Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Neurosci Lett ; 496(2): 70-4, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21501659

RESUMO

Nerve growth factor (NGF) activates multiple downstream effectors, including Ras, phosphoinositide-3 kinase, and sphingomyelins. However, pathway mediating the NGF-induced augmentation of sensory neuronal excitability remains largely unknown. We previously reported that small-diameter sensory neurons with a heterozygous mutation of the Nf1 gene (Nf1+/-) exhibited increased excitability. The protein product of the Nf1 gene is neurofibromin, a guanosine triphosphatase-activating protein (GAP) for p21ras (Ras) that accelerates the conversion of active Ras-GTP to inactive Ras-GDP. Thus, Nf1+/- cells have augmented basal and stimulated Ras activity. To investigate whether NGF-induced increases in excitability of small-diameter sensory neurons are dependent on Ras signaling, an antibody that blocks the activation of Ras, Y13-259, was perfused into the cell. Under these conditions, the enhanced excitability produced by NGF was suppressed in wildtype neurons but the excitability of Nf1+/- neurons was unaltered. In addition, expression of a dominant-negative form of Ras abolished the ability of NGF to increase the excitability of small-diameter sensory neurons. These results demonstrate that NGF enhances excitability of small-diameter sensory neurons in a Ras-dependent manner while the consequences of decreased expression of neurofibromin cannot be restored by blocking Ras signaling; suggesting that Ras-initiated signaling pathways can regulate both transcriptional and posttranslational control of ion channels important in neuronal excitability.


Assuntos
Potenciais de Ação/fisiologia , Capsaicina/farmacologia , Fatores de Transcrição NFI/metabolismo , Fator de Crescimento Neural/farmacologia , Células Receptoras Sensoriais/fisiologia , Transdução de Sinais/fisiologia , Proteínas ras/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Tamanho Celular , Camundongos , Camundongos Knockout , Fatores de Transcrição NFI/genética , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Especificidade da Espécie
2.
Neurotoxicology ; 32(1): 140-9, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20932997

RESUMO

Peripheral neuropathy is a major side effect following treatment with the cancer chemotherapeutic drug paclitaxel. Whether paclitaxel-induced peripheral neuropathy is secondary to altered function of small diameter sensory neurons remains controversial. To ascertain whether the function of the small diameter sensory neurons was altered following systemic administration of paclitaxel, we injected male Sprague Dawley rats with 1mg/kg paclitaxel every other day for a total of four doses and examined vasodilatation in the hindpaw at day 14 as an indirect measure of calcitonin gene related peptide (CGRP) release. In paclitaxel-treated rats, the vasodilatation induced by either intradermal injection of capsaicin into the hindpaw or electrical stimulation of the sciatic nerve was significantly attenuated in comparison to vehicle-injected animals. Paclitaxel treatment, however, did not affect direct vasodilatation induced by intradermal injection of methacholine or CGRP, demonstrating that the blood vessels' ability to dilate was intact. Paclitaxel treatment did not alter the compound action potentials or conduction velocity of C-fibers. The stimulated release of CGRP from the central terminals in the spinal cord was not altered in paclitaxel-injected animals. These results suggest that paclitaxel affects the peripheral endings of sensory neurons to alter transmitter release, and this may contribute to the symptoms seen in neuropathy.


Assuntos
Membro Posterior/irrigação sanguínea , Membro Posterior/efeitos dos fármacos , Paclitaxel/toxicidade , Células Receptoras Sensoriais/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Membro Posterior/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/fisiologia , Vasodilatação/fisiologia
3.
Neuroscience ; 171(3): 910-23, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20870010

RESUMO

Nerve growth factor (NGF) can augment transmitter release in sensory neurons by acutely sensitizing sensory neurons and by increasing the expression of calcitonin gene-related peptide (CGRP) over time. The current study examined the intracellular signaling pathways that mediate these two temporally distinct effects of NGF to augment CGRP release from sensory neurons. Growing sensory neurons in 30 or 100 ng/mL of NGF for 7 days increases CGRP content and this increase augments the amount of CGRP that is released by high extracellular potassium. Overexpressing a dominant negative Ras, Ras(17N) or treatment with a farnesyltransferase inhibitor attenuates the NGF-induced increase in CGRP content. Conversely, overexpressing a constitutively active Ras augments the NGF-induced increase in content of CGRP. Inhibiting mitogen activated protein kinase (MEK) activity also blocks the ability of NGF to increase CGRP expression. In contrast to the ability of chronic NGF to increase peptide content, acute exposure of sensory neurons to 100 ng/mL NGF augments capsaicin-evoked release of CGRP without affecting the content of CGRP. This sensitizing action of NGF is not affected by inhibiting Ras, MEK, or PI3 kinases. In contrast, the NGF-induced increase in capsaicin-evoked release of CGRP is blocked by the protein kinase C (PKC) inhibitor, BIM and the Src family kinases inhibitor, PP2. These data demonstrate that different signaling pathways mediate the alterations in expression of CGRP by chronic NGF and the acute actions of the neurotrophin to augment capsaicin-evoked release of CGRP in the absence of a change in the content of the peptide.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/genética , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Fator de Crescimento Neural/fisiologia , Nociceptores/metabolismo , Células Receptoras Sensoriais/metabolismo , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia , Animais , Peptídeo Relacionado com Gene de Calcitonina/biossíntese , Células Cultivadas , Gânglios Espinais/metabolismo , Gânglios Espinais/fisiologia , Masculino , Fator de Crescimento Neural/metabolismo , Nociceptores/fisiologia , Radioimunoensaio , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/fisiologia , Transdução de Sinais/genética , Regulação para Cima/genética
4.
Am J Transplant ; 8(3): 537-46, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18294150

RESUMO

Type V collagen (col[V])-reactive lymphocytes contribute to lung transplant rejection, but the mechanisms for emigration into the graft are unknown. Sphingosine-1-phosphate-1 receptors (S1P(1R)) are believed to be required for lymphocyte emigration in other studies, but their role in col(V)-reactive lymphocyte rejection responses is not known. Utilizing small interfering RNA (siRNA) to reduce S1P(1R) expression on col(V)-reactive lymphocytes, we examined the role of S1P(1R) in the rejection response. Quantitative polymerase chain reaction (PCR) revealed strong expression of S1P(1R) messenger RNA (mRNA)on col(V)-reactive lymphocytes isolated from immunized rats. S1P(1R)-specific siRNA (S1P(1R) siRNA) reduced expression of S1P(1R) mRNA and protein, whereas scramble siRNA (SC siRNA) had no effect. Adoptive transfer of lymphocytes treated with S1P(1R) siRNA to rat Wistar Kyoto (WKY) lung isograft recipients resulted in retention of cells within the liver with fewer cells in mediastinal lymph nodes when compared to cells exposed to SC siRNA. S1P(1R)-deficient cells proliferated in response to alloantigens, but not in response to col(V), and produced less interferon (IFN)-gamma in response to col(V) compared to controls. Downregulating S1P(1R) did not affect production of interleukin (IL)-10and tumor necrosis factor (TNF)-alpha, or expression of adhesion molecules critical for migration, but prevented rejection pathology and lowered local levels of IFN-gamma post adoptive transfer. These data demonstrate novel roles of S1P(1R,) which include regulating emigration and modulating lymphocyte activation.


Assuntos
Movimento Celular/genética , Colágeno Tipo V/imunologia , Rejeição de Enxerto/imunologia , Transplante de Pulmão , Receptores de Lisoesfingolipídeo/fisiologia , Linfócitos T/imunologia , Transferência Adotiva , Animais , Rejeição de Enxerto/patologia , Masculino , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Endogâmicos WKY , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/genética , Linfócitos T/efeitos dos fármacos , Linfócitos T/transplante , Transcrição Gênica/efeitos dos fármacos
5.
Neuroscience ; 152(1): 119-27, 2008 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-18222611

RESUMO

Dorsal horn N-methyl-D-aspartate (NMDA) receptors contribute significantly to spinal nociceptive processing through an effect postsynaptic to non-primary glutamatergic axons, and perhaps presynaptic to the primary afferent terminals. The present study sought to examine the regulatory effects of NMDA receptors on primary afferent release of substance P (SP), as measured by neurokinin 1 receptor (NK1r) internalization in the spinal dorsal horn of rats. The effects of intrathecal NMDA alone or in combination with D-serine (a glycine site agonist) were initially examined on basal levels of NK1r internalization. NMDA alone or when co-administered with D-serine failed to induce NK1r internalization, whereas activation of spinal TRPV1 receptors by capsaicin resulted in a notable NK1r internalization. To determine whether NMDA receptor activation could potentiate NK1r internalization or pain behavior induced by a peripheral noxious stimulus, intrathecal NMDA was given prior to an intraplantar injection of formalin. NMDA did not alter the formalin-induced NK1r internalization nor did it enhance the formalin paw flinching behavior. To further characterize the effects of presynaptic NMDA receptors, the NMDA antagonists DL-2-amino-5-phosphonopentanoic acid (AP-5) and MK-801 were intrathecally administered to assess their regulatory effects on formalin-induced NK1r internalization and pain behavior. AP-5 had no effect on formalin-induced NK1r internalization, whereas MK-801 produced only a modest reduction. Both antagonists, however, reduced the formalin paw flinching behavior. In subsequent in vitro experiments, perfusion of NMDA in spinal cord slice preparations did not evoke basal release of SP or calcitonin gene-related peptide (CGRP). Likewise, perfusion of NMDA did not enhance capsaicin-evoked release of the two peptides. These results suggest that presynaptic NMDA receptors in the spinal cord play little if any role on the primary afferent release of SP.


Assuntos
Neurônios Aferentes/metabolismo , Dor/metabolismo , Células do Corno Posterior/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Substância P/metabolismo , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Processamento de Imagem Assistida por Computador , Masculino , Microscopia Confocal , N-Metilaspartato/metabolismo , N-Metilaspartato/farmacologia , Neurônios Aferentes/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores da Neurocinina-1/efeitos dos fármacos , Receptores da Neurocinina-1/metabolismo
6.
J Physiol ; 575(Pt 1): 101-13, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16740613

RESUMO

Our previous studies found that nerve growth factor (NGF), via ceramide, enhanced the number of action potentials (APs) evoked by a ramp of depolarizing current in capsaicin-sensitive sensory neurons. Ceramide can be metabolized by ceramidase to sphingosine (Sph), and Sph to sphingosine 1-phosphate (S1P) by sphingosine kinase. It is well established that each of these products of sphingomyelin metabolism can act as intracellular signalling molecules. This raises the question as to whether the enhanced excitability produced by NGF was mediated directly by ceramide or required additional metabolism to Sph and/or S1P. Sph applied externally did not affect the neuronal excitability, whereas internally perfused Sph augmented the number of APs evoked by the depolarizing ramp. Furthermore, internally perfused S1P enhanced the number of evoked APs. This sensitizing action of NGF, ceramide and internally perfused Sph was abolished by dimethylsphingosine (DMS), an inhibitor of sphingosine kinase. In contrast, internally perfused S1P enhanced the number of evoked APs in the presence of DMS. These observations support the idea that the metabolism of ceramide/Sph to S1P is critical for the sphingolipid-induced modulation of excitability. Both internally perfused Sph and S1P inhibited the outward K+ current by 25-35% for the step to +60 mV. The Sph- and S1P-sensitive currents had very similar current-voltage relations, suggesting that they were likely to be the same. In addition, the Sph-induced suppression of the K+ current was blocked by pretreatment with DMS. These findings demonstrate that intracellular S1P derived from ceramide acts as an internal second messenger to regulate membrane excitability; however, the effector system whereby S1P modulates excitability remains undetermined.


Assuntos
Gânglios Espinais/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Fator de Crescimento Neural/farmacologia , Neurônios Aferentes/efeitos dos fármacos , Esfingosina/análogos & derivados , Potenciais de Ação , Animais , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Gânglios Espinais/citologia , Gânglios Espinais/enzimologia , Masculino , Neurônios Aferentes/enzimologia , Técnicas de Patch-Clamp , Perfusão , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Esfingosina/farmacologia , Fatores de Tempo
7.
J Neurophysiol ; 96(3): 1042-52, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16723416

RESUMO

Sphingosine-1-phosphate (S1P) is released by immune cells and is thought to play a key role in chemotaxis and the onset of the inflammatory response. The question remains whether this lipid mediator also contributes to the enhanced sensitivity of nociceptive neurons that is associated with inflammation. Therefore we examined whether S1P alters the excitability of small diameter, capsaicin-sensitive sensory neurons by measuring action potential (AP) firing and two of the membrane currents critical in regulating the properties of the AP. External application of S1P augments the number of APs evoked by a depolarizing current ramp. The enhanced firing is associated with a decrease in the rheobase and an increase in the resistance at firing threshold although neither the firing threshold nor the resting membrane potential are changed. Treatment with S1P enhanced the tetrodotoxin-resistant sodium current and decreased the total outward potassium current (IK). When sensory neurons were internally perfused with GDP-beta-S, a blocker of G protein activation, the S1P-induced increase in APs was completely blocked and suggests the excitatory actions of S1P are mediated through G-protein-coupled receptors called endothelial differentiation gene or S1PR. In contrast, internal perfusion with GDP-beta-S and S1P increased the number of APs evoked by the current ramp. These results and our finding that the mRNAs for S1PRs are expressed in both the intact dorsal root ganglion and cultures of adult sensory neurons supports the notion that S1P acts on S1PRs linked to G proteins. Together these findings demonstrate that S1P can regulate the excitability of small diameter sensory neurons by acting as an external paracrine-type ligand through activation of G-protein-coupled receptors and thus may contribute to the hypersensitivity during inflammation.


Assuntos
Lisofosfolipídeos/farmacologia , Neurônios Aferentes/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Lisoesfingolipídeo/fisiologia , Esfingosina/análogos & derivados , Animais , Células Cultivadas , Gânglios Espinais/fisiologia , Masculino , Neurônios Aferentes/efeitos dos fármacos , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/genética , Esfingosina/farmacologia , Tetrodotoxina/farmacologia
8.
J Pharmacol Exp Ther ; 306(3): 1137-44, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12829729

RESUMO

ATP has recently emerged as an important proinflammatory mediator that has direct excitatory actions on sensory neurons through activation of ion channel-coupled P2X receptors. The purpose of the current work is to assess whether ATP alters the release of neuropeptides from sensory neurons and the receptors mediating this putative action. Exposing embryonic sensory neurons in culture to concentrations of ATP up to 300 microm did not increase the release of immunoreactive substance P or calcitonin gene-related peptide from sensory neurons. However, pre-exposing sensory neurons to 0.1 to 100 microm ATP prior to and throughout administration of 30 nM capsaicin resulted in a significant augmentation of release evoked by the vanilloid. This sensitizing action of ATP is blocked by suramin but not pyridoxal phosphate-6-azobenzene-2,4-disulfonic acid and is mimicked by the P2Y receptor agonists, 2-2-chloroadenosine triphosphate and UTP, but not by 2-(methylthio)adenosine 5'-triphosphate or alpha,beta-methyleneadenosine 5'-diphosphate. This profile of drug actions suggests that the sensitizing actions of ATP are mediated by P2Y receptors. Pretreating sensory neurons with bisindolylmaleimide I, a selective protein kinase C (PKC) inhibitor, attenuates the augmentation of capsaicin-induced peptide release by ATP, further implicating P2Y receptors in the actions of ATP. Immunoblotting also indicates the presence of P2Y2-like immunoreactive substance in embryonic dorsal root ganglia neurons. Together, these data support the notion that ATP acts at P2Y receptors in sensory neurons in a PKC-dependent manner to augment their sensitivity to other stimuli.


Assuntos
Trifosfato de Adenosina/farmacologia , Neurônios Aferentes/efeitos dos fármacos , Fosfato de Piridoxal/análogos & derivados , Receptores Purinérgicos P2/metabolismo , Análise de Variância , Animais , Células Cultivadas , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Indóis/farmacologia , Maleimidas/farmacologia , Neurônios Aferentes/metabolismo , Peptídeos/metabolismo , Inibidores da Agregação Plaquetária/farmacologia , Antagonistas do Receptor Purinérgico P2 , Fosfato de Piridoxal/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2/biossíntese , Receptores Purinérgicos P2Y2
9.
J Physiol ; 544(2): 385-402, 2002 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-12381813

RESUMO

Because nerve growth factor (NGF) is elevated during inflammation and is known to activate the sphingomyelin signalling pathway, we examined whether NGF and its putative second messenger, ceramide, could modulate the excitability of capsaicin-sensitive adult and embryonic sensory neurons. Using the whole-cell patch-clamp recording technique, exposure of isolated sensory neurons to either 100 ng ml(-1) NGF or 1 microM N-acetyl sphingosine (C2-ceramide) produced a 3- to 4-fold increase in the number of action potentials (APs) evoked by a ramp of depolarizing current in a time-dependent manner. Intracellular perfusion with bacterial sphingomyelinase (SMase) also increased the number of APs suggesting that the release of native ceramide enhanced neuronal excitability. Glutathione, an inhibitor of neutral SMase, completely blocked the NGF-induced augmentation of AP firing, whereas dithiothreitol, an inhibitor of acidic SMase, was without effect. In the presence of glutathione and NGF, exogenous ceramide still enhanced the number of evoked APs, indicating that the sensitizing action of ceramide was downstream of NGF. To investigate the mechanisms of action for NGF and ceramide, isolated membrane currents were examined. Both NGF and ceramide facilitated the peak amplitude of the TTX-resistant sodium current (TTX-R I(Na)) by approximately 1.5-fold and shifted the activation to more hyperpolarized voltages. In addition, NGF and ceramide suppressed an outward potassium current (I(K)) by approximately 35 %. Ceramide reduced I(K) in a concentration-dependent manner. Isolation of the NGF- and ceramide-sensitive currents indicates that they were delayed rectifier types of I(K). The inflammatory prostaglandin, PGE(2), produced an additional suppression of I(K) after exposure to ceramide (approximately 35 %), suggesting that these agents might act on different targets. Thus, our findings indicate that the pro-inflammatory agent, NGF, can rapidly enhance the excitability of sensory neurons. This NGF-induced sensitization is probably mediated by activation of the sphingomyelin signalling pathway to liberate ceramide(s), wherein ceramide appears to be the second messenger involved in modulating neuronal excitability.


Assuntos
Ceramidas/fisiologia , Fator de Crescimento Neural/fisiologia , Neurônios Aferentes/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/fisiologia , Sistemas do Segundo Mensageiro/fisiologia , Canais de Sódio/fisiologia , Tetrodotoxina/farmacologia , Animais , Células Cultivadas , Ceramidas/farmacologia , Canais de Potássio de Retificação Tardia , Dinoprostona/farmacologia , Condutividade Elétrica , Inibidores Enzimáticos/farmacologia , Masculino , Fator de Crescimento Neural/farmacologia , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/fisiologia , Canais de Potássio/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Canais de Sódio/efeitos dos fármacos , Esfingomielina Fosfodiesterase/antagonistas & inibidores
10.
Pain ; 96(3): 285-296, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11973001

RESUMO

Although the tissue levels of prostaglandins are elevated for a relatively long period during injury or inflammation, few studies have been performed to assess the effects of prolonged prostaglandin exposure on receptor binding and activity in sensory neurons. Consequently, we examined whether unilateral inflammation or a 24 h exposure to prostaglandin E2 (PGE2) altered binding of this prostanoid in spinal cord tissue or in isolated sensory neurons, respectively. To assess functional changes in EP receptors, we also examined PGE2-induced cAMP production and the prostanoid-mediated augmentation of substance P release from isolated sensory neurons after acute and 24 h pretreatment with PGE2. Injection of complete Freund's adjuvant into the hindpaw decreased binding of PGE2 in ipsilateral, but not contralateral dorsal spinal cord 24 h after injection. This decrease in Bmax was blocked by administration of intrathecal ketorolac (10 nmol/microl/h) for 24 h prior to and throughout the period of inflammation, suggesting that the inflammation-induced decrease in binding is dependent on prostaglandin synthesis. In an analogous manner, treating sensory neurons grown in culture with 1 microM PGE2 for 24 h decreased [3H]-PGE2 binding by approximately 50% without altering binding affinity. Exposing neuronal cultures to 1 microM PGE2 for 24 h also reduced, but did not abolish the ability of the prostanoid to increase the production of cAMP. This treatment, however, did not significantly alter the ability of PGE2 to augment the evoked release of immunoreactive substance P from sensory neurons. These results demonstrate that under conditions that significantly downregulate PGE2 binding, sensory neurons are still capable of maintaining PGE2-mediated sensitization.


Assuntos
Dinoprostona/farmacologia , Neurônios Aferentes/metabolismo , Receptores de Prostaglandina E/metabolismo , Animais , Células Cultivadas , AMP Cíclico/metabolismo , Dinoprostona/metabolismo , Regulação para Baixo/efeitos dos fármacos , Feminino , Gânglios Espinais/citologia , Masculino , Inflamação Neurogênica/metabolismo , Gravidez , Ratos , Ratos Sprague-Dawley , Medula Espinal/citologia , Substância P/metabolismo , Trítio
11.
J Biol Chem ; 276(19): 16083-91, 2001 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-11278900

RESUMO

Although a number of prostaglandin E(2) (PGE(2)) receptor subtypes have been cloned, limited studies have been performed to elucidate subtypes that subserve specific actions of this eicosanoid, in part because of a paucity of selective receptor antagonists. Using reverse transcription-polymerase chain reaction (PCR) and antisense oligonucleotides, we examined which prostaglandin E(2) receptor (EP receptor) subtypes are expressed in sensory neurons and which mediate the PGE(2)-induced increase in cAMP production and augmentation of peptide release. Reverse transcription-PCR of cDNA isolated from rat sensory neurons grown in culture revealed PCR products for the EP1, EP2, EP3C, and EP4 receptor subtypes but not the EP3A or EP3B. Preexposing neuronal cultures for 48 h to antisense oligonucleotides of EP3C and EP4 mRNA diminished expression of the respective receptors by approximately 80%, abolished the PGE(2)-stimulated production of cAMP, and blocked the ability of PGE(2) to augment release of immunoreactive substance P and calcitonin gene-related peptide. Pretreating with individual antisense against the EP2, EP3C, or EP4 receptors or combinations of missense oligonucleotides had no effect on PGE(2)-induced activity. Treatment with antisense to EP3C and EP4 receptor subtypes did not alter the ability of forskolin to increase cAMP or enhance peptide release. These results demonstrate that sensory neurons are capable of expressing multiple EP receptor subtypes but that only the EP3C and EP4 receptors mediate PGE(2)-induced sensitization of sensory neurons.


Assuntos
AMP Cíclico/metabolismo , Dinoprostona/farmacologia , Neurônios Aferentes/fisiologia , Receptores de Prostaglandina E/fisiologia , Receptores de Prostaglandina/fisiologia , Animais , Células Cultivadas , Colforsina/farmacologia , Embrião de Mamíferos , Gânglios Espinais/citologia , Gânglios Espinais/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Neurônios Aferentes/efeitos dos fármacos , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , Ratos , Receptores de Epoprostenol , Receptores de Prostaglandina/efeitos dos fármacos , Receptores de Prostaglandina/genética , Receptores de Prostaglandina E/efeitos dos fármacos , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E Subtipo EP4 , Transcrição Gênica
12.
Neurosci Lett ; 287(1): 33-6, 2000 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-10841984

RESUMO

To ascertain whether chronic exposure to nerve growth factor (NGF) alters the responsiveness of sensory neurons to prostaglandin E(2) (PGE(2)), sensory neurons taken from adult rats were grown in culture in the presence or absence of NGF for 7 days. Neurons then were exposed to PGE(2) and release of immunoreactive calcitonin gene-related peptide (iCGRP) and production of immunoreactive cAMP (icAMP) were examined. Growing neurons in the presence of 250 ng/ml NGF increased the content and the release of iCGRP from sensory neurons. Independent of NGF treatment, exposure to 100 nM PGE(2) augmented capsaicin- or potassium-stimulated release of iCGRP by 1. 5-fold compared with cells not exposed to PGE(2). In a similar manner, NGF treatment did not alter the ability of PGE(2) to increase the content of icAMP. These data suggest that prostaglandin-induced sensitization of sensory neurons is not influenced by NGF.


Assuntos
Dinoprostona/metabolismo , Dinoprostona/farmacologia , Fator de Crescimento Neural/metabolismo , Fator de Crescimento Neural/farmacologia , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/metabolismo , Animais , Peptídeo Relacionado com Gene de Calcitonina/efeitos dos fármacos , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Capsaicina/farmacologia , Células Cultivadas , AMP Cíclico/metabolismo , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Hiperalgesia/etiologia , Hiperalgesia/fisiopatologia , Inflamação/etiologia , Inflamação/fisiopatologia , Masculino , Nociceptores/efeitos dos fármacos , Nociceptores/metabolismo , Ratos , Ratos Sprague-Dawley
13.
J Pharmacol Exp Ther ; 293(3): 912-20, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10869392

RESUMO

Isoprostanes are a novel class of eicosanoids primarily formed by peroxidation of arachidonic acid. Because of their potential as inflammatory and/or hyperalgesic agents whose formation is largely independent of cyclooxygenases, we examined whether 8-iso prostaglandin E(2) (8-iso PGE(2)) or 8-iso prostaglandin F(2alpha) (8-iso PGF(2alpha)) reduces mechanical and thermal withdrawal threshold in rats, and whether they sensitize rat sensory neurons. Injection of 1 microg of 8-iso PGE(2) (in 2.5 microl) into the hindpaw of rats significantly reduced mechanical and thermal withdrawal thresholds, whereas 1 microg of 8-iso PGF(2alpha) elicited a transient decrease in only the mechanical withdrawal threshold. Both isoprostanes enhanced the firing of C-nociceptors in a concentration-dependent manner when injected into peripheral receptive fields. Exposing sensory neurons grown in culture to 1 microM 8-iso PGE(2) or 8-iso PGF(2alpha) augmented the number of action potentials elicited by a ramp of depolarizing current. In contrast, 8-iso PGE(2) but not 8-iso PGF(2alpha) enhanced the release of substance P- and calcitonin gene-related peptide-like immunoreactivity from isolated sensory neurons. Ten micromolar 8-iso PGE(2) stimulated peptide release directly, whereas treatment with 1 microM 8-iso PGE(2) augmented the release evoked by either bradykinin or capsaicin. Pretreating neuronal cultures with the nonsteroidal anti-inflammatory drug ketorolac did not alter the sensitizing action of 8-iso PGE(2) on peptide release, suggesting that this action of the isoprostane was not secondary to the production of prostaglandins via the cyclooxygenase pathway. These data support the notion that isoprostanes are an important class of inflammatory mediators that augment nociception.


Assuntos
Dinoprosta/análogos & derivados , Dinoprostona/análogos & derivados , Isoprostanos , Neurônios Aferentes/efeitos dos fármacos , Dor/etiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Células Cultivadas , AMP Cíclico/fisiologia , Dinoprosta/farmacologia , Dinoprostona/farmacologia , F2-Isoprostanos , Feminino , Masculino , Fibras Nervosas/efeitos dos fármacos , Fibras Nervosas/fisiologia , Neurônios Aferentes/fisiologia , Prostaglandinas/biossíntese , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Substância P/metabolismo
15.
Neurosci Lett ; 265(1): 17-20, 1999 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-10327195

RESUMO

One possible mechanism to account for the pain enhancing effects of protein kinase C (PKC) activation may be a facilitation of neurotransmitter release from terminals of nociceptive sensory neurons in the spinal cord. To examine this notion, we studied whether treatment with a phorbol ester enhanced the resting and capsaicin-evoked release of immunoreactive substance P (iSP) and immunoreactive calcitonin gene-related peptide (iCGRP) using an in vitro spinal cord slice preparation. Exposing the spinal cord tissue to 100 nM phorbol 12,13 dibutyrate (PDBu), an activator of PKC, results in a two-fold increase in the basal and the capsaicin-evoked release of iSP and iCGRF compared to evoked peptide release without PDBu. When the tissue was perfused with 1 microM 4-alpha PDBu, an analog of PDBu that does not activate PKC, the peptide release was not significantly different from control. Pre-exposing slices to 1 microM bisindolylmaleimide I, an inhibitor of PKC activity, prevents the facilitation of peptide release induced by PDBu. These results suggest that activation of PKC can augment the release of peptides in the spinal cord, which could increase nociceptive sensory transmission and contribute to hyperalgesia.


Assuntos
Neuropeptídeos/metabolismo , Proteína Quinase C/metabolismo , Medula Espinal/metabolismo , Animais , Peptídeo Relacionado com Gene de Calcitonina/análise , Ativação Enzimática , Imuno-Histoquímica , Técnicas In Vitro , Masculino , Dibutirato de 12,13-Forbol/farmacologia , Ratos , Ratos Sprague-Dawley , Medula Espinal/efeitos dos fármacos , Substância P/análise
16.
J Physiol ; 516 ( Pt 1): 163-78, 1999 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-10066931

RESUMO

1. The role of the cyclic AMP (cAMP) transduction cascade in mediating the prostaglandin E2 (PGE2)-induced decrease in potassium current (IK) was investigated in isolated embryonic rat sensory neurones using the whole-cell patch-clamp recording technique. 2. Exposure to 100 microM chlorophenylthio-adenosine cyclic 3', 5'-monophosphate (cpt-cAMP) or 1 microM PGE2 caused a slow suppression of the whole-cell IK by 34 and 36 %, respectively (measured after 20 min), without a shift in the voltage dependence of activation for this current. Neither of these agents altered the shape of the voltage-dependent inactivation curve indicating that the suppression of IK did not result from alterations in the inactivation properties. 3. To determine whether the PGE2-mediated suppression of IK depended on activation of the cAMP pathway, cells were exposed to this prostanoid in the presence of the protein kinase A (PKA) inhibitor, PKI. The PGE2-induced suppression of IK was prevented by PKI. In the absence of PGE2, PKI had no significant effect on the magnitude of IK. 4. Results obtained from protocols using different conditioning prepulse voltages indicated that the extent of cpt-cAMP- and PGE2-mediated suppression of IK was independent of the prepulse voltage. The subtraction of control and treated currents revealed that the cpt-cAMP- and PGE2-sensitive currents exhibited little time-dependent inactivation. Taken together, these results suggest that the modulated currents may be delayed rectifier-like IK. 5. Exposure to the inhibitors of IK, tetraethylammonium (TEA) or 4-aminopyridine (4-AP), reduced the control current elicited by a voltage step to +60 mV by 40-50 %. In the presence of 10 mM TEA, treatment with cpt-cAMP did not result in any further inhibition of IK. In contrast, cpt-cAMP reduced IK by an additional 25-30 % in the presence of 1 mM 4-AP. This effect was independent of the conditioning prepulse voltage. 6. These results establish that PGE2 inhibits an outward IK in sensory neurones via activation of PKA and are consistent with the idea that the PGE2-mediated sensitization of sensory neurones results, in part, from an inhibition of delayed rectifier-like IK.


Assuntos
AMP Cíclico/fisiologia , Dinoprostona/farmacologia , Neurônios Aferentes/efeitos dos fármacos , Bloqueadores dos Canais de Potássio , Transdução de Sinais/fisiologia , 4-Aminopiridina/farmacologia , Animais , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Dinoprostona/antagonistas & inibidores , Eletrofisiologia , Inibidores Enzimáticos/farmacologia , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Ratos , Tetraetilamônio/farmacologia , Tionucleotídeos/farmacologia
17.
J Pharmacol Exp Ther ; 287(3): 1119-27, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9864301

RESUMO

[3H]5-HT revealed the presence of serotonin receptors in cultured rat sensory neurons. [3H]5-CT binding was inhibited by cyanopindolol with an IC50 of 0.87 +/- 0.30 nM, suggesting the expression of the 5-HT1B receptor in these neurons. The presence of 5-HT1B receptors was confirmed by the displacement of [125I]Iodocyanopindolol binding by cyanopindolol with an IC50 of 2.43 +/- 0.81 nM. 5-HT1B receptors are the predominant type of serotonin receptors labeled by [3H]5-HT in cultured DRG neurons, representing approximately 60% of the specific [3H]5-HT binding sites. In addition, 5-HT1D and 5-HT2A receptor binding was also found in these neurons. RT-PCR analysis of RNA isolated from embryonic sensory neurons in culture confirmed the expression of 5-HT1B, 5-HT1D and 5-HT2A receptor mRNA. It also demonstrated the presence of 5-HT1F, 5-HT2C, 5-HT3, 5-HT4, 5-HT5A and 5-HT5B receptor mRNA and the absence of 5-HT1A, 5-HT1E, 5-HT2B, 5-HT6 and 5-HT7 mRNA. The identification of multiple subtypes of serotonin receptors expressed in cultured embryonic sensory neurons suggests that DRG neuronal cultures may be an excellent model to examine the direct effects of serotonin on the activity of these sensory neurons.


Assuntos
Gânglios Espinais/metabolismo , Neurônios Aferentes/metabolismo , Receptores de Serotonina/biossíntese , Serotonina/metabolismo , Animais , Southern Blotting , Células Cultivadas , Isoformas de Proteínas/biossíntese , RNA Mensageiro/biossíntese , RNA Mensageiro/isolamento & purificação , Ensaio Radioligante , Ratos , Receptores de Serotonina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serotonina/análogos & derivados , Serotonina/farmacologia , Antagonistas da Serotonina/farmacologia
18.
Mol Pharmacol ; 51(4): 666-73, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9106633

RESUMO

To determine whether opioid receptors in sensory neurons are regulated by chronic exposure to opioids, we assessed the binding of various opioid ligands to membranes derived from isolated rat dorsal root ganglia neurons grown in culture. Equilibrium binding of [3H]diprenorphine onto membranes from cells grown for 13-15 days revealed a saturable binding site with a Kd value of 0.3 +/- 0.2 nM and an approximate Bmax value of 1300 +/- 200 fmol/mg of protein. [3H]Diprenorphine binding increased 3-fold from 1-15 days in culture. The mu receptors represent approximately 70 +/- 11% of the [3H]diprenorphine binding sites, as indicated by saturation binding of [3H]DAMGO. The kappa and delta receptors represent approximately 10 +/- 3% and approximately 5 +/- 2% of the [3H]diprenorphine binding sites, respectively. Preexposure of neurons to 10 microM naloxone for 48 hr up-regulated the receptors by 40%, whereas incubation with 100 nM to 10 microM DAMGO for 48 hr resulted in a significant decrease in the Bmax value of opioid receptors, with a maximum reduction of 70%. The identification of a high level of opioid receptors expressed in isolated sensory neurons and their modulation by opioids demonstrates that cultured sensory neurons are an excellent model with which to study opioid receptor regulation.


Assuntos
Neurônios Aferentes/ultraestrutura , Receptores Opioides/metabolismo , Analgésicos/farmacologia , Animais , Células Cultivadas , Diprenorfina/metabolismo , Regulação para Baixo/efeitos dos fármacos , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Encefalinas/farmacologia , Feminino , Cinética , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/metabolismo , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores Opioides/classificação , Receptores Opioides/efeitos dos fármacos , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/efeitos dos fármacos , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/metabolismo , Trítio , Regulação para Cima/efeitos dos fármacos
19.
J Neurophysiol ; 77(1): 167-76, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9120557

RESUMO

The cellular mechanisms giving rise to the enhanced excitability induced by prostaglandin E2 (PGE2) and carba prostacyclin (CPGI2) in embryonic rat sensory neurons were investigated using the whole cell patch-clamp recording technique. Exposing sensory neurons to 1 microM PGE2 produced a twofold increase in the number of action potentials elicited by a ramp of depolarizing current, but this eicosanoid had no effect on the resting membrane potential or the amplitude of the slow afterhyperpolarization. Characterization of the outward potassium currents in the embryonic sensory neurons indicated that the composition of the total current was variable among these neurons. A steady-state inactivation protocol was used to determine the extent of residual noninactivating current. A conditioning prepulse to +20 mV demonstrated that some of these neurons exhibited only a sustained potassium current with little steady-state inactivation whereas other exhibited some combination of a sustained as well as a rapidly inactivating IA-type current. Treatment with 1 microM PGE2 or 1 microM CPGI2, but not 1 microM prostaglandin F2 alpha (PGF2 alpha) produced a time-dependent suppression of the total potassium current. After a 20-min exposure, PGE2 and CPGI2 inhibited the maximal current obtained at +60 mV by 48 and 40%, respectively. The prostaglandin-induced suppression of the potassium current was not associated with a shift in the voltage dependence for activation. Subtraction of the currents remaining after PGE2 or CPGI2 treatment from their respective control recordings revealed that the prostaglandin-sensitive current had characteristics that were consistent with a sustained-type of potassium current. This idea is supported by the following observation. The steady-state inactivation protocol revealed that for prepulse voltages activating both rapidly inactivating and sustained currents, the relaxation of the current was accelerated after treatment with PGE2 or CPGI2 suggesting the removal of a slower component. This effect was not observed in neurons exhibiting only the sustained type current. These results suggest that pro-inflammatory prostaglandins enhance the excitability of rat sensory neurons, in part, through the suppression of an outward potassium current that may modulate the firing threshold for generation of the action potential.


Assuntos
Neurônios Aferentes/metabolismo , Canais de Potássio/metabolismo , Prostaglandinas/farmacologia , Animais , Células Cultivadas , Dinoprosta/farmacologia , Dinoprostona/farmacologia , Eletrofisiologia , Epoprostenol/análogos & derivados , Epoprostenol/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Neurônios Aferentes/efeitos dos fármacos , Técnicas de Patch-Clamp , Canais de Potássio/efeitos dos fármacos , Ratos
20.
J Neurochem ; 67(1): 72-80, 1996 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8667028

RESUMO

To determine whether protein kinase C (PKC) mediates release of peptides from sensory neurons, we examined the effects of altering PKC activity on resting and evoked release of substance P (SP) and calcitonin gene-related peptide (CGRP). Exposing rat sensory neurons in culture to 10 or 50 nM phorbol 12,13-dibutyrate (PDBu) significantly increased SP and CGRP release at least 10-fold above resting levels, whereas the inactive 4alpha-PDBu analogue at 100 nM had no effect on release. Furthermore, 100 nM bradykinin increased peptide release approximately fivefold. Down-regulation of PKC significantly attenuated the release of peptides evoked by either PDBu or bradykinin. PDBu at 1 nM or 1-oleoyl-2-acetyl-sn-glycerol at 50 microM did not alter resting release of peptides, but augmented potassium- and capsaicin-stimulated release of both SP and CGRP approximately twofold. This sensitizing action of PKC activators on peptide release was significantly reduced by PKC down-regulation or by pretreating cultures with 10 nM staurosporine. These results establish that activation of PKC is important in the regulation of peptide release from sensory neurons. The PKC-induced enhancement of peptide release may be a mechanism underlying the neuronal sensitization that produces hyperalgesia.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Neurônios Aferentes/enzimologia , Proteína Quinase C/metabolismo , Substância P/metabolismo , Animais , Bradicinina/farmacologia , Capsaicina/farmacologia , Carcinógenos/farmacologia , Células Cultivadas/enzimologia , Regulação para Baixo , Ativação Enzimática , Feminino , Camundongos , Neurônios Aferentes/citologia , Neuropeptídeos/metabolismo , Dibutirato de 12,13-Forbol/farmacologia , Potássio/farmacologia , Gravidez , Ratos , Ratos Sprague-Dawley , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...