1
|
GM1 ganglioside prevents axonal regeneration inhibition and cognitive deficits in a mouse model of traumatic brain injury. Sci Rep 2018; 8:13340. [PMID: 30190579 PMCID: PMC6127193 DOI: 10.1038/s41598-018-31623-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/13/2018] [Indexed: 11/08/2022] Open
Abstract
Traumatic Brain Injury (TBI) is one of the most common causes of neurological damage in young populations. It has been previously suggested that one of the mechanisms that underlie brain injury is Axonal Outgrowth Inhibition (AOI) that is caused by altered composition of the gangliosides on the axon surface. In the present study, we have found a significant reduction of GM1 ganglioside levels in the cortex in a closed head traumatic brain injury model of a mouse, induced by a weight drop device. In addition, axonal regeneration in the brains of the injured mice was affected as seen by the expression of the axonal marker pNF-H and the growth cones (visualized by F-actin and β-III-tubulin). NeuN immunostaining revealed mTBI-induced damage to neuronal survival. Finally, as expected, spatial and visual memories (measured by the Y-maze and the Novel Object Recognition tests, respectively) were also damaged 7 and 30 days post injury. A single low dose of GM1 shortly after the injury (2 mg/kg; IP) prevented all of the deficits mentioned above. These results reveal additional insights into the neuroprotective characteristics of GM1 in prevention of biochemical, cellular and cognitive changes caused by trauma, and may suggest a potential intervention for mTBI.
Collapse
|
2
|
Cell Transplantation for Spinal Cord Injury: Tumorigenicity of Induced Pluripotent Stem Cell-Derived Neural Stem/Progenitor Cells. Stem Cells Int 2018. [PMID: 29535771 PMCID: PMC5817265 DOI: 10.1155/2018/5653787] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury (SCI) is an intractable and worldwide difficult medical challenge with limited treatments. Neural stem/progenitor cell (NS/PC) transplantation derived from fetal tissues or embryonic stem cells (ESCs) has demonstrated therapeutic effects via replacement of lost neurons and severed axons and creation of permissive microenvironment to promote repair of spinal cord and axon regeneration but causes ethnical concerns and immunological rejections as well. Thus, the implementation of induced pluripotent stem cells (iPSCs), which can be generated from adult somatic cells and differentiated into NS/PCs, provides an effective alternation in the treatment of SCI. However, as researches further deepen, there is accumulating evidence that the use of iPSC-derived NS/PCs shows mounting concerns of safety, especially the tumorigenicity. This review discusses the tumorigenicity of iPSC-derived NS/PCs focusing on the two different routes of tumorigenicity (teratomas and true tumors) and underlying mechanisms behind them, as well as possible solutions to circumvent them.
Collapse
|
3
|
Anthonymuthu TS, Kenny EM, Bayır H. Therapies targeting lipid peroxidation in traumatic brain injury. Brain Res 2016; 1640:57-76. [PMID: 26872597 PMCID: PMC4870119 DOI: 10.1016/j.brainres.2016.02.006] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/01/2016] [Accepted: 02/02/2016] [Indexed: 02/06/2023]
Abstract
Lipid peroxidation can be broadly defined as the process of inserting a hydroperoxy group into a lipid. Polyunsaturated fatty acids present in the phospholipids are often the targets for peroxidation. Phospholipids are indispensable for normal structure of membranes. The other important function of phospholipids stems from their role as a source of lipid mediators - oxygenated free fatty acids that are derived from lipid peroxidation. In the CNS, excessive accumulation of either oxidized phospholipids or oxygenated free fatty acids may be associated with damage occurring during acute brain injury and subsequent inflammatory responses. There is a growing body of evidence that lipid peroxidation occurs after severe traumatic brain injury in humans and correlates with the injury severity and mortality. Identification of the products and sources of lipid peroxidation and its enzymatic or non-enzymatic nature is essential for the design of mechanism-based therapies. Recent progress in mass spectrometry-based lipidomics/oxidative lipidomics offers remarkable opportunities for quantitative characterization of lipid peroxidation products, providing guidance for targeted development of specific therapeutic modalities. In this review, we critically evaluate previous attempts to use non-specific antioxidants as neuroprotectors and emphasize new approaches based on recent breakthroughs in understanding of enzymatic mechanisms of lipid peroxidation associated with specific death pathways, particularly apoptosis. We also emphasize the role of different phospholipases (calcium-dependent and -independent) in hydrolysis of peroxidized phospholipids and generation of pro- and anti-inflammatory lipid mediators. This article is part of a Special Issue entitled SI:Brain injury and recovery.
Collapse
Affiliation(s)
- Tamil Selvan Anthonymuthu
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA 15219, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Elizabeth Megan Kenny
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA 15219, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Hülya Bayır
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15219, USA; Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA 15219, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15260, USA; Childrens׳s Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA 15224, USA.
| |
Collapse
|
4
|
Dunbar GL, Sandstrom MI, Rossignol J, Lescaudron L. Neurotrophic Enhancers as Therapy for Behavioral Deficits in Rodent Models of Huntington's Disease: Use of Gangliosides, Substituted Pyrimidines, and Mesenchymal Stem Cells. ACTA ACUST UNITED AC 2016; 5:63-79. [PMID: 16801683 DOI: 10.1177/1534582306289367] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The interest in using neurotrophic factors as potential treatments for neurodegenerative disorders, such as Huntington's disease, has grown in the past decade. A major impediment for the clinical utility of neurotrophic factors is their inability to cross the blood-brain barrier in therapeutically significant amounts. Although several novel mechanisms for delivering exogenous neurotrophins to the brain have been developed, most of them involve invasive procedures or present significant risks. One approach to circumventing these problems is using therapeutic agents that can be administered systemically and have the ability to enhance the activity of neurotrophic factors. This review highlights the use of gangliosides, substituted pyrimidines, and mesenchymal stem cells as neurotrophic enhancers that have significant therapeutic potential while avoiding the pitfalls of delivering exogenous neurotrophic factors through the blood-brain barrier. The review focuses on the potential of these neurotrophic enhancers for treating the behavioral deficits in rodent models of Huntington's disease.
Collapse
|
5
|
Fujimura M, Usuki F. Methylmercury causes neuronal cell death through the suppression of the TrkA pathway: In vitro and in vivo effects of TrkA pathway activators. Toxicol Appl Pharmacol 2015; 282:259-66. [DOI: 10.1016/j.taap.2014.12.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/11/2014] [Accepted: 12/15/2014] [Indexed: 10/24/2022]
|
6
|
Silva NA, Sousa N, Reis RL, Salgado AJ. From basics to clinical: a comprehensive review on spinal cord injury. Prog Neurobiol 2013; 114:25-57. [PMID: 24269804 DOI: 10.1016/j.pneurobio.2013.11.002] [Citation(s) in RCA: 555] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 11/12/2013] [Accepted: 11/12/2013] [Indexed: 12/15/2022]
Abstract
Spinal cord injury (SCI) is a devastating neurological disorder that affects thousands of individuals each year. Over the past decades an enormous progress has been made in our understanding of the molecular and cellular events generated by SCI, providing insights into crucial mechanisms that contribute to tissue damage and regenerative failure of injured neurons. Current treatment options for SCI include the use of high dose methylprednisolone, surgical interventions to stabilize and decompress the spinal cord, and rehabilitative care. Nonetheless, SCI is still a harmful condition for which there is yet no cure. Cellular, molecular, rehabilitative training and combinatorial therapies have shown promising results in animal models. Nevertheless, work remains to be done to ascertain whether any of these therapies can safely improve patient's condition after human SCI. This review provides an extensive overview of SCI research, as well as its clinical component. It starts covering areas from physiology and anatomy of the spinal cord, neuropathology of the SCI, current clinical options, neuronal plasticity after SCI, animal models and techniques to assess recovery, focusing the subsequent discussion on a variety of promising neuroprotective, cell-based and combinatorial therapeutic approaches that have recently moved, or are close, to clinical testing.
Collapse
Affiliation(s)
- Nuno A Silva
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4806-909 Caldas das Taipas, Guimarães, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
7
|
Varma AK, Das A, Wallace G, Barry J, Vertegel AA, Ray SK, Banik NL. Spinal cord injury: a review of current therapy, future treatments, and basic science frontiers. Neurochem Res 2013; 38:895-905. [PMID: 23462880 DOI: 10.1007/s11064-013-0991-6] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 01/17/2013] [Accepted: 01/29/2013] [Indexed: 12/12/2022]
Abstract
The incidence of acute and chronic spinal cord injury (SCI) in the United States is more than 10,000 per year, resulting in 720 cases per million persons enduring permanent disability each year. The economic impact of SCI is estimated to be more than 4 billion dollars annually. Preclinical studies, case reports, and small clinical trials suggest that early treatment may improve neurological recovery. To date, no proven therapeutic modality exists that has demonstrated a positive effect on neurological outcome. Emerging data from recent preclinical and clinical studies offer hope for this devastating condition. This review gives an overview of current basic research and clinical studies for the treatment of SCI.
Collapse
Affiliation(s)
- Abhay K Varma
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | | | | | | | | | | | |
Collapse
|
8
|
Chen X, Wang R, Yin Y, Røe O, Li J, Zhu L, Guo R, Wu T, Shu Y. The effect of monosialotetrahexosylganglioside (GM1) in prevention of oxaliplatin induced neurotoxicity: A retrospective study. Biomed Pharmacother 2012; 66:279-84. [DOI: 10.1016/j.biopha.2012.01.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2011] [Accepted: 01/04/2012] [Indexed: 11/28/2022] Open
|
9
|
She JQ, Wang M, Zhu DM, Tang M, Chen JT, Wang L, Ruan DY. Monosialoanglioside (GM1) prevents lead-induced neurotoxicity on long-term potentiation, SOD activity, MDA levels, and intracellular calcium levels of hippocampus in rats. Naunyn Schmiedebergs Arch Pharmacol 2008; 379:517-24. [DOI: 10.1007/s00210-008-0379-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Accepted: 11/11/2008] [Indexed: 10/21/2022]
|
10
|
Kakinoki B, Sekimoto S, Yuki S, Ohgami T, Sejima M, Yamagami K, Saito KI. Orally active neurotrophin-enhancing agent protects against dysfunctions of the peripheral nerves in hyperglycemic animals. Diabetes 2006; 55:616-21. [PMID: 16505223 DOI: 10.2337/diabetes.55.03.06.db05-1091] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Biological substances with neurotrophic activities, such as nerve growth factor (NGF) and monosialoganglioside GM1, have been considered as agents for diabetic peripheral neuropathy. Because recent studies have suggested that decreased availability of these substances might contribute to the pathogenesis of diabetic peripheral neuropathy, some clinical trials of NGF for diabetic peripheral neuropathy have been conducted and have led to mixed conclusions. The major reasons were its limited delivery to the nervous system and adverse effects induced by subcutaneous injection, which was necessary because NGF is a polypeptide. The current study investigates whether an orally active sialic acid derivative, MCC-257, has neuroprotective properties in diabetic peripheral nerves. MCC-257 augmented NGF activity in cultured dorsal root ganglia and PC12 (pheochromocytoma 12) cells. Treatment with MCC-257 elevated NGF levels in the sciatic nerve, accompanied by improvement in nerve conduction velocity in streptozotocin-induced diabetic animals. More importantly, MCC-257 ameliorated small fiber dysfunctions, including thermal hypoalgesia, substance P content, and histopathological innervation in the plantar skin of diabetic animals. Thus, the orally active neurotrophin enhancer provides a new option for the clinical treatment of diabetic peripheral neuropathy.
Collapse
Affiliation(s)
- Bunpei Kakinoki
- Research Laboratory I, Pharmaceutical Research Unit, Research and Development Division, Mitsubishi Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan.
| | | | | | | | | | | | | |
Collapse
|
11
|
She JQ, Wang M, Zhu DM, Sun LG, Ruan DY. Effect of ganglioside on synaptic plasticity of hippocampus in lead-exposed rats in vivo. Brain Res 2005; 1060:162-9. [PMID: 16229825 DOI: 10.1016/j.brainres.2005.08.044] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2005] [Revised: 08/29/2005] [Accepted: 08/30/2005] [Indexed: 10/25/2022]
Abstract
Synaptic plasticity, including long-term potentiation (LTP), long-term depression (LTD) and depotentiation (DP), is important for learning and memory. Previous studies proved that chronic lead exposure especially during early post-natal development induced impairment on synapse plasticity. The purpose of this study is to evaluate the effect of ganglioside on the lead-induced impairments of LTP and DP in rat dentate gyrus in vivo. The experiments were carried out in three groups of rats (control, lead-exposed, ganglioside-treated lead-exposed, respectively). The input-output (I/O) function, pair pulses reaction, excitatory post-synaptic potential (EPSP) and population spike (PS) amplitude were measured in the dentate gyrus (DG) of adult rats (70-90 days) in response to stimulation applied to the lateral perforant path. The results show that (1) chronic lead exposure impaired LTP/DP measured on both EPSP slope and PS amplitude in DG area of the hippocampus. (2) The amplitudes of LTP/DP of lead-exposed group were significantly increased by supplying ganglioside. These results suggest intraperitoneally injection with ganglioside could reverse the lead-induced impairments of synaptic plasticity in rats and might be effective in attenuating the cognitive deficits induced by lead.
Collapse
Affiliation(s)
- Jia-Qi She
- School of Life Science and Institute of Polar Environment, University of Science and Technology of China, Hefei, Anhui 230027, PR China
| | | | | | | | | |
Collapse
|
12
|
|
13
|
Rabin SJ, Bachis A, Mocchetti I. Gangliosides activate Trk receptors by inducing the release of neurotrophins. J Biol Chem 2002; 277:49466-72. [PMID: 12388556 DOI: 10.1074/jbc.m203240200] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We used NIH-3T3 fibroblasts expressing the different Trk receptors to examine whether GM1 ganglioside and its semisynthetic derivative LIGA20 activate various neurotrophin receptors. GM1 induced autophosphorylation of TrkC more potently than TrkA or TrkB receptors. In contrast, LIGA20 activated TrkB tyrosine phosphorylation only. Therefore, Scatchard analysis was performed to determine whether GM1 binds to TrkC. GM1 failed to displace neurotrophin-3 binding, suggesting that this ganglioside does not act as a ligand for Trk receptors. In addition, GM1 failed to induce autophosphorylation of a chimeric receptor consisting of the extracellular domain of the tumor necrosis factor receptor and the intracellular domain of TrkA, suggesting that GM1 does not affect the tyrosine kinase domain. We next determined whether GM1 induces the release of neurotrophins from fibroblast cells. GM1 induced a rapid and significant increase in the amount of neurotrophin-3, but not other neurotrophins. This effect was independent of the presence of Trk because K252a did not prevent GM1-mediated release of neurotrophin-3. Moreover, GM1-mediated TrkC autophosphorylation was blocked by TrkC-IgG (but not TrkB-IgG) receptor bodies, further suggesting that GM1 activates TrkC by inducing the release of neurotrophin-3. This hypothesis was also tested in cultured cerebellar granule cells. GM1 induced neurotrophin-3 (but not brain-derived neurotrophic factor or nerve growth factor) release. In contrast, LIGA20 increased the secretion of brain-derived neurotrophic factor. Our data show that gangliosides may activate different Trk receptors by differentially affecting the release of neurotrophins.
Collapse
Affiliation(s)
- Stuart J Rabin
- Department of Neuroscience, Georgetown University Medical Center, Washington, D. C. 20057, USA
| | | | | |
Collapse
|
14
|
Abstract
STUDY DESIGN Randomized, double-blind, sequential, multicenter clinical trial of two doses of Sygen versus placebo. OBJECTIVES To determine efficacy and safety of Sygen in acute spinal cord injury. SUMMARY OF BACKGROUND DATA An earlier, single-center trial in 28 patients showed an improvement (50.0% vs. 7.1%, P = 0.034) in marked recovery with Sygen. METHODS Standard clinical trial techniques. RESULTS The prospectively planned analysis at the prespecified endpoint time for all patients was negative. There was a significant effect in all patients in the primary outcome variable (the percentage of marked recovery) at week 8, the end of the dosing period. There was a significant effect in all patients in the time at which marked recovery is first achieved. Restricted to severity Group B, which has small sample size, the primary efficacy analysis showed a trend but did not reach significance. There is a large, consistent and, at some time points, significant effect in the primary outcome variable in the nonoperated patients through week 26. The American Spinal Injury Association motor, light touch, and pinprick scores showed a consistent trend in favor of Sygen, as also did bowel function, bladder function, sacral sensation, and anal contraction. The less severely injured patients appeared to have a greater beneficial drug effect. Evidence against an effect of Sygen was minimal and scattered. CONCLUSIONS Although not proven in the primary efficacy analysis of this trial, Sygen appears to be beneficial in patients with severe spinal cord injury.
Collapse
Affiliation(s)
- F H Geisler
- Medical Group, S.C., Chicago Institute of Neurosurgery and Neuroresearch, 2515 North Clark Street, Suite 800, Chicago, Illinois 60614, USA.
| | | | | | | |
Collapse
|
15
|
Abstract
The search for a pharmacologic treatment of acute spinal cord injury (SCI) dates back to the 1960s. It was not until 1990 that the pharmacologic agent methylprednisolone demonstrated improved outcomes in humans. Methylprednisolone has shown superiority to placebo in humans in two large, multicenter trials, and is the standard of care thus far. Other potentially useful agents include tirilazad, ganglioside (GM-1), and naloxone. Additional studies are needed for these agents to determine the optimal dose and timing of administration.
Collapse
Affiliation(s)
- E C Seidl
- Department of Pharmacy, Allegheny General Hospital, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
16
|
Panni MK, Cooper JD, Sofroniew MV. Ganglioside GM1 potentiates NGF action on axotomised medial septal cholinergic neurons. Brain Res 1998; 812:76-80. [PMID: 9813248 DOI: 10.1016/s0006-8993(98)00948-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Transection of the fimbria fornix leads to retrograde degeneration of axotomised septal cholinergic neurons as manifested by loss of choline acetyltransferase and p75NGFR immunoreactivity. Intracerebroventricularly administered nerve growth factor initiated at the time of axotomy can prevent these changes. We have shown that concurrent intraperitoneal administration of GM1 with a low and otherwise unprotective intracerebroventricular dose of nerve growth factor, can also prevent the loss of these fimbria fornix axotomised cholinergic neurons, where GM1 alone does not have this effect. This study further confirms the neuroprotective actions of GM1 and suggests that it may interact to potentiate the effect of nerve growth factor on these axotomised septal cholinergic neurons.
Collapse
Affiliation(s)
- M K Panni
- Medical Research Council Cambridge Centre for Brain Repair, Department of Anatomy, University of Cambridge, England, UK.
| | | | | |
Collapse
|
17
|
Abstract
Spinal cord injury remains with limited natural recovery and only a few general ineffective treatment options. Recent publications have reported enhanced neurologic recovery with the use of methylprednisolone and GM-1 ganglioside. The results of the Maryland GM-1 Ganglioside Study reported a significant drug effect with respect to the fraction of patients that had a change of two or more Frankel grades from entrance into the study to 1-year follow-up. This study formed the basis for the currently ongoing larger placebo-controlled multicentered study using Sygen GM-1 following acute spinal cord injury. This study has entered 797 patients and is expected to present results in early 1998.
Collapse
Affiliation(s)
- F H Geisler
- Chicago Institute of Neurosurgery and Neuroresearch, Illinois 60614, USA
| |
Collapse
|
18
|
Miyazaki H, Fukumoto S, Okada M, Hasegawa T, Furukawa K. Expression cloning of rat cDNA encoding UDP-galactose:GD2 beta1,3-galactosyltransferase that determines the expression of GD1b/GM1/GA1. J Biol Chem 1997; 272:24794-9. [PMID: 9312075 DOI: 10.1074/jbc.272.40.24794] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Using an anti-GD1b monoclonal antibody, expression cloning of a cDNA for the beta1,3-galactosyltransferase gene (EC 2.4.1.62) was performed. KF4C, mouse melanoma B16 transfected with polyoma T antigen gene, and GM2/GD2 synthase cDNA was used as a recipient cell line for the cDNA library transfection. A cDNA clone of GD3 synthase, pD3T-31 was co-transfected with a cDNA library prepared from rat brain RNA using the pcDNAI expression vector. The isolated cDNA clone pM1T-9 predicted a type II membrane protein with 4 amino acids of cytoplasmic domain, 21 amino acids of transmembrane region, and a large catalytic domain with 346 amino acids. Introduction of the cDNA clone into a mouse melanoma line B16 previously transfected with a GM2/GD2 synthase gene resulted in the neo-synthesis of GM1. Co-transfection of the cell line with pM1T-9 and a GD3 synthase cDNA resulted in the expression of GD1b as well as GM1. Moreover, introduction of pM1T-9 into L cell (lacking GM3 synthase), previously transfected with GM2/GD2 synthase gene, resulted in the definite expression of asialo-GM1. These results indicated that GD1b/GM1/GA1 synthases were identical, as previously suggested based on enzymological analysis. In Northern blots of the beta1, 3-galactosyltransferase gene with total RNA from various rat tissues, a 1.6-kilobase mRNA was strongly expressed in spleen, thymus, kidney, and testis. However, the expression level of the gene in the adult brain tissue was not especially high. On the other hand, this gene was expressed at high levels in the rat brain of embryonal day 12, and reached a peak at around birth, then fell to low level in the adult brain.
Collapse
Affiliation(s)
- H Miyazaki
- Department of Biochemistry II, Nagoya University School of Medicine, 65 Tsuramai, Nagoya 466, Japan
| | | | | | | | | |
Collapse
|
19
|
Abstract
Although at present there is no definitive treatment or cure for Alzheimer's disease, different pharmacological strategies are being actively investigated. At present, cholinergic therapy and nootropics and some neuronotrophic agents represent the available approaches to symptomatic treatment of Alzheimer's disease. The use of cholinesterase inhibitors (ChEI) constitutes the best cholinergic approach to increase acetylcholine levels. Available data suggest that about 15 to 40% of Alzheimer's disease patients show a varying degree of cognitive improvement while taking these medications; however, haematological complications (neutropenia or agranulocytosis), together with hepatotoxicity, need to be considered carefully. Recent data suggest that long term administration of nootropics may lead to a significant improvement of cognitive functions in Alzheimer's disease patients compared with untreated individuals, having excellent tolerability. Protocols for the intracerebroventricular administration of neuronotrophic substances are also ongoing. The most promising approaches for the future currently undergoing investigation involve attempts to slow the production of beta-amyloid and/or to inhibit beta-amyloid aggregation. Another rational therapeutic approach would be to inhibit the formation of paired helical filaments (PHF) by increasing and/or modulating the activities of protein phosphatases and kinases. Antioxidant therapy should disrupt or prevent the free radical/beta-amyloid recirculating cascade and the progressive neurodegeneration. Idebenone, a synthetic compound acting as an 'electron trapper' and free radical scavenger, has shown some efficacy in degenerative and vascular dementia; at present, other different molecules having antioxidative properties [lazaroids (21-aminosteroids), pyrrolopyrimidines, nitric oxide blockers, selegiline, some vitamins] are under investigation. Lowering absorption or brain tissue concentrations of aluminium also offers possible therapeutic opportunities for slowing the rate of clinical progression of the disease; in this sense, some evidence exists using the aluminium chelating agent deferoxamine (desferrioxamine). Inflammation also may play a significant pathogenetic role in Alzheimer's disease. As shown by several retrospective analyses, there is an inverse association of anti-inflammatory drug use with the frequency of Alzheimer's disease diagnosis. Consequently, clinical trials using both nonsteroidal and steroidal molecules have been proposed. These lines of pharmacological intervention represent an important premise for future therapeutic strategies capable of counteracting the pathogenesis of Alzheimer's disease.
Collapse
|
20
|
Silva RH, Felicio LF, Nasello AG, Vital MA, Frussa-Filho R. Effect of ganglioside (GM1) on memory in senescent rats. Neurobiol Aging 1996; 17:583-6. [PMID: 8832633 DOI: 10.1016/0197-4580(96)00065-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Monosialoganglioside GM1 (GM1) has been found to alleviate genetic and lesion-induced memory deficits. The purpose of this study was to investigate the effects of 7-day treatment with GM1 (50 mg/kg IP) on acquisition and retention performance of senescent rats in a passive avoidance situation. Saline-treated old rats showed a decreased performance in acquisition and retention tests as compared to saline-treated adult rats. GM1 improved both acquisition and retention performance of old animals, and there was no significant difference between GM1-treated old rats and saline-treated adult rats. These data suggest that GM1 treatment can improve memory deficits in intact senescent animals.
Collapse
Affiliation(s)
- R H Silva
- Departamento de Farmacologia, Escola Paulista de Medicina, São Paulo, Brasil
| | | | | | | | | |
Collapse
|
21
|
Abstract
Traumatic injuries to the brain or spinal cord cause tissue damage, in part by initiating reactive biochemical changes. Pharmacological approaches aim to modify this delayed injury response by blocking one or more components of the reactive biochemical/metabolic cascade. This minireview summarizes both historical and recent developments in experimental and clinical treatment of CNS trauma. Potential treatments include: corticosteroids, antioxidants or free radical scavengers; drugs that modify arachidonic acid metabolism, platelet-activating factor antagonists; gangliosides; modulators of monoamine actions; opioid receptor antagonists; thyrotropin-releasing hormone and thyrotropin-releasing hormone analogues; glutamase receptor antagonists; calcium channel blockers; agents that modify the inflammatory/immune response; and trophic factors. Understanding the mechanisms of action for these compounds can permit rational drug development/application, delineation of the therapeutic window, and laying of the ground-work for evaluating potential synergistic effects of combination treatment strategies.
Collapse
Affiliation(s)
- A I Faden
- Department of Neurology, Georgetown University Medical Center, Washington, DC 20007-2197, USA
| |
Collapse
|
22
|
Money MK, Pippin GW, Weaver KE, Kirsch JP, Webster DB. Auditory brainstem responses of CBA/J mice with neonatal conductive hearing losses and treatment with GM1 ganglioside. Hear Res 1995; 87:104-13. [PMID: 8567428 DOI: 10.1016/0378-5955(95)00083-g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Exogenous administration of GM1 ganglioside to CBA/J mice with a neonatal conductive hearing loss ameliorates the atrophy of spiral ganglion neurons, ventral cochlear nucleus neurons, and ventral cochlear nucleus volume. The present investigation demonstrates the extent of a conductive loss caused by atresia and tests the hypothesis that GM1 ganglioside treatment will ameliorate the conductive hearing loss. Auditory brainstem responses were recorded from four groups of seven mice each: two groups received daily subcutaneous injections of saline (one group had normal hearing; the other had a conductive hearing loss); the other two groups received daily subcutaneous injections of GM1 ganglioside (one group had normal hearing; the other had a conductive hearing loss). In mice with a conductive loss, decreases in hearing sensitivity were greatest at high frequencies. The decreases were determined by comparing mean ABR thresholds of the conductive loss mice with those of normal hearing mice. The conductive hearing loss induced in the mice in this study was similar to that seen in humans with congenital aural atresias. GM1 ganglioside treatment had no significant effect on ABR wave I thresholds or latencies in either group.
Collapse
Affiliation(s)
- M K Money
- Louisiana State University Medical Center, Department of Otorhinolaryngology and Biocommunication Kresge Hearing Research Laboratory of the South, New Orleans 70112-2234, USA
| | | | | | | | | |
Collapse
|
23
|
Stein DG, Glasier MM, Hoffman SW. Pharmacological treatments for brain-injury repair: Progress and prognosis. Neuropsychol Rehabil 1994. [DOI: 10.1080/09602019408401605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
24
|
Young W, Kume-Kick J, Constantini S. Glucocorticoid therapy of spinal cord injury. Ann N Y Acad Sci 1994; 743:241-63; discussion 263-5. [PMID: 7802417 DOI: 10.1111/j.1749-6632.1994.tb55796.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- W Young
- Department of Neurosurgery, New York University Medical Center, New York 10016
| | | | | |
Collapse
|
25
|
Sommer BR, Cohen BM, Satlin A, Cole JO, Jandorf L, Dorsey F. Changes in tardive dyskinesia symptoms in elderly patients treated with ganglioside GM1 or placebo. J Geriatr Psychiatry Neurol 1994; 7:234-7. [PMID: 7826493 DOI: 10.1177/089198879400700408] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The unique role of ganglioside GM1 in neuronal plasticity led two centers, New York University and McLean Hospital, to study the effect of GM1 or placebo in patients with tardive dyskinesia. Results from the NYU cohort have already been published. We now present data from the entire cohort, allowing us to evaluate the effects of GM1 in the elderly compared to young adults. Subjects with tardive dyskinesia were randomly assigned to single-blind placebo injections for 1 week, followed by 1 month of double-blind intramuscular placebo or GM1 100 mg. The final sample included 29 patients: 12 younger than 55 years of age and 17 older. There was no GM1-versus-placebo difference observed in either age group, or in the total group. However, whether on placebo or GM1, repeated measures analysis of variance (RANOVA) found a significant difference in response between Abnormal Involuntary Movement Scale scores, taken baseline and week 4, in the elderly compared to young adults. Scores for the young adults show initial improvement then deterioration back to baseline, and those for the elderly show continuing improvement during the 4-week trial. The importance of the placebo effect in the elderly and its meaning for studies of GM1 in tardive dyskinesia are discussed.
Collapse
Affiliation(s)
- B R Sommer
- Department of Psychiatry, Mount Sinai School of Medicine, New York, New York
| | | | | | | | | | | |
Collapse
|
26
|
Okada Y, Rahmann H, Hirai H, Terashima A. Exogenously applied gangliosides (GM1, GD1a and Gmix) fail to facilitate the induction of long-term potentiation (LTP) in the slices of hippocampus and superior colliculus of the guinea pig. Neurosci Lett 1994; 170:269-72. [PMID: 8058199 DOI: 10.1016/0304-3940(94)90335-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
To confirm the effect of gangliosides on the facilitation of the induction of long-term potentiation (LTP), slices of hippocampus and superior colliculus from guinea pig were prepared. One group of slices was incubated in the standard medium containing gangliosides (GM1, GD1a, or a mixture of gangliosides from bovine brain, each at a concentration of 70 microM) for 2 h. The other group of slices was incubated in the standard medium only. In both groups of slices, tetanic stimulation induced LTP in a similar manner. In low Ca2+ (1 mM) medium, LTP was not induced in either group of slices. Thus, the present experiments could not confirm previous reports indicating that exogenously applied gangliosides facilitate the induction of LTP.
Collapse
Affiliation(s)
- Y Okada
- Department of Physiology, School of Medicine, Kobe University, Japan
| | | | | | | |
Collapse
|
27
|
Stull ND, Schneider JS, Iacovitti L. GM1 ganglioside partially rescues cultured dopaminergic neurons from MPP(+)-induced damage: dependence on initial damage and time of treatment. Brain Res 1994; 640:308-15. [PMID: 7911728 DOI: 10.1016/0006-8993(94)91886-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
GM1 ganglioside is believed to be important in promoting the recovery of neurons from injury. The present study assesses the ability of GM1 to repair or prevent the damage of dopamine neurons caused by the neurotoxin 1-methyl-4-phenylpyridinium (MPP+). Treatment of mesencephalic cell cultures with 2.5 microM MPP+ resulted in the loss of 30% of tyrosine hydroxylase (TH) immunoreactive neurons. In contrast, cultures administered 100 microM GM1 ganglioside for 3 days after toxin treatment contained nearly control numbers of TH+ neurons (97%). This reparative effect of GM1 was reflected in parallel increases in TH enzyme activity, dopamine and dopac levels. Cultures sustaining greater insult from higher doses of MPP+ (5.0-10.0 microM) did not benefit from ganglioside treatment, suggesting that rescue by GM1 depended on the degree of initial damage to cells. Moreover, the timing of ganglioside treatment was critical; pretreatment with GM1 alone did not prevent or attenuate the damage caused by subsequent incubation in 2.5 microM MPP+.
Collapse
Affiliation(s)
- N D Stull
- Department of Neurology, Hahnemann University, Philadelphia, PA 19102
| | | | | |
Collapse
|
28
|
Pepeu G, Oderfeld-Nowak B, Casamenti F. CNS pharmacology of gangliosides. PROGRESS IN BRAIN RESEARCH 1994; 101:327-35. [PMID: 8029462 DOI: 10.1016/s0079-6123(08)61960-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- G Pepeu
- Department of Preclinical and Clinical Pharmacology, University of Florence, Italy
| | | | | |
Collapse
|
29
|
Abstract
Gangliosides are glycosphingolipids localized to the outer leaflet of the plasma membrane of vertebrate cells. The highest ganglioside concentration of any organ is found in the mammalian brain, where the gangliosides are enriched in the neuronal membrane, particularly in the synapses. There are four major brain gangliosides with the same neutral tetrasaccharide core to which one to three sialic acids are linked--the simplest being the GM1-ganglioside. These gangliosides have been shown to have neuritogenic and neuronotrophic activity and to facilitate repair of neuronal tissue after mechanical, biochemical or toxic injuries. Mixtures of native bovine brain gangliosides were adopted for pharmacological use in the treatment of peripheral nerve damage, and GM1-ganglioside has been applied for the treatment of CNS injuries and diseases. Beneficial effects of GM1 have been documented in the treatment of stroke and spinal cord injuries, particularly when the treatment has been initiated within a few hours of the acute event. Continuous intraventricular infusion of GM1 has recently been shown to have a significant beneficial effect in Alzheimer disease of early onset (AD Type I).
Collapse
Affiliation(s)
- L Svennerholm
- Department of Clinical Neuroscience, University of Göteborg, Mölndal Hospital, Sweden
| |
Collapse
|
30
|
Siegel DA, Suzuki K. A GM1-ganglioside-binding protein in rat brain. PROGRESS IN BRAIN RESEARCH 1994; 101:149-62. [PMID: 8029447 DOI: 10.1016/s0079-6123(08)61946-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- D A Siegel
- Howard Hughes Medical Institute, Rockefeller University, New York, NY 10021
| | | |
Collapse
|
31
|
Constantini S, Young W. The effects of methylprednisolone and the ganglioside GM1 on acute spinal cord injury in rats. J Neurosurg 1994; 80:97-111. [PMID: 8271028 DOI: 10.3171/jns.1994.80.1.0097] [Citation(s) in RCA: 245] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Recent clinical trials have reported that methylprednisolone sodium succinate (MP) or the monosialic ganglioside GM1 improves neurological recovery in human spinal cord injury. Because GM1 may have additive or synergistic effects when used with MP, the authors compared MP, GM1, and MP+GM1 treatments in a graded rat spinal cord contusion model. Spinal cord injury was caused by dropping a rod weighing 10 gm from a height of 1.25, 2.5, or 5.0 cm onto the rat spinal cord at T-10, which had been exposed via laminectomy. The lesion volumes were quantified from spinal cord Na and K shifts at 24 hours after injury and the results were verified histologically in separate experiments. A single dose of MP (30 mg/kg), given 5 minutes after injury, reduced 24-hour spinal cord lesion volumes by 56% (p = 0.0052), 28% (p = 0.0065), and 13% (p > 0.05) in the three injury-severity groups, respectively, compared to similarly injured control groups treated with vehicle only. Methylprednisolone also prevented injury-induced hyponatremia and increased body weight loss in the spine-injured rats. When used alone, GM1 (10 to 30 mg/kg) had little or no effect on any measured variable compared to vehicle controls; when given concomitantly with MP, GM1 blocked the neuroprotective effects of MP. At a dose of 3 mg/kg, GM1 partially prevented MP-induced reductions in lesion volumes, while 10 to 30 mg/kg of GM1 completely blocked these effects of MP. The effects of MP on injury-induced hyponatremia and body weight loss were also blocked by GM1. Thus, GM1 antagonized both central and peripheral effects of MP in spine-injured rats. Until this interaction is clarified, the authors recommend that MP and GM1 not be used concomitantly to treat acute human spinal cord injury. Because GM1 modulates protein kinase activity, protein kinases inhibit lipocortins, and lipocortins mediate anti-inflammatory effects of glucocorticoids, it is proposed that the neuroprotective effects of MP are partially due to anti-inflammatory effects and that GM1 antagonizes the effects of MP by inhibiting lipocortin. Possible beneficial effects of GM1 reported in central nervous system injury may be related to the effects on neural recovery rather than acute injury processes.
Collapse
Affiliation(s)
- S Constantini
- Department of Neurosurgery, New York University Medical Center, New York
| | | |
Collapse
|
32
|
Abstract
On the basis of a taxonomy of functions an outline is given on potential deficits after brain lesions. The taxonomy distinguishes between "what"- and "how"-functions. Whereas for what-functions the localizing principle may apply, the how-functions being responsible for the logistics of the brain are non-locally represented. After brain injury the what- and how-functions may be differentially effected. On this basis 4 domains of functional rehabilitation can be distinguished, namely restitution and substitution of function after partly or complete losses of what-functions, activation and integration of function after alterations of activation or a disruption of functional coordination. Different strategies of functional rehabilitation are related to basic neurobiological principles which have been made responsible for restitution of function. Neuropsychological rehabilitation remains a challenge for neurobiology.
Collapse
Affiliation(s)
- N von Steinbüchel
- Institut für medizinische Psychologie, Ludwig-Maximilians-Universität, München, Germany
| | | |
Collapse
|
33
|
Dunbar GL, Lescaudron LL, Stein DG. Comparison of GM1 ganglioside, AGF2, and D-amphetamine as treatments for spatial reversal and place learning deficits following lesions of the neostriatum. Behav Brain Res 1993; 54:67-79. [PMID: 8504013 DOI: 10.1016/0166-4328(93)90049-v] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
These experiments tested the effectiveness of parenterally administered gangliosides and amphetamine as treatments for spatial learning deficits caused by bilateral lesions of the neostriatum. In Expt. 1, rats were tested postsurgically for 30 days on a shock-avoidance, spatial reversal task. Treatments of gangliosides (GM1 at 30 mg/kg, and AGF2 at 20 mg/kg and 30 mg/kg) and D-amphetamine (2 mg/kg) significantly decreased lesion-induced learning deficits on this task, while treatments of 10 mg/kg AGF2 and the combination of GM1 (30 mg/kg) and D-amphetamine (2 mg/kg) were ineffective. In Expt. 2, rats were given bilateral neostriatal lesions and treated with GM1 (30 mg/kg), AGF2 (20 mg/kg) or D-amphetamine (2 mg/kg) and tested postsurgically for 5 days on a place learning task in the Morris water maze. Only the GM1-treated rats showed a reduction in lesion-induced place learning deficits on this task. Since in both experiments, cell counts near the area of the lesion revealed no differences among any of the brain-damaged groups, it was suggested that the treatments exert their behavioral effects by biochemically activating spared neurons, independent of any ultimate effects they may have on neuronal survival.
Collapse
Affiliation(s)
- G L Dunbar
- Department of Psychology, Central Michigan University, Mt. Pleasant 48859
| | | | | |
Collapse
|
34
|
McIntosh TK. Novel pharmacologic therapies in the treatment of experimental traumatic brain injury: a review. J Neurotrauma 1993; 10:215-61. [PMID: 8258838 DOI: 10.1089/neu.1993.10.215] [Citation(s) in RCA: 197] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Delayed or secondary neuronal damage following traumatic injury to the central nervous system (CNS) may result from pathologic changes in the brain's endogenous neurochemical systems. Although the precise mechanisms mediating secondary damage are poorly understood, posttraumatic neurochemical changes may include overactivation of neurotransmitter release or re-uptake, changes in presynaptic or postsynaptic receptor binding, or the pathologic release or synthesis of endogenous "autodestructive" factors. The identification and characterization of these factors and the timing of the neurochemical cascade after CNS injury provides a window of opportunity for treatment with pharmacologic agents that modify synthesis, release, receptor binding, or physiologic activity with subsequent attenuation of neuronal damage and improvement in outcome. Over the past decade, a number of studies have suggested that modification of postinjury events through pharmacologic intervention can promote functional recovery in both a variety of animal models and clinical CNS injury. This article summarizes recent work suggesting that pharmacologic manipulation of endogenous systems by such diverse pharmacologic agents as anticholinergics, excitatory amino acid antagonists, endogenous opioid antagonists, catecholamines, serotonin antagonists, modulators of arachidonic acid, antioxidants and free radical scavengers, steroid and lipid peroxidation inhibitors, platelet activating factor antagonists, anion exchange inhibitors, magnesium, gangliosides, and calcium channel antagonists may improve functional outcome after brain injury.
Collapse
Affiliation(s)
- T K McIntosh
- Department of Surgery, University of Pennsylvania, Philadelphia
| |
Collapse
|
35
|
|
36
|
Schneider JS, Pope A, Simpson K, Taggart J, Smith MG, DiStefano L. Recovery from Experimental Parkinsonism in Primates with GM
1
Ganglioside Treatment. Science 1992. [DOI: 10.1126/science.256.5058.843] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- J. S. Schneider
- Center for Neurological Research, Department of Neurology, and Institute of Neuroscience, Hahnemann University School of Medicine, Philadelphia, PA 19102
| | - A. Pope
- Center for Neurological Research, Department of Neurology, and Institute of Neuroscience, Hahnemann University School of Medicine, Philadelphia, PA 19102
| | - K. Simpson
- Center for Neurological Research, Department of Neurology, and Institute of Neuroscience, Hahnemann University School of Medicine, Philadelphia, PA 19102
| | - J. Taggart
- Center for Neurological Research, Department of Neurology, and Institute of Neuroscience, Hahnemann University School of Medicine, Philadelphia, PA 19102
| | - M. G. Smith
- Center for Neurological Research, Department of Neurology, and Institute of Neuroscience, Hahnemann University School of Medicine, Philadelphia, PA 19102
| | - L. DiStefano
- Center for Neurological Research, Department of Neurology, and Institute of Neuroscience, Hahnemann University School of Medicine, Philadelphia, PA 19102
| |
Collapse
|
37
|
Raiteri M, Marchi M, Bonanno G, Fedele E, Versace P. Dopamine release and dopaminergic inhibition of acetylcholine release in rat striatal slices after nigro-striatal hemitransection and parenteral ganglioside administration. Eur J Pharmacol 1992; 213:17-24. [PMID: 1499655 DOI: 10.1016/0014-2999(92)90227-u] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Hemitransection of the nigro-striatal bundle in adult rats reduced [3H]dopamine ([3H]DA) uptake into striatal slices from the lesioned side to about 20% of that in the contralateral side 5 days after surgery. Spontaneous recovery of [3H]DA uptake was observed at days 8 and 15 post-lesion (42 and 67% of the unoperated side, respectively). After a short treatment (3 days) with the GM1 ganglioside inner ester (AGF2, 30 mg/kg i.p., daily, starting on day 2 after surgery) [3H]DA uptake amounted to 52% of that in the unoperated side. The electrically evoked fractional overflow of [3H]DA was increased by 500% in slices prepared from the lesioned side 5 days after injury, largely due to the reduced re-uptake by the DA axon terminals. The increase on day 5 was only about 350% in AGF2-treated animals. The DA D2 receptor antagonist, (-)-sulpiride, potentiated the stimulus-evoked overflow of [14C]acetylcholine in slices from the unoperated side prelabelled with [14C]choline. The effect of (-)-sulpiride was much reduced (by about 80%) in the lesioned striata at days 5 and 8 after surgery. Partial recovery was seen at day 15. The lesion did not modify the (-)-sulpiride effect in animals treated with AGF2 from the 2nd to the 5th day post-lesion. Thus early ganglioside administration slows the loss of endogenous dopaminergic control of acetylcholine release caused by partial hemitransection of the nigro-striatal bundle.
Collapse
Affiliation(s)
- M Raiteri
- Istituto di Farmacologia e Farmacognosia, Università degli Studi di Genova, Italy
| | | | | | | | | |
Collapse
|
38
|
Abstract
BACKGROUND AND PURPOSE Reports of improved short-term (less than 72 hours) outcome in experimental models of mechanical and ischemic central nervous system injury suggest that exogenous ganglioside administration may confer a protective effect on neural tissue. We studied the effect of the monosialoganglioside GM1 on cerebral infarction and edema in spontaneously hypertensive rats subjected to permanent focal cerebral ischemia. METHODS GM1 or normal saline was injected intramuscularly once a day for 3 days before and 30 and 120 minutes after occlusion of the right middle and common carotid arteries. Following a 24-hour survival period, the volume of infarction was measured by computer-assisted image analysis, and the extent of edema was assessed by measurements of tissue water content and hemispheric volume. RESULTS Infarct volume was similar among the GM1-treated (n = 10) and saline-treated (n = 10) rats (212 +/- 10 versus 220 +/- 13 microliters, respectively). In a second series of experiments, the brain water content and edema volume of the ischemic right hemisphere in GM1-treated rats (n = 10) did not differ from saline-treated controls (n = 10). CONCLUSIONS GM1 ganglioside does not effectively reduce cerebral infarction caused by permanent focal ischemia.
Collapse
Affiliation(s)
- S A Mayer
- Department of Neurology, Cornell University Medical College, New York, N.Y
| | | |
Collapse
|
39
|
Merlo Pich E, Grimaldi R, Zoli M, Biagini G, Solfrini V, Toffano G, Fuxe K, Agnati LF. Siagoside selectively attenuates morphological and functional striatal impairments induced by transient forebrain ischemia in rats. Stroke 1992; 23:234-41. [PMID: 1561654 DOI: 10.1161/01.str.23.2.234] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND PURPOSE Transient forebrain ischemia induced in rats by the four-vessel occlusion method is known to produce severe neural damage in the hippocampus and striatum and a behavioral syndrome the major symptom of which is a working memory deficit. Recent evidence suggests that monosialogangliosides can ameliorate postischemic symptoms. Our purpose was to study the effect of siagoside, the inner ester of GM1 ganglioside, on some behavioral and morphological impairments induced by four-vessel occlusion in rats. METHODS Rats were injected daily with 5 mg/kg i.p. siagoside starting 4 hours after the cerebral ischemia. After 14 days the rats were tested for working memory in a water T maze or scored for apomorphine-induced stereotypy. The rats were killed 21 days after the cerebral ischemia. Histological and computer-assisted morphometric analyses were performed on cresyl violet-stained brain sections, which were graded according to a neuropathologic score, and on sections stained with a monoclonal antiserum against dopamine and cyclic adenosine-3',5'-monophosphate-regulated phosphoprotein, a marker for striatal dopaminoceptive neurons. RESULTS Siagoside treatment reduced the stereotypy score induced by low doses of apomorphine and the extent of striatal lesions but did not affect the working memory deficit or the extent of hippocampal lesions. CONCLUSION Daily siagoside treatment after acute cerebral ischemia attenuates some morphological and functional deficits related to striatal damage. These effects can be interpreted as a selective protective action on striatal neural populations or as a modulatory action on neural systems involved in striatal control. These data are consistent with preliminary clinical reports showing that monosialogangliosides enhance motor recovery after acute ischemic stroke.
Collapse
Affiliation(s)
- E Merlo Pich
- Institute of Human Physiology, University of Modena, Italy
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Delayed biochemical changes play an important role in tissue damage resulting from traumatic injuries to the central nervous system. Identification of such 'secondary' injury factors has led to the development of various pharmacological strategies aimed at limiting this progressive tissue destruction. In this review, Alan Faden and Steven Salzman discuss the pharmacological approaches that have the most experimental support. These include corticosteroids, antioxidants and free-radical scavengers, modulators of arachidonate metabolism, gangliosides, monoamine modulators, opioid receptor antagonists, TRH and its analogs, NMDA receptor antagonists, Ca2+ channel antagonists and platelet-activating factor antagonists.
Collapse
Affiliation(s)
- A I Faden
- Georgetown University School of Medicine, Washington, DC 20007
| | | |
Collapse
|
41
|
Neuropsychological Rehabilitation from a Theoretical Point of View. Neuropsychol Rehabil 1992. [DOI: 10.1007/978-3-642-77067-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
42
|
Stein DG, Glasier MM. An overview of developments in research on recovery from brain injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1992; 325:1-22. [PMID: 1337820 DOI: 10.1007/978-1-4615-3420-4_1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- D G Stein
- Brain Research Laboratory, Rutgers, the State University, Newark, New Jersey
| | | |
Collapse
|
43
|
Antal J, d'Amore A, Nerozzi D, Palazzesi S, Pezzini G, Loizzo A. An EEG analysis of drug effects after mild head injury in mice. Life Sci 1992; 51:185-93. [PMID: 1614284 DOI: 10.1016/0024-3205(92)90074-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
An electroencephalographic (EEG) and behavioral model of head injury in unanesthetized, free moving mice has been used to test the effects of TRH and GM1. In our experimental conditions a mechanical head injury capable of inducing loss of righting reflex for 2 to 60 sec, also induces a consistent decrease of the total power of the spectrum of EEG and a decrease of the power of fast beta band (20-40 Hz) for at least 120 min. TRH, injected after trauma in dose of 10 mg/kg, caused improvement of EEG total power of the spectrum. GM1 in high (30 mg/kg) but not in low dose (5 mg/kg) caused more rapid restoration of both the total power and fast as well as slow beta band power. These results suggest that GM1 has favorable effects on post-concussive neurophysiological symptoms in head injured animals.
Collapse
Affiliation(s)
- J Antal
- Istituto Superiore di Sanità, Roma, Italy
| | | | | | | | | | | |
Collapse
|
44
|
Sautter J, Schwartz M, Duvdevani R, Sabel BA. GM1 ganglioside treatment reduces visual deficits after graded crush of the rat optic nerve. Brain Res 1991; 565:23-33. [PMID: 1773355 DOI: 10.1016/0006-8993(91)91732-g] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Despite numerous reports of beneficial effects of GM1 ganglioside treatment following brain lesions in animals, the underlying neurobiological mechanism of ganglioside-induced functional restoration is still unclear. In order to obtain a better insight into this question, we have made use of a newly developed animal model of brain injury that would potentially permit us to determine the causal relationship(s) among behavioral and neuroanatomical/neurochemical parameters of restoration of function. Following graded crush of the adult rat optic nerve, we have treated the rats with intraperitoneally injected gangliosides and studied the functional outcome with electrophysiological and behavioral parameters. The electrophysiological recording of the compound action potential (CAP) from excised rat optic nerve revealed a significant loss of CAP throughout the first 2 weeks after the injury. However, when rats were treated daily for 7 days with GM1-gangliosides, the CAP measured 10 days after the crush was significantly larger compared to operated controls without treatment. Thus, GM1 appeared to be capable of delaying or partially preventing retinal ganglion cells or their axons from secondary degeneration. Loss of visual function was also evident on the behavioral level of analysis: when rats with unilateral optic nerve crush were evaluated in a visual orienting paradigm, the rats revealed deficits in their ability to orient towards small, moving visual stimuli. However, within about 2 weeks, the animals recovered spontaneously to near normal performance. Daily treatment with GM1-gangliosides was found to significantly improve outcome, largely due to a reduction of the immediate post-lesion deficit. In a second behavioral experiment we also created graded crush in rats bilaterally and evaluated the animals visual capacities in a two-choice brightness discrimination task. In this task, an initial loss of function was followed by recovery within about 2 weeks, but GM1 treatment was without beneficial effects in this paradigm. It is concluded that GM1 improves outcome after graded crush of the adult rat optic nerve, although it appears that improved function needs to be documented with sufficiently sensitive behavioral assays.
Collapse
Affiliation(s)
- J Sautter
- Institute of Medical Psychology, Ludwig-Maximilians University of Munich, Medical Faculty, F.R.G
| | | | | | | |
Collapse
|
45
|
Geisler FH, Dorsey FC, Coleman WP. Recovery of motor function after spinal-cord injury--a randomized, placebo-controlled trial with GM-1 ganglioside. N Engl J Med 1991; 324:1829-38. [PMID: 2041549 DOI: 10.1056/nejm199106273242601] [Citation(s) in RCA: 369] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Spinal-cord injury is devastating; until recently, there was no medical treatment to improve recovery of the initial neurologic deficit. Studies in animals have shown that monosialotetrahexosylganglioside (GM-1) ganglioside enhances the functional recovery of damaged neurons. METHODS A prospective, randomized, placebo-controlled, double-blind trial of GM-1 ganglioside was conducted in patients with spinal-cord injuries. Of 37 patients entered into the study, 34 (23 with cervical injuries and 11 with thoracic injuries) completed the test-drug protocol (100 mg of GM-1 sodium salt or placebo intravenously per day for 18 to 32 doses, with the first dose taken within 72 hours of the injury) and a one-year follow-up period. Neurologic recovery was assessed with the Frankel scale (comprising five categories) and the American Spinal Injury Association (ASIA) motor score (a scale of scores from 0 to 100, derived from strength tests of 20 specific muscles, each scored from 0 to 5). RESULTS There was a significant difference between groups in the distribution of improvement of Frankel grades from base line to the one-year follow-up (improvement of 0, 1, 2, and 3 grades in 13, 4, 1, and 0 patients, respectively, in the placebo group and 8, 1, 6, and 1 patients, respectively, in the GM-1 group; P = 0.034 by the Cochran-Mantel-Haenszel chi-square test). The GM-1-treated patients also had a significantly greater mean improvement in ASIA motor score from base line to the one-year follow-up than the placebo-treated patients (36.9 vs. 21.6 points; P = 0.047 by analysis of covariance with the base-line ASIA motor score as the covariate). An analysis of individual muscle recoveries revealed that the increased recovery in the GM-1 group was attributable to initially paralyzed muscles that regained useful motor strength rather than to strengthening of paretic muscles. CONCLUSIONS This small study provides evidence that GM-1 enhances the recovery of neurologic function after one year. A larger study must be conducted, however, before GM-1 is considered efficacious and safe in treating spinal-cord injury.
Collapse
Affiliation(s)
- F H Geisler
- Shock Trauma Center, Maryland Institute for Emergency Medical Services Systems, Baltimore
| | | | | |
Collapse
|
46
|
Rodden FA, Wiegandt H, Bauer BL. Gangliosides: the relevance of current research to neurosurgery. J Neurosurg 1991; 74:606-19. [PMID: 2002375 DOI: 10.3171/jns.1991.74.4.0606] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Gangliosides are complex glycolipids found on the outer surface of most cell membranes: they are particularly concentrated in tissues of the nervous system. Gangliosides form part of the immunological identity of mammalian cells and are involved in a variety of cell-surface phenomena such as cell-substrate binding and receptor functions. In tumorous tissue, the ganglioside composition is altered, sometimes in direct proportion to the degree of malignancy. The literature on the glycosphingolipid composition and immunology of intracranial tumors is reviewed. Some gangliosides induce neuritogenesis and exhibit a trophic effect on nerve cells grown in vitro. In vivo, a particular ganglioside, GM1, reduces cerebral edema and accelerates recovery from injury (traumatic and ischemic) to the peripheral and central nervous systems of laboratory animals. Preliminary clinical studies have shown that treatment with gangliosides may have corresponding effects on lesions of the human peripheral nervous system. Gangliosides have not been tested in human subjects with brain injury.
Collapse
Affiliation(s)
- F A Rodden
- Department of Neurosurgery, Philipps University Medical Center, Marburg/Lahn, Federal Republic of Germany
| | | | | |
Collapse
|
47
|
Dietz MA, McDowell FH. Potentiation of rehabilitation: Medication effects on the recovery of function after brain injury and stroke. J Stroke Cerebrovasc Dis 1991; 1:37-48. [DOI: 10.1016/s1052-3057(11)80018-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
48
|
Skaper SD, Mazzari S, Vantini G, Facci L, Toffano G, Leon A. Monosialoganglioside GM1 and modulation of neuronal plasticity in CNS repair processes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1991; 296:257-66. [PMID: 1781332 DOI: 10.1007/978-1-4684-8047-4_24] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- S D Skaper
- Fidia Research Laboratories, Abano Terme, Italy
| | | | | | | | | | | |
Collapse
|
49
|
Fagioli S, Rossi-Arnaud C, Ammassari-Teule M. Open field behaviours and spatial learning performance in C57BL/6 mice: early stage effects of chronic GM1 ganglioside administration. Psychopharmacology (Berl) 1991; 105:209-12. [PMID: 1796128 DOI: 10.1007/bf02244311] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
One month intact C57BL/6 mice were treated with GM1 ganglioside for 3 consecutive weeks. At 2 months of age, treated and control mice were observed in the open-field situation and tested for spatial learning in a radial eight-arm maze. The results showed that, in the open-field, treated mice displayed less freezing but more rearing behavior than control animals. In the radial maze, GM1-treated mice made more correct path choices before the first error within each trial than control mice. However, this improvement was limited to the first stage of training. These results suggest an early stimulating effect of the GM1 ganglioside treatment which could facilitate adaptive reactions to new situations.
Collapse
Affiliation(s)
- S Fagioli
- Department of Genetics and Molecular Biology, University La Sapienza, Rome, Italy
| | | | | |
Collapse
|
50
|
Fazzini E, Durso R, Davoudi H, Szabo GK, Albert ML. GM1 gangliosides alter acute MPTP-induced behavioral and neurochemical toxicity in mice. J Neurol Sci 1990; 99:59-68. [PMID: 2250172 DOI: 10.1016/0022-510x(90)90199-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We investigated the effect of GM1 gangliosides on a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) animal model of Parkinson disease. Five groups of mice (saline, GM1 (30 mg/kg), MPTP, MPTP + GM1 (15 mg/kg), MPTP + GM1 (30 mg/kg] were compared. GM1 was given daily via intraperitoneal injection before and during 13 daily doses of MPTP (30 mg/kg). Mice were tested for locomotion (1) within 2 h of an MPTP dose (to measure reduced motor activity), and (2) within 24 h of an MPTP dose (after animals had recovered and exhibited hyperactivity). We found that mice given GM1 gangliosides exhibited significantly less MPTP-induced behavior. This effect was most evident with the 15 mg/kg GM1 dose. GM1 also appeared to attenuate MPTP-induced neurochemical changes. GM1 effects indicating enhancement of DA turnover and preservation of DA, DOPAC and HVA concentrations in the striatum were found after the 8th MPTP dose. These latter neurochemical changes, however, were transient and not present after the 13th MPTP dose. Our data would suggest that gangliosides may reduce acute MPTP-induced neurotoxicity in mice either through an increase in DA neuron survival and/or the augmentation of striatal DA activity.
Collapse
Affiliation(s)
- E Fazzini
- Department of Neurology, Boston University School of Medicine, MA
| | | | | | | | | |
Collapse
|