Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Glia ; 72(8): 1402-1417, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38591338

RESUMEN

It is well-established that spinal microglia and peripheral macrophages play critical roles in the etiology of neuropathic pain; however, growing evidence suggests sex differences in pain hypersensitivity owing to microglia and macrophages. Therefore, it is crucial to understand sex- and androgen-dependent characteristics of pain-related myeloid cells in mice with nerve injury-induced neuropathic pain. To deplete microglia and macrophages, pexidartinib (PLX3397), an inhibitor of the colony-stimulating factor 1 receptor, was orally administered, and mice were subjected to partial sciatic nerve ligation (PSL). Following PSL induction, healthy male and female mice and male gonadectomized (GDX) mice exhibited similar levels of spinal microglial activation, peripheral macrophage accumulation, and mechanical allodynia. Treatment with PLX3397 significantly suppressed mechanical allodynia in normal males; this was not observed in female and GDX male mice. Sex- and androgen-dependent differences in the PLX3397-mediated preventive effects were observed on spinal microglia and dorsal root ganglia (DRG) macrophages, as well as in expression patterns of pain-related inflammatory mediators in these cells. Conversely, no sex- or androgen-dependent differences were detected in sciatic nerve macrophages, and inhibition of peripheral CC-chemokine receptor 5 prevented neuropathic pain in both sexes. Collectively, these findings demonstrate the presence of considerable sex- and androgen-dependent differences in the etiology of neuropathic pain in spinal microglia and DRG macrophages but not in sciatic nerve macrophages. Given that the mechanisms of neuropathic pain may differ among experimental models and clinical conditions, accumulating several lines of evidence is crucial to comprehensively clarifying the sex-dependent regulatory mechanisms of pain.


Asunto(s)
Microglía , Neuralgia , Pirroles , Caracteres Sexuales , Animales , Masculino , Femenino , Ratones , Neuralgia/metabolismo , Neuralgia/tratamiento farmacológico , Neuralgia/etiología , Microglía/efectos de los fármacos , Microglía/metabolismo , Pirroles/farmacología , Aminopiridinas/farmacología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Nervio Ciático/lesiones , Nervio Ciático/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/patología , Traumatismos de los Nervios Periféricos/complicaciones , Traumatismos de los Nervios Periféricos/metabolismo , Modelos Animales de Enfermedad
2.
J Pharmacol Sci ; 153(4): 183-187, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37973215

RESUMEN

Although microglia are associated with chronic pain, the role of spinal microglia in the regulation of itch remains unclear. In this study, we characterized spinal microglial activation in a mouse model of imiquimod (IMQ)-induced psoriasis. Hypertrophic (activated) microglia were observed throughout the spinal cord after the topical application of IMQ. Furthermore, the mRNA expression of microglial markers and inflammatory mediators was upregulated. Ablation of itch-related sensory neurons using resiniferatoxin decreased itch-related scratching behavior and the number of hypertrophic microglia in the spinal dorsal horn. Conclusively, sensory neuron input may partially contribute to spinal microglial activation after IMQ application.


Asunto(s)
Microglía , Psoriasis , Ratones , Animales , Imiquimod/efectos adversos , Imiquimod/metabolismo , Microglía/metabolismo , Médula Espinal/metabolismo , Modelos Animales de Enfermedad , Prurito/inducido químicamente , Psoriasis/inducido químicamente , Psoriasis/genética
3.
Drugs ; 83(9): 771-793, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37209211

RESUMEN

As clinical use of currently available opioid analgesics is often impeded by dose-limiting adverse effects, such as abuse liability and respiratory depression, new approaches have been pursued to develop safe, effective, and non-addictive pain medications. After the identification of the nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor more than 25 years ago, NOP receptor-related agonists have emerged as a promising target for developing novel and effective opioids that modulate the analgesic and addictive properties of mu-opioid peptide (MOP) receptor agonists. In this review, we highlight the effects of the NOP receptor-related agonists compared with those of MOP receptor agonists in experimental rodent and more translational non-human primate (NHP) models and the development status of key NOP receptor-related agonists as potential safe and non-addictive analgesics. Several lines of evidence demonstrated that peptidic and non-peptidic NOP receptor agonists produce potent analgesic effects by intrathecal delivery in NHPs. Moreover, mixed NOP/MOP receptor partial agonists (e.g., BU08028, BU10038, and AT-121) display potent analgesic effects when administered intrathecally or systemically, without eliciting adverse effects, such as respiratory depression, itch behavior, and signs of abuse liability. More importantly, cebranopadol, a mixed NOP/opioid receptor agonist with full efficacy at NOP and MOP receptors, produces robust analgesic efficacy with reduced adverse effects, conferring promising outcomes in clinical studies. A balanced coactivation of NOP and MOP receptors is a strategy that warrants further exploration and refinement for the development of novel analgesics with a safer and effective profile.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Insuficiencia Respiratoria , Animales , Humanos , Receptor de Nociceptina , Receptores Opioides/agonistas , Dolor/tratamiento farmacológico , Receptores Opioides mu/agonistas , Analgésicos Opioides/efectos adversos , Analgésicos/efectos adversos , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/tratamiento farmacológico
4.
Neuropharmacology ; 223: 109328, 2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36356937

RESUMEN

Opioids provide pain relief but are associated with several adverse effects. Researchers are exploring cannabis-based medicine as an alternative. However, little is known about the tendency for physical dependence on cannabinoids in comparison with that on opioids in primates. The aim of this study was to compare the potency of heroin and delta-9-tetrahydrocannabinol (THC) in eliciting analgesic effects and the development of physical dependence between opioids and cannabinoids in both male and female rhesus monkeys. Systemic administration of either heroin (0.03-0.18 mg/kg) or THC (0.3-1.8 mg/kg) in a dose-dependent manner produced antinociceptive effects against an acute thermal nociceptive stimulus. The µ-opioid receptor antagonist naltrexone (0.01 mg/kg) and the cannabinoid receptor antagonist SR141716A (0.3 mg/kg) produced the same degree of rightward shift in the dose-response curves for heroin- and THC-induced antinociception, respectively. Monkeys implanted with telemetry devices were subjected to short-term repeated administrations (two injections per day for 1-3 days) of either heroin (0.18 mg/kg), morphine (1.8 mg/kg), THC (1.8 mg/kg), or CP 55,940 (0.032 mg/kg). Administration of naltrexone (0.01 mg/kg) increased respiration, heart rate, and blood pressure in heroin- or morphine-treated monkeys. In contrast, administration of SR141716A (0.3 mg/kg) did not cause a significant change in these physiological parameters in THC- or CP 55,940-treated monkeys. Additionally, morphine, but not CP 55,940, enhanced the monkeys' hypersensitivity to the algogen capsaicin. Collectively, these results demonstrate that in nonhuman primates, both opioids and cannabinoids exert comparable antinociception; however, physical dependence on opioids, but not cannabinoids, at their antinociceptive doses, occurs following short-term exposures.


Asunto(s)
Analgésicos Opioides , Cannabinoides , Femenino , Masculino , Animales , Analgésicos Opioides/farmacología , Cannabinoides/farmacología , Dronabinol/farmacología , Morfina/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Heroína/farmacología , Naltrexona/farmacología , Rimonabant , Relación Dosis-Respuesta a Droga
5.
Biochem Pharmacol ; 198: 114972, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35189108

RESUMEN

Despite accumulating evidence in rodents, the functional role of neuromedin B (NMB) in regulating somatosensory systems in primate spinal cord is unknown. We aimed to compare the expression patterns of NMB and its receptor (NMBR) and the behavioral effects of intrathecal (i.t.) NMB with gastrin-releasing peptide (GRP) on itch or pain in non-human primates (NHPs). We used six adult rhesus monkeys. The mRNA or protein expressions of NMB, GRP, and their receptors were evaluated by quantitative reverse transcription polymerase chain reaction, immunohistochemistry, or in situ hybridization. We determined the behavioral effects of NMB or GRP via acute thermal nociception, capsaicin-induced thermal allodynia, and itch scratching response assays. NMB expression levels were greater than those of GRP in the dorsal root ganglia and spinal dorsal horn. Conversely, NMBR expression was significantly lower than GRP receptor (GRPR). I.t. NMB elicited only mild scratching responses, whereas GRP caused robust scratching responses. GRP- and NMB-elicited scratching responses were attenuated by GRPR (RC-3095) and NMBR (PD168368) antagonists, respectively. Moreover, i.t. NMB and GRP did not induce thermal hypersensitivity and GRPR and NMBR antagonists did not affect peripherally elicited thermal allodynia. Consistently, NMBR expression was low in both itch- and pain-responsive neurons in the spinal dorsal horn. Spinal NMB-NMBR system plays a minimal functional role in the neurotransmission of itch and pain in primates. Unlike the functional significance of the GRP-GRPR system in itch, drugs targeting the spinal NMB-NMBR system may not effectively alleviate non-NMBR-mediated itch.


Asunto(s)
Hiperalgesia , Prurito , Animales , Péptido Liberador de Gastrina/genética , Péptido Liberador de Gastrina/metabolismo , Péptido Liberador de Gastrina/farmacología , Hiperalgesia/metabolismo , Neuroquinina B/análogos & derivados , Dolor/metabolismo , Primates/metabolismo , Prurito/inducido químicamente , Prurito/metabolismo , Receptores de Bombesina/genética , Receptores de Bombesina/metabolismo , Médula Espinal , Asta Dorsal de la Médula Espinal/metabolismo
6.
Pharmacol Res Perspect ; 9(3): e00790, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34000759

RESUMEN

Several lines of evidence have clarified that the key transmission pathways of itching sensation travel from the periphery to the central nervous system (CNS). Despite the functional significance of gastrin-releasing peptide (GRP) and its cognate receptor in the itch processing mechanism in the spinal dorsal horn (SDH), the roles of GRP-expressing (GRP+ ) neurons in different regions remain unclear. This study aimed to determine whether GRP+ neurons in the CNS directly modulated itch processing. To specifically activate spinal and supraspinal GRP neurons by the designer receptors exclusively activated by designer drugs (DREADDs) system, CAG-LSL-Gq-DREADD mice were crossed with GRP-Cre mice, resulting in the development of GRP-hM3Dq mice. Immunohistochemistry showed that hM3Dq was highly expressed in the SDH and brainstem closely related to sensory processing. The intraperitoneal, intrathecal, or intracerebroventricular administration of clozapine-N-oxide, an agonist of hM3Dq, strongly elicited dermatome-dependent itch-related scratching behavior, but did not change pain sensitivity. Importantly, GRP-Gq-DREADD-mediated scratching behavior in GRP-hM3Dq mice was not affected by the ablation of transient receptor potential vanilloid 1+ sensory C-fibers, and it was also observed to a similar degree under chronic itch conditions. Furthermore, there were no significant sex differences in the scratching behavior elicited by GRP-Gq-DREADD, suggesting that itch-dominant roles of central GRP+ neurons might be common in both sexes, at least under normal physiological conditions. These novel findings not only contribute to understanding the functional roles of central GRP+ neurons further, but also propose the development of future effective therapeutics for intractable itching.


Asunto(s)
Péptido Liberador de Gastrina/fisiología , Neuronas/fisiología , Prurito/fisiopatología , Animales , Conducta Animal , Clozapina/análogos & derivados , Clozapina/farmacología , Ciclopropanos , Dermatitis por Contacto , Femenino , Haptenos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos
7.
Nihon Yakurigaku Zasshi ; 156(3): 139-144, 2021.
Artículo en Japonés | MEDLINE | ID: mdl-33952840

RESUMEN

After the identification of nociceptin/orphanin FQ (N/OFQ) peptide (NOP) and its cognate receptor, the unique functional profiles of the N/OFQ-NOP receptor system have been uncovered. NOP receptors are distributed in the key regions that regulate pain and reward processing in the central nervous system. In non-human primates (NHPs), activation of the NOP receptor causes antinociception and anti-hypersensitivity via spinal and supraspinal effects. Moreover, activation of the NOP receptor attenuates dopaminergic transmission and potentiates mu-opioid peptide (MOP) receptor-mediated analgesia. Here, we highlight the functional profiles of bifunctional NOP and MOP receptor agonists based on their promising effects for the treatment of pain and drug abuse. Bifunctional NOP/MOP receptor "partial" agonists, such as AT-121, BU08028, and BU10038, exert potent analgesic effects without MOP receptor-related side effects such as abuse liability, respiratory depression, physical dependence, and itching in NHPs. These novel NOP/MOP receptor agonists reduce rewarding and the reinforcing effects of abused drugs. Furthermore, a mixed NOP/opioid receptor "full" agonist, cebranopadol, is undergoing several clinical trials, and the therapeutic advantage of the coactivation of NOP and MOP receptors has also been confirmed in humans. Therefore, this class of drugs that coactivate NOP and MOP receptors proposes a wide therapeutic range with fewer side effects, indicating a greater potential for the development of novel safer opioid analgesics.


Asunto(s)
Analgésicos Opioides , Receptores Opioides , Analgésicos , Analgésicos Opioides/efectos adversos , Animales , Péptidos Opioides/uso terapéutico , Dolor/tratamiento farmacológico , Nociceptina
8.
Cells ; 10(4)2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33921365

RESUMEN

It is important to investigate the sex-dependent roles of microglia in pain hypersensitivity as reactive microglia within the spinal dorsal horn (DH) have been reported to be pivotal in neuropathic pain induction in male rodents upon nerve injury. Here, we aimed at determining the role of sex differences in the behavioral and functional outcomes of the chemogenetic activation of spinal microglia using Gq-designer receptors exclusively activated by designer drugs (Gq-DREADD) driven by the microglia-specific Cx3cr1 promoter. CAG-LSL-human Gq-coupled M3 muscarinic receptors (hM3Dq)-DREADD mice were crossed with CX3C chemokine receptor 1 (CX3CR1)-Cre mice, and immunohistochemistry images revealed that hM3Dq was selectively expressed on Iba1+ microglia, but not on astrocytes and neurons. Intrathecal (i.t.) administration of clozapine-N-oxide (CNO) elicited mechanical allodynia exclusively in male mice. Furthermore, the reactive microglia-dominant molecules that contributed to pain hypersensitivity in CX3CR1-hM3Dq were upregulated in mice of both sexes. The degree of upregulation was greater in male than in female mice. Depletion of spinal microglia using pexidartinib (PLX3397), a colony stimulating factor-1 receptor inhibitor, alleviated the male CX3CR1-hM3Dq mice from pain hypersensitivity and compromised the expression of inflammatory molecules. Thus, the chemogenetic activation of spinal microglia resulted in pain hypersensitivity in male mice, suggesting the sex-dependent molecular aspects of spinal microglia in the regulation of pain.


Asunto(s)
Receptor 1 de Quimiocinas CX3C/metabolismo , Drogas de Diseño/química , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Hiperalgesia/patología , Microglía/metabolismo , Médula Espinal/metabolismo , Animales , Clozapina/análogos & derivados , Mediadores de Inflamación/metabolismo , Masculino , Ratones Transgénicos , Regulación hacia Arriba
9.
Front Pharmacol ; 11: 925, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32636748

RESUMEN

Despite growing evidence suggesting that spinal microglia play an important role in the molecular mechanism underlying experimental neuropathic pain (NP) in male rodents, evidence regarding the sex-dependent role of these microglia in NP is insufficient. In this study, we evaluated the effects of microglial regulation on NP using Gi-designer receptors exclusively activated by designer drugs (Gi-DREADD) driven by the microglia-specific Cx3cr1 promoter. For the Cre-dependent expression of human Gi-coupled M4 muscarinic receptors (hM4Di) in CX3C chemokine receptor 1-expressing (CX3CR1+) cells, R26-LSL-hM4Di-DREADD mice were crossed with CX3CR1-Cre mice. Mouse models of NP were generated by partial sciatic nerve ligation (PSL) and treatment with anti-cancer agent paclitaxel (PTX) or oxaliplatin (OXA), and mechanical allodynia was evaluated using the von Frey test. Immunohistochemistry revealed that hM4Di was specifically expressed on Iba1+ microglia, but not on astrocytes or neurons in the spinal dorsal horn of CX3CR1-hM4Di mice. PSL-induced mechanical allodynia was significantly attenuated by systemic (intraperitoneal, i.p.) administration of 10 mg/kg of clozapine N-oxide (CNO), a hM4Di-selective ligand, in male CX3CR1-hM4Di mice. The mechanical threshold in naive CX3CR1-hM4Di mice was not altered by i.p. administration of CNO. Consistently, local (intrathecal, i.t.) administration of CNO (20 nmol) significantly relieved PSL-induced mechanical allodynia in male CX3CR1-hM4Di mice. However, neither i.p. nor i.t. administration of CNO affected PSL-induced mechanical allodynia in female CX3CR1-hM4Di mice. Both i.p. and i.t. administration of CNO relieved PTX-induced mechanical allodynia in male CX3CR1-hM4Di mice, and a limited effect of i.p. CNO was observed in female CX3CR1-hM4Di mice. Unlike PTX-induced allodynia, OXA-induced mechanical allodynia was slightly improved, but not significantly relieved, by i.p. administration of CNO in both male and female CX3CR1-hM4Di mice. These results suggest that spinal microglia can be regulated by Gi-DREADD and support the notion that CX3CR1+ spinal microglia play sex-dependent roles in nerve injury-induced NP; however, their roles may vary among different models of NP.

10.
Neuropsychopharmacol Rep ; 40(3): 287-290, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32584520

RESUMEN

AIM: Ample evidence indicates that gastrin-releasing peptide receptor (GRPR)-expressing neurons play a critical role in the transmission of acute itch. However, the pathophysiology of spinal mechanisms underlying intractable itch such as psoriasis remains unclear. In this study, we aimed to determine whether itch-responsive GRPR+ neurons contribute to the spinal transmission of imiquimod (IMQ)-induced psoriatic itch. METHODS: To generate a psoriasis model, C57BL/6J mice received a daily topical application of 5% IMQ cream on their shaved back skin for 7-10 consecutive days. GRP+ neurons were inhibited using Cre-dependent expression of Gi-designer receptors exclusively activated by designer drugs (DREADDs), while GRPR+ neurons were ablated by intrathecal administration of bombesin-saporin. RESULTS: Repeated topical application of IMQ elicited psoriasis-like dermatitis and scratching behaviors. The mRNA expression levels of GRP and GRPR were upregulated in the cervical spinal dorsal horn (SDH) on days 7 and 10 after IMQ application. Either chemogenetic silencing of GRP+ neurons by Gi-DREADD or ablation of GRPR+ neurons significantly attenuated IMQ-induced scratching behaviors. CONCLUSION: The GRP-GRPR system might be enhanced in the SDH, and itch-responsive GRPR+ neurons largely contribute to intractable itch in a mouse model of psoriasis.


Asunto(s)
Imiquimod/toxicidad , Neuronas/metabolismo , Células del Asta Posterior/metabolismo , Prurito/inducido químicamente , Prurito/metabolismo , Receptores de Bombesina/biosíntesis , Adyuvantes Inmunológicos , Animales , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Células del Asta Posterior/efectos de los fármacos , Prurito/genética , Receptores de Bombesina/genética
11.
Curr Top Med Chem ; 20(31): 2878-2888, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32384033

RESUMEN

Despite similar distribution patterns and intracellular events observed in the nociceptin/ orphanin FQ peptide (NOP) receptor and other opioid receptors, NOP receptor activation displays unique pharmacological profiles. Several researchers have identified a variety of peptide and nonpeptide ligands to determine the functional roles of NOP receptor activation and observed that NOP receptor- related ligands exhibit pain modality-dependent pain processing. Importantly, NOP receptor activation results in anti-nociception and anti-hypersensitivity at the spinal and supraspinal levels regardless of the experimental settings in non-human primates (NHPs). Given that the NOP receptor agonists synergistically enhance mu-opioid peptide (MOP) receptor agonist-induced anti-nociception, it has been hypothesized that dual NOP and MOP receptor agonists may display promising functional properties as analgesics. Accumulating evidence indicates that the mixed NOP/opioid receptor agonists demonstrate favorable functional profiles. In NHP studies, bifunctional NOP/MOP partial agonists (e.g., AT-121, BU08028, and BU10038) exerted potent anti-nociception via NOP and MOP receptor activation; however, dose-limiting adverse effects associated with the MOP receptor activation, including respiratory depression, itch sensation, physical dependence, and abuse liability, were not observed. Moreover, a mixed NOP/opioid receptor agonist, cebranopadol, presented promising outcomes in clinical trials as a novel analgesic. Collectively, the dual agonistic actions on NOP and MOP receptors, with appropriate binding affinities and efficacies, may be a viable strategy to develop innovative and safe analgesics.


Asunto(s)
Analgésicos Opioides/farmacología , Isoquinolinas/farmacología , Naltrexona/análogos & derivados , Fenilpropionatos/farmacología , Receptores Opioides/metabolismo , Analgésicos Opioides/química , Humanos , Isoquinolinas/química , Ligandos , Naltrexona/química , Naltrexona/farmacología , Dolor/tratamiento farmacológico , Fenilpropionatos/química
12.
Neuropharmacology ; 170: 108025, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32142790

RESUMEN

Gastrin-releasing peptide (GRP) receptor-expressing (GRPR)+ neurons have a central role in the spinal transmission of itch. Because their fundamental regulatory mechanisms are not yet understood, it is important to determine how such neurons are excited and integrate itch sensation. In this study, we investigated the mechanisms for the activation of itch-responsive GRPR+ neurons in the spinal dorsal horn (SDH). GRPR+ neurons expressed the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) containing the GluR2 subunit. In mice, peripherally elicited histaminergic and non-histaminergic itch was prevented by intrathecal (i.t.) administration of the AMPAR antagonist NBQX, which was consistent with the fact that firing of GRPR+ neurons in SDH under histaminergic and non-histaminergic itch was completely blocked by NBQX, but not by the GRPR antagonist RC-3095. Because GRP+ neurons in SDH contain glutamate, we investigated the role of GRP+ (GRP+/Glu+) neurons in regulating itch. Chemogenetic inhibition of GRP+ neurons suppressed both histaminergic and non-histaminergic itch without affecting the mechanical pain threshold. In nonhuman primates, i.t. administration of NBQX also attenuated peripherally elicited itch without affecting the thermal pain threshold. In a mouse model of diphenylcyclopropenone (DCP)-induced contact dermatitis, GRP, GRPR, and AMPAR subunits were upregulated in SDH. DCP-induced itch was prevented by either silencing GRP+ neurons or ablation of GRPR+ neurons. Altogether, these findings demonstrate that GRP and glutamate cooperatively regulate GRPR+ AMPAR+ neurons in SDH, mediating itch sensation. GRP-GRPR and the glutamate-AMPAR system may play pivotal roles in the spinal transmission of itch in rodents and nonhuman primates.


Asunto(s)
Neuronas/metabolismo , Prurito/metabolismo , Receptores AMPA/metabolismo , Receptores de Bombesina/metabolismo , Asta Dorsal de la Médula Espinal/metabolismo , Animales , Bombesina/análogos & derivados , Bombesina/farmacología , Ciclopropanos/toxicidad , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/farmacología , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Prurito/inducido químicamente , Receptores AMPA/antagonistas & inhibidores , Receptores de Bombesina/antagonistas & inhibidores , Asta Dorsal de la Médula Espinal/efectos de los fármacos
13.
Artículo en Inglés | MEDLINE | ID: mdl-32087968

RESUMEN

Peripheral nerve injury typically leads to chronic inflammation through recruitment of immune cells, which may induce neuropathic pain. We previously reported that M1-like macrophages at sites of peripheral nerve injury induced neuropathic pain; however, the involvement of other immune cells (e.g. M2-like macrophages) in the progression of neuropathic pain remains unclear. In addition, the immune responses that occur at sites of nerve injury have not been well characterized. In this study, we show that M2-like macrophages accumulate in injured nerves to participate in the clearance of dead or dying cells (i.e., efferocytosis). Because MerTK (a receptor of dead or dying cells) levels on the surface of macrophages are limited, it seems to induce the insufficient of efferocytosis, such that the levels of dead or dying cells cannot be controlled in injured nerves. Given that efferocytosis is pivotal for resolution of inflammation, our data suggest that insufficient efferocytosis is a contributing factor in the development of chronic inflammation in injured nerves.

14.
J Pharmacol Exp Ther ; 368(3): 535-544, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30602591

RESUMEN

Despite the requirement for effective medication against neuropathic pain associated with type 2 diabetes mellitus (T2DM), mechanism-based pharmacotherapy has yet to be established. Given that long-lasting neuroinflammation, driven by inflammatory macrophages in the peripheral nerves, plays a pivotal role in intractable pain, it is important to determine whether inflammatory macrophages contribute to neuropathic pain associated with T2DM. To generate an experimental model of T2DM, C57BL/6J mice were fed a high-fat diet (HFD) ad libitum. Compared with control diet feeding, obesity and hyperglycemia were observed after HFD feeding, and the mechanical pain threshold evaluated using the von Frey test was found to be decreased, indicating the development of mechanical allodynia. The expression of mRNA markers for macrophages, inflammatory cytokines, and chemokines were significantly upregulated in the sciatic nerve (SCN) after HFD feeding. Perineural administration of saporin-conjugated anti-Mac1 antibody (Mac1-Sap) improved HFD-induced mechanical allodynia. Moreover, treatment of Mac1-Sap decreased the accumulation of F4/80+ macrophages and the upregulation of inflammatory mediators in the SCN after HFD feeding. Inoculation of lipopolysaccharide-activated peritoneal macrophages in tissue surrounding the SCN elicited mechanical allodynia. Furthermore, pharmacological inhibition of inflammatory macrophages by either perineural or systemic administration of TC-2559 [4-(5-ethoxy-3-pyridinyl)-N-methyl-(3E)-3-buten-1-amine difumarate], a α4ß2 nicotinic acetylcholine receptor-selective agonist, relieved HFD-induced mechanical allodynia. Taken together, inflammatory macrophages that accumulate in the SCN mediate the pathophysiology of neuropathic pain associated with T2DM. Inhibitory agents for macrophage-driven neuroinflammation could be potential candidates for novel pharmacotherapy against intractable neuropathic pain.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Neuralgia/metabolismo , Nervio Ciático/metabolismo , Animales , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/patología , Inflamación/inmunología , Inflamación/metabolismo , Mediadores de Inflamación/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuralgia/inmunología , Neuralgia/patología , Nervio Ciático/inmunología , Nervio Ciático/patología
15.
Neuropsychopharmacol Rep ; 38(3): 145-148, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30175527

RESUMEN

AIM: We have previously demonstrated that upregulation of CC chemokines through dopamine receptor signaling in the prefrontal cortex (PFC) underlies methamphetamine (Meth)-induced reward. Given the common pharmacological property of Meth and cocaine (Coca), which are highly addictive psychostimulants, we hypothesized that chemokines may also contribute to Coca-induced reward. The aim of this study was to identify a key chemokine-mediating Coca-induced reward in mice. METHODS: The mRNA expression levels of chemokines were measured by reverse transcription-quantitative polymerase chain reaction. Coca-induced reward was evaluated by conditioned place preference test. RESULTS: We found that mRNA expression levels of CC chemokine ligand 2 (CCL2), CCL7, and CXC chemokine ligand 1 (CXCL1) were upregulated in the PFC after a single administration of Coca (20 mg/kg, s.c.). Upregulation of CXCL1, but not CCL2 and CCL7, mRNA in the PFC was also observed after repeated administration of Coca. A single administration of dopamine D1 receptor agonist SKF 81297 (10 mg/kg, s.c.), but not D2 receptor agonist sumanirole, upregulated CXCL1 mRNA in the PFC. Coca-induced reward was attenuated by the pretreatment of SB 225002 (5 mg/kg, s.c.), a selective antagonist of CXC chemokine receptor 2 (CXCR2, cognate receptor for CXCL1). CONCLUSIONS: Collectively, we identified CXCL1 as a key regulator in Coca-induced reward and propose that pharmacological approach targeting CXCL1 could be a novel pharmacotherapy for Coca-induced reward.


Asunto(s)
Quimiocina CXCL1/genética , Trastornos Relacionados con Cocaína/metabolismo , Recompensa , Animales , Benzazepinas/farmacología , Bencimidazoles/farmacología , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CXCL1/antagonistas & inhibidores , Quimiocina CXCL1/metabolismo , Cocaína/farmacología , Trastornos Relacionados con Cocaína/fisiopatología , Agonistas de Dopamina/farmacología , Inhibidores de Captación de Dopamina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Compuestos de Fenilurea/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
16.
Sci Transl Med ; 10(456)2018 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-30158150

RESUMEN

Misuse of prescription opioids, opioid addiction, and overdose underscore the urgent need for developing addiction-free effective medications for treating severe pain. Mu opioid peptide (MOP) receptor agonists provide very effective pain relief. However, severe side effects limit their use in the clinical setting. Agonists of the nociceptin/orphanin FQ peptide (NOP) receptor have been shown to modulate the antinociceptive and reinforcing effects of MOP agonists. We report the discovery and development of a bifunctional NOP/MOP receptor agonist, AT-121, which has partial agonist activity at both NOP and MOP receptors. AT-121 suppressed oxycodone's reinforcing effects and exerted morphine-like analgesic effects in nonhuman primates. AT-121 treatment did not induce side effects commonly associated with opioids, such as respiratory depression, abuse potential, opioid-induced hyperalgesia, and physical dependence. Our results in nonhuman primates suggest that bifunctional NOP/MOP agonists with the appropriate balance of NOP and MOP agonist activity may provide a dual therapeutic action for safe and effective pain relief and treating prescription opioid abuse.


Asunto(s)
Analgésicos Opioides/farmacología , Péptidos Opioides/farmacología , Receptores Opioides mu/agonistas , Analgésicos Opioides/química , Analgésicos Opioides/uso terapéutico , Animales , Diseño de Fármacos , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/patología , Hiperalgesia/fisiopatología , Ligandos , Morfina/administración & dosificación , Morfina/farmacología , Morfina/uso terapéutico , Nocicepción/efectos de los fármacos , Péptidos Opioides/administración & dosificación , Péptidos Opioides/química , Péptidos Opioides/uso terapéutico , Oxicodona/farmacología , Oxicodona/uso terapéutico , Primates , Receptores Opioides mu/metabolismo , Relación Estructura-Actividad , Nociceptina
17.
J Diabetes ; 10(11): 886-895, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29749036

RESUMEN

BACKGROUND: Given that diabetes-associated complications are closely associated with neuroinflammation, it is imperative to study potential changes in neuroinflammatory modulators in the central nervous system of diabetic primates. METHODS: The mRNA levels of pro- and anti-inflammatory cytokines, toll-like receptors (TLRs), growth factors, and cannabinoid receptors were compared in the spinal dorsal horn (SDH) and thalamus of naturally occurring type 2 diabetic monkeys and an age-matched control group using reverse transcription and quantitative real-time polymerase chain reaction. RESULTS: In the SDH of diabetic monkeys, mRNA levels of proinflammatory cytokines (i.e. interleukin [IL]-1ß and tumor necrosis factor [TNF] α), TLR1, and TLR2 were increased, whereas mRNA levels of IL-10, an anti-inflammatory cytokine, were decreased. No changes were observed in the mRNA levels of growth factors and cannabinoid receptors. In line with the mRNA data, TNFα immunoreactivity was significantly increased in diabetic monkeys. Moreover, mRNA expression levels of IL-1ß, TNFα, TLR1, and TLR2 in the SDH were positively correlated with plasma glucose concentrations in all monkeys. CONCLUSIONS: Several ligands and receptors involved in neuroinflammation are simultaneously dysregulated in the spinal cord of diabetic monkeys. This primate disease model will facilitate the design of novel treatment approaches to ameliorate neuroinflammation-driven adverse effects in diabetic patients.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Mediadores de Inflamación/metabolismo , Proteínas del Tejido Nervioso/genética , ARN Mensajero/genética , Asta Dorsal de la Médula Espinal/metabolismo , Tálamo/metabolismo , Animales , Glucemia/metabolismo , Citocinas/genética , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Macaca fascicularis , Masculino , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso/metabolismo , ARN Mensajero/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
18.
J Neuroinflammation ; 15(1): 96, 2018 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-29587798

RESUMEN

BACKGROUND: Neuro-immune interaction underlies chronic neuroinflammation and aberrant sensory processing resulting in neuropathic pain. Despite the pathological significance of both neuroinflammation-driven peripheral sensitization and spinal sensitization, the functional relationship between these two distinct events has not been understood. METHODS: In this study, we determined whether inhibition of inflammatory macrophages by administration of α4ß2 nicotinic acetylcholine receptor (nAChR) agonists improves neuropathic pain and affects microglial activation in the spinal dorsal horn (SDH) in mice following partial sciatic nerve ligation (PSL). Expression levels of neuroinflammatory molecules were evaluated by RT-qPCR and immunohistochemistry, and PSL-induced mechanical allodynia was defined by the von Frey test. RESULTS: Flow cytometry revealed that CD11b+ F4/80+ macrophages were accumulated in the injured sciatic nerve (SCN) after PSL. TC-2559, a full agonist for α4ß2 nAChR, suppressed the upregulation of interleukin-1ß (IL-1ß) in the injured SCN after PSL and attenuated lipopolysaccharide-induced upregulation of IL-1ß in cultured macrophages. Systemic (subcutaneous, s.c.) administration of TC-2559 during either the early (days 0-3) or middle/late (days 7-10) phase of PSL improved mechanical allodynia. Moreover, local (perineural, p.n.) administration of TC-2559 and sazetidine A, a partial agonist for α4ß2 nAChR, during either the early or middle phase of PSL improved mechanical allodynia. However, p.n. administration of sazetidine A during the late (days 21-24) phase did not show the attenuating effect, whereas p.n. administration of TC-2559 during this phase relieved mechanical allodynia. Most importantly, p.n. administration of TC-2559 significantly suppressed morphological activation of Iba1+ microglia and decreased the upregulation of inflammatory microglia-dominant molecules, such as CD68, interferon regulatory factor 5, and IL-1ß in the SDH after PSL. CONCLUSION: These findings support the notion that pharmacological inhibition of inflammatory macrophages using an α4ß2 nAChR agonist exhibit a wide therapeutic window on neuropathic pain after nerve injury, and it could be nominated as a novel pharmacotherapy to relieve intractable pain.


Asunto(s)
Macrófagos/efectos de los fármacos , Microglía/efectos de los fármacos , Agonistas Nicotínicos/uso terapéutico , Neuropatía Ciática/tratamiento farmacológico , Neuropatía Ciática/patología , Médula Espinal/patología , Animales , Citocinas/genética , Citocinas/metabolismo , Dihidro-beta-Eritroidina/uso terapéutico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Lateralidad Funcional/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hiperalgesia/tratamiento farmacológico , Activación de Macrófagos , Masculino , Ratones , Ratones Endogámicos ICR , Agonistas Nicotínicos/farmacología , Umbral del Dolor/efectos de los fármacos , Piridinas/farmacología , Piridinas/uso terapéutico , ARN Mensajero/metabolismo , Factores de Tiempo
19.
Neurosci Lett ; 665: 33-37, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29174638

RESUMEN

We previously showed that the CC-chemokine ligand 2 (CCL2)-CC-chemokine receptor 2 (CCR2) system is responsible for conditioned place preference (CPP) by methamphetamine (Meth). In this study, we investigated the roles for other chemokines mediating Meth-induced CPP and the upstream factors upregulating chemokines in mice. We found that CCL7 mRNA level was upregulated in the prefrontal cortex (PFC) after Meth administration (3mg/kg, subcutaneous), and increased CCL7 immunoreactivity was localized to the PFC NeuN-positive neurons. Meth-induced CPP was blocked by the dopamine D1 receptor antagonist SCH 23390 but not by the D2 receptor antagonists raclopride or haloperidol. The D1 receptor agonist SKF 81297 alone elicited CPP, suggesting a critical role of D1 receptor signaling in Meth-induced reward. Consistent with these results, the Meth-induced upregulation of CCL7 and CCL2 were attenuated by SCH 23390, and a single administration of SKF 81297 upregulated mRNA expression levels of CCL7 and CCL2 in the PFC. Furthermore, Meth-induced CPP was prevented by INCB 3284, a selective antagonist of CCR2, a receptor that binds both CCL7 and CCL2. Collectively, we identified two CC-chemokines (i.e., CCL7 and CCL2) as key regulatory factors in Meth-induced reward. Pharmacological inhibitors of these chemokines may warrant development as novel therapeutics for ameliorating Meth addiction.


Asunto(s)
Conducta Adictiva/tratamiento farmacológico , Quimiocina CCL2/metabolismo , Quimiocina CCL7/metabolismo , Receptores de Dopamina D1/metabolismo , Animales , Estimulantes del Sistema Nervioso Central/farmacología , Condicionamiento Operante/efectos de los fármacos , Antagonistas de Dopamina/farmacología , Masculino , Metanfetamina/farmacología , Ratones Endogámicos C57BL , Recompensa , Regulación hacia Arriba/efectos de los fármacos
20.
Int J Mol Sci ; 18(11)2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-29104252

RESUMEN

Neuropathic pain can have a major effect on quality of life but current therapies are often inadequate. Growing evidence suggests that neuropathic pain induced by nerve damage is caused by chronic inflammation. Upon nerve injury, damaged cells secrete pro-inflammatory molecules that activate cells in the surrounding tissue and recruit circulating leukocytes to the site of injury. Among these, the most abundant cell type is macrophages, which produce several key molecules involved in pain enhancement, including cytokines and chemokines. Given their central role in the regulation of peripheral sensitization, macrophage-derived cytokines and chemokines could be useful targets for the development of novel therapeutics. Inhibition of key pro-inflammatory cytokines and chemokines prevents neuroinflammation and neuropathic pain; moreover, recent studies have demonstrated the effectiveness of pharmacological inhibition of inflammatory (M1) macrophages. Nicotinic acetylcholine receptor ligands and T helper type 2 cytokines that reduce M1 macrophages are able to relieve neuropathic pain. Future translational studies in non-human primates will be crucial for determining the regulatory mechanisms underlying neuroinflammation-associated neuropathic pain. In turn, this knowledge will assist in the development of novel pharmacotherapies targeting macrophage-driven neuroinflammation for the treatment of intractable neuropathic pain.


Asunto(s)
Descubrimiento de Drogas , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Neuralgia/complicaciones , Neuralgia/tratamiento farmacológico , Animales , Citocinas/inmunología , Descubrimiento de Drogas/métodos , Humanos , Inflamación/patología , Ligandos , Macrófagos/inmunología , Macrófagos/patología , Neuralgia/patología , Receptores Nicotínicos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA