Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 49
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
eNeuro ; 10(8)2023 08.
Article de Anglais | MEDLINE | ID: mdl-37558465

RÉSUMÉ

Traumatic brain injury (TBI) elicits neuronal loss at the site of injury and progressive neuronal loss in the penumbra. However, the consequences of TBI on afferent neurons projecting to the injured tissue from distal locations is unknown. Basal forebrain cholinergic neurons (BFCNs) extend long projections to multiple brain regions including the cortex, regulate many cognitive functions, and are compromised in numerous neurodegenerative disorders. To determine the consequence of cortical injury on these afferent neurons, we used the fluid percussion injury model of traumatic brain injury and assessed the effects on BFCN survival and axon integrity in male and female mice. Survival or death of BF neurons can be regulated by neurotrophins or proneurotrophins, respectively. The injury elicited an induction of proNGF and proBDNF in the cortex and a loss of BFCNs ipsilateral to the injury compared with sham uninjured mice. The p75NTR knock-out mice did not show loss of BFCN neurons, indicating a retrograde degenerative effect of the cortical injury on the afferent BFCNs mediated through p75NTR. In contrast, locus ceruleus neurons, which also project throughout the cortex, were unaffected by the injury, suggesting specificity in retrograde degeneration after cortical TBI. Proneurotrophins (proNTs) provided directly to basal forebrain axons in microfluidic cultures triggered retrograde axonal degeneration and cell death, which did not occur in the absence of p75NTR. This study shows that after traumatic brain injury, proNTs induced in the injured cortex promote BFCN axonal degeneration and retrograde neuron loss through p75NTR.


Sujet(s)
Prosencéphale basal , Lésions traumatiques de l'encéphale , Récepteurs facteur croissance nerf , Animaux , Femelle , Mâle , Souris , Lésions traumatiques de l'encéphale/métabolisme , Neurones cholinergiques/métabolisme , Neurones afférents , Dégénérescence rétrograde/métabolisme , Récepteurs facteur croissance nerf/métabolisme
2.
Acta Neuropathol ; 146(4): 611-629, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37555859

RÉSUMÉ

Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by TDP-43 inclusions in the cortical and spinal motor neurons. It remains unknown whether and how pathogenic TDP-43 spreads across neural connections to progress degenerative processes in the cortico-spinal motor circuitry. Here we established novel mouse ALS models that initially induced mutant TDP-43 inclusions in specific neuronal or cell types in the motor circuits, and investigated whether TDP-43 and relevant pathological processes spread across neuronal or cellular connections. We first developed ALS models that primarily induced TDP-43 inclusions in the corticospinal neurons, spinal motor neurons, or forelimb skeletal muscle, by using adeno-associated virus (AAV) expressing mutant TDP-43. We found that TDP-43 induced in the corticospinal neurons was transported along the axons anterogradely and transferred to the oligodendrocytes along the corticospinal tract (CST), coinciding with mild axon degeneration. In contrast, TDP-43 introduced in the spinal motor neurons did not spread retrogradely to the cortical or spinal neurons; however, it induced an extreme loss of spinal motor neurons and subsequent degeneration of neighboring spinal neurons, suggesting a degenerative propagation in a retrograde manner in the spinal cord. The intraspinal degeneration further led to severe muscle atrophy. Finally, TDP-43 induced in the skeletal muscle did not propagate pathological events to spinal neurons retrogradely. Our data revealed that mutant TDP-43 spread across neuro-glial connections anterogradely in the corticospinal pathway, whereas it exhibited different retrograde degenerative properties in the spinal circuits. This suggests that pathogenic TDP-43 may induce distinct antero- and retrograde mechanisms of degeneration in the motor system in ALS.


Sujet(s)
Sclérose latérale amyotrophique , Dégénérescence rétrograde , Animaux , Souris , Sclérose latérale amyotrophique/anatomopathologie , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Motoneurones/métabolisme , Dégénérescence rétrograde/métabolisme , Dégénérescence rétrograde/anatomopathologie , Moelle spinale/anatomopathologie
3.
Transl Neurodegener ; 10(1): 43, 2021 11 02.
Article de Anglais | MEDLINE | ID: mdl-34727977

RÉSUMÉ

OBJECTIVE: The dopaminergic nigrostriatal neurons (DA cells) in healthy people present a slow degeneration with aging, which produces cellular debris throughout life. About 2%-5% of people present rapid cell degeneration of more than 50% of DA cells, which produces Parkinson's disease (PD). Neuroinflammation accelerates the cell degeneration and may be critical for the transition between the slow physiological and the rapid pathological degeneration of DA cells, particularly when it activates microglial cells of the medial forebrain bundle near dopaminergic axons. As synaptic debris produced by DA cell degeneration may trigger the parkinsonian neuroinflammation, this study investigated the removal of axonal debris produced by retrograde degeneration of DA cells, paying particular attention to the relative roles of astrocytes and microglia. METHODS: Rats and mice were injected in the lateral ventricles with 6-hydroxydopamine, inducing a degeneration of dopaminergic synapses in the striatum which was not accompanied by non-selective tissue damage, microgliosis or neuroinflammation. The possible retrograde degeneration of dopaminergic axons, and the production and metabolization of DA-cell debris were studied with immunohistochemical methods and analyzed in confocal and electron microscopy images. RESULTS: The selective degeneration of dopaminergic synapses in the striatum was followed by a retrograde degeneration of dopaminergic axons whose debris was found within spheroids of the medial forebrain bundle. These spheroids retained mitochondria and most (e.g., tyrosine hydroxylase, the dopamine transporter protein, and amyloid precursor protein) but not all (e.g., α-synuclein) proteins of the degenerating dopaminergic axons. Spheroids showed initial (autophagosomes) but not late (lysosomes) components of autophagy (incomplete autophagy). These spheroids were penetrated by astrocytic processes of the medial forebrain bundle, which provided the lysosomes needed to continue the degradation of dopaminergic debris. Finally, dopaminergic proteins were observed in the cell somata of astrocytes. No microgliosis or microglial phagocytosis of debris was observed in the medial forebrain bundle during the retrograde degeneration of dopaminergic axons. CONCLUSIONS: The present data suggest a physiological role of astrocytic phagocytosis of axonal debris for the medial forebrain bundle astrocytes, which may prevent the activation of microglia and the spread of retrograde axonal degeneration in PD.


Sujet(s)
Neurones dopaminergiques , Maladie de Parkinson , Animaux , Astrocytes/métabolisme , Axones/anatomopathologie , Neurones dopaminergiques/anatomopathologie , Humains , Souris , Maladie de Parkinson/métabolisme , Rats , Rat Sprague-Dawley , Dégénérescence rétrograde/métabolisme , Dégénérescence rétrograde/anatomopathologie
4.
J Pharmacol Sci ; 141(2): 91-96, 2019 Oct.
Article de Anglais | MEDLINE | ID: mdl-31679963

RÉSUMÉ

Neurons extend axons far from cell bodies, and retrograde communications from distal axons to cell bodies and/or dendrites play critical roles in the development and maintenance of neuronal circuits. In neurotrophin signaling, the retrograde axonal transport of endosomes containing active ligand-receptor complexes from distal axons to somatodendrite compartments mediates retrograde signaling. However, the generality and specificity of these endosome-based transportations called "signaling endosomes" remain to be elucidated. Here, I summarize the discovery of semaphorin3A signaling endosomes, the first example other than neurotrophins to regulate dendritic development via AMPA receptor GluA2 localization in dendrites. The molecular components of Sema3A and neurotrophin signaling endosomes are distinct, but partially overlap to regulate specific and common cellular events. Because receptors are transported back to the cell bodies, neurons must replenish receptors on the growth cone surface to ensure continued response to the target-derived ligands. Recent findings have demonstrated that retrograde signaling endosomes also induce anterograde delivery of nascent receptors in neurotrophin signaling. The coupling between anterograde and retrograde axonal transport via signaling endosomes therefore plays a critical role in regulating proper neuronal network formation.


Sujet(s)
Transport axonal/physiologie , Axones/métabolisme , Endosomes/métabolisme , Dégénérescence rétrograde/métabolisme , Animaux , Communication cellulaire , Humains , Facteurs de croissance nerveuse/métabolisme , Transport des protéines/physiologie , Récepteur de l'AMPA/métabolisme , Sémaphorine-3A/métabolisme , Transduction du signal
5.
Dev Neurobiol ; 78(10): 1011-1024, 2018 10.
Article de Anglais | MEDLINE | ID: mdl-30027624

RÉSUMÉ

After axonal injury, chromatolysis (fragmentation of Nissl substance) can occur in the soma. Electron microscopy shows that chromatolysis involves fission of the rough endoplasmic reticulum. In CNS neurons (which do not regenerate axons back to their original targets) or in motor neurons or dorsal root ganglion neurons denied axon regeneration (e.g., by transection and ligation), chromatolysis is often accompanied by degranulation (loss of ribosomes from rough endoplasmic reticulum), disaggregation of polyribosomes and degradation of monoribosomes into dust-like particles. Ribosomes and rough endoplasmic reticulum may also be degraded in autophagic vacuoles by ribophagy and reticulophagy, respectively. In other words, chromatolysis is disruption of parts of the protein synthesis infrastructure. Whereas some neurons may show transient or no chromatolysis, severely injured neurons can remain chromatolytic and never again synthesize normal levels of protein; some may atrophy or die. Ribonuclease(s) might cause the following features of chromatolysis: fragmentation and degranulation of rough endoplasmic reticulum, disaggregation of polyribosomes and degradation of monoribosomes. For example, ribonucleases in the EndoU/PP11 family can modify rough endoplasmic reticulum; many ribonucleases can degrade mRNA causing polyribosomes to unchain and disperse, and they can disassemble monoribosomes; Ribonuclease 5 can control rRNA synthesis and degrade tRNA; Ribonuclease T2 can degrade ribosomes, endoplasmic reticulum and RNA within autophagic vacuoles; and Ribonuclease IRE1α acts as a stress sensor within the endoplasmic reticulum. Regeneration might be improved after axonal injury by protecting the protein synthesis machinery from catabolism; targeting ribonucleases using inhibitors can enhance neurite outgrowth and could be a profitable strategy in vivo. © 2018 Wiley Periodicals, Inc. Develop Neurobiol, 2018.


Sujet(s)
Axones/métabolisme , Axones/anatomopathologie , Réticulum endoplasmique rugueux/métabolisme , Régénération nerveuse/physiologie , ARN/métabolisme , Dégénérescence rétrograde/métabolisme , Ribonucléases/métabolisme , Ribosomes/métabolisme , Traumatismes du système nerveux/métabolisme , Animaux , Humains
6.
Sci Rep ; 5: 9185, 2015 Mar 18.
Article de Anglais | MEDLINE | ID: mdl-25784190

RÉSUMÉ

Neurodegenerative processes are preceded by neuronal dysfunction and synaptic disconnection. Disconnection between spinal motoneuron (MN) soma and synaptic target leads either to a retrograde degenerative process or to a regenerative reaction, depending injury proximity among other factors. Distinguished key events associated with one or other processes may give some clues towards new therapeutical approaches based on boosting endogenous neuroprotective mechanisms. Root mechanical traction leads to retrograde MN degeneration, but share common initial molecular mechanisms with a regenerative process triggered by distal axotomy and suture. By 7 days post-injury, key molecular events starts to diverge and sign apart each destiny. We used comparative unbiased proteomics to define these signatures, coupled to a novel network-based analysis to get biological meaning. The procedure implicated the previous generation of combined topological information from manual curated 19 associated biological processes to be contrasted with the proteomic list using gene enrichment analysis tools. The novel and unexpected results suggested that motoneurodegeneration is better explained mainly by the concomitant triggering of anoikis, anti-apoptotic and neuropathic-pain related programs. In contrast, the endogenous neuroprotective mechanisms engaged after distal axotomy included specifically rather anti-anoikis and selective autophagy. Validated protein-nodes and processes are highlighted across discussion.


Sujet(s)
Axones/physiologie , Protéomique , Animaux , Anoïkis , Axotomie , Mâle , Motoneurones/métabolisme , Maladies neurodégénératives/métabolisme , Maladies neurodégénératives/anatomopathologie , Douleur/métabolisme , Douleur/anatomopathologie , Cartes d'interactions protéiques , Rats , Rat Sprague-Dawley , Dégénérescence rétrograde/métabolisme , Dégénérescence rétrograde/anatomopathologie
7.
Mol Neurobiol ; 49(3): 1327-37, 2014 Jun.
Article de Anglais | MEDLINE | ID: mdl-24390474

RÉSUMÉ

Atrophy of upper motor neurons hampers axonal regeneration and functional recovery following spinal cord injury (SCI). Apart from the severity of primary injury, a series of secondary pathological damages including spinal cord edema and glial scar formation affect the fate of injured upper motor neurons. The aquaporin-4 (AQP4) water channel plays a critical role in water homeostasis and migration of astrocytes in the central nervous system, probably offering a new therapeutic target for protecting against upper motor neuron degeneration after SCI. To test this hypothesis, we examined the effect of AQP4 deficiency on atrophy of rubrospinal neurons after unilateral rubrospinal tract transection at the fourth cervical level in mice. AQP4 gene knockout (AQP4-/-) mice exhibited high extent of spinal cord edema at 72 h after lesion compared with wild-type littermates. AQP4-/- mice showed impairments in astrocyte migration toward the transected site with a greater lesion volume at 1 week after surgery and glial scar formation with a larger cyst volume at 6 weeks. More severe atrophy and loss of axotomized rubrospinal neurons as well as axonal degeneration in the rubrospinal tract rostral to the lesion were observed in AQP4-/- mice at 6 weeks after SCI. AQP4 expression was downregulated at the lesioned spinal segment at 3 days and 1 week after injury, but upregulated at 6 weeks. These results demonstrated that AQP4 not only mitigates spinal cord damage but also ameliorates retrograde degeneration of rubrospinal neurons by promoting edema clearance and glial scar formation after laceration SCI. This finding supports the notion that AQP4 may be a promising therapeutic target for SCI.


Sujet(s)
Aquaporine-4/déficit , Cicatrice/métabolisme , Oedème/métabolisme , Névroglie/métabolisme , Dégénérescence rétrograde/métabolisme , Traumatismes de la moelle épinière/métabolisme , Animaux , Vertèbres cervicales/métabolisme , Vertèbres cervicales/anatomopathologie , Cicatrice/anatomopathologie , Oedème/anatomopathologie , Femelle , Souris , Souris knockout , Névroglie/anatomopathologie , Neurones/métabolisme , Neurones/anatomopathologie , Noyau rouge/métabolisme , Noyau rouge/anatomopathologie , Dégénérescence rétrograde/anatomopathologie , Traumatismes de la moelle épinière/anatomopathologie
8.
Clin Neuropathol ; 33(1): 68-75, 2014.
Article de Anglais | MEDLINE | ID: mdl-24216149

RÉSUMÉ

OBJECTIVE: Pathogenesis of pseudohypertrophy of the inferior olivary nucleus (PH-IO) was analyzed based on immunohistochemical study. METHODS: Immunostained medullas with PH-IO were observed with confocal laser microscopy. RESULTS: αB-crystallin (αBC) was frequently expressed in the neurons and co-localized with microtubule-associated protein 2 (MAP2). The neurons were occasionally positive for SMI-31. αBC and SMI-31 were co-localized in some neurons. Synaptophysin (SYP)-immunoreactive dots were present around MAP2-positive hypertrophic neurons and hypertrophic thick neurites. Periphery-stained Lys-Asp-Glu-Leu (KDEL)-positive neurons were shown. Central chromatolytic neurons were found with Klüver-Barrera staining, which indicated that the rough endoplasmic reticulum (ER) was distributed to the periphery of the cytoplasm. CONCLUSIONS: αBC prevents microtubule disassembly and phosphorylation of the neurofilaments under stressful conditions. Our results indicated that αBC protected microtubules and neurofilaments in PH-IO. The retrograde transport of KDEL receptors from the Golgi complex to the ER is increased under stressful conditions. We considered that KDEL receptors were retro-transported to ER, and then the ER containing KDEL receptors was distributed to the periphery of the cytoplasm. PH-IO showed various immunohistochemical changes due to trans-synaptic degeneration.


Sujet(s)
Noyau olivaire/métabolisme , Noyau olivaire/anatomopathologie , Réticulum endoplasmique/physiologie , Appareil de Golgi/physiologie , Humains , Hypertrophie/étiologie , Hypertrophie/métabolisme , Hypertrophie/anatomopathologie , Microscopie confocale , Récepteurs peptidiques/métabolisme , Dégénérescence rétrograde/complications , Dégénérescence rétrograde/métabolisme , Dégénérescence rétrograde/anatomopathologie , Chaîne B de la cristalline alpha/métabolisme
9.
Neurosci Lett ; 519(1): 56-61, 2012 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-22583768

RÉSUMÉ

Suppression of inhibition of axonal outgrowth and promotion of axonal protection from progressive axonal degeneration are both therapeutic strategies for the treatment of neuronal diseases characterized by axonal loss. Myelin-associated inhibitors (MAIs) have been shown to suppress axonal outgrowth, but a specific MAI, myelin-associated glycoprotein (MAG), has also been shown to protect neurons from axonal degeneration through activation of the small GTPase protein RhoA. Recent in vitro studies have shown that collapsin response mediator protein 4 (CRMP4) interacts with RhoA and that the CRMP4b/RhoA complex mediates MAG-induced inhibitory signaling against axonal outgrowth. However, whether CRMP4 is involved in MAG-mediated axon protection signaling remains unclear. Here, we show involvement of CRMP4 in MAG-induced inhibition of axonal outgrowth and axonal protection using the CRMP4-/- mouse model. In dorsal root ganglion (DRG) neurons, loss of CRMP4 prevents MAG-induced inhibition of axonal outgrowth and growth cone collapse and increases sensitivity to microtubule destabilizing factor Vincristine (VNC)-induced axonal degeneration. MAG-mediated axon protection against VNC is suppressed in CRMP4-/- DRG neurons. Understanding the molecular mechanism of MAG-mediated inhibition and protection via CRMP4 may provide novel opportunities to control axonal degeneration and regeneration.


Sujet(s)
Amidohydrolases/métabolisme , Axones/métabolisme , Glycoprotéine associée à la myéline/métabolisme , Dégénérescence rétrograde/induit chimiquement , Dégénérescence rétrograde/métabolisme , Vincristine , Amidohydrolases/génétique , Animaux , Axones/effets des médicaments et des substances chimiques , Hydrolases , Souris , Souris knockout , Protéines associées aux microtubules , Inhibition nerveuse , Transduction du signal , Modulateurs de la polymérisation de la tubuline
10.
Cell Death Differ ; 18(10): 1617-27, 2011 Oct.
Article de Anglais | MEDLINE | ID: mdl-21436843

RÉSUMÉ

Disconnection of the axon from the soma of spinal motoneurons (MNs) leads either to a retrograde degenerative process or to a regenerative reaction, depending on the severity and the proximity to the soma of the axonal lesion. The endoplasmic reticulum (ER) is a continuous membranous network that extends from the nucleus to the entire cytoplasm of the neuronal soma, axon and dendrites. We investigated whether axonal injury is sensed by the ER and triggers the activation of protective mechanisms, such as the unfolded protein response (UPR) and autophagy. We found early (at 3 days) accumulation of beclin1, LC3II and Lamp-1, hallmarks of autophagy, in both degenerating MNs after spinal root avulsion and in non-degenerating MNs after distal nerve section, although Lamp-1 disappeared by 5 days only in the former. In contrast, only degenerating MNs presented early activation of IRE1α, revealed by an increase of the spliced isoform of Xbp1 and accumulation of ATF4 in their nucleus, two branches of the UPR, and late BiP downregulation in association with cytoskeletal and organelle disorganization. We conclude that BiP decrease is a signature of the degenerating process, as its overexpression led to an increase in MN survival after root avulsion. Besides, Bcl2 is strongly implicated in the survival pathway activated by BiP overexpression.


Sujet(s)
Autophagie/physiologie , Protéines du choc thermique/métabolisme , Motoneurones/métabolisme , Dégénérescence rétrograde/métabolisme , Facteur de transcription ATF-4/métabolisme , Animaux , Protéines régulatrices de l'apoptose/métabolisme , Autophagie/génétique , Bécline-1 , Technique de Western , Protéines de liaison à l'ADN/métabolisme , Réticulum endoplasmique/métabolisme , Femelle , Protéines du choc thermique/génétique , Immunohistochimie , Protéines lysosomales membranaires/métabolisme , Rats , Rat Sprague-Dawley , Facteurs de transcription des facteurs régulateurs X , Racines des nerfs spinaux/métabolisme , Racines des nerfs spinaux/anatomopathologie , Facteurs de transcription/métabolisme , Protéine-1 liant la boite X
11.
J Neurosci ; 31(6): 2125-35, 2011 Feb 09.
Article de Anglais | MEDLINE | ID: mdl-21307249

RÉSUMÉ

Axon degeneration is a hallmark of neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Such degeneration is not a passive event but rather an active process mediated by mechanisms that are distinct from the canonical pathways of programmed cell death that mediate destruction of the cell soma. Little is known of the diverse mechanisms involved, particularly those of retrograde axon degeneration. We have previously observed in living animal models of degeneration in the nigrostriatal projection that a constitutively active form of the kinase, myristoylated Akt (Myr-Akt), demonstrates an ability to suppress programmed cell death and preserve the soma of dopamine neurons. Here, we show in both neurotoxin and physical injury (axotomy) models that Myr-Akt is also able to preserve dopaminergic axons due to suppression of acute retrograde axon degeneration. This cellular phenotype is associated with increased mammalian target of rapamycin (mTor) activity and can be recapitulated by a constitutively active form of the small GTPase Rheb, an upstream activator of mTor. Axon degeneration in these models is accompanied by the occurrence of macroautophagy, which is suppressed by Myr-Akt. Conditional deletion of the essential autophagy mediator Atg7 in adult mice also achieves striking axon protection in these acute models of retrograde degeneration. The protection afforded by both Myr-Akt and Atg7 deletion is robust and lasting, because it is still observed as protection of both axons and dopaminergic striatal innervation weeks after injury. We conclude that acute retrograde axon degeneration is regulated by Akt/Rheb/mTor signaling pathways.


Sujet(s)
Autophagie/physiologie , Axones/métabolisme , Dopamine/métabolisme , Neurones/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Dégénérescence rétrograde/métabolisme , Dégénérescence rétrograde/anatomopathologie , Animaux , Autophagie/effets des médicaments et des substances chimiques , Protéine-7 associée à l'autophagie , Axones/effets des médicaments et des substances chimiques , Axones/ultrastructure , Dependovirus/génétique , Modèles animaux de maladie humaine , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/physiologie , Protéines à fluorescence verte/génétique , Faisceau télencéphalique médial/anatomopathologie , Souris , Souris de lignée C57BL , Microscopie confocale , Microscopie électronique à transmission/méthodes , Protéines associées aux microtubules/métabolisme , Oxidopamine/effets indésirables , Protéines proto-oncogènes c-akt/génétique , Dégénérescence rétrograde/étiologie , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/génétique , Substantia nigra/anatomopathologie , Sérine-thréonine kinases TOR/métabolisme , Tyrosine 3-monooxygenase/métabolisme
12.
FASEB J ; 25(5): 1461-73, 2011 May.
Article de Anglais | MEDLINE | ID: mdl-21248239

RÉSUMÉ

Even though VEGF-B is a homologue of the potent angiogenic factor VEGF, its angiogenic activities have been controversial. Intrigued by findings that VEGF-B may also affect neuronal cells, we assessed the neuroprotective and vasculoprotective effects of VEGF-B in the skin, in which vessels and nerves are functionally intertwined. Although VEGF-B and its FLT1 receptor were prominently expressed in dorsal root ganglion (DRG) neurons innervating the hindlimb skin, they were not essential for nerve function or vascularization of the skin. However, primary DRG cultures lacking VEGF-B or FLT1 exhibited increased neuronal stress and were more susceptible to paclitaxel-induced cell death. Concomitantly, mice lacking VEGF-B or a functional FLT1 developed more retrograde degeneration of sensory neurons in a model of distal neuropathy. On the other hand, the addition of the VEGF-B isoform, VEGF-B(186), to DRG cultures antagonized neuronal stress, maintained the mitochondrial membrane potential and stimulated neuronal survival. Mice overexpressing VEGF-B(186) or FLT1 selectively in neurons were protected against the distal neuropathy, whereas exogenous VEGF-B(186), either delivered by gene transfer or as a recombinant factor, was protective by directly affecting sensory neurons and not the surrounding vasculature. Overall, this indicates that VEGF-B, instead of acting as an angiogenic factor, exerts direct neuroprotective effects through FLT1. These findings also suggest a clinically relevant role for VEGF-B in preventing distal neuropathies.


Sujet(s)
Dégénérescence rétrograde/métabolisme , Cellules réceptrices sensorielles/métabolisme , Cellules réceptrices sensorielles/anatomopathologie , Facteur de croissance endothéliale vasculaire de type B/métabolisme , Récepteur-1 au facteur croissance endothéliale vasculaire/métabolisme , Animaux , Immunohistochimie , Potentiel de membrane mitochondriale/génétique , Potentiel de membrane mitochondriale/physiologie , Souris , Souris de lignée C57BL , Souris knockout , Polyneuropathies/génétique , Polyneuropathies/métabolisme , Rats , Rat Sprague-Dawley , Dégénérescence rétrograde/génétique , RT-PCR , Facteur de croissance endothéliale vasculaire de type B/génétique , Récepteur-1 au facteur croissance endothéliale vasculaire/génétique
13.
Exp Neurol ; 225(1): 94-103, 2010 Sep.
Article de Anglais | MEDLINE | ID: mdl-20570589

RÉSUMÉ

The mechanism underlying axotomy-induced motoneuron loss is not fully understood, but appears to involve molecular changes within the injured motoneuron and the surrounding local microenvironment (neuropil). The mouse facial nucleus consists of six subnuclei which respond differentially to facial nerve transection at the stylomastoid foramen. The ventromedial (VM) subnucleus maintains virtually full facial motoneuron (FMN) survival following axotomy, whereas the ventrolateral (VL) subnucleus results in significant FMN loss with the same nerve injury. We hypothesized that distinct molecular phenotypes of FMN existed within the two subregions, one responsible for maintaining cell survival and the other promoting cell death. In this study, we used laser microdissection to isolate VM and VL facial subnuclear regions for molecular characterization. We discovered that, regardless of neuronal fate after injury, FMN in either subnuclear region respond vigorously to injury with a characteristic "regenerative" profile and additionally, the surviving VL FMN appear to compensate for the significant FMN loss. In contrast, significant differences in the expression of pro-inflammatory cytokine mRNA in the surrounding neuropil response were found between the two subnuclear regions of the facial nucleus that support a causative role for glial and/or immune-derived molecules in directing the contrasting responses of the FMN to axonal transection.


Sujet(s)
Nerf facial/anatomopathologie , Nerf facial/physiopathologie , Thérapie laser/méthodes , Microdissection/méthodes , Motoneurones/métabolisme , Neuropile/métabolisme , Phénotype , Animaux , Axotomie/instrumentation , Axotomie/méthodes , Modèles animaux de maladie humaine , Nerf facial/métabolisme , Femelle , Souris , Souris de lignée C57BL , Microdissection/instrumentation , Motoneurones/anatomopathologie , Neuropile/anatomopathologie , Cortex préfrontal/métabolisme , Cortex préfrontal/anatomopathologie , Cortex préfrontal/physiopathologie , Dégénérescence rétrograde/métabolisme , Dégénérescence rétrograde/anatomopathologie , Dégénérescence rétrograde/physiopathologie , Noyaux ventraux du thalamus/métabolisme , Noyaux ventraux du thalamus/anatomopathologie , Noyaux ventraux du thalamus/physiopathologie
14.
J Neurosci ; 28(46): 11980-8, 2008 Nov 12.
Article de Anglais | MEDLINE | ID: mdl-19005063

RÉSUMÉ

G-protein-coupled receptors (GPCRs) form the largest superfamily of membrane proteins, and several GPCRs have been implicated in signaling between neurons and glia to protect neurons from pathological stresses. Here, we have used a screening strategy to investigate GPCRs that are involved in neuronal protection. The real-time PCR was performed using 274 primers targeting nonsensory GPCR mRNAs, which were listed on the database. The cDNAs from control and nerve-injured hypoglossal nuclei of mouse brain were used, and the alterations of PCR products were compared. This screen and the subsequent in situ hybridization screen exhibited six GPCR mRNAs which were prominently and convincingly induced in nerve-injured hypoglossal nuclei. Among these candidates, the chemokine receptor CCR5 was selected, based on the marked induction in CCR5 mRNA in microglia after nerve injury. The mRNA expression of ligands for CCR5, such as regulated on activation normal T-cell expressed and secreted (RANTES/CCL5), MIP-1alpha, and MIP-1beta, were induced in injured motor neurons, indicating that CCR5 and its ligands were expressed in microglia and neurons, respectively, in response to nerve injury. In vitro, lipopolysaccharide (LPS)-induced expression of mRNAs for inflammatory cytokines (IL-1beta, IL-6, and tumor necrosis factor-alpha) and inducible nitric oxide synthase (iNOS) in microglia were all suppressed by RANTES. Those suppressions were not observed in microglia from CCR5 null mice. In addition, nerve injury-induced motor neuron death seen in wild type C56BL/6J mice was accelerated in CCR5 knock-out C57BL/6J. These results may suggest that CCR5-mediated neuron-glia signaling functions to protect neurons by suppressing microglia toxicity.


Sujet(s)
Microglie/métabolisme , Neurotoxines/métabolisme , Récepteurs CCR5/métabolisme , Récepteurs couplés aux protéines G/génétique , Dégénérescence rétrograde/métabolisme , Animaux , Communication cellulaire/génétique , Communication cellulaire/immunologie , Mort cellulaire/génétique , Mort cellulaire/immunologie , Cellules cultivées , Chimiokine CCL3/génétique , Chimiokine CCL3/immunologie , Chimiokine CCL3/métabolisme , Chimiokine CCL4/génétique , Chimiokine CCL4/immunologie , Chimiokine CCL4/métabolisme , Chimiokine CCL5/génétique , Chimiokine CCL5/métabolisme , Chimiokine CCL5/pharmacologie , Cytokines/immunologie , Cytokines/métabolisme , Dépistage génétique , Lésions du nerf hypoglosse , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Microglie/immunologie , Motoneurones/immunologie , Motoneurones/métabolisme , Motoneurones/anatomopathologie , Nitric oxide synthase type II/immunologie , Nitric oxide synthase type II/métabolisme , ARN messager/métabolisme , Récepteurs CCR5/génétique , Récepteurs CCR5/immunologie , Dégénérescence rétrograde/immunologie , Dégénérescence rétrograde/physiopathologie , Transduction du signal/génétique , Transduction du signal/immunologie
15.
Neurosci Lett ; 417(3): 255-60, 2007 May 07.
Article de Anglais | MEDLINE | ID: mdl-17382469

RÉSUMÉ

We investigate whether Nogo-A is involved in the secondary axonal degeneration in the thalamus after distal middle cerebral artery occlusion (MCAO) in stroke-prone renovascular hypertensive rats (RHRSP). The expression of Nogo-A in ipsilateral ventroposterior nucleus (VPN) of the thalamus in RHRSP was observed at 1, 2 and 4 weeks after distal MCAO. In addition, intracerebroventricular infusion of NEP1-40, a Nogo-66 receptor (NgR) antagonist peptide, was administered starting 24 h after MCAO and continued for 1, 2 and 4 weeks, respectively. Axonal damage and regeneration were evaluated by analysis of the immunoreactivity (IR) of amyloid betaA4 precursor protein (APP), growth associated protein 43 (GAP-43) and microtubule associated protein 2 (MAP-2) in ipsilateral VPN of the thalamus at 1, 2 and 4 weeks after distal MCAO. Following ischemia, the expression of Nogo-A in oligodendrocytes increased persistently and its localization became redistributed around damaged axons and dendrites. Administration of NEP1-40 downregulated the expression of Nogo-A, reduced axonal injury and enhanced axonal regeneration. Our data suggest that Nogo-A is involved in secondary axonal degeneration and that inhibition of Nogo-A can reduce neuronal damage in the thalamus after distal MCAO.


Sujet(s)
Infarctus cérébral/métabolisme , Hypertension artérielle/complications , Protéines de la myéline/métabolisme , Dégénérescence rétrograde/métabolisme , Thalamus/métabolisme , Dégénérescence wallerienne/métabolisme , Animaux , Axones/métabolisme , Axones/anatomopathologie , Marqueurs biologiques/métabolisme , Infarctus cérébral/anatomopathologie , Infarctus cérébral/physiopathologie , Hypertension artérielle/physiopathologie , Immunohistochimie , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne/anatomopathologie , Infarctus du territoire de l'artère cérébrale moyenne/physiopathologie , Mâle , Protéines de la myéline/pharmacologie , Protéines de la myéline/usage thérapeutique , Facteurs de croissance nerveuse/pharmacologie , Facteurs de croissance nerveuse/usage thérapeutique , Régénération nerveuse/effets des médicaments et des substances chimiques , Régénération nerveuse/physiologie , Protéines de tissu nerveux/métabolisme , Protéines Nogo , Oligodendroglie/métabolisme , Fragments peptidiques/pharmacologie , Fragments peptidiques/usage thérapeutique , Rats , Rat Sprague-Dawley , Dégénérescence rétrograde/anatomopathologie , Dégénérescence rétrograde/physiopathologie , Thalamus/anatomopathologie , Thalamus/physiopathologie , Régulation positive/physiologie , Noyaux ventraux du thalamus/métabolisme , Noyaux ventraux du thalamus/anatomopathologie , Noyaux ventraux du thalamus/physiopathologie , Dégénérescence wallerienne/anatomopathologie , Dégénérescence wallerienne/physiopathologie
16.
Exp Neurol ; 203(1): 185-95, 2007 Jan.
Article de Anglais | MEDLINE | ID: mdl-16970940

RÉSUMÉ

Antegrade, target-directed axonal regeneration is the explicit goal of nerve repair. However, aberrant and dysfunctional regrowth is commonly observed as well. At the site of surgical nerve coaptation, axonal sprouts encounter fibrotic connective tissue rich in growth-inhibiting chondroitin sulfate proteoglycan that may contribute to misdirection of axonal regrowth. In the present study, we tested the hypothesis that degradation of chondroitin sulfate proteoglycan by application of chondroitinase at the site of nerve repair can decrease aberrant axonal growth. Adult rats received bilateral sciatic nerve transection and end-to-end repair. One nerve was injected with chondroitinase ABC and the contralateral nerve treated with vehicle alone. After 28 weeks, retrograde axonal regeneration was assessed proximal to the repair by scoring neurofilament-immunopositive axons within the nerve (intrafascicular) and outside the nerve proper (extrafascicular). Intrafascicular retrograde axonal growth was equivalent in both control and chondroitinase treatment conditions. In contrast, chondroitinase treatment caused a pronounced (93%) reduction in extrafascicular retrograde axonal growth. The decrease in axon egress from the nerve was coincident with an increase in antegrade regeneration and improved recovery of motor function. Based on these findings, we conclude that chondroitinase applied at the site of nerve transection repair averts dysfunctional extrafascicular retrograde axonal growth.


Sujet(s)
Chondroitinases et chondroitin lyases/métabolisme , Cônes de croissance/métabolisme , Inhibiteurs de croissance/métabolisme , Régénération nerveuse/physiologie , Lésions des nerfs périphériques , Nerfs périphériques/métabolisme , Animaux , Protéoglycanes à chondroïtine sulfate/antagonistes et inhibiteurs , Protéoglycanes à chondroïtine sulfate/métabolisme , Chondroitinases et chondroitin lyases/pharmacologie , Chondroitinases et chondroitin lyases/usage thérapeutique , Modèles animaux de maladie humaine , Femelle , Cônes de croissance/effets des médicaments et des substances chimiques , Cônes de croissance/ultrastructure , Inhibiteurs de croissance/pharmacologie , Inhibiteurs de croissance/usage thérapeutique , Motoneurones/cytologie , Motoneurones/effets des médicaments et des substances chimiques , Motoneurones/métabolisme , Régénération nerveuse/effets des médicaments et des substances chimiques , Neurones afférents/cytologie , Neurones afférents/effets des médicaments et des substances chimiques , Neurones afférents/métabolisme , Nerfs périphériques/physiopathologie , Rats , Rat Sprague-Dawley , Dégénérescence rétrograde/traitement médicamenteux , Dégénérescence rétrograde/métabolisme , Dégénérescence rétrograde/physiopathologie , Neuropathie du nerf sciatique/traitement médicamenteux , Neuropathie du nerf sciatique/métabolisme , Neuropathie du nerf sciatique/physiopathologie
17.
Brain Res ; 1132(1): 36-41, 2007 Feb 09.
Article de Anglais | MEDLINE | ID: mdl-17166487

RÉSUMÉ

We examined changes in the expression of Smad family members, which transduce signals from TGF-beta superfamily ligands, following hypoglossal nerve injury. RT-PCR and in situ hybridization revealed that Smad1, 2, 3 and 4 mRNAs were significantly up-regulated in injured side, whereas Smad8 mRNA was down-regulated. Immunohistochemistry and Western blotting analysis confirmed the alterations of Smad1, 2 and 4 in injured neurons. These results suggest that the Smad signaling may be important for nerve regeneration.


Sujet(s)
Atteintes du nerf hypoglosse/métabolisme , Lésions du nerf hypoglosse , Nerf hypoglosse/métabolisme , Motoneurones/métabolisme , Régénération nerveuse , Protéines Smad/métabolisme , Animaux , Axotomie , Régulation négative/génétique , Régulation de l'expression des gènes/génétique , Nerf hypoglosse/physiopathologie , Atteintes du nerf hypoglosse/physiopathologie , Immunohistochimie , Mâle , Rats , Rat Wistar , Dégénérescence rétrograde/génétique , Dégénérescence rétrograde/métabolisme , Dégénérescence rétrograde/physiopathologie , Protéines Smad/génétique , Protéine Smad-1/génétique , Protéine Smad-1/métabolisme , Protéine Smad-3/génétique , Protéine Smad-3/métabolisme , Protéine Smad-4/génétique , Protéine Smad-4/métabolisme , Protéine Smad-8/génétique , Protéine Smad-8/métabolisme , Régulation positive/génétique
18.
J Comp Neurol ; 498(2): 252-65, 2006 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-16856127

RÉSUMÉ

Mutations in the major peripheral nervous system (PNS) myelin protein, myelin protein zero (MPZ), cause Charcot-Marie-Tooth Disease type 1B (CMT1B), typically thought of as a demyelinating peripheral neuropathy. Certain MPZ mutations, however, cause adult onset neuropathy with minimal demyelination but pronounced axonal degeneration. Mechanism(s) for this phenotype are unknown. We performed an autopsy of a 73-year-old woman with a late-onset neuropathy caused by an H10P MPZ mutation whose nerve conduction studies suggested severe axonal loss but no demyelination. The autopsy demonstrated axonal loss and reorganization of the molecular architecture of the axolemma. Segmental demyelination was negligible. In addition, we identified focal nerve enlargements containing MPZ and ubiquitin either in the inner myelin intralaminar and/or periaxonal space that separates axons from myelinating Schwann cells. Taken together, these data confirmed that a mutation in MPZ can cause axonal neuropathy, in the absence of segmental demyelination, thus uncoupling the two pathological processes. More important, it also provided potential molecular mechanisms as to how the axonal degeneration occurred: either by disruption of glial-axon interaction by protein aggregates or by alterations in the molecular architecture of internodes and paranodes. This report represents the first study in which the molecular basis of axonal degeneration in the late-onset CMT1B has been explored in human tissue.


Sujet(s)
Mutation , Protéine P0 de la myéline/génétique , Dégénérescence rétrograde/génétique , Adulte , Sujet âgé , Animaux , Autopsie , Marqueurs biologiques/métabolisme , Maladie de Charcot-Marie-Tooth/génétique , Maladie de Charcot-Marie-Tooth/anatomopathologie , Maladie de Charcot-Marie-Tooth/physiopathologie , Femelle , Humains , Protéine P0 de la myéline/métabolisme , Gaine de myéline/métabolisme , Gaine de myéline/anatomopathologie , Gaine de myéline/ultrastructure , Neurofibres myélinisées/métabolisme , Neurofibres myélinisées/anatomopathologie , Neurofibres myélinisées/ultrastructure , Nerfs périphériques/métabolisme , Nerfs périphériques/anatomopathologie , Nerfs périphériques/ultrastructure , Dégénérescence rétrograde/métabolisme , Dégénérescence rétrograde/anatomopathologie
19.
J Neurosci ; 26(12): 3281-91, 2006 Mar 22.
Article de Anglais | MEDLINE | ID: mdl-16554478

RÉSUMÉ

To address the hypothesis that retinoids produced by synthesizing enzymes present in the primary olfactory system influence the mouse olfactory sensory map, we expressed a dominant-negative retinoic acid receptor selectively in olfactory sensory neurons. We show that neurons deficient in nuclear retinoid signaling are responsive to odors and form correct odorant receptor-specific axonal projections to target neurons in the olfactory bulb of the brain. Subsequent to the formation of the map, the neurons die prematurely by retrograde-driven caspase-3 activation, which resembles the previously described mechanism of neural death after olfactory bulb ablation. This neurodegenerative event is initiated the second postnatal week and occurs in the adult animal without a compensatory increase of progenitor cell proliferation. In addition, we find that nuclear retinoid signaling is required for the expression of a retinoic acid-degrading enzyme, Cyp26B1, in a small fraction of mature neurons. Collectively, the results provide evidence for a role of locally regulated retinoid metabolism in neuroprotection and in determining population size of neurons at a late stage of neural circuit formation.


Sujet(s)
Apoptose/génétique , Bulbe olfactif/croissance et développement , Muqueuse olfactive/croissance et développement , Voies olfactives/croissance et développement , Neurorécepteurs olfactifs/métabolisme , Récepteurs à l'acide rétinoïque/génétique , Vieillissement/physiologie , Animaux , Animaux nouveau-nés , Caspase-3 , Caspases/métabolisme , Différenciation cellulaire/physiologie , Survie cellulaire/physiologie , Cytochrome P-450 enzyme system/métabolisme , Femelle , Cônes de croissance/métabolisme , Cônes de croissance/ultrastructure , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Bulbe olfactif/cytologie , Bulbe olfactif/métabolisme , Muqueuse olfactive/cytologie , Muqueuse olfactive/métabolisme , Nerf olfactif/cytologie , Nerf olfactif/croissance et développement , Nerf olfactif/métabolisme , Voies olfactives/cytologie , Voies olfactives/métabolisme , Neurorécepteurs olfactifs/cytologie , Retinoic acid 4-hydroxylase , Dégénérescence rétrograde/génétique , Dégénérescence rétrograde/métabolisme , Dégénérescence rétrograde/physiopathologie , Transduction du signal/physiologie , Odorat/génétique , Trétinoïne/métabolisme
20.
Hand Surg ; 10(1): 7-15, 2005 Jul.
Article de Anglais | MEDLINE | ID: mdl-16106495

RÉSUMÉ

The purpose of this study is to clarify the reactions of the neuro-muscular junction and nerve cell body to gradual nerve elongation. The sciatic nerves of Japanese white rabbits were lengthened by 30 mm in increments of 0.8 mm/day, 2.0 mm/day and 4.0 mm/day. A scanning electron microscopic examination showed no degenerative change at the neuro-muscular junction, even eight weeks after elongation in the 4-mm group. Hence, neuro-muscular junction is not critical for predicting damage from gradual nerve elongation. There were no axon reaction cells in the 0.8-mm group, a small amount in the 2-mm group, and a large amount in the 4-mm group. The rate of growth associated protein-43 positive nerve cells was significant in the 4-mm group. Hence, the safe speed for nerve cells appeared to be 0.8-mm/day, critical speed to be 2.0-mm/day, and dangerous speed to be 4.0-mm/day in this elongation model.


Sujet(s)
Jonction neuromusculaire/physiologie , Neurones/métabolisme , Procédures de neurochirurgie/méthodes , Nerf ischiatique/chirurgie , Animaux , Protéine GAP-43/métabolisme , Microscopie électronique à balayage , Modèles animaux , Muscles squelettiques/anatomopathologie , Jonction neuromusculaire/anatomopathologie , Neurones/anatomopathologie , Taille d'organe , Lapins , Dégénérescence rétrograde/métabolisme , Nerf ischiatique/métabolisme , Nerf ischiatique/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...