Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
JAMA Facial Plast Surg ; 21(5): 426-433, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31219545

ABSTRACT

IMPORTANCE: Aberrant synkinetic movement after facial nerve injury can lead to prominent facial asymmetry and resultant psychological distress. The current practices of neuroinhibition to promote greater facial symmetry are often temporary in nature and require repeated procedures. OBJECTIVE: To determine whether myelin-associated glycoprotein (MAG), a specific neuroinhibitor, can prevent neuroregeneration with efficacy comparable with that of vincristine, a well-established neurotoxin. DESIGN, SETTING, AND PARTICIPANTS: Rats transgenic for Thy-1 cell surface antigen-green fluorescent protein (Thy1-Gfp) were randomized into 3 groups. Each rat received bilateral crush axotomy injuries to the buccal and marginal mandibular branches of the facial nerves. The animals received intraneural injection of saline, MAG, or vincristine. MAIN OUTCOMES AND MEASURES: The animals were imaged via fluorescent microscopy at weeks 1, 3, 4, and 5 after surgery. Quantitative fluorescent data were generated as mean intensities of nerve segments proximal and distal to the axotomy site. Electrophysiological analysis, via measurement of compound muscle action potentials, was performed at weeks 0, 3, 4, and 5 after surgery. RESULTS: A total of 12 rats were included in the study. Administration of MAG significantly reduced fluorescent intensity of the distal nerve in comparison with the control group at week 3 (mean [SD], MAG group: 94 [11] intensity units vs control group: 130 [11] intensity units; P < .001), week 4 (MAG group: 81 [19] intensity units vs control group: 103 [9] intensity units; P = .004), and week 5 (MAG group: 76 [10] intensity units vs control group: 94 [10] intensity units; P < .001). In addition, rats treated with MAG had greater fluorescent intensity than those treated with vincristine at week 3 (mean [SD], MAG group: 94 [11] intensity units vs vincristine group: 76 [6] intensity units; P = .03), although there was no significant difference for weeks 4 and 5. At week 5, both MAG and vincristine demonstrated lower distal nerve to proximal nerve intensity ratios than the control group (control group, 0.94; vs MAG group, 0.82; P = .01; vs vincristine group; 0.77; P < .001). There was no significant difference in amplitude between the experimental groups at week 5 of electrophysiological testing. CONCLUSIONS AND RELEVANCE: Lower facial asymmetry and synkinesis are common persistent concerns to patients after facial nerve injury. Using the Thy1-Gfp rat, this study demonstrates effective inhibition of neuroregeneration via intraneural application of MAG in a crush axotomy model, comparable with results with vincristine. By potentially avoiding systemic toxic effects of vincristine, MAG demonstrates potential as an inhibitor of neural regeneration for patients with synkinesis. LEVEL OF EVIDENCE: NA.


Subject(s)
Facial Nerve , Myelin-Associated Glycoprotein , Synkinesis , Vincristine , Animals , Rats , Disease Models, Animal , Facial Nerve/drug effects , Facial Nerve/surgery , Myelin-Associated Glycoprotein/pharmacology , Rats, Transgenic , Synkinesis/drug therapy , Synkinesis/surgery , Vincristine/pharmacology
2.
J Cell Biol ; 218(6): 1871-1890, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31068376

ABSTRACT

Inhibition of histone deacetylase 6 (HDAC6) was shown to support axon growth on the nonpermissive substrates myelin-associated glycoprotein (MAG) and chondroitin sulfate proteoglycans (CSPGs). Though HDAC6 deacetylates α-tubulin, we find that another HDAC6 substrate contributes to this axon growth failure. HDAC6 is known to impact transport of mitochondria, and we show that mitochondria accumulate in distal axons after HDAC6 inhibition. Miro and Milton proteins link mitochondria to motor proteins for axon transport. Exposing neurons to MAG and CSPGs decreases acetylation of Miro1 on Lysine 105 (K105) and decreases axonal mitochondrial transport. HDAC6 inhibition increases acetylated Miro1 in axons, and acetyl-mimetic Miro1 K105Q prevents CSPG-dependent decreases in mitochondrial transport and axon growth. MAG- and CSPG-dependent deacetylation of Miro1 requires RhoA/ROCK activation and downstream intracellular Ca2+ increase, and Miro1 K105Q prevents the decrease in axonal mitochondria seen with activated RhoA and elevated Ca2+ These data point to HDAC6-dependent deacetylation of Miro1 as a mediator of axon growth inhibition through decreased mitochondrial transport.


Subject(s)
Histone Deacetylase 6/genetics , Mitochondria/metabolism , Neurons/metabolism , rho GTP-Binding Proteins/genetics , rho-Associated Kinases/genetics , Acetylation/drug effects , Animals , Axonal Transport/drug effects , Axonal Transport/genetics , Calcium/metabolism , Chondroitin Sulfate Proteoglycans/pharmacology , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Gene Expression Regulation , Histone Deacetylase 6/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Myelin-Associated Glycoprotein/pharmacology , Neurons/cytology , Neurons/drug effects , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Signal Transduction , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
3.
J Neurosci ; 34(21): 7165-78, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24849351

ABSTRACT

Extracellular molecular cues guide migrating growth cones along specific routes during development of axon tracts. Such processes rely on asymmetric elevation of cytosolic Ca(2+) concentrations across the growth cone that mediates its attractive or repulsive turning toward or away from the side with Ca(2+) elevation, respectively. Downstream of these Ca(2+) signals, localized activation of membrane trafficking steers the growth cone bidirectionally, with endocytosis driving repulsion and exocytosis causing attraction. However, it remains unclear how Ca(2+) can differentially regulate these opposite membrane-trafficking events. Here, we show that growth cone turning depends on localized imbalance between exocytosis and endocytosis and identify Ca(2+)-dependent signaling pathways mediating such imbalance. In embryonic chicken dorsal root ganglion neurons, repulsive Ca(2+) signals promote clathrin-mediated endocytosis through a 90 kDa splice variant of phosphatidylinositol-4-phosphate 5-kinase type-1γ (PIPKIγ90). In contrast, attractive Ca(2+) signals facilitate exocytosis but suppress endocytosis via Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and cyclin-dependent kinase 5 (Cdk5) that can inactivate PIPKIγ90. Blocking CaMKII or Cdk5 leads to balanced activation of both exocytosis and endocytosis that causes straight growth cone migration even in the presence of guidance signals, whereas experimentally perturbing the balance restores the growth cone's turning response. Remarkably, the direction of this resumed turning depends on relative activities of exocytosis and endocytosis, but not on the type of guidance signals. Our results suggest that navigating growth cones can be redirected by shifting the imbalance between exocytosis and endocytosis, highlighting the importance of membrane-trafficking imbalance for axon guidance and, possibly, for polarized cell migration in general.


Subject(s)
Endocytosis/physiology , Exocytosis/physiology , Growth Cones/physiology , Neurons/cytology , Animals , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cells, Cultured , Chick Embryo , Clathrin/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Endocytosis/drug effects , Enzyme Inhibitors/pharmacology , Exocytosis/drug effects , Ganglia, Spinal/cytology , Growth Cones/drug effects , Myelin-Associated Glycoprotein/pharmacology , Neurons/drug effects , Organophosphonates/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Photolysis , Piperazines/pharmacology , Vesicle-Associated Membrane Protein 2/metabolism
4.
J Biol Chem ; 288(29): 20837-20842, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23775074

ABSTRACT

Negatively targeting the tumor suppressor and phosphoinositide phosphatase PTEN (phosphatase and tensin homologue) promotes axon regrowth after injury. How PTEN functions in axon guidance has remained unknown. Here we report the differential role of PTEN in chemotactic guidance of axonal growth cones. Down-regulating PTEN expression in Xenopus laevis spinal neurons selectively abolished growth cone chemorepulsion but permitted chemoattraction. These findings persisted during cAMP-dependent switching of turning behaviors. Live cell imaging using a GFP biosensor revealed rapid PTEN-dependent depression of phosphatidylinositol 3,4,5-trisphosphate levels in the growth cone induced by the repellent myelin-associated glycoprotein. Moreover, down-regulating PTEN expression blocked negative remodeling of ß1-integrin adhesions triggered by myelin-associated glycoprotein, yet permitted integrin clustering by a positive chemotropic treatment. Thus, PTEN negatively regulates growth cone phosphatidylinositol 3,4,5-trisphosphate levels and mediates chemorepulsion, whereas chemoattraction is PTEN-independent. Regenerative therapies targeting PTEN may therefore suppress growth cone repulsion to soluble cues while permitting attractive guidance, an essential feature for re-forming functional neural circuits.


Subject(s)
Chemotaxis , Growth Cones/enzymology , Phosphoric Monoester Hydrolases/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/metabolism , Animals , Chemotaxis/drug effects , Cluster Analysis , Cyclic AMP/pharmacology , Down-Regulation/drug effects , Endocytosis/drug effects , Growth Cones/drug effects , Integrin beta1/metabolism , Myelin-Associated Glycoprotein/pharmacology , Phosphatidylinositol Phosphates/metabolism
5.
PLoS One ; 8(3): e57539, 2013.
Article in English | MEDLINE | ID: mdl-23469201

ABSTRACT

Zebrafish (Danio rerio) is a widely used model organism in genetics and developmental biology research. Genetic screens have proven useful for studying embryonic development of the nervous system in vivo, but in vitro studies utilizing zebrafish have been limited. Here, we introduce a robust zebrafish primary neuron culture system for functional nerve growth and guidance assays. Distinct classes of central nervous system neurons from the spinal cord, hindbrain, forebrain, and retina from wild type zebrafish, and fluorescent motor neurons from transgenic reporter zebrafish lines, were dissociated and plated onto various biological and synthetic substrates to optimize conditions for axon outgrowth. Time-lapse microscopy revealed dynamically moving growth cones at the tips of extending axons. The mean rate of axon extension in vitro was 21.4±1.2 µm hr(-1) s.e.m. for spinal cord neurons, which corresponds to the typical ∼0.5 mm day(-1) growth rate of nerves in vivo. Fluorescence labeling and confocal microscopy demonstrated that bundled microtubules project along axons to the growth cone central domain, with filamentous actin enriched in the growth cone peripheral domain. Importantly, the growth cone surface membrane expresses receptors for chemotropic factors, as detected by immunofluorescence microscopy. Live-cell functional assays of axon extension and directional guidance demonstrated mammalian brain-derived neurotrophic factor (BDNF)-dependent stimulation of outgrowth and growth cone chemoattraction, whereas mammalian myelin-associated glycoprotein inhibited outgrowth. High-resolution live-cell Ca(2+)-imaging revealed local elevation of cytoplasmic Ca(2+) concentration in the growth cone induced by BDNF application. Moreover, BDNF-induced axon outgrowth, but not basal outgrowth, was blocked by treatments to suppress cytoplasmic Ca(2+) signals. Thus, this primary neuron culture model system may be useful for studies of neuronal development, chemotropic axon guidance, and mechanisms underlying inhibition of neural regeneration in vitro, and complement observations made in vivo.


Subject(s)
Axons/ultrastructure , Calcium/metabolism , Motor Neurons/cytology , Primary Cell Culture/methods , Zebrafish/growth & development , Animals , Animals, Genetically Modified , Axons/drug effects , Brain-Derived Neurotrophic Factor/pharmacology , Calcium Signaling/drug effects , Gene Expression Regulation, Developmental/drug effects , Microscopy, Fluorescence , Motor Neurons/drug effects , Myelin-Associated Glycoprotein/pharmacology , Prosencephalon/cytology , Prosencephalon/drug effects , Prosencephalon/growth & development , Retina/cytology , Retina/drug effects , Retina/growth & development , Rhombencephalon/cytology , Rhombencephalon/drug effects , Rhombencephalon/growth & development , Spinal Cord/cytology , Spinal Cord/drug effects , Spinal Cord/growth & development , Time-Lapse Imaging , Zebrafish/anatomy & histology , Zebrafish/genetics
6.
Neuron ; 74(3): 490-503, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22578501

ABSTRACT

Correct guidance of axons to their targets depends on an intricate network of signaling molecules in the growth cone. Calcium and cAMP are two key regulators of whether axons are attracted or repelled by molecular gradients, but how these molecules interact to determine guidance responses remains unclear. Here, we constructed a mathematical model for the relevant signaling network, which explained a large range of previous biological data and made predictions for when axons will be attracted or repelled. We then confirmed these predictions experimentally, in particular showing that while small increases in cAMP levels promote attraction large increases do not, and that under some circumstances reducing cAMP levels promotes attraction. Together, these results show that a relatively simple mathematical model can quantitatively predict guidance decisions across a wide range of conditions, and that calcium and cAMP levels play a more complex role in these decisions than previously determined.


Subject(s)
Axons/physiology , Calcium/metabolism , Chemotaxis/physiology , Cyclic AMP/metabolism , Neurons/cytology , Animals , Animals, Newborn , Axons/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Carbazoles/pharmacology , Cells, Cultured , Chemotaxis/drug effects , Computer Simulation , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Growth Cones/drug effects , Growth Cones/physiology , Models, Neurological , Myelin-Associated Glycoprotein/pharmacology , Nerve Growth Factor/pharmacology , Neurites/drug effects , Neurites/physiology , Neurons/drug effects , Potassium/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , Superior Cervical Ganglion/cytology , Thionucleotides/pharmacology
7.
Neurochem Res ; 36(12): 2363-72, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21822922

ABSTRACT

NogoA, myelin-associated glycoprotein (MAG) and oligodendrocyte myelin glycoprotein are CNS myelin molecules that bind to the neuronal Nogo-66 receptor (NgR) and inhibit axon growth. The NgR antagonist, soluble NgR1-Fc protein (sNgR-Fc), facilitates axon regeneration by neutralizing the inhibitory effects of myelin proteins in experimental models of CNS injury. Here we aim to investigate the effect of sNgR-Fc on the proliferation of neural progenitor cells (NPCs). The hippocampus cells of embryonic rats were isolated and cultured in vitro. The expression of nestin, ßIII-Tubulin, GFAP and Nogo-A on these cells was observed using immunocytochemistry. In order to investigate the effect on proliferation of NPCs, sNgR-Fc, MAG-Fc chimera and Notch1 blocker were added respectively. The total cell number for the proliferated NPCs was counted. BrdU was applied and the rate of proliferating cells was examined. The level of Notch1 was analyzed using Western blotting. We identified that NogoA is expressed in NPCs. sNgR-Fc significantly enhanced the proliferation of NPCs in vitro as indicated by BrdU labeling and total cell count. This proliferation effect was abolished by the administration of MAG suggesting specificity. In addition, we demonstrate that sNgR-Fc is a potent activator for Notch1 and Notch1 antagonist reversed the effect of sNgR-Fc on NPC proliferation. Our results suggest that sNgR-Fc may modulate Nogo activity to induce NPC proliferation via the Notch pathway.


Subject(s)
Hippocampus/cytology , Myelin Proteins/metabolism , Myelin-Associated Glycoprotein/pharmacology , Receptor, Notch1/physiology , Recombinant Fusion Proteins/pharmacology , Stem Cells/physiology , Animals , Cell Proliferation/drug effects , Myelin Proteins/biosynthesis , Myelin-Oligodendrocyte Glycoprotein , Nogo Proteins , Rats , Stem Cells/drug effects
8.
J Biol Chem ; 286(38): 33310-21, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21799010

ABSTRACT

Decreased expression of prosurvival and progrowth-stimulatory pathways, in addition to an environment that inhibits neuronal growth, contribute to the limited regenerative capacity in the central nervous system following injury or neurodegeneration. Membrane/lipid rafts, plasmalemmal microdomains enriched in cholesterol, sphingolipids, and the protein caveolin (Cav) are essential for synaptic development/stabilization and neuronal signaling. Cav-1 concentrates glutamate and neurotrophin receptors and prosurvival kinases and regulates cAMP formation. Here, we show that primary neurons that express a synapsin-driven Cav-1 vector (SynCav1) have increased raft formation, neurotransmitter and neurotrophin receptor expression, NMDA- and BDNF-mediated prosurvival kinase activation, agonist-stimulated cAMP formation, and dendritic growth. Moreover, expression of SynCav1 in Cav-1 KO neurons restores NMDA- and BDNF-mediated signaling and enhances dendritic growth. The enhanced dendritic growth occurred even in the presence of inhibitory cytokines (TNFα, IL-1ß) and myelin-associated glycoproteins (MAG, Nogo). Targeting of Cav-1 to neurons thus enhances prosurvival and progrowth signaling and may be a novel means to repair the injured and neurodegenerative brain.


Subject(s)
Caveolin 1/metabolism , Neurons/metabolism , Signal Transduction , Animals , Axons/drug effects , Axons/metabolism , Cell Survival/drug effects , Cells, Cultured , Cholesterol/metabolism , Cytokines/pharmacology , Dendrites/drug effects , Dendrites/metabolism , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Associated Glycoprotein/pharmacology , Neurons/cytology , Neurons/drug effects , Organ Specificity/drug effects , Signal Transduction/drug effects , Synapsins/metabolism
9.
J Immunol ; 186(2): 675-84, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21160051

ABSTRACT

The ability of IFN-ß to induce IL-10 production from innate immune cells is important for its anti-inflammatory properties and is believed to contribute to its therapeutic value in treating multiple sclerosis patients. In this study, we identified that IFN-ß stimulates IL-10 production by activating the JAK1- and PI3K-signaling pathways. JAK1 activity was required for IFN-ß to activate PI3K and Akt1 that resulted in repression of glycogen synthase kinase 3 (GSK3)-ß activity. IFN-ß-mediated suppression of GSK3-ß promoted IL-10, because IL-10 production by IFN-ß-stimulated dendritic cells (DC) expressing an active GSK3-ß knockin was severely reduced, whereas pharmacological or genetic inhibition of GSK3-ß augmented IL-10 production. IFN-ß increased the phosphorylated levels of CREB and STAT3 but only CREB levels were affected by PI3K. Also, a knockdown in CREB, but not STAT3, affected the capacity of IFN-ß to induce IL-10 from DC. IL-10 production by IFN-ß-stimulated DC was shown to suppress IFN-γ and IL-17 production by myelin oligodendrocyte glycoprotein-specific CD4(+) T cells, and this IL-10-dependent anti-inflammatory effect was enhanced by directly targeting GSK3 in DC. These findings highlight how IFN-ß induces IL-10 production and the importance that IL-10 plays in its anti-inflammatory properties, as well as identify a therapeutic target that could be used to increase the IL-10-dependent anti-inflammatory properties of IFN-ß.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Glycogen Synthase Kinase 3/physiology , Interferon-beta/physiology , Interleukin-10/biosynthesis , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Dendritic Cells/enzymology , Enzyme Activation/genetics , Enzyme Activation/immunology , Epitopes, T-Lymphocyte/immunology , Gene Knock-In Techniques , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Interleukin-10/physiology , Interleukin-17/antagonists & inhibitors , Interleukin-17/biosynthesis , Intracellular Fluid/enzymology , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Janus Kinase 1/metabolism , Janus Kinase 1/physiology , Mice , Mice, Inbred C57BL , Myelin Proteins , Myelin-Associated Glycoprotein/immunology , Myelin-Associated Glycoprotein/pharmacology , Myelin-Oligodendrocyte Glycoprotein , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/immunology
10.
J Neuroinflammation ; 7: 60, 2010 Oct 07.
Article in English | MEDLINE | ID: mdl-20929574

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) is the endpoint of a complex and still poorly understood process which results in inflammation, demyelination and axonal and neuronal degeneration. Since the first description of MS, psychological stress has been suggested to be one of the trigger factors in the onset and/or relapse of symptoms. However, data from animal models of MS, such as experimental autoimmune encephalomyelitis (EAE) are inconsistent and the effect of stress on EAE onset and severity depends on duration and time of application of the stress protocol and the underlying mechanisms. METHODS: Dark Agouti rats were inoculated with MOG/CFA to induce EAE, and an immobilisation stress protocol with two different durations (12 and 21 days, starting at the moment of MOG-inoculation) was applied in order to analyse the effect of stress on disease onset and neuroinflammation. RESULTS: Twelve days of stress exposure increased EAE clinical score in Dark Agouti rats. In addition, these animals presented higher levels of MMP-9 and proinflammatory PGE2 in spinal cord. In contrast, animals chronically exposed to stress (21 days) showed a significantly lower incidence of EAE clinical signs and reduced myelin loss, leukocyte infiltration and accumulation of inflammatory/oxidative mediators in spinal cord. Interestingly, chronically stressed animals showed a parallel increase in levels of the anti-inflammatory prostaglandin 15d-PGJ2, the main endogenous agonist of PPARγ. CONCLUSIONS: Our results demonstrate that, depending on duration, stress exposure elicits opposite effects on PGE2/15d-PGJ2 ratios in spinal cord of EAE-induced Dark Agouti rats. Further studies are needed to elucidate if these changes in prostaglandin balance are sufficient to mediate the differences in clinical score and inflammation here reported, and to establish the potential utility of pharmacological intervention in MS directed toward anti-inflammatory pathways.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Myelin-Associated Glycoprotein/pharmacology , Spinal Cord/pathology , Stress, Physiological , Analysis of Variance , Animals , Blotting, Western , Corticosterone/blood , Dinoprostone/metabolism , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Lipid Peroxidation , Male , Myelin Proteins , Myelin-Associated Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein , Peroxidase/metabolism , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/metabolism , Radioimmunoassay , Rats , Restraint, Physical , Spinal Cord/immunology , Spinal Cord/metabolism
11.
Proc Natl Acad Sci U S A ; 107(45): 19555-60, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-20978211

ABSTRACT

The neuropeptide vasoactive intestinal peptide (VIP) has been shown to inhibit macrophage proinflammatory actions, promote a positive Th2/Th1 balance, and stimulate regulatory T-cell production. The fact that this peptide is highly efficacious in animal models of inflammatory diseases such as collagen-induced arthritis and experimental autoimmune encephalomyelitis (EAE) suggests that the endogenous peptide might normally provide protection against such pathologies. We thus studied the response of VIP-deficient (i.e., VIP KO) mice to myelin oligodendrocyte protein-induced EAE. Surprisingly, VIP KO mice were almost completely resistant to EAE, with delayed onset and mild or absent clinical profile. Despite this, flow cytometric analyses and antigen-rechallenge experiments indicated that myelin oligodendrocyte protein-treated VIP KO mice exhibited robust Th1/Th17 cell inductions and antigen-specific proliferation and cytokine responses. Moreover, adoptive transfer of lymphocytes from immunized VIP KO mice to WT recipients resulted in full-blown EAE, supporting their encephalitogenic potential. In contrast, transfer of encephalitogenic WT cells to VIP KO hosts did not produce EAE, suggesting that loss of VIP specifically affected the effector phase of the disease. Histological analyses indicated that CD4 T cells entered the meningeal and perivascular areas of VIP-deficient mice, but that parenchymal infiltration was strongly impaired. Finally, VIP pretreatment of VIP KO mice before immunization was able to restore their sensitivity to EAE. These results indicate that VIP plays an unanticipated permissive and/or proinflammatory role in the propagation of the inflammatory response in the CNS, a finding with potential therapeutic relevance in autoimmune neuroinflammatory diseases such as multiple sclerosis.


Subject(s)
Cell Movement/immunology , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , T-Lymphocytes/immunology , Vasoactive Intestinal Peptide/deficiency , Vasoactive Intestinal Peptide/immunology , Animals , Autoimmune Diseases/etiology , Cytokines/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/etiology , Inflammation/etiology , Lymphocyte Activation , Mice , Mice, Knockout , Myelin-Associated Glycoprotein/pharmacology , Th1 Cells , Th17 Cells
12.
J Neurosci ; 30(37): 12432-45, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20844138

ABSTRACT

In the adult mammalian CNS, the growth inhibitors oligodendrocyte-myelin glycoprotein (OMgp) and the reticulon RTN4 (Nogo) are broadly expressed in oligodendrocytes and neurons. Nogo and OMgp complex with the neuronal cell surface receptors Nogo receptor-1 (NgR1) and paired Ig-like receptor-B (PirB) to regulate neuronal morphology. In the healthy CNS, NgR1 regulates dendritic spine shape and attenuates activity-driven synaptic plasticity at Schaffer collateral-CA1 synapses. Here, we examine whether Nogo and OMgp influence functional synaptic plasticity, the efficacy by which synaptic transmission occurs. In acute hippocampal slices of adult mice, Nogo-66 and OMgp suppress NMDA receptor-dependent long-term potentiation (LTP) when locally applied to Schaffer collateral-CA1 synapses. Neither Nogo-66 nor OMgp influences basal synaptic transmission or paired-pulse facilitation, a form of short-term synaptic plasticity. PirB(-/-) and NgR1(-/-) single mutants and NgR1(-/-);PirB(-/-) double mutants show normal LTP, indistinguishable from wild-type controls. In juvenile mice, LTD in NgR1(-/-), but not PirB(-/-), slices is absent. Mechanistic studies revealed that Nogo-66 and OMgp suppress LTP in an NgR1-dependent manner. OMgp inhibits LTP in part through PirB but independently of p75. This suggests that NgR1 and PirB participate in ligand-dependent inhibition of synaptic plasticity. Loss of NgR1 leads to increased phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), signaling intermediates known to regulate neuronal growth and synaptic function. In primary cortical neurons, BDNF elicited phosphorylation of AKT and p70S6 kinase is attenuated in the presence of myelin inhibitors. Collectively, we provide evidence that mechanisms of neuronal growth inhibition and inhibition of synaptic strength are related. Thus, myelin inhibitors and their receptors may coordinate structural and functional neuronal plasticity in CNS health and disease.


Subject(s)
Down-Regulation/physiology , Myelin Proteins/physiology , Myelin-Associated Glycoprotein/physiology , Neural Inhibition/physiology , Neuronal Plasticity/physiology , Animals , Cell Line , Down-Regulation/genetics , GPI-Linked Proteins , Humans , Long-Term Potentiation/genetics , Long-Term Potentiation/physiology , Mice , Mice, Knockout , Mice, Transgenic , Myelin Proteins/genetics , Myelin Proteins/metabolism , Myelin-Associated Glycoprotein/genetics , Myelin-Associated Glycoprotein/pharmacology , Myelin-Oligodendrocyte Glycoprotein , Neural Inhibition/genetics , Neuronal Plasticity/genetics , Nogo Proteins , Nogo Receptor 1 , Rats , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/physiology , Receptors, Immunologic/deficiency , Receptors, Immunologic/metabolism , Receptors, Immunologic/physiology
13.
Exp Mol Pathol ; 89(2): 126-34, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20599940

ABSTRACT

IL-12Rß2(-/-) mice, which are unresponsive to IL-12, develop severe experimental autoimmune encephalomyelitis (EAE). The mechanisms for enhanced autoimmunity are incompletely understood. We report that in IL-12Rß2(-/-) mice, thymocytes undergo markedly accelerated maturation. This occurs at the transition from a double positive (DP) to a single positive (SP) phenotype, resulting in higher numbers of CD4 and CD8 SP cells, and to a lesser extent at the transition from double negative (DN) to DP cells. Accelerated maturation is observed in mice injected with anti-CD3 to mimic pre-T-cell receptor stimulation, and also in mice immunized with myelin oligodendrocyte glycoprotein (MOG) peptide to induce EAE.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Animals , CD3 Complex/immunology , Cells, Cultured , Disease Susceptibility , Female , Interleukin-12/immunology , Lymphocyte Activation , Mice , Mice, Knockout , Myelin Proteins , Myelin-Associated Glycoprotein/pharmacology , Myelin-Oligodendrocyte Glycoprotein , Thymus Gland/immunology
14.
J Neurochem ; 113(5): 1331-42, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20345749

ABSTRACT

One of the major barriers to successful axon regeneration in the adult CNS is the presence of inhibitory molecules that originate from the myelin sheath and glial scar. So far, only a small number of pharmacological compounds have exhibited functional activity against CNS inhibitors in promoting axon regeneration after injury. To search for novel compounds that enhance neurite outgrowth in vitro, we initiated a screen of a collection of natural products. We identified four compounds with the potential to promote growth over a myelin substrate. Of these, Amphotericin B (AmB) was shown to enhance neurite outgrowth and antagonize activities of major myelin associated inhibitors and glial-scar-derived chondroitin sulfate proteoglycans. AmB was found to activate Akt and thereby suppress the activity of glycogen synthase kinase 3 beta. Also, a cell permeable peptide that inhibits Akt activity was shown to block the effect of AmB in promoting axonal growth, while another peptide that increases Akt activity stimulated axonal growth in the presence of the myelin associated inhibitors. Our results suggest that AmB can promote neurite outgrowth over a wide range of inhibitory substrates via a mechanism that involves activation of Akt.


Subject(s)
Amphotericin B/pharmacology , Antifungal Agents/pharmacology , Axons/drug effects , Biological Products/pharmacology , Neurons/drug effects , Oncogene Protein v-akt/metabolism , Animals , Blotting, Western , Cell Proliferation/drug effects , Chondroitin Sulfate Proteoglycans/antagonists & inhibitors , Chondroitin Sulfate Proteoglycans/pharmacology , Drug Evaluation, Preclinical , Myelin-Associated Glycoprotein/antagonists & inhibitors , Myelin-Associated Glycoprotein/pharmacology , Nerve Regeneration/drug effects , Neurites/drug effects , Principal Component Analysis , Rats , Signal Transduction/drug effects
15.
J Neurosci ; 30(2): 739-48, 2010 Jan 13.
Article in English | MEDLINE | ID: mdl-20071539

ABSTRACT

An ideal therapeutic for stroke or spinal cord injury should promote survival and regeneration in the CNS. Arginase 1 (Arg1) has been shown to protect motor neurons from trophic factor deprivation and allow sensory neurons to overcome neurite outgrowth inhibition by myelin proteins. To identify small molecules that capture Arg1's protective and regenerative properties, we screened a hippocampal cell line stably expressing the proximal promoter region of the arginase 1 gene fused to a reporter gene against a library of compounds containing clinically approved drugs. This screen identified daidzein as a transcriptional inducer of Arg1. Both CNS and PNS neurons primed in vitro with daidzein overcame neurite outgrowth inhibition from myelin-associated glycoprotein, which was mirrored by acutely dissociated and cultured sensory neurons primed in vivo by intrathecal or subcutaneous daidzein infusion. Further, daidzein was effective in promoting axonal regeneration in vivo in an optic nerve crush model when given intraocularly without lens damage, or most importantly, when given subcutaneously after injury. Mechanistically, daidzein requires transcription and induction of Arg1 activity for its ability to overcome myelin inhibition. In contrast to canonical Arg1 activators, daidzein increases Arg1 without increasing CREB phosphorylation, suggesting its effects are cAMP-independent. Accordingly, it may circumvent known CNS side effects of some cAMP modulators. Indeed, daidzein appears to be safe as it has been widely consumed in soy products, crosses the blood-brain barrier, and is effective without pretreatment, making it an ideal candidate for development as a therapeutic for spinal cord injury or stroke.


Subject(s)
Arginase/genetics , Cyclic AMP/metabolism , Isoflavones/pharmacology , Nerve Regeneration/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Promoter Regions, Genetic/physiology , Analysis of Variance , Animals , Animals, Newborn , Arginase/metabolism , CHO Cells , Cells, Cultured , Cerebellum/cytology , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , GAP-43 Protein/metabolism , Ganglia, Spinal/cytology , High-Throughput Screening Assays/methods , Hippocampus/cytology , Male , Myelin-Associated Glycoprotein/pharmacology , Nerve Regeneration/physiology , Neurons/cytology , Optic Nerve Diseases/drug therapy , Optic Nerve Diseases/pathology , Oxidative Stress/drug effects , Promoter Regions, Genetic/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/metabolism , Small Molecule Libraries
16.
J Immunol ; 182(12): 7776-83, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494301

ABSTRACT

Genes controlling immunopathologic diseases of differing etiopathology may also influence susceptibility to autoimmune disease. B10.D1-H2(q)/SgJ mice with a 2538 G-->A missense mutation in the tyrosine kinase-2 gene (Tyk2) are susceptible to Toxoplasma gondii yet resistant to autoimmune arthritis, unlike the wild-type B10.Q/Ai substrain. To understand whether Tyk2 is also important in a second autoimmune model, experimental allergic encephalomyelitis (EAE) was induced in B10.D1-H2(q)/SgJ (Tyk2(A)) and B10.Q/Ai (Tyk2(G)) mice with the myelin oligodendrocyte glycoprotein peptide 79-96. B10.D1-H2(q)/SgJ mice were resistant to EAE whereas B10.Q/Ai mice were susceptible, and a single copy of the Tyk2(G) allele conferred EAE susceptibility in F(1) hybrids. Furthermore, EAE resistance in B10.D1-H2(q)/SgJ mice was overridden when pertussis toxin (PTX) was used to mimic the effects of environmental factors derived from infectious agents. Numerous cytokines and chemokines were increased when PTX was included in the immunization protocol. However, only RANTES, IL-6, and IFN-gamma increased significantly with both genetic compensation and PTX treatment. These data indicate that Tyk2 is a shared autoimmune disease susceptibility gene whose genetic contribution to disease susceptibility can be modified by environmental factors. Single nucleotide polymorphisms like the one that distinguishes Tyk2 alleles are of considerable significance given the potential role of gene-by-environment interactions in autoimmune disease susceptibility.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/genetics , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide/genetics , TYK2 Kinase/genetics , TYK2 Kinase/metabolism , Alleles , Animals , Cells, Cultured , Cytokines/biosynthesis , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Myelin Proteins , Myelin-Associated Glycoprotein/pharmacology , Myelin-Oligodendrocyte Glycoprotein , Pertussis Toxin/pharmacology
17.
J Neuroimmunol ; 213(1-2): 60-8, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19540601

ABSTRACT

XIncreasing evidence supports a role for CD8+ T cells in multiple sclerosis. In an attempt to isolate the contribution of CD8+ T cells in a murine model of MS, we immunized mice with a dominant CD8 epitope MOG37-46, a truncated version of MOG35-55. The data presented here show mild disease induced with MOG37-46, characterized by lower clinical scores, a decrease in CNS infiltration and a decrease in microglial activation. CD8+ T cells reactive to MOG37-46 are pro-inflammatory and traffic to the CNS; however, the presence of CD4+ T cells elicits more severe disease and sustained inflammation of the CNS.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Lymphocyte Activation/immunology , Myelin-Associated Glycoprotein/immunology , Animals , Cell Line , Cells, Cultured , Central Nervous System/pathology , Central Nervous System/physiopathology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Encephalitis/chemically induced , Encephalitis/immunology , Encephalitis/physiopathology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Gliosis/chemically induced , Gliosis/immunology , Gliosis/physiopathology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Microglia/immunology , Molecular Weight , Myelin Proteins , Myelin-Associated Glycoprotein/pharmacology , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/immunology , Peptide Fragments/pharmacology
18.
J Neurosci Res ; 87(10): 2222-36, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19301432

ABSTRACT

Elucidating the mechanisms that regulate the survival and outgrowth of corticospinal tract (CST) neurons and other CNS tracts will be a key component in developing novel approaches for the treatment of central nervous system (CNS) disorders, including stroke, spinal cord injury (SCI), and motor neuron disease (MND). However, the in vivo complexities of these diseases make a systematic evaluation of potential therapeutics that directly affect corticospinal regeneration or survival very challenging. Here, we use Thy1.2 transgenic mice expressing yellow fluorescent protein (YFP) in postnatal day 8 (P8) corticospinal neurons, as a source of CST neurons that have already established synapses in the spinal cord, to assess factors that influence neurite outgrowth and survival of axotomized CST neurons. After culture, YFP-positive corticospinal neurons represent an enriched neuronal population over other glia and interneurons, survive, and extend processes over time. YFP-positive CST neurons also continue to express the corticospinal markers CTIP2 and Otx1. CST neurons display different degrees of axon extension, dendritic branch length and elaboration, and neurite elongation in response to neurotrophin-3 and ciliary neurotrophic factor, and an inhibitory outgrowth response when cultured on myelin-associated glycoprotein. Some CST neurons are lost with extended culture, which provides a baseline from which we can also assess factors that enhance CST neuron survival. This assay thus allows us to assess independent aspects of CST axonal and dendritic outgrowth kinetics, which allows for the rapid and sensitive investigation of new therapies to address corticospinal neuron outgrowth in the context of CNS injury and neurodegenerative disorders.


Subject(s)
Cell Differentiation/physiology , Myelin-Associated Glycoprotein/pharmacology , Nerve Growth Factors/pharmacology , Neurons/drug effects , Pyramidal Tracts/cytology , Animals , Animals, Newborn , Astrocytes/drug effects , CHO Cells , Cell Death , Cells, Cultured , Cricetinae , Cricetulus , Genotype , In Vitro Techniques , Luminescent Proteins/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurites/drug effects , Neurons/cytology , Thy-1 Antigens/genetics , Time Factors
19.
Mol Brain ; 1: 10, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18922173

ABSTRACT

Several myelin-associated factors that inhibit axon growth of mature neurons, including Nogo66, myelin-associated glycoprotein (MAG) and oligodendrocyte myelin glycoprotein (OMgp), can associate with a common GPI-linked protein Nogo-66 receptor (NgR). Accumulating evidence suggests that myelin inhibitors also signal through unknown NgR-independent mechanisms. Here we show that MAG, a RGD tri-peptide containing protein, forms a complex with ß1-integrin to mediate axonal growth cone turning responses of several neuronal types. Mutations that alter the RGD motif in MAG or inhibition of ß1-integrin function, but not removal of NgRs, abolish these MAG-dependent events. In contrast, OMgp-induced repulsion is not affected by inhibition of b1-integrin function. We further show that MAG stimulates tyrosine phosphorylation of focal adhesion kinase (FAK), which in turn is required for MAG-induced growth cone turning. These studies identify ß1-integrin as a specific mediator for MAG in growth cone turning responses, acting through FAK activation.


Subject(s)
Growth Cones/metabolism , Integrin beta1/metabolism , Myelin-Associated Glycoprotein/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Embryo, Mammalian/metabolism , Enzyme Activation/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , GPI-Linked Proteins/metabolism , Growth Cones/drug effects , Growth Cones/enzymology , Mice , Molecular Sequence Data , Myelin Proteins/metabolism , Myelin-Associated Glycoprotein/chemistry , Myelin-Associated Glycoprotein/pharmacology , Nogo Receptor 1 , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Binding/drug effects , Rats , Receptors, Cell Surface/metabolism , Signal Transduction/drug effects
20.
Am J Pathol ; 172(4): 980-92, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18349128

ABSTRACT

The Pten tumor suppressor gene is critical for normal intrathymic development of T cells; however, its role in mature antigen-activated T cells is less well defined. A genetically crossed mouse line, Pten(fl/fl) GBC, in which Pten gene deletions could be primarily confined to antigen-activated CD8+ T cells, enabled us to evaluate the consequences of Pten loss on the course of experimental autoimmune encephalomyelitis. Compared with Pten(fl/fl) controls, myelin oligodendrocyte glycoprotein (MOG) peptide-immunized Pten(fl/fl) GBC mice developed more severe and protracted disease. This was accompanied by increased spinal cord white matter myelin basic protein depletion and axonal damage, as well as a striking persistence of macrophage and granzyme B-expressing cellular neuroinfiltrates in the chronic phase of the disease. This persistence may be explained by the observation that anti-CD3 activated Pten(fl/fl) GBC T cells were more resistant to proapoptotic stimuli. Consistent with the predicted consequences of Pten loss, purified CD8+ T cells from Pten(fl/fl) GBC mice displayed augmented proliferative responses to anti-T-cell receptor stimulation, and MOG-primed Pten(fl/fl) GBC T cells exhibited a reduced activation threshold to MOG peptide. Pten(fl/fl) GBC mice also developed atypical central nervous system disease, manifested by prominent cervical cord and forebrain involvement. Collectively, our findings indicate that the phosphatidylinositol 3-kinase signaling pathway is an essential regulator of CD8+ T-cell effector function in experimental autoimmune encephalomyelitis.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Deletion , Myelin-Associated Glycoprotein/immunology , PTEN Phosphohydrolase/genetics , Animals , Apoptosis/drug effects , Axons/drug effects , Axons/immunology , CD3 Complex/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Chronic Disease , Genes, Reporter , Granzymes/metabolism , Immunization , Inflammation , Integrases/metabolism , Interleukin-2/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Myelin Proteins , Myelin-Associated Glycoprotein/administration & dosage , Myelin-Associated Glycoprotein/pharmacology , Myelin-Oligodendrocyte Glycoprotein , Peptides/administration & dosage , Peptides/pharmacology , Prosencephalon/drug effects , Prosencephalon/immunology , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...