30 results on '"Thomas H. Hutson"'
Search Results
2. Enriched conditioning expands the regenerative ability of sensory neurons after spinal cord injury via neuronal intrinsic redox signaling
- Author
-
Francesco De Virgiliis, Thomas H. Hutson, Ilaria Palmisano, Sarah Amachree, Jian Miao, Luming Zhou, Rositsa Todorova, Richard Thompson, Matt C. Danzi, Vance P. Lemmon, John L. Bixby, Ilka Wittig, Ajay M. Shah, and Simone Di Giovanni
- Subjects
Science - Abstract
Pre conditioning injury or environmental enrichment have been shown to promote axon regeneration. Here the authors show that environmental enrichment, combined with preconditioning injury promotes regeneration via a redox signalling dependent mechanism.
- Published
- 2020
- Full Text
- View/download PDF
3. Wireless closed-loop optogenetics across the entire dorsoventral spinal cord in mice
- Author
-
Sadaf Soloukey, Andreas Rowald, Claudia Kathe, Ivan Furfaro, Philipp Schonle, Valentina Paggi, Quentin Barraud, Stéphanie P. Lacour, Jimmy Ravier, Qiuting Huang, Kyungjin Kim, Thomas H. Hutson, Ileana O. Jelescu, Antoine Philippides, Noaf Salah Ali Alwahab, Chris I. De Zeeuw, Daniel Huber, Frédéric Michoud, Leonie Asboth, Noe Brun, Jerome Gandar, Grégoire Courtine, Neurosciences, and Netherlands Institute for Neuroscience (NIN)
- Subjects
Opsin ,optoelectronics ,Dura mater ,brain ,Biomedical Engineering ,Bioengineering ,Sensory system ,Optogenetics ,Spinal cord ,Closed-loop system ,Integrated circuit ,Wireless sensor & stimulation node ,Biology ,Applied Microbiology and Biotechnology ,Photostimulation ,Mice ,circuit reorganization ,medicine ,Animals ,Wireless ,locomotor recovery ,Neurons ,business.industry ,medicine.anatomical_structure ,Spinal Cord ,Molecular Medicine ,business ,Wireless Technology ,Closed loop ,Neuroscience ,Biotechnology - Abstract
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., Optogenetics is applied to the entire mouse spinal cord.
- Published
- 2022
- Full Text
- View/download PDF
4. Overexpression of the Fibroblast Growth Factor Receptor 1 (FGFR1) in a Model of Spinal Cord Injury in Rats.
- Author
-
Barbara Haenzi, Katharina Gers-Barlag, Halima Akhoundzadeh, Thomas H Hutson, Sean C Menezes, Mary Bartlett Bunge, and Lawrence D F Moon
- Subjects
Medicine ,Science - Abstract
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.
- Published
- 2016
- Full Text
- View/download PDF
5. AMPK controls the axonal regenerative ability of dorsal root ganglia sensory neurons after spinal cord injury
- Author
-
Simone Di Giovanni, Eilidh McLachlan, Ilaria Palmisano, Radhika Puttagunta, Paolo La Montanara, Kirill Shkura, Thomas H. Hutson, Guiping Kong, Luming Zhou, Elisabeth Serger, Francesco De Virgiliis, Anja Freiwald, International Spinal Research Trust, Wings for Life Spinal Cord Research Foundation, and The Weizmann Institute of Science
- Subjects
Endocrinology, Diabetes and Metabolism ,Regeneration (biology) ,medicine.medical_treatment ,Central nervous system ,AMPK ,Cell Biology ,Biology ,medicine.disease ,medicine.anatomical_structure ,Dorsal root ganglion ,Axoplasm ,nervous system ,Physiology (medical) ,Peripheral nervous system ,Internal Medicine ,medicine ,Axotomy ,Spinal cord injury ,Neuroscience - 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.
- Published
- 2020
6. Cyclin-dependent-like kinase 5 is required for pain signaling in human sensory neurons and mouse models
- Author
-
Nikos Gorgoraptis, Kingsley Wong, Arnau Hervera, Sila K. Ultanir, Guiping Kong, Lucas L. Baltussen, Francesco De Virgiliis, Hongwei Yu, Yunan Gao, Jenny Downs, Jessica Chadwick, Paolo La Montanara, Tommaso Pizzorusso, Simone Di Giovanni, Qasim A. Majid, Helen Leonard, Thomas H. Hutson, Nagy Istvan, Ilaria Palmisano, David Stuart Millar, Nicholas D. Mazarakis, Katalin Bartus, Imperial College Healthcare NHS Trust- BRC Funding, National Institute for Health Research, La Montanara, Paolo, Hervera, Arnau, Baltussen, Lucas L, Hutson, Thomas H, Palmisano, Ilaria, De Virgiliis, Francesco, Kong, Guiping, Chadwick, Jessica, Gao, Yunan, Bartus, Katalin, Majid, Qasim A, Gorgoraptis, Niko, Wong, Kingsley, Downs, Jenny, Pizzorusso, Tommaso, Ultanir, Sila K, Leonard, Helen, Yu, Hongwei, Millar, David S, Istvan, Nagy, Mazarakis, Nicholas D, and Di Giovanni, Simone
- Subjects
CDKL5 ,CHILDREN ,CYTOSKELETON ,Gene mutation ,Research & Experimental Medicine ,Settore BIO/09 - Fisiologia ,Transient receptor potential channel ,Mice ,animal ,pain ,Axon ,11 Medical and Health Sciences ,disease models, animal ,protein serine-threonine kinase ,General Medicine ,AXON ,medicine.anatomical_structure ,Nociception ,Medicine, Research & Experimental ,Nociceptor ,GROWTH ,Life Sciences & Biomedicine ,signal transduction ,Signal Transduction ,CAMKII ,mice ,Sensory Receptor Cells ,TRPV1 ,Pain ,PHENOTYPES ,Biology ,Protein Serine-Threonine Kinases ,Article ,cyclin ,Ca2+/calmodulin-dependent protein kinase ,Cyclins ,CYTOPLASMIC DYNEIN ,medicine ,Animals ,Humans ,human ,Science & Technology ,sensory receptor cell ,Cell Biology ,06 Biological Sciences ,RETT-SYNDROME ,TRANSPORT ,Disease Models, Animal ,nervous system ,Neuroscience - Abstract
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.
- Published
- 2020
7. Cell type prioritization in single-cell data
- Author
-
Michael A. Skinnider, Jordan W. Squair, Aaron A. Phillips, Matthieu Gautier, Gioele La Manno, Leonard J. Foster, Kaya J.E. Matson, Thomas H. Hutson, Quentin Barraud, Grégoire Courtine, Ariel J. Levine, Marco Milano, Mark Anderson, and Claudia Kathe
- Subjects
Prioritization ,noise ,Cell type ,architecture ,Computer science ,medicine.medical_treatment ,defines ,enables ,Cell ,Biomedical Engineering ,Bioengineering ,Walking ,Computational biology ,Applied Microbiology and Biotechnology ,Article ,Machine Learning ,Transcriptome ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Databases, Genetic ,Biological neural network ,medicine ,Animals ,atlas ,Neurostimulation ,030304 developmental biology ,0303 health sciences ,Sequence Analysis, RNA ,Gene Expression Profiling ,Computational Biology ,Spinal cord ,gene-expression ,Chromatin ,Rats ,medicine.anatomical_structure ,Molecular Medicine ,Nerve Net ,Single-Cell Analysis ,Neuroscience ,030217 neurology & neurosurgery ,Biotechnology - Abstract
The cell types affected by biological perturbations in complex tissues are uncovered by single-cell analysis., 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.
- Published
- 2019
- Full Text
- View/download PDF
8. Reactive oxygen species regulate axonal regeneration through the release of exosomal NADPH oxidase 2 complexes into injured axons
- Author
-
Celio X.C. Santos, Matt C. Danzi, Francesco De Virgiliis, Thomas H. Hutson, Arnau Hervera, Elena Tantardini, José Antonio del Río, Mike Fainzilber, Roland A. Fleck, John L. Bixby, Ajay M. Shah, Rotem Ben-Tov Perry, Vance Lemmon, Alexander N. Kapustin, Thomas L. Carroll, Simone Di Giovanni, Luming Zhou, Ilaria Palmisano, and Guiping Kong
- Subjects
0301 basic medicine ,Endosome ,CX3C Chemokine Receptor 1 ,Endosomes ,Exosomes ,Endocytosis ,Exocytosis ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Peripheral Nerve Injuries ,Ganglia, Spinal ,Animals ,Spinal Cord Injuries ,Mice, Knockout ,chemistry.chemical_classification ,Reactive oxygen species ,NADPH oxidase ,biology ,Chemistry ,Macrophages ,Regeneration (biology) ,PTEN Phosphohydrolase ,Dyneins ,Nuclear Proteins ,Cell Biology ,beta Karyopherins ,Sciatic Nerve ,Axons ,Nerve Regeneration ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,NADPH Oxidase 2 ,Nerve Degeneration ,biology.protein ,Beta Karyopherins ,Sciatic nerve ,Phosphatidylinositol 3-Kinase ,Reactive Oxygen Species ,Proto-Oncogene Proteins c-akt ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Reactive oxygen species (ROS) contribute to tissue damage and remodelling mediated by the inflammatory response after injury. Here we show that ROS, which promote axonal dieback and degeneration after injury, are also required for axonal regeneration and functional recovery after spinal injury. We find that ROS production in the injured sciatic nerve and dorsal root ganglia requires CX3CR1-dependent recruitment of inflammatory cells. Next, exosomes containing functional NADPH oxidase 2 complexes are released from macrophages and incorporated into injured axons via endocytosis. Once in axonal endosomes, active NOX2 is retrogradely transported to the cell body through an importin-β1-dynein-dependent mechanism. Endosomal NOX2 oxidizes PTEN, which leads to its inactivation, thus stimulating PI3K-phosporylated (p-)Akt signalling and regenerative outgrowth. Challenging the view that ROS are exclusively involved in nerve degeneration, we propose a previously unrecognized role of ROS in mammalian axonal regeneration through a NOX2-PI3K-p-Akt signalling pathway.
- Published
- 2018
- Full Text
- View/download PDF
9. The translational landscape in spinal cord injury: focus on neuroplasticity and regeneration
- Author
-
Thomas H, Hutson and Simone, Di Giovanni
- Subjects
Translational Research, Biomedical ,Neuronal Plasticity ,Animals ,Humans ,Recovery of Function ,Axons ,Spinal Cord Injuries ,Nerve Regeneration - Abstract
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.
- Published
- 2019
10. The translational landscape in spinal cord injury: focus on neuroplasticity and regeneration
- Author
-
Thomas H. Hutson, Simone Di Giovanni, Wings for Life Spinal Cord Research Foundation, Rosetrees Trust, and Brain Research UK
- Subjects
0301 basic medicine ,Clinical Neurology ,CORTICOSPINAL TRACT REGENERATION ,PROMOTES FUNCTIONAL RECOVERY ,RHO-ASSOCIATED KINASE ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Neuroplasticity ,medicine ,Biological neural network ,Premovement neuronal activity ,Axon ,Spinal cord injury ,MOTOR CORTEX ,ANTI-NOGO-A ,AXON REGENERATION ,Science & Technology ,Neurology & Neurosurgery ,business.industry ,Regeneration (biology) ,CENTRAL-NERVOUS-SYSTEM ,STEM-CELL TRANSPLANTATION ,1103 Clinical Sciences ,medicine.disease ,Spinal cord ,CIRCUIT REORGANIZATION ,Neural stem cell ,1113 Opthalmology and Optometry ,030104 developmental biology ,medicine.anatomical_structure ,CHONDROITINASE ABC ,Neurology (clinical) ,Neurosciences & Neurology ,business ,1109 Neurosciences ,Neuroscience ,Life Sciences & Biomedicine ,030217 neurology & neurosurgery - Abstract
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. Spinal cord injury leads to disruption of neural circuitry and connectivity, resulting in permanent neurological disability. Hutson and Di Giovanni assess the clinical potential of emerging strategies that are designed to augment neuroplasticity and promote sensorimotor recovery after spinal cord injury.
- Published
- 2019
11. PP4‐dependent HDAC3 dephosphorylation discriminates between axonal regeneration and regenerative failure
- Author
-
José Antonio del Río, Radhika Puttagunta, John L. Bixby, Matt C. Danzi, Guiping Kong, Ilaria Palmisano, Dina P. Matheos, Thomas H. Hutson, Simone Di Giovanni, Andreu Matamoros-Angles, Kirsi Forsberg, Janine L. Kwapis, Vance Lemmon, Eilidh McLachlan, Luming Zhou, Marcelo A. Wood, Francesco De Virgiliis, Arnau Hervera, Wings for Life Spinal Cord Research Foundation, Rosetrees Trust, and The Henry Smith Charity
- Subjects
Male ,GAP-43 EXPRESSION ,Neurodegenerative ,Regenerative Medicine ,Medical and Health Sciences ,Epigenesis, Genetic ,Mice ,0302 clinical medicine ,Injury - Trauma - (Head and Spine) ,Peripheral Nerve Injuries ,Ganglia, Spinal ,Gene expression ,Phosphoprotein Phosphatases ,2.1 Biological and endogenous factors ,NERVE ,Aetiology ,Phosphorylation ,nerve regeneration ,Spinal Cord Injury ,NEURONS ,Spinal cord injury ,11 Medical and Health Sciences ,Cells, Cultured ,0303 health sciences ,Cultured ,General Neuroscience ,Articles ,Biological Sciences ,Cell biology ,Histone ,Neurological ,Peripheral nerve injury ,GROWTH ,Female ,Signal transduction ,transcription ,Life Sciences & Biomedicine ,Biotechnology ,Signal Transduction ,Biochemistry & Molecular Biology ,Physical Injury - Accidents and Adverse Effects ,Spinal ,1.1 Normal biological development and functioning ,Cells ,INHIBITION ,Biology ,NEURITE OUTGROWTH ,General Biochemistry, Genetics and Molecular Biology ,Histone Deacetylases ,Dephosphorylation ,Small Molecule Libraries ,03 medical and health sciences ,Genetic ,Underpinning research ,Information and Computing Sciences ,Genetics ,INJURY ,medicine ,Animals ,Epigenetics ,Molecular Biology ,Traumatic Head and Spine Injury ,030304 developmental biology ,P53 ,Science & Technology ,calcium ,General Immunology and Microbiology ,Animal ,Regeneration (biology) ,Neurosciences ,HDAC3 ,Cell Biology ,06 Biological Sciences ,medicine.disease ,spinal cord injury ,Axons ,Nerve Regeneration ,Disease Models, Animal ,Disease Models ,Injury (total) Accidents/Adverse Effects ,biology.protein ,Ganglia ,08 Information and Computing Sciences ,030217 neurology & neurosurgery ,Epigenesis ,Developmental Biology - 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.
- Published
- 2019
12. Trans-neuronal transduction of spinal neurons following cortical injection and anterograde axonal transport of a bicistronic AAV1 vector
- Author
-
Thomas H. Hutson, Claudia Kathe, and Lawrence D. F. Moon
- Subjects
0301 basic medicine ,viruses ,Central nervous system ,Gene delivery ,Biology ,Bioinformatics ,Axonal Transport ,Article ,03 medical and health sciences ,Transduction (genetics) ,Transduction, Genetic ,Genetics ,medicine ,Animals ,Molecular Biology ,Neurons ,Reporter gene ,Gene Transfer Techniques ,Dependovirus ,Spinal cord ,Anterograde axonal transport ,Rats ,030104 developmental biology ,medicine.anatomical_structure ,Spinal Cord ,nervous system ,Corticospinal tract ,Axoplasmic transport ,Molecular Medicine ,Female ,Sensorimotor Cortex ,Neuroscience - Abstract
Adeno-associated viral (AAV) vectors are one of the most promising gene delivery systems to the central nervous system. We now report, that AAV1 can be used to express transgenes trans-neuronally in neurons distant from the injection site. Specifically, intracortical injection of a bicistronic AAV1 vector trans-neuronally transduced spinal neurons as shown by fluorescence microscopy, the presence of AAV genome and AAV transcript in the contralateral spinal cord. Prior pyramidotomy abolished spinal transduction, confirming anterograde axonal transport of AAV1 in the corticospinal tract. These observations demonstrate the potential of bicistronic AAV1 for trans-neuronal expression of therapeutic transgenes in neurological disorders or reporter genes in connectivity studies.
- Published
- 2015
- Full Text
- View/download PDF
13. Epigenomic signatures underpin the axonal regenerative ability of dorsal root ganglia sensory neurons
- Author
-
Miroslav Kubat, Elisabeth Serger, Evan Elliott, Matthias Merkenschlager, John L. Bixby, Thomas H. Hutson, Hassen Dhrif, Prashant K. Srivastava, Vance Lemmon, Kirill Shkura, Arnau Hervera, Stefan Wuchty, Tong Liu, Simone Di Giovanni, Nick O’ Neill, Zheng Wang, Matt C. Danzi, Ilaria Palmisano, Liron Levi, Luming Zhou, Eilidh McLachlan, Wings for Life Spinal Cord Research Foundation, Rosetrees Trust, Wellcome Trust, and Medical Research Council (MRC)
- Subjects
0301 basic medicine ,Epigenomics ,Male ,CCCTC-Binding Factor ,Sensory Receptor Cells ,medicine.medical_treatment ,1702 Cognitive Sciences ,Gene Expression ,Sensory system ,Mice, Transgenic ,Histones ,Machine Learning ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Ganglia, Spinal ,medicine ,Animals ,Epigenetics ,Axon ,Neurology & Neurosurgery ,biology ,Sequence Analysis, RNA ,General Neuroscience ,Acetylation ,Sciatic Nerve ,Axons ,Chromatin ,Nerve Regeneration ,030104 developmental biology ,Histone ,medicine.anatomical_structure ,1701 Psychology ,biology.protein ,Female ,Axotomy ,1109 Neurosciences ,Neuroscience ,Chromatin immunoprecipitation ,030217 neurology & neurosurgery ,Algorithms - Abstract
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. This manuscript describes the systematic investigation of epigenomic signatures discriminating between regenerative success and failure in dorsal root ganglia sensory neurons following axonal injury. This epigenomic map offers a tool to design novel approaches for neuronal repair.
- Published
- 2018
14. Lentiviral Vector-Mediated RNA Silencing in the Central Nervous System
- Author
-
Thomas H. Hutson, Edmund Foster, Rafael J. Yáñez-Muñoz, and Lawrence D. F. Moon
- Subjects
Pharmacology ,Small interfering RNA ,Genetic enhancement ,Genetic Vectors ,Lentivirus ,RNA ,Biology ,Applied Microbiology and Biotechnology ,Virology ,Cell biology ,Viral vector ,Small hairpin RNA ,RNA silencing ,Central Nervous System Diseases ,RNA interference ,Genetics ,Humans ,Molecular Medicine ,RNA Interference ,RNA, Small Interfering ,Gene ,Research Articles ,Genetics (clinical) - Abstract
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.
- Published
- 2014
- Full Text
- View/download PDF
15. Intramuscular Neurotrophin-3 normalizes low threshold spinal reflexes, reduces spasms and improves mobility after bilateral corticospinal tract injury in rats
- Author
-
Thomas H. Hutson, Stephen B. McMahon, Lawrence D. F. Moon, and Claudia Kathe
- Subjects
0301 basic medicine ,Spasm ,central nervous system injury ,Pyramidal Tracts ,neurotrophins ,0302 clinical medicine ,Neurotrophin 3 ,Biology (General) ,Spinal cord injury ,biology ,General Neuroscience ,spasticity ,General Medicine ,Recombinant Proteins ,3. Good health ,Treatment Outcome ,medicine.anatomical_structure ,Medicine ,movement disorder ,medicine.symptom ,Locomotion ,Research Article ,Neurotrophin ,medicine.medical_specialty ,QH301-705.5 ,Science ,Neurotrophin-3 ,Injections, Intramuscular ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Physical medicine and rehabilitation ,medicine ,Animals ,Nerve Growth Factors ,Spasticity ,Reflex, Abnormal ,General Immunology and Microbiology ,business.industry ,Genetic Therapy ,Spinal cord ,medicine.disease ,Rats ,Disease Models, Animal ,030104 developmental biology ,nervous system ,Corticospinal tract ,Reflex ,biology.protein ,Rat ,Forelimb ,business ,030217 neurology & neurosurgery ,Neuroscience - Abstract
Brain and spinal injury reduce mobility and often impair sensorimotor processing in the spinal cord leading to spasticity. Here, we establish that complete transection of corticospinal pathways in the pyramids impairs locomotion and leads to increased spasms and excessive mono- and polysynaptic low threshold spinal reflexes in rats. Treatment of affected forelimb muscles with an adeno-associated viral vector (AAV) encoding human Neurotrophin-3 at a clinically-feasible time-point after injury reduced spasticity. Neurotrophin-3 normalized the short latency Hoffmann reflex to a treated hand muscle as well as low threshold polysynaptic spinal reflexes involving afferents from other treated muscles. Neurotrophin-3 also enhanced locomotor recovery. Furthermore, the balance of inhibitory and excitatory boutons in the spinal cord and the level of an ion co-transporter in motor neuron membranes required for normal reflexes were normalized. Our findings pave the way for Neurotrophin-3 as a therapy that treats the underlying causes of spasticity and not only its symptoms. DOI: http://dx.doi.org/10.7554/eLife.18146.001, eLife digest Injuries to the brain and spinal cord cause disability in millions of people worldwide. Physical rehabilitation can restore some muscle control and improve mobility in affected individuals. However, no current treatments provide long-term relief from the unwanted muscle contractions and spasms that affect as many as 78% of people with a spinal cord injury. These spasms can seriously hamper a person’s ability to carry out day-to-day tasks and get around independently. A few treatments can help in the short term but have side effects; indeed while Botox injections are used to paralyse the muscle, these also reduce the chances of useful improvements. As such, better therapies for muscle spasms are needed; especially ones that reduce spasms in the arms. Rats with injuries to the spinal cord between their middle to lower back typically develop spasms in their legs or tail, and rat models have helped scientists begin to understand why these involuntary movements occur. Now, Kathe et al. report that cutting one specific pathway that connects the brain to the spinal cord in anesthetised rats leads to the development of spasms in the forelimbs as well. Several months after the surgery, the rats had spontaneous muscle contractions in their forelimbs and walked abnormally. Further experiments showed that some other neural pathways in the rats became incorrectly wired and hyperactive and that this resulted in the abnormal movements. Next, Kathe et al. asked whether using gene therapy to deliver a protein that is required for neural circuits to form between muscles and the spinal cord (called neurotrophin-3) would stop the involuntary movements in the forelimbs. Delivering the gene therapy directly into the forelimb muscles of the disabled rats a day after their injury increased the levels of neurotrophin-3 in these muscles. Rats that received this treatment had fewer spasms and walked better than those that did not. Further experiments confirmed that this was because the rats’ previously hyperactive and abnormally wired neural circuits became more normal after the treatment. Together these results suggest that neurotrophin-3 might be a useful treatment for muscle spasms in people with spinal injury. There have already been preliminary studies in people showing that treatment with neurotrophin-3 is safe and well tolerated. Future studies are needed to confirm that it could be useful in humans. DOI: http://dx.doi.org/10.7554/eLife.18146.002
- Published
- 2016
- Full Text
- View/download PDF
16. The Adaptor Protein CD2AP Is a Coordinator of Neurotrophin Signaling-Mediated Axon Arbor Plasticity
- Author
-
Jeffrey C. Petruska, Kristofer K. Rau, Fred H. Gage, Benjamin J. Harrison, Caitlin E. Hill, Cassa Drury, Richard D. Johnson, Gayathri Venkat, Thomas H. Hutson, Lorne M. Mendell, Eric C. Rouchka, James L. Lamb, Mary Barlett Bunge, and Lawrence D. F. Moon
- Subjects
Male ,0301 basic medicine ,Neurite ,MAP Kinase Signaling System ,Cellular differentiation ,Endosomes ,Rats, Sprague-Dawley ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Nerve Growth Factors ,Pseudopodia ,RNA, Messenger ,RNA, Small Interfering ,Receptor, trkA ,Axon ,Growth cone ,Adaptor Proteins, Signal Transducing ,Neuronal Plasticity ,biology ,General Neuroscience ,Signal transducing adaptor protein ,Cell Differentiation ,Articles ,Collateral sprouting ,Axons ,Rats ,Cell biology ,Class Ia Phosphatidylinositol 3-Kinase ,Mice, Inbred C57BL ,Cytoskeletal Proteins ,030104 developmental biology ,medicine.anatomical_structure ,Nerve growth factor ,nervous system ,biology.protein ,Female ,Neuroscience ,030217 neurology & neurosurgery ,Signal Transduction ,Neurotrophin - Abstract
Growth of intact axons of noninjured neurons, often termed collateral sprouting, contributes to both adaptive and pathological plasticity in the adult nervous system, but the intracellular factors controlling this growth are largely unknown. An automated functional assay of genes regulated in sensory neurons from the ratin vivospared dermatome model of collateral sprouting identified the adaptor protein CD2-associated protein (CD2AP; human CMS) as a positive regulator of axon growth. In non-neuronal cells, CD2AP, like other adaptor proteins, functions to selectively control the spatial/temporal assembly of multiprotein complexes that transmit intracellular signals. Although CD2AP polymorphisms are associated with increased risk of late-onset Alzheimer's disease, its role in axon growth is unknown. Assessments of neurite arbor structurein vitrorevealed CD2AP overexpression, and siRNA-mediated knockdown, modulated (1) neurite length, (2) neurite complexity, and (3) growth cone filopodia number, in accordance with CD2AP expression levels. We show, for the first time, that CD2AP forms a novel multiprotein complex with the NGF receptor TrkA and the PI3K regulatory subunit p85, with the degree of TrkA:p85 association positively regulated by CD2AP levels. CD2AP also regulates NGF signaling through AKT, but not ERK, and regulates long-range signaling though TrkA+/RAB5+signaling endosomes. CD2AP mRNA and protein levels were increased in neurons during collateral sprouting but decreased following injury, suggesting that, although typically considered together, these two adult axonal growth processes are fundamentally different. These data position CD2AP as a major intracellular signaling molecule coordinating NGF signaling to regulate collateral sprouting and structural plasticity of intact adult axons.SIGNIFICANCE STATEMENTGrowth of noninjured axons in the adult nervous system contributes to adaptive and maladaptive plasticity, and dysfunction of this process may contribute to neurologic pathologies. Functional screening of genes regulated during growth of noninjured axons revealed CD2AP as a positive regulator of axon outgrowth. A novel association of CD2AP with TrkA and p85 suggests a distinct intracellular signaling pathway regulating growth of noninjured axons. This may also represent a novel mechanism of generating specificity in multifunctional NGF signaling. Divergent regulation of CD2AP in different axon growth conditions suggests that separate mechanisms exist for different modes of axon growth. CD2AP is the first signaling molecule associated with adult sensory axonal collateral sprouting, and this association may offer new insights for NGF/TrkA-related Alzheimer's disease mechanisms.
- Published
- 2016
- Full Text
- View/download PDF
17. Overexpression of the Fibroblast Growth Factor Receptor 1 (FGFR1) in a Model of Spinal Cord Injury in Rats
- Author
-
Halima Akhoundzadeh, Lawrence D. F. Moon, Barbara Haenzi, Sean Christopher Menezes, Mary Bartlett Bunge, Thomas H. Hutson, and Katharina Gers-Barlag
- Subjects
Male ,0301 basic medicine ,Critical Care and Emergency Medicine ,Fibroblast Growth Factor ,Physiology ,Pyramidal Tracts ,lcsh:Medicine ,Artificial Gene Amplification and Extension ,Fibroblast growth factor ,Polymerase Chain Reaction ,Mechanical Treatment of Specimens ,Endocrinology ,Nerve Fibers ,Animal Cells ,Medicine and Health Sciences ,Spinal Cord Injury ,lcsh:Science ,Spinal cord injury ,Trauma Medicine ,Neurons ,Multidisciplinary ,Anatomy ,Electroporation ,medicine.anatomical_structure ,Neurology ,Specimen Disruption ,Hyperexpression Techniques ,Cellular Types ,Signal transduction ,Traumatic Injury ,Research Article ,Signal Transduction ,medicine.medical_specialty ,Neurite ,Research and Analysis Methods ,03 medical and health sciences ,In vivo ,Growth Factors ,Internal medicine ,Neurites ,Gene Expression and Vector Techniques ,medicine ,Animals ,Humans ,Receptor, Fibroblast Growth Factor, Type 1 ,Molecular Biology Techniques ,Molecular Biology ,Spinal Cord Injuries ,Molecular Biology Assays and Analysis Techniques ,Pyramidal tracts ,Endocrine Physiology ,business.industry ,Fibroblast growth factor receptor 1 ,lcsh:R ,Biology and Life Sciences ,Cell Biology ,Neuronal Dendrites ,medicine.disease ,Axons ,Nerve Regeneration ,Rats ,Disease Models, Animal ,stomatognathic diseases ,030104 developmental biology ,Specimen Preparation and Treatment ,Cellular Neuroscience ,Corticospinal tract ,lcsh:Q ,business ,Neuroscience - Abstract
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.
- Published
- 2016
- Full Text
- View/download PDF
18. Efficient gene expression from integration-deficient lentiviral vectors in the spinal cord
- Author
-
G Olmos, Hugo Peluffo, V. Caraballo-Miralles, Sherif G. Ahmed, Rafael J. Yáñez-Muñoz, Lawrence D. F. Moon, J. Llado, Klaus Wanisch, Thomas H. Hutson, Ping K. Yip, Stephen B. McMahon, Edmund Foster, and N Lago
- Subjects
Virus Integration ,Genetic Vectors ,Gene Expression ,Mice ,03 medical and health sciences ,Transduction (genetics) ,0302 clinical medicine ,Neurotrophic factors ,Genetics ,medicine ,Glial cell line-derived neurotrophic factor ,Animals ,Humans ,Molecular Biology ,Spinal cord injury ,Spinal Cord Injuries ,030304 developmental biology ,0303 health sciences ,biology ,Amyotrophic Lateral Sclerosis ,Lentivirus ,Gene Transfer Techniques ,Spinal muscular atrophy ,Motor neuron ,medicine.disease ,Spinal cord ,Molecular biology ,Rats ,Cell biology ,Muscular Atrophy ,Mutagenesis, Insertional ,medicine.anatomical_structure ,Spinal Cord ,nervous system ,GDF7 ,biology.protein ,Molecular Medicine ,030217 neurology & neurosurgery - Abstract
Gene transfer to spinal cord cells may be crucial for therapy in spinal muscular atrophy, amyotrophic lateral sclerosis and spinal cord injury. Lentiviral vectors are efficient for transduction of a variety of cells, but like all integrating vectors they pose a risk of insertional mutagenesis. Integration-deficient lentiviral vectors (IDLVs) remain episomal but retain the transduction efficiency of standard integrating lentiviral vectors, particularly when the episomes are not diluted out through repeated cell division. We have now applied IDLVs for transduction of spinal cord in vitro, in explants and in vivo. Our results demonstrate similar efficiency of eGFP expression from integrating lentiviral vectors and IDLVs in most cell types analyzed, including motor neurons, interneurons, dorsal root ganglia (DRG) neurons and astroglia. IDLV-mediated expression of pro-glial-cell-derived neurotrophic factor (Gdnf) rescues motor neuron cultures from death caused by removal of exogenous trophic support. IDLVs also mediate efficient RNA interference in DRG neuron cultures. After intraparenchymal injection in the rat and mouse cervical and lumbar regions in vivo, transduction is mainly neuronal, with both motor neurons and interneurons being efficiently targeted. These results suggest that IDLVs could be efficient and safer tools for spinal cord transduction in future therapeutic strategies.
- Published
- 2012
- Full Text
- View/download PDF
19. Lentiviral vectors encoding short hairpin RNAs efficiently transduce and knockdown LINGO-1 but induce an interferon response and cytotoxicity in central nervous system neurones
- Author
-
Thomas H. Hutson, John M. Dawes, Lawrence D. F. Moon, Robert Hindges, Rafael J. Yáñez-Muñoz, and Edmund Foster
- Subjects
0303 health sciences ,Gene knockdown ,Biology ,Protein kinase R ,Molecular biology ,Viral vector ,Cell biology ,Small hairpin RNA ,03 medical and health sciences ,Transduction (genetics) ,0302 clinical medicine ,Interferon ,RNA interference ,Drug Discovery ,Genetics ,medicine ,Molecular Medicine ,Gene silencing ,Molecular Biology ,030217 neurology & neurosurgery ,Genetics (clinical) ,030304 developmental biology ,medicine.drug - Abstract
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 p75NTR 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. Copyright © 2012 John Wiley & Sons, Ltd.
- Published
- 2012
- Full Text
- View/download PDF
20. Transcriptional changes in sensory ganglia associated with primary afferent axon collateral sprouting in spared dermatome model
- Author
-
Lawrence D. F. Moon, Gayathri Venkat, Mary Bartlett Bunge, Lorne M. Mendell, Kristofer K. Rau, Caitlin E. Hill, Fred H. Gage, Jeffrey C. Petruska, Thomas H. Hutson, Eric C. Rouchka, Benjamin J. Harrison, and Richard D. Johnson
- Subjects
Nerve injury ,lcsh:QH426-470 ,Pain ,Sensory system ,Biology ,Stimulus (physiology) ,Biochemistry ,Data in Brief ,Genetics ,medicine ,Axon ,Transcriptomics ,Spinal cord injury ,Denervation ,Anatomy ,Collateral sprouting ,medicine.disease ,Axon growth ,lcsh:Genetics ,Axonal plasticity ,medicine.anatomical_structure ,Dermatome ,Molecular Medicine ,medicine.symptom ,Neuroscience ,Biotechnology - Abstract
Primary afferent collateral sprouting is a process whereby non-injured primary afferent neurons respond to some stimulus and extend new branches from existing axons. Neurons of both the central and peripheral nervous systems undergo this process, which contributes to both adaptive and maladaptive plasticity (e.g., [1], [2], [3], [4], [5], [6], [7], [8], [9]). In the model used here (the "spared dermatome" model), the intact sensory neurons respond to the denervation of adjacent areas of skin by sprouting new axon branches into that adjacent denervated territory. Investigations of gene expression changes associated with collateral sprouting can provide a better understanding of the molecular mechanisms controlling this process. Consequently, it can be used to develop treatments to promote functional recovery for spinal cord injury and other similar conditions. This report includes raw gene expression data files from microarray experiments in order to study the gene regulation in spared sensory ganglia in the initiation (7 days) and maintenance (14 days) phases of the spared dermatome model relative to intact ("naïve") sensory ganglia. Data has been deposited into GEO (GSE72551).
- Published
- 2015
- Full Text
- View/download PDF
21. Corticospinal tract transduction: a comparison of seven adeno-associated viral vector serotypes and a non-integrating lentiviral vector
- Author
-
Joost Verhaagen, Rafael J. Yáñez-Muñoz, Lawrence D. F. Moon, Thomas H. Hutson, and Netherlands Institute for Neuroscience (NIN)
- Subjects
Genetic enhancement ,Virus Integration ,Spinal cord Injury ,Central nervous system ,Genetic Vectors ,Green Fluorescent Proteins ,Pyramidal Tracts ,Cell Count ,Integration-deficient lentiviral vector ,Biology ,Transfection ,Article ,Viral vector ,Rats, Sprague-Dawley ,03 medical and health sciences ,Transduction (genetics) ,Transduction ,0302 clinical medicine ,Gene therapy ,Genetics ,medicine ,Animals ,Humans ,Serotyping ,Molecular Biology ,Tropism ,030304 developmental biology ,Neurons ,0303 health sciences ,Pyramidal tracts ,Lentivirus ,Gene Transfer Techniques ,AAV ,Genetic Therapy ,Dependovirus ,Virology ,Corticospinal tract ,Cell biology ,Rats ,medicine.anatomical_structure ,HEK293 Cells ,Astrocytes ,Molecular Medicine ,Microglia ,030217 neurology & neurosurgery ,Cellular Tropism - Abstract
The corticospinal tract (CST) is extensively used as a model system for assessing potential therapies to enhance neuronal regeneration and functional recovery following spinal cord injury (SCI). However, efficient transduction of the CST is challenging and remains to be optimised. Recombinant adeno-associated viral (AAV) vectors and integration-deficient lentiviral vectors are promising therapeutic delivery systems for gene therapy to the central nervous system (CNS). In the present study the cellular tropism and transduction efficiency of seven AAV vector serotypes (AAV1, 2, 3, 4, 5, 6, 8) and an integration-deficient lentiviral vector were assessed for their ability to transduce corticospinal neurons (CSNs) following intracortical injection. AAV1 was identified as the optimal serotype for transducing cortical and CSNs with green fluorescent protein (GFP) expression detectable in fibres projecting through the dorsal corticospinal tract (dCST) of the cervical spinal cord. In contrast, AAV3 and AAV4 demonstrated a low efficacy for transducing CNS cells and AAV8 presented a potential tropism for oligodendrocytes. Furthermore, it was shown that neither AAV nor lentiviral vectors generate a significant microglial response. The identification of AAV1 as the optimal serotype for transducing CSNs should facilitate the design of future gene therapy strategies targeting the CST for the treatment of SCI.
- Published
- 2011
- Full Text
- View/download PDF
22. Publisher Correction: Reactive oxygen species regulate axonal regeneration through the release of exosomal NADPH oxidase 2 complexes into injured axons
- Author
-
Guiping Kong, José Antonio del Río, Vance Lemmon, Francesco De Virgiliis, Ajay M. Shah, Luming Zhou, Rotem Ben-Tov Perry, Arnau Hervera, Roland A. Fleck, Thomas H. Hutson, Matt C. Danzi, Elena Tantardini, John L. Bixby, Simone Di Giovanni, Ilaria Palmisano, Alexander N. Kapustin, Thomas L. Carroll, Celio X.C. Santos, and Mike Fainzilber
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,03 medical and health sciences ,Reactive oxygen species ,030104 developmental biology ,NADPH oxidase ,biology ,chemistry ,Regeneration (biology) ,biology.protein ,Cell Biology ,Microbiology ,Cell biology - Abstract
In the version of this Article originally published, the affiliations for Roland A. Fleck and Jose Antonio Del Rio were incorrect due to a technical error that resulted in affiliations 8 and 9 being switched. The correct affiliations are: Roland A. Fleck: 8Centre for Ultrastructural Imaging, Kings College London, London, UK. Jose Antonio Del Rio: 2Cellular and Molecular Neurobiotechnology, Institute for Bioengineering of Catalonia, Barcelona, Spain; 9Department of Cell Biology, Physiology and Immunology, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; 10Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain. This has now been amended in all online versions of the Article.
- Published
- 2018
- Full Text
- View/download PDF
23. Delayed intramuscular human neurotrophin-3 improves recovery in adult and elderly rats after stroke
- Author
-
Denise A, Duricki, Thomas H, Hutson, Claudia, Kathe, Sara, Soleman, Daniel, Gonzalez-Carter, Jeffrey C, Petruska, H David, Shine, Qin, Chen, Tobias C, Wood, Michel, Bernanos, Diana, Cash, Steven C R, Williams, Fred H, Gage, and Lawrence D F, Moon
- Subjects
Time Factors ,Endothelin-1 ,Microinjections ,Genetic Vectors ,Age Factors ,Pyramidal Tracts ,Neuroimaging ,corticospinal ,Recovery of Function ,Original Articles ,sprouting ,Magnetic Resonance Imaging ,stroke ,Injections, Intramuscular ,Rats ,Adenoviridae ,Spinal Cord ,Neurotrophin 3 ,neurotrophin-3 ,plasticity ,Animals ,Humans ,Female ,Muscle, Skeletal ,Locomotion - Abstract
Duricki et al. show that intramuscular delivery of human neurotrophin-3 induces corticospinal plasticity and locomotor recovery in adult and elderly rats 24 hours post-stroke. This time-frame would be clinically feasible for most stroke victims, and the safety and tolerability of neurotrophin-3 in humans have been established for other disorders., 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.
- Published
- 2015
24. Unilateral Pyramidotomy of the Corticospinal Tract in Rats for Assessment of Neuroplasticity-inducing Therapies
- Author
-
Harold D Shine, Stephen B. McMahon, Claudia Kathe, Qin Chen, Lawrence D. F. Moon, and Thomas H. Hutson
- Subjects
Male ,General Chemical Engineering ,Pyramidal Tracts ,General Biochemistry, Genetics and Molecular Biology ,forelimb function ,Lesion ,Forelimb ,Animals ,Medicine ,Issue 94 ,Spinal Cord Injuries ,unilateral spinal cord injury ,Denervation ,Neuronal Plasticity ,Pyramidal tracts ,General Immunology and Microbiology ,business.industry ,General Neuroscience ,Anatomy ,corticospinal tract lesion ,Spinal cord ,Rats ,Disease Models, Animal ,in vivo ,medicine.anatomical_structure ,pyramids ,Corticospinal tract ,Medulla oblongata ,central nervous system lesions ,Brainstem ,medicine.symptom ,business ,Brain Stem ,Neuroscience - Abstract
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.
- Published
- 2014
- Full Text
- View/download PDF
25. The Use of an Adeno-Associated Viral Vector for Efficient Bicistronic Expression of Two Genes in the Central Nervous System
- Author
-
Lawrence D. F. Moon, Thomas H. Hutson, Sean Christopher Menezes, Claudia Kathe, Marie-Claire Rooney, and Hansruedi Büeler
- Subjects
Reporter gene ,Transduction (genetics) ,medicine.anatomical_structure ,Genetic enhancement ,Transgene ,Central nervous system ,Gene expression ,medicine ,Biology ,Gene ,Viral vector ,Cell biology - Abstract
Recombinant adeno-associated viral (AAV) vectors are one of the most promising therapeutic delivery systems for gene therapy to the central nervous system (CNS). Preclinical testing of novel gene therapies requires the careful design and production of AAV vectors and their successful application in a model of CNS injury. One major limitation of AAV vectors is their limited packaging capacity (
- Published
- 2014
- Full Text
- View/download PDF
26. Peripheral nervous system genes expressed in central neurons induce growth on inhibitory substrates
- Author
-
John L. Bixby, Lawrence D. F. Moon, Jose R. Pardinas, Stanley Hoffman, Candace L. Haddox, Thomas H. Hutson, William Buchser, Vance Lemmon, and Robin P. Smith
- Subjects
Cerebellum ,Glycobiology ,Gene Expression ,lcsh:Medicine ,Biochemistry ,Myelin ,0302 clinical medicine ,Molecular Cell Biology ,Neurobiology of Disease and Regeneration ,Axon ,lcsh:Science ,Neurons ,0303 health sciences ,Multidisciplinary ,Systems Biology ,Cell biology ,medicine.anatomical_structure ,Phenotype ,Peripheral nervous system ,Proteoglycans ,Research Article ,Signal Transduction ,DNA, Complementary ,animal structures ,Neurite ,Central nervous system ,Nerve Tissue Proteins ,Biology ,Inhibitory postsynaptic potential ,Molecular Genetics ,03 medical and health sciences ,Peripheral Nervous System ,medicine ,Genetics ,Neurites ,Humans ,030304 developmental biology ,Computational Neuroscience ,lcsh:R ,Computational Biology ,Microarray Analysis ,Molecular biology ,Nerve Regeneration ,Chondroitin Sulfate Proteoglycans ,Gene Expression Regulation ,nervous system ,Cellular Neuroscience ,lcsh:Q ,Neuron ,Laminin ,Molecular Neuroscience ,030217 neurology & neurosurgery ,Neuroscience - Abstract
Trauma to the spinal cord and brain can result in irreparable loss of function. This failure of recovery is in part due to inhibition of axon regeneration by myelin and chondroitin sulfate proteoglycans (CSPGs). Peripheral nervous system (PNS) neurons exhibit increased regenerative ability compared to central nervous system neurons, even in the presence of inhibitory environments. Previously, we identified over a thousand genes differentially expressed in PNS neurons relative to CNS neurons. These genes represent intrinsic differences that may account for the PNS’s enhanced regenerative ability. Cerebellar neurons were transfected with cDNAs for each of these PNS genes to assess their ability to enhance neurite growth on inhibitory (CSPG) or permissive (laminin) substrates. Using high content analysis, we evaluated the phenotypic profile of each neuron to extract meaningful data for over 1100 genes. Several known growth associated proteins potentiated neurite growth on laminin. Most interestingly, novel genes were identified that promoted neurite growth on CSPGs (GPX3, EIF2B5, RBMX). Bioinformatic approaches also uncovered a number of novel gene families that altered neurite growth of CNS neurons.
- Published
- 2012
27. Optimization of a 96-Well Electroporation Assay for Postnatal Rat CNS Neurons Suitable for Cost–Effective Medium-Throughput Screening of Genes that Promote Neurite Outgrowth
- Author
-
Thomas H. Hutson, William Buchser, Lawrence D. F. Moon, John L. Bixby, and Vance Lemmon
- Subjects
Neurite ,CNS neuron ,Central nervous system ,Biology ,Transfection ,Inhibitory postsynaptic potential ,lcsh:RC321-571 ,Cellular and Molecular Neuroscience ,Methods Article ,medicine ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Molecular Biology ,chemistry.chemical_classification ,neuronal regeneration ,Electroporation ,Neurite outgrowth ,Granule (cell biology) ,RAG ,Cell biology ,medicine.anatomical_structure ,chemistry ,Rho kinase inhibitor ,Inhibitory substrate ,96-well ,Glycoprotein ,96 well ,Neuroscience - Abstract
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 characterised 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 optimised a 96-well electroporation and neurite outgrowth assay for postnatal rat cerebellar granule neurons cultured upon an inhibitory cellular substrate expressing myelin-associated glycoprotein or a mixture of growth-inhibitory chondroitin sulphate proteoglycans. Optimal electroporation parameters resulted in 25% transfection efficiency and 50% viability for postnatal rat cerebellar granule neurons (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.
- Published
- 2011
- Full Text
- View/download PDF
28. Natural and targeted circuit reorganization after spinal cord injury
- Author
-
Mark A. Anderson, Jordan W. Squair, Matthieu Gautier, Thomas H. Hutson, Claudia Kathe, Quentin Barraud, Jocelyne Bloch, and Grégoire Courtine
- Subjects
functional recovery ,General Neuroscience ,electrical-stimulation ,Sensation ,axon regeneration ,Brain ,corticospinal tract lesion ,fibroblasts form ,motor recovery ,scar formation ,critical regulator ,Humans ,neural stem-cells ,fibrotic scar ,Spinal Cord Injuries - Abstract
The authors summarize changes in circuits after spinal cord injury and current strategies to target these circuits in order to improve recovery, but also advocate for new concepts of reorganizing circuits informed by multi-omic single-cell atlases., 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.
29. The neurons that restore walking after paralysis
- Author
-
Claudia Kathe, Michael A. Skinnider, Thomas H. Hutson, Nicola Regazzi, Matthieu Gautier, Robin Demesmaeker, Salif Komi, Steven Ceto, Nicholas D. James, Newton Cho, Laetitia Baud, Katia Galan, Kaya J. E. Matson, Andreas Rowald, Kyungjin Kim, Ruijia Wang, Karen Minassian, John O. Prior, Leonie Asboth, Quentin Barraud, Stéphanie P. Lacour, Ariel J. Levine, Fabien Wagner, Jocelyne Bloch, Jordan W. Squair, and Grégoire Courtine
- Subjects
Neurons ,Multidisciplinary ,nervous-system ,architecture ,Sequence Analysis, RNA ,Gene Expression Profiling ,Lumbosacral Region ,Neurological Rehabilitation ,Walking ,organization ,stimulation ,Electric Stimulation ,motor ,locomotion ,Mice ,circuit reorganization ,interrogation ,Spinal Cord ,Animals ,Humans ,Paralysis ,spinal-cord-injury ,v2a interneurons ,Spinal Cord Injuries - Abstract
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.
30. Cbp-dependent histone acetylation mediates axon regeneration induced by environmental enrichment in rodent spinal cord injury models
- Author
-
Piyush Chaturbedy, Alejandro Medrano-Fernandez, Angel Barco, John L. Bixby, Thomas H. Hutson, Eilidh McLachlan, Anne Laurence Boutillier, Kay Bartholdi, Luming Zhou, Lawrence D. F. Moon, Ilaria Palmisano, Claudia Kathe, Tapas K. Kundu, Quentin Barraud, Vance Lemmon, Arnau Hervera, Simone Di Giovanni, Sarmistha H. Sinha, Matt C. Danzi, Grégoire Courtine, José P. López-Atalaya, Guiping Komg, Akash Kumar Singh, Francesco De Virgiliis, Grup de Neurofarmacologia Molecular, Universitat Autònoma de Barcelona (UAB), Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Instituto de Neurociencias de Alicante, Laboratoire de neurosciences cognitives et adaptatives (LNCA), Université de Strasbourg (UNISTRA)-Centre National de la Recherche Scientifique (CNRS), Department of Computing Sciences [South Africa], Nelson Mandela Metropolitan University [Port Elizabeth, South Africa], Chemistry and Physics of Materials Unit [Bangalore, India], Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Molecular Biology and Genetics Unit [Bangalore, India] (Transcription and Disease Laboratory), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR)-Indian Institute of Science, Universidad Miguel Herández, Centre Hospitalier Universitaire Vaudois [Lausanne] (CHUV), Neurology, Wings for Life Spinal Cord Research Foundation, Rosetrees Trust, and Brain Research UK
- Subjects
mice ,Sensory Receptor Cells ,functional recovery ,[SDV]Life Sciences [q-bio] ,Environment ,Research & Experimental Medicine ,Article ,Histones ,03 medical and health sciences ,circuit reorganization ,0302 clinical medicine ,Political science ,Ganglia, Spinal ,expression ,Animals ,Spinal Cord Injuries ,11 Medical and Health Sciences ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,Motor Neurons ,0303 health sciences ,Science & Technology ,exercise ,European research ,sciatic-nerve ,Acetylation ,General Medicine ,Recovery of Function ,Cell Biology ,06 Biological Sciences ,Proprioception ,CREB-Binding Protein ,Axons ,Management ,Nerve Regeneration ,Disease Models, Animal ,sensory neurons ,Medicine, Research & Experimental ,proprioceptive feedback ,Christian ministry ,Calcium ,[SDV.NEU]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,central-nervous-system ,transcription ,E1A-Associated p300 Protein ,Life Sciences & Biomedicine ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
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.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.