ABSTRACT
Parkinson's disease (PD) is one of the most prevalent neurodegenerative disorders worldwide. It is caused by the degeneration of dopaminergic neurons from the substantia nigra pars compacta. This neuronal loss causes the dopamine deficiency that leads to a series of functional changes within the basal ganglia, producing motor control abnormalities. L-DOPA is considered the gold standard for PD treatment, and it may alleviate its clinical manifestations for some time. However, its prolonged administration produces tolerance and several severe side effects, including dyskinesias and gastrointestinal disorders. Thus, there is an urgent need to find effective medications, and current trends have proposed some natural products as emerging options for this purpose. Concerning this, curcumin represents a promising bioactive compound with high therapeutic potential. Diverse studies in cellular and animal models have suggested that curcumin could be employed for the treatment of PD. Therefore, the objective of this narrative mini-review is to present an overview of the possible therapeutic effects of curcumin and the subjacent molecular mechanisms. Moreover, we describe several possible nanocarrier-based approaches to improve the bioavailability of curcumin and enhance its biological activity.
Subject(s)
Brain/drug effects , Curcumin/administration & dosage , Nanoparticles/administration & dosage , Parkinson Disease/drug therapy , Animals , Biological Availability , Brain/metabolism , Curcumin/chemistry , Curcumin/pharmacokinetics , Drug Liberation , Glutathione Peroxidase/metabolism , Humans , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Nanoparticles/chemistry , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Parkinson Disease/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Treatment Outcome , Up-Regulation/drug effectsABSTRACT
Extensive data have reported the involvement of oxidative stress in the pathogenesis of neuropsychiatric disorders, prompting the pursuit of antioxidant molecules that could become adjuvant pharmacological agents for the management of oxidative stress-associated disorders. The 3-[(4-chlorophenyl)selanyl]-1-methyl-1H-indole (CMI) has been reported as an antioxidant and immunomodulatory compound that improves depression-like behavior and cognitive impairment in mice. However, the exact effect of CMI on specific brain cells is yet to be studied. In this context, the present study aimed to evaluate the antioxidant activity of CMI in H2O2-induced oxidative stress on human dopaminergic neuroblastoma cells (SH-SY5Y) and to shed some light into its possible mechanism of action. Our results demonstrated that the treatment of SH-SY5Y cells with 4 µM CMI protected them against H2O2 (343 µM)-induced oxidative stress. Specifically, CMI prevented the increased number of reactive oxygen species (ROS)-positive cells induced by H2O2 exposure. Furthermore, CMI treatment increased the levels of reduced glutathione in SH-SY5Y cells. Molecular docking studies demonstrated that CMI might interact with enzymes involved in glutathione metabolism (i.e., glutathione peroxidase and glutathione reductase) and H2O2 scavenging (i.e., catalase). In silico pharmacokinetics analysis predicted that CMI might be well absorbed, metabolized, and excreted, and able to cross the blood-brain barrier. Also, CMI was not considered toxic overall. Taken together, our results suggest that CMI protects dopaminergic neurons from H2O2-induced stress by lowering ROS levels and boosting the glutathione system. These results will facilitate the clinical application of CMI to treat nervous system diseases associated with oxidative stress.
Subject(s)
Hydrogen Peroxide/toxicity , Indoles/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Selenium Compounds/pharmacology , Catalytic Domain , Cell Line, Tumor , Glutathione/metabolism , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Humans , Indoles/chemistry , Indoles/metabolism , Indoles/pharmacokinetics , Molecular Docking Simulation , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacokinetics , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Protein Binding , Reactive Oxygen Species/metabolism , Selenium Compounds/chemistry , Selenium Compounds/metabolism , Selenium Compounds/pharmacokineticsABSTRACT
A new series of ten multifunctional Cinnamoyl-N-acylhydrazone-donepezil hybrids was synthesized and evaluated as multifunctional ligands against neurodegenerative diseases. The molecular hybridization approach was based on the combination of 1-benzyl-4-piperidine fragment from the anti-Alzheimer AChE inhibitor donepezil (1) and the cinnamoyl subunit from curcumin (2), a natural product with remarkable antioxidant, neuroprotective and anti-inflammatory properties, using a N-acylhydrazone fragment as a spacer subunit. Compounds 4a and 4d showed moderate inhibitory activity towards AChE with IC50 values of 13.04 and 9.1 µM, respectively. In addition, compound 4a and 4d showed a similar predicted binding mode to that observed for donepezil in the molecular docking studies. On the other hand, compounds 4a and 4c exhibited significant radical scavenging activity, showing the best effects on the DPPH test and also exhibited a significant protective neuronal cell viability exposed to t-BuOOH and against 6-OHDA insult to prevent the oxidative stress in Parkinson's disease. Similarly, compound 4c was capable to prevent the ROS formation, with indirect antioxidant activity increasing intracellular GSH levels and the ability to counteract the neurotoxicity induced by both OAß1-42 and 3-NP. In addition, ADMET in silico prediction indicated that both compounds 4a and 4c did not show relevant toxic effects. Due to their above-mentioned biological properties, compounds 4a and 4c could be explored as lead compounds in search of more effective and low toxic small molecules with multiple neuroprotective effects for neurodegenerative diseases.
Subject(s)
Cinnamates/pharmacology , Donepezil/pharmacology , Hydrazones/pharmacology , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Acetylcholinesterase/metabolism , Butyrylcholinesterase/metabolism , Cell Line, Tumor , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/pharmacokinetics , Cholinesterase Inhibitors/pharmacology , Cinnamates/chemical synthesis , Cinnamates/metabolism , Cinnamates/pharmacokinetics , Donepezil/chemical synthesis , Donepezil/metabolism , Donepezil/pharmacokinetics , Free Radical Scavengers/chemical synthesis , Free Radical Scavengers/metabolism , Free Radical Scavengers/pharmacokinetics , Free Radical Scavengers/pharmacology , Humans , Hydrazones/chemical synthesis , Hydrazones/metabolism , Hydrazones/pharmacokinetics , Ligands , Molecular Docking Simulation , Molecular Structure , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacokinetics , Protein Binding , Structure-Activity RelationshipABSTRACT
Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder that involves different pathogenic mechanisms. In this regard, the goal of this study was the design and synthesis of new compounds with multifunctional pharmacological activity by molecular hybridization of structural fragments of curcumin and resveratrol connected by an N-acyl-hydrazone function linked to a 1,4-disubstituted triazole system. Among these hybrid compounds, derivative 3e showed the ability to inhibit acetylcholinesterase activity, the intracellular formation of reactive oxygen species as well as the neurotoxicity elicited by Aß42 oligomers in neuronal SH-SY5Y cells. In parallel, compound 3e showed a good profile of safety and ADME parameters. Taken together, these results suggest that 3e could be considered a lead compound for the further development of AD therapeutics.
Subject(s)
Alzheimer Disease/drug therapy , Triazoles/chemistry , Triazoles/pharmacology , Acetylcholinesterase/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cells, Cultured , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacokinetics , Cholinesterase Inhibitors/pharmacology , Curcumin/pharmacokinetics , Curcumin/pharmacology , Humans , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Pharmacokinetics , Reactive Oxygen Species/metabolism , Resveratrol/pharmacokinetics , Resveratrol/pharmacology , Triazoles/pharmacokineticsABSTRACT
Stroke is the second cause of death and first cause of physical disability around the world; it affects the brain parenchyma through oxygen deficiency and spreads excitotoxicity. The complexity of the disease has made it difficult to find effective therapies. It is necessary to identify new treatments that effectively act within the narrow therapeutic window but also offer long-term protection poststroke. Our previous work found that oral linalool reversed the hippocampal and peripheral pro-inflammatory phospholipidomic biomarkers in ischemic rats; based on these observations, the "proof of concept" was to demonstrate that intranasal administration of linalool has a faster delivery to the central nervous system to protect it after focal ischemia in Wistar rats. The ischemic animals treated with linalool (25â¯mg/kg) showed a decrease in infarct volume at 24â¯h and seven days, and the treated animals had better neurological and motor skills at both poststroke times. Additionally, one month after daily intranasal administration of linalool, the ischemic rats showed improved relearning performance in the Morris water maze test. They also exhibited a reduction in microgliosis and decreased COX2, IL-1Beta and Nrf2 markers in the cerebral cortex and hippocampus. In astrocyte and microglial cultures, linalool reduced pro-inflammation and had a potent effect on microglial cells, generating Nrf2 subcellular redistribution under glutamate excitotoxicity conditions. Together, our findings indicate an acute and chronic recovery after ischemia induced by a daily intranasal puff of linalool, which mainly acts on microglial populations with anti-inflammatory actions.
Subject(s)
Acyclic Monoterpenes/administration & dosage , Acyclic Monoterpenes/pharmacology , Brain Ischemia/pathology , Microglia/drug effects , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Acyclic Monoterpenes/pharmacokinetics , Administration, Intranasal , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Brain Ischemia/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Intracellular Space/drug effects , Intracellular Space/metabolism , Male , Microglia/metabolism , Microglia/pathology , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacokinetics , Protein Transport/drug effects , Rats , Rats, Wistar , Tissue DistributionABSTRACT
OBJECTIVES: To evaluate the effects of combined treatment with granulocyte colony-stimulating factor (G-CSF) and methylprednisolone in rats subjected to experimental spinal cord injury. METHODS: Forty Wistar rats received a moderate spinal cord injury and were divided into four groups: control (no treatment); G-CSF (G-CSF at the time of injury and daily over the next five days); methylprednisolone (methylprednisolone for 24 h); and G-CSF/Methylprednisolone (methylprednisolone for 24 h and G-CSF at the time of injury and daily over the next five days). Functional evaluation was performed using the Basso, Beattie and Bresnahan score on days 2, 7, 14, 21, 28, 35 and 42 following injury. Motor-evoked potentials were evaluated. Histological examination of the spinal cord lesion was performed immediately after euthanasia on day 42. RESULTS: Eight animals were excluded (2 from each group) due to infection, a normal Basso, Beattie and Bresnahan score at their first evaluation, or autophagy, and 32 were evaluated. The combination of methylprednisolone and G-CSF promoted greater functional improvement than methylprednisolone or G-CSF alone (p<0.001). This combination also exhibited a synergistic effect, with improvements in hyperemia and cellular infiltration at the injury site (p<0.001). The groups displayed no neurophysiological differences (latency p=0.85; amplitude p=0.75). CONCLUSION: Methylprednisolone plus G-CSF promotes functional and histological improvements superior to those achieved by either of these drugs alone when treating spinal cord contusion injuries in rats. Combining the two drugs did have a synergistic effect.
Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacokinetics , Methylprednisolone/pharmacokinetics , Neuroprotective Agents/pharmacokinetics , Spinal Cord Injuries/drug therapy , Animals , Disease Models, Animal , Drug Combinations , Male , Random Allocation , Rats, Wistar , Recovery of Function/drug effects , Reference Values , Reproducibility of Results , Spinal Cord Injuries/pathology , Spinal Cord Injuries/rehabilitation , Time Factors , Treatment OutcomeABSTRACT
OBJECTIVES: To evaluate the effects of combined treatment with granulocyte colony-stimulating factor (G-CSF) and methylprednisolone in rats subjected to experimental spinal cord injury. METHODS: Forty Wistar rats received a moderate spinal cord injury and were divided into four groups: control (no treatment); G-CSF (G-CSF at the time of injury and daily over the next five days); methylprednisolone (methylprednisolone for 24 h); and G-CSF/Methylprednisolone (methylprednisolone for 24 h and G-CSF at the time of injury and daily over the next five days). Functional evaluation was performed using the Basso, Beattie and Bresnahan score on days 2, 7, 14, 21, 28, 35 and 42 following injury. Motor-evoked potentials were evaluated. Histological examination of the spinal cord lesion was performed immediately after euthanasia on day 42. RESULTS: Eight animals were excluded (2 from each group) due to infection, a normal Basso, Beattie and Bresnahan score at their first evaluation, or autophagy, and 32 were evaluated. The combination of methylprednisolone and G-CSF promoted greater functional improvement than methylprednisolone or G-CSF alone (p<0.001). This combination also exhibited a synergistic effect, with improvements in hyperemia and cellular infiltration at the injury site (p<0.001). The groups displayed no neurophysiological differences (latency p=0.85; amplitude p=0.75). CONCLUSION: Methylprednisolone plus G-CSF promotes functional and histological improvements superior to those achieved by either of these drugs alone when treating spinal cord contusion injuries in rats. Combining the two drugs did have a synergistic effect.
Subject(s)
Animals , Male , Spinal Cord Injuries/drug therapy , Methylprednisolone/pharmacokinetics , Granulocyte Colony-Stimulating Factor/pharmacokinetics , Neuroprotective Agents/pharmacokinetics , Reference Values , Spinal Cord Injuries/pathology , Spinal Cord Injuries/rehabilitation , Time Factors , Random Allocation , Reproducibility of Results , Treatment Outcome , Rats, Wistar , Recovery of Function/drug effects , Disease Models, Animal , Drug CombinationsABSTRACT
This study investigated the toxicity of rats exposed to lead acetate (AcPb) during the second phase of brain development (8-12 days postnatal) in hematological and cerebral parameters. Moreover, the preventive effect of zinc chloride (ZnCl2) and N-acetylcysteine (NAC) was investigated. Pups were injected subcutaneously with saline (0.9% NaCl solution), ZnCl2 (27 mg/kg/day), NAC (5 mg/kg/day) or ZnCl2 plus NAC for 5 days (3rd-7th postnatal days), and with saline (0.9% NaCl solution) or AcPb (7 mg/kg/day) in the five subsequent days (8th-12th postnatal days). Animals were sacrificed 21 days after the last AcPb exposure. Pups exposed to AcPb presented inhibition of blood porphobilinogen-synthase (PBG-synthase) activity without changes in hemoglobin content. ZnCl2 pre-exposure partially prevented PBG-synthase inhibition. Regarding neurotoxicity biomarkers, animals exposed to AcPb presented a decrease in cerebrum acetylcholinesterase (AChE) activity and an increase in Pb accumulation in blood and cerebrum. These changes were prevented by pre-treatment with ZnCl2, NAC, and ZnCl2 plus NAC. AcPb exposure caused no alteration in behavioral tasks. In short, results show that AcPb inhibited the activity of two important enzymatic biomarkers up to 21 days after the end of the exposure. Moreover, ZnCl2 and NAC prevented the alterations induced by AcPb.
Subject(s)
Acetylcysteine/therapeutic use , Cerebrum/drug effects , Chlorides/therapeutic use , Lead Poisoning, Nervous System/prevention & control , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Zinc Compounds/therapeutic use , Acetylcholinesterase/metabolism , Acetylcysteine/administration & dosage , Animals , Animals, Newborn , Biomarkers/blood , Biomarkers/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cerebrum/enzymology , Cerebrum/metabolism , Chlorides/administration & dosage , Chlorides/metabolism , Chlorides/pharmacokinetics , Drug Therapy, Combination , Environmental Pollutants/blood , Environmental Pollutants/metabolism , Environmental Pollutants/toxicity , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Injections, Subcutaneous , Lead/blood , Lead/metabolism , Lead/toxicity , Lead Poisoning, Nervous System/blood , Lead Poisoning, Nervous System/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/enzymology , Neurons/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacokinetics , Organometallic Compounds/administration & dosage , Porphobilinogen Synthase/antagonists & inhibitors , Porphobilinogen Synthase/blood , Random Allocation , Rats, Wistar , Tissue Distribution/drug effects , Toxicokinetics , Zinc Compounds/administration & dosage , Zinc Compounds/metabolism , Zinc Compounds/pharmacokineticsABSTRACT
Quercetin has been identified as a promising compound with a neuroprotective potential against age-related neurodegenerative diseases such as Alzheimer's disease (AD). Nevertheless, the clinical application of quercetin is hampered by its low oral bioavailability. The aim of this work was to evaluate the capability of nanoencapsulated quercetin in zein nanoparticles (NPQ), that significantly improves the oral absorption and bioavailability of the flavonoid, as potential oral treatment for AD. For this purpose, SAMP8 mice were orally treated for two months with either NPQ (25mg/kg every 48h) or a solution of quercetin (Q; 25mg/kg daily). NPQ displayed a size of 260nm and a payload of about 70µg/mg. For Q, no significant effects were observed in animals. On the contrary, the oral administration of NPQ improved the cognition and memory impairments characteristics of SAMP8 mice. These observations appeared to be related with a decreased expression of the hippocampal astrocyte marker GFAP. Furthermore, significant levels of quercetin were quantified in the brain of mice treated with nanoparticles. These findings highlight the potential of zein nanoparticles to promote the oral absorption of quercetin as well as the therapeutic potential of this flavonoid in AD pathogenesis.
Subject(s)
Alzheimer Disease/drug therapy , Behavior, Animal/drug effects , Drug Carriers/chemistry , Motor Activity/drug effects , Neuroprotective Agents/therapeutic use , Quercetin/therapeutic use , 2-Hydroxypropyl-beta-cyclodextrin , Administration, Oral , Alzheimer Disease/psychology , Animals , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Male , Maze Learning/drug effects , Mice, Inbred Strains , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacokinetics , Particle Size , Quercetin/administration & dosage , Quercetin/pharmacokinetics , Rotarod Performance Test , Zein/chemistry , beta-Cyclodextrins/chemistryABSTRACT
In addition to its intracellular roles, the nucleoside guanosine (GUO) also has extracellular effects that identify it as a putative neuromodulator signaling molecule in the central nervous system. Indeed, GUO can modulate glutamatergic neurotransmission, and it can promote neuroprotective effects in animal models involving glutamate neurotoxicity, which is the case in brain ischemia. In the present study, we aimed to investigate a new in vivo GUO administration route (intranasal, IN) to determine putative improvement of GUO neuroprotective effects against an experimental model of permanent focal cerebral ischemia. Initially, we demonstrated that IN [(3)H] GUO administration reached the brain in a dose-dependent and saturable pattern in as few as 5 min, presenting a higher cerebrospinal GUO level compared with systemic administration. IN GUO treatment started immediately or even 3 h after ischemia onset prevented behavior impairment. The behavior recovery was not correlated to decreased brain infarct volume, but it was correlated to reduced mitochondrial dysfunction in the penumbra area. Therefore, we showed that the IN route is an efficient way to promptly deliver GUO to the CNS and that IN GUO treatment prevented behavioral and brain impairment caused by ischemia in a therapeutically wide time window.
Subject(s)
Brain Ischemia/drug therapy , Guanosine/administration & dosage , Guanosine/therapeutic use , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Stroke/drug therapy , Administration, Intranasal , Animals , Behavior, Animal , Brain Ischemia/psychology , Cerebral Infarction/pathology , Cerebral Infarction/prevention & control , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Guanosine/cerebrospinal fluid , Guanosine/pharmacokinetics , Male , Mitochondria/drug effects , Neuroprotective Agents/cerebrospinal fluid , Neuroprotective Agents/pharmacokinetics , Rats , Rats, Wistar , Stroke/psychologyABSTRACT
OBJECTIVE: To determine the pharmacokinetics (PK) and placental transfer of intravenous (i.v.) N-acetylcysteine (NAC) in mothers with a clinical diagnosis of chorioamnionitis (CA) and determine the PK of i.v. NAC in their infants. STUDY DESIGN: In this prospective, double-blind study i.v. NAC 100 mg/kg/dose or saline was administered within 4 hours of CA diagnosis to pregnant women ≥24 weeks' gestation and then every 6 hours until delivery. Maternal PK and placental transfer were determined with maternal blood and matched maternal and cord venous blood. Neonatal PK estimates were determined from i.v. NAC (12.5-25 mg/kg/dose) administered every 12 hours for 5 doses. Noncompartmental analyses were performed for maternal and neonatal PK estimates. RESULTS: Eleven mothers (5 preterm, 6 near-term) and 12 infants (1 set of twins) received NAC. Maternal clearance (CL) of NAC was faster than in nonpregnant adults, with a terminal elimination half-life of 1.2 ± 0.2 hours. The NAC cord to maternal ratio was 1.4 ± 0.8, suggesting rapid placental transfer and slower rate of fetal CL. Neonatal PK estimates for near-term compared with preterm infants showed a significantly shorter terminal elimination half-life (5.1 vs 7.5 hours, respectively) and greater CL (53.7 vs 45.0 mL/h/kg, respectively). CONCLUSIONS: Maternal CL and placental transfer of NAC was rapid, with umbilical cord concentrations frequently exceeding maternal concentrations. The administration of NAC to mothers with CA achieves predictable NAC plasma concentrations in the fetus, indicating that antenatal neuroprotection may be possible for these newborns at high risk for neuroinflammation.
Subject(s)
Acetylcysteine/pharmacokinetics , Chorioamnionitis/drug therapy , Neuroprotective Agents/pharmacokinetics , Placenta/drug effects , Double-Blind Method , Female , Fetal Blood/drug effects , Free Radical Scavengers/pharmacokinetics , Humans , Inflammation/drug therapy , Infusions, Intravenous , Male , Maternal-Fetal Exchange , Mothers , Pilot Projects , Pregnancy , Prospective Studies , Time FactorsABSTRACT
Although historically used for the treatment of anemia, erythropoietin (EPO) has emerged as a neurotrophic and neuroprotective agent in different conditions of neuronal damage (traumatic brain injury, ischemia, spinal cord compression, peripheral neuropathy, retinal damage, epilepsy, Parkinson's Disease, among others). Nonetheless, EPO's therapeutic application is limited due to its hematological side-effects. With the aim of obtaining EPO derivatives resembling the hormone isolated from cells and tissues of neural origin, a novel combination of less acidic EPO glycoforms -designated as neuroepoetin (rhNEPO)- was purified to homogeneity from the supernatant of a CHO-producing cell line by a four-step chromatographic procedure. This simple and single process allowed us to prepare two EPO derivatives with distinct therapeutic expectations: the hematopoietic version and a minimally hematopoietic, but mainly in vitro cytoprotective, alternative. Further biological characterization showed that the in vivo erythropoietic activity of rhNEPO was 25-times lower than that of rhEPO. Interestingly, using different in vitro cytoprotective assays we found that this molecule exerts cytoprotection equivalent to, or better than, that of rhEPO in cells of neural phenotype. Furthermore, despite its shorter plasma half-life, rhNEPO was rapidly absorbed and promptly detected in the cerebrospinal fluid after intravenous administration in rats (5 min postinjection, in comparison with 30 min for rhEPO). Therefore, our results support the study of neuroepoetin as a potential drug for the treatment of neurological diseases, combining high cytoprotective activity with reduced hematological side-effects.
Subject(s)
Erythropoietin/isolation & purification , Protein Isoforms/isolation & purification , Animals , CHO Cells , Cell Survival/drug effects , Cricetinae , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Erythropoietin/metabolism , Erythropoietin/pharmacokinetics , Erythropoietin/pharmacology , Female , Humans , Isoelectric Focusing , Neurons/cytology , Neurons/drug effects , Neuroprotective Agents/isolation & purification , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , PC12 Cells , Protein Isoforms/metabolism , Protein Isoforms/pharmacology , Rats , Rats, WistarABSTRACT
In this work we describe the protective effects of quercetin against H(2)O(2) in 24-h-pretreated neuronal cultures. We explored quercetin availability and subcellular fate through the use of HPLC-Diode Array Detection (DAD), epifluorescence, and confocal microscopy. We focused on quercetin modulation of thiol-redox systems by evaluating changes in mitochondrial thioredoxin Trx2, the levels of total glutathione (GSH), and the expression of the gamma-glutamate-cysteine ligase catalytic subunit (GCLC), the rate-limiting enzyme of GSH synthesis, by the use of Western blot, HPLC, and real-time PCR techniques, respectively. We further explored the activation of the protective NF-E2-related factor 2 (Nrf2)-dependent signaling pathway by quercetin using immunocytochemistry techniques. Our results showed rapid quercetin internalization into neurons, reaching the nucleus after its addition to the culture. Quercetin pretreatment increased total GSH levels, but did not increase Trx2. Interestingly it caused Nrf2 nuclear translocation and significantly increased GCLC gene expression. At the moment of H(2)O(2) addition, intracellular quercetin or related metabolites were undetectable in the cultures although quercetin pretreatment prevented neuronal death from the oxidant exposure. Our findings suggest alternative mechanisms of quercetin neuroprotection beyond its long-established ROS scavenging properties, involving Nrf2-dependent modulation of the GSH redox system.
Subject(s)
Cell Nucleus/drug effects , Cytoprotection/drug effects , Glutathione/metabolism , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Quercetin/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Antioxidants/pharmacokinetics , Antioxidants/pharmacology , Cell Death/drug effects , Cell Nucleus/metabolism , Drug Evaluation, Preclinical , Hydrogen Peroxide/toxicity , Neurons/metabolism , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Protein Transport/drug effects , Quercetin/pharmacokinetics , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effectsABSTRACT
Retinal ischemia could provoke blindness and there is no effective treatment against retinal ischemic damage. Brief intermittent ischemia applied during the onset of reperfusion (i.e., post-conditioning) protects the retina from ischemia/reperfusion injury. Multiple evidences support that glutamate is implicated in retinal ischemic damage. We investigated the involvement of glutamate clearance in post-conditioning-induced protection. For this purpose, ischemia was induced by increasing intra-ocular pressure for 40 min, and 5 min after reperfusion, animals underwent seven cycles of 1 min/1 min ischemia/reperfusion. One, three, or seven days after ischemia, animals were subjected to electroretinography and histological analysis. The functional and histological protection induced by post-conditioning was evident at 7 (but not 1 or 3) days post-ischemia. An increase in Müller cell glial fibrillary acidic protein (GFAP) levels was observed at 1, 3, and 7 days after ischemia, whereas post-conditioning reduced GFAP levels of Müller cells at 3 and 7 days post-ischemia. Three days after ischemia, a significant decrease in glutamate uptake and glutamine synthetase activity was observed, whereas post-conditioning reversed the effect of ischemia. The intravitreal injection of supraphysiological levels of glutamate mimicked electroretinographic and histological alterations provoked by ischemia, which were abrogated by post-conditioning. These results support the involvement of glutamate in retinal protection against ischemia/reperfusion damage induced by post-conditioning.
Subject(s)
Glutamic Acid/pharmacokinetics , Ischemic Preconditioning , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Retina/pathology , Animals , Electroretinography , Glial Fibrillary Acidic Protein/metabolism , Glutamate-Ammonia Ligase/metabolism , Glutaminase/metabolism , Glutamine/pharmacokinetics , Intraocular Pressure , Male , Neuroprotective Agents/pharmacokinetics , Rats , Rats, Wistar , Reperfusion Injury/metabolism , Retina/physiology , TritiumABSTRACT
Resveratrol is a phytoalexin structurally related to stilbenes, which is synthesized in considerable amounts in the skin of grapes, raspberries, mulberries, pistachios and peanuts, and by at least 72 medicinal and edible plant species in response to stress conditions. It was isolated in 1940 and did not maintain much interest for around five decades until its role in treatment of cardiovascular diseases was suggested. To date, resveratrol has been identified as an agent that may be useful to treat cancer, pain, inflammation, tissue injury, and other diseases. However, currently the attention is being focused in analyzing its properties against neurodegenerative diseases and as antiaging compound. It has been reported that resveratrol shows effects in in vitro models of epilepsy, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and nerve injury. However, evidences in vivo as well as in human beings are still lacking. Thus, further investigations on the pharmacological effects of resveratrol in vivo are necessary before any conclusions on its effects on neurodegenerative diseases can be obtained.