Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Mol Pain ; 16: 1744806920969476, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33121353

RESUMEN

The endocannabinoid system (ECS) is known to modulate not only food intake but also pain, especially via the cannabinoid type 1 receptor (CB1R) expressed throughout the central nervous system and the peripheral tissues. Our previous study demonstrated that fasting produces an analgesic effect in adult male mice, which is reversed by intraperitoneal (i.p.) administration of CB1R antagonist (SR 141716). In the present study, we further examined the effect of CB1R expressed in the peripheral tissues. In the formalin-induced inflammatory pain model, i.p. administration of peripherally restricted CB1R antagonist (AM 6545) reversed fasting-induced analgesia. However, intraplantar administration of SR 141716 did not affect fasting-induced analgesia. Furthermore, mRNA expression of CB1R did not change in the formalin model by fasting in the dorsal root ganglia. The formalin-induced c-Fos expression at the spinal cord level was not affected by fasting, and in vivo recording from the superficial dorsal horn of the lumbar spinal cord revealed that fasting did not affect formalin-induced neural activity, which indicates minimal involvement of the spinal cord in fasting-induced analgesia. Finally, when we performed subdiaphragmatic vagotomy to block the hunger signal from the gastrointestinal (GI) system, AM 6545 did not affect fasting-induced analgesia, but SR 141716 still reversed fasting-induced analgesia. Taken together, our results suggest that both peripheral and central CB1Rs contribute to fasting-induced analgesic effects and the CB1Rs in the GI system which transmit fasting signals to the brain, rather than those in the peripheral sensory neurons, may contribute to fasting-induced analgesic effects.


Asunto(s)
Analgesia/métodos , Antagonistas de Receptores de Cannabinoides/farmacología , Ayuno/fisiología , Manejo del Dolor/métodos , Receptor Cannabinoide CB1/antagonistas & inhibidores , Rimonabant/farmacología , Animales , Modelos Animales de Enfermedad , Formaldehído/toxicidad , Ganglios Espinales/metabolismo , Tracto Gastrointestinal/fisiología , Inmunohistoquímica , Inflamación/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB1/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Vagotomía
2.
Korean J Physiol Pharmacol ; 22(2): 173-182, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29520170

RESUMEN

Recent studies have provided several lines of evidence that peripheral administration of oxytocin induces analgesia in human and rodents. However, the exact underlying mechanism of analgesia still remains elusive. In the present study, we aimed to identify which receptor could mediate the analgesic effect of intraperitoneal injection of oxytocin and its cellular mechanisms in thermal pain behavior. We found that oxytocin-induced analgesia could be reversed by d(CH2)5[Tyr(Me)2,Dab5] AVP, a vasopressin-1a (V1a) receptor antagonist, but not by desGly-NH2-d(CH2)5[DTyr2, Thr4]OVT, an oxytocin receptor antagonist. Single cell RT-PCR analysis revealed that V1a receptor, compared to oxytocin, vasopressin-1b and vasopressin-2 receptors, was more profoundly expressed in dorsal root ganglion (DRG) neurons and the expression of V1a receptor was predominant in transient receptor potential vanilloid 1 (TRPV1)-expressing DRG neurons. Fura-2 based calcium imaging experiments showed that capsaicin-induced calcium transient was significantly inhibited by oxytocin and that such inhibition was reversed by V1a receptor antagonist. Additionally, whole cell patch clamp recording demonstrated that oxytocin significantly increased potassium conductance via V1a receptor in DRG neurons. Taken together, our findings suggest that analgesic effects produced by peripheral administration of oxytocin were attributable to the activation of V1a receptor, resulting in reduction of TRPV1 activity and enhancement of potassium conductance in DRG neurons.

3.
Brain Struct Funct ; 229(3): 759-773, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38411929

RESUMEN

Malformation during cortical development can disrupt the balance of excitatory and inhibitory neural circuits, contributing to various psychiatric and developmental disorders. One of the critical factors of cortical neural networks is the fine regulation of neurogenesis through mechanical cues, such as shear stress and substrate stiffness. Piezo1, a mechanically-activated channel, serves as a transducer for these mechanical cues, regulating embryogenesis. However, specific cell-type expression patterns of this channel during cortical development have not yet been characterized. In the present study, we conducted an RNAscope experiment to visualize the location of Piezo1 transcripts with embryonic neuronal/glial lineage cell markers. Our analysis covered coronal sections of the mouse forebrain on embryonic day 12.5 (E12.5), E14.5, E16.5, and E18.5. In addition, applying Yoda1, a specific Piezo1 agonist, evoked distinct calcium elevation in piriform cortices of E16.5 and E18.5 embryonic slices. Furthermore, pharmacological activation or inhibition of this channel significantly modulated the migration of neurosphere-derived cells in vitro. These findings contribute valuable insights to the field of mechanobiology and provide an understanding of the intricate processes underlying embryonic brain development.


Asunto(s)
Canales Iónicos , Neurogénesis , Animales , Ratones , Canales Iónicos/genética , Canales Iónicos/metabolismo , Mecanotransducción Celular/fisiología , Neurogénesis/genética , Prosencéfalo/metabolismo
4.
Int J Oral Sci ; 15(1): 45, 2023 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-37749100

RESUMEN

Dental primary afferent (DPA) neurons and proprioceptive mesencephalic trigeminal nucleus (MTN) neurons, located in the trigeminal ganglion and the brainstem, respectively, are essential for controlling masticatory functions. Despite extensive transcriptomic studies on various somatosensory neurons, there is still a lack of knowledge about the molecular identities of these populations due to technical challenges in their circuit-validated isolation. Here, we employed high-depth single-cell RNA sequencing (scRNA-seq) in combination with retrograde tracing in mice to identify intrinsic transcriptional features of DPA and MTN neurons. Our transcriptome analysis revealed five major types of DPA neurons with cell type-specific gene enrichment, some of which exhibit unique mechano-nociceptive properties capable of transmitting nociception in response to innocuous mechanical stimuli in the teeth. Furthermore, we discovered cellular heterogeneity within MTN neurons that potentially contribute to their responsiveness to mechanical stretch in the masseter muscle spindles. Additionally, DPA and MTN neurons represented sensory compartments with distinct molecular profiles characterized by various ion channels, receptors, neuropeptides, and mechanoreceptors. Together, our study provides new biological insights regarding the highly specialized mechanosensory functions of DPA and MTN neurons in pain and proprioception.


Asunto(s)
Neuronas , Propiocepción , Animales , Ratones , Perfilación de la Expresión Génica , Dolor , Análisis de Secuencia de ARN
5.
Neurosci Bull ; 38(4): 373-385, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35294713

RESUMEN

Mitochondrial reactive oxygen species (mROS) that are overproduced by mitochondrial dysfunction are linked to pathological conditions including sensory abnormalities. Here, we explored whether mROS overproduction induces itch through transient receptor potential canonical 3 (TRPC3), which is sensitive to ROS. Intradermal injection of antimycin A (AA), a selective inhibitor of mitochondrial electron transport chain complex III for mROS overproduction, produced robust scratching behavior in naïve mice, which was suppressed by MitoTEMPO, a mitochondria-selective ROS scavenger, and Pyr10, a TRPC3-specific blocker, but not by blockers of TRPA1 or TRPV1. AA activated subsets of trigeminal ganglion neurons and also induced inward currents, which were blocked by MitoTEMPO and Pyr10. Besides, dry skin-induced chronic scratching was relieved by MitoTEMPO and Pyr10, and also by resveratrol, an antioxidant. Taken together, our results suggest that mROS elicit itch through TRPC3, which may underlie chronic itch, representing a potential therapeutic target for chronic itch.


Asunto(s)
Mitocondrias , Prurito , Animales , Antioxidantes/farmacología , Ratones , Prurito/inducido químicamente , Especies Reactivas de Oxígeno/metabolismo , Canal Catiónico TRPA1
6.
Exp Neurobiol ; 30(5): 329-340, 2021 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-34737238

RESUMEN

Pulpitis (toothache) is a painful inflammation of the dental pulp and is a prevalent problem throughout the world. This pulpal inflammation occurs in the cells inside the dental pulp, which have host defense mechanisms to combat oral microorganisms invading the pulp space of exposed teeth. This innate immunity has been well studied, with a focus on Toll-like receptors (TLRs). The function of TLR4, activated by Gram-negative bacteria, has been demonstrated in trigeminal ganglion (TG) neurons for dental pain. Although Gram-positive bacteria predominate in the teeth of patients with caries and pulpitis, the role of TLR2, which is activated by Gram-positive bacteria, is poorly understood in dental primary afferent (DPA) neurons that densely innervate the dental pulp. Using Fura-2 based Ca2+ imaging, we observed reproducible intracellular Ca2+ responses induced by Pam3CSK4 and Pam2CSK4 (TLR2-specific agonists) in TG neurons of adult wild-type (WT) mice. The response was completely abolished in TLR2 knock-out (KO) mice. Single-cell RT-PCR detected Tlr2 mRNA in DPA neurons labeled with fluorescent retrograde tracers from the upper molars. Using the mouse pulpitis model, real-time RT-PCR revealed that Tlr2 and inflammatory-related molecules were upregulated in injured TG, compared to non-injured TG, from WT mice, but not from TLR2 KO mice. TLR2 protein expression was also upregulated in injured DPA neurons, and the change was corresponded with a significant increase in calcitonin gene-related peptide (CGRP) expression. Our results provide a better molecular understanding of pulpitis by revealing the potential contribution of TLR2 to pulpal inflammatory pain.

7.
Eur J Pharmacol ; 854: 320-327, 2019 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-31009638

RESUMEN

Alpha 2 (α2-) adrenoceptor agonists, such as clonidine or dexmedetomidine, have been found to inhibit hyperpolarization-activated, cyclic nucleotide-modulated (HCN) channels, not only by reducing intracellular cyclic AMP levels but also by directly blocking HCN channels. In this study, we examined the inhibitory effect of guanabenz, a centrally acting α2-adrenoceptor agonist with high specificity for α2A-subtype, on HCN channels in mesencephalic trigeminal nucleus (MTN) neurons which robustly express HCN channels and have been suggested to coexpress α2A-adrenoceptors. By performing whole-cell patch-clamp recording on MTN neurons in brainstem slices, hyperpolarization-activated inward current (Ih) was examined during guanabenz treatment. Guanabenz inhibited Ih in a dose-dependent manner, which was likely to be ZD7288-sensitive HCN current as it did not affect barium-sensitive inward rectifying potassium current. Guanabenz not only inhibited Ih but also shifted the voltage-dependent activation curve to hyperpolarizing potentials. Interestingly, Ih inhibition by guanabenz was not reversed by α2-adrenoceptor antagonist atipamezole treatment or by intracellular cyclic AMP perfusion, suggesting that the inhibition may not result from α2A-adrenoceptor signalling pathway but from direct inhibition of HCN channels. Coherent to our electrophysiological results, single-cell RT-PCR revealed that most MTN neurons lack α2A-adrenoceptor mRNA. Our study demonstrates that guanabenz can directly inhibit HCN channels in addition to its primary role of activating α2A-adrenoceptors.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Guanabenzo/farmacología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Tegmento Mesencefálico/citología , Animales , Relación Dosis-Respuesta a Droga , Fenómenos Electrofisiológicos/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Neuronas/citología , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos alfa 2/genética
8.
Sci Rep ; 9(1): 16873, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31727949

RESUMEN

Pain is susceptible to various cognitive factors. Suppression of pain by hunger is well known, but the effect of food intake after fasting (i.e. refeeding) on pain remains unknown. In the present study, we examined whether inflammatory pain behavior is affected by 24 h fasting and 2 h refeeding. In formalin-induced acute inflammatory pain model, fasting suppressed pain behavior only in the second phase and the analgesic effect was also observed after refeeding. Furthermore, in Complete Freund's adjuvant-induced chronic inflammatory pain model, both fasting and refeeding reduced spontaneous pain response. Refeeding with non-calorie agar produced an analgesic effect. Besides, intraperitoneal (i.p.) administration of glucose after fasting, which mimics calorie recovery following refeeding, induced analgesic effect. Administration of opioid receptor antagonist (naloxone, i.p.) and cannabinoid receptor antagonist (SR 141716, i.p.) reversed fasting-induced analgesia, but did not affect refeeding-induced analgesia in acute inflammatory pain model. Taken together, our results show that refeeding produce analgesia in inflammatory pain condition, which is associated with eating behavior and calorie recovery effect.


Asunto(s)
Dolor Agudo/dietoterapia , Dolor Crónico/dietoterapia , Ingestión de Alimentos/psicología , Glucosa/administración & dosificación , Hiperalgesia/dietoterapia , Manejo del Dolor/métodos , Dolor Agudo/etiología , Dolor Agudo/fisiopatología , Dolor Agudo/psicología , Analgésicos Opioides/farmacología , Animales , Dolor Crónico/etiología , Dolor Crónico/fisiopatología , Dolor Crónico/psicología , Modelos Animales de Enfermedad , Ingestión de Alimentos/fisiología , Privación de Alimentos/fisiología , Formaldehído/administración & dosificación , Adyuvante de Freund/administración & dosificación , Calor/efectos adversos , Hiperalgesia/etiología , Hiperalgesia/fisiopatología , Hiperalgesia/psicología , Inflamación , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Dimensión del Dolor , Rimonabant/farmacología
9.
Neuropharmacology ; 135: 572-580, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29634983

RESUMEN

Unlike in the central nervous system (CNS), in the adult peripheral nervous system (PNS), activation of GABAA receptors (GABAAR) is excitatory because of the relatively high concentration of intracellular chloride in these neurons. Indeed, exogenous GABA and muscimol, a GABAAR agonist, exacerbate acute inflammatory hypersensitivity in rodents. However, it remains unclear whether peripheral GABAAR and the endogenous GABA play an important role in persistent inflammatory hypersensitivity. In this study, we thus investigated how peripheral GABAAR affects pain hypersensitivity by using the complete Freund's adjuvant (CFA)-induced persistent inflammatory pain mouse model. We found that intraplantar (i.pl.) administration of GABAAR antagonists, picrotoxin, and 1(S),9(R)-(-)-bicuculline methiodide significantly inhibited both spontaneous nociceptive (paw licking and flinching) behavior and mechanical hypersensitivity in CFA-injected mice at day 3 (D3), but not in naïve mice. Interestingly, CFA-induced mechanical hypersensitivity was significantly reversed by anti-GABA antibody (anti-GABA, i.pl.). In addition, RT-qPCR revealed that glutamate decarboxylase Gad1 (GAD 67) and Gad2 (GAD 65) mRNA expression was also upregulated in the ipsilateral hind paw of CFA-injected mice at D3. Finally, 5α-pregnan-3α-ol-20-one (3α,5α-THP), a selective positive allosteric modulator of GABAAR, produced mechanical hypersensitivity in naïve mice in a dose-dependent manner. Taken together, our results indicate that peripheral GABAAR and endogenous GABA, possibly produced by the inflamed tissue, potentiate CFA-induced persistent inflammatory hypersensitivity, suggesting that they can be used as a therapeutic target for alleviating inflammatory pain.


Asunto(s)
Hiperalgesia/metabolismo , Inflamación/metabolismo , Receptores de GABA-A/metabolismo , Animales , Anticuerpos/farmacología , Adyuvante de Freund , Agonistas de Receptores de GABA-A/farmacología , Antagonistas de Receptores de GABA-A/farmacología , Glutamato Descarboxilasa/metabolismo , Hiperalgesia/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Masculino , Ratones Endogámicos C57BL , Dolor Nociceptivo/tratamiento farmacológico , Dolor Nociceptivo/metabolismo , ARN Mensajero/metabolismo , Distribución Aleatoria , Tacto , Ácido gamma-Aminobutírico/inmunología , Ácido gamma-Aminobutírico/metabolismo
10.
Physiol Rep ; 5(8)2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28438981

RESUMEN

Gamma-aminobutyric acid (GABA) depolarizes dorsal root ganglia (DRG) primary afferent neurons through activation of Cl- permeable GABAA receptors but the physiologic role of GABAA receptors in the peripheral terminals of DRG neurons remains unclear. In this study, we investigated the role of peripheral GABAA receptors in nociception using a mouse model of acute inflammation. In vivo, peripheral administration of the selective GABAA receptor agonist muscimol evoked spontaneous licking behavior, as well as spinal wide dynamic range (WDR) neuron firing, after pre-conditioning with formalin but had no effect in saline-treated mice. GABAA receptor-mediated pain behavior after acute formalin treatment was abolished by the GABAA receptor blocker picrotoxin and cyclooxygenase inhibitor indomethacin. In addition, treatment with prostaglandin E2 (PGE2) was sufficient to reveal muscimol-induced licking behavior. In vitro, GABA induced sub-threshold depolarization in DRG neurons through GABAA receptor activation. Both formalin and PGE2 potentiated GABA-induced Ca2+ transients and membrane depolarization in capsaicin-sensitive nociceptive DRG neurons; these effects were blocked by the prostaglandin E2 receptor 4 (EP4) antagonist AH23848 (10 µmol/L). Furthermore, potentiation of GABA responses by PGE2 was prevented by the selective Nav1.8 antagonist A887826 (100 nmol/L). Although the function of the Na+-K+-2Cl- co-transporter NKCC1 was required to maintain the Cl- ion gradient in isolated DRG neurons, NKCC1 was not required for GABAA receptor-mediated nociceptive behavior after acute inflammation. Taken together, these results demonstrate that GABAA receptors may contribute to the excitation of peripheral sensory neurons in inflammation through a combined effect involving PGE2-EP4 signaling and Na+ channel sensitization.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Nocicepción , Receptores de GABA-A/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Células Receptoras Sensoriales/metabolismo , Potenciales de Acción , Animales , Señalización del Calcio , Células Cultivadas , Dinoprostona/farmacología , Femenino , Agonistas del GABA/farmacología , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/fisiología , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Ganglios Espinales/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Muscimol/farmacología , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/fisiología , Bloqueadores de los Canales de Sodio/farmacología , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA