Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Brain Pathol ; 32(3): e13022, 2022 05.
Article in English | MEDLINE | ID: mdl-34585475

ABSTRACT

A preclinical strategy to broaden the search of potentially effective treatments in amyotrophic lateral sclerosis (ALS) relies on identifying factors controlling motor neuron (MN) excitability. These partners might be part of still unknown pathogenic pathways and/or useful for the design of new interventions to affect disease progression. In this framework, the bioactive membrane-derived phospholipid lysophosphatidic acid (LPA) affects MN excitability through LPA receptor 1 (LPA1 ). Furthermore, LPA1  knockdown is neuroprotective in transgenic ALS SOD1-G93A mice. On this basis, we raised the hypothesis that the major LPA-synthesizing ectoenzyme, autotaxin (ATX), regulates MN excitability and is a potential target to modulate disease development in ALS mice. We show here that PF-8380, a specific ATX inhibitor, reduced intrinsic membrane excitability (IME) of hypoglossal MNs in brainstem slices, supporting that baseline ATX activity regulates MN IME. PF-8380-induced alterations were prevented by a small-interfering RNA directed against mRNA for lpa1 . These outcomes support that impact of ATX-originated lysophospholipids on MN IME engages, at least, the G-protein-coupled receptor LPA1 . Interestingly, mRNAatx levels increased in the spinal cord of pre-symptomatic (1-2 months old) SOD1-G93A mice, thus preceding MN loss. The rise in transcripts levels also occurred in cultured spinal cord MNs from SOD1-G93A embryos, suggesting that mRNAatx upregulation in MNs is an etiopathogenic event in the ALS cell model. Remarkably, chronic administration in the drinking water of the orally bioavailable ATX inhibitor PF-8380 delayed MN loss, motor deterioration and prolonged life span in ALS mice. Treatment also led to a reduction in LPA1 -immunoreactive patches in transgenic animals mostly in MNs. These outcomes support that neuroprotective effects of interfering with ATX in SOD1-G93A mice rely, at least in part, on LPA1  knockdown in MNs. Therefore, we propose ATX as a potential target and/or a biomarker in ALS and highlight ATX inhibitors as reasonable tools with therapeutic usefulness for this lethal pathology.


Subject(s)
Amyotrophic Lateral Sclerosis , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Disease Models, Animal , Mice , Mice, Transgenic , Motor Neurons/metabolism , Nerve Degeneration/pathology , RNA, Messenger/metabolism , Spinal Cord/pathology , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/metabolism
2.
Front Mol Neurosci ; 14: 788039, 2021.
Article in English | MEDLINE | ID: mdl-34938160

ABSTRACT

Intrinsic membrane excitability (IME) sets up neuronal responsiveness to synaptic drive. Several neurotransmitters and neuromodulators, acting through G-protein-coupled receptors (GPCRs), fine-tune motoneuron (MN) IME by modulating background K+ channels TASK1. However, intracellular partners linking GPCRs to TASK1 modulation are not yet well-known. We hypothesized that isoform 2 of rho-kinase (ROCK2), acting as downstream GPCRs, mediates adjustment of MN IME via TASK1. Electrophysiological recordings were performed in hypoglossal MNs (HMNs) obtained from adult and neonatal rats, neonatal knockout mice for TASK1 (task1 -/-) and TASK3 (task3 -/-, the another highly expressed TASK subunit in MNs), and primary cultures of embryonic spinal cord MNs (SMNs). Small-interfering RNA (siRNA) technology was also used to knockdown either ROCK1 or ROCK2. Furthermore, ROCK activity assays were performed to evaluate the ability of various physiological GPCR ligands to stimulate ROCK. Microiontophoretically applied H1152, a ROCK inhibitor, and siRNA-induced ROCK2 knockdown both depressed AMPAergic, inspiratory-related discharge activity of adult HMNs in vivo, which mainly express the ROCK2 isoform. In brainstem slices, intracellular constitutively active ROCK2 (aROCK2) led to H1152-sensitive HMN hyper-excitability. The aROCK2 inhibited pH-sensitive and TASK1-mediated currents in SMNs. Conclusively, aROCK2 increased IME in task3 -/-, but not in task1 -/- HMNs. MN IME was also augmented by the physiological neuromodulator lysophosphatidic acid (LPA) through a mechanism entailing Gαi/o-protein stimulation, ROCK2, but not ROCK1, activity and TASK1 inhibition. Finally, two neurotransmitters, TRH, and 5-HT, which are both known to increase MN IME by TASK1 inhibition, stimulated ROCK2, and depressed background resting currents via Gαq/ROCK2 signaling. These outcomes suggest that LPA and several neurotransmitters impact MN IME via Gαi/o/Gαq-protein-coupled receptors, downstream ROCK2 activation, and subsequent inhibition of TASK1 channels.

3.
Neuropathol Appl Neurobiol ; 47(7): 1004-1018, 2021 12.
Article in English | MEDLINE | ID: mdl-33508894

ABSTRACT

AIMS: Alterations in excitability represent an early hallmark in Amyotrophic Lateral Sclerosis (ALS). Therefore, deciphering the factors that impact motor neuron (MN) excitability offers an opportunity to uncover further aetiopathogenic mechanisms, neuroprotective agents, therapeutic targets, and/or biomarkers in ALS. Here, we hypothesised that the lipokine lysophosphatidic acid (lpa) regulates MN excitability via the G-protein-coupled receptor lpa1 . Then, modulating lpa1 -mediated signalling might affect disease progression in the ALS SOD1-G93A mouse model. METHODS: The influence of lpa-lpa1 signalling on the electrical properties, Ca2+ dynamic and survival of MNs was tested in vitro. Expression of lpa1 in cultured MNs and in the spinal cord of SOD1-G93A mice was analysed. ALS mice were chronically treated with a small-interfering RNA against lpa1 (siRNAlpa1 ) or with the lpa1 inhibitor AM095. Motor skills, MN loss, and lifespan were evaluated. RESULTS: AM095 reduced MN excitability. Conversely, exogenous lpa increased MN excitability by modulating task1 'leak' potassium channels downstream of lpa1 . Lpa-lpa1 signalling evoked an excitotoxic response in MNs via voltage-sensitive calcium channels. Cultured SOD1-G93A MNs displayed lpa1 upregulation and heightened vulnerability to lpa. In transgenic mice, lpa1 was upregulated mostly in spinal cord MNs before cell loss. Chronic administration of either siRNAlpa1 or AM095 reduced lpa1 expression at least in MNs, delayed MN death, improved motor skills, and prolonged life expectancy of ALS mice. CONCLUSIONS: These results suggest that stressed lpa-lpa1 signalling contributes to MN degeneration in SOD1-G93A mice. Consequently, disrupting lpa1 slows down disease progression. This highlights LPA1 signalling as a potential target and/or biomarker in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Motor Neurons/pathology , Receptors, Lysophosphatidic Acid/metabolism , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis/genetics , Animals , Disease Models, Animal , Disease Progression , Mice, Transgenic , Microglia/pathology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Spinal Cord/pathology
4.
Brain Struct Funct ; 225(9): 2871-2884, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33130922

ABSTRACT

Synaptic remodeling during early postnatal development lies behind neuronal networks refinement and nervous system maturation. In particular, the respiratory system is immature at birth and is subjected to significant postnatal development. In this context, the excitatory/inhibitory balance dramatically changes in the respiratory-related hypoglossal nucleus (HN) during the 3 perinatal weeks. Since, development abnormalities of hypoglossal motor neurons (HMNs) are associated with sudden infant death syndrome and obstructive sleep apnea, deciphering molecular partners behind synaptic remodeling in the HN is of basic and clinical relevance. Interestingly, a transient expression of the neuronal isoform of nitric oxide (NO) synthase (NOS) occurs in HMNs at neonatal stage that disappears before postnatal day 21 (P21). NO, in turn, is a determining factor for synaptic refinement in several physiopathological conditions. Here, intracerebroventricular chronic administration (P7-P21) of the broad spectrum NOS inhibitor L-NAME (N(ω)-nitro-L-arginine methyl ester) differentially affected excitatory and inhibitory rearrangement during this neonatal interval in the rat. Whilst L-NAME led to a reduction in the number of excitatory structures, inhibitory synaptic puncta were increased at P21 in comparison to administration of the inactive stereoisomer D-NAME. Finally, L-NAME decreased levels of the phosphorylated form of myosin light chain in the nucleus, which is known to regulate the actomyosin contraction apparatus. These outcomes indicate that physiologically synthesized NO modulates excitatory/inhibitory balance during early postnatal development by acting as an anti-synaptotrophic and/or synaptotoxic factor for inhibitory synapses, and as a synaptotrophin for excitatory ones. The mechanism of action could rely on the modulation of the actomyosin contraction apparatus.


Subject(s)
Brain Stem/growth & development , Motor Neurons/metabolism , Nitric Oxide Synthase Type I/metabolism , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , Animals , Brain Stem/metabolism , Female , Membrane Glycoproteins , Rats, Wistar , Receptors, Interleukin-1
5.
Nat Commun ; 10(1): 3784, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31439839

ABSTRACT

Disruption in membrane excitability contributes to malfunction and differential vulnerability of specific neuronal subpopulations in a number of neurological diseases. The adaptor protein p11, and background potassium channel TASK1, have overlapping distributions in the CNS. Here, we report that the transcription factor Sp1 controls p11 expression, which impacts on excitability by hampering functional expression of TASK1. In the SOD1-G93A mouse model of ALS, Sp1-p11-TASK1 dysregulation contributes to increased excitability and vulnerability of motor neurons. Interference with either Sp1 or p11 is neuroprotective, delaying neuron loss and prolonging lifespan in this model. Nitrosative stress, a potential factor in human neurodegeneration, stimulated Sp1 expression and human p11 promoter activity, at least in part, through a Sp1-binding site. Disruption of Sp1 or p11 also has neuroprotective effects in a traumatic model of motor neuron degeneration. Together our work suggests the Sp1-p11-TASK1 pathway is a potential target for treatment of degeneration of motor neurons.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Annexin A2/metabolism , Motor Neurons/pathology , Nerve Degeneration/pathology , Nerve Tissue Proteins/genetics , Potassium Channels, Tandem Pore Domain/genetics , S100 Proteins/metabolism , Sp1 Transcription Factor/metabolism , Amyotrophic Lateral Sclerosis/etiology , Animals , Cell Membrane/pathology , Disease Models, Animal , Female , Gene Expression Regulation , Gene Knockdown Techniques , HEK293 Cells , Humans , Male , Membrane Potentials , Mice , Mice, Transgenic , Motor Neurons/cytology , Nerve Degeneration/etiology , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Primary Cell Culture , Promoter Regions, Genetic , Rats , Sp1 Transcription Factor/genetics , Spinal Cord/cytology , Spinal Cord/pathology
6.
PLoS Biol ; 13(5): e1002153, 2015 May.
Article in English | MEDLINE | ID: mdl-25996636

ABSTRACT

Synaptic communication is a dynamic process that is key to the regulation of neuronal excitability and information processing in the brain. To date, however, the molecular signals controlling synaptic dynamics have been poorly understood. Membrane-derived bioactive phospholipids are potential candidates to control short-term tuning of synaptic signaling, a plastic event essential for information processing at both the cellular and neuronal network levels in the brain. Here, we showed that phospholipids affect excitatory and inhibitory neurotransmission by different degrees, loci, and mechanisms of action. Signaling triggered by lysophosphatidic acid (LPA) evoked rapid and reversible depression of excitatory and inhibitory postsynaptic currents. At excitatory synapses, LPA-induced depression depended on LPA1/Gαi/o-protein/phospholipase C/myosin light chain kinase cascade at the presynaptic site. LPA increased myosin light chain phosphorylation, which is known to trigger actomyosin contraction, and reduced the number of synaptic vesicles docked to active zones in excitatory boutons. At inhibitory synapses, postsynaptic LPA signaling led to dephosphorylation, and internalization of the GABAAγ2 subunit through the LPA1/Gα12/13-protein/RhoA/Rho kinase/calcineurin pathway. However, LPA-induced depression of GABAergic transmission was correlated with an endocytosis-independent reduction of GABAA receptors, possibly by GABAAγ2 dephosphorylation and subsequent increased lateral diffusion. Furthermore, endogenous LPA signaling, mainly via LPA1, mediated activity-dependent inhibitory depression in a model of experimental synaptic plasticity. Finally, LPA signaling, most likely restraining the excitatory drive incoming to motoneurons, regulated performance of motor output commands, a basic brain processing task. We propose that lysophospholipids serve as potential local messengers that tune synaptic strength to precedent activity of the neuron.


Subject(s)
Lysophospholipids/metabolism , Motor Neurons/physiology , Neuronal Plasticity , Synaptic Transmission , Animals , Calcineurin/metabolism , Female , Male , Mice , Patch-Clamp Techniques , Pregnancy , Rats, Wistar , Receptors, GABA-A/metabolism , Synapses/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
7.
Neuropharmacology ; 92: 69-79, 2015 May.
Article in English | MEDLINE | ID: mdl-25595101

ABSTRACT

Impairment of motor skills is one of the most common acute adverse effects of cannabis. Related studies have focused mainly on psychomotor alterations, and little is known about the direct impact of cannabinoids (CBs) on motoneuron physiology. As key modulators of synaptic function, CBs regulate multiple neuronal functions and behaviors. Presynaptic CB1 mediates synaptic strength depression by inhibiting neurotransmitter release, via a poorly understood mechanism. The present study examined the effect of CB agonists on excitatory synaptic inputs incoming to hypoglossal motoneurons (HMNs) in vitro and in vivo. The endocannabinoid anandamide (AEA) and the synthetic CB agonist WIN 55,212-2 rapidly and reversibly induced short-term depression (STD) of glutamatergic synapses on motoneurons by a presynaptic mechanism. Presynaptic effects were fully reversed by the CB1-selective antagonist AM281. Electrophysiological and electron microscopy analysis showed that WIN 55,212-2 reduced the number of synaptic vesicles (SVs) docked to active zones in excitatory boutons. Given that AM281 fully abolished depolarization-induced depression of excitation, motoneurons can be feasible sources of CBs, which in turn act as retrograde messengers regulating synaptic function. Finally, microiontophoretic application of the CB agonist O-2545 reversibly depressed, presumably via CB1, glutamatergic inspiratory-related activity of HMNs in vivo. Therefore, evidence support that CBs, via presynaptic CB1, induce excitatory STD by reducing the readily releasable pool of SVs at excitatory synapses, then attenuating motoneuron activity. These outcomes contribute a possible mechanistic basis for cannabis-associated motor performance disturbances such as ataxia, dysarthria and dyscoordination.


Subject(s)
Brain Stem/cytology , Cannabinoid Receptor Agonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Synapses/drug effects , Synaptic Vesicles/drug effects , Animals , Animals, Newborn , Benzopyrans/pharmacology , Brain Stem/ultrastructure , Dose-Response Relationship, Drug , Electric Stimulation , Excitatory Amino Acid Agents/pharmacology , Female , Imidazoles/pharmacology , In Vitro Techniques , Iontophoresis , Male , Microscopy, Electron, Transmission , Motor Neurons/cytology , Motor Neurons/drug effects , Motor Neurons/physiology , Patch-Clamp Techniques , Rats , Rats, Wistar , Synapses/ultrastructure
8.
J Neurosci ; 32(1): 68-84, 2012 Jan 04.
Article in English | MEDLINE | ID: mdl-22219271

ABSTRACT

Rho-associated kinase (ROCK) regulates neural cell migration, proliferation and survival, dendritic spine morphology, and axon guidance and regeneration. There is, however, little information about whether ROCK modulates the electrical activity and information processing of neuronal circuits. At neonatal stage, ROCKα is expressed in hypoglossal motoneurons (HMNs) and in their afferent inputs, whereas ROCKß is found in synaptic terminals on HMNs, but not in their somata. Inhibition of endogenous ROCK activity in neonatal rat brainstem slices failed to modulate intrinsic excitability of HMNs, but strongly attenuated the strength of their glutamatergic and GABAergic synaptic inputs. The mechanism acts presynaptically to reduce evoked neurotransmitter release. ROCK inhibition increased myosin light chain (MLC) phosphorylation, which is known to trigger actomyosin contraction, and reduced the number of synaptic vesicles docked to active zones in excitatory boutons. Functional and ultrastructural changes induced by ROCK inhibition were fully prevented/reverted by MLC kinase (MLCK) inhibition. Furthermore, ROCK inhibition drastically reduced the phosphorylated form of p21-associated kinase (PAK), which directly inhibits MLCK. We conclude that endogenous ROCK activity is necessary for the normal performance of motor output commands, because it maintains afferent synaptic strength, by stabilizing the size of the readily releasable pool of synaptic vesicles. The mechanism of action involves a tonic inhibition of MLCK, presumably through PAK phosphorylation. This mechanism might be present in adults since unilateral microinjection of ROCK or MLCK inhibitors into the hypoglossal nucleus reduced or increased, respectively, whole XIIth nerve activity.


Subject(s)
Hypoglossal Nerve/enzymology , Motor Neurons/enzymology , Presynaptic Terminals/enzymology , Synaptic Transmission/physiology , Synaptic Vesicles/enzymology , rho-Associated Kinases/physiology , Animals , Animals, Newborn , Female , Hypoglossal Nerve/growth & development , Hypoglossal Nerve/ultrastructure , MAP Kinase Signaling System/physiology , Male , Motor Neurons/drug effects , Motor Neurons/ultrastructure , Organ Culture Techniques , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Rats , Rats, Wistar , Synaptic Transmission/drug effects , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure , rho-Associated Kinases/antagonists & inhibitors
9.
Methods Mol Biol ; 704: 197-223, 2011.
Article in English | MEDLINE | ID: mdl-21161639

ABSTRACT

Nitric oxide, a free gaseous signalling molecule, has attracted the attention of numerous biologists and has been implicated in the regulation of the cardiovascular, nervous and immune system. However, the cellular mechanisms mediating nitric oxide modulation remain unclear. Upregulation by gene over-expression or down-regulation by gene inactivation of nitric oxide synthase has generated quantitative changes in abundance thereby permitting functional insights. We have tested and proved that genetic nitric oxide synthase antagonism using viral vectors, particularly with dominant negative mutants and microRNA 30-based short hairpin RNA, is an efficient and effective experimental approach to manipulate nitric oxide synthase expression both in vitro and in vivo.


Subject(s)
Cardiovascular Diseases/pathology , Cardiovascular Diseases/therapy , Genetic Therapy/methods , Genetic Vectors/genetics , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Peripheral Nervous System Diseases/therapy , Adenoviridae/genetics , Animals , Cardiovascular Diseases/enzymology , Cardiovascular Diseases/genetics , Cell Line , Humans , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Peripheral Nervous System Diseases/enzymology , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/pathology , Rats
10.
Mol Neurobiol ; 43(1): 41-66, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21190141

ABSTRACT

Synapse elimination is the main factor responsible for the cognitive decline accompanying many of the neuropathological conditions affecting humans. Synaptic stripping of motoneurons is also a common hallmark of several motor pathologies. Therefore, knowledge of the molecular basis underlying this plastic process is of central interest for the development of new therapeutic tools. Recent advances from our group highlight the role of nitric oxide (NO) as a key molecule triggering synapse loss in two models of motor pathologies. De novo expression of the neuronal isoform of NO synthase (nNOS) in motoneurons commonly occurs in response to the physical injury of a motor nerve and in the course of amyotrophic lateral sclerosis. In both conditions, this event precedes synaptic withdrawal from motoneurons. Strikingly, nNOS-synthesized NO is "necessary" and "sufficient" to induce synaptic detachment from motoneurons. The mechanism involves a paracrine/retrograde action of NO on pre-synaptic structures, initiating a downstream signaling cascade that includes sequential activation of (1) soluble guanylyl cyclase, (2) cyclic guanosine monophosphate-dependent protein kinase, and (3) RhoA/Rho kinase (ROCK) signaling. Finally, ROCK activation promotes phosphorylation of regulatory myosin light chain, which leads to myosin activation and actomyosin contraction. This latter event presumably contributes to the contractile force to produce ending axon retraction. Several findings support that this mechanism may operate in the most prevalent neurodegenerative diseases.


Subject(s)
Motor Neurons/pathology , Motor Neurons/ultrastructure , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Nitric Oxide/metabolism , Presynaptic Terminals/metabolism , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Cell Communication , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Motor Neurons/metabolism , Nitric Oxide Synthase Type I/metabolism , Peripheral Nervous System Diseases/pathology , Peripheral Nervous System Diseases/physiopathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
11.
Hippocampus ; 21(8): 827-34, 2011 Aug.
Article in English | MEDLINE | ID: mdl-20607795

ABSTRACT

Tau is a neuronal microtubule-associated protein implicated in microtubules stabilization, axonal establishment and elongation during neuronal morphogenesis. Because of its elevated expression in neocortical regions and hippocampus, tau might play a role in sculpting collective neural responses underlying slow and fast brain oscillations and/or long-range synchronization patterns between hippocampus and neocortex. To test this hypothesis, local field potentials were recorded in tau-deficient (tau(-/-) ) and wild-type mice from different neocortical regions and from the hippocampus during spontaneous motor exploratory behavior. We found that tau(-/-) mice showed hippocampal theta slowing and reduced levels of gamma long-range synchronization involving the frontal cortex. We hypothesize that the lack of normal phosphorylated tau during early stages of development might influence the maturation of parvalbumin interneurons affecting the spatiotemporal structure of long-range gamma synchronization. Also, the proper functioning of gap-junction channels might be compromised by the absence of tau in hippocampal networks. Altogether, these results provide novel insights into the functional role of tau protein in the formation of collective neural responses and emergence of neocortical-hippocampal interactions in the mammalian brain.


Subject(s)
Hippocampus/physiology , Neocortex/physiology , Theta Rhythm/physiology , tau Proteins , Animals , Electrophysiology , Evoked Potentials, Motor , Exploratory Behavior , Interneurons/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Neurological , Parvalbumins/metabolism , tau Proteins/deficiency , tau Proteins/immunology , tau Proteins/metabolism
12.
Brain Pathol ; 21(1): 1-15, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20653686

ABSTRACT

Excitotoxicity is a widely studied mechanism underlying motoneuron degeneration in amyotrophic lateral sclerosis (ALS). Synaptic alterations that produce an imbalance in the ratio of inhibitory/excitatory synapses are expected to promote or protect against motoneuron excitotoxicity. In ALS patients, motoneurons suffer a reduction in their synaptic coverage, as in the transition from the presymptomatic (2-month-old) to early-symptomatic (3-month-old) stage of the hSOD1(G93A) mouse model of familial ALS. Net synapse loss resulted from inhibitory bouton loss and excitatory synapse gain. Furthermore, in 3-month-old transgenic mice, remaining inhibitory but not excitatory boutons attached to motoneurons showed reduction in the active zone length and in the spatial density of synaptic vesicles in the releasable pool near the active zone. Bouton degeneration/loss seems to be mediated by bouton vacuolization and by mechanical displacement due to swelling vacuolated dendrites. In addition, chronic treatment with a nitric oxide (NO) synthase inhibitor avoided inhibitory loss but not excitatory gain. These results indicate that NO mediates inhibitory loss occurring from the pre- to early-symptomatic stage of hSOD1(G93A) mice. This work contributes new insights on ALS pathogenesis, recognizing synaptic re-arrangement onto motoneurons as a mechanism favoring disease progression rather than as a protective homeostatic response against excitotoxic events.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Motor Neurons/pathology , Nerve Degeneration/pathology , Superoxide Dismutase/genetics , Synapses/pathology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Analysis of Variance , Animals , Blotting, Western , Disease Models, Animal , Immunohistochemistry , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron , Motor Neurons/metabolism , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nitric Oxide/metabolism , Superoxide Dismutase-1 , Synapses/genetics , Synapses/metabolism , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
13.
J Physiol ; 588(Pt 18): 3425-43, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20660560

ABSTRACT

Dysregulation of protein expression, function and/or aggregation is a hallmark of a number of neuropathological conditions. Among them, upregulation and/or de novo expression of the neuronal isoform of nitric oxide (NO) synthase (nNOS) commonly occurs in diverse neurodegenerative diseases and in axotomized motoneurons. We used adenoviral (AVV) and lentiviral (LVV) vectors to study the effects of de novo nNOS expression on the functional properties and synaptic array of motoneurons. AVV-nNOS injection into the genioglossus muscle retrogradely transduced neonatal hypoglossal motoneurons (HMNs). Ratiometric real-time NO imaging confirmed that transduced HMNs generated NO gradients in brain parenchyma (space constant: 12.3 µm) in response to a glutamatergic stimulus. Unilateral AVV-nNOS microinjection in the hypoglossal nucleus of adult rats induced axotomy-like changes in HMNs. Specifically, we found alterations in axonal conduction properties and the recruitment order of motor units and reductions in responsiveness to synaptic drive and in the linear density of synaptophysin-positive puncta opposed to HMN somata. Functional alterations were fully prevented by chronic treatment with nNOS or soluble guanylyl cyclase inhibitors. Synaptic and functional changes were also completely avoided by prior intranuclear injection of a neuron-specific LVV system for miRNA-mediated nNOS knock-down (LVV-miR-shRNA/nNOS). Furthermore, synaptic and several functional changes evoked by XIIth nerve injury were to a large extent prevented by intranuclear administration of LVV-miR-shRNA/nNOS. We suggest that nNOS up-regulation creates a repulsive NO gradient for synaptic boutons underlying most of the functional impairment undergone by injured motoneurons. This further strengthens the case for nNOS targeting as a plausible strategy for treatment of peripheral neuropathies and neurodegenerative disorders.


Subject(s)
Axotomy , Motor Neurons/enzymology , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Animals , Animals, Genetically Modified , Animals, Newborn , Carbon Dioxide , Endothelium/metabolism , Gene Expression Regulation, Enzymologic , Hypoglossal Nerve/metabolism , Hypoglossal Nerve/pathology , Male , Motor Neurons/cytology , Motor Neurons/physiology , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , Rats , Rats, Wistar , Synapses , Time Factors
14.
J Alzheimers Dis ; 21(2): 411-21, 2010.
Article in English | MEDLINE | ID: mdl-20555133

ABSTRACT

Evidence has shown that the lack of tau produces subtle changes in neuronal structure and modest impairment in complex behaviors, suggesting compensatory mechanisms carried out by other neuronal microtubule-associated proteins. Here we show major abnormalities in sleep-wake cycle of tau-deficient animals including increased wakefulness duration and decreased non-rapid eye movement (NREM) sleep time, a higher number of state transitions between NREM and wake, and shortened sleep bouts. Altered sleep structure in tau-/- mice was accompanied by a significant decline in delta power together with an enhanced spectral density of sleep spindles during NREM sleep. No significant differences were observed in rapid eye movement (REM) sleep between the two mouse strains. Taken together, these results suggest that tau indirectly participates in the regulation of the sleep-wake cycle modulating not only the control and maintenance of global brain states but also the cerebral oscillatory patterns underlying sleep-wake states.


Subject(s)
Sleep, REM/physiology , Wakefulness/physiology , tau Proteins/physiology , Animals , Behavior, Animal/physiology , Brain/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Mutant Strains , Microtubules/physiology , Periodicity , Photoperiod , tau Proteins/genetics
15.
J Neurosci Res ; 88(9): 1846-57, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20143424

ABSTRACT

Physical injury to a nerve is the most frequent cause of acquired peripheral neuropathy, which is responsible for loss of motor, sensory and/or autonomic functions. Injured axons in the peripheral nervous system maintain the capacity to regenerate in adult mammals. However, after nerve transection, stumps of damaged nerves must be surgically joined to guide regenerating axons into the distal nerve stump. Even so, severe functional limitations persist after restorative surgery. Therefore, the identification of molecules that regulate degenerative and regenerative processes is indispensable in developing therapeutic tools to accelerate and improve functional recovery. Here, I consider the role of nitric oxide (NO) synthesized by the three major isoforms of NO synthases (NOS) in motor neuropathy. Neuronal NOS (nNOS) seems to be the primary source of NO that is detrimental to the survival of injured motoneurons. Endothelial NOS (eNOS) appears to be the major source of NO that interferes with axonal regrowth, at least soon after injury. Finally, NO derived from inducible NOS (iNOS) or nNOS is critical to the process of lipid breakdown for Wallerian degeneration and thereby benefits axonal regrowth. Specific inhibitors of these isoforms can be used to protect injured neurons from degeneration and promote axonal regeneration. A cautious proposal for the treatment of acquired motor neuropathy using therapeutic tools that locally interfere with eNOS/nNOS activities seems to merit consideration.


Subject(s)
Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Peripheral Nervous System Diseases/enzymology , Peripheral Nervous System/injuries , Recovery of Function/physiology , Animals , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Peripheral Nervous System/enzymology , Peripheral Nervous System Diseases/drug therapy , Recovery of Function/drug effects
16.
Neurosci Lett ; 471(2): 119-24, 2010 Mar 03.
Article in English | MEDLINE | ID: mdl-20093168

ABSTRACT

The loss of intimate contact with axons triggers Schwann cells (SCs) to switch from a myelin-producing phenotype to a dedifferentiated, proliferating non-myelin-forming state after nerve injury. SC dedifferentiation is required for effective nerve regeneration. Negative regulators of SC dedifferentiation are promising targets to accelerate function recovery in acquired peripheral neuropathies. We recently reported that nitric oxide (NO) synthesized by endothelial NO synthase (eNOS) slows down functional recovery and axon regeneration after XIIth nerve crushing. This harmful action could be effected by a NO-delaying action on SC dedifferentiation. Adenoviral vectors directing the expression of a dominant negative mutant for eNOS (AVV-TeNOS) or the enhanced green fluorescent protein (AVV-eGFP) were individually injected into the distal stump just after XIIth nerve crushing. Growth-associated protein 43 (GAP-43), strongly over-expressed in dedifferentiated SCs and regenerating axons, was up-regulated in AVV-TeNOS-transduced nerves relative to AVV-eGFP-treated nerves. AVV-TeNOS increased the number of GAP-43-positive cells and bands of Bungner but did not alter the number of Hoechst-positive nuclei relative to AVV-eGFP. These results signal endothelial NO as a negative regulator of the SC dedifferentiation process, but not of SC proliferation rate, after nerve injury. Vascular-derived factors should be taken into account as feasible extrinsic regulators of SC plasticity.


Subject(s)
Endothelium, Vascular/metabolism , Nitric Oxide/physiology , Peripheral Nerves/pathology , Schwann Cells/physiology , Animals , Axons/metabolism , Cell Dedifferentiation , Cell Proliferation , GAP-43 Protein/biosynthesis , Green Fluorescent Proteins/genetics , Male , Nerve Crush , Nitric Oxide Synthase Type III/genetics , Peripheral Nerve Injuries , Peripheral Nerves/metabolism , Rats , Rats, Wistar , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Schwann Cells/metabolism
17.
J Neurosci ; 30(3): 973-84, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-20089906

ABSTRACT

The molecular signaling that underpins synapse loss in neuropathological conditions remains unknown. Concomitant upregulation of the neuronal nitric oxide (NO) synthase (nNOS) in neurodegenerative processes places NO at the center of attention. We found that de novo nNOS expression was sufficient to induce synapse loss from motoneurons at adult and neonatal stages. In brainstem slices obtained from neonatal animals, this effect required prolonged activation of the soluble guanylyl cyclase (sGC)/protein kinase G (PKG) pathway and RhoA/Rho kinase (ROCK) signaling. Synapse elimination involved paracrine/retrograde action of NO. Furthermore, before bouton detachment, NO increased synapse myosin light chain phosphorylation (p-MLC), which is known to trigger actomyosin contraction and neurite retraction. NO-induced MLC phosphorylation was dependent on cGMP/PKG-ROCK signaling. In adulthood, motor nerve injury induced NO/cGMP-dependent synaptic stripping, strongly affecting ROCK-expressing synapses, and increased the percentage of p-MLC-expressing inputs before synapse destabilization. We propose that this molecular cascade could trigger synapse loss underlying early cognitive/motor deficits in several neuropathological states.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Motor Neurons/pathology , Myosin Light Chains/metabolism , Nitric Oxide Synthase Type I/metabolism , Synapses/pathology , rho-Associated Kinases/metabolism , Analysis of Variance , Animals , Animals, Newborn , Brain Stem/cytology , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , DNA-Binding Proteins/genetics , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins/genetics , Humans , Hypoglossal Nerve Diseases/pathology , In Vitro Techniques , Male , Microscopy, Immunoelectron/methods , Motor Neurons/drug effects , Motor Neurons/ultrastructure , Nitric Oxide/pharmacology , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/pharmacology , Nuclear Proteins/genetics , Patch-Clamp Techniques , Phosphorylation/drug effects , Phosphorylation/physiology , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Rats , Rats, Wistar , Synapses/drug effects , Synapses/ultrastructure , Synaptic Potentials/drug effects , Synaptic Potentials/genetics , Synaptophysin/metabolism , Transfection , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , rho-Associated Kinases/antagonists & inhibitors
18.
Eur J Neurosci ; 28(1): 107-16, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18616563

ABSTRACT

Motoneurons integrate interneuronal activity into commands for skeletal muscle contraction and relaxation to perform motor actions. Hypoglossal motoneurons (HMNs) are involved in essential motor functions such as breathing, mastication, swallowing and phonation. We have investigated the role of the gaseous molecule nitric oxide (NO) in the regulation of the inspiratory-related activity of HMNs in order to further understand how neural activity is transformed into motor activity. In adult rats, we observed nitrergic fibers and bouton-like structures in close proximity to motoneurons, which normally lack the molecular machinery to synthesize NO. In addition, immunohistochemistry studies demonstrated that perfusion of animals with a NO donor resulted in an increase in the levels of cyclic guanosine monophosphate (cGMP) in motoneurons, which express the soluble guanylyl cyclase (sGC) in the hypoglossal nucleus. Modulators of the NO/cGMP pathway were micro-iontophoretically applied while performing single-unit extracellular recordings in the adult decerebrated rat. Application of a NO synthase inhibitor or a sGC inhibitor induced a statistically significant reduction in the inspiratory-related activity of HMNs. However, excitatory effects were observed by ejection of a NO donor or a cell-permeable analogue of cGMP. In slice preparations, application to the bath of a NO donor evoked membrane depolarization and a decrease in rheobase, which were prevented by co-addition to the bath of a sGC inhibitor. These effects were not prevented by reduction of the spontaneous synaptic activity. We conclude that NO from afferent fibers anterogradely modulates the inspiratory-related activity of HMNs by a cGMP-dependent mechanism in physiological conditions.


Subject(s)
Guanosine Monophosphate/metabolism , Hypoglossal Nerve/cytology , Inhalation/physiology , Motor Neurons/metabolism , Nitric Oxide/metabolism , Signal Transduction/physiology , Animals , Cell Membrane/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Enzyme Inhibitors/metabolism , Iontophoresis , Male , Motor Neurons/cytology , Patch-Clamp Techniques , Rats , Rats, Wistar
19.
J Comp Neurol ; 506(2): 339-46, 2008 Jan 10.
Article in English | MEDLINE | ID: mdl-18022945

ABSTRACT

Nitric oxide (NO) synthase (NOS) is developmentally regulated in the embryonic brain, where NO participates in cell proliferation, survival, and differentiation. In adults, NO inhibits neurogenesis under physiological conditions. This work investigates whether the NO action is preserved all along development up to adulthood or whether its effects in adults are a new feature acquired during brain maturation. The relationship between nitrergic neurons and precursors, as well as the functional consequences of pharmacological NOS inhibition, were comparatively analyzed in the subventricular zone (SVZ) and olfactory bulb (OB) of postnatal (P7) and adult (>P60) mouse brains. The SVZ was markedly reduced between P7 and adults, and, at both ages, neurons expressing neuronal NOS (nNOS) were found in its striatal limits. In postnatal mice, these nitrergic neurons contained PSA-NCAM, and their projections were scarce, whereas, in adults, mature nitrergic neurons, devoid of PSA-NCAM, presented abundant neuropil. In the OB, local proliferation almost disappeared in the transition to adulthood, and periglomerular nitrergic neurons, some of which were PSA-NCAM positive, were found in postnatal and adult mice. Administration of the NOS inhibitor L-NAME did not affect cell proliferation in the SVZ or in the OB of postnatal mice, whereas it significantly enhanced the number of mitotic cells in both regions in adults. Thus, the NO action on SVZ neurogenesis is a phenomenon that appears after the postnatal age, which is probably due to the germinal layer size reduction, allowing exposure of the NO-sensitive neural precursors to the NO produced in the SVZ-striatum limits.


Subject(s)
Aging/physiology , Cell Proliferation , Lateral Ventricles/cytology , Neurons/metabolism , Nitric Oxide/metabolism , Olfactory Bulb/cytology , Stem Cells/physiology , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Glutathione/metabolism , Green Fluorescent Proteins/metabolism , Mice , NADPH Dehydrogenase/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Neural Cell Adhesion Molecule L1/metabolism , Neurons/drug effects , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I/metabolism , Sialic Acids/metabolism , Stem Cells/drug effects
20.
J Neurosci ; 27(23): 6302-12, 2007 Jun 06.
Article in English | MEDLINE | ID: mdl-17554004

ABSTRACT

Glutamate-induced excitotoxicity, the most common pathological mechanism leading to neuronal death, may occur even with normal levels of glutamate if it coincides with a persistent enhancement of neuronal excitability. Neurons expressing nitric oxide (NO) synthase (NOS-I), which is upregulated in many human chronic neurodegenerative diseases, are highly susceptible to neurodegeneration. We hypothesized that chronic production of NO in damaged neurons may increase their intrinsic excitability via modulation of resting or "leak" K+ currents. Peripheral XIIth nerve injury in adult rats induced de novo NOS-I expression and an increased incidence of low-threshold motor units, the latter being prevented by chronic inhibition of the neuronal NO/cGMP pathway. Accordingly, sustained synthesis of NO maintained an enhanced basal activity in injured motoneurons that was slowly reverted (over the course of 2-3 h) by NOS-I inhibitors. In slice preparations, persistent, but not acute, activation of the NO/cGMP pathway evoked a robust augment in motoneuron excitability independent of synaptic activity. Furthermore, chronic activation of the NO/cGMP pathway fully suppressed TWIK-related acid-sensitive K+ (TASK) currents through a protein kinase G (PKG)-dependent mechanism. Finally, we found evidence for the involvement of this long-term mechanism in regulating membrane excitability of motoneurons, because their pH-sensitive currents were drastically reduced by nerve injury. This NO/cGMP/PKG-mediated modulation of TASK conductances might represent a new pathological mechanism that leads to hyperexcitability and sensitizes neurons to excitotoxic damage. It could explain why de novo expression of NOS-I and/or its overexpression makes them susceptible to neurodegeneration under pathological conditions.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Long-Term Potentiation/physiology , Neurons/metabolism , Nitric Oxide/metabolism , Potassium Channels/metabolism , Animals , Enzyme Inhibitors/pharmacology , Hypoglossal Nerve/drug effects , Hypoglossal Nerve/enzymology , Hypoglossal Nerve/pathology , Long-Term Potentiation/drug effects , Male , Neurons/drug effects , Neurons/enzymology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL