1
|
Li H, Dan QQ, Chen YJ, Chen L, Zhang HT, Mu DZ, Wang TH. Cellular Localization and Distribution of TGF-β1, GDNF and PDGF-BB in the Adult Primate Central Nervous System. Neurochem Res 2023; 48:2406-2423. [PMID: 36976393 DOI: 10.1007/s11064-023-03909-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/29/2023]
Abstract
The available data on the localization of transforming growth factor beta1 (TGF-β1), glial cell line-derived neurotrophic factor (GDNF), and platelet-derived growth factor-BB (PDGF-BB) in the adult primate and human central nervous system (CNS) are limited and lack comprehensive and systematic information. This study aimed to investigate the cellular localization and distribution of TGF-β1, GDNF, and PDGF-BB in the CNS of adult rhesus macaque (Macaca mulatta). Seven adult rhesus macaques were included in the study. The protein levels of TGF-β1, PDGF-BB, and GDNF in the cerebral cortex, cerebellum, hippocampus, and spinal cord were analyzed by western blotting. The expression and location of TGF-β1, PDGF-BB, and GDNF in the brain and spinal cord was examined by immunohistochemistry and immunofluorescence staining, respectively. The mRNA expression of TGF-β1, PDGF-BB, and GDNF was detected by in situ hybridization. The molecular weight of TGF-β1, PDGF-BB, and GDNF in the homogenate of spinal cord was 25 KDa, 30 KDa, and 34 KDa, respectively. Immunolabeling revealed GDNF was ubiquitously distributed in the cerebral cortex, hippocampal formation, basal nuclei, thalamus, hypothalamus, brainstem, cerebellum, and spinal cord. TGF-β1 was least distributed and found only in the medulla oblongata and spinal cord, and PDGF-BB expression was also limited and present only in the brainstem and spinal cord. Besides, TGF-β1, PDGF-BB, and GDNF were localized in the astrocytes and microglia of spinal cord and hippocampus, and their expression was mainly found in the cytoplasm and primary dendrites. The mRNA of TGF-β1, PDGF-BB, and GDNF was localized to neuronal subpopulations in the spinal cord and cerebellum. These findings suggest that TGF-β1, GDNF and PDGF-BB may be associated with neuronal survival, neural regeneration and functional recovery in the CNS of adult rhesus macaques, providing the potential insights into the development or refinement of therapies based on these factors.
Collapse
Affiliation(s)
- Hui Li
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Department of Intensive Care Unit of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Qi-Qin Dan
- Institute of Neurological Disease, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China
| | - Yan-Jun Chen
- Institute of Neurological Disease, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China
| | - Li Chen
- Institute of Neurological Disease, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China
| | - Hong-Tian Zhang
- Institute of Neurological Disease, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China
| | - De-Zhi Mu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Ting-Hua Wang
- Institute of Neurological Disease, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
2
|
Onesto MM, Short CA, Rempel SK, Catlett TS, Gomez TM. Growth Factors as Axon Guidance Molecules: Lessons From in vitro Studies. Front Neurosci 2021; 15:678454. [PMID: 34093120 PMCID: PMC8175860 DOI: 10.3389/fnins.2021.678454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
Growth cones at the tips of extending axons navigate through developing organisms by probing extracellular cues, which guide them through intermediate steps and onto final synaptic target sites. Widespread focus on a few guidance cue families has historically overshadowed potentially crucial roles of less well-studied growth factors in axon guidance. In fact, recent evidence suggests that a variety of growth factors have the ability to guide axons, affecting the targeting and morphogenesis of growth cones in vitro. This review summarizes in vitro experiments identifying responses and signaling mechanisms underlying axon morphogenesis caused by underappreciated growth factors.
Collapse
Affiliation(s)
| | | | | | | | - Timothy M. Gomez
- Neuroscience Training Program and Cell and Molecular Biology Program, Department of Neuroscience, University of Wisconsin–Madison, Madison, WI, United States
| |
Collapse
|
3
|
Cortés D, Carballo-Molina OA, Castellanos-Montiel MJ, Velasco I. The Non-Survival Effects of Glial Cell Line-Derived Neurotrophic Factor on Neural Cells. Front Mol Neurosci 2017; 10:258. [PMID: 28878618 PMCID: PMC5572274 DOI: 10.3389/fnmol.2017.00258] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 07/31/2017] [Indexed: 01/23/2023] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) was first characterized as a survival-promoting molecule for dopaminergic neurons (DANs). Afterwards, other cells were also discovered to respond to GDNF not only as a survival factor but also as a protein supporting other cellular functions, such as proliferation, differentiation, maturation, neurite outgrowth and other phenomena that have been less studied than survival and are now more extendedly described here in this review article. During development, GDNF favors the commitment of neural precursors towards dopaminergic, motor, enteric and adrenal neurons; in addition, it enhances the axonal growth of some of these neurons. GDNF also induces the acquisition of a dopaminergic phenotype by increasing the expression of Tyrosine Hydroxylase (TH), Nurr1 and other proteins that confer this identity and promote further dendritic and electrical maturation. In motor neurons (MNs), GDNF not only promotes proliferation and maturation but also participates in regenerating damaged axons and modulates the neuromuscular junction (NMJ) at both presynaptic and postsynaptic levels. Moreover, GDNF modulates the rate of neuroblastoma (NB) and glioblastoma cancer cell proliferation. Additionally, the presence or absence of GDNF has been correlated with conditions such as depression, pain, muscular soreness, etc. Although, the precise role of GDNF is unknown, it extends beyond a survival effect. The understanding of the complete range of properties of this trophic molecule will allow us to investigate its broad mechanisms of action to accelerate and/or improve therapies for the aforementioned pathological conditions.
Collapse
Affiliation(s)
- Daniel Cortés
- Instituto de Fisiología Celular—Neurociencias, Universidad Nacional Autónoma de MéxicoMéxico City, Mexico
- Laboratorio de Reprogramación Celular del IFC-UNAM, Instituto Nacional de Neurología y NeurologíaMéxico City, Mexico
| | - Oscar A. Carballo-Molina
- Instituto de Fisiología Celular—Neurociencias, Universidad Nacional Autónoma de MéxicoMéxico City, Mexico
- Laboratorio de Reprogramación Celular del IFC-UNAM, Instituto Nacional de Neurología y NeurologíaMéxico City, Mexico
| | - María José Castellanos-Montiel
- Instituto de Fisiología Celular—Neurociencias, Universidad Nacional Autónoma de MéxicoMéxico City, Mexico
- Laboratorio de Reprogramación Celular del IFC-UNAM, Instituto Nacional de Neurología y NeurologíaMéxico City, Mexico
| | - Iván Velasco
- Instituto de Fisiología Celular—Neurociencias, Universidad Nacional Autónoma de MéxicoMéxico City, Mexico
- Laboratorio de Reprogramación Celular del IFC-UNAM, Instituto Nacional de Neurología y NeurologíaMéxico City, Mexico
| |
Collapse
|
4
|
Soderstrom K, O'Malley J, Steece-Collier K, Kordower JH. Neural Repair Strategies for Parkinson's Disease: Insights from Primate Models. Cell Transplant 2017; 15:251-65. [PMID: 16719060 DOI: 10.3727/000000006783982025] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Nonhuman primate models of Parkinson's disease (PD) have been invaluable to our understanding of the human disease and in the advancement of novel therapies for its treatment. In this review, we attempt to give a brief overview of the animal models of PD currently used, with a more comprehensive focus on the advantages and disadvantages presented by their use in the nonhuman primate. In particular, discussion addresses the 6-hydroxydopamine (6-OHDA), 1-methyl-1,2,3,6-tetrahydopyridine (MPTP), rotenone, paraquat, and maneb parkinsonian models. Additionally, the role of primate PD models in the development of novel therapies, such as trophic factor delivery, grafting, and deep brain stimulation, are described. Finally, the contribution of primate PD models to our understanding of the etiology and pathology of human PD is discussed.
Collapse
Affiliation(s)
- Katherine Soderstrom
- Department of Neurological Science, Research Center for Brain Repair, Rush University Medical Center, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
5
|
Marsh SE, Blurton-Jones M. Neural stem cell therapy for neurodegenerative disorders: The role of neurotrophic support. Neurochem Int 2017; 106:94-100. [PMID: 28219641 PMCID: PMC5446923 DOI: 10.1016/j.neuint.2017.02.006] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 12/19/2016] [Accepted: 02/14/2017] [Indexed: 12/17/2022]
Abstract
Neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and Huntington's disease currently affect tens of millions of people worldwide. Unfortunately, as the world's population ages, the incidence of many of these diseases will continue to rise and is expected to more than double by 2050. Despite significant research and a growing understanding of disease pathogenesis, only a handful of therapies are currently available and all of them provide only transient benefits. Thus, there is an urgent need to develop novel disease-modifying therapies to prevent the development or slow the progression of these debilitating disorders. A growing number of pre-clinical studies have suggested that transplantation of neural stem cells (NSCs) could offer a promising new therapeutic approach for neurodegeneration. While much of the initial excitement about this strategy focused on the use of NSCs to replace degenerating neurons, more recent studies have implicated NSC-mediated changes in neurotrophins as a major mechanism of therapeutic efficacy. In this mini-review we will discuss recent work that examines the ability of NSCs to provide trophic support to disease-effected neuronal populations and synapses in models of neurodegeneration. We will then also discuss some of key challenges that remain before NSC-based therapies for neurodegenerative diseases can be translated toward potential clinical testing.
Collapse
Affiliation(s)
- Samuel E Marsh
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Mathew Blurton-Jones
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA; Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
6
|
Sarchielli P, Alberti A, Candeliere A, Floridi A, Capocchi G, Calabresi P. Glial Cell Line-Derived Neurotrophic Factor and Somatostatin Levels in Cerebrospinal Fluid of Patients Affected by Chronic Migraine and Fibromyalgia. Cephalalgia 2016; 26:409-15. [PMID: 16556241 DOI: 10.1111/j.1468-2982.2005.01048.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The aim of the present study was to verify cerebrospinal fluid (CSF) levels of glial cell line-derived neurotrophic factor (GDNF) and somatostatin, both measured by sensitive immunoassay, in: 16 chronic migraine (CM) patients, 15 patients with an antecedent history of migraine without aura diagnosed as having probable chronic migraine (PCM) and probable analgesic-abuse headache (PAAH), 20 patients affected by primary fibromyalgia syndrome (PFMS), and 20 control subjects. Significantly lower levels of GDNF and somatostatin were found in the CSF of both CM and PCM + PAAH patients compared with controls (GDNF = P < 0.001, P < 0.002; somatostatin = P < 0.002, P < 0.0003), without significant difference between the two groups. PFMS patients, with and without analgesic abuse, also had significantly lower levels of both somatostatin and GDNF ( P < 0.0002, P < 0.001), which did not differ from those of CM and PCM + PAAH patients. A significant positive correlation emerged between CSF values of GDNF and those of somatostatin in CM ( r = 0.70, P < 0.02), PCM + PAAH ( r = 0.78, P < 0.004), and PFMS patients ( r = 0.68, P < 0.008). Based on experimental findings, it can be postulated that reduced CSF levels of GDNF and somatostatin in both CM and PCM + PAAH patients can contribute to sustained central sensitization underlying chronic head pain. The abuse of simple or combination analgesics does not seem to influence the biochemical changes investigated, which appear to be more strictly related to the chronic pain state, as demonstrated also for fibromyalgia.
Collapse
Affiliation(s)
- P Sarchielli
- Neurologic Clinic, Department of Medical and Surgical Specialties and Public Health, University of Perugia, Perugia, Italy.
| | | | | | | | | | | |
Collapse
|
7
|
Hypothalamic proteoglycan syndecan-3 is a novel cocaine addiction resilience factor. Nat Commun 2013; 4:1955. [PMID: 23736082 PMCID: PMC3709481 DOI: 10.1038/ncomms2955] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 04/29/2013] [Indexed: 11/08/2022] Open
Abstract
Proteoglycans like syndecan-3 have complex signaling roles in addition to their function as structural components of the extracellular matrix. Here, we show that syndecan-3 in the lateral hypothalamus has an unexpected new role in limiting compulsive cocaine intake. In particular, we observe that syndecan-3 null mice self-administer greater amounts of cocaine than wild-type mice. This effect can be rescued by re-expression of syndecan-3 in the lateral hypothalamus with an adeno-associated viral vector. Adeno-associated viral vector delivery of syndecan-3 to the lateral hypothalamus also reduces motivation for cocaine in normal mice. Syndecan-3 limits cocaine intake by modulating the effects of glial-cell-line-derived neurotrophic factor, which uses syndecan-3 as an alternative receptor. Our findings indicate syndecan-3-dependent signaling as a novel therapeutic target for the treatment of cocaine addiction.
Collapse
|
8
|
Del Fiacco M, Quartu M, Serra MP, Boi M, Demontis R, Poddighe L, Picci C, Melis T. The human cuneate nucleus contains discrete subregions whose neurochemical features match those of the relay nuclei for nociceptive information. Brain Struct Funct 2013; 219:2083-101. [PMID: 23975345 PMCID: PMC4223579 DOI: 10.1007/s00429-013-0625-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 08/04/2013] [Indexed: 12/21/2022]
Abstract
The present paper is aimed at defining distinctive subdivisions of the human cuneate nucleus (Cu), evident from prenatal to old life, whose occurrence has never been clearly formalized in the human brain, or described in other species so far. It extends our early observations on the presence of gray matter areas that host strong substance P (SP) immunoreactivity in the territory of the human Cu and adjacent cuneate fascicle. Here we provide a three-dimensional reconstruction of the Cu fields rich in SP and further identify those areas by means of their immunoreactivity to the neuropeptides SP, calcitonin gene-related peptide, methionine- and leucine-enkephalin, peptide histidine-isoleucine, somatostatin and galanin, to the trophins glial cell line-derived neurotrophic factor and brain-derived neurotrophic factor, and to the neuroplasticity proteins polysialylated neural cell adhesion molecule and growth-associated protein-43. The presence, density and distribution of immunoreactivity for each of these molecules closely resemble those occurring in the superficial layers of the caudal spinal trigeminal nucleus (Sp5C). Myelin and Nissl stainings suggest that those Cu subregions and the Sp5C superficial layers share a similar histological aspect. This work establishes the existence of definite subregions, localized within the Cu territory, that bear the neurochemical and histological features of sensory nuclei committed to the neurotransmission of protopathic stimuli, including pain. These findings appear of particular interest when considering that functional, preclinical and clinical studies show that the dorsal column nuclei, classical relay station of fine somatic tactile and proprioceptive sensory stimuli, are also involved in pain neurotransmission.
Collapse
Affiliation(s)
- Marina Del Fiacco
- Department of Biomedical Sciences, Section of Cytomorphology, University of Cagliari, Cittadella Universitaria, 09042, Monserrato, Italy,
| | | | | | | | | | | | | | | |
Collapse
|
9
|
McCullough MJ, Gyorkos AM, Spitsbergen JM. Short-term exercise increases GDNF protein levels in the spinal cord of young and old rats. Neuroscience 2013; 240:258-68. [PMID: 23500094 DOI: 10.1016/j.neuroscience.2013.02.063] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 02/12/2013] [Accepted: 02/27/2013] [Indexed: 12/29/2022]
Abstract
Neurotrophic factors may play a role in exercise-induced neuroprotective effects, however it is not known if exercise mediates changes in glial cell line-derived neurotrophic factor (GDNF) protein levels in the spinal cord. The aim of the current study was to determine if 2 weeks of exercise alters GDNF protein content in the lumbar spinal cord of young and old rats. GDNF protein was quantified via an enzyme-linked immunosorbent assay and Western blot. Immunohistochemical analysis localized GDNF in choline acetyltransferase (ChAT)-positive motor neurons and cell body areas were measured. Involuntary running in the young animals appeared to elicit the greatest increase in GDNF protein content (sixfold increase), followed by swimming (threefold increase) and voluntary running (twofold increase); however there was no significant difference between the modalities of exercise. Low-intensity running of the old animals significantly increased GDNF protein content in the spinal cord. Both young and old exercised animals showed a doubling in ChAT-positive motor neuron cell body areas. These results suggest that GDNF protein content in the spinal cord is modulated by exercise.
Collapse
Affiliation(s)
- M J McCullough
- Department of Biological Sciences, Western Michigan University, 1903 W. Michigan Avenue, Kalamazoo, MI 49008-5410, USA.
| | | | | |
Collapse
|
10
|
Injury-induced accumulation of glial cell line-derived neurotrophic factor in the rostral part of the injured rat spinal cord. Int J Mol Sci 2012. [PMID: 23202963 PMCID: PMC3497337 DOI: 10.3390/ijms131013484] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The spinal cord of a 7-week-old female Wistar rat was hemi-transected at thoracic position 10 with a razor blade, and changes in glial cell line-derived neurotrophic factor (GDNF) protein and mRNA expression levels in the spinal cord were examined. GDNF protein and mRNA expression levels were evaluated by enzyme immunoassay and reverse transcription polymerase chain reaction, respectively. Although GDNF is distributed in the healthy spinal cord from 150 to 400 pg/g tissue in a regionally dependent manner, hemi-transection (left side) of the spinal cord caused a rapid increase in GDNF content in the ipsilateral rostral but not in the caudal part of the spinal cord. On the other hand, injury-induced GDNF mRNA was distributed limitedly in both rostral and caudal stumps. These observations suggest the possibility that increased GDNF in the rostral part is responsible for the accumulation of GDNF that may be constitutively transported from the rostral to caudal side within the spinal cord. Although such local increase of endogenous GDNF protein may not be sufficient for nerve regeneration and locomotor improvement, it may play a physiological role in supporting spinal neurons including motoneurons.
Collapse
|
11
|
Geng Z, Xu FY, Huang SH, Chen ZY. Sorting protein-related receptor SorLA controls regulated secretion of glial cell line-derived neurotrophic factor. J Biol Chem 2011; 286:41871-41882. [PMID: 21994944 DOI: 10.1074/jbc.m111.246413] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF), after secreted from cells, plays a critical role in central and peripheral neuron survival and function. The secretion of GDNF can be either constitutive or regulated by physiological stimuli; however, the detailed mechanism driving GDNF secretion is still unknown. Here, we report that sorting protein-related receptor with A-type repeats (SorLA), a member of the mammal Vps10p domain receptor, interacts with GDNF and is localized to GDNF-containing vesicles. Overexpression of SorLA significantly increases, and knockdown of SorLA by siRNA decreases, the regulated secretion of GDNF in PC12 and MN9D cells but has no effect on GDNF constitutive secretion. In addition, overexpression of a truncated form of SorLA also impairs GDNF-regulated secretion. Finally, we found that the prodomain of GDNF mediates the interaction of GDNF with SorLA under acidic conditions. Moreover, overexpression of SorLA could enhance the regulated secretion of the GDNF prodomain-GFP fusion protein, suggesting that the prodomain of GDNF is responsible for its regulated secretion. Together, these findings will advance our understanding of the molecular mechanism underlying GDNF-regulated secretion.
Collapse
Affiliation(s)
- Zhao Geng
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Feng-Yi Xu
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Shu-Hong Huang
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Zhe-Yu Chen
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China.
| |
Collapse
|
12
|
Anastasía A, Wojnacki J, de Erausquin GA, Mascó DH. Glial cell-line derived neurotrophic factor is essential for electroconvulsive shock-induced neuroprotection in an animal model of Parkinson's disease. Neuroscience 2011; 195:100-11. [PMID: 21871541 DOI: 10.1016/j.neuroscience.2011.08.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 07/18/2011] [Accepted: 08/09/2011] [Indexed: 01/25/2023]
Abstract
Sustained motor improvement in human patients with idiopathic Parkinson's disease has been described following electroconvulsive shock (ECS) treatment. In rats, ECS stimulates the expression of various trophic factors (TFs), some of which have been proposed to exert neuroprotective actions. We previously reported that ECS protects the integrity of the rat nigrostriatal dopaminergic system against 6-hydroxydopamine (6-OHDA)-induced toxicity; in order to shed light into its neuroprotective mechanism, we studied glial cell-line derived neurotrophic factor (GDNF) levels (the most efficient TF for dopaminergic neurons) in the substantia nigra (SN) and striatum of 6-OHDA-injected animals with or without ECS treatment. 6-OHDA injection decreased GDNF levels in the SN control animals, but not in those receiving chronic ECS, suggesting that changes in GDNF expression may participate in the ECS neuroprotective mechanism. To evaluate this possibility, we inhibit GDNF by infusion of GDNF function blocking antibodies in the SN of 6-OHDA-injected animals treated with ECS (or sham ECS). Animals were sacrificed 7 days after 6-OHDA infusion, and the integrity of the nigrostriatal system was studied by tyrosine hydroxylase immunohistochemistry and Cresyl Violet staining. Neuroprotection observed in ECS-treated animals was inhibited by GDNF antibodies in the SN. These results robustly demonstrate that GDNF is essential for the ECS neuroprotective effect observed in 6-OHDA-injected animals.
Collapse
Affiliation(s)
- A Anastasía
- Facultad de Ciencias Exactas, Físicas y Naturales, Centro de Biología Celular y Molecular, Universidad Nacional de Córdoba, Av. Vélez Sarsfield 1611, ZC: X5016GCA, Córdoba, Argentina
| | | | | | | |
Collapse
|
13
|
Takeda M, Kitagawa J, Nasu M, Takahashi M, Iwata K, Matsumoto S. Glial cell line-derived neurotrophic factor acutely modulates the excitability of rat small-diameter trigeminal ganglion neurons innervating facial skin. Brain Behav Immun 2010; 24:72-82. [PMID: 19679180 DOI: 10.1016/j.bbi.2009.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Revised: 07/14/2009] [Accepted: 08/05/2009] [Indexed: 12/31/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) plays an important role in adult sensory neuron function. However, the acute effects of GDNF on primary sensory neuron excitability remain to be elucidated. The aim of the present study was to investigate whether GDNF acutely modulates the excitability of adult rat trigeminal ganglion (TRG) neurons that innervate the facial skin by using perforated-patch clamping, retrograde-labeling and immunohistochemistry techniques. Fluorogold (FG) retrograde labeling was used to identify the TRG neurons innervating the facial skin. The FG-labeled small- and medium-diameter GDNF immunoreactive TRG neurons, and most of these neurons also expressed the GDNF family receptor alpha-1 (GFRalpha-1). In whole-cell voltage-clamp mode, GDNF application significantly inhibited voltage-gated K(+) transient (I(A)) and sustained (I(K)) currents in most dissociated FG-labeled small-diameter TRG neurons. This effect was concentration-dependent and was abolished by co-application of the protein tyrosine kinase inhibitor, K252b. Under current-clamp conditions, the repetitive firing during a depolarizing pulse were significantly increased by GDNF application. GDNF application also increased the duration of the repolarization phase and decreased the duration of the depolarization phase of the action potential, and these characteristic effects were also abolished by co-application of K252b. These results suggest that acute application of GDNF enhances the neuronal excitability of adult rat small-diameter TRG neurons innervating the facial skin, via activation of GDNF-induced intracellular signaling pathway. We therefore conclude that a local release of GDNF from TRG neuronal soma and/or nerve terminals may regulate normal sensory function, including nociception.
Collapse
Affiliation(s)
- Mamoru Takeda
- Department of Physiology, School of Life Dentistry at Tokyo, Nippon Dental University, 1-9-20, Fujimi-cho, Chiyoda-ku, Tokyo 102-8159, Japan.
| | | | | | | | | | | |
Collapse
|
14
|
Facello B, Castaldo L, De Martino L, Lucini C. Glial cell line-derived neurotrophic factor in Purkinje cells of adult zebrafish: An autocrine mode of action? Neurosci Lett 2009; 465:133-7. [DOI: 10.1016/j.neulet.2009.08.068] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Revised: 07/29/2009] [Accepted: 08/25/2009] [Indexed: 11/28/2022]
|
15
|
Wang Y, Geng Z, Zhao L, Huang SH, Sheng AL, Chen ZY. GDNF isoform affects intracellular trafficking and secretion of GDNF in neuronal cells. Brain Res 2008; 1226:1-7. [PMID: 18598685 DOI: 10.1016/j.brainres.2008.05.087] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2008] [Revised: 05/14/2008] [Accepted: 05/24/2008] [Indexed: 11/19/2022]
Abstract
Glial cell line derived neurotrophic factor (GDNF) plays a critical role in central and peripheral neuron survival and function. In human and rodents, GDNF exists in an alternative spliced isoform (GDNF Delta 78), which has a 78 bp deletion in the pro-region of the GDNF encoding sequence. Whether the GDNF isoform affects GDNF function is unknown. Here, we investigated the secretion and intracellular localization of the GDNF Delta 78 isoform in neuronal cell populations. Our data indicate that a decreased secretion and an abnormal intracellular distribution of GDNF Delta 78 occurred in neuronal cells. The colocalization studies revealed much more localization of GDNF Delta 78 with Golgi marker-TGN38, which indicates that the accumulation of GDNF Delta 78 in the Golgi apparatus might in part account for its intracellular trafficking and secretion deficit. To our knowledge, it is reported for the first time that the GDNF Delta 78 isoform has a deficit in GDNF intracellular trafficking and secretion.
Collapse
Affiliation(s)
- Yue Wang
- Department of Neurobiology, School of Medicine, Shandong University, No.44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | | | | | | | | | | |
Collapse
|
16
|
Stefanova N, Poewe W, Wenning GK. Rasagiline is neuroprotective in a transgenic model of multiple system atrophy. Exp Neurol 2007; 210:421-7. [PMID: 18222424 DOI: 10.1016/j.expneurol.2007.11.022] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2007] [Revised: 11/14/2007] [Accepted: 11/16/2007] [Indexed: 10/22/2022]
Abstract
Rasagiline is a novel selective irreversible monoamine oxidase-B (MAO-B) inhibitor recently introduced for the symptomatic treatment of Parkinson disease. Like other propargylamines rasagiline has also shown neuroprotective effects independent of MAO-B-inhibition in various in vitro and in vivo models. The present study was performed to test the potential of rasagiline as a disease-modifying agent in multiple system atrophy (MSA) using a transgenic mouse model previously described by our group. (PLP)-alpha-synuclein transgenic mice featuring glial cytoplasmic inclusion pathology underwent 3-nitropropionic acid intoxication to model full-blown MSA-like neurodegeneration. Two doses of rasagiline were used (0.8 and 2.5 mg/kg) for a treatment period of 4 weeks. Rasagiline-treated animals were compared to placebo saline-treated mice by evaluation of motor behaviour and neuropathology. Motor behavioural tests including pole test, stride length test and general motor score evaluation showed improvements in motor deficits associated with 2.5 mg/kg rasagiline therapy. Immunohistochemistry and histology showed significant reduction of 3-NP-induced neuronal loss in striatum, substantia nigra pars compacta, cerebellar cortex, pontine nuclei and inferior olives of MSA mice receiving 2.5 mg/kg rasagiline. The results of the study indicate that rasagiline confers neuroprotection in a transgenic mouse model of MSA and may therefore be considered a promising disease-modifying candidate for human MSA.
Collapse
Affiliation(s)
- Nadia Stefanova
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria.
| | | | | |
Collapse
|
17
|
Abstract
The mainstays of Parkinson's disease (PD) treatment remain symptomatic, including initial dopamine replacement and subsequent deep brain stimulation, however, neither of these approaches is neuroprotective. Neurotrophic factors - proteins that activate cell signalling pathways regulating neuronal survival, differentiation, growth and regeneration - represent an alternative for treating dopaminergic neurons in PD but are difficult to administer clinically because they do not pass through the blood-brain barrier. Glial cell line-derived neurotrophic factor (GDNF) has potent neurotrophic effects particularly but not exclusively on dopaminergic neurons; in animal models of PD, it has consistently demonstrated both neuroprotective and neuroregenerative effects when provided continuously, either by means of a viral vector or through continuous infusion either into the cerebral ventricles (ICV) or directly into the denervated putamen. This led to a human PD study in which GDNF was administered by monthly bolus intracerebroventricular injections, however, no clinical benefit resulted, probably because of the limited penetration to the target brain areas, and instead significant side effects occurred. In an open-label study of continuous intraputamenal GDNF infusion in five patients (one unilaterally and four bilaterally), we reported excellent tolerance, few side effects and clinical benefit evident within three months of the commencement of treatment. The clinical improvement was sustained and progressive, and by 24-months patients demonstrated a 57 and 63% improvement in their off-medication motor and activities of daily living UPDRS subscores, respectively, with clear benefit in dyskinesias. The benefit was associated with a significant increase in putamenal 18F-dopa uptake on positron emission tomography (PET), and in one patient coming to autopsy after 43 months of unilateral infusion there was evident increased tyrosine hydroxylase immunopositive nerve fibres in the infused putamen. A second open trial in 10 patients using unilateral intraputamenal GDNF infusions has also demonstrated a greater than 30% bilateral benefit in both on- and off-medication scores at 24 weeks. Based on our 6-month results, a randomized controlled clinical trial was conducted to confirm the open-label results, however, GDNF infusion over 6-months did not confer the predetermined level of clinical benefit to patients with PD despite increased 18F-dopa uptake surrounding the catheter tip. It is possible that technical differences between this trial and the positive open label studies contributed to this negative outcome.
Collapse
Affiliation(s)
- N K Patel
- Institute of Neurosciences, Frenchay Hospital, Bristol, UK
| | | |
Collapse
|
18
|
Katoh-Semba R, Tsuzuki M, Miyazaki N, Yoshida A, Nakajima H, Nakagawa C, Kitajima S, Matsuda M. Distribution and immunohistochemical localization of GDNF protein in selected neural and non-neural tissues of rats during development and changes in unilateral 6-hydroxydopamine lesions. Neurosci Res 2007; 59:277-87. [PMID: 17765347 DOI: 10.1016/j.neures.2007.07.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Revised: 07/17/2007] [Accepted: 07/18/2007] [Indexed: 01/15/2023]
Abstract
The tissue distribution of glial cell line-derived neurotrophic factor (GDNF) during development and changes in GDNF levels by unilateral 6-hydroxydopamine lesions were investigated in rats using a newly established enzyme immunoassay system and by immunohistochemistry. The detection limit of the assay was 0.3 pg/0.2 ml and the system recognized glycosylated mature GDNF. Concentrations of GDNF were relatively high in the kidney and testis during the embryonic and neonatal periods, respectively, and decreased with age. In the striatum, hippocampus and brain stem, GDNF reached a maximal level at around postnatal day 14. However, brain levels were generally lower than those in non-neural tissues. In the CNS, GDNF immunoreactivity was observed in striatal neurons, pyramidal neurons in the hippocampus and the Vth layer of the cortex, large neurons in the diagonal band and brain stem, and spinal motor neurons. It was also evident in several non-neural, tissue-specific cells, such as cells in the renal collecting ducts and distal tubules, and testicular Sertoli cells. Destruction of nigral dopaminergic neurons by 6-hydroxydopamine enhanced the levels of striatal GDNF protein, with apparent involvement of astrocytes. These results suggest that GDNF is normally synthesized in neurons, but may also be produced by astroglial cells in damaged brains.
Collapse
Affiliation(s)
- Ritsuko Katoh-Semba
- Institute for Developmental Research, Aichi Human Service Center, Kasugai, Aichi 480-0392, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Gotohda T, Tokunaga I, Kitamura O, Kubo SI. Toluene inhalation induced neuronal damage in the spinal cord and changes of neurotrophic factors in rat. Leg Med (Tokyo) 2007; 9:123-7. [PMID: 17197224 DOI: 10.1016/j.legalmed.2006.11.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2006] [Revised: 11/14/2006] [Accepted: 11/16/2006] [Indexed: 10/23/2022]
Abstract
We investigated the effects of toluene inhalation on neurons and neurotrophic factors in the spinal cord and the relationship between them. Male Wistar rats were exposed to toluene (1500ppm for 4h per day) for 7 days. To observe damage of the neurons in spinal cord with the toluene, expression of microtubule associated protein 2 (MAP2) and 70kDa heat shock protein (HSP70) in spinal cord were performed by immunohistochemistry. MAP2 was degraded and HSP70-immunoreactivity was enhanced in nerve cell bodies of the gray matter in toluene inhalation group. Immunoreactivity of glial fibrillary acidic protein (GFAP), a marker of astrocytes, was enhanced in the toluene-treated group. Furthermore, glial cell line-derived neurotrophic factor (GDNF)- and brain-derived neurotrophic factor (BDNF)-immunoreactivity in spinal cord were slightly decreased in the treated group. In addition, the concentrations of GDNF and BDNF in the spinal cord were determined using enzyme linked immunosorbent assay (ELISA). Concentration of GDNF was reduced significantly by toluene exposure. BDNF also reduced, but not significantly. The toluene inhalation caused the damage of the neuron in the spinal cord, which was accompanied by the decrease in the neurotrophic factors, such as BDNF and GDNF.
Collapse
Affiliation(s)
- Takako Gotohda
- Department of Forensic Medicine, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8503, Japan
| | | | | | | |
Collapse
|
20
|
Dong ZQ, Ma F, Xie H, Wang YQ, Wu GC. Changes of expression of glial cell line-derived neurotrophic factor and its receptor in dorsal root ganglions and spinal dorsal horn during electroacupuncture treatment in neuropathic pain rats. Neurosci Lett 2005; 376:143-8. [PMID: 15698937 DOI: 10.1016/j.neulet.2004.11.044] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2004] [Revised: 11/15/2004] [Accepted: 11/16/2004] [Indexed: 10/25/2022]
Abstract
Injury to the nervous system occasionally leads to intense and persistent neuropathic pain, which is resistant to conventional analgesic methods. It was reported that electroacupuncture (EA) had potent analgesic effect on neuropathic pain by activating various endogenous transmitters such as the opioid peptides. Glial cell line-derived neurotrophic factor (GDNF) has been hypothesized to play an important role in modulation of nociceptive signals especially during neuropathic pain state. Using immunohistochemistry, Western blot, and RT-PCR analysis techniques, the present study observed the effects of EA on the expression of GDNF and GDNF family receptor alpha-1 (GFRalpha-1, the high-affinity receptor of GDNF) in neuropathic pain rats. The results showed that both protein and mRNA levels of GDNF and GFRalpha-1 in the dorsal root ganglions (DRG), as well as GDNF protein in the spinal dorsal horn, were significantly increased after chronic constriction injury (CCI) of the rats' sciatic nerve and could be further enhanced by EA treatment. The present data demonstrated that EA could activate endogenous GDNF and GFRalpha-1 system of neuropathic pain rats and this might underlie the effectiveness of EA in the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Zhi-Qiang Dong
- Department of Integrative Medicine and Neurobiology, Institute of Acupuncture Research, Shanghai Medical College, Fudan University, P.O. Box 291, 138 Yi Xue Yuan Road, Shanghai 200032, China
| | | | | | | | | |
Collapse
|
21
|
Eslamboli A, Cummings RM, Ridley RM, Baker HF, Muzyczka N, Burger C, Mandel RJ, Kirik D, Annett LE. Recombinant adeno-associated viral vector (rAAV) delivery of GDNF provides protection against 6-OHDA lesion in the common marmoset monkey (Callithrix jacchus). Exp Neurol 2004; 184:536-48. [PMID: 14637123 DOI: 10.1016/j.expneurol.2003.08.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) has shown potential as a treatment for Parkinson's disease. Recombinant adeno-associated viral vectors expressing the GDNF protein (rAAV-GDNF) have been used in rodent models of Parkinson's disease to promote functional regeneration after 6-OHDA lesions of the nigrostriatal system. The goal of the present study was to assess the anatomical and functional efficacy of rAAV-GDNF in the common marmoset monkey (Callithrix jacchus). rAAV-GDNF was injected into the striatum and substantia nigra 4 weeks prior to a unilateral 6-OHDA lesion of the nigrostriatal bundle. Forty percent of the dopamine cells in the lesioned substantia nigra of the rAAV-GDNF-treated monkeys survived, compared with 21% in the untreated monkeys. Fine dopaminergic fibres were observed microscopically in the injected striatum of some rAAV-GDNF-treated monkeys, suggesting that rAAV-GDNF treatment may have prevented, at least in part, the loss of dopaminergic innervation of the striatum. Protection of dopamine cells and striatal fibre innervation was associated with amelioration of the lesion-induced behavioural deficits. rAAV-GDNF-treated monkeys showed partial or complete protection not only in the amphetamine and apomorphine rotation but also in head position and the parkinsonian disability rating scale. Therefore, our study provides evidence for the behavioural and anatomical efficacy of GDNF delivered via an rAAV vector as a possible treatment for Parkinson's disease.
Collapse
Affiliation(s)
- Andisheh Eslamboli
- Department of Experimental Psychology, Downing Street, University of Cambridge, CB2 3EB, Cambridge, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Alberch J, Pérez-Navarro E, Canals JM. Neurotrophic factors in Huntington's disease. PROGRESS IN BRAIN RESEARCH 2004; 146:195-229. [PMID: 14699966 DOI: 10.1016/s0079-6123(03)46014-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Huntington's disease is a neurodegenerative disorder characterized by the selective loss of striatal neurons and, to a lesser extent, cortical neurons. The neurodegenerative process is caused by the mutation of huntingtin gene. Recent studies have established a link between mutant huntingtin, excitotoxicity and neurotrophic factors. Neurotrophic factors prevent cell death in degenerative processes but they can also enhance growth and function of neurons that are affected in Huntington's disease. The endogenous regulation of the expression of neurotrophic factors and their receptors in the striatum and its connections can be important to protect striatal cells and maintains basal ganglia connectivity. The administration of exogenous neurotrophic factors, in animal models of Huntington's disease, has been used to characterize the trophic requirements of striatal and cortical neurons. Neurotrophins, glial cell line-derived neurotrophic factor family members and ciliary neurotrophic factor have shown a potent neuroprotective effects on different neuronal populations of the striatum. Furthermore, they are also useful to maintain the integrity of the corticostriatal pathway. Thus, these neurotrophic factors may be suitable for the development of a neuroprotective therapy for neurodegenerative disorders of the basal ganglia.
Collapse
Affiliation(s)
- Jordi Alberch
- Department of Cell Biology and Pathology, Medical School, IDIBAPS, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain.
| | | | | |
Collapse
|
23
|
Koyama Y, Tsujikawa K, Matsuda T, Baba A. Intracerebroventricular administration of an endothelin ETB receptor agonist increases expressions of GDNF and BDNF in rat brain. Eur J Neurosci 2003; 18:887-94. [PMID: 12925014 DOI: 10.1046/j.1460-9568.2003.02797.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Endothelins (ETs) are suggested to be involved in functional alterations of astrocytes after brain injury, including proliferation, hypertrophy and production of neurotrophic factors. In this study, effects of Ala1,3,11,15-endothelin-1 (Ala1,3,11,15-ET-1), an ETB receptor selective agonist, on neurotrophic factor production were examined in rat brain. A continuous intracerebroventricular administration of Ala1,3,11,15-ET-1 (500 pmol/day for 7 days) increased the numbers of GFAP- and vimentin-positive astrocytes in the hippocampus, caudate putamen and cerebrum. Ala1,3,11,15-ET-1 did not induce neuronal degeneration and activation of microglia/macrophage in these brain regions. The intracerebroventricular administration of Ala1,3,11,15-ET-1 for 7 days caused two- to three-fold increases in glial cell line-derived neurotrophic factors (GDNF) mRNA in the hippocampus and cerebrum. The mRNA levels of brain-derived neurotrophic factors (BDNF) in caudate putamen were increased by Ala1,3,11,15-ET-1. Expressions of nerve growth factor (NGF) and basic fibroblast growth factor (bFGF) mRNA in these regions were not largely affected by Ala1,3,11,15-ET-1, except cerebral NGF mRNA level was increased. The Ala1,3,11,15-ET-1-induced increases in GDNF and BDNF mRNA levels were accompanied by increases in immunoreactive GDNF and BDNF. Immunohistochemical observations showed that GFAP-positive astrocytes expressed GDNF and BDNF in the brain regions of Ala1,3,11,15-ET-1-infused rats. In cultured rat astrocytes, Ala1,3,11,15-ET-1 (100 nm) increased mRNA levels of GDNF and BDNF. These results suggest that activation of brain ETB receptors induced GDNF and BDNF expression in astrocytes.
Collapse
Affiliation(s)
- Yutaka Koyama
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-Oka 1-6 Suita, Osaka 565-0871, Japan
| | | | | | | |
Collapse
|
24
|
Fang M, Wang Y, He QH, Sun YX, Deng LB, Wang XM, Han JS. Glial cell line-derived neurotrophic factor contributes to delayed inflammatory hyperalgesia in adjuvant rat pain model. Neuroscience 2003; 117:503-12. [PMID: 12617957 DOI: 10.1016/s0306-4522(02)00958-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Neurotrophic factors, such as nerve growth factor and brain-derived neurotrophic factor, are members of the structurally related neurotrophin family that play important roles in pain modulation. Although there are also indications for the involvement of glial cell line-derived neurotrophic factor (GDNF), it is unclear whether and how GDNF is involved in inflammatory pain. In the present study, we studied the expression pattern of GDNF in dorsal root ganglia (DRG) and spinal cord, using confocal microscopy. We demonstrate that GDNF is well associated with nonpeptidergic pain pathway and that GDNF could possibly be anterogradely transported from DRG neurons to superficial spinal cord dorsal horn. We also studied the dynamic changes of GDNF expression in rats during chronic inflammation using injection of complete Freund's adjuvant as a model of chronic pain. We found that GDNF was down-regulated in both dorsal root ganglia and spinal cords 2 weeks after arthritis induction. To assess the impact of this down-regulation on pain transmission, we used a function-blocking antibody against GDNF delivered intrathecally in the same chronic-pain animal models. Injection of this antibody to GDNF produced no immediate effect, but decreased the delayed, bilateral hyperalgesia induced from a unilateral injection of complete Freund's adjuvant. The effect of this antibody coincided with the down-regulation of GDNF immunoreactivity in response to inflammation, suggesting that GDNF supports biochemical changes that contribute to hyperalgesia.
Collapse
Affiliation(s)
- M Fang
- Neuroscience Research Institute, Peking University Health Science Center, Beijing 100083, PR China
| | | | | | | | | | | | | |
Collapse
|
25
|
Nomura H, Furuta A, Iwaki T. Dorsal root rupture injury induces extension of astrocytic processes into the peripheral nervous system and expression of GDNF in astrocytes. Brain Res 2002; 950:21-30. [PMID: 12231225 DOI: 10.1016/s0006-8993(02)02982-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Preganglionic brachial plexus injuries fall into two categories according to the lesion site, root avulsion injury and root rupture injury. The latter type of injury involves part of the peripheral nervous system (PNS) component at the injured spinal cord surface. Previous investigators have used rhizotomy of experimental animals as a model for dorsal root rupture injury. However, the effect on the central nervous system (CNS)-PNS junction accompanied by the mechanical stress from traction force is hard to estimate in this model. The current study aimed to demonstrate temporal molecular alterations from the CNS-PNS junction to the ruptured dorsal root after traction injury by immunohistochemical procedures. At 28 days after dorsal rupture injury, GFAP-positive structures could be clearly identified showing rather straight lines from the centro-peripheral junction toward the peripheral stump in the ruptured dorsal root. Immunoelectron microscopy for GFAP verified GFAP IR within the astrocytic processes at the injured dorsal root at 28 days after dorsal rupture injury. Glial cell line-derived neurotrophic factor immunoreactivity (GDNF IR) was slightly upregulated within the Schwann cell bodies on the injured dorsal root at 24-48 h after rupture injury. However, GDNF IR had appeared showing a process-like profile on the ruptured dorsal root by 28 days, and it was closely related with GFAP-positive structures. In contrast, a small increase in GFAP IR was only detected on the proximal side on the rhizotomized dorsal root at 28 days after rhizotomy. A marked decrease in NF IR and S-100 IR was observed at the ruptured dorsal root from 7 days. On the other hand, laminin IR was strongly upregulated on the ruptured dorsal root from 48 h to 7 days, and was still evident at 28 days. We therefore conclude that the astrocytes show a unique ability to extend their processes toward the stump. This ability may provide a new medium for the study of axonal regeneration in future clinical experiments.
Collapse
Affiliation(s)
- Hiroshi Nomura
- Department of Neuropathology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 812-8582, Fukuoka, Japan.
| | | | | |
Collapse
|
26
|
Pina Serra M, Quartu M, Ambu R, Follesa P, Del Fiacco M. Immunohistochemical localization of GDNF in the human hippocampal formation from prenatal life to adulthood. Brain Res 2002; 928:138-46. [PMID: 11844480 DOI: 10.1016/s0006-8993(01)03377-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In this study, we examined the immunohistochemical occurrence and distribution of glial cell line-derived neurotrophic factor (GDNF) in autoptic specimens of normal human hippocampus at different ages, from 22 weeks of gestation (w.g.) to adult life. Two different anti-GDNF polyclonal antibodies were used. Western blot analysis on homogenates of human and rat brain and recombinant human GDNF resulted in differential detection of monomeric and dimeric forms of the proteins. The ABC immunohistochemical technique revealed that in the Ammon's horn, numerous positive cell bodies occurred in the pyramidal layer, the majority of them being present in the proximal CA1 and in CA2. Sparse positive neurons could be observed in the stratum oriens and moleculare. In the fascia dentata many granule cells showed a light punctate staining, whereas more heavily labelled neurons occurred in the polymorphic layer and, occasionally, in the molecular layer. The distribution pattern of GDNF-like immunoreactivity appeared consistently similar throughout life stages from 29 w.g. to adult age. However, intensity of labelling and frequency of neuronal cell bodies was highest in the neonate and decreased in adulthood. The present data provide a comprehensive map of the localization of GDNF-like immunoreactive neurons in the human archicortex at developmental ages and in the mature tissue and represent a first step towards the identification of hippocampal neurons which express the protein and/or are responsive to it. They further suggest that GDNF may play a role in the development of intrahippocampal circuitry and in neuronal function and maintenance throughout life.
Collapse
Affiliation(s)
- Maria Pina Serra
- Department of Cytomorphology, University of Cagliari, Cittadella Universitaria di Monserrato, 09042 Monserrat, Italy
| | | | | | | | | |
Collapse
|
27
|
Del Fiacco M, Quartu M, Serra MP, Follesa P, Lai ML, Bachis A. Topographical localization of glial cell line-derived neurotrophic factor in the human brain stem: an immunohistochemical study of prenatal, neonatal and adult brains. J Chem Neuroanat 2002; 23:29-48. [PMID: 11756008 DOI: 10.1016/s0891-0618(01)00139-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
As a step towards the identification of the neuronal populations responsive to glial cell line-derived neurotrophic factor (GDNF) in the human nervous system and their changes with age, this study reports on the immunohistochemical localization of the protein GDNF in the autoptic normal human brain stem of pre- and full-term newborns and adult subjects. Two different anti-GDNF polyclonal antibodies were used. Western blot analysis on homogenates of human and rat brain and recombinant human GDNF resulted in differential detection of monomeric and dimeric forms of the proteins. The ABC immunohistochemical technique on cryostat tissue sections showed an uneven distribution of GDNF-like immunoreactive nerve fibers and terminals and neuronal cell bodies. Immunoreactive elements were mainly localized to the spinal trigeminal, cuneate, solitary, vestibular, and cochlear sensory nuclei, dorsal motor nucleus of the vagus nerve, ventral grey column, hypoglossal nucleus, dorsal and ventrolateral medullary reticular formation, pontine subventricular grey and locus coeruleus, lateral regions of the rostral pontine tegmentum, tectal plate, trochlear nucleus, dorsal and median raphe nuclei, caudal and rostral linear nuclei, cuneiform nucleus, and substantia nigra. Comparison between pre- and full-term newborns and adult subjects revealed changes with age in density of positive innervation and frequency of immunoreactive perikarya. The results obtained provide detailed information on the occurrence of GDNF-like immunoreactive neurons in the human brain stem and suggest that the protein is present in a variety of neuronal systems, which subserve different functional activities, at developmental ages and in adult brains.
Collapse
Affiliation(s)
- Marina Del Fiacco
- Department of Cytomorphology, University of Cagliari, Cittadella Universitaria di Monserrato, 09042 Monserrato, Italy.
| | | | | | | | | | | |
Collapse
|
28
|
Koo H, Choi BH. Expression of glial cell line-derived neurotrophic factor (GDNF) in the developing human fetal brain. Int J Dev Neurosci 2001; 19:549-58. [PMID: 11600317 DOI: 10.1016/s0736-5748(01)00042-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
GDNF expression was examined immunocytochemically in developing human fetal brains obtained from aborted fetuses ranging from 7 to 39 weeks in gestational age. At 7-8 weeks, strong immunoreactivity was noted within radial glial processes, glia limitans and choroid plexus of the telencephalic vesicle. By 10 weeks, ependymal cells, primitive matrix cells and early developing cortical plate neurons showed positive staining. By 15-16 weeks, migrating neurons in the subventricular and intermediate zones and in the cortical plate were strongly positive for GDNF. The glia limitans of the cerebral cortex and subependymal astrocytes remained positive at this time. As fetal age increased, GDNF expression shifted to neurons and glial cells in the deeper structures of the brain. The most prominent GDNF staining was observed in the cytoplasm and dendrites of Purkinje cells of the cerebellum by 25 weeks and thereafter. Pyramidal neurons of the CA1 region and granule cells of the dentate fascia of the hippocampus, neurons of the entorhinal cortex, and scattered neurons within the brain stem, medulla and spinal cord all showed strong GDNF staining by 25-35 weeks. Widespread GDNF expression in neuronal and non-neuronal cells with distinct developmental shifts suggests that GDNF may play a critical role in the survival, differentiation and maintenance of neurons at different stages of development in the developing human fetal brain.
Collapse
Affiliation(s)
- H Koo
- Department of Pathology, College of Medicine, Ewha Womans University, and Ewha Medical Research Center, Seoul, South
| | | |
Collapse
|