Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Pain ; 165(6): 1336-1347, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38739766

ABSTRACT

ABSTRACT: Evidence from previous studies supports the concept that spinal cord injury (SCI)-induced neuropathic pain (NP) has its neural roots in the peripheral nervous system. There is uncertainty about how and to which degree mechanoreceptors contribute. Sensorimotor activation-based interventions (eg, treadmill training) have been shown to reduce NP after experimental SCI, suggesting transmission of pain-alleviating signals through mechanoreceptors. The aim of the present study was to understand the contribution of mechanoreceptors with respect to mechanical allodynia in a moderate mouse contusion SCI model. After genetic ablation of tropomyosin receptor kinase B expressing mechanoreceptors before SCI, mechanical allodynia was reduced. The identical genetic ablation after SCI did not yield any change in pain behavior. Peptidergic nociceptor sprouting into lamina III/IV below injury level as a consequence of SCI was not altered by either mechanoreceptor ablation. However, skin-nerve preparations of contusion SCI mice 7 days after injury yielded hyperexcitability in nociceptors, not in mechanoreceptors, which makes a substantial direct contribution of mechanoreceptors to NP maintenance unlikely. Complementing animal data, quantitative sensory testing in human SCI subjects indicated reduced mechanical pain thresholds, whereas the mechanical detection threshold was not altered. Taken together, early mechanoreceptor ablation modulates pain behavior, most likely through indirect mechanisms. Hyperexcitable nociceptors seem to be the main drivers of SCI-induced NP. Future studies need to focus on injury-derived factors triggering early-onset nociceptor hyperexcitability, which could serve as targets for more effective therapeutic interventions.


Subject(s)
Disease Models, Animal , Hyperalgesia , Mechanoreceptors , Mice, Inbred C57BL , Spinal Cord Injuries , Animals , Spinal Cord Injuries/complications , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Mice , Hyperalgesia/physiopathology , Hyperalgesia/etiology , Hyperalgesia/metabolism , Mechanoreceptors/metabolism , Mechanoreceptors/physiology , Male , Humans , Pain Threshold/physiology , Female , Pain Measurement , Mice, Transgenic , Neuralgia/etiology , Neuralgia/metabolism , Neuralgia/physiopathology
2.
PLoS One ; 18(10): e0291740, 2023.
Article in English | MEDLINE | ID: mdl-37906544

ABSTRACT

Locomotion is a complex task involving excitatory and inhibitory circuitry in spinal gray matter. While genetic knockouts examine the function of individual spinal interneuron (SpIN) subtypes, the phenotype of combined SpIN loss remains to be explored. We modified a kainic acid lesion to damage intermediate gray matter (laminae V-VIII) in the lumbar spinal enlargement (spinal L2-L4) in female rats. A thorough, tailored behavioral evaluation revealed deficits in gross hindlimb function, skilled walking, coordination, balance and gait two weeks post-injury. Using a Random Forest algorithm, we combined these behavioral assessments into a highly predictive binary classification system that strongly correlated with structural deficits in the rostro-caudal axis. Machine-learning quantification confirmed interneuronal damage to laminae V-VIII in spinal L2-L4 correlates with hindlimb dysfunction. White matter alterations and lower motoneuron loss were not observed with this KA lesion. Animals did not regain lost sensorimotor function three months after injury, indicating that natural recovery mechanisms of the spinal cord cannot compensate for loss of laminae V-VIII neurons. As gray matter damage accounts for neurological/walking dysfunction in instances of spinal cord injury affecting the cervical or lumbar enlargement, this research lays the groundwork for new neuroregenerative therapies to replace these lost neuronal pools vital to sensorimotor function.


Subject(s)
Gray Matter , Spinal Cord Injuries , Rats , Female , Animals , Gray Matter/pathology , Spinal Cord/pathology , Motor Neurons/physiology , Interneurons/physiology , Locomotion/physiology
3.
Cells ; 11(19)2022 09 30.
Article in English | MEDLINE | ID: mdl-36231048

ABSTRACT

Physical activity-based rehabilitative interventions represent the main treatment concept for people suffering from spinal cord injury (SCI). The role such interventions play in the relief of neuropathic pain (NP) states is emerging, along with underlying mechanisms resulting in SCI-induced NP (SCI-NP). Animal models have been used to investigate the benefits of activity-based interventions (ABI), such as treadmill training, wheel running, walking, swimming, and bipedal standing. These activity-based paradigms have been shown to modulate inflammatory-related alterations as well as induce functional and structural changes in the spinal cord gray matter circuitry correlated with pain behaviors. Thus far, the research available provides an incomplete picture of the cellular and molecular pathways involved in this beneficial effect. Continued research is essential for understanding how such interventions benefit SCI patients suffering from NP and allow the development of individualized rehabilitative therapies. This article reviews preclinical studies on this specific topic, goes over mechanisms involved in SCI-NP in relation to ABI, and then discusses the effectiveness of different activity-based paradigms as they relate to different forms, intensity, initiation times, and duration of ABI. This article also summarizes the mechanisms of respective interventions to ameliorate NP after SCI and provides suggestions for future research directions.


Subject(s)
Neuralgia , Spinal Cord Injuries , Animals , Gray Matter , Hyperalgesia/metabolism , Motor Activity , Neuralgia/metabolism , Spinal Cord Injuries/complications , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/therapy
4.
Science ; 376(6594): eabd5926, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35549409

ABSTRACT

Aging is associated with increased prevalence of axonal injuries characterized by poor regeneration and disability. However, the underlying mechanisms remain unclear. In our experiments, RNA sequencing of sciatic dorsal root ganglia (DRG) revealed significant aging-dependent enrichment in T cell signaling both before and after sciatic nerve injury (SNI) in mice. Lymphotoxin activated the transcription factor NF-κB, which induced expression of the chemokine CXCL13 by neurons. This in turn recruited CXCR5+CD8+ T cells to injured DRG neurons overexpressing major histocompatibility complex class I. CD8+ T cells repressed the axonal regeneration of DRG neurons via caspase 3 activation. CXCL13 neutralization prevented CXCR5+CD8+ T cell recruitment to the DRG and reversed aging-dependent regenerative decline, thereby promoting neurological recovery after SNI. Thus, axonal regeneration can be facilitated by antagonizing cross-talk between immune cells and neurons.


Subject(s)
Aging , Axons , CD8-Positive T-Lymphocytes , Ganglia, Spinal , Nerve Regeneration , Neurons , Sciatic Nerve , Aging/metabolism , Animals , Axons/physiology , CD8-Positive T-Lymphocytes/metabolism , Ganglia, Spinal/metabolism , Mice , Neurons/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/physiology
5.
eNeuro ; 8(2)2021.
Article in English | MEDLINE | ID: mdl-33593735

ABSTRACT

In many preclinical spinal cord injury (SCI) studies, assessment of locomotion recovery is key to understanding the effectiveness of the experimental intervention. In such rat SCI studies, the most basic locomotor recovery scoring system is a subjective observation of animals freely roaming in an open field, the Basso Beattie Bresnahan (BBB) score. In comparison, CatWalk is an automated gait analysis system, providing further parameter specifications. Although together the CatWalk parameters encompass gait, studies consistently report single parameters, which differ in significance from other behavioral assessments. Therefore, we believe no single parameter produced by the CatWalk can represent the fully-coordinated motion of gait. Typically, other locomotor assessments, such as the BBB score, combine several locomotor characteristics into a representative score. For this reason, we ranked the most distinctive CatWalk parameters between uninjured and SC injured rats. Subsequently, we combined nine of the topmost parameters into an SCI gait index score based on linear discriminant analysis (LDA). The resulting combination was applied to assess gait recovery in SCI experiments comprising of three thoracic contusions, a thoracic dorsal hemisection, and a cervical dorsal column lesion model. For thoracic lesions, our unbiased machine learning model revealed gait differences in lesion type and severity. In some instances, our LDA was found to be more sensitive in differentiating recovery than the BBB score alone. We believe the newly developed gait parameter combination presented here should be used in CatWalk gait recovery work with preclinical thoracic rat SCI models.


Subject(s)
Spinal Cord Injuries , Animals , Disease Models, Animal , Gait , Locomotion , Rats , Recovery of Function , Spinal Cord
6.
Cells ; 10(1)2020 12 28.
Article in English | MEDLINE | ID: mdl-33379276

ABSTRACT

The central nervous system (CNS) does not recover from traumatic axonal injury, but the peripheral nervous system (PNS) does. We hypothesize that this fundamental difference in regenerative capacity may be based upon the absence of stimulatory mechanical forces in the CNS due to the protective rigidity of the vertebral column and skull. We developed a bioreactor to apply low-strain cyclic axonal stretch to adult rat dorsal root ganglia (DRG) connected to either the peripheral or central nerves in an explant model for inducing axonal growth. In response, larger diameter DRG neurons, mechanoreceptors and proprioceptors showed enhanced neurite outgrowth as well as increased Activating Transcription Factor 3 (ATF3).


Subject(s)
Central Nervous System/cytology , Ganglia, Spinal/cytology , Neurons/cytology , Activating Transcription Factor 3/metabolism , Animals , Cells, Cultured , Female , Male , Neuronal Outgrowth , Rats , Tensile Strength
7.
Nat Metab ; 2(9): 918-933, 2020 09.
Article in English | MEDLINE | ID: mdl-32778834

ABSTRACT

Regeneration after injury occurs in axons that lie in the peripheral nervous system but fails in the central nervous system, thereby limiting functional recovery. Differences in axonal signalling in response to injury that might underpin this differential regenerative ability are poorly characterized. Combining axoplasmic proteomics from peripheral sciatic or central projecting dorsal root ganglion (DRG) axons with cell body RNA-seq, we uncover injury-dependent signalling pathways that are uniquely represented in peripheral versus central projecting sciatic DRG axons. We identify AMPK as a crucial regulator of axonal regenerative signalling that is specifically downregulated in injured peripheral, but not central, axons. We find that AMPK in DRG interacts with the 26S proteasome and its CaMKIIα-dependent regulatory subunit PSMC5 to promote AMPKα proteasomal degradation following sciatic axotomy. Conditional deletion of AMPKα1 promotes multiple regenerative signalling pathways after central axonal injury and stimulates robust axonal growth across the spinal cord injury site, suggesting inhibition of AMPK as a therapeutic strategy to enhance regeneration following spinal cord injury.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Axons , Ganglia, Spinal/metabolism , Nerve Regeneration , Sensory Receptor Cells/metabolism , Spinal Cord Injuries/metabolism , ATPases Associated with Diverse Cellular Activities/metabolism , Animals , Axonal Transport , Axotomy , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Female , Ganglia, Spinal/pathology , Mice , Mice, Inbred C57BL , Proteasome Endopeptidase Complex/metabolism , Proteomics , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Sensory Receptor Cells/pathology , Spinal Cord Injuries/pathology
8.
EMBO J ; 38(13): e101032, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31268609

ABSTRACT

The molecular mechanisms discriminating between regenerative failure and success remain elusive. While a regeneration-competent peripheral nerve injury mounts a regenerative gene expression response in bipolar dorsal root ganglia (DRG) sensory neurons, a regeneration-incompetent central spinal cord injury does not. This dichotomic response offers a unique opportunity to investigate the fundamental biological mechanisms underpinning regenerative ability. Following a pharmacological screen with small-molecule inhibitors targeting key epigenetic enzymes in DRG neurons, we identified HDAC3 signalling as a novel candidate brake to axonal regenerative growth. In vivo, we determined that only a regenerative peripheral but not a central spinal injury induces an increase in calcium, which activates protein phosphatase 4 that in turn dephosphorylates HDAC3, thus impairing its activity and enhancing histone acetylation. Bioinformatics analysis of ex vivo H3K9ac ChIPseq and RNAseq from DRG followed by promoter acetylation and protein expression studies implicated HDAC3 in the regulation of multiple regenerative pathways. Finally, genetic or pharmacological HDAC3 inhibition overcame regenerative failure of sensory axons following spinal cord injury. Together, these data indicate that PP4-dependent HDAC3 dephosphorylation discriminates between axonal regeneration and regenerative failure.


Subject(s)
Ganglia, Spinal/physiology , Histone Deacetylases/metabolism , Peripheral Nerve Injuries/metabolism , Phosphoprotein Phosphatases/metabolism , Small Molecule Libraries/pharmacology , Animals , Axons , Cells, Cultured , Disease Models, Animal , Epigenesis, Genetic/drug effects , Female , Male , Mice , Nerve Regeneration , Phosphorylation/drug effects , Signal Transduction
9.
Tissue Eng Part A ; 25(7-8): 522-537, 2019 04.
Article in English | MEDLINE | ID: mdl-30351234

ABSTRACT

IMPACT STATEMENT: Axonal bridging across a lesion in the injured spinal cord requires a growth substrate and guidance cues. Using alginate hydrogels with capillary channels we show that poly-l-ornithine and laminin can be stably bound and improve cell adhesion and neurite growth in vitro, and axon growth in vivo by enhancing host cell infiltration in the injured spinal cord. Filling of coated hydrogels with postnatal astrocytes further increases short-distance axon growth and results in a continuous astroglial substrate across the host/graft interface. Thus, positively charged bioactive molecules can be stably bound to anisotropic capillary alginate hydrogels and early astrocytes further promote tissue integration.


Subject(s)
Alginates/chemistry , Hydrogels/chemistry , Peptides/chemistry , Spinal Cord Injuries/therapy , Animals , Axons , Biocompatible Materials/chemistry , Guided Tissue Regeneration/methods , Humans
10.
J Neurotrauma ; 35(18): 2222-2238, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29706124

ABSTRACT

A large proportion of patients suffering from spinal cord injury (SCI) develop chronic central neuropathic pain. Previously, we and others have shown that sensorimotor training early after SCI can prevent the development of mechanical allodynia. To determine whether training initiated in the subchronic/chronic phase remains effective, correlates of below-level neuropathic pain were analyzed in the hindpaws 5-10 weeks after a moderate T11 contusion SCI (50 kDyn) in adult female C57BL/6 mice. In a comparison of SCI and sham mice 5 weeks post-injury, about 80% of injured animals developed mechanical hypersensitivity to light mechanical stimuli, whereas testing of noxious stimuli revealed hypo-responsiveness. Thermal sensitivity testing showed a decreased response latency after injury. Without intervention, mechanical and thermal hyper-responsiveness were evident until the end of the experiment (10 weeks). In contrast, treadmill training (2 × 15 min/day; 5 × /week) initiated 6 weeks post-injury resulted in partial amelioration of pain behavior and this effect remained stable. Analysis of calcitonin gene-related peptide (CGRP)-labeled fibers in lamina III-IV of the lumbar dorsal horn revealed an increase in labeling density after SCI. This was not due to changes in the number or size distribution of CGRP-labeled lumbar dorsal root ganglion neurons. Treadmill training reduced the CGRP-labeling density in the spinal cord of injured mice, whereas the density of non-peptidergic isolectin-B4 (IB4)+ fibers showed no changes in lamina IIi and a slight reduction of sparse IB4 labeling in laminae III-IV. Thus, sensorimotor activity initiated in the subchronic/chronic phase of SCI remains effective in ameliorating pain behavior and influencing structural changes of the nociceptive system.


Subject(s)
Neuralgia/physiopathology , Nociceptors/pathology , Physical Conditioning, Animal/methods , Spinal Cord Injuries/physiopathology , Animals , Chronic Disease , Female , Hyperalgesia/etiology , Hyperalgesia/physiopathology , Mice , Mice, Inbred C57BL , Neuralgia/etiology , Spinal Cord Injuries/complications , Spinal Cord Injuries/pathology
11.
Exp Neurol ; 306: 250-259, 2018 08.
Article in English | MEDLINE | ID: mdl-29408734

ABSTRACT

Following a spinal cord injury (SCI) a growth aversive environment forms, consisting of a fibroglial scar and inhibitory factors, further restricting the already low intrinsic growth potential of injured adult central nervous system (CNS) neurons. Previous studies have shown that local administration of the microtubule-stabilizing drug paclitaxel or epothilone B (Epo B) reduce fibrotic scar formation and axonal dieback as well as induce axonal growth/sprouting after SCI. Likewise, systemic administration of Epo B promoted functional recovery. In this study, we investigated the effects of epothilone D (Epo D), an analog of Epo B with a possible greater therapeutic index, on fibrotic scarring, axonal sprouting and functional recovery after SCI. Delayed systemic administration of Epo D after a moderate contusion injury (150 kDyn) in female Fischer 344 rats resulted in a reduced number of footfalls when crossing a horizontal ladder at 4 and 8 weeks post-injury. Hindlimb motor function assessed with the BBB open field locomotor rating scale and Catwalk gait analysis were not significantly altered. Moreover, formation of laminin positive fibrotic scar tissue and 5-HT positive serotonergic fiber length caudal to the lesion site were not altered after treatment with Epo D. These findings recapitulate a functional benefit after systemic administration of a microtubule-stabilizing drug in rat contusion SCI.


Subject(s)
Epothilones/therapeutic use , Hindlimb/physiopathology , Spinal Cord Injuries/drug therapy , Tubulin Modulators/therapeutic use , Animals , Axons/drug effects , Contusions/drug therapy , Contusions/physiopathology , Female , Fibrosis , Locomotion , Microtubules/drug effects , Nerve Fibers/drug effects , Nerve Regeneration , Psychomotor Performance , Rats , Rats, Inbred F344 , Recovery of Function , Serotonin/metabolism , Spinal Cord Injuries/physiopathology
12.
Acta Biomater ; 60: 167-180, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28735026

ABSTRACT

Grafting of cell-seeded alginate capillary hydrogels into a spinal cord lesion site provides an axonal bridge while physically directing regenerating axonal growth in a linear pattern. However, without an additional growth stimulus, bridging axons fail to extend into the distal host spinal cord. Here we examined whether a combinatory strategy would support regeneration of descending axons across a cervical (C5) lateral hemisection lesion in the rat spinal cord. Following spinal cord transections, Schwann cell (SC)-seeded alginate hydrogels were grafted to the lesion site and AAV5 expressing brain-derived neurotrophic factor (BDNF) under control of a tetracycline-regulated promoter was injected caudally. In addition, we examined whether SC injection into the caudal spinal parenchyma would further enhance regeneration of descending axons to re-enter the host spinal cord. Our data show that both serotonergic and descending axons traced by biotinylated dextran amine (BDA) extend throughout the scaffolds. The number of regenerating axons is significantly increased when caudal BDNF expression is activated and transient BDNF delivery is able to sustain axons after gene expression is switched off. Descending axons are confined to the caudal graft/host interface even with continuous BDNF expression for 8weeks. Only with a caudal injection of SCs, a pathway facilitating axonal regeneration through the host/graft interface is generated allowing axons to successfully re-enter the caudal spinal cord. STATEMENT OF SIGNIFICANCE: Recovery from spinal cord injury is poor due to the limited regeneration observed in the adult mammalian central nervous system. Biomaterials, cell transplantation and growth factors that can guide axons across a lesion site, provide a cellular substrate, stimulate axon growth and have shown some promise in increasing the growth distance of regenerating axons. In the present study, we combined an alginate biomaterial with linear channels with transplantation of Schwann cells within and beyond the lesion site and injection of a regulatable vector for the transient expression of brain-derived neurotrophic factor (BDNF). Our data show that only with the full combination axons extend across the lesion site and that expression of BDNF beyond 4weeks does not further increase the number of regenerating axons.


Subject(s)
Alginates , Axons/physiology , Brain-Derived Neurotrophic Factor , Dependovirus , Hydrogels , Regeneration , Schwann Cells , Spinal Cord Injuries/therapy , Transduction, Genetic , Alginates/chemistry , Alginates/pharmacology , Animals , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , Female , Genetic Vectors , Glucuronic Acid/chemistry , Glucuronic Acid/pharmacology , Hexuronic Acids/chemistry , Hexuronic Acids/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Rats , Rats, Inbred F344 , Rats, Transgenic , Schwann Cells/metabolism , Schwann Cells/pathology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
13.
Front Cell Neurosci ; 11: 430, 2017.
Article in English | MEDLINE | ID: mdl-29375316

ABSTRACT

Spinal cord injury (SCI), resulting in para- and tetraplegia caused by the partial or complete disruption of descending motor and ascending sensory neurons, represents a complex neurological condition that remains incurable. Following SCI, numerous obstacles comprising of the loss of neural tissue (neurons, astrocytes, and oligodendrocytes), formation of a cavity, inflammation, loss of neuronal circuitry and function must be overcome. Given the multifaceted primary and secondary injury events that occur with SCI treatment options are likely to require combinatorial therapies. While several methods have been explored, only the intersection of two, cell transplantation and biomaterial implantation, will be addressed in detail here. Owing to the constant advance of cell culture technologies, cell-based transplantation has come to the forefront of SCI treatment in order to replace/protect damaged tissue and provide physical as well as trophic support for axonal regrowth. Biomaterial scaffolds provide cells with a protected environment from the surrounding lesion, in addition to bridging extensive damage and providing physical and directional support for axonal regrowth. Moreover, in this combinatorial approach cell transplantation improves scaffold integration and therefore regenerative growth potential. Here, we review the advances in combinatorial therapies of Schwann cells (SCs), astrocytes, olfactory ensheathing cells (OECs), mesenchymal stem cells, as well as neural stem and progenitor cells (NSPCs) with various biomaterial scaffolds.

14.
Front Mol Neurosci ; 10: 445, 2017.
Article in English | MEDLINE | ID: mdl-29379413

ABSTRACT

Adult mammalian central nervous system (CNS) neurons are unable to regenerate following axonal injury, leading to permanent functional impairments. Yet, the reasons underlying this regeneration failure are not fully understood. Here, we studied the transcriptome and translatome shortly after spinal cord injury. Profiling of the total and ribosome-bound RNA in injured and naïve spinal cords identified a substantial post-transcriptional regulation of gene expression. In particular, transcripts associated with nervous system development were down-regulated in the total RNA fraction while remaining stably loaded onto ribosomes. Interestingly, motif association analysis of post-transcriptionally regulated transcripts identified the cytoplasmic polyadenylation element (CPE) as enriched in a subset of these transcripts that was more resistant to injury-induced reduction at the transcriptome level. Modulation of these transcripts by overexpression of the CPE binding protein, Cpeb1, in mouse and Drosophila CNS neurons promoted axonal regeneration following injury. Our study uncovered a global evolutionarily conserved post-transcriptional mechanism enhancing regeneration of injured CNS axons.

15.
Brain ; 138(Pt 7): 1843-62, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25981963

ABSTRACT

Regeneration of injured central nervous system axons is highly restricted, causing neurological impairment. To date, although the lack of intrinsic regenerative potential is well described, a key regulatory molecular mechanism for the enhancement of both axonal regrowth and functional recovery after central nervous system injury remains elusive. While ubiquitin ligases coordinate neuronal morphogenesis and connectivity during development as well as after axonal injury, their role specifically in axonal regeneration is unknown. Following a bioinformatics network analysis combining ubiquitin ligases with previously defined axonal regenerative proteins, we found a triad composed of the ubiquitin ligases MDM4, MDM2 and the transcription factor p53 (encoded by TP53) as a putative central signalling complex restricting the regeneration program. Indeed, conditional deletion of MDM4 or pharmacological inhibition of MDM2/p53 interaction in the eye and spinal cord promote axonal regeneration and sprouting of the optic nerve after crush and of supraspinal tracts after spinal cord injury. The double conditional deletion of MDM4-p53 as well as MDM2 inhibition in p53-deficient mice blocks this regenerative phenotype, showing its dependence upon p53. Genome-wide gene expression analysis from ex vivo fluorescence-activated cell sorting in MDM4-deficient retinal ganglion cells identifies the downstream target IGF1R, whose activity and expression was found to be required for the regeneration elicited by MDM4 deletion. Importantly, we demonstrate that pharmacological enhancement of the MDM2/p53-IGF1R axis enhances axonal sprouting as well as functional recovery after spinal cord injury. Thus, our results show MDM4-MDM2/p53-IGF1R as an original regulatory mechanism for CNS regeneration and offer novel targets to enhance neurological recovery.media-1vid110.1093/brain/awv125_video_abstractawv125_video_abstract.


Subject(s)
Nerve Regeneration/physiology , Optic Nerve Injuries/metabolism , Recovery of Function/physiology , Signal Transduction/physiology , Spinal Cord Injuries/metabolism , Animals , Axons/metabolism , Axons/pathology , Computational Biology , Disease Models, Animal , Flow Cytometry , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nerve Crush , Optic Nerve Injuries/pathology , Optic Nerve Injuries/physiopathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Transcriptome , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism
16.
Nat Commun ; 5: 3527, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24686445

ABSTRACT

Axonal regenerative failure is a major cause of neurological impairment following central nervous system (CNS) but not peripheral nervous system (PNS) injury. Notably, PNS injury triggers a coordinated regenerative gene expression programme. However, the molecular link between retrograde signalling and the regulation of this gene expression programme that leads to the differential regenerative capacity remains elusive. Here we show through systematic epigenetic studies that the histone acetyltransferase p300/CBP-associated factor (PCAF) promotes acetylation of histone 3 Lys 9 at the promoters of established key regeneration-associated genes following a peripheral but not a central axonal injury. Furthermore, we find that extracellular signal-regulated kinase (ERK)-mediated retrograde signalling is required for PCAF-dependent regenerative gene reprogramming. Finally, PCAF is necessary for conditioning-dependent axonal regeneration and also singularly promotes regeneration after spinal cord injury. Thus, we find a specific epigenetic mechanism that regulates axonal regeneration of CNS axons, suggesting novel targets for clinical application.


Subject(s)
Axons/enzymology , Central Nervous System/physiology , Epigenesis, Genetic , Nerve Regeneration , Spinal Cord Injuries/enzymology , p300-CBP Transcription Factors/metabolism , Acetylation , Animals , Female , Histones/metabolism , Humans , Male , Mice , Mice, Knockout/genetics , Spinal Cord Injuries/genetics , Spinal Cord Injuries/physiopathology , p300-CBP Transcription Factors/genetics
17.
Sci Data ; 1: 140038, 2014.
Article in English | MEDLINE | ID: mdl-25977793

ABSTRACT

The regulatory mechanisms responsible for the gene expression pattern associated with axotomy-dependent signaling affecting the neuronal phenotype, including the axonal regenerative program, remain unclear. To further this understanding, we recently performed DNA methylation temporal profiling in lumbar dorsal root ganglia (DRG) after axotomy of the central spinal (non-regenerating) and of the peripheral sciatic nerve (regenerating) axonal branches. DNA methylation microarrays for mouse gene promoters and CpG islands (Roche/NimbleGen) were employed after immunoprecipitation of 5-methylcytosine-DNA. Here we provide a detailed data descriptor of this DNA methylation dataset, which allows in depth evaluation of the experimental design, assessment of data reproducibility and a full interactive operator-based systematic data analysis. In fact, we offer a methylation 'hit' scoring map of the whole microarray data in a workable spreadsheet that allows data sorting by genes, conditions or hits of interests that is ready for functional gene annotation and classification. This dataset allows investigators bioinformatic comparison to other epigenetic and gene expression datasets and further experimental characterization of the role of DNA methylation in axotomy-dependent pathways.


Subject(s)
DNA Methylation , Ganglia, Spinal , Sciatic Nerve , Animals , Axons/metabolism , Axotomy , Central Nervous System/metabolism , Ganglia, Spinal/metabolism , Mice , Sciatic Nerve/metabolism , Sciatic Nerve/pathology
18.
Neurotherapeutics ; 10(4): 771-81, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23881454

ABSTRACT

Inadequate axonal sprouting and lack of regeneration limit functional recovery following neurologic injury, such as stroke, brain, and traumatic spinal cord injury. Recently, the enhancement of the neuronal regenerative program has led to promising improvements in axonal sprouting and regeneration in animal models of axonal injury. However, precise knowledge of the essential molecular determinants of this regenerative program remains elusive, thus limiting the choice of fully effective therapeutic strategies. Given that molecular regulation of axonal outgrowth and regeneration requires carefully orchestrated waves of gene expression, both temporally and spatially, epigenetic changes may be an ideal regulatory mechanism to address this unique need. While recent evidence suggests that epigenetic modifications could contribute to the regulation of axonal outgrowth and regeneration following axonal injury in models of stroke, and spinal cord and optic nerve injury, a number of unanswered questions remain. Such questions require systematic investigation of the epigenetic landscape between regenerative and non-regenerative conditions for the potential translation of this knowledge into regenerative strategies in human spinal and brain injury, as well as stroke.


Subject(s)
Axons/physiology , Brain Injuries/genetics , Epigenesis, Genetic , Nerve Regeneration/genetics , Spinal Cord Injuries/genetics , Brain Injuries/physiopathology , Humans , Spinal Cord Injuries/physiopathology
19.
J Cell Biol ; 193(7): 1147-56, 2011 Jun 27.
Article in English | MEDLINE | ID: mdl-21690307

ABSTRACT

After an acute central nervous system injury, axonal regeneration is limited as the result of a lack of neuronal intrinsic competence and the presence of extrinsic inhibitory signals. The injury fragments the myelin neuronal insulating layer, releasing extrinsic inhibitory molecules to signal through the neuronal membrane-bound Nogo receptor (NgR) complex. In this paper, we show that a neuronal transcriptional pathway can interfere with extrinsic inhibitory myelin-dependent signaling, thereby promoting neurite outgrowth. Specifically, retinoic acid (RA), acting through the RA receptor ß (RAR-ß), inhibited myelin-activated NgR signaling through the transcriptional repression of the NgR complex member Lingo-1. We show that suppression of Lingo-1 was required for RA-RAR-ß to counteract extrinsic inhibition of neurite outgrowth. Furthermore, we confirm in vivo that RA treatment after a dorsal column overhemisection injury inhibited Lingo-1 expression, specifically through RAR-ß. Our findings identify a novel link between RA-RAR-ß-dependent proaxonal outgrowth and inhibitory NgR complex-dependent signaling, potentially allowing for the development of molecular strategies to enhance axonal regeneration after a central nervous system injury.


Subject(s)
Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Neurites/metabolism , Receptors, Retinoic Acid/physiology , Tretinoin/physiology , Animals , Cells, Cultured , Gene Expression Regulation , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Myelin Sheath/physiology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Neurites/ultrastructure , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Tretinoin/metabolism
20.
Front Mol Neurosci ; 4: 59, 2011.
Article in English | MEDLINE | ID: mdl-22287943

ABSTRACT

Following an acute central nervous system (CNS) injury, axonal regeneration and functional recovery are extremely limited. This is due to an extrinsic inhibitory growth environment and the lack of intrinsic growth competence. Retinoic acid (RA) signaling, essential in developmental dorsoventral patterning and specification of spinal motor neurons, has been shown through its receptor, the transcription factor RA receptor ß2 (RARß2), to induce axonal regeneration following spinal cord injury (SCI). Recently, it has been shown that in dorsal root ganglion neurons (DRGs), cAMP levels were greatly increased by lentiviral RARß2 expression and contributed to neurite outgrowth. Moreover, RARßagonists, in cerebellar granule neurons (CGN) and in the brain in vivo, induced phosphoinositide 3-kinase dependent phosphorylation of AKT that was involved in RARß-dependent neurite outgrowth. More recently, RA-RARßpathways were shown to directly transcriptionally repress a member of the inhibitory Nogo receptor (NgR) complex, Lingo-1, under an axonal growth inhibitory environment in vitro as well as following spinal injury in vivo. This perspective focuses on these newly discovered molecular mechanisms and future directions in the field.

SELECTION OF CITATIONS
SEARCH DETAIL
...