Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
J Pain ; 23(2): 326-336, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34547430

RESUMEN

Nociceptive pain involves the activation of nociceptors without damage to the nervous system, whereas neuropathic pain is related to an alteration in the central or peripheral nervous system. Chronic pain itself and the transition from acute to chronic pain may be epigenetically controlled. In this cross-sectional study, a genome-wide DNA methylation analysis was performed using the blood DNA reduced representation bisulfite sequencing (RRBS) technique. Three prospective cohorts including 20 healthy controls (CTL), 18 patients with chronic nociceptive pain (NOCI), and 19 patients with chronic neuropathic pain (NEURO) were compared at both the single CpG and differentially methylated region (DMR) levels. Genes with DMRs were seen in the NOCI and NEURO groups belonged to the neuro-musculoskeletal system and differed between NOCI and NEURO patients. Our results demonstrate that the epigenetic disturbances accompanying nociceptive pain are very different from those accompanying neuropathic pain. In the former, among others, the epigenetic disturbance observed would affect the function of the opioid analgesic system, whereas in the latter it would affect that of the GABAergic reward system. This study presents biological findings that help to characterize NOCI- and NEURO-affected pathways and opens the possibility of developing epigenetic diagnostic assays. PERSPECTIVE: Our results help to explain the various biological pathways modifications underlying the different clinical manifestations of nociceptive and neuropathic pains. Furthermore, the new targets identified in our study might help to discover more specific treatments for nociceptive or neuropathic pains.


Asunto(s)
Dolor Crónico/genética , Epigenoma/genética , Estudios de Asociación Genética , Neuralgia/genética , Dolor Nociceptivo/genética , Adulto , Estudios de Cohortes , Metilación de ADN/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Sistema Musculoesquelético/metabolismo , Sistema Nervioso/metabolismo
2.
Nat Commun ; 12(1): 2936, 2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-34006861

RESUMEN

Host protection against cutaneous herpes simplex virus 1 (HSV-1) infection relies on the induction of a robust adaptive immune response. Here, we show that Nav1.8+ sensory neurons, which are involved in pain perception, control the magnitude of CD8 T cell priming and expansion in HSV-1-infected mice. The ablation of Nav1.8-expressing sensory neurons is associated with extensive skin lesions characterized by enhanced inflammatory cytokine and chemokine production. Mechanistically, Nav1.8+ sensory neurons are required for the downregulation of neutrophil infiltration in the skin after viral clearance to limit the severity of tissue damage and restore skin homeostasis, as well as for eliciting robust CD8 T cell priming in skin-draining lymph nodes by controlling dendritic cell responses. Collectively, our data reveal an important role for the sensory nervous system in regulating both innate and adaptive immune responses to viral infection, thereby opening up possibilities for new therapeutic strategies.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Dolor Nociceptivo/inmunología , Células Receptoras Sensoriales/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Herpes Simple/genética , Herpes Simple/virología , Herpesvirus Humano 1/fisiología , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Canal de Sodio Activado por Voltaje NAV1.8/genética , Canal de Sodio Activado por Voltaje NAV1.8/inmunología , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Infiltración Neutrófila/inmunología , Dolor Nociceptivo/genética , Dolor Nociceptivo/metabolismo , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/virología , Piel/inmunología , Piel/metabolismo , Piel/virología
3.
Eur J Pharmacol ; 899: 174007, 2021 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-33705801

RESUMEN

G-protein-biased agonists with reduced ß-arrestin-2 activation are being investigated as safer alternatives to clinically-used opioids. ß-arrestin-2 has been implicated in the mechanism of opioid-induced antinociceptive tolerance. Opioid-induced analgesic tolerance is classically considered as centrally-mediated, but recent reports implicate nociceptive dorsal root ganglia neurons as critical mediators in this process. Here, we investigated the role of ß-arrestin-2 in the mechanism of opioid tolerance in dorsal root ganglia nociceptive neurons using ß-arrestin-2 knockout mice and the G-protein-biased µ-opioid receptor agonist, TRV130. Whole-cell current-clamp electrophysiology experiments revealed that 15-18-h overnight exposure to 10 µM morphine in vitro induced acute tolerance in ß-arrestin-2 wild-type but not knockout neurons. Furthermore, in wild-type neurons circumventing ß-arrestin-2 activation by overnight treatment with 200 nM TRV130 attenuated tolerance. Similarly, acute morphine tolerance in vivo in ß-arrestin-2 knockout mice was prevented in the warm-water tail-withdrawal assay. Treatment with 30 mg/kg TRV130 s.c. also inhibited acute antinociceptive tolerance in vivo in wild-type mice. Alternately, in ß-arrestin-2 knockout neurons tolerance induced by 7-day in vivo exposure to 50 mg morphine pellet was conserved. Likewise, ß-arrestin-2 deletion did not mitigate in vivo antinociceptive tolerance induced by 7-day exposure to 25 mg or 50 mg morphine pellet in both female or male mice, respectively. Consequently, these results indicated that ß-arrestin-2 mediates acute but not chronic opioid tolerance in dorsal root ganglia neurons and to antinociception in vivo. This suggests that opioid-induced antinociceptive tolerance may develop even in the absence of ß-arrestin-2 activation, and thus significantly affect the clinical utility of biased agonists.


Asunto(s)
Analgésicos Opioides/farmacología , Tolerancia a Medicamentos , Ganglios Espinales/efectos de los fármacos , Morfina/farmacología , Neuronas/efectos de los fármacos , Dolor Nociceptivo/prevención & control , Receptores Opioides mu/agonistas , Compuestos de Espiro/farmacología , Tiofenos/farmacología , Arrestina beta 2/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Ganglios Espinales/metabolismo , Ganglios Espinales/fisiopatología , Masculino , Ratones Noqueados , Neuronas/metabolismo , Dolor Nociceptivo/genética , Dolor Nociceptivo/metabolismo , Dolor Nociceptivo/fisiopatología , Umbral del Dolor/efectos de los fármacos , Receptores Opioides mu/metabolismo , Factores de Tiempo , Arrestina beta 2/deficiencia , Arrestina beta 2/genética
4.
Toxicol Appl Pharmacol ; 414: 115428, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33524449

RESUMEN

Researches have shown that calcitonin gene-related peptide (CGRP) plays a pivotal role in pain modulation. Nociceptive information from the periphery is relayed from parabrachial nucleus (PBN) to brain regions implicated involved in pain. This study investigated the effects and mechanisms of CGRP and CGRP receptors in pain regulation in the PBN of naive and neuropathic pain rats. Chronic sciatic nerve ligation was used to model neuropathic pain, CGRP and CGRP 8-37 were injected into the PBN of the rats, and calcitonin receptor-like receptor (CLR), a main structure of CGRP receptor, was knocked down by lentivirus-coated CLR siRNA. The hot plate test (HPT) and the Randall Selitto Test (RST) was used to determine the latency of the rat hindpaw response. The expression of CLR was detected with RT-PCR and western blotting. We found that intra-PBN injecting of CGRP induced an obvious anti-nociceptive effect in naive and neuropathic pain rats in a dose-dependent manner, the CGRP-induced antinociception was significantly reduced after injection of CGRP 8-37, Moreover, the mRNA and protein levels of CLR, in PBN decreased significantly and the antinociception CGRP-induced was also significantly lower in neuropathic pain rats than that in naive rats. Knockdown CLR in PBN decreased the expression of CLR and the antinociception induced by CGRP was observably decreased. Our results demonstrate that CGRP induced antinociception in PBN of naive or neuropathic pain rats, CGRP receptor mediates this effect. Neuropathic pain induced decreases in the expression of CGRP receptor, as well as in CGRP-induced antinociception in PBN.


Asunto(s)
Analgésicos/farmacología , Péptido Relacionado con Gen de Calcitonina/farmacología , Proteína Similar al Receptor de Calcitonina/agonistas , Dolor Nociceptivo/prevención & control , Umbral del Dolor/efectos de los fármacos , Núcleos Parabraquiales/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Receptores de Péptido Relacionado con el Gen de Calcitonina/agonistas , Ciática/prevención & control , Animales , Proteína Similar al Receptor de Calcitonina/genética , Proteína Similar al Receptor de Calcitonina/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Masculino , Dolor Nociceptivo/genética , Dolor Nociceptivo/metabolismo , Dolor Nociceptivo/fisiopatología , Núcleos Parabraquiales/metabolismo , Núcleos Parabraquiales/fisiopatología , Ratas Sprague-Dawley , Receptores de Péptido Relacionado con el Gen de Calcitonina/genética , Receptores de Péptido Relacionado con el Gen de Calcitonina/metabolismo , Ciática/genética , Ciática/metabolismo , Ciática/fisiopatología
5.
J Biol Chem ; 296: 100326, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33493520

RESUMEN

Human voltage-gated sodium channel Nav1.7 (hNav1.7) is involved in the generation and conduction of neuropathic and nociceptive pain signals. Compelling genetic and preclinical studies have validated that hNav1.7 is a therapeutic target for the treatment of pain; however, there is a dearth of currently available compounds capable of targeting hNav1.7 with high potency and specificity. Hainantoxin-III (HNTX-III) is a 33-residue polypeptide from the venom of the spider Ornithoctonus hainana. It is a selective antagonist of neuronal tetrodotoxin-sensitive voltage-gated sodium channels. Here, we report the engineering of improved potency and Nav selectivity of hNav1.7 inhibition peptides derived from the HNTX-III scaffold. Alanine scanning mutagenesis showed key residues for HNTX-III interacting with hNav1.7. Site-directed mutagenesis analysis indicated key residues on hNav1.7 interacting with HNTX-III. Molecular docking was conducted to clarify the binding interface between HNTX-III and Nav1.7 and guide the molecular engineering process. Ultimately, we obtained H4 [K0G1-P18K-A21L-V] based on molecular docking of HNTX-III and hNav1.7 with a 30-fold improved potency (IC50 0.007 ± 0.001 µM) and >1000-fold selectivity against Nav1.4 and Nav1.5. H4 also showed robust analgesia in the acute and chronic inflammatory pain model and neuropathic pain model. Thus, our results provide further insight into peptide toxins that may prove useful in guiding the development of inhibitors with improved potency and selectivity for Nav subtypes with robust analgesia.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/genética , Dolor Nociceptivo/tratamiento farmacológico , Péptidos/genética , Venenos de Araña/química , Animales , Humanos , Simulación del Acoplamiento Molecular , Canal de Sodio Activado por Voltaje NAV1.4/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.4/genética , Canal de Sodio Activado por Voltaje NAV1.5/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.7/química , Canal de Sodio Activado por Voltaje NAV1.7/efectos de los fármacos , Dolor Nociceptivo/genética , Dolor Nociceptivo/patología , Péptidos/química , Péptidos/farmacología , Venenos de Araña/genética
6.
Gene ; 742: 144583, 2020 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-32184167

RESUMEN

BACKGROUND: Studies showed that increased let-7b-5p microRNA during repeated electroacupuncture (EA) treatment was associated the formation of EA tolerance, which manifested as gradually decreased nociceptive threshold. Proenkephalin (PENK) is the precursor of enkephalin which is a pivot neuropeptide responsible for the decreased nociceptive threshold in EA. The aim of this study was to evaluate the relationship between let-7b-5p and PENK in EA tolerance. METHODS: The target gene of let-7b-5p microRNA was determined through the dual-luciferase reporter assay in cortical neurons. Seventy-two Sprague Dawley rats received a combination of EA and intracerebroventricular injection of microRNA (let-7b-5p agomir, antagomir or their controls). The nociceptive thresholds were assessed with radiant heat tail-flick method. PENK and let-7b-5p were measured with Western Blot and qPCR, respectively, after administration of let-7b-5p agomir, antagomir, and their controls at day 1, 4 and 7. RESULTS: Let-7b-5p targeted the 3' untranslated region of Penk1. The nociceptive thresholds in Let-7b-5p agomir + EA group were decreased (p < 0.05) compared with those in Let-7b-5p antagomir + EA group at day 1 to 7. Compared with Let-7b-5p agomir + EA group, the expression level of PENK in Let-7b-5p antagomir + EA group was increased at days 1, 4, and 7 (p < 0.05) CONCLUSION: Let-7b-5p may be a new potential target for decreasing the EA tolerance effect and facilitating the application of EA in treating chronic nociception of patients.


Asunto(s)
Electroacupuntura , Encefalinas/genética , MicroARNs/metabolismo , Dolor Nociceptivo/terapia , Precursores de Proteínas/genética , Animales , Antagomirs/administración & dosificación , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Adyuvante de Freund/administración & dosificación , Adyuvante de Freund/inmunología , Humanos , Inyecciones Intraventriculares , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , Nocicepción/efectos de los fármacos , Dolor Nociceptivo/diagnóstico , Dolor Nociceptivo/genética , Dolor Nociceptivo/inmunología , Umbral del Dolor/efectos de los fármacos , Ratas
7.
Artículo en Inglés | MEDLINE | ID: mdl-32117068

RESUMEN

Various types of acute/chronic nociceptive stimuli cause neuroendocrine responses such as activation of the hypothalamo-neurohypophysial [oxytocin (OXT) and arginine vasopressin (AVP)] system and hypothalamo-pituitary adrenal (HPA) axis. Chronic multiple-arthritis activates the OXT/AVP system, but the effects of acute mono-arthritis on the OXT/AVP system in the same animals has not been simultaneously evaluated. Further, AVP, not corticotropin-releasing hormone (CRH), predominantly activates the HPA axis in chronic multiple-arthritis, but the participation of AVP in HPA axis activation in acute mono-arthritis remains unknown. Therefore, we aimed to simultaneously evaluate the effects of acute mono-arthritis on the activity of the OXT/AVP system and the HPA axis. In the present study, we used an acute mono-arthritic model induced by intra-articular injection of carrageenan in a single knee joint of adult male Wistar rats. Acute mono-arthritis was confirmed by a significant increase in knee diameter in the carrageenan-injected knee and a significant decrease in the mechanical nociceptive threshold in the ipsilateral hind paw. Immunohistochemical analysis revealed that the number of Fos-immunoreactive (ir) cells in the ipsilateral lamina I-II of the dorsal horn was significantly increased, and the percentage of OXT-ir and AVP-ir neurons expressing Fos-ir in both sides of the supraoptic (SON) and paraventricular nuclei (PVN) was increased in acute mono-arthritic rats. in situ hybridization histochemistry revealed that levels of OXT mRNA and AVP hnRNA in the SON and PVN, CRH mRNA in the PVN, and proopiomelanocortin mRNA in the anterior pituitary were also significantly increased in acute mono-arthritic rats. Further, plasma OXT, AVP, and corticosterone levels were significantly increased in acute mono-arthritic rats. These results suggest that acute mono-arthritis activates ipsilateral nociceptive afferent pathways at the spinal level and causes simultaneous and integrative activation of the OXT/AVP system. In addition, the HPA axis is activated by both AVP and CRH in acute mono-arthritis with a distinct pattern compared to that in chronic multiple-arthritis.


Asunto(s)
Artritis/fisiopatología , Sistema Hipotálamo-Hipofisario/fisiopatología , Sistema Hipófiso-Suprarrenal/fisiopatología , Enfermedad Aguda , Vías Aferentes/fisiología , Animales , Arginina Vasopresina/sangre , Arginina Vasopresina/genética , Artritis/genética , Artritis/metabolismo , Artritis/patología , Hormona Liberadora de Corticotropina/sangre , Hormona Liberadora de Corticotropina/genética , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/patología , Masculino , Neuronas/fisiología , Dolor Nociceptivo/etiología , Dolor Nociceptivo/genética , Dolor Nociceptivo/metabolismo , Dolor Nociceptivo/fisiopatología , Osteoartritis de la Rodilla/genética , Osteoartritis de la Rodilla/metabolismo , Osteoartritis de la Rodilla/patología , Osteoartritis de la Rodilla/fisiopatología , Oxitocina/sangre , Oxitocina/genética , Sistema Hipófiso-Suprarrenal/metabolismo , Sistema Hipófiso-Suprarrenal/patología , Proopiomelanocortina/sangre , Proopiomelanocortina/genética , Ratas , Ratas Wistar
8.
Science ; 364(6443): 852-859, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31147513

RESUMEN

Noxious substances, called algogens, cause pain and are used as defensive weapons by plants and stinging insects. We identified four previously unknown instances of algogen-insensitivity by screening eight African rodent species related to the naked mole-rat with the painful substances capsaicin, acid (hydrogen chloride, pH 3.5), and allyl isothiocyanate (AITC). Using RNA sequencing, we traced the emergence of sequence variants in transduction channels, like transient receptor potential channel TRPA1 and voltage-gated sodium channel Nav1.7, that accompany algogen insensitivity. In addition, the AITC-insensitive highveld mole-rat exhibited overexpression of the leak channel NALCN (sodium leak channel, nonselective), ablating AITC detection by nociceptors. These molecular changes likely rendered highveld mole-rats immune to the stings of the Natal droptail ant. Our study reveals how evolution can be used as a discovery tool to find molecular mechanisms that shut down pain.


Asunto(s)
Evolución Molecular , Ratas Topo/fisiología , Canal de Sodio Activado por Voltaje NAV1.7/genética , Dolor Nociceptivo/genética , Umbral del Dolor , Canal Catiónico TRPA1/genética , Animales , Sitios de Unión , Capsaicina/farmacología , Ácido Clorhídrico/farmacología , Mordeduras y Picaduras de Insectos/genética , Mordeduras y Picaduras de Insectos/inmunología , Isotiocianatos/farmacología , Ratas Topo/genética , Ratas Topo/inmunología , Dolor Nociceptivo/inducido químicamente , Nociceptores/efectos de los fármacos , Nociceptores/fisiología , Conformación Proteica , Análisis de Secuencia de ARN , Especificidad de la Especie , Canal Catiónico TRPA1/química
9.
Am J Physiol Renal Physiol ; 317(1): F90-F98, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31091120

RESUMEN

Altered Toll-like receptor (TLR)4 activation has been identified in several chronic pain conditions but has not been well studied in interstitial cystitis/bladder pain syndrome (IC/BPS). Our previously published human studies indicated that patients with IC/BPS present altered systemic TLR4-mediated inflammatory responses, which were significantly correlated with reported pain severity. In the present study, we sought to determine whether altered TLR4 activation plays a role in pelvic/bladder pain seen in patients with IC/BPS using our validated IC/BPS-like transgenic autoimmune cystitis model (URO-OVA). URO-OVA mice developed responses consistent with pelvic and bladder pain after cystitis induction, which was associated with increased splenocyte production of TLR4-mediated proinflammatory cytokines IL-1ß, IL-6, and TNF-α. Increased spinal expression of mRNAs for proinflammatory cytokines IL-6 and TNF-α, glial activation markers CD11b and glial fibrillary acidic protein, and endogenous TLR4 ligand high mobility group box 1 was also observed after cystitis induction. Compared with URO-OVA mice, TLR4-deficient URO-OVA mice developed significantly reduced nociceptive responses, although similar bladder inflammation and voiding dysfunction, after cystitis induction. Intravenous administration of TAK-242 (a TLR4-selective antagonist) significantly attenuated nociceptive responses in cystitis-induced URO-OVA mice, which was associated with reduced splenocyte production of TLR4-mediated IL-1ß, IL-6, and TNF-α as well as reduced spinal expression of mRNAs for IL-6, TNF-α, CD11b, glial fibrillary acidic protein, and high mobility group box 1. Our results indicate that altered TLR4 activation plays a critical role in bladder nociception independent of inflammation and voiding dysfunction in the URO-OVA model, providing a potential mechanistic insight and therapeutic target for IC/BPS pain.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Cistitis Intersticial/metabolismo , Dolor Nociceptivo/metabolismo , Umbral del Dolor , Receptor Toll-Like 4/metabolismo , Vejiga Urinaria/metabolismo , Analgésicos/farmacología , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/fisiopatología , Células Cultivadas , Cistitis Intersticial/genética , Cistitis Intersticial/inmunología , Cistitis Intersticial/fisiopatología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Dolor Nociceptivo/genética , Dolor Nociceptivo/inmunología , Dolor Nociceptivo/fisiopatología , Ovalbúmina/genética , Ovalbúmina/inmunología , Ovalbúmina/metabolismo , Umbral del Dolor/efectos de los fármacos , Transducción de Señal , Columna Vertebral/inmunología , Columna Vertebral/metabolismo , Bazo/inmunología , Bazo/metabolismo , Sulfonamidas/farmacología , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/inmunología , Vejiga Urinaria/fisiopatología , Urodinámica
10.
Sci Rep ; 9(1): 2430, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30787340

RESUMEN

Current medications inadequately treat the symptoms of chronic pain experienced by over 50 million people in the United States, and may come with substantial adverse effects signifying the need to find novel treatments. One novel therapeutic target is the Transient Receptor Potential A1 channel (TRPA1), an ion channel that mediates nociception through calcium influx of sensory neurons. Drug discovery still relies heavily on animal models, including zebrafish, a species in which TRPA1 activation produces hyperlocomotion. Here, we investigated if this hyperlocomotion follows zebrafish TRPA1 pharmacology and evaluated the strengths and limitations of using TRPA1-mediated hyperlocomotion as potential preclinical screening tool for drug discovery. To support face validity of the model, we pharmacologically characterized mouse and zebrafish TRPA1 in transfected HEK293 cells using calcium assays as well as in vivo. TRPA1 agonists and antagonists respectively activated or blocked TRPA1 activity in HEK293 cells, mice, and zebrafish in a dose-dependent manner. However, our results revealed complexities including partial agonist activity of TRPA1 antagonists, bidirectional locomotor activity, receptor desensitization, and off-target effects. We propose that TRPA1-mediated hyperlocomotion in zebrafish larvae has the potential to be used as in vivo screening tool for novel anti-nociceptive drugs but requires careful evaluation of the TRPA1 pharmacology.


Asunto(s)
Descubrimiento de Drogas , Locomoción/efectos de los fármacos , Dolor Nociceptivo/genética , Canal Catiónico TRPA1/genética , Proteínas de Pez Cebra/genética , Animales , Células HEK293 , Humanos , Locomoción/genética , Masculino , Ratones , Nocicepción/efectos de los fármacos , Dolor Nociceptivo/tratamiento farmacológico , Dolor Nociceptivo/patología , Canal Catiónico TRPA1/antagonistas & inhibidores , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/antagonistas & inhibidores
11.
Sci Rep ; 8(1): 6809, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29717160

RESUMEN

The spinal dorsal horn (SDH) is comprised of distinct neuronal populations that process different somatosensory modalities. Somatostatin (SST)-expressing interneurons in the SDH have been implicated specifically in mediating mechanical pain. Identifying the transcriptomic profile of SST neurons could elucidate the unique genetic features of this population and enable selective analgesic targeting. To that end, we combined the Isolation of Nuclei Tagged in Specific Cell Types (INTACT) method and Fluorescence Activated Nuclei Sorting (FANS) to capture tagged SST nuclei in the SDH of adult male mice. Using RNA-sequencing (RNA-seq), we uncovered more than 13,000 genes. Differential gene expression analysis revealed more than 900 genes with at least 2-fold enrichment. In addition to many known dorsal horn genes, we identified and validated several novel transcripts from pharmacologically tractable functional classes: Carbonic Anhydrase 12 (Car12), Phosphodiesterase 11 A (Pde11a), and Protease-Activated Receptor 3 (F2rl2). In situ hybridization of these novel genes showed differential expression patterns in the SDH, demonstrating the presence of transcriptionally distinct subpopulations within the SST population. Overall, our findings provide new insights into the gene repertoire of SST dorsal horn neurons and reveal several novel targets for pharmacological modulation of this pain-mediating population and treatment of pathological pain.


Asunto(s)
Interneuronas/metabolismo , Proteínas del Tejido Nervioso/genética , ARN Mensajero/genética , Somatostatina/genética , Asta Dorsal de la Médula Espinal/metabolismo , Transcripción Genética , 3',5'-GMP Cíclico Fosfodiesterasas/genética , 3',5'-GMP Cíclico Fosfodiesterasas/metabolismo , Animales , Anhidrasas Carbónicas/genética , Anhidrasas Carbónicas/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Perfilación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Interneuronas/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/clasificación , Proteínas del Tejido Nervioso/metabolismo , Dolor Nociceptivo/genética , Dolor Nociceptivo/metabolismo , Dolor Nociceptivo/fisiopatología , Células del Asta Posterior/metabolismo , Células del Asta Posterior/ultraestructura , ARN Mensajero/clasificación , ARN Mensajero/metabolismo , Receptores de Trombina/genética , Receptores de Trombina/metabolismo , Somatostatina/metabolismo , Asta Dorsal de la Médula Espinal/citología
12.
J Pharmacol Sci ; 136(1): 46-49, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29289470

RESUMEN

We studied the pronociceptive role of proteinase-activated receptor-2 (PAR2) in mouse bladder. In female mice, intravesical infusion of the PAR2-activating peptide, SLIGRL-amide (SL), caused delayed mechanical hypersensitivity in the lower abdomen, namely 'referred hyperalgesia', 6-24 h after the administration. The PAR2-triggered referred hyperalgesia was prevented by indomethacin or a selective TRPV1 blocker, and restored by a T-type Ca2+ channel blocker. In human urothelial T24 cells, SL caused delayed prostaglandin E2 production and COX-2 upregulation. Our data suggest that luminal PAR2 stimulation in the bladder causes prostanoid-dependent referred hyperalgesia in mice, which involves the activation of TRPV1 and T-type Ca2+ channels.


Asunto(s)
Canales de Calcio Tipo T/fisiología , Dinoprostona/metabolismo , Hiperalgesia/inducido químicamente , Hiperalgesia/genética , Dolor Nociceptivo/inducido químicamente , Dolor Nociceptivo/genética , Oligopéptidos/farmacología , Receptor PAR-2/metabolismo , Receptor PAR-2/fisiología , Canales Catiónicos TRPV/fisiología , Vejiga Urinaria , Animales , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo T/metabolismo , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Femenino , Humanos , Hiperalgesia/prevención & control , Indometacina , Ratones Endogámicos , Dolor Nociceptivo/prevención & control , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/metabolismo
13.
Am J Physiol Gastrointest Liver Physiol ; 314(2): G188-G200, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28971837

RESUMEN

The role of calcitonin gene-related peptide (CGRP) in visceral and somatic nociception is incompletely understood. CGRPα is highly expressed in sensory neurons of dorsal root ganglia and particularly in neurons that also express the transient receptor potential cation channel subfamily V member 1 (Trpv1). Therefore, we investigated changes in visceral and somatic nociception following deletion of CGRPα from the Trpv1-Cre population using the Cre/lox system. In control mice, acetic acid injection (0.6%, ip) caused significant immobility (time stationary), an established indicator of visceral pain. In CGRPα-mCherrylx/lx;Trpv1-Cre mice, the duration of immobility was significantly less than controls, and the distance CGRPα-mCherrylx/lx;Trpv1-Cre mice traveled over 20 min following acetic acid was significantly greater than controls. However, following acetic acid injection, there was no difference between genotypes in the writhing reflex, number of abdominal licks, or forepaw wipes of the cheek. CGRPα-mCherrylx/lx;Trpv1-Cre mice developed more pronounced inflammation-induced heat hypersensitivity above baseline values compared with controls. However, analyses of noxious acute heat or cold transmission revealed no difference between genotypes. Also, odor avoidance test, odor preference test, and buried food test for olfaction revealed no differences between genotypes. Our findings suggest that CGRPα-mediated transmission within the Trpv1-Cre population plays a significant role in visceral nociceptive pathways underlying voluntary movement. Monitoring changes in movement over time is a sensitive parameter to identify differences in visceral nociception, compared with writhing reflexes, abdominal licks, or forepaw wipes of the cheek that were unaffected by deletion of CGRPα- from Trpv1-Cre population and likely utilize different mechanisms. NEW & NOTEWORTHY The neuropeptide calcitonin gene-related peptide (CGRP) is highly colocalized with transient receptor potential cation channel subfamily V member 1 (TRPV1)-expressing primary afferent neurons, but the functional role of CGRPα specifically in these neurons is unknown in pain processing from visceral and somatic afferents. We used cre-lox recombination to conditionally delete CGRPα from TRPV1-expressing neurons in mice. We show that CGRPα from within TRPV1-cre population plays an important role in visceral nociception but less so in somatic nociception.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/metabolismo , Ganglios Espinales/metabolismo , Integrasas/metabolismo , Nocicepción , Dolor Nociceptivo/metabolismo , Canales Catiónicos TRPV/metabolismo , Dolor Visceral/metabolismo , Ácido Acético , Animales , Conducta Animal , Péptido Relacionado con Gen de Calcitonina/deficiencia , Péptido Relacionado con Gen de Calcitonina/genética , Modelos Animales de Enfermedad , Ganglios Espinales/fisiopatología , Calor , Integrasas/genética , Masculino , Ratones Noqueados , Actividad Motora , Dolor Nociceptivo/etiología , Dolor Nociceptivo/genética , Dolor Nociceptivo/fisiopatología , Tiempo de Reacción , Células Receptoras Sensoriales/metabolismo , Canales Catiónicos TRPV/genética , Dolor Visceral/inducido químicamente , Dolor Visceral/genética , Dolor Visceral/fisiopatología
14.
Eur J Immunol ; 48(2): 239-249, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29067676

RESUMEN

MYCBP2 is an E3 ubiquitin ligase, which is well characterized as a key element in the inhibition of neuronal growth, synapse formation and synaptic strength by regulating several signaling pathways. Although MYCBP2 was suspected to be expressed also in immune cells, to date nothing is known about its role in inflammation. We used Multi-epitope ligand cartography (MELC), a method for multiple sequential immunohistology, to show that MYCBP2 is strongly expressed in monocyte-derived macrophages during zymosan-induced inflammation. We generated a myeloid-specific knockout mouse and found that loss of MYCBP2 in myeloid cells reduced nociceptive (painful) behavior during the resolution phase (1-3 days after zymosan injection). Quantitative MELC analyses and flow cytometric analysis showed an increased number of CD206-expressing macrophages in the inflamed paw tissue. Fittingly, CD206 and arginase 1 expression was upregulated in MYCBP2-deficient bone marrow-derived macrophages after polarization with IL10 or IL4. The regulation of protein expression in these macrophages by MYCBP2 varied depending on the polarization signal. The increased IL10-induced CD206 expression in MYCBP2-deficient macrophages was mediated by p38 MAPK, while IL4-induced CD206 expression in MYCBP2-deficient macrophages was mediated by protein kinase A.


Asunto(s)
Proteínas Portadoras/metabolismo , Inflamación/inmunología , Macrófagos/inmunología , Animales , Arginasa/genética , Arginasa/metabolismo , Proteínas Portadoras/genética , Diferenciación Celular , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citocinas/metabolismo , Citometría de Flujo , Inflamación/genética , Lectinas Tipo C/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Dolor Nociceptivo/genética , Fenotipo , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Células Th2/inmunología , Ubiquitina-Proteína Ligasas , Zimosan/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
Anesth Analg ; 127(1): 263-266, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-28991117

RESUMEN

We investigated the effect of isoflurane on 2 main types of thermal nociceptors: A-δ and C-fibers. Surprisingly, 1% inhaled isoflurane led to a hyperalgesic response to C-fiber thermal stimulation, whereas responses to A-δ thermal stimulation were blunted. We explored the hypothesis that differences in withdrawal behavior are mediated by differential expression of isoflurane-sensitive proteins between these types of thermal nociceptors. Multiple transcriptomic databases of peripheral neurons were integrated to reveal that isoflurane-susceptible proteins Htr3a, Kcna2, and Scn8a were enriched in thermosensitive A-δ neurons. This exploratory analysis highlights the differing role that volatile anesthetics might have on nociceptors in the peripheral nervous system.


Asunto(s)
Anestésicos por Inhalación/administración & dosificación , Perfilación de la Expresión Génica/métodos , Isoflurano/administración & dosificación , Fibras Nerviosas Mielínicas/efectos de los fármacos , Fibras Nerviosas Amielínicas/efectos de los fármacos , Dolor Nociceptivo/prevención & control , Nociceptores/efectos de los fármacos , Administración por Inhalación , Anestésicos por Inhalación/toxicidad , Animales , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Calor , Hiperalgesia/inducido químicamente , Hiperalgesia/genética , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Isoflurano/toxicidad , Canal de Potasio Kv.1.2/genética , Canal de Potasio Kv.1.2/metabolismo , Masculino , Canal de Sodio Activado por Voltaje NAV1.6/genética , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Fibras Nerviosas Mielínicas/metabolismo , Fibras Nerviosas Amielínicas/metabolismo , Dolor Nociceptivo/genética , Dolor Nociceptivo/metabolismo , Dolor Nociceptivo/fisiopatología , Nociceptores/metabolismo , Umbral del Dolor/efectos de los fármacos , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Receptores de Serotonina 5-HT3/genética , Receptores de Serotonina 5-HT3/metabolismo
16.
Sci Rep ; 7(1): 15865, 2017 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-29158567

RESUMEN

Patients with interstitial cystitis/bladder pain syndrome (IC/BPS) suffer from chronic pain that severely affects quality of life. Although the underlying pathophysiology is not well understood, inhibition of bladder sensory afferents temporarily relieves pain. Here, we explored the possibility that optogenetic inhibition of nociceptive sensory afferents could be used to modulate bladder pain. The light-activated inhibitory proton pump Archaerhodopsin (Arch) was expressed under control of the sensory neuron-specific sodium channel (sns) gene to selectively silence these neurons. Optically silencing nociceptive sensory afferents significantly blunted the evoked visceromotor response to bladder distension and led to small but significant changes in bladder function. To study of the role of nociceptive sensory afferents in freely behaving mice, we developed a fully implantable, flexible, wirelessly powered optoelectronic system for the long-term manipulation of bladder afferent expressed opsins. We found that optogenetic inhibition of nociceptive sensory afferents reduced both ongoing pain and evoked cutaneous hypersensitivity in the context of cystitis, but had no effect in uninjured, naïve mice. These results suggest that selective optogenetic silencing of nociceptive bladder afferents may represent a potential future therapeutic strategy for the treatment of bladder pain.


Asunto(s)
Hiperalgesia/fisiopatología , Dolor Nociceptivo/fisiopatología , Dolor Pélvico/fisiopatología , Vejiga Urinaria/fisiopatología , Vías Aferentes/metabolismo , Animales , Proteínas Arqueales/genética , Cistitis Intersticial/genética , Cistitis Intersticial/fisiopatología , Ganglios Espinales , Humanos , Hiperalgesia/genética , Ratones , Neuronas Aferentes/patología , Dolor Nociceptivo/genética , Optogenética/métodos , Dolor Pélvico/genética , Calidad de Vida , Canales de Sodio/genética
17.
J Pain ; 18(9): 1046-1059, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28456745

RESUMEN

Cancer patients in pain require high doses of opioids and quickly become opioid-tolerant. Previous studies have shown that chronic cancer pain as well as high-dose opioid use lead to mu-opioid receptor downregulation. In this study we explore downregulation of the mu-opioid receptor gene (OPRM1), as a mechanism for opioid tolerance in the setting of opioid use for cancer pain. We demonstrate in a cohort of 84 cancer patients that high-dose opioid use correlates with OPRM1 hypermethylation in peripheral leukocytes of these patients. We then reverse-translate our clinical findings by creating a mouse cancer pain model; we create opioid tolerance in the mouse cancer model to mimic opioid tolerance in the cancer patients. Using this model we determine the functional significance of OPRM1 methylation on cancer pain and opioid tolerance. We focus on 2 main cells within the cancer microenvironment: the cancer cell and the neuron. We show that targeted re-expression of mu-opioid receptor on cancer cells inhibits mechanical and thermal hypersensitivity, and prevents opioid tolerance, in the mouse model. The resultant analgesia and protection against opioid tolerance are likely due to preservation of mu-opioid receptor expression on the cancer-associated neurons. PERSPECTIVE: We demonstrate that epigenetic regulation of OPRM1 contributes to opioid tolerance in cancer patients, and that targeted gene therapy could treat cancer-induced nociception and opioid tolerance in a mouse cancer model.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Dolor en Cáncer/tratamiento farmacológico , Dolor en Cáncer/metabolismo , Metilación de ADN , Tolerancia a Medicamentos/genética , Receptores Opioides mu/metabolismo , Adulto , Anciano , Analgésicos Opioides/farmacología , Animales , Dolor en Cáncer/genética , Línea Celular Tumoral , Estudios de Cohortes , Tolerancia a Medicamentos/fisiología , Epigénesis Genética , Femenino , Humanos , Masculino , Ratones Endogámicos BALB C , Persona de Mediana Edad , Morfina/farmacología , Morfina/uso terapéutico , Trasplante de Neoplasias , Neoplasias/genética , Neoplasias/fisiopatología , Dolor Nociceptivo/tratamiento farmacológico , Dolor Nociceptivo/genética , Dolor Nociceptivo/metabolismo , Trastornos Relacionados con Opioides/genética , Trastornos Relacionados con Opioides/metabolismo , Manejo del Dolor/métodos , Educación del Paciente como Asunto , Pruebas de Farmacogenómica , Receptores Opioides mu/genética
18.
Sci Rep ; 7: 45930, 2017 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-28378856

RESUMEN

The present study was performed to explore the role of galanin and galanin receptor 2 in nociceptive modulation in anterior cingulate cortex (ACC) of normal rats and rats with mononeuropathy. Intra-ACC injection of galanin induced significant increases in hindpaw withdrawal latencies (HWLs) to thermal and mechanical stimulations in both normal rats and rats with mononeuropathy, the increased HWLs were attenuated significantly by intra-ACC injection of galanin receptor 2 antagonist M871, indicating an involvement of galanin receptor 2 in nociceptive modulation in ACC. Interestingly, the galanin-induced HWL was significant higher in rats with mononeuropathy than that in normal rats tested by Randall Selitto test. Furthermore, both the galanin mRNA expression and galanin content increased significantly in ACC in rats with mononeuropathy than that in normal rats. Moreover, both the mRNA levels of galanin receptor 2 and the content of galanin receptor 2 in ACC increased significantly in rats with mononeuropathy than that in normal rats. These results found that galanin induced antinociception in ACC in both normal rats and rats with mononeuropathy. And there may be plastic changes in the expression of galanin and galanin receptor 2 in rats with mononeuropathy, as well as in the galanin-induced antinociception.


Asunto(s)
Galanina/metabolismo , Giro del Cíngulo/metabolismo , Mononeuropatías/metabolismo , Nociceptores/metabolismo , Precursores de Proteínas/metabolismo , Receptor de Galanina Tipo 2/metabolismo , Animales , Galanina/genética , Galanina/farmacología , Expresión Génica/efectos de los fármacos , Giro del Cíngulo/efectos de los fármacos , Giro del Cíngulo/fisiopatología , Miembro Posterior , Masculino , Mononeuropatías/genética , Mononeuropatías/fisiopatología , Nocicepción/efectos de los fármacos , Dolor Nociceptivo/genética , Dolor Nociceptivo/metabolismo , Dolor Nociceptivo/fisiopatología , Nociceptores/efectos de los fármacos , Dimensión del Dolor/métodos , Péptidos/farmacología , Precursores de Proteínas/genética , Precursores de Proteínas/farmacología , Ratas Sprague-Dawley , Receptor de Galanina Tipo 2/antagonistas & inhibidores , Receptor de Galanina Tipo 2/genética
19.
Mol Genet Genomics ; 292(4): 729-739, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28389699

RESUMEN

Migraine is a common neurological disorder that affects approximately 12-20% of the general adult population. Migraine pathogenesis is complex and not wholly understood. Molecular genetic investigations, imaging and biochemical studies, have unveiled a number of interconnected neurological pathways which seem to have a cause and effect component integral to its cause. Much weight of migraine attack initiation can be placed on the initial trigger and the pathways involved in its neuronal counter reaction. Ion channels play a large role in the generation, portrayal and mitigation of the brains response to external triggers. Several genetic studies have identified and implicated a number of ion channelopathy genes which may contribute to this generalised process. This review will focus on the genetics of migraine with particular emphasis placed on the potentially important role genes HEPH (responsible for iron transport and homeostasis) and KCNK18 (important for the transport and homeostasis of potassium) play in migraine cause.


Asunto(s)
Canalopatías/genética , Proteínas de la Membrana/genética , Trastornos Migrañosos/genética , Dolor Nociceptivo/genética , Canales de Potasio/genética , Adulto , Canalopatías/patología , Femenino , Humanos , Masculino , Trastornos Migrañosos/patología , Dolor Nociceptivo/patología , Canales de Potasio/fisiología
20.
Anesthesiology ; 126(5): 967-977, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28248712

RESUMEN

BACKGROUND: The authors and others have previously shown that the up-regulation of spinal ephrin type-b receptor 1 plays an essential role in the pathologic process of nerve injury-induced nociceptive hypersensitivity, but the regulatory mechanism remains unclear. METHODS: Radiant heat and von Frey filaments were applied to assess nociceptive behaviors. Real-time quantitative polymerase chain reaction, Western blotting, fluorescence in situ hybridization, immunofluorescence, immunohistochemistry, dual-luciferase reporter gene assays, recombinant lentivirus, and small interfering RNA were used to characterize the likely mechanisms. RESULTS: Periphery nerve injury induced by chronic constriction injury of the sciatic nerve significantly reduced spinal microRNA-182-5p (miR-182-5p) expression levels, which were inversely correlated with spinal ephrin type-b receptor 1 expression (R = 0.90; P < 0.05; n = 8). The overexpression of miR-182-5p in the spinal cord prevented and reversed the nociceptive behaviors induced by sciatic nerve injury, accompanied by a decreased expression of spinal ephrin type-b receptor 1 (recombinant lentiviruses containing pre-microRNA-182: 1.91 ± 0.34 vs. 1.24 ± 0.31, n = 4; miR-182-5p mimic: 2.90 ± 0.48 vs. 1.51 ± 0.25, n = 4). In contrast, the down-regulation of spinal miR-182-5p facilitated the nociceptive behaviors induced by sciatic nerve injury and increased the expression of spinal ephrin type-b receptor 1 (1.0 ± 0.26 vs. 1.74 ± 0.31, n = 4). Moreover, the down-regulation of miR-182-5p and up-regulation of ephrin type-b receptor 1 caused by sciatic nerve injury were mediated by the N-methyl-D-aspartate receptor. CONCLUSIONS: Collectively, our findings reveal that the spinal ephrin type-b receptor 1 is regulated by miR-182-5p in nerve injury-induced nociceptive hypersensitivity.


Asunto(s)
MicroARNs/metabolismo , Dolor Nociceptivo/fisiopatología , Receptores de la Familia Eph/metabolismo , Nervio Ciático/fisiopatología , Regulación hacia Arriba/fisiología , Animales , Western Blotting , Regulación hacia Abajo , Efrinas , Fluorescencia , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Masculino , Ratones , MicroARNs/genética , Dolor Nociceptivo/genética , Dolor Nociceptivo/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de la Familia Eph/genética , Nervio Ciático/metabolismo , Médula Espinal/metabolismo , Regulación hacia Arriba/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA