Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Front Mol Neurosci ; 10: 60, 2017.
Article in English | MEDLINE | ID: mdl-28386216

ABSTRACT

In the adult mammalian brain, new neurons continue to be produced throughout life in two main regions in the brain, the subgranular zone (SGZ) in the hippocampus and the subventricular zone in the walls of the lateral ventricles. Neural stem cells (NSCs) proliferate in these niches, and migrate as neuroblasts, to further differentiate in locations where new neurons are needed, either in normal or pathological conditions. However, the endogenous attempt of brain repair is not very efficient. Calpains are proteases known to be involved in neuronal damage and in cell proliferation, migration and differentiation of several cell types, though their effects on neurogenesis are not well known. Previous work by our group has shown that the absence of calpastatin (CAST), the endogenous inhibitor of calpains, impairs early stages of neurogenesis. Since the hippocampus is highly associated with learning and memory, we aimed to evaluate whether calpain inhibition would help improve cognitive recovery after lesion and efficiency of post-injury neurogenesis in this region. For that purpose, we used the kainic acid (KA) model of seizure-induced hippocampal lesion and mice overexpressing CAST. Selected cognitive tests were performed on the 3rd and 8th week after KA-induced lesion, and cell proliferation, migration and differentiation in the dentate gyrus (DG) of the hippocampus of adult mice were analyzed using specific markers. Cognitive recovery was evaluated by testing the animals for recognition, spatial and associative learning and memory. Cognitive function was preserved by CAST overexpression following seizures, while modulation of post-injury neurogenesis was similar to wild type (WT) mice. Calpain inhibition could still be potentially able to prevent the impairment in the formation of new neurons, given that the levels of calpain activity could be reduced under a certain threshold and other harmful effects from the pathological environment could also be controlled.

2.
Oxid Med Cell Longev ; 2015: 451512, 2015.
Article in English | MEDLINE | ID: mdl-26587180

ABSTRACT

Hippocampal neurogenesis is changed by brain injury. When neuroinflammation accompanies injury, activation of resident microglial cells promotes the release of inflammatory cytokines and reactive oxygen/nitrogen species like nitric oxide (NO). In these conditions, NO promotes proliferation of neural stem cells (NSC) in the hippocampus. However, little is known about the role of NO in the survival and differentiation of newborn cells in the injured dentate gyrus. Here we investigated the role of NO following seizures in the regulation of proliferation, migration, differentiation, and survival of NSC in the hippocampus using the kainic acid (KA) induced seizure mouse model. We show that NO increased the proliferation of NSC and the number of neuroblasts following seizures but was detrimental to the survival of newborn neurons. NO was also required for the maintenance of long-term neuroinflammation. Taken together, our data show that NO positively contributes to the initial stages of neurogenesis following seizures but compromises survival of newborn neurons.


Subject(s)
Hippocampus/metabolism , Nitric Oxide/metabolism , Seizures/pathology , Animals , Cell Proliferation , Dentate Gyrus/metabolism , Disease Models, Animal , Doublecortin Domain Proteins , Immunohistochemistry , Kainic Acid/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/immunology , Microtubule-Associated Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neuropeptides/immunology , Neuropeptides/metabolism , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/genetics , Seizures/chemically induced , Seizures/metabolism
3.
Front Cell Neurosci ; 9: 22, 2015.
Article in English | MEDLINE | ID: mdl-25698931

ABSTRACT

Calpains are ubiquitous proteases involved in cell proliferation, adhesion and motility. In the brain, calpains have been associated with neuronal damage in both acute and neurodegenerative disorders, but their physiological function in the nervous system remains elusive. During brain ischemia, there is a large increase in the levels of intracellular calcium, leading to the activation of calpains. Inhibition of these proteases has been shown to reduce neuronal death in a variety of stroke models. On the other hand, after stroke, neural stem cells (NSC) increase their proliferation and newly formed neuroblasts migrate towards the site of injury. However, the process of forming new neurons after injury is not efficient and finding ways to improve it may help with recovery after lesion. Understanding the role of calpains in the process of neurogenesis may therefore open a new window for the treatment of stroke. We investigated the involvement of calpains in NSC proliferation and neuroblast migration in two highly neurogenic regions in the mouse brain, the dentate gyrus (DG) and the subventricular zone (SVZ). We used mice that lack calpastatin, the endogenous calpain inhibitor, and calpains were also modulated directly, using calpeptin, a pharmacological calpain inhibitor. Calpastatin deletion impaired both NSC proliferation and neuroblast migration. Calpain inhibition increased NSC proliferation, migration speed and migration distance in cells from the SVZ. Overall, our work suggests that calpains are important for neurogenesis and encourages further research on their neurogenic role. Prospective therapies targeting calpain activity may improve the formation of new neurons following stroke, in addition to affording neuroprotection.

4.
Front Cell Neurosci ; 8: 343, 2014.
Article in English | MEDLINE | ID: mdl-25389386

ABSTRACT

Neuroinflammation is characterized by activation of microglial cells, followed by production of nitric oxide (NO), which may have different outcomes on neurogenesis, favoring or inhibiting this process. In the present study, we investigated how the inflammatory mediator NO can affect proliferation of neural stem cells (NSCs), and explored possible mechanisms underlying this effect. We investigated which mechanisms are involved in the regulation of NSC proliferation following treatment with an inflammatory stimulus (lipopolysaccharide plus IFN-γ), using a culture system of subventricular zone (SVZ)-derived NSCs mixed with microglia cells obtained from wild-type mice (iNOS(+/+)) or from iNOS knockout mice (iNOS(-/-)). We found an impairment of NSC cell proliferation in iNOS(+/+) mixed cultures, which was not observed in iNOS(-/-) mixed cultures. Furthermore, the increased release of NO by activated iNOS(+/+) microglial cells decreased the activation of the ERK/MAPK signaling pathway, which was concomitant with an enhanced nitration of the EGF receptor. Preventing nitrogen reactive species formation with MnTBAP, a scavenger of peroxynitrite (ONOO(-)), or using the ONOO(-) degradation catalyst FeTMPyP, cell proliferation and ERK signaling were restored to basal levels in iNOS(+/+) mixed cultures. Moreover, exposure to the NO donor NOC-18 (100 µM), for 48 h, inhibited SVZ-derived NSC proliferation. Regarding the antiproliferative effect of NO, we found that NOC-18 caused the impairment of signaling through the ERK/MAPK pathway, which may be related to increased nitration of the EGF receptor in NSC. Using MnTBAP nitration was prevented, maintaining ERK signaling, rescuing NSC proliferation. We show that NO from inflammatory origin leads to a decreased function of the EGF receptor, which compromised proliferation of NSC. We also demonstrated that NO-mediated nitration of the EGF receptor caused a decrease in its phosphorylation, thus preventing regular proliferation signaling through the ERK/MAPK pathway.

5.
Stem Cells Int ; 2014: 878397, 2014.
Article in English | MEDLINE | ID: mdl-24550991

ABSTRACT

The involvement of nitric oxide (NO) and cyclic GMP (cGMP) in neurogenesis has been progressively unmasked over the last decade. Phosphodiesterase 5 (PDE5) specifically degrades cGMP and is highly abundant in the mammalian brain. Inhibition of cGMP hydrolysis by blocking PDE5 is a possible strategy to enhance the first step of neurogenesis, proliferation of neural stem cells (NSC). In this work, we have studied the effect on cell proliferation of 3 inhibitors with different selectivity and potency for PDE5, T0156, sildenafil, and zaprinast, using subventricular zone-(SVZ-) derived NSC cultures. We observed that a short- (6 h) or a long-term (24 h) treatment with PDE5 inhibitors increased SVZ-derived NSC proliferation. Cell proliferation induced by PDE5 inhibitors was dependent on the activation of the mitogen-activated protein kinase (MAPK) and was abolished by inhibitors of MAPK signaling, soluble guanylyl cyclase, and protein kinase G. Moreover, sildenafil neither activated ERK1/2 nor altered p27(Kip1) levels, suggesting the involvement of pathways different from those activated by T0156 or zaprinast. In agreement with the present results, PDE5 inhibitors may be an interesting therapeutic approach for enhancing the proliferation stage of adult neurogenesis.

6.
Toxicol In Vitro ; 27(8): 2193-202, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24055897

ABSTRACT

In this study we evaluated the neurotoxicity of eslicarbazepine acetate (ESL), and of its in vivo metabolites eslicarbazepine (S-Lic) and R-licarbazepine (R-Lic), as compared to the structurally-related compounds carbamazepine (CBZ) and oxcarbazepine (OXC), in an in vitro model of cultured rat hippocampal neurons. The non-related antiepileptic drugs (AEDs) lamotrigine (LTG) and sodium valproate (VPA) were also studied. We assessed whether AEDs modulate pro-survival/pro-apoptotic pathways, such as extracellular-regulated kinase (ERK1/2), Akt and stress activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK). We found that neither ESL nor its metabolites, CBZ or LTG, up to 0.3mM, for 24h of exposure, decreased cell viability. OXC was the most toxic drug decreasing cell viability in a concentration-dependent manner, leading to activation of caspase-3 and PARP cleavage. VPA caused the appearance of the apoptotic markers, but did not alter cell viability. ESL, S-Lic and OXC decreased the levels of phospho-ERK1/2 and of phospho-Akt, when compared to basal levels, whereas CBZ decreased phospho-SAPK/JNK and phospho-Akt levels. LTG and VPA increased the phosphorylation levels of SAPK/JNK. These results suggest that ESL and its main metabolite S-Lic, as well as CBZ, LTG and VPA, are less toxic to hippocampal neurons than OXC, which was the most toxic agent.


Subject(s)
Anticonvulsants/pharmacology , Hippocampus/cytology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Caspase 3/metabolism , Cell Survival/drug effects , Cells, Cultured , Mitogen-Activated Protein Kinases/metabolism , Neurons/metabolism , Rats , Rats, Wistar
7.
Curr Protoc Stem Cell Biol ; Chapter 2: Unit 2D.14, 2013.
Article in English | MEDLINE | ID: mdl-23404673

ABSTRACT

This unit describes two basic protocols for the detection of the proliferation of neural stem cells (NSC). The first one addresses cell proliferation in cultures, starting with primary cell cultures isolated from the mouse subventricular zone (SVZ), in which SVZ-derived NSC are kept in culture as neurospheres. By using this culture system, we are able to study different stages of adult neurogenesis, such as proliferation, differentiation, migration, and survival. Thus, in the first basic protocol, we describe two different techniques to evaluate cell proliferation based on EdU incorporation: (a) immunocytochemistry and (b) flow cytometry. EdU, a new thymidine analog, which is detected by a reproducible and sensitive method based on click chemistry, does not require DNA denaturation, as is the case with BrdU. Thus, co-labeling of EdU with other specific antibodies of extracellular or intracellular targets, as well as other DNA dyes, is possible. In the second basic protocol, we describe an in vivo assay to evaluate proliferation of NSC in the dentate gyrus of hippocampus of adult mice, by both BrdU and EdU detection. With this approach, it is also possible to study different stages of adult neurogenesis, by co-labeling thymidine analogs with other specific markers, such as doublecortin (DCX) or neuronal nuclei protein (NeuN).


Subject(s)
Cell Culture Techniques/methods , Neural Stem Cells/cytology , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cerebral Ventricles/cytology , Doublecortin Protein , Flow Cytometry , Formaldehyde , Gelatin/pharmacology , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Polymers , Tissue Fixation
8.
Neurosignals ; 21(1-2): 1-13, 2013.
Article in English | MEDLINE | ID: mdl-22378242

ABSTRACT

Nitric oxide (NO) is an important inflammatory mediator involved in the initial boost in the proliferation of neural stem cells following brain injury. However, the mechanisms underlying the proliferative effect of NO are still unclear. The aim of this work was to investigate whether cyclic GMP (cGMP) and the cGMP-dependent kinase (PKG) are involved in the proliferative effect triggered by NO in neural stem cells. For this purpose, cultures of neural stem cells isolated from the mouse subventricular zone (SVZ) were used. We observed that long-term exposure to the NO donor (24 h), NOC-18, increased the proliferation of SVZ cells in a cGMP-dependent manner, since the guanylate cyclase inhibitor, ODQ, prevented cell proliferation. Similarly to NOC-18, the cGMP analogue, 8-Br-cGMP, also increased cell proliferation. Interestingly, shorter exposures to NO (6 h) increased cell proliferation in a cGMP-independent manner via the ERK/MAP kinase pathway. The selective inhibitor of PKG, KT5823, prevented the proliferative effect induced by NO at 24 h but not at 6 h. In conclusion, the proliferative effect of NO is initially mediated by the ERK/MAPK pathway, and at later stages by the GC/cGMP/PKG pathway. Thus, our work shows that NO induces neural stem cell proliferation by targeting these two pathways in a biphasic manner.


Subject(s)
Cell Proliferation , Cyclic GMP-Dependent Protein Kinases/physiology , Guanylate Cyclase/physiology , Neural Stem Cells/physiology , Nitric Oxide/physiology , Signal Transduction/physiology , Animals , Carbazoles/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Guanylate Cyclase/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Neural Stem Cells/drug effects , Nitric Oxide Donors/pharmacology , Signal Transduction/drug effects
9.
Stem Cells Int ; 2012: 895659, 2012.
Article in English | MEDLINE | ID: mdl-22997523

ABSTRACT

The finding that neural stem cells (NSCs) are able to divide, migrate, and differentiate into several cellular types in the adult brain raised a new hope for restorative neurology. Nitric oxide (NO), a pleiotropic signaling molecule in the central nervous system (CNS), has been described to be able to modulate neurogenesis, acting as a pro- or antineurogenic agent. Some authors suggest that NO is a physiological inhibitor of neurogenesis, while others described NO to favor neurogenesis, particularly under inflammatory conditions. Thus, targeting the NO system may be a powerful strategy to control the formation of new neurons. However, the exact mechanisms by which NO regulates neural proliferation and differentiation are not yet completely clarified. In this paper we will discuss the potential interest of the modulation of the NO system for the treatment of neurodegenerative diseases or other pathological conditions that may affect the CNS.

10.
Neurochem Res ; 35(12): 1966-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21110090

ABSTRACT

Overactivation of glutamate receptors results in neurodegeneration in a variety of brain pathologies, including ischemia, epilepsy, traumatic brain injury and slow-progressing neurodegenerative disorders. In all these pathologies, it is well accepted that the calcium-dependent cysteine proteases calpains are key players in the mechanisms of neuronal cell death. Many research groups have been actively pursuing to establish a link between the deregulation of intracellular Ca(2+) homeostasis associated with excitotoxicity and calpain activity. It is well established that these two events are connected and interact synergistically to promote neurodegeneration, but whether calpain activity depends on or contributes to Ca(2+) deregulation is still under debate.


Subject(s)
Calcium/physiology , Calpain/physiology , Receptors, Glutamate/physiology , Animals , Calpain/metabolism , Enzyme Activation , Homeostasis , Humans
11.
Neurochem Res ; 33(8): 1501-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18368487

ABSTRACT

We investigated the contribution of L-, N- and P/Q-type Ca(2+) channels to the [Ca(2+)](i) changes, evoked by kainate, in the cell bodies of hippocampal neurons, using a pharmacological approach and Ca(2+) imaging. Selective Ca(2+) channel blockers, namely nitrendipine, omega-Conotoxin GVIA (omega-GVIA) and omega-Agatoxin IVA (omega-AgaIVA) were used. The [Ca(2+)](i) changes evoked by kainate presented a high variability, and were abolished by NBQX, a AMPA/kainate receptor antagonist, but the N-methyl-D-aspartate (NMDA) receptor antagonist, D-AP5, was without effect. Each Ca(2+) channel blocker caused differential inhibitory effects on [Ca(2+)](i) responses evoked by kainate. We grouped the neurons for each blocker in three subpopulations: (1) neurons with responses below 60% of the control; (2) neurons with responses between 60% and 90% of the control, and (3) neurons with responses above 90% of the control. The inhibition caused by nitrendipine was higher than the inhibition caused by omega-GVIA or omega-AgaIVA. Thus, in the presence of nitrendipine, the percentage of cells with responses below 60% of the control was 41%, whereas in the case of omega-GVIA or omega-AgaIVA the values were 9 or 17%, respectively. The results indicate that hippocampal neurons differ in what concerns their L-, N- and P/Q-type Ca(2+) channels activated by stimulation of the AMPA/kainate receptors.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Channels, N-Type/metabolism , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/metabolism , Excitatory Amino Acid Agonists/pharmacology , Hippocampus , Kainic Acid/pharmacology , Neurons/drug effects , Animals , Calcium/metabolism , Calcium Channel Blockers/metabolism , Cells, Cultured , Excitatory Amino Acid Antagonists/metabolism , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Neurons/cytology , Neurons/metabolism , Nitrendipine/metabolism , Quinoxalines/metabolism , Rats , Rats, Wistar , omega-Agatoxin IVA/metabolism , omega-Conotoxin GVIA/metabolism
12.
J Neurochem ; 105(3): 666-76, 2008 May.
Article in English | MEDLINE | ID: mdl-18088374

ABSTRACT

Evidence for increased calpain activity has been described in the hippocampus of rodent models of temporal lobe epilepsy. However, it is not known whether calpains are involved in the cell death that accompanies seizures. In this work, we characterized calpain activation by examining the proteolysis of calpain substrates and in parallel we followed cell death in the hippocampus of epileptic rats. Male Wistar rats were injected with kainic acid (10 mg/kg) intraperitoneally and killed 24 h later, after development of grade 5 seizures. We observed a strong Fluoro-Jade labeling in the CA1 and CA3 areas of the hippocampus in the rats that received kainic acid, when compared with saline-treated rats. Immunohistochemistry and western blot analysis for the calpain-derived breakdown products of spectrin showed evidence of increased calpain activity in the same regions of the hippocampus where cell death is observed. No evidence was found for caspase activation, in the same conditions. Treatment with the calpain inhibitor MDL 28170 significantly prevented the neurodegeneration observed in CA1. Taken together, our data suggest that early calpain activation, but not caspase activation, is involved in neurotoxicity in the hippocampus after status epilepticus.


Subject(s)
Calpain/metabolism , Epilepsy/enzymology , Hippocampus/enzymology , Nerve Degeneration/enzymology , Status Epilepticus/enzymology , Animals , Caspases/metabolism , Convulsants , Dipeptides/pharmacology , Disease Models, Animal , Enzyme Activation , Enzyme Inhibitors/pharmacology , Epilepsy/chemically induced , Epilepsy/physiopathology , Fluoresceins , Hippocampus/pathology , Hippocampus/physiopathology , Kainic Acid , Male , Nerve Degeneration/etiology , Nerve Degeneration/physiopathology , Organic Chemicals , Rats , Rats, Wistar , Spectrin/metabolism , Status Epilepticus/chemically induced , Status Epilepticus/physiopathology , Time Factors
13.
Neurobiol Dis ; 23(3): 533-42, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16766196

ABSTRACT

The effect of selective injury to dopaminergic neurons on the expression of glial cell line-derived neurotrophic factor (GDNF) was examined in substantia nigra cell cultures. H(2)O(2), mimicking increased oxidative stress, or l-DOPA, the main symptomatic treatment for Parkinson's disease, increased GDNF mRNA and protein levels in a time-dependent mode in neuron-glia mixed cultures. The concentration dependence indicated that mild, but not extensive, injury induced GDNF up-regulation. GDNF neutralization with an antibody decreased dopaminergic cell viability in H(2)O(2)-treated cultures, showing that up-regulation of GDNF was protecting dopaminergic neurons. Neither H(2)O(2) nor l-DOPA directly affected GDNF expression in astrocyte cultures, but conditioned media from challenged mixed cultures increased GDNF mRNA and protein levels in astrocyte cultures, indicating that GDNF up-regulation was mediated by neuronal factors. Since pretreatment with 6-OHDA completely abolished H(2)O(2)-induced GDNF up-regulation, we propose that GDNF up-regulation is triggered by failing dopaminergic neurons that signal astrocytes to increase GDNF expression.


Subject(s)
Astrocytes/metabolism , Cytoprotection/physiology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Neurons/metabolism , Oxidative Stress/physiology , Substantia Nigra/metabolism , Animals , Animals, Newborn , Antibodies/pharmacology , Cell Communication/drug effects , Cell Communication/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Coculture Techniques , Cytoprotection/drug effects , Dopamine/metabolism , Dopamine Agents/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/drug effects , Glial Cell Line-Derived Neurotrophic Factor/genetics , Hydrogen Peroxide/toxicity , Levodopa/pharmacology , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neurons/drug effects , Oxidative Stress/drug effects , Oxidopamine/pharmacology , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Substantia Nigra/growth & development , Substantia Nigra/physiopathology , Sympatholytics/pharmacology
14.
Free Radic Biol Med ; 39(12): 1611-9, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16298686

ABSTRACT

Heme oxygenase-1 (HO-1) has been strongly highlighted because of its induction in many cell types by toxic stimuli, including oxidative stress. The intense HO-1 immunostaining in the substantia nigra of Parkinson disease (PD) patients suggests its involvement in the pathogenesis of this neurodegenerative disease. In this work we investigated HO-1 expression in rat substantia nigra postnatal cell cultures under conditions mimicking dopamine toxicity and its modulation by glial cell line-derived neurotrophic factor (GDNF), a potent neuroprotective factor for dopaminergic neurons. In neuron-glia cultures, we found that H2O2, a product of dopamine metabolism, or l-3,4-dihydroxyphenylalanine (L-DOPA), the dopamine precursor used in the therapy of PD, induced a fast up-regulation of HO-1 mRNA and protein levels, followed by a secondary down-regulation. H2O2 and L-DOPA also increased HO-1 expression in astrocyte cultures, but with a delayed time course in H2O2-treated cultures. HO-1 expression was decreased in neuron-glia cultures under conditions under which GDNF up-regulation was observed. Because exogenously applied GDNF prevented HO-1 up-regulation in cultures treated with H2O2 or l-DOPA, and antibody neutralization of GDNF prevented the secondary HO-1 down-regulation observed in neuron-glia cultures, we propose that GDNF negatively modulates HO-1 expression induced by oxidative stress. To our knowledge, this is the first report showing the modulation of HO-1 expression by GDNF.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Substantia Nigra/drug effects , Substantia Nigra/enzymology , Animals , Antibodies/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Down-Regulation , Gene Expression Regulation, Enzymologic/drug effects , Glial Cell Line-Derived Neurotrophic Factor/antagonists & inhibitors , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Heme Oxygenase-1/drug effects , Hydrogen Peroxide/pharmacology , Levodopa/pharmacology , Neurons/cytology , Neurons/drug effects , Neurons/enzymology , Rats , Rats, Wistar , Substantia Nigra/cytology , Up-Regulation
15.
Curr Drug Targets CNS Neurol Disord ; 4(4): 319-24, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16101551

ABSTRACT

Dysregulation of intracellular calcium homeostasis is a common hallmark of degenerating neurons, at some point in the cell death cascade. It is also a feature of many neurological disorders, including stroke, epilepsy, trauma and several neurodegenerative diseases, commonly associated with the phenomenon of excitotoxicity. Nitric oxide (NO) is a signaling gaseous molecule formed in the brain as a part of the normal intracellular calcium signalling, playing highly diversified roles in cellular physiology. For the past 20 years, numerous studies have demonstrated that NO can acts as a neurotoxin in several disorders of the nervous system. More recent evidence shows that NO can also act as a neuroprotective agent. Calcium-dependent proteases, like calpains, were also shown to be activated in several conditions of the nervous system that involve excitotoxic neurodegeneration, and have been receiving increasing attention as therapeutical targets in recent years. In this review, we bring together the recent literature concerning the involvement of NO and calpains in neuronal survival and death. The biological pathways involved with NO and calpains may be good drug targets to alter neurodegenerative diseases.


Subject(s)
Calpain/metabolism , Nerve Degeneration/metabolism , Neurons/enzymology , Nitric Oxide/metabolism , Animals , Cell Death/physiology , Cell Survival/physiology , Enzyme Activation/physiology , Humans , Nerve Degeneration/chemically induced , Neurons/pathology , Neurotoxins , Nitric Oxide/adverse effects , Signal Transduction/physiology
16.
Neuroreport ; 16(4): 393-6, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15729144

ABSTRACT

Overactivation of N-methyl-D-aspartate receptors is known to mediate excitotoxicity due to excessive entry of calcium, leading to the activation of several calcium-dependent enzymes. Calpains are calcium-activated proteases that appear to play a role in excitotoxic neuronal death. Several cellular proteins are substrates for these proteases, particularly the N-methyl-D-aspartate receptor. Recently, cleavage of NR2B subunits has been implicated in excitotoxic neurodegeneration in ischemia. In this work, we investigated the proteolysis by calpains of NR2B subunits of the N-methyl-D-aspartate receptor in the hippocampus of epileptic rats. Our results show that cleaved forms of NR2B subunits are formed after status epilepticus, in the same areas of the hippocampus where calpain activation was detected by immunohistochemical staining of calpain-specific spectrin breakdown products.


Subject(s)
Calpain/metabolism , Epilepsy/metabolism , Hippocampus/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Enzyme Activation/drug effects , Enzyme Activation/physiology , Hippocampus/drug effects , Kainic Acid/pharmacology , Male , Peptide Hydrolases/metabolism , Rats , Rats, Wistar
17.
J Neurochem ; 91(6): 1322-31, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15584909

ABSTRACT

In this work, we investigated the involvement of calpains in the neurotoxicity induced by short-term exposure to kainate (KA) in non-desensitizing conditions of AMPA receptor activation (cyclothiazide present, CTZ), in cultured rat hippocampal neurons. The calpain inhibitor MDL28170 had a protective effect in cultures treated with KA plus CTZ (p < 0.01), preventing the decrease in MTT reduction caused by exposure to KA (p < 0.001). Caspase inhibition by ZVAD-fmk was not neuroprotective against the toxic effect of KA. At 1 h after treatment, we could already observe significantly increased calpain activity, which was prevented by MDL 28170 and NBQX. Western blot analysis of calpain substrates, GluR1, neuronal nitric oxide synthase (nNOS) and nonerythroid spectrin (fodrin), showed a time-dependent and MDL 28170-sensitive proteolysis of these proteins. This effect was due to calpains, but not caspases, since ZVAD-fmk was ineffective in preventing proteolytic events. Breakdown products of fodrin (BDPs) were detected as early as 15 min after exposure to KA. Overall, these results show early activation of calpains following activation of AMPA receptors as well as compromise of neuronal survival, likely due to proteolytic events that affect proteins involved in neuronal signaling.


Subject(s)
Calpain/metabolism , Hippocampus/physiology , Neurons/physiology , Receptors, AMPA/physiology , Animals , Calpain/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Survival/physiology , Cells, Cultured , Embryo, Mammalian , Hippocampus/cytology , Hippocampus/drug effects , Kainic Acid/poisoning , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurotoxins/poisoning , Rats , Rats, Wistar
18.
Neuroreport ; 15(13): 2007-11, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15486472

ABSTRACT

We investigated the role of nitric oxide (NO) on mitochondrial complexes activity, following short-term non-desensitizing activation of AMPA receptors with kainate (KA) plus cyclothiazide (CTZ), in cultured rat hippocampal neurons. In these conditions, we observed a decrease in the activity of mitochondrial complexes I, II/III, and IV. A selective neuronal nitric oxide synthase inhibitor, 7-Nitroindazole, prevented the decrease in the activity of mitochondrial complex I, but not for the other complexes. Exposure to KA plus CTZ also increased cyclic GMP levels significantly, and led to increased levels of 3-nitrotyrosine, a biomarker for peroxynitrite production. Taken together, our results suggest that non-desensitizing activation of AMPA receptors causes inhibition of mitochondrial complex I via peroxynitrite.


Subject(s)
Neurons/drug effects , Nitric Oxide/pharmacology , Peroxynitrous Acid/pharmacology , Proton Pumps/metabolism , Receptors, AMPA/metabolism , Tyrosine/analogs & derivatives , Analysis of Variance , Animals , Antihypertensive Agents/pharmacology , Benzothiadiazines/pharmacology , Cells, Cultured , Cyclic GMP/metabolism , Drug Interactions , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/cytology , Immunohistochemistry/methods , Kainic Acid/pharmacology , Microtubule-Associated Proteins/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Proton Pumps/classification , Quinoxalines/pharmacology , Rats , Tyrosine/metabolism
19.
J Neurochem ; 85(3): 791-800, 2003 May.
Article in English | MEDLINE | ID: mdl-12694405

ABSTRACT

In this work, we investigated the role of nitric oxide (NO) in neurotoxicity triggered by alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA) receptor activation in cultured hippocampal neurons. In the presence of cyclothiazide (CTZ), short-term exposures to kainate (KA; 5 and 15 min, followed by 24-h recovery) decreased cell viability. Both NBQX and d-AP-5 decreased the neurotoxicity caused by KA plus CTZ. Long-term exposures to KA plus CTZ (24 h) resulted in increased toxicity. In short-, but not in long-term exposures, the presence of NO synthase (NOS) inhibitors (l-NAME and 7-NI) decreased the toxicity induced by KA plus CTZ. We also found that KA plus CTZ (15-min exposure) significantly increased cGMP levels. Furthermore, short-term exposures lead to decreased intracellular ATP levels, which was prevented by NBQX, d-AP-5 and NOS inhibitors. Immunoblot analysis revealed that KA induced neuronal NOS (nNOS) proteolysis, gradually lowering the levels of nNOS according to the time of exposure. Calpain, but not caspase-3 inhibitors, prevented this effect. Overall, these results show that NO is involved in the neurotoxicity caused by activation of non-desensitizing AMPA receptors, although to a limited extent, since AMPA receptor activation triggers mechanisms that lead to nNOS proteolysis by calpains, preventing a further contribution of NO to the neurotoxic process.


Subject(s)
Hippocampus/cytology , Kainic Acid/toxicity , Neurons/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Adenosine Triphosphate/metabolism , Animals , Antibodies/pharmacology , Calpain/metabolism , Cells, Cultured , Cyclic GMP/metabolism , Enzyme Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Neurons/cytology , Neurons/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type I , Peptide Hydrolases/metabolism , Rats , Rats, Wistar , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Time Factors
20.
FASEB J ; 17(9): 1118-20, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12692082

ABSTRACT

Glutamate and NPY have been implicated in hippocampal neuropathology in temporal lobe epilepsy. Thus, we investigated the involvement of NPY receptors in mediating neuroprotection against excitotoxic insults in organotypic cultures of rat hippocampal slices. Exposure of hippocampal slice cultures to 2 microM AMPA (alpha-amino-3-hydroxy-5-methyl-isoxazole-4-propionate) induced neuronal degeneration, monitored by propidium iodide uptake, of granule cells and CA1 pyramidal cells. For dentate granule cells, selective activation of Y1, Y2, or Y5 receptors with 1 microM [Leu31,Pro34]NPY, 300 nM NPY13-36 or 1 microM 500 nM NPY(19-23)-(Gly1,Ser3,Gln4,Thr6,Ala31,Aib32,Gln34)-PP, respectively, had a neuroprotective effect against AMPA, whereas only the activation of Y2 receptors was effective for CA1 pyramidal cells. When the slice cultures were exposed to 6 microM kainate, the CA3 pyramidal cells displayed significant degeneration, and in this case the activation of Y1, Y2, and Y5 receptors was neuroprotective. For the kainic acid-induced degeneration of CA1 pyramidal cells, it was again found that only the Y2 receptor activation was effective. Based on the present findings, it was concluded that Y1, Y2, and Y5 receptors effectively can modify glutamate receptor-mediated neurodegeneration in the hippocampus.


Subject(s)
Hippocampus/drug effects , Nerve Degeneration/prevention & control , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/physiology , Animals , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Hippocampus/cytology , Kainic Acid/toxicity , Nerve Degeneration/chemically induced , Neuroprotective Agents/metabolism , Organ Culture Techniques , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Rats , Receptors, Neuropeptide/physiology , Receptors, Neuropeptide Y/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...