Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Exp Gerontol ; 102: 3-11, 2018 02.
Article in English | MEDLINE | ID: mdl-29174969

ABSTRACT

With upcoming age, the capability to fight against harmful stimuli decreases and the organism becomes more susceptible to infections and diseases. Here, the objective was to demonstrate the effect of dietary resveratrol in aged mice in potentiating brain defenses against LipoPolySaccharide (LPS). Acute LPS injection induced a strong proinflammatory effect in 24-months-old C57/BL6 mice hippocampi, increasing InterLeukin (Il)-6, Tumor Necrosis Factor-alpha (Tnf-α), Il-1ß, and C-X-C motif chemokine (Cxcl10) gene expression levels. Resveratrol induced higher expression in those cytokines regarding to LPS. Oxidative Stress (OS) markers showed not significant changes after LPS or resveratrol, although for resveratrol treated groups a slight increment in most of the parameters studies was observed, reaching signification for NF-kB protein levels and iNOS expression. However, Endoplasmic Reticulum (ER) stress markers demonstrated significant changes in resveratrol-treated mice after LPS treatment, specifically in eIF2α, BIP, and ATF4. Moreover, as described, resveratrol is able to inhibit the mechanistic Target of Rapamycin (mTOR) pathway and this effect could be linked to (eIF2α) phosphorylation and the increase in the expression of the previously mentioned proinflammatory genes as a response to LPS treatment in aged animals. In conclusion, resveratrol treatment induced a different cellular response in aged animals when they encountered acute inflammatory stimuli.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Hippocampus/drug effects , Inflammation Mediators/metabolism , Inflammation/prevention & control , Resveratrol/pharmacology , Age Factors , Aging , Animals , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factor-2B/metabolism , Gene Expression Regulation , Hippocampus/metabolism , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Lipopolysaccharides , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Phosphorylation , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
2.
Mol Neurobiol ; 54(3): 1661-1676, 2017 04.
Article in English | MEDLINE | ID: mdl-26873850

ABSTRACT

Metabolic stress induced by high-fat (HF) diet leads to cognitive dysfunction and aging, but the physiological mechanisms are not fully understood. Senescence-accelerated prone mouse (SAMP8) models were conducted under metabolic stress conditions by feeding HF for 15 weeks, and the preventive effect of resveratrol was studied. This dietary strategy demonstrates cognitive impairment in SAMP8-HF and significant preventive effect by resveratrol-treated animals. Hippocampal changes in the proteins involved in mitochondrial dynamics optic atrophy-1 protein (OPA1) and mitofusin 2 (MFN2) comprised a differential feature found in SAMP8-HF that was prevented by resveratrol. Electronic microscopy showed a larger mitochondria in SAMP8-HF + resveratrol (SAMP8-HF + RV) than in SAMP8-HF, indicating increases in fusion processes in resveratrol-treated mice. According to the mitochondrial morphology, significant increases in the I-NDUFB8, II-SDNB, III-UQCRC2, and V-ATPase complexes, in addition to that of voltage-dependent anion channel 1 (VDAC1)/porin, were found in resveratrol-treated animals with regard to SAMP8-HF, reaching control-animal levels. Moreover, tumor necrosis factor alpha (TNF-α) and interleukin (IL-6) were increased after HF, and resveratrol prevents its increase. Moreover, we found that the HF diet affected the Wnt pathway, as demonstrated by ß-catenin inactivation and modification in the expression of several components of this pathway. Resveratrol induced strong activation of ß-catenin. The metabolic stress rendered in the cognitive and cellular pathways altered in SAMP8 focus on different targets in order to act on preventing cognitive impairment in neurodegeneration, and resveratrol can offer therapeutic possibilities for preventive strategies in aging or neurodegenerative conditions.


Subject(s)
Brain/drug effects , Cellular Senescence/drug effects , Mitochondria/drug effects , Stilbenes/pharmacology , Stress, Physiological/drug effects , Wnt Signaling Pathway/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Brain/physiology , Cellular Senescence/physiology , Diet, High-Fat/adverse effects , Male , Mice , Mitochondria/physiology , Neuroprotective Agents/pharmacology , Random Allocation , Resveratrol , Wnt Signaling Pathway/physiology
3.
Age (Dordr) ; 37(1): 9747, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25663420

ABSTRACT

The amyloid precursor protein/presenilin 1 (APP/PS1) mouse model of Alzheimer's disease (AD) has provided robust neuropathological hallmarks of familial AD-like pattern at early ages, whereas senescence-accelerated mouse prone 8 (SAMP8) has a remarkable early senescence phenotype with pathological similarities to AD. The aim of this study was the investigation and characterization of cognitive and neuropathological AD markers in a novel mouse model that combines the characteristics of the APP/PS1 transgenic mouse model with a senescence-accelerated background of SAMP8 mice. Initially, significant differences were found regarding amyloid plaque formation and cognitive abnormalities. Bearing these facts in mind, we determined a general characterization of the main AD brain molecular markers, such as alterations in amyloid pathway, neuroinflammation, and hyperphosphorylation of tau in these mice along their lifetimes. Results from this analysis revealed that APP/PS1 in SAMP8 background mice showed alterations in the pathways studied in comparison with SAMP8 and APP/PS1, demonstrating that a senescence-accelerated background exacerbated the amyloid pathology and maintained the cognitive dysfunction present in APP/PS1 mice. Changes in tau pathology, including the activity of cyclin-dependent kinase 5 (CDK5) and glycogen synthase kinase 3 ß (GSK3ß), differs, but not in a parallel manner, with amyloid disturbances.


Subject(s)
Aging/physiology , Alzheimer Disease/etiology , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/physiology , Presenilin-1/physiology , tau Proteins/physiology , Aging/pathology , Alzheimer Disease/metabolism , Animals , Disease Models, Animal , Mice , Mice, Transgenic , Phenotype
4.
J Physiol Pharmacol ; 65(2): 229-39, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24781732

ABSTRACT

The autophagic process is a lysosomal degradation pathway, which is activated during stress conditions, such as starvation or exercise. Regular exercise has beneficial effects on human health, including neuroprotection. However, the cellular mechanisms underlying these effects are incompletely understood. Endurance and a single bout of exercise induce autophagy not only in brain but also in peripheral tissues. However, little is known whether autophagy could be modulated in brain and peripheral tissues by long-term moderate exercise. Here, we examined the effects on macroautophagy process of long-term moderate treadmill training (36 weeks) in adult rats both in brain (hippocampus and cerebral cortex) and peripheral tissues (skeletal muscle, liver and heart). We assessed mTOR activation and the autophagic proteins Beclin 1, p62, LC3B (LC3B-II/LC3B-I ratio) and the lysosomal protein LAMP1, as well as the ubiquitinated proteins. Our results showed in the cortex of exercised rats an inactivation of mTOR, greater autophagy flux (increased LC3-II/LC3-I ratio and reduced p62) besides increased LAMP1. Related with these effects a reduction in the ubiquitinated proteins was observed. No significant changes in the autophagic pathway were found either in hippocampus or in skeletal and cardiac muscle by exercise. Only in the liver of exercised rats mTOR phosphorylation and p62 levels increased, which could be related with beneficial metabolic effects in this organ induced by exercise. Thus, our findings suggest that long-term moderate exercise induces autophagy specifically in the cortex.


Subject(s)
Autophagy/physiology , Cerebral Cortex/metabolism , Liver/metabolism , Physical Conditioning, Animal/physiology , Animals , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Heat-Shock Proteins/metabolism , Hippocampus/metabolism , Lysosomal Membrane Proteins/metabolism , Male , Microtubule-Associated Proteins/metabolism , Muscle, Skeletal/metabolism , Myocardium/metabolism , Rats, Sprague-Dawley , Sequestosome-1 Protein , TOR Serine-Threonine Kinases/metabolism
5.
Brain Res ; 1543: 38-48, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24183784

ABSTRACT

An active lifestyle involving regular exercise reduces the deleterious effects of the aging process. At the cerebral level, both synaptic plasticity and neurogenesis are modulated by exercise, although the molecular mechanisms underlying these effects are not clearly understood. In the mature nervous system, the canonical Wnt (Wnt/ß-catenin) signaling pathway is implicated in neuroprotection and synaptic plasticity. Here, we examined whether the Wnt pathway could be modulated in adult male rat hippocampus by long-term moderate exercise (treadmill running) or enrichment (handling/environmental stimulation). Sedentary animals showed higher protein levels of the Wnt antagonist, Dkk-1, the lowest levels being found in the exercised group. Although there was no evidence of any changes in activation of the LRP6 receptor, the total levels of LRP6 were higher in exercised and enriched animals. Analysis of some of the components implicated in the phosphorylation of ß-catenin, which leads ultimately to its proteasomal degradation, revealed higher levels and activation of Axin1 and GSK-3α/ß respectively in sedentary animals. However neither different phosphorylated forms nor total ß-catenin protein levels differed between the experimental groups. Higher protein levels of Axin2 and the antiapoptotic protein, Bcl-2, were found with exercise and handling, whereas the proapototic, Bax, was unaffected. Thus, our results suggest activation of the Wnt pathway not only with moderate exercise, but also with the handling of the animals.


Subject(s)
Gene Expression Regulation/physiology , Physical Conditioning, Animal/physiology , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , Adaptor Proteins, Signal Transducing , Analysis of Variance , Animals , Carrier Proteins/metabolism , Exercise Test , Intercellular Signaling Peptides and Proteins/metabolism , Male , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Time , Wnt Proteins/genetics , beta Catenin/metabolism
6.
Exp Gerontol ; 47(12): 925-35, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22940286

ABSTRACT

The protein deacetylase, sirtuin 1, is suggested as a master regulator of exercise-induced beneficial effects. Sirtuin 1 modulates mitochondrial biogenesis, primarily via its ability to deacetylate and activate proliferator-activated receptor-γ coactivator-1α (PGC-1α), interacting with AMPK kinase. Redox cell status can also influence this regulatory axis and together they form an important convergence point in hormesis during the aging process. Here, we tested whether treadmill training (36weeks), as a paradigm of long-term moderate exercise, modifies the AMPK-sirtuin 1-PGC-1α axis and redox balance in rat gastrocnemius muscle, liver and heart. Physical activity induced increases in sirtuin 1 protein levels in all the aged rat tissues studied, as well as total PGC-1α levels. However, no changes in AMPK activation or significant differences in mitochondrial biogenesis (by measuring electron transport chain protein content) were found after exercise training. Parallel to these changes, we observed an improvement of oxidative stress defenses, mainly in muscle, with modification of the antioxidant enzyme machinery resulting in a reduction in lipid peroxidation and protein carbonylation. Thus, we demonstrate that moderate long-term exercise promotes tissue adaptations, increasing muscle, liver and heart sirtuin 1 protein content and activity and increasing PGC-1α protein expression. However, AMPK activation or mitochondrial biogenesis is not modified, but it cannot be discarded that its participation in the adaptive mechanism which prevents the development of the deleterious effects of age.


Subject(s)
Oxidative Stress/physiology , Physical Conditioning, Animal/physiology , Sirtuin 1/biosynthesis , AMP-Activated Protein Kinase Kinases , Animals , Liver/metabolism , Male , Mitochondrial Turnover/physiology , Muscle, Skeletal/metabolism , Myocardium/metabolism , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Kinases/metabolism , RNA-Binding Proteins/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Transcription Factors/metabolism
7.
J Appl Physiol (1985) ; 111(5): 1380-90, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21817108

ABSTRACT

Exercise enhances general health. However, its effects on neurodegeneration are controversial, and the molecular pathways in the brain involved in this enhancement are poorly understood. Here, we examined the effect of long-term moderate treadmill training on adult male rat cortex and hippocampus to identify the cellular mechanisms behind the effects of exercise. We compared three animal groups: exercised (30 min/day, 12 m/min, 5 days/wk, 36 wk), handled but nonexercised (treadmill handling procedure, 0 m/min), and sedentary (nonhandled and nonexercised). Moderate long-term exercise induced an increase in IGF-1 levels and also in energy parameters, such as PGC-1α and the OXPHOS system. Moreover, the sirtuin 1 pathway was activated in both the exercised and nonexercised groups but not in sedentary rats. This induction could be a consequence of exercise as well as the handling procedure. To determine whether the long-term moderate treadmill training had neuroprotective effects, we studied tau hyperphosphorylation and GSK3ß activation. Our results showed reduced levels of phospho-tau and GSK3ß activation mainly in the hippocampus of the exercised animals. In conclusion, in our rodent model, exercise improved several major brain parameters, especially in the hippocampus. These improvements induced the upregulation of sirtuin 1, a protein that extends life, the stimulation of mitochondrial biogenesis, the activation of AMPK, and the prevention of signs of neurodegeneration. These findings are consistent with other reports showing that physical exercise has positive effects on hormesis.


Subject(s)
Cerebral Cortex/metabolism , Hippocampus/metabolism , Neuroprotective Agents/metabolism , Physical Conditioning, Animal/physiology , AMP-Activated Protein Kinase Kinases , Animals , Body Weight/physiology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Insulin-Like Growth Factor I/metabolism , Male , Mitochondria/metabolism , Neurodegenerative Diseases/prevention & control , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Kinases/metabolism , RNA-Binding Proteins/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Sirtuin 1/metabolism , Transcription Factors/metabolism , tau Proteins/metabolism
8.
Hippocampus ; 20(8): 962-70, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19714564

ABSTRACT

Glycogen synthase kinase-3beta (GSK-3beta) is a crucial component in the cascade of events that culminate in a range of neurodegenerative diseases. It is controlled by several pathways, including calpain-mediated cleavage. Calpain mediates in cell death induced by 3-nitropropionic acid (3-NP), but GSK-3beta regulation has not been demonstrated. Here we studied changes in total GSK-3beta protein levels and GSK-3beta phosphorylation at Ser-9 in this model. The 3-NP treatment induced GSK-3beta truncation. This regulation was dependent on calpain activation, since addition of calpeptin to the medium prevented this cleavage. While calpain inhibition prevented 3-NP-induced neuronal loss, inhibition of GSK-3beta by SB-415286 did not. Furthermore, inhibition of cdk5, a known target of calpain involved in 3-NP-induced cell death, also failed to rescue neurons in our model. Our results point to a new target of calpain and indicate possible cross-talk between calpain and GSK-3beta in the 3-NP toxicity pathway. On the basis of our findings, we propose that calpain may modulate 3-NP-induced neuronal loss.


Subject(s)
Calpain/metabolism , Convulsants/toxicity , Glycogen Synthase Kinase 3/metabolism , Neurodegenerative Diseases , Neurons/drug effects , Nitro Compounds/toxicity , Propionates/toxicity , Amino Acid Chloromethyl Ketones/pharmacology , Aminophenols/pharmacology , Animals , Calpain/pharmacology , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Glycogen Synthase Kinase 3 beta , Hippocampus/cytology , Male , Maleimides/pharmacology , Mice , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neuroprotective Agents/pharmacology , Purines/pharmacology , Rats , Roscovitine , Signal Transduction/drug effects , Time Factors
9.
Neuropharmacology ; 56(2): 422-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18948125

ABSTRACT

Lithium reduced striatal neurodegeneration induced in rats by 3-nitropropionic acid inhibiting calpain activation. Lithium prevented an increase in cdk5 activity, as shown by the levels of the co-activator p35. Myocite enhancer factor 2 (MEF2), a downstream substrate for cdk5 with pro-survival activity, showed increased phosphorylation. In primary cultures of neurons treated with 3-NP, lithium also reduced protease activity mediated by calpain, cdk5 activation and cellular death. These observations indicate that lithium has a neuroprotective effect. Lithium treatment also reduced the intracellular increase in calcium induced by 3-NP. The finding that lithium mediates the modulation of the calpain/cdk5 pathway further supports its use in the treatment of neurodegenerative diseases.


Subject(s)
Calpain/metabolism , Cyclin-Dependent Kinase 5/metabolism , Huntington Disease , Lithium Chloride/therapeutic use , Neuroprotective Agents/therapeutic use , Nitro Compounds , Propionates , Signal Transduction/drug effects , Animals , Calcium/metabolism , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Gene Expression Regulation/drug effects , Hippocampus/cytology , Huntington Disease/chemically induced , Huntington Disease/drug therapy , Huntington Disease/metabolism , Male , Mice , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Succinate Dehydrogenase/metabolism
10.
Neuroscience ; 154(4): 1388-97, 2008 Jul 17.
Article in English | MEDLINE | ID: mdl-18538940

ABSTRACT

We examined the expression of SIRT1 in several experimental paradigms of human pathologies. We used a neuroblastoma cell line (B65), neuronal primary cultures (hippocampus and cerebellar granule cells) and in vivo approaches in rat and senescence murine models (SAM). Cell cultures and rats were treated with several well-know neurotoxins, i.e. rotenone, MPP(+), kainate and 3-nitropropionic acid. Subsequently, SIRT1 expression was compared in these different paradigms of neurotoxicity. The pattern of expression of SIRT1 in proliferating cell cultures (B65) was different to that in quiescent cell cultures. In the murine model of senescence (senescence-accelerated mice prone, SAMP8), SIRT1 expression progressively decreased, while in the control strain (senescence-accelerated mice resistant, SAMR1) it increased. Finally, we studied human samples of Parkinson's disease (PD), dementia with Lewy bodies (DLB) and Huntington's diseases (HD). SIRT1 expression decreased dramatically in HD, but there were no significant changes in Parkinson-related illnesses. In conclusion, SIRT1 expression may be a good sensor of toxic neuronal processes.


Subject(s)
Aging/metabolism , Neurodegenerative Diseases/metabolism , Sirtuins/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Blotting, Western , Cells, Cultured , Flow Cytometry , Humans , Male , Mice , Neurons/drug effects , Neurons/metabolism , Neurotoxins/pharmacology , Rats , Rats, Sprague-Dawley , Sirtuin 1 , Sirtuins/drug effects
11.
Neuroscience ; 147(3): 746-56, 2007 Jul 13.
Article in English | MEDLINE | ID: mdl-17583434

ABSTRACT

The mechanism involved in neuronal apoptosis is largely unknown. Studies performed on neuronal cell cultures provide information about the pathways which orchestrate the process of neuronal loss and potential drugs for the treatment of neurological disorders. In the present study we select resveratrol, a natural antioxidant, as a potential drug for the treatment of neurodegenerative diseases. We evaluate the neuroprotective effects of resveratrol in two apoptotic models in rat cerebellar granule neurons (CGNs): the inhibition of mitochondrial complex I using 1-methyl-4-phenylpyridinium (MPP(+)) (an in vitro model of Parkinson's disease) and serum potassium withdrawal. We study the role of the mammalian silent information regulator 2 (SIRT1) in the process of neuroprotection mediated by resveratrol. Because recent studies have demonstrated that SIRT1 is involved in cell survival and has antiaging properties, we also measured changes in the expression of this protein after the addition of these two apoptotic stimuli. MPP(+)--induced loss of cell viability and apoptosis in CGNs was prevented by the addition of RESV (1 microM to 100 microM). However, the neuroprotective effects were not mediated by the activation of SIRT1, since sirtinol-an inhibitor of this enzyme--did not attenuate them. Furthermore MPP(+) decreases the protein expression of SIRT1. RESV did not prevent serum potassium withdrawal-induced apoptosis although it did completely attenuate oxidative stress production by these apoptotic stimuli. Furthermore, serum potassium withdrawal increases the expression of SIRT1. Our results indicate that the antiapoptotic effects of RESV in MPP(+) are independent of the stimulation of SIRT1 and depend on its antioxidant properties. Furthermore, because SIRT1 is involved in neuronal survival depending on the apoptotic stimuli, changes in the expression of SIRT1 could be involved in the regulation of the apoptotic route.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Cerebellum/cytology , Electron Transport Complex I/metabolism , Neurons/drug effects , Potassium/metabolism , Stilbenes/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/genetics , Gene Expression Regulation/drug effects , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Resveratrol , Reverse Transcriptase Polymerase Chain Reaction/methods , Sirtuin 1 , Sirtuins/metabolism , Time Factors
12.
Neurochem Int ; 50(2): 435-42, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17116346

ABSTRACT

Acute treatment with kainate 30 mg/kg (KA) produced behavioral alterations and reactive gliosis. However, it did not produce major death of mouse hippocampal neurons, indicating that concentrations were not cytotoxic. KA caused rapid and temporal Erk phosphorylation (at 6h) and Akt dephosphorylation (1-3 days). Concomitantly, the activation of GSK3beta was increased 1-3 days after KA. After 7 days, a reduction in GSK3beta activation was observed. Caspase-3 activity increased, but to a lesser extent than calpain activation (measured by fluorimetry and calpain-cleaved alpha-spectrin). As calpain is involved in cdk5 activation, and cdk5 is related to GSK3beta, the cdk5/p25 pathway was examined. Results showed that the p25/p35 ratio in KA-injected mice for 3 days was 73.6% higher than control levels. However, no changes in cdk5 expression were detected. Both Western blot and immunohistochemistry against p-Tau(Thr(231)) indicated an increase at this phosphorylated site of tau protein. Indeed an increase in p-Tau(Ser(199)) and p-Tau(Ser(396)) was observed by Western blot. Our results demonstrate that tau hyperphosphorylation, induced by KA, is due to an increase in GSK3beta/cdk5 activity in combination with an inactivation of Akt. This indicates that the calpain/cdk5 pathway for tau phosphorylation has a potential role in delayed apoptotic death evoked by excitotoxicity. Moreover, the subsequent activation of caspase and calpain proteases leads to dephosphorylation of tau, thus increasing microtubular destructuration. Taken together, our results provide new insights in the activation of several kinase-pathways implicated in cytoskeletal alterations that are a common feature of neurodegenerative diseases.


Subject(s)
Cyclin-Dependent Kinase 5/biosynthesis , Extracellular Signal-Regulated MAP Kinases/biosynthesis , Glycogen Synthase Kinase 3/biosynthesis , Hippocampus/metabolism , Kainic Acid/pharmacology , Oncogene Protein v-akt/biosynthesis , tau Proteins/metabolism , Actins/biosynthesis , Animals , Apoptosis/drug effects , Blotting, Western , Calpain/metabolism , Caspases/metabolism , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Hippocampus/drug effects , Immunohistochemistry , Male , Mice , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/metabolism , Phosphorylation
13.
Neuroscience ; 134(3): 1001-11, 2005.
Article in English | MEDLINE | ID: mdl-15979805

ABSTRACT

Although numerous studies have demonstrated a neuroprotective and anti-apoptotic role of lithium in neuronal cell cultures, the precise mechanism by which this occurs, remains to be elucidated. In this study, we evaluated the lithium-mediated neuroprotection against colchicine-induced apoptosis in cultured cerebellar granule neurons. Previously, it has been demonstrated that colchicine mediates apoptosis in cerebellar granule neurons through cytoskeletal alteration and activation of an intrinsic pro-apoptotic pathway. Recently we also demonstrated a potential role of cyclin-dependent kinase 5 (cdk5) in this pathway. Here we report that colchicine induces dephosphorylation in Ser-9 and phosphorylation in Tyr-216, and thus activation, of glycogen synthase kinase-3beta in cerebellar granule neurons, and that this modification is inhibited by the presence of 5 mM lithium. However, the selective glycogen synthase kinase-3beta inhibitors SB-415286 and SB-216763 were unable to prevent colchicine-induced apoptosis in these cells, suggesting that the anti-apoptotic activity of lithium is not mediated by glycogen synthase kinase-3beta under these conditions. On the other hand, 5 mM lithium prevented the colchicine-induced increase in cdk5 expression and breakdown of cdk5/p35 to cdk5/p25. In addition, we show that up-regulation of cdk5/p25 is unrelated to inhibition of the activity of myocyte enhancer factor 2, a pro-survival transcription factor. These data suggest a previously undescribed neuroprotective mechanism of lithium associated with the modulation of cdk5/p35 or cdk5/p25 expression.


Subject(s)
Cerebellum/cytology , Cyclin-Dependent Kinases/metabolism , Lithium/administration & dosage , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Aminophenols/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Apoptosis/drug effects , Blotting, Western/methods , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Colchicine/pharmacology , Culture Media, Serum-Free/pharmacology , Cyclin-Dependent Kinase 5 , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Flow Cytometry/methods , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Indoles/pharmacology , MEF2 Transcription Factors , Maleimides/pharmacology , Microscopy, Electron, Transmission/methods , Myogenic Regulatory Factors , Neurons/ultrastructure , Potassium Deficiency , Rats , Rats, Sprague-Dawley , Serine/metabolism , Threonine/metabolism , Time Factors , Transcription Factors/metabolism
14.
Cell Death Differ ; 12(8): 1124-33, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15947794

ABSTRACT

The use of neural progenitor cells (NPCs) is limited by the incomplete knowledge of the extracellular signals regulating their proliferation and survival. We report that cultured mouse NPCs express functional mGlu3 and mGlu5 metabotropic glutamate receptors. Pharmacological blockade of both receptors reduced NPC proliferation and survival, whereas activation of mGlu5 receptors substantially enhanced cell proliferation. Adult mice lacking mGlu5 receptors or treated with mGlu5 or mGlu3 receptor antagonists showed a dramatic reduction in the number of dividing neuroprogenitors present in the subventricular zone and in the dentate gyrus of the hippocampus. These data disclose a novel function of mGlu receptors and offer new potential strategies for the optimization of cell replacement therapy in neurodegenerative disorders.


Subject(s)
Neurons/cytology , Receptors, Metabotropic Glutamate/physiology , Stem Cells/cytology , Animals , Blotting, Western , Cell Cycle/physiology , Cell Growth Processes/physiology , Cell Survival/drug effects , Cells, Cultured , Immunohistochemistry , Mice , Mice, Knockout , Neurons/metabolism , Prosencephalon/cytology , Prosencephalon/metabolism , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism
15.
J Neurochem ; 93(5): 1345-52, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15934953

ABSTRACT

Antidepressant drugs have a clinical latency that correlates with the development of neuroadaptive changes, including down-regulation of beta-adrenergic receptors in different brain regions. The identification of drugs that shorten this latency will have a great impact on the treatment of major depressive disorders. We report that the time required for the antidepressant imipramine to reduce the expression of beta-adrenergic receptors in the hippocampus is reduced by a co-administration with centrally active ligands of type 2/3 metabotropic glutamate (mGlu2/3) receptors. Daily treatment of mice with imipramine alone (10 mg/kg, i.p.) reduced the expression of beta-adrenergic receptors in the hippocampus after 21 days, but not at shorter times, as assessed by western blot analysis of beta1-adrenergic receptors and by the amount of specifically bound [3H]CGP-12177, a selective beta-adrenergic receptor ligand. Down-regulation of beta-adrenergic receptors occurred at shorter times (i.e. after 14 days) when imipramine was combined with low doses (0.5 mg/kg, i.p.) of the selective mGlu2/3 receptor agonist LY379268, or with the preferential mGlu2/3 receptor antagonist LY341495 (1 mg/kg, i.p.). Higher doses of LY379268 (2 mg/kg, i.p.) were inactive. This intriguing finding suggests that neuroadaptation to imipramine--at least as assessed by changes in the expression of beta1-adrenergic receptors--is influenced by drugs that interact with mGlu2/3 receptors and stimulates further research aimed at establishing whether any of these drugs can shorten the clinical latency of classical antidepressants.


Subject(s)
Adaptation, Physiological , Antidepressive Agents, Tricyclic/pharmacology , Imipramine/pharmacology , Nervous System Physiological Phenomena , Receptors, Adrenergic, beta/metabolism , Receptors, Metabotropic Glutamate/metabolism , Amino Acids/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Down-Regulation , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/metabolism , Ligands , Male , Mice , Mice, Inbred Strains , Reaction Time/drug effects , Receptors, Metabotropic Glutamate/administration & dosage , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Xanthenes/pharmacology
16.
Apoptosis ; 10(1): 91-104, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15711925

ABSTRACT

The mitochondrial peripheral benzodiazepine receptor (PBR) is involved in a functional structure designated as the mitochondrial permeability transition (MPT) pore, which controls apoptosis. PBR expression in nervous system has been reported in glial and immune cells. We now show expression of both PBR mRNA and protein, and the appearance of binding of a synthetic ligand fluo-FGIN-1-27 in mitochondria of rat cerebellar granule cells (CGCs). Additionally, the effect of PBR ligands on colchicine-induced apoptosis was investigated. Colchicine-induced neurotoxicity in CGCs was measured at 24 h. We show that, in vitro, PBR ligands 1-(2-chlorophenyl-N-methylpropyl)-3-isoquinolinecarboxamide (PK11195), 7-chloro-5-(4-chlorophenyl)-1,3-dihydro-1-methyl-2H-1,4- benzodiazepin-2-one (Ro5-4864) and diazepam (25- 50 microM) enhanced apoptosis induced by colchicine, as demonstrated by viability experiments, flow cytometry and nuclear chromatin condensation. Enhancement of colchicine-induced apoptosis was characterized by an increase in mitochondrial release of cytochrome c and AIF proteins and an enhanced activation of caspase-3, suggesting mitochondrion dependent mechanism that is involved in apoptotic process. Our results indicate that exposure of neural cells to PBR ligands generates an amplification of apoptotic process induced by colchicine and that the MPT pore may be involved in this process.


Subject(s)
Apoptosis/drug effects , Benzodiazepinones/pharmacology , Diazepam/pharmacology , Isoquinolines/pharmacology , Neurons/drug effects , Receptors, GABA-A/drug effects , Animals , Animals, Newborn , Cell Survival/drug effects , Cells, Cultured , Cerebellum/cytology , Colchicine/toxicity , Ligands , Mitochondria/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Time Factors
17.
Med Hypotheses ; 64(1): 120-3, 2005.
Article in English | MEDLINE | ID: mdl-15533627

ABSTRACT

Several lines of evidence show that cyclin-dependent kinases (CDKs) contribute to neurodegenerative disorders such as Alzheimer's and Parkinson's diseases, and amyotrophic lateral sclerosis. Given their role in the neuronal apoptosis, the inhibition of CDKs by specific drugs such as flavopiridol may be a valid therapeutic approach. Expression of CDKs was observed in rodent models of excitotoxicity and stroke, and CDK inhibitors showed neuroprotective effects. Flavopiridol may provide significant improvement in neurodegenerative diseases in humans.


Subject(s)
Apoptosis/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Flavoproteins/administration & dosage , Models, Neurological , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Animals , Antineoplastic Agents/administration & dosage , Clinical Trials as Topic , Humans , Neurons/drug effects , Neuroprotective Agents/administration & dosage
18.
J Neurosci ; 24(46): 10343-52, 2004 Nov 17.
Article in English | MEDLINE | ID: mdl-15548648

ABSTRACT

Exposure of immature rat cerebellar granule cell cultures to the type 4 metabotropic glutamate (mGlu4) receptor enhancer N-phenyl-7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxamide (PHCCC) reduced [3H]thymidine incorporation. Its action was sensitive to the growth conditions and was attenuated by two mGlu4 receptor antagonists. An antiproliferative action of PHCCC was also seen in cultures from wild-type, but not mGlu4, knock-out mice. At least in rat cultures, PHCCC was not neurotoxic and enhanced neuritogenesis. Although PHCCC reduced the increase in cAMP formation and phospho-AKT levels induced by forskolin, none of these transduction pathways significantly contributed to the reduction of [3H]thymidine incorporation. Interestingly, PHCCC reduced the expression of Gli-1, a transcription factor that mediates the mitogenic action of Sonic hedgehog. Finally, we treated newborn rats with PHCCC either intracerebrally (infusion of 5 nmol/2 microl in the cerebellar region once every other day) or systemically (5 mg/kg, i.p., once daily) from postnatal days 3-9. Local infusion of PHCCC induced substantial changes in the morphology of the developing cerebellum. In contrast, systemic injection of PHCCC induced only morphological abnormalities of the cerebellar lobule V, which became visible 11 d after the end of the treatment. These data suggest that mGlu4 receptors are involved in the regulation of cerebellar development.


Subject(s)
Benzopyrans/pharmacology , Cerebellum/cytology , Neurons/cytology , Receptors, Metabotropic Glutamate/agonists , Stem Cells/cytology , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cerebellum/drug effects , Cerebellum/growth & development , Cyclic AMP/biosynthesis , Depression, Chemical , Mice , Mice, Knockout , Neurites/drug effects , Neurites/physiology , Neurons/drug effects , Organ Size/drug effects , Phosphorylation , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/genetics , Signal Transduction , Stem Cells/drug effects , Thymidine/metabolism
19.
J Endocrinol Invest ; 27(6 Suppl): 171-6, 2004.
Article in English | MEDLINE | ID: mdl-15481819

ABSTRACT

Glutamate is the major neurotransmitter in the mammalian central nervous system and plays a pivotal role in both acute and chronic pain. The actions of glutamate are mediated by two receptor families: ionotropic glutamate receptors (iGluRs), and metabotropic glutamate receptors (mGluRs). Activation of glutamate receptor can elicit both hyperalgesic and analgesic effects. Eight mGluRs subtypes (mGluR1-mGluR8) have been identified and classified into three groups. Among these, group I mGluRs (mGlu1 and -5) have been implicated in the processes of central sensitization and persistent nociception, whereas activation of group II mGluRs (mGlu2/3) is effective against neuropathic or inflammatory pain. In this review we focus on the role of mGlu2/3 in the modulation of persistent pain, and on their potential use as drug targets in pain management.


Subject(s)
Pain/drug therapy , Peripheral Nervous System Diseases/drug therapy , Receptors, Metabotropic Glutamate/drug effects , Acetylcarnitine/therapeutic use , Analgesics/therapeutic use , Animals , Humans , Nootropic Agents/therapeutic use , Pain/etiology , Pain/physiopathology , Peripheral Nervous System Diseases/complications , Peripheral Nervous System Diseases/physiopathology , Receptors, Metabotropic Glutamate/physiology
20.
Bipolar Disord ; 6(2): 144-9, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15005753

ABSTRACT

OBJECTIVES: Here we evaluated the neuroprotective effects of two well-known mood stabilizers, lithium and valproic acid (VPA), against colchicine neurotoxicity in cerebellar granule cells (CGNs). METHODS: The CGNs were differentiated for 7 days, pretreated with lithium or VPA for 24 h and after colchicine 1 microM was added. Cellular damage was assessed with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium (MTT) method and apoptosis in CGNs was characterized by chromatin condensation and DNA fragmentation. RESULTS: Incubation with lithium (1-5 mM) attenuated this apoptosis markedly, in a dose-dependent way however, the addition of VPA (0.5-2 mM) did not protect CGNs. Colchicine-induced apoptosis is mediated through the activation of caspase-3. An increase in caspase-3 activity was detected within 18 h and was blocked in presence of lithium 5 mM. CONCLUSIONS: Our data indicate that lithium treatment is selectively neuroprotective; however, in our experimental conditions VPA did not protect CGNs from apoptosis induced by colchicine. Our results support the hypothesis that distinct pathways mediate the neuroprotective effects of lithium and VPA.


Subject(s)
Apoptosis/drug effects , Cerebellum/drug effects , Cerebellum/pathology , Colchicine/antagonists & inhibitors , Colchicine/pharmacology , Gout Suppressants/pharmacology , Lithium Carbonate/pharmacology , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Animals , Colchicine/administration & dosage , Colorimetry , Flow Cytometry , Gout Suppressants/administration & dosage , Lithium Carbonate/administration & dosage , Neuroprotective Agents/administration & dosage , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...