Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Eur J Neurol ; 28(7): 2280-2293, 2021 07.
Article in English | MEDLINE | ID: mdl-33547839

ABSTRACT

BACKGROUND AND PURPOSE: Foveal changes were reported in aquaporin-4 antibody (AQP4-Ab) seropositive neuromyelitis optica spectrum disorder (NMOSD) patients; however, it is unclear whether they are independent of optic neuritis (ON), stem from subclinical ON or crossover from ON in fellow eyes. Fovea morphometry and a statistical classification approach were used to investigate if foveal changes in NMOSD are independent of ON and progressive. METHODS: This was a retrospective longitudinal study of 27 AQP4-IgG + NMOSD patients (49 eyes; 15 ON eyes and 34 eyes without a history of ON [NON eyes]), follow-up median (first and third quartile) 2.32 (1.33-3.28), and 38 healthy controls (HCs) (76 eyes), follow-up median (first and third quartile) 1.95 (1.83-2.54). The peripapillary retinal nerve fibre layer thickness and the volume of combined ganglion cell and inner plexiform layer as measures of neuroaxonal damage from ON were determined by optical coherence tomography. Nineteen foveal morphometry parameters were extracted from macular optical coherence tomography volume scans. Data were analysed using orthogonal partial least squares discriminant analysis and linear mixed effects models. RESULTS: At baseline, foveal shape was significantly altered in ON eyes and NON eyes compared to HCs. Discriminatory analysis showed 81% accuracy distinguishing ON vs. HCs and 68% accuracy in NON vs. HCs. NON eyes were distinguished from HCs by foveal shape parameters indicating widening. Orthogonal partial least squares discriminant analysis discriminated ON vs. NON with 76% accuracy. In a follow-up of 2.4 (20.85) years, no significant time-dependent foveal changes were found. CONCLUSION: The parafoveal area is altered in AQP4-Ab seropositive NMOSD patients suggesting independent neuroaxonal damage from subclinical ON. Longer follow-ups are needed to confirm the stability of the parafoveal structure over time.


Subject(s)
Neuromyelitis Optica , Optic Neuritis , Aquaporin 4 , Humans , Longitudinal Studies , Neuromyelitis Optica/complications , Neuromyelitis Optica/diagnostic imaging , Retrospective Studies , Tomography, Optical Coherence
2.
J Neurotrauma ; 35(19): 2259-2271, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29609523

ABSTRACT

Traumatic brain injury (TBI) is a leading cause of death and disability, particularly among the young. Despite this, no disease-specific treatments exist. Recently, blood-brain barrier disruption and parenchymal fibrinogen deposition have been reported in acute traumatic brain injury and in long-term survival; however, their contribution to the neuropathology of TBI remains unknown. The presence of fibrinogen-a well-documented activator of microglia/macrophages-may be associated with neuroinflammation, and neuronal/axonal injury. To test this hypothesis, cases of human TBI with survival times ranging from 12 h to 13 years (survival <2 months, n = 15; survival >1 year, n = 6) were compared with uninjured controls (n = 15). Tissue was selected from the frontal lobe, temporal lobe, corpus callosum, cingulate gyrus, and brainstem, and the extent of plasma protein (fibrinogen and immunoglobulin G [IgG]) deposition, microglial/macrophage activation (CD68 and ionized calcium-binding adapter molecule 1 [Iba-1] immunoreactivity), neuronal density, and axonal transport impairment (ß-amyloid precursor protein [ßAPP] immunoreactivity) were assessed. Quantitative analysis revealed a significant increase in parenchymal fibrinogen and IgG deposition following acute TBI compared with long-term survival and control. Fibrinogen, but not IgG, was associated with microglial/macrophage activation and a significant reduction in neuronal density. Perivascular fibrinogen deposition also was associated with microglial/macrophage clustering and accrual of ßAPP in axonal spheroids, albeit rarely. These findings mandate the future exploration of causal relationships between fibrinogen deposition, microglia/macrophage activation, and potential neuronal loss in acute TBI.


Subject(s)
Brain Injuries, Traumatic/pathology , Inflammation/pathology , Neurons/pathology , Adult , Aged , Brain Injuries, Traumatic/metabolism , Female , Fibrinogen/metabolism , Humans , Macrophage Activation/physiology , Male , Middle Aged
3.
Am J Pathol ; 178(2): 794-802, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21281812

ABSTRACT

IL-17-producing CD4(+) T cells (Th-17) contribute to the pathogenesis of experimental autoimmune encephalomyelitis and are associated with active disease in multiple sclerosis (MS). In addition to IL-17, Th-17 cells can also express IL-21, IL-22, and IL-6 under Th-17-polarizing conditions (IL-6 and transforming growth factor-ß). In this study we investigated IL-21 and IL-21 receptor (IL-21R) expression in MS lesions by in situ hybridization and immunohistochemistry. We detected strongly IL-21(+) infiltrating cells predominantly in acute but also in chronic active white matter MS lesions in which IL-21 expression was restricted to CD4(+) cells. In contrast, IL-21R was much more broadly distributed on CD4(+), CD19(+), and CD8(+) lymphocytes but not major histocompatibility complex class-II(+) macrophages/microglia. Interestingly, in cortical areas we detected both IL-21 and IL-21R expression by neurons. These findings suggest role(s) for IL-21 in both the acute and chronic stages of MS via direct effects on T and B lymphocytes and, demonstrated for the first time, also on neurons.


Subject(s)
Brain/pathology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Interleukin-21 Receptor alpha Subunit/metabolism , Interleukins/metabolism , Multiple Sclerosis/metabolism , Neurons/metabolism , Acute Disease , Antigens, CD19/metabolism , B-Lymphocytes/metabolism , Brain/metabolism , Chronic Disease , Humans , Interleukin-21 Receptor alpha Subunit/genetics , Interleukins/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Neurons/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation/genetics
4.
Brain ; 134(Pt 2): 571-84, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21233144

ABSTRACT

Although there is growing evidence for a role of excess intracellular cations, particularly calcium ions, in neuronal and glial cell injury in multiple sclerosis, as well as in non-inflammatory neurological conditions, the molecular mechanisms involved are not fully determined. We previously showed that the acid-sensing ion channel 1 which, when activated under the acidotic tissue conditions found in inflammatory lesions opens to allow influx of sodium and calcium ions, contributes to axonal injury in experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis. However, the extent and cellular distribution of acid-sensing ion channel 1 expression in neurons and glia in inflammatory lesions is unknown and, crucially, acid-sensing ion channel 1 expression has not been determined in multiple sclerosis lesions. Here we studied acute and chronic experimental autoimmune encephalomyelitis and multiple sclerosis spinal cord and optic nerve tissues to describe in detail the distribution of acid-sensing ion channel 1 and its relationship with neuronal and glial damage. We also tested the effects of amiloride treatment on tissue damage in the mouse models. We found that acid-sensing ion channel 1 was upregulated in axons and oligodendrocytes within lesions from mice with acute experimental autoimmune encephalomyelitis and from patients with active multiple sclerosis. The expression of acid-sensing ion channel 1 was associated with axonal damage as indicated by co-localization with the axonal injury marker beta amyloid precursor protein. Moreover, blocking acid-sensing ion channel 1 with amiloride protected both myelin and neurons from damage in the acute model, and when given either at disease onset or, more clinically relevant, at first relapse, ameliorated disability in mice with chronic-relapsing experimental autoimmune encephalomyelitis. Together these findings suggest that blockade of acid-sensing ion channel 1 has the potential to provide both neuro- and myelo-protective benefits in multiple sclerosis.


Subject(s)
Axons/metabolism , Demyelinating Diseases/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Nerve Tissue Proteins/biosynthesis , Sodium Channels/biosynthesis , Acid Sensing Ion Channels , Aged , Amiloride/pharmacology , Amiloride/therapeutic use , Animals , Axons/drug effects , Axons/pathology , Cell Culture Techniques , Demyelinating Diseases/drug therapy , Demyelinating Diseases/prevention & control , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Mice , Mice, Inbred C57BL , Middle Aged , Multiple Sclerosis/pathology , Nerve Tissue Proteins/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oligodendroglia/metabolism , Optic Nerve/drug effects , Optic Nerve/metabolism , Optic Nerve/pathology , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , Up-Regulation
5.
Nat Med ; 14(11): 1227-35, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18953350

ABSTRACT

The major known genetic risk factors in multiple sclerosis reside in the major histocompatibility complex (MHC) region. Although there is strong evidence implicating MHC class II alleles and CD4(+) T cells in multiple sclerosis pathogenesis, possible contributions from MHC class I genes and CD8(+) T cells are controversial. We have generated humanized mice expressing the multiple sclerosis-associated MHC class I alleles HLA-A(*)0301 (encoding human leukocyte antigen-A3 (HLA-A3)) and HLA-A(*)0201 (encoding HLA-A2) and a myelin-specific autoreactive T cell receptor (TCR) derived from a CD8(+) T cell clone from an individual with multiple sclerosis to study mechanisms of disease susceptibility. We demonstrate roles for HLA-A3-restricted CD8(+) T cells in induction of multiple sclerosis-like disease and for CD4(+) T cells in its progression, and we also define a possible mechanism for HLA-A(*)0201-mediated protection. To our knowledge, these data provide the first direct evidence incriminating MHC class I genes and CD8(+) T cells in the pathogenesis of human multiple sclerosis and reveal a network of MHC interactions that shape the risk of multiple sclerosis.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/immunology , Multiple Sclerosis/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Central Nervous System/immunology , Gene Expression Regulation , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Mice , Mice, Transgenic , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Thymus Gland/immunology
6.
Am J Pathol ; 172(1): 146-55, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18156204

ABSTRACT

Recent findings in the animal model for multiple sclerosis (MS), experimental autoimmune encephalomyelitis, implicate a novel CD4+ T-cell subset (TH17), characterized by the secretion of interleukin-17 (IL-17), in disease pathogenesis. To elucidate its role in MS, brain tissues from patients with MS were compared to controls. We detected expression of IL-17 mRNA (by in situ hybridization) and protein (by immunohistochemistry) in perivascular lymphocytes as well as in astrocytes and oligodendrocytes located in the active areas of MS lesions. Further, we found a significant increase in the number of IL-17+ T cells in active rather than inactive areas of MS lesions. Specifically, double immunofluorescence showed that IL-17 immunoreactivity was detected in 79% of T cells in acute lesions, 73% in active areas of chronic active lesions, but in only 17% of those in inactive lesions and 7% in lymph node control tissue. CD8+, as well as CD4+, T cells were equally immunostained for IL-17 in MS tissues. Interestingly, and in contrast to lymph node T cells, no perivascular T cells showed FoxP3 expression, a marker of regulatory T cells, at any stage of MS lesions. These observations suggest an enrichment of both IL-17+CD4+ and CD8+ T cells in active MS lesions as well as an important role for IL-17 in MS pathogenesis, with some remarkable differences from the experimental autoimmune encephalomyelitis model.


Subject(s)
Central Nervous System/metabolism , Interleukin-17/metabolism , Multiple Sclerosis/immunology , Neuroglia/immunology , T-Lymphocytes/immunology , Adult , Aged , Brain/pathology , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Case-Control Studies , Cell Differentiation , Humans , Lymph Nodes/pathology , Middle Aged , Multiple Sclerosis/metabolism , Neuroglia/metabolism , T-Lymphocytes/metabolism
7.
Nat Med ; 13(12): 1483-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17994101

ABSTRACT

Multiple sclerosis is a neuroinflammatory disease associated with axonal degeneration. The neuronally expressed, proton-gated acid-sensing ion channel-1 (ASIC1) is permeable to Na+ and Ca2+, and excessive accumulation of these ions is associated with axonal degeneration. We tested the hypothesis that ASIC1 contributes to axonal degeneration in inflammatory lesions of the central nervous system (CNS). After induction of experimental autoimmune encephalomyelitis (EAE), Asic1-/- mice showed both a markedly reduced clinical deficit and reduced axonal degeneration compared to wild-type mice. Consistently with acidosis-mediated injury, pH measurements in the spinal cord of EAE mice showed tissue acidosis sufficient to open ASIC1. The acidosis-related protective effect of Asic1 disruption was also observed in nerve explants in vitro. Amiloride, a licensed and clinically safe blocker of ASICs, was equally neuroprotective in nerve explants and in EAE. Although ASICs are also expressed by immune cells, this expression is unlikely to explain the neuroprotective effect of Asic1 inactivation, as CNS inflammation was similar in wild-type and Asic1-/- mice. In addition, adoptive transfer of T cells from wild-type mice did not affect the protection mediated by Asic1 disruption. These results suggest that ASIC1 blockers could provide neuroprotection in multiple sclerosis.


Subject(s)
Autoimmunity , Axons/pathology , Central Nervous System/pathology , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Sodium Channels/physiology , Acid Sensing Ion Channels , Amiloride/pharmacology , Animals , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Inflammation/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Nerve Degeneration , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/pharmacology , Sodium Channels/metabolism , Spinal Cord/pathology
8.
Eur J Neurosci ; 22(1): 39-49, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16029194

ABSTRACT

Contactin, a glycosyl-phosphatidylinositol (GPI)-anchored predominantly neuronal cell surface glycoprotein, associates with sodium channels Nav1.2, Nav1.3 and Nav1.9, and enhances the density of these channels on the plasma membrane in mammalian expression systems. However, a detailed functional analysis of these interactions and of untested putative interactions with other sodium channel isoforms in mammalian neuronal cells has not been carried out. We examined the expression and function of sodium channels in small-diameter dorsal root ganglion (DRG) neurons from contactin-deficient (CNTN-/-) mice, compared to CNTN+/+ litter mates. Nav1.9 is preferentially expressed in isolectin B4 (IB4)-positive neurons and thus we used this marker to subdivide small-diameter DRG neurons. Using whole-cell patch-clamp recording, we observed a greater than two-fold reduction of tetrodotoxin-resistant (TTX-R) Nav1.8 and Nav1.9 current densities in IB4+ DRG neurons cultured from CNTN-/- vs. CNTN+/+ mice. Current densities for TTX-sensitive (TTX-S) sodium channels were unaffected. Contactin's effect was selective for IB4+ neurons as current densities for both TTX-R and TTX-S channels were not significantly different in IB4- DRG neurons from the two genotypes. Consistent with these results, we have demonstrated a reduction in Nav1.8 and Nav1.9 immunostaining on peripherin-positive unmyelinated axons in sciatic nerves from CNTN-/- mice but detected no changes in the expression for the two major TTX-S channels Nav1.6 and Nav1.7. These data provide evidence of a role for contactin in selectively regulating the cell surface expression and current densities of TTX-R but not TTX-S Na+ channel isoforms in nociceptive DRG neurons; this regulation could modulate the membrane properties and excitability of these neurons.


Subject(s)
Axons/metabolism , Cell Adhesion Molecules, Neuronal/physiology , Cell Membrane/metabolism , Ganglia, Spinal/metabolism , Neurons, Afferent/metabolism , Sodium Channels/metabolism , Animals , Axons/drug effects , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Membrane/drug effects , Cells, Cultured , Contactins , Down-Regulation/drug effects , Down-Regulation/genetics , Ganglia, Spinal/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , NAV1.8 Voltage-Gated Sodium Channel , NAV1.9 Voltage-Gated Sodium Channel , Nerve Fibers, Unmyelinated/drug effects , Nerve Fibers, Unmyelinated/metabolism , Neurons, Afferent/drug effects , Neuropeptides/drug effects , Neuropeptides/metabolism , Nociceptors/drug effects , Nociceptors/metabolism , Patch-Clamp Techniques , Plant Lectins , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Tetrodotoxin/pharmacology
9.
Glia ; 49(2): 220-9, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15390090

ABSTRACT

Loss of axons is a major contributor to nonremitting deficits in the inflammatory demyelinating disease multiple sclerosis (MS). Based on biophysical studies showing that activity of axonal sodium channels can trigger axonal degeneration, recent studies have tested sodium channel-blocking drugs in experimental autoimmune encephalomyelitis (EAE), an animal model of MS, and have demonstrated a protective effect on axons. However, it is possible that, in addition to a direct effect on axons, sodium channel blockers may also interfere with inflammatory mechanisms. We therefore examined the novel hypothesis that sodium channels contribute to activation of microglia and macrophages in EAE and acute MS lesions. In this study, we demonstrate a robust increase of sodium channel Nav1.6 expression in activated microglia and macrophages in EAE and MS. We further demonstrate that treatment with the sodium channel blocker phenytoin ameliorates the inflammatory cell infiltrate in EAE by 75%. Supporting a role for sodium channels in microglial activation, we show that tetrodotoxin, a specific sodium channel blocker, reduces the phagocytic function of activated rat microglia by 40%. To further confirm a role of Nav1.6 in microglial activation, we examined the phagocytic capacity of microglia from med mice, which lack Nav1.6 channels, and show a 65% reduction in phagocytic capacity compared with microglia from wildtype mice. Our findings indicate that sodium channels are important for activation and phagocytosis of microglia and macrophages in EAE and MS and suggest that, in addition to a direct neuroprotective effect on axons, sodium channel blockade may ameliorate neuroinflammatory disorders via anti-inflammatory mechanisms.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Gliosis/metabolism , Macrophages/metabolism , Microglia/metabolism , Multiple Sclerosis/metabolism , Nerve Tissue Proteins/metabolism , Sodium Channels/metabolism , Animals , Axons/drug effects , Axons/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Gliosis/drug therapy , Gliosis/physiopathology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Multiple Sclerosis/drug therapy , Multiple Sclerosis/physiopathology , NAV1.6 Voltage-Gated Sodium Channel , Nerve Degeneration/chemically induced , Nerve Degeneration/drug therapy , Nerve Degeneration/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neuroprotective Agents/pharmacology , Phagocytosis/drug effects , Phagocytosis/genetics , Phenytoin/pharmacology , Phenytoin/therapeutic use , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/therapeutic use , Sodium Channels/genetics , Tetrodotoxin/pharmacology , Up-Regulation/drug effects , Up-Regulation/physiology
10.
Trends Pharmacol Sci ; 25(11): 584-91, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15491781

ABSTRACT

Following the loss of myelin from axons in multiple sclerosis, some axons recover the ability to conduct impulses despite the absence of an insulating sheath, providing a basis for remission of clinical deficits. By contrast, other axons degenerate and contribute to non-remitting clinical deficits and, thus, disability. Investigations using laboratory models of multiple sclerosis indicate that altered expression of two distinct isoforms of Na+ channels underlies these two processes, and the study of human tissue reveals similar changes in multiple sclerosis.


Subject(s)
Axons/metabolism , Multiple Sclerosis/metabolism , Sodium Channels/metabolism , Animals , Axons/pathology , Disease Models, Animal , Humans , Multiple Sclerosis/pathology , Protein Isoforms , Sodium Channels/classification
11.
Neurosci Lett ; 369(3): 186-90, 2004 Oct 21.
Article in English | MEDLINE | ID: mdl-15464262

ABSTRACT

Recent studies have indicated that, in addition to demyelination and axonal degeneration, a third factor, dysregulated ion channel expression, contributes to the pathophysiology of experimental autoimmune encephalomyelitis (EAE) and multiple sclerosis (MS). Consistent with this suggestion, upregulated expression of sodium channel Na(v)1.8 is observed in Purkinje neurons in EAE and MS, and biophysical studies indicate that aberrant expression of Na(v)1.8 produces abnormal Purkinje cell firing which may contribute to the development of cerebellar ataxia. However, the molecular mechanisms that contribute to the upregulation of Na(v)1.8 in Purkinje cells in EAE and MS have not yet been determined. Previous studies have shown that neurotrophic factors can modulate sodium channel expression and that elevated levels of NGF are present in EAE and MS. Using immunocytochemical methods, we examined the relationship between the upregulation of Na(v)1.8 and the expression of the NGF receptors p75 and TrkA in EAE. Here we demonstrate that upregulation of Na(v)1.8 is associated with expression of p75 and low levels of TrkA in the majority of Purkinje cells in EAE. These findings, together with previous studies demonstrating a modulatory role of NGF on sodium channel expression, suggest that NGF acting via p75 contributes to the upregulation of Na(v)1.8 in Purkinje cells in EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Purkinje Cells/metabolism , Sodium Channels/metabolism , Trans-Activators/metabolism , Adaptor Proteins, Signal Transducing , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Fluorescent Antibody Technique/methods , Glycoproteins/metabolism , Mice , Myelin-Oligodendrocyte Glycoprotein , NAV1.8 Voltage-Gated Sodium Channel , Peptide Fragments/metabolism , Receptor, trkA/metabolism , Transcription Factors , Up-Regulation
12.
J Neurosci ; 24(30): 6765-75, 2004 Jul 28.
Article in English | MEDLINE | ID: mdl-15282281

ABSTRACT

Voltage-gated sodium channels interact with cytosolic proteins that regulate channel trafficking and/or modulate the biophysical properties of the channels. Na(v)1.6 is heavily expressed at the nodes of Ranvier along adult CNS and PNS axons and along unmyelinated fibers in the PNS. In an initial yeast two-hybrid screen using the C terminus of Na(v)1.6 as a bait, we identified FHF2B, a member of the FGF homologous factor (FHF) subfamily, as an interacting partner of Na(v)1.6. Members of the FHF subfamily share approximately 70% sequence identity, and individual members demonstrate a cell- and tissue-specific expression pattern. FHF2 is abundantly expressed in the hippocampus and DRG neurons and colocalizes with Na(v)1.6 at mature nodes of Ranvier in myelinated sensory fibers in the dorsal root of the sciatic nerve. However, retinal ganglion cells and spinal ventral horn motor neurons show very low levels of FHF2 expression, and their axons exhibit no nodal FHF2 staining within the optic nerve and ventral root, respectively. Thus, FHF2 is selectively localized at nodes of dorsal root sensory but not ventral root motor axons. The coexpression of FHF2B and Na(v)1.6 in the DRG-derived cell line ND7/23 significantly increases the peak current amplitude and causes a 4 mV depolarizing shift of voltage-dependent inactivation of the channel. The preferential expression of FHF2B in sensory neurons may provide a basis for physiological differences in sodium currents that have been reported at the nodes of Ranvier in sensory versus motor axons.


Subject(s)
Fibroblast Growth Factors/metabolism , Ganglia, Spinal/chemistry , Hippocampus/chemistry , Nerve Tissue Proteins/metabolism , Neurons, Afferent/chemistry , Ranvier's Nodes/chemistry , Sodium Channels/metabolism , Animals , Anterior Horn Cells/chemistry , Axons/chemistry , Axons/ultrastructure , Brain Chemistry , Cells, Cultured/chemistry , Fibroblast Growth Factors/analysis , Fibroblast Growth Factors/genetics , Ganglia, Spinal/cytology , Humans , Mice , NAV1.6 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Neurons, Afferent/physiology , Organ Specificity , Protein Binding , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Recombinant Fusion Proteins/metabolism , Retinal Ganglion Cells/chemistry , Sciatic Nerve/chemistry , Sciatic Nerve/cytology , Sodium Channels/analysis , Sodium Channels/genetics , Transfection , Two-Hybrid System Techniques
13.
J Neurosci ; 24(20): 4832-9, 2004 May 19.
Article in English | MEDLINE | ID: mdl-15152043

ABSTRACT

Peripheral nerve injury is known to upregulate the rapidly repriming Na(v)1.3 sodium channel within first-order spinal sensory neurons. In this study, we hypothesized that (1) after peripheral nerve injury, second-order dorsal horn neurons abnormally express Na(v)1.3, which (2) contributes to the responsiveness of these dorsal horn neurons and to pain-related behaviors. To test these hypotheses, adult rats underwent chronic constriction injury (CCI) of the sciatic nerve. Ten days after CCI, allodynia and hyperalgesia were evident. In situ hybridization, quantitative reverse transcription-PCR, and immunocytochemical analysis revealed upregulation of Na(v)1.3 in dorsal horn nociceptive neurons but not in astrocytes or microglia, and unit recordings demonstrated hyperresponsiveness of dorsal horn sensory neurons. Intrathecal antisense oligodeoxynucleotides targeting Na(v)1.3 decreased the expression of Na(v)1.3 mRNA and protein, reduced the hyperresponsiveness of dorsal horn neurons, and attenuated pain-related behaviors after CCI, all of which returned after cessation of antisense delivery. These results demonstrate for the first time that sodium channel expression is altered within higher-order spinal sensory neurons after peripheral nerve injury and suggest a link between misexpression of the Na(v)1.3 sodium channel and central mechanisms that contribute to neuropathic pain after peripheral nerve injury.


Subject(s)
Nerve Tissue Proteins/metabolism , Neurons, Afferent/metabolism , Pain/physiopathology , Sciatic Neuropathy/physiopathology , Sodium Channels/metabolism , Spinal Cord/metabolism , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Disease Models, Animal , Hyperalgesia/etiology , Hyperalgesia/metabolism , Immunohistochemistry , In Situ Hybridization , Ligation , Male , NAV1.3 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neurons, Afferent/drug effects , Oligonucleotides, Antisense/pharmacology , Pain/etiology , Pain Measurement/drug effects , Posterior Horn Cells/drug effects , Posterior Horn Cells/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sciatic Neuropathy/complications , Sciatic Neuropathy/metabolism , Sodium Channels/genetics , Spinal Cord/drug effects , Up-Regulation
14.
Proc Natl Acad Sci U S A ; 101(21): 8168-73, 2004 May 25.
Article in English | MEDLINE | ID: mdl-15148385

ABSTRACT

Although voltage-gated sodium channels are known to be deployed along experimentally demyelinated axons, the molecular identities of the sodium channels expressed along axons in human demyelinating diseases such as multiple sclerosis (MS) have not been determined. Here we demonstrate changes in the expression of sodium channels in demyelinated axons in MS, with Nav1.6 confined to nodes of Ranvier in controls but with diffuse distribution of Nav1.2 and Nav1.6 along extensive regions of demyelinated axons within acute MS plaques. Using triple-labeled fluorescent immunocytochemistry, we also show that Nav1.6, which is known to produce a persistent sodium current, and the Na+/Ca2+ exchanger, which can be driven by persistent sodium current to import damaging levels of calcium into axons, are colocalized with beta-amyloid precursor protein, a marker of axonal injury, in acute MS lesions. Our results demonstrate the molecular identities of the sodium channels expressed along demyelinated and degenerating axons in MS and suggest that coexpression of Nav1.6 and Na+/Ca2+ exchanger is associated with axonal degeneration in MS.


Subject(s)
Axons/metabolism , Axons/pathology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Neurons/metabolism , Sodium Channels/metabolism , Sodium-Calcium Exchanger/metabolism , Acute Disease , Amyloid beta-Protein Precursor/metabolism , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Humans , Middle Aged , NAV1.2 Voltage-Gated Sodium Channel , NAV1.6 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/metabolism , Neurons/pathology , Sodium/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology
15.
Exp Brain Res ; 158(1): 1-8, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15118796

ABSTRACT

Cerebellar deficits in multiple sclerosis (MS) tend to persist and can produce significant disability. Although the pathophysiological basis for these deficits is not clear, it was recently reported that the expression of the sensory neuron-specific sodium channel Nav1.8 (which is not normally expressed within the cerebellum) is aberrantly upregulated within Purkinje cells in experimental allergic encephalomyelitis (EAE) and in human MS. The expression of Nav1.8 in cultured Purkinje cells has been shown to alter the activity pattern of these cells in vitro by decreasing the number of spikes per conglomerate action potential and by contributing to the production of sustained, pacemaker-like activity upon depolarization, suggesting the hypothesis that, in pathophysiological situations where Nav1.8 is upregulated within Purkinje cells, the pattern of activity in these cells will be altered. In the present study, we examined this hypothesis in vivo in mice with EAE. Our results demonstrate a reduction in the number of secondary spikes per complex spike and irregularity in the temporal organization of secondary spikes in Purkinje cells from mice with EAE in which Nav1.8 is upregulated. We also observed abnormal bursting activity in Purkinje cells from mice with EAE, which was not observed in control animals. These results demonstrate functional changes in Purkinje cells in vivo within their native cerebellar environment in EAE, a model of MS, and support the hypothesis that misexpression of Nav1.8 can contribute to cerebellar deficits in neuroinflammatory disorders by altering the pattern of electrical activity within the cerebellum.


Subject(s)
Cerebellar Cortex/physiopathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Multiple Sclerosis/physiopathology , Purkinje Cells/metabolism , Sodium Channels/metabolism , Action Potentials/physiology , Afferent Pathways/immunology , Afferent Pathways/metabolism , Afferent Pathways/physiopathology , Animals , Cell Shape/physiology , Cerebellar Cortex/immunology , Cerebellar Cortex/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Mice , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , NAV1.8 Voltage-Gated Sodium Channel , Purkinje Cells/immunology , RNA, Messenger/metabolism , Sodium/metabolism , Sodium Channels/genetics , Synaptic Transmission/physiology , Up-Regulation/physiology
16.
Neurobiol Dis ; 15(2): 221-8, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15006692

ABSTRACT

The hyperosmolality associated with diabetes mellitus triggers an increase in neuronal activity and vasopressin production within magnocellular neurosecretory cells (MNCs) of the hypothalamic supraoptic nucleus (SON). In this study, we examined the effect of chronic diabetes on the function and survival of these neurons. After 6 months, but not 6 weeks, of streptozotocin (STZ)-induced diabetes, we observed an increase in the appearance of small hyperchromatic neurons and a decrease in SON neuronal density. A subpopulation of neurons within the SON at this time point demonstrated positive staining for cleaved caspase-3 and TUNEL, two markers of apoptosis. In addition, the number of vasopressin-positive neurons was decreased. Markers for apoptosis did not colocalize with vasopressin immunopositivity; this was probably due to a diabetes-induced degenerative process causing downregulation of vasopressin expression or depletion of neuropeptide. Although the phenotypes of the apoptotic neurons were not identified, other SON neurons including oxytocin-producing neurons are unlikely to be affected by chronic hyperglycemia. Microglial hypertrophy and condensation were also observed in the 6-month diabetic SON. Although upregulation of vasopressin production in response to acute hyperosmolality is adaptive, prolonged overstimulation of vasopressin-producing neurons in chronic diabetes results in neurodegeneration and apoptosis.


Subject(s)
Antigens, CD , Antigens, Neoplasm , Antigens, Surface , Apoptosis/physiology , Avian Proteins , Blood Proteins , Diabetes Mellitus, Experimental/pathology , Nerve Degeneration/pathology , Neurons/pathology , Supraoptic Nucleus/pathology , Vasopressins/metabolism , Animals , Basigin , Caspase 3 , Caspases/metabolism , Chronic Disease , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Down-Regulation/physiology , Glial Fibrillary Acidic Protein/metabolism , Gliosis/pathology , Gliosis/physiopathology , In Situ Nick-End Labeling , Male , Membrane Glycoproteins/metabolism , Microglia/pathology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Supraoptic Nucleus/metabolism , Water-Electrolyte Balance/physiology
17.
Brain ; 127(Pt 2): 294-303, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14662515

ABSTRACT

Axonal degeneration contributes to the development of non-remitting neurological deficits and disability in multiple sclerosis, but the molecular mechanisms that underlie axonal loss in multiple sclerosis are not clearly understood. Studies of white matter axonal injury have demonstrated that voltage-gated sodium channels can provide a route for sodium influx into axons that triggers reverse operation of the Na(+)/Ca(2+) exchanger (NCX) and subsequent influx of damaging levels of intra-axonal calcium. The molecular identities of the involved sodium channels have, however, not been determined. We have previously demonstrated extensive regions of diffuse expression of Na(v)1.6 and Na(v)1.2 sodium channels along demyelinated axons in experimental allergic encephalomyelitis (EAE). Based on the hypothesis that the co-localization of Na(v)1.6 and NCX along extensive regions of demyelinated axons may predispose these axons to injury, we examined the expression of myelin basic protein, Na(v)1.2, Na(v)1.6, NCX and beta-amyloid precursor protein (beta-APP), a marker of axonal injury, in the spinal cord dorsal columns of mice with EAE. We demonstrate a significant increase in the number of demyelinated axons demonstrating diffuse Na(v)1.6 and Na(v)1.2 sodium channel immunoreactivity in EAE (92.2 +/- 2.1% of beta-APP positive axons were Na(v)1.6-positive). Only 38.0 +/- 2.9% of beta-APP positive axons were Na(v)1.2 positive, and 95% of these co-expressed Na(v)1.6 together with Na(v)1.2. Using triple-labelled fluorescent immunohistochemistry, we demonstrate that 73.5 +/- 4.3% of beta-APP positive axons co-express Na(v)1.6 and NCX, compared with 4.4 +/- 1.0% in beta-APP negative axons. Our results indicate that co-expression of Na(v)1.6 and NCX is associated with axonal injury in the spinal cord in EAE.


Subject(s)
Axons/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Nerve Degeneration/metabolism , Nerve Tissue Proteins/metabolism , Sodium Channels/metabolism , Sodium-Calcium Exchanger/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Axons/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Male , Mice , Myelin Sheath/pathology , NAV1.2 Voltage-Gated Sodium Channel , NAV1.6 Voltage-Gated Sodium Channel , Nerve Degeneration/pathology , Spinal Cord/metabolism , Spinal Cord/pathology
18.
J Neuropathol Exp Neurol ; 62(9): 968-75, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14533785

ABSTRACT

Multiple sclerosis (MS) is recognized to involve demyelination and axonal atrophy but accumulating evidence suggests that dysregulated sodium channel expression may also contribute to its pathophysiology. Recent studies have demonstrated that the expression of Na(v)1.8 voltage-gated sodium channels, which are normally undetectable within the CNS, is upregulated in cerebellar Purkinje cells in experimental allergic encephalomyelitis (EAE) and MS, and suggest that the aberrant expression of these channels contributes to clinical dysfunction by distorting the firing pattern of these neurons. In this study we examined the temporal pattern of upregulation for Na(v)1.8 mRNA and protein in chronic relapsing EAE by in situ hybridization and immunocytochemistry, respectively. Our results demonstrate a positive correlation between disease duration and degree of upregulation of Na(v)1.8 mRNA and protein in Purkinje neurons in chronic-relapsing EAE. The progressive deterioration in clinical baseline scores (i.e. in clinical scores during remissions) is paralleled by a continued increase in Na(v)1.8 mRNA and protein expression, but temporary worsening during relapses is not associated with transient changes in Na(v)1.8 expression. These results provide evidence that the expression of sodium channel Na(v)1.8 contributes to the development of clinical deficits in an in vivo model of neuroinflammatory disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Nerve Tissue Proteins/biosynthesis , Purkinje Cells/metabolism , Sodium Channels/biosynthesis , Up-Regulation/physiology , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Gene Expression Regulation/physiology , Mice , Mice, Biozzi , NAV1.8 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sodium Channels/genetics
19.
J Neurosci ; 23(26): 8881-92, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14523090

ABSTRACT

Spinal cord injury (SCI) can result in hyperexcitability of dorsal horn neurons and central neuropathic pain. We hypothesized that these phenomena are consequences, in part, of dysregulated expression of voltage-gated sodium channels. Because the rapidly repriming TTX-sensitive sodium channel Nav1.3 has been implicated in peripheral neuropathic pain, we investigated its role in central neuropathic pain after SCI. In this study, adult male Sprague Dawley rats underwent T9 spinal contusion injury. Four weeks after injury when extracellular recordings demonstrated hyperexcitability of L3-L5 dorsal horn multireceptive nociceptive neurons, and when pain-related behaviors were evident, quantitative RT-PCR, in situ hybridization, and immunocytochemistry revealed an upregulation of Nav1.3 in dorsal horn nociceptive neurons. Intrathecal administration of antisense oligodeoxynucleotides (ODNs) targeting Nav1.3 resulted in decreased expression of Nav1.3 mRNA and protein, reduced hyperexcitability of multireceptive dorsal horn neurons, and attenuated mechanical allodynia and thermal hyperalgesia after SCI. Expression of Nav1.3 protein and hyperexcitability in dorsal horn neurons as well as pain-related behaviors returned after cessation of antisense delivery. Responses to normally noxious stimuli and motor function were unchanged in SCI animals administered Nav1.3 antisense, and administration of mismatch ODNs had no effect. These results demonstrate for the first time that Nav1.3 is upregulated in second-order dorsal horn sensory neurons after nervous system injury, showing that SCI can trigger changes in sodium channel expression, and suggest a functional link between Nav1.3 expression and neuronal hyperexcitability associated with central neuropathic pain.


Subject(s)
Nerve Tissue Proteins/metabolism , Neuralgia/physiopathology , Neurons/metabolism , Posterior Horn Cells/physiopathology , Sodium Channels/metabolism , Spinal Cord Injuries/physiopathology , Animals , Behavior, Animal/drug effects , Cell Count , Disease Models, Animal , Electrophysiology , Immunohistochemistry , In Situ Hybridization , Male , NAV1.3 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/genetics , Neuralgia/complications , Neurons/drug effects , Neurons/pathology , Nociceptors/pathology , Nociceptors/physiopathology , Oligodeoxyribonucleotides, Antisense/metabolism , Oligodeoxyribonucleotides, Antisense/pharmacology , Pain Measurement , Posterior Horn Cells/drug effects , Posterior Horn Cells/pathology , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sodium Channels/genetics , Spinal Cord Injuries/complications , Up-Regulation/drug effects , Up-Regulation/physiology
20.
Brain ; 126(Pt 7): 1552-61, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12805113

ABSTRACT

Myelinated fibres are characterized by the aggregation of Nav1.6 sodium channels within the axon membrane at nodes of Ranvier, where their presence supports saltatory conduction. In this study, we used immunocytochemical methods to study the organization of sodium channels along axons in experimental allergic encephalomyelitis (EAE), a model of multiple sclerosis. We studied axons within the optic nerve, a CNS tract commonly affected in multiple sclerosis, and their cell bodies of origin (retinal ganglion cells), using subtype-specific antibodies generated against sodium channel subtypes Nav1.1, Nav1.2, Nav1.3 and Nav1.6, which previously have been shown to be expressed by retinal ganglion cells. We demonstrate a significant switch from Nav1.6 to Nav1.2 expression in the optic nerve in EAE; there was a reduction in frequency of Nav1.6-positive nodes (84.5% Nav1.6-immunopositive nodes in control versus 32.9% in EAE) and increased frequency of Nav1.2-positive nodes (11.8% Nav1.2 immunopositive nodes in control versus 74.9% in EAE). Moreover, we observed a significant increase in the number of linear (presumably demyelinated) axonal profiles demonstrating extended diffuse immunostaining for Nav1.2 in EAE versus control optic nerves. These changes within the optic nerve are paralleled by decreased levels of Nav1.6 and increased Nav1.2 protein, together with increased levels of Nav1.2 mRNA, within retinal ganglion cells in EAE. Our findings of a loss of Nav1.6 and increased expression of Nav1.2 suggest that electrogenesis in EAE may revert to a stage similar to that observed in immature retinal ganglion cells in which Nav1.2 channels support conduction of action potentials along axons.


Subject(s)
Axons/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Optic Nerve/metabolism , Sodium Channels/metabolism , Animals , Disease Models, Animal , Down-Regulation , Mice , Multiple Sclerosis/metabolism , Ranvier's Nodes/metabolism , Retinal Ganglion Cells/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...