Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 5294, 2022 09 08.
Article in English | MEDLINE | ID: mdl-36075894

ABSTRACT

Interferon-induced transmembrane protein 3 (IFITM3) is a restriction factor that limits viral pathogenesis and exerts poorly understood immunoregulatory functions. Here, using human and mouse models, we demonstrate that IFITM3 promotes MyD88-dependent, TLR-mediated IL-6 production following exposure to cytomegalovirus (CMV). IFITM3 also restricts IL-6 production in response to influenza and SARS-CoV-2. In dendritic cells, IFITM3 binds to the reticulon 4 isoform Nogo-B and promotes its proteasomal degradation. We reveal that Nogo-B mediates TLR-dependent pro-inflammatory cytokine production and promotes viral pathogenesis in vivo, and in the case of TLR2 responses, this process involves alteration of TLR2 cellular localization. Nogo-B deletion abrogates inflammatory cytokine responses and associated disease in virus-infected IFITM3-deficient mice. Thus, we uncover Nogo-B as a driver of viral pathogenesis and highlight an immunoregulatory pathway in which IFITM3 fine-tunes the responsiveness of myeloid cells to viral stimulation.


Subject(s)
COVID-19 , Interleukin-6 , Nogo Proteins/metabolism , Animals , Cytokines/metabolism , Humans , Interleukin-6/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , SARS-CoV-2 , Toll-Like Receptor 2/metabolism
2.
Mol Psychiatry ; 21(9): 1281-9, 2016 09.
Article in English | MEDLINE | ID: mdl-26619810

ABSTRACT

Critical periods are temporary windows of heightened neural plasticity early in development. For example, fear memories in juvenile rodents are subject to erasure following extinction training, while after closure of this critical period, extinction training only temporarily and weakly suppresses fear memories. Persistence of fear memories is important for survival, but the inability to effectively adapt to the trauma is a characteristic of post-traumatic stress disorder (PTSD). We examined whether Nogo Receptor 1 (NgR1) regulates the plasticity associated with fear extinction. The loss of NgR1 function in adulthood eliminates spontaneous fear recovery and fear renewal, with a restoration of fear reacquisition rate equal to that of naive mice; thus, mimicking the phenotype observed in juvenile rodents. Regional gene disruption demonstrates that NgR1 expression is required in both the basolateral amygdala (BLA) and infralimbic (IL) cortex to prevent fear erasure. NgR1 expression by parvalbumin expressing interneurons is essential for limiting extinction-dependent plasticity. NgR1 gene deletion enhances anatomical changes of inhibitory synapse markers after extinction training. Thus, NgR1 robustly inhibits elimination of fear expression in the adult brain and could serve as a therapeutic target for anxiety disorders, such as PTSD.


Subject(s)
Fear/physiology , Neuronal Plasticity/physiology , Nogo Receptor 1/genetics , Nogo Receptor 1/metabolism , Amygdala/physiology , Animals , Brain/drug effects , Brain/metabolism , Cerebral Cortex/drug effects , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Fear/psychology , Gene Expression , Interneurons , Male , Memory/physiology , Mice , Mice, Inbred C57BL , Neuronal Plasticity/genetics , Parvalbumins , Prefrontal Cortex/drug effects
3.
Neuroimage ; 84: 1070-81, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23685159

ABSTRACT

A first-ever spinal cord imaging meeting was sponsored by the International Spinal Research Trust and the Wings for Life Foundation with the aim of identifying the current state-of-the-art of spinal cord imaging, the current greatest challenges, and greatest needs for future development. This meeting was attended by a small group of invited experts spanning all aspects of spinal cord imaging from basic research to clinical practice. The greatest current challenges for spinal cord imaging were identified as arising from the imaging environment itself; difficult imaging environment created by the bone surrounding the spinal canal, physiological motion of the cord and adjacent tissues, and small cross-sectional dimensions of the spinal cord, exacerbated by metallic implants often present in injured patients. Challenges were also identified as a result of a lack of "critical mass" of researchers taking on the development of spinal cord imaging, affecting both the rate of progress in the field, and the demand for equipment and software to manufacturers to produce the necessary tools. Here we define the current state-of-the-art of spinal cord imaging, discuss the underlying theory and challenges, and present the evidence for the current and potential power of these methods. In two review papers (part I and part II), we propose that the challenges can be overcome with advances in methods, improving availability and effectiveness of methods, and linking existing researchers to create the necessary scientific and clinical network to advance the rate of progress and impact of the research.


Subject(s)
Neuroimaging/methods , Spinal Cord Injuries/diagnosis , Spinal Cord , Humans , Spinal Cord/pathology
4.
Neuroimage ; 84: 1082-93, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23859923

ABSTRACT

A first-ever spinal cord imaging meeting was sponsored by the International Spinal Research Trust and the Wings for Life Foundation with the aim of identifying the current state-of-the-art of spinal cord imaging, the current greatest challenges, and greatest needs for future development. This meeting was attended by a small group of invited experts spanning all aspects of spinal cord imaging from basic research to clinical practice. The greatest current challenges for spinal cord imaging were identified as arising from the imaging environment itself; difficult imaging environment created by the bone surrounding the spinal canal, physiological motion of the cord and adjacent tissues, and small crosssectional dimensions of the spinal cord, exacerbated by metallic implants often present in injured patients. Challenges were also identified as a result of a lack of "critical mass" of researchers taking on the development of spinal cord imaging, affecting both the rate of progress in the field, and the demand for equipment and software to manufacturers to produce the necessary tools. Here we define the current state-of-the-art of spinal cord imaging, discuss the underlying theory and challenges, and present the evidence for the current and potential power of these methods. In two review papers (part I and part II), we propose that the challenges can be overcome with advances in methods, improving availability and effectiveness of methods, and linking existing researchers to create the necessary scientific and clinical network to advance the rate of progress and impact of the research.


Subject(s)
Magnetic Resonance Imaging/methods , Magnetic Resonance Imaging/trends , Spinal Cord Diseases/diagnosis , Spinal Cord Injuries/diagnosis , Animals , Humans , Spinal Cord/pathology
5.
Behav Brain Res ; 238: 69-78, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23089646

ABSTRACT

The LRRTM family proteins have been shown to act as synaptogenic cell adhesion molecules via interaction with presynaptic neurexins and are associated with neuropsychiatric disorders. LRRTM1-knockout mice have subtle morphological deficits in excitatory hippocampal synapses and were suggested to have impaired cognitive function. Here we report that LRRTM1-knockout mice exhibit an extraordinary phenotype of avoiding small enclosures. In the light-dark box, the knockout mice escape to dark through a standard opening as quickly as wild-type littermates but avoid escaping through a small doorway. While all wild-type mice spontaneously enter a small tube, most knockout mice do not. This apparent aversion to enter narrow space may explain other abnormalities such as increased time in open arms in the elevated plus maze and less visits through a tunnel in the IntelliCage. Moreover, LRRTM1-knockout mice show increased social interaction, reduced nest building and MK801-induced locomotion, and slower swim speed but normal water maze learning. Since LRRTM1 is predominantly expressed in thalamus, hippocampus and limbic cortex, specific synaptic defects in those areas presumably cause these behavioural abnormalities.


Subject(s)
Behavior, Animal/physiology , Motor Activity/physiology , Neural Cell Adhesion Molecules/genetics , Phobic Disorders/genetics , Animals , Disease Models, Animal , Maze Learning/physiology , Membrane Proteins , Mice , Mice, Knockout , Nerve Tissue Proteins , Neural Cell Adhesion Molecules/metabolism , Phenotype , Phobic Disorders/metabolism , Social Behavior , Swimming/physiology
6.
Cell Tissue Res ; 349(1): 119-32, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22350947

ABSTRACT

Limited axonal plasticity within the central nervous system (CNS) is a major restriction for functional recovery after CNS injury. The small GTPase RhoA is a key molecule of the converging downstream cascade that leads to the inhibition of axonal re-growth. The Rho-pathway integrates growth inhibitory signals derived from extracellular cues, such as chondroitin sulfate proteoglycans, Nogo-A, myelin-associated glycoprotein, oligodendrocyte-myelin glycoprotein, Ephrins and repulsive guidance molecule-A, into the damaged axon. Consequently, the activation of RhoA results in growth cone collapse and finally outgrowth failure. In turn, the inhibition of RhoA-activation blinds the injured axon to its growth inhibitory environment resulting in enhanced axonal sprouting and plasticity. This has been demonstrated in various CNS-injury models for direct RhoA-inhibition and for downstream/upstream blockade of the RhoA-associated pathway. In addition, RhoA-inhibition reduces apoptotic cell death and secondary damage and improves locomotor recovery in clinically relevant models after experimental spinal cord injury (SCI). Unexpectedly, a subset of "small molecules" from the group of non-steroid anti-inflammatory drugs, particularly the FDA-approved ibuprofen, has recently been identified as (1) inhibiting RhoA-activation, (2) enhancing axonal sprouting/regeneration, (3) protecting "tissue at risk" (neuroprotection) and (4) improving motor recovery confined to realistic therapeutical time-frames in clinically relevant SCI models. Here, we survey the effect of small-molecule-induced RhoA-inhibition on axonal plasticity and neurofunctional outcome in CNS injury paradigms. Furthermore, we discuss the body of preclinical evidence for a possible clinical translation with a focus on ibuprofen and illustrate putative risks and benefits for the treatment of acute SCI.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Small Molecule Libraries/pharmacology , Spinal Cord Injuries/drug therapy , rho GTP-Binding Proteins/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Humans , Neuronal Plasticity/drug effects , Signal Transduction/drug effects , rho GTP-Binding Proteins/metabolism
7.
Neuroscience ; 163(2): 540-51, 2009 Oct 06.
Article in English | MEDLINE | ID: mdl-19555742

ABSTRACT

Increasing age is associated with a poor prognosis following traumatic brain injury (TBI). CNS axons may recover poorly following TBI due to expression of myelin-derived inhibitors to axonal outgrowth such as Nogo-A. To study the role of Nogo-A/B in the pathophysiological response of the elderly to TBI, 1-year-old mice deficient in Nogo-A/B (Nogo-A/B homozygous(-/-) mice), Nogo-A/B heterozygous(-/+) mice, and age-matched wild-type (WT) littermate controls were subjected to a controlled cortical impact (CCI) TBI. Sham-injured WT mice (7 months old) and 12 month old naïve Nogo-A/B(-/-) and Nogo-A/B(-/+) served as controls. Neurological motor function was evaluated up to 3 weeks, and cognitive function, hemispheric tissue loss, myelin staining and hippocampal beta-amyloid (A beta) immunohistochemistry were evaluated at 4 weeks post-injury. In WT littermates, TBI significantly impaired learning ability at 4 weeks and neurological motor function up to 2 weeks post-injury and caused a significant loss of hemispheric tissue. Following TBI, Nogo-A/B(-/-) mice showed significantly less recovery from neurological motor and cognitive deficits compared to brain-injured WT mice. Naïve Nogo-A/B(-/-) and Nogo-A/B(-/+) mice quickly learned the MWM task in contrast to brain-injured Nogo-A/B(-/-) mice who failed to learn the MWM task at 4 weeks post-injury. Hemispheric tissue loss and cortical lesion volume were similar among the brain-injured genotypes. Neither TBI nor the absence of NogoA/B caused an increased A beta expression. Myelin staining showed a reduced area and density in the corpus callosum in brain-injured Nogo-A/B(-/-) animals compared to their littermate controls. These novel and unexpected behavioral results demonstrate that the absence of Nogo-A/B may negatively influence outcome, possibly related to hypomyelination, following TBI in mice and suggest a complex role for this myelin-associated axonal growth inhibitor following TBI.


Subject(s)
Aging , Brain Injuries/physiopathology , Myelin Proteins/deficiency , Recovery of Function/physiology , Amyloid beta-Peptides/metabolism , Animals , Brain Injuries/pathology , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Proteins/genetics , Myelin Proteins/metabolism , Myelin Sheath/metabolism , Myelin Sheath/pathology , Neuropsychological Tests , Nogo Proteins , Organ Size , Random Allocation , Time Factors , Treatment Outcome
8.
Br J Ophthalmol ; 93(9): 1134-40, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19286686

ABSTRACT

Maintenance of ocular viability is one of the major impediments to successful whole-eye transplantation. This review provides a comprehensive understanding of the current literature to help guide future studies in order to overcome this hurdle. A systematic multistage review of published literature was performed. Three specific questions were addressed: (1) Is recovery of visual function following eye transplantation greater in cold-blooded vertebrates when compared with mammals? (2) Is outer retina function following enucleation and reperfusion improved compared with enucleation alone? (3) Following optic-nerve transection, is there a correlation between retinal ganglion cell (RGC) survival and either time after transection or proximity of the transection to the globe? In a majority of the studies performed in the literature, recovery of visual function can occur after whole-eye transplantation in cold-blooded vertebrates. Following enucleation (and reperfusion), outer retinal function is maintained from 4 to 9 h. RGC survival following optic-nerve transection is inversely related to both the time since transection and the proximity of transection to the globe. Lastly, neurotrophins can increase RGC survival following optic-nerve transection. This review of the literature suggests that the use of a donor eye is feasible for whole-eye transplantation.


Subject(s)
Eye Enucleation/adverse effects , Eye/transplantation , Optic Nerve Injuries/complications , Retinal Ganglion Cells/physiology , Animals , Eye/physiopathology , Optic Nerve Injuries/physiopathology , Retina/physiology , Tissue Donors , Tissue Survival/physiology , Visual Acuity
9.
Mol Psychiatry ; 12(12): 1129-39, 1057, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17667961

ABSTRACT

Left-right asymmetrical brain function underlies much of human cognition, behavior and emotion. Abnormalities of cerebral asymmetry are associated with schizophrenia and other neuropsychiatric disorders. The molecular, developmental and evolutionary origins of human brain asymmetry are unknown. We found significant association of a haplotype upstream of the gene LRRTM1 (Leucine-rich repeat transmembrane neuronal 1) with a quantitative measure of human handedness in a set of dyslexic siblings, when the haplotype was inherited paternally (P=0.00002). While we were unable to find this effect in an epidemiological set of twin-based sibships, we did find that the same haplotype is overtransmitted paternally to individuals with schizophrenia/schizoaffective disorder in a study of 1002 affected families (P=0.0014). We then found direct confirmatory evidence that LRRTM1 is an imprinted gene in humans that shows a variable pattern of maternal downregulation. We also showed that LRRTM1 is expressed during the development of specific forebrain structures, and thus could influence neuronal differentiation and connectivity. This is the first potential genetic influence on human handedness to be identified, and the first putative genetic effect on variability in human brain asymmetry. LRRTM1 is a candidate gene for involvement in several common neurodevelopmental disorders, and may have played a role in human cognitive and behavioral evolution.


Subject(s)
Chromosomes, Human, Pair 2 , Functional Laterality/genetics , Genetic Predisposition to Disease , Membrane Proteins/genetics , Schizophrenia/genetics , Animals , Brain/metabolism , Brain/pathology , Cell Line, Transformed , Family Health , Female , Gene Expression Regulation, Developmental/physiology , Genotype , Humans , In Situ Hybridization/methods , Karyotyping , Male , Membrane Proteins/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Schizophrenia/pathology , Subcellular Fractions/metabolism , Subcellular Fractions/pathology , Subcellular Fractions/ultrastructure
10.
Neuroscientist ; 7(5): 377-86, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11597097

ABSTRACT

Following injury, axons of the adult mammalian central nervous system (CNS) fail to regenerate. As a result, CNS trauma generally results in severe and persistent functional deficits. The inability of CNS axons to regenerate is largely associated with nonneuronal aspects of the CNS environment that are inhibitory to axonal elongation. This inhibition is mediated by the glial scar, including reactive astrocytes, and by the myelin-associated neurite outgrowth inhibitors chondroitin sulfate proteoglycans, myelin-associated glycoprotein, and Nogo. Nogo is an integral membrane protein that localizes to CNS, but not peripheral nervous system, myelin. In vitro characterization of Nogo has demonstrated its function as a potent inhibitor of axon elongation. In vivo neutralization of Nogo activity results in enhanced axonal regeneration and functional recovery following CNS injury as well as increased plasticity in uninjured CNS fibers. These findings suggest that Nogo may be a major contributor to the nonpermissive nature of the CNS environment.


Subject(s)
Axons/physiology , Growth Inhibitors/physiology , Myelin Proteins/physiology , Nerve Regeneration/physiology , Animals , Brain/physiology , Brain/physiopathology , Brain Injuries/physiopathology , Humans , Mammals , Models, Neurological , Neuronal Plasticity , Nogo Proteins
11.
Curr Opin Cell Biol ; 13(5): 619-26, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11544032

ABSTRACT

For many growing axons, interaction with an extracelluar Semaphorin signal leads to growth cone collapse and axon repulsion. Semaphorin receptors composed of Neuropilins and Plexins transduce extracellular cues into changes in the growth cone actin cytoskeleton. The data implicating Rho family G proteins in Semaphorin signaling and in other axon guidance events are considered here. Recent work makes it clear that Rac1 is required for this process. In particular, there is intriguing new evidence that the Plexin receptors communicate directly with members of the Rho family GTPases, although uncertainties remain concerning how Plexins alter Rac1 function. The CRMP (collapsin response mediator protein) family is also required for Plexin-based Semaphorin signaling, and new data demonstrate direct links to Rho and Rac1-based signaling.


Subject(s)
Axons/physiology , Carrier Proteins/physiology , Nerve Tissue Proteins/physiology , rho GTP-Binding Proteins/physiology , Animals , GTPase-Activating Proteins/physiology , Growth Cones/physiology , Guanine Nucleotide Exchange Factors/physiology , Intercellular Signaling Peptides and Proteins , Models, Neurological , Nerve Tissue Proteins/metabolism , Neurons/physiology , Neuropilin-1 , Receptors, Cell Surface/metabolism
12.
Neuron ; 29(2): 429-39, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11239433

ABSTRACT

Semaphorin 3A (Sema3A) binds to neuropilin-1 (NP1) and activates the transmembrane Plexin to transduce a repulsive axon guidance signal. Here, we show that Sema3 signals are transduced equally effectively by PlexinA1 or PlexinA2, but not by PlexinA3. Deletion analysis of the PlexinA1 ectodomain demonstrates that the sema domain prevents PlexinA1 activation in the basal state. Sema-deleted PlexinA1 is constitutively active, producing cell contraction, growth cone collapse, and inhibition of neurite outgrowth. The sema domain of PlexinA1 physically associates with the remainder of the PlexinA1 ectodomain and can reverse constitutive activation. Both the sema portion and the remainder of the ectodomain of PlexinA1 associate with NP1 in a Sema3A-independent fashion. Plexin A1 is autoinhibited by its sema domain, and Sema3A/NP1 releases this inhibition.


Subject(s)
Carrier Proteins/metabolism , Growth Cones/metabolism , Mutation/genetics , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , Animals , COS Cells , Chick Embryo , Ganglia, Spinal/metabolism , Nerve Tissue Proteins/genetics , Neural Inhibition/physiology , Neuropilin-1 , Receptors, Cell Surface/genetics , Signal Transduction/physiology
13.
Curr Opin Neurobiol ; 11(1): 89-94, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11179877

ABSTRACT

During the past year, a major advance in the study of axon regeneration was the molecular cloning of Nogo. The expression of Nogo protein by CNS myelin may be a major factor in the failure of CNS axon regeneration. The effect of disrupting Nogo-dependent axon inhibition can now be studied conclusively. In related work, immunization with a Nogo-containing CNS myelin preparation was shown to promote regeneration and dramatic functional recovery after spinal cord trauma.


Subject(s)
Axons/physiology , Central Nervous System/physiology , Nerve Growth Factors/physiology , Nerve Regeneration/physiology , Central Nervous System/cytology , Humans , Myelin Proteins/physiology , Myelin Sheath/physiology , Nogo Proteins
14.
Nature ; 409(6818): 341-6, 2001 Jan 18.
Article in English | MEDLINE | ID: mdl-11201742

ABSTRACT

Nogo has been identified as a component of the central nervous system (CNS) myelin that prevents axonal regeneration in the adult vertebrate CNS. Analysis of Nogo-A has shown that an axon-inhibiting domain of 66 amino acids is expressed at the extracellular surface and at the endoplasmic reticulum lumen of transfected cells and oligodendrocytes. The acidic amino terminus of Nogo-A is detected at the cytosolic face of cellular membranes and may contribute to inhibition of axon regeneration at sites of oligodendrocyte injury. Here we show that the extracellular domain of Nogo (Nogo-66) inhibits axonal extension, but does not alter non-neuronal cell morphology. In contrast, a multivalent form of the N terminus of Nogo-A affects the morphology of both neurons and other cell types. Here we identify a brain-specific, leucine-rich-repeat protein with high affinity for soluble Nogo-66. Cleavage of the Nogo-66 receptor and other glycophosphatidylinositol-linked proteins from axonal surfaces renders neurons insensitive to Nogo-66. Nogo-66 receptor expression is sufficient to impart Nogo-66 axonal inhibition to unresponsive neurons. Disruption of the interaction between Nogo-66 and its receptor provides the potential for enhanced recovery after human CNS injury.


Subject(s)
Axons/physiology , Myelin Proteins/physiology , Nerve Regeneration , Receptors, Cell Surface/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Cell Division , Cell Line , Chickens , Cloning, Molecular , DNA, Complementary , GPI-Linked Proteins , Gene Expression , Growth Cones , Humans , Mice , Molecular Sequence Data , Myelin Proteins/chemistry , Nogo Proteins , Nogo Receptor 1 , Protein Structure, Tertiary , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
16.
J Neurobiol ; 44(2): 219-29, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10934324

ABSTRACT

The semaphorin family of proteins constitute one of the major cues for axonal guidance. The prototypic member of this family is Sema3A, previously designated semD/III or collapsin-1. Sema3A acts as a diffusible, repulsive guidance cue in vivo for the peripheral projections of embryonic dorsal root ganglion neurons. Sema3A binds with high affinity to neuropilin-1 on growth cone filopodial tips. Although neuropilin-1 is required for Sema3A action, it is incapable of transmitting a Sema3A signal to the growth cone interior. Instead, the Sema3A/neuropilin-1 complex interacts with another transmembrane protein, plexin, on the surface of growth cones. Certain semaphorins, other than Sema3A, can bind directly to plexins. The intracellular domain of plexin is responsible for initiating the signal transduction cascade leading to growth cone collapse, axon repulsion, or growth cone turning. This intracellular cascade involves the monomeric G-protein, Rac1, and a family of neuronal proteins, the CRMPs. Rac1 is likely to be involved in semaphorin-induced rearrangements of the actin cytoskeleton, but how plexin controls Rac1 activity is not known. Vertebrate CRMPs are homologous to the Caenorhabditis elegans unc-33 protein, which is required for proper axon morphology in worms. CRMPs are essential for Sema3A-induced, neuropilin-plexin-mediated growth cone collapse, but the molecular interactions of growth cone CRMPs are not well defined. Mechanistic aspects of plexin-based signaling for semaphorin guidance cues may have implications for other axon guidance events and for the basis of growth cone motility.


Subject(s)
Axons/physiology , Growth Cones/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Gene Expression/physiology , Intercellular Signaling Peptides and Proteins , Neuropilin-1 , Semaphorin-3A
17.
J Cell Biol ; 149(2): 411-22, 2000 Apr 17.
Article in English | MEDLINE | ID: mdl-10769032

ABSTRACT

Axonal growth cone collapse is accompanied by a reduction in filopodial F-actin. We demonstrate here that semaphorin 3A (Sema3A) induces a coordinated rearrangement of Sema3A receptors and F-actin during growth cone collapse. Differential interference contrast microscopy reveals that some sites of Sema3A-induced F-actin reorganization correlate with discrete vacuoles, structures involved in endocytosis. Endocytosis of FITC-dextran by the growth cone is enhanced during Sema3A treatment, and sites of dextran accumulation colocalize with actin-rich vacuoles and ridges of membrane. Furthermore, the Sema3A receptor proteins, neuropilin-1 and plexin, and the Sema3A signaling molecule, rac1, also reorganize to vacuoles and membrane ridges after Sema3A treatment. These data support a model whereby Sema3A stimulates endocytosis by focal and coordinated rearrangement of receptor and cytoskeletal elements. Dextran accumulation is also increased in retinal ganglion cell (RGC) growth cones, in response to ephrin A5, and in RGC and DRG growth cones, in response to myelin and phorbol-ester. Therefore, enhanced endocytosis may be a general principle of physiologic growth cone collapse. We suggest that growth cone collapse is mediated by both actin filament rearrangements and alterations in membrane dynamics.


Subject(s)
Actins/physiology , Glycoproteins/pharmacology , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/physiology , Neurons/cytology , Neurons/physiology , Actins/drug effects , Animals , Cell Adhesion Molecules/physiology , Cell Membrane/drug effects , Cell Membrane/physiology , Cell Membrane/ultrastructure , Chick Embryo , Endocytosis/drug effects , Endocytosis/physiology , Ganglia, Spinal/cytology , Microscopy, Interference/methods , Nerve Tissue Proteins/drug effects , Neurons/ultrastructure , Neuropilin-1 , Receptors, Cell Surface/physiology , Retina/embryology , Semaphorin-3A , Signal Transduction , rac1 GTP-Binding Protein/physiology
19.
Nature ; 403(6768): 439-44, 2000 Jan 27.
Article in English | MEDLINE | ID: mdl-10667797

ABSTRACT

Adult mammalian axon regeneration is generally successful in the peripheral nervous system (PNS) but is dismally poor in the central nervous system (CNS). However, many classes of CNS axons can extend for long distances in peripheral nerve grafts. A comparison of myelin from the CNS and the PNS has revealed that CNS white matter is selectively inhibitory for axonal outgrowth. Several components of CNS white matter, NI35, NI250(Nogo) and MAG, that have inhibitory activity for axon extension have been described. The IN-1 antibody, which recognizes NI35 and NI250(Nogo), allows moderate degrees of axonal regeneration and functional recovery after spinal cord injury. Here we identify Nogo as a member of the Reticulon family, Reticulon 4-A. Nogo is expressed by oligodendrocytes but not by Schwann cells, and associates primarily with the endoplasmic reticulum. A 66-residue lumenal/extracellular domain inhibits axonal extension and collapses dorsal root ganglion growth cones. In contrast to Nogo, Reticulon 1 and 3 are not expressed by oligodendrocytes, and the 66-residue lumenal/extracellular domains from Reticulon 1, 2 and 3 do not inhibit axonal regeneration. These data provide a molecular basis to assess the contribution of Nogo to the failure of axonal regeneration in the adult CNS.


Subject(s)
Axons/physiology , Central Nervous System/physiology , Growth Inhibitors/physiology , Membrane Proteins/physiology , Myelin Proteins , Nerve Regeneration , Amino Acid Sequence , Animals , Cattle , Cell Division , Cell Line , Chick Embryo , Cloning, Molecular , Consensus Sequence , Escherichia coli , Growth Inhibitors/chemistry , Growth Inhibitors/genetics , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Nogo Proteins , Oligodendroglia/metabolism , PC12 Cells , Rats , Recombinant Proteins , Sequence Homology, Amino Acid
20.
J Neurochem ; 74(2): 582-95, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10646509

ABSTRACT

Neurotrophic factors (NTFs) can protect against or sensitize neurons to excitotoxicity. We studied the role played by various NTFs in the excitotoxic death of purified embryonic rat motor neurons. Motor neurons cultured in brain-derived neurotrophic factor, but not neurotrophin 3, glial-derived neurotrophic factor, or cardiotrophin 1, were sensitive to excitotoxic insult. BDNF also induces excitotoxic sensitivity (ES) in motor neurons when BDNF is combined with these other NTFs. The effect of BDNF depends on de novo protein and mRNA synthesis. Reagents that either activate or inhibit the 75-kDa NTF receptor p75NTR do not affect BDNF-induced ES. The low EC50 for BDNF-induced survival and ES suggests that TrkB mediates both of these biological activities. BDNF does not alter glutamate-evoked rises of intracellular Ca2+, suggesting BDNF acts downstream. Both wortmannin and LY294002, which specifically block the phosphatidylinositol 3-kinase (PI3K) intracellular signaling pathway in motor neurons, inhibit BDNF-induced ES. We confirm this finding using a herpes simplex virus (HSV) that expresses the dominant negative p85 subunit of PI3K. Infecting motor neurons with this HSV, but not a control HSV, blocks activation of the PI3K pathway and BDNF-induced ES. Through the activation of TrkB and the PI3K signaling pathway, BDNF renders developing motor neurons susceptible to glutamate receptor-mediated cell death.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Motor Neurons/drug effects , Neurotoxins/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Spinal Cord/drug effects , Spinal Cord/enzymology , Animals , Calcium/metabolism , Cells, Cultured , Drug Synergism , Enzyme Activation/physiology , Glutamic Acid/pharmacology , Motor Neurons/enzymology , Motor Neurons/metabolism , Protein Biosynthesis , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor/physiology , Receptor, trkB/physiology , Spinal Cord/cytology , Spinal Cord/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...