Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Science ; 381(6664): 1338-1345, 2023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37733871

RESUMO

Axon regeneration can be induced across anatomically complete spinal cord injury (SCI), but robust functional restoration has been elusive. Whether restoring neurological functions requires directed regeneration of axons from specific neuronal subpopulations to their natural target regions remains unclear. To address this question, we applied projection-specific and comparative single-nucleus RNA sequencing to identify neuronal subpopulations that restore walking after incomplete SCI. We show that chemoattracting and guiding the transected axons of these neurons to their natural target region led to substantial recovery of walking after complete SCI in mice, whereas regeneration of axons simply across the lesion had no effect. Thus, reestablishing the natural projections of characterized neurons forms an essential part of axon regeneration strategies aimed at restoring lost neurological functions.


Assuntos
Axônios , Regeneração Nervosa , Paralisia , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal , Caminhada , Animais , Camundongos , Axônios/fisiologia , Regeneração Nervosa/genética , Regeneração Nervosa/fisiologia , Neurônios/fisiologia , Paralisia/fisiopatologia , Traumatismos da Medula Espinal/fisiopatologia , Conectoma
2.
Nat Neurosci ; 25(12): 1584-1596, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36396975

RESUMO

A spinal cord injury disrupts communication between the brain and the circuits in the spinal cord that regulate neurological functions. The consequences are permanent paralysis, loss of sensation and debilitating dysautonomia. However, the majority of circuits located above and below the injury remain anatomically intact, and these circuits can reorganize naturally to improve function. In addition, various neuromodulation therapies have tapped into these processes to further augment recovery. Emerging research is illuminating the requirements to reconstitute damaged circuits. Here, we summarize these natural and targeted reorganizations of circuits after a spinal cord injury. We also advocate for new concepts of reorganizing circuits informed by multi-omic single-cell atlases of recovery from injury. These atlases will uncover the molecular logic that governs the selection of 'recovery-organizing' neuronal subpopulations, and are poised to herald a new era in spinal cord medicine.


Assuntos
Traumatismos da Medula Espinal , Humanos , Traumatismos da Medula Espinal/terapia , Sensação , Encéfalo
3.
Nature ; 611(7936): 540-547, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36352232

RESUMO

A spinal cord injury interrupts pathways from the brain and brainstem that project to the lumbar spinal cord, leading to paralysis. Here we show that spatiotemporal epidural electrical stimulation (EES) of the lumbar spinal cord1-3 applied during neurorehabilitation4,5 (EESREHAB) restored walking in nine individuals with chronic spinal cord injury. This recovery involved a reduction in neuronal activity in the lumbar spinal cord of humans during walking. We hypothesized that this unexpected reduction reflects activity-dependent selection of specific neuronal subpopulations that become essential for a patient to walk after spinal cord injury. To identify these putative neurons, we modelled the technological and therapeutic features underlying EESREHAB in mice. We applied single-nucleus RNA sequencing6-9 and spatial transcriptomics10,11 to the spinal cords of these mice to chart a spatially resolved molecular atlas of recovery from paralysis. We then employed cell type12,13 and spatial prioritization to identify the neurons involved in the recovery of walking. A single population of excitatory interneurons nested within intermediate laminae emerged. Although these neurons are not required for walking before spinal cord injury, we demonstrate that they are essential for the recovery of walking with EES following spinal cord injury. Augmenting the activity of these neurons phenocopied the recovery of walking enabled by EESREHAB, whereas ablating them prevented the recovery of walking that occurs spontaneously after moderate spinal cord injury. We thus identified a recovery-organizing neuronal subpopulation that is necessary and sufficient to regain walking after paralysis. Moreover, our methodology establishes a framework for using molecular cartography to identify the neurons that produce complex behaviours.


Assuntos
Neurônios , Paralisia , Traumatismos da Medula Espinal , Medula Espinal , Caminhada , Animais , Humanos , Camundongos , Neurônios/fisiologia , Paralisia/genética , Paralisia/fisiopatologia , Paralisia/terapia , Medula Espinal/citologia , Medula Espinal/fisiologia , Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/terapia , Caminhada/fisiologia , Estimulação Elétrica , Região Lombossacral/inervação , Reabilitação Neurológica , Análise de Sequência de RNA , Perfilação da Expressão Gênica
4.
Nat Biotechnol ; 40(2): 198-208, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34580478

RESUMO

Optoelectronic systems can exert precise control over targeted neurons and pathways throughout the brain in untethered animals, but similar technologies for the spinal cord are not well established. In the present study, we describe a system for ultrafast, wireless, closed-loop manipulation of targeted neurons and pathways across the entire dorsoventral spinal cord in untethered mice. We developed a soft stretchable carrier, integrating microscale light-emitting diodes (micro-LEDs), that conforms to the dura mater of the spinal cord. A coating of silicone-phosphor matrix over the micro-LEDs provides mechanical protection and light conversion for compatibility with a large library of opsins. A lightweight, head-mounted, wireless platform powers the micro-LEDs and performs low-latency, on-chip processing of sensed physiological signals to control photostimulation in a closed loop. We use the device to reveal the role of various neuronal subtypes, sensory pathways and supraspinal projections in the control of locomotion in healthy and spinal-cord injured mice.


Assuntos
Optogenética , Tecnologia sem Fio , Animais , Encéfalo/fisiologia , Camundongos , Neurônios/fisiologia , Medula Espinal/fisiologia
5.
Nat Commun ; 12(1): 5692, 2021 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34584091

RESUMO

Differential expression analysis in single-cell transcriptomics enables the dissection of cell-type-specific responses to perturbations such as disease, trauma, or experimental manipulations. While many statistical methods are available to identify differentially expressed genes, the principles that distinguish these methods and their performance remain unclear. Here, we show that the relative performance of these methods is contingent on their ability to account for variation between biological replicates. Methods that ignore this inevitable variation are biased and prone to false discoveries. Indeed, the most widely used methods can discover hundreds of differentially expressed genes in the absence of biological differences. To exemplify these principles, we exposed true and false discoveries of differentially expressed genes in the injured mouse spinal cord.


Assuntos
Confiabilidade dos Dados , Modelos Estatísticos , RNA-Seq/métodos , Análise de Célula Única/métodos , Animais , Variação Biológica Individual , Variação Biológica da População , Conjuntos de Dados como Assunto , Regulação da Expressão Gênica , Humanos , Camundongos , RNA-Seq/estatística & dados numéricos , Coelhos , Ratos , Análise de Célula Única/estatística & dados numéricos , Suínos
6.
Nat Biotechnol ; 39(1): 30-34, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32690972

RESUMO

We present Augur, a method to prioritize the cell types most responsive to biological perturbations in single-cell data. Augur employs a machine-learning framework to quantify the separability of perturbed and unperturbed cells within a high-dimensional space. We validate our method on single-cell RNA sequencing, chromatin accessibility and imaging transcriptomics datasets, and show that Augur outperforms existing methods based on differential gene expression. Augur identified the neural circuits restoring locomotion in mice following spinal cord neurostimulation.


Assuntos
Biologia Computacional/métodos , Aprendizado de Máquina , Análise de Célula Única/métodos , Transcriptoma , Animais , Cromatina/genética , Cromatina/metabolismo , Bases de Dados Genéticas , Perfilação da Expressão Gênica/métodos , Camundongos , Rede Nervosa/metabolismo , Ratos , Análise de Sequência de RNA , Transcriptoma/genética , Transcriptoma/fisiologia , Caminhada/fisiologia
7.
Nat Commun ; 11(1): 6425, 2020 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-33349630

RESUMO

Overcoming the restricted axonal regenerative ability that limits functional repair following a central nervous system injury remains a challenge. Here we report a regenerative paradigm that we call enriched conditioning, which combines environmental enrichment (EE) followed by a conditioning sciatic nerve axotomy that precedes a spinal cord injury (SCI). Enriched conditioning significantly increases the regenerative ability of dorsal root ganglia (DRG) sensory neurons compared to EE or a conditioning injury alone, propelling axon growth well beyond the spinal injury site. Mechanistically, we established that enriched conditioning relies on the unique neuronal intrinsic signaling axis PKC-STAT3-NADPH oxidase 2 (NOX2), enhancing redox signaling as shown by redox proteomics in DRG. Finally, NOX2 conditional deletion or overexpression respectively blocked or phenocopied enriched conditioning-dependent axon regeneration after SCI leading to improved functional recovery. These studies provide a paradigm that drives the regenerative ability of sensory neurons offering a potential redox-dependent regenerative model for mechanistic and therapeutic discoveries.


Assuntos
Regeneração Nervosa , Células Receptoras Sensoriais/metabolismo , Células Receptoras Sensoriais/patologia , Transdução de Sinais , Traumatismos da Medula Espinal/fisiopatologia , Animais , Axônios/patologia , Axotomia , Gânglios Espinais/patologia , Camundongos Endogâmicos C57BL , NADPH Oxidase 2/metabolismo , Crescimento Neuronal , Plasticidade Neuronal , Oxirredução , Fosforilação , Regiões Promotoras Genéticas/genética , Proteína Quinase C/metabolismo , Subunidades Proteicas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT3/metabolismo , Nervo Isquiático/fisiopatologia , Regulação para Cima
8.
Nat Metab ; 2(9): 918-933, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32778834

RESUMO

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.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Axônios , Gânglios Espinais/metabolismo , Regeneração Nervosa , Células Receptoras Sensoriais/metabolismo , Traumatismos da Medula Espinal/metabolismo , ATPases Associadas a Diversas Atividades Celulares/metabolismo , Animais , Transporte Axonal , Axotomia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Feminino , Gânglios Espinais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteômica , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia , Células Receptoras Sensoriais/patologia , Traumatismos da Medula Espinal/patologia
9.
Sci Transl Med ; 12(551)2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32641489

RESUMO

Cyclin-dependent-like kinase 5 (CDKL5) gene mutations lead to an X-linked disorder that is characterized by infantile epileptic encephalopathy, developmental delay, and hypotonia. However, we found that a substantial percentage of these patients also report a previously unrecognized anamnestic deficiency in pain perception. Consistent with a role in nociception, we found that CDKL5 is expressed selectively in nociceptive dorsal root ganglia (DRG) neurons in mice and in induced pluripotent stem cell (iPS)-derived human nociceptors. CDKL5-deficient mice display defective epidermal innervation, and conditional deletion of CDKL5 in DRG sensory neurons impairs nociception, phenocopying CDKL5 deficiency disorder in patients. Mechanistically, CDKL5 interacts with calcium/calmodulin-dependent protein kinase II α (CaMKIIα) to control outgrowth and transient receptor potential cation channel subfamily V member 1 (TRPV1)-dependent signaling, which are disrupted in both CDKL5 mutant murine DRG and human iPS-derived nociceptors. Together, these findings unveil a previously unrecognized role for CDKL5 in nociception, proposing an original regulatory mechanism for pain perception with implications for future therapeutics in CDKL5 deficiency disorder.


Assuntos
Células Receptoras Sensoriais , Transdução de Sinais , Animais , Ciclinas , Modelos Animais de Doenças , Humanos , Camundongos , Dor , Proteínas Serina-Treonina Quinases/genética
10.
Nat Rev Neurol ; 15(12): 732-745, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31728042

RESUMO

Over the past decade, we have witnessed a flourishing of novel strategies to enhance neuroplasticity and promote axon regeneration following spinal cord injury, and results from preclinical studies suggest that some of these strategies have the potential for clinical translation. Spinal cord injury leads to the disruption of neural circuitry and connectivity, resulting in permanent neurological disability. Recovery of function relies on augmenting neuroplasticity to potentiate sprouting and regeneration of spared and injured axons, to increase the strength of residual connections and to promote the formation of new connections and circuits. Neuroplasticity can be fostered by exploiting four main biological properties: neuronal intrinsic signalling, the neuronal extrinsic environment, the capacity to reconnect the severed spinal cord via neural stem cell grafts, and modulation of neuronal activity. In this Review, we discuss experimental evidence from rodents, nonhuman primates and patients regarding interventions that target each of these four properties. We then highlight the strengths and challenges of individual and combinatorial approaches with respect to clinical translation. We conclude by considering future developments and providing views on how to bridge the gap between preclinical studies and clinical translation.


Assuntos
Regeneração Nervosa/fisiologia , Plasticidade Neuronal/fisiologia , Recuperação de Função Fisiológica/fisiologia , Traumatismos da Medula Espinal/metabolismo , Pesquisa Translacional Biomédica/métodos , Animais , Axônios/fisiologia , Humanos , Traumatismos da Medula Espinal/diagnóstico , Traumatismos da Medula Espinal/genética
11.
Nat Neurosci ; 22(11): 1913-1924, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31591560

RESUMO

Axonal injury results in regenerative success or failure, depending on whether the axon lies in the peripheral or the CNS, respectively. The present study addresses whether epigenetic signatures in dorsal root ganglia discriminate between regenerative and non-regenerative axonal injury. Chromatin immunoprecipitation for the histone 3 (H3) post-translational modifications H3K9ac, H3K27ac and H3K27me3; an assay for transposase-accessible chromatin; and RNA sequencing were performed in dorsal root ganglia after sciatic nerve or dorsal column axotomy. Distinct histone acetylation and chromatin accessibility signatures correlated with gene expression after peripheral, but not central, axonal injury. DNA-footprinting analyses revealed new transcriptional regulators associated with regenerative ability. Machine-learning algorithms inferred the direction of most of the gene expression changes. Neuronal conditional deletion of the chromatin remodeler CCCTC-binding factor impaired nerve regeneration, implicating chromatin organization in the regenerative competence. Altogether, the present study offers the first epigenomic map providing insight into the transcriptional response to injury and the differential regenerative ability of sensory neurons.


Assuntos
Axônios/fisiologia , Epigenômica , Gânglios Espinais/fisiologia , Regeneração Nervosa/fisiologia , Células Receptoras Sensoriais/fisiologia , Acetilação , Algoritmos , Animais , Fator de Ligação a CCCTC/genética , Cromatina/metabolismo , Feminino , Gânglios Espinais/lesões , Expressão Gênica , Histonas/metabolismo , Aprendizado de Máquina , Masculino , Camundongos , Camundongos Transgênicos , Nervo Isquiático/lesões , Análise de Sequência de RNA
12.
EMBO J ; 38(13): e101032, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31268609

RESUMO

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.


Assuntos
Gânglios Espinais/fisiologia , Histona Desacetilases/metabolismo , Traumatismos dos Nervos Periféricos/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Axônios , Células Cultivadas , Modelos Animais de Doenças , Epigênese Genética/efeitos dos fármacos , Feminino , Masculino , Camundongos , Regeneração Nervosa , Fosforilação/efeitos dos fármacos , Transdução de Sinais
13.
Sci Transl Med ; 11(487)2019 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-30971452

RESUMO

After a spinal cord injury, axons fail to regenerate in the adult mammalian central nervous system, leading to permanent deficits in sensory and motor functions. Increasing neuronal activity after an injury using electrical stimulation or rehabilitation can enhance neuronal plasticity and result in some degree of recovery; however, the underlying mechanisms remain poorly understood. We found that placing mice in an enriched environment before an injury enhanced the activity of proprioceptive dorsal root ganglion neurons, leading to a lasting increase in their regenerative potential. This effect was dependent on Creb-binding protein (Cbp)-mediated histone acetylation, which increased the expression of genes associated with the regenerative program. Intraperitoneal delivery of a small-molecule activator of Cbp at clinically relevant times promoted regeneration and sprouting of sensory and motor axons, as well as recovery of sensory and motor functions in both the mouse and rat model of spinal cord injury. Our findings showed that the increased regenerative capacity induced by enhancing neuronal activity is mediated by epigenetic reprogramming in rodent models of spinal cord injury. Understanding the mechanisms underlying activity-dependent neuronal plasticity led to the identification of potential molecular targets for improving recovery after spinal cord injury.


Assuntos
Axônios/fisiologia , Proteína de Ligação a CREB/metabolismo , Meio Ambiente , Histonas/metabolismo , Regeneração Nervosa , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/fisiopatologia , Acetilação , Animais , Cálcio/metabolismo , Modelos Animais de Doenças , Proteína p300 Associada a E1A/metabolismo , Gânglios Espinais/patologia , Gânglios Espinais/fisiopatologia , Camundongos , Neurônios Motores/patologia , Propriocepção , Recuperação de Função Fisiológica , Células Receptoras Sensoriais/patologia , Transdução de Sinais , Traumatismos da Medula Espinal/patologia
14.
PLoS One ; 11(3): e0150541, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27015635

RESUMO

Spinal cord injury (SCI) is a severe condition that affects many people and results in high health care costs. Therefore, it is essential to find new targets for treatment. The fibroblast growth factor receptor 1 (FGFR1) signalling pathway has a history of being explored for SCI treatment. Several groups have examined the effect of high availability of different FGFR1 ligands at the injury site and reported corticospinal tract (CST) regeneration as well as improved motor functions. In this study, we investigated overexpression of the FGFR1 in rat corticospinal neurons in vivo after injury (unilateral pyramidotomy) and in cerebellar granule neurons (CGNs) in vitro. We show that overexpression of FGFR1 using AAV1 intracortical injections did not increase sprouting of the treated corticospinal tract and did not improve dexterity or walking in a rat model of SCI. Furthermore, we show that overexpression of FGFR1 in vitro resulted in decreased neurite outgrowth compared to control. Thus, our results suggest that the FGFR1 is not a suitable therapeutic target after SCI.


Assuntos
Regeneração Nervosa/genética , Tratos Piramidais/crescimento & desenvolvimento , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/biossíntese , Traumatismos da Medula Espinal/genética , Animais , Axônios/metabolismo , Axônios/patologia , Modelos Animais de Doenças , Humanos , Masculino , Neurônios/metabolismo , Neurônios/patologia , Tratos Piramidais/metabolismo , Tratos Piramidais/patologia , Ratos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Transdução de Sinais , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/terapia
15.
Brain ; 139(Pt 1): 259-75, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26614754

RESUMO

There is an urgent need for a therapy that reverses disability after stroke when initiated in a time frame suitable for the majority of new victims. We show here that intramuscular delivery of neurotrophin-3 (NT3, encoded by NTF3) can induce sensorimotor recovery when treatment is initiated 24 h after stroke. Specifically, in two randomized, blinded preclinical trials, we show improved sensory and locomotor function in adult (6 months) and elderly (18 months) rats treated 24 h following cortical ischaemic stroke with human NT3 delivered using a clinically approved serotype of adeno-associated viral vector (AAV1). Importantly, AAV1-hNT3 was given in a clinically-feasible timeframe using a straightforward, targeted route (injections into disabled forelimb muscles). Magnetic resonance imaging and histology showed that recovery was not due to neuroprotection, as expected given the delayed treatment. Rather, treatment caused corticospinal axons from the less affected hemisphere to sprout in the spinal cord. This treatment is the first gene therapy that reverses disability after stroke when administered intramuscularly in an elderly body. Importantly, phase I and II clinical trials by others show that repeated, peripherally administered high doses of recombinant NT3 are safe and well tolerated in humans with other conditions. This paves the way for NT3 as a therapy for stroke.


Assuntos
Neurotrofina 3/administração & dosagem , Neurotrofina 3/uso terapêutico , Recuperação de Função Fisiológica/efeitos dos fármacos , Acidente Vascular Cerebral/tratamento farmacológico , Adenoviridae , Fatores Etários , Animais , Endotelina-1/administração & dosagem , Feminino , Vetores Genéticos/administração & dosagem , Humanos , Injeções Intramusculares , Locomoção/efeitos dos fármacos , Imageamento por Ressonância Magnética , Microinjeções , Músculo Esquelético/metabolismo , Neuroimagem , Neurotrofina 3/sangue , Neurotrofina 3/metabolismo , Tratos Piramidais/efeitos dos fármacos , Ratos , Medula Espinal/metabolismo , Acidente Vascular Cerebral/induzido quimicamente , Fatores de Tempo
16.
J Vis Exp ; (94)2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25549050

RESUMO

The corticospinal tract (CST) can be completely severed unilaterally in the medullary pyramids of the rodent brainstem. The CST is a motor tract that has great importance for distal muscle control in humans and, to a lesser extent, in rodents. A unilateral cut of one pyramid results in loss of CST innervation of the spinal cord mainly on the contralateral side of the spinal cord leading to transient motor disability in the forelimbs and sustained loss of dexterity. Ipsilateral projections of the corticospinal tract are minor. We have refined our surgical method to increase the chances of lesion completeness. We describe postsurgical care. Deficits on the Montoya staircase pellet reaching test and the horizontal ladder test shown here are detected up to 8 weeks postinjury. Deficits on the cylinder rearing test are only detected transiently. Therefore, the cylinder test may only be suitable for detection of short term recovery. We show how, electrophysiologically and anatomically, one may assess lesions and plastic changes. We also describe how to analyse fibers from the uninjured CST sprouting across the midline into the deprived areas. It is challenging to obtain >90% complete lesions consistently due to the proximity to the basilar artery in the medulla oblongata and survival rates can be low. Alternative surgical approaches and behavioural testing are described in this protocol. The pyramidotomy model is a good tool for assessing neuroplasticity-inducing treatments, which increase sprouting of intact fibers after injury.


Assuntos
Modelos Animais de Doenças , Plasticidade Neuronal/fisiologia , Tratos Piramidais/cirurgia , Ratos , Traumatismos da Medula Espinal/cirurgia , Animais , Tronco Encefálico/fisiopatologia , Denervação/métodos , Membro Anterior , Masculino
17.
Hum Gene Ther Methods ; 25(1): 14-32, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24090197

RESUMO

RNA silencing is an established method for investigating gene function and has attracted particular interest because of the potential for generating RNA-based therapeutics. Using lentiviral vectors as an efficient delivery system that offers stable, long-term expression in postmitotic cells further enhances the applicability of an RNA-based gene therapy for the CNS. In this review we provide an overview of both lentiviral vectors and RNA silencing along with design considerations for generating lentiviral vectors capable of RNA silencing. We go on to describe the current preclinical data regarding lentiviral vector-mediated RNA silencing for CNS disorders and discuss the concerns of side effects associated with lentiviral vectors and small interfering RNAs and how these might be mitigated.


Assuntos
Doenças do Sistema Nervoso Central/genética , Doenças do Sistema Nervoso Central/metabolismo , Vetores Genéticos/metabolismo , Lentivirus/genética , Interferência de RNA , Doenças do Sistema Nervoso Central/patologia , Vetores Genéticos/química , Vetores Genéticos/genética , Humanos , RNA Interferente Pequeno/metabolismo
18.
J Gene Med ; 14(5): 299-315, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22499506

RESUMO

BACKGROUND: Knocking down neuronal LINGO-1 using short hairpin RNAs (shRNAs) might enhance axon regeneration in the central nervous system (CNS). Integration-deficient lentiviral vectors have great potential as a therapeutic delivery system for CNS injuries. However, recent studies have revealed that shRNAs can induce an interferon response resulting in off-target effects and cytotoxicity. METHODS: CNS neurones were transduced with integration-deficient lentiviral vectors in vitro. The transcriptional effect of shRNA expression was analysed using quantitative real time-polymerase chain reaction and northern blots were used to assess shRNA production. RESULTS: Integration-deficient lentiviral vectors efficiently transduced CNS neurones and knocked down LINGO-1 mRNA in vitro. However, an increase in cell death was observed when lentiviral vectors encoding an shRNA were applied or when high vector concentrations were used. We demonstrate that high doses of vector or the use of vectors encoding shRNAs can induce an up-regulation of interferon-stimulated genes (2',5'-oligoadenylate synthase 1 and protein kinase R although not myxovirus resistance 1) and a down-regulation of off-target genes (including p75(NTR) and Nogo receptor 1). Furthermore, the northern blot demonstrated that these negative consequences occur even when lentiviral vectors express low levels of shRNAs. Taken together, these results may explain why neurite outgrowth was not enhanced on an inhibitory substrate following transduction with lentiviral vectors encoding an shRNA targeting LINGO-1. CONCLUSIONS: These findings highlight the importance of including appropriate controls to verify silencing specificity and the requirement to check for an interferon response when conducting RNA interference experiments. However, the potential benefits that RNA interference and viral vectors offer to gene-based therapies to CNS injuries cannot be overlooked and demand further investigation.


Assuntos
Sistema Nervoso Central , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuritos/metabolismo , RNA Interferente Pequeno/genética , Regeneração/genética , Animais , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Transferência de Genes/efeitos adversos , Vetores Genéticos , Células HEK293 , Células HeLa , Humanos , Interferons/metabolismo , Lentivirus , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/efeitos adversos , Ratos
19.
Front Mol Neurosci ; 4: 55, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22207835

RESUMO

Following an injury, central nervous system (CNS) neurons show a very limited regenerative response which results in their failure to successfully form functional connections with their original target. This is due in part to the reduced intrinsic growth state of CNS neurons, which is characterized by their failure to express key regeneration-associated genes (RAGs) and by the presence of growth inhibitory molecules in CNS environment that form a molecular and physical barrier to regeneration. Here we have optimized a 96-well electroporation and neurite outgrowth assay for postnatal rat cerebellar granule neurons (CGNs) cultured upon an inhibitory cellular substrate expressing myelin-associated glycoprotein or a mixture of growth inhibitory chondroitin sulfate proteoglycans. Optimal electroporation parameters resulted in 28% transfection efficiency and 51% viability for postnatal rat CGNs. The neurite outgrowth of transduced neurons was quantitatively measured using a semi-automated image capture and analysis system. The neurite outgrowth was significantly reduced by the inhibitory substrates which we demonstrated could be partially reversed using a Rho Kinase inhibitor. We are now using this assay to screen large sets of RAGs for their ability to increase neurite outgrowth on a variety of growth inhibitory and permissive substrates.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...