Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Mov Disord ; 34(6): 832-844, 2019 06.
Article in English | MEDLINE | ID: mdl-30759320

ABSTRACT

OBJECTIVE: Spreading depolarization (SD) is a transient self-propagating wave of neuronal and glial depolarization coupled with large membrane ionic changes and a subsequent depression of neuronal activity. Spreading depolarization in the cortex is implicated in migraine, stroke, and epilepsy. Conversely, spreading depolarization in the striatum, a brain structure deeply involved in motor control and in Parkinson's disease (PD) pathophysiology, has been poorly investigated. METHODS: We characterized the participation of glutamatergic and dopaminergic transmission in the induction of striatal spreading depolarization by using a novel approach combining optical imaging, measurements of endogenous DA levels, and pharmacological and molecular analyses. RESULTS: We found that striatal spreading depolarization requires the concomitant activation of D1-like DA and N-methyl-d-aspartate receptors, and it is reduced in experimental PD. Chronic l-dopa treatment, inducing dyskinesia in the parkinsonian condition, increases the occurrence and speed of propagation of striatal spreading depolarization, which has a direct impact on one of the signaling pathways downstream from the activation of D1 receptors. CONCLUSION: Striatal spreading depolarization might contribute to abnormal basal ganglia activity in the dyskinetic condition and represents a possible therapeutic target. © 2019 International Parkinson and Movement Disorder Society.


Subject(s)
Corpus Striatum/physiopathology , Dopaminergic Neurons/physiology , Dyskinesia, Drug-Induced/physiopathology , Levodopa/pharmacology , Neurons/physiology , Parkinsonian Disorders/physiopathology , Synaptic Transmission/physiology , Animals , Antineoplastic Combined Chemotherapy Protocols/metabolism , Antiparkinson Agents/pharmacology , Corpus Striatum/drug effects , Nitrogen Mustard Compounds/metabolism , Prednisolone/metabolism , Procarbazine/metabolism , Rats , Rats, Wistar , Vincristine/metabolism
2.
Neurobiol Dis ; 113: 97-108, 2018 05.
Article in English | MEDLINE | ID: mdl-29325869

ABSTRACT

During multiple sclerosis (MS), a close link has been demonstrated to occur between inflammation and neuro-axonal degeneration, leading to the hypothesis that immune mechanisms may promote neurodegeneration, leading to irreversible disease progression. Energy deficits and inflammation-driven mitochondrial dysfunction seem to be involved in this process. In this work we investigated, by the use of striatal electrophysiological field-potential recordings, if the inflammatory process associated with experimental autoimmune encephalomyelitis (EAE) is able to influence neuronal vulnerability to the blockade of mitochondrial complex IV, a crucial component for mitochondrial activity responsible of about 90% of total cellular oxygen consumption. We showed that during the acute relapsing phase of EAE, neuronal susceptibility to mitochondrial complex IV inhibition is markedly enhanced. This detrimental effect was counteracted by the pharmacological inhibition of microglia, of nitric oxide (NO) synthesis and its intracellular pathway (involving soluble guanylyl cyclase, sGC, and protein kinase G, PKG). The obtained results suggest that mitochondrial complex IV exerts an important role in maintaining neuronal energetic homeostasis during EAE. The pathological processes associated with experimental MS, and in particular the activation of microglia and of the NO pathway, lead to an increased neuronal vulnerability to mitochondrial complex IV inhibition, representing promising pharmacological targets.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Microglia/metabolism , Mitochondria/metabolism , Nitric Oxide/metabolism , Animals , Cyclic GMP/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/pathology , Mitochondria/drug effects , Mitochondria/pathology , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nitric Oxide/antagonists & inhibitors , Organ Culture Techniques , Signal Transduction/drug effects , Signal Transduction/physiology , Sodium Azide/pharmacology , Sodium Azide/therapeutic use
3.
Blood ; 121(25): 5088-97, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23589671

ABSTRACT

Thrombolytic therapy is the cornerstone of treatment of acute atherothrombotic ischemic stroke but is associated with brain hemorrhage; antiplatelet therapy has limited efficacy and is still associated with intracranial bleeding. Therefore, new antithrombotic approaches with a better efficacy/safety ratio are required. We have assessed the effect of ALX-0081, a Nanobody against the A1 domain of von Willebrand factor (VWF) that blocks VWF binding to GPIb, of the thrombolytic agent recombinant tissue plasminogen activator (rtPA), and of the GPIIb/IIIa antagonist tirofiban, in a middle cerebral artery (MCA) thrombosis model in guinea pigs. Drugs were administered before, immediately after, or 15 or 60 minutes after the total occlusion of the MCA. ALX-0081 prevented MCA thrombosis and induced reperfusion when given immediately after and 15 minutes after complete occlusion and reduced brain damage without inducing hemorrhage, whereas tirofiban prevented thrombosis but did not induce reperfusion and induced striking brain hemorrhage. rtPA also induced reperfusion when given 60 minutes after occlusion but provoked brain hemorrhage. Skin bleeding time was not modified or was moderately prolonged by ALX-0081, whereas tirofiban and rtPA prolonged it. The inhibition of the GPIb-VWF axis in guinea pigs prevents cerebral artery thrombosis and induces early reperfusion without provoking intracerebral bleeding thus reducing brain infarct area.


Subject(s)
Brain Infarction/prevention & control , Fibrinolytic Agents/pharmacology , Intracranial Thrombosis/drug therapy , Single-Domain Antibodies/pharmacology , Thrombolytic Therapy/methods , Animals , Cerebral Arteries/pathology , Disease Models, Animal , Guinea Pigs , Male , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Reperfusion/methods , von Willebrand Factor/antagonists & inhibitors
4.
Neurobiol Dis ; 62: 387-93, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24135008

ABSTRACT

Multiple sclerosis, one of the main causes of non-traumatic neurological disability in young adults, is an inflammatory and neurodegenerative disorder of the central nervous system. Although the pathogenesis of neuroaxonal damage occurring during the course of the disease is still largely unknown, there is accumulating evidence highlighting the potential role of mitochondria in multiple sclerosis-associated neuronal degeneration. The aim of the present study was to investigate, by utilizing electrophysiological techniques in brain striatal slices, the potential protective effects of interferon-ß1a, one of the most widely used medication for multiple sclerosis, against acute neuronal dysfunction induced by mitochondrial toxins. Interferon-ß1a was found to exert a dose-dependent protective effect against the progressive loss of striatal field potential amplitude induced by the mitochondrial complex I inhibitor rotenone. Interferon-ß1a also reduced the generation of the rotenone-induced inward current in striatal spiny neurons. Conversely, interferon-ß1a did not influence the electrophysiological effects of the mitochondrial complex II inhibitor 3-nitropropionic acid. The protective effect of interferon-ß1a against mitochondrial complex I inhibition was found to be dependent on the activation of STAT1 signaling. Conversely, endogenous dopamine depletion and the modulation of the p38 MAPK and mTOR pathways did not influence the effects of interferon-ß1a. During experimental autoimmune encephalomyelitis (EAE) striatal rotenone toxicity was enhanced but the protective effect of interferon-ß1a was still evident. These results support future studies investigating the role played by specific intracellular signaling pathways in mediating the potential link among inflammation, mitochondrial impairment and neuroaxonal degeneration in multiple sclerosis.


Subject(s)
Interferon-beta/therapeutic use , Mitochondria/drug effects , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Animals , Corpus Striatum/drug effects , Corpus Striatum/physiology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Interferon beta-1a , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Neurons/physiology , Rats , Rotenone/toxicity , STAT1 Transcription Factor/metabolism , Uncoupling Agents/toxicity
5.
Neurobiol Dis ; 52: 229-36, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23295855

ABSTRACT

The central nervous system (CNS) and the immune system are known to be engaged in an intense bidirectional crosstalk. In particular, the immune system has the potential to influence the induction of brain plastic phenomena and neuronal networks functioning. During direct CNS inflammation, as well as during systemic, peripheral, inflammation, the modulation exerted by neuroinflammatory mediators on synaptic plasticity might negatively influence brain neuronal networks functioning. The aim of the present study was to investigate, by using electrophysiological techniques, the ability of hippocampal excitatory synapses to undergo synaptic plasticity during the initial clinical phase of an experimental model of CNS (experimental autoimmune encephalomyelitis, EAE) as well as following a systemic inflammatory trigger. Moreover, we compared the morphologic, synaptic and molecular consequences of central neuroinflammation with those accompanying peripheral inflammation. Hippocampal long-term potentiation (LTP) has been studied by extracellular field potential recordings in the CA1 region. Immunohistochemistry was performed to investigate microglia activation. Western blot and ELISA assays have been performed to assess changes in the subunit composition of the synaptic glutamate NMDA receptor and the concentration of pro-inflammatory cytokines in the hippocampus. Significant microglial activation together with an impairment of CA1 LTP was present in the hippocampus of mice with central as well as peripheral inflammation. Interestingly, exclusively during EAE but not during systemic inflammation, the impairment of hippocampal LTP was paralleled by a selective reduction of the NMDA receptor NR2B subunit levels and a selective increase of interleukin-1ß (IL1ß) levels. Both central and peripheral inflammation-triggered mechanisms can activate CNS microglia and influence the function of CNS synapses. During direct CNS inflammation these events are accompanied by detectable changes in synaptic glutamate receptors subunit composition and in the levels of the pro-inflammatory cytokine IL1ß.


Subject(s)
Hippocampus/physiopathology , Inflammation/physiopathology , Long-Term Potentiation/physiology , Synapses/physiology , Animals , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Excitatory Postsynaptic Potentials/physiology , Mice , Synaptic Transmission/physiology
6.
Brain ; 135(Pt 6): 1884-99, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22561640

ABSTRACT

Although patients with Parkinson's disease show impairments in cognitive performance even at the early stage of the disease, the synaptic mechanisms underlying cognitive impairment in this pathology are unknown. Hippocampal long-term potentiation represents the major experimental model for the synaptic changes underlying learning and memory and is controlled by endogenous dopamine. We found that hippocampal long-term potentiation is altered in both a neurotoxic and transgenic model of Parkinson's disease and this plastic alteration is associated with an impaired dopaminergic transmission and a decrease of NR2A/NR2B subunit ratio in synaptic N-methyl-d-aspartic acid receptors. Deficits in hippocampal-dependent learning were also found in hemiparkinsonian and mutant animals. Interestingly, the dopamine precursor l-DOPA was able to restore hippocampal synaptic potentiation via D1/D5 receptors and to ameliorate the cognitive deficit in parkinsonian animals suggesting that dopamine-dependent impairment of hippocampal long-term potentiation may contribute to cognitive deficits in patients with Parkinson's disease.


Subject(s)
Hippocampus/physiopathology , Long-Term Potentiation/physiology , Memory Disorders/etiology , Parkinson Disease/complications , Parkinson Disease/pathology , Analysis of Variance , Animals , Antiparkinson Agents/pharmacology , Antiparkinson Agents/therapeutic use , Benserazide/pharmacology , Benserazide/therapeutic use , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , Disease Models, Animal , Dopamine/metabolism , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Exploratory Behavior/drug effects , Humans , Levodopa/pharmacology , Levodopa/therapeutic use , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Male , Memory Disorders/drug therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microdialysis/methods , Mutation/genetics , Oxidopamine/toxicity , Parkinson Disease/drug therapy , Parkinson Disease/etiology , Patch-Clamp Techniques , Radionuclide Imaging , Rats , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Sympatholytics/toxicity , Synaptosomes/diagnostic imaging , Synaptosomes/drug effects , Tritium/metabolism , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/genetics
7.
J Neurosci ; 31(5): 1850-62, 2011 Feb 02.
Article in English | MEDLINE | ID: mdl-21289195

ABSTRACT

A(2A) adenosine receptor antagonists are currently under investigation as potential therapeutic agents for Parkinson's disease (PD). However, the molecular mechanisms underlying this therapeutic effect is still unclear. A functional antagonism exists between A(2A) adenosine and D(2) dopamine (DA) receptors that are coexpressed in striatal medium spiny neurons (MSNs) of the indirect pathway. Since this interaction could also occur in other neuronal subtypes, we have analyzed the pharmacological modulation of this relationship in murine MSNs of the direct and indirect pathways as well in striatal cholinergic interneurons. Under physiological conditions, endogenous cannabinoids (eCBs) play a major role in the inhibitory effect on striatal glutamatergic transmission exerted by the concomitant activation of D(2) DA receptors and blockade of A(2A) receptors in both D(2)- and D(1)-expressing striatal MSNs. In experimental models of PD, the inhibition of striatal glutamatergic activity exerted by D(2) receptor activation did not require the concomitant inhibition of A(2A) receptors, while it was still dependent on the activation of CB(1) receptors in both D(2)- and D(1)-expressing MSNs. Interestingly, the antagonism of M1 muscarinic receptors blocked the effects of D(2)/A(2A) receptor modulation on MSNs. Moreover, in cholinergic interneurons we found coexpression of D(2) and A(2A) receptors and a reduction of the firing frequency exerted by the same pharmacological agents that reduced excitatory transmission in MSNs. This evidence supports the hypothesis that striatal cholinergic interneurons, projecting to virtually all MSN subtypes, are involved in the D(2)/A(2A) and endocannabinoid-mediated effects observed on both subpopulations of MSNs in physiological conditions and in experimental PD.


Subject(s)
Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Neurons/metabolism , Parkinson Disease/metabolism , Receptor, Adenosine A2A/metabolism , Receptors, Dopamine D2/metabolism , Signal Transduction , Adrenergic Uptake Inhibitors/pharmacology , Animals , Cells, Cultured , Cholinergic Agents/pharmacology , Corpus Striatum/drug effects , Dendritic Spines/metabolism , Disease Models, Animal , Electrophysiology , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials , Fluorescent Antibody Technique , Immunohistochemistry , Interneurons/metabolism , Male , Mice , Mice, Inbred C57BL , Microelectrodes , Neurons/drug effects , Oxidopamine , Parkinson Disease/physiopathology , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Receptor, Muscarinic M1/antagonists & inhibitors , Receptors, Dopamine D1/metabolism , Reserpine/pharmacology , Signal Transduction/drug effects
8.
Neurobiol Dis ; 44(2): 185-91, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21749921

ABSTRACT

Nitric oxide (NO) is an intercellular retrograde messenger involved in several physiological processes such as synaptic plasticity, hippocampal long-term potentiation (LTP), and learning and memory. Moreover NO signaling is implicated in the pathophysiology of brain ischemia. In this study, we have characterized the role of NO/cGMP signaling cascade in the induction and maintenance of post-ischemic LTP (iLTP) in rat brain slices. Moreover, we have investigated the possible inhibitory action of zonisamide (ZNS) on this pathological form of synaptic plasticity as well as the effects of this antiepileptic drug (AED) on physiological activity-dependent LTP. Finally, we have characterized the possible interaction between ZNS and the NO/cGMP/PKG-dependent pathway involved in iLTP. Here, we provided the first evidence that an oxygen and glucose deprivation episode can induce, in CA1 hippocampal slices, iLTP by modulation of the NO/cGMP/PKG pathway. Additionally, we found that while ZNS application did not affect short-term synaptic plasticity and LTP induced by high-frequency stimulation, it significantly reduced iLTP. This reduction was mimicked by bath application of NO synthase inhibitors and a soluble guanyl cyclase inhibitor. The effect of ZNS was prevented by either the application of a NO donor or drugs increasing intracellular levels of cGMP and activating PKG. These findings are in line with the possible use of AEDs, such as ZNS, as a possible neuroprotective strategy in brain ischemia. Moreover, these findings strongly suggest that NO/cGMP/PKG intracellular cascade might represent a physiological target for neuroprotection in pathological forms of synaptic plasticity such as hippocampal iLTP.


Subject(s)
Brain Ischemia/physiopathology , Cyclic GMP-Dependent Protein Kinases/physiology , Cyclic GMP/physiology , Hippocampus/physiopathology , Isoxazoles/pharmacology , Long-Term Potentiation/physiology , Nitric Oxide/physiology , Signal Transduction/physiology , Animals , Anticonvulsants/pharmacology , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Long-Term Potentiation/drug effects , Male , Neuroprotective Agents/pharmacology , Organ Culture Techniques , Rats , Rats, Wistar , Signal Transduction/drug effects , Zonisamide
9.
Mov Disord ; 26(9): 1639-47, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21542017

ABSTRACT

Among heat shock proteins, mortalin has been linked to the pathogenesis of Parkinson's disease. In the present work a rat model of Parkinson's disease was used to analyze the expression of striatal proteins and, more specifically, mortalin expression. The possible involvement of mortalin in Parkinson's disease pathogenesis was further investigated by utilizing an electrophysiological approach and pharmacological inhibition of mortalin in both the physiological and the parkinsonian states. Proteomic analysis was used to investigate changes in striatal protein expression in the 6-hydroxydopamine rat model of Parkinson's disease. The electrophysiological effects of MKT-077, a rhodamine-123 analogue acting as an inhibitor of mortalin, were measured by field potential recordings from corticostriatal brain slices obtained from control, sham-operated, and 6-hydroxydopamine-denervated animals. Slices in the presence of rotenone, an inhibitor of mitochondrial complex I, were also analyzed. Proteomic analysis revealed downregulation of mortalin in the striata of 6-hydroxydopamine-treated rats in comparison with sham-operated animals. MKT-077 reduced corticostriatal field potential amplitude in physiological conditions, inducing membrane depolarization and inward current in striatal medium spiny neurons. In addition, we observed that concentrations of MKT-077 not inducing any electrophysiological effect in physiological conditions caused significant changes in striatal slices from parkinsonian animals as well as in slices treated with a submaximal concentration of rotenone. These findings suggest a critical link between mortalin function and mitochondrial activity in both physiological and pathological conditions mimicking Parkinson's disease.


Subject(s)
Antiparasitic Agents/therapeutic use , HSP70 Heat-Shock Proteins/therapeutic use , Parkinson Disease/drug therapy , Action Potentials/drug effects , Animals , Cerebral Cortex/pathology , Corpus Striatum/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Electron Transport Complex I/metabolism , Gene Expression Regulation/drug effects , HSP70 Heat-Shock Proteins/antagonists & inhibitors , In Vitro Techniques , Male , Neurons/drug effects , Oxidopamine/toxicity , Parkinson Disease/etiology , Parkinson Disease/pathology , Proteomics/methods , Pyridines/pharmacology , Rats , Rats, Wistar , Thiazoles/pharmacology
10.
Cell Rep ; 37(10): 110094, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34879272

ABSTRACT

Cognitive impairment (CI) is a disabling concomitant of multiple sclerosis (MS) with a complex and controversial pathogenesis. The cytokine interleukin-17A (IL-17A) is involved in the immune pathogenesis of MS, but its possible effects on synaptic function and cognition are still largely unexplored. In this study, we show that the IL-17A receptor (IL-17RA) is highly expressed by hippocampal neurons in the CA1 area and that exposure to IL-17A dose-dependently disrupts hippocampal long-term potentiation (LTP) through the activation of its receptor and p38 mitogen-activated protein kinase (MAPK). During experimental autoimmune encephalomyelitis (EAE), IL-17A overexpression is paralleled by hippocampal LTP dysfunction. An in vivo behavioral analysis shows that visuo-spatial learning abilities are preserved when EAE is induced in mice lacking IL-17A. Overall, this study suggests a key role for the IL-17 axis in the neuro-immune cross-talk occurring in the hippocampal CA1 area and its potential involvement in synaptic dysfunction and MS-related CI.


Subject(s)
Behavior, Animal , CA1 Region, Hippocampal/metabolism , Cognition , Encephalomyelitis, Autoimmune, Experimental/metabolism , Interleukin-17/metabolism , Neuronal Plasticity , Receptors, Interleukin-17/metabolism , Synapses/metabolism , Animals , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/physiopathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Encephalomyelitis, Autoimmune, Experimental/psychology , Interleukin-17/genetics , Long-Term Potentiation , Male , Mice, Biozzi , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-17/genetics , Signal Transduction , Spatial Learning , Synapses/pathology , p38 Mitogen-Activated Protein Kinases
11.
J Neurosci ; 28(32): 8040-52, 2008 Aug 06.
Article in English | MEDLINE | ID: mdl-18685029

ABSTRACT

Reduced activity of the mitochondrial respiratory chain and in particular of complex I is implicated not only in the etiology of Parkinson's disease but also in other forms of parkinsonism in which striatal neurodegeneration occurs, such as progressive supranuclear palsy. The pesticide rotenone inhibits mitochondrial complex I and reproduces features of these basal ganglia neurological disorders in animal models. We have characterized the electrophysiological effects of rotenone in the striatum as well as potential neuroprotective strategies to counteract the detrimental effects of this neurotoxin. We found that rotenone causes a dose-dependent and irreversible loss of the corticostriatal field potential amplitude, which was related to the development of a membrane depolarization/inward current in striatal spiny neurons, coupled to an increased release of both excitatory amino acids and dopamine (DA). In particular, we have investigated whether glutamate, DA, and GABA systems might represent possible targets for neuroprotection against rotenone-induced striatal neuronal dysfunction. Interestingly, whereas modulation of glutamatergic transmission was not neuroprotective, blockade of D(2)-like but not D(1)-like DA receptors significantly reduced the rotenone-induced effects via a GABA-mediated mechanism. In addition, because antiepileptic drugs (AEDs) modulate multiple transmitter systems, we have analyzed the possible neuroprotective effects of some AEDs against rotenone. We found that carbamazepine, unlike other tested AEDs, exerts a potent neuroprotective action against rotenone-induced striatal neuronal dysfunction. This neuroprotection was observed at therapeutically relevant concentrations requiring endogenous GABA. Differential targeting of GABAergic transmission may represent a possible therapeutic strategy against basal ganglia neurodegenerative disorders involving mitochondrial complex I dysfunction.


Subject(s)
Corpus Striatum/physiopathology , Electron Transport Complex I/antagonists & inhibitors , Mitochondria/metabolism , Neurons , Rotenone/pharmacology , Action Potentials/drug effects , Animals , Anticonvulsants/pharmacology , Corpus Striatum/pathology , Dopamine/metabolism , Dopamine Antagonists/pharmacology , Dopamine D2 Receptor Antagonists , Dose-Response Relationship, Drug , Electrophysiology , Extracellular Space , Glutamic Acid/metabolism , Hippocampus/physiopathology , In Vitro Techniques , Intracellular Space , Male , Mice , Mice, Transgenic , Neuroprotective Agents/pharmacology , Patch-Clamp Techniques , Rats , Rats, Wistar , Rotenone/administration & dosage , Synaptosomes/metabolism , gamma-Aminobutyric Acid/metabolism
12.
Neuropharmacology ; 55(3): 353-62, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18359495

ABSTRACT

Stroke is the second commonest cause of death and the principal cause of adult disability in the world. In most cases ischemic injuries have been reported to induce mild to severe permanent deficits. Nevertheless, recovery is often dynamic, reflecting the ability of the injured neuronal networks to adapt. Plastic phenomena occurring in the cerebral cortex and in subcortical structures after ischemic injuries have been documented at the synaptic, cellular, and network level and several findings suggest that they may play a key role during neurorehabilitation in human stroke survivors. In particular, in vitro studies have demonstrated that oxygen and glucose deprivation (in vitro ischemia) exerts long-term effects on the efficacy of synaptic transmission via the induction of a post-ischemic long-term potentiation (i-LTP). i-LTP may deeply influence the plastic reorganization of cortical representational maps occurring after cerebral ischemia, inducing a functional connection of previously non-interacting neurons. On the other hand, there is evidence that i-LTP may exert a detrimental effect in the peri-infarct area, facilitating excitotoxic processes via the sustained, long-term enhancement of glutamate mediated neurotransmission. In the present work we will review the molecular and synaptic mechanisms underlying ischemia-induced synaptic plastic changes taking into account their potential adaptive and/or detrimental effects on the neuronal network in which they occur. Thereafter, we will consider the implications of brain plastic phenomena in the post-stroke recovery phase as well as during the rehabilitative and therapeutic intervention in human subjects.


Subject(s)
Brain Ischemia/pathology , Brain/pathology , Brain/physiopathology , Neuronal Plasticity/physiology , Animals , Humans
13.
Cell Death Dis ; 9(2): 204, 2018 02 12.
Article in English | MEDLINE | ID: mdl-29434188

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder in which genetic and environmental factors synergistically lead to loss of midbrain dopamine (DA) neurons. Mutation of leucine-rich repeated kinase2 (Lrrk2) genes is responsible for the majority of inherited familial cases of PD and can also be found in sporadic cases. The pathophysiological role of this kinase has to be fully understood yet. Hyperactivation of Lrrk2 kinase domain might represent a predisposing factor for both enhanced striatal glutamatergic release and mitochondrial vulnerability to environmental factors that are observed in PD. To investigate possible alterations of striatal susceptibility to mitochondrial dysfunction, we performed electrophysiological recordings from the nucleus striatum of a G2019S Lrrk2 mouse model of PD, as well as molecular and morphological analyses of G2019S Lrrk2-expressing SH-SY5Y neuroblastoma cells. In G2019S mice, we found reduced striatal DA levels, according to the hypothesis of alteration of dopaminergic transmission, and increased loss of field potential induced by the mitochondrial complex I inhibitor rotenone. This detrimental effect is reversed by the D2 DA receptor agonist quinpirole via the inhibition of the cAMP/PKA intracellular pathway. Analysis of mitochondrial functions in G2019S Lrrk2-expressing SH-SY5Y cells revealed strong rotenone-induced oxidative stress characterized by reduced Ca2+ buffering capability and ATP synthesis, production of reactive oxygen species, and increased mitochondrial fragmentation. Importantly, quinpirole was able to prevent all these changes. We suggest that the G2019S-Lrrk2 mutation is a predisposing factor for enhanced striatal susceptibility to mitochondrial dysfunction induced by exposure to mitochondrial environmental toxins and that the D2 receptor stimulation is neuroprotective on mitochondrial function, via the inhibition of cAMP/PKA intracellular pathway. We suggest new possible neuroprotective strategies for patients carrying this genetic alteration based on drugs specifically targeting Lrrk2 kinase domain and mitochondrial functionality.


Subject(s)
Corpus Striatum/metabolism , Dopaminergic Neurons/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Mesencephalon/metabolism , Neuroprotection , Parkinson Disease/metabolism , Receptors, Dopamine D2/metabolism , Animals , Corpus Striatum/pathology , Disease Models, Animal , Dopamine/genetics , Dopamine/metabolism , Dopaminergic Neurons/pathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Male , Mesencephalon/pathology , Mice , Mice, Mutant Strains , Parkinson Disease/genetics , Parkinson Disease/pathology , Receptors, Dopamine D2/genetics
14.
Neuropharmacology ; 135: 424-430, 2018 06.
Article in English | MEDLINE | ID: mdl-29614316

ABSTRACT

Lacosamide ([(R)-2-acetamido-N-benzyl-3-methoxypropanamide], LCM), is an antiepileptic that exerts anticonvulsant activity by selectively enhancing slow sodium channel inactivation. By inhibiting seizures and neuronal excitability it might therefore be a good candidate to stabilize neurons and protect them from energetic insults. Using electrophysiological analyses, we have investigated in mice the possible neuroprotective effect of LCM against in vitro ischemia obtained by oxygen and glucose deprivation (ODG), in striatal and hippocampal tissues, two brain structures particularly susceptible to ischemic injury and of pivotal importance for different form of learning and memory. We also explored in these regions the influence of LCM on firing discharge and on long-term synaptic plasticity. We found that in both areas LCM reduced the neuronal firing activity in a use-dependent manner without influencing the physiological synaptic transmission, confirming its anticonvulsant effects. Moreover, we found that this AED is able to protect, in a dose dependent manner, striatal and hippocampal neurons from energy metabolism failure produced by OGD. This neuroprotective effect does not imply impairment of long-term potentiation of striatal and hippocampal synapses and suggests that LCM might exert additional beneficial therapeutic effects beyond its use as antiepileptic.


Subject(s)
Brain Ischemia/drug therapy , CA1 Region, Hippocampal/drug effects , Corpus Striatum/drug effects , Lacosamide/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Brain Ischemia/physiopathology , CA1 Region, Hippocampal/physiopathology , Corpus Striatum/physiopathology , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Membrane Potentials/drug effects , Mice, Inbred C57BL , Neurons/physiology , Synapses/drug effects , Synapses/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Tissue Culture Techniques
15.
Stroke ; 38(5): 1614-20, 2007 May.
Article in English | MEDLINE | ID: mdl-17395860

ABSTRACT

BACKGROUND AND PURPOSE: A prominent feature of cerebral ischemia is the excessive intracellular accumulation of both Na(+) and Ca(2+) ions, which results in subsequent cell death. The plasma membrane Na(+)/Ca(2+) exchanger (NCX), regulates the distribution of these ions acting either in the forward mode or in its reverse mode and it can play a critical role in brain ischemia. However, it is unclear whether the activity of NCX leads to detrimental or beneficial effects. METHODS: Extracellular field potentials and whole-cell patch clamp recordings were obtained from rat corticostriatal brain-slice preparations in the peri-infarct area 24 hours after the permanent middle cerebral artery occlusion. Ischemia was induced in rats by permanent middle cerebral artery occlusion. RESULTS: Bepridil, an inhibitor of NCX, reduced in a concentration-dependent manner (IC(50)=68 micromol/L) the field potential amplitude recorded from the peri-infarct area of corticostriatal slices. Conversely, no change was observed in sham-operated animals. The effect of bepridil was mimicked by 5-(N-4-chlorobenzyl)-2',4'-dimethylbenzamil (CB-DMB) (IC(50)=6 micromol/L), a more selective inhibitor of NCX. In whole-cell patch clamp experiments, bepridil and CB-DMB caused an inward current in spiny neurons recorded from the peri-infarct area but not in the same cells recorded from controls. Interestingly, cholinergic interneurons recorded from the striatal peri-infarct area did not develop an inward current after the application of NCX inhibitors, suggesting that the electrophysiological alterations induced by NCX inhibition are cell-type specific. Bepridil and CB-DMB also induced a suppression of excitatory synaptic currents in most of spiny neurons recorded from the peri-infarct area. This effect was not coupled to a significant change of paired-pulse facilitation suggesting a postsynaptic site of action. CONCLUSIONS: Our data indicate that NCX plays a critical role in the maintenance of ionic homeostasis in the peri-infarct area.


Subject(s)
Brain Ischemia/physiopathology , Infarction, Middle Cerebral Artery/physiopathology , Sodium-Calcium Exchanger/physiology , Animals , Homeostasis , Ions/metabolism , Male , Rats , Rats, Sprague-Dawley
16.
Neuropharmacology ; 53(6): 783-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17889039

ABSTRACT

Adenosine and dopamine (DA) strongly modulate the neuronal activity in the striatum by pre- and postsynaptic mechanisms. As several behavioral and molecular studies indicate a functional antagonism between A2A adenosine and D2 DA receptors, compounds that are able to block A2A receptors are of particular interest as antiparkinsonian agents. To study the interaction of A2A and D2 receptors in the striatum, we performed intracellular recordings with sharp microelectrodes and whole-cell patch clamp recordings from spiny neurons in rat corticostriatal slices. The amplitude of the evoked excitatory postsynaptic potentials (EPSPs), as well as the frequency and the amplitude of spontaneous excitatory postsynaptic currents (sEPSCs), were affected neither by the A2A receptor antagonists ST1535 and ZM241385, nor by the D2 receptor agonist quinpirole when applied in isolation. However, co-application of quinpirole and ST1535 or ZM241385 significantly reduced the EPSPs amplitude. This inhibitory effect was associated with an increased paired-pulse facilitation suggesting a presynaptic mechanism of action. Accordingly, whole-cell recordings showed that the concomitant activation of D2 receptors and the antagonism of A2A receptors decreased the frequency of sEPSCs without affecting their amplitude. These results suggest that A2A and D2 receptors converge in the control of corticostriatal glutamatergic transmission by exerting an opposite function.


Subject(s)
Cerebral Cortex/physiology , Corpus Striatum/physiology , Glutamic Acid/metabolism , Neurons/metabolism , Receptor, Adenosine A2A/metabolism , Receptors, Dopamine D2/metabolism , Synaptic Transmission/physiology , Adenine/analogs & derivatives , Adenine/pharmacology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine Agonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Male , Organ Culture Techniques , Patch-Clamp Techniques , Phenethylamines/pharmacology , Quinpirole/pharmacology , Rats , Rats, Wistar , Receptor, Adenosine A2A/drug effects , Receptors, Dopamine D2/drug effects , Synaptic Transmission/drug effects , Triazines/pharmacology , Triazoles/pharmacology
17.
Neurosci Lett ; 415(1): 77-80, 2007 Mar 19.
Article in English | MEDLINE | ID: mdl-17240064

ABSTRACT

Transient focal ischemia induced in rat brain by occlusion of the middle cerebral artery (MCAo) elicits a generalized induction of the 72 kDa heat-shock protein (hsp72) heralding functional recovery. As this effect implies activation of protein synthesis, and local systems of protein synthesis are present in brain synapses, and may be analyzed in preparations of brain synaptosomes, we evaluated hsp72 expression and protein synthesis in synaptosomal fractions of spontaneously hypertensive rats (SHRs) subjected to permanent MCAo. SHRs were randomly divided in ischemics and sham controls, anaesthesia controls and passive controls. Focal ischemia was induced under chloral hydrate anaesthesia by unilateral permanent MCAo. Protein synthesis was determined by [35S]methionine incorporation into synaptosomal proteins from ischemic and contralateral cortex/striatum, and from cerebellum. Hsp72 expression was measured in the same fractions by immunoblotting. Our data demonstrate that under these conditions synaptic hsp72 markedly increases in the ischemic hemisphere 1 and 2 days after MCAo, progressively declining in the following 2 days, while no significant change occurs in control rats. In addition, in the ischemic hemisphere the rate of synaptic protein synthesis increases more than two-fold between 1 and 4 days after MCAo, without showing signs of an impending decline. The present data provide the first demonstration that synaptic protein synthesis is massively involved in brain plastic events elicited by permanent focal ischemia.


Subject(s)
Brain Ischemia/metabolism , HSP72 Heat-Shock Proteins/metabolism , Nerve Tissue Proteins/biosynthesis , Neuronal Plasticity/physiology , Presynaptic Terminals/metabolism , Telencephalon/metabolism , Adaptation, Physiological/physiology , Animals , Biomarkers/metabolism , Brain Ischemia/physiopathology , Infarction, Middle Cerebral Artery/metabolism , Male , Rats , Rats, Inbred SHR , Recovery of Function/physiology , Stress, Physiological/metabolism , Stress, Physiological/physiopathology , Synaptosomes/metabolism , Telencephalon/blood supply , Telencephalon/physiopathology , Time Factors , Up-Regulation/physiology
18.
Eur J Pharmacol ; 545(1): 65-72, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16854409

ABSTRACT

The basal ganglia circuit plays a key role in the regulation of voluntary movements as well as in behavioural control and cognitive functions. The main pathogenic role of mitochondrial dysfunctions is now accepted in the neurodegenerative process and the mitochondria have been successfully used as subcellular targets to obtain relevant experimental models of basal ganglia neurodegenerative disorders. Mitochondrial toxins act through an inhibition of the respiratory chain complexes. These toxins, by uncoupling cellular respiration, shift the cell into a state of oxidative stress and trigger several bidirectional links with the excitotoxic process. Moreover, the in vitro inhibition of the respiratory chain complexes alters the electrophysiological properties of the neurons. The downstream effects triggered by mitochondrial complexes inhibition provide a model integrating genetic and environmental pathogenic factors to explain the selective neuronal vulnerability.


Subject(s)
Basal Ganglia Diseases/metabolism , Mitochondria/physiology , Nerve Degeneration/metabolism , Animals , Disease Models, Animal , Electron Transport Complex I/metabolism , Electron Transport Complex II/metabolism , Humans , Mitochondria/metabolism
19.
Neurobiol Aging ; 48: 161-171, 2016 12.
Article in English | MEDLINE | ID: mdl-27701029

ABSTRACT

Experimental and clinical observations indicate that amyloid-ß1-42 (Aß1-42) peptide not only represents a major actor in neurodegenerative mechanisms but also induce hyperexcitation in individual neurons and neural circuits. In this abnormal excitability, possibly leading to seizures, the D1 dopamine (DA) receptors may play a role. Cerebrospinal fluid levels of Aß1-42 were measured in patients with late-onset epilepsy of unknown etiology. Moreover, the effect of amyloid peptide on the hippocampal epileptic threshold and synaptic plasticity and its link to D1 receptor function were tested in experimental mouse model of cerebral amyloidosis and in acute model of Aß1-42-induced neurotoxicity. Among 272 evaluated epileptic patients, aged >55 years, 35 suffered from late-onset epilepsy of unknown etiology. In these subjects, cerebrospinal fluid Aß1-42 levels were measured. The effects of Aß1-42, amyloid oligomers, and D1 receptor modulation on epileptic threshold were analyzed by electrophysiological recordings in the dentate gyrus of mice hippocampal slices. We found that Aß1-42 levels were significantly decreased in cerebrospinal fluid of patients with late-onset epilepsy of unknown etiology with respect to controls suggesting the cerebral deposition of this peptide in these patients. Aß1-42 enhanced epileptic activity in mice through a mechanism involving increased surface expression of D1 receptor, and this effect was mimicked by D1 receptor stimulation and blocked by SCH 23390, a D1 receptor antagonist. Aß1-42 may contribute to the pathophysiology of late-onset epilepsy of unknown origin. Our preclinical findings indicate that the D1 receptor is involved in mediating the epileptic effects of Aß1-42. This novel link between Aß1-42 and D1 receptor signaling might represent a potential therapeutic target.


Subject(s)
Amyloid beta-Peptides/metabolism , Epilepsy/etiology , Peptide Fragments/metabolism , Receptors, Dopamine D1/physiology , Aged , Aged, 80 and over , Alzheimer Disease/etiology , Alzheimer Disease/genetics , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Benzazepines/pharmacology , Disease Models, Animal , Epilepsy/genetics , Female , Humans , Male , Mice, Transgenic , Middle Aged , Peptide Fragments/cerebrospinal fluid , Receptors, Dopamine D1/antagonists & inhibitors
20.
Sci Rep ; 6: 20926, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26887636

ABSTRACT

Cognitive impairment is common in multiple sclerosis (MS). Unfortunately, the synaptic and molecular mechanisms underlying MS-associated cognitive dysfunction are largely unknown. We explored the presence and the underlying mechanism of cognitive and synaptic hippocampal dysfunction during the remission phase of experimental MS. Experiments were performed in a chronic-relapsing experimental autoimmune encephalomyelitis (EAE) model of MS, after the resolution of motor deficits. Immunohistochemistry and patch-clamp recordings were performed in the CA1 hippocampal area. The hole-board was utilized as cognitive/behavioural test. In the remission phase of experimental MS, hippocampal microglial cells showed signs of activation, CA1 hippocampal synapses presented an impaired long-term potentiation (LTP) and an alteration of spatial tests became evident. The activation of hippocampal microglia mediated synaptic and cognitive/behavioural alterations during EAE. Specifically, LTP blockade was found to be caused by the reactive oxygen species (ROS)-producing enzyme nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. We suggest that in the remission phase of experimental MS microglia remains activated, causing synaptic dysfunctions mediated by NADPH oxidase. Inhibition of microglial activation and NADPH oxidase may represent a promising strategy to prevent neuroplasticity impairment associated with active neuro-inflammation, with the aim to improve cognition and counteract MS disease progression.


Subject(s)
Behavior, Animal , CA1 Region, Hippocampal , Cognition , Long-Term Potentiation , Microglia , Multiple Sclerosis , NADPH Oxidases/metabolism , Animals , CA1 Region, Hippocampal/enzymology , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/physiopathology , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Enzyme Activation , Female , Mice , Microglia/enzymology , Microglia/pathology , Multiple Sclerosis/enzymology , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL