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











Publication year range
1.
Transl Stroke Res ; 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37326791

ABSTRACT

Repulsive guidance molecule A (RGMa) is an inhibitor of neuronal growth and survival which is upregulated in the damaged central nervous system following acute spinal cord injury (SCI), traumatic brain injury, acute ischemic stroke (AIS), and other neuropathological conditions. Neutralization of RGMa is neuroprotective and promotes neuroplasticity in several preclinical models of neurodegeneration and injury including multiple sclerosis, AIS, and SCI. Given the limitations of current treatments for AIS due to narrow time windows to intervention (TTI), and restrictive patient selection criteria, there is significant unmet need for therapeutic agents that enable tissue survival and repair following acute ischemic damage for a broader population of stroke patients. In this preclinical study, we evaluated whether elezanumab, a human anti-RGMa monoclonal antibody, could improve neuromotor function and modulate neuroinflammatory cell activation following AIS with delayed intervention times up to 24 h using a rabbit embolic permanent middle cerebral artery occlusion model (pMCAO). In two replicate 28-day pMCAO studies, weekly intravenous infusions of elezanumab, over a range of doses and TTIs of 6 and 24 h after stroke, significantly improved neuromotor function in both pMCAO studies when first administered 6 h after stroke. All elezanumab treatment groups, including the 24 h TTI group, had significantly less neuroinflammation as assessed by microglial and astrocyte activation. The novel mechanism of action and potential for expanding TTI in human AIS make elezanumab distinct from current acute reperfusion therapies, and support evaluation in clinical trials of acute CNS damage to determine optimal dose and TTI in humans. A: Ramified/resting astrocytes and microglia in a normal, uninjured rabbit brain. B: Rabbit pMCAO brain illustrating lesion on right side of brain (red), surrounded by penumbra (pink) during acute phase post stroke, with minimal injury to left brain hemisphere. Penumbra characterized by activated astrocytes and microglia (region in crosshair within circle), with upregulation of free and bound RGMa. C: Elezanumab binds to both free and bound RGMa, preventing full activation of astrocytes and microglia. D: Elezanumab is efficacious in rabbit pMCAO with a 4 × larger TTI window vs. tPA (6 vs. 1.5 h, respectively). In human AIS, tPA is approved for a TTI of 3-4.5 h. Elezanumab is currently being evaluated in a clinical Ph2 study of AIS to determine the optimal dose and TTI (NCT04309474).

2.
Stem Cell Reports ; 18(3): 672-687, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36764296

ABSTRACT

The adult spinal cord contains a population of ependymal-derived neural stem/progenitor cells (epNSPCs) that are normally quiescent, but are activated to proliferate, differentiate, and migrate after spinal cord injury. The mechanisms that regulate their response to injury cues, however, remain unknown. Here, we demonstrate that excitotoxic levels of glutamate promote the proliferation and astrocytic fate specification of adult spinal cord epNSPCs. We show that glutamate-mediated calcium influx through calcium-permeable alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors (CP-AMPARs) in concert with Notch signaling increases the proliferation of epNSPCs via pCREB, and induces astrocytic differentiation through Hes1 upregulation. Furthermore, the in vivo targeting of this pathway via positive modulation of AMPARs after spinal cord injury enhances epNSPC proliferation, astrogliogenesis, neurotrophic factor production and increases neuronal survival. Our study uncovers an important mechanism by which CP-AMPARs regulate the growth and phenotype of epNSPCs, which can be targeted therapeutically to harness the regenerative potential of these cells after injury.


Subject(s)
Glutamic Acid , Spinal Cord Injuries , Humans , Glutamic Acid/metabolism , Calcium/metabolism , Spinal Cord , Receptors, AMPA/metabolism , Spinal Cord Injuries/metabolism , Cell Proliferation
3.
Neurobiol Dis ; 172: 105812, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35810963

ABSTRACT

Spinal cord injury (SCI) elicits a cascade of degenerative events including cell death, axonal degeneration, and the upregulation of inhibitory molecules which limit repair. Repulsive guidance molecule A (RGMa) is an axon growth inhibitor which is also involved in neuronal cell death and differentiation. SCI causes upregulation of RGMa in the injured rodent, non-human primate, and human spinal cord. Recently, we showed that delayed administration of elezanumab, a high affinity human RGMa-specific monoclonal antibody, promoted neuroprotective and regenerative effects following thoracic SCI. Since most human traumatic SCI is at the cervical level, and level-dependent anatomical and molecular differences may influence pathophysiological responses to injury and treatment, we examined the efficacy of elezanumab and its therapeutic time window of administration in a clinically relevant rat model of cervical impact-compression SCI. Pharmacokinetic analysis of plasma and spinal cord tissue lysate showed comparable levels of RGMa antibodies with delayed administration following cervical SCI. At 12w after SCI, elezanumab promoted long term benefits including perilesional sparing of motoneurons and increased neuroplasticity of key descending pathways involved in locomotion and fine motor function. Elezanumab also promoted growth of corticospinal axons into spinal cord gray matter and enhanced serotonergic innervation of the ventral horn to form synaptic connections caudal to the cervical lesion. Significant recovery in grip and trunk/core strength, locomotion and gait, and spontaneous voiding ability was found in rats treated with elezanumab either immediately post-injury or at 3 h post-SCI, and improvements in specific gait parameters were found when elezanumab was delayed to 24 h post-injury. We also developed a new locomotor score, the Cervical Locomotor Score, a simple and sensitive measure of trunk/core and limb strength and stability during dynamic locomotion.


Subject(s)
Cervical Cord , Spinal Cord Injuries , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Cervical Cord/metabolism , GPI-Linked Proteins , Humans , Membrane Proteins , Nerve Tissue Proteins/metabolism , Rats , Recovery of Function/physiology , Spinal Cord/pathology , Spinal Cord Injuries/pathology
4.
Neurobiol Dis ; 155: 105385, 2021 07.
Article in English | MEDLINE | ID: mdl-33991647

ABSTRACT

Spinal cord injury (SCI) is a devastating condition characterized by loss of function, secondary to damaged spinal neurons, disrupted axonal connections, and myelin loss. Spontaneous recovery is limited, and there are no approved pharmaceutical treatments to reduce ongoing damage or promote repair. Repulsive guidance molecule A (RGMa) is upregulated following injury to the central nervous system (CNS), where it is believed to induce neuronal apoptosis and inhibit axonal growth and remyelination. We evaluated elezanumab, a human anti-RGMa monoclonal antibody, in a novel, newly characterized non-human primate (NHP) hemicompression model of thoracic SCI. Systemic intravenous (IV) administration of elezanumab over 6 months was well tolerated and associated with significant improvements in locomotor function. Treatment of animals for 16 weeks with a continuous intrathecal infusion of elezanumab below the lesion was not efficacious. IV elezanumab improved microstructural integrity of extralesional tissue as reflected by higher fractional anisotropy and magnetization transfer ratios in treated vs. untreated animals. IV elezanumab also reduced SCI-induced increases in soluble RGMa in cerebrospinal fluid, and membrane bound RGMa rostral and caudal to the lesion. Anterograde tracing of the corticospinal tract (CST) from the contralesional motor cortex following 20 weeks of IV elezanumab revealed a significant increase in the density of CST fibers emerging from the ipsilesional CST into the medial/ventral gray matter. There was a significant sprouting of serotonergic (5-HT) fibers rostral to the injury and in the ventral horn of lower thoracic regions. These data demonstrate that 6 months of intermittent IV administration of elezanumab, beginning within 24 h after a thoracic SCI, promotes neuroprotection and neuroplasticity of key descending pathways involved in locomotion. These findings emphasize the mechanisms leading to improved recovery of neuromotor functions with elezanumab in acute SCI in NHPs.


Subject(s)
Antibodies, Monoclonal/administration & dosage , GPI-Linked Proteins/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Neuronal Plasticity/drug effects , Neuroprotection/drug effects , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , Chlorocebus aethiops , Exercise Test/methods , Humans , Injections, Spinal , Male , Neuronal Plasticity/physiology , Neuroprotection/physiology , Primates , Recovery of Function/physiology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Thoracic Vertebrae/injuries
5.
Neurobiol Dis ; 150: 105259, 2021 03.
Article in English | MEDLINE | ID: mdl-33434618

ABSTRACT

Neuronal regeneration in the injured central nervous system is hampered by multiple extracellular proteins. These proteins exert their inhibitory action through interactions with receptors that are located in cholesterol rich compartments of the membrane termed lipid rafts. Here we show that cholesterol-synthesis inhibition prevents the association of the Neogenin receptor with lipid rafts. Furthermore, we show that cholesterol-synthesis inhibition enhances axonal growth both on inhibitory -myelin and -RGMa substrates. Following optic nerve injury, lowering cholesterol synthesis with both drugs and siRNA-strategies allows for robust axonal regeneration and promotes neuronal survival. Cholesterol inhibition also enhanced photoreceptor survival in a model of Retinitis Pigmentosa. Our data reveal that Lovastatin leads to several opposing effects on regenerating axons: cholesterol synthesis inhibition promotes regeneration whereas altered prenylation impairs regeneration. We also show that the lactone prodrug form of lovastatin has differing effects on regeneration when compared to the ring-open hydroxy-acid form. Thus the association of cell surface receptors with lipid rafts contributes to axonal regeneration inhibition, and blocking cholesterol synthesis provides a potential therapeutic approach to promote neuronal regeneration and survival in the diseased Central Nervous System. SIGNIFICANCE STATEMENT: Statins have been intensively used to treat high levels of cholesterol in humans. However, the effect of cholesterol inhibition in both the healthy and the diseased brain remains controversial. In particular, it is unclear whether cholesterol inhibition with statins can promote regeneration and survival following injuries. Here we show that late stage cholesterol inhibition promotes robust axonal regeneration following optic nerve injury. We identified distinct mechanisms of action for activated vs non-activated Lovastatin that may account for discrepancies found in the literature. We show that late stage cholesterol synthesis inhibition alters Neogenin association with lipid rafts, thereby i) neutralizing the inhibitory function of its ligand and ii) offering a novel opportunity to promote CNS regeneration and survival following injuries.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , Nerve Regeneration/drug effects , Neurons/drug effects , Optic Nerve/drug effects , Animals , Anticholesteremic Agents/pharmacology , Axons/drug effects , Axons/pathology , Cell Survival , Chick Embryo , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Myelin Sheath , Neurons/metabolism , Optic Nerve/metabolism , Optic Nerve/pathology , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Photoreceptor Cells , Prenylation , Prodrugs , Rats , Retina , Retinitis Pigmentosa , trans-1,4-Bis(2-chlorobenzaminomethyl)cyclohexane Dihydrochloride/pharmacology
6.
Stem Cells Transl Med ; 9(12): 1509-1530, 2020 12.
Article in English | MEDLINE | ID: mdl-32691994

ABSTRACT

Spinal cord injuries (SCIs) are associated with tremendous physical, social, and financial costs for millions of individuals and families worldwide. Rapid delivery of specialized medical and surgical care has reduced mortality; however, long-term functional recovery remains limited. Cell-based therapies represent an exciting neuroprotective and neuroregenerative strategy for SCI. This article summarizes the most promising preclinical and clinical cell approaches to date including transplantation of mesenchymal stem cells, neural stem cells, oligodendrocyte progenitor cells, Schwann cells, and olfactory ensheathing cells, as well as strategies to activate endogenous multipotent cell pools. Throughout, we emphasize the fundamental biology of cell-based therapies, critical features in the pathophysiology of spinal cord injury, and the strengths and limitations of each approach. We also highlight salient completed and ongoing clinical trials worldwide and the bidirectional translation of their findings. We then provide an overview of key adjunct strategies such as trophic factor support to optimize graft survival and differentiation, engineered biomaterials to provide a support scaffold, electrical fields to stimulate migration, and novel approaches to degrade the glial scar. We also discuss important considerations when initiating a clinical trial for a cell therapy such as the logistics of clinical-grade cell line scale-up, cell storage and transportation, and the delivery of cells into humans. We conclude with an outlook on the future of cell-based treatments for SCI and opportunities for interdisciplinary collaboration in the field.


Subject(s)
Nerve Regeneration/physiology , Neuroprotection/physiology , Spinal Cord Injuries/therapy , Humans , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
7.
Neurobiol Dis ; 143: 104995, 2020 09.
Article in English | MEDLINE | ID: mdl-32590037

ABSTRACT

Spinal cord injury (SCI) often results in permanent functional loss due to a series of degenerative events including cell death, axonal damage, and the upregulation of inhibitory proteins that impede regeneration. Repulsive Guidance Molecule A (RGMa) is a potent inhibitor of axonal growth that is rapidly upregulated following injury in both the rodent and human central nervous system (CNS). Previously, we showed that monoclonal antibodies that specifically block inhibitory RGMa signaling promote neuroprotective and regenerative effects when administered acutely in a clinically relevant rat model of thoracic SCI. However, it is unknown whether systemic administration of RGMa blocking antibodies are effective for SCI after delayed administration. Here, we administered elezanumab, a human monoclonal antibody targeting RGMa, intravenously either acutely or at 3 h or 24 h following thoracic clip impact-compression SCI. Rats treated with elezanumab acutely and at 3 h post-injury showed improvements in overground locomotion and fine motor function and gait. Rats treated 24 h post-SCI trended towards better recovery demonstrating significantly greater stride length and swing speed. Treated rats also showed greater tissue preservation with reduced lesion areas. As seen with acute treatment, delayed administration of elezanumab at 3 h post-SCI also increased perilesional neuronal sparing and serotonergic and corticospinal axonal plasticity. In addition, all elezanumab treated rats showed earlier spontaneous voiding ability and less post-trauma bladder wall hypertrophy. Together, our data demonstrate the therapeutic efficacy of delayed systemic administration of elezanumab in a rat model of SCI, and uncovers a new role for RGMa inhibition in bladder recovery following SCI.


Subject(s)
Antibodies, Monoclonal/administration & dosage , GPI-Linked Proteins/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Recovery of Function/drug effects , Spinal Cord Injuries/physiopathology , Animals , Female , Humans , Rats , Rats, Wistar , Urination/drug effects
8.
Stem Cells ; 38(2): 187-194, 2020 02.
Article in English | MEDLINE | ID: mdl-31648407

ABSTRACT

Nearly a century ago, the concept of the secondary injury in spinal cord trauma was first proposed to explain the complex cascade of molecular and cellular events leading to widespread neuronal and glial cell death after trauma. In recent years, it has been established that the ependymal region of the adult mammalian spinal cord contains a population of multipotent neural stem/progenitor cells (NSPCs) that are activated after spinal cord injury (SCI) and likely play a key role in endogenous repair and regeneration. How these cells respond to the various components of the secondary injury remains poorly understood. Emerging evidence suggests that many of the biochemical components of the secondary injury cascade which have classically been viewed as deleterious to host neuronal and glial cells may paradoxically trigger NSPC activation, proliferation, and differentiation thus challenging our current understanding of secondary injury mechanisms in SCI. Herein, we highlight new findings describing the response of endogenous NSPCs to spinal cord trauma, redefining the secondary mechanisms of SCI through the lens of the endogenous population of stem/progenitor cells. Moreover, we outline how these insights can fuel novel stem cell-based therapeutic strategies to repair the injured spinal cord.


Subject(s)
Neural Stem Cells/metabolism , Spinal Cord Injuries/physiopathology , Humans
9.
Neuroscientist ; 25(1): 8-21, 2019 02.
Article in English | MEDLINE | ID: mdl-29283022

ABSTRACT

The ideal biomarker for central nervous system (CNS) trauma in patients would be a molecular marker specific for injured nervous tissue that would provide a consistent and reliable assessment of the presence and severity of injury and the prognosis for recovery. One candidate biomarker is the protein tau, a microtubule-associated protein abundant in the axonal compartment of CNS neurons. Following axonal injury, tau becomes modified primarily by hyperphosphorylation of its various amino acid residues and cleavage into smaller fragments. These posttrauma products can leak into the cerebrospinal fluid or bloodstream and become candidate biomarkers of CNS injury. This review examines the primary molecular changes that tau undergoes following traumatic brain injury and spinal cord injury, and reviews the current literature in traumatic CNS biomarker research with a focus on the potential for hyperphosphorylated and cleaved tau as sensitive biomarkers of injury.


Subject(s)
Brain Injuries, Traumatic/diagnosis , Brain Injuries, Traumatic/metabolism , Protein Processing, Post-Translational , Spinal Cord Injuries/diagnosis , Spinal Cord Injuries/metabolism , tau Proteins/metabolism , Animals , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Humans , Phosphorylation
10.
J Neurotrauma ; 35(16): 1929-1941, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29644915

ABSTRACT

Current biomarker research in spinal cord injury (SCI) and traumatic brain injury has focused on a number of structural protein candidates, including the microtubule-associated protein tau. Evidence from models of traumatic brain injury has demonstrated that hyperphosphorylation of tau (p-tau) occurs in injured axons and demonstrates its utility as a biomarker for brain injury; however, the potential of p-tau as a biomarker for SCI is not yet known. Therefore, the present study determined whether tau is hyperphosphorylated in injured spinal cord axons, and then examined cerebrospinal fluid (CSF) and serum concentrations of p-tau and total-tau protein after a clinically relevant severe impact-compression SCI in rats. We found that severe SCI at T8 showed the presence of p-tau in damaged axons with a similar time course and distribution pattern to ß-APP, a biomarker of axonal injury. The presence of p-tau and ß-APP positive axons extended no farther than 5000 µm rostral and caudal to the injury epicenter, and was at its maximum at one day post-SCI. CSF levels of p-tau and total-tau significantly increased at one day post-SCI; however, only serum p-tau levels were significantly elevated in rats with SCI compared with naïve rats. These results suggest that CSF and serum p-tau may be a useful biomarker for severe traumatic SCI.


Subject(s)
Biomarkers/analysis , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , tau Proteins/analysis , Animals , Axons/metabolism , Axons/pathology , Female , Phosphorylation , Rats , Rats, Wistar , tau Proteins/metabolism
11.
Sci Rep ; 7(1): 10529, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28874746

ABSTRACT

Traumatic spinal cord injury (SCI) causes a cascade of degenerative events including cell death, axonal damage, and the upregulation of inhibitory molecules which prevent regeneration and limit recovery. Repulsive guidance molecule A (RGMa) is a potent neurite growth inhibitor in the central nervous system, exerting its repulsive activity by binding the Neogenin receptor. Here, we show for the first time that inhibitory RGMa is markedly upregulated in multiple cell types after clinically relevant impact-compression SCI in rats, and importantly, also in the injured human spinal cord. To neutralize inhibitory RGMa, clinically relevant human monoclonal antibodies were systemically administered after acute SCI, and were detected in serum, cerebrospinal fluid, and in the injured tissue. Rats treated with RGMa blocking antibodies showed significantly improved recovery of motor function and gait. Furthermore, RGMa blocking antibodies promoted neuronal survival, and enhanced the plasticity of descending serotonergic pathways and corticospinal tract axonal regeneration. RGMa antibody also attenuated neuropathic pain responses, which was associated with fewer activated microglia and reduced CGRP expression in the dorsal horn caudal to the lesion. These results show the therapeutic potential of the first human RGMa antibody for SCI and uncovers a new role for the RGMa/Neogenin pathway on neuropathic pain.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Membrane Proteins/immunology , Nerve Tissue Proteins/immunology , Neuralgia/therapy , Spinal Cord Injuries/therapy , Spinal Cord Regeneration , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Cells, Cultured , Female , GPI-Linked Proteins , Humans , Mice , Neuronal Plasticity , Rats , Rats, Wistar
12.
Stem Cells Dev ; 26(23): 1675-1681, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28950762

ABSTRACT

Transplantation of neural stem/progenitor cells (NSPCs) following spinal cord injury (SCI) is a promising strategy to enhance regeneration but is limited by poor survival of grafted cells. Determining methods to enhance survival of NSPCs is therefore essential. Positive modulation of AMPA receptors has been shown to enhance neurogenesis in various models of brain injury. Here we examined the effect of selective AMPA receptor modulation in adult rat spinal cord-derived NSPCs using a class of allosteric AMPA receptor modulators known as ampakines. NSPCs from the periventricular region of the adult rat spinal cord were treated with ampakines CX614 and CX546 for 72 h either alone or in the presence of low-dose glutamate (50 µM). Treatment with either agent in the presence of glutamate significantly increased cell survival and proliferation and reduced cell death. Moreover, ampakine/glutamate treatment reduced cell death in the setting of oxidative stress. Treatment with ampakines did not significantly alter cell phenotype. These findings offer important insight into a potential therapeutic strategy to positively regulate transplanted and endogenous adult spinal cord-derived NSPCs after SCI.


Subject(s)
Cell Death , Cell Proliferation , Dioxoles/pharmacology , Neural Stem Cells/drug effects , Oxazines/pharmacology , Piperidines/pharmacology , Receptors, AMPA/metabolism , Spinal Cord/cytology , Animals , Cells, Cultured , Female , Glutamic Acid/pharmacology , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Oxidative Stress , Rats , Rats, Wistar
13.
J Neurotrauma ; 34(6): 1209-1226, 2017 03 15.
Article in English | MEDLINE | ID: mdl-27775474

ABSTRACT

The immune system plays a critical and complex role in the pathobiology of spinal cord injury (SCI), exerting both beneficial and detrimental effects. Increasing evidence suggests that there are injury level-dependent differences in the immune response to SCI. Patients with traumatic SCI have elevated levels of circulating autoantibodies against components of the central nervous system, but the role of these antibodies in SCI outcomes remains unknown. In rodent models of mid-thoracic SCI, antibody-mediated autoimmunity appears to be detrimental to recovery. However, whether autoantibodies against the spinal cord are generated following cervical SCI (cSCI), the most common level of injury in humans, remains undetermined. To address this knowledge gap, we investigated the antibody responses following cSCI in a rat model of injury. We found increased immunoglobulin G (IgG) and IgM antibodies in the spinal cord in the subacute phase of injury (2 weeks), but not in more chronic phases (10 and 20 weeks). At 2 weeks post-cSCI, antibodies were detected at the injury epicenter and co-localized with the astroglial scar and neurons of the ventral horn. These increased levels of antibodies corresponded with enhanced activation of immune responses in the spleen. Higher counts of antibody-secreting cells were observed in the spleen of injured rats. Further, increased levels of secreted IgG antibodies and enhanced proliferation of T-cells in splenocyte cultures from injured rats were found. These findings suggest the potential development of autoantibody responses following cSCI in the rat. The impact of the post-traumatic antibody responses on functional outcomes of cSCI is a critical topic that requires further investigation.


Subject(s)
Autoantibodies/immunology , Cervical Cord/injuries , Spinal Cord Injuries/immunology , Animals , Antibody-Producing Cells/immunology , Astrocytes/immunology , Disease Models, Animal , Female , Rats , Rats, Wistar , Spleen/immunology
14.
Stem Cells Dev ; 25(16): 1223-33, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27316370

ABSTRACT

Traumatic spinal cord injury (SCI) leads to a cascade of secondary chemical insults, including oxidative stress and glutamate excitotoxicity, which damage host neurons and glia. Transplantation of exogenous neural stem/progenitor cells (NSPCs) has shown promise in enhancing regeneration after SCI, although survival of transplanted cells remains poor. Understanding the response of NSPCs to the chemical mediators of secondary injury is essential in finding therapies to enhance survival. We examined the in vitro effects of glutamate and glutamate receptor agonists on adult rat spinal cord-derived NSPCs. NSPCs isolated from the periventricular region of the adult rat spinal cord were exposed to various concentrations of glutamate for 96 h. We found that glutamate treatment (500 µM) for 96 h significantly increased live cell numbers, reduced cell death, and increased proliferation, but did not significantly alter cell phenotype. Concurrent glutamate treatment (500 µM) in the setting of H2O2 exposure (500 µM) for 10 h increased NSPC survival compared to H2O2 exposure alone. The effects of glutamate on NSPCs were blocked by the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)/kainate receptor antagonist GYKI-52466, but not by the N-methyl-D-aspartic acid receptor antagonist MK-801 or DL-AP5, or the mGluR3 antagonist LY-341495. Furthermore, treatment of NSPCs with AMPA, kainic acid, or the kainate receptor-specific agonist (RS)-2-amino-3-(3-hydroxy-5-tert-butylisoxazol-4-yl)propanoic acid mimicked the responses seen with glutamate both alone and in the setting of oxidative stress. These findings offer important insights into potential mechanisms to enhance NSPC survival and implicate a potential role for glutamate in promoting NSPC survival and proliferation after traumatic SCI.


Subject(s)
Adult Stem Cells/cytology , Glutamic Acid/pharmacology , Neural Stem Cells/cytology , Oxidative Stress/drug effects , Receptors, Ionotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord/cytology , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Aging , Animals , Brain/cytology , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Phenotype , Rats, Transgenic , Rats, Wistar , Receptors, AMPA/metabolism , Receptors, Ionotropic Glutamate/antagonists & inhibitors , Receptors, Kainic Acid/metabolism
15.
J Neurotrauma ; 33(3): 278-89, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26414192

ABSTRACT

Traumatic injury to the lumbar spinal cord results in complex central and peripheral nervous tissue damage causing significant neurobehavioral deficits and personal/social adversity. Although lumbar cord injuries are common in humans, there are few clinically relevant models of lumbar spinal cord injury (SCI). This article describes a novel lumbar SCI model in the rat. The effects of moderate (20 g), moderate-to-severe (26 g) and severe (35 g, and 56 g) clip impact-compression injuries at the lumbar spinal cord level L1-L2 (vertebral level T11-T12) were assessed using several neurobehavioral, neuroanatomical, and electrophysiological outcome measures. Lesions were generated after meticulous anatomical landmarking using microCT, followed by laminectomy and extradural inclusion of central and radicular elements to generate a traumatic SCI. Clinically relevant outcomes, such as MR and ultrasound imaging, were paired with robust morphometry. Analysis of the lesional tissue demonstrated that pronounced tissue loss and cavitation occur throughout the acute to chronic phases of injury. Behavioral testing revealed significant deficits in locomotion, with no evidence of hindlimb weight-bearing or hindlimb-forelimb coordination in any injured group. Evaluation of sensory outcomes revealed highly pathological alterations including mechanical allodynia and thermal hyperalgesia indicated by increasing avoidance responses and decreasing latency in the tail-flick test. Deficits in spinal tracts were confirmed by electrophysiology showing increased latency and decreased amplitude of both sensory and motor evoked potentials (SEP/MEP), and increased plantar H-reflex indicating an increase in motor neuron excitability. This is a comprehensive lumbar SCI model and should be useful for evaluation of translationally oriented pre-clinical therapies.


Subject(s)
Evoked Potentials, Motor/physiology , Evoked Potentials, Somatosensory/physiology , Hyperalgesia/physiopathology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Animals , Behavior, Animal , Disease Models, Animal , Female , H-Reflex/physiology , Hyperalgesia/etiology , Lumbar Vertebrae , Magnetic Resonance Imaging , Motor Activity/physiology , Rats, Wistar , Spinal Cord Injuries/complications , Spinal Cord Injuries/diagnostic imaging , Ultrasonography
16.
Int J Dev Neurosci ; 47(Pt B): 140-6, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26390954

ABSTRACT

Riluzole, a sodium/glutamate antagonist, has shown significant neuroprotective effects in experimental models of spinal cord injury (SCI) and is currently under clinical trial for patients with SCI. However, the effect of riluzole on adult spinal cord-derived NSPCs remains unknown. In this study, we examined the effects of riluzole on NSPC survival both in vitro and in vivo. NSPCs harvested from the adult rat spinal cord were exposed to riluzole (1-30 µM) either alone or in combination with hydrogen peroxide or glutamate in vitro. Measures of intracellular reactive oxygen species (ROS), cell viability and proliferation were assessed. To examine the effects of riluzole on transplanted NSPCs in vivo, a rodent clip compression model of SCI was used. One week after injury, NSPCs were transplanted into the spinal cord and rats received either riluzole or vehicle treatment for two weeks (similar to the clinically accepted dosing regimen) at which time cords were processed for analysis. Exposure to riluzole (≥ 10 µM) for more than 48 h in vitro reduced NSPC viability. Riluzole treatment (1-10 µM) did not significantly affect intracellular ROS levels or cell viability in the setting of in vitro oxidative stress. While glutamate (500 µM) exposure for 96 h significantly increased adult NSPC proliferation and survival, this response was not blocked by concurrent treatment with riluzole (1-10 µM) thus supporting the notion that the known anti-glutamatergic properties of riluzole are not mediated through direct inhibition of glutamate receptors. Furthermore, riluzole treatment did not impair the survival of transplanted NSPCs in a rodent model of SCI. These results suggest that although NSPCs may have a narrow tolerance to riluzole treatment in vitro, riluzole does not impair NSPC survival at doses that would be used clinically.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Neural Stem Cells/drug effects , Riluzole/pharmacology , Spinal Cord/cytology , Animals , Cell Count , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Female , Glutamic Acid/pharmacology , Hydro-Lyases/metabolism , Hydrogen Peroxide/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Oxidants/pharmacology , Rats , Rats, Transgenic , Rats, Wistar , Reactive Oxygen Species/metabolism , Time Factors
17.
Biores Open Access ; 4(1): 146-59, 2015.
Article in English | MEDLINE | ID: mdl-26309791

ABSTRACT

Transplantation of neural stem/progenitor cells (NSPCs) is a promising strategy in spinal cord injury (SCI). However, poor survival of transplanted stem cells remains a major limitation of this therapy due to the hostile environment of the injured cord. Oxidative stress is a hallmark in the pathogenesis of SCI; however, its effects on NSPCs from the adult spinal cord have yet to be examined. We therefore developed in vitro models of mild and severe oxidative stress of adult spinal cord-derived NSPCs and used these models to examine potential cell survival factors. NSPCs harvested from the adult rat spinal cord were treated with hydrogen peroxide (H2O2) in vitro to induce oxidative stress. A mild 4 h exposure to H2O2 (500 µM) significantly increased the level of intracellular reactive oxygen species with minimal effect on viability. In contrast, 24 h of oxidative stress led to a marked reduction in cell survival. Pretreatment with brain-derived neurotrophic factor (BDNF) for 48 h attenuated the increase in intracellular reactive oxygen species and enhanced survival. This survival effect was associated with a significant reduction in the number of apoptotic cells and a significant increase in the activity of the antioxidant enzymes glutathione reductase and superoxide dismutase. BDNF treatment had no effect on NSPC differentiation or proliferation. In contrast, cyclosporin A and thyrotropin-releasing hormone had minimal or no effect on NSPC survival. Thus, these models of in vitro oxidative stress may be useful for screening neuroprotective factors administered prior to transplantation to enhance survival of stem cell transplants.

18.
J Vis Exp ; (99): e52732, 2015 May 14.
Article in English | MEDLINE | ID: mdl-26067928

ABSTRACT

Adult rat and human spinal cord neural stem/progenitor cells (NSPCs) cultured in growth factor-enriched medium allows for the proliferation of multipotent, self-renewing, and expandable neural stem cells. In serum conditions, these multipotent NSPCs will differentiate, generating neurons, astrocytes, and oligodendrocytes. The harvested tissue is enzymatically dissociated in a papain-EDTA solution and then mechanically dissociated and separated through a discontinuous density gradient to yield a single cell suspension which is plated in neurobasal medium supplemented with epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), and heparin. Adult rat spinal cord NSPCs are cultured as free-floating neurospheres and adult human spinal cord NSPCs are grown as adherent cultures. Under these conditions, adult spinal cord NSPCs proliferate, express markers of precursor cells, and can be continuously expanded upon passage. These cells can be studied in vitro in response to various stimuli, and exogenous factors may be used to promote lineage restriction to examine neural stem cell differentiation. Multipotent NSPCs or their progeny can also be transplanted into various animal models to assess regenerative repair.


Subject(s)
Neural Stem Cells/cytology , Spinal Cord/cytology , Adolescent , Adult , Adult Stem Cells/cytology , Animals , Astrocytes/cytology , Cell Culture Techniques/methods , Cell Differentiation/physiology , Cell Separation/methods , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Multipotent Stem Cells/cytology , Neurons/cytology , Oligodendroglia/cytology , Rats , Tissue and Organ Harvesting/methods , Young Adult
19.
Exp Neurol ; 269: 154-68, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25902036

ABSTRACT

Large animal and primate models of spinal cord injury (SCI) are being increasingly utilized for the testing of novel therapies. While these represent intermediary animal species between rodents and humans and offer the opportunity to pose unique research questions prior to clinical trials, the role that such large animal and primate models should play in the translational pipeline is unclear. In this initiative we engaged members of the SCI research community in a questionnaire and round-table focus group discussion around the use of such models. Forty-one SCI researchers from academia, industry, and granting agencies were asked to complete a questionnaire about their opinion regarding the use of large animal and primate models in the context of testing novel therapeutics. The questions centered around how large animal and primate models of SCI would be best utilized in the spectrum of preclinical testing, and how much testing in rodent models was warranted before employing these models. Further questions were posed at a focus group meeting attended by the respondents. The group generally felt that large animal and primate models of SCI serve a potentially useful role in the translational pipeline for novel therapies, and that the rational use of these models would depend on the type of therapy and specific research question being addressed. While testing within these models should not be mandatory, the detection of beneficial effects using these models lends additional support for translating a therapy to humans. These models provides an opportunity to evaluate and refine surgical procedures prior to use in humans, and safety and bio-distribution in a spinal cord more similar in size and anatomy to that of humans. Our results reveal that while many feel that these models are valuable in the testing of novel therapies, important questions remain unanswered about how they should be used and how data derived from them should be interpreted.


Subject(s)
Spinal Cord Injuries , Translational Research, Biomedical , Animals , Cell- and Tissue-Based Therapy/methods , Disease Models, Animal , Focus Groups , Humans , Primates , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/therapy , Surveys and Questionnaires , Translational Research, Biomedical/methods
20.
Cell Rep ; 8(4): 1146-59, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25127134

ABSTRACT

Ideal strategies to ameliorate CNS damage should promote both neuronal survival and axon regeneration. The receptor Neogenin promotes neuronal apoptosis. Its ligand prevents death, but the resulting repulsive guidance molecule a (RGMa)-Neogenin interaction also inhibits axonal growth, countering any prosurvival benefits. Here, we explore strategies to inhibit Neogenin, thus simultaneously enhancing survival and regeneration. We show that bone morphogenetic protein (BMP) and RGMa-dependent recruitment of Neogenin into lipid rafts requires an interaction between RGMa and Neogenin subdomains. RGMa or Neogenin peptides that prevent this interaction, BMP inhibition by Noggin, or reduction of membrane cholesterol all block Neogenin raft localization, promote axon outgrowth, and prevent neuronal apoptosis. Blocking Neogenin raft association influences axonal pathfinding, enhances survival in the developing CNS, and promotes survival and regeneration in the injured adult optic nerve and spinal cord. Moreover, lowering cholesterol disrupts rafts and restores locomotor function after spinal cord injury. These data reveal a unified strategy to promote both survival and regeneration in the CNS.


Subject(s)
Membrane Microdomains/physiology , Nerve Regeneration , Animals , Axons/physiology , Chickens , Female , Growth Cones/physiology , Macrolides , Membrane Proteins/metabolism , Motor Neurons/physiology , Nerve Tissue Proteins/physiology , Optic Nerve/pathology , Optic Nerve/physiopathology , Rats, Wistar , Retinal Ganglion Cells/physiology , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Cord Injuries/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL