Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Cell Tissue Res ; 382(1): 173-183, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32725425

ABSTRACT

The last decade has been a frustrating time for investigators who had envisioned major advances in the treatment of Parkinson's disease using neurotrophic factors. The first trials of glial cell line-derived neurotrophic factor for treating Parkinson's disease were very promising. Later blinded control trials were disappointing, not reaching the predetermined outcomes for improvement in motor function. Consideration of the problems in the studies as well as the biology of the neurotrophins used can potentially lead to more effective therapies. Parkinson's disease presents a multitude of opportunities for the cell biologist wanting to understand its pathology and to find possible new avenues for treatment.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/genetics , Animals , Humans , Ligands , Signal Transduction
2.
Neurobiol Dis ; 96: 335-345, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27425888

ABSTRACT

In Parkinson's disease midbrain dopaminergic neurons degenerate and die. Oral medications and deep brain stimulation can relieve the initial symptoms, but the disease continues to progress. Growth factors that might support the survival, enhance the activity, or even regenerate degenerating dopamine neurons have been tried with mixed results in patients. As growth factors do not pass the blood-brain barrier, they have to be delivered intracranially. Therefore their efficient diffusion in brain tissue is of crucial importance. To improve the diffusion of the growth factor neurturin (NRTN), we modified its capacity to attach to heparan sulfates in the extracellular matrix. We present four new, biologically fully active variants with reduced heparin binding. Two of these variants are more stable than WT NRTN in vitro and diffuse better in rat brains. We also show that one of the NRTN variants diffuses better than its close homolog GDNF in monkey brains. The variant with the highest stability and widest diffusion regenerates dopamine fibers and improves the conditions of rats in a 6-hydroxydopamine model of Parkinson's disease more potently than GDNF, which previously showed modest efficacy in clinical trials. The new NRTN variants may help solve the major problem of inadequate distribution of NRTN in human brain tissue.


Subject(s)
Drug Design , Genetic Variation/genetics , Neurturin/chemistry , Neurturin/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Amphetamine/pharmacology , Animals , CHO Cells , Cricetulus , Disease Models, Animal , Humans , Macaca fascicularis , Male , Models, Molecular , Neurturin/genetics , Oxidopamine/toxicity , Parkinson Disease/complications , Parkinson Disease/etiology , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Rats , Rats, Wistar , Stereotyped Behavior/drug effects , Sympatholytics/toxicity , Tyrosine 3-Monooxygenase/metabolism
4.
Biochem J ; 387(Pt 3): 817-24, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15610063

ABSTRACT

The GDNF (glial cell line-derived neurotrophic factor)-binding receptor GFRalpha1 (GDNF family receptor alpha1) is attached to the membrane by a GPI (glycosylphosphatidylinositol) anchor and consists of three cysteine-rich domains. The region corresponding to the second and third domains has been shown previously to participate in ligand binding, and to interact with the transmembrane tyrosine kinase receptor RET. No function has so far been found for the N-terminal, first domain (D1). Here we show that the GPI-anchored full-length receptor binds 125I-GDNF two times more tightly than does a GPI-anchored truncated receptor lacking D1. Scintillation proximity assays with purified receptor proteins also show that the GDNF-binding capacity of the soluble full-length GFRalpha1 is two times higher than the GDNF-binding capacity of the soluble D1-truncated GFRalpha1. As RET stabilizes the binding of GDNF equally well to the full-length and truncated receptors, D1 seems not to be involved in the interaction between GFRalpha1 and RET. Moreover, soluble full-length GFRalpha1 mediates GDNF-promoted neurite outgrowth in PC6-3 cells more efficiently than the soluble truncated GFRalpha1 protein. At low concentrations, the soluble fulllength receptor mediates the phosphorylation of RET more efficiently than the soluble truncated receptor. However, when the receptors are overexpressed on the cell surface as GPI-anchored proteins, or added to the growth medium at high concentrations as soluble proteins, full-length and truncated GFRalpha1 are indistinguishable in GDNF-dependent RET-phosphorylation assays. High levels of the receptors can thus mask a slightly impaired function in the phosphorylation assay. Based on assays with both GPI-anchored and soluble receptors, we therefore conclude that D1 contributes to the optimal function of GFRalpha1 by stabilizing the interaction between GFRalpha1 and GDNF.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor Receptors/chemistry , Glial Cell Line-Derived Neurotrophic Factor/chemistry , Amino Acid Sequence , Animals , Cell Line , Cysteine/chemistry , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Mutation , Phosphorylation , Protein Binding , Protein Structure, Tertiary
5.
FEBS Lett ; 569(1-3): 267-71, 2004 Jul 02.
Article in English | MEDLINE | ID: mdl-15225646

ABSTRACT

Previously, it was shown that the recruitment of RET into lipid rafts by glial cell line-derived neurotrophic factor (GDNF)/GFRalpha1 is crucial for efficient signal transduction. Here, we show that the mouse GFRalpha4 is a functional, N-glycosylated, glycosylphosphatidylinositol (GPI)-anchored protein, which mediates persephin (PSPN)-induced phosphorylation of RET, but has an almost undetectable capacity to recruit RET into the 0.1% Triton X-100 insoluble membrane fraction. In spite of this, PSPN/mGFRalpha4 promotes neurite outgrowth in PC6-3 cells and survival of cerebellar granule neurons. As we show that also human PSPN/GFRalpha4 is unable to recruit RET into lipid rafts, we propose that the mammalian GFRalpha4 in this respect differs from GFRalpha1.


Subject(s)
Membrane Glycoproteins/metabolism , Membrane Microdomains/metabolism , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Neurites/physiology , Neurons/physiology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Cell Line , Cell Membrane/physiology , Cloning, Molecular , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Humans , Membrane Glycoproteins/genetics , Mice , Neurons/cytology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ret , Rats , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Nerve Growth Factor/genetics , Recombinant Proteins/metabolism , Transfection
6.
Ann Med ; 45(1): 66-73, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23305235

ABSTRACT

INTRODUCTION: Although glial cell line-derived neurotrophic factor (GDNF) has a strong clinical potential, little is known of how the posttranslational modifications of GDNF affect its biological activity and therapeutic potential. In mammalian cells GDNF is synthesized as a preproprotein. During secretion GDNF dimerizes, folds with -S-S- bonds, is modified by N-linked glycosylation, and undergoes proteolytic processing. After production in E. coli, unglycosylated GDNF is renaturated in vitro. Nevertheless, GDNF from E. coli was used in Parkinson's disease-related clinical trials. MATERIAL AND METHODS: Constructs encoding variants of human GDNF were generated and expressed in mammalian cells. The proteins were analysed by SDS-PAGE, Western blotting, RET-phosphorylation assays, and N-terminal sequencing. The stability of mammalian GDNF was compared to commercial GDNF produced in E. coli. RESULTS: Posttranslational processing of mammalian GDNF depends on the expression conditions. Two forms of GDNF with different N-termini are formed. GDNF without a prosequence is secreted and biologically active. GDNF is modified by N-linked glycosylation at one (Asn(49)) out of two consensus sites. N-linked glycosylation aids proteolytic processing of GDNF. Both glycosylated and unglycosylated GDNF from mammalian cells are more stable than GDNF from E. coli. DISCUSSION: Posttranslational modifications of GDNF influence its stability, which may be critical for its clinical use.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/metabolism , Protein Processing, Post-Translational , Animals , CHO Cells , Cells, Cultured , Cricetinae , Culture Media , Escherichia coli , Glial Cell Line-Derived Neurotrophic Factor/chemistry , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Glycosylation , HEK293 Cells , Humans , Polysaccharides/metabolism , Protein Precursors/metabolism , Protein Stability
7.
J Biol Chem ; 283(50): 35164-72, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-18845535

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF), a neuronal survival factor, binds its co-receptor GDNF family receptor alpha1 (GFR alpha 1) in a 2:2 ratio and signals through the receptor tyrosine kinase RET. We have solved the GDNF(2).GFR alpha 1(2) complex structure at 2.35 A resolution in the presence of a heparin mimic, sucrose octasulfate. The structure of our GDNF(2).GFR alpha 1(2) complex and the previously published artemin(2).GFR alpha 3(2) complex are unlike in three ways. First, we have experimentally identified residues that differ in the ligand-GFR alpha interface between the two structures, in particular ones that buttress the key conserved Arg(GFR alpha)-Glu(ligand)-Arg(GFR alpha) interaction. Second, the flexible GDNF ligand "finger" loops fit differently into the GFR alphas, which are rigid. Third, and we believe most importantly, the quaternary structure of the two tetramers is dissimilar, because the angle between the two GDNF monomers is different. This suggests that the RET-RET interaction differs in different ligand(2)-co-receptor(2)-RET(2) heterohexamer complexes. Consistent with this, we showed that GDNF(2).GFR alpha1(2) and artemin(2).GFR alpha 3(2) signal differently in a mitogen-activated protein kinase assay. Furthermore, we have shown by mutagenesis and enzyme-linked immunosorbent assays of RET phosphorylation that RET probably interacts with GFR alpha 1 residues Arg-190, Lys-194, Arg-197, Gln-198, Lys-202, Arg-257, Arg-259, Glu-323, and Asp-324 upon both domains 2 and 3. Interestingly, in our structure, sucrose octasulfate also binds to the Arg(190)-Lys(202) region in GFR alpha 1 domain 2. This may explain how GDNF.GFR alpha 1 can mediate cell adhesion and how heparin might inhibit GDNF signaling through RET.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor Receptors/chemistry , Glial Cell Line-Derived Neurotrophic Factor/chemistry , Heparin/chemistry , Animals , Binding Sites , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Humans , Ligands , MAP Kinase Signaling System , Molecular Conformation , Protein Binding , Protein Structure, Quaternary , Proto-Oncogene Proteins c-ret/chemistry , Rats , Sucrose/analogs & derivatives , Sucrose/chemistry , Thrombin/chemistry
8.
Ann Med ; 39(8): 572-80, 2007.
Article in English | MEDLINE | ID: mdl-17934909

ABSTRACT

RET (REarranged during Transfection) is a transmembrane receptor tyrosine kinase that is activated by a complex consisting of a soluble glial cell line-derived neurotrophic factor (GDNF) family ligand (GFL) and a glycosyl phosphatidylinositol-anchored co-receptor, GDNF family receptors alpha (GFRalpha). RET signalling is crucial for the development of the enteric nervous system. RET also regulates the development of sympathetic, parasympathetic, motor, and sensory neurons, and is necessary for the postnatal maintenance of dopaminergic neurons. The effect of GFLs on sensory, motor, and dopaminergic neurons has raised clinical interest towards these ligands. Outside the nervous system, RET is crucial for development of the kidney and plays a key role in spermatogenesis. Inactivating mutations in RET cause the Hirschsprung's disease characterized by megacolon aganglionosis. In contrast, activating mutations give rise to different types of cancer, multiple endocrine neoplasia type 2A and type 2B, familial medullary thyroid carcinoma, and papillary thyroid carcinoma. The multiple disease phenotypes correlate with differences in the molecular and cell biological functions of different oncogenic RET proteins. In this review we summarize how the different domains of the RET protein contribute to its normal function and how mutations in these domains affect the function of the receptor.


Subject(s)
Proto-Oncogene Proteins c-ret/physiology , Receptors, Nerve Growth Factor/physiology , Hirschsprung Disease/genetics , Humans , Multiple Endocrine Neoplasia/genetics , Mutation , Proto-Oncogene Proteins c-ret/genetics , Signal Transduction/physiology , Transcriptional Activation/physiology
9.
EMBO J ; 23(7): 1452-62, 2004 Apr 07.
Article in English | MEDLINE | ID: mdl-15044950

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) binds to the GDNF family co-receptor alpha1 (GFRalpha1) and activates RET receptor tyrosine kinase. GFRalpha1 has a putative domain structure of three homologous cysteine-rich domains, where domains 2 and 3 make up a central domain responsible for GDNF binding. We report here the 1.8 A crystal structure of GFRalpha1 domain 3 showing a new protein fold. It is an all-alpha five-helix bundle with five disulfide bridges. The structure was used to model the homologous domain 2, the other half of the GDNF-binding fragment, and to construct the first structural model of the GDNF-GFRalpha1 interaction. Using site-directed mutagenesis, we identified closely spaced residues, Phe213, Arg224, Arg225 and Ile229, comprising a putative GDNF-binding surface. Mutating each one of them had slightly different effects on GDNF binding and RET phosphorylation. In addition, the R217E mutant bound GDNF equally well in the presence and absence of RET. Arg217 may thus be involved in the allosteric properties of GFRalpha1 or in binding RET.


Subject(s)
Nerve Growth Factors/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Enzyme Activation , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding , Protein Folding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ret , Rats , Receptor Protein-Tyrosine Kinases/genetics , Sequence Alignment
10.
J Gen Virol ; 83(Pt 5): 1211-1221, 2002 May.
Article in English | MEDLINE | ID: mdl-11961277

ABSTRACT

Processing of the polyprotein encoded by Potato virus A (PVA; genus Potyvirus) was studied using expression of the complete PVA polyprotein or its mutants from recombinant baculoviruses in insect cells. The time-course of polyprotein processing by the main viral proteinase (NIaPro) was examined with the pulse-chase method. The sites at the P3/6K1, CI-6K2 and VPg/NIaPro junctions were processed slowly, in contrast to other proteolytic cleavage sites which were processed at a high rate. The CI-6K2 polyprotein was observed in the baculovirus system and in infected plant cells. In both cell types the majority of CI-6K2 was found in the membrane fraction, in contrast to fully processed CI. Deletion of the genomic region encoding the 6K1 protein prevented proper proteolytic separation of P3 from CI, but did not affect processing of VPg, NIaPro, NIb or CP from the polyprotein. The 6K2-encoding sequence could be removed without any detectable effect on polyprotein processing. However, deletion of either the 6K1 or 6K2 protein-encoding regions rendered PVA non-infectious. Mutations at the 6K2/VPg cleavage site reduced virus infectivity in plants, but had a less pronounced, albeit detectable, effect on proteolytic processing in the baculovirus system. The results of this study indicate that NIaPro catalyses proteolytic cleavages preferentially in cis, and that the 6K1/CI and NIb/CP sites can also be processed in trans. Both 6K peptides are indispensable for virus replication, and proteolytic separation of the 6K2 protein from the adjacent proteins by NIaPro is important for the rate of virus replication and movement.


Subject(s)
Endopeptidases/physiology , Plants/virology , Polyproteins/metabolism , Potyvirus/metabolism , Viral Proteins/metabolism , Animals , Baculoviridae/metabolism , Cells, Cultured , Spodoptera , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL