Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
J Neurosci ; 43(47): 7929-7945, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37748862

ABSTRACT

The corticospinal tract (CST) forms a central part of the voluntary motor apparatus in all mammals. Thus, injury, disease, and subsequent degeneration within this pathway result in chronic irreversible functional deficits. Current strategies to repair the damaged CST are suboptimal in part because of underexplored molecular heterogeneity within the adult tract. Here, we combine spinal retrograde CST tracing with single-cell RNA sequencing (scRNAseq) in adult male and female mice to index corticospinal neuron (CSN) subtypes that differentially innervate the forelimb and hindlimb. We exploit publicly available datasets to confer anatomic specialization among CSNs and show that CSNs segregate not only along the forelimb and hindlimb axis but also by supraspinal axon collateralization. These anatomically defined transcriptional data allow us to use machine learning tools to build classifiers that discriminate between CSNs and cortical layer 2/3 and nonspinally terminating layer 5 neurons in M1 and separately identify limb-specific CSNs. Using these tools, CSN subtypes can be differentially identified to study postnatal patterning of the CST in vivo, leveraged to screen for novel limb-specific axon growth survival and growth activators in vitro, and ultimately exploited to repair the damaged CST after injury and disease.SIGNIFICANCE STATEMENT Therapeutic interventions designed to repair the damaged CST after spinal cord injury have remained functionally suboptimal in part because of an incomplete understanding of the molecular heterogeneity among subclasses of CSNs. Here, we combine spinal retrograde labeling with scRNAseq and annotate a CSN index by the termination pattern of their primary axon in the cervical or lumbar spinal cord and supraspinal collateral terminal fields. Using machine learning we have confirmed the veracity of our CSN gene lists to train classifiers to identify CSNs among all classes of neurons in primary motor cortex to study the development, patterning, homeostasis, and response to injury and disease, and ultimately target streamlined repair strategies to this critical motor pathway.


Subject(s)
Pyramidal Tracts , Spinal Cord Injuries , Mice , Female , Male , Animals , Pyramidal Tracts/physiology , Spinal Cord Injuries/genetics , Neurons/physiology , Axons/physiology , Mammals
2.
J Neurosci ; 42(11): 2190-2204, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35135857

ABSTRACT

Failure of CNS neurons to mount a significant growth response after trauma contributes to chronic functional deficits after spinal cord injury. Activator and repressor screening of embryonic cortical neurons and retinal ganglion cells in vitro and transcriptional profiling of developing CNS neurons harvested in vivo have identified several candidates that stimulate robust axon growth in vitro and in vivo Building on these studies, we sought to identify novel axon growth activators induced in the complex adult CNS environment in vivo We transcriptionally profiled intact sprouting adult corticospinal neurons (CSNs) after contralateral pyramidotomy (PyX) in nogo receptor-1 knock-out mice and found that intact CSNs were enriched in genes in the 3-phosphoinositide degradation pathway, including six 5-phosphatases. We explored whether inositol polyphosphate-5-phosphatase K (Inpp5k) could enhance corticospinal tract (CST) axon growth in preclinical models of acute and chronic CNS trauma. Overexpression of Inpp5k in intact adult CSNs in male and female mice enhanced the sprouting of intact CST terminals after PyX and cortical stroke and sprouting of CST axons after acute and chronic severe thoracic spinal contusion. We show that Inpp5k stimulates axon growth in part by elevating the density of active cofilin in labile growth cones, thus stimulating actin polymerization and enhancing microtubule protrusion into distal filopodia. We identify Inpp5k as a novel CST growth activator capable of driving compensatory axon growth in multiple complex CNS injury environments and underscores the veracity of using in vivo transcriptional screening to identify the next generation of cell-autonomous factors capable of repairing the damaged CNS.SIGNIFICANCE STATEMENT Neurologic recovery is limited after spinal cord injury as CNS neurons are incapable of self-repair post-trauma. In vitro screening strategies exploit the intrinsically high growth capacity of embryonic CNS neurons to identify novel axon growth activators. While promising candidates have been shown to stimulate axon growth in vivo, concomitant functional recovery remains incomplete. We identified Inpp5k as a novel axon growth activator using transcriptional profiling of intact adult corticospinal tract (CST) neurons that had initiated a growth response after pyramidotomy in plasticity sensitized nogo receptor-1-null mice. Here, we show that Inpp5k overexpression can stimulate CST axon growth after pyramidotomy, stroke, and acute and chronic contusion injuries. These data support in vivo screening approaches to identify novel axon growth activators.


Subject(s)
Pyramidal Tracts , Spinal Cord Injuries , Animals , Axons/metabolism , Female , Inositol/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Regeneration/physiology , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Polyphosphates/metabolism , Pyramidal Tracts/physiology
3.
STAR Protoc ; 2(4): 100941, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34877546

ABSTRACT

This protocol provides an improved pipeline for dissociating intact projection neurons from adult mouse cortex for applications including droplet and plate-based single-cell RNA sequencing, qPCR, immunocytochemistry, and long-term in vitro cell culture. This protocol provides a robust and reproducible dissociation pipeline that uses exclusively off-the-shelf reagents, not requiring the use of expensive dissociation kits. The unique incubation steps, in combination with the FACS gating strategy, results in unparalleled enrichment for intact cortical neurons from the adult brain. For complete details on the use and execution of this protocol, please refer to Golan et al. (2021).


Subject(s)
Cerebral Cortex/cytology , Neurites , Neurons/cytology , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Animals , Female , Male , Mice , Mice, Inbred C57BL , Neurites/chemistry , Neurites/metabolism
4.
Brain ; 143(6): 1697-1713, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32375169

ABSTRACT

After CNS trauma such as spinal cord injury, the ability of surviving neural elements to sprout axons, reorganize neural networks and support recovery of function is severely restricted, contributing to chronic neurological deficits. Among limitations on neural recovery are myelin-associated inhibitors functioning as ligands for neuronal Nogo receptor 1 (NgR1). A soluble decoy (NgR1-Fc, AXER-204) blocks these ligands and provides a means to promote recovery of function in multiple preclinical rodent models of spinal cord injury. However, the safety and efficacy of this reagent in non-human primate spinal cord injury and its toxicological profile have not been described. Here, we provide evidence that chronic intrathecal and intravenous administration of NgR1-Fc to cynomolgus monkey and to rat are without evident toxicity at doses of 20 mg and greater every other day (≥2.0 mg/kg/day), and far greater than the projected human dose. Adult female African green monkeys underwent right C5/6 lateral hemisection with evidence of persistent disuse of the right forelimb during feeding and right hindlimb during locomotion. At 1 month post-injury, the animals were randomized to treatment with vehicle (n = 6) or 0.10-0.17 mg/kg/day of NgR1-Fc (n = 8) delivered via intrathecal lumbar catheter and osmotic minipump for 4 months. One animal was removed from the study because of surgical complications of the catheter, but no treatment-related adverse events were noted in either group. Animal behaviour was evaluated at 6-7 months post-injury, i.e. 1-2 months after treatment cessation. The use of the impaired forelimb during spontaneous feeding and the impaired hindlimb during locomotion were both significantly greater in the treatment group. Tissue collected at 7-12 months post-injury showed no significant differences in lesion size, fibrotic scar, gliosis or neuroinflammation between groups. Serotoninergic raphespinal fibres below the lesion showed no deficit, with equal density on the lesioned and intact side below the level of the injury in both groups. Corticospinal axons traced from biotin-dextran-amine injections in the left motor cortex were equally labelled across groups and reduced caudal to the injury. The NgR1-Fc group tissue exhibited a significant 2-3-fold increased corticospinal axon density in the cervical cord below the level of the injury relative to the vehicle group. The data show that NgR1-Fc does not have preclinical toxicological issues in healthy animals or safety concerns in spinal cord injury animals. Thus, it presents as a potential therapeutic for spinal cord injury with evidence for behavioural improvement and growth of injured pathways in non-human primate spinal cord injury.


Subject(s)
Nogo Receptor 1/metabolism , Recombinant Fusion Proteins/pharmacology , Spinal Cord Injuries/drug therapy , Animals , Axons/pathology , Cervical Cord/pathology , Chlorocebus aethiops , Disease Models, Animal , Female , Male , Motor Activity/physiology , Myelin Proteins/metabolism , Myelin Sheath/metabolism , Nerve Regeneration/physiology , Neurons/metabolism , Neurons/pathology , Nogo Receptor 1/genetics , Pyramidal Tracts/pathology , Rats , Receptors, Fc/genetics , Receptors, Fc/metabolism , Recovery of Function , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
5.
J Neurosci ; 39(17): 3204-3216, 2019 04 24.
Article in English | MEDLINE | ID: mdl-30804090

ABSTRACT

After brain or spinal cord trauma, interaction of Nogo-A with neuronal NgR1 limits regenerative axonal sprouting and functional recovery. Cellular signaling by lipid-anchored NgR1 requires a coreceptor but the relevant partner in vivo is not clear. Here, we examined proteins enriched in NgR1 immunoprecipitates by Nogo-A exposure, identifying CRMP2, a cytosolic protein implicated in axon growth inhibition by Semaphorin/Plexin complexes. The Nogo-A-induced association of NgR1 with CRMP2 requires PlexinA2 as a coreceptor. Non-neuronal cells expressing both NgR1 and PlexinA2, but not either protein alone, contract upon Nogo-A exposure. Inhibition of cortical axon regeneration by Nogo-A depends on a NgR1/PlexinA2 genetic interaction because double-heterozygous NgR1+/-, PlexinA2+/- neurons, but not single-heterozygote neurons, are rescued from Nogo-A inhibition. NgR1 and PlexinA2 also interact genetically in vivo to restrict corticospinal sprouting in mouse cervical spinal cord after unilateral pyramidotomy. Greater post-injury sprouting in NgR1+/-, PlexinA2+/- mice supports enhanced neurological recovery of a mixed female and male double-heterozygous cohort. Thus, a NgR1/PlexinA2/CRMP2 ternary complex limits neural repair after adult mammalian CNS trauma.SIGNIFICANCE STATEMENT Several decades of molecular research have suggested that developmental regulation of axon growth is distinct in most regards from titration of axonal regenerative growth after adult CNS trauma. Among adult CNS pathways, the oligodendrocyte Nogo-A inhibition of growth through NgR1 is thought to have little molecular relationship to axonal guidance mechanisms active embryonically. Here, biochemical analysis of NgR1 function uncovered a physical complex with CRMP cytoplasmic mediators, and this led to appreciation of a role for PlexinA2 in concert with NgR1 after adult trauma. The data extend molecular understanding of neural repair after CNS trauma and link it to developmental processes.


Subject(s)
Axons/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Regeneration/physiology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nogo Proteins/metabolism , Nogo Receptor 1/metabolism , Pyramidal Tracts/metabolism , Receptors, Cell Surface/metabolism , Animals , COS Cells , Chlorocebus aethiops , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Motor Activity/physiology , Nerve Tissue Proteins/genetics , Nogo Proteins/genetics , Pyramidal Tracts/injuries , Receptors, Cell Surface/genetics , Recovery of Function/physiology , Spinal Cord Injuries/metabolism
6.
Nat Commun ; 9(1): 3419, 2018 08 24.
Article in English | MEDLINE | ID: mdl-30143638

ABSTRACT

Traumatic spinal cord injury results in persistent disability due to disconnection of surviving neural elements. Neural stem cell transplantation has been proposed as a therapeutic option, but optimal cell type and mechanistic aspects remain poorly defined. Here, we describe robust engraftment into lesioned immunodeficient mice of human neuroepithelial stem cells derived from the developing spinal cord and maintained in self-renewing adherent conditions for long periods. Extensive elongation of both graft and host axons occurs. Improved functional recovery after transplantation depends on neural relay function through the grafted neurons, requires the matching of neural identity to the anatomical site of injury, and is accompanied by expression of specific marker proteins. Thus, human neuroepithelial stem cells may provide an anatomically specific relay function for spinal cord injury recovery.


Subject(s)
Neural Stem Cells/cytology , Spinal Cord Regeneration/physiology , Animals , Axons/metabolism , Cell Differentiation/physiology , Cell Line , Cell Survival/physiology , Cells, Cultured , Female , Humans , Male , Mice , Neural Stem Cells/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/therapy , Stem Cell Transplantation
8.
Sci Signal ; 11(524)2018 04 03.
Article in English | MEDLINE | ID: mdl-29615517

ABSTRACT

Axonal growth after traumatic spinal cord injury is limited by endogenous inhibitors, selective blockade of which promotes partial neurological recovery. The partial repair phenotypes suggest that compensatory pathways limit improvement. Gene expression profiles of mice deficient in Ngr1, which encodes a receptor for myelin-associated inhibitors of axonal regeneration such as Nogo, revealed that trauma increased the mRNA expression of ORL1, which encodes the receptor for the opioid-related peptide nociceptin. Endogenous and overexpressed ORL1 coimmunoprecipitated with immature NgR1 protein, and ORL1 enhanced the O-linked glycosylation and surface expression of NgR1 in HEK293T and Neuro2A cells and primary neurons. ORL1 overexpression inhibited cortical neuron axon regeneration independently of NgR1. Furthermore, regeneration was inhibited by an ORL1 agonist and enhanced by the ORL1 antagonist J113397 through a ROCK-dependent mechanism. Mice treated with J113397 after dorsal hemisection of the mid-thoracic spinal cord recovered greater locomotor function and exhibited lumbar raphespinal axon sprouting. These effects were further enhanced by combined Ngr1 deletion and ORL1 inhibition. Thus, ORL1 limits neural repair directly and indirectly by enhancing NgR1 maturation, and ORL1 antagonists enhance recovery from traumatic CNS injuries in wild-type and Ngr1 null mice.


Subject(s)
Axons/physiology , Nerve Regeneration/physiology , Nogo Receptor 1/metabolism , Receptors, Opioid/metabolism , Spinal Cord Injuries/metabolism , Animals , Axons/metabolism , COS Cells , Cell Line, Tumor , Cells, Cultured , Chlorocebus aethiops , HEK293 Cells , Humans , Mice, Inbred C57BL , Mice, Knockout , Nerve Regeneration/drug effects , Nerve Regeneration/genetics , Neurons/cytology , Neurons/metabolism , Neurons/physiology , Nogo Receptor 1/genetics , Opioid Peptides/pharmacology , Receptors, Opioid/genetics , Spinal Cord Injuries/genetics , Spinal Cord Injuries/physiopathology , Nociceptin Receptor , Nociceptin
9.
Cell Rep ; 23(2): 415-428, 2018 Apr 10.
Article in English | MEDLINE | ID: mdl-29642001

ABSTRACT

Axonal regrowth is crucial for recovery from CNS injury but is severely restricted in adult mammals. We used a genome-wide loss-of-function screen for factors limiting axonal regeneration from cerebral cortical neurons in vitro. Knockdown of 16,007 individual genes identified 580 significant phenotypes. These molecules share no significant overlap with those suggested by previous expression profiles. There is enrichment for genes in pathways related to transport, receptor binding, and cytokine signaling, including Socs4 and Ship2. Among transport-regulating proteins, Rab GTPases are prominent. In vivo assessment with C. elegans validates a cell-autonomous restriction of regeneration by Rab27. Mice lacking Rab27b show enhanced retinal ganglion cell axon regeneration after optic nerve crush and greater motor function and raphespinal sprouting after spinal cord trauma. Thus, a comprehensive functional screen reveals multiple pathways restricting axonal regeneration and neurological recovery after injury.


Subject(s)
Axons/metabolism , Central Nervous System/physiology , Genome , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Female , Gene Regulatory Networks , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Regeneration , Optic Nerve/physiology , RNA Interference , RNA, Small Interfering/metabolism , Recovery of Function , Retinal Ganglion Cells/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Suppressor of Cytokine Signaling Proteins/antagonists & inhibitors , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , rab GTP-Binding Proteins/antagonists & inhibitors , rab GTP-Binding Proteins/deficiency , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
10.
Cell Rep ; 18(11): 2687-2701, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28297672

ABSTRACT

Functional deficits persist after spinal cord injury (SCI) because axons in the adult mammalian central nervous system (CNS) fail to regenerate. However, modest levels of spontaneous functional recovery are typically observed after trauma and are thought to be mediated by the plasticity of intact circuitry. The mechanisms underlying intact circuit plasticity are not delineated. Here, we characterize the in vivo transcriptome of sprouting intact neurons from Ngr1 null mice after partial SCI. We identify the lysophosphatidic acid signaling modulators LPPR1 and LPAR1 as intrinsic axon growth modulators for intact corticospinal motor neurons after adjacent injury. Furthermore, in vivo LPAR1 inhibition or LPPR1 overexpression enhances sprouting of intact corticospinal tract axons and yields greater functional recovery after unilateral brainstem lesion in wild-type mice. Thus, the transcriptional profile of injury-induced sprouting of intact neurons reveals targets for therapeutic enhancement of axon growth initiation and new synapse formation.


Subject(s)
Axons/pathology , Central Nervous System/injuries , Central Nervous System/pathology , Animals , Gene Expression Profiling , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/pathology , Neurites/metabolism , Neurogenesis , Protein Binding , Signal Transduction , Spinal Cord/pathology , Transcription, Genetic
11.
eNeuro ; 3(6)2016.
Article in English | MEDLINE | ID: mdl-28032120

ABSTRACT

After traumatic damage of the brain or spinal cord, many surviving neurons are disconnected, and recovery of function is limited by poor axon regeneration. Recent data have suggested that poly ADP-ribosylation plays a role in limiting axonal regrowth such that inhibition of poly (ADP-ribose) polymerase (PARP) may have therapeutic efficacy for neurological recovery after trauma. Here, we tested systemic administration of the PARP inhibitor, veliparib, and showed effective suppression of PARylation in the mouse CNS. After optic nerve crush injury or dorsal hemisection of the thoracic spinal cord in mice, treatment with veliparib at doses with pharmacodynamic action had no benefit for axonal regeneration or functional recovery. We considered whether PARP gene family specificity might play a role. In vitro mouse cerebral cortex axon regeneration experiments revealed that short hairpin RNA (shRNA)-mediated suppression of PARP1 promoted axonal regeneration, whereas suppression of other PARP isoforms either had no effect or decreased regeneration. Therefore, we examined recovery from neurological trauma in mice lacking PARP1. No increase of axonal regeneration was observed in Parp1-/- mice after optic nerve crush injury or dorsal hemisection of the thoracic spinal cord, and there was no improvement in motor function recovery. Thus, comprehensive in vivo analysis reveals no indication that clinical PARP inhibitors will on their own provide benefit for recovery from CNS trauma.


Subject(s)
Axons/drug effects , Benzimidazoles/pharmacology , Nerve Regeneration/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Recovery of Function/drug effects , Animals , Axons/enzymology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Disease Models, Animal , Female , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Nerve Regeneration/physiology , Optic Nerve Injuries/drug therapy , Optic Nerve Injuries/enzymology , Optic Nerve Injuries/pathology , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Recovery of Function/physiology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/enzymology , Spinal Cord Injuries/pathology , Thoracic Vertebrae
12.
Neurotherapeutics ; 13(2): 370-81, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26846379

ABSTRACT

Neurons have a limited capacity to regenerate in the adult central nervous system (CNS). The inability of damaged axons to re-establish original circuits results in permanent functional impairment after spinal cord injury (SCI). Despite abortive regeneration of axotomized CNS neurons, limited spontaneous recovery of motor function emerges after partial SCI in humans and experimental rodent models of SCI. It is hypothesized that this spontaneous functional recovery is the result of the reorganization of descending motor pathways spared by the injury, suggesting that plasticity of intact circuits is a potent alternative conduit to enhance functional recovery after SCI. In support of this hypothesis, several studies have shown that after unilateral corticospinal tract (CST) lesion (unilateral pyramidotomy), the intact CST functionally sprouts into the denervated side of the spinal cord. Furthermore, pharmacologic and genetic methods that enhance the intrinsic growth capacity of adult neurons or block extracellular growth inhibitors are effective at significantly enhancing intact CST reorganization and recovery of motor function. Owing to its importance in controlling fine motor behavior in primates, the CST is the most widely studied descending motor pathway; however, additional studies in rodents have shown that plasticity within other spared descending motor pathways, including the rubrospinal tract, raphespinal tract, and reticulospinal tract, can also result in restoration of function after incomplete SCI. Identifying the molecular mechanisms that drive plasticity within intact circuits is crucial in developing novel, potent, and specific therapeutics to restore function after SCI. In this review we discuss the evidence supporting a focus on exploring the capacity of intact motor circuits to functionally repair the damaged CNS after SCI.


Subject(s)
Efferent Pathways/injuries , Animals , Efferent Pathways/physiopathology , Humans , Motor Neurons/physiology , Nerve Regeneration/physiology , Neuronal Plasticity/physiology , Spinal Cord/physiopathology , Spinal Cord Injuries/physiopathology
13.
J Neurosci ; 35(46): 15403-18, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26586827

ABSTRACT

Spinal cord injury interrupts descending motor tracts and creates persistent functional deficits due to the absence of spontaneous axon regeneration. Of descending pathways, the corticospinal tract (CST) is thought to be the most critical for voluntary function in primates. Even with multiple tracer injections and genetic tools, the CST is visualized to only a minor degree in experimental studies. Here, we identify and validate the mu-crystallin (crym) gene as a high-fidelity marker of the CST. In transgenic mice expressing green fluorescent protein (GFP) under crym regulatory elements (crym-GFP), comprehensive and near complete CST labeling is achieved throughout the spinal cord. Bilateral pyramidotomy eliminated the 17,000 GFP-positive CST axons that were reproducibly labeled in brainstem from the spinal cord. We show that CST tracing with crym-GFP is 10-fold more efficient than tracing with biotinylated dextran amine (BDA). Using crym-GFP, we reevaluated the CST in mice lacking nogo receptor 1 (NgR1), a protein implicated in limiting neural repair. The number and trajectory of CST axons in ngr1(-/-) mice without injury was indistinguishable from ngr1(+/+) mice. After dorsal hemisection in the midthoracic cord, CST axons did not significantly regenerate in ngr1(+/+) mice, but an average of 162 of the 6000 labeled thoracic CST axons (2.68%) regenerated >100 µm past the lesion site in crym-GFP ngr1(-/-) mice. Although traditional BDA tracing cannot reliably visualize regenerating ngr1(-/-) CST axons, their regenerative course is clear with crym-GFP. Therefore the crym-GFP transgenic mouse is a useful tool for studies of CST anatomy in experimental studies of motor pathways. SIGNIFICANCE STATEMENT: Axon regeneration fails in the adult CNS, resulting in permanent functional deficits. Traditionally, inefficient extrinsic tracers such a biotinylated dextran amine (BDA) are used to label regenerating fibers after therapeutic intervention. We introduce crym-green fluorescent protein (GFP) transgenic mice as a comprehensive and specific tool with which to study the primary descending motor tract, the corticospinal tract (CST). CST labeling with crym-GFP is 10 times more efficient compared with BDA. The enhanced sensitivity afforded by crym-GFP revealed significant CST regeneration in NgR1 knock-out mice. Therefore, crym-GFP can be used as a standardized tool for future CST spinal cord injury studies.


Subject(s)
Crystallins/metabolism , Gene Expression Regulation/genetics , Myelin Proteins/deficiency , Nerve Regeneration/genetics , Pyramidal Tracts/pathology , Receptors, Cell Surface/deficiency , Spinal Cord Injuries/complications , Amidines/metabolism , Analysis of Variance , Animals , Axons/pathology , Biotin/analogs & derivatives , Biotin/metabolism , Crystallins/biosynthesis , Crystallins/genetics , Dextrans/metabolism , Disease Models, Animal , Functional Laterality , GPI-Linked Proteins/deficiency , GPI-Linked Proteins/genetics , Glial Fibrillary Acidic Protein/metabolism , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Proteins/genetics , Nogo Receptor 1 , Pyramidal Tracts/metabolism , Receptors, Cell Surface/genetics , Recovery of Function/genetics , Spinal Cord Injuries/pathology , mu-Crystallins
14.
J Neurosci ; 35(29): 10429-39, 2015 Jul 22.
Article in English | MEDLINE | ID: mdl-26203138

ABSTRACT

Axonal growth and neuronal rewiring facilitate functional recovery after spinal cord injury. Known interventions that promote neural repair remain limited in their functional efficacy. To understand genetic determinants of mammalian CNS axon regeneration, we completed an unbiased RNAi gene-silencing screen across most phosphatases in the genome. We identified one known and 17 previously unknown phosphatase suppressors of injury-induced CNS axon growth. Silencing Inpp5f (Sac2) leads to robust enhancement of axon regeneration and growth cone reformation. Results from cultured Inpp5f(-/-) neurons confirm lentiviral shRNA results from the screen. Consistent with the nonoverlapping substrate specificity between Inpp5f and PTEN, rapamycin does not block enhanced regeneration in Inpp5f(-/-) neurons, implicating mechanisms independent of the PI3K/AKT/mTOR pathway. Inpp5f(-/-) mice develop normally, but show enhanced anatomical and functional recovery after mid-thoracic dorsal hemisection injury. More serotonergic axons sprout and/or regenerate caudal to the lesion level, and greater numbers of corticospinal tract axons sprout rostral to the lesion. Functionally, Inpp5f-null mice exhibit enhanced recovery of motor functions in both open-field and rotarod tests. This study demonstrates the potential of an unbiased high-throughput functional screen to identify endogenous suppressors of CNS axon growth after injury, and reveals Inpp5f (Sac2) as a novel suppressor of CNS axon repair after spinal cord injury. Significance statement: The extent of axon regeneration is a critical determinant of neurological recovery from injury, and is extremely limited in the adult mammalian CNS. We describe an unbiased gene-silencing screen that uncovered novel molecules suppressing axonal regeneration. Inpp5f (Sac2) gene deletion promoted recovery from spinal cord injury with no side effects. The mechanism of action is distinct from another lipid phosphatase implicated in regeneration, PTEN. This opens new pathways for investigation in spinal cord injury research. Furthermore the screening methodology can be applied on a genome wide scale to discovery the entire set of mammalian genes contributing to axonal regeneration.


Subject(s)
Axons/pathology , Nerve Regeneration/genetics , Phosphoric Monoester Hydrolases/genetics , Spinal Cord Injuries/pathology , Animals , Axons/metabolism , Disease Models, Animal , Gene Knockdown Techniques , Immunohistochemistry , Inositol Polyphosphate 5-Phosphatases , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoric Monoester Hydrolases/deficiency , Phosphoric Monoester Hydrolases/metabolism , Recovery of Function/physiology , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord Injuries/metabolism
15.
J Neurosci ; 35(4): 1443-57, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25632122

ABSTRACT

Axons in the adult CNS fail to regenerate after injury, and therefore recovery from spinal cord injury (SCI) is limited. Although full recovery is rare, a modest degree of spontaneous recovery is observed consistently in a broad range of clinical and nonclinical situations. To define the mechanisms mediating spontaneous recovery of function after incomplete SCI, we created bilaterally complete medullary corticospinal tract lesions in adult mice, eliminating a crucial pathway for voluntary skilled movement. Anatomic and pharmacogenetic tools were used to identify the pathways driving spontaneous functional recovery in wild-type and plasticity-sensitized mice lacking Nogo receptor 1. We found that plasticity-sensitized mice recovered 50% of normal skilled locomotor function within 5 weeks of lesion. This significant, yet incomplete, spontaneous recovery was accompanied by extensive sprouting of intact rubrofugal and rubrospinal projections with the emergence of a de novo circuit between the red nucleus and the nucleus raphe magnus. Transient silencing of this rubro-raphe circuit in vivo via activation of the inhibitory DREADD (designer receptor exclusively activated by designer drugs) receptor hM4di abrogated spontaneous functional recovery. These data highlight the pivotal role of uninjured motor circuit plasticity in supporting functional recovery after trauma, and support a focus of experimental strategies on enhancing intact circuit rearrangement to promote functional recovery after SCI.


Subject(s)
Neuronal Plasticity/physiology , Pyramidal Tracts/pathology , Raphe Nuclei/pathology , Recovery of Function/physiology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Animals , Designer Drugs/pharmacology , Functional Laterality , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Strength/genetics , Myelin Proteins/deficiency , Myelin Proteins/genetics , Nogo Proteins , Psychomotor Disorders/etiology , Stereotyped Behavior/physiology , Time Factors
16.
J Neurotrauma ; 31(24): 1955-66, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-24964223

ABSTRACT

Axonal growth and neurological recovery after traumatic spinal cord injury (SCI) is limited by the presence of inhibitory proteins in myelin, several of which act via the NgR1 protein in neurons. A truncated soluble ligand-binding fragment of NgR1 serves as a decoy and promotes recovery in acute and chronic rodent SCI models. To develop the translational potential of these observations, we created a human sequence-derived NgR1(310)-Fc protein. This protein is active in vitro. When the human NgR1 decoy is administered by continuous intracerebroventricular infusion to rats with a spinal contusion injury at doses of 0.09-0.53 mg/kg/d, neurological recovery is improved. Effective doses double the percentage of rats able to bear weight on their hindlimbs. Next, we considered the half-life and distribution of NgR1(310)-Fc after bolus delivery to the lumbar intrathecal space. The protein is found throughout the neuraxis and has a tissue half-life of approximately 2 days in the rat, and 5 days in the nonhuman primate. At an intermittent, once every 4 day, lumbar bolus dosing schedule of 0.14 mg/kg/d, NgR1(310)-Fc promoted locomotor rat recovery from spinal cord contusion at least as effectively as continuous infusion in open field and grid walking tasks. Moreover, the intermittent lumbar NgR1(310)-Fc treatment increased the growth of raphespinal axons into the lumbar spinal cord after injury. Thus, human NgR1(310)-Fc provides effective treatment for recovery from traumatic SCI in this preclinical model with a simplified administration regimen that facilitates clinical testing.


Subject(s)
Myelin Proteins/administration & dosage , Neuroprotective Agents/administration & dosage , Receptors, Cell Surface/administration & dosage , Receptors, Fc/administration & dosage , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Animals , Disease Models, Animal , Female , GPI-Linked Proteins/administration & dosage , Humans , Injections, Spinal , Nogo Receptor 1 , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage
17.
J Neurotrauma ; 31(15): 1362-73, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24779685

ABSTRACT

Traumatic spinal cord injury (SCI) causes long-term disability with limited functional recovery linked to the extent of axonal connectivity. Quantitative diffusion tensor imaging (DTI) of axonal integrity has been suggested as a potential biomarker for prognostic and therapeutic evaluation after trauma, but its correlation with functional outcomes has not been clearly defined. To examine this application, female Sprague-Dawley rats underwent midthoracic laminectomy followed by traumatic spinal cord contusion of differing severities or laminectomy without contusion. Locomotor scores and hindlimb kinematic data were collected for 4 weeks post-injury. Ex vivo DTI was then performed to assess axonal integrity using tractography and fractional anisotropy (FA), a numerical measure of relative white matter integrity, at the injury epicenter and at specific intervals rostral and caudal to the injury site. Immunohistochemistry for tissue sparing was also performed. Statistical correlation between imaging data and functional performance was assessed as the primary outcome. All injured animals showed some recovery of locomotor function, while hindlimb kinematics revealed graded deficits consistent with injury severity. Standard T2 magnetic resonance sequences illustrated conventional spinal cord morphology adjacent to contusions while corresponding FA maps indicated graded white matter pathology within these adjacent regions. Positive correlations between locomotor (Basso, Beattie, and Bresnahan score and gait kinematics) and imaging (FA values) parameters were also observed within these adjacent regions, most strongly within caudal segments beyond the lesion. Evaluation of axonal injury by DTI provides a mechanism for functional recovery assessment in a rodent SCI model. These findings suggest that focused DTI analysis of caudal spinal cord should be studied in human cases in relationship to motor outcome to augment outcome biomarkers for clinical cases.


Subject(s)
Diffusion Tensor Imaging , Motor Activity , Recovery of Function , Spinal Cord Injuries/pathology , Animals , Disease Models, Animal , Female , Image Processing, Computer-Assisted , Immunohistochemistry , Prognosis , Rats , Rats, Sprague-Dawley
18.
Brain Res ; 1538: 17-25, 2013 Nov 13.
Article in English | MEDLINE | ID: mdl-24055330

ABSTRACT

In the context of injury to the corticospinal tract (CST), brainstem-origin circuits may provide an alternative system of descending motor influence. However, subcortical circuits are largely under subconscious control. To improve volitional control over spared fibers after CST injury, we hypothesized that a combination of physical exercises simultaneously stimulating cortical and brainstem pathways above the injury would strengthen corticobulbar connections through Hebbian-like mechanisms. We sought to test this hypothesis in mice with unilateral CST lesions. Ten days after pyramidotomy, mice were randomized to four training groups: (1) postural exercises designed to stimulate brainstem pathways (BS); (2) distal limb-grip exercises preferentially stimulating CST pathways (CST); (3) simultaneous multimodal exercises (BS+CST); or (4) no training (NT). Behavioral and anatomical outcomes were assessed after 20 training sessions over 4 weeks. Mice in the BS+CST training group showed a trend toward greater improvements in skilled limb performance than mice in the other groups. There were no consistent differences between training groups in gait kinematics. Anatomically, multimodal BS+CST training neither increased corticobulbar fiber density of the lesioned CST rostral to the lesion nor collateral sprouting of the unlesioned CST caudal to the lesion. Further studies should incorporate electrophysiological assessment to gauge changes in synaptic strength of direct and indirect pathways between the cortex and spinal cord in response to multimodal exercises.


Subject(s)
Brain Stem/physiopathology , Cerebral Cortex/physiopathology , Motor Skills , Physical Conditioning, Animal , Pyramidal Tracts/pathology , Pyramidal Tracts/physiopathology , Animals , Female , Gait/physiology , Mice , Mice, Inbred C57BL , mu-Crystallins
19.
Neuron ; 77(5): 859-66, 2013 Mar 06.
Article in English | MEDLINE | ID: mdl-23473316

ABSTRACT

Experience rearranges anatomical connectivity in the brain, but such plasticity is suppressed in adulthood. We examined the turnover of dendritic spines and axonal varicosities in the somatosensory cortex of mice lacking Nogo Receptor 1 (NgR1). Through adolescence, the anatomy and plasticity of ngr1 null mice are indistinguishable from control, but suppression of turnover after age 26 days fails to occur in ngr1-/- mice. Adolescent anatomical plasticity can be restored to 1-year-old mice by conditional deletion of ngr1. Suppression of anatomical dynamics by NgR1 is cell autonomous and is phenocopied by deletion of Nogo-A ligand. Whisker removal deprives the somatosensory cortex of experience-dependent input and reduces dendritic spine turnover in adult ngr1-/- mice to control levels, while an acutely enriched environment increases dendritic spine dynamics in control mice to the level of ngr1-/- mice in a standard environment. Thus, NgR1 determines the low set point for synaptic turnover in adult cerebral cortex.


Subject(s)
Brain Chemistry/physiology , Brain/anatomy & histology , Myelin Proteins/physiology , Neuronal Plasticity/physiology , Aging/physiology , Animals , Behavior/physiology , Brain/growth & development , Brain Chemistry/genetics , Cerebral Cortex/growth & development , Cerebral Cortex/physiology , Dendritic Spines/physiology , Denervation , Fear/psychology , Green Fluorescent Proteins/genetics , Image Processing, Computer-Assisted , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Mutation/genetics , Mutation/physiology , Myelin Proteins/genetics , Neuronal Plasticity/genetics , Nogo Proteins , Postural Balance/genetics , Postural Balance/physiology , Vibrissae/innervation
20.
Exp Neurol ; 237(1): 55-69, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22728374

ABSTRACT

Several pharmacological approaches to promote neural repair and recovery after CNS injury have been identified. Blockade of either astrocyte-derived chondroitin sulfate proteoglycans (CSPGs) or oligodendrocyte-derived NogoReceptor (NgR1) ligands reduces extrinsic inhibition of axonal growth, though combined blockade of these distinct pathways has not been tested. The intrinsic growth potential of adult mammalian neurons can be promoted by several pathways, including pre-conditioning injury for dorsal root ganglion (DRG) neurons and macrophage activation for retinal ganglion cells (RGCs). Singly, pharmacological interventions have restricted efficacy without foreign cells, mechanical scaffolds or viral gene therapy. Here, we examined combinations of pharmacological approaches and assessed the degree of axonal regeneration. After mouse optic nerve crush injury, NgR1-/- neurons regenerate RGC axons as extensively as do zymosan-injected, macrophage-activated WT mice. Synergistic enhancement of regeneration is achieved by combining these interventions in zymosan-injected NgR1-/- mice. In rats with a spinal dorsal column crush injury, a preconditioning peripheral sciatic nerve axotomy, or NgR1(310)ecto-Fc decoy protein treatment or ChondroitinaseABC (ChABC) treatment independently support similar degrees of regeneration by ascending primary afferent fibers into the vicinity of the injury site. Treatment with two of these three interventions does not significantly enhance the degree of axonal regeneration. In contrast, triple therapy combining NgR1 decoy, ChABC and preconditioning, allows axons to regenerate millimeters past the spinal cord injury site. The benefit of a pre-conditioning injury is most robust, but a peripheral nerve injury coincident with, or 3 days after, spinal cord injury also synergizes with NgR1 decoy and ChABC. Thus, maximal axonal regeneration and neural repair are achieved by combining independently effective pharmacological approaches.


Subject(s)
Axons/physiology , Neural Inhibition/physiology , Neuroglia/physiology , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/rehabilitation , Animals , Axons/pathology , Chondroitin ABC Lyase/administration & dosage , Chondroitin ABC Lyase/therapeutic use , Female , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/deficiency , GPI-Linked Proteins/therapeutic use , Macrophage Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Proteins/antagonists & inhibitors , Myelin Proteins/deficiency , Myelin Proteins/therapeutic use , Neural Inhibition/genetics , Neuroglia/pathology , Nogo Receptor 1 , Optic Nerve Injuries/genetics , Optic Nerve Injuries/pathology , Optic Nerve Injuries/physiopathology , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/therapeutic use , Spinal Cord Injuries/genetics , Zymosan/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...