Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
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.
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
7.
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
8.
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
9.
Bioorg Med Chem Lett ; 19(17): 5176-81, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19635667

ABSTRACT

A series of 30 RCO-HfR-NH(2) derivatives show preference for the mouse MC1R vs MC3-5Rs. trans-4-HOC(6)H(4)CH=CHCO-HfR-NH(2) (13) [EC(50) (nM): MC1R 83, MC3R 20500, MC4R 18130 and MC5R 935; ratio 1:246:217:11] is 11 times more potent than the lead compound LK-394 Ph(CH(2))(3)CO-HfR-NH(2) (2) and only 11 times less potent than the native tridecapeptide alpha-MSH at mMC1R. Differences in conformations of 2 and 13 are discussed.


Subject(s)
Peptides/chemistry , Receptors, Melanocortin/agonists , Amino Acid Sequence , Animals , Binding Sites , Computer Simulation , Mice , Molecular Conformation , Peptides/chemical synthesis , Peptides/pharmacology , Protein Isoforms/agonists , Protein Isoforms/metabolism , Receptors, Melanocortin/metabolism , alpha-MSH/chemistry
11.
J Med Chem ; 51(18): 5585-93, 2008 Sep 25.
Article in English | MEDLINE | ID: mdl-18800761

ABSTRACT

The melanocortin-3 and -4 receptors (MC3R, MC4R) have been implicated in energy homeostasis and obesity. Whereas the physiological role of the MC4R is extensively studied, little is known about the MC3R. One caveat is the limited availability of ligands that are selective for the MC3R. Previous studies identified Ac-His-DPhe(p-I)-Arg-Trp-NH 2, which possessed partial agonist/antagonist pharmacology at the mMC3R while retaining full nanomolar agonist pharmacology at the mMC4R. These data allowed for the hypothesis that the DPhe position in melanocortin tetrapeptides can be used to examine ligand side-chain determinants important for differentiation of mMC3R agonist versus antagonist activity. A series of 15 DPhe (7) modified Ac-His-DPhe (7)-Arg-Trp-NH 2 tetrapeptides has been synthesized and pharmacologically characterized. Most notable results include the identification of modifications that resulted in potent antagonists/partial agonists at the mMC3R and full, potent agonists at the mMC4R. These SAR studies provide experimental evidence that the molecular mechanism of antagonism at the mMC3R differentiates this subtype from the mMC4R.


Subject(s)
Melanocortins/chemistry , Receptor, Melanocortin, Type 3/agonists , Animals , Melanocortins/pharmacology , Mice , Receptor, Melanocortin, Type 3/antagonists & inhibitors , Static Electricity , Structure-Activity Relationship
12.
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
13.
Biopolymers ; 90(5): 671-82, 2008.
Article in English | MEDLINE | ID: mdl-18655141

ABSTRACT

Backbone cyclization (BC) and N-methylation have been shown to enhance the activity and/or selectivity of biologically active peptides and improve metabolic stability and intestinal permeability. In this study, we describe the synthesis, structure-activity relationship (SAR) and intestinal metabolic stability of a backbone cyclic peptide library, BL3020, based on the linear alpha-Melanocyte stimulating hormone analog Phe-D-Phe-Arg-Trp-Gly. The drug lead, BL3020-1, selected from the BL3020 library (compound 1) has been shown to inhibit weight gain in mice following oral administration. Another member of the BL3020 library, BL3020-17, showed improved biological activity towards the mMC4R, in comparison to BL3020-1, although neither were selective for MC4R or MC5R. N-methylation, which restrains conformational freedom while increasing metabolic stability beyond that which is imparted by BC, was used to find analogs with increased selectivity. N-methylated backbone cyclic libraries were synthesized based on the BL3020 library. SAR studies showed that all the N-methylated backbone cyclic peptides demonstrated reduced biological activity and selectivity for all the analyzed receptors. N-methylation of active backbone cyclic peptides destabilized the active conformation or stabilized an inactive conformation, rendering the peptides biologically inactive. N-methylation of backbone cyclic peptides maintained stability to degradation by intestinal enzymes.


Subject(s)
Melanocortins/chemistry , Melanocortins/metabolism , Peptide Library , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/metabolism , Animals , Cell Line , Cyclization , Humans , Methylation , Mice , Protein Conformation , Receptors, Melanocortin/biosynthesis , Receptors, Melanocortin/genetics , Structure-Activity Relationship
14.
J Med Chem ; 51(4): 1026-34, 2008 Feb 28.
Article in English | MEDLINE | ID: mdl-18220330

ABSTRACT

The tetrapeptide sequence His-Phe-Arg-Trp, derived from melanocyte-stimulating hormone (alphaMSH) and its analogs, causes a decrease in food intake and elevates energy utilization upon binding to the melanocortin-4 receptor (MC4R). To utilize this sequence as an effective agent for treating obesity, we improved its metabolic stability and intestinal permeability by synthesizing a library of backbone cyclic peptidomimetic derivatives. One analog, peptide 1 (BL3020-1), was selected according to its selectivity in activating the MC4R, its favorable transcellular penetration through enterocytes and its enhanced intestinal metabolic stability. This peptide was detected in the brain following oral administration to rats. A single oral dose of 0.5 mg/kg in mice led to reduced food consumption (up to 48% vs the control group) that lasted for 5 h. Repetitive once daily oral dosing (0.5 mg/kg/day) for 12 days reduced weight gain. Backbone cyclization was shown to produce a potential drug lead for treating obesity.


Subject(s)
Anti-Obesity Agents/chemical synthesis , Peptides, Cyclic/chemical synthesis , Receptor, Melanocortin, Type 4/agonists , Administration, Oral , Animals , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/pharmacology , Biological Availability , Brain/metabolism , Cell Line , Humans , Injections, Intravenous , Intestinal Absorption , Ligands , Magnetic Resonance Spectroscopy , Male , Mice , Molecular Mimicry , Peptides, Cyclic/pharmacokinetics , Peptides, Cyclic/pharmacology , Rats , Rats, Wistar , Structure-Activity Relationship , Tissue Distribution
15.
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
16.
J Neurosci ; 25(6): 1448-58, 2005 Feb 09.
Article in English | MEDLINE | ID: mdl-15703399

ABSTRACT

In adult mammals, learning, memory, and restoration of sensorimotor lost functions imply synaptic reorganization that requires diffusible messengers-mediated communication between presynaptic and postsynaptic structures. A candidate molecule to accomplish this function is the gaseous intercellular messenger nitric oxide (NO), which is involved in synaptogenesis and projection refinement during development; however, the role of NO in synaptic reorganization processes in adulthood remains to be established. In this work, we tested the hypothesis that this free radical is a mediator in the adult mammal CNS synaptic remodeling processes using a model of hypoglossal axonal injury recently developed by us. Axonal injury-induced disconnection of motoneurons from myocytes produces withdrawal of synaptic inputs to motoneurons and concomitant upregulation of the neuronal isoform of NO synthase (NOS-I). After recovery of the neuromuscular function, synaptic coverage is reestablished and NOS-I is downregulated. We also report, by using functional and morphological approaches, that chronic inhibition of the NO/cGMP pathway prevents synaptic withdrawal evoked by axon injury, despite the persistent muscle disconnection. After successful withdrawal of synaptic boutons, inhibition of NO synthesis, but not of cGMP, accelerated the recovery of synaptic coverage, although neuromuscular disconnection was maintained. Furthermore, protein S-nitrosylation was upregulated after nerve injury, and this effect was reversed by NOS-I inhibition. Our results suggest that during synaptic remodeling in the adult CNS, NO acts as a signal for synaptic detachment and inhibits synapse formation by cGMP-dependent and probably S-nitrosylation-mediated mechanisms, respectively. We also suggest a feasible role of NO in neurological disorders coursing with NOS-I upregulation.


Subject(s)
Cyclic GMP/physiology , Hypoglossal Nerve/physiology , Motor Neurons/physiology , Nerve Regeneration/physiology , Nerve Tissue Proteins/physiology , Neuronal Plasticity/physiology , Nitric Oxide Synthase/physiology , Nitric Oxide/physiology , Synapses/physiology , Animals , Enzyme Induction , Hypoglossal Nerve/enzymology , Hypoglossal Nerve Injuries , Male , Motor Neurons/enzymology , Motor Neurons/ultrastructure , NG-Nitroarginine Methyl Ester/pharmacology , Nerve Crush , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Rats , Rats, Wistar , Signal Transduction , Synapses/enzymology
17.
J Physiol ; 557(Pt 3): 991-1011, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15090609

ABSTRACT

The effects of peripheral nerve lesions on the membrane and synaptic properties of motoneurones have been extensively studied. However, minimal information exists about how these alterations finally influence discharge activity and motor output under physiological afferent drive. The aim of this work was to evaluate the effect of hypoglossal (XIIth) nerve crushing on hypoglossal motoneurone (HMN) discharge in response to the basal inspiratory afferent drive and its chemosensory modulation by CO(2). The evolution of the lesion was assessed by recording the compound muscle action potential evoked by XIIth nerve stimulation, which was lost on crushing and then recovered gradually to control values from the second to fourth weeks post-lesion. Basal inspiratory activities recorded 7 days post-injury in the nerve proximal to the lesion site, and in the nucleus, were reduced by 51.6% and 35.8%, respectively. Single unit antidromic latencies were lengthened by lesion, and unusually high stimulation intensities were frequently required to elicit antidromic spikes. Likewise, inspiratory modulation of unitary discharge under conditions in which chemoreceptor drive was varied by altering end-tidal CO(2) was reduced by more than 60%. Although the general recruitment scheme was preserved after XIIth nerve lesion, we noticed an increased proportion of low-threshold units and a reduced recruitment gain across the physiological range. Immunohistochemical staining of synaptophysin in the hypoglossal nuclei revealed significant reductions of this synaptic marker after nerve injury. Morphological and functional alterations recovered with muscle re-innervation. Thus, we report here that nerve lesion induced changes in the basal activity and discharge modulation of HMNs, concurrent with the loss of afferent inputs. Nevertheless, we suggest that an increase in membrane excitability, reported by others, and in the proportion of low-threshold units, could serve to preserve minimal electrical activity, prevent degeneration and favour axonal regeneration.


Subject(s)
Chemoreceptor Cells/physiology , Hypoglossal Nerve Injuries , Motor Neurons/physiology , Respiratory Mechanics/physiology , Action Potentials/drug effects , Animals , Axotomy , Carbon Dioxide/pharmacology , Cell Differentiation , Electromyography , Hypoglossal Nerve/cytology , Immunohistochemistry , Male , Nerve Crush , Neuromuscular Junction/physiology , Rats , Rats, Wistar , Synaptophysin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...