Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Mol Neurobiol ; 60(1): 171-182, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36251233

RESUMEN

We have previously shown that pituitary adenylate cyclase-activating polypeptide (PACAP) in the ventromedial hypothalamus (VMH) enhances feeding during the dark cycle and after fasting, and inhibits feeding during the light cycle. On the other hand, galanin is highly expressed in the hypothalamus and has been reported to be involved in feeding regulation. In this study, we investigated the involvement of the VMH-PACAP to the dorsomedial hypothalamus (DMH)-galanin signaling in the regulation of feeding. Galanin expression in the hypothalamus was significantly increased with fasting, but this increment was canceled in PACAP-knockout (KO) mice. Furthermore, overexpression of PACAP in the VMH increased the expression of galanin, while knockdown (KD) of PACAP in the VMH decreased the expression of galanin, indicating that the expression of galanin in the hypothalamus might be regulated by PACAP in the VMH. Therefore, we expressed the synaptophysin-EGFP fusion protein (SypEGFP) in PACAP neurons in the VMH and visualized the neural projection to the hypothalamic region where galanin was highly expressed. A strong synaptophysin-EGFP signal was observed in the DMH, indicating that PACAP-expressing cells of the VMH projected to the DMH. Furthermore, galanin immunostaining in the DMH showed that galanin expression was weak in PACAP-KO mice. When galanin in the DMH was knocked down, food intake during the dark cycle and after fasting was decreased, and food intake during the light cycle was increased, as in PACAP-KO mice. These results indicated that galanin in the DMH may regulate the feeding downstream of PACAP in the VMH.


Asunto(s)
Hipotálamo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa , Animales , Ratones , Regulación del Apetito , Galanina/metabolismo , Hipotálamo/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Sinaptofisina/metabolismo
2.
Biomolecules ; 12(12)2022 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-36551287

RESUMEN

We have previously shown that spinal pituitary adenylate cyclase-activating polypeptide (PACAP)/PACAP type 1 (PAC1) receptor signaling triggered long-lasting nociceptive behaviors through astroglial activation in mice. Since astrocyte-neuron lactate shuttle (ANLS) could be essential for long-term synaptic facilitation, we aimed to elucidate a possible involvement of spinal ANLS in the development of the PACAP/PAC1 receptor-induced nociceptive behaviors. A single intrathecal administration of PACAP induced short-term spontaneous aversive behaviors, followed by long-lasting mechanical allodynia in mice. These nociceptive behaviors were inhibited by 1,4-dideoxy-1,4-imino-d-arabinitol (DAB), an inhibitor of glycogenolysis, and this inhibition was reversed by simultaneous L-lactate application. In the cultured spinal astrocytes, the PACAP-evoked glycogenolysis and L-lactate secretion were inhibited by DAB. In addition, a protein kinase C (PKC) inhibitor attenuated the PACAP-induced nociceptive behaviors as well as the PACAP-evoked glycogenolysis and L-lactate secretion. Finally, an inhibitor for the monocarboxylate transporters blocked the L-lactate secretion from the spinal astrocytes and inhibited the PACAP- and spinal nerve ligation-induced nociceptive behaviors. These results suggested that spinal PAC1 receptor-PKC-ANLS signaling contributed to the PACAP-induced nociceptive behaviors. This signaling system could be involved in the peripheral nerve injury-induced pain-like behaviors.


Asunto(s)
Astrocitos , Ácido Láctico , Neuronas , Nocicepción , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria , Animales , Ratones , Astrocitos/metabolismo , Ácido Láctico/metabolismo , Neuronas/metabolismo , Nocicepción/fisiología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Transporte Biológico
3.
Biochem Biophys Res Commun ; 631: 146-151, 2022 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-36194909

RESUMEN

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a highly conserved pleiotropic neuropeptide, implicated in emotional stress responses and anxiety-related disorders. Here, we examined whether our recently developed small-molecule non-peptide PACAP receptor antagonists could ameliorate anxiety-like behaviors induced by acute restraint stress in mice. The antagonists PA-9 and its derivative PA-915 improved anxiety-like behaviors in mice subjected to restraint stress. An anxiolytic effect was observed with single acute dose, suggesting their fast-acting properties. PA-915 demonstrated a statistically significant anxiolytic effect whereas fluoxetine did not. These results indicate the potential of PAC1 antagonists as a novel treatment for anxiety.


Asunto(s)
Ansiolíticos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa , Animales , Ansiolíticos/farmacología , Ansiolíticos/uso terapéutico , Ansiedad/tratamiento farmacológico , Fluoxetina , Ratones , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/uso terapéutico , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria
4.
Sci Rep ; 12(1): 12604, 2022 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-35871167

RESUMEN

Fractalkine is one of the CX3C chemokine family, and it is widely expressed in the brain including the hypothalamus. In the brain, fractalkine is expressed in neurons and binds to a CX3C chemokine receptor 1 (CX3CR1) in microglia. The hypothalamus regulates energy homeostasis of which dysregulation is associated with obesity. Therefore, we examined whether fractalkine-CX3CR1 signalling involved in regulating food intake and hypothalamic inflammation associated with obesity pathogenesis. In the present study, fractalkine significantly reduced food intake induced by several experimental stimuli and significantly increased brain-derived neurotrophic factor (BDNF) mRNA expression in the hypothalamus. Moreover, tyrosine receptor kinase B (TrkB) antagonist impaired fractalkine-induced anorexigenic actions. In addition, compared with wild-type mice, CX3CR1-deficient mice showed a significant increase in food intake and a significant decrease in BDNF mRNA expression in the hypothalamus. Mice fed a high-fat diet (HFD) for 16 weeks showed hypothalamic inflammation and reduced fractalkine mRNA expression in the hypothalamus. Intracerebroventricular administration of fractalkine significantly suppressed HFD-induced hypothalamic inflammation in mice. HFD intake for 4 weeks caused hypothalamic inflammation in CX3CR1-deficient mice, but not in wild-type mice. These findings suggest that fractalkine-CX3CR1 signalling induces anorexigenic actions via activation of the BDNF-TrkB pathway and suppresses HFD-induced hypothalamic inflammation in mice.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Quimiocina CX3CL1 , Animales , Antiinflamatorios , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Quimiocina CX3CL1/genética , Quimiocina CX3CL1/metabolismo , Dieta Alta en Grasa/efectos adversos , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/etiología , Obesidad/metabolismo , ARN Mensajero
5.
Eur J Med Chem ; 231: 114160, 2022 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-35124531

RESUMEN

Since PA-8 (5-(4-(Allyloxy)-3-methoxyphenyl)-2-amino-5,8-dihydro-3H,6H-pyrido[2,3-d]pyrimidine-4,7-dione) was recently identified as a novel small-molecule antagonist of the pituitary adenylate cyclase-activating polypeptide (PACAP) type I (PAC1) receptor, a series of pyrido[2,3-d]pyrimidine derivatives have been designed, synthesized and subsequently evaluated for antagonistic activity on the PAC1 receptor. In this study, we synthesized 21 derivatives based on the PA-8 structure. Among them, the compound 2o (2-Amino-5-(3-trifluoromethoxy-phenyl)-5,8-dihydro-3H,6H-pyrido[2,3-d]pyrimidine-4,7-dione) showed more potent antagonistic activities than PA-8. Intrathecal (i.t.) injection of 2o blocked the induction of PACAP-induced aversive behaviors and mechanical allodynia in mice, and the effects were more potent than those of PA-8. A single i.t. injection of 2o also inhibited spinal nerve ligation (SNL)-induced mechanical allodynia. Repeated intraperitoneal administration of 2o gradually reduced the SNL-induced mechanical allodynia, and this effect appeared earlier than for PA-8. In addition, 2o exhibited a favorable ADME and pharmacokinetics profiles. These results suggest that 2o may become an analgesic for the treatment of neuropathic pain.


Asunto(s)
Neuralgia , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria , Animales , Hiperalgesia , Ratones , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Pirimidinas/farmacología
6.
J Pharmacol Sci ; 148(1): 108-115, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34924114

RESUMEN

Brain glycogen metabolism is known to be involved in the learning and memory processes. Protein targeting to glycogen (PTG) is a crucial molecule for glycogenesis, and its expression level is shown to be increased in the dorsal hippocampus during fear memory acquisition and recall, suggesting that PTG may contribute to the memory process. However, its detailed role in the dorsal hippocampus remains unclear. Therefore, we knocked down the expression of PTG in the dorsal hippocampus and attempted to analyze its function behaviorally. PTG expression was found to be enriched in astrocytes. Furthermore, short hairpin RNA against PTG suppressed the expression of PTG in astrocytes. Mice with knockdown of PTG in the dorsal hippocampus showed suppressed alternation behavior in the Y-maze test and reduced memory recall at the first hour after acquisition in the passive avoidance test. Knockdown of mouse dorsal hippocampal astrocyte-specific PTG also impaired working memory in the Y-maze test. GluR1, GluR2, and NR2a subunits expressions were significantly down-regulated in the dorsal hippocampus of mice in which PTG was knocked down. These results indicate that PTG in the dorsal hippocampal astrocytes may contribute to working and short-term memories by maintaining the expression of glutamate receptor subunits.


Asunto(s)
Expresión Génica , Glucógeno/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Memoria a Corto Plazo/fisiología , Receptores AMPA/genética , Receptores AMPA/metabolismo , Animales , Astrocitos/metabolismo , Hipocampo/citología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones Endogámicos
7.
Front Pharmacol ; 12: 699026, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34489696

RESUMEN

The free fatty acid receptor 1 (FFAR1) is suggested to function as a G protein-coupled receptor (GPR40) for medium-to-long-chain free fatty acids. Previous studies on the expression of FFAR1 revealed that the nigrostriatal region is one of the areas which express abundant FFAR1 mRNA/protein in the central nervous system (CNS). However, the role of FFAR1 in the CNS has been still largely unclarified. Here, we examined a possible functional role of FFAR1 in the control of extracellular concentrations of striatal monoamines and cocaine-induced locomotor activity. Microdialysis analysis revealed that the basal level of extracellular dopamine (DA) was significantly elevated, while the basal serotonin (5-HT) level tended to be reduced in the striatum of FFAR1 knockout (-/-) mice. Interestingly, local application of a FFAR1 agonist, GW9508, markedly augmented the striatal 5-HT release in FFAR1 wild-type (+/+) mice, whereas topical application of a FFAR1 antagonist, GW1100, significantly reduced the 5-HT release. However, the enhanced 5-HT release was completely lost in -/- mice. Although acute administration of cocaine enhanced the locomotor activity in both +/+ and -/- mice, the magnitude of the enhancement was significantly reduced in -/- mice. In addition, intraperitoneal injection of GW1100 significantly decreased the cocaine-induced locomotor enhancement. These results suggest that FFAR1 has a facilitatory role in striatal 5-HT release, and the evoked 5-HT release might contribute to enhance cocaine-induced locomotor activity.

8.
Pharmacol Rep ; 73(4): 1109-1121, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33835466

RESUMEN

BACKGROUND: Pituitary adenylate cyclase-activating polypeptide (PACAP) plays an essential role in the modulation of astrocyte functions. Although lactate secretion from astrocytes contributes to many forms of neuronal plasticity in the central nervous system, including fear learning and memory, the role of PACAP in lactate secretion from astrocytes is unclear. METHODS: The amygdala and hippocampus of PACAP (+ / +) and PACAP (-/-) mice were acquired 1 h after memory acquisition and recall in the passive avoidance test. The concentration of glycogen and lactate in these regions was measured. The concentration of lactate in the hippocampus's extracellular fluid was also measured by microdialysis during memory acquisition or intracerebroventricular administration of PACAP. RESULTS: We observed that memory acquisition caused a significant decrease in glycogen concentration and increased lactate concentration in the PACAP (+ / +) mice's hippocampus. However, memory acquisition did not increase in the lactate concentration in PACAP (-/-) mice's hippocampus. Further, memory retrieval evoked lactate production in the amygdala and the hippocampus of PACAP (+ / +) mice. Still, there was no significant increase in lactate concentration in the same regions of PACAP (-/-) mice. In vivo microdialysis in rats revealed that the hippocampus's extracellular lactate concentration increased after a single PACAP intracerebroventricular injection. Additionally, the hippocampus's extracellular lactate concentration increased with the memory acquisition in PACAP (+ / +) mice, but not in PACAP (-/-) mice. CONCLUSIONS: PACAP may enhance lactate production and secretion in astrocytes during the acquisition and recall of fear memories.


Asunto(s)
Astrocitos/metabolismo , Miedo/fisiología , Ácido Láctico/metabolismo , Memoria/fisiología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/fisiología , Animales , Astrocitos/fisiología , Glucógeno/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiología , Masculino , Ratones , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley
9.
Curr Mol Pharmacol ; 14(2): 115-122, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32329707

RESUMEN

BACKGROUND: Major depressive disorder (MDD) is a common psychological disorder worldwide. However, one-third of patients with MDD are resistant to the present anti-depressant medicine, which regulates monoamine contents in the brain. Thus, another drug target is strongly required. Much evidence strongly suggests that sirtuin1, which is the key factor in regulating the mitochondrial activity, may be implicated in MDD. OBJECTIVE: Since it is suggested that royal jelly (RJ) ameliorated depressive-like behavior and affected mitochondrial activity in mice, we hypothesized that RJ could be an alternative medicine against MDD, which acts via sirtuin1 signaling to improve mitochondrial activity. METHODS: In the present study, we applied a mouse model of MDD to investigate the effect of RJ on the depressive-like behavior and the sirtuin1 signaling on mitochondrial activity. RESULTS: Our results indicated that either the oral administration of RJ for 12 days or single intracerebroventricular (i.c.v.) injection decreased the duration of immobility in the tail suspension test, which suggested that RJ had an antidepressant-like effect. Moreover, sirtuin1 protein expression increased in mice following RJ treatment in the amygdala region, but not in the other brain regions. Similarly, the expressions of oxidative phosphorylation (OXPHOS) related proteins increased in the amygdala regions, but not in the hippocampal regions. CONCLUSION: The increase of sirtuin1 and OXPHOS protein expression may at least in part contribute to the antidepressant-like effect of the RJ pathway, and RJ may have the potential to be a novel anti-depressant drug.


Asunto(s)
Antidepresivos/química , Trastorno Depresivo Mayor/tratamiento farmacológico , Ácidos Grasos/química , Sirtuina 1/metabolismo , Amígdala del Cerebelo/metabolismo , Animales , Antidepresivos/farmacología , Conducta Animal , Ácidos Grasos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Histona Desacetilasas/metabolismo , Humanos , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Proteómica , Transducción de Señal , Sirtuina 1/genética
10.
Mol Neurobiol ; 57(4): 2101-2114, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31927724

RESUMEN

Pituitary adenylate cyclase-activating polypeptide (PACAP) is abundantly expressed in the hypothalamus and contributes to hypothalamic functions, including appetite regulation. Although food intake is suggested to be decreased in PACAP (-/-) mice, the detailed mechanisms are still being discussed. We sought to investigate this link. The food consumption at 8 h after refeeding in the (-/-) mice who had fasted for 2 days was significantly lower than in the PACAP (+/+) mice. The nocturnal and daily food intake of (-/-) mice was significantly lower than those of (+/+) mice, but the diurnal food intake showed a tendency to increase. mRNA expression levels of agouti-related peptide (AgRP) were decreased, but those of proopiomelanocortin (POMC) were increased in the hypothalamus of (-/-) mice 4 h after refeeding. Furthermore, intracerebroventricular administration of a PACAP receptor antagonist, PACAP6-38 (1 nmol/4 µL/mouse), decreased food intake and body weight 1, 2, and 4 h after refeeding, as well as expression levels of AgRP at 4 h after refeeding in (+/+) mice. The selective overexpression of PACAP by the infection of an adeno-associated virus in the ventromedial hypothalamus (VMH) resulted in an increase in food intake and AgRP expression in the nocturnal period in addition to the increased food intake at 8 h after refeeding. These results suggest that food intake behavior in mice is triggered by the increase in PACAP expression in the VMH via modulation of AgRP expression in the hypothalamus, pointing to PACAP inhibition as a potential strategy for the development of anti-obesity drugs.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Conducta Alimentaria , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Núcleo Hipotalámico Ventromedial/metabolismo , Proteína Relacionada con Agouti/genética , Animales , Ritmo Circadiano , Ayuno , Ratones , Neuropéptidos/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/deficiencia , Proopiomelanocortina/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal
11.
Eur J Med Chem ; 186: 111902, 2020 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-31771828

RESUMEN

We recently identified novel small-molecule antagonists of the PACAP type I (PAC1) receptor using docking-based in silico screening followed by in vitro/vivo pharmacological assays. In the present study, we synthesized 18 novel derivatives based on the structure of PA-9, a recently developed antagonist of the PAC1 receptor, with a view to obtain a panel of compounds with more potent antagonistic and analgesic activities. Among them, compound 3d showed improved antagonistic activities. Intrathecal injection of 3d inhibited both pituitary adenylate cyclase-activating polypeptide (PACAP) and spinal nerve ligation-induced mechanical allodynia. The effects were more potent than PA-9. Compound 3d also showed anti-allodynic effects following oral administration. Hence, our results suggest that 3d may become an orally available analgesic in the treatment of the neuropathic pain.


Asunto(s)
Analgésicos/farmacología , Neuralgia/tratamiento farmacológico , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Analgésicos/síntesis química , Analgésicos/química , Animales , Conducta Animal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos , Estructura Molecular , Neuralgia/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
12.
J Pharmacol Sci ; 139(2): 129-132, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30552012

RESUMEN

We recently developed PA-8, a novel small-molecule antagonist of PACAP type 1 (PAC1) receptor. In the present study, we examined whether PA-8 was effective against formalin-induced inflammatory pain in mice. Both intrathecal and oral administration of PA-8 resulted in the dose-dependent attenuation of the second phase of formalin-induced nociceptive responses. PA-8 also inhibited c-fos upregulation in the ipsilateral dorsal horn of the spinal cord. The results suggested that PACAP-PAC1 receptor signaling system in the spinal cord were primarily involved in the transmission of inflammatory pain, and PA-8 could be useful for the development of novel analgesics for treating inflammatory pain.


Asunto(s)
Analgésicos/uso terapéutico , Dolor/tratamiento farmacológico , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/antagonistas & inhibidores , Animales , Antiinflamatorios/uso terapéutico , Conducta Animal/efectos de los fármacos , Formaldehído , Inyecciones Espinales , Masculino , Ratones , Dolor/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo
13.
Nihon Yakurigaku Zasshi ; 151(6): 239-243, 2018.
Artículo en Japonés | MEDLINE | ID: mdl-29887572

RESUMEN

Transfer of lactate from astrocytes to neurons is activated when synaptic activity is increased, and this mechanism is now known as the astrocyte-neuron lactate shuttle (ANLS), that could account for the coupling between synaptic activity and energy delivery. Many lines of evidence suggested that ANLS contributes to neuronal activation or synaptic plasticity at the cellular level as well as learning/memory and cocaine addiction at the behavioral level. However, the candidate neurotransmitters which evoke ANLS activation are still under discussion. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neurotransmitter which distributed widely in central nervous system. Since PACAP might activate ANLS from very low concentration in cultured forebrain astrocytes, PACAP might be one of the candidates for the endogenous ANLS activator. In the present study, we investigated the potential relevance of PACAP/ANLS signaling in the learning/memory and spinal nociceptive transmission. In this study, we made the following findings: 1) PACAP could be an endogenous inducer for ANLS activation in central nervous system; 2) ANLS activation by PACAP/PAC1 receptor signaling contributed to learning/memory and induced long-lasting nociceptive behaviors; 3) PKC activation played an important role in the PACAP/PAC1 receptor-evoked ANLS.


Asunto(s)
Astrocitos , Sistema Nervioso Central , Ácido Láctico , Neuronas , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria
14.
J Pharmacol Exp Ther ; 365(1): 1-8, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29363578

RESUMEN

Pituitary adenylate cyclase-activating polypeptide (PACAP) and its receptors are present in the spinal dorsal horn and dorsal root ganglia, suggesting an important role of PACAP signaling systems in the modulation of spinal nociceptive transmission. Previously, we found that intrathecal injection of PACAP or maxadilan, a selective PACAP type I (PAC1) receptor agonist, induced transient aversive responses followed by a long-lasting mechanical allodynia in mice, suggesting that PACAP-PAC1 receptor systems are involved in chronic pain and that selective PAC1 antagonists may become a new class of analgesics. Although several PAC1 antagonists, such as PACAP 6-38, have been reported, all of them are peptide compounds. In the present study, we identified new small-molecule antagonists of the PAC1 receptor using in silico screening and in vitro/vivo pharmacological assays. The identified small-molecule compounds, named PA-8 and PA-9, dose dependently inhibited the phosphorylation of CREB induced by PACAP in PAC1-, but not VPAC1- or VPAC2-receptor-expressing CHO cells. PA-8 and PA-9 also dose dependently inhibited PACAP-induced cAMP elevation with an IC50 of 2.0 and 5.6 nM, respectively. In vivo pharmacological assays showed that intrathecal injection of these compounds blocked the induction of PACAP-induced aversive responses and mechanical allodynia in mice. In contrast, the compounds when administered alone exerted neither agonistic nor algesic actions in the in vitro/vivo assays. The compounds identified in the present study are new and the first small-molecule antagonists of the PAC1 receptor; they may become seed compounds for developing novel analgesics.


Asunto(s)
Simulación por Computador , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/química , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/antagonistas & inhibidores , Animales , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Evaluación Preclínica de Medicamentos , Hiperalgesia/tratamiento farmacológico , Masculino , Ratones , Simulación del Acoplamiento Molecular , Nocicepción/efectos de los fármacos , Fosforilación/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/uso terapéutico , Dominios Proteicos , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/química , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo
16.
Biol Pharm Bull ; 40(8): 1255-1259, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28769007

RESUMEN

The free fatty acid receptor 1 (GPR40/FFAR1) is activated by polyunsaturated fatty acids (PUFAs) such as docosahexaenoic acids (DHA). This receptor has been the focus of many studies regarding physiological functions of the central nervous system. PUFAs are essential for neuronal development and maintenance of neuronal function; thus, the decrease of PUFAs in the brain is closely related to the induction of psychiatric diseases associated with emotional disorder, such as anxiety, depression, and schizophrenia. However, details of the mechanisms remain unclear. In this study, we investigated changes of maternal and/or emotional behavior caused by a deficiency of GPR40/FFAR1 signaling. GPR40/FFAR1 deficient (FFAR1-/-) female mice exhibited impaired maternal care such as retrieving behaviors and an increased rate of neglect and infanticide when compared to wild type (WT) female mice. Furthermore, FFAR1-/- female mice showed increased time spent in the open arms in an elevated plus maze test, reduction of locomotor activity and social interaction behavior, and decreased sucrose intake, when compared to WT female mice. In conclusion, these findings suggest that PUFAs-GPR40/FFAR1 signaling might function, at least in part, as a regulatory factor of emotional and maternal behavior in mice.


Asunto(s)
Conducta Animal , Emociones , Conducta Materna , Receptores Acoplados a Proteínas G/genética , Animales , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Conducta Social
17.
PLoS One ; 12(7): e0180610, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28723961

RESUMEN

We previously showed that activation of G protein-coupled receptor 40/free fatty acid receptor 1 (GPR40/FFAR1) signaling modulates descending inhibition of pain. In this study, we investigated the involvement of fatty acid-GPR40/FFAR1 signaling in the transition from acute to chronic pain. We used GPR40/FFAR1-knockout (GPR40KO) mice and wild-type (WT) mice. A plantar incision was performed, and mechanical allodynia and thermal hyperalgesia were evaluated with a von Frey filament test and plantar test, respectively. Immunohistochemistry was used to localize GPR40/FFAR1, and the levels of free fatty acids in the hypothalamus were analyzed with liquid chromatography-tandem mass spectrometry. The repeated administration of GW1100, a GPR40/FFAR1 antagonist, exacerbated the incision-induced mechanical allodynia and significantly increased the levels of phosphorylated extracellular signal-regulated kinase in the spinal cord after low-threshold touch stimulation in the mice compared to vehicle-treated mice. The levels of long-chain free fatty acids, such as docosahexaenoic acid, oleic acid, and palmitate, which are GPR40/FFAR1 agonists, were significantly increased in the hypothalamus two days after the surgery compared to levels in the sham group. Furthermore, the incision-induced mechanical allodynia was exacerbated in the GPR40KO mice compared to the WT mice, while the response in the plantar test was not changed. These findings suggested that dysfunction of the GPR40/FFAR1 signaling pathway altered the endogenous pain control system and that this dysfunction might be associated with the development of chronic pain.


Asunto(s)
Conducta Animal/fisiología , Hiperalgesia/genética , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/fisiología , Animales , Conducta Animal/efectos de los fármacos , Benzoatos/farmacología , Ácidos Docosahexaenoicos/metabolismo , Hiperalgesia/metabolismo , Hipotálamo/metabolismo , Ratones , Ratones Noqueados , Ácido Oléico/metabolismo , Dimensión del Dolor , Ácido Palmítico/metabolismo , Fosforilación , Pirimidinas/farmacología , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos
18.
J Pharmacol Sci ; 133(2): 88-95, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28215474

RESUMEN

We prepared a DIC model by administrating LPS to cynomolgus monkeys, and investigated its potential for evaluations of new medicines for DIC therapy. Peripheral blood mononuclear cells (PBMC) collected from cynomolgus monkeys were incubated with LPS (8 types), and TNF-α levels in the media were measured. LPS from Escherichia coli (K-235) was most appropriate in terms of larger increases and smaller variation in TNF-α levels. PBMC from rats, cynomolgus monkeys or humans were incubated with LPS (K-235), and the TNF-α response to LPS was investigated. The response was comparable between cynomolgus monkeys and humans but small in rats. In an in vivo experiment, LPS (K-235) was administered once intravenously to cynomolgus monkeys with or without recombinant human thrombomodulin (rhTM) to investigate any changes in coagulation and fibrinolysis biomarkers and the suppressive effect of rhTM. The liver, kidney, and lung were examined histopathologically. Almost all of the changes resembled the pathophysiological status of human DIC and were suppressed by co-administration of rhTM. The DIC model resembling human DIC was established by LPS (K-235) treatment in cynomolgus monkeys, and therapeutic effect of rhTM was noted, suggesting that this model is useful in evaluations of the efficacy of new medicines for DIC therapy.


Asunto(s)
Modelos Animales de Enfermedad , Coagulación Intravascular Diseminada/tratamiento farmacológico , Leucocitos Mononucleares/efectos de los fármacos , Trombomodulina/uso terapéutico , Adulto , Animales , Coagulación Sanguínea , Células Cultivadas , Coagulación Intravascular Diseminada/inducido químicamente , Coagulación Intravascular Diseminada/fisiopatología , Escherichia coli , Humanos , Lipopolisacáridos , Macaca fascicularis , Masculino , Ratas , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Trombomodulina/administración & dosificación , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
19.
J Pharmacol Sci ; 132(4): 249-254, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27979701

RESUMEN

The free fatty acid receptor 1 (GPR40/FFAR1) is a G protein-coupled receptor, which is activated by long chain fatty acids. We have previously demonstrated that activation of brain GPR40/FFAR1 exerts an antinociceptive effect that is mediated by the modulation of the descending pain control system. However, it is unclear whether brain GPR40/FFAR1 contributes to emotional function. In this study, we investigated the involvement of GPR40/FFAR1 in emotional behavior using GPR40/FFAR1 deficient (knockout, KO) mice. The emotional behavior in wild and KO male mice was evaluated at 9-10 weeks of age by the elevated plus-maze test, open field test, social interaction test, and sucrose preference test. Brain monoamines levels were measured using LC-MS/MS. The elevated plus-maze test and open field tests revealed that the KO mice reduced anxiety-like behavior. There were no differences in locomotor activity or social behavior between the wild and KO mice. In the sucrose preference test, the KO mice showed reduction in sucrose preference and intake. The level of noradrenaline was higher in the hippocampus, medulla oblongata, hypothalamus and midbrain of KO mice. Therefore, these results suggest that brain GPR40/FFAR1 is associated with anxiety- and depression-related behavior regulated by the increment of noradrenaline in the brain.


Asunto(s)
Conducta Animal/fisiología , Encéfalo/fisiología , Emociones/fisiología , Norepinefrina/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Animales , Ansiedad/metabolismo , Encéfalo/metabolismo , Depresión/metabolismo , Conducta Alimentaria/fisiología , Locomoción/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Acoplados a Proteínas G/metabolismo , Conducta Social , Sacarosa/administración & dosificación
20.
Mol Pain ; 122016.
Artículo en Inglés | MEDLINE | ID: mdl-27175011

RESUMEN

BACKGROUND: Pituitary adenylate cyclase-activating polypeptide (PACAP) and its receptors are present in the spinal dorsal horn and dorsal root ganglia, suggesting an important role of PACAP-PACAP receptors signaling system in the modulation of spinal nociceptive transmission. We have previously reported that a single intrathecal injection of PACAP or a PACAP specific (PAC1) receptor selective agonist, maxadilan, in mice induced dose-dependent aversive behaviors, which lasted more than 30 min, and suggested that the maintenance of the nociceptive behaviors was associated with the spinal astrocytic activation. RESULTS: We found that a single intrathecal administration of PACAP or maxadilan also produced long-lasting hind paw mechanical allodynia, which persisted at least 84 days without affecting thermal nociceptive threshold. In contrast, intrathecal application of vasoactive intestinal polypeptide did not change mechanical threshold, and substance P, calcitonin gene-related peptide, or N-methyl-D-aspartate induced only transient mechanical allodynia, which disappeared within 21 days. Western blot and immunohistochemical analyses with an astrocytic marker, glial fibrillary acidic protein, revealed that the spinal PAC1 receptor stimulation caused sustained astrocytic activation, which also lasted more than 84 days. Intrathecal co-administration of L-α-aminoadipate, an astroglial toxin, with PACAP or maxadilan almost completely prevented the induction of the mechanical allodynia. Furthermore, intrathecal treatment of L-α-aminoadipate at 84 days after the PAC1 stimulation transiently reversed the mechanical allodynia accompanied by the reduction of glial fibrillary acidic protein expression level. CONCLUSION: Our data suggest that spinal astrocytic activation triggered by the PAC1 receptor stimulation contributes to both induction and maintenance of the long-term mechanical allodynia.


Asunto(s)
Astrocitos/patología , Hiperalgesia/metabolismo , Hiperalgesia/patología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Médula Espinal/patología , Adipatos/farmacología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente , Proteína Ácida Fibrilar de la Glía/metabolismo , Inyecciones Espinales , Proteínas de Insectos/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Ratones , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/agonistas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA