Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neurosci ; 42(40): 7513-7529, 2022 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-36658457

RESUMEN

Migraine is believed to be initiated by neuronal activity in the CNS, that triggers excitation of nociceptive trigeminal ganglion (TG) nerve fibers innervating the meninges and thus causes a unilateral throbbing headache. Drugs that precipitate or potentiate migraine are known to elevate intracellular levels of the cyclic nucleotides cAMP or cGMP, while anti-migraine treatments couple to signaling pathways that reduce cAMP or cGMP, suggesting an involvement of these cyclic nucleotides in migraine. Members of the HCN ion channel family are activated by direct binding of cAMP or cGMP, suggesting in turn that a member of this family may be a critical trigger of migraine. Here, we show that pharmacological block or targeted genetic deletion of HCN2 abolishes migraine-like pain in three rodent migraine models (in both sexes). Induction of migraine-like pain in these models triggered expression of the protein C-FOS, a marker of neuronal activity, in neurons of the trigeminocervical complex (TCC), where TG neurons terminate, and C-FOS expression was reversed by peripheral HCN2 inhibition. HCN2 block in vivo inhibited both evoked and spontaneous neuronal activity in nociceptive TG neurons. The NO donor glyceryl trinitrate (GTN) caused an increase in cGMP in the TG in vivo Exposing isolated TG neurons to GTN caused a rightward shift in the voltage dependence of HCN currents and thus increased neuronal excitability. This work identifies HCN2 as a novel target for the development of migraine treatments.SIGNIFICANCE STATEMENT Migraine is believed to be initiated by localized excitability of neurons within the CNS, but the most disturbing symptom, the characteristic throbbing migraine headache pain, is widely agreed to be caused by activity in afferent pain-sensitive (nociceptive) nerve fibers of the trigeminal nerve. Using a variety of preclinical models of migraine, we identify the HCN2 ion channel as the molecular source of trigeminal hyperexcitability in migraine and we show that pharmacological or genetic inhibition of HCN2 can relieve migraine-like pain symptoms. The work highlights the HCN2 ion channel as a potential pharmacological target for the development of novel analgesics effective in migraine.


Asunto(s)
Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Trastornos Migrañosos , Animales , Masculino , Femenino , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Roedores , Dolor/metabolismo , Trastornos Migrañosos/genética , Cefalea , Nucleótidos Cíclicos
2.
Int J Mol Sci ; 20(18)2019 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-31540178

RESUMEN

Ion channels contribute fundamental properties to cell membranes. Although highly diverse in conductivity, structure, location, and function, many of them can be regulated by common mechanisms, such as voltage or (de-)phosphorylation. Primarily considering ion channels involved in the nociceptive system, this review covers more novel and less known features. Accordingly, we outline noncanonical operation of voltage-gated sodium, potassium, transient receptor potential (TRP), and hyperpolarization-activated cyclic nucleotide (HCN)-gated channels. Noncanonical features discussed include properties as a memory for prior voltage and chemical exposure, alternative ion conduction pathways, cluster formation, and silent subunits. Complementary to this main focus, the intention is also to transfer knowledge between fields, which become inevitably more separate due to their size.


Asunto(s)
Canales Iónicos/metabolismo , Dolor/etiología , Dolor/metabolismo , Animales , Susceptibilidad a Enfermedades , Descubrimiento de Drogas , Humanos , Activación del Canal Iónico , Canales Iónicos/química , Canales Iónicos/genética , Dolor/tratamiento farmacológico
3.
J Physiol ; 597(17): 4661-4675, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31290157

RESUMEN

KEY POINTS: HCN ion channels conducting the Ih current control the frequency of firing in peripheral sensory neurons signalling pain. Previous studies have demonstrated a major role for the HCN2 subunit in chronic pain but the potential involvement of HCN3 in pain has not been investigated. HCN3 was found to be widely expressed in all classes of sensory neurons (small, medium, large) where it contributes to Ih . HCN3 deletion increased the firing rate of medium but not small, sensory neurons. Pain sensitivity both acutely and following neuropathic injury was largely unaffected by HCN3 deletion, with the exception of a small decrease of mechanical hyperalgesia in response to a pinprick. We conclude that HCN3 plays little role in either acute or chronic pain sensation. ABSTRACT: HCN ion channels govern the firing rate of action potentials in the pacemaker region of the heart and in pain-sensitive (nociceptive) nerve fibres. Intracellular cAMP promotes activation of the HCN4 and HCN2 isoforms, whereas HCN1 and HCN3 are relatively insensitive to cAMP. HCN2 modulates action potential firing rate in nociceptive neurons and plays a critical role in all modes of inflammatory and neuropathic pain, although the role of HCN3 in nociceptive excitability and pain is less studied. Using antibody staining, we found that HCN3 is expressed in all classes of somatosensory neurons. In small nociceptive neurons, genetic deletion of HCN2 abolished the voltage shift of the Ih current carried by HCN isoforms following cAMP elevation, whereas the voltage shift was retained following deletion of HCN3, consistent with the sensitivity of HCN2 but not HCN3 to cAMP. Deletion of HCN3 had little effect on the evoked firing frequency in small neurons but enhanced the firing of medium-sized neurons, showing that HCN3 makes a significant contribution to the input resistance only in medium-sized neurons. Genetic deletion of HCN3 had no effect on acute thresholds to heat or mechanical stimuli in vivo and did not affect inflammatory pain measured with the formalin test. Nerve-injured HCN3 knockout mice exhibited similar levels of mechanical allodynia and thermal hyperalgesia to wild-type mice but reduced mechanical hyperalgesia in response to a pinprick. These results show that HCN3 makes some contribution to excitability, particularly in medium-sized neurons, although it has no major influence on acute or neuropathic pain processing.


Asunto(s)
Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Potenciales de la Membrana/fisiología , Neuralgia/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , AMP Cíclico/metabolismo , Femenino , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fibras Nerviosas/metabolismo , Fibras Nerviosas/fisiología , Neuralgia/fisiopatología , Nociceptores/metabolismo , Nociceptores/fisiología , Dimensión del Dolor/métodos , Umbral del Dolor/fisiología
4.
Pain ; 159(8): 1641-1651, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29697531

RESUMEN

Voltage-gated potassium (Kv) channels are increasingly recognised as key regulators of nociceptive excitability. Kcns1 is one of the first potassium channels to be associated with neuronal hyperexcitability and mechanical sensitivity in the rat, as well as pain intensity and risk of developing chronic pain in humans. Here, we show that in mice, Kcns1 is predominantly expressed in the cell body and axons of myelinated sensory neurons positive for neurofilament-200, including Aδ-fiber nociceptors and low-threshold Aß mechanoreceptors. In the spinal cord, Kcns1 was detected in laminae III to V of the dorsal horn where most sensory A fibers terminate, as well as large motoneurons of the ventral horn. To investigate Kcns1 function specifically in the periphery, we generated transgenic mice in which the gene is deleted in all sensory neurons but retained in the central nervous system. Kcns1 ablation resulted in a modest increase in basal mechanical pain, with no change in thermal pain processing. After neuropathic injury, Kcns1 KO mice exhibited exaggerated mechanical pain responses and hypersensitivity to both noxious and innocuous cold, consistent with increased A-fiber activity. Interestingly, Kcns1 deletion also improved locomotor performance in the rotarod test, indicative of augmented proprioceptive signalling. Our results suggest that restoring Kcns1 function in the periphery may be of some use in ameliorating mechanical and cold pain in chronic states.


Asunto(s)
Neuralgia/metabolismo , Umbral del Dolor/fisiología , Células del Asta Posterior/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Animales , Ratones , Ratones Noqueados , Destreza Motora/fisiología , Neuralgia/genética , Estimulación Física , Canales de Potasio con Entrada de Voltaje/genética , Propiocepción/fisiología
5.
Sci Transl Med ; 9(409): eaam6072, 2017 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-28954930

RESUMEN

Diabetic patients frequently suffer from continuous pain that is poorly treated by currently available analgesics. We used mouse models of type 1 and type 2 diabetes to investigate a possible role for the hyperpolarization-activated cyclic nucleotide-gated 2 (HCN2) ion channels as drivers of diabetic pain. Blocking or genetically deleting HCN2 channels in small nociceptive neurons suppressed diabetes-associated mechanical allodynia and prevented neuronal activation of second-order neurons in the spinal cord in mice. In addition, we found that intracellular cyclic adenosine monophosphate (cAMP), a positive HCN2 modulator, is increased in somatosensory neurons in an animal model of painful diabetes. We propose that the increased intracellular cAMP drives diabetes-associated pain by facilitating HCN2 activation and consequently promoting repetitive firing in primary nociceptive nerve fibers. Our results suggest that HCN2 may be an analgesic target in the treatment of painful diabetic neuropathy.


Asunto(s)
Neuropatías Diabéticas/complicaciones , Neuropatías Diabéticas/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Dolor/complicaciones , Dolor/metabolismo , Canales de Potasio/metabolismo , Analgésicos , Animales , Benzazepinas/farmacología , Benzazepinas/uso terapéutico , AMP Cíclico/metabolismo , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Neuropatías Diabéticas/tratamiento farmacológico , Neuropatías Diabéticas/patología , Modelos Animales de Enfermedad , Eliminación de Gen , Hiperalgesia/complicaciones , Hiperalgesia/tratamiento farmacológico , Ivabradina , Nocicepción , Dolor/tratamiento farmacológico , Dolor/patología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Células Receptoras Sensoriales/metabolismo , Piel/inervación , Asta Dorsal de la Médula Espinal/metabolismo , Estreptozocina
6.
Biochem J ; 473(18): 2717-36, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27621481

RESUMEN

Nociception - the ability to detect painful stimuli - is an invaluable sense that warns against present or imminent damage. In patients with chronic pain, however, this warning signal persists in the absence of any genuine threat and affects all aspects of everyday life. Neuropathic pain, a form of chronic pain caused by damage to sensory nerves themselves, is dishearteningly refractory to drugs that may work in other types of pain and is a major unmet medical need begging for novel analgesics. Hyperpolarisation-activated cyclic nucleotide (HCN)-modulated ion channels are best known for their fundamental pacemaker role in the heart; here, we review data demonstrating that the HCN2 isoform acts in an analogous way as a 'pacemaker for pain', in that its activity in nociceptive neurons is critical for the maintenance of electrical activity and for the sensation of chronic pain in pathological pain states. Pharmacological block or genetic deletion of HCN2 in sensory neurons provides robust pain relief in a variety of animal models of inflammatory and neuropathic pain, without any effect on normal sensation of acute pain. We discuss the implications of these findings for our understanding of neuropathic pain pathogenesis, and we outline possible future opportunities for the development of efficacious and safe pharmacotherapies in a range of chronic pain syndromes.


Asunto(s)
Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Neuralgia/metabolismo , Humanos
7.
Pain ; 157 Suppl 1: S7-S14, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26785158

RESUMEN

A workshop of the 2015 International Neuropathic Pain Congress was focused on potassium channels to propose emerging ideas on the role of these channels on pain modulation and to determine whether they can become relevant targets for designing novel analgesic compounds. Two kinds of potassium channels were particularly evoked: selected subunits of the voltage-gated potassium (Kv) and of the K2P channel families. In this review, the role of the former is described with a focus first on silent subunits as modulators of Kv and second on the Kv7 subunits. The physiological, pathophysiological, and pharmacological involvement of the K2P in pain modulation is then described. Throughout this review, the role of potassium channels in pain is obvious, which renders them potential targets for innovative analgesics with peripheral and/or central action depending on the channel. Clearly, some preliminary results obtained with known or novel potassium channel openers suggest that they might represent a novel class of analgesics in neuropathic pain or other pathological contexts.


Asunto(s)
Neuralgia/metabolismo , Canales de Potasio/metabolismo , Animales , Humanos , Canales de Potasio/genética
8.
Curr Opin Support Palliat Care ; 9(2): 147-54, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25872119

RESUMEN

PURPOSE OF REVIEW: Poor management of chronic pain remains a significant cause of misery with huge socioeconomic costs. Accumulating research in potassium (K+) channel physiology has uncovered several promising leads for the development of novel analgesics. RECENT FINDINGS: We now recognize that certain K+ channel subunits are directly gated to pain-relevant stimuli (Kv1.1, K2P) whereas others are specifically modulated by inflammatory processes (Kv7, BKCA, K2P). Genetic analyses illustrate that K+ channel gene variation can predict pain sensitivity (KCNS1, GIRKs), risk for persistent pain (KCNS1, GIRKs, TRESK) and analgesic effectiveness (GIRK2). Importantly, preclinical studies confirm that K+ channel dysfunction can be a pain trigger in traumatic neuropathies (Kv9.1/Kv2.1, Kv7, Kv1.2) and migraine (TRESK). Finally, emerging data suggest that even pain in diabetes, bone cancer and autoimmune neuropathies may have K+ channel dysfunction constituents. SUMMARY: There is a long-sought need for superior pharmacotherapy of pain syndromes. Although universal enhancement of K+ channel function in the periphery can decrease nociceptive excitability irrespective of the underlying cause, a more refined targeting of subunits with dominant nociceptive roles could yield highly efficacious treatments with fewer side-effects. The ongoing characterization of molecular interactions linking K+ channel dysfunction to pain is instrumental for identifying candidates with the most therapeutic potential.


Asunto(s)
Analgésicos/farmacología , Dolor Crónico/tratamiento farmacológico , Diseño de Fármacos , Neuralgia/tratamiento farmacológico , Canales de Potasio/uso terapéutico , Dolor Crónico/etiología , Neuropatías Diabéticas/tratamiento farmacológico , Neuropatías Diabéticas/fisiopatología , Humanos , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Inflamación/fisiopatología , Trastornos Migrañosos/tratamiento farmacológico , Trastornos Migrañosos/fisiopatología , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Neoplasias/fisiopatología , Neuralgia/etiología , Neuralgia/fisiopatología , Dolor Postoperatorio/tratamiento farmacológico , Dolor Postoperatorio/fisiopatología , Canales de Potasio/farmacología
10.
Pain ; 155(9): 1708-1719, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24861581

RESUMEN

Previous studies have shown that hyperpolarisation-activated cyclic nucleotide-gated (HCN)-2 ion channels regulate the firing frequency of nociceptive sensory neurons and thus play a central role in both inflammatory and neuropathic pain conditions. Here we use ivabradine, a clinically approved anti-anginal agent that blocks all HCN channel isoforms approximately equally, to investigate the effect on inflammatory and neuropathic pain of HCN ion channel block. We show that ivabradine does not have major off-target effects on a sample group of Na, Ca, and K ion channels, and that it is peripherally restricted because it is a substrate for the P-glycoprotein (PgP) multidrug transporter that is expressed in the blood-brain barrier. Its effects are therefore likely to be due to an action on HCN ion channels in peripheral sensory neurons. Using patch clamp electrophysiology, we found that ivabradine was a use-dependent blocker of native HCN channels expressed in small sensory neurons. Ivabradine suppressed the action potential firing that is induced in nociceptive neurons by elevation of intracellular cAMP. In the formalin model of inflammatory pain, ivabradine reduced pain behaviour only in the second (inflammatory) phase. In nerve injury and chemotherapy models of neuropathic pain, we observed rapid and effective analgesia as effective as that with gabapentin. We conclude that both inflammatory and neuropathic pain are rapidly inhibited by blocking HCN-dependent repetitive firing in peripheral nociceptive neurons.


Asunto(s)
Benzazepinas/uso terapéutico , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/antagonistas & inhibidores , Inflamación/tratamiento farmacológico , Neuralgia/tratamiento farmacológico , Animales , Benzazepinas/farmacología , Células Cultivadas , Femenino , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Inflamación/metabolismo , Ivabradina , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Neuralgia/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Técnicas de Placa-Clamp
11.
Trends Neurosci ; 37(3): 146-58, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24461875

RESUMEN

Chronic pain is associated with abnormal excitability of the somatosensory system and remains poorly treated in the clinic. Potassium (K⁺) channels are crucial determinants of neuronal activity throughout the nervous system. Opening of these channels facilitates a hyperpolarizing K⁺ efflux across the plasma membrane that counteracts inward ion conductance and therefore limits neuronal excitability. Accumulating research has highlighted a prominent involvement of K⁺ channels in nociceptive processing, particularly in determining peripheral hyperexcitability. We review salient findings from expression, pharmacological, and genetic studies that have untangled a hitherto undervalued contribution of K⁺ channels in maladaptive pain signaling. These emerging data provide a framework to explain enigmatic pain syndromes and to design novel pharmacological treatments for these debilitating states.


Asunto(s)
Analgésicos/farmacología , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/metabolismo , Terapia Molecular Dirigida/métodos , Canales de Potasio/metabolismo , Animales , Dolor Crónico/fisiopatología , Diseño de Fármacos , Humanos
12.
Exp Neurol ; 251: 115-26, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24252178

RESUMEN

Peripheral nerve injuries caused by trauma are associated with increased sensory neuron excitability and debilitating chronic pain symptoms. Axotomy-induced alterations in the function of ion channels are thought to largely underlie the pathophysiology of these phenotypes. Here, we characterise the mRNA distribution of Kv2 family members in rat dorsal root ganglia (DRG) and describe a link between Kv2 function and modulation of sensory neuron excitability. Kv2.1 and Kv2.2 were amply expressed in cells of all sizes, being particularly abundant in medium-large neurons also immunoreactive for neurofilament-200. Peripheral axotomy led to a rapid, robust and long-lasting transcriptional Kv2 downregulation in the DRG, correlated with the onset of mechanical and thermal hypersensitivity. The consequences of Kv2 loss-of-function were subsequently investigated in myelinated neurons using intracellular recordings on ex vivo DRG preparations. In naïve neurons, pharmacological Kv2.1/Kv2.2 inhibition by stromatoxin-1 (ScTx) resulted in shortening of action potential (AP) after-hyperpolarization (AHP). In contrast, ScTx application on axotomized neurons did not alter AHP duration, consistent with the injury-induced Kv2 downregulation. In accordance with a shortened AHP, ScTx treatment also reduced the refractory period and improved AP conduction to the cell soma during high frequency stimulation. These results suggest that Kv2 downregulation following traumatic nerve lesion facilitates greater fidelity of repetitive firing during prolonged input and thus normal Kv2 function is postulated to limit neuronal excitability. In summary, we have profiled Kv2 expression in sensory neurons and provide evidence for the contribution of Kv2 dysfunction in the generation of hyperexcitable phenotypes encountered in chronic pain states.


Asunto(s)
Enfermedades del Sistema Nervioso Periférico/metabolismo , Células Receptoras Sensoriales/metabolismo , Canales de Potasio Shab/metabolismo , Animales , Axotomía/métodos , Péptido Relacionado con Gen de Calcitonina/metabolismo , Ganglios Espinales/citología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Proteínas del Tejido Nervioso/metabolismo , Dolor/etiología , Dolor/metabolismo , Dimensión del Dolor , Enfermedades del Sistema Nervioso Periférico/complicaciones , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Células Receptoras Sensoriales/fisiología , Canales de Potasio Shab/genética
13.
PLoS One ; 8(11): e80722, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24278311

RESUMEN

Pathological changes in axonal function are integral features of many neurological disorders, yet our knowledge of the molecular basis of axonal dysfunction remains limited. Microfluidic chambers (MFCs) can provide unique insight into the axonal compartment independent of the soma. Here we demonstrate how an MFC based cell culture system can be readily adapted for the study of axonal function in vitro. We illustrate the ease and versatility to assay electrogenesis and conduction of action potentials (APs) in naïve, damaged or sensitized DRG axons using calcium imaging at the soma for pharmacological screening or patch-clamp electrophysiology for detailed biophysical characterisation. To demonstrate the adaptability of the system, we report by way of example functional changes in nociceptor axons following sensitization by neurotrophins and axotomy in vitro. We show that NGF can locally sensitize axonal responses to capsaicin, independent of the soma. Axotomizing neurons in MFC results in a significant increase in the proportion of neurons that respond to axonal stimulation, and interestingly leads to accumulation of Nav1.8 channels in regenerating axons. Axotomy also augmented AP amplitude following axotomy and altered activation thresholds in a subpopulation of regenerating axons. We further show how the system can readily be used to study modulation of axonal function by non-neuronal cells such as keratinocytes. Hence we describe a novel in vitro platform for the study of axonal function and a surrogate model for nerve injury and sensitization.


Asunto(s)
Axones/fisiología , Microfluídica/métodos , Nocicepción , Potenciales de Acción/efectos de los fármacos , Animales , Axones/efectos de los fármacos , Axotomía , Bioensayo , Calcio/metabolismo , Capsaicina/farmacología , Comunicación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Estimulación Eléctrica , Femenino , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/lesiones , Ganglios Espinales/patología , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Microfluídica/instrumentación , Modelos Biológicos , Factor de Crecimiento Nervioso/farmacología , Nocicepción/efectos de los fármacos , Técnicas de Placa-Clamp , Ratas Wistar , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/patología , Bloqueadores de los Canales de Sodio/farmacología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Canales Catiónicos TRPV/metabolismo
14.
J Neurosci ; 32(48): 17502-13, 2012 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-23197740

RESUMEN

Chronic neuropathic pain affects millions of individuals worldwide, is typically long-lasting, and remains poorly treated with existing therapies. Neuropathic pain arising from peripheral nerve lesions is known to be dependent on the emergence of spontaneous and evoked hyperexcitability in damaged nerves. Here, we report that the potassium channel subunit Kv9.1 is expressed in myelinated sensory neurons, but is absent from small unmyelinated neurons. Kv9.1 expression was strongly and rapidly downregulated following axotomy, with a time course that matches the development of spontaneous activity and pain hypersensitivity in animal models. Interestingly, siRNA-mediated knock-down of Kv9.1 in naive rats led to neuropathic pain behaviors. Diminished Kv9.1 function also augmented myelinated sensory neuron excitability, manifested as spontaneous firing, hyper-responsiveness to stimulation, and persistent after-discharge. Intracellular recordings from ex vivo dorsal root ganglion preparations revealed that Kv9.1 knock-down was linked to lowered firing thresholds and increased firing rates under physiologically relevant conditions of extracellular potassium accumulation during prolonged activity. Similar neurophysiological changes were detected in animals subjected to traumatic nerve injury and provide an explanation for neuropathic pain symptoms, including poorly understood conditions such as hyperpathia and paresthesias. In summary, our results demonstrate that Kv9.1 dysfunction leads to spontaneous and evoked neuronal hyperexcitability in myelinated fibers, coupled with development of neuropathic pain behaviors.


Asunto(s)
Regulación hacia Abajo , Neuralgia/metabolismo , Traumatismos de los Nervios Periféricos/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Axotomía , Conducta Animal/fisiología , Hiperalgesia/etiología , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Masculino , Fibras Nerviosas Mielínicas/metabolismo , Neuralgia/etiología , Neuralgia/fisiopatología , Dimensión del Dolor , Traumatismos de los Nervios Periféricos/complicaciones , Traumatismos de los Nervios Periféricos/fisiopatología , Canales de Potasio con Entrada de Voltaje/genética , ARN Interferente Pequeño , Ratas , Ratas Wistar
15.
J Neurosci ; 31(9): 3225-33, 2011 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-21368034

RESUMEN

Neuregulin-1 (NRG1) plays a crucial role in axoglial signaling during the development of the peripheral nervous system, but its importance in adulthood after peripheral nerve injury remains unclear. We used single-neuron labeling with inducible Cre-mediated knock-out animals, which enabled visualization of a subset of adult myelinated sensory and motoneurons neurons in which Nrg1 was inducibly mutated by tamoxifen treatment. In uninjured mice, NRG1-deficient axons and the associated myelin sheath were normal, and the neuromuscular junction demonstrated normal apposition of presynaptic and postsynaptic components. After sciatic nerve crush, NRG1 ablation resulted in severe defects in remyelination: axons were either hypomyelinated or had no myelin sheath. NRG1-deficient axons were also found to regenerate at a slower rate. After nerve injury, the neuromuscular junction was reinnervated, but excess terminal sprouting was observed. Juxtacrine Neuregulin-1 signaling is therefore dispensable for maintenance of the myelin sheath in adult animals but has a key role in reparative processes after nerve injury.


Asunto(s)
Envejecimiento/fisiología , Axones/fisiología , Vaina de Mielina/fisiología , Fibras Nerviosas Mielínicas/fisiología , Regeneración Nerviosa/fisiología , Neurregulina-1/fisiología , Neuropatía Ciática/fisiopatología , Envejecimiento/genética , Animales , Axones/metabolismo , Axones/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fibras Nerviosas Mielínicas/patología , Regeneración Nerviosa/genética , Neurregulina-1/deficiencia , Neurregulina-1/genética , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Neuropatía Ciática/genética , Neuropatía Ciática/metabolismo
16.
J Neurosci ; 30(15): 5437-50, 2010 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-20392965

RESUMEN

A key component in the response of the nervous system to injury is the proliferation and switch to a "proinflammatory" phenotype by microglia (microgliosis). In situations where the blood-brain barrier is intact, microglial numbers increase via the proliferation and chemotaxis of resident microglia; however, there is limited knowledge regarding the factors mediating this response. After peripheral nerve injury, a dorsal horn microgliosis develops, which directly contributes to the development of neuropathic pain. Neuregulin-1 (NRG-1) is a growth and differentiation factor with a well characterized role in neural and cardiac development. Microglia express the NRG1 receptors erbB2, 3, and 4, and NRG1 signaling via the erbB2 receptor stimulated microglial proliferation, chemotaxis, and survival, as well as interleukin-1beta release in vitro. Intrathecal treatment with NRG1 resulted in microglial proliferation within the dorsal horn, and these cells developed an activated morphology. This microglial response was associated with the development of both mechanical and cold pain-related hypersensitivity. Primary afferents express NRG1, and after spinal nerve ligation (SNL) we observed both an increase in NRG1 within the dorsal horn as well as activation of erbB2 specifically within microglia. Blockade of the erbB2 receptor or sequestration of endogenous NRG after SNL reduced the proliferation, the number of microglia with an activated morphology, and the expression of phospho-P38 by microglia. Furthermore, consequent to such changes, the mechanical pain-related hypersensitivity and cold allodynia were reduced. NRG1-erbB signaling therefore represents a novel pathway regulating the injury response of microglia.


Asunto(s)
Gliosis/fisiopatología , Microglía/fisiología , Neurregulina-1/metabolismo , Dolor/fisiopatología , Receptor ErbB-2/metabolismo , Nervios Espinales/lesiones , Animales , Proliferación Celular , Supervivencia Celular/fisiología , Quimiotaxis/fisiología , Gliosis/etiología , Masculino , Neuronas Aferentes/metabolismo , Dolor/etiología , Células del Asta Posterior/patología , Células del Asta Posterior/fisiopatología , Ratas , Ratas Wistar , Receptor ErbB-2/antagonistas & inhibidores , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
J Neurosci ; 29(24): 7667-78, 2009 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-19535578

RESUMEN

Neuregulin-1 has a key role in mediating signaling between axons and Schwann cells during development. A limitation to studying its role in adulthood is the embryonic lethality of global Nrg1 gene deletion. We used the Cre-loxP system to generate transgenic mice in which neuregulin-1 is conditionally ablated in the majority of small-diameter and a proportion of large-diameter sensory neurons that have axons conducting in the C- and Adelta-fiber range, respectively. Sensory neuron-specific neuregulin-1 ablation resulted in abnormally large Remak bundles with axons clustered in "polyaxonal" pockets. The total number of axons in the sural nerve was unchanged, but a greater proportion was unmyelinated. In addition, we observed large-diameter axons that were in a 1:1 relationship with Schwann cells, surrounded by a basal lamina but not myelinated. There was no evidence of DRG or Schwann cell death; the markers of different DRG cell populations and cutaneous innervation were unchanged. These anatomical changes were reflected in a slowing of conduction velocity at the lower end of the A-fiber conduction velocity range and a new population of more rapidly conducting C-fibers that are likely to represent large-diameter axons that have failed to myelinate. Conditional neuregulin-1 ablation resulted in a reduced sensitivity to noxious mechanical stimuli. These findings emphasize the importance of neuregulin-1 in mediating the signaling between axons and both myelinating and nonmyelinating Schwann cells required for normal sensory function. Sensory neuronal survival and axonal maintenance, however, are not dependent on axon-derived neuregulin-1 signaling in adulthood.


Asunto(s)
Axones/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Umbral del Dolor/fisiología , Sensación/fisiología , Células Receptoras Sensoriales/citología , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Animales Recién Nacidos , Axones/ultraestructura , Péptido Relacionado con Gen de Calcitonina/metabolismo , Células Cultivadas , Estimulación Eléctrica , Embrión de Mamíferos , Ganglios Espinales/citología , Regulación de la Expresión Génica/genética , Etiquetado Corte-Fin in Situ/métodos , Indoles , Lectinas/metabolismo , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión/métodos , Canal de Sodio Activado por Voltaje NAV1.8 , Fibras Nerviosas/fisiología , Proteínas del Tejido Nervioso/deficiencia , Conducción Nerviosa/efectos de los fármacos , Conducción Nerviosa/genética , Neurregulina-1 , Proteínas de Neurofilamentos/metabolismo , Neuroglía/fisiología , Dimensión del Dolor/métodos , Estimulación Física/métodos , Tiempo de Reacción/genética , Células de Schwann/metabolismo , Células de Schwann/fisiología , Sensación/genética , Transducción de Señal/genética , Piel/inervación , Canales de Sodio/genética , Nervio Sural/patología , Nervio Sural/ultraestructura
18.
Eur J Pain ; 13(7): 673-81, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18849175

RESUMEN

Loss of function is usually considered the major consequence of spinal cord injury (SCI). However, pain severely compromises the quality of life in nearly 70% of SCI patients. The principal aim of this study was to assess the contribution of Tumor necrosis factor alpha (TNF-alpha) to SCI pain. TNF-alpha blockers have already been successfully used to treat inflammatory disorders but there are few studies on its effect on neuropathic pain, especially following SCI. Following T13 spinal cord hemisection, we examined the effects on mechanical allodynia and microglial activation of immediate and delayed chronic intrathecal treatment with etanercept, a fusion protein blocker of TNF-alpha. Immediate treatment (starting at the time of injury) with etanercept resulted in markedly reduced mechanical allodynia 1, 2, 3 and 4 weeks after SCI. Delayed treatment had no effect. Immediate etanercept treatment also reduced spinal microglial activation assessed by OX-42 immunostaining, a putative marker of activated microglia. To assess whether the effects of etanercept were mediated via decreased microglial activation, we examined the effects of the microglial inhibitor, minocycline which significantly reduced the development of pain behaviours at 1 and 2 weeks after SCI compared to saline treatment. Minocycline also significantly reduced microglial OX-42 expression. Furthermore, minocycline decreased the expression of noxious-stimulation-induced c-Fos, suggesting an effect on evoked neuronal activity. This study demonstrates that TNF-alpha plays an important role in the establishment of neuropathic pain following SCI, seemingly dependent on microglial activation. Pharmacological targeting of TNF-alpha may offer therapeutic opportunities for treating SCI pain.


Asunto(s)
Antibacterianos/uso terapéutico , Inmunoglobulina G/uso terapéutico , Inmunosupresores/uso terapéutico , Minociclina/uso terapéutico , Dolor/tratamiento farmacológico , Dolor/etiología , Receptores del Factor de Necrosis Tumoral/uso terapéutico , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Recuento de Células , Etanercept , Lateralidad Funcional/fisiología , Calor , Inmunohistoquímica , Inyecciones Espinales , Masculino , Microglía/efectos de los fármacos , Microglía/fisiología , Estimulación Física , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Factores de Necrosis Tumoral/fisiología
19.
J Virol ; 81(24): 13578-86, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17928342

RESUMEN

Reactivation of lytic replication from viral latency is a defining property of all herpesviruses. Despite this, the authentic physiological cues for the latent-lytic switch are unclear. Such cues should ensure that viral lytic replication occurs under physiological conditions, predominantly in sites which facilitate transmission to permissive uninfected cells and new susceptible hosts. Kaposi's sarcoma-associated herpesvirus (KSHV) is associated with the B-cell neoplasm primary effusion lymphoma (PEL), in which the virus remains latent. We have previously shown that PEL cells have the gene expression profile and immunophenotype of cycling preplasma cells (plasmablasts). Here, we show that the highly active spliced isoform of plasma cell transcription factor X box binding protein 1 (XBP-1s) is a lytic switch for KSHV. XBP-1s is normally absent in PEL, but the induction of endoplasmic reticulum stress leads to XBP-1s generation, plasma cell-like differentiation, and lytic reactivation of KSHV. XBP-1s binds to and activates the KSHV immediate-early gene ORF50 and synergizes with the ORF50 gene product RTA to induce a full lytic cycle. These data suggest that KSHV remains latent until B-cell terminal differentiation into plasma cells, the transcriptional environment of which provides the physiological "lytic switch" through XBP-1s. This links B-cell terminal differentiation to KSHV lytic reactivation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Herpesvirus Humano 8/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Nucleares/metabolismo , Células Plasmáticas/citología , Regiones Promotoras Genéticas , Transactivadores/metabolismo , Activación Transcripcional , Proteínas Virales/metabolismo , Activación Viral , Animales , Diferenciación Celular , Línea Celular , Chlorocebus aethiops , Proteínas de Unión al ADN/genética , Retículo Endoplásmico/ultraestructura , Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Proteínas Inmediatas-Precoces/genética , Microscopía Confocal , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Transcripción del Factor Regulador X , Transactivadores/genética , Factores de Transcripción , Células Vero , Proteínas Virales/genética , Latencia del Virus , Replicación Viral , Proteína 1 de Unión a la X-Box
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA