Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Eur J Neurosci ; 14(7): 1153-63, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11683907

ABSTRACT

Exogenous glial cell line-derived neurotrophic factor (GDNF) exhibits potent survival-promoting effects on dopaminergic neurons of the nigrostriatal pathway that is implicated in Parkinson's disease and also protects neurons in forebrain ischemia of animal models. However, a role for endogenous GDNF in brain function has not been established. Although mice homozygous for a targeted deletion of the GDNF gene have been generated, these mice die within hours of birth because of deficits in kidney morphogenesis, and, thus, the effect of the absence of GDNF on brain function could not be studied. Herein, we sought to determine whether adult mice, heterozygous for a GDNF mutation on two different genetic backgrounds, demonstrate alterations in the nigrostriatal dopaminergic system or in cognitive function. While both neurochemical and behavioural measures suggested that reduction of GDNF gene expression in the mutant mice does not alter the nigrostriatal dopaminergic system, it led to a significant and selective impairment of performance in the spatial version of the Morris water maze. A standard panel of blood chemistry tests and basic pathological analyses did not reveal alterations in the mutants that could account for the observed performance deficit. These results suggest that endogenous GDNF may not be critical for the development and functioning of the nigrostriatal dopaminergic system but it plays an important role in cognitive abilities.


Subject(s)
Brain/metabolism , Dopamine/metabolism , Heterozygote , Learning Disabilities/genetics , Maze Learning/physiology , Mutation/physiology , Nerve Growth Factors , Nerve Tissue Proteins/deficiency , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Brain/physiopathology , Gene Expression/genetics , Glial Cell Line-Derived Neurotrophic Factor , Learning Disabilities/metabolism , Learning Disabilities/physiopathology , Mice , Mice, Knockout , Motor Activity/genetics , Neostriatum/metabolism , Neostriatum/physiopathology , Nerve Tissue Proteins/genetics , Neural Pathways/metabolism , Neural Pathways/physiopathology , Organ Size/genetics , Substantia Nigra/metabolism , Substantia Nigra/physiopathology
2.
J Neurosci ; 21(4): 1283-91, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11160399

ABSTRACT

Developing motoneurons require trophic support from their target, the skeletal muscle. Despite a large number of neurotrophic molecules with survival-promoting activity for isolated embryonic motoneurons, those factors that are required for motoneuron survival during development are still not known. Cytokines of the ciliary neurotrophic factor (CNTF)-leukemia inhibitory factor (LIF) family have been shown to play a role in motoneuron (MN) survival. Importantly, in mice lacking the LIFRbeta or the CNTFRalpha there is a significant loss of MNs during embryonic development. Because genetic deletion of either (or both) CNTF or LIF fails, by contrast, to perturb MN survival before birth, it was concluded that another ligand exists that is functionally inactivated in the receptor deleted mice, resulting in MN loss during development. One possible candidate for this ligand is the CNTF-LIF family member cardiotrophin-1 (CT-1). CT-1 is highly expressed in embryonic skeletal muscle, secreted by myotubes, and promotes the survival of cultured embryonic mouse and rat MNs. Here we show that ct-1 deficiency causes increased motoneuron cell death in spinal cord and brainstem nuclei of mice during a period between embryonic day 14 and the first postnatal week. Interestingly, no further loss was detectable during the subsequent postnatal period, and nerve lesion in young adult ct-1-deficient mice did not result in significant additional loss of motoneurons, as had been previously observed in mice lacking both CNTF and LIF. CT-1 is the first bona fide muscle-derived neurotrophic factor to be identified that is required for the survival of subgroups of developing motoneurons.


Subject(s)
Cytokines/metabolism , Interleukin-6 , Motor Neurons/metabolism , Muscle, Skeletal/metabolism , Neurodegenerative Diseases/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Axotomy , Brain Stem/embryology , Brain Stem/metabolism , Brain Stem/pathology , Cell Death , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Chick Embryo , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/metabolism , Cytokine Receptor gp130 , Cytokines/deficiency , Cytokines/genetics , Cytokines/pharmacology , Dose-Response Relationship, Drug , Facial Nerve , Growth Inhibitors/genetics , Growth Inhibitors/metabolism , Leukemia Inhibitory Factor , Lymphokines/genetics , Lymphokines/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Motor Neurons/drug effects , Motor Neurons/pathology , Muscle, Skeletal/embryology , Muscle, Skeletal/innervation , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , RNA, Messenger/biosynthesis , Receptor, Ciliary Neurotrophic Factor/genetics , Receptor, Ciliary Neurotrophic Factor/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Spinal Cord/pathology
3.
Hybridoma ; 19(4): 303-15, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11001403

ABSTRACT

Neurturin (NTN) a structural and functional relative of glial cell line-derived neurotrophic factor, was originally identified based on its ability to support the survival of sympathetic neurons in culture. Similar to glial cell line-derived neurotrophic factor (GDNF), Neurturin has been shown to bind to a high affinity glycosylphosphatidylinositol (GPI)-linked receptor (GFRalpha2) and induce phosphorylation of the tyrosine kinase receptor Ret, resulting in the activation of the mitogen activated protein kinase (MAPK) signalling pathway. A panel of six novel murine monoclonal antibodies (MAbs) specific to human Neurturin has been developed and characterized. Four of the MAbs tested inhibit, to varying degrees, binding of NTN to the GPI-linked GFRalpha2 receptor. Three MAbs cross-react with the murine homolog. These antibodies have been shown to be useful reagents for Western blotting, immunohistochemistry, and also for the development of a sensitive, quantitative enzyme-linked immunosorbent assay (ELISA) for human NTN. Novel, specific MAbs with varying epitope specificities and blocking activity will be valuable tools for both the in vitro and in vivo characterization of NTN and its relationship to the GFRalpha2 and Ret receptors.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Nerve Growth Factors/immunology , Animals , Antibodies, Monoclonal/isolation & purification , Antibody Affinity/immunology , Binding, Competitive/immunology , Blotting, Western , Cell Survival/physiology , Cricetinae , Cross Reactions/immunology , Enzyme Inhibitors/immunology , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Glial Cell Line-Derived Neurotrophic Factor Receptors , Humans , Immunization , Immunohistochemistry , Mice , Mice, Inbred BALB C , Nerve Tissue Proteins/immunology , Neurites/physiology , Neuroblastoma/immunology , Neuroblastoma/pathology , Neuroblastoma/ultrastructure , Neurturin , Rats , Substantia Nigra/cytology , Substantia Nigra/immunology , Superior Cervical Ganglion/immunology
5.
Neuron ; 25(1): 239-52, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10707987

ABSTRACT

alpha-Synuclein (alpha-Syn) is a 14 kDa protein of unknown function that has been implicated in the pathophysiology of Parkinson's disease (PD). Here, we show that alpha-Syn-/- mice are viable and fertile, exhibit intact brain architecture, and possess a normal complement of dopaminergic cell bodies, fibers, and synapses. Nigrostriatal terminals of alpha-Syn-/- mice display a standard pattern of dopamine (DA) discharge and reuptake in response to simple electrical stimulation. However, they exhibit an increased release with paired stimuli that can be mimicked by elevated Ca2+. Concurrent with the altered DA release, alpha-Syn-/- mice display a reduction in striatal DA and an attenuation of DA-dependent locomotor response to amphetamine. These findings support the hypothesis that alpha-Syn is an essential presynaptic, activity-dependent negative regulator of DA neurotransmission.


Subject(s)
Corpus Striatum/physiopathology , Dopamine/metabolism , Nerve Tissue Proteins/genetics , Substantia Nigra/physiopathology , Amphetamine/pharmacology , Animals , Autoreceptors/physiology , Calbindins , Calcium/pharmacokinetics , Corpus Striatum/chemistry , Corpus Striatum/cytology , Dopamine/analysis , Dopamine Agents/pharmacology , Female , Gene Expression/physiology , Glutamic Acid/physiology , Hippocampus/chemistry , Hippocampus/cytology , Hippocampus/physiology , Locomotion/drug effects , Locomotion/genetics , Long-Term Potentiation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/genetics , Neurons/chemistry , Neurons/physiology , Presynaptic Terminals/chemistry , Presynaptic Terminals/metabolism , S100 Calcium Binding Protein G/analysis , Substantia Nigra/chemistry , Substantia Nigra/cytology , Synaptic Transmission/physiology , Synucleins , alpha-Synuclein , rab3A GTP-Binding Protein/genetics
6.
J Neurosci ; 20(1): 427-37, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10627618

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) has potent trophic effects on adult sensory neurons after nerve injury and is one of a family of proteins that includes neurturin, persephin, and artemin. Sensitivity to these factors is conferred by a receptor complex consisting of a ligand binding domain (GFRalpha1-GFRalpha4) and a signal transducing domain RET. We have investigated the normal expression of GDNF family receptor components within sensory neurons and the response to nerve injury. In normal rats, RET and GFRalpha1 were expressed in a subpopulation of both small- and large-diameter afferents projecting through the sciatic nerve [60 and 40% of FluoroGold (FG)-labeled cells, respectively]. GFRalpha2 and GFRalpha3 were both expressed principally within small-diameter DRG cells (30 and 40% of FG-labeled cells, respectively). Two weeks after sciatic axotomy, the expression of GFRalpha2 was markedly reduced (to 12% of sciatic afferents). In contrast, the proportion of sciatic afferents that expressed GFRalpha1 increased (to 66% of sciatic afferents) so that virtually all large-diameter afferents expressed this receptor component, and the expression of GFRalpha3 also increased (to 66% of sciatic afferents) so that almost all of the small-diameter afferents expressed this receptor component after axotomy. There was little change in RET expression. The changes in the proportions of DRG cells expressing different receptor components were mirrored by alterations in the total RNA levels within the DRG. The changes in GFRalpha1 and GFRalpha2 expression after axotomy could be largely reversed by treatment with GDNF.


Subject(s)
Drosophila Proteins , Nerve Growth Factors , Nerve Tissue Proteins/pharmacology , Posterior Horn Cells/chemistry , Posterior Horn Cells/physiology , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Animals , Axotomy , Gene Expression/physiology , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , In Situ Hybridization , Ligation , Male , Nerve Regeneration/physiology , Neurofilament Proteins/analysis , Neurofilament Proteins/metabolism , Oligonucleotide Probes , Phosphorylation , Posterior Horn Cells/drug effects , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins c-ret , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptor Protein-Tyrosine Kinases/analysis , Sciatic Nerve/chemistry , Sciatic Nerve/physiology , Up-Regulation/genetics
7.
J Cell Sci ; 112 ( Pt 23): 4437-48, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10564661

ABSTRACT

Drosophila Suppressor of fused (Su(fu)) encodes a novel 468-amino-acid cytoplasmic protein which, by genetic analysis, functions as a negative regulator of the Hedgehog segment polarity pathway. Here we describe the primary structure, tissue distribution, biochemical and functional analyses of a human Su(fu) (hSu(fu)). Two alternatively spliced isoforms of hSu(fu) were identified, predicting proteins of 433 and 484 amino acids, with a calculated molecular mass of 48 and 54 kDa, respectively. The two proteins differ only by the inclusion or exclusion of a 52-amino-acid extension at the carboxy terminus. Both isoforms were expressed in multiple embryonic and adult tissues, and exhibited a developmental profile consistent with a role in Hedgehog signaling. The hSu(fu) contains a high-scoring PEST-domain, and exhibits an overall 37% sequence identity (63% similarity) with the Drosophila protein and 97% sequence identity with the mouse Su(fu). The hSu(fu) locus mapped to chromosome 10q24-q25, a region which is deleted in glioblastomas, prostate cancer, malignant melanoma and endometrial cancer. HSu(fu) was found to repress activity of the zinc-finger transcription factor Gli, which mediates Hedgehog signaling in vertebrates, and to physically interact with Gli, Gli2 and Gli3 as well as with Slimb, an F-box containing protein which, in the fly, suppresses the Hedgehog response, in part by stimulating the degradation of the fly Gli homologue. Coexpression of Slimb with Su(fu) potentiated the Su(fu)-mediated repression of Gli. Taken together, our data provide biochemical and functional evidence for the hypothesis that Su(fu) is a key negative regulator in the vertebrate Hedgehog signaling pathway. The data further suggest that Su(fu) can act by binding to Gli and inhibiting Gli-mediated transactivation as well as by serving as an adaptor protein, which links Gli to the Slimb-dependent proteasomal degradation pathway.


Subject(s)
Chromosomes, Human, Pair 10 , Drosophila Proteins , Gene Expression Regulation, Developmental , Oncogene Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/metabolism , Adult , Alternative Splicing , Amino Acid Sequence , Animals , Cell Line , Chromosome Mapping , Cloning, Molecular , Drosophila , Female , Fetus , Gene Expression Regulation , Humans , Luciferases/genetics , Male , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Fusion Proteins/biosynthesis , Repressor Proteins/chemistry , Sequence Alignment , Sequence Homology, Amino Acid , Trans-Activators , Zinc Finger Protein GLI1 , Zinc Fingers
8.
J Neurosci ; 19(21): 9538-49, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10531456

ABSTRACT

EphA family receptor tyrosine kinases and their ephrin-A ligands are involved in patterning axonal connections during brain development, but until now a role for these molecules in the mature brain had not been elucidated. Here, we show that both the EphA5 receptor and its ephrin-A ligands (2 and 5) are expressed in the adult mouse hippocampus, and the EphA5 protein is present in a phosphorylated form. Because there are no pharmacological agents available for EphA receptors, we designed recombinant immunoadhesins that specifically bind to the receptor binding site of the ephrin-A ligand (antagonist) or the ligand binding site of the EphA receptor (agonist) and thus target EphA function. We demonstrate that intrahippocampal infusion of an EphA antagonist immunoadhesin leads to impaired performance in two behavioral paradigms, T-maze spontaneous alternation and context-dependent fear conditioning, sensitive to hippocampal function, whereas activation of EphA by infusion of an agonist immunoadhesin results in enhanced performance on these tasks. Because the two behavioral tasks have different motivational, perceptual, and motor requirements, we infer the changes were not caused by these performance factors but rather to cognitive alterations. We also find bidirectional changes in gene expression and in electrophysiological measures of synaptic efficacy that correlate with the behavioral results. Thus, EphA receptors and their ligands are implicated as mediators of plasticity in the adult mammalian brain.


Subject(s)
Conditioning, Operant/physiology , Hippocampus/physiology , Learning/physiology , Membrane Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Transcription Factors/physiology , Animals , Binding Sites , Ephrin-A2 , Ephrin-A5 , Fear , Immunoglobulin G/pharmacology , In Situ Hybridization , In Vitro Techniques , Infusions, Parenteral , Ligands , Male , Maze Learning/physiology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Phosphorylation , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/immunology , Receptor, EphA5 , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/immunology
9.
Proc Natl Acad Sci U S A ; 95(23): 13630-4, 1998 Nov 10.
Article in English | MEDLINE | ID: mdl-9811851

ABSTRACT

The multitransmembrane protein Patched (PTCH) is the receptor for Sonic Hedgehog (Shh), a secreted molecule implicated in the formation of embryonic structures and in tumorigenesis. Current models suggest that binding of Shh to PTCH prevents the normal inhibition of the seven-transmembrane-protein Smoothened (SMO) by PTCH. According to this model, the inhibition of SMO signaling is relieved after mutational inactivation of PTCH in the basal cell nevus syndrome. Recently, PTCH2, a molecule with sequence homology to PTCH, has been identified. To characterize both PTCH molecules with respect to the various Hedgehog proteins, we have isolated the human PTCH2 gene. Biochemical analysis of PTCH and PTCH2 shows that they both bind to all hedgehog family members with similar affinity and that they can form a complex with SMO. However, the expression patterns of PTCH and PTCH2 do not fully overlap. While PTCH is expressed throughout the mouse embryo, PTCH2 is found at high levels in the skin and in spermatocytes. Because Desert Hedgehog (Dhh) is expressed specifically in the testis and is required for germ cell development, it is likely that PTCH2 mediates its activity in vivo. Chromosomal localization of PTCH2 places it on chromosome 1p33-34, a region deleted in some germ cell tumors, raising the possibility that PTCH2 may be a tumor suppressor in Dhh target cells.


Subject(s)
Chromosomes, Human, Pair 1 , Drosophila Proteins , Insect Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Amino Acid Sequence , Animals , Chromosome Mapping , Cloning, Molecular , Hedgehog Proteins , Humans , Intracellular Signaling Peptides and Proteins , Mice , Molecular Sequence Data , Patched Receptors , Patched-1 Receptor , Patched-2 Receptor , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Sequence Alignment , Vertebrates
10.
Neuron ; 21(1): 53-62, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9697851

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) is a distant member of the TGFbeta protein family that is essential for neuronal survival and renal morphogenesis. We show that mice who are deficient in the glycosyl-phosphatidyl inositol (GPI) -linked protein GFRalpha1 (GDNFRalpha) display deficits in the kidneys, the enteric nervous system, and spinal motor and sensory neurons that are strikingly similar to those of the GDNF- and Ret-deficient mice. GFRalpha1-deficient dopaminergic and nodose sensory ganglia neurons no longer respond to GDNF or to the structurally related protein neurturin (NTN) but can be rescued when exposed to GDNF or neurturin in the presence of soluble GFRalpha1. In contrast, GFRalpha1-deficient submandibular parasympathetic neurons retain normal response to these two factors. Taken together with the available genetic and biochemical data, these findings support the idea that GFRalpha1 and the transmembrane tyrosine kinase Ret are both necessary receptor components for GDNF in the developing kidney and nervous system, and that GDNF and neurturin can mediate some of their activities through a second receptor.


Subject(s)
Aging/physiology , Drosophila Proteins , Kidney/embryology , Nerve Tissue Proteins/metabolism , Nervous System/embryology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Animals , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Embryonic and Fetal Development/physiology , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Intestines/innervation , Kidney/growth & development , Mice , Nerve Growth Factors/pharmacology , Nervous System/growth & development , Nervous System/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neurturin , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/deficiency
11.
Mol Cell Neurosci ; 11(5-6): 247-59, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9698392

ABSTRACT

The Eph-related tyrosine kinase receptor, REK7/EphA5, mediates the effects of AL-1/Ephrin-A5 and related ligands and is involved in the guidance of retinal, cortical, and hippocampal axons during development. The continued expression of REK7/EphA5 in the adult brain, in particular in areas associated with a high degree of synaptic plasticity such as the hippocampus, raises the question of its function in the mature nervous system. In this report we examined the role of REK7/EphA5 in synaptic remodeling by asking if agents that either block or activate REK7/EphA5 affect synaptic strength in hippocampal slices from adult mouse brain. We show that a REK7/EphA5 antagonist, soluble REK7/EphA5-IgG, impairs the induction of long-term potentiation (LTP) without affecting other synaptic parameters such as normal synaptic transmission or paired-pulse facilitation. In contrast, perfusion with AL-1/Ephrin-A5-IgG, an activator of REK7/EphA5, induces a sustained increase in normal synaptic transmission that partially mimics LTP. The sustained elevation of normal synaptic transmission could be attributable to a long-lasting binding of the AL-1/Ephrin-A5-IgG to the endogenous REK7/EphA5 receptor, as revealed by immunohistochemistry. Furthermore, maximal electrical induction of LTP occludes the potentiating effects of subsequent treatment with AL-1/Ephrin-A5-IgG. Taken together these results implicate REK7/EphA5 in the regulation of synaptic plasticity in the mature hippocampus and suggest that REK7/EphA5 activation is recruited in the LTP induced by tetanization.


Subject(s)
Dentate Gyrus/chemistry , Dentate Gyrus/enzymology , Neuronal Plasticity/physiology , Receptor Protein-Tyrosine Kinases/genetics , Transcription Factors/genetics , Animals , Axons/chemistry , Axons/drug effects , CD4 Antigens/pharmacology , Cells, Cultured , Dendrites/chemistry , Dendrites/drug effects , Electric Stimulation , Ephrin-A2 , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Fluorescent Antibody Technique , Gene Expression Regulation, Enzymologic , Immunoglobulin G/pharmacology , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Organ Culture Techniques , RNA, Messenger/analysis , Rats , Receptor Protein-Tyrosine Kinases/analysis , Receptor Protein-Tyrosine Kinases/pharmacology , Receptor, EphA5 , Solubility , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Time Factors , Transcription Factors/pharmacology
12.
J Neurosci ; 18(13): 4929-37, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9634558

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) exhibits potent effects on survival and function of midbrain dopaminergic (DA) neurons in a variety of models. Although other growth factors expressed in the vicinity of developing DA neurons have been reported to support survival of DA neurons in vitro, to date none of these factors duplicate the potent and selective actions of GDNF in vivo. We report here that neurturin (NTN), a homolog of GDNF, is expressed in the nigrostriatal system, and that NTN exerts potent effects on survival and function of midbrain DA neurons. Our findings indicate that NTN mRNA is sequentially expressed in the ventral midbrain and striatum during development and that NTN exhibits survival-promoting actions on both developing and mature DA neurons. In vitro, NTN supports survival of embryonic DA neurons, and in vivo, direct injection of NTN into the substantia nigra protects mature DA neurons from cell death induced by 6-OHDA. Furthermore, administration of NTN into the striatum of intact adult animals induces behavioral and biochemical changes associated with functional upregulation of nigral DA neurons. The similarity in potency and efficacy of NTN and GDNF on DA neurons in several paradigms stands in contrast to the differential distribution of the receptor components GDNF Family Receptor alpha1 (GFRalpha1) and GFRalpha2 within the ventral mesencephalon. These results suggest that NTN is an endogenous trophic factor for midbrain DA neurons and point to the possibility that GDNF and NTN may exert redundant trophic influences on nigral DA neurons acting via a receptor complex that includes GFRalpha1.


Subject(s)
Corpus Striatum/cytology , Dopamine/physiology , Nerve Growth Factors/genetics , Neurons/cytology , Substantia Nigra/cytology , 3,4-Dihydroxyphenylacetic Acid/analysis , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Corpus Striatum/embryology , Disease Models, Animal , Dopamine/analysis , Gene Expression Regulation, Developmental/physiology , Glial Cell Line-Derived Neurotrophic Factor , Mice , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/pharmacology , Neurons/chemistry , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neurturin , Nucleus Accumbens/cytology , Nucleus Accumbens/embryology , Oxidopamine , Parkinson Disease, Secondary/drug therapy , Parkinson Disease, Secondary/pathology , RNA, Messenger/analysis , Substantia Nigra/embryology , Sympatholytics
13.
J Neurosci ; 17(19): 7288-96, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9295375

ABSTRACT

Administration of nerve growth factor (NGF) to aged or lesioned animals has been shown to reverse the atrophy of basal forebrain cholinergic neurons and ameliorate behavioral deficits. To examine the importance of endogenous NGF in the survival of basal forebrain cholinergic cells and in spatial memory, mice bearing a disruption mutation in one allele of the NGF gene were studied. Heterozygous mutant mice (ngf+/-) have reduced levels of NGF mRNA and protein within the hippocampus and were found to display significant deficits in memory acquisition and retention in the Morris water maze. The behavioral deficits observed in NGF-deficient mice were accompanied by both shrinkage and loss of septal cells expressing cholinergic markers and by a decrease in cholinergic innervation of the hippocampus. Infusions of NGF into the lateral ventricle of adult ngf+/- mice abolished the deficits on the water maze task. Prolonged exposure to NGF may be required to induce cognitive effects, because reversal of the acquisition deficit was seen after long (5 weeks) but not short (3 d) infusion. Although NGF administration did not result in any improvement in the number of septal cells labeled for choline acetyltransferase, this treatment did effectively correct the deficits in both size of cholinergic neurons and density of cholinergic innervation of the hippocampus. These findings demonstrate the importance of endogenous NGF for survival and function of basal forebrain cholinergic neurons and reveal that partial depletion of this trophic factor is associated with measurable deficits in learning and memory.


Subject(s)
Alleles , Memory Disorders/genetics , Nerve Growth Factors/genetics , Neurons/pathology , Parasympathetic Nervous System/pathology , Prosencephalon/pathology , Acetylcholinesterase/metabolism , Animals , Atrophy , Behavior, Animal/drug effects , Hippocampus/drug effects , Injections, Intraventricular , Maze Learning/drug effects , Mice , Mice, Mutant Strains , Motor Activity/drug effects , Nerve Growth Factors/deficiency , Nerve Growth Factors/pharmacology , Neurons/drug effects , Prosencephalon/drug effects , Prosencephalon/enzymology , Septum Pellucidum/pathology , Swimming , Time Factors
14.
Nature ; 387(6634): 717-21, 1997 Jun 12.
Article in English | MEDLINE | ID: mdl-9192898

ABSTRACT

Glial-cell-line-derived neurotrophic factor (GDNF) and neurturin (NTN) are two structurally related, potent survival factors for sympathetic, sensory and central nervous system neurons. GDNF mediates its actions through a multicomponent receptor system composed of a ligand-binding glycosyl-phosphatidylinositol (GPI)-linked protein (designated GDNFR-alpha) and the transmembrane protein tyrosine kinase Ret. In contrast, the mechanism by which the NTN signal is transmitted is not well understood. Here we describe the identification and tissue distribution of a GPI-linked protein (designated NTNR-alpha) that is structurally related to GDNFR-alpha. We further demonstrate that NTNR-alpha binds NTN (K[d] approximately 10 pM) but not GDNF with high affinity; that GDNFR-alpha binds to GDNF but not NTN with high affinity; and that cellular responses to NTN require the presence of NTNR-alpha. Finally, we show that NTN, in the presence of NTNR-alpha, induces tyrosine-phosphorylation of Ret, and that NTN, NTNR-alpha and Ret form a physical complex on the cell surface. These findings identify Ret and NTNR-alpha as signalling and ligand-binding components, respectively, of a receptor for NTN and define a novel family of receptors for neurotrophic and differentiation factors composed of a shared transmembrane protein tyrosine kinase and a ligand-specific GPI-linked protein.


Subject(s)
Drosophila Proteins , Glycosylphosphatidylinositols/metabolism , Nerve Growth Factors/metabolism , Neurons/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Binding, Competitive , Cell Membrane/metabolism , Cell Survival/drug effects , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Humans , In Situ Hybridization , Ligands , Mice , Molecular Sequence Data , Motor Neurons/cytology , Motor Neurons/metabolism , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurturin , Phosphorylation , Phosphotyrosine/metabolism , Proto-Oncogene Proteins c-ret , Rats , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Recombinant Proteins/metabolism , Signal Transduction
15.
J Biol Chem ; 272(11): 7264-77, 1997 Mar 14.
Article in English | MEDLINE | ID: mdl-9054423

ABSTRACT

Here we describe a novel member of the receptor-like protein-tyrosine phosphatases (PTPs) termed PTP lambda, which is homologous to the homotypically adherent PTPs kappa and mu. Murine PTP lambda contains MAM, IgG, fibronectin type III, and dual phosphatase domains. As has been demonstrated for PTPs kappa and mu, PTP lambda mediates homotypic adhesion in vitro, and PTP lambda is associated with beta catenin in kidney epithelial cells. The extracellular domain of PTP lambda is proteolytically processed in cell culture as well as in vivo. Northern blot analysis reveals that PTP lambda is expressed throughout embryonic development and is predominately found in adult brain, lung, and kidney. In situ hybridization to 15.5-day old rat embryos reveals that PTP lambda is expressed in a variety of embryonic neuronal sites as well as in the esophagus, lung bronchiolar epithelium, kidney glomerular epithelium, olfactory epithelium, and various cartilagenous sites. Analysis of neonatal brain demonstrates expression in cells of the hippocampus, cortex, and the substantia nigra. Finally, immunohistochemical analysis reveals expression of this PTP on specific neurons of the spinal cord as well as on isolated cortical neurons.


Subject(s)
Embryo, Mammalian/enzymology , Protein Tyrosine Phosphatases/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Gene Expression Regulation, Developmental , Mice , Molecular Sequence Data , Protein Tyrosine Phosphatases/biosynthesis , Protein Tyrosine Phosphatases/isolation & purification , Rats , Receptor-Like Protein Tyrosine Phosphatases, Class 2 , Sequence Homology, Amino Acid
16.
J Mol Endocrinol ; 18(1): 77-85, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9061609

ABSTRACT

Leptin, the product of the ob gene, is a hormone secreted by fat cells which is primarily involved in the regulation of body weight. We have generated a leptin immunoadhesin (leptin-IgG) which was more potent than leptin alone at reducing body weight and food intake when injected into ob/ob mice. This molecule was used to identify high affinity binding sites on human embryonic 293 kidney cells and subsequently to isolate a cDNA encoding the leptin receptor from this cell line by expression cloning. This receptor corresponds to the short form of the recently isolated leptin receptor. Analysis of the expression pattern of the two forms of receptor by Northern blot, in situ hybridization and quantitative PCR showed that the receptor is expressed in most tissues but that the long form is prevalent in the hypothalamus.


Subject(s)
Carrier Proteins/genetics , Cell Adhesion Molecules/metabolism , Proteins/metabolism , Receptors, Cell Surface , Recombinant Fusion Proteins/metabolism , Animals , COS Cells , Carrier Proteins/metabolism , Cloning, Molecular , DNA, Complementary , Humans , Immunoglobulin G/metabolism , In Situ Hybridization , Leptin , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Receptors, Leptin
17.
Eur J Neurosci ; 9(11): 2383-93, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9464932

ABSTRACT

BatK is a second member of the Csk family of regulatory kinases that phosphorylate a key inhibitory tyrosine on Src family kinases, leading to down-regulation. To investigate the roles of BatK and Csk, both of which are expressed in the brain, we compared their temporal expression patterns during development of the central nervous system (CNS) in rats. BatK mRNA is undetectable at embryonic day 12 (E12), appears in the developing nervous system at approximately E15, and its expression progressively increases up to the time of birth, thereafter remaining high throughout the adult brain. In striking contrast, Csk is highly expressed throughout embryonic development and remains high in the CNS until birth. It is then dramatically down-regulated in the adult brain except in the olfactory bulb. BatK and Csk thus exhibit complementary temporal expression patterns. Since BatK expression correlates with late-stage development and terminal differentiation, we speculated that it might be involved in regulating neuronal differentiation. Using PC12 cells as a model system, we show that overexpression of BatK is sufficient to induce neurite outgrowth in the absence of nerve growth factor. Further, overexpression of BatK activates the mitogen-activated protein kinase cascade. We propose a model suggesting that, despite overlapping in vitro activities, BatK and Csk regulate different targets in vivo and have different functions during and after neuronal development, BatK being the dominant regulator of Src kinases in the fully differentiated adult brain.


Subject(s)
Central Nervous System/growth & development , Central Nervous System/metabolism , Nerve Tissue Proteins/biosynthesis , Protein-Tyrosine Kinases/biosynthesis , src Homology Domains , Animals , Antimetabolites/pharmacology , Blotting, Southern , Bromodeoxyuridine/pharmacology , CSK Tyrosine-Protein Kinase , Cell Differentiation/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Fluorescent Antibody Technique, Indirect , Genetic Vectors , In Situ Hybridization , Nerve Growth Factors/physiology , Neurons/metabolism , PC12 Cells , Polymerase Chain Reaction , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/pharmacology , Rats , Transfection , src-Family Kinases
18.
Nature ; 384(6605): 129-34, 1996 Nov 14.
Article in English | MEDLINE | ID: mdl-8906787

ABSTRACT

The protein Sonic hedgehog (Shh) controls patterning and growth during vertebrate development. Here we demonstrate that it binds Patched (vPtc), which has been identified as a tumour-suppressor protein in basal cell carcinoma, with high affinity. We show that Ptc can form a physical complex with a newly cloned vertebrate homologue of the Drosophila protein Smoothened (vSmo), and that vSmo is coexpressed with vPtc in many tissues but does not bind Shh directly. These findings, combined with available genetic evidence from Drosophila, support the hypothesis that Ptc is a receptor for Shh, and that vSmo could be a signalling component that is linked to Ptc.


Subject(s)
Drosophila Proteins , Genes, Tumor Suppressor , Insect Hormones/genetics , Membrane Proteins/genetics , Proteins/genetics , Proteins/metabolism , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled , Trans-Activators , Amino Acid Sequence , Animals , COS Cells , Cell Line , Cloning, Molecular , Drosophila , Hedgehog Proteins , Humans , Insect Hormones/metabolism , Intracellular Signaling Peptides and Proteins , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Patched Receptors , Patched-1 Receptor , Rats , Receptors, Cell Surface/metabolism , Sequence Homology, Amino Acid , Smoothened Receptor , Tissue Distribution
19.
Neuron ; 17(1): 63-74, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8755479

ABSTRACT

The muscle-derived factors required for survival of embryonic motoneurons are not clearly identified. Cardiotrophin-1 (CT-1), a cytokine related to ciliary neurotrophic factor (CNTF), is expressed at high levels in embryonic limb bud and is secreted by differentiated myotubes. In vitro, CT-1 kept 43% of purified E14 rat motoneurons alive for 2 weeks (EC50 = 20 pM). In vivo, CT-1 protected neonatal sciatic motoneurons against the effects of axotomy. CT-1 action on motoneurons was inhibited by phosphatidylinositol-specific phospholipase C (PIPLC), suggesting that CT-1 may act through a GPI-linked component. Since no binding of CT-1 to CNTFR alpha was detected, CT-1 may use a novel cytokine receptor alpha subunit. CT-1 may be important in normal motoneuron development and as a potential tool for slowing motoneuron degeneration in human diseases.


Subject(s)
Cytokines/physiology , Motor Neurons/physiology , Muscles/embryology , Muscles/metabolism , Spinal Cord/cytology , Animals , Animals, Newborn , Axons/physiology , Base Sequence , Cell Survival , Cytokines/genetics , Denervation , Embryo, Mammalian/metabolism , Mice/embryology , Molecular Probes/genetics , Molecular Sequence Data , RNA, Messenger/metabolism , Rats/embryology , Receptor, Ciliary Neurotrophic Factor , Receptors, Nerve Growth Factor/metabolism , Time Factors
20.
Nature ; 382(6586): 76-9, 1996 Jul 04.
Article in English | MEDLINE | ID: mdl-8657308

ABSTRACT

Glial cell-line derived neurotrophic factor (GDNF) is a potent survival factor for embryonic midbrain dopaminergic, spinal motor, cranial sensory, sympathetic, and hindbrain noradrenergic neurons, and is available to these cells in vivo. It is therefore considered a physiological trophic factor and a potential therapeutic agent for Parkinson's disease, amyotrophic lateral sclerosis, and Alzheimer's disease. Here we show that at postnatal day 0 (P0), GDNF-deficient mice have deficits in dorsal root ganglion, sympathetic and nodose neurons, but not in hindbrain noradrenergic or midbrain dopaminergic neurons. These mice completely lack the enteric nervous system (ENS), ureters and kidneys. Thus GDNF is important for the development and/or survival of enteric, sympathetic and sensory neurons and the renal system, but is not essential for catecholaminergic neurons in the central nervous system (CNS).


Subject(s)
Digestive System/embryology , Kidney/embryology , Nerve Growth Factors/physiology , Nerve Tissue Proteins/physiology , Neurons/cytology , Animals , Brain/cytology , Brain/embryology , Cell Count , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cell Survival , Digestive System/innervation , Digestive System Abnormalities , Dopamine/metabolism , Gene Targeting , Glial Cell Line-Derived Neurotrophic Factor , In Situ Hybridization , Kidney/abnormalities , Mice , Mice, Inbred C57BL , Motor Neurons/cytology , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurons/metabolism , RNA, Messenger/metabolism , Ureter/embryology
SELECTION OF CITATIONS
SEARCH DETAIL