Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Neurochem ; 123(4): 589-601, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22891703

RESUMEN

Many extracellular factors sensitize nociceptors. Often they act simultaneously and/or sequentially on nociceptive neurons. We investigated if stimulation of the protein kinase C epsilon (PKCε) signaling pathway influences the signaling of a subsequent sensitizing stimulus. Central in activation of PKCs is their transient translocation to cellular membranes. We found in cultured nociceptive neurons that only a first stimulation of the PKCε signaling pathway resulted in PKCε translocation. We identified a novel inhibitory cascade to branch off upstream of PKCε, but downstream of Epac via IP3-induced calcium release. This signaling branch actively inhibited subsequent translocation and even attenuated ongoing translocation. A second 'sensitizing' stimulus was rerouted from the sensitizing to the inhibitory branch of the signaling cascade. Central for the rerouting was cytoplasmic calcium increase and CaMKII activation. Accordingly, in behavioral experiments, activation of calcium stores switched sensitizing substances into desensitizing substances in a CaMKII-dependent manner. This mechanism was also observed by in vivo C-fiber electrophysiology corroborating the peripheral location of the switch. Thus, we conclude that the net effect of signaling in nociceptors is defined by the context of the individual cell's signaling history.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calcio/metabolismo , Neuronas/metabolismo , Nociceptores/fisiología , Umbral del Dolor/fisiología , Agonistas Adrenérgicos beta/farmacología , Análisis de Varianza , Animales , Células Cultivadas , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Estimulación Eléctrica , Inhibidores Enzimáticos/farmacología , Ganglios Espinales/citología , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/fisiopatología , Inositol 1,4,5-Trifosfato/farmacología , Isoproterenol/farmacología , Masculino , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/fisiología , Neuronas/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Proteína Quinasa C-epsilon/metabolismo , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Rianodina/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Canales Catiónicos TRPV/metabolismo , Tionucleótidos/farmacología , Uridina Trifosfato/farmacología
2.
J Neurochem ; 117(6): 995-1008, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21480900

RESUMEN

Recently, we described estrogen and agonists of the G-protein coupled estrogen receptor GPR30 to induce protein kinase C (PKC)ε-dependent pain sensitization. PKCε phosphorylates the ion channel transient receptor potential, vanilloid subclass I (TRPV1) close to a novel microtubule-TRPV1 binding site. We now modeled the binding of tubulin to the TRPV1 C-terminus. The model suggests PKCε phosphorylation of TRPV1-S800 to abolish the tubulin-TRPV1 interaction. Indeed, in vitro PKCε phosphorylation of TRPV1 hindered tubulin-binding to TRPV1. In vivo, treatment of sensory neurons and F-11 cells with estrogen and the GPR30 agonist, G-1, resulted in microtubule destabilization and retraction of microtubules from filopodial structures. We found estrogen and G-1 to regulate the stability of the microtubular network via PKC phosphorylation of the PKCε-phosphorylation site TRPV1-S800. Microtubule disassembly was not, however, dependent on TRPV1 ion conductivity. TRPV1 knock-down in rats inverted the effect of the microtubule-modulating drugs, Taxol and Nocodazole, on estrogen-induced and PKCε-dependent mechanical pain sensitization. Thus, we suggest the C-terminus of TRPV1 to be a signaling intermediate downstream of estrogen and PKCε, regulating microtubule-stability and microtubule-dependent pain sensitization.


Asunto(s)
Estradiol/farmacología , Estrógenos/farmacología , Microtúbulos/efectos de los fármacos , Canales Catiónicos TRPV/metabolismo , Animales , Unión Competitiva , Línea Celular , Ciclopentanos/farmacología , Estrógenos/fisiología , Ganglios Espinales/citología , Técnicas de Silenciamiento del Gen , Activación del Canal Iónico , Ligandos , Masculino , Microtúbulos/ultraestructura , Modelos Moleculares , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Dolor/fisiopatología , Fosforilación , Unión Proteica , Proteína Quinasa C-epsilon/fisiología , Seudópodos/ultraestructura , Quinolinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Transducción de Señal , Canales Catiónicos TRPV/genética , Tubulina (Proteína)/metabolismo
3.
J Neurosci ; 29(19): 6217-28, 2009 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-19439599

RESUMEN

The transient receptor potential vanilloid 4 (TRPV4) contributes to mechanical hyperalgesia of diverse etiologies, presumably as part of a mechanoreceptor signaling complex (Alessandri-Haber et al., 2008). To investigate the hypothesis that a functional interaction between TRPV4 and stretch-activated ion channels (SACs) is involved in this mechanical transduction mechanism, we used a selective SACs inhibitor, GsMTx-4. Intradermal injection of GsMTx-4 in the rat hindpaw reversed the mechanical hyperalgesia induced by intradermal injection of inflammatory mediators. In vivo single fiber recordings showed that GsMTx-4 reversed inflammatory mediator-induced decrease in mechanical threshold in half of sensitized C-fibers. Furthermore, GsMTx-4 reduced hyperalgesia to both mechanical and hypotonic stimuli in different models of inflammatory and neuropathic pain, although it had no effect on baseline mechanical nociceptive thresholds. TRPC1 and TRPC6, two GsMTx-4-sensitive SACs, are expressed in dorsal root ganglion (DRG) neurons. Single-cell reverse transcription-PCR showed that messenger RNAs for TRPV4, TRPC1, and TRPC6 are frequently coexpressed in DRG neurons. Spinal intrathecal administration of oligodeoxynucleotides antisense to TRPC1 and TRPC6, like that to TRPV4, reversed the hyperalgesia to mechanical and hypotonic stimuli induced by inflammatory mediators without affecting baseline mechanical nociceptive threshold. However, antisense to TRPC6, but not to TRPC1, reversed the mechanical hyperalgesia induced by a thermal injury or the TRPV4-selective agonist 4alpha-PDD (4 alpha-phorbol 12,13-didecanoate). We conclude that TRPC1 and TRPC6 channels cooperate with TRPV4 channels to mediate mechanical hyperalgesia and primary afferent nociceptor sensitization, although they may have distinctive roles.


Asunto(s)
Ganglios Espinales/fisiología , Hiperalgesia/fisiopatología , Neuronas/fisiología , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Pie , Expresión Génica , Miembro Posterior , Hiperalgesia/inducido químicamente , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oligodesoxirribonucleótidos Antisentido/metabolismo , Umbral del Dolor/fisiología , Péptidos/farmacología , Ésteres del Forbol , Estimulación Física , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Venenos de Araña/farmacología , Canal Catiónico TRPC6 , Canales Catiónicos TRPV/genética
4.
Mol Pain ; 6: 98, 2010 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-21187008

RESUMEN

BACKGROUND: Dorsal root ganglia (DRG)-neurons are commonly characterized immunocytochemically. Cells are mostly grouped by the experimenter's eye as "marker-positive" and "marker-negative" according to their immunofluorescence intensity. Classification criteria remain largely undefined. Overcoming this shortfall, we established a quantitative automated microscopy (QuAM) for a defined and multiparametric analysis of adherent heterogeneous primary neurons on a single cell base.The growth factors NGF, GDNF and EGF activate the MAP-kinase Erk1/2 via receptor tyrosine kinase signalling. NGF and GDNF are established factors in regeneration and sensitization of nociceptive neurons. If also the tissue regenerating growth factor, EGF, influences nociceptors is so far unknown. We asked, if EGF can act on nociceptors, and if QuAM can elucidate differences between NGF, GDNF and EGF induced Erk1/2 activation kinetics. Finally, we evaluated, if the investigation of one signalling component allows prediction of the behavioral response to a reagent not tested on nociceptors such as EGF. RESULTS: We established a software-based neuron identification, described quantitatively DRG-neuron heterogeneity and correlated measured sample sizes and corresponding assay sensitivity. Analysing more than 70,000 individual neurons we defined neuronal subgroups based on differential Erk1/2 activation status in sensory neurons. Baseline activity levels varied strongly already in untreated neurons. NGF and GDNF subgroup responsiveness correlated with their subgroup specificity on IB4(+)- and IB4(-)-neurons, respectively. We confirmed expression of EGF-receptors in all sensory neurons. EGF treatment induced STAT3 translocation into the nucleus. Nevertheless, we could not detect any EGF induced Erk1/2 phosphorylation. Accordingly, intradermal injection of EGF resulted in a fundamentally different outcome than NGF/GDNF. EGF did not induce mechanical hyperalgesia, but blocked PGE2-induced sensitization. CONCLUSIONS: QuAM is a suitable if not necessary tool to analyze activation of endogenous signalling in heterogeneous cultures. NGF, GDNF and EGF stimulation of DRG-neurons shows differential Erk1/2 activation responses and a corresponding differential behavioral phenotype. Thus, in addition to expression-markers also signalling-activity can be taken for functional subgroup differentiation and as predictor of behavioral outcome. The anti-nociceptive function of EGF is an intriguing result in the context of tissue damage but also for understanding pain resulting from EGF-receptor block during cancer therapy.


Asunto(s)
Factor de Crecimiento Epidérmico/farmacología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Microscopía/métodos , Dolor/metabolismo , Transducción de Señal/fisiología , Animales , Células Cultivadas , Ganglios Espinales , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Masculino , Factor de Crecimiento Nervioso/farmacología , Dolor/fisiopatología , Ratas , Ratas Sprague-Dawley , Proteínas Tirosina Quinasas Receptoras/metabolismo , Células Receptoras Sensoriales , Programas Informáticos
5.
J Neurosci ; 28(5): 1046-57, 2008 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-18234883

RESUMEN

Although the transient receptor potential vanilloid 4 (TRPV4) has been implicated in the process of osmomechanical transduction, it appears to make little contribution to the normal somatosensory detection of mechanical stimuli. However, evidence suggests that it may play an important role in mechanical hyperalgesia. In the present study, we examined the common requirement for TRPV4 in mechanical hyperalgesia associated with diverse pain models and investigated whether the very close association observed between TRPV4 and mechanical hyperalgesia, regardless of etiology, reflects a close functional connection of TRPV4 with other molecules implicated in mechanical transduction. In models of painful peripheral neuropathy associated with vincristine chemotherapy, alcoholism, diabetes, and human immunodeficiency virus/acquired immune deficiency syndrome therapy, mechanical hyperalgesia was markedly reduced by spinal intrathecal administration of oligodeoxynucleotides antisense to TRPV4. Similarly, mechanical hyperalgesia induced by paclitaxel, vincristine, or diabetes was strongly reduced in TRPV4 knock-out mice. We also show that alpha2beta1 integrin and Src tyrosine kinase, which have been implicated in mechanical transduction, are important for the development of mechanical hyperalgesia, and that their contribution requires TRPV4. Furthermore, we establish a direct interaction between TRPV4, alpha2 integrin, and the Src tyrosine kinase Lyn in sensory neurons. We suggest that TRPV4 plays a role in mechanotransduction, as a component of a molecular complex that functions only in the setting of inflammation or nerve injury.


Asunto(s)
Hiperalgesia/metabolismo , Integrinas/metabolismo , Canales Catiónicos TRPV/metabolismo , Familia-src Quinasas/metabolismo , Animales , Células Cultivadas , Hiperalgesia/genética , Mediadores de Inflamación/metabolismo , Integrina alfa2/genética , Integrina alfa2/metabolismo , Integrina alfa2beta1/genética , Integrina alfa2beta1/metabolismo , Integrinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nociceptores/metabolismo , Dimensión del Dolor/métodos , Estimulación Física/métodos , Ratas , Ratas Sprague-Dawley , Canales Catiónicos TRPV/genética , Familia-src Quinasas/genética
6.
J Neurosci ; 28(22): 5721-30, 2008 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-18509033

RESUMEN

Stress dramatically exacerbates pain in diseases such as fibromyalgia and rheumatoid arthritis, but the underlying mechanisms are unknown. We tested the hypothesis that stress causes generalized hyperalgesia by enhancing pronociceptive effects of immune mediators. Rats exposed to nonhabituating sound stress exhibited no change in mechanical nociceptive threshold, but showed a marked increase in hyperalgesia evoked by local injections of prostaglandin E(2) or epinephrine. This enhancement, which developed more than a week after exposure to stress, required concerted action of glucocorticoids and catecholamines at receptors located in the periphery on sensory afferents. The altered response to pronociceptive mediators involved a switch in coupling of their receptors from predominantly stimulatory to inhibitory G-proteins (G(s) to G(i)), and for prostaglandin E(2), emergence of novel dependence on protein kinase C epsilon. Thus, an important mechanism in generalized pain syndromes may be stress-induced coactivation of the hypothalamo-pituitary-adrenal and sympathoadrenal axes, causing a long-lasting alteration in intracellular signaling pathways, enabling normally innocuous levels of immune mediators to produce chronic hyperalgesia.


Asunto(s)
Neuronas Aferentes/fisiología , Dolor/patología , Transducción de Señal/fisiología , Estrés Fisiológico/fisiopatología , Adrenalectomía/métodos , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Corticosterona/farmacología , Dinoprostona , Modelos Animales de Enfermedad , Epinefrina/efectos adversos , Epinefrina/sangre , Antagonistas de Hormonas/farmacología , Hiperalgesia/inducido químicamente , Hiperalgesia/fisiopatología , Masculino , Mifepristona/farmacología , Músculo Esquelético/inervación , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Ratas , Ratas Sprague-Dawley , Piel/inervación , Sonido/efectos adversos , Estrés Fisiológico/etiología , Factores de Tiempo
7.
Neuron ; 39(4): 613-24, 2003 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-12925276

RESUMEN

Prostaglandin E(2) (PGE(2)) and epinephrine act directly on nociceptors to produce mechanical hyperalgesia through protein kinase A (PKA) alone or through a combination of PKA, protein kinase C epsilon (PKCepsilon), and extracellular signal-regulated kinase (ERK), respectively. Disruptors of the cytoskeleton (microfilaments, microtubules, and intermediate filaments) markedly attenuated the hyperalgesia in rat paws caused by injection of epinephrine or its downstream mediators. In contrast, the hyperalgesia induced by PGE(2) or its mediators was not affected by any of the cytoskeletal disruptors. These effects were mimicked in vitro, as measured by enhancement of the tetrodotoxin-resistant sodium current. When PGE(2) hyperalgesia was shifted to dependence on PKCepsilon and ERK as well as PKA, as when the tissue is "primed" by prior treatment with carrageenan, it too became dependent on an intact cytoskeleton. Thus, inflammatory mediator-induced mechanical hyperalgesia was differentially dependent on the cytoskeleton such that cytoskeletal dependence correlated with mediation by PKCepsilon and ERK.


Asunto(s)
Citoesqueleto/fisiología , Neuronas Aferentes/fisiología , Dolor/fisiopatología , Sistemas de Mensajero Secundario/fisiología , Transducción de Señal/fisiología , Animales , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico , Dinoprostona/metabolismo , Electrofisiología , Epinefrina/metabolismo , Hiperalgesia/inducido químicamente , Inmunohistoquímica , Inflamación/fisiopatología , MAP Quinasa Quinasa Quinasa 3 , Quinasas Quinasa Quinasa PAM/metabolismo , Masculino , Microscopía Confocal , Nociceptores/fisiología , Técnicas de Placa-Clamp , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Quimiocina/fisiología
8.
Eur J Neurosci ; 27(1): 83-92, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18093169

RESUMEN

Chronic alcohol consumption induces a painful small-fiber peripheral neuropathy, the severity of which increases during alcohol withdrawal. Chronic alcohol consumption also produces a sustained increase in stress hormones, epinephrine and corticosterone, that is exacerbated during alcohol withdrawal. We report that adrenal medullectomy and administration of a glucocorticoid receptor antagonist, mifepristone (RU 38486), both prevented and reversed a model of painful peripheral neuropathy in alcohol binge-drinking rats. Chronic administration of stress levels of epinephrine to rats that had undergone adrenal medullectomy and were being fed the alcohol diet reconstituted this phenotype. Intrathecal administration of oligodeoxynucleotides antisense to the beta(2)-adrenergic- or glucocorticoid-receptor also prevented and reversed the pro-nociceptive effects of ethanol. Our results suggest a convergence of the effects of mediators of the hypothalamic-pituitary- and sympathoadrenal-stress axes on sensory neurons in the induction and maintenance of alcohol-induced painful peripheral neuropathy.


Asunto(s)
Neuropatía Alcohólica/complicaciones , Alcoholes/efectos adversos , Neuralgia/etiología , Estrés Fisiológico/inducido químicamente , Adrenalectomía/métodos , Análisis de Varianza , Animales , Interacciones Farmacológicas , Epinefrina/administración & dosificación , Epinefrina/sangre , Antagonistas de Hormonas/administración & dosificación , Hiperalgesia/prevención & control , Masculino , Mifepristona/administración & dosificación , Neuralgia/prevención & control , Oligonucleótidos Antisentido/farmacología , Paclitaxel/administración & dosificación , Dimensión del Dolor/métodos , Umbral del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos beta 2/genética , Receptores de Glucocorticoides/genética , Factores de Tiempo , Zalcitabina/administración & dosificación
9.
Eur J Neurosci ; 27(7): 1700-9, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18371086

RESUMEN

We evaluated the signalling pathway by which estrogen acts in peripheral tissue to produce protein kinase Cepsilon (PKCepsilon)-dependent mechanical hyperalgesia. Specific agonists for the classical estrogen receptors (ER), ERalpha and ERbeta, did not result in activation of PKCepsilon in neurons of dissociated rat dorsal root ganglia. In contrast, G-1, a specific agonist of the recently identified G-protein-coupled estrogen receptor, GPR30, induced PKCepsilon translocation. Involvement of GPR30 and independence of ERalpha and ERbeta was confirmed using the GPR30 agonist and simultaneous ERalpha and ERbeta antagonist ICI 182,780 (fulvestrant). The GPR30 transcript could be amplified from dorsal root ganglia tissue. We found estrogen-induced as well as GPR30-agonist-induced PKCepsilon translocation to be restricted to the subgroup of nociceptive neurons positive for isolectin IB4 from Bandeiraea simplicifolia. Corroborating the cellular results, both GPR30 agonists, G-1 as well as ICI 182,780, resulted in the onset of PKCepsilon-dependent mechanical hyperalgesia if injected into paws of adult rats. We therefore suggest that estrogen acts acutely at GPR30 in nociceptors to produce mechanical hyperalgesia.


Asunto(s)
Moduladores de los Receptores de Estrógeno/farmacología , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/fisiología , Animales , Células Cultivadas , Ciclopentanos/farmacología , Estradiol/análogos & derivados , Estradiol/farmacología , Fulvestrant , Masculino , Estimulación Física/métodos , Proteína Quinasa C-epsilon/metabolismo , Quinolinas/farmacología , Ratas , Ratas Sprague-Dawley
10.
Eur J Neurosci ; 28(6): 1180-90, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18783367

RESUMEN

The neurotoxic effects of catecholamine metabolites have been implicated in neurodegenerative diseases. As some sensory neurons express tyrosine hydroxylase and monoamine oxidase (MAO), we investigated the potential contribution of catecholamine metabolites to neuropathic pain in a model of alcoholic neuropathy. The presence of catecholamines in sensory neurons is supported by capsaicin-stimulated epinephrine release, an effect enhanced in ethanol-fed rats. mRNA for enzymes in dorsal root ganglia involved in catecholamine uptake and metabolism, dopamine beta-hydroxylase and MAO-A, were decreased by neonatal administration of capsaicin. Ethanol-induced hyperalgesia was attenuated by systemic and local peripheral administration of inhibitors of MAO-A, reduction of norepinephrine transporter (NET) in sensory neurons and a NET inhibitor. Finally, intradermal injection of 3,4-dihydroxyphenylglycolaldehyde (DOPEGAL), a neurotoxic MAO-A catecholamine metabolite, produced robust mechanical hyperalgesia. These observations suggest that catecholamines in nociceptors are metabolized to neurotoxic products by MAO-A, which can cause neuronal dysfunction underlying neuropathic pain.


Asunto(s)
Neuropatía Alcohólica/metabolismo , Catecolaminas/metabolismo , Etanol/administración & dosificación , Hiperalgesia/metabolismo , Neurotoxinas/metabolismo , Nociceptores/metabolismo , Neuropatía Alcohólica/fisiopatología , Animales , Conducta Animal/fisiología , Capsaicina/farmacología , Clorgilina/farmacología , Desipramina/farmacología , Inhibidores Enzimáticos/farmacología , Etanol/metabolismo , Ganglios Espinales/metabolismo , Hiperalgesia/fisiopatología , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , Inhibidores de la Monoaminooxidasa/farmacología , Nociceptores/efectos de los fármacos , Nociceptores/fisiopatología , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/antagonistas & inhibidores , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Pargilina/farmacología , Ratas , Ratas Sprague-Dawley , Fármacos del Sistema Sensorial/farmacología
11.
J Pain ; 9(5): 457-62, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18342576

RESUMEN

UNLABELLED: Skeletal muscle injuries can induce chronic pain, but the underlying mechanism is unknown. One possible cause has been suggested to be an increased sensitivity to inflammatory mediators. We demonstrate that self-limited inflammatory hyperalgesia induced by intramuscular carrageenan (lasting approximately 5 days) results in a state of chronic-latent hyperalgesia, revealed by injection of prostaglandin E(2) (PGE(2)) 10 days after carrageenan at the same site. In carrageenan-pretreated muscle, PGE(2) produced hyperalgesia that was unattenuated even 14 days after injection, markedly longer than the 4-hour hyperalgesia induced by PGE(2) in naive rats. This chronic-latent hyperalgesia was reversed as well as prevented by spinal intrathecal injection of oligodeoxynucleotide antisense to protein kinase Cepsilon, a second messenger implicated in long-lasting plasticity in cutaneous nociceptors. PERSPECTIVE: We describe a novel experimental model for chronic muscle pain, produced by mild acute muscle inflammation, that has clinical significance since it has the potential to reveal cellular processes by which acute inflammation or muscle trauma underlies chronic muscle pain.


Asunto(s)
Modelos Animales de Enfermedad , Músculo Esquelético/enzimología , Miositis/enzimología , Nociceptores/enzimología , Dolor/enzimología , Proteína Quinasa C-epsilon/metabolismo , Animales , Carragenina/efectos adversos , Enfermedad Crónica , Dinoprostona/metabolismo , Dinoprostona/farmacología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Hiperalgesia/inducido químicamente , Hiperalgesia/enzimología , Hiperalgesia/fisiopatología , Inyecciones Intramusculares , Inyecciones Espinales , Masculino , Músculo Esquelético/inervación , Músculo Esquelético/fisiopatología , Miositis/inducido químicamente , Miositis/fisiopatología , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/enzimología , Nociceptores/efectos de los fármacos , Nociceptores/fisiopatología , Oligodesoxirribonucleótidos Antisentido/farmacología , Dolor/inducido químicamente , Dolor/fisiopatología , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley
12.
J Neurosci ; 26(14): 3864-74, 2006 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-16597741

RESUMEN

The transient receptor potential vanilloid 4 (TRPV4) is a primary afferent transducer that plays a crucial role in neuropathic hyperalgesia for osmotic and mechanical stimuli, as well as in inflammatory mediator-induced hyperalgesia for osmotic stimuli. In view of the clinical importance of mechanical hyperalgesia in inflammatory states, the present study investigated the role of TRPV4 in mechanical hyperalgesia induced by inflammatory mediators and the second-messenger pathways involved. Intradermal injection of either the inflammogen carrageenan or a soup of inflammatory mediators enhanced the nocifensive paw-withdrawal reflex elicited by hypotonic or mechanical stimuli in rat. Spinal administration of TRPV4 antisense oligodeoxynucleotide blocked the enhancement without altering baseline nociceptive threshold. Similarly, in TRPV4(-/-) knock-out mice, inflammatory soup failed to induce any significant mechanical or osmotic hyperalgesia. In vitro investigation showed that inflammatory mediators engage the TRPV4-mediated mechanism of sensitization by direct action on dissociated primary afferent neurons. Additional behavioral observations suggested that multiple mediators are necessary to achieve sufficient activation of the cAMP pathway to engage the TRPV4-dependent mechanism of hyperalgesia. In addition, direct activation of protein kinase A or protein kinase C epsilon, two pathways that mediate inflammation-induced mechanical hyperalgesia, also induced hyperalgesia for both hypotonic and mechanical stimuli that was decreased by TRPV4 antisense and absent in TRPV4(-/-) mice. We conclude that TRPV4 plays a crucial role in the mechanical hyperalgesia that is generated by the concerted action of inflammatory mediators present in inflamed tissues.


Asunto(s)
Ganglios Espinales/inmunología , Hiperalgesia/inmunología , Mediadores de Inflamación/inmunología , Mecanotransducción Celular/inmunología , Canales Catiónicos TRPV/inmunología , Tacto/inmunología , Animales , Células Cultivadas , Masculino , Ratas , Ratas Sprague-Dawley
13.
Burns ; 33(8): 1021-6, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17707592

RESUMEN

Severe burn induces severe pain. While chronic as well as acute pain syndromes are reported, the peripheral mechanisms of burn-induced chronic pain syndromes have not been studied. We tested the hypothesis that burn induces plastic changes in primary afferent nociceptors that predispose to chronic pain states. Mechanical nociceptive thresholds were measured using the Randall-Selitto paw-withdrawal test in male Sprague-Dawley rats, before and following a small (<1% total body surface area) partial-thickness thermal injury to the dorsal surface of one hind paw. This burn induced mechanical hyperalgesia, which lasted over 2 weeks. After recovery, local injection of prostaglandin E2 (PGE2), to mimic re-injury, induced an enhanced and markedly prolonged mechanical hyperalgesia compared to the hyperalgesic effect of PGE2 in the control contralateral paw. This prolonged PGE2-induced hyperalgesia was reversed by a selective inhibitor of protein kinase C-epsilon (PKCepsilon). Our findings suggest PKCepsilon as a peripheral mechanism for burn-induced chronic pain syndromes.


Asunto(s)
Quemaduras/complicaciones , Hiperalgesia/etiología , Animales , Dinoprostona , Inhibidores Enzimáticos/farmacología , Hiperalgesia/inducido químicamente , Hiperalgesia/enzimología , Masculino , Nociceptores/fisiología , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Proteína Quinasa C-epsilon/fisiología , Ratas , Ratas Sprague-Dawley
14.
J Neurosci ; 25(26): 6119-26, 2005 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-15987941

RESUMEN

The epsilon isoform of protein kinase C (PKCepsilon) has emerged as a critical second messenger in sensitization toward mechanical stimulation in models of neuropathic (diabetes, alcoholism, and cancer therapy) as well as acute and chronic inflammatory pain. Signaling pathways leading to activation of PKCepsilon remain unknown. Recent results indicate signaling from cAMP to PKC. A mechanism connecting cAMP and PKC, two ubiquitous, commonly considered separate pathways, remains elusive. We found that, in cultured DRG neurons, signaling from cAMP to PKCepsilon is not mediated by PKA but by the recently identified cAMP-activated guanine exchange factor Epac. Epac, in turn, was upstream of phospholipase C (PLC) and PLD, both of which were necessary for translocation and activation of PKCepsilon. This signaling pathway was specific to isolectin B4-positive [IB4(+)] nociceptors. Also, in a behavioral model, cAMP produced mechanical hyperalgesia (tenderness) through Epac, PLC/PLD, and PKCepsilon. By delineating this signaling pathway, we provide a mechanism for cAMP-to-PKC signaling, give proof of principle that the mitogen-activated protein kinase pathway-activating protein Epac also stimulates PKC, describe the first physiological function unique for the IB4(+) subpopulation of sensory neurons, and find proof of principle that G-protein-coupled receptors can activate PKC not only through the G-proteins alpha(q) and betagamma but also through alpha(s).


Asunto(s)
AMP Cíclico/fisiología , Glicoproteínas/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Inflamación/fisiopatología , Lectinas/metabolismo , Neuronas/fisiología , Dolor/fisiopatología , Proteína Quinasa C/metabolismo , Animales , Ganglios Espinales/fisiología , Masculino , Técnicas de Cultivo de Órganos , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Versicanos
15.
J Pain ; 7(12): 884-91, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17157774

RESUMEN

UNLABELLED: Although mechanical hyperalgesia associated with medical procedures is the major source of severe pain in burn-injured patients, little is known about its underlying mechanism. One reason for this has been the lack of a model for mechanical hyperalgesia at the site of injury. We have modified an established partial-thickness burn model in the rat to produce long-lasting primary mechanical hyperalgesia, which is present from the first measurement at 0.5 h, reaches a maximum at 3 days, and is still significant after 7 days. Because nerve growth factor (NGF), which is elevated in burn-injured tissue, produces mechanical hyperalgesia and activates protein kinase C (PKC)-epsilon, a key mediator in inflammatory and neuropathic pain, we used this model to evaluate the role of the NGF receptor, tyrosine-receptor kinase A (TrkA), and PKC-epsilon in burn-induced primary mechanical hyperalgesia. Intrathecal administration of antisense oligodeoxynucleotides to TrkA and PKC-epsilon, starting 3 days before inducing a burn injury, caused dose-related decrease of burn-induced primary mechanical hyperalgesia. In addition, intradermal injection of a PKC-epsilon-selective inhibitor eliminated hyperalgesia. Our model provides a method to elucidate the underlying mechanism of burn-injury pain as well as to screen for targets for novel analgesic treatments of this important clinical condition. PERSPECTIVE: This manuscript presents the first model of thermal injury-induced mechanical hyperalgesia which mimics prolonged duration of clinical burn injury pain. We also perform proof of concept experiments demonstrating that our model provides a method to elucidate the mechanism of this important clinical condition.


Asunto(s)
Quemaduras/complicaciones , Hiperalgesia/etiología , Hiperalgesia/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Receptor trkA/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/administración & dosificación , Hiperalgesia/tratamiento farmacológico , Masculino , Oligodesoxirribonucleótidos Antisentido/administración & dosificación , Dimensión del Dolor/métodos , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Péptidos/administración & dosificación , Proteína Quinasa C-epsilon/genética , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/fisiología , Receptor trkA/genética , Factores de Tiempo
16.
J Neurosci ; 24(18): 4444-52, 2004 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-15128858

RESUMEN

The development of treatments for neuropathic pain has been hindered by our limited understanding of the basic mechanisms underlying abnormalities in nociceptor hyperexcitability. We recently showed that the polymodal receptor transient receptor potential vanilloid 4 (TRPV4), a member of the transient receptor potential (TRP) family of ion channels, may play a role in inflammatory pain (Alessandri-Haber et al., 2003). The present study tested whether TRVP4 also contributes to neuropathic pain, using a rat model of Taxol-induced painful peripheral neuropathy. Taxol is the most widely used drug for the treatment of a variety of tumor types, but the dose of Taxol that can be tolerated is limited by the development of a small-fiber painful peripheral neuropathy. We found that Taxol treatment enhanced the nociceptive behavioral responses to both mechanical and hypotonic stimulation of the hind paw. Spinal administration of antisense oligodeoxynucleotides to TRPV4, which reduced the expression of TRPV4 in sensory nerve, abolished Taxol-induced mechanical hyperalgesia and attenuated hypotonic hyperalgesia by 42%. The enhancement of osmotic nociception involves sensitization of osmotransduction in primary afferents because osmotransduction was enhanced in cultured sensory neurons isolated from Taxol-treated rats. Taxol-induced TRPV4-mediated hyperalgesia and the enhanced osmotransduction in cultured nociceptors were dependent on integrin/Src tyrosine kinase signaling. These results suggest that TRPV4 plays a crucial role in a painful peripheral neuropathy, making it a very promising target for the development of a novel class of analgesics.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Canales Iónicos/metabolismo , Neuralgia/fisiopatología , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Animales , Antineoplásicos Fitogénicos , Conducta Animal/efectos de los fármacos , Calcio/metabolismo , Proteínas de Transporte de Catión/antagonistas & inhibidores , Proteínas de Transporte de Catión/genética , Células Cultivadas , Modelos Animales de Enfermedad , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/fisiopatología , Soluciones Hipotónicas , Integrinas/metabolismo , Canales Iónicos/antagonistas & inhibidores , Canales Iónicos/genética , Masculino , Neuralgia/inducido químicamente , Neuralgia/tratamiento farmacológico , Nociceptores/citología , Nociceptores/efectos de los fármacos , Nociceptores/metabolismo , Oligonucleótidos Antisentido/farmacología , Paclitaxel , Dimensión del Dolor , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética , Transducción de Señal/fisiología , Canales Catiónicos TRPV , Familia-src Quinasas/metabolismo
17.
Pain ; 118(1-2): 70-9, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16213085

RESUMEN

The ligand-gated ion channel, TRPV4, functions as a transducer of hypotonic stimuli in primary afferent nociceptive neurons and contributes to inflammatory and neuropathic pain. Hypertonic saline also stimulates primary afferent nociceptors and the injection of mild hypertonic saline (2-5%) is widely used as an experimental model of pain in humans. Therefore, we tested whether TRPV4 participates in the transduction of hypertonic stimuli. Intradermal injection of 2% (607 mOsm) or 10% (3,250 mOsm) saline solution in the hind paw of rats induced a concentration-dependent pain-related behavior, flinching. Sensitization with prostaglandin E(2) (PGE(2)) caused a 7-fold increase in the number of flinches induced by 2% saline but failed to increase those caused by 10% saline. Spinal administration of antisense oligodeoxynucleotides to TRPV4 caused a 46% decrease in the number of flinches induced by 2% saline, but there was no change in flinching induced by 10% saline. Similarly, only the nociceptive behavior caused by 2% saline was reduced in TRPV4(-/-) knockout mice. The TRPV4-mediated nociceptive behaviors induced by hyper- and hypotonic stimuli were dependent on Src tyrosine kinase. We suggest TRPV4 is a transducer in primary afferents that mediates nociceptive behavior induced by small increases or decreases in osmolarity. Such changes in osmolarity might contribute to pain in inflammatory and neuropathic states.


Asunto(s)
Inflamación/fisiopatología , Neuralgia/fisiopatología , Nociceptores/fisiología , Presión Osmótica , Dolor/fisiopatología , Canales Catiónicos TRPV/fisiología , Animales , Soluciones Hipertónicas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuralgia/etiología , Nociceptores/efectos de los fármacos , Presión Osmótica/efectos de los fármacos , Estimulación Física , Ratas , Ratas Sprague-Dawley , Canales Catiónicos TRPV/efectos de los fármacos
18.
Pain ; 115(1-2): 191-203, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15836982

RESUMEN

We recently reported that hyperalgesia induced by the inflammatory mediator prostaglandin E(2) (PGE(2)) requires intact alpha1, alpha3 and beta1 integrin subunit function, whereas epinephrine-induced hyperalgesia depends on alpha5 and beta1. PGE(2)-induced hyperalgesia is mediated by protein kinase A (PKA), while epinephrine-induced hyperalgesia is mediated by a combination of PKA, protein kinase Cepsilon (PKCepsilon) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK). We hypothesized that inflammatory mediator-induced hyperalgesia involves specific interactions between different subsets of integrin subunits and particular second messenger species. In the present study, function-blocking anti-integrin antibodies and antisense oligodeoxynucleotides were used to elucidate these interactions in rat. Hyperalgesia produced by an activator of adenylate cyclase (forskolin) depended on alpha1, alpha3 and beta1 integrins. However, hyperalgesia induced by activation of the cascade at a point farther downstream (by cAMP analog or PKA catalytic subunit) was independent of any integrins tested. In contrast, hyperalgesia induced by a specific PKCepsilon agonist depended only on alpha5 and beta1 integrins. Hyperalgesia induced by agonism of MAPK/ERK depended on all four integrin subunits tested (alpha1, alpha3, alpha5 and beta1). Finally, disruption of lipid rafts antagonized hyperalgesia induced by PGE(2) and by forskolin, but not that induced by epinephrine. Furthermore, alpha1 integrin, but not alpha5, was present in detergent-resistant membrane fractions (which retain lipid raft components). These observations suggest that integrins play a critical role in inflammatory pain by interacting with components of second messenger cascades that mediate inflammatory hyperalgesia, and that such interaction with the PGE(2)-activated pathway may be organized by lipid rafts.


Asunto(s)
Ganglios Espinales/metabolismo , Hiperalgesia/metabolismo , Inflamación/metabolismo , Integrinas/antagonistas & inhibidores , Integrinas/metabolismo , Transducción de Señal , Animales , Colforsina , Dinoprostona , Epinefrina , Ganglios Espinales/efectos de los fármacos , Hiperalgesia/inducido químicamente , Hiperalgesia/etiología , Inflamación/inducido químicamente , Inflamación/etiología , Masculino , Ratas , Ratas Sprague-Dawley
19.
Eur J Pain ; 15(8): 796-800, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21419675

RESUMEN

Stress exacerbates both experimental and clinical pain, most well-characterized in irritable bowel and fibromyalgia syndromes. Since it has been hypothesized that cytokines play an etiopathogenic role in fibromyalgia and other chronic widespread pain conditions, we investigated the relationship between stress and cytokines in a model of stress-induced chronic somatic pain. A series of experiments were performed to evaluate the impact of stress on the hyperalgesia-induced by endotoxin (lipopolysaccharide, LPS) and the role of two pro-inflammatory cytokines, interleukin-6 (IL-6) and tumor necrosis α (TNFα). Fourteen days after exposure to a 4-day protocol of unpredictable sound stress, the ability of systemic LPS (100 µg/kg, i.p) to elicit cytokine-mediated mechanical hyperalgesia was measured in gastrocnemius muscle. LPS-induced hyperalgesia was significantly greater in stressed rats, but when rats were treated intrathecally with antisense oligodeoxynucleotide (ODN), to decrease either the gp130 subunit of the IL-6 receptor or the TNFα receptor, in nociceptors, skeletal muscle hyperalgesia in sound stressed, but not control, rats was prevented. These data suggest that chronic stress alters signaling in the primary afferent nociceptor for the hyperalgesia induced by endogenously produced pro-inflammatory cytokines.


Asunto(s)
Citocinas/fisiología , Hiperalgesia/metabolismo , Músculo Esquelético/metabolismo , Nociceptores/fisiología , Sonido/efectos adversos , Estrés Psicológico/metabolismo , Estimulación Acústica/efectos adversos , Animales , Modelos Animales de Enfermedad , Hiperalgesia/fisiopatología , Masculino , Músculo Esquelético/inervación , Músculo Esquelético/fisiopatología , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/etiología , Estrés Psicológico/fisiopatología
20.
J Pain ; 11(4): 369-77, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19962353

RESUMEN

UNLABELLED: While occupational exposure to vibration is a common cause of acute and chronic musculoskeletal pain, eliminating exposure produces limited symptomatic improvement, and reexposure precipitates rapid recurrence or exacerbation. To evaluate mechanisms underlying these pain syndromes, we have developed a model in the rat, in which exposure to vibration (60-80Hz) induces, in skeletal muscle, both acute mechanical hyperalgesia as well as long-term changes characterized by enhanced hyperalgesia to a proinflammatory cytokine or reexposure to vibration. Exposure of a hind limb to vibration-produced mechanical hyperalgesia measured in the gastrocnemius muscle of the exposed hind limb, which persisted for approximately 2 weeks. When nociceptive thresholds had returned to baseline, exposure to a proinflammatory cytokine or reexposure to vibration produced markedly prolonged hyperalgesia. The chronic prolongation of vibration- and cytokine-hyperalgesia was prevented by spinal intrathecal injection of oligodeoxynucleotide (ODN) antisense to protein kinase Cepsilon, a second messenger in nociceptors implicated in the induction and maintenance of chronic pain. Vibration-induced hyperalgesia was inhibited by spinal intrathecal administration of ODN antisense to receptors for the type-1 tumor necrosis factor-alpha (TNFalpha) receptor. Finally, in TNFalpha-pretreated muscle, subsequent vibration-induced hyperalgesia was markedly prolonged. PERSPECTIVE: These studies establish a model of vibration-induced acute and chronic musculoskeletal pain, and identify the proinflammatory cytokine TNFalpha and the second messenger protein kinase Cepsilon as targets against which therapies might be directed to prevent and/or treat this common and very debilitating chronic pain syndrome.


Asunto(s)
Enfermedades Musculoesqueléticas/etiología , Enfermedades Musculoesqueléticas/fisiopatología , Exposición Profesional/efectos adversos , Dolor/etiología , Dolor/fisiopatología , Vibración/efectos adversos , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Miembro Posterior/efectos de los fármacos , Miembro Posterior/fisiopatología , Hiperalgesia/etiología , Hiperalgesia/fisiopatología , Inflamación/etiología , Inflamación/fisiopatología , Mediadores de Inflamación/farmacología , Inyecciones Espinales , Masculino , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiopatología , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/uso terapéutico , Umbral del Dolor/fisiología , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Proteína Quinasa C-epsilon/genética , Ratas , Ratas Sprague-Dawley , Receptores Tipo I de Factores de Necrosis Tumoral/antagonistas & inhibidores , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Sistemas de Mensajero Secundario/efectos de los fármacos , Sistemas de Mensajero Secundario/fisiología , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA