for manufacturing compositions useful as male contraceptives .
  • One other objective of the present invention is to provide tools for regulating and studying spermatogenesis. This is facilitated by inhibiting the differentiation of sperm cells.
  • a third objective of the present invention is to provide tools, which have an effect on infertility caused by disturbed differentiation of sperm cells.
  • the objective of the present invention is also to provide tools for regulating and studying spermatogenesis as well as for inhibiting the differentiation of sperm cells.
  • the objective of the present invention is to provide transgenic animals, especially mice, which due to the testis targeted GDNF-family-related compounds provide a useful tool for studying spermatogenesis.
  • the characterstics of the present invention are as defined in the claims.
  • the present invention describes a method, whereby it is possible to study sterility caused by disturbed differentiation of sperm cells and secondly it provides a system for inhibiting the differentiation of sperm cells in a contraceptive sense, whereby the male (man) is sterile (infertile) .
  • the GDNF-transgene carrying transgenic mouse strain, the GDNF t-mouse strain is a model for infertility caused by disturbances in the differentiation of sperm cells.
  • GDNF compounds acting like, said GDNF, another cRet receptor, another GDNF-receptor activating compound or a compound, which activates the cRet receptor signal transmitting tubules in spermatogonia
  • the GDNF t-mouse model is suitable as an animal model for studying the regulation of spermatogenesis and spermatogenesis.
  • Figure la Testicular morphology in wild-type mice at 3 -weeks of age. Scale bar 100 ⁇ m.
  • Figure lb Testicular morphology in transgenic mice at 3 -weeks of age. Scale bar 100 ⁇ m. Note the undifferentiated spermatogonia formed cell cluster.
  • Figure lc Testicular morphology in wild-type mice at 8 -weeks of age. Scale bar 100 ⁇ m.
  • Figure Id Testicular morphology in transgenic mice at 8 -weeks of age. Note in d the remnants of type A spermatogonia clusters that are abundant in the young mice. Scale bar 100 ⁇ m.
  • Figure le Testicular morphology in wild-type mice at 6-months of age. Scale bar 100 ⁇ m.
  • Figure If Testicular morphology in transgenic mice at 6- months of age. Note the advanced germ cell atrophy and Leydig cell hyperplasia (star) . Scale bar 100 ⁇ m.
  • Figure lh Testicular morphology in transgenic epididymal ducts at 8-weeks. Note the lack of sperm. Scale bar 100m.
  • FIG. 2a Northern blotting for GDNF RNAs in wild-type mice at various ages . GDNF levels are strongly downregulated after the first or second postnatal week.
  • FIG. 2b Northern blotting for Ret mRNAs in wild-type mice testes at various ages. GDNF mRNA levels are strongly downregulated after the first or second postnatal week.
  • Figure 2c Northern blotting for GFRcel mRNAs in wild-type mice at various ages. GFR ⁇ l mR ⁇ A levels are strongly downregulated after the first or second postnatal week.
  • Figure 2d Northern blotting for GDNF mRNAs in transgenic testes at various ages. The hGDNF levels remain high after the first or second postnatal week.
  • Figure 2e Northern blotting for Ret mRNAs in transgenic mouse testis at various ages.
  • FIG. 2f Northern blotting for GFR ⁇ l mRNAs in transgenic testes at various ages. GFR ⁇ l RNA levels are high in the transgenic mice at all stages analysed.
  • Figure 2g depicts immunoprecipitation of GDNF from wild-type (WT) and transgenic (TG) testes at 3 and 6 weeks of age. In wild-type testes, the GDNF protein is not detectable. The control, 25 ng of reco binant human GDNF protein is shown on the right side.
  • FIG. 3a The distribution of GDNF transcripts in testes of wild-type (WT) mice by cRNA in si tu hybridisation. Sense controls did not show grain density above the background. 1- week-old wild-type testis. Scale bar 100 ⁇ m.
  • FIG. 3b The distribution of GDNF transcripts in 8 -weeks-old testis of wild-type (WT) mouse by cRNA in si tu hybridisation. Sense controls did not show grain density above the background. Scale bar 100 ⁇ m.
  • FIG. 3c The distribution of GDNF transcripts in testes of transgenic mice (TG) by cRNA in si tu hybridisation. Sense controls did not show grain density above the background. 8-weeks-old transgenic testis. Note that the spermatogonia in the transgenic mice continuously express human GDNF at high levels, but the number of these cells is clearly diminishing at 8 weeks of age. Scale bar 100 ⁇ m.
  • FIG. 3d The distribution of Ret transcripts in testes of wild-type (WT) mice by cRNA in situ hybridisation. 2-weeks-old wild-type testis. At two weeks of age, Ret expression is localised to some spermatogonia in the basal layer of the seminiferous tubules (arrows) .
  • FIG. 3e The distribution of Ret transcripts in testes of wild-type (WT) mice by cRNA in si tu hybridisation. 8 -weeks-old wild-type testis. At 8 weeks of age, Ret is expressed in both some spermatogonia and spermatides.
  • FIG. 3f The distribution of Ret transcripts in testes of transgenic mice (TG) by cRNA in si tu hybridisation. 8 -weeks- old transgenic testis. Note that the spermatogonia in the transgenic mice continuously express Ret at high levels, but the number of these cells is clearly diminishing at 8 weeks of age. In transgenic mice, Ret is expressed in the clusters of spermatogonia. Scale bar 100 ⁇ m.
  • FIG. 3g The distribution of GFR ⁇ l transcripts in testes of wild-type mice (WT) . 1- to 2-weeks-old wild-type testis. GFR ⁇ l is expressed by some spermatogonia, morphologically similar to those with Ret (arrows) .
  • FIG. 3h The distribution of GFR ⁇ l transcripts in testes of wild-type by cRNA in si tu hybridisation. 8 -weeks-old wild- type testis. GFR ⁇ l mRNA distribution follows that of Ret, but is also highly expressed by spermatocytes. Scale bar 100 ⁇ m.
  • FIG. 3i The distribution of GFR ⁇ l transcripts in testes of transgenic mice (TG) by cRNA in situ hybridisation. 8-weeks- old transgenic testis. Note that the spermatogonia in the transgenic mice continuously express human GDNF at high levels, but the number of these cells is clearly diminishing at 8 weeks of age. In transgenic mice, GFR ⁇ l is expressed in the clusters of spermatogonia. Scale bar 100 ⁇ m.
  • Figure 4a Cell proliferation in wild-type mice testes. In 3 -weeks-old testis, BrdU labelling of S-phase cells is seen only in the periphery of wild-type seminiferous tubules. Note the absence of labelling in some transsections of tubules, reflecting the segmented distribution of S-phase cells during spermatogenesis (stars) . Scale bar 100 ⁇ m.
  • Figure 4b Cell proliferation in transgenic mice testes.
  • many BrdU-labelled cells are seen in the luminal area of the seminiferous tubules, reflecting the abnormal distribution of spermatogonia (arrow) .
  • the segmental distribution of S-phase cells is not seen in the transgenic tubules.
  • Scale bar 100 ⁇ m.
  • Figure 4c Distribution of cell line proliferation indexes in wild type and transgenic testes. The number of BrdU labeled cells / 100 spermatogonia was counted in 100 transections of wild type and transgenic seminiferous tubules.
  • FIG. 4d Apoptosis in wild-type mice testes. TUNEL- labelling for apoptosis in 4-week-old mice testes. Only a few apoptotic cells are seen. Scale bar (c) 100 ⁇ m.
  • Figure 4e Apoptosis in transgenic mouse testis. TUNEL-label- ling for apoptosis in 4-week-old testes. Note the increase of labelling in spermatogonia clusters in ( Figure 4e) , as compared to wild-type ( Figure 4d) seminiferous tubules. Scale bar 100 ⁇ m.
  • FIG. 4f Live morphology in both a wild-type and transgenic seminiferous tubule. Live preparation of seminiferous tubules from 15-week-old wild-type (WT) and transgenic (TG) mice. Note the reduced diameter of the transgenic tubule and the increase of Leydig cells (arrows) around it. Scale bar 200 ⁇ m.
  • FIG. 4g Live morphology in transgenic testes. High magnification of living cells from a transgenic seminiferous tubule in an unfixed squash preparation showing large numbers of dead cells (arrow) , some of which resemble type A spermatogonia, in a Sertoli cell (arrow head) . Scale bar 10 ⁇ m.
  • GDNF glial cell line-derived neurotrophic factor family-related compounds
  • GDNF-family-related compounds comprises glial cell line-derived neurotrophic factor (GDNF) , neurturin, persephin, artemin and other similar growth factors, but also other compounds which have the same effect as GDNF-like compounds on the GDNF-receptor (s) or co-receptors.
  • Said receptor (s) or co-receptor (s) comprises Ret tyrosine kinase and GDNF-family-receptor a : s , respectively.
  • glial cell line-derived neurotrophic factor means the glial cell line-derived neurotrophic factor (GDNF) , which is a member of the transforming growth factor- ⁇ family (Lin, L. F., et al . , Science 260, 1130-1132 (1993).
  • GDNF maintains the dopaminergic , noradrenergic, cholinergic and motor neurons in the nervous system and also protects peripheral, parasympathetic, ciliary and sensory nerve cells (Lin, L.F., Neural Notes. 2, 3-7 (1996)).
  • GDNF is expressed also in other tissues, such as embryonic kidney and testis (Suvanto P, et al .
  • GDNF-like factors or GDNF-like compounds mean growth factors which have a similar structure or act like GDNF on its receptor or co-receptors.
  • GDNF-like factor or "GDNF-like compound” above all relates to three GDNF-like growth factors, persephin, artemin and neurturin, which are described in (Milbrandt, J., et al . , Neuron 20, 1-20 (1998); Kotzbauer P. T., et al . , Nature 384, 467-470 (1996)).
  • GDNF-family All four growth factors of the GDNF-family are structurally similar and cRet receptor tyrosine kinase (Durbec, P., et al., Nature 381, 789-793 (1996); Buj-Bello, A., et al . , Nature. 387, 721-724, (1997)) acts as the common signal transmitting receptor of GDNF, artemin, persephin and neurturin.
  • the "compounds which act like said GDNF” means compounds which act as GDNF on the receptor transmitting signals of GDNF or co-receptors thereof.
  • receptor means GDNF-family binding substances, i.e. substances or compounds belonging to the GDNF-family-related compounds which are capable of transmitting the signals of GDNF-family-related compounds.
  • receptor above all means the GDNF-family signal mediatory receptor is cRet receptor tyrosine kinase.
  • co-receptors means receptors, which do not transmit the signals of GDNF-family related compounds, but activate the receptor transmitting the signals of the GDNF-family-related compounds.
  • Such compounds are above all the GDNF-family receptor a : s (GFRcn) , which is a new receptor class, the members of which are called GDNF-family-receptor c.s (GFRc . These also belong to the GDNF, persephin, artemin and neur- turin signalling receptor complex.
  • GFR ⁇ l-4 Jing, S., et al . , Cell 85, 9-10 (1996), Jing, S. Q., et al., J. Biol.
  • Chem. 272, 33111-3311 7 (1997); Treanor, J. J., et al., Nature. 382, 80-83, (1996); Suvanto, P., et al., Human Molecular Genetics. 6, 1267-1273, (1997)) of said family are known. They may transmit signals independently, but they are essential for ligand binding and cRet activation.
  • concentration of GDNF-family-related compounds in such amounts that the differentiation of sperm cells is prevented, spermatogenesis can be regulated and infertility caused by disturbed differentiation of sperm cells or that the male is rendered unable to fertilize the female means a concentration of GDNF-family-related compounds, which is approximately 50 - 5000, preferably 50 - 500, most preferably 50 - 100 times higher than the concentration of endogenous GDNF in the testis of said male. Said amount can be calculated from the test results obtained when measuring the GDNF mRNA levels of transgenic mice shown in the Northern blots ( Figures 2) and which indicate an elevated amount of mRNA from which the expressed protein levels can be calculated.
  • the amount of GD ⁇ F in transgenic mice is 50-500 times higher than the endogenic level in three to six week old mice, but only a little bit higher than in new born mice, when the GD ⁇ F levels are at their highest and no spermatogenesis is presented. From these results it is possible for those skilled in the art to calculate the amounts needed for obtaining the desired effects in man.
  • derivatives means iso ers of GD ⁇ F-family-related compounds . It also includes hybrid molecules constructed based on the structures of the compounds GD ⁇ F, persephin, neurturin and artemin, as well as truncated forms, complexes and fragments of said compounds. The only prerequisite for said compounds is that they still have substantially the same properties and effects as the compounds listed above, determined by the same methods as described in the present inven- tion. Naturally, the term “derivatives” includes derivatives of the compounds listed above in a more conventional sense, e.g.
  • amino- and/or hydroxy- groups may be substituted by alkylation, esterification, etherification and amidation with e.g. alkyl groups, including methyl, ethyl, propyl, etc.
  • alkyl groups including methyl, ethyl, propyl, etc.
  • Glial cell line-derived neurotrophic factor is a potent survival factor for various sets of neuronal cells, it regulates kidney differentiation (Lin, L. F., et al . , Science 260, 1130-1132 (1993); Tomac, A., et al . , Nature 373, 335-339 (1995); Winkler, C, et al . , J. Neurosci . 16, 7206-7215 (1996), Oppenheim, R. W. , et al . , Nature 373, 344-346 (1995), Nguyen, Q. T., et al .
  • GDNF Glial cell line-derived neurotrophic factor
  • GDNF a distant member of the transforming growth factor- ⁇ (TGF- ⁇ ) superfamily (Lin, L. F., et al . , Science 260, 1130- 1132 (1993)) is a potent survival factor for dopaminergic neurones of the substantia nigra, and some other sets of neurones in the central and peripheral nervous systems (Lin, L. F., et al., Science 260, 1130-1132 (1993); Tomac, A., et al., Nature 373, 335-339 (1995); Winkler, C, et al . , J. Neurosci. 16, 7206-7215 (1996), Oppenheim, R. W., et al .
  • TGF- ⁇ transforming growth factor- ⁇
  • GNDF also acts as a signalling molecule in ureteric branching during kidney morphogenesis (Schuchardt, A., et al . , Nature 367, 380-383 (1994); Pichel, J. G., et al . , Nature 382, 73-75 (1996)) . It is synthesised by several neuronal and non-neu- ronal embryonic organs including the embryonic and postnatal testis (Trupp, M. , et al . , J. Cell Biol. 130, 137-148 (1995); Hellmich, H. L., et al . , Mech. Dev. 54, 95-105 (1996); Suvanto, P., et al .
  • the signalling receptor-complex for GDNF includes Ret receptor tyrosine kinase (Durbec, P., et al . , Nature 381, 789-793 (1996); Trupp, M., et al . , Nature 381, 785-789 (1996)) and a member of the glycosylphosphatidylinositol- linked co-receptors, GDNF-family-receptor a : s (GFRo.1-4) (Jing, S., et al., Cell 85, 9-10 (1996); Jing, S. Q., et al . , J. Biol. Chem.
  • mice exhibit remarkably similar phenotypes with absent or hypodysplastic kidneys and severe defects in enteric innervation (Schuchardt, A., et al . , Nature 367, 380-383 (1994); Pichel, J. G., et al., Nature 382, 73-75 (1996)). As they die during the first postnatal day, spermatogenesis could not be analysed in these mutant mice.
  • the present inventors studied transgenic mice with targeted overexpression of GDNF in testicular germ cells.
  • the transgenic males were infertile and showed an early arrest in the differentiation of spermatogenic cells, the type A spermatogonia. They formed clusters within seminiferous tubules, finally resulting in testicular atrophy.
  • the data obtained showed a novel function for GDNF as a paracrine inhibitory signal during early spermatogenesis.
  • GDNF is believed to regulate the pool of spermatogonia that is gradually committed to sperm differentiation.
  • the activation of the GDNF signalling cascade possibly provides us with means to develop contraceptives for man.
  • the effects of GDNF on spermatogenesis should also be taken into consideration, when GDNF-like molecules are used in clinical trials of neurodegenerative disorders, such as Parkinson's disease.
  • GDNF targeting to testis provides a new animal model for male infertility.
  • the activation of the GDNF signalling cascade by GDNF, Ret agonists or downstream substrates possibly provides us with means whereby sperm production in man could be prevented and male contraceptives developed.
  • the effects of GDNF on spermatogenesis should also be taken into consideration, when GDNF-like molecules are designed for and used in clinical trials of neurodegenerative disorders, such as Parkinson's disease .
  • transgenic mice overexpressing human GDNF under human translation elongation factor-1 (EF-1) promoter (Mizushima, S. & Nagata, S., Nucleic Acids Res. 18, 5322 (1990)), which targets the transgene expression specifically to testicular germ cells (Furuchi, T., et al . , Development 122, 1703-1709 (1996) ) .
  • EF-1 human translation elongation factor-1
  • Other promoters targetting the transgene expression specifically to testicular germ cells can be used as well.
  • Four independent transgenic founders were analysed, two males CIO, C12 and two females S6, E19 with transgene copy numbers 10, 3, 20, and 4, respectively.
  • the male founders CIO and C12 were infertile and further analysis of the male phenotype was done with the offspring of the two female founders S6 and E19.
  • the transgenic mice developed normally to adulthood and showed a similar phenotype. The body weight was normal, and they did not have any defects in the weights and morphology of paren- chymal organs (data not shown), except in the testis. After four weeks of age, a reduction in weight of testes was observed in the transgenic mice as compared ' to wild-type controls (Table 1) . All transgenic males in the offspring of the female founders S6 and E19 were infertile.
  • transgenic males were caged with wild-type FVB and NMRI females in three to four day intervals.
  • the wild-type females mated with transgenic males at normal frequency, as shown by the appearance of vaginal plugs, but none became pregnant.
  • 20 transgenic males at breeding age were subjected to wild-type FVB and NMRI females in short term breeding test.
  • approximately 200 females from both strains were plugged, but none became pregnant.
  • Strain dependence of the infertile phenotype was excluded by crossbreeding the transgenic FVB mice to NMRI background.
  • those of the FI generation were also infertile.
  • testicular histology of one-week-old transgenic mice was normal . From two to three weeks of age (Fig. la, lb) , they showed clusters of cells morphologically similar to type A spermatogonia, partially detached from the basal lamina of the seminiferous tubules. These cells underwent gradual degeneration during subsequent weeks so that by ten weeks the tubules showed already advanced atrophy with Sertoli cells and only spermatogonia (Fig. lc-lf) . The Leydig cells, the testicular interstitial cells, showed progressive hyperplasia from the fourth postnatal week (Fig. Id, If) .
  • GDNF mouse and human GDNF and its receptor mRNAs were elevated in the transgenic mice as determined by Northern blotting in wild-type and transgenic testes at various ages (Fig. 2) .
  • GDNF, Ret and GFR ⁇ l transcripts were highly expressed at birth and during the first postnatal week (Fig.2a-c) .
  • GDNF mRNA was strongly downregulated shortly before the initiation of puberty (at two weeks of age) (Fig2a) .
  • the Ret the Ret
  • NTN Neurturin
  • GFR ⁇ 2 GFR ⁇ 2
  • Both NTN and GFR ⁇ 2 transcripts are expressed by postnatal testes (Widenfalk, J. , et al., J. Neurosci. 17, 8506-8519 (1997)), however preferentially after puberty.
  • the third member of GDNF-family, persephin Movator-like substance
  • the transgenic testes showed a continuous and high expression of the human GDNF mRNA by type A spermatogonia clusters from two weeks of age to adulthood (Fig. 3c) .
  • Ret mRNA was expressed by both spermatogonia and spermatids in wild-type testes (Fig. 3d, 3e) and in the clusters of spermatogonia in transgenic testes (Fig. 3f) .
  • GFR ⁇ l mRNA was expressed by spermatogonia, spermatocytes and round spermatids in wild-type testes (Fig. 3g, 3h) and also intensively by the spermatogonia clusters in transgenic testes (Fig. 3i) .
  • the signalling receptor complex for GD ⁇ F is present in the spermatogonia of both wild-type and transgenic mice.
  • GD ⁇ F is obviously acting as a paracrine Sertoli-cell-derived regulator of spermatogenesis, but in transgenic mice this regulation is disrupted by the continuous autocrine expression of G ⁇ DF by germ cells.
  • spermatogonia are the only proliferating germ cells in testis and the mitotic activity is highly segmented in normal semini- ferous tubules (Fig. 4a) .
  • the BrdU- labelled spermatogonia showed an abnormal distribution within the seminiferous tubules, reflecting the spermatogonia clusters in the lumina of seminiferous tubules (Fig. 4b) . Because of the highly segmental nature of cell proliferation in wild-type testis, it is difficult to compare the net proliferation rates of wild-type and transgenic testes.
  • Sertoli cells Fig. 4f, 4g
  • increased apoptosis overriding cell proliferation may explain the gradual testicular atrophy in the GDNF overexpressing mice.
  • the accumulation of spermatogonia in young mice seems to be caused by the block of their differentiation and not enhanced cell proliferation.
  • mice or rats are sterile in the absence of the vitamin A metabolite, retinoic acid (RA) , as shown by feeding them with vitamin A deficient (VAD) diet.
  • VAD vitamin A deficient
  • Differentiation of type A spermatogonia is completely blocked by VAD diet and restored by reintroduction of vitamin A or its active metabolites (Kim, K.H., et al., Molec . Endocrinology 4: 1679-1688, 1990).
  • Retinoic acid receptor a (RAR ⁇ ) mRNA and protein levels decline upon a VAD diet (Akmal, K. , et al . , Endocrinology 139, 1239-1248, 1998).
  • RAR ⁇ -deficient transgenic mice are infertile and develop testicular atrophy (Lufkin, T., et al., Devel . Biol 90, 7225-7229, 1993). Therefore, the expression of RAR proteins was analysed in wild type and GD ⁇ F overexpressing transgenic mice. The RAR expression practically disappeared in the spermatogonial clusters of the transgenic mice, as show by indirect immunoperoxidase staining with antibodies to RAR proteins. The data indicate that one of the mechanisms how GD ⁇ F acts as a negative regulator of spermatogonia is downregulation of RAR expression in Ret expressing spermatogonia .
  • transgenic mice overexpressing neurturin in testis To analyse whether the other members of the GD ⁇ F family would act as negative regulators of spermatogenesis, the investigators constructed transgenic mice overexpressing neurturin in testis. Indeed, the transgenic mice expressing neurturin under the translation elongation factor l ⁇ promoter that targets the transgene expression specifically to testis show also infertility during the first four postpubertal weeks as described by mating tests identical to those with the GD ⁇ F overexpressing mice .
  • sperm differentiation in mammals can be divided into three distinct phases that are regulated by both local cellular interactions and hormonal control (Russell, L. D., et al . , Histological and histopathological evaluation of the testes. Clearwater, F. L., Cache River, pp. 1-40 (1990)).
  • the first phase of spermatogenesis is the mitosis phase, when the spermatogonia give rise to the spermatocytes . Beyond the spermatogonial stage, spermatogenesis proceeds without mitotic cell divisions.
  • Spermatogonia are further classified to subtypes, in which the type A spermatogonia subtypes include the spermatogenic stem cells.
  • the second phase is the meiosis phase of spermatocytes resulting in haploid spermatids .
  • the third phase is the spermiogenic phase, in which the spermatids transform into spermatozoa, the sperm cells, structurally equipped to reach and fertilise the egg.
  • type A spermatogonia proliferate and are triggered to differentiate. However, some of them remain undifferentiated and are considered as the reservoir of stem cells (Russell, L. D., et al . , Histological and histopathological evaluation of the testes. Clearwater, F. L., Cache River, pp. 1-40 (1990)).
  • Sertoli and germ cells may involve a number of molecules, such as stem cell factor (c-kit ligand) , insulin-like growth factor- I, three members of the TGF- ⁇ family (M ⁇ llerian inhibiting substance, activin, and inhibin) , and transferrin (Pescovitz, 0. H. , et al . , Trends Endocrinol . Metab. 5, 126-131 (1994) ) .
  • Sertoli cells secrete all these molecules and germ cells possess their receptors. Conversely, Sertoli cells express receptors for ligands secreted by germ cells.
  • Sertoli cells for instance express p75 nerve growth factor receptor and fibroblast growth factor receptors, whose cognate ligands are secreted by testicular germ cells (Pescovitz, 0. H., et al., Trends Endocrinol. Metab. 5, 126-131 (1994); Parvinen, M., et al., J. Cell Biol. 117, 629-41 (1992); Djakiew, D., et al., Biol. Reprod. 51, 214-21 (1994)). Furthermore, Sertoli cells also interact with Leydig cells. Desert hedgehog (Dhh) gene is expressed by Sertoli cells and its putative receptor, patched, predominantly by Leydig cells (Bitgood, M. J.
  • the human GDNF cDNA was cloned to the p ⁇ F-BOS vector with the EFl ⁇ -promoter (Migushi a, S. and Nagaca, S., Nucleic Acid Res. 18, 5322 (1990)) by replacing Xbal-Xbal stuffer fragment with entire coding sequence of human GDNF cDNA (GenBank accession number L15306) .
  • the correctness of the expression construct was verified by sequencing. Proper expression GDNF protein with the construct was verified by Cos cell transfection followed by i munoprecipitation of GDNF from lysate and culture supernatant.
  • Transgenic mice were produced by micro- injecting the 2.7kb Pvul-Hindlll fragment of the GDNF cDNA construct into the pronuclei of newly fertilised FVB inbred mouse eggs as described (Hogan, B., et al . , Manipulating the Mouse Embryo. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press (1994) ) .
  • the founders were identified by Southern blotting with a human GDNF cDNA and the offspring of the transgene positive mice by PCR from tail DNA.
  • Primers for human GDNF were from exon 1 (5' ⁇ TGT CGT GGC TGT CTG CCT GGT GC-3') and exon 2 (5'-AAG GCG ATG GGT CTG CAA CAT GCC-3 ' ) . Histology, cell proliferation and apoptosis. For histology, freshly dissected testes and epididymides were fixed in Bouins fixative or 4% PFA for 2-24 hours depending on the size, processed in paraffin, cut at 7 ⁇ m and stained by hemato- xylin/eosin.
  • mice were injected intraperitoneally with a BrdU and 5-fluoro2 -deoxy- uridine (FrdU) cocktail (Amersham) at a concentration of 10 mg/kg in PBS. After 2 hr, the mice were sacrificed by neck dislocation, autopsy was done, testes were removed and fixed in Bouin solution. Testes were processed in paraffin, 7 ⁇ m sections were cut and depara finised. BrdU incorporation was detected by indirect immunofluorescence with monoclonal antibodies to anti-bromodeoxyuridine (Amersham) and rhoda- mine-conjugated goat anti-mouse IgG (Jackson) . Apoptotic cells were detected in deparaffinised sections with ApopTag in situ cell death detection kit (Oncor) according to manuf cturer's instructions .
  • RhdU 5-fluoro2 -deoxy- uridine
  • RNA of different tissues were isolated with TRIZOL Reagent kit (Life Technology) according to the manufacturer's instructions and 30g of total RNA was used in each lane.
  • Northern blotting was carried out according to Maniatis protocol (Sambrook, J. , et al . , T. Molecular Cloning: A Laboratory Manual. Second Edition. Cold Spring Harbor Laboratory Press 1989 pp. 7.43-7.50).
  • the cDNA probes (the same as in situ hybridisation, see below) were labelled with [ 32 P]dCTP.
  • the filters were exposed in a FujiBAS phospho- i ager . Im unoprecipitation and Western blotting with anti-GDNF antibody.
  • Freshly dissected testes were homogenized and lysed in high salt lysis buffer (300 mg protein/ml; high salt lysis buffer: 1M NaCl, 100 mM Tris HCl, pH 8 , 2 % BSA, 4 mM EDTA, 0.2 % Triton X-100, 2 mM PMSF, 1 mM sodium orthovanadate and 1 tabl. /10 ml of protease inhibitor cocktail tablets (Complete, Mini EDTA, Boehringer Mannheim) ) .
  • the lysates were immuno- precipitated with a polyclonal antibody to human recombinant GDNF peptide that crossreacts with mouse GDNF (R & D Systems) and Protein A-Sepharose.
  • the immunoprecipitates were run on a 20 % SDS-PAGE and transferred to Hybond ECL nitrocellulose membranes (Amersham Life Science) .
  • the membranes were blocked with 5 % bovine serum albumin and immunoprecipitated GDNF was detected with a polyclonal anti-GDNF antibody (Santa Cruz Biotechnology) at room temperature for 2 h.
  • Detection was accomplished by using anti-rabbit secondary antibody conjugated to horseradish peroxidase (Sigma) and ECL chemilumine- scence (Amersham) according to the manufacturer's instructions
  • In situ hybridization In situ hybridisation was performed as described (Wilkinson, D. & Green, P., In situ hybridization and the three-dimensional reconstruction of serial sections. In: Postimplantation Mammalian Embryos. A practical approach (ed. A. Copp & D. Cockroft) , pp. 155-171. Oxford University Press: London (1990)) .
  • Antisense and sense cRNA probes (mouse GDNF 328 bp from exon 3, human GDNF 513 bp from exon 1 and exon 2, mouse Ret from 3 end 714 bp, mouse GFR ⁇ l 777 bp) were synthesized using appropriate RNA polymerases and 35g_]_ a b )e- L-L e ci UTP.
  • Hybridisation temperature was 52°C and autoradiography slides were exposed at +4°C for 2-4 weeks. The slides were photographed with CCR camera hooked with computer. In PhotoShop graphics programme, the dark field images were inverted, artificially stained red and combined with the bright field images .
  • Example 1 Example 1
  • GDNF human GDNF
  • the 2.7 kb HindiII -EcoRI fragment isolated from the pEF-BOS vector contains EFl ⁇ (translation elongation factor l ⁇ ) promoter and the 0.7 kb Xba-EcoRI fragment having a ply (A) adenylation signal.
  • the size of the entire coding region of human GDNF cDNA is 636 bp .
  • Transgenic mice are produced by microinjecting the 2.7 kb PVuI-Hindlll fragment into the pronucleus of fertilized egg cells. The injection is at first made into a FVB/NIH-mice strain and later the transgene positive mice are mated with a FVB/NIH- and NMRI-mice strains.
  • Transgenic founder mice are identified by Southern hybridization with a hGDNF-probe and the transgenic offspring is identified by polymerase chain reaction (PCR) from tail DNA (the transgene transferring mouse strain is later called GDNF t-mice) .
  • Primers used in PCR are from exon 1 (5'-TGT CGT GGC TGT CTG CCT GGT GC-3') and exon 2 (5'-AAG GCG ATG GGT CTG CAA CAT GCC-3'). The results indicated that GDNF t-mice have a coding region of human GDNF cDNA and they transfer it to their offspring.
  • RNA from the testis of a three week old mouse with which the expression levels of endogenic GDNF mRNA and the human transgenic GD ⁇ F R ⁇ A of a mouse is compared.
  • RT-PCR reverse transcription polymerase chain reaction
  • 1 ⁇ g entire R ⁇ A is copied by reverse transcrition in a random-primed reaction (20 ⁇ l) using AMV reversed copy enzyme (Finnzymes, Helsinki) according to the instructions of the manufacturer. 2 ⁇ l of the reaction product is used as template when copying the endogenic mouse GD ⁇ F cD ⁇ A or the transgenic human GD ⁇ F cD ⁇ A.
  • the first primer (5 ' GCTGAG/CCAGTGACTCA/CAATATGCC3 ' ) recognizes both the mouse and human GD ⁇ F alleles and covers the intron region in order to avoid genomic contamination.
  • the primers of the other end were selective to endogenic and transgenic GD ⁇ F (GD ⁇ F specific region in mouse: 5 ' TGTTAGCCTTCTACTCCGAGACAG3 ' ) and the transgenic primer (transgenic GD ⁇ F specific region 5 ' CACCAGCCTTCTATTTCTGGATAA3 ' ) .
  • primers are used to copy in identical conditions (l ⁇ M primer, lmM, d ⁇ TP and Dynazyme polymerase (Finnzymes, Helsinki) in their own buffer) fragments of 373 bp length from endogenic and transgenic GD ⁇ F cD ⁇ A, which has been reversed copied either from RNA from the testis of a normal or transgenic mouse.
  • the cycle is 30s 95°, 30s 62° ja 45s 72°, and it is renewed 20-36 times after a "hot starting" 95°C for 2 minutes.
  • the copying is ended after the last cycle for 5 minutes in 72°C.
  • the PCR product is run in an agarose gel containing ethidium bromide, is studied under UV-light and is photographed with a Polaroid-camera.
  • Northern blot hybridization to identify transgenic hGDNF and endogenic mouse GDNF mRNA expression is carried out with species specific [ ⁇ - 32 P] -labelled GDNF cDNA probes from total-RNA isolated from testis with a TRIzol Reagent-kit (Life Technologies) according to the manufacturer's instructions.
  • the probes are: mouse GDNF (328 bp exon 3 (NCB1 Gene Bank, accession number U37459) and human GNDF (536 bp from exon 1 ja 2, (Lin et al .
  • Northern blot hybridization is carried out according to Sambrook, J. , et al., (Molecular Cloning: A Laboratory Manual. Second Edi- tion. Cold Spring Harbor Laboratory Press 1989 pp. 7.43-7.50). In each sample the total RNA used is 30 ⁇ g.
  • the hybridization filter is kept on a Fuji phosphoimager plate and exposed to light overnight and the image is recorded with a FujiBAS phosphoimager.
  • a actin-probe is used as a probe to control even loading of R ⁇ A, which is hybridized on each filter after the hybridization of the G ⁇ DF-probe.
  • GDNF-transgene Due to the testis targeting elongation factor l ⁇ promoter the GDNF-transgene is not expressed in the male elsewhere than in testis (Furuchi, T., et al . , Development 122, 1703-1709 (1996) ) , which is studied with Northern blot hybridization from different female and male tissues. Also, cRNA in si tu hybridization is carried out from testis, ovary and kidney according to Wilkinson, D. & Green, P., (Tn situ hybridisation and the three-dimensional reconstruction of serial sections. In: Postimplantation Mammalian Embryos. A practical approach (ed. A. Copp & D. Cockroft) , pp. 155-171. Oxford University Press: London (1990)).
  • the probes are: mouse GDNF-probe (328 bp) , human GDNF-probe (536 bp) , GFR ⁇ l-probe (777 bp) and GFR ⁇ 2-probe (approximately 500 bp) .
  • mouse GDNF-probe 328 bp
  • human GDNF-probe 536 bp
  • GFR ⁇ l-probe 777 bp
  • GFR ⁇ 2-probe approximately 500 bp
  • mice GDNF-receptor Ret is expressed in prepubertal mice spermatogonia and in adult mice also in spermatides, but not any more in differentiated spermatozoa and the co-receptors of GFR ⁇ l and GFR ⁇ 2 are expressed during spermatogenesis also in germ line cells ( Figure 3) .
  • transgenic GDNF The morphological results of the expression of transgenic GDNF is followed by histological studies of the testis. Normal and transgenic mouse tissues are fixed with 4 % paraformaldehyde overnight, processed in paraffin, cut in 4-7 ⁇ m sections, deparaffinized and stained with hematoxylineosin- stain.
  • GDNF can prevent the formation of sperm cells
  • the fertilization ability of male GDNF t-mouse strain was studied by allowing them to mate with fertile, more than 8 week old females. 20 males (more than six weeks old) were kept with females for two weeks. Calculated together they mated with 200 females. Additionally, three males (from the age of eight weeks) were kept half a year with females so that the females were changed with three to four days intervals . They were mating as often as normal mice. After mating the uterus and egg leaders of some females were studied in order to find sperm cells.
  • Figure 1 Testicular morphology in wild-type (left panel: a,c,e) and transgenic (right panel: b,d,f) mice at three different ages, and wild-type (g) and transgenic (h) epididymal ducts at 8-weeks. (a and b) 3 -weeks of age. (c and d) 8 weeks, (e and f) 6 months. Note in d the remnants of type A spermatogonia clusters that are abundant in the young mice (d) , and the advanced germ cell atrophy and Leydig cell hyperplasia (star) in f. Epididymal ducts from a wild-type (g) and transgenic (h) mouse show the lack of sperm in the transgenic mice. Scale bar 100 ⁇ m.
  • FIG. 1 Northern blotting for GDNF (a,d), Ret (b,e) and GFR ⁇ l (c,f) mRNAs in wild-type (left panel) and transgenic testes (right panel) at various ages.
  • Mouse GD ⁇ F cD ⁇ A and human GD ⁇ F cD ⁇ A probes were used in a and d, respectively.
  • GD ⁇ F (a) , Ret (b) , and GFR ⁇ l (c) R ⁇ A levels are strongly downregulated after the first or second postnatal week.
  • the levels of human GD ⁇ F mR ⁇ A transgene (d) remain high to adulthood.
  • Figure 3 The distribution of GD ⁇ F (a,b,c), Ret (d,e,f) and GFR ⁇ l (g,h,i) transcripts in testes of wild-type (WT, left and middle panel) and transgenic mice (TG, right panel) by cR ⁇ A in situ hybridisation.
  • (a,b) mouse GD ⁇ F cD ⁇ A template and, in (c) , human GD ⁇ F cD ⁇ A template were used. All other probes were to mouse transcripts. Sense controls did not show grain density above the background.
  • (a,d,g) 1- to 2-weeks-old wild-type testes.
  • GFR ⁇ l mRNA distribution follows that of Ret, but is also highly expressed by spermatocytes. In transgenic mice, both Ret (f) and GFR ⁇ l (i) are expressed in the clusters of spermatogonia. Scale bar 100 ⁇ m.
  • Figure 4. Cell proliferation (a,b,c), apoptosis (d,e), and live morphology (f,g) in wild-type (a,d) and transgenic (b,e,g) testes.
  • (f) depicts both a wild-type and transgenic seminiferous tubule
  • Testis weights (average + SD) in wild-type and transgenic mice at various ages
  • n the number of testes in each group

Abstract

The present invention is related to glial cell line-derived neurotrophic factor (GDNF) family-related compounds, such as glial cell line-derived neurotrophic factor (GDNF), GDNF-like factors, such as persephin, artemin and neurturin or other compounds which act like GDNF on its signal transmitting receptor, i.e. cRet receptor tyrosine kinase and/or co-receptors, e.g. GDNF-family-receptor α:s (GFRα1-4) and their use for regulating and studying spermatogenesis, for inhibting the differentiation of sperm cells, for developing male contraceptives or as a male contraceptive as well as their use for manufacturing male contraceptives.

Description

THE USE OF GLIAL CELL LINE-DERIVED NEUROTROPHIC FACTOR FAMILY- RELATED COMPOUNDS FOR REGULATING SPERMATOGENESIS AND FOR PREPARING MALE CONTRACEPTIVES
The Field of the Invention
The present invention is related to the use of glial cell line-derived neurotrophic factor (GDNF) family-related compounds, such as glial cell line-derived neurotrophic factor (GDNF) , GDNF-like factors and/or other compounds which act like GDNF on its receptor and/or co-receptors, for regulating and studying spermatogenesis, for inhibiting the differentiation of sperm cells, for developing male contraceptives or as a male contraceptive as well as their use for manufacturing male contraceptive compositions.
The Background of the Invention
Spermatogenesis is a complex sequence of events involving both hormonal regulators and local interactions between Sertoli and germ cells (Bellve, A. R. & Zhang, W. J., Reprod. Fertil. 85, 771-793 (1989); Skinner, M. K. , Endocr. Rev. 12, 45-72 (1991); Pescovitz, 0. H., et al . , Trends Endocrinol . Metab. 5, 126-131 (1994). Parvinen, M., et al . , J. Cell Biol . 117, 629-41 (1992), Djakiew, D., et al . , Biol. Reprod. 51, 214-21 (1994), Cancilla, B. & Risbridger, G. P., Biol. Reprod. 58, 1138-45 (1998); Bitgood, M. J., et al . , Curr . Biol. 6, 298-304 (1996); Zhao, G. Q., et al . , Genes Dev. 10, 1657-1669 (1996); Zhao, G. Q., et al., Development 125, 1103-1112 (1998)). The differentiation of sperm cells or spermatogenesis can be divided in three phases. In the first phase of the spermatogenesis undifferentiated stem cells from the germ line, the spermato- gonia, are divided mitotically and differentiate into sper- matocytes . Spermatogonia are very resistant against different types of damages and after damage spermatogenesis starts from said cells. At present, it is not known by which mechanism the spermatogonia are maintained and secondly by which signals they are differentiated into spermatocytes . Spermatogonia are located in the peripheral parts of the seminiferous tubules and are in contact with the Sertoli cells, which represent the regulating cells of the seminiferous tubules and are classified as somatic cells of the organism.
In the next phase the spermatogonia are at first differentiated into primary and thereafter into secondary spermatocytes, whereby their chromosomes become haploid by two meiotic cell divisions after which they are called spermatides. In the third phase the spermatides mature into spermazoa or mature sperm cells.
The luteinizing hormone, the follicle stimulating hormone and testosterone are the most important hormonal regulators of spermatogenesis. The cells of the germ line in the seminiferous tubules of testis are not direct targets of the hormonal regulation but the hormonal regulation is transmitted by the Sertoli cells. They modulate the hormonal signals into paracrine signals which are believed to be signal molecules and growth factors, such as stem cell factor (c-kit ligand) , insulin like growth factor- 1 and three members of the transforming growth factor-β family (transforming growth factor-β, activin and inhibin) (Bellve A. R. & Zhang . J., Reprod. Fertil. 85, 771-793 (1989); Skinner, . K. , Endocr. Rev. 12, 45-72 (1991); Pescovitz, 0. H., et al . , Trends Endocrinol . Metab. 5, 126-131 (1994)). All said ligands are expressed by Sertoli cells and their receptors are located in the cells of the germ line. Functional proof of said regulating system is scare. The probably best experimental evidence of the paracrine regulation of spermatogenesis is obtained with desert hedgehog-deficient transgenic mice (Bitgood, M. J., et al . , Curr. Biol. 6, 298-304 (1996)). Desert hedgehog is a signal molecule, which is expressed in the Sertoli cells. The patched-receptor thereof is located in the Leydig cells, i.e. the testosterone producing interstitial cells of the testis. When the desert hedgehog molecule is missing the differentia- tion of sperm stops, sperm cells are not formed and the male mice are all sterile.
The spermatogenetic cells can also send the signals back to the Sertoli cells (Bellve, A. R. & Zhang W. J., Reprod. Fertil. 85, 771-793 (1989); Skinner, M. K. , Endocr. Rev. 12, 45-72 (1991); Pescovitz, 0. H., et al . , Trends Endocrinol . Metab. 5, 126-131 (1994)). This is indicated for example by the fact that nerve growth factor and fibroblast growth factor- 1 are expressed in the cells of the germ line and the p75 nerve growth factor receptor and fibroblast growth factor receptor are located in the Sertoli cells. Hints of auto- crine regulation of spermatogenesis also exist. Bone morphoge- netic protein 8a and 8b and their receptors are expressed in the same spermatogenetic cells (Zhao, G. Q., et al . , Gene Dev . 10, 1657-1669 (1996); Zhao, G. Q., et al . , Dev. 125, 1103-1112 (1998)) . The functional evidence for the autocrine regulation is still lacking.
The glial cell line-derived neurotrophic factor (GDNF) is a member of the transforming growth factor-β superfamily (Lin, L. F., et al., Science 260, 1130-1132 (1993)). GDNF maintains the dopaminergic, noradrenergic, cholinergic and motor neurons in the nervous system and also protects peripheral parasympa- thetic ciliary and sensory nerve cells (Lin, L.F., Neural Notes. 2, 3-7, (1996). During the development of mammals, GDNF is expressed also in other tissues, such as embryonic kidney and testis (Suvanto, P., et al . , European Journal of Neuro- science 8, 816-822, (1996); Trupp M, et al . , Journal of Cell Biology 130, 137-148, (1996)). The role of GDNF as a regulator of the differentiation of the kidney has been shown with gene knockout or gene deletion techniques in mice (Pichel, J.G., et al., Nature 382, 73-75 (1996). Because said mice die as newborn, it has not been possible to study with said deletion gene model - or other experimental models - the function of GDNF in spermatogenesis, which is initiated much later, only after birth. Studies with different model systems indicate that there is a multitude of ways for interfering with spermatogenesis and for developing male contraceptives . So far the methods suggested have been very rough with severe side effects for the male subject. Thus, the problem yet to be solved is how to select from the multitude of hormonal growth regulators one which can sufficiently specifically be targeted to the testis and at the same time provides a system for regulating spermatogenesis without disturbing side-effects and which system does not interfere negatively with the male mating activity.
The problem was solved by the present inventors when studying transgenic mice with human GDNF incorporated into their testis. It was shown that mice with an elevated level of human GDNF were capable of mating but not capable of fertilizing the female. Thus, the solution to the problem is to use glial cell line-derived neurotrophic factor family-related compounds as defined in the claims of the present invention for regulating spermatogenesis and inhibiting or preventing spermatogenesis followed by infertility and as active ingredients for manufacturing compositions useful as male contraceptives, which are both effective and without severe side-effects .
Thus, the first objective of the present invention is to provide an effective male contraceptive without severe, undesired side-effects by using GDNF-family-related compounds as male contraceptives.
Another objective of the present invention is to use the GDNF-family-related compounds as active ingredients for manufacturing compositions useful as male contraceptives .
One other objective of the present invention is to provide tools for regulating and studying spermatogenesis. This is facilitated by inhibiting the differentiation of sperm cells. A third objective of the present invention is to provide tools, which have an effect on infertility caused by disturbed differentiation of sperm cells.
The objective of the present invention is also to provide tools for regulating and studying spermatogenesis as well as for inhibiting the differentiation of sperm cells.
Further the objective of the present invention is to provide transgenic animals, especially mice, which due to the testis targeted GDNF-family-related compounds provide a useful tool for studying spermatogenesis.
The Summary of the Invention
The characterstics of the present invention are as defined in the claims. The present invention describes a method, whereby it is possible to study sterility caused by disturbed differentiation of sperm cells and secondly it provides a system for inhibiting the differentiation of sperm cells in a contraceptive sense, whereby the male (man) is sterile (infertile) . The GDNF-transgene carrying transgenic mouse strain, the GDNF t-mouse strain, is a model for infertility caused by disturbances in the differentiation of sperm cells. It is characterized in that GDNF, compounds acting like, said GDNF, another cRet receptor, another GDNF-receptor activating compound or a compound, which activates the cRet receptor signal transmitting tubules in spermatogonia, are suitable as a male contraceptive and it is also characterized in that the GDNF t-mouse model is suitable as an animal model for studying the regulation of spermatogenesis and spermatogenesis.
A Short Description of the Drawings
Figure la. Testicular morphology in wild-type mice at 3 -weeks of age. Scale bar 100 μm. Figure lb. Testicular morphology in transgenic mice at 3 -weeks of age. Scale bar 100 μm. Note the undifferentiated spermatogonia formed cell cluster.
Figure lc. Testicular morphology in wild-type mice at 8 -weeks of age. Scale bar 100 μm.
Figure Id. Testicular morphology in transgenic mice at 8 -weeks of age. Note in d the remnants of type A spermatogonia clusters that are abundant in the young mice. Scale bar 100 μm.
Figure le. Testicular morphology in wild-type mice at 6-months of age. Scale bar 100 μm.
Figure If. Testicular morphology in transgenic mice at 6- months of age. Note the advanced germ cell atrophy and Leydig cell hyperplasia (star) . Scale bar 100 μm.
Figure lg. Testicular morphology in wild-type epididymal ducts at 8-weeks. Scale bar 100 μm.
Figure lh. Testicular morphology in transgenic epididymal ducts at 8-weeks. Note the lack of sperm. Scale bar 100m.
Figure 2a. Northern blotting for GDNF RNAs in wild-type mice at various ages . GDNF levels are strongly downregulated after the first or second postnatal week.
Figure 2b. Northern blotting for Ret mRNAs in wild-type mice testes at various ages. GDNF mRNA levels are strongly downregulated after the first or second postnatal week.
Figure 2c. Northern blotting for GFRcel mRNAs in wild-type mice at various ages. GFRαl mRΝA levels are strongly downregulated after the first or second postnatal week. Figure 2d. Northern blotting for GDNF mRNAs in transgenic testes at various ages. The hGDNF levels remain high after the first or second postnatal week.
Figure 2e. Northern blotting for Ret mRNAs in transgenic mouse testis at various ages.
Figure 2f. Northern blotting for GFRαl mRNAs in transgenic testes at various ages. GFRαl RNA levels are high in the transgenic mice at all stages analysed.
Figure 2g depicts immunoprecipitation of GDNF from wild-type (WT) and transgenic (TG) testes at 3 and 6 weeks of age. In wild-type testes, the GDNF protein is not detectable. The control, 25 ng of reco binant human GDNF protein is shown on the right side.
Figure 3a. The distribution of GDNF transcripts in testes of wild-type (WT) mice by cRNA in si tu hybridisation. Sense controls did not show grain density above the background. 1- week-old wild-type testis. Scale bar 100 μm.
Figure 3b. The distribution of GDNF transcripts in 8 -weeks-old testis of wild-type (WT) mouse by cRNA in si tu hybridisation. Sense controls did not show grain density above the background. Scale bar 100 μm.
Figure 3c. The distribution of GDNF transcripts in testes of transgenic mice (TG) by cRNA in si tu hybridisation. Sense controls did not show grain density above the background. 8-weeks-old transgenic testis. Note that the spermatogonia in the transgenic mice continuously express human GDNF at high levels, but the number of these cells is clearly diminishing at 8 weeks of age. Scale bar 100 μm.
Figure 3d. The distribution of Ret transcripts in testes of wild-type (WT) mice by cRNA in situ hybridisation. 2-weeks-old wild-type testis. At two weeks of age, Ret expression is localised to some spermatogonia in the basal layer of the seminiferous tubules (arrows) .
Figure 3e. The distribution of Ret transcripts in testes of wild-type (WT) mice by cRNA in si tu hybridisation. 8 -weeks-old wild-type testis. At 8 weeks of age, Ret is expressed in both some spermatogonia and spermatides.
Figure 3f. The distribution of Ret transcripts in testes of transgenic mice (TG) by cRNA in si tu hybridisation. 8 -weeks- old transgenic testis. Note that the spermatogonia in the transgenic mice continuously express Ret at high levels, but the number of these cells is clearly diminishing at 8 weeks of age. In transgenic mice, Ret is expressed in the clusters of spermatogonia. Scale bar 100 μm.
Figure 3g. The distribution of GFRαl transcripts in testes of wild-type mice (WT) . 1- to 2-weeks-old wild-type testis. GFRαl is expressed by some spermatogonia, morphologically similar to those with Ret (arrows) .
Figure 3h. The distribution of GFRαl transcripts in testes of wild-type by cRNA in si tu hybridisation. 8 -weeks-old wild- type testis. GFRαl mRNA distribution follows that of Ret, but is also highly expressed by spermatocytes. Scale bar 100 μm.
Figure 3i. The distribution of GFRαl transcripts in testes of transgenic mice (TG) by cRNA in situ hybridisation. 8-weeks- old transgenic testis. Note that the spermatogonia in the transgenic mice continuously express human GDNF at high levels, but the number of these cells is clearly diminishing at 8 weeks of age. In transgenic mice, GFRαl is expressed in the clusters of spermatogonia. Scale bar 100 μm.
Figure 4a. Cell proliferation in wild-type mice testes. In 3 -weeks-old testis, BrdU labelling of S-phase cells is seen only in the periphery of wild-type seminiferous tubules. Note the absence of labelling in some transsections of tubules, reflecting the segmented distribution of S-phase cells during spermatogenesis (stars) . Scale bar 100 μm.
Figure 4b. Cell proliferation in transgenic mice testes. In a 3 -week-old transgenic mouse testis, many BrdU-labelled cells are seen in the luminal area of the seminiferous tubules, reflecting the abnormal distribution of spermatogonia (arrow) . The segmental distribution of S-phase cells is not seen in the transgenic tubules. Scale bar 100 μm.
Figure 4c. Distribution of cell line proliferation indexes in wild type and transgenic testes. The number of BrdU labeled cells / 100 spermatogonia was counted in 100 transections of wild type and transgenic seminiferous tubules.
Figure 4d. Apoptosis in wild-type mice testes. TUNEL- labelling for apoptosis in 4-week-old mice testes. Only a few apoptotic cells are seen. Scale bar (c) 100 μm.
Figure 4e. Apoptosis in transgenic mouse testis. TUNEL-label- ling for apoptosis in 4-week-old testes. Note the increase of labelling in spermatogonia clusters in (Figure 4e) , as compared to wild-type (Figure 4d) seminiferous tubules. Scale bar 100 μm.
Figure 4f. Live morphology in both a wild-type and transgenic seminiferous tubule. Live preparation of seminiferous tubules from 15-week-old wild-type (WT) and transgenic (TG) mice. Note the reduced diameter of the transgenic tubule and the increase of Leydig cells (arrows) around it. Scale bar 200 μm.
Figure 4g. Live morphology in transgenic testes. High magnification of living cells from a transgenic seminiferous tubule in an unfixed squash preparation showing large numbers of dead cells (arrow) , some of which resemble type A spermatogonia, in a Sertoli cell (arrow head) . Scale bar 10 μm.
The Detailed Description of the Invention
Definitions
In the present invention the terms used have the meaning they generally have in the fields of biochemistry, pharmacology, recombinant DNA technology, including transgenic animal production, but some terms are used with a somewhat deviating or broader meaning than in the normal context. Accordingly, in order to avoid uncertainty caused by terms with unclear meaning some of the terms used in this specification and in the claims are defined in more detail below.
The term "glial cell line-derived neurotrophic factor (GDNF) family-related compounds" i.e. GDNF-family-related compounds comprises glial cell line-derived neurotrophic factor (GDNF) , neurturin, persephin, artemin and other similar growth factors, but also other compounds which have the same effect as GDNF-like compounds on the GDNF-receptor (s) or co-receptors. Said receptor (s) or co-receptor (s) comprises Ret tyrosine kinase and GDNF-family-receptor a : s , respectively.
The term "glial cell line-derived neurotrophic factor (GDNF) " means the glial cell line-derived neurotrophic factor (GDNF) , which is a member of the transforming growth factor-β family (Lin, L. F., et al . , Science 260, 1130-1132 (1993). GDNF maintains the dopaminergic , noradrenergic, cholinergic and motor neurons in the nervous system and also protects peripheral, parasympathetic, ciliary and sensory nerve cells (Lin, L.F., Neural Notes. 2, 3-7 (1996)). During the development of mammals, GDNF is expressed also in other tissues, such as embryonic kidney and testis (Suvanto P, et al . , European Journal of Neuroscience 8, 816-822, (1996); Trupp M, et al . , Journal of Cell Biology 130, 137-148, (1996)). The properties and structure, amino acid sequence is disclosed by Lin, L. F., et al. (Science 260, 1130-1132 (1993). Said protein is coded by a gene characterized by the GDNF cDNA sequence deposited as Genebank accession number L15306.
The term "GDNF-like factors" or GDNF-like compounds mean growth factors which have a similar structure or act like GDNF on its receptor or co-receptors. The term "GDNF-like factor" or "GDNF-like compound" above all relates to three GDNF-like growth factors, persephin, artemin and neurturin, which are described in (Milbrandt, J., et al . , Neuron 20, 1-20 (1998); Kotzbauer P. T., et al . , Nature 384, 467-470 (1996)).
All four growth factors of the GDNF-family are structurally similar and cRet receptor tyrosine kinase (Durbec, P., et al., Nature 381, 789-793 (1996); Buj-Bello, A., et al . , Nature. 387, 721-724, (1997)) acts as the common signal transmitting receptor of GDNF, artemin, persephin and neurturin.
The "compounds which act like said GDNF" means compounds which act as GDNF on the receptor transmitting signals of GDNF or co-receptors thereof.
The term "receptor" means GDNF-family binding substances, i.e. substances or compounds belonging to the GDNF-family-related compounds which are capable of transmitting the signals of GDNF-family-related compounds. The term "receptor" above all means the GDNF-family signal mediatory receptor is cRet receptor tyrosine kinase.
The term "co-receptors" means receptors, which do not transmit the signals of GDNF-family related compounds, but activate the receptor transmitting the signals of the GDNF-family-related compounds. Such compounds are above all the GDNF-family receptor a : s (GFRcn) , which is a new receptor class, the members of which are called GDNF-family-receptor c.s (GFRc . These also belong to the GDNF, persephin, artemin and neur- turin signalling receptor complex. At present four members (GFRαl-4) (Jing, S., et al . , Cell 85, 9-10 (1996), Jing, S. Q., et al., J. Biol. Chem. 272, 33111-3311 7 (1997); Treanor, J. J., et al., Nature. 382, 80-83, (1996); Suvanto, P., et al., Human Molecular Genetics. 6, 1267-1273, (1997)) of said family are known. They may transmit signals independently, but they are essential for ligand binding and cRet activation.
The term "concentration of GDNF-family-related compounds in such amounts that the differentiation of sperm cells is prevented, spermatogenesis can be regulated and infertility caused by disturbed differentiation of sperm cells or that the male is rendered unable to fertilize the female" means a concentration of GDNF-family-related compounds, which is approximately 50 - 5000, preferably 50 - 500, most preferably 50 - 100 times higher than the concentration of endogenous GDNF in the testis of said male. Said amount can be calculated from the test results obtained when measuring the GDNF mRNA levels of transgenic mice shown in the Northern blots (Figures 2) and which indicate an elevated amount of mRNA from which the expressed protein levels can be calculated. From these results it can be concluded that the amount of GDΝF in transgenic mice is 50-500 times higher than the endogenic level in three to six week old mice, but only a little bit higher than in new born mice, when the GDΝF levels are at their highest and no spermatogenesis is presented. From these results it is possible for those skilled in the art to calculate the amounts needed for obtaining the desired effects in man.
The term "derivatives" means iso ers of GDΝF-family-related compounds . It also includes hybrid molecules constructed based on the structures of the compounds GDΝF, persephin, neurturin and artemin, as well as truncated forms, complexes and fragments of said compounds. The only prerequisite for said compounds is that they still have substantially the same properties and effects as the compounds listed above, determined by the same methods as described in the present inven- tion. Naturally, the term "derivatives" includes derivatives of the compounds listed above in a more conventional sense, e.g. the free groups in the listed compounds, such as car- boxy- , amino- and/or hydroxy- groups may be substituted by alkylation, esterification, etherification and amidation with e.g. alkyl groups, including methyl, ethyl, propyl, etc. The only prerequisite in this case, too is that the substitutions do not substantially alter the properties or effectivity of said molecules or compounds listed above in this paragraph.
General Description of the Invention
Glial cell line-derived neurotrophic factor (GDNF) is a potent survival factor for various sets of neuronal cells, it regulates kidney differentiation (Lin, L. F., et al . , Science 260, 1130-1132 (1993); Tomac, A., et al . , Nature 373, 335-339 (1995); Winkler, C, et al . , J. Neurosci . 16, 7206-7215 (1996), Oppenheim, R. W. , et al . , Nature 373, 344-346 (1995), Nguyen, Q. T., et al . , Science 279, 1725-1729 (1998); Trupp, M., et al., J. Cell Biol. 130, 137-148 (1995); Hellmich, H. L., et al., Mech. Dev. 54, 95-105 (1996); Schuchardt , A., et al., Nature 367, 380-383 (1994); Pichel, J. G., et al . , Nature 382, 73-75 (1996) and is expressed in testis (Trupp, M. , et al., J. Cell Biol. 130, 137-148 (1995); Hellmich, H. L., et al., Mech. Dev. 54, 95-105 (1996); Suvanto, P., et al . , European Journal of Neuroscience 8, 816-822 (1996)) where its function, however, remains unknown.
GDNF, a distant member of the transforming growth factor-β (TGF-β) superfamily (Lin, L. F., et al . , Science 260, 1130- 1132 (1993)) is a potent survival factor for dopaminergic neurones of the substantia nigra, and some other sets of neurones in the central and peripheral nervous systems (Lin, L. F., et al., Science 260, 1130-1132 (1993); Tomac, A., et al., Nature 373, 335-339 (1995); Winkler, C, et al . , J. Neurosci. 16, 7206-7215 (1996), Oppenheim, R. W., et al . , Nature 373, 344-346 (1995), Nguyen, Q. T., et al . , Science 279, 1725-1729 (1998); Trupp, M., et al . , J. Cell Biol. 130, 137-148 (1995); Hellmich, H. L. , et al . , Mech. Dev. 54, 95-105 (1996); Schuchardt , A., et al . , Nature 367, 380-383 (1994); Pichel, J. G., et al . , Nature 382, 73-75 (1996)). GNDF also acts as a signalling molecule in ureteric branching during kidney morphogenesis (Schuchardt, A., et al . , Nature 367, 380-383 (1994); Pichel, J. G., et al . , Nature 382, 73-75 (1996)) . It is synthesised by several neuronal and non-neu- ronal embryonic organs including the embryonic and postnatal testis (Trupp, M. , et al . , J. Cell Biol. 130, 137-148 (1995); Hellmich, H. L., et al . , Mech. Dev. 54, 95-105 (1996); Suvanto, P., et al . , European Journal of Neuroscience 8, 816 - 822 (1996) ) The signalling receptor-complex for GDNF includes Ret receptor tyrosine kinase (Durbec, P., et al . , Nature 381, 789-793 (1996); Trupp, M., et al . , Nature 381, 785-789 (1996)) and a member of the glycosylphosphatidylinositol- linked co-receptors, GDNF-family-receptor a : s (GFRo.1-4) (Jing, S., et al., Cell 85, 9-10 (1996); Jing, S. Q., et al . , J. Biol. Chem. 272, 33111-3311 7 (1997); Enokido, Y., et al . , Curr. Biol. 18, 1019-1022 (1998)). The GDNF-, GFRcl and Ret-null mice exhibit remarkably similar phenotypes with absent or hypodysplastic kidneys and severe defects in enteric innervation (Schuchardt, A., et al . , Nature 367, 380-383 (1994); Pichel, J. G., et al., Nature 382, 73-75 (1996)). As they die during the first postnatal day, spermatogenesis could not be analysed in these mutant mice.
In order to approach the role of GDNF in spermatogenesis, the present inventors studied transgenic mice with targeted overexpression of GDNF in testicular germ cells. The transgenic males were infertile and showed an early arrest in the differentiation of spermatogenic cells, the type A spermatogonia. They formed clusters within seminiferous tubules, finally resulting in testicular atrophy. The data obtained showed a novel function for GDNF as a paracrine inhibitory signal during early spermatogenesis. In wild-type mice, GDNF is believed to regulate the pool of spermatogonia that is gradually committed to sperm differentiation. These infertile transgenic mice serve as a model for male infertility with spermatogenic deficiency. The activation of the GDNF signalling cascade possibly provides us with means to develop contraceptives for man. The effects of GDNF on spermatogenesis should also be taken into consideration, when GDNF-like molecules are used in clinical trials of neurodegenerative disorders, such as Parkinson's disease.
When studying transgenic mice overexpressing human GDNF under human translation elongation factor-1 (EF-1) promoter (Mizu- shima, S. & Nagata, S., Nucleic Acids Res. 18, 5322 (1990), which targets the transgene expression specifically to testicular germ cells (Furuchi, T., et al . , Development 122, 1703-1709 (1996)), a novel function for GDNF was found. The disturbance of spermatogenesis in transgenic testis, targeted to overexpress GDNF, suggested that GDNF acts as a local inhibitory signal in spermatogonia differentiation. The gradual degeneration of germ cells and the hyperplasia of Leydig cells in the transgenic mice are apparently secondary to the spermatogenic deficiency, because Sertoli cells eliminate poorly differentiated and abnormal germ cells, and interplay between the cell types is disrupted. Therefore, GDNF targeting to testis provides a new animal model for male infertility. The activation of the GDNF signalling cascade by GDNF, Ret agonists or downstream substrates possibly provides us with means whereby sperm production in man could be prevented and male contraceptives developed. The effects of GDNF on spermatogenesis should also be taken into consideration, when GDNF-like molecules are designed for and used in clinical trials of neurodegenerative disorders, such as Parkinson's disease .
To study the function of GDNF in testis, the inventors generated transgenic mice overexpressing human GDNF under human translation elongation factor-1 (EF-1) promoter (Mizushima, S. & Nagata, S., Nucleic Acids Res. 18, 5322 (1990)), which targets the transgene expression specifically to testicular germ cells (Furuchi, T., et al . , Development 122, 1703-1709 (1996) ) . Other promoters targetting the transgene expression specifically to testicular germ cells can be used as well. Four independent transgenic founders were analysed, two males CIO, C12 and two females S6, E19 with transgene copy numbers 10, 3, 20, and 4, respectively. The male founders CIO and C12 were infertile and further analysis of the male phenotype was done with the offspring of the two female founders S6 and E19. The transgenic mice developed normally to adulthood and showed a similar phenotype. The body weight was normal, and they did not have any defects in the weights and morphology of paren- chymal organs (data not shown), except in the testis. After four weeks of age, a reduction in weight of testes was observed in the transgenic mice as compared' to wild-type controls (Table 1) . All transgenic males in the offspring of the female founders S6 and E19 were infertile. During six months of continuous breeding, three transgenic males were caged with wild-type FVB and NMRI females in three to four day intervals. The wild-type females mated with transgenic males at normal frequency, as shown by the appearance of vaginal plugs, but none became pregnant. Further 20 transgenic males at breeding age were subjected to wild-type FVB and NMRI females in short term breeding test. During two weeks of continuing breeding, approximately 200 females from both strains were plugged, but none became pregnant. Strain dependence of the infertile phenotype was excluded by crossbreeding the transgenic FVB mice to NMRI background. Like the transgenic males of the FVB strain, those of the FI generation were also infertile.
It was shown that the testicular histology of one-week-old transgenic mice was normal . From two to three weeks of age (Fig. la, lb) , they showed clusters of cells morphologically similar to type A spermatogonia, partially detached from the basal lamina of the seminiferous tubules. These cells underwent gradual degeneration during subsequent weeks so that by ten weeks the tubules showed already advanced atrophy with Sertoli cells and only spermatogonia (Fig. lc-lf) . The Leydig cells, the testicular interstitial cells, showed progressive hyperplasia from the fourth postnatal week (Fig. Id, If) . Occasional germ cells outside the spermatogonial clusters developed further into spermatocytes and rarely to spermatids, but they arrested postmeiotically and degenerated. Mature sperm was not observed in seminiferous tubules or epididymal ducts, although no obstruction was found in the ductuli efferens, which conduct sperm from the testis to the epidi- dy is (Fig. lg, lh) .
It was also shown that the levels of mouse and human GDNF and its receptor mRNAs were elevated in the transgenic mice as determined by Northern blotting in wild-type and transgenic testes at various ages (Fig. 2) . In wild-type testis, GDNF, Ret and GFRαl transcripts were highly expressed at birth and during the first postnatal week (Fig.2a-c) . GDNF mRNA was strongly downregulated shortly before the initiation of puberty (at two weeks of age) (Fig2a) . Simultaneously, the Ret
(Fig.2b) and GFRαl (Fig.2c) mRNA levels declined but were still detectable in adult testes. The forth number artemin
(ART) binds to GFRα3 and also signals via Ret.
Neurturin (NTN) is a GDNF-related molecule that utilises the same multicomponent receptor complex as GDNF but prefers GFRα2 (Klein, R. D., et al . , Nature 387, 717-721 (1997)) as a co-receptor, a homologue to GFRαl. Both NTN and GFRα2 transcripts are expressed by postnatal testes (Widenfalk, J. , et al., J. Neurosci. 17, 8506-8519 (1997)), however preferentially after puberty. The third member of GDNF-family, persephin (Milbrandt, J. et al . , Neuron 20, 1-20 (1998)), binds to chicken GFRα4 (Enokido, Y., et al . , Curr . Biol. 18, 1019-1022 (1998)), whose homologue has, to date, not been found in mammals .
Northern blot with a human GDNF mRNA specific probe showed that the transgene was expressed only in testis, and not in other organs of both sexes, such as ovary, penis, brain, liver, kidney, lung, and skin (data not shown) . In contrast to the declining of the levels of GDNF mRNA in wild-type mice, the transgenic GDNF mRNA expression remained high in testis from neonate to adulthood (Fig. 2d) . Also, Ret and GFRαl mRNA remained high at all ages analysed (Fig. 2e,2f) . Immunoprecipi- tation of GDNF from 3 and 6 -week-old wild-type and transgenic testes verified that the GDNF protein levels were strongly increased in transgenic testes as compared to wild type testes (Fig. 2g) -
Previously, embryonic Sertoli cells and a Sertoli cell lineage have been shown to express GDNF (Hellmich, H. L., et al . , Mech. Dev. 54, 95-105 (1996); Trupp, M. , et al . , J. Cell Biology 130, 138 - 148, (1995)). By in si tu hybridisation, GDNF mRNA was detectable in wild-type seminiferous tubules during the first postnatal week but not thereafter (Fig. 3a, 3b) and the label for GDNF mRNA was distributed over Sertoli cells. The transgenic testes showed a continuous and high expression of the human GDNF mRNA by type A spermatogonia clusters from two weeks of age to adulthood (Fig. 3c) . Ret mRNA was expressed by both spermatogonia and spermatids in wild-type testes (Fig. 3d, 3e) and in the clusters of spermatogonia in transgenic testes (Fig. 3f) . GFRαl mRNA was expressed by spermatogonia, spermatocytes and round spermatids in wild-type testes (Fig. 3g, 3h) and also intensively by the spermatogonia clusters in transgenic testes (Fig. 3i) . Thus, the signalling receptor complex for GDΝF is present in the spermatogonia of both wild-type and transgenic mice. In wild- ype mice, GDΝF is obviously acting as a paracrine Sertoli-cell-derived regulator of spermatogenesis, but in transgenic mice this regulation is disrupted by the continuous autocrine expression of GΝDF by germ cells.
Spermatogonia are the only proliferating germ cells in testis and the mitotic activity is highly segmented in normal semini- ferous tubules (Fig. 4a) . In transgenic mice, the BrdU- labelled spermatogonia showed an abnormal distribution within the seminiferous tubules, reflecting the spermatogonia clusters in the lumina of seminiferous tubules (Fig. 4b) . Because of the highly segmental nature of cell proliferation in wild-type testis, it is difficult to compare the net proliferation rates of wild-type and transgenic testes. However, the peak cell proliferation index of spermatogonia (BrdU- labelled nuclei/100 spermatogonia) was clearly lower in transgenic than wild-type mice: 0.26 +/-0.11 (n=714) and 0.72 +/-0.14 (n=436 cells, counted from the proliferative segments of seminiferous tubules) , respectively (Fig. 4c) .
Apoptosis was prominent in the clusters of spermatogonia and other poorly differentiated germ cells from the second postnatal week, as shown by TUNEL- labelling (Fig. 4d, 4e) . Large numbers of dead cells, some of which resembled type A spermatogonia, were engulfed and phagocytically removed by Sertoli cells (Fig. 4f, 4g) . Thus, increased apoptosis overriding cell proliferation may explain the gradual testicular atrophy in the GDNF overexpressing mice. On the other hand, the accumulation of spermatogonia in young mice seems to be caused by the block of their differentiation and not enhanced cell proliferation.
Three GDNF overexpression males were followed for 5 months and in that time neither macroscopic testicular tumours nor microinvasive cancer cells were observed. Furthermore, we have not seen testicular malignancies in any of the younger male mice autopsies (n=103), showing that the differentiation defect of spermatogonia in GDNF overexpressing mice does not ultimately lead to germ line malignancies but results in testicular atrophy. However, more mice should be followed in long term to exclude any increased risk of testicular cancer.
Male mice or rats are sterile in the absence of the vitamin A metabolite, retinoic acid (RA) , as shown by feeding them with vitamin A deficient (VAD) diet. Differentiation of type A spermatogonia is completely blocked by VAD diet and restored by reintroduction of vitamin A or its active metabolites (Kim, K.H., et al., Molec . Endocrinology 4: 1679-1688, 1990). Retinoic acid receptor a (RARα) mRNA and protein levels decline upon a VAD diet (Akmal, K. , et al . , Endocrinology 139, 1239-1248, 1998). Accordingly, RARα-deficient transgenic mice are infertile and develop testicular atrophy (Lufkin, T., et al., Devel . Biol 90, 7225-7229, 1993). Therefore, the expression of RAR proteins was analysed in wild type and GDΝF overexpressing transgenic mice. The RAR expression practically disappeared in the spermatogonial clusters of the transgenic mice, as show by indirect immunoperoxidase staining with antibodies to RAR proteins. The data indicate that one of the mechanisms how GDΝF acts as a negative regulator of spermatogonia is downregulation of RAR expression in Ret expressing spermatogonia .
To analyse whether the other members of the GDΝF family would act as negative regulators of spermatogenesis, the investigators constructed transgenic mice overexpressing neurturin in testis. Indeed, the transgenic mice expressing neurturin under the translation elongation factor lα promoter that targets the transgene expression specifically to testis show also infertility during the first four postpubertal weeks as described by mating tests identical to those with the GDΝF overexpressing mice .
Sperm differentiation in mammals can be divided into three distinct phases that are regulated by both local cellular interactions and hormonal control (Russell, L. D., et al . , Histological and histopathological evaluation of the testes. Clearwater, F. L., Cache River, pp. 1-40 (1990)). The first phase of spermatogenesis is the mitosis phase, when the spermatogonia give rise to the spermatocytes . Beyond the spermatogonial stage, spermatogenesis proceeds without mitotic cell divisions. Spermatogonia are further classified to subtypes, in which the type A spermatogonia subtypes include the spermatogenic stem cells. The second phase is the meiosis phase of spermatocytes resulting in haploid spermatids . The third phase is the spermiogenic phase, in which the spermatids transform into spermatozoa, the sperm cells, structurally equipped to reach and fertilise the egg. During the mitosis phase, type A spermatogonia proliferate and are triggered to differentiate. However, some of them remain undifferentiated and are considered as the reservoir of stem cells (Russell, L. D., et al . , Histological and histopathological evaluation of the testes. Clearwater, F. L., Cache River, pp. 1-40 (1990)). The precise mechanisms by which the stem cells are maintained or transformed into differentiating spermatogonia have remained unresolved. Our Northern blotting showed that the down- regulation of GDNF coincides with the initiation of the sperm differentiation in normal mice and GDNF overexpression arrests spermatogonial differentiation. Therefore, local GDNF signalling may be essential for spermatogonia to maintain their undifferentiated stage.
There is increasing evidence that each step of spermatogenesis requires paracrine interactions between somatic Sertoli cells and germ cells, and these interactions may be mediated by growth factors and their receptors (Bellve A. R. & Zhang W. J., Reprod. Fertil. 85, 771-793 (1989); Skinner, M. K. , Endocr. Rev. 12, 45-72 (1991); Pescovitz, 0. H. , et al . , Trends Endocrinol . Metab. 5, 126-131 (1994). Parvinen, M., et al., J. Cell Biol. 117, 629-41 (1992), Djakiew, D., et al., Biol. Reprod. 51, 214-21 (1994), Cancilla, B. & Risbridger, G. P., Biol. Reprod. 58, 1138-45 (1998); Bitgood, M. J., et al., Curr. Biol. 6, 298-304 (1996); Zhao, G. Q., et al . , Genes Dev. 10, 1657-1669 (1996); Zhao, G. Q., et al . , Development 125, 1103-1112 (1998)). However, direct functional data regarding the intercellular communication between testicular somatic cells and germ cells are still scarce. The interplay between Sertoli and germ cells may involve a number of molecules, such as stem cell factor (c-kit ligand) , insulin-like growth factor- I, three members of the TGF-β family (Mύllerian inhibiting substance, activin, and inhibin) , and transferrin (Pescovitz, 0. H. , et al . , Trends Endocrinol . Metab. 5, 126-131 (1994) ) . Sertoli cells secrete all these molecules and germ cells possess their receptors. Conversely, Sertoli cells express receptors for ligands secreted by germ cells. Sertoli cells for instance express p75 nerve growth factor receptor and fibroblast growth factor receptors, whose cognate ligands are secreted by testicular germ cells (Pescovitz, 0. H., et al., Trends Endocrinol. Metab. 5, 126-131 (1994); Parvinen, M., et al., J. Cell Biol. 117, 629-41 (1992); Djakiew, D., et al., Biol. Reprod. 51, 214-21 (1994)). Furthermore, Sertoli cells also interact with Leydig cells. Desert hedgehog (Dhh) gene is expressed by Sertoli cells and its putative receptor, patched, predominantly by Leydig cells (Bitgood, M. J. , et al., Curr. Biol. 6, 298-304 (1996). Indeed, Dhh null mice show a severe differentiation defect of spermatogenesis (Bitgood, M. J., et al., Curr. Biol. 6, 298-304 (1996)) . Recently, bone morphogenetic protein 8a and 8b together with their putative receptors were found in the germ cells and inactivation of these ligands resulted in abnormal spermatogenesis and male infertility (Zhao, G. Q., et al . , Genes Dev. 10, 1657-1669 (1996); Zhao, G. Q., et al . , Development 125, 1103-1112 (1998)). The co-localisation of those growth factors and their receptors in germ cells suggests an autocrine regulation of germ cells .
Our data demonstrate a novel function for GDNF. The disturbance of spermatogenesis in transgenic testis, targeted to overexpress GDNF, suggests that GDNF acts as a local inhibitory signal in spermatogonia differentiation. The gradual testicular athrophy and the hyperplasia of Leydig cells in the transgenic mice are apparently secondary to the spermatogenic deficiency. Therefore, GDNF targeting to testis provides a new animal model for male infertility. The activation of the GDNF signalling cascade by GDNF, Ret agonists or downstream substrates possibly provides us with means whereby sperm pro- duction in man could be prevented and male contraceptives developed. The effects of GDNF on spermatogenesis should also be taken into consideration, when GDNF-like molecules are designed for and used in clinical trials of neurodegenerative disorders, such as Parkinson's disease.
The invention is described in more detail in the following experimental part, which discloses the material and methods used the preparation of transgenetic mice and the experiments used to demonstrate the effect of GDNF in the animal model. Said experimental part should not be construed to limit the scope of the invention. Those skilled in the art can foresee a multitude of different application based on the results obtained in the following experimental part .
Methods
GDNF transgene construction and production of transgenic mice.
The human GDNF cDNA was cloned to the pΞF-BOS vector with the EFlα-promoter (Migushi a, S. and Nagaca, S., Nucleic Acid Res. 18, 5322 (1990)) by replacing Xbal-Xbal stuffer fragment with entire coding sequence of human GDNF cDNA (GenBank accession number L15306) . The correctness of the expression construct was verified by sequencing. Proper expression GDNF protein with the construct was verified by Cos cell transfection followed by i munoprecipitation of GDNF from lysate and culture supernatant. Transgenic mice were produced by micro- injecting the 2.7kb Pvul-Hindlll fragment of the GDNF cDNA construct into the pronuclei of newly fertilised FVB inbred mouse eggs as described (Hogan, B., et al . , Manipulating the Mouse Embryo. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press (1994) ) . The founders were identified by Southern blotting with a human GDNF cDNA and the offspring of the transgene positive mice by PCR from tail DNA. Primers for human GDNF were from exon 1 (5'~TGT CGT GGC TGT CTG CCT GGT GC-3') and exon 2 (5'-AAG GCG ATG GGT CTG CAA CAT GCC-3 ' ) . Histology, cell proliferation and apoptosis. For histology, freshly dissected testes and epididymides were fixed in Bouins fixative or 4% PFA for 2-24 hours depending on the size, processed in paraffin, cut at 7 μm and stained by hemato- xylin/eosin. For the cell proliferation assay, the mice were injected intraperitoneally with a BrdU and 5-fluoro2 -deoxy- uridine (FrdU) cocktail (Amersham) at a concentration of 10 mg/kg in PBS. After 2 hr, the mice were sacrificed by neck dislocation, autopsy was done, testes were removed and fixed in Bouin solution. Testes were processed in paraffin, 7 μm sections were cut and depara finised. BrdU incorporation was detected by indirect immunofluorescence with monoclonal antibodies to anti-bromodeoxyuridine (Amersham) and rhoda- mine-conjugated goat anti-mouse IgG (Jackson) . Apoptotic cells were detected in deparaffinised sections with ApopTag in situ cell death detection kit (Oncor) according to manuf cturer's instructions .
Living cells. Seminiferous tubules were dissected and squashed on the preparation slides with PBS buffer. Computer-aided analysis of cells within the seminiferous tubules was carried out according to Parvinen M. , et al . (Histochem. & Biol. 108, 77-81 (1997) ) .
Northern blotting. Total RNA of different tissues were isolated with TRIZOL Reagent kit (Life Technology) according to the manufacturer's instructions and 30g of total RNA was used in each lane. Northern blotting was carried out according to Maniatis protocol (Sambrook, J. , et al . , T. Molecular Cloning: A Laboratory Manual. Second Edition. Cold Spring Harbor Laboratory Press 1989 pp. 7.43-7.50). The cDNA probes (the same as in situ hybridisation, see below) were labelled with [32P]dCTP. The filters were exposed in a FujiBAS phospho- i ager . Im unoprecipitation and Western blotting with anti-GDNF antibody. Freshly dissected testes were homogenized and lysed in high salt lysis buffer (300 mg protein/ml; high salt lysis buffer: 1M NaCl, 100 mM Tris HCl, pH 8 , 2 % BSA, 4 mM EDTA, 0.2 % Triton X-100, 2 mM PMSF, 1 mM sodium orthovanadate and 1 tabl. /10 ml of protease inhibitor cocktail tablets (Complete, Mini EDTA, Boehringer Mannheim) ) . The lysates were immuno- precipitated with a polyclonal antibody to human recombinant GDNF peptide that crossreacts with mouse GDNF (R & D Systems) and Protein A-Sepharose. The immunoprecipitates were run on a 20 % SDS-PAGE and transferred to Hybond ECL nitrocellulose membranes (Amersham Life Science) . The membranes were blocked with 5 % bovine serum albumin and immunoprecipitated GDNF was detected with a polyclonal anti-GDNF antibody (Santa Cruz Biotechnology) at room temperature for 2 h. Detection was accomplished by using anti-rabbit secondary antibody conjugated to horseradish peroxidase (Sigma) and ECL chemilumine- scence (Amersham) according to the manufacturer's instructions
In situ hybridization. In situ hybridisation was performed as described (Wilkinson, D. & Green, P., In situ hybridization and the three-dimensional reconstruction of serial sections. In: Postimplantation Mammalian Embryos. A practical approach (ed. A. Copp & D. Cockroft) , pp. 155-171. Oxford University Press: London (1990)) . Antisense and sense cRNA probes (mouse GDNF 328 bp from exon 3, human GDNF 513 bp from exon 1 and exon 2, mouse Ret from 3 end 714 bp, mouse GFRαl 777 bp) were synthesized using appropriate RNA polymerases and 35g_]_ab)e-L-Leci UTP. Hybridisation temperature was 52°C and autoradiography slides were exposed at +4°C for 2-4 weeks. The slides were photographed with CCR camera hooked with computer. In PhotoShop graphics programme, the dark field images were inverted, artificially stained red and combined with the bright field images . Example 1
Preparation of transgenic mice
Targeting GDNF expression under the translation elongation factor 1 alpha-promoter to testis, wherewith the function of GDNF in testis is studied. The entire coding region of human GDNF (hGDNF) cDNA is cloned to the pEF-BOS vector by replacing the Zbal-Xbal fragment with hGDNF cDNA (Mizushima, S. & Nagata, S., Nucleic Acids Res. 18, 5322 (1990)). The 2.7 kb HindiII -EcoRI fragment isolated from the pEF-BOS vector contains EFlα (translation elongation factor lα) promoter and the 0.7 kb Xba-EcoRI fragment having a ply (A) adenylation signal. The size of the entire coding region of human GDNF cDNA is 636 bp . Transgenic mice are produced by microinjecting the 2.7 kb PVuI-Hindlll fragment into the pronucleus of fertilized egg cells. The injection is at first made into a FVB/NIH-mice strain and later the transgene positive mice are mated with a FVB/NIH- and NMRI-mice strains. Transgenic founder mice are identified by Southern hybridization with a hGDNF-probe and the transgenic offspring is identified by polymerase chain reaction (PCR) from tail DNA (the transgene transferring mouse strain is later called GDNF t-mice) . Primers used in PCR are from exon 1 (5'-TGT CGT GGC TGT CTG CCT GGT GC-3') and exon 2 (5'-AAG GCG ATG GGT CTG CAA CAT GCC-3'). The results indicated that GDNF t-mice have a coding region of human GDNF cDNA and they transfer it to their offspring.
Example 2
Expression of the transgene is studied by Northern blot hybridization and RT-PCR
Expression of the transgene is studied both by Northern blot hybridization in mice of different ages (0-13 weeks old) , with which endogenic and transgenic GDNF mRNA levels at different ages are detected, and by reverse transcription polymerase chain reaction (RT-PCR) from RNA from the testis of a three week old mouse, with which the expression levels of endogenic GDNF mRNA and the human transgenic GDΝF RΝA of a mouse is compared. For the reverse transcription PCR 1 μg entire RΝA is copied by reverse transcrition in a random-primed reaction (20μl) using AMV reversed copy enzyme (Finnzymes, Helsinki) according to the instructions of the manufacturer. 2μl of the reaction product is used as template when copying the endogenic mouse GDΝF cDΝA or the transgenic human GDΝF cDΝA.
The first primer (5 ' GCTGAG/CCAGTGACTCA/CAATATGCC3 ' ) recognizes both the mouse and human GDΝF alleles and covers the intron region in order to avoid genomic contamination. The primers of the other end were selective to endogenic and transgenic GDΝF (GDΝF specific region in mouse: 5 ' TGTTAGCCTTCTACTCCGAGACAG3 ' ) and the transgenic primer (transgenic GDΝF specific region 5 ' CACCAGCCTTCTATTTCTGGATAA3 ' ) . These primers are used to copy in identical conditions (lμM primer, lmM, dΝTP and Dynazyme polymerase (Finnzymes, Helsinki) in their own buffer) fragments of 373 bp length from endogenic and transgenic GDΝF cDΝA, which has been reversed copied either from RNA from the testis of a normal or transgenic mouse. The cycle is 30s 95°, 30s 62° ja 45s 72°, and it is renewed 20-36 times after a "hot starting" 95°C for 2 minutes. The copying is ended after the last cycle for 5 minutes in 72°C. The PCR product is run in an agarose gel containing ethidium bromide, is studied under UV-light and is photographed with a Polaroid-camera. Northern blot hybridization to identify transgenic hGDNF and endogenic mouse GDNF mRNA expression is carried out with species specific [α-32P] -labelled GDNF cDNA probes from total-RNA isolated from testis with a TRIzol Reagent-kit (Life Technologies) according to the manufacturer's instructions. The probes are: mouse GDNF (328 bp exon 3 (NCB1 Gene Bank, accession number U37459) and human GNDF (536 bp from exon 1 ja 2, (Lin et al . , Science 260, 1130 - 1132, (1993)). Northern blot hybridization is carried out according to Sambrook, J. , et al., (Molecular Cloning: A Laboratory Manual. Second Edi- tion. Cold Spring Harbor Laboratory Press 1989 pp. 7.43-7.50). In each sample the total RNA used is 30 μg. The hybridization filter is kept on a Fuji phosphoimager plate and exposed to light overnight and the image is recorded with a FujiBAS phosphoimager. A actin-probe is used as a probe to control even loading of RΝA, which is hybridized on each filter after the hybridization of the GΝDF-probe. The results showed a strong expression of GDΝF mRNA in the testis of a mouse at birth (Figure 2a) , which expression is weakened (declines) during the first postnatal weeks, especially when the mouse reaches puberty, and is barely detectable in an adult mouse. Based on Northern blotting it can be estimated that in the testis of a three-week-old GDNF t-mouse (Fig. 2d) there is transgenic human GDNF mRNA 50-500 times more than endogenic GDNF mRNA (Figure 2a) .
Example 3
Location of GDNF to testis by Northern blot hybridization and in situ hybridization
Due to the testis targeting elongation factor lα promoter the GDNF-transgene is not expressed in the male elsewhere than in testis (Furuchi, T., et al . , Development 122, 1703-1709 (1996) ) , which is studied with Northern blot hybridization from different female and male tissues. Also, cRNA in si tu hybridization is carried out from testis, ovary and kidney according to Wilkinson, D. & Green, P., (Tn situ hybridisation and the three-dimensional reconstruction of serial sections. In: Postimplantation Mammalian Embryos. A practical approach (ed. A. Copp & D. Cockroft) , pp. 155-171. Oxford University Press: London (1990)). The probes are: mouse GDNF-probe (328 bp) , human GDNF-probe (536 bp) , GFRαl-probe (777 bp) and GFRα2-probe (approximately 500 bp) . For the in si tu hybridization temperature with all probe is 52° C and exposure times are at +4°C 1.5-4 weeks. From each probe at least three different exposures are developed. The results obtained with both Northern blot hybridization and cRNA in si tu hybridization showed that the human GDNF-probe hybridizes only to RNA from testis, in which the in si tu hybridization signal is located into the spermatogonia and some early spermatocytes (Figure 3) .
It was also shown that in both transgenic GDNF t-mice and normal, mice GDNF-receptor Ret is expressed in prepubertal mice spermatogonia and in adult mice also in spermatides, but not any more in differentiated spermatozoa and the co-receptors of GFRαl and GFRα2 are expressed during spermatogenesis also in germ line cells (Figure 3) .
Example 4
Morphology of expression shown by histology
The morphological results of the expression of transgenic GDNF is followed by histological studies of the testis. Normal and transgenic mouse tissues are fixed with 4 % paraformaldehyde overnight, processed in paraffin, cut in 4-7 μm sections, deparaffinized and stained with hematoxylineosin- stain.
In the histological studies of 1-4 week old testis a strong elevation of the amount of spermatogonia can be seen in males of GDNF t-mouse strains, which elevation is related to the fact that they do not continue their differentiation to spermatids and spermatozoa. Based on their histological structure, the cell clusters formed within the seminiferous tubules can be identified as spermatogonia of A-type (Figure IB) . After four weeks the number of said spermatogonia cell clusters in the seminiferous tubules are decreased and after 11 weeks only few spermatogonia are left. A mature and free spermatozoa is not formed in any stage and sperm cells cannot be detected in epididymal ducts. Example 5
Demonstration of ability of GDNF to cause infertility
Because GDNF can prevent the formation of sperm cells, the fertilization ability of male GDNF t-mouse strain was studied by allowing them to mate with fertile, more than 8 week old females. 20 males (more than six weeks old) were kept with females for two weeks. Calculated together they mated with 200 females. Additionally, three males (from the age of eight weeks) were kept half a year with females so that the females were changed with three to four days intervals . They were mating as often as normal mice. After mating the uterus and egg leaders of some females were studied in order to find sperm cells.
The results obtained indicated that not a single female, which had mated with a transgene carrying male from GDNF t-mouse strain became pregnant, and from their uterus or egg leaders no sperm cells were found. The transgene carrying males of the GDNF t-mouse strain are totally infertile and are not capable of breeding litter.
The results are also discussed in the Figures as described in detail below:
Figure 1. Testicular morphology in wild-type (left panel: a,c,e) and transgenic (right panel: b,d,f) mice at three different ages, and wild-type (g) and transgenic (h) epididymal ducts at 8-weeks. (a and b) 3 -weeks of age. (c and d) 8 weeks, (e and f) 6 months. Note in d the remnants of type A spermatogonia clusters that are abundant in the young mice (d) , and the advanced germ cell atrophy and Leydig cell hyperplasia (star) in f. Epididymal ducts from a wild-type (g) and transgenic (h) mouse show the lack of sperm in the transgenic mice. Scale bar 100 μm.
Figure 2. Northern blotting for GDNF (a,d), Ret (b,e) and GFRαl (c,f) mRNAs in wild-type (left panel) and transgenic testes (right panel) at various ages. Mouse GDΝF cDΝA and human GDΝF cDΝA probes were used in a and d, respectively. GDΝF (a) , Ret (b) , and GFRαl (c) RΝA levels are strongly downregulated after the first or second postnatal week. In contrast to the declining endogenous GDΝF mRΝA levels in wild-type mice, the levels of human GDΝF mRΝA transgene (d) remain high to adulthood. Also, Ret (e) and GFRαl (f) mRΝA levels are high in the transgenic mice at all stages analysed, (g) Immunoprecipitation of GDΝF from wild-type (WT) and transgenic (TG) testes at 3 and 6,5 weeks of age. In wild-type testes, the GDΝF protein is not detectable. On the right, 25 ng of recombinant human GDΝF protein as a control.
Figure 3. The distribution of GDΝF (a,b,c), Ret (d,e,f) and GFRαl (g,h,i) transcripts in testes of wild-type (WT, left and middle panel) and transgenic mice (TG, right panel) by cRΝA in situ hybridisation. In (a,b) mouse GDΝF cDΝA template and, in (c) , human GDΝF cDΝA template were used. All other probes were to mouse transcripts. Sense controls did not show grain density above the background. (a,d,g) 1- to 2-weeks-old wild-type testes. (b,e,h) 8-weeks-old wild-type testes. (c,f,i) 8-weeks-old transgenic testes. Note that the spermatogonia in the transgenic mice continuously express human GDNF at high levels, but the number of these cells is clearly diminishing at 8 weeks of age. (d) At two weeks of age, Ret expression is localised to some spermatogonia in the basal layer of the seminiferous tubules (arrows) . (g) Also, GFRαl is expressed by some spermatogonia, morphologically similar to those with Ret (arrows) . (e) At 8 weeks of age, Ret is expressed in both some spermatogonia and spermatids. (h) GFRαl mRNA distribution follows that of Ret, but is also highly expressed by spermatocytes. In transgenic mice, both Ret (f) and GFRαl (i) are expressed in the clusters of spermatogonia. Scale bar 100 μm. Figure 4. Cell proliferation (a,b,c), apoptosis (d,e), and live morphology (f,g) in wild-type (a,d) and transgenic (b,e,g) testes. (f) depicts both a wild-type and transgenic seminiferous tubule, (a) In 3 -weeks-old testis, BrdU labelling of S-phase cells is seen only in the periphery of wild-type seminiferous tubules. Note the absence of labelling in some transsections of tubules, reflecting the segmented distribution of S-phase cells during spermatogenesis. (b) In a 3 -week- old transgenic testis, many BrdU-labelled cells are seen in the luminal area of the seminiferous tubules, reflecting the abnormal distribution of spermatogonia (arrow) . The segmental distribution of S-phase cells is not seen in the transgenic tubules. Figure 4c. Distribution of cell proliferation indexes in wild type and transgenic testes. The number of BrdU labeled cells / 100 spermatogonia was counted in 100 transections of wild type and transgenic siminiferous tubules. The normal segmental distribution of BrdU labeled cells is pertured in transgenic testes. (d and e) TUNEL- labelling for apoptosis in 4-week-old testes. Note the increase of labelling in spermatogonia clusters (arrow) (e) , as compared to wild-type (d) seminiferous tubules. (f) Live preparation of seminiferous tubules from 15 -week-old wild-type (WT) and transgenic (TG) mice. Note the reduced diameter of the transgenic tubule and the increase of Leydig cells (arrows) around it. (g) High magnification of living cells from a transgenic seminiferous tubule in an unfixed squash preparation showing large numbers of dead cells (arrow) , some of which resemble type A spermatogonia, in a Sertoli cell (arrow head) . Scale bar (a-d) 100 μm, (e) 200 μm, (f) 10 μm. Table 1
Testis weights (average + SD) in wild-type and transgenic mice at various ages
n=the number of testes in each group
Age Wild- type Transgenic
Weeks g n mg n
1 5.5 ± 2. 6 20 4.8 ± 2. 5 20
2 27.8 + 6. ,0 20 22.7 ±12 20
3 50.2 + 6. .5 20 58.7 ± 8. .6 20
4 68.8 ± 8. .0 10 40.3 +10. .0 10
5 79.9 + 7. .3 10 30.5 ± 9, .3 10
8 102.2 + 4 .6 10 34.7 ± 8. .0 8
13 100.9 + 6 .0 10 38.3 ± 8 .3 6
24 90.0 ± 4 .6 10 43.2 ± 8 .9 6

Claims

1. The use of glial cell line-derived neurotrophic factor (GDNF) family-related compounds, c h a r a c t e r i z e d in that the compounds related to the GDNF-family are glial cell line-derived neurotrophic factor (GDNF) , GDNF-like factors or compounds as well as derivatives thereof, which compounds or derivatives act like said GDNF on the receptor transmitting signals of GDNF, of GDNF-like factors or compounds or co- receptors thereof are used for regulating and studying spermatogenesis, for inhibiting the differentiation of sperm cells, for developing male contraceptives or as a male contraceptive.
2. The use of the GDNF-family-related compounds according to claim 1, c h a r a c t e r i z e d in that the GDNF-family- related compounds or derivatives thereof are GDNF, persephin, neurturin and/or artemin, GDNF- family like compounds or compounds acting like the GDNF-family-related com ounds.
3. The use of the GDNF-family-related compounds according to claim 1, c h a r a c t e r i z e d in that the GDNF- family- related compound is GDNF and derivatives thereof.
4. The use of the GDNF-family-related compounds according to claim 1, c h a r a c t e r i z e d in that the GDNF-family- related compound is persephin and derivatives thereof.
5. The use of the GDNF-family-related compounds according to claim 1, c h a r a c t e r i z e d in that the GDNF-family- related compound is neurturin and derivatives thereof.
6. The use of the GDNF-family-related compounds according to claim 1, c h a r a c t e r i z e d in that the GDNF- family- related compound is artemin and derivatives thereof.
7. The use of the GDNF-family-related compounds according to claims 1, c h a r a c t e r i z e d in that the receptor transmitting the signals of GDNF-family-related compounds is cRet receptor tyrosine kinase.
8. The use of the GDNF-family-related compounds according to claim 1, c h a r a c t e r i z e d in that the co-receptors activating the receptor transmitting the signals or transmitting the signal of the GDNF-family-related compounds are the GDNF-family-receptor ╬▒:s (GFR╬▒) .
9. The use of the GDNF-family-related compounds according to claim 1 for manufacturing compositions for male use, c h a r a c t e r i z e d in that the composition comprises GDNF, GDNF-related compounds or compounds acting like GDNF on its receptor or co-receptors in such amounts that the differentiation of sperm cells is prevented, spermatogenesis can be regulated and infertility caused by disturbed differentiation of sperm cells can be effected.
10. The use of the GDNF-family-related compounds according to claim 1, for manufacturing composition active as male contraceptives, c h a r a c t e r i z e d in that the composition comprises GDNF, GDNF-related compounds or compounds acting like GDNF on its receptor or co-receptors in such amounts the male is rendered unable to fertilize the female.
11. The use of the GDNF-family-related compounds for manufacturing composition active as male contraceptives according to claim 10, c h a r a c t e r i z e d in that the composition comprises such an amount of GDNF, GDNF-related compounds or compounds acting like GDNF on its receptor and/or co-receptors that the concentration of said compounds in the male is 50 - 5000, preferably 50 - 500, most preferably 50 - 100 times higher than the concentration of endogenous GDNF in the testis of the male .
PCT/FI1999/000687 1998-08-21 1999-08-20 The use of glial cell line-derived neurotrophic factor family-related compounds for regulating spermatogenesis and for preparing male contraceptives WO2000010594A1 (en)

Priority Applications (12)

Application Number Priority Date Filing Date Title
EEP200100103A EE200100103A (en) 1998-08-21 1999-08-20 Use of compounds related to the glial cell line neurotrophic factor family for the regulation of spermatogenesis and for the preparation of contraceptives for use in males
NZ509592A NZ509592A (en) 1998-08-21 1999-08-20 The use of GLIAL cell line-derived neurotrophic factor familty-related compounds for regulating spermatogenesis and for preparing male contraceptives
PL99346298A PL346298A1 (en) 1998-08-21 1999-08-20 The use of glial cell line-derived neurotrophic factor family-related compounds for regulating spermatogenesis and for preparing male contraceptives
CA002338644A CA2338644A1 (en) 1998-08-21 1999-08-20 The use of glial cell line-derived neurotrophic factor family-related compounds for regulating spermatogenesis and for preparing male contraceptives
IL14103799A IL141037A0 (en) 1998-08-21 1999-08-20 The use of glial cell line-derived neurotrophic factor family related compounds for regulating spermatogenesis and for preparing male contraceptives
EP99939468A EP1105151A1 (en) 1998-08-21 1999-08-20 The use of glial cell line-derived neurotrophic factor family-related compounds for regulating spermatogenesis and for preparing male contraceptives
JP2000565914A JP2002523374A (en) 1998-08-21 1999-08-20 Use of glial cell line-induced neurotrophic factor family related compounds to modulate spermatogenesis and produce male contraceptives
HU0103419A HUP0103419A3 (en) 1998-08-21 1999-08-20 The use of glial cell line-derived neurotrophic factor family-related compounds for regulating spermatogenesis and for preparing male contraceptives
AU53752/99A AU763678B2 (en) 1998-08-21 1999-08-20 The use of glial cell line-derived neurotrophic factor family-related compounds for regulating spermatogenesis and for preparing male contraceptives
SK247-2001A SK2472001A3 (en) 1998-08-21 1999-08-20 The use of glial cell line-derived neurotrophic factor family-related compounds for regulating spermatogenesis and for preparing male contraceptives
KR1020017002234A KR20010088793A (en) 1998-08-21 1999-08-20 The use of glial cell line-derived neurotrophic factor family-related compounds for regulating spermatogenesis and for preparing male contraceptives
NO20010859A NO20010859L (en) 1998-08-21 2001-02-20 The use of glial cell line-derived neurotrophic factor family-related compounds to regulate spermatogenesis and to produce contraceptives for men

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
FI981793A FI981793A0 (en) 1998-08-21 1998-08-21 Inhibition of sperm differentiation by GDNF
FI981793 1998-08-21
FI990415A FI990415A0 (en) 1999-02-26 1999-02-26 Use of Compounds Related to Glial Cell Line Neurotrophic Factor Family for the Control of Spermatogenesis and Preparation of Male Contraceptives
FI990415 1999-09-30

Publications (1)

Publication Number Publication Date
WO2000010594A1 true WO2000010594A1 (en) 2000-03-02

Family

ID=26160633

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/FI1999/000687 WO2000010594A1 (en) 1998-08-21 1999-08-20 The use of glial cell line-derived neurotrophic factor family-related compounds for regulating spermatogenesis and for preparing male contraceptives

Country Status (14)

Country Link
EP (1) EP1105151A1 (en)
JP (1) JP2002523374A (en)
KR (1) KR20010088793A (en)
CN (1) CN1323218A (en)
AU (1) AU763678B2 (en)
CA (1) CA2338644A1 (en)
EE (1) EE200100103A (en)
HU (1) HUP0103419A3 (en)
IL (1) IL141037A0 (en)
NO (1) NO20010859L (en)
NZ (1) NZ509592A (en)
PL (1) PL346298A1 (en)
SK (1) SK2472001A3 (en)
WO (1) WO2000010594A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001062273A1 (en) * 2000-02-22 2001-08-30 Hannu Sariola The use of gdnf family-related compounds for manufacturing products for treating testicular tumors
CZ303358B6 (en) * 1998-07-06 2012-08-15 Nsgene A/S Neurotrophic polypeptide, process for its preparation and use, encoding polynucleotide, corresponding vector and host cell, synthetic gene and antibody

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006028278A1 (en) * 2004-09-09 2006-03-16 Kyoto University Method of in-vitro proliferation of androgenetic line stem cell

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997022697A1 (en) * 1995-12-20 1997-06-26 Ontogeny, Inc. TGFβ SIGNAL TRANSDUCTION PROTEINS, GENES, AND USES RELATED THERETO
WO1997033911A1 (en) * 1996-03-14 1997-09-18 Washington University Persephin and related growth factors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997022697A1 (en) * 1995-12-20 1997-06-26 Ontogeny, Inc. TGFβ SIGNAL TRANSDUCTION PROTEINS, GENES, AND USES RELATED THERETO
WO1997033911A1 (en) * 1996-03-14 1997-09-18 Washington University Persephin and related growth factors

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
CHEMICAL ABSTRACTS, 1 January 1900, Columbus, Ohio, US; abstract no. 126:302072, CHEN Y, DICOU E, DJAKIEW D: "CHARACTERIZATION OF NERVE GROWTH FACTOR PRECURSOR PROTEIN EXPRESSION IN RAT ROUND SPERMATIDS AND THE TROPHIC EFFECTS OF NERVE GROWTH FACTOR IN THE MAINTENANCE OF SERTOLI CELL VIABILITY" XP002943863 *
DATABASE CA [online] RUSSO, MARIO A.: "Expression of the p75 Neurotrophin Receptor in the Developing and Adult Testis of the Rat", XP002943865, accession no. STN International Database accession no. 126:234027 *
DATABASE DIALOG EMBASE 1 January 1900 (1900-01-01), LAMB D J: "GROWTH FACTORS AND TESTICULAR DEVELOPMENT", XP002943866, Database accession no. 1993213709 *
DATABASE DIALOG EMBASE 1 January 1900 (1900-01-01), SAEZ J M, ET AL: "CELL-CELL COMMUNICATION IN THE TESTIS", XP002943868, Database accession no. 1992141185 *
DATABASE DIALOG EMBASE 1 January 1900 (1900-01-01), SPITERI-GRECH J, NIESCHLAG E: "PARACRINE FACTOR RELEVANT TO THE REGULATION OF SPERMATOGENESIS - A REVIEW", XP002943867, Database accession no. 1993204753 *
DATABASE DIALOG MEDLINE 1 January 1900 (1900-01-01), BITGOOD M J, SHEN L, MCMAHON A P: "SERTOLI CELL SIGNALING BY DESERT HEDGEHOG REGULATE THE MALE GERMLINE", XP002943864, Database accession no. 96398786 *
DATABASE DIALOG MEDLINE 1 January 1900 (1900-01-01), MASURE S, ET AL: "MOLECULAR CLONING, EXPRESSION AND TISSUE DISTRIBUTION OF GLIAL-CELL-LINE DERIVED NEUROTROPHIC FACTOR FAMILY RECEPTOR ALPHA-3 (GFRALPHA-3)", XP002943869, Database accession no. 98149678 *
DATABASE STN CAPLUS 1 January 1900 (1900-01-01), WIDENFALK J, ET AL: "NEURTURIN AND GLIAL CELL LINE-DERIVED NEUROTROPHIC FACTOR RECEPTOR.BETA. (GDNFR-BETA.), NOVEL PROTEINS RELATED TO GDNF AND GDNFR-.ALPHA. WITH SPECIFIC CELLULAR PATTERNS OF EXPRESSION SUGGESTING ROLES IN THE DEVELOPING AND ADULT NERVOUS SYSTEM AND IN PERIPHERAL ORGANS", XP002943862, Database accession no. 1997:708091 *
INT. J. DEV. BIOL. (SUPPL. 1, PROCEEDINGS OF THE FIRST CONGRESS OF THE SPANISH SOCIETY OF DEVELOPMENTAL BIOLOGY), 1996, pages 227S - 228S *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CZ303358B6 (en) * 1998-07-06 2012-08-15 Nsgene A/S Neurotrophic polypeptide, process for its preparation and use, encoding polynucleotide, corresponding vector and host cell, synthetic gene and antibody
WO2001062273A1 (en) * 2000-02-22 2001-08-30 Hannu Sariola The use of gdnf family-related compounds for manufacturing products for treating testicular tumors

Also Published As

Publication number Publication date
HUP0103419A3 (en) 2004-06-28
AU763678B2 (en) 2003-07-31
JP2002523374A (en) 2002-07-30
AU5375299A (en) 2000-03-14
CA2338644A1 (en) 2000-03-02
IL141037A0 (en) 2002-02-10
NZ509592A (en) 2003-01-31
HUP0103419A2 (en) 2002-01-28
NO20010859D0 (en) 2001-02-20
PL346298A1 (en) 2002-01-28
EE200100103A (en) 2002-06-17
EP1105151A1 (en) 2001-06-13
NO20010859L (en) 2001-04-19
KR20010088793A (en) 2001-09-28
CN1323218A (en) 2001-11-21
SK2472001A3 (en) 2001-09-11

Similar Documents

Publication Publication Date Title
Harvey Extrapituitary growth hormone
Michaelidis et al. Inactivation of bcl-2 results in progressive degeneration of motoneurons, sympathetic and sensory neurons during early postnatal development
Solloway et al. Mice lacking Bmp6 function
Vassalli et al. Activin/inhibin beta B subunit gene disruption leads to defects in eyelid development and female reproduction.
Allard et al. A rat G protein‐coupled receptor selectively expressed in myelin‐forming cells
Spears et al. The role of neurotrophin receptors in female germ-cell survival in mouse and human
Zhao et al. Characterization of GDF-10 expression patterns and null mice
McFarlane et al. A role for the fibroblast growth factor receptor in cell fate decisions in the developing vertebrate retina
Hendershot et al. Conditional deletion of Hand2 reveals critical functions in neurogenesis and cell type-specific gene expression for development of neural crest-derived noradrenergic sympathetic ganglion neurons
Koibuchi et al. Effect of altered thyroid status on neurotrophin gene expression during postnatal development of the mouse cerebellum
Gao et al. Impaired dendritic development and synaptic formation of postnatal-born dentate gyrus granular neurons in the absence of brain-derived neurotrophic factor signaling
Wu et al. Microphthalmia resulting from MSX2-induced apoptosis in the optic vesicle
Duncan et al. Ectopic Pax6 expression disturbs lens fiber cell differentiation
McDowell et al. Gremlin induces ocular hypertension in mice through Smad3-dependent signaling
Song et al. Spatial and temporal expression of basic fibroblast growth factor (FGF-2) mRNA and protein in early Xenopus development
Patel et al. Overexpression of FGF-2 alters cell fate specification in the developing retina of Xenopus laevis
Hung et al. Retinal ablation and altered lens differentiation induced by ocular overexpression of BMP7
Streichert et al. A diffusible factor from normal retinal cells promotes rod photoreceptor survival in an in vitro model of retinitis pigmentosa
Phelps et al. Pituitary Hormones as Neurotrophic Signals: Update on Hypothalamic Differentiation in Genetic Models of Altered Feedback2
Kuny et al. Differential gene expression in eyecup and retina of a mouse model of Stargardt-like macular dystrophy (STGD3)
Zhu et al. Carboxypeptidase E is required for normal synaptic transmission from photoreceptors to the inner retina
Kreidberg et al. Coordinate action of Wt1 and a modifier gene supports embryonic survival in the oviduct
AU763678B2 (en) The use of glial cell line-derived neurotrophic factor family-related compounds for regulating spermatogenesis and for preparing male contraceptives
Harrison et al. Cell Death during Development of Testis and Cerebellum in the Mutant MouseWeaver
US20040223952A1 (en) Generation and/or reduction of new lung tissue in an affected lung

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 99812289.0

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ CZ DE DE DK DK DM EE EE ES FI FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

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

Ref document number: 141037

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 509592

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: PV2001-623

Country of ref document: CZ

WWE Wipo information: entry into national phase

Ref document number: 2472001

Country of ref document: SK

Ref document number: 53752/99

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: PA/a/2001/001892

Country of ref document: MX

Ref document number: 1020017002234

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 1999939468

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: PV2001-623

Country of ref document: CZ

Ref document number: 1999939468

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1020017002234

Country of ref document: KR

ENP Entry into the national phase

Ref document number: 2338644

Country of ref document: CA

Ref document number: 2338644

Country of ref document: CA

Kind code of ref document: A

WWG Wipo information: grant in national office

Ref document number: 53752/99

Country of ref document: AU

WWW Wipo information: withdrawn in national office

Ref document number: 1999939468

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1020017002234

Country of ref document: KR

WWR Wipo information: refused in national office

Ref document number: PV2001-623

Country of ref document: CZ