Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Mol Ther ; 29(10): 3059-3071, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34111562

ABSTRACT

Parkinson's disease (PD) is characterized by Lewy bodies (composed predominantly of alpha-synuclein [aSyn]) and loss of pigmented midbrain dopaminergic neurons comprising the nigrostriatal pathway. Most PD patients show significant deficiency of gangliosides, including GM1, in the brain, and GM1 ganglioside appears to keep dopaminergic neurons functioning properly. Thus, supplementation of GM1 could potentially provide some rescuing effects. In this study, we demonstrate that intranasal infusion of GD3 and GM1 gangliosides reduces intracellular aSyn levels. GM1 also significantly enhances expression of tyrosine hydroxylase (TH) in the substantia nigra pars compacta of the A53T aSyn overexpressing mouse, following restored nuclear expression of nuclear receptor related 1 (Nurr1, also known as NR4A2), an essential transcription factor for differentiation, maturation, and maintenance of midbrain dopaminergic neurons. GM1 induces epigenetic activation of the TH gene, including augmentation of acetylated histones and recruitment of Nurr1 to the TH promoter region. Our data indicate that intranasal administration of gangliosides could reduce neurotoxic proteins and restore functional neurons via modulating chromatin status by nuclear gangliosides.


Subject(s)
G(M1) Ganglioside/administration & dosage , Gangliosides/administration & dosage , Parkinson Disease/drug therapy , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/metabolism , Administration, Intranasal , Animals , Cell Line , Disease Models, Animal , Down-Regulation , Epigenesis, Genetic/drug effects , G(M1) Ganglioside/pharmacology , Gangliosides/pharmacology , Gene Expression Regulation/drug effects , Humans , Male , Mice , Parkinson Disease/genetics , Parkinson Disease/metabolism , Substantia Nigra/drug effects , Substantia Nigra/enzymology , Tyrosine 3-Monooxygenase/genetics
2.
Exp Neurol ; 329: 113284, 2020 07.
Article in English | MEDLINE | ID: mdl-32165255

ABSTRACT

Parkinson's disease (PD) is a major neurodegenerative disorder characterized by a variety of non-motor symptoms in addition to the well-recognized motor dysfunctions that have commanded primary interest. We previously described a new PD mouse model based on heterozygous disruption of the B4galnt1 gene leading to partial deficiency of the GM1 family of gangliosides that manifested several nigrostriatal neuropathological features of PD as well as movement impairment. We now show this mouse also suffers three non-motor symptoms characteristic of PD involving the gastrointestinal, sympathetic cardiac, and cerebral cognitive systems. Treatment of these animals with a synthetic form of GM1 ganglioside, produced by transfected E. coli, proved ameliorative of these symptoms as well as the motor defect. These findings further suggest subnormal GM1 to be a systemic defect constituting a major risk factor in sporadic PD and indicate the B4galnt1(+/-) (HT) mouse to be a true neuropathological model that recapitulates both motor and non-motor lesions of this condition.


Subject(s)
Disease Models, Animal , G(M1) Ganglioside/administration & dosage , G(M1) Ganglioside/deficiency , N-Acetylgalactosaminyltransferases/deficiency , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Animals , Female , G(M1) Ganglioside/genetics , Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/metabolism , Male , Memory Disorders/drug therapy , Memory Disorders/genetics , Memory Disorders/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Motor Skills Disorders/drug therapy , Motor Skills Disorders/genetics , Motor Skills Disorders/metabolism , N-Acetylgalactosaminyltransferases/genetics , Parkinson Disease/genetics
3.
Anesth Analg ; 130(2): 505-517, 2020 02.
Article in English | MEDLINE | ID: mdl-31934908

ABSTRACT

BACKGROUND: A prolonged exposure to ketamine triggers significant neurodegeneration and long-term neurocognitive deficits in the developing brain. Monosialotetrahexosylganglioside (GM1) can limit the neuronal damage from necrosis and apoptosis in neurodegenerative conditions. We aimed to assess whether GM1 can prevent ketamine-induced developmental neurotoxicity. METHODS: Postnatal day 7 (P7) rat pups received 5 doses of intraperitoneal ketamine (20 mg/kg per dose) at 90-minute intervals for 6 hours. Cognitive functions, determined by using Morris water maze (MWM) including escape latency (at P32-36) and platform crossing (at P37), were compared among the ketamine-exposed pups treated with or without exogenous GM1 (30 mg/kg; n = 12/group). The effect of GM1 on apoptosis in hippocampus was determined by terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'-triphosphate nick end labeling (TUNEL) staining and activated caspase 3 measurement. The hippocampal expression of brain-derived neurotrophic factor (BDNF), along with the phosphorylation of protein kinase B (AKT) and extracellular signal-related kinases 1 and 2 (ERK1/2), was detected by western blotting (n = 6/group). Anti-BDNF antibody (2 µg per rat) administered before GM1 treatment was applied to determine the neuroprotective mechanisms of GM1. RESULTS: The rats receiving ketamine exposure experinced cognitive impairment in MWM test compared to the control rats, indicated by prolonged escape latency at P34 (P = .006), P35 (P = .002), and P36 (P = .005). However, in GM1-pretreated rats, ketamine exposure did not induce prolonged escape latency. The exogenous GM1 increased the platform-crossing times at P37 (3.00 ± 2.22 times vs 5.40 ± 1.53 times, mean ± standard deviation; P = .041) and reduced the hippocampal TUNEL-positive cells and cleaved-caspase 3 expression in ketamine-exposed young rats. Ketamine decreased BDNF expression and phosphorylation of AKT and ERK in the hippocampus, whereas exogenous GM1 blocked these ketamine-caused effects. However, for the ketamine-exposed rat pups receiving exogenous GM1, compared to immunoglobulin Y (IgY) isotype control, the BDNF-neutralizing antibody treatment counteracted the exogenous GM1-induced improvement of the escape latency at P36 (41.32 ± 12.37 seconds vs 25.14 ± 8.97 seconds, mean ± standard deviation; P = .036), platform-crossing times at P37 (2.16 ± 1.12 times vs 3.92 ± 1.97 times, mean ± standard deviation; P < .036), apoptotic activity, as well as AKT and ERK1/2 phosphorylation in the hippocampus of ketamine-challenged young rats. CONCLUSIONS: Our data suggest that the exogenous GM1 acts on BDNF signaling pathway to ameliorate the cognitive impairment and hippocampal apoptosis induced by ketamine in young rats. Our study may indicate a potential use of GM1 in preventing the cognitive deficits induced by ketamine in the young per se.


Subject(s)
Anesthetics, Dissociative/toxicity , Brain/drug effects , G(M1) Ganglioside/administration & dosage , Ketamine/toxicity , Neurocognitive Disorders/chemically induced , Neurocognitive Disorders/prevention & control , Animals , Animals, Newborn , Brain/growth & development , Maze Learning/drug effects , Maze Learning/physiology , Neurocognitive Disorders/pathology , Random Allocation , Rats , Rats, Sprague-Dawley
4.
Cancer Med ; 9(1): 151-159, 2020 01.
Article in English | MEDLINE | ID: mdl-31724334

ABSTRACT

BACKGROUND: Monosialotetrahexosylganglioside (GM1) is a neuroprotective glycosphingolipid that repairs nerves. Oxaliplatin-based chemotherapy is neurotoxic. This study assessed the efficacy of GM1 for preventing oxaliplatin-induced peripheral neurotoxicity (OIPN) in colorectal cancer (CRC) patients receiving oxaliplatin-based chemotherapy. METHODS: In total, 196 patients with stage II/III CRC undergoing adjuvant chemotherapy with mFOLFOX6 were randomly assigned to intravenous GM1 or a placebo. The primary endpoint was the rate of grade 2 or worse cumulative neurotoxicity (NCI-CTCAE). The secondary endpoints were chronic cumulative neurotoxicity (EORTC QLQ-CIPN20), time to grade 2 neurotoxicity (NCI-CTCAE or the oxaliplatin-specific neuropathy scale), acute neurotoxicity (analog scale), rates of dose reduction or withdrawal due to OIPN, 3-year disease-free survival (DFS) and adverse events. RESULTS: There were no significant differences between the arms in the rate of NCI-CTCAE grade 2 or worse neurotoxicity (GM1: 33.7% vs placebo: 31.6%; P = .76) or neuropathy measured by the EORTC QLQ-CIPN20 or time to grade 2 neurotoxicity using NCI-CTCAE and the oxaliplatin-specific neuropathy scale. GM1 substantially decreased participant-reported acute neurotoxicity (sensitivity to cold items [P < .01], discomfort swallowing cold liquids [P < .01], throat discomfort [P < .01], muscle cramps [P < .01]). The rates of dose reduction or withdrawal were not significantly different between the arms (P = .08). The 3-year DFS rates were 85% and 83% in the GM1 and placebo arms, respectively (P = .19). There were no differences in toxicity between the arms. CONCLUSION: Patients receiving GM1 were less troubled by the symptoms of acute neuropathy. However, we do not support the use of GM1 to prevent cumulative neurotoxicity. (ClinicalTrials.gov number, NCT02251977).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Capecitabine/adverse effects , Colorectal Neoplasms/drug therapy , G(M1) Ganglioside/administration & dosage , Oxaliplatin/adverse effects , Oxaloacetates/adverse effects , Peripheral Nervous System Diseases/epidemiology , Adult , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Capecitabine/administration & dosage , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease-Free Survival , Dose-Response Relationship, Drug , Double-Blind Method , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Humans , Leucovorin/administration & dosage , Leucovorin/adverse effects , Male , Middle Aged , Neoplasm Staging , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/adverse effects , Oxaliplatin/administration & dosage , Oxaloacetates/administration & dosage , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/diagnosis , Peripheral Nervous System Diseases/prevention & control , Placebos/administration & dosage , Severity of Illness Index
5.
J Mol Neurosci ; 68(1): 78-90, 2019 May.
Article in English | MEDLINE | ID: mdl-30863991

ABSTRACT

Long-term memory impairment is reported in more than 50% of cardiac arrest survivors. Monosialoganglioside (GM1) provided neuroprotection in experimental models of stroke but failed to replicate its promise clinically for unknown reasons. GM1 stimulates the release of nerve growth factor (NGF), which is synthesized as a precursor protein (pro-NGF) that either mediates apoptosis through the p75 neurotrophin receptor (p75NTR) or is cleaved by the protease furin (FUR) to yield mature NGF, the latter supporting survival through tropomyosin kinase receptor (Trk). The flavanol epicatechin (EPI) inhibits p75NTR-mediated signaling and apoptosis by pro-NGF. The aim of the current work is to test whether these two drugs affect, or communicate with, each other in the setting of CNS injuries. Using the two-vessel occlusion model of global ischemia/reperfusion (I/R), we tested if pharmacological modulation of Trk, p75NTR, and NGF balance with GM1, EPI, and their combination, can correct the memory deficit that follows this insult. Finally, we tested if FUR insufficiency and/or p75NTR-mediated apoptosis negatively affect the neurotherapeutic effect of GM1. Key proteins for Trk and p75NTR, FUR, and both forms of NGF were assessed. All treatment regiments successfully improved spatial memory retention and acquisition. A week after the insult, most Trk and p75NTR proteins were normal, but pro/mature NGF ratio remained sharply elevated and was associated with the poorest memory performance. Pharmacological correction of this balance was achieved by reinforcing Trk and p75NTR signaling. GM1 increased FUR levels, while concomitant administration of EPI weakened GM1 effect on pro-survival Trk and p75NTR mediators. GM1 neuroprotection is therefore not limited by FUR but could be dependent on p75NTR. Graphical Abstract "."


Subject(s)
Brain Ischemia/drug therapy , Memory Disorders/drug therapy , Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Brain Ischemia/complications , Catechin/administration & dosage , Catechin/pharmacology , Catechin/therapeutic use , G(M1) Ganglioside/administration & dosage , G(M1) Ganglioside/pharmacology , G(M1) Ganglioside/therapeutic use , Male , Memory Disorders/etiology , Nerve Tissue Proteins , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Rats, Wistar , Receptors, Growth Factor , Receptors, Nerve Growth Factor/metabolism
6.
J Mol Neurosci ; 67(2): 305-311, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30604381

ABSTRACT

To study the clinical effectiveness and mechanism of GM1 acupoint injection therapy on mental retardation for children with cerebral palsy (CP). A total of 90 children with CP were divided into acupoint injection group (group A), subcutaneous injection group (group B), and control group (group C). Another 30 healthy children were set as a healthy control group (group D). The Mental Developmental Index (MDI), Psychomotor Developmental Index (PDI), and hemodynamic parameters in the cerebral arteries were measured before and after treatment. After three treatment courses, the MDI and PDI in groups A, B, and C were increased, and the increase in group A was most obvious (P < 0.05). Peak systolic velocity, mean velocity, and end-diastolic velocity were also elevated in group A, and after three treatment courses, resistance index decreased with a statistical significance (P < 0.05). However, there were no significant changes in groups B and C (P > 0.05). For all groups, neuron-specific enolase levels decreased and total superoxide dismutase increased after treatment. Acupoint injection therapy combined with conventional rehabilitation therapy demonstrated significant effects on cerebral hemodynamic conditions for children with CP.


Subject(s)
Acupuncture Therapy/methods , Cerebral Palsy/complications , G(M1) Ganglioside/therapeutic use , Intellectual Disability/therapy , Acupuncture Points , Cerebral Arteries/physiology , Child, Preschool , Female , G(M1) Ganglioside/administration & dosage , Hemodynamics , Humans , Infant , Injections, Subcutaneous/methods , Intellectual Disability/drug therapy , Intellectual Disability/etiology , Male , Phosphopyruvate Hydratase/blood , Superoxide Dismutase/blood
7.
Macromol Biosci ; 19(2): e1800340, 2019 02.
Article in English | MEDLINE | ID: mdl-30536989

ABSTRACT

Alzheimer's disease (AD) is an incurable neurodegenerative brain disorder that exhibits clear pathologic changes in the hippocampus. Traditional drug delivery systems are ineffective due to the existence of the blood-brain barrier (BBB). In this study, an efficient, stable, and easily constructed nanosystem (CB-Gd-Cy5.5) based on the cholera toxin B subunit (CB) is designed to improve the efficiency of drug delivery to the brain, especially the hippocampus. Through intranasal administration, CB-Gd-Cy5.5 is easily delivered to the brain without intervention by the BBB. The CB in CB-Gd-Cy5.5 is used for specifically combining with the monosialoganglioside GM1, which is widely found in the hippocampus. This nanosystem exhibits impressive performance in accumulating in the hippocampus. In addition, the good magnetic resonance imaging (MRI) capability of CB-Gd-Cy5.5 can satisfy the monitoring of AD in the different stages.


Subject(s)
Alzheimer Disease/drug therapy , Cholera Toxin/pharmacology , Drug Carriers/pharmacology , G(M1) Ganglioside/chemistry , Hippocampus/physiology , Nanoparticles/chemistry , Administration, Intranasal , Animals , Blood-Brain Barrier , Carbocyanines , Cell Line , Cholera Toxin/chemistry , Drug Carriers/chemistry , G(M1) Ganglioside/administration & dosage , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred BALB C , Nanoparticles/administration & dosage
8.
Sci Rep ; 7: 41269, 2017 01 23.
Article in English | MEDLINE | ID: mdl-28112258

ABSTRACT

Blast induced traumatic brain injury (B-TBI) may cause various degrees of cognitive and behavioral disturbances but the exact brain pathophysiology involved is poorly understood. It was previously suggested that ganglioside alteration on the axon surface as well as axonal regenerating inhibitors (ARIs) such as myelin associated glycoprotein (MAG) were involved in axonal outgrowth inhibition (AOI), leading to brain damage. GM1 ganglioside content in the brain was significantly reduced while GD1 ganglioside was not affected. The axonal regeneration was also reduced as seen by the phosphorylated NF-H expression. Moreover, B-TBI induced a significant elevation in MAG expression in the brains of the injured mice. The blast injured mice exhibited a significant decline in spatial memory as seen by the Y-maze test. In addition, the injured mice showed pronounced damage to the visual memory (as evaluated by the Novel object recognition test). A single low dose of GM1 (2 mg/kg; IP), shortly after the injury, prevented both the cognitive and the cellular changes in the brains of the injured mice. These results enlighten part of the complicated mechanism that underlies the damage induced by B-TBI and may also suggest a potential new treatment strategy for brain injuries.


Subject(s)
Axons/metabolism , Brain Injuries, Traumatic/complications , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Explosions , G(M1) Ganglioside/metabolism , Actins/metabolism , Animals , Biomarkers/metabolism , Brain Injuries, Traumatic/drug therapy , Cognitive Dysfunction/drug therapy , G(M1) Ganglioside/administration & dosage , Gangliosides/metabolism , Growth Cones/drug effects , Growth Cones/metabolism , Hippocampus/metabolism , Male , Mice, Inbred ICR , Myelin Sheath/metabolism , Neurofilament Proteins/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Time Factors
9.
J Coll Physicians Surg Pak ; 26(7): 633-4, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27504562

ABSTRACT

Postherpetic neuralgia (PHN) is a commonest and difficult-to-manage complication of Herpes zoster. This comparative study included 140 cases of PHN admitted in the department of dermatology in Renmin Hospital of Wuhan University, Wuhan, China, from March 2014 to February 2015, divided into a control and a study group. In addition to the combination of antiviral, analgesic, and neurotrophic agents given to the control group, additional ganglioside GM1 was given to patients in the study group. Pain assessment was performed at the time of admission, and then on the third, seventh and tenth day of treatment, on both groups, using a 10 cm visual analogue scale (VAS). There was a significant statistical difference between the pain VAS score of the two groups, on the seventh day (3.73 ±1.66 vs. 3.03 ±1.86, p=0.024) and on the tenth day (3.25 ±1.78 vs. 2.20 ±1.59, p=0.006) of treatment. The number of patients who have good /and complete response (37.5%) were largely higher in the study group than those in the control group (15%, p < 0.05). This finding demonstrates that the administration of ganglioside GM1 may potentially serve as a neoadjuvant therapy to reduce the severity and duration of pain in PHN patients.


Subject(s)
Analgesics/administration & dosage , Antiviral Agents/administration & dosage , G(M1) Ganglioside/administration & dosage , Herpes Zoster/drug therapy , Neuralgia, Postherpetic/drug therapy , Vitamin B Complex/administration & dosage , Administration, Intravenous , Adult , China , Female , Herpes Zoster/complications , Herpes Zoster/immunology , Humans , Injections, Intramuscular , Male , Middle Aged , Neuralgia, Postherpetic/etiology , Pain Measurement , Thiamine/administration & dosage , Treatment Outcome , Vitamin B 12
10.
ACS Nano ; 9(11): 10801-16, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26440073

ABSTRACT

Alzheimer's disease (AD) exerts a heavy health burden for modern society and has a complicated pathological background. The accumulation of extracellular ß-amyloid (Aß) is crucial in AD pathogenesis, and Aß-initiated secondary pathological processes could independently lead to neuronal degeneration and pathogenesis in AD. Thus, the development of combination therapeutics that can not only accelerate Aß clearance but also simultaneously protect neurons or inhibit other subsequent pathological cascade represents a promising strategy for AD intervention. Here, we designed a nanostructure, monosialotetrahexosylganglioside (GM1)-modified reconstituted high density lipoprotein (GM1-rHDL), that possesses antibody-like high binding affinity to Aß, facilitates Aß degradation by microglia, and Aß efflux across the blood-brain barrier (BBB), displays high brain biodistribution efficiency following intranasal administration, and simultaneously allows the efficient loading of a neuroprotective peptide, NAP, as a nanoparticulate drug delivery system for the combination therapy of AD. The resulting multifunctional nanostructure, αNAP-GM1-rHDL, was found to be able to protect neurons from Aß(1-42) oligomer/glutamic acid-induced cell toxicity better than GM1-rHDL in vitro and reduced Aß deposition, ameliorated neurologic changes, and rescued memory loss more efficiently than both αNAP solution and GM1-rHDL in AD model mice following intranasal administration with no observable cytotoxicity noted. Taken together, this work presents direct experimental evidence of the rational design of a biomimetic nanostructure to serve as a safe and efficient multifunctional nanoplatform for the combination therapy of AD.


Subject(s)
Alzheimer Disease/therapy , G(M1) Ganglioside/metabolism , Lipoproteins, HDL/metabolism , Nanoparticles/chemistry , Administration, Intranasal , Alzheimer Disease/complications , Alzheimer Disease/pathology , Amino Acid Sequence , Amyloid beta-Peptides/metabolism , Animals , Apolipoproteins E/metabolism , Brain/drug effects , Brain/metabolism , Combined Modality Therapy , Disease Models, Animal , Endocytosis/drug effects , G(M1) Ganglioside/administration & dosage , G(M1) Ganglioside/pharmacokinetics , Glutamates/toxicity , Lipoproteins, HDL/administration & dosage , Lipoproteins, HDL/pharmacokinetics , Memory Disorders/complications , Memory Disorders/drug therapy , Memory Disorders/pathology , Mice, Inbred ICR , Microglia/drug effects , Microglia/metabolism , Molecular Sequence Data , Nanoparticles/ultrastructure , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Particle Size , Peptide Fragments/metabolism , Peptides/chemistry , Rats, Sprague-Dawley , Static Electricity , Tissue Distribution/drug effects
11.
Behav Brain Res ; 278: 303-6, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25453740

ABSTRACT

Gangliosides are compounds that are abundant throughout the CNS, participating actively in neuroplasticity. We previously described that exogenous GM1 ganglioside pretreatment enhances the rewarding properties of cocaine, evidenced by a lower number of sessions and/or dosage necessary to induce conditioned place preference (CPP). Since GM1 pretreatment did not modify cocaine's pharmacokinetic parameters, we suspected that the increased rewarding effect found might be mediated by BDNF, a neurotrophic factor closely related to cocaine addiction. This study was performed to investigate the possibility that GM1 may induce changes in BDNF levels in the nucleus accumbens (NAc), a core structure in the brain's reward circuitry, of rats submitted to three conditioning sessions with cocaine (10 mg/kg, i.p.). The results demonstrate that GM1 administration, which showed no rewarding effect by itself in the CPP, induced a significant increase of BDNF protein levels in the NAc, which may account for the increased rewarding effect of cocaine shown in the CPP paradigm.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cocaine/pharmacology , Conditioning, Operant/drug effects , G(M1) Ganglioside/pharmacology , Nucleus Accumbens/metabolism , Reward , Animals , G(M1) Ganglioside/administration & dosage , Male , Rats , Rats, Wistar
12.
Brain Res ; 1592: 73-81, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25285892

ABSTRACT

The progressive pathogenesis and prevention of Parkinson's disease (PD) remains unknown at present. Therefore, the present study aimed to investigate the possible progressive pathogenesis and prevention of PD. Our study investigated the content of glutamate, mitochondria calcium, calmodulin, malonaldehyde and trace elements in striatum, cerebral cortex and hippocampus tissues; and the expression of bcl-2, bax and neuronal nitric oxide synthase (nNOS) in substantia nigra and striatum; and the change of apomorphine induced rotation behavior; and the treatmental effect of monosialotetrahexosylganglioside (GM1) intraperitoneal administration for 14 days in a PD rat model induced by 6-hydroxydopamine. The results revealed that the content of glutamate significantly decreased, and that of mitochondria calcium, calmodulin, malonaldehyde and ferrum significantly increased in striatum, cerebral cortex and hippocampus tissues; the content of magnesium significantly decreased, and that of cuprum and zinc significantly increased in cerebral cortex; the expression of bcl-2 significantly decreased, and that of bax and nNOS significantly increased in substantia nigra and striatum in PD rat. GM1 can partially improve the apomorphine induced rotation behavior and changes of glutamate, mitochondria calcium, calmodulin content in striatum of PD rat. Data suggested that dysfunction of excitatory amino acids neurotransmitter, calcium homeostasis disorder, abnormal metabolism of oxygen free radicals, abnormal trace elements distribution and/or deposition and excessive apoptosis participated in the progressive process of PD, and that GM1 could partially prevent the progressive damage.


Subject(s)
Brain/drug effects , Brain/physiopathology , G(M1) Ganglioside/administration & dosage , Neuroprotective Agents/administration & dosage , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/physiopathology , Animals , Calcium/metabolism , Calmodulin/metabolism , Disease Progression , Glutamic Acid/metabolism , Infusions, Parenteral , Male , Malondialdehyde/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Nitric Oxide Synthase Type I/metabolism , Oxidopamine , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats, Wistar , Trace Elements/metabolism , bcl-2-Associated X Protein/metabolism
13.
Mol Pharm ; 9(6): 1620-7, 2012 Jun 04.
Article in English | MEDLINE | ID: mdl-22536790

ABSTRACT

Due to the aging of the population, the incidence of neurodegenerative diseases, such as Parkinson's and Alzheimer's, is expected to grow and, hence, the demand for adequate treatment modalities. However, the delivery of medicines into the brain for the treatment of brain-related diseases is hampered by the presence of a tight layer of endothelial cells that forms the blood-brain barrier (BBB). Furthermore, most conventional drugs lack stability and/or bioavailability. These obstacles can be overcome by the application of nanocarriers, in which the therapeutic entity has been incorporated, provided that they are effectively targeted to the brain endothelial cell layer. Drug nanocarriers decorated with targeting ligands that bind BBB receptors may accumulate efficiently at/in brain microvascular endothelium and hence represent a promising tool for brain drug delivery. Following the accumulation of drug nanocarriers at the brain vasculature, the drug needs to be transported across the brain endothelial cells into the brain. Transport across brain endothelial cells can occur via passive diffusion, transport proteins, and the vesicular transport pathways of receptor-mediated and adsorptive-mediated transcytosis. When a small lipophilic drug is released from its carrier at the brain vasculature, it may enter the brain via passive diffusion. On the other hand, the passage of intact nanocarriers, which is necessary for the delivery of larger and more hydrophilic drugs into brain, may occur via active transport by means of transcytosis. In previous work we identified GM1 ganglioside and prion protein as potential transcytotic receptors at the BBB. GM1 is a glycosphingolipid that is ubiquitously present on the endothelial surface and capable of acting as the transcytotic receptor for cholera toxin B. Likewise, prion protein has been shown to have transcytotic capacity at brain endothelial cells. Here we determine the transcytotic potential of polymersome nanocarriers functionalized with GM1- and prion-targeting peptides (G23, P50 and P9), that were identified by phage display, in an in vitro BBB model. In addition, the biodistribution of polymersomes functionalized with either the prion-targeting peptide P50 or the GM1-targeting peptide G23 is determined following intravenous injection in mice. We show that the prion-targeting peptides do not induce efficient transcytosis of polymersomes across the BBB in vitro nor induce accumulation of polymersomes in the brain in vivo. In contrast, the G23 peptide is shown to have transcytotic capacity in brain endothelial cells in vitro, as well as a brain-targeting potential in vivo, as reflected by the accumulation of G23-polymersomes in the brain in vivo at a level comparable to that of RI7217-polymersomes, which are targeted toward the transferrin receptor. Thus the G23 peptide seems to serve both of the requirements that are needed for efficient brain drug delivery of nanocarriers. An unexpected finding was the efficient accumulation of G23-polymersomes in lung. In conclusion, because of its combined brain-targeting and transcytotic capacity, the G23 peptide could be useful in the development of targeted nanocarriers for drug delivery into the brain, but appears especially attractive for specific drug delivery to the lung.


Subject(s)
G(M1) Ganglioside/administration & dosage , G(M1) Ganglioside/pharmacokinetics , Prions/administration & dosage , Prions/pharmacokinetics , Administration, Intravenous , Animals , Blood-Brain Barrier/metabolism , Drug Delivery Systems/methods , Male , Mice , Mice, Inbred BALB C
14.
Proc Natl Acad Sci U S A ; 109(9): 3528-33, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22331905

ABSTRACT

Huntington disease (HD) is a progressive neurodegenerative monogenic disorder caused by expansion of a polyglutamine stretch in the huntingtin (Htt) protein. Mutant huntingtin triggers neural dysfunction and death, mainly in the corpus striatum and cerebral cortex, resulting in pathognomonic motor symptoms, as well as cognitive and psychiatric decline. Currently, there is no effective treatment for HD. We report that intraventricular infusion of ganglioside GM1 induces phosphorylation of mutant huntingtin at specific serine amino acid residues that attenuate huntingtin toxicity, and restores normal motor function in already symptomatic HD mice. Thus, our studies have identified a potential therapy for HD that targets a posttranslational modification of mutant huntingtin with critical effects on disease pathogenesis.


Subject(s)
G(M1) Ganglioside/therapeutic use , Motor Activity/drug effects , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Protein Processing, Post-Translational/drug effects , Animals , Codon/drug effects , Corpus Striatum/metabolism , Dimerization , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/biosynthesis , Dopamine and cAMP-Regulated Phosphoprotein 32/genetics , Drug Evaluation, Preclinical , G(M1) Ganglioside/administration & dosage , Huntingtin Protein , Infusion Pumps, Implantable , Infusions, Parenteral , Mice , Mice, Neurologic Mutants , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/metabolism , Phosphorylation/drug effects , Phosphoserine/analysis , Psychomotor Performance/drug effects
15.
Cell Mol Neurobiol ; 30(1): 43-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19582569

ABSTRACT

The neuropeptide-immunoreactive (IR) and neurofilament-IR neurons are two major phenotypical classes in dorsal root ganglion (DRG). Targets of neuronal innervation play a vital role in regulating the survival and differentiation of innervating neurotrophin-responsive neurons. Monosialoganglioside (GM1) has been considered to have a neurotrophic factor-like activity. Both GM1 and target skeletal muscle (SKM) cells are essential for the maintenance of the function of neurons. However, whether target SKM cells and GM1, alone or associated, generate neuropeptide or neurofilament expression remains unclear. The aim of the present study is to investigate the effects of GM1 or/and SKM on DRG neuronal phenotypes. DRG neurons containing the neuropeptide substance P (SP) and neurofilament 200 (NF-200) were quantified using immunofluorescent labeling in cultures of DRG, which was dissected out at times before (at embryonic days 12.5, E12.5) and after (at E19.5) sensory neurons contact peripheral targets in vivo. DRG neurons were cultured in absence or presence of GM1 or/and SKM cells. In this experiment, we found that: (1) GM1 promoted expression of SP and NF-200 in E12.5 DRG cultures; (2) SKM cells promoted expression of NF-200 but not SP in E12.5 DRG cultures; (3) GM1 and target SKM cells had additive effects on expression of SP and NF-200 in E12.5 DRG cultures; and (4) SKM or/and GM1 did not have effects on expression of SP and NF-200 in E19.5 DRG cultures. These results suggested that GM1 could influence DRG, two major neuronal phenotypes, before sensory neurons contact peripheral targets in vivo. Target SKM cells could only influence neurofilament-expressed neuronal phenotype before sensory neurons contact peripheral targets in vivo. GM1 and SKM cells had the additive effects on two major DRG neuronal classes, which express neuropeptide or neurofilament when DRG cells were harvested before sensory neurons contact peripheral targets in vivo. These results offered new clues for a better understanding of the association of GM1 or/and SKM with neuronal phenotypes.


Subject(s)
G(M1) Ganglioside/administration & dosage , G(M1) Ganglioside/pharmacology , Ganglia, Spinal/cytology , Muscle Cells/cytology , Muscle, Skeletal/cytology , Neurons/cytology , Animals , Cells, Cultured , Microscopy, Fluorescence , Muscle Cells/drug effects , Neurofilament Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Phenotype , Rats , Rats, Wistar , Substance P/metabolism
16.
Brain Res Bull ; 79(1): 85-8, 2009 Apr 06.
Article in English | MEDLINE | ID: mdl-19133317

ABSTRACT

Monosialoganglioside (GM1) has been considered to have a neurotrophic factor-like activity. Nerve growth factor (NGF), a member of the neurotrophin family, is essential for neuronal survival, differentiation and maturation. The aim of the present study was to investigate whether co-administration of GM1 and NGF reverses glutamate (Glu) neurotoxicity in primary cultured rat embryonic spinal cord neurons. Spinal cord neurons were exposed to Glu (2 mmol/l), Glu (2 mmol/l) plus GM1 (10 mg/ml), Glu (2 mmol/l) plus NGF (10 ng/ml), Glu (2 mmol/l) plus GM1 (5 mg/ml) and NGF (5 ng/ml) and then processed for detecting intracellular concentrations of Ca2+([Ca2+]i) by confocal laser scanning microscopy and growth associated protein 43 (GAP43) mRNA by RT-PCR. The fluorescent intensity in Glu plus GM1 and NGF incubated neurons was the lowest as compared with that in other groups. The expression of GAP43 mRNA in Glu plus GM1 and NGF incubated neurons was the highest as compared with that in other groups. These results implicated that GM1 and NGF have synergistic neuroprotective effects on spinal cord neurons with excitotoxicity induced by Glu in vitro.


Subject(s)
G(M1) Ganglioside/administration & dosage , Nerve Growth Factor/administration & dosage , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Spinal Cord/cytology , Analysis of Variance , Animals , Calcium/metabolism , Cells, Cultured , Drug Therapy, Combination , GAP-43 Protein/metabolism , Microscopy, Confocal , Neurons/physiology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/drug effects
17.
Cytokine ; 42(1): 92-104, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18329889

ABSTRACT

NOD (non-obese diabetic) mice develop type 1 diabetes mellitus spontaneously and with a strong similarity to the human disease. Differentiation and function of pancreas beta cells are regulated by a variety of hormones and growth factors, including the nerve growth factor (NGF). Gangliosides have multiple immunomodulatory activities with immunosuppressive properties, decreasing lymphoproliferative responses and modulating cytokine production. In the present study, serum, pancreas islets and spleen mononuclear cells from NOD mice treated with monosialic ganglioside GM1 (100 mg/kg/day) and the group control which received saline solution were isolated to investigate the proinflammatory cytokines (IL-1beta, IFN-gamma, IL-12, TNF-alpha), NGF and its high-affinity receptor TrkA, peri-islet Schwann cells components (GFAP, S100-beta) expression and the relationship with diabetes onset and morphological aspects. Our results suggest that GM1 administration to female NOD mice beginning at the 4th week of life is able to reduce the index of inflammatory infiltrate and consequently the expression of diabetes, modulating the expression of proinflammatory cytokines (IL-12, IFN-gamma, TNF-alpha and IL-1beta). Furthermore, GM1 increases GFAP, S-100beta and NGF in pancreas islets, factors involved in beta cell survival.


Subject(s)
Cytokines/metabolism , Diabetes Mellitus, Type 1/immunology , G(M1) Ganglioside/metabolism , Mice, Inbred NOD , Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Age of Onset , Animals , Cytokines/genetics , Female , G(M1) Ganglioside/administration & dosage , Glial Fibrillary Acidic Protein , Humans , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Male , Mice , Nerve Growth Factor/genetics , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptor, trkA/genetics , S100 Calcium Binding Protein beta Subunit , S100 Proteins/genetics , S100 Proteins/metabolism , Schwann Cells/cytology , Schwann Cells/metabolism , Spleen/cytology , Spleen/metabolism
18.
Brain Res ; 1058(1-2): 193-7, 2005 Oct 05.
Article in English | MEDLINE | ID: mdl-16153613

ABSTRACT

Cytidine-5'-diphosphocholine (CDP-choline, Citicoline, Somazina) is in clinical use (intravenous administration) for stroke treatment in Europe and Japan, while USA phase III stroke clinical trials (oral administration) were disappointing. Others showed that CDP-choline liposomes significantly increased brain uptake over the free drug in cerebral ischemia models. Liposomes were formulated as DPPC, DPPS, cholesterol, GM(1) ganglioside; 7/4/7/1.57 molar ratio or 35.8/20.4/35.8/8.0 mol%. GM(1) ganglioside confers long-circulating properties to the liposomes by suppressing phagocytosis. CDP-choline liposomes deliver the agent intact to the brain, circumventing the rate-limiting, cytidine triphosphate:phosphocholine cytidylyltransferase in phosphatidylcholine synthesis. Our data show that CDP-choline liposomes significantly ( P < 0.01) decreased cerebral infarction (by 62%) compared to the equivalent dose of free CDP-choline (by 26%) after 1 h focal cerebral ischemia and 24 h reperfusion in spontaneously hypertensive rats. Beneficial effects of CDP-choline liposomes in stroke may derive from a synergistic effect between the phospholipid components of the liposomes and the encapsulated CDP-choline.


Subject(s)
Brain Infarction/drug therapy , Brain/drug effects , Choline/administration & dosage , Cytidine Diphosphate Choline/administration & dosage , Liposomes/pharmacology , Stroke/drug therapy , Animals , Brain/metabolism , Brain/pathology , Brain Infarction/metabolism , Brain Infarction/physiopathology , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Cholesterol/administration & dosage , Disease Models, Animal , Drug Combinations , Drug Synergism , G(M1) Ganglioside/administration & dosage , Liposomes/therapeutic use , Male , Phagocytosis/drug effects , Phagocytosis/physiology , Phosphatidylcholines/biosynthesis , Rats , Rats, Inbred SHR , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology , Stroke/metabolism , Stroke/physiopathology , Treatment Outcome
19.
Brain Res ; 1054(2): 125-34, 2005 Aug 30.
Article in English | MEDLINE | ID: mdl-16084500

ABSTRACT

We investigated the ability of GM1 to induce phosphorylation/activation of the extracellular-regulated protein kinases (ERKs) in the striatum, hippocampus and frontal cortex of aged male Sprague-Dawley rats. Three different treatment paradigms were used: a single application of GM1 to brain slices in situ, a single intracerebroventricular (icv) administration of GM1 in vivo, and chronic administration of GM1 in vivo. In situ, GM1 induced a rapid and transient activation of ERK1 and ERK 2 in both young and aged rats, and a similar effect was observed after stimulation with the neurotrophins NGF and BDNF. The aged brain appeared to respond more robustly to neurotrophic stimulation with the pERK2 response being significantly greater in the hippocampus and frontal cortex. Acute icv administration of GM1 resulted in short-lasting phosphorylation of ERKs in both aged groups, while chronic administration of GM1 induced a protracted phosphorylation of ERKs. Following chronic GM1 treatment, pERK2 levels in the aged hippocampus were elevated over young control animals. In agreement with reports that GM1 phosphorylates TrkA in vitro or in situ, treatment with GM1 increased the phosphorylation of TrkA in hippocampus of both young and aged animals. These observations indicate that the aged brain maintains the ability to respond to neurotrophic stimuli and put forward the proposition that the ERK cascade is associated with the action(s) of GM1 ganglioside in vivo.


Subject(s)
Aging/physiology , Brain/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , G(M1) Ganglioside/administration & dosage , Signal Transduction/drug effects , Age Factors , Animals , Blotting, Western/methods , Brain/anatomy & histology , Brain/metabolism , Drug Administration Routes , Drug Administration Schedule , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/classification , Flavonoids/pharmacology , In Vitro Techniques , Male , Myelin Basic Protein/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley
20.
J Neurosci ; 23(1): 29-33, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12514198

ABSTRACT

Plaques containing beta-amyloid (Abeta) peptides are one of the pathological features of Alzheimer's disease, and the reduction of Abeta is considered a primary therapeutic target. Amyloid clearance by anti-Abeta antibodies has been reported after immunization, and recent data have shown that the antibodies may act as a peripheral sink for Abeta, thus altering the periphery/brain dynamics. Here we show that peripheral treatment with an agent that has high affinity for Abeta (gelsolin or GM1) but that is unrelated to an antibody or immune modulator reduced the level of Abeta in the brain, most likely because of a peripherally acting effect. We propose that in general, compounds that sequester plasma Abeta could reduce or prevent brain amyloidosis, which would enable the development of new therapeutic agents that are not limited by the need to penetrate the brain or evoke an immune response.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloidosis/drug therapy , G(M1) Ganglioside/pharmacology , Gelsolin/pharmacology , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/blood , Amyloid beta-Protein Precursor/genetics , Amyloidosis/metabolism , Amyloidosis/pathology , Animals , Brain/metabolism , Female , G(M1) Ganglioside/administration & dosage , G(M1) Ganglioside/metabolism , Gelsolin/administration & dosage , Gelsolin/metabolism , Injections, Intraperitoneal , Male , Membrane Proteins/genetics , Mice , Mutation , Presenilin-1
SELECTION OF CITATIONS
SEARCH DETAIL
...