Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Nat Rev Neurosci ; 16(9): 511-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26243570

RESUMO

The voltage-gated sodium channel Na(V)1.9 is preferentially expressed in nociceptors and has been shown in rodent models to have a major role in inflammatory and neuropathic pain. These studies suggest that by selectively targeting Na(V)1.9, it might be possible to ameliorate pain without inducing adverse CNS side effects such as sedation, confusion and addictive potential. Three recent studies in humans--two genetic and functional studies in rare genetic disorders, and a third study showing a role for Na(V)1.9 in painful peripheral neuropathy--have demonstrated that Na(V)1.9 plays an important part both in regulating sensory neuron excitability and in pain signalling. With this human validation, attention is turning to this channel as a potential therapeutic target for pain.


Assuntos
Dor/metabolismo , Células Receptoras Sensoriais/metabolismo , Animais , Humanos , Mutação/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.9/biossíntese , Nociceptores/metabolismo , Nociceptores/patologia , Dor/diagnóstico , Dor/tratamento farmacológico , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/patologia , Bloqueadores dos Canais de Sódio/farmacologia , Bloqueadores dos Canais de Sódio/uso terapêutico
2.
Glia ; 66(10): 2174-2187, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30194875

RESUMO

Astrogliosis is a hallmark of neuroinflammatory disorders such as multiple sclerosis (MS). A detailed understanding of the underlying molecular mechanisms governing astrogliosis might facilitate the development of therapeutic targets. We investigated whether Nav1.5 expression in astrocytes plays a role in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), a murine model of MS. We created a conditional knockout of Nav1.5 in astrocytes and determined whether this affects the clinical course of EAE, focal macrophage and T cell infiltration, and diffuse activation of astrocytes. We show that deletion of Nav1.5 from astrocytes leads to significantly worsened clinical outcomes in EAE, with increased inflammatory infiltrate in both early and late stages of disease, unexpectedly, in a sex-specific manner. Removal of Nav1.5 in astrocytes leads to increased inflammation in female mice with EAE, including increased astroglial response and infiltration of T cells and phagocytic monocytes. These cellular changes are consistent with more severe EAE clinical scores. Additionally, we found evidence suggesting possible dysregulation of the immune response-particularly with regard to infiltrating macrophages and activated microglia-in female Nav1.5 KO mice compared with WT littermate controls. Together, our results show that deletion of Nav1.5 from astrocytes leads to significantly worsened clinical outcomes in EAE, with increased inflammatory infiltrate in both early and late stages of disease, in a sex-specific manner.


Assuntos
Astrócitos/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Esclerose Múltipla/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Caracteres Sexuais , Animais , Astrócitos/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Progressão da Doença , Encefalomielite Autoimune Experimental/patologia , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Monócitos/metabolismo , Monócitos/patologia , Esclerose Múltipla/patologia , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Medula Espinal/metabolismo , Medula Espinal/patologia , Linfócitos T/metabolismo , Linfócitos T/patologia
3.
Annu Rev Neurosci ; 33: 325-47, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20367448

RESUMO

Nociception is essential for survival whereas pathological pain is maladaptive and often unresponsive to pharmacotherapy. Voltage-gated sodium channels, Na(v)1.1-Na(v)1.9, are essential for generation and conduction of electrical impulses in excitable cells. Human and animal studies have identified several channels as pivotal for signal transmission along the pain axis, including Na(v)1.3, Na(v)1.7, Na(v)1.8, and Na(v)1.9, with the latter three preferentially expressed in peripheral sensory neurons and Na(v)1.3 being upregulated along pain-signaling pathways after nervous system injuries. Na(v)1.7 is of special interest because it has been linked to a spectrum of inherited human pain disorders. Here we review the contribution of these sodium channel isoforms to pain.


Assuntos
Nociceptores/metabolismo , Dor/metabolismo , Dor/fisiopatologia , Canais de Sódio/metabolismo , Animais , Modelos Animais de Doenças , Predisposição Genética para Doença/genética , Humanos , Dor/genética , Células Receptoras Sensoriais/metabolismo , Canais de Sódio/genética
4.
Nat Rev Neurosci ; 14(1): 49-62, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23232607

RESUMO

The voltage-gated sodium channel Na(V)1.7 is preferentially expressed in peripheral somatic and visceral sensory neurons, olfactory sensory neurons and sympathetic ganglion neurons. Na(V)1.7 accumulates at nerve fibre endings and amplifies small subthreshold depolarizations, poising it to act as a threshold channel that regulates excitability. Genetic and functional studies have added to the evidence that Na(V)1.7 is a major contributor to pain signalling in humans, and homology modelling based on crystal structures of ion channels suggests an atomic-level structural basis for the altered gating of mutant Na(V)1.7 that causes pain.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.7/genética , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Dor , Animais , Biofísica , Humanos , Modelos Moleculares , Mutação/genética , Dor/genética , Dor/patologia , Dor/fisiopatologia , Nervos Periféricos/efeitos dos fármacos , Nervos Periféricos/metabolismo , Nervos Periféricos/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia
5.
Glia ; 64(10): 1628-45, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26919466

RESUMO

Voltage-gated sodium channels are required for electrogenesis in excitable cells. Their activation, triggered by membrane depolarization, generates transient sodium currents that initiate action potentials in neurons, cardiac, and skeletal muscle cells. Cells that have not traditionally been considered to be excitable (nonexcitable cells), including glial cells, also express sodium channels in physiological conditions as well as in pathological conditions. These channels contribute to multiple functional roles that are seemingly unrelated to the generation of action potentials. Here, we discuss the dynamics of sodium channel expression in astrocytes and microglia, and review evidence for noncanonical roles in effector functions of these cells including phagocytosis, migration, proliferation, ionic homeostasis, and secretion of chemokines/cytokines. We also examine possible mechanisms by which sodium channels contribute to the activity of glial cells, with an eye toward therapeutic implications for central nervous system disease. GLIA 2016;64:1628-1645.


Assuntos
Astrócitos/metabolismo , Microglia/metabolismo , Canais de Sódio/metabolismo , Potenciais de Ação/fisiologia , Animais , Humanos , Sistema Nervoso/citologia
6.
Mol Pain ; 122016.
Artigo em Inglês | MEDLINE | ID: mdl-27821467

RESUMO

Small fiber neuropathy is a painful sensory nervous system disorder characterized by damage to unmyelinated C- and thinly myelinated Aδ- nerve fibers, clinically manifested by burning pain in the distal extremities and dysautonomia. The clinical onset in adulthood suggests a time-dependent process. The mechanisms that underlie nerve fiber injury in small fiber neuropathy are incompletely understood, although roles for energetic stress have been suggested. In the present study, we report time-dependent degeneration of neurites from dorsal root ganglia neurons in culture expressing small fiber neuropathy-associated G856D mutant Nav1.7 channels and demonstrate a time-dependent increase in intracellular calcium levels [Ca2+]i and reactive oxygen species, together with a decrease in ATP levels. Together with a previous clinical report of burning pain in the feet and hands associated with reduced levels of Na+/K+-ATPase in humans with high altitude sickness, the present results link energetic stress and reactive oxygen species production with the development of a painful neuropathy that preferentially affects small-diameter axons.


Assuntos
Trifosfato de Adenosina/metabolismo , Axônios/patologia , Cálcio/metabolismo , Mutação/genética , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Degeneração Neural/metabolismo , Neurônios/citologia , Animais , Células Cultivadas , Gânglios Espinais/citologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Degeneração Neural/genética , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo , Transfecção , Proteína Vermelha Fluorescente
7.
Mol Pain ; 11: 26, 2015 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-25957174

RESUMO

BACKGROUND: The skin is a morphologically complex organ that serves multiple complementary functions, including an important role in thermoregulation, which is mediated by a rich vasculature that is innervated by sympathetic and sensory endings. Two autosomal dominant disorders characterized by episodes of severe pain, inherited erythromelalgia (IEM) and paroxysmal extreme pain disorder (PEPD) have been directly linked to mutations that enhance the function of sodium channel Nav1.7. Pain attacks are accompanied by reddening of the skin in both disorders. Nav1.7 is known to be expressed at relatively high levels within both dorsal root ganglion (DRG) and sympathetic ganglion neurons, and mutations that enhance the activity of Nav1.7 have been shown to have profound effects on the excitability of both cell-types, suggesting that dysfunction of sympathetic and/or sensory fibers, which release vasoactive peptides at skin vasculature, may contribute to skin reddening in IEM and PEPD. RESULTS: In the present study, we demonstrate that smooth muscle cells of cutaneous arterioles and arteriole-venule shunts (AVS) in the skin express sodium channel Nav1.7. Moreover, Nav1.7 is expressed by endothelial cells lining the arterioles and AVS and by sensory and sympathetic fibers innervating these vascular elements. CONCLUSIONS: These observations suggest that the activity of mutant Nav1.7 channels in smooth muscle cells of skin vasculature and innervating sensory and sympathetic fibers contribute to the skin reddening and/or pain in IEM and PEPD.


Assuntos
Axônios/metabolismo , Endotélio/metabolismo , Células Musculares/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Pele/inervação , Pele/metabolismo , Eritromelalgia/genética , Gânglios Espinais/metabolismo , Humanos , Mutação/genética
8.
J Neurosci ; 33(49): 19250-61, 2013 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-24305821

RESUMO

Axonal degeneration occurs in multiple neurodegenerative disorders of the central and peripheral nervous system. Although the underlying molecular pathways leading to axonal degeneration are incompletely understood, accumulating evidence suggests contributions of impaired mitochondrial function, disrupted axonal transport, and/or dysfunctional intracellular Ca(2+)-homeostasis in the injurious cascade associated with axonal degeneration. Utilizing an in vitro model of axonal degeneration, we studied a subset of mouse peripheral sensory neurons in which neurites were exposed selectively to conditions associated with the pathogenesis of axonal neuropathies in vivo. Rotenone-induced mitochondrial dysfunction resulted in neurite degeneration accompanied by reduced ATP levels and increased ROS levels in neurites. Blockade of voltage-gated sodium channels with TTX and reverse (Ca(2+)-importing) mode of the sodium-calcium exchanger (NCX) with KB-R7943 partially protected rotenone-treated neurites from degeneration, suggesting a contribution of sodium channels and reverse NCX activity to the degeneration of neurites resulting from impaired mitochondrial function. Pharmacological inhibition of the Na(+)/K(+)-ATPase with ouabain induced neurite degeneration, which was attenuated by TTX and KB-R7943, supporting a contribution of sodium channels in axonal degenerative pathways accompanying impaired Na(+)/K(+)-ATPase activity. Conversely, oxidant stress (H2O2)-induced neurite degeneration was not attenuated by TTX. Our results demonstrate that both energetic and oxidative stress targeted selectively to neurites induces neurite degeneration and that blockade of sodium channels and of reverse NCX activity blockade partially protects neurites from injury due to energetic stress, but not from oxidative stress induced by H2O2.


Assuntos
Axônios/fisiologia , Gânglios Espinais/fisiologia , Doenças Mitocondriais/fisiopatologia , Degeneração Neural/fisiopatologia , Neuritos/fisiologia , Canais de Sódio/fisiologia , Animais , Axotomia , Morte Celular/fisiologia , Células Cultivadas , Gânglios Espinais/citologia , Humanos , Peróxido de Hidrogênio/toxicidade , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Microtúbulos/fisiologia , Neuritos/ultraestrutura , Oxidantes/toxicidade , Rotenona/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Trocador de Sódio e Cálcio/antagonistas & inibidores , Trocador de Sódio e Cálcio/metabolismo , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , ATPase Trocadora de Sódio-Potássio/fisiologia , Tetrodotoxina/toxicidade , Tioureia/análogos & derivados , Tioureia/farmacologia , Desacopladores/farmacologia
9.
Glia ; 62(7): 1162-75, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24740847

RESUMO

Astrogliosis is a prominent feature of many, if not all, pathologies of the brain and spinal cord, yet a detailed understanding of the underlying molecular pathways involved in the transformation from quiescent to reactive astrocyte remains elusive. We investigated the contribution of voltage-gated sodium channels to astrogliosis in an in vitro model of mechanical injury to astrocytes. Previous studies have shown that a scratch injury to astrocytes invokes dual mechanisms of migration and proliferation in these cells. Our results demonstrate that wound closure after mechanical injury, involving both migration and proliferation, is attenuated by pharmacological treatment with tetrodotoxin (TTX) and KB-R7943, at a dose that blocks reverse mode of the Na(+) /Ca(2+) exchanger (NCX), and by knockdown of Nav 1.5 mRNA. We also show that astrocytes display a robust [Ca(2+) ]i transient after mechanical injury and demonstrate that this [Ca(2+) ]i response is also attenuated by TTX, KB-R7943, and Nav 1.5 mRNA knockdown. Our results suggest that Nav 1.5 and NCX are potential targets for modulation of astrogliosis after injury via their effect on [Ca(2+) ]i .


Assuntos
Astrócitos/fisiologia , Gliose/fisiopatologia , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Ferimentos e Lesões/fisiopatologia , Animais , Astrócitos/efeitos dos fármacos , Cálcio/metabolismo , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Células Cultivadas , Córtex Cerebral , Técnicas de Silenciamento de Genes , Gliose/tratamento farmacológico , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Estimulação Física , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Bloqueadores dos Canais de Sódio/farmacologia , Trocador de Sódio e Cálcio/antagonistas & inibidores , Tetrodotoxina/farmacologia , Tioureia/análogos & derivados , Tioureia/farmacologia , Cicatrização/efeitos dos fármacos , Cicatrização/fisiologia , Ferimentos e Lesões/tratamento farmacológico
10.
Glia ; 62(12): 2080-95, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25043721

RESUMO

Microglia are motile resident immune cells of the central nervous system (CNS) that continuously explore their territories for threats to tissue homeostasis. Following CNS insult (e.g., cellular injury, infection, or ischemia), microglia respond to signals such as ATP, transform into an activated state, and migrate towards the threat. Directed migration is a complex and highly-coordinated process involving multiple intersecting cellular pathways, including signal transduction, membrane adhesion and retraction, cellular polarization, and rearrangement of cytoskeletal elements. We previously demonstrated that the activity of sodium channels contributes to ATP-induced migration of microglia. Here we show that TTX-sensitive sodium channels, specifically NaV 1.6, participate in an initial event in the migratory process, i.e., the formation in ATP-stimulated microglia of polymerized actin-rich membrane protrusions, lamellipodia, containing accumulations of Rac1 and phosphorylated ERK1/2. We also examined Ca(2+) transients in microglia and found that blockade of sodium channels with TTX produced a downward shift in the level of [Ca(2+) ]i during the delayed, slower recovery of [Ca(2+) ]i following ATP stimulation. These observations demonstrate a modulatory role of sodium channels on Ca(2+) transients in microglia that are likely to affect down-stream signaling cascades. Consistent with these observations, we demonstrate that ATP-induced microglial migration is mediated via Rac1 and ERK1/2, but not p38α/ß and JNK, dependent pathways, and that activation of both Rac1 and ERK1/2 is modulated by sodium channel activity. Our results provide evidence for a direct link between sodium channel activity and modulation of Rac1 and ERK1/2 activation in ATP-stimulated microglia, possibly by regulating Ca(2+) transients.


Assuntos
Trifosfato de Adenosina/farmacologia , Microglia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Pseudópodes/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Camundongos Transgênicos , Microglia/citologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Pseudópodes/efeitos dos fármacos , Pseudópodes/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Bloqueadores dos Canais de Sódio/farmacologia
11.
Ann Neurol ; 73(1): 140-5, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23280954

RESUMO

Small-fiber neuropathy (SFN) is characterized by injury to small-diameter peripheral nerve axons and intraepidermal nerve fibers (IENF). Although mechanisms underlying loss of IENF in SFN are poorly understood, available data suggest that it results from axonal degeneration and reduced regenerative capacity. Gain-of-function variants in sodium channel Na(V)1.7 that increase firing frequency and spontaneous firing of dorsal root ganglion (DRG) neurons have recently been identified in ∼30% of patients with idiopathic SFN. In the present study, to determine whether these channel variants can impair axonal integrity, we developed an in vitro assay of DRG neurite length, and examined the effect of 3 SFN-associated variant Na(V)1.7 channels, I228M, M932L/V991L (ML/VL), and I720K, on DRG neurites in vitro. At 3 days after culturing, DRG neurons transfected with I228M channels exhibited ∼20% reduced neurite length compared to wild-type channels; DRG neurons transfected with ML/VL and I720K variants displayed a trend toward reduced neurite length. I228M-induced reduction in neurite length was ameliorated by the use-dependent sodium channel blocker carbamazepine and by a blocker of reverse Na-Ca exchange. These in vitro observations provide evidence supporting a contribution of the I228M variant Na(V)1.7 channel to impaired regeneration and/or degeneration of sensory axons in idiopathic SFN, and suggest that enhanced sodium channel activity and reverse Na-Ca exchange can contribute to a decrease in length of peripheral sensory axons.


Assuntos
Axônios/fisiologia , Gânglios Espinais/fisiologia , Variação Genética/genética , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Doenças do Sistema Nervoso Periférico/genética , Animais , Axônios/patologia , Morte Celular/genética , Células Cultivadas , Gânglios Espinais/patologia , Humanos , Doenças do Sistema Nervoso Periférico/patologia , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/patologia , Células Receptoras Sensoriais/fisiologia
12.
Mol Pain ; 9: 39, 2013 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-23924059

RESUMO

BACKGROUND: NaV1.7 is preferentially expressed, at relatively high levels, in peripheral neurons, and is often referred to as a "peripheral" sodium channel, and NaV1.7-specific blockers are under study as potential pain therapeutics which might be expected to have minimal CNS side effects. However, occasional reports of patients with NaV1.7 gain-of-function mutations and apparent hypothalamic dysfunction have appeared. The two sodium channels previously studied within the rat hypothalamic supraoptic nucleus, NaV1.2 and NaV1.6, display up-regulated expression in response to osmotic stress. RESULTS: Here we show that NaV1.7 is present within vasopressin-producing neurons and oxytocin-producing neurons within the rat hypothalamus, and demonstrate that the level of Nav1.7 immunoreactivity is increased in these cells in response to osmotic stress. CONCLUSIONS: NaV1.7 is present within neurosecretory neurons of rat supraoptic nucleus, where the level of immunoreactivity is dynamic, increasing in response to osmotic stress. Whether NaV1.7 levels are up-regulated within the human hypothalamus in response to environmental factors or stress, and whether NaV1.7 plays a functional role in human hypothalamus, is not yet known. Until these questions are resolved, the present findings suggest the need for careful assessment of hypothalamic function in patients with NaV1.7 mutations, especially when subjected to stress, and for monitoring of hypothalamic function as NaV1.7 blocking agents are studied.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Neurônios/metabolismo , Pressão Osmótica/fisiologia , Núcleo Supraóptico/metabolismo , Animais , Hipotálamo/metabolismo , Imuno-Histoquímica , Masculino , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Dor/metabolismo , Ratos , Ratos Sprague-Dawley , Regulação para Cima
13.
Mol Pain ; 9: 52, 2013 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-24144460

RESUMO

BACKGROUND: Voltage-gated sodium channels Nav1.8 and Nav1.9 are expressed preferentially in small diameter sensory neurons, and are thought to play a role in the generation of ectopic activity in neuronal cell bodies and/or their axons following peripheral nerve injury. The expression of Nav1.8 and Nav1.9 has been quantified in human lingual nerves that have been previously injured inadvertently during lower third molar removal, and any correlation between the expression of these ion channels and the presence or absence of dysaesthesia investigated. RESULTS: Immunohistochemical processing and quantitative image analysis revealed that Nav1.8 and Nav1.9 were expressed in human lingual nerve neuromas from patients with or without symptoms of dysaesthesia. The level of Nav1.8 expression was significantly higher in patients reporting pain compared with no pain, and a significant positive correlation was observed between levels of Nav1.8 expression and VAS scores for the symptom of tingling. No significant differences were recorded in the level of expression of Nav1.9 between patients with or without pain. CONCLUSIONS: These results demonstrate that Nav1.8 and Nav1.9 are present in human lingual nerve neuromas, with significant correlations between the level of expression of Nav1.8 and symptoms of pain. These data provide further evidence that changes in expression of Nav1.8 are important in the development and/or maintenance of nerve injury-induced pain, and suggest that Nav1.8 may be a potential therapeutic target.


Assuntos
Regulação Neoplásica da Expressão Gênica , Nervo Lingual/metabolismo , Nervo Lingual/patologia , Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Neuralgia/metabolismo , Neuroma/metabolismo , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Canal de Sódio Disparado por Voltagem NAV1.9/metabolismo , Neuroma/fisiopatologia
14.
Ann Neurol ; 71(2): 186-94, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22367990

RESUMO

OBJECTIVE: Cerebellar dysfunction in multiple sclerosis (MS) contributes significantly to disability, is relatively refractory to symptomatic therapy, and often progresses despite treatment with disease-modifying agents. We previously observed that sodium channel Nav1.8, whose expression is normally restricted to the peripheral nervous system, is present in cerebellar Purkinje neurons in a mouse model of MS (experimental autoimmune encephalomyelitis [EAE]) and in humans with MS. Here, we tested the hypothesis that upregulation of Nav1.8 in cerebellum in MS and EAE has functional consequences contributing to symptom burden. METHODS: Electrophysiology and behavioral assessment were performed in a new transgenic mouse model overexpressing Nav1.8 in Purkinje neurons. We also measured EAE symptom progression in mice lacking Nav1.8 compared to wild-type littermates. Finally, we administered the Nav1.8-selective blocker A803467 in the context of previously established EAE to determine reversibility of MS-like deficits. RESULTS: We report that, in the context of an otherwise healthy nervous system, ectopic expression of Nav1.8 in Purkinje neurons alters their electrophysiological properties, and disrupts coordinated motor behaviors. Additionally, we show that Nav1.8 expression contributes to symptom development in EAE. Finally, we demonstrate that abnormal patterns of Purkinje neuron firing and MS-like deficits in EAE can be partially reversed by pharmacotherapy using a Nav1.8-selective blocker. INTERPRETATION: Our results add to the evidence that a channelopathy contributes to cerebellar dysfunction in MS. Our data suggest that Nav1.8-specific blockers, when available for humans, merit study in MS.


Assuntos
Doenças Cerebelares/fisiopatologia , Canalopatias/fisiopatologia , Encefalomielite Autoimune Experimental/fisiopatologia , Esclerose Múltipla/fisiopatologia , Compostos de Anilina/uso terapêutico , Animais , Doenças Cerebelares/genética , Cerebelo/citologia , Cerebelo/metabolismo , Cerebelo/patologia , Canalopatias/genética , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/tratamento farmacológico , Furanos/uso terapêutico , Camundongos , Camundongos Transgênicos , Esclerose Múltipla/genética , Canal de Sódio Disparado por Voltagem NAV1.8 , Células de Purkinje/patologia , Células de Purkinje/fisiologia , Bloqueadores dos Canais de Sódio/uso terapêutico , Canais de Sódio/biossíntese , Canais de Sódio/genética , Canais de Sódio/metabolismo , Regulação para Cima/genética
15.
Mult Scler ; 19(5): 532-42, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22951351

RESUMO

BACKGROUND: Macrophages are dynamic participants in destruction of white matter in active multiple sclerosis (MS) plaques. Regulation of phagocytosis and myelin degradation along endosomal pathways in macrophages is highly-orchestrated and critically-dependent upon acidification of endosomal lumena. Evidence from in vitro studies with macrophages and THP-1 cells suggests that sodium channel Nav1.5 is present in the limiting membrane of maturing endosomes where it plays a prominent role in the accumulation of protons. However, a contribution of the Nav1.5 channel to macrophage-mediated events in vivo has not been demonstrated. METHOD: We examined macrophages within active MS lesions by immunohistochemistry to determine whether Nav1.5 is expressed in these cells in situ and, if expressed, whether it is localized to specific compartments along the endocytic pathway. RESULTS: Our results demonstrate that Nav1.5 is expressed within macrophages in active MS lesions, and that it is preferentially expressed in late endosomes and phagolysosomes (Rab7(+), LAMP-1(+)), and sparsely expressed in early (EEA-1(+)) endosomes. Triple-immunolabeling studies showed localization of Nav1.5 within Rab7(+) endosomes containing proteolipid protein, a myelin marker, in macrophages within active MS plaques. CONCLUSIONS: These observations support the suggestion that Nav1.5 contributes to the phagocytic pathway of myelin degradation in macrophages in vivo within MS lesions.


Assuntos
Encéfalo/metabolismo , Macrófagos/metabolismo , Esclerose Múltipla Crônica Progressiva/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Adulto , Idoso , Estudos de Casos e Controles , Endocitose/fisiologia , Endossomos/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla Crônica Progressiva/patologia , Esclerose Múltipla Crônica Progressiva/fisiopatologia , Fagocitose/fisiologia , Fagossomos/metabolismo
16.
Mol Pain ; 8: 82, 2012 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-23134641

RESUMO

BACKGROUND: Sodium channel Nav1.7 has emerged as a target of considerable interest in pain research, since loss-of-function mutations in SCN9A, the gene that encodes Nav1.7, are associated with a syndrome of congenital insensitivity to pain, gain-of-function mutations are linked to the debiliting chronic pain conditions erythromelalgia and paroxysmal extreme pain disorder, and upregulated expression of Nav1.7 accompanies pain in diabetes and inflammation. Since Nav1.7 has been implicated as playing a critical role in pain pathways, we examined by immunocytochemical methods the expression and distribution of Nav1.7 in rat dorsal root ganglia neurons, from peripheral terminals in the skin to central terminals in the spinal cord dorsal horn. RESULTS: Nav1.7 is robustly expressed within the somata of peptidergic and non-peptidergic DRG neurons, and along the peripherally- and centrally-directed C-fibers of these cells. Nav1.7 is also expressed at nodes of Ranvier in a subpopulation of Aδ-fibers within sciatic nerve and dorsal root. The peripheral terminals of DRG neurons within skin, intraepidermal nerve fibers (IENF), exhibit robust Nav1.7 immunolabeling. The central projections of DRG neurons in the superficial lamina of spinal cord dorsal horn also display Nav1.7 immunoreactivity which extends to presynaptic terminals. CONCLUSIONS: The expression of Nav1.7 in DRG neurons extends from peripheral terminals in the skin to preterminal central branches and terminals in the dorsal horn. These data support a major contribution for Nav1.7 in pain pathways, including action potential electrogenesis, conduction along axonal trunks and depolarization/invasion of presynaptic axons. The findings presented here may be important for pharmaceutical development, where target engagement in the right compartment is essential.


Assuntos
Gânglios Espinais/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Neurônios/metabolismo , Dor/metabolismo , Pele/inervação , Animais , Imuno-Histoquímica , Masculino , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Terminações Pré-Sinápticas/metabolismo , Ratos , Ratos Sprague-Dawley , Medula Espinal/citologia
18.
J Neurosci ; 30(5): 1637-47, 2010 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-20130174

RESUMO

Na(v)1.7 sodium channels can amplify weak stimuli in neurons and act as threshold channels for firing action potentials. Neurotrophic factors and pro-nociceptive cytokines that are released during development and under pathological conditions activate mitogen-activated protein kinases (MAPKs). Previous studies have shown that MAPKs can transduce developmental or pathological signals by regulating transcription factors that initiate a gene expression response, a long-term effect, and directly modulate neuronal ion channels including sodium channels, thus acutely regulating dorsal root ganglion (DRG) neuron excitability. For example, neurotrophic growth factor activates (phosphorylates) ERK1/2 MAPK (pERK1/2) in DRG neurons, an effect that has been implicated in injury-induced hyperalgesia. However, the acute effects of pERK1/2 on sodium channels are not known. We have shown previously that activated p38 MAPK (pp38) directly phosphorylates Na(v)1.6 and Na(v)1.8 sodium channels and regulates their current densities without altering their gating properties. We now report that acute inhibition of pERK1/2 regulates resting membrane potential and firing properties of DRG neurons. We also show that pERK1 phosphorylates specific residues within L1 of Na(v)1.7, inhibition of pERK1/2 causes a depolarizing shift of activation and fast inactivation of Na(v)1.7 without altering current density, and mutation of these L1 phosphoacceptor sites abrogates the effect of pERK1/2 on this channel. Together, these data are consistent with direct phosphorylation and modulation of Na(v)1.7 by pERK1/2, which unlike the modulation of Na(v)1.6 and Na(v)1.8 by pp38, regulates gating properties of this channel but not its current density and contributes to the effects of MAPKs on DRG neuron excitability.


Assuntos
Ativação do Canal Iônico/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Canais de Sódio/metabolismo , Animais , Células Cultivadas , Gânglios Espinais/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Fator de Crescimento Neural/metabolismo , Neurônios/metabolismo , Técnicas de Patch-Clamp , Fosforilação , Ratos , Ratos Sprague-Dawley
19.
Mol Pain ; 7: 32, 2011 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-21569247

RESUMO

BACKGROUND: Voltage-gated sodium channel Nav1.7 is preferentially expressed in dorsal root ganglion (DRG) and sympathetic neurons within the peripheral nervous system. Homozygous or compound heterozygous loss-of-function mutations in SCN9A, the gene which encodes Nav1.7, cause congenital insensitivity to pain (CIP) accompanied by anosmia. Global knock-out of Nav1.7 in mice is neonatal lethal reportedly from starvation, suggesting anosmia. These findings led us to hypothesize that Nav1.7 is the main sodium channel in the peripheral olfactory sensory neurons (OSN, also known as olfactory receptor neurons). METHODS: We used multiplex PCR-restriction enzyme polymorphism, in situ hybridization and immunohistochemistry to determine the identity of sodium channels in rodent OSNs. RESULTS: We show here that Nav1.7 is the predominant sodium channel transcript, with low abundance of other sodium channel transcripts, in olfactory epithelium from rat and mouse. Our in situ hybridization data show that Nav1.7 transcripts are present in rat OSNs. Immunostaining of Nav1.7 and Nav1.6 channels in rat shows a complementary accumulation pattern with Nav1.7 in peripheral presynaptic OSN axons, and Nav1.6 primarily in postsynaptic cells and their dendrites in the glomeruli of the olfactory bulb within the central nervous system. CONCLUSIONS: Our data show that Nav1.7 is the dominant sodium channel in rat and mouse OSN, and may explain anosmia in Nav1.7 null mouse and patients with Nav1.7-related CIP.


Assuntos
Mucosa Olfatória/inervação , Mucosa Olfatória/metabolismo , Neurônios Receptores Olfatórios/metabolismo , Canais de Sódio/metabolismo , Animais , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Regulação da Expressão Gênica , Hibridização In Situ , Ativação do Canal Iônico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Canal de Sódio Disparado por Voltagem NAV1.6 , Canal de Sódio Disparado por Voltagem NAV1.7 , Mucosa Olfatória/citologia , Neurônios Receptores Olfatórios/citologia , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Canais de Sódio/genética
20.
Brain ; 133(Pt 3): 835-46, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20147455

RESUMO

Astrocytes are prominent participants in the response of the central nervous system to injury, including neuroinflammatory insults. Rodent astrocytes in vitro have been shown to express voltage-gated sodium channels in a dynamic manner, with a switch in expression of tetrodotoxin-sensitive to tetrodotoxin-resistant channels in reactive astrocytes. However, the expression of sodium channels in human astrocytes has not been studied, and it is not known whether there are changes in the expression of sodium channels in reactive astrocytes of the human central nervous system. Here, we demonstrate a focal and robust upregulation of sodium channel Nav1.5 in reactive astrocytes at the borders of, and within, active and chronic multiple sclerosis lesions. Nav1.5 was only detectable at very low levels in astrocytes within multiple sclerosis macroscopically normal-appearing white matter or in normal control brain. Nav1.1, Nav1.2, Nav1.3 and Nav1.6 showed little or no expression in astrocytes within normal control tissue and limited upregulation in active multiple sclerosis lesions. Nav1.5 was also expressed at high levels in astrocytes in tissue surrounding new and old cerebrovascular accidents and brain tumours. These results demonstrate the expression of Nav1.5 in human astrocytes and show that Nav1.5 expression is dynamic in these cells. Our observations suggest that the upregulated expression of Nav1.5 in astrocytes may provide a compensatory mechanism, which supports sodium/potassium pump-dependent ionic homoeostasis in areas of central nervous system injury.


Assuntos
Astrócitos/metabolismo , Encéfalo/metabolismo , Esclerose Múltipla Crônica Progressiva/metabolismo , Proteínas Musculares/metabolismo , Fibras Nervosas Mielinizadas/metabolismo , Canais de Sódio/metabolismo , Doença Aguda , Adulto , Idoso , Neoplasias Encefálicas/metabolismo , Doença Crônica , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Canal de Sódio Disparado por Voltagem NAV1.1 , Canal de Sódio Disparado por Voltagem NAV1.2 , Canal de Sódio Disparado por Voltagem NAV1.3 , Canal de Sódio Disparado por Voltagem NAV1.5 , Canal de Sódio Disparado por Voltagem NAV1.6 , Proteínas do Tecido Nervoso/metabolismo , Acidente Vascular Cerebral/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA