EP0879247A1 - Recepteurs du facteur neurotrophique derive de lignees de cellules gliales - Google Patents

Recepteurs du facteur neurotrophique derive de lignees de cellules gliales

Info

Publication number
EP0879247A1
EP0879247A1 EP96942748A EP96942748A EP0879247A1 EP 0879247 A1 EP0879247 A1 EP 0879247A1 EP 96942748 A EP96942748 A EP 96942748A EP 96942748 A EP96942748 A EP 96942748A EP 0879247 A1 EP0879247 A1 EP 0879247A1
Authority
EP
European Patent Office
Prior art keywords
gdnf
cells
ret
receptors
compounds
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP96942748A
Other languages
German (de)
English (en)
Other versions
EP0879247A4 (fr
Inventor
Carlos Ibanez
Petro Suvanto
Miles Trupp
Mart Saarma
Urmas Arumae
Hannu Sariola
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of EP0879247A1 publication Critical patent/EP0879247A1/fr
Publication of EP0879247A4 publication Critical patent/EP0879247A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/566Immunoassay; Biospecific binding assay; Materials therefor using specific carrier or receptor proteins as ligand binding reagents where possible specific carrier or receptor proteins are classified with their target compounds

Definitions

  • the present invention relates to the identification of receptors for and functions of GDNF, and cell lines expressing the receptors.
  • Glial cell line-derived neurotrophic factor is a trophic polypeptide.
  • GDNF was shown to promote survival of adult substantia nigra neurons in vivo following pharmacological treatments and lesions that mimic Parkinsonian syndromes (Beck et al. , 377 Nature 339, 1995; Tomac et al. , 373 Nature 335, 1995)
  • GDNF was originally reported to be highly specific for dopaminergic neurons, several other potent activities of this molecule have subsequently been demonstrated, including survival and phenotypic responses in facial and spinal motor neurons (Henderson et al. , 266 Science 30 1062, 1994; Oppenheim et al. , 373 Nature 344, 1995; Yan et al.
  • noradrenergic neurons of the locus coeruleus (Arenas et al. , Neuron, in press, 1995), cerebellar Purkinjie cells (Mount et al. , 92 PNAS 9092, 1995), sympathetic and sensory neurons in peripheral ganglia (Trupp et al. , 130 J. Cell Biol. 137, 1995) and for populations of peripheral neurons with target-derived and paracrine mode of action (Trapp, M. et. al., /. Cell Biol, 130, 137-148 (1995); Pitchel, J. ,
  • GDNF may have potent therapeutical applications.
  • GDNF maintains dopaminergic neurons of the substantia nigra in experimentally induced Parkinsons disease in rodents (Beck et al. (1995) Nature, 373, 339-341; Tomac et al. (1995) Nature, 373, 335-339) and leads to functional recovery in Parkinsonian rhesus monkeys (Gash et al. (1996) Nature, 380, 252-255).
  • GDNF treatment also rescues about half of the experimentally axotomized murine motoneurons (Oppenheim et al. (1995) Nature, 373, 344-346; Li et al. (1995) Proc. Natl, Acad. Sci.
  • GDNF appears to be a distant member of the transforming growth factor-beta (TGF- 13) superfamily of multifunctional cytokines, which includes TGF- ⁇ s, activins, bone-morphogenetic proteins (BMPs) and growth and differentiation factors (GDFS) (Roberts et al. , 327 Philos. Trans. R. Soc. Land.
  • TGF- 13 transforming growth factor-beta
  • BMPs bone-morphogenetic proteins
  • GDFS growth and differentiation factors
  • TGF-j ⁇ and related ligands are known to suppress proliferation in epithelial and immune cells, to function as morphogens in early development, to induce ectopic expression of skeletal tissue, and to promote survival and differentiation of neurons.
  • TGF- 3 superfamily proteins interact with numerous receptor subunits on the surface of responsive cells (Attisano et al. , 1222 Mol.Cell
  • Type II receptors which represent binding proteins of 55kD, 70kD and 3OOkD, respectively.
  • Type III receptors are abundantly expressed transmembrane proteoglycans of approximately 3OOkD with a short cytoplasmic tail, and are thought to function in recruitment of ligand to an oligomeric receptor complex (Lopez-Casillas et al., 67 Cell 785,
  • Type III receptor is required on some cell lines for TGF-32 binding to the signaling receptors.
  • Type I and type II receptors are transmembrane proteins with an intracellular serine-threonine kinase domain and can therefore transmit downstream signals upon ligand binding (Attisano et al. , 75 Cell 671 , 1993; Derynck, 1994 supra).
  • Type II receptors are constitutively activated kinases which upon ligand binding recruit type I receptors to a signaling complex.
  • type I receptors are phosphorylated by type II receptors on a juxtamembrane domain rich in serine residues, this phosphorylation is thought to result in the activation of the ser-thr kinase activity of type I receptors and in downstream signaling (Wrana et al. , 370 Nature 341, 1994).
  • TGF-3 superfamily proteins can not bind to type I receptors in the absence of type II receptors, although in some cases, type I receptors are necessary for efficient binding to type II receptors (Letsou et al., 80 Cell 899, 1995).
  • cDNA clones of type I, II and III receptors for TGF-3S, activins and BMPs have been isolated by either expression or homology cloning, including seven mammalian type I receptors, four type II receptors and one type III betaglycan receptor. Additional membrane proteins binding different members of this family include glycosylphosphatidyl inositol (GPS)-linked 150kD and 180kD proteins of unknown structure and function (MacKay and Danielpour, 266 _Biol. Chem. 9907, 1991), and endoglin, a 180kD disulphide linked dimer which binds TGF-/31 but not TGF- 32.
  • GPS glycosylphosphatidyl inositol
  • GDNF receptors The isolation and characterization of GDNF receptors is a prerequisite for the understanding of the full range of biological actions of GDNF and the signaling events that take place upon GDNF binding to responsive cells. Until now, progress in this area has been hampered by the lack of cell lines responsive to
  • GDNF that is, cell lines comprising GDNF receptors.
  • Receptors for GDNF are disclosed herein, as are cell lines expressing the same. Methods for identifying and isolating these receptors are also disclosed.
  • the present invention relates to isolated receptors which bind
  • the present invention relates a method for determining compounds or compositions which bind GDNF receptors.
  • the present invention relates to methods for identifying homologs of GDNF by screening for compounds or compositions which have similar biological effects, such as tyrosine phosphorylation, increase in c-fos mRNA, and and increases in cell survival.
  • the present invention relates to method for identifying analogs of GDNF by screening for compounds or compositions which are antagonistic for the biological effects of GDNF, such as are listed above.
  • Fig. 1 Binding of 125 I-GDNF to receptors on chick sympathetic neurons, (a) Saturation steady-state binding of 125 I-GDNF to ElO embryonic chick sympathetic neurons. Data are expressed as mean ⁇ SD of triplicate determinations, (b)
  • Fig.2 Affinity labeling of GDNF receptors on chick sympathetic neurons.
  • 125 I- GDNF was cross-linked to ElO embryonic chick sympathetic neurons, receptor complexes were fractionated by SDS/PAGE and visualized by gel autoradiography (middle lane). A doublet at lOOkD and a 300kD complex are indicated by arrows. Excess cold GDNF prevented cross-linking of 125 I-GDNF (right lane). For comparison, crosslinking of 1 5 I-TGF-0 to mink lung epithelial cells MvILu is also shown (left lane). Molecular weight markers are indicated in kD.
  • Fig.3 Affinity labeling of GDNF receptors on cell lines.
  • 125 I-GDNF was cross- -5- linked to C6 glioma, RN33B raphe nucleus, L6 myoblast and MN-1 motor neuron cell lines with either DSS or EDAC as crosslinker agents.
  • Receptor complexes were fractionated by SDS/PAGE and visualized by gel autoradiography. Excess cold GDNF prevented cross-linking of 125 I-GDNF (cold). Molecular weight markers are indicated in kD.
  • Fig. 4 Individual constituent affinities of GDNF receptor subunits in RN33B and MN-1 cells, (a) Sizes of different GDNF receptor complexes on RN33B and MN-1 cells after cross linking with EDAC or DSS. (b) and (c) 125 I-GDNF was cross ⁇ linked to RN33B (b) or MN-1 (c) cells in the presence of increasing concentrations of cold GDNF. The percentage of 125 I-GDNF binding to the indicated receptor subunit is plotted as a function of the concentration of cold GDNF used during binding.
  • Fig. 5 Expression of GDNF mRNA in cell lines expressing GDNF receptors, (a) Autoradiogram of an RNAse protection assay using equal amounts of total RNA from the indicated cell lines. Kidney post natal day 1 and yeast tRNA were used as positive and negative controls, respectively, (b) Quantification of the level of GDNF mRNA in different cell lines relative to the level in PI kidney, undiff
  • RN33B undifferentiated RN33B cells; diff RN33B, differentiated RN33B cells; diff RN338+GDNF, RN33B cells differentiated in the presence of GDNF.
  • Fig. 6 Expression of mRNA for c-ret in different cell lines.
  • Fig. 7 GDNF stimulation of tyrosine phosphorylation of ERKs in RN33B and MN-1 cells.
  • RN33B (a) or MN-1 (b) cell monolayers were exposed to 50 ng/ml GDNF during the indicated periods of time (in minutes), cell lysates were fractionated by SDS/PAGE and Western blots probed with an anti-phosphotyrosine antibody (aP-Tyr). The blots were stripped and reprobed with an anti ERK2 antibody (a-ERK2) that recognizes both p42 crk 12 and p44 er l (arrows to the right).
  • Molecular weight markers are indicated in kD.
  • GDNF stimulation of c-fos mRNA expression in RN33B and MN-1 cells GDNF stimulation of c-fos mRNA expression in RN33B and MN-1 cells.
  • RN33B (a) or MN-1 (b) cell monolayers were exposed to 50 ng/ml GDNF during the indicated periods of times, total RNA was extracted and fractionated in agarose gels and Northern blots probed with a 32 P-labeled rat c-fos probe. Shown are x-ray autoradiograms of filters washed at high stringency.
  • GDNF increased the survival of RN46A cells.
  • GDNF stimulates survival of serum-deprived MN-1 cells.
  • GDNF stimulates rapid and transient tyrosine phosphorylation of several proteins (asterisks) in MN-1 cells. Time of GDNF treatment (in minutes), and molecular weight markers are indicated,
  • c Rapid and sustained ERK1 and ERK2 tyrosine phosphorylation stimulated by GDNF in MN-1 cells.
  • c-RET is a signal transducing receptor for GDNF.
  • GDNF induces tyrosine phosphorylation of c-RET in MN-1 cells.
  • c-RET tyrosine phosphorylation was detected already 5 minutes after addition of GDNF (upper panel). Saturation was observed at 30 ng/ml GDNF (lower panel).
  • c-ret expression is sufficient to mediate binding and biological responses to GDNF in fibroblasts.
  • Iodinated GDNF could be cross linked to 3T3 cells stably transfected with MEN2a-ret or wild type c-ret expression plasmids. Untransfected 3T3 cells (3T3) did not bind GDNF. The specificity of the binding was demonstrated by displacement of the labeling with 50X excess cold GDNF.
  • GDNF promotes survival and growth responses in 3T3 fibroblasts stably transfected with a c-ret expression plasmid. Untransfected cells did not respond to GDNF.
  • RPA Ribonuclease protection analysis
  • GDNF mRNA expression in the developing striatum mRNA expression is indicated in arbitrary units where 100 corresponds to the level of expression in the respective regions in newborn animals.
  • c-RET is expressed in GDNF-responsive substantia nigra dopaminergic neuron
  • Fig. 15 a-c. PC 12 and NB2/a cells respond to GDNF and bind GDNF.
  • GDNF promotes survival of serum-deprived PC12 cells
  • GDNF increases the number of NB2/a cells
  • 125 I-GDNF binds to PC 12 and NB2/a cells in the absence (open column) or presence (filled column) of 50-fold unlabeled GDNF.
  • Fig. 16 Affinity crosslinking of 125 I-GDNF to cell lines.
  • 125 I-GDNF was crosslinked to PC12 cells (lane 1), SY5Y cells (lane 2), E20 rat kidney cells (lane 3) and NB2/a cells (lane 4), and the resulting complexes were precipitated from detergent lysates by anti-GDNF antibodies (Santa Cruz).
  • GDNF specifically binds to c-RET.
  • a 125 I-GDNF was crosslinked to NB2/a cells in the presence (+) or absence (-) of 1000-fold excess of unlabeled GDNF (PeproTech EC Ltd.), and the resulting complexes were precipitated from detergent lysates by cocktail of monoclonal and polyclonal (Santa Cruz) anti-c-RET antibodies recognizing the extracellular and intracellular domain of cRET, respectively. Lysates were also precipitated by monoclonal anti-neurof ilament antibodies 13AA8 (lane 3), by Protein A-Sepharose (lane 4) and by WGA- Agarose (lane 5).
  • I25 I-GDNF binds to COS cells transiently expressing c-RET, but not to mock-transfected (with pBK-CNV plasmid) COS cells. Open column represents binding in the presence, and filled column in the absence of 50- fold excess of unlabeled GDNF.
  • GDNF increases tyrosine phosphorylation of c-RET in transfected COS cells.
  • c-RET was immunoprecipitated from detergent lysates of GDNF-treated ( + ) (lane 1) or untreated (-) (lane 2) COS cells transfected (lane 3) with c-ret cDNA or mock-transfected with PBK-CMV plasmid. (a) immunoblot probed with anti-c-RET antibodies (Santa Cruz), (b) the same filter reprobed with anti-phosphotyrosine antibodies.
  • GDNF binds in situ to c-ret-positive developing enteric neurons,
  • Fig. 20 a - b Crosslinked GDNF-c-RET-complexes are obtained from GDNF- responsive cell lines and from c-ret-transfected cells
  • I25 I-GDNF was crosslinked with EDAC to PC 12 cells, NB2/a cells, dissociated E20 rat kidney cells, and COS cells, and the resulting complexes were precipitated by anti-GDNF antibodies
  • EDAC-crosslinked 125 I-GDNF-c-RET complexes were immunoprecipitated with anti-c-RET antibodies from the extracts of PC 12 cells, stably c-rct-transfected (Ret.-3T3) or mock-transfected (mock-3T3) 3T3 cells, as well as from dissociated
  • the ⁇ 50K bands in all gels are the crosslinked dimers of GDNF.
  • GDNF increases c-RET autophosphorylation in stably transfected
  • 3T3 cell line (a) GDNF dose-dependently increases tyrosine phosphorylation of 160 kD isoform of c-RET in c-ret-transfected (ret-3T3) but not in mock-transfected (mock) cells, (b) GDNF time-dependently increases tyrosine phosphorylation of 160 kD isoform of cRET in c-ret-transfected 3T3 cells.
  • Upper panels (Ret.-PTyr) are the immunoblots stained with anti-phosphotyrosine antibodies, and lower panels
  • GDNF increases the number of trkC-3T3 fibroblasts transiently expressing c-RET (open squares), but not mock-transfected cells (filled squares).
  • c-ret and mock-transfected cells in five parallels were treated with rat GDNF at indicated concentrations, or with NT-3, for five days.
  • Cell number quantified with AbacusTM Cell Proliferation Kit (Clontech), is expressed as a percent of the control cells without growth factors. *, p ⁇ 0.001 compared to mock transfected cells.
  • Fig. 23 a-b Purification of receptor from L6 myeloblast cells, (a) Plasmon resonance analysis of fractions obtained from anion exchange chromatography of
  • Fig. 23 Autoradiographic film of the ligand blot 125 I-GDNF with proteins from adult rat brain (lane 2) and liver (lane 3). 50-fold excess of unlabeled GDNF (lane 1)
  • GDNF receptors A prerequisite for the understanding of the full range and mechanisms of action of GDNF is the characterization of GDNF receptors and their signaling pathways. Although receptors for other members of the TGF- ⁇ superfamily are well characterized, GDNF receptors remained undefined until this disclosure. Disclosed herein is the biochemical characterization of GDNF receptors and their downstream responses in sympathetic neurons and responsive cell lines. Using affinity labeling, multiple GDNF binding subunits that mediate cooperative binding of GDNF to embryonic sympathetic neurons are identified. Screening of over thirty cell lines initially revealed high expression of GDNF binding proteins of 55 kD, 70 kD, 135 kD and 300 kD in conditionally immortalized neuronal precursors from the raphe nucleus.
  • GDNF receptors were highly induced after neuronal differentiation of these cells, which then became sensitive to the survival-promoting effects of GDNF.
  • Different combinations of these subunits were also seen in glioma, myoblast and Sertoli cells.
  • a different receptor pattern was found in a motor neuron hybrid cell line, where the predominant component was a CPI-anchored protein of 155kD.
  • GDNF receptor subunits of 55kD, 70kD, 135kD, and 300kD are novel proteins.
  • 155kD subunit was subsequently determined to be the product of the c-ret proto ⁇ oncogene, c-RET, a receptor tyrosine kinase crucial for the development of parts of the excretory and nervous systems.
  • GDNF stimulated ERK tyrosine phosphorylation and c-fos mRNA expression with different time-courses in raphe nucleus and motor neuron cell lines, suggesting that different complements of
  • GDNF receptor subunits can form distinct signaling complexes.
  • c-RET was identified as receptor for GDNF on additional cell lines. GDNF rescues c-RET-positive dopaminergic and noradrenergic neurons in lesion models of Parkinson's disease, suggesting that cRET may mediate the anti-Parkinsonian effects of GDNF in the adult brain.
  • c-ret proto-oncogene (Takahashi et al. (1985) Cell, 42, 581-588) encodes a protein that is structurally related to receptor tyrosine kinases (Takahashi et al. (1988) Oncogene, 3,571-578).
  • c-ret- encoded proteins with molecular weights of 160 kD and 140 kD are described, representing the fully and partially glycosylated isoforms of 120 kD core protein, respectively (Takahashi et al, 1988).
  • c- RET is activated by homodimerization followed by phosphorylation of its tyrosine residues.
  • c-ret mRNA is expressed primarily in the nervous and excretory systems, c-ret mRNA is found in dorsal root, sympathetic, enteric and cranial ganglia (Pachnis et al. , Development 119, 1005-17 (1993), as well as in post migratory neural crest cells and in various tumors of neural crest origin, including pheochromocytoma, medullary thyroid carcinoma and neuroblastoma (Ikeda, I. , et al Oncogene 5, 1291-6 (1990); Santoro, M. , et al. Oncogene 5, 1595-1598 (1990).
  • NGF and GDNF promote survival of PC12 cells, whereas only NGF induces their differentiation, suggesting only a partial overlap in the signaling pathways of c- RET and trkA, a receptor for NGF.
  • Binding of an adaptor protein Grb2 to oncogenic forms of c-RET has been demonstrated (Borrello et al. (1994) Oncogene, 9, 1661-1668). However, the details of the pathways are completely unknown.
  • GDNF as a ligand, it is possible to address the intracellular signaling of c-RET upon GDNF binding.
  • GDNF is abundantly expressed in the muscle layer of the gastrointestinal tract and in the condensing mesenchyme of the kidney (Suvanto et al. (1996) Eur. J. Neurosci. , 8, 816-822). Further, as disclosed herein, GDNF specifically binds c-RET-positive cells in developing gut, GDNF can be crosslinked to c-RET in several cell lines and in developing kidney, GDNF specifically induces tyrosine phosphorylation of c-RET, and ectopical expression of c-RET in 3T3 cells confers a biological response of these cells to GDNF. Thus, c-RET is activated by GDNF and mediates its functions.
  • the product of the c-ret proto-oncogene plays important roles in human disease. Rearrangements and mutations in the c-ret gene are associated with several tumors e.g. familial medullary thyroid carcinoma, multiple endocrine neoplasia type 2, etc. , but also with Hirschsprung disease, a disorder that is characterized by the absence of enteric neurons in the hindgut, resulting in obstipation and megacolon in infants and adults (reviewed in Mak, Y. F. and Ponder, B.A. J. (1996)C «rr. Op. Genet. Dev., 6, 82-86). Identification of GDNF as a ligand for c-RET further enables the analysis of the molecular basis of these diseases.
  • GDNF receptor and "receptor for GDNF” as used herein each refer to a single subunit which binds GDNF as well as combinations of the receptor subunits which bind GDNF.
  • effect means an alteration or change.
  • An effect can be positive, such as causing an increase in some material, or negative, e.g. , antagonistic or inhibiting.
  • homolog refers to a compound or composition having a similar biological effects as GDNF, such as are disclosed herein.
  • analog refers to a compound or composition having an antagonistic effect on the biological effects of GDNF.
  • isolated as used herein in reference to a GDNF receptor means a compound which has been separated from its native environment or, if recombinantly expressed, from its expression environment.
  • substantially pure as used herein in reference to a compound means an isolated compound which has been separated from other components which naturally accompany it. Typically, a compound is substantially pure when it is at least 75 %, more preferably at least 90%, and most preferably 99% of the total material as measured, for example, by volume, by wet or dry weight, or by mole percent or mole fraction.
  • non-permissive culture conditions refers to conditions which do not normally support survival of the cells being cultured in vitro, e.g. , temperature, media components, etc.
  • an excess as used in reference to the addition of labeled GDNF in a competitive assay refers to an amount of labeled GDNFsufficient to facilitate the detection of a competing compound - for example, an amount of labeled GDNF which is twice the amount of the compound to be tested.
  • binding refers to the interaction between the GDNF ligand and its receptor, the binding being of a sufficient strength and for a sufficient time to allow the detection of said binding under the conditions of the assays disclosed herein.
  • GDNF receptor is present in multiple neuronal and non-neuronal cell types
  • GDNF receptor is composed of multiple subunits which cooperate to achieve high affinity binding
  • members of the ERK/MAP kinase family are components of the GDNF signaling mechanism
  • c-RET is a functional receptor for GDNF.
  • GDNF receptors are identified herein as GDNF receptors. These novel GDNF receptors were found in cells lines of different origins, although they appeared to be most abundant in neuronal cells. Preferably, the cell lines are selected from the group consisting of RN33B, RN46A, and C6 (see Table I), with RN33B being most preferred.
  • RN33B is selected from the group consisting of RN33B, RN46A, and C6 (see Table I), with RN33B being most preferred.
  • the identification of GDNF receptors in many of these cell types suggests novel cellular populations responsive to GDNF in vivo. GDNF has been shown to promote survival and phenotype of distinct subpopulations of neurons, in particular dopaminergic and noradrenergic central neurons, as well as spinal and facial motor neurons.
  • GDNF receptors in cell lines derived from the medullary raphe indicate that serotonergic neurons may also respond to GDNF in vivo.
  • the endogenous expression of GDNF by these cells suggests that this factor may act in a paracrine/autocrine fashion within the raphe nucleus.
  • Expression of GDNF receptors in Sertoli TM4 cells suggests non- neuronal roles for GDNF in developing testis.
  • the temporal expression of GDNF mRNA in testis correlates with the expansion of the Sertoli cell population (Trupp et al. , supra) which, together with the discovery of GDNF receptors on the
  • TM4 cell line suggest an autocrine action of GDNF during Sertoli cell maturation.
  • the presence of GDNF receptors in rat myoblast L6 cells indicates a potential paracrine role of GDNF during myogenesis.
  • PC 12 cells which had been differentiated into sympathetic- like neurons with NGF did not express GDNF receptors under initial experimental conditions.
  • GDNF receptors were ultimately identified on PC 12 cells. GDNF receptors are absent in the pons noradrenergic cell line CATH.a.
  • GDNF elicits a more profound induction of the phenotype of noradrenergic neurons following 6-OH-dopamine injection than in the non-lesioned locus coeruleus.
  • Treanor et al. recently reported upregulation of GDNF binding in sections of the substantia nigra after medical forebrain bundle transaction (Treanor et al. , 21
  • GDNF receptor upregulation was also observed during in vitro differentiation of raphe nucleus cells. These lines have recently been shown to retain the ability to respond to local microenvironmental signals after transplantation into the adult brain, where they differentiate in a direction that is consistent with that of endogenous neurons in the transplantation site (Shihabuddin et al., 15 J. Neurosci. 6666, 1995). In vitro, however, a shift to the non- permissive temperature differentiates them along default pathways into glutamatergic (RN33B) or serotonergic (RN46A) phenotypes, respectively.
  • R33B glutamatergic
  • R46A serotonergic
  • GDNF receptor is composed of multiple subunits which cooperate to achieve high affinity binding.
  • the cooperative binding of GDNF to embryonic sympathetic neurons may thus be an indication of a multi- step mechanism of receptor assembly. Because binding assays were performed at 4°C, binding cooperativity is unlikely to have resulted from substantial lateral mobility of transmembrane receptor proteins, suggesting that GDNF binding induces conformational changes on receptor complexes that are partially preformed on the membrane.
  • the nearly identical affinities of the different GDNF receptor subunits obtained by crosslinking also support the notion of cooperative binding of GDNF to a partially pre-assembled receptor complex.
  • TGF-/3 receptors I, type II and type III TGF-/3 receptors.
  • GDNF has been detected in COS cells transfected with different combinations of known type I and type II TGF-3 superfamily receptors (Ibanez, C, unpublished; P. ten Dijke, personal communication), including the recently isolated type II receptor for BMPs (Rosenzweig et al. , 92 PNAS USA 7632, 1995) and a novel brain- specific type I receptor (Ryden et al. , 21 Abs. Soc. Neurosci 1754, 1995).
  • type I and type II TGF-3 superfamily receptors Ibanez, C, unpublished; P. ten Dijke, personal communication
  • c-RET is a receptor for GDNF
  • GDNF receptors were found in a motor neuron-neuroblastoma hybrid cell line, but not in a basal forebrain cell which was also a hybrid with the same neuroblastoma, suggesting that the receptors detected on MN-1 cells represent physiologically relevant motor neuron GDNF receptors.
  • GDNF expression could not be detected in the motor neuron cell line, consistent with a target-derived mode of action for muscle-derived GDNF in vivo (Henderson et al., 266 Science 1062, 1994; Trupp et al. , supra).
  • GDNF binds to and induces tyrosine phosphorylation of the these receptor which were identified as the product of c-ret.
  • c-ret was also able to mediate GDNF binding and survival/growth responses to GDNF upon transfection into naive fibroblasts.
  • dopaminergic neurons of the adult substantia nigra were found to express high levels of c-ret mRNA, and c-RET expressing dopaminergic and noradrenergic neurons in the CNS responded to the protective effects of exogenous GDNF in vivo.
  • c-RET receptor tyrosine kinase is a signal transducing receptor for GDNF. This finding is surprising, given that all receptors for members of the TGF- ⁇ superfamily characterized so far are receptor serine-threonine kinases (Derynck, R. Trends Biochem Sci 19, 548-553
  • GDNF is in fact a very divergent member of the TGF-0 superfamily, with which it shares primarily the spacing between conserved cysteine residues in the amino acid sequencer. Its ability to interact with a receptor tyrosine kinase indicates a further functional divergence from other members, of the TGF-0 superfamily. Conversely, these findings could suggest that other TGF-/3 superfamily members may also utilize receptor tyrosine kinases.
  • GDNF binds to COS cells ectopically expressing the c-ret proto-oncogene; ii) GDNF can be chemically crosslinked to the product of the c-ret proto-oncogene ectopically expressed in COS cells or from NB2/a and PC 12 cells; iii) the c-ret proto-oncogene product ectopically expressed in COS cells, but also in NB2/a cells, becomes rapidly phosphorylated on tyrosine residues upon GDNF binding; iv) GDNF promotes biological effects i.e.
  • GDNF specifically binds to RET-expressing ( Figure 19 c, d, h) enteric neurons and the tips of ureteric buds in developing kidney. These tissues were absent or severely reduced in c-rct-def icient mice (Schuchardt et al. (1994) Nature, 367,380-383; Durbec et al. (1996) Development, 122, 349-358).
  • the data disclosed herein further demonstrate GDNF-c-RET complexes from GDNF- responsive and c-ret-transfected cells and from embryonic kidney cells.
  • GDNF signal transducing mechanisms in raphe nucleus and motor neuron cell lines has been conducted.
  • the downstream responses elicited by GDNF in these cells demonstrate that the GDNF binding proteins identified herein represent functional GDNF receptors.
  • the initial biochemical characterization of GDNF signal transduction pathways has identified members of the ERK/MAP kinase family as components of the GDNF signaling mechanism.
  • ERK/MAP kinase activation by phosphorylation is the final step in a cascade of kinases that is set in motion after activation of the Ras pathway by various growth factors, including TGF-/J (Yan et al., 269 J. Biol Chem. 13231, 1994; Hartsough and Mulder, 270 J. Biol. Chem. 7117, 1995) and nerve growth factor (Thomas et al. , 68 Cell 1031 ,
  • ERK2 has also been shown to form part of the signal transduction pathway activated by several cytokines, such as interferons and interleukins, which are not known to activate Ras (David et al. , 269 Science 1721 , 1995). Whether or not Ras activation is one of the steps in the signaling transduction mechanism of GDNF is an area of further interest.
  • ERK2 in RN33B cells but relatively slower (maximum at 15 min) and more sustained (still detectable after 120 min) phosphorylation of ERK2, but not ERKI, in MN-1 cells. That these differences may have functional significance is suggested by recent observations made in PC 12 cells treated with different growth factors. Exposure of PC 12 cells to NGF or fibroblast growth factor (FGF) results in neuronal differentiation and in sustained elevation of Ras activity and ERK tyrosine phosphorylation (Qiu and Green, 7 Neuron 977, 1991). In contrast, treatment with epidermal growth factor, which stimulates DNA synthesis and proliferation of PC 12 cells, results in only transient ( ⁇ 1 hr) activation of Ras and ERKs (Qiu and Green, 1991).
  • NGF fibroblast growth factor
  • ERK activation underlie different biological responses in PC 12 cells.
  • the different patterns of GDNF receptors and GDNF- induced ERK phosphorylation in RN33B and MN-1 cells suggest that different GDNF receptor subunits can cooperate to assemble distinct signaling complexes in different cell types. Whether different GDNF signal transduction pathways underlie the different biological effects of GDNF is an area of further interest.
  • ERKs Upon activation, ERKs translocate to the nucleus where they phosphorylate and thereby regulate the activity of transcription factors which, in turn, control gene expression.
  • c-fos forms part of the AP-1 transcription factor complex, which is thought to be involved in the regulation of multiple genes, including growth factor, neuropeptide and neurotransmitter synthesizing enzyme genes (Gizang-Ginsberg and
  • c-fos could mediate the increase in the tyrosine hydroxylase (TH) expression observed upon GDNF treatment of central noradrenergic neurons, or the GDNF- induced upregulation of vasoactive intestinal peptide (VIP) and preprotachykinin-A (PPTA) mRNAs in cultured sympathetic neurons from the superior cervical ganglion (Trupp et al. , 130 J. Cell Biol. 137, 1995).
  • VIP vasoactive intestinal peptide
  • PPTA preprotachykinin-A
  • GDNF receptors identified on these cells are able to elicit relevant biological responses.
  • cessation of proliferation, differentiation, and GDNF responsiveness were concomitant with increased GDNF receptor expression in these cells, suggests that GDNF may be a survival factor for developing serotonergic raphe neurons in vivo.
  • the data of this patent disclosure suggest a role for ERKs and c-fos in GDNF-mediated neuron survival. This can be directly established using dominant negatives or antisense oligonucleotides.
  • the GDNF receptor subunits and complexes disclosed herein have wide- range applicability.
  • GDNF receptor facilitates rational drug design for drugs useful in treating, for example, neuronal disorders, particularly those involving neuronal cell death.
  • GDNF has been shown to promote survival of adult substantia nigra neurons in vivo following pharmacological treatments and lesions that mimic Parkinsonian syndromes, as well as survival responses in other neuronal cell lines.
  • the drugs can be tested for binding affinity to gdnf receptor, and for their influence on the downstream effect of GDNF disclosed below — i.e. , the phosphorylation of ERK2 and ERKI.
  • GDNF receptor has also been identified on malignant cell lines, design of drugs for use in cancer therapy is also evident.
  • the development of drugs to be used in treating bone-related diseases, i.e. , osteoporosis, and for promoting the healing of fractures is also contemplated.
  • isolated receptors according to the present invention can be used, inter alia, to screen for compounds or compositions which are analogs and homologs of GDNF.
  • the potential analogs and homologs can be screened initially in competitive binding assays employing either isolated receptor or cell lines expressing the receptor - i.e. , NB2/a cells - and I25 I-labeled GDNF. Methods such as those disclosed in Example 13 can be used. Analog or homolog activity can then be ascertained by further identifying those compounds or compositions which, for example, effect a decrease or increase, respectively, in the tyrosine phosphorylation of the RET proto-oncogene. Methods such as those disclosed in Example 17 can be used.
  • GDNF can be used to screen for and identify other receptors using the above-reference procedures, or variations thereof.
  • GDNF receptor also facilitates the development of antibodies, both polyclonal and monoclonal, against the receptor. These antibodies can be used to purify the receptors themselves, identify other cells expressing
  • the antibodies can initially be produced using the ligand/receptor complexes disclosed herein as the immunogens. Antibodies specific for the ligand can be eliminated from the polyclonal serum by absorption with the ligand. Hybridomas for monoclonal production can be selected on the basis of binding of ligand, with the expansion of only those clones which do not bind the ligand uncomplexed with the receptor.
  • the antibodies can be prepared by methods well known to those skilled in the art.
  • nucleic acids for the expression of recombinant GDNF receptor, both in vitro and in vivo, for diagnostic and therapeutic applications.
  • nucleic acids includes, for example, genomic DNA, mRNA, and cDNA.
  • oligonucleotide primers for isolating genomic DNA for GDNF receptor and receptor mRNA can be developed.
  • cDNA can be prepared from isolated mRNA.
  • the isolation and production of nucleic acids can be accomplished utilizing methods well known to those skilled in the art using standard molecular biology techniques such as are set forth in Maniatis et al. , Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 1982, inco ⁇ orated herein by reference.
  • Recombinantly produced receptors can be used in crystallography studies for rational drug design.
  • Recombinant extracellular domain can be produced and used as a drug in ligand sink applications, e.g. , for ligands with antagonistic properties.
  • the nucleic acids as set forth above can be utilized for gene therapy, using both in vivo and ex vivo techniques.
  • the nucleic acids can also be used to clone other related receptors using, for example, low stringency screens and reversed transcriptase PCR; and to produce cells overexpressing the receptors to screen for other ligands, e.g. , by panning, and other materials serving as receptor agonists, antagonists, or partial agonists and antagonists.
  • recombinantly produced receptor itself can be used for the screening assays.
  • cells expressing chimeric receptors can be produced using other TGF-0 receptor family members to elucidate signal pathways.
  • Intracellular targets of GDNF receptor can be identified using, for example, the yeast two-hybrid system. (Chen, et al. , 377
  • transgenic animals can be developed for testing the effects of the overexpression of GDNF receptor. Procedures can be utilized such as are described in Hogan et al. , Manipulating the Mouse Emblvo: A
  • cell lines and transgenic animals unable to express GDNFreceptor can be prepared to ascertain the effects of blocking signaling by
  • GDNF GDNF. Procedures such as are set forth in Wurst et al., Gene Targeting Vol. 126, edited by A. L. Joyner, IRL Press, Oxford University Press, Oxford, England, pp. 33-61, 1993, inco ⁇ orated herein by reference, can be utilized.
  • GDNF protein was quantified after silver staining of SDS/PAGE gels using standard curves obtained with commercial samples of proteins of molecular weight similar to that of GDNF.
  • Purified human TGF- ⁇ l was generously provided by Jun-ichi Koumegawa, Kirin Brewery, Tokyo, Japan. Proteins were labeled with Na- 125 I by the chloramine-T method to a specific activity of approximately 1 x 10 8 cpm/ ⁇ g.
  • binding assays were performed as follows. Cells were incubated with iodinated GDNF in Dulbecco's phosphatebuffered saline and 2 mg/ml bovine serum albumin (BSA) on Millipore Hydrophilic Durapore 96- well filtration plates. Following two hours of vigorous shaking at 4°C, the cells were washed twice with ice-cold binding buffer under vacuum. Dried filters were liberated and bound 125 I-GDNF quantified in a gamma counter. Non-specific binding was determined by addition of 500-fold excess of cold ligand to the binding mixtures. For affinity labeling, iodinated proteins were bound to monolayer cultures of primary neurons or cell lines.
  • BSA bovine serum albumin
  • dissociated chick sympathetic neurons Prior to binding, dissociated chick sympathetic neurons were cultured for 48 hours in the presence of NGF on polyornithine/laminin coated dishes. Plated cells were incubated with 10 ng/ml 125 I- GDNF at 4°C in binding buffer as described above. Ligand/receptor complexes were chemically cross-linked for thirty minutes at room temperature using either disuccinimidyl suberate (DSS) or l-Ethyl-3(-3-dimethylaminopropyl)-carbodiimide hydrochloride (EDAC) (Pierce Chemical, Rockland, IL).
  • DSS disuccinimidyl suberate
  • EDAC l-Ethyl-3(-3-dimethylaminopropyl)-carbodiimide hydrochloride
  • cells were washed twice with lOmM Tris/HCI buffered saline, 2 mM EDTA, 10% glycerol, 1 % NP-40, 1 % Triton x- 100, 10 ⁇ g/ml leupeptin, 10 ⁇ g/ml antipain, 50 ug/ml aprotinin, 100 ⁇ g/ml benzamidine hydrochloride, 10 ⁇ g/ml pepstatin and 1 mM PMSF(proteinase inhibitors from Sigma).
  • GDNF nerve growth factor
  • NGF nerve growth factor
  • Chicken sympathetic neurons isolated and prepared as previously described (Trupp et al. , supra). Saturation binding with iodinated GDNF was carried out on neurons isolated from embryonic day 10 (ElO) chick paravertebral sympathetic ganglia mechanically dissociated in the presence of trypsin. The preparation was preplated for two hours on untreated tissue culmre plastic in order to enrich in neurons and allow for re expression of receptors. Plots of saturation binding data produced a sigmoidal curve from which a Kd of 400 pM could be approximated (Fig. la).
  • GDNF receptors Over thirty cell lines were screened for expression of GDNF receptors using affinity labeling with iodinated GDNF (Table I, infra). Except as otherwise noted, all cell lines used in this study are available from and described by the American Type Culture Collection, Rockville, MD. A875 human neuroblastoma was provided by Mart Saarma, University of Helsinki, Finland.
  • CATH.A a noradrenergic cell line isolated from a tumor in the pons of transgenic mice expressing SV40 T antigen under the transcriptional control of a tyrosine hydroxylase promoter (Suri et al., 1993), was generated and provided by Dona Chikaraishi, Tufts University School of Medicine, Boston, MA.
  • the rat neural stem cell line C17-2 (Snyder et al., 68 Cell 33, 1992) was generated and provided by Evan Snyder, HarvardMedical School, Boston, MA.
  • LAN5 human neuroblastoma was provided by Sven Pahlman, Uppsala University, Sweden. David
  • SN6 cells a hybrid of mouse basal forebrain chohnergic neurons and the mouse neuroblastoma N18TG2 (Hammond et al. , 1986).
  • Human neuroblastoma SY5Y was a provided by David Kaplan, ABL-Basic Research Program, NCI-Frederick Cancer Research and Development Center, Frederick, MD.
  • ST15A rat neural stem cell line was kindly provided by Ron McKay, National Instituted of Health, MD.
  • the generation and characterization of raphe nucleus cell lines RN33B and RN46A has been described elsewhere (Whittemore and White, 1993). RN33B and RN46A cells were obtained from Dr. Scott Whittemore of the University of Miami.
  • the motor neuron hybrid cell line 2FI.10.14 (referred to here as MN-1) has been previously described (Salazar-Grueso et al. , 2 Neuroreport 505, 1991). Multiple GDNF receptor subunits were detected in various glial, neuronal and non-neuronal cells (Table I). A large molecular weight band of 300kD appeared to be the most prevalent species in several cell lines after crosslinking with disuccymidyl suberate (DSS), and it was the only receptor which appeared to bind ligand in the absence of all other receptors (Table I).
  • DSS disuccymidyl suberate
  • the consensus pattern in these cells after cross-linking with DSS consisted of the large molecular weight band of 300kD, and two other receptor subunits with molecular weights at 50-55kD and 65-70kD, respectively (Fig. 3 and Table I).
  • the 50-55kD component often ran as a doublet or triplet.
  • the smeary appearance and heterogeneous range of sizes displayed by Lhe large molecular weight component suggests a post-translational modification, presumably glycosylation, and appears similar to that previously described for type III betaglycan TGF-3 receptors. This species was somewhat smaller in the cells derived from the raphe nucleus, which could indicate either a distinct core protein or difference levels of glycosylation.
  • GDNF receptors could not be detected in pheochromocytoma PC 12 cells under the present assay conditions of 4°C, even after NGF-induced differentiation into a sympathetic neuron-like phenotype (Table I, and data not shown). No or very low GDNF receptor expression could be seen in various neuroblastomas, and in two pluripotent neuronal stem cells (Table I).
  • Presence (+) or absence (-) of specific GDNF receptor complexes in the designated cell line Presence (+) or absence (-) of specific GDNF receptor complexes in the designated cell line.
  • DSS cross-linked complexes. Like DSS, EDAC also cross-linked GDNF to receptors of 50-55kD and 65-70kD; the high molecular weight subunit of 300kD was, however, not as efficiently cross-linked by EDAC (Fig.3).
  • the raphe nucleus cell lines are only conditionally immortalized and do not show signs of transformation. At Lhe non-permissive temperature and in defined medium, they stop proliferating and differentiate into postmitotic neurons
  • FIG. 3 A distinct receptor complex was found on an embryonic mouse spinal cord motor neuron hybrid cell (Fig. 3). This line was obtained by fusion of E14 mouse spinal cord motor neurons and the N18TG2 mouse neuroblastoma, followed by selection of clones expressing high levels of choline acetyltransferase activity
  • MN-1 GDNF binding proteins seen on the motor neuron cell
  • Fig. 3 the predominant receptor in MN-1 cells was preferentially cross-linked with EDAC, although in these cells it was a larger protein of 155kD (Fig. 3). This was subsequently identified to be a c-RET receptor (see Example 9 below).
  • MN-1 cells also expressed 65-70kD binding proteins and low amounts of the 300kD receptor (Fig. 3 and Table 1).
  • GDNF receptor subunits display similar binding affinities or, whether they are all required to assemble a high affinity receptor complex.
  • 125 I-TGF-/31 was cross-linked to type I, type II and type III receptors on the mink lung epithelial cell line MvILu followed by immunoprecipitation with antisera against TBRI (ALK-5), TBRII and betaglycan, respectively.
  • type I, type II and type III TGF-3 receptors were recovered in the control experiment, none of the GDNF receptor components in differentiated RN33B cells could be immunoprecipitated by any of the tested antisera (not shown). These data confirmed that the GDNF receptor subunits expressed on these cells are novel proteins.
  • RNAse protection assay Ten micrograms of total RNA from the cell lines indicated wasa analyzed using a riboprobe complementary to 400 nucleotides of coding sequence from the kinase domain of the mouse c-ret mRNA. Although high expression was seen in MN-1 cells, no c-ret mRNA was detected in either the RN33B or L6 cells (Fig. 6). These results indicate that a signaling receptor for GDNF other than c-RET must be present in these cells.
  • GDNF binding proteins characterized in cell lines were able to form ligand-dependent signaling complexes.
  • Cell monolayers in 10 cm plates were incubated at 37°C in the presence of 50 ng/ml GDNF for the indicated time periods and immediately lysed with 1 ml of ice cold lysis buffer (as above) with the addition of 1 mM sodium othovanadate.
  • Whole cell lysates were fractionated by SDS-PAGE (10% polyacrylamide) and blotted to nitrocellulose filters.
  • GDNF receptor subunits Because of their distinct patterns of GDNF receptor subunits, intracellular signaling responses were initially characterized in the raphe nucleus cell line RN33B and in the motor neuron cell line MN-1. Changes in the pattern of tyrosine-phosphorylated proteins elicited by GDNF treatment of RN33B or MN-1 cells were investigated. Tyrosine phophorylation is a universal mechanism of regulation of intracellular signaling proteins that is stimulated by numerous cytokines and growth factors.
  • RN33B and MN-1 monolayers were exposed to a saturating concentration of GDNF (5 ng/ml) for different periods of tine, and total cell lysates were analysed for tyrosine phosphorylation by SDS/PAGE and Western blotting with an anti-phosphotyrosine monoclonal antibody.
  • Two proteins with mobilities corresponding to 42kD and 44kD, respectively, were phosphorylated on tyrosine within 5 minutes of GDNF treatment of RN33B cells (Fig. 7A).
  • a similar result was obtained in differentiated RN33B cells (not shown) of exposure to GDNF.
  • the 42kD and 44kD species would appear to be, respectively, p42 .k2 and p44 ,kl , two protein serine -threonine kinases members of the extracellular signal- regulated kinase (ERK, also termed microtubule-associated protein kinase) family (Boulton et al. , 65 Cell 663, 1991).
  • ERK extracellular signal- regulated kinase
  • ERK2 and ERKI protein blots which had been reacted with the anti- phosphotyrosine antibody were stripped and reprobed with a rabbit polyclonal antibody raised against recombinant ERK2 that recognizes both ERKI and ERK2 in protein blots.
  • Comparison of autoradiograms of blots probed with the anti- phosphotyrosine antibody and the anti-ERK2 antibody identified the p42 and p44 proteins as ERK2 and ERKI, respectively (Fig. 7a).
  • GDNF treatment of MN-1 cells appeared to only stimulate phosphorylation of ERK2, both ERKI and
  • ERK2 were present in MN-1 cell lysates (Fig. 7b).
  • GDNF treatment stimulated very rapid and transient tyrosine phosphorylation of ERKI and ERK2 in RN33B cells, but relatively slower and more sustained phosphorylation of ERK2 and MN-1 cells.
  • Activation of the ERK pathway has previously been shown to induce rapid and transient increase in transcription of immediate early genes, including the c-fos protooncogene (Gille et al. , 358 Nature 414, 1992).
  • the ability of GDNF to induce c-fos mRNA in differentiated raphe nucleus RN33B cells and in motor neuron MN-1 cells was investigated.
  • RNA extracted as previously described Twenty micrograms of total RNA was fractionated on 1 % agarose gels containing 0.7% formaldehyde and transferred to Hybond-C membranes (Amersham, UK). Northern blots were hybridized with an a- 32 P-dCTP labeled rat c-fos gene fragment (Curran et al. , 2 Oncogene 79, 1987), washed at high stringency and visualized by autoradiography on x-ray films.
  • RNA upregulation induced by GDNF treatment was very rapid and transient in RN33B cells, but somewhat slower in MN-1 cells.
  • GDNF may be a survival factor for differentiated raphe nucleus neurons.
  • Survival assays were performed as previously described (Eaton et al., 1995, supra). Briefly, 10 s RN cells were seeded to collagen fibronectin coated 8-welI glass slides and incubated at 33°C (growth permissive temperature) until 75-90% confluent. The slides were then shifted to 39° (non-permissive temperature) and serum containing medium was replaced by B16 defined medium (Brewer and Cotman, 494 Brain
  • Hoechst dye 33342 Hoechst dye 33342
  • RN46A cells were cultured at the non-permissive temperamre in defined medium in the presence of increasing concentrations of GDNF.
  • surviving cells were counted and compared with cultures established in the absence of GDNF.
  • a 3-fold increase in the number of surviving cells was observed in culmres grown in the presence of GDNF (Fig. 9).
  • Viable cells from the Sp2/0 murine cell line were adjusted to 2xlO 5 cells / ml with complete DMEM (10% fetal calf serum, 1 % L-glutamine, 100 U/ml penicillin and 100 ug/ streptomycin sulphate).
  • Cells from a non-immunised mouse were obtained from the peritoneal cavity by injection of 0.34M sucrose solution. The cells were resuspended in complete DMEM containing: hypoxanthine lOO ⁇ M; aminopterin 0.4M; and thymidine 16 ⁇ M, (HAT medium), to lxlO 5 cells /ml. 100 ⁇ l of the cell suspension was added to the 60 inner wells of 96 well plates and incubated overnight at 37°C in an atmosphere of 5% C0 2 in air. These cells were the source of growth factors. b) Fusion
  • Spleen cells from the mouse exhibiting the highest serum titer were homogenized in 10 ml DMEM removing surface fat and other adhering tissue in a sterile hood.
  • 4.2xl0 7 Sp2/0 cells were fused with 8.4 x IO 7 spleen cells in a solution of melted PEG (3000-3700, Hybri-Max, Sigma).
  • the cells were then grown in HAT medium at 37° C in an atmosphere of 5% CO 2 in air. After one week of culmre, the wells were inspected. When hybrids cells covered 10 to 50% of the surface area of the well, the culmre supernatants were assayed for antibody by ELISA.
  • PBS-T 0.05M phosphate buffered saline, pH 7.2, containing 0.05% Tween
  • PBS-T 0.05M phosphate buffered saline, pH 7.2, containing 0.05% Tween
  • Nonspecific binding was blocked with PBS-T containing 3 % non-fatty milk and 1 % goat normal serum.
  • Supernatant samples were incubated 4 hours at room temperature.
  • Peroxidase goat anti-mouse antibody was used and the substrate was o-phenylenediamine dihydrochloride (OPD). Plates were read at 492 nm in an
  • Negative controls included completed medium and normal mouse serum.
  • hybrids were grown in HAT medium up to two weeks after fusion. Cells were subsequently grown in HT medium until the completion of two cloning procedures, using Lhe limiting dilution method. After each step (when cells reached 10 to 50% confluence), assays for specific antibody in supernatants were done by ELISA. Upon recloning, 5 positive hybridoma clones were chosen and the cells were maintained in complete DMEM for 30 days.
  • Monoclonal antibodies from culmre supernatants were purified by Protein G Sepharose fast flow (Pharmacia, Biotech) according to manufacmrer's instructions.
  • Culture supernatants were concentrated and filtered through a 0.45 ⁇ m membrane (Schleicheer and, Schull, Germany) and then pumped overnight through the column previously equilibrated with 2OmM sodium phosphate, pH 7.0. Ig was eluted with 0.05M glycine buffer.
  • MN-1 cell monolayers were exposed to increasing concentrations of GDNF in serum-free medium and assayed 3 days later for cell survival and growth by measurement of acid phosphatase activity (Clontech).
  • GDNF was produced and purified from baculovirus infected insect cells as previously described (Trupp et al. , supra).
  • GDNF treatment of serum deprived-MN-1 monolayers increased cell number in a dose-dependent manner (Fig. 10a).
  • the biological response of MN-1 cells correlated with biochemical and transcriptional responses to GDNF treatment.
  • MN-1 cell monolayers were exposed to 50 ng/ml GDNF for increasing periods of time, cell lysates were fractionated by SDS/PAGE and Western blots probed with an anti-phosphotyrosine antibody (UBI).
  • UBI anti-phosphotyrosine antibody
  • the 42K species would appear to be p42 erk2 , a serine- threonine kinase member of the extracellular signal-regulated kinase (ERK) family .
  • ERK extracellular signal-regulated kinase
  • the identity of this protein as ERK2 was confirmed after immunoprecipitation with an antiERK2 polyclonal antiserum followed by analysis of tyrosine phosphorylation (Fig. 10c). Lysates of GDNF-stimulated MN-1 cells were immunoprecipitated with an anti-ERK2 antiserum (Santa Cruz) that also recognizes ERKI followed by antiphosphotyrosine Western blotting.
  • Example 9 The product of the c-ret proto-oncogene as a signal transducing receptor for
  • GDNF receptor complexes from MN-1 cells could be recovered by immunoprecipitation with anti-GDNF antibodies or by binding to lectin-Sepharose beads (Fig. lla).
  • the GDNF binding protein in this complex could be a receptor tyrosine kinase.
  • the product of the c-ret proto-oncogene is highly expressed in primary motor neurons (Pachnis et al. , supra, and Tsuzuki, T. , et al. Oncogene 10, 191-8 (1995) and is of similar molecular weight as the major GDNF receptor component detected in MN-1 cells (Takahashi, M. , et al. Oncogene 3, 571-578 (1988). We tested whether this species represented a C-RET-GDNF cross-linked complex by immunoprecipitation with anti-c-RET antibodies.
  • 125 I-GDNF was cross-linked to MN-1 cells using EDAC and receptor complexes were precipitated with antibodies against GDNF (Trupp et al. , supra), lectin Sepharose beads (Formica), anti-phosphotyrosine antibodies (UBI), anti-c-
  • RET antibodies (Santa Cruz) and control antibodies from non- immune rabbits.
  • An antipeptide c-RET rabbit antiserum readily immunoprecipitated the major 180kD ligand-receptor complex in MN-1 cells (Fig. lla), while a number of unrelated monoclonal and polyclonal antibodies used as controls failed to immunoprecipitate this complex (Fig. l l a and data not shown).
  • MN-1 cell monolayers were exposed to GDNF at different concentrations or for different periods of time and cell lysates were immunoprecipitated with anti-c-RET antibodies and analyzed by SDS/PAGE and
  • GDNF (Fig. lib), which is similar to the response of both serum deprived MN-1 cells (Fig. 10a) and embryonic sympathetic neurons (Trupp et al. , supra) to GDNF.
  • Fig. 10a serum deprived MN-1 cells
  • Fig. 10a embryonic sympathetic neurons
  • c-RET receptor may be an important component in the signal transduction mechanism of GDNF.
  • Example 10 c-ret transfection reconstitutes GDNF binding and biological activities to GDNF
  • GDNF-labeled receptor complexes of approximately 180K were detected in both MEN2a-ret and c-ret transfected 3T3 fibroblasts, but not in untransfected cells (Fig. 12a). The labeling could be displaced by excess cold GDNF, indicating that it represented specific
  • c-ret product may mediate the neurotrophic effects of GDNF in the brain by examining the expression of c-ret in different regions of the rat central nervous system.
  • a rat c-ret riboprobe was generated using as template a cDNA fragment obtained by PCR wi primers based on sequences U22513 and U22514 (Genbank accession numbers). High levels of c-ret mRNA were found in MN-1 cells and in rat spinal cord (data not shown). High c-ret mRNA expression was also found in the adult pons, medulla, locus coeruleus and hypomalamus (Fig.
  • c-ret mRNA was expressed at barely detectable levels in striatum, hippocampus and cerebral cortex (Fig. 13a).
  • Fig. 13a In the ventral mesencephalon, containing the cell bodies of GDNF-responsive dopaminergic neurons, c-ret mRNA levels increased progressively during post-natal development (Fig. 13b).
  • a peak of expression was detected between post natal day 6 (P6) and P8, at which time axons of dopaminergic neurons of the substantia nigra begin innervation of the striatum, and coincident with an increase in GDNF mRNA expression in this target region (Fig. 13b).
  • glyceraldehyde-3-P dehydrogenase (GAPDH) riboprobe was included in the RPA, and values of relative mRNA expression, obtained after densitometric scanning of gel autoradiograms, were normalised using the GAPDH signal of each RNA sample.
  • GAPDH glyceraldehyde-3-P dehydrogenase
  • c-RET protein was detected using a hamster monoclonal anti-mouse c-RET antibody which also recognises rat c-RET (Lo, supra) followed by fluoresce in-conjugated rabbit anti-hamster secondary antibodies (Southern Biotechnologies). In situ hybridization on sections through the adult substantia nigra revealed strong labelling over neurons throughout this strucmre (Figs. 14 a-b). In addition, cells positive for c-RET-like immunoreactivity (c-RET-LI) were found throughout the adult substantia nigra, with strong labelling over cell bodies (Fig. 14c).
  • GDNF-expressing fibroblast cells in 3 ⁇ l of medium were injected supranigrally at the following coordinates: 3,1 mm from interaural line, 2 mm lateral to midline, and 7 mm under the dural surface, with the incisor bar at -3.3 mm.
  • Lesion and grafting in the locus coeruleus were as previously described (Arenas et al. , Neuron 15, 1465-1473 (1995). The generation and characterisation of GDNF expressing fibroblasts have been described previously (Arenas et al. , supra).
  • GDNF rescues c-RET-positive dopaminergic and noradrenergic neurons
  • PC 12 cells and NB2/a cells were washed three times with serum free RPMI- 1640 or DMEM, respectively, plated on noncoated (NB2/a cells) or collagen-coated (PC 12 cells) dishes (5000-6000 cells per dish) in the presence or absence of 50 ng/ml of GDNF (Peprotech EC Ltd.) and the number of cells was microscopically counted after 48 hours.
  • PC12 and NB2/a cells were harvested (100,000 cells, five parallels), incubated with 10 ng/ml human 125 I-GDNF (iodinated by Chloramine T method, 100 ⁇ Ci/ ⁇ g) in the presence or absence of 50- fold unlabeled GDNF for 120-150 min on ice, the unbound factor was removed by centrifugation through 30% sucrose cushion, and the cell-associated radioactivity counted on 1271 RIAGAMMA counter (LKB Wallac).
  • GDNF nerve growth factor
  • PCI2 cells Upon treatment with (NGF), PCI2 cells also stop dividing and differentiate into sympathetic neuron-like cells with long neurites.
  • NGF nerve growth factor
  • PCI2 cells Upon treatment with (NGF), PCI2 cells also stop dividing and differentiate into sympathetic neuron-like cells with long neurites.
  • NGF nerve growth factor
  • GDNF is a survival- promoting factor for PCI2 cells, although less potent than NGF, but it does not induce differentiation of PC12 cells at the concentrations studied, presumably because of the differences in signal transduction of NGFactivated trkA receptors and GDNF receptors.
  • Human neuroblastoma NB2/a cells were plated in serum-free medium in the presence or absence of 50 ng/ml of GDNF and the number of cells was counted after 48 hr of culmre.
  • GDNF significantly increased the number of NB2/a cells (Fig. 15b).
  • GDNF exerts biological effects on rat PC 12 cells and human NB2/a cells, indicating that both cell lines express functional GDNF receptors.
  • PC12 cells SY5Y neuroblastoma cells and NB2/a cells where chemically cross-linked to 125 I-GDNF with EDC.
  • 3-5 10° cells or mechanically dissociated cells from 2 E20 rat kidneys were incubated with 10 ng/ml of 12S I-GDNF for I hour on ice and cross linked with 30 mM EDAC (Pierce) for 30 minutes on ice.
  • Detergent lysates were immunoprecipitated, the precipitates collected by Protein A- Sepharose, separated on 7% SDS-PAGE, and visualized by Phosphorimager SI (Molecular Dynamics).
  • the resulting complexes were immunoprecipitated with rabbit antibodies to GDNF, analyzed by SDS-PAGE and visualized by autoradiography. Embryonic kidney cells were also studied as the source of putative GDNF receptor (Suvanto, P. et. al., Eur. J. Neurosci, 8, 101-107 (1996); Sainio, K. et. al., Nature, (1996) submitted).
  • Cross-linked complexes of 170 and 190 kD were obtained from the extracts of PC 12 cells, SY5Y cells and NB2/a cells and a 190 kD complex from embryonic kidney extracts. (Fig. 16).
  • the molecular weights of the crosslinked proteins minus GDNF of approximately 25-30 kD substantially, if not exactly, correspond to the molecular weights of c-RET protooncogene, an o ⁇ han receptor tyrosine kinase (Takahashi, M. , Ritz, J. & Cooper, G. M. Cell, 42, 581-588, 1985; Takahashi, M. et.al., Oncogene, 3, 571-578 (1988)) (140 kD and 160 kD, representing differently glycosylated forms of c-RET., Tsuzuki, T. , Takahashi, M. , Asai, N., lwashita, T. , Matsuyama, M. & Asai, J. Oncogene, 10, 191-198 (1995).
  • the cross linked complexes were immunoprecipitated from the NB2/a cells with the cocktail of antibodies recognizing extracellular and intracellular part of the c-RET receptor. As shown in Fig. 17a (lane 1), the complexes of 170 kD and 190 kD were precipitated by anti-c-RET antibodies, which thus correspond to cross linked GDNFc-RET complexes. Binding of 125 I-GDNF to c-RET proteins was completely abolished by 500-fold excess of unlabeled GDNF (lane 2). No proteins were precipitated by monoclonal anti-neurofilament antibodies (lane 3) or by
  • COS cells were transiently transfected with c-ret cDNA or with empty plasmid by electroporation (Bio Rad) with - 30 % efficiency by fluorescence of cotransfected Red Shift Green Fluorescent Protein in PEF-BOS vector. 48 hours later, 10 x 10' transfected COS cells or 3-5 x 106 parental COS cells or NB2/a cells were treated With 125 I-GDNF, cross linked and analysed as specified in legends of Fig. 15 and Fig. 16.
  • c-RET protein by was examined Western blotting, c-rct-transfected COS cells (Fig. 18a) and NB2/a cells (not shown) expressed detectable amounts of the c-RET protein, whereas no c-RET protein was detected in mock-transfected (with PBK-CMV plasmid) COS cells (Fig. 18a).
  • PC 12 cells also express c-RET protein, albeit at considerably lower level than NB2/a cells or c-ret-transfected COS cells (not shown).
  • COS cells, transiently expressing mouse c- ret proto-oncogene were incubated with 125 I-GDNF. As shown in Fig. 17b, those cells bound GDNF, and binding of 125 I-GDNF can be competed with excess of unlabeled GDNF. In contrast, no significant binding-of GDNF was observed in mock-transfected COS cells.
  • Example 17 12S 1-GDNF binds to c-ret-positive enteric neurons
  • 125 I-GDNF was bound to developing rat tissue explants in situ.
  • In situ binding of human 125 I-GDNF (PeproTech. EC Ltd.), iodinated by Chloramine T Method, was carried out essentially as described (Partanen and Thesleff, 1987). Briefly, explants of E15 rat gut were incubated with 10 ng/ml of 125 I-GDNF in Eagle's minimal essential medium on the Nuclepore filter (Costar) for 90 min at room temperamre. 250-fold excess of unlabeled GDNF was applied as a competitor to control explants. After careful washing, the explants were fixed with
  • the gastrointestinal tract was chosen as it strongly expresses GDNF mRNA (Suvanto et al, 1996); Figure 19 a and b) and c-RET-positive neurons are absent in the gastrointestinal tract in c- -deficient mice (Schuchart et al. , 1994; Durbec et al. , 1996).
  • I25 I-GDNF binds to a group of cells within the muscle layer of embryonic day (E)15 rat gut ( Figure 19 c and d). This binding was specific as it was totally competed with 250-fold excess of unlabeled GDNF (Figure 19h).
  • the cells that bind GDNF were the enteric neurons of the myenteric plexus, as revealed by peripherin immunoreactivity (Figure 19f). Moreover, these neurons also expressed c-ret mRNA, as demonstrated by in situ hybridization (Figure 19e).
  • cRNAs in antisense and sense orientation were labeled with digoxigenin-UTP (Boehringer-Mannheim), hybridized to cryosections through El 5 rat gut and visualized with alkaline phosphatase-conjugated anti-digoxigenin antibodies according to manufacmrers instructions. In both cases, only background labeling was obtained with hybridization of corresponding probes in sense orientation ( Figure 19g).
  • Polyclonal anti-peripherin antibodies (Bio-Rad) were applied to cryosections of E15 rat gut at a dilution of 1 : 100 for 1 hr and visualized by FITC-conjugated secondary antibodies (Jackson).
  • GDNF specifically binds to c-RET-expressing enteric neurons of developing rat.
  • Affinity-crosslinking of GDNF to c-RET PC12 cells and NB2/a cells were washed three times with serum- free RPMI- 1640 or DMEM, respectively, plated on uncoated (NB2/a cells) or collagen- coated (PC 12 cells) dishes (5000-6000 cells per dish in triplicate) in the presence or absence of 50 ng/ml of GDNF (PeproTech EC Ltd.), and the number of cells microscopically counted after 48 h.
  • c-RET expression in transfected cells the shorter form (Takahashi el al. , 1988) of human wild-type c-ret cDNA was subcloned in pcDNA3 (Invitrogen). 3T3 fibroblasts were stably transfected with c-ret expression plasmid or with empty vector (mock-transfected cells) and positive cells lines selected with G418. Transient transfection of trkC 3T3 fibroblasts (Ip et al. (1993)
  • c-ret cDNA in pcDNA3 vector or with empty vector was performed by the lipofectin method (Gibco-BRL).
  • c-ret and mock- transfected cells (10.000 - 15.000 cells per well) in five parallels were treated with rat GDNF (Trupp et al. (1995) J. Cell. Biol. 130, 137-148) at indicated concentrations for five days.
  • NT-3 was used as positive control at 30 ng/ml.
  • Cell number was quantified by measurement of acid phosphatase activity using AbacusTM Cell Proliferation Kit (Clontech).
  • PeproTech EC Ltd. or rat GDNF from C. F. Ibanez) (Trupp et al. , 1995), iodinated by Chloramine T method, for 1 hour on ice.
  • 250-fold excess of unlabeled GDNF (PeproTech EC) or TGF- ⁇ T was applied to control sample.
  • 125 I-GDNF was then crosslinked to the cells with 30 mM of ethyl-dimethylarninopropyl carbodiimide (EDAC) (Pierce) for 30 minutes on ice.
  • EDAC ethyl-dimethylarninopropyl carbodiimide
  • Detergent lysates of the cells were immunoprecipitated with polyclonal anti-GDNF antibodies (Santa Cruz) or with the cocktail of monoclonal (kindly provided by Dr. D. Anderson, Lo and Anderson, 1995) and polyclonal (Santa Cruz) anti-c-RET antibodies to neurofilament proteins (a gift of Dr. I. Virtanen) were used as control antibodies.
  • the precipitates were collected by Protein A-Sepharose (Pharmacia) or by WGA-agarose (a gift from Dr. O Renkonen), separated on 7% SDS-PACE, and visualized with a Phosphorimager SI (Molecular DynanLics).
  • 125 I-GDNF was crosslinked to PC 12 cells, NB2/a cells and COS cells with ethyl-dimethyiaminopropyl carbodiimide (EDAC), and the complexes were precipitated with anti-GDNF antibodies.
  • EDAC ethyl-dimethyiaminopropyl carbodiimide
  • complexes with molecular weight of 190 kD and 170 kD were obtained from PC 12 and NB2/A cells, but not from COS cells.
  • EDAC-crosslink approach was also used to reveal GDNF-c-RET complexes from El 5 embryonic kidney cells, where c-ret mRNA is strongly expressed in Lhe tips of growing ureter branches.
  • GDNF specifically increases tyrosine phosphorylation of c-RET c-ret-3T3 cells and mock-3T3 cells were treated with GDNF and the proteins from these cells were immunoprecipitated with anti-c-RET antibodies. The precipitated proteins were then analyzed by Western blotting with anti- phosphotyrosine antibodies. 10 x 10° c-ret-3T3 cells were treated with different doses of GDNF (PeproTech LC Ltd. or from C. F.
  • Ibanez (Trupp et al , 1995) for 5 min, or with 50 ng/ml of GDNF for indicated times in serum-free Dulbecco's modified Eagle's medium containing 1 mM Na 3 VO 4 and then quickly washed with the same medium.
  • c-RET proteins were immunoprecipitated from detergent extracts, containing 1 mM Na 3 VO 4 by cocktail of monoclonal (Lo, L. and Anderson, D.J.
  • trkC-3T3 Mouse 3T3 fibroblast cell line expressing trkC (trkC-3T3) (Ip et al, 1993) were transiently transformed with c-ret expression plasmid. trkC-3T3 cells die within 2-3 days in serum-free medium in the absence of trkC ligand neurotrophin-3
  • GDNF-nonresponsive cells is sufficient to bring about the biological response to GDNF.
  • L6 myoblast cells were lysed with 1 % NP40 and cell lysates were fractionated by anionic exchange on a Q-Sepharose column. Fractions eluted at different ionic strength were dialyzed and assayed for binding to GDNF immoliblized on a chip in a Biacore device (Pharmacia). A distinct binding component was detected in a fraction of L6 cell lystaes (Figure 23a). In the Figure, solid bars indicate total protein (as absorbance at 280 nm); hatched bars indicate GDNF finding (in resonance units). This fraction was not particularly rich in protein, indicating a substantial purification over the total protein mixmre. The equivalent fraction of a COS cell lysate did not show binding under the same conditions (data not shown).
  • Example 22 A novel GDNF-binding protein in brain
  • c-RET might not bind GDNF directly, but might first bind to another nonsignalling receptor that thereafter presents the ligand to c-RET, a signaling receptor.
  • a 50 kD GDNF-binding protein is a good candidate for the putative presenting receptor.
  • 3T3 fibroblasts can be made dependent on a given exogenous growth factor provided appropriate receptors are expressed on the cell surface.
  • An expression library can be made using RN33B cDNA, which can then be transfected into 3T3 fibroblasts by procedures well known in the art (Maniatis et al. , supra). Stable transfectants can be selected in serum-free media supplemented with GDNF. Fibroblast clones that express signaling GDNF receptors will selectively grow in the presence of GDNF in serum-free media. The selection step may allow detection of even very reare clones due to their differential growth advantage. Further analysis of the recovered clones in media with or without GDNF would help to distinguish GDNF-dependent from GDNF-independent survival of clones.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • Biophysics (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Hematology (AREA)
  • Food Science & Technology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Compounds Of Unknown Constitution (AREA)
  • Medicines Containing Plant Substances (AREA)

Abstract

L'invention concerne des récepteurs du factor neurotrophique dérivé de lignées de cellules gliales (GDNF), leur expression cellulaire, leur isolement et leur caractérisation biochimique. Le récepteur c-RET est présenté comme un récepteur de GDNF. Des récepteurs nouveaux supplémentaires sont décrits ainsi que la préparation d'anticorps monoclonaux dirigés contre GDNF.
EP96942748A 1995-11-13 1996-11-13 Recepteurs du facteur neurotrophique derive de lignees de cellules gliales Withdrawn EP0879247A4 (fr)

Applications Claiming Priority (11)

Application Number Priority Date Filing Date Title
US661995P 1995-11-13 1995-11-13
US6619P 1995-11-13
US1576796P 1996-04-16 1996-04-16
US15767P 1996-04-16
US2063896P 1996-06-27 1996-06-27
US2196596P 1996-06-27 1996-06-27
US2063996P 1996-06-27 1996-06-27
US21965P 1996-06-27
US20638P 1996-06-27
PCT/US1996/018197 WO1997018240A1 (fr) 1995-11-13 1996-11-13 Recepteurs du facteur neurotrophique derive de lignees de cellules gliales
US20639P 2008-01-11

Publications (2)

Publication Number Publication Date
EP0879247A1 true EP0879247A1 (fr) 1998-11-25
EP0879247A4 EP0879247A4 (fr) 2002-01-30

Family

ID=27533181

Family Applications (1)

Application Number Title Priority Date Filing Date
EP96942748A Withdrawn EP0879247A4 (fr) 1995-11-13 1996-11-13 Recepteurs du facteur neurotrophique derive de lignees de cellules gliales

Country Status (8)

Country Link
EP (1) EP0879247A4 (fr)
JP (1) JP2000501924A (fr)
KR (1) KR19990067508A (fr)
AU (1) AU718979B2 (fr)
CA (1) CA2237641A1 (fr)
NO (1) NO982141L (fr)
NZ (1) NZ324511A (fr)
WO (1) WO1997018240A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10975154B2 (en) 2016-03-31 2021-04-13 Ngm Biopharmaceuticals, Inc. Binding proteins and methods of use thereof

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2270465T3 (es) * 1996-05-08 2007-04-01 Biogen Idec Ma Inc. Ligando 3 ret para estimular el crecimiento neural y renal.
US6677135B1 (en) 1996-05-08 2004-01-13 Biogen, Inc. Ret ligand (RetL) for stimulating neutral and renal growth
WO2000020867A1 (fr) * 1998-10-01 2000-04-13 Alexey Vladimirovich Titievsky Nouvelle voie de signalisation independante de ret pour gdnf
US6905817B1 (en) 1998-10-01 2005-06-14 Licestia Ltd. Ret-independent signaling pathway for GDNF
FI20000403A0 (fi) * 2000-02-22 2000-02-22 Hannu Sariola GDNF perhesukuisten yhdisteiden käyttö kivessyövän hoitoon tarkoitettujen tuotteiden valmistamiseksi

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
DURBEC P ET AL: "GDNF SIGNALLING THROUGH THE Ret RECEPTOR TYROSINE KINASE" NATURE, MACMILLAN JOURNALS LTD. LONDON, GB, vol. 381, 27 June 1996 (1996-06-27), pages 789-793, XP002092160 ISSN: 0028-0836 *
IBANEZ CARLOS F: "Biochemical characterisation of GDNF receptors and downstream responses." INTERNATIONAL JOURNAL OF DEVELOPMENTAL NEUROSCIENCE, vol. 14, no. SUPPL. 1, 1996, page 76 XP001041850 11th Biennial Meeting of the International Society for Developmental Neuroscience;Tampere, Finland; July 30-August 3, 1996 ISSN: 0736-5748 *
See also references of WO9718240A1 *
TREANO J J S ET AL: "CHARACTERIZATION OF A MULTICOMPONENT RECEPTOR FOR GDNF" NATURE, MACMILLAN JOURNALS LTD. LONDON, GB, vol. 382, no. 6586, 4 July 1996 (1996-07-04), pages 80-83, XP002047616 ISSN: 0028-0836 *
TRUPP M ET AL: "Characterization of GDNF receptors on primary neurons and cell lines." SOCIETY FOR NEUROSCIENCE ABSTRACTS, vol. 21, no. 1-3, 1995, page 1302 XP001041846 25th Annual Meeting of the Society for Neuroscience;San Diego, California, USA; November 11-16, 1995 ISSN: 0190-5295 *
TRUPP M ET AL: "FUNCTIONAL RECEPTOR FOR GDNF ENCODED BY THE C-RET PROTO-ONCOGENE" NATURE, MACMILLAN JOURNALS LTD. LONDON, GB, vol. 381, 27 June 1996 (1996-06-27), pages 785-789, XP002071817 ISSN: 0028-0836 *
TRUPP M ET AL: "PERIPHERAL EXPRESSION AND BIOLOGICAL ACTIVITIES OF GDNF, A NEW NEUROTROPHIC FACTOR FOR AVIAN AND MAMMALIAN PERIPHERAL NEURONS" THE JOURNAL OF CELL BIOLOGY, ROCKEFELLER UNIVERSITY PRESS, US, vol. 130, no. 1, 1 July 1995 (1995-07-01), pages 137-148, XP002047358 ISSN: 0021-9525 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10975154B2 (en) 2016-03-31 2021-04-13 Ngm Biopharmaceuticals, Inc. Binding proteins and methods of use thereof

Also Published As

Publication number Publication date
NZ324511A (en) 1999-09-29
KR19990067508A (ko) 1999-08-25
NO982141D0 (no) 1998-05-12
EP0879247A4 (fr) 2002-01-30
AU718979B2 (en) 2000-05-04
NO982141L (no) 1998-07-13
AU1158897A (en) 1997-06-05
CA2237641A1 (fr) 1997-05-22
WO1997018240A1 (fr) 1997-05-22
JP2000501924A (ja) 2000-02-22

Similar Documents

Publication Publication Date Title
US10717775B2 (en) Methods for cardiomyocyte survival, proliferation or differentiation by NRG-2
Trupp et al. Multiple GPI-anchored receptors control GDNF-dependent and independent activation of the c-Ret receptor tyrosine kinase
Lin et al. Expression cloning of the TGF-β type II receptor, a functional transmembrane serine/threonine kinase
Tsuchida et al. Molecular cloning of a novel type I receptor serine/threonine kinase for the TGFβ superfamily from rat brain
AU2001274947A1 (en) NRG-2 nucleic acid molecules, polypeptides, and diagnostic and therapeutic methods
IE20010175A1 (en) HEREGULINS (HRGs), BINDING PROTEINS OF P185erb2
AU718979B2 (en) Glial cell line-derived neurotrophic factor receptors
US6696259B1 (en) Assays using glial cell line-derived neurotrophic factor receptors
AU741208B2 (en) Glial cell line-derived neurotrophic factor receptors
MXPA98003767A (en) Receivers of the neurotrophic factor derived from cell line gl
JP2003510029A (ja) Egfh2遺伝子および遺伝子産物
JPH05273208A (ja) 神経刺激性物質の検出法
US6441133B1 (en) Thyrotropin-releasing hormone receptor 2(TRHR-2)
MXPA99010773A (en) Glial cell line-derived neurotrophic factor receptors
CA2887616A1 (fr) Molecules d'acide nucleique et polypeptides nrg-2, et methodes diagnostiques et therapeutiques associees

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19980609

AK Designated contracting states

Kind code of ref document: A1

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

A4 Supplementary search report drawn up and despatched

Effective date: 20011219

AK Designated contracting states

Kind code of ref document: A4

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

17Q First examination report despatched

Effective date: 20030918

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1016992

Country of ref document: HK

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

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

18D Application deemed to be withdrawn

Effective date: 20060221