Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 38(13): 110588, 2022 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-35354022

RESUMO

This review highlights recent findings of different amplitude ranges, roles, and modulations of A-type K+ currents (IA) in excitatory (GAD67-GFP-) and inhibitory (GAD67-GFP+) interneurons in mouse spinal cord pain pathways. Endogenous neuropeptides, such as TAFA4, oxytocin, and dynorphin in particular, have been reported to modulate IA in these pain pathways, but only TAFA4 has been shown to fully reverse the opposing modulations that occur selectively in LIIo GAD67-GFP- and LIIi GAD67-GFP+ interneurons following both neuropathic and inflammatory pain. If, as hypothesized here, Kv4 subunits underlie IA in both GAD67-GFP- and GAD67-GFP+ interneurons, then IA diversity in spinal cord pain pathways may depend on the interneuron-subtype-selective expression of Kv4 auxiliary subunits with functionally different N-terminal variants. Thus, IA emerges as a good candidate for explaining the mechanisms underlying injury-induced mechanical hypersensitivity.


Assuntos
Dor , Medula Espinal , Animais , Citocinas/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Interneurônios/metabolismo , Camundongos , Dor/metabolismo , Medula Espinal/metabolismo
2.
Cell Rep ; 37(4): 109884, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34706225

RESUMO

Pain, whether acute or persistent, is a serious medical problem worldwide. However, its management remains unsatisfactory, and new analgesic molecules are required. We show here that TAFA4 reverses inflammatory, postoperative, and spared nerve injury (SNI)-induced mechanical hypersensitivity in male and female mice. TAFA4 requires functional low-density lipoprotein receptor-related proteins (LRPs) because their inhibition by RAP (receptor-associated protein) dose-dependently abolishes its antihypersensitive actions. SNI selectively decreases A-type K+ current (IA) in spinal lamina II outer excitatory interneurons (L-IIo ExINs) and induces a concomitant increase in IA and decrease in hyperpolarization-activated current (Ih) in lamina II inner inhibitory interneurons (L-IIi InhINs). Remarkably, SNI-induced ion current alterations in both IN subtypes were rescued by TAFA4 in an LRP-dependent manner. We provide insights into the mechanism by which TAFA4 reverses injury-induced mechanical hypersensitivity by restoring normal spinal neuron activity and highlight the considerable potential of TAFA4 as a treatment for injury-induced mechanical pain.


Assuntos
Citocinas/metabolismo , Hiperalgesia/metabolismo , Dor/metabolismo , Potássio/metabolismo , Receptores de LDL/metabolismo , Corno Dorsal da Medula Espinal/metabolismo , Animais , Células CHO , Cricetulus , Células HEK293 , Humanos , Camundongos , Células RAW 264.7
3.
Front Cell Neurosci ; 11: 6, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28184187

RESUMO

The axon initial segment (AIS) is a highly specialized neuronal compartment that plays a key role in neuronal development and excitability. It concentrates multiple membrane proteins such as ion channels and cell adhesion molecules (CAMs) that are recruited to the AIS by the scaffold protein ankyrin G (ankG). The crucial function of ankG in the anchoring of AIS membrane components is well established, but a reciprocal role of membrane partners in ankG targeting and stabilization remained elusive. In rat cultured hippocampal neurons and cortical organotypic slices, we found that shRNA-mediated knockdown of ankG membrane partners (voltage-gated sodium channels (Nav) or neurofascin-186) led to a decrease of ankG concentration and perturbed the AIS formation and maintenance. These effects were rescued by expressing a recombinant AIS-targeted Nav or by a minimal construct containing the ankyrin-binding domain of Nav1.2 and a membrane anchor (mABD). Moreover, overexpressing mABD in mature neurons led to ankG mislocalization. Altogether, these results demonstrate that a tight and precocious association of ankG with its membrane partners is a key step for the establishment and maintenance of the AIS.

4.
Pflugers Arch ; 467(4): 789-803, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24872163

RESUMO

In the nucleus of the tractus solitarii (NTS), a large proportion of neurones express transient A-type potassium currents (I KA) having deep influence on the fidelity of the synaptic transmission of the visceral primary afferent inputs to second-order neurones. Up to now, the strong impact of I KA within the NTS was considered to result exclusively from its variation in amplitude, and its molecular correlate(s) remained unknown. In order to identify which Kv channels underlie I KA in NTS neurones, the gating properties and the pharmacology of this current were determined using whole cell patch clamp recordings in slices. Complementary information was brought by immunohistochemistry. Strikingly, two neurone subpopulations characterized by fast or slow inactivation time courses (respectively about 50 and 200 ms) were discriminated. Both characteristics matched those of the Kv4 channel subfamily. The other gating properties, also matching the Kv4 channel ones, were homogeneous through the NTS. The activation and inactivation occurred at membrane potentials around the threshold for generating action potentials, and the time course of recovery from inactivation was rapid. Pharmacologically, I KA in NTS neurones was found to be resistant to tetraethylammonium (TEA), sea anemone toxin blood-depressing substance (BDS) and dendrotoxin (DTX), whereas Androctonus mauretanicus mauretanicus toxin 3 (AmmTX3), a scorpion toxin of the α-KTX 15 family that has been shown to block all the members of the Kv4 family, inhibited 80 % of I KA irrespectively of its inactivation time course. Finally, immunohistochemistry data suggested that, among the Kv4 channel subfamily, Kv4.3 is the prevalent subunit expressed in the NTS.


Assuntos
Ativação do Canal Iônico , Canais de Potássio Shal/metabolismo , Núcleo Solitário/metabolismo , Potenciais de Ação , Animais , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Wistar , Núcleo Solitário/citologia , Núcleo Solitário/fisiologia
5.
Front Neurosci ; 7: 58, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23596386

RESUMO

The colonic migrating motor complex (CMMC) is a major pattern of motility that is entirely generated and organized by the enteric nervous system. We have previously demonstrated that the Nav1.9 channel underlies a tetrodotoxin-resistant sodium current which modulates the excitability of enteric neurons. The aim of this study was to observe the effect of loss of the Nav1.9 channel in enteric neurons on mouse colonic motility in vitro. The mechanical activity of the circular muscle was simultaneously recorded from three sites, namely, proximal, mid- and distal, along the whole colon of male, age-matched wild-type and Nav1.9 null mice. Spontaneous CMMCs were observed in all preparations. The mean frequency of CMMCs was significantly higher in the Nav1.9 null mice (one every 2.87 ± 0.1 min compared to one every 3.96 ± 0.23 min in the wild type). The mean duration of CMMCs was shorter and the mean area-under-contraction was larger in the Nav1.9 null mice compared to the wild type. In addition, CMMCs propagated preferentially in an aboral direction in the Nav1.9 null mice. Our study demonstrates that CMMCs do occur in mice lacking the Nav1.9 channel, but their characteristics are significantly different from controls. Up to now, the Nav1.9 channel was mainly associated with nociceptive neurons and involved in their hyperexcitability after inflammation. Our result shows for the first time a role for the Nav1.9 channel in a complex colonic motor pattern.

6.
J Physiol ; 587(Pt 7): 1461-79, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19204045

RESUMO

The intrinsic primary afferent neurons (IPANs) of the guinea pig enteric nervous system express Na(v)1.9 sodium channels that produce a persistent TTX-resistant current having a low activation threshold and slow gating kinetics. These neurons receive slow EPSPs induced mainly by the activation of neurokinin 3 receptors (NK3r). Here, we demonstrate that senktide, a specific NK3r agonist, potentiates the Na(v)1.9 current (I(Nav1.9)) in IPANs. Using whole-cell patch-clamp recordings from IPANs in duodenum longitudinal muscle/myenteric plexus preparations, we show that short (1-5 s) and long (up to 1 min) applications of senktide, increase the I(Nav1.9) peak current up to 13-fold. The effect, blocked by a NK3r antagonist SB235375 is transient, lasting approximately 2 min and is due to a negative shift of the activation voltage by approximately 20 mV and of fast inactivation by approximately 10 mV. As a consequence, the window current resulting from the product of the activation and fast inactivation curves is shifted and enlarged. The transient effect of senktide is likely to be due to the fast desensitization of NK3r. Protein kinase C (PKC) activation with phorbol or oleoyl acetylglycerol also increases I(Nav1.9), although persistently, by inducing similar voltage-dependent changes. Current-clamp experiments showed that I(Nav1.9) modulation by senktide lowers action potential threshold and increases excitability. The increase in I(Nav1.9) by NK3r activation is also likely to amplify slow EPSPs generated in the IPANs. These changes in excitability potentially have a profound effect on the entire enteric synaptic circuit and ultimately on gut motility and secretion.


Assuntos
Duodeno/inervação , Sistema Nervoso Entérico/metabolismo , Músculo Liso/inervação , Receptores da Neurocinina-3/metabolismo , Células Receptoras Sensoriais/metabolismo , Canais de Sódio/metabolismo , Sódio/metabolismo , Acetatos/farmacologia , Animais , Diglicerídeos/farmacologia , Sistema Nervoso Entérico/efeitos dos fármacos , Sistema Nervoso Entérico/enzimologia , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Potenciais Pós-Sinápticos Excitadores , Motilidade Gastrointestinal , Cobaias , Técnicas In Vitro , Ativação do Canal Iônico , Cinética , Técnicas de Patch-Clamp , Fragmentos de Peptídeos/farmacologia , Dibutirato de 12,13-Forbol/farmacologia , Proteína Quinase C/metabolismo , Quinolinas/farmacologia , Receptores da Neurocinina-3/efeitos dos fármacos , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/enzimologia , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/efeitos dos fármacos , Substância P/análogos & derivados , Substância P/farmacologia , Tetrodotoxina/farmacologia
7.
J Gen Physiol ; 131(3): 211-25, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18270172

RESUMO

Altered function of Na+ channels is responsible for increased hyperexcitability of primary afferent neurons that may underlie pathological pain states. Recent evidence suggests that the Nav1.9 subunit is implicated in inflammatory but not acute pain. However, the contribution of Nav1.9 channels to the cellular events underlying nociceptor hyperexcitability is still unknown, and there remains much uncertainty as to the biophysical properties of Nav1.9 current and its modulation by inflammatory mediators. Here, we use gene targeting strategy and computer modeling to identify Nav1.9 channel current signature and its impact on nociceptors' firing patterns. Recordings using internal fluoride in small DRG neurons from wild-type and Nav1.9-null mutant mice demonstrated that Nav1.9 subunits carry the TTX-resistant "persistent" Na+ current called NaN. Nav1.9(-/-) nociceptors showed no significant change in the properties of the slowly inactivating TTX-resistant SNS/Nav1.8 current. The loss in Nav1.9-mediated Na+ currents was associated with the inability of small DRG neurons to generate a large variety of electrophysiological behaviors, including subthreshold regenerative depolarizations, plateau potentials, active hyperpolarizing responses, oscillatory bursting discharges, and bistable membrane behaviors. We further investigated, using CsCl- and KCl-based pipette solutions, whether G-protein signaling pathways and inflammatory mediators upregulate the NaN/Nav1.9 current. Bradykinin, ATP, histamine, prostaglandin-E2, and norepinephrine, applied separately at maximal concentrations, all failed to modulate the Nav1.9 current. However, when applied conjointly as a soup of inflammatory mediators they rapidly potentiated Nav1.9 channel activity, generating subthreshold amplification and increased excitability. We conclude that Nav1.9 channel, the molecular correlate of the NaN current, is potentiated by the concerted action of inflammatory mediators that may contribute to nociceptors' hyperexcitability during peripheral inflammation.


Assuntos
Neuropeptídeos/metabolismo , Nociceptores/metabolismo , Canais de Sódio/metabolismo , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Bradicinina/metabolismo , Bradicinina/farmacologia , Dinoprostona/metabolismo , Dinoprostona/farmacologia , Relação Dose-Resposta a Droga , Eletrofisiologia , Gânglios Espinais/citologia , Regulação da Expressão Gênica , Histamina/metabolismo , Histamina/farmacologia , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.9 , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuropeptídeos/genética , Norepinefrina/metabolismo , Norepinefrina/farmacologia , Canais de Sódio/genética , Tetrodotoxina/metabolismo , Tetrodotoxina/farmacologia , Regulação para Cima
8.
Mol Cell Neurosci ; 35(1): 138-52, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17363266

RESUMO

The Nav1.9 sodium channel is expressed in nociceptive DRG neurons where it contributes to spontaneous pain behavior after peripheral inflammation. Here, we used a newly developed antibody to investigate the distribution of Nav1.9 in rat and mouse trigeminal ganglion (TG) nerve endings and in enteric nervous system (ENS). In TGs, Nav1.9 was expressed in the soma of small- and medium-sized, peripherin-positive neurons. Nav1.9 was present along trigeminal afferent fibers and at terminals in lip skin and dental pulp. In the ENS, Nav1.9 was detected within the soma and proximal axons of sensory, Dogiel type II, myenteric and submucosal neurons. Immunological data were correlated with the detection of persistent TTX-resistant Na(+) currents sharing similar properties in DRG, TG and myenteric neurons. Collectively, our data support a potential role of Nav1.9 in the transmission of trigeminal pain and the regulation of intestinal reflexes. Nav1.9 might therefore constitute a molecular target for therapeutic treatments of orofacial pain and gastrointestinal syndromes.


Assuntos
Dor Facial/fisiopatologia , Plexo Mientérico/citologia , Neurônios Aferentes/fisiologia , Neuropeptídeos/genética , Canais de Sódio/genética , Plexo Submucoso/citologia , Gânglio Trigeminal/citologia , Sequência de Aminoácidos , Animais , Axônios/fisiologia , Polpa Dentária/inervação , Dor Facial/metabolismo , Lábio/inervação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Canal de Sódio Disparado por Voltagem NAV1.9 , Neurônios Aferentes/ultraestrutura , Neuropeptídeos/química , Neuropeptídeos/metabolismo , Nociceptores/fisiologia , Técnicas de Patch-Clamp , Estrutura Terciária de Proteína , Ratos , Ratos Wistar , Reflexo/fisiologia , Pele/inervação , Canais de Sódio/química , Canais de Sódio/metabolismo
9.
Histochem Cell Biol ; 126(5): 537-48, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16733665

RESUMO

In many organs, different protein kinase C (PKC) isoforms are expressed in specific cell types, suggesting that the different PKCs have cell-specific roles, and also that drugs acting on a particular PKC may have effects on the whole organ that are distinguishable from drugs that target other isoforms. Previous studies of the guinea-pig and mouse intestine indicate that there are cell-specific expressions of PKC isoforms in neurons, muscle and the interstitial cells of Cajal. In the present study we have investigated the expression of different PKCs in human intestine. Immunohistochemical studies showed that the forms that are prominent in human enteric neurons are PKCs gamma and epsilon and in muscle the dominant form is PKCdelta. Neurons were weakly stained for PKCbetaI. These observations parallel findings in guinea-pig and mouse, except that in human PKCgamma-IR was not present in the same types of neurons that express it in the guinea-pig. Enteric glial cells were strongly immunoreactive for PKCalpha, which is also the major isoform in enteric glial cells of guinea-pig. In human and guinea-pig, glial cells also express PKCbetaI. Spindle-shaped cells in the mucosa were immunoreactive for PKCalpha and PKCgamma and in the muscle layers similar cells had PKCgamma-IR and PKCtheta-IR. The spindle-shaped cells were similar in morphology to interstitial cells of Cajal. Western analysis and RT-PCR confirmed the presence of the PKC isoform proteins and mRNA in the tissue. We conclude that there is cell-type specific expression of different PKCs in enteric neurons and intestinal muscle in human tissue, and that there are strong similarities in patterns of expression between laboratory animals and human, but some clear differences are also observed.


Assuntos
Intestinos/enzimologia , Neurônios/enzimologia , Proteína Quinase C/biossíntese , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Western Blotting , Criança , Pré-Escolar , Feminino , Humanos , Imuno-Histoquímica , Lactente , Intestinos/citologia , Isoenzimas/biossíntese , Isoenzimas/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Músculo Liso/citologia , Músculo Liso/enzimologia , Plexo Mientérico/enzimologia , Proteína Quinase C/imunologia , Proteína Quinase C beta , Coelhos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Auton Neurosci ; 115(1-2): 1-6, 2004 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-15507400

RESUMO

We have investigated the effects of protein kinase inhibitors on the sustained slow postsynaptic excitation (SSPE) that is evoked by prolonged stimulation of synaptic inputs to intrinsic primary afferent neurons (IPANs) in the small intestines of guinea pigs. Stimulation of synaptic inputs to the IPANs caused depolarisation, increased input resistance, and increased excitation that continued after the cessation of stimulation. The excitation was substantially reduced by the broad-spectrum kinase inhibitor staurosporine (1 microM), PKC inhibitors Ro 31-8220 (3.3 microM) and calphostin C (1 microM), but not by the PKA inhibitor H89 (1 microM). At a higher concentration, 10 microM Ro 31-8220 reduced the excitability of axons to electrical stimulation. Phorbol dibutyrate (1 microM) caused excitability increases, membrane depolarisation, and increased input resistance that mimicked the SSPE. We conclude that the generation of the SSPE requires a phosphorylation step that is mediated by protein kinase C.


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , Íleo/enzimologia , Potenciação de Longa Duração/fisiologia , Neurônios Aferentes/enzimologia , Proteínas Quinases/fisiologia , Animais , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Cobaias , Íleo/efeitos dos fármacos , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Neurônios Aferentes/efeitos dos fármacos
11.
Prog Neurobiol ; 72(2): 143-64, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15063530

RESUMO

Intrinsic primary afferent neurons (IPANs) of the enteric nervous system are quite different from all other peripheral neurons. The IPANs are transducers of physiological stimuli, including movement of the villi or distortion of the mucosa, contraction of intestinal muscle and changes in the chemistry of the contents of the gut lumen. They are the first neurons in intrinsic reflexes that influence the patterns of motility, secretion of fluid across the mucosal epithelium and local blood flow in the small and large intestines. In the guinea pig small intestine, where they have been characterized in detail, IPANs have Dogiel type II morphology, that is they are large round or oval neurons with multiple processes, some of which end close to the luminal surface of the intestine, and some of which form synapses with enteric interneurons, motor neurons and with other IPANs. The IPANs have well-defined ionic currents through which their excitability, and their functions in enteric nerve circuits, is determined. These include voltage-gated Na(+) and Ca(2+) currents, a long lasting calcium-activated K(+) current, and a hyperpolarization-activated cationic current. The IPANs exhibit long-term changes in their states of excitation that can be induced by extended periods of low frequency activity in synaptic inputs and by inflammatory mediators, either applied directly or released during an inflammatory challenge. The IPANs may be involved in pathological changes in enteric function following inflammation.


Assuntos
Sistema Nervoso Entérico/fisiologia , Intestinos/inervação , Vias Neurais/fisiologia , Neurônios Aferentes/citologia , Neurônios Aferentes/fisiologia , Animais , Sistema Nervoso Entérico/citologia , Humanos , Intestinos/citologia , Vias Neurais/citologia , Sinapses/fisiologia
12.
Pflugers Arch ; 447(3): 298-304, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14556073

RESUMO

Low-frequency stimulation of synaptic inputs to after-hyperpolarising (AH) neurons in the guinea-pig small intestine causes sustained increases in excitability that far outlast the stimulus period. This excitation has been called sustained, slow, post-synaptic excitation (SSPE). Intracellular microelectrodes were used to record the effects of the protein kinase C (PKC) stimulant, phorbol dibutyrate (PDBu), and compare these with changes seen during the SSPE, in AH neurons of the small intestine of the guinea-pig. PDBu (1 nM-1 microM) increased excitability, depolarised the membrane and increased input resistance concentration dependently, mimicking the effects of low-frequency stimulation of pre-synaptic inputs. These changes developed slowly after the start of infusion and were only slowly reversible after wash out. PDBu suppressed a late after-hyperpolarising potential (AHP) that depends on Ca2+ entry via voltage-gated Ca2+ channels during the action potential. The effects of PDBu (10 nM) on the late AHP were indistinguishable from those observed during the SSPE. PDBu, at a concentration that inhibited the AHP, had no effect on the action potential half-width or the slope of its first repolarisation phase (the early phase of repolarisation is slowed by the Ca2+ influx of the action potential). Thus PDBu inhibited K+ channel opening underlying the late AHP, but did not suppress Ca2+ entry during the action potential. The hyperpolarisation-activated cation current (Ih) in intrinsic primary afferent neurons (IPANs) was not affected by PDBu. We conclude that PDBu mimics the sustained excitation caused by low-frequency stimulation of synaptic inputs to IPANs by closing IK channels responsible for the AHP or restricting their opening by Ca2+ and by reducing the current carried by K+ channels that are active at rest. IK channels, the opening of which results in the AHP, have consensus sites for PKC and are likely targets for phosphorylation during the SSPE.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Plexo Mientérico/efeitos dos fármacos , Dibutirato de 12,13-Forbol/farmacologia , Sinapses/fisiologia , Potenciais de Ação/fisiologia , Animais , Relação Dose-Resposta a Droga , Estimulação Elétrica/métodos , Cobaias , Plexo Mientérico/fisiologia
13.
J Neurosci ; 23(7): 2715-25, 2003 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-12684457

RESUMO

Voltage-gated Na(+) currents play critical roles in shaping electrogenesis in neurons. Here, we have identified a TTX-resistant Na(+) current (TTX-R I(Na)) in duodenum myenteric neurons of guinea pig and rat and have sought evidence regarding the molecular identity of the channel producing this current from the expression of Na(+) channel alpha subunits and the biophysical and pharmacological properties of TTX-R I(Na). Whole-cell patch-clamp recording from in situ neurons revealed the presence of a voltage-gated Na(+) current that was highly resistant to TTX (IC(50), approximately 200 microm) and selectively distributed in myenteric sensory neurons but not in interneurons and motor neurons. TTX-R I(Na) activated slowly in response to depolarization and exhibited a threshold for activation at -50 mV. V(1/2) values of activation and steady-state inactivation were -32 and -31 mV in the absence of fluoride, respectively, which, as predicted from the window current, generated persistent currents. TTX-R I(Na) also had prominent ultraslow inactivation, which turns off 50% of the conductance at rest (-60 mV). Substituting CsF for CsCl in the intracellular solution shifted the voltage-dependent parameters of TTX-R I(Na) leftward by approximately 20 mV. Under these conditions, TTX-R I(Na) had voltage-dependent properties similar to those reported previously for NaN/Na(V)1.9 in dorsal root ganglion neurons. Consistent with this, reverse transcription-PCR, single-cell profiling, and immunostaining experiments indicated that Na(V)1.9 transcripts and subunits, but not Na(V)1.8, were expressed in the enteric nervous system and restricted to myenteric sensory neurons. TTX-R I(Na) may play an important role in regulating subthreshold electrogenesis and boosting synaptic stimuli, thereby conferring distinct integrative properties to myenteric sensory neurons.


Assuntos
Plexo Mientérico/citologia , Neurônios Aferentes/metabolismo , Neurônios Aferentes/fisiologia , Neuropeptídeos/metabolismo , Canais de Sódio/metabolismo , Tetrodotoxina/farmacologia , Potenciais de Ação , Sequência de Aminoácidos , Animais , Cádmio/farmacologia , Células Cultivadas , Condutividade Elétrica , Cobaias , Imuno-Histoquímica , Cinética , Dados de Sequência Molecular , Canal de Sódio Disparado por Voltagem NAV1.9 , Neurônios Aferentes/efeitos dos fármacos , Neuropeptídeos/genética , Neuropeptídeos/fisiologia , Técnicas de Patch-Clamp , Subunidades Proteicas , RNA Mensageiro/análise , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Canais de Sódio/genética , Canais de Sódio/fisiologia , Transcrição Gênica
14.
Curr Opin Pharmacol ; 2(6): 657-64, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12482727

RESUMO

Recent studies have shown that intrinsic primary afferent neurons (IPANs) express a much larger range of ionic currents than non-sensory neurons of the enteric nervous system. These ionic currents can be modulated by neurotransmitters that are synaptically released onto the soma (unlike cranial and spinal sensory neurons). The membrane receptors and ionic channels that are involved in the sensory transduction processes of IPANS are beginning to be defined. IPANS can move between a large range of excitability states that are influenced by neurotransmitters and hormones. An additional cause of variability in excitability is the actions of inflammatory mediators. It is becoming apparent that the variation in excitability of IPANS might play a critical role in determining the physiological state of the intestine.


Assuntos
Sistema Digestório/inervação , Gastroenteropatias/tratamento farmacológico , Gastroenteropatias/metabolismo , Neurônios Aferentes/fisiologia , Animais , Sistema Digestório/efeitos dos fármacos , Sistema Digestório/metabolismo , Condutividade Elétrica , Gastroenteropatias/fisiopatologia , Humanos , Inflamação/metabolismo , Canais Iônicos/metabolismo , Sinapses/metabolismo
15.
J Physiol ; 538(Pt 2): 447-63, 2002 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11790812

RESUMO

Whole-cell patch-clamp recordings taken from guinea-pig duodenal myenteric neurones within intact ganglia were used to determine the properties of S and AH neurones. Major currents that determine the states of AH neurones were identified and quantified. S neurones had resting potentials of -47 +/- 6 mV and input resistances (R(in)) of 713 +/- 49 MOmega at voltages ranging from -90 to -40 mV. At more negative levels, activation of a time-independent, caesium-sensitive, inward-rectifier current (I(Kir)) decreased R(in) to 103 +/- 10 MOmega. AH neurones had resting potentials of -57 +/- 4 mV and R(in) was 502 +/- 27 MOmega. R(in) fell to 194 +/- 16 MOmega upon hyperpolarization. This decrease was attributable mainly to the activation of a cationic h current, I(h), and to I(Kir). Resting potential and R(in) exhibited a low sensitivity to changes in [K(+)](o) in both AH and S neurones. This indicates that both cells have a low background K(+) permeability. The cationic current, I(h), contributed about 20 % to the resting conductance of AH neurones. It had a half-activation voltage of -72 +/- 2 mV, and a voltage sensitivity of 8.2 +/- 0.7 mV per e-fold change. I(h) has relatively fast, voltage-dependent kinetics, with on and off time constants in the range of 50-350 ms. AH neurones had a previously undescribed, low threshold, slowly inactivating, sodium-dependent current that was poorly sensitive to TTX. In AH neurones, the post-action-potential slow hyperpolarizing current, I(AHP), displayed large variation from cell to cell. I(AHP) appeared to be highly Ca(2+) sensitive, since its activation with either membrane depolarization or caffeine (1 mM) was not prevented by perfusing the cell with 10 mM BAPTA. We determined the identity of the Ca(2+) channels linked to I(AHP). Action potentials of AH neurones that were elongated by TEA (10 mM) were similarly shortened and I(AHP) was suppressed with each of the three omega-conotoxins GVIA, MVIIA and MVIIC (0.3-0.5 microM), but not with omega-agatoxin IVA (0.2 microM). There was no additivity between the effects of the three conotoxins, which indicates the presence of N- but not of P/Q-type Ca(2+) channels. A residual Ca(2+) current, resistant to all toxins, but blocked by 0.5 mM Cd(2+), could not generate I(AHP). This patch-clamp study, performed on intact ganglia, demonstrates that the AH neurones of the guinea-pig duodenum are under the control of four major currents, I(AHP), I(h), an N-type Ca(2+) current and a slowly inactivating Na(+) current.


Assuntos
Gânglios/fisiologia , Plexo Mientérico/fisiologia , Neurônios/fisiologia , Potenciais de Ação , Animais , Cátions/metabolismo , Condutividade Elétrica , Estimulação Elétrica , Gânglios/citologia , Cobaias , Técnicas In Vitro , Modelos Neurológicos , Plexo Mientérico/citologia , Técnicas de Patch-Clamp , Tempo de Reação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...