Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
J Chem Neuroanat ; 136: 102391, 2024 Mar.
Article En | MEDLINE | ID: mdl-38219812

BACKGROUND: Maternal diabetes during pregnancy can affect the neurological development of offspring. Glial cell-derived neurotrophic factor (GDNF), neurturin (NRTN), and neural cell adhesion molecules (NCAM) are three important proteins for brain development. Therefore, this study aimed to investigate the impacts of the mentioned neurotrophic factors in the hippocampal dentate gyrus (DG) of rat offspring born to diabetic mothers. METHODS: Wistar female rats were randomly allocated into diabetic (STZ-D) [(45 mg/kg BW, STZ (Streptozotocin), i.p)], diabetic + NPH insulin (STZ-INS) [(4-6 unit/kg/day SC)], and control groups. The animals in all groups were mated by non-diabetic male rats. Two weeks after birth, male pups from each group were sacrificed and then protein contents of GDNF, NRTN, and NCAM were evaluated using immunohistochemistry. RESULTS: The study found that the expression of GDNF and NRTN in the hippocampus of diabetic rat offspring was significantly higher compared to the diabetic+ insulin and control groups, respectively (P < 0.01, P < 0.001). Additionally, the expression of NCAM was significantly higher in the diabetic group the diabetic+ insulin and control groups (P < 0.01, P < 0.001). CONCLUSIONS: The results of the study revealed that diabetes during pregnancy significantly impacts the distribution pattern of GDNF, NRTN, and NCAM in the hippocampus of rat neonates.


Diabetes, Gestational , Insulins , Humans , Pregnancy , Rats , Animals , Male , Female , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Neurturin/metabolism , Neurturin/pharmacology , Rats, Wistar , Neural Cell Adhesion Molecules/metabolism , Dentate Gyrus/metabolism
2.
Cell Metab ; 33(11): 2215-2230.e8, 2021 11 02.
Article En | MEDLINE | ID: mdl-34592133

Endurance exercise promotes skeletal muscle vascularization, oxidative metabolism, fiber-type switching, and neuromuscular junction integrity. Importantly, the metabolic and contractile properties of the muscle fiber must be coupled to the identity of the innervating motor neuron (MN). Here, we show that muscle-derived neurturin (NRTN) acts on muscle fibers and MNs to couple their characteristics. Using a muscle-specific NRTN transgenic mouse (HSA-NRTN) and RNA sequencing of MN somas, we observed that retrograde NRTN signaling promotes a shift toward a slow MN identity. In muscle, NRTN increased capillary density and oxidative capacity and induced a transcriptional reprograming favoring fatty acid metabolism over glycolysis. This combination of effects on muscle and MNs makes HSA-NRTN mice lean with remarkable exercise performance and motor coordination. Interestingly, HSA-NRTN mice largely recapitulate the phenotype of mice with muscle-specific expression of its upstream regulator PGC-1ɑ1. This work identifies NRTN as a myokine that couples muscle oxidative capacity to slow MN identity.


Motor Neurons , Neurturin , Animals , Mice , Mice, Transgenic , Motor Neurons/metabolism , Muscle, Skeletal/metabolism , Neurturin/genetics , Neurturin/metabolism , Neurturin/pharmacology , Oxidative Stress
3.
PLoS One ; 16(2): e0245663, 2021.
Article En | MEDLINE | ID: mdl-33534843

Parkinson's disease is associated with the loss of dopamine (DA) neurons in ventral mesencephalon. We have previously reported that no single neurotrophic factor we tested protected DA neurons from the dopaminergic toxin 1-methyl-4-phenylpyridinium (MPP+) in dissociated cultures isolated from the P0 rat substantia nigra, but that a combination of five neurotrophic factors was protective. We now report that cerebral DA neurotrophic factor (CDNF) and a variant of neurturin (NRTN), N4, were also not protective when provided alone but were protective when added together. In cultures isolated from the substantia nigra, MPP+ (10 µM) decreased tyrosine hydroxylase-positive cells to 41.7 ± 5.4% of vehicle control. Although treatment of cultures with 100 ng/ml of either CDNF or N4 individually before and after toxin exposure did not significantly increase survival in MPP+-treated cultures, when the two trophic factors were added together at 100 ng/ml each, survival of cells was increased 28.2 ± 6.1% above the effect of MPP+ alone. In cultures isolated from the ventral tegmental area, another DA rich area, a higher dose of MPP+ (1 mM) was required to produce an EC50 in TH-positive cells but, as in the substantia nigra, only the combination of CDNF and N4 (100 ng/ml each) was successful at increasing the survival of these cells compared to MPP+ alone (by 22.5 ± 3.5%). These data support previous findings that CDNF and N4 may be of therapeutic value for treatment of PD, but suggest that they may need to be administered together.


Dopaminergic Neurons/metabolism , Mesencephalon/metabolism , Nerve Growth Factors/pharmacology , Neuroprotective Agents/pharmacology , Neurturin/pharmacology , 1-Methyl-4-phenylpyridinium , Animals , CHO Cells , Cell Survival/drug effects , Cells, Cultured , Cricetulus , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopaminergic Neurons/drug effects , Humans , Nomifensine/pharmacology , Rats, Sprague-Dawley , Substantia Nigra/cytology , Tritium/metabolism , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/cytology
4.
Life Sci Alliance ; 3(12)2020 12.
Article En | MEDLINE | ID: mdl-33020210

Lung-resident macrophages are crucial to the maintenance of health and in the defence against lower respiratory tract infections. Macrophages adapt to local environmental cues that drive their appropriate function; however, this is often dysregulated in many inflammatory lung pathologies. In mucosal tissues, neuro-immune interactions enable quick and efficient inflammatory responses to pathogenic threats. Although a number of factors that influence the antimicrobial response of lung macrophages are known, the role of neuronal factors is less well understood. Here, we show an intricate circuit involving the neurotrophic factor, neurturin (NRTN) on human lung macrophages that dampens pro-inflammatory cytokine release and modulates the type of matrix metalloproteinases produced in response to viral stimuli. This circuit involves type 1 interferon-induced up-regulation of RET that when combined with the glial cell line-derived neurotrophic factor (GDNF) receptor α2 (GFRα2) allows binding to epithelial-derived NRTN. Our research highlights a non-neuronal immunomodulatory role for NRTN and a novel process leading to a specific antimicrobial immune response by human lung-resident macrophages.


Lung/immunology , Macrophages, Alveolar/metabolism , Neurturin/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Humans , Lung/metabolism , Lung/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages, Alveolar/immunology , Neurons/metabolism , Neurturin/metabolism , Proto-Oncogene Proteins c-ret/metabolism , RNA, Messenger/metabolism , Virus Diseases/immunology , Virus Diseases/metabolism
5.
Neuropharmacology ; 147: 28-36, 2019 03 15.
Article En | MEDLINE | ID: mdl-29857941

Parkinson's disease (PD) is a disorder affecting dopamine neurons for which there is no cure. Glial cell line-derived neurotrophic factor (GDNF) and the closely related protein neurturin are two trophic factors with demonstrated neuroprotective and neurorestorative properties on dopamine neurons in multiple animal species. However, GDNF and neurturin Phase-2 clinical trials have failed to demonstrate a significant level of improvement over placebo controls. Insufficient drug distribution in the brain parenchyma has been proposed as a major contributing factor for the lack of clinical efficacy in the Phase-2 trial patients. To address this issue, a novel mammalian cell-derived variant form of GDNF (GDNFv) was designed to promote better tissue distribution by reducing its heparin binding to the extracellular matrix and key amino acids were substituted to enhance its chemical stability. Administration of this fully glycosylated GDNFv in the normal rat striatum increased dopamine turnover and produced significantly greater brain distribution than E. coli-produced wildtype GDNF (GDNFwt). Intrastriatal GDNFv also protected midbrain dopamine neuron function in 6-hydroxydopamine-lesioned rats. Studies conducted in normal adult rhesus macaques support that GDNFv was well tolerated in all animals and demonstrated a greater volume of distribution than GDNFwt in the brain following intrastriatal infusion. Importantly, favorable physiological activity of potential therapeutic value was maintained in this variant trophic factor with significant target activation in GDNFv recipients as indicated by dopamine turnover modulation. These data suggest that GDNFv may be a promising drug candidate for the treatment of PD. Additional studies are needed in non-human primates with dopamine depletion. This article is part of the Special Issue entitled 'Drug Repurposing: old molecules, new ways to fast track drug discovery and development for CNS disorders'.


Brain/metabolism , Dopamine/metabolism , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Neurturin/pharmacology , Animals , Brain/drug effects , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Humans , Macaca mulatta , Neurturin/pharmacokinetics , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Rats , Rats, Sprague-Dawley , Tissue Distribution
6.
Diabetes ; 66(7): 2007-2018, 2017 07.
Article En | MEDLINE | ID: mdl-28408435

Neurturin (NRTN), a member of the glial-derived neurotrophic factor family, was identified from an embryonic chicken pancreatic cDNA library in a screen for secreted factors. In this study, we assessed the potential antidiabetic activities of NRTN relative to liraglutide, a glucagon-like peptide 1 receptor agonist, in Zucker diabetic fatty (ZDF) rats. Subcutaneous administration of NRTN to 8-week-old male ZDF rats prevented the development of hyperglycemia and improved metabolic parameters similar to liraglutide. NRTN treatment increased pancreatic insulin content and ß-cell mass and prevented deterioration of islet organization. However, unlike liraglutide-treated rats, NRTN-mediated improvements were not associated with reduced body weight or food intake. Acute NRTN treatment did not activate c-Fos expression in key feeding behavior and metabolic centers in ZDF rat brain or directly enhance glucose-stimulated insulin secretion from pancreatic ß-cells. Treating 10-week-old ZDF rats with sustained hyperglycemia with liraglutide resulted in some alleviation of hyperglycemia, whereas NRTN was not as effective despite improving plasma lipids and fasting glucose levels. Interestingly, coadministration of NRTN and liraglutide normalized hyperglycemia and other metabolic parameters, demonstrating that combining therapies with distinct mechanism(s) can alleviate advanced diabetes. This emphasizes that therapeutic combinations can be more effective to manage diabetes in individuals with uncontrolled hyperglycemia.


Blood Glucose/drug effects , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Liraglutide/pharmacology , Neurturin/pharmacology , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Disease Models, Animal , Eating/drug effects , Feeding Behavior/drug effects , Glucagon-Like Peptide-1 Receptor/agonists , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Organ Size , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Zucker
7.
Brain Res Bull ; 115: 37-44, 2015 Jun.
Article En | MEDLINE | ID: mdl-25931391

Progenitors were discovered in the corpus striatum several years ago, but little is known about their proliferation and differentiation. The aim of this study was to analyze embryonic progenitor cells from the corpus striatum using a bioassay with trophic stimulation. Primary cells obtained from brains of rat embryos at E13-14 were dissected from striatum niches and cultured in stem cell media. These floating dispersed cells clumped together to forming floating bodies like irregular spheres (spheroids), which were placed in type I collagen gel and cultured under basal conditions or with the addition of NGF, NT-3, or NTN. Optimum growth of neurites was obtained, and after 24 and 48 h, they were measured for number and length. The expression of proliferation markers such as PCNA and Ki67, and of neural progenitor markers such as GFAP, nestin, vimentin, O4, A2B5, Pax6, S100, TubIII, and NeuN, was then analyzed. The initial behavior in cell cultures showed distinguishable spheroids that, when placed in 3D gels and with trophic support, generated neurites. A similar effect was observed in glial cell outgrowth from the spheroids. Our assay showed high reproducibility, short culture time, and high resolution for tracing neuron-neurite outgrowth or visualizing glial outgrowth in a few hours.


Biological Assay/methods , Central Nervous System Agents/pharmacology , Embryonic Stem Cells/drug effects , Neural Stem Cells/physiology , Neurogenesis , Neurons/physiology , Animals , Biological Assay/instrumentation , Cell Culture Techniques , Cell Enlargement , Cells, Cultured , Collagen , Corpus Striatum/cytology , Corpus Striatum/drug effects , Corpus Striatum/embryology , Corpus Striatum/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Gels , Nerve Growth Factor/pharmacology , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neurites/drug effects , Neurites/physiology , Neurons/cytology , Neurons/drug effects , Neurotrophin 3/pharmacology , Neurturin/pharmacology , Rats, Sprague-Dawley
8.
J Cereb Blood Flow Metab ; 35(4): 611-22, 2015 Mar 31.
Article En | MEDLINE | ID: mdl-25586140

The blood-brain barrier (BBB) constitutes a major obstacle in brain drug delivery. Focused ultrasound (FUS) in conjunction with microbubbles has been shown to open the BBB noninvasively, locally, and transiently to allow large molecules diffusion. Neurturin (NTN), a member of the glial-derived neurotrophic factor (GDNF) family, has been demonstrated to have neuroprotective and regenerative effects on dopaminergic neurons in vivo using invasive drug delivery methods. The brain's ascending nigrostriatal pathway is severely damaged in Parkinson's disease (PD), and therefore the substantia nigra (SN) and striatal caudoputamen (CP) were selected as the target areas. The objective of the study was to investigate whether safe and efficient NTN delivery can be achieved through FUS-induced BBB opening via intravenous administration, and thus trigger the neuroregeneration cascade in the nigrostriatal pathway. After the optimization of FUS parameters and target locations in the murine brain, NTN bioavailability and downstream signaling were detected and characterized through immunostaining. FUS significantly enhanced the delivery of NTN compared with the direct injection technique, whereas triggering of the signaling cascade was detected downstream to the neuronal nuclei. These findings thus indicate the potential of the FUS method to mediate transport of proteins through the blood-brain barrier in a PD animal model.


Blood-Brain Barrier/metabolism , Drug Delivery Systems/instrumentation , Neuroprotective Agents/administration & dosage , Neurturin/administration & dosage , Sonication/instrumentation , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Equipment Design , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Neurturin/pharmacokinetics , Neurturin/pharmacology , Parkinson Disease/drug therapy , Rats, Sprague-Dawley , Signal Transduction/drug effects
9.
Carcinogenesis ; 35(1): 103-13, 2014 Jan.
Article En | MEDLINE | ID: mdl-24067900

UNLABELLED: Neurotrophic factors possess an emerging role in the pathophysiology of several gastrointestinal disorders, regulating innervation, pain sensation and disease-associated neuroplasticity. Here, we aimed at characterizing the role of the neurotrophic factor neurturin (NRTN) and its receptor glial-cell-line-derived neurotrophic factor receptor alpha-2 (GFRα-2) in pancreatic cancer (PCa) and pancreatic neuropathy. For this purpose, NRTN and GFRα-2 were studied in normal human pancreas and PCa tissues via immunohistochemistry, quantitative reverse transcription-polymerase chain reaction, immunoblotting and correlated to abdominal pain. The impact of NRTN/GFRα-2 on PCa cell (PCC) biology was investigated via exposure to hypoxia, 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyl tetrazolium bromide viability and matrigel invasion assays in native and specific small interfering RNA-silenced PCCs. To assess the influence of NRTN on pancreatic neuroplasticity and neural invasion (NI), its impact was explored via an in vitro 'neuroplasticity assay' and a 3D neural migration assay. NRTN and GFRα-2 demonstrated a site-specific upregulation in PCa, predominantly in nerves, PCCs and extracellular matrix. Patients with severe pain demonstrated higher intraneural GFRα-2 immunoreactivity than patients with no pain. PCa tissue and PCCs contained increased amounts of NRTN, which was suppressed under hypoxia. NRTN promoted PCC invasiveness, and silencing of NRTN limited both PCC proliferation and invasion. Depletion of NRTN from PCa tissue extracts and PCC supernatants decreased axonal sprouting in neuronal cultures but did not influence glial density. Silencing of NRTN in PCCs boosted NI. We conclude that increased NRTN/GFRα-2 in PCa seems to promote an aggressive PCC phenotype and neuroplasticity in PCa. Accelerated NI following NRTN suppression constitutes a novel explanation for the attraction of PCC to nerves in the hypoxic PCa tumor microenvironment. SUMMARY: PCa is characterized by intrapancreatic neuroplasticity and NI. Here, we show that PCC produce the neurotrophic factor NRTN, which reinforces their biological properties, triggers neuroplastic alterations, NI and influences pain sensation via the GFRα-2 receptor.


Abdominal Pain/metabolism , Neuronal Plasticity , Neurturin/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/physiopathology , Animals , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/pathology , Gene Expression Regulation, Neoplastic , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Humans , Male , Middle Aged , Neuronal Plasticity/physiology , Neurturin/genetics , Neurturin/pharmacology , Protein Isoforms/metabolism , Rats
10.
Biochim Biophys Acta ; 1833(12): 2789-2802, 2013 Dec.
Article En | MEDLINE | ID: mdl-23872421

Neurturin (NRTN), a member of the GDNF family of ligands (GFL), is currently investigated in a series of clinical trials for Parkinson's disease. NRTN signals through its cognate receptor GFRα2 and co-receptor RET to induce neurite outgrowth, but the underlying mechanism remains to be better understood. STAT3 was previously shown to be activated by oncogenic RET, independent of ligand and GFRα. In this study, we demonstrated that NRTN induced serine(727) but not tyrosine(705) phosphorylation of STAT3 in primary cortical neuron and neuronal cell lines. Remarkably, STAT3 phosphorylation was found to be mediated specifically by GFRα2c and RET9 isoforms. Furthermore, serine but not tyrosine dominant negative mutant of STAT3 impaired NRTN induced neurite outgrowth, indicative of the role of STAT3 as a downstream mediator of NRTN function. Similar to NGF, the NRTN induced P-Ser-STAT3 was localized to the mitochondria but not to the nucleus. Mitochondrial STAT3 was further found to be intimately involved in NRTN induced neurite outgrowth. Collectively, these findings demonstrated the hitherto unrecognized and novel role of specific GFRα2 and RET isoforms in mediating NRTN activation of STAT3 and the transcription independent mechanism whereby the mitochondria localized P-Ser-STAT3 mediated NRTN induced neurite outgrowth.


Alternative Splicing/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Neurturin/pharmacology , Proto-Oncogene Proteins c-ret/metabolism , STAT3 Transcription Factor/metabolism , Alternative Splicing/drug effects , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Ligands , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Neural Cell Adhesion Molecules/metabolism , Neurites/drug effects , Neurites/metabolism , PC12 Cells , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport/drug effects , Proto-Oncogene Proteins c-ret/genetics , Rats , src-Family Kinases/metabolism
11.
Nat Commun ; 4: 1494, 2013.
Article En | MEDLINE | ID: mdl-23422662

Parasympathetic nerves are a vital component of the progenitor cell niche during development, maintaining a pool of progenitors for organogenesis. Injured adult organs do not regenerate after parasympathectomy, and there are few treatments to improve organ regeneration, particularly after damage by therapeutic irradiation. Here we show that restoring parasympathetic function with the neurotrophic factor neurturin increases epithelial organ regeneration after damage. We use mouse salivary gland explant culture containing fluorescently labelled progenitors, and injure the tissue with irradiation. The progenitors survive, parasympathetic function is diminished and epithelial apoptosis reduces the expression of neurturin, which increases neuronal apoptosis. Treatment with neurturin reduces neuronal apoptosis, restores parasympathetic function and increases epithelial regeneration. Furthermore, adult human salivary glands damaged by irradiation also have reduced parasympathetic innervation. We propose that neurturin will protect the parasympathetic nerves from damage and improve organ regeneration. This concept may be applicable for other organs where parasympathetic innervation influences their function.


Epithelium/innervation , Epithelium/physiology , Organogenesis , Parasympathetic Nervous System/physiology , Regeneration , Submandibular Gland/innervation , Submandibular Gland/physiology , Adult , Aged , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Epithelium/growth & development , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Neurites/drug effects , Neurites/metabolism , Neurturin/pharmacology , Organogenesis/drug effects , Organogenesis/radiation effects , Parasympathetic Nervous System/drug effects , Parasympathetic Nervous System/radiation effects , Radiation, Ionizing , Regeneration/drug effects , Regeneration/radiation effects , Submandibular Gland/drug effects , Submandibular Gland/radiation effects
12.
J Mol Neurosci ; 49(3): 480-90, 2013 Mar.
Article En | MEDLINE | ID: mdl-22847529

Neurons and glial cells can protect each other from stress and following death by mutual exchange with neurotrophins. In order to examine involvement of different neurotrophic factors in neuroglial interactions in a photosensitized crayfish stretch receptor, a simple model object consisting of only two sensory neurons enveloped by glial cells, we studied the influence of glial cell line-derived neurotrophic factor (GDNF), neurturin, and ciliary neurotrophic factor (CNTF) on its photodynamic injury. Photodynamic treatment, which causes strong oxidative stress, induced firing abolition and necrosis of neurons, necrosis, and apoptosis of glial cells. GDNF significantly reduced photoinduced neuronal necrosis and neurturin but not CNTF showed a similar tendency. Both of them significantly reduced necrosis and apoptosis of glial cells. At the ultrastructural level, neurons and glial cells treated with GDNF in the darkness contained large mitochondria with well-developed cristae, numerous ribosomes, polysomes, rough endoplasmic reticulum (ER), and dictyosomes. This indicated the high level of bioenergetic, biosynthetic, and transport processes. Photodynamic treatment caused swelling and vacuolization of mitochondria, dictyosomes, and ER. It also impaired formation of glial protrusions and double membrane vesicles that transfer glial material into the neuron. GDNF prevented photoinduced mitochondria swelling that disturbed the cellular bioenergetics and cytoplasm vacuolization associated with injury of intracellular organelles. It also preserved the structures involved in protein synthesis and transport: rough ER, dictyosomes, polysomes, microtubule bundles, submembrane cisterns, and double membrane vesicles. GDNF-mediated maintenance of metabolism and ultrastructure of photosensitized neurons and glial cells may be the basis of its neuro- and glia protective effects.


Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Mechanoreceptors/drug effects , Neuroglia/drug effects , Neurons/drug effects , Neurturin/pharmacology , Photosensitivity Disorders/drug therapy , Radiation-Protective Agents/pharmacology , Action Potentials/drug effects , Animals , Astacoidea , Cell Death , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Ciliary Neurotrophic Factor/pharmacology , Humans , In Vitro Techniques , Indoles/toxicity , Lasers , Mechanoreceptors/radiation effects , Mechanoreceptors/ultrastructure , Mitochondria/drug effects , Mitochondria/ultrastructure , Neuroglia/radiation effects , Neuroglia/ultrastructure , Neurons/radiation effects , Neurons/ultrastructure , Organometallic Compounds/toxicity , Oxidative Stress/drug effects , Photochemotherapy/adverse effects , Photosensitizing Agents/toxicity , Recombinant Proteins/pharmacology
13.
Cell Signal ; 23(11): 1727-37, 2011 Nov.
Article En | MEDLINE | ID: mdl-21723942

Cyclic AMP (cAMP) and neurotrophic factors are known to interact closely to promote neurite outgrowth and neuronal regeneration. Glial cell line-derived neurotrophic factor (GDNF) and its family member neurturin (NTN) transduce signal through a multi-component receptor complex consisting of GDNF family receptor alpha 2 (GFRα2) and Ret receptor tyrosine kinase. Neurons from GFRα2-deficient mice do not promote axonal initiation when stimulated by NTN, consistent with the role of GFRα2 in neuronal outgrowth. Multiple alternatively spliced isoforms of GFRα2 are known to be expressed in the nervous system. GFRα2a and GFRα2c but not GFRα2b promoted neurite outgrowth. It is currently unknown if cAMP signalling is differentially regulated by these isoforms. In this study, NTN activation of GFRα2a and GFRα2c but not GFRα2b induced biphasic ERK1/2 activation and phosphorylation of the major cAMP target CREB. Interestingly, inhibition of cAMP signalling significantly impaired GFRα2a and GFRα2c-mediated neurite outgrowth while cAMP agonists cooperated with GFRα2b to induce neurite outgrowth. Importantly, the specific cAMP effector PKA but not Epac was essential for NTN-induced neurite outgrowth, through transcription and translation-dependent activation of late phase ERK1/2. Taken together, these results not only demonstrated the essential role of cAMP-PKA signalling in NTN-induced biphasic ERK1/2 activation and neurite outgrowth, but also suggested cAMP-PKA signalling as a hitherto unrecognized underlying mechanism contributing to the differential neuritogenic activities of GFRα2 isoforms.


Cyclic AMP-Dependent Protein Kinases/pharmacology , Cyclic AMP/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurites/physiology , Neurturin , Protein Isoforms/metabolism , Signal Transduction/physiology , Acetylcysteine/analogs & derivatives , Acetylcysteine/metabolism , Acetylcysteine/pharmacology , Animals , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Erythromycin/analogs & derivatives , Erythromycin/metabolism , Erythromycin/pharmacology , Gene Expression Regulation/physiology , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Humans , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 3/genetics , Nerve Regeneration , Neurturin/metabolism , Neurturin/pharmacology , Phosphorylation/drug effects , Protein Isoforms/genetics , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Rats , Real-Time Polymerase Chain Reaction , Transfection
14.
Brain Res ; 1381: 31-7, 2011 Mar 24.
Article En | MEDLINE | ID: mdl-21241671

Activation of endothelin receptors expressed in DRG neurons is functionally coupled to translocation of PKCε from cytoplasm to the plasma membrane. Using immunocytochemistry we show that in DRG cultured neurons PKCε translocation induced by endothelin-1 was prominently seen in a peptidergic subpopulation of cultured DRG neurons largely negative for isolectin B4 staining, indicating that in basal conditions functional expression of endothelin receptors does not occur in non-peptidergic, RET-expressing nociceptors. Translocation was blocked by the specific ETA-R antagonist BQ-123 while it was unaffected by the ETB-R antagonist BQ-788. No calcium response in response to endothelin-1 was observed in sensory neurons, while large and long-lasting responses were observed in the majority of non-neuronal cells present in DRG cultures, which are ensheathing Schwann cells and satellite cells, identified with the glial marker S-100. Calcium responses in non-neuronal cells were abolished by BQ-788. The fraction of peptidergic PKCε-translocated neurons was significantly increased by nerve growth factor, while in the presence of neurturin or glia-derived neurotropic factor (GDNF), an IB4-positive subpopulation of small- and medium-sized neurons showed PKCε translocation induced by endothelin-1 which could be blocked by BQ-123 but not by BQ-788. Our in vitro results show that the level of expression of functional endothelin receptors coupled to PKCε is different in peptidergic and non-peptidergic nociceptors and is modulated with different mechanisms in distinct neuronal subpopulations.


Ganglia, Spinal/metabolism , Glycoproteins/metabolism , Lectins/metabolism , Receptors, Endothelin/metabolism , Sensory Receptor Cells/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Endothelin-1/pharmacology , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Immunohistochemistry , Nerve Growth Factor/pharmacology , Neurturin/pharmacology , Protein Kinase C-epsilon/metabolism , Protein Transport/drug effects , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/cytology , Sensory Receptor Cells/drug effects , Up-Regulation/drug effects , Up-Regulation/physiology , Versicans
15.
Folia Histochem Cytobiol ; 48(3): 434-41, 2010 Sep 30.
Article En | MEDLINE | ID: mdl-21071351

Parkinson's disease (PD) is the second most common neurodegenerative disorder marked by cell death in the Substantia nigra (SN). Docosahexaenoic acid (DHA) is the major polyunsaturated fatty acid (PUFA) in the phospholipid fraction of the brain and is required for normal cellular function. Glial cell line derived neurotrophic factor (GDNF) and neurturin (NTN) are very potent trophic factors for PD. The aim of the study was to evaluate the neuroprotective effects of GDNF and NTN by investigating their immunostaining levels after administration of DHA in a model of PD. For this reason we hypothesized that DHA administration of PD might alter GDNF, NTN expression in SN. MPTP neurotoxin that induces dopaminergic neurodegeneration was used to create the experimental Parkinsonism model. Rats were divided into; control, DHA-treated (DHA), MPTP-induced (MPTP), MPTP-induced+DHA-treated (MPTP+DHA) groups. Dopaminergic neuron numbers were clearly decreased in MPTP, but showed an increase in MPTP+DHA group. As a result of this, DHA administration protected dopaminergic neurons as shown by tyrosine hydroxylase immunohistochemistry. In the MPTP+DHA group, GDNF, NTN immunoreactions in dopaminergic neurons were higher than that of the MPTP group. In conclusion, the characterization of GDNF and NTN will certainly help elucidate the mechanism of DHA action, and lead to better strategies for the use of DHA to treat neurodegenerative diseases.


Docosahexaenoic Acids/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Neurturin/metabolism , Parkinson Disease, Secondary/pathology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Disease Models, Animal , Dopamine/metabolism , Glial Cell Line-Derived Neurotrophic Factor/adverse effects , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Immunohistochemistry , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neurotoxins , Neurturin/adverse effects , Neurturin/pharmacology , Random Allocation , Rats , Rats, Wistar , Substantia Nigra/cytology , Substantia Nigra/metabolism , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/immunology , Tyrosine 3-Monooxygenase/metabolism
16.
Neurochem Int ; 57(5): 540-6, 2010 Nov.
Article En | MEDLINE | ID: mdl-20615442

Neurturin (NTN), a member of the glial cell line-derived neurotrophic factor (GDNF) family, has substantial effects on normal and lesioned nigrostriatal dopamine systems. However, its ability to protect against toxin-induced loss of striatal dopamine release has not been previously reported. The goal of the present study was to determine if NTN could protect against 6-hydroxydopamine (6-OHDA)-induced reductions in striatal dopamine overflow and tissue levels of dopamine and to compare the effects of NTN with those of GDNF. Male Fischer-344 rats were given a single injection of vehicle, or 5 microg NTN or GDNF, into the right striatum. The following day the animals were given a single injection of 12 microg 6-OHDA into the striatum at the same site where the trophic factor was injected. Microdialysis experiments conducted three weeks later indicated that the 6-OHDA decreased basal levels of dopamine and metabolites in the lesioned striatum compared to the contralateral striatum, and NTN was able to partially protect against the 6-OHDA-induced reductions. Injection of NTN one day prior to 6-OHDA also led to significant protection against loss of both potassium- and amphetamine-evoked overflow of dopamine. The NTN treatments partially protected against 6-OHDA-induced reductions in striatal tissue levels of dopamine and completely protected against loss of nigral dopamine content. The protective effects of NTN were similar in magnitude to those of GDNF. These results support that within the experimental parameters used in this study, NTN is as effective as GDNF in protecting against the dopamine-depleting effects of intrastriatal 6-OHDA.


Corpus Striatum/metabolism , Dopamine/metabolism , Neuroprotective Agents , Neurturin/pharmacology , Oxidopamine/antagonists & inhibitors , Oxidopamine/toxicity , Sympatholytics/toxicity , 3,4-Dihydroxyphenylacetic Acid/metabolism , Amphetamine/pharmacology , Animals , Chromatography, High Pressure Liquid , Corpus Striatum/drug effects , Dopamine Uptake Inhibitors/pharmacology , Functional Laterality/physiology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Homovanillic Acid/metabolism , Male , Microdialysis , Nerve Growth Factors/metabolism , Potassium Chloride/pharmacology , Rats , Rats, Inbred F344 , Substantia Nigra/drug effects , Substantia Nigra/metabolism
17.
Neurochem Res ; 35(5): 727-34, 2010 May.
Article En | MEDLINE | ID: mdl-20119638

Neurturin (NTN) is a member of the glial cell line-derived neurotrophic factor (GDNF) family; and, while GDNF has been shown to increase dopamine (DA) release in normal animals, the ability of NTN to alter DA release has not been previously reported. The purpose of the present study was to determine if NTN could alter striatal DA release, and to compare the effects of NTN to GDNF. Male Fischer-344 rats were given a single injection of vehicle or 5 microg NTN or GDNF into the right substantia nigra. Three weeks later microdialysis experiments were conducted to assess striatal DA release. Basal extracellular levels of striatal DA were not affected by either NTN or GDNF. However, both NTN and GDNF led to increases in amphetamine-evoked overflow of DA from the ipsilateral striatum, and there was a trend for potassium-evoked overflow to be augmented. Postmortem tissue levels of DA were decreased by approximately 20% in the striatum, and increased by approximately 100% in the substantia nigra, on the ipsilateral side of the brain compared to the contralateral side following both NTN and GDNF injection. Thus, NTN, like GDNF, can augment striatal DA release, and the magnitude of the NTN effects are similar to those of GDNF.


Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Neurturin/pharmacology , Substantia Nigra/drug effects , Substantia Nigra/metabolism , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Homovanillic Acid/metabolism , Male , Rats , Rats, Inbred F344
18.
Neuropharmacology ; 58(7): 1114-21, 2010 Jun.
Article En | MEDLINE | ID: mdl-20153340

Convection-enhanced delivery (CED) of GDNF and NTN was employed to determine the tissue clearance of these factors from the rat striatum and the response of the dopaminergic system to a single infusion. Two doses of GDNF (15 and 3 microg) and NTN (10 microg and 2 microg) were infused into the rat striatum. Animals were euthanized 3, 7, 14, 21, and 28 days post-infusion. Brains were processed for ELISA, HPLC, and immunohistochemistry (IHC). Both doses of the infused GDNF resulted in a sharp increase in striatal GDNF levels followed by a rapid decrease between day 3 and 7. Interestingly, IHC revealed GDNF in the septum and the base of the brain 14 days after GDNF administration. Dopamine (DA) turnover was significantly increased in a dose-dependent manner for more than 7 days after a single GDNF infusion. NTN persisted in the brain for at least two weeks longer than GDNF. It also had more persistent effects on DA turnover, probably due to its precipitation in the brain at neutral pH after infusion. Our data suggest that daily or continuous dosing may not be necessary for delivering growth factors into the CNS.


Brain/metabolism , Dopamine/metabolism , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Neurturin/pharmacology , Neurturin/pharmacokinetics , Animals , Brain/drug effects , Chromatography, High Pressure Liquid , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Immunohistochemistry , Male , Neurturin/administration & dosage , Neurturin/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
19.
Mol Cell Neurosci ; 41(4): 464-73, 2009 Aug.
Article En | MEDLINE | ID: mdl-19463953

Glial cell line-derived neurotrophic factor (GDNF) transduces signal and promotes neurite outgrowths in diverse neurons through the interactions of GDNF family receptor alpha 1 (GFRalpha1) and other co-receptors including Ret receptor tyrosine kinase and NCAM. GFRalpha1 is alternatively spliced into two isoforms, GFRalpha1a and GFRalpha1b, with five amino acids difference. In this study, we found that both GFRalpha1a and GFRalpha1b were expressed in various human tissues. Interestingly, when stimulated with GDNF, GFRalpha1a but not GFRalpha1b promoted neurite outgrowth in neuroblastoma cells through the activations of ERK1/2, Rac1 and Cdc42. Remarkably, in cells co-expressing GFRalpha1a and GFRalpha1b, GDNF inhibited neurite outgrowths. The inhibitory activity of GFRalpha1b was dependent on RhoA and ROCK activation. Furthermore, GFRalpha1b but not GFRalpha1a activated Rho and various ROCK downstream effectors LIMK1/2, cofilin and MLC2. This study demonstrates the hitherto unrecognized roles of GFRalpha1 isoforms in the activation of distinct signaling pathways and in neurite outgrowths.


Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Neurites/physiology , Protein Isoforms/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Central Nervous System/metabolism , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Humans , Mice , Neuroblastoma , Neurturin/pharmacology , Rho Factor/metabolism , Transfection/methods , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism
20.
Neurosurgery ; 64(4): 602-12; discussion 612-3, 2009 Apr.
Article En | MEDLINE | ID: mdl-19349823

OBJECTIVE: Parkinson's disease is characterized by profound motor deficits that result mainly as a consequence of degeneration of midbrain dopaminergic neurons. No current therapy slows or halts disease progression. Neurturin (NTN) and glial cell line-derived neurotrophic factor have potent neuroprotective and neurorestorative effects on dopaminergic neurons, but their use in treating Parkinson's disease has been limited by significant delivery obstacles. In this study, we examined the long-term expression, bioactivity, and safety/tolerability of CERE-120, an adeno-associated virus type 2 vector encoding human NTN, after bilateral stereotactic delivery to the striatum of nonhuman primates. METHODS: Twelve naïve rhesus macaques received bilateral stereotactic injections of 1 of 2 CERE-120 doses or vehicle to the caudate and putamen. Neurological and clinical parameters were monitored for up to 1 year postadministration, after which animals were sacrificed for histological analyses. RESULTS: Dose-related NTN expression was observed at 1 year and was associated with enhanced tyrosine hydroxylase immunolabeling in the striatum, hypertrophy of tyrosine hydroxylase-positive cells in the substantia nigra, and induction of extracellular signal-regulated kinase signaling in the substantia nigra. Extensive, formal analyses, conducted in accordance with Good Laboratory Practice Regulations, across multiple time points revealed no evidence of clinical, neurological, or systemic toxicity. CONCLUSION: The present study provides evidence of long-term expression and bioactivity of NTN on the dopaminergic nigrostriatal system after bilateral stereotactic delivery of CERE-120 to the striatum. Furthermore, no evidence of any adverse effects for up to 1 year postadministration was observed. These findings reveal a wide safety margin for CERE-120 and collectively support the ongoing clinical testing of the efficacy and safety of CERE-120 in patients with Parkinson's disease.


Corpus Striatum/drug effects , Dependovirus/genetics , Gene Expression/physiology , Neurturin/administration & dosage , Neurturin/pharmacology , Parkinson Disease/drug therapy , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Behavior, Animal/drug effects , Behavior, Animal/physiology , Body Weight/drug effects , Brain/metabolism , Calcitonin Gene-Related Peptide/metabolism , Cell Count , Corpus Striatum/metabolism , Corpus Striatum/virology , Dependovirus/metabolism , Dose-Response Relationship, Drug , Eating/drug effects , Enzyme-Linked Immunosorbent Assay/methods , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Transfer Techniques , Genetic Vectors , Humans , Longitudinal Studies , Macaca mulatta , Neurturin/genetics , Neurturin/metabolism , Spinal Cord/metabolism , Time Factors , Tissue Distribution , Tyrosine 3-Monooxygenase/metabolism , Viral Matrix Proteins/metabolism
...