Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 849
Filtrar
1.
Sci Adv ; 10(25): eadj9173, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38905344

RESUMEN

Sensory neurons in the dorsal root ganglion (DRG) and trigeminal ganglion (TG) are specialized to detect and transduce diverse environmental stimuli to the central nervous system. Single-cell RNA sequencing has provided insights into the diversity of sensory ganglia cell types in rodents, nonhuman primates, and humans, but it remains difficult to compare cell types across studies and species. We thus constructed harmonized atlases of the DRG and TG that describe and facilitate comparison of 18 neuronal and 11 non-neuronal cell types across six species and 31 datasets. We then performed single-cell/nucleus RNA sequencing of DRG from both human and the highly regenerative axolotl and found that the harmonized atlas also improves cell type annotation, particularly of sparse neuronal subtypes. We observed that the transcriptomes of sensory neuron subtypes are broadly similar across vertebrates, but the expression of functionally important neuropeptides and channels can vary notably. The resources presented here can guide future studies in comparative transcriptomics, simplify cell-type nomenclature differences across studies, and help prioritize targets for future analgesic development.


Asunto(s)
Ganglios Espinales , Transcriptoma , Ganglio del Trigémino , Animales , Humanos , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Ganglio del Trigémino/citología , Ganglio del Trigémino/metabolismo , Análisis de la Célula Individual/métodos , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/citología , Especificidad de la Especie , Ratones , Atlas como Asunto , Perfilación de la Expresión Génica , Ratas
2.
J Virol ; 98(6): e0042324, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38771044

RESUMEN

Bovine alphaherpesvirus 1 (BoHV-1) infection causes respiratory tract disorders and immune suppression and may induce bacterial pneumonia. BoHV-1 establishes lifelong latency in sensory neurons after acute infection. Reactivation from latency consistently occurs following stress or intravenous injection of the synthetic corticosteroid dexamethasone (DEX), which mimics stress. The immediate early transcription unit 1 (IEtu1) promoter drives expression of infected cell protein 0 (bICP0) and bICP4, two viral transcriptional regulators necessary for productive infection and reactivation from latency. The IEtu1 promoter contains two glucocorticoid receptor (GR) responsive elements (GREs) that are transactivated by activated GR. GC-rich motifs, including consensus binding sites for specificity protein 1 (Sp1), are in the IEtu1 promoter sequences. E2F family members bind a consensus sequence (TTTCCCGC) and certain specificity protein 1 (Sp1) sites. Consequently, we hypothesized that certain E2F family members activate IEtu1 promoter activity. DEX treatment of latently infected calves increased the number of E2F2+ TG neurons. GR and E2F2, but not E2F1, E2F3a, or E2F3b, cooperatively transactivate a 436-bp cis-regulatory module in the IEtu1 promoter that contains both GREs. A luciferase reporter construct containing a 222-bp fragment downstream of the GREs was transactivated by E2F2 unless two adjacent Sp1 binding sites were mutated. Chromatin immunoprecipitation studies revealed that E2F2 occupied IEtu1 promoter sequences when the BoHV-1 genome was transfected into mouse neuroblastoma (Neuro-2A) or monkey kidney (CV-1) cells. In summary, these findings revealed that GR and E2F2 cooperatively transactivate IEtu1 promoter activity, which is predicted to influence the early stages of BoHV-1 reactivation from latency. IMPORTANCE: Bovine alpha-herpesvirus 1 (BoHV-1) acute infection in cattle leads to establishment of latency in sensory neurons in the trigeminal ganglia (TG). A synthetic corticosteroid dexamethasone consistently initiates BoHV-1 reactivation in latently infected calves. The BoHV-1 immediate early transcription unit 1 (IEtu1) promoter regulates expression of infected cell protein 0 (bICP0) and bICP4, two viral transcriptional regulators. Hence, the IEtu1 promoter must be activated for the reactivation to occur. The number of TG neurons expressing E2F2, a transcription factor and cell cycle regulator, increased during early stages of reactivation from latency. The glucocorticoid receptor (GR) and E2F2, but not E2F1, E2F3a, or E2F3b, cooperatively transactivated a 436-bp cis-regulatory module (CRM) in the IEtu1 promoter that contains two GR responsive elements. Chromatin immunoprecipitation studies revealed that E2F2 occupies IEtu1 promoter sequences in cultured cells. GR and E2F2 mediate cooperative transactivation of IEtu1 promoter activity, which is predicted to stimulate viral replication following stressful stimuli.


Asunto(s)
Ciclo Celular , Factor de Transcripción E2F2 , Regulación Viral de la Expresión Génica , Herpesvirus Bovino 1 , Proteínas Inmediatas-Precoces , Regiones Promotoras Genéticas , Receptores de Glucocorticoides , Activación Transcripcional , Animales , Bovinos , Ratones , Sitios de Unión , Línea Celular , Dexametasona/farmacología , Factor de Transcripción E2F2/metabolismo , Regulación Viral de la Expresión Génica/efectos de los fármacos , Regulación Viral de la Expresión Génica/genética , Infecciones por Herpesviridae/virología , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/genética , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 1/fisiología , Proteínas Inmediatas-Precoces/genética , Neuronas/virología , Receptores de Glucocorticoides/metabolismo , Elementos de Respuesta/genética , Factor de Transcripción Sp1/metabolismo , Transactivadores/metabolismo , Ganglio del Trigémino/citología , Ganglio del Trigémino/virología , Activación Viral , Latencia del Virus
3.
Toxins (Basel) ; 14(2)2022 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-35202143

RESUMEN

Chimeras of botulinum neurotoxin (BoNT) serotype A (/A) combined with /E protease might possess improved analgesic properties relative to either parent, due to inheriting the sensory neurotropism of the former with more extensive disabling of SNAP-25 from the latter. Hence, fusions of /E protease light chain (LC) to whole BoNT/A (LC/E-BoNT/A), and of the LC plus translocation domain (HN) of /E with the neuronal acceptor binding moiety (HC) of /A (BoNT/EA), created previously by gene recombination and expression in E. coli., were used. LC/E-BoNT/A (75 units/kg) injected into the whisker pad of rats seemed devoid of systemic toxicity, as reflected by an absence of weight loss, but inhibited the nocifensive behavior (grooming, freezing, and reduced mobility) induced by activating TRPV1 with capsaicin, injected at various days thereafter. No sex-related differences were observed. c-Fos expression was increased five-fold in the trigeminal nucleus caudalis ipsi-lateral to capsaicin injection, relative to the contra-lateral side and vehicle-treated controls, and this increase was virtually prevented by LC/E-BoNT/A. In vitro, LC/E-BoNT/A or /EA diminished CGRP exocytosis from rat neonate trigeminal ganglionic neurons stimulated with up to 1 µM capsaicin, whereas BoNT/A only substantially reduced the release in response to 0.1 µM or less of the stimulant, in accordance with the /E protease being known to prevent fusion of exocytotic vesicles.


Asunto(s)
Analgésicos/farmacología , Toxinas Botulínicas Tipo A/farmacología , Capsaicina/farmacología , Neurotoxinas/farmacología , Células Receptoras Sensoriales/efectos de los fármacos , Animales , Animales Recién Nacidos , Conducta Animal/efectos de los fármacos , Péptido Relacionado con Gen de Calcitonina/metabolismo , Femenino , Masculino , Dolor/inducido químicamente , Dolor/tratamiento farmacológico , Dolor/metabolismo , Ratas Sprague-Dawley , Células Receptoras Sensoriales/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Ganglio del Trigémino/citología
4.
Int J Mol Sci ; 23(3)2022 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-35163452

RESUMEN

The pro-nociceptive role of glutamate in the CNS in migraine pathophysiology is well established. Glutamate, released from trigeminal afferents, activates second order nociceptive neurons in the brainstem. However, the function of peripheral glutamate receptors in the trigeminovascular system suggested as the origin site for migraine pain, is less known. In the current project, we used calcium imaging and patch clamp recordings from trigeminal ganglion (TG) neurons, immunolabelling, CGRP assay and direct electrophysiological recordings from rat meningeal afferents to investigate the role of glutamate in trigeminal nociception. Glutamate, aspartate, and, to a lesser extent, NMDA under free-magnesium conditions, evoked calcium transients in a fraction of isolated TG neurons, indicating functional expression of NMDA receptors. The fraction of NMDA sensitive neurons was increased by the migraine mediator CGRP. NMDA also activated slowly desensitizing currents in 37% of TG neurons. However, neither glutamate nor NMDA changed the level of extracellular CGRP. TG neurons expressed both GluN2A and GluN2B subunits of NMDA receptors. In addition, after removal of magnesium, NMDA activated persistent spiking activity in a fraction of trigeminal nerve fibers in meninges. Thus, glutamate activates NMDA receptors in somas of TG neurons and their meningeal nerve terminals in magnesium-dependent manner. These findings suggest that peripherally released glutamate can promote excitation of meningeal afferents implicated in generation of migraine pain in conditions of inherited or acquired reduced magnesium blockage of NMDA channels and support the usage of magnesium supplements in migraine.


Asunto(s)
Calcio/metabolismo , Ácido Glutámico/farmacología , Nocicepción/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Ganglio del Trigémino/citología , Animales , Ácido Aspártico/farmacología , Células Cultivadas , Masculino , Trastornos Migrañosos/metabolismo , N-Metilaspartato/farmacología , Técnicas de Placa-Clamp , Ratas , Ganglio del Trigémino/efectos de los fármacos , Ganglio del Trigémino/metabolismo
5.
Theranostics ; 11(19): 9342-9357, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34646374

RESUMEN

Background: Neuromedin B (Nmb) is implicated in the regulation of nociception of sensory neurons. However, the underlying cellular and molecular mechanisms remain unknown. Methods: Using patch clamp recording, western blot analysis, immunofluorescent labelling, enzyme-linked immunosorbent assays, adenovirus-mediated shRNA knockdown and animal behaviour tests, we studied the effects of Nmb on the sensory neuronal excitability and peripheral pain sensitivity mediated by Cav3.2 T-type channels. Results: Nmb reversibly and concentration-dependently increased T-type channel currents (IT) in small-sized trigeminal ganglion (TG) neurons through the activation of neuromedin B receptor (NmbR). This NmbR-mediated IT response was Gq protein-coupled, but independent of protein kinase C activity. Either intracellular application of the QEHA peptide or shRNA-mediated knockdown of Gß abolished the NmbR-induced IT response. Inhibition of protein kinase A (PKA) or AMP-activated protein kinase (AMPK) completely abolished the Nmb-induced IT response. Analysis of phospho-AMPK (p-AMPK) revealed that Nmb significantly activated AMPK, while AMPK inhibition prevented the Nmb-induced increase in PKA activity. In a heterologous expression system, activation of NmbR significantly enhanced the Cav3.2 channel currents, while the Cav3.1 and Cav3.3 channel currents remained unaffected. Nmb induced TG neuronal hyperexcitability and concomitantly induced mechanical and thermal hypersensitivity, both of which were attenuated by T-type channel blockade. Moreover, blockade of NmbR signalling prevented mechanical hypersensitivity in a mouse model of complete Freund's adjuvant-induced inflammatory pain, and this effect was attenuated by siRNA knockdown of Cav3.2. Conclusions: Our study reveals a novel mechanism by which NmbR stimulates Cav3.2 channels through a Gßγ-dependent AMPK/PKA pathway. In mouse models, this mechanism appears to drive the hyperexcitability of TG neurons and induce pain hypersensitivity.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Dolor/metabolismo , Receptores de Bombesina/metabolismo , Potenciales de Acción , Animales , Canales de Calcio Tipo T/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Adyuvante de Freund/farmacología , Ganglios Espinales/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Neuroquinina B/análogos & derivados , Neuroquinina B/metabolismo , Dolor/fisiopatología , Receptores de Bombesina/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología , Transducción de Señal/efectos de los fármacos , Ganglio del Trigémino/citología , Ganglio del Trigémino/metabolismo
6.
Sci Rep ; 11(1): 17813, 2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34497285

RESUMEN

Trigeminal (TG), dorsal root (DRG), and nodose/jugular (NG/JG) ganglia each possess specialized and distinct functions. We used RNA sequencing of two-cycle sorted Pirt-positive neurons to identify genes exclusively expressing in L3-L5 DRG, T10-L1 DRG, NG/JG, and TG mouse ganglion neurons. Transcription factor Phox2b and Efcab6 are specifically expressed in NG/JG while Hoxa7 is exclusively present in both T10-L1 and L3-L5 DRG neurons. Cyp2f2, Krt18, and Ptgds, along with pituitary hormone prolactin (Prl), growth hormone (Gh), and proopiomelanocortin (Pomc) encoding genes are almost exclusively in TG neurons. Immunohistochemistry confirmed selective expression of these hormones in TG neurons and dural nerves; and showed GH expression in subsets of TRPV1+ and CGRP+ TG neurons. We next examined GH roles in hypersensitivity in the spinal versus trigeminal systems. Exogenous GH produced mechanical hypersensitivity when injected intrathecally, but not intraplantarly. GH-induced thermal hypersensitivity was not detected in the spinal system. GH dose-dependently generated orofacial and headache-like periorbital mechanical hypersensitivity after administration into masseter muscle and dura, respectively. Periorbital mechanical hypersensitivity was reversed by a GH receptor antagonist, pegvisomant. Overall, pituitary hormone genes are selective for TG versus other ganglia somatotypes; and GH has distinctive functional significance in the trigeminal versus spinal systems.


Asunto(s)
Hormona del Crecimiento/metabolismo , Dolor/metabolismo , Proopiomelanocortina/metabolismo , Prolactina/metabolismo , Células Receptoras Sensoriales/metabolismo , Ganglio del Trigémino/metabolismo , Animales , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Ratones , Ratones Transgénicos , Ganglio Nudoso/citología , Ganglio Nudoso/metabolismo , Ganglio del Trigémino/citología
7.
J Pharmacol Sci ; 146(4): 200-205, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34116733

RESUMEN

Gentle touch such as stroking of the skin produces a pleasant feeling, which is detected by a rare subset of sensory neurons that express Mas-related G protein-coupled receptor B4 (MrgprB4) in mice. We examined small populations of MrgprB4-positive neurons in the trigeminal ganglion and the dorsal root ganglion, and most of these were sensitive to transient receptor potential ankyrin 1 (TRPA1) agonist but not TRPV1, TRPM8, or TRPV4 agonists. Deficiency of MrgprB4 did not affect noxious pain or itch behaviors in the hairless plantar and hairy cheek. Although behavior related to acetone-induced cold sensing in the hind paw was not changed, unpleasant sensory behaviors in response to acetone application or sucrose splash to the cheek were significantly enhanced in Mrgprb4-knockout mice as well as in TRPA1-knockout mice. These results suggest that MrgprB4 in the trigeminal neurons produces pleasant sensations in cooperation with TRPA1, rather than noxious or cold sensations. Pleasant sensations may modulate unpleasant sensations on the cheek via MrgprB4.


Asunto(s)
Expresión Génica/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Sensación/genética , Sensación/fisiología , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología , Canal Catiónico TRPA1/genética , Canal Catiónico TRPA1/fisiología , Ganglio del Trigémino/citología , Animales , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenómenos Fisiológicos de la Piel/genética , Canal Catiónico TRPA1/metabolismo
8.
Sci Rep ; 10(1): 19694, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-33184395

RESUMEN

Evidence indicates that dental pulp stem cells (DPSC) secrete neurotrophic factors which play an important role in neurogenesis, neural maintenance and repair. In this study we investigated the trophic potential of DPSC-derived conditioned medium (CM) to protect and regenerate isolated primary trigeminal ganglion neuronal cells (TGNC). DPSC and TGNC were harvested by enzymatic digestion from Wister-Hann rats. CM was collected from 72 h serum-free DPSC cultures and neurotrophic factors; nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and glial cell line-derived neurotrophic factor (GDNF) were analysed by specific enzyme-linked immunosorbent assays (ELISAs). Primary co-cultures of DPSC and TGNC were established to evaluate the paracrine effects of DPSC. In comparison, NGF was used to evaluate its neurotrophic and neuritogenic effect on TGNC. Immunocytochemistry was performed to detect the neuronal-markers; neuronal nuclei (NeuN), microtubule-associated protein-2 (MAP-2) and ßIII-tubulin. Quantitative real time polymerase chain reaction (qRT-PCR) was used to analyse neuronal-associated gene expression of NeuN, MAP-2, ßIII-tubulin in addition to growth-associated protein-43 (GAP-43), Synapsin-I and thermo-sensitive transient receptor potential vanilloid channel-1 (TRPV1). DPSC-CM contained significant levels of NGF, BDNF, NT-3 and GDNF. DPSC and DPSC-CM significantly enhanced TGNC survival with extensive neurite outgrowth and branching as evaluated by immunocytochemistry of neuronal markers. DPSC-CM was more effective in stimulating TGNC survival than co-cultures or NGF treated culture. In comparison to controls, DPSC-CM significantly upregulated gene expression of several neuronal markers as well as TRPV1. This study demonstrated that DPSC-derived factors promoted survival and regeneration of isolated TGNC and may be considered as cell-free therapy for TG nerve repair.


Asunto(s)
Técnicas de Cocultivo/métodos , Pulpa Dental/citología , Factores de Crecimiento Nervioso/metabolismo , Ganglio del Trigémino/citología , Animales , Biomarcadores/metabolismo , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Pulpa Dental/metabolismo , Masculino , Neurogénesis , Cultivo Primario de Células , Ratas , Células Madre/citología
9.
Biomolecules ; 10(11)2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33167484

RESUMEN

Local anesthetics (LAs) can completely block nociception by inhibiting voltage-gated sodium channels (VGSCs), and thus, blocking action potentials (APs) within sensory neurons. As one of the several LAs, eugenol is used for dental pain treatment. It reportedly features multiple functions in regulating diverse ion channels. This study aimed to investigate the long-lasting analgesic effect of eugenol alone, as well as that of the combination of eugenol as a noxious-heat-sensitive transient receptor potential vanilloid 1 (TRPV1) channel agonist and a permanently charged sodium channel blocker (QX-314), on neuronal excitability in trigeminal ganglion (TG) neurons. Eugenol alone increased inward current in a dose-dependent manner in capsaicin-sensitive TG neurons. Eugenol also inhibited the VGSC current and AP. These effects were reversed through wash-out. The combination of eugenol and QX-314 was evaluated in the same manner. The combination completely inhibited the VGSC current and AP. However, these effects were not reversed and were continuously blocked even after wash-out. Taken together, our results suggest that, in contrast to the effect of eugenol alone, the combination of eugenol and QX-314 irreversibly and selectively blocked VGSCs in TG neurons expressing TRPV1.


Asunto(s)
Eugenol/farmacología , Lidocaína/análogos & derivados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ganglio del Trigémino/citología , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Canales de Sodio Activados por Voltaje/metabolismo , Animales , Interacciones Farmacológicas , Lidocaína/farmacología , Masculino , Neuronas/citología , Nocicepción/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
10.
Mol Pain ; 16: 1744806920960856, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32985330

RESUMEN

Capsaicin is an agonist of transient receptor potential cation channel subfamily V member 1 (TRPV1). Strong TRPV1 stimulation with capsaicin causes mitochondrial damage in primary sensory neurons. However, the effect of repetitive and moderate exposure to capsaicin on the integrity of neuronal mitochondria remains largely unknown. Our electron microscopic analysis revealed that repetitive stimulation of the facial skin of mice with 10 mM capsaicin induced short-term damage to the mitochondria in small-sized trigeminal ganglion neurons. Further, capsaicin-treated mice exhibited decreased sensitivity to noxious heat stimulation, indicating TRPV1 dysfunction, in parallel with the mitochondrial damage in the trigeminal ganglion neurons. To analyze the capsaicin-induced mitochondrial damage and its relevant cellular events in detail, we performed cell-based assays using TRPV1-expressing PC12 cells. Dose-dependent capsaicin-mediated mitochondrial toxicity was observed. High doses of capsaicin caused rapid destruction of mitochondrial internal structure, while low doses induced mitochondrial swelling. Further, capsaicin induced a dose-dependent loss of mitochondria and autophagy-mediated degradation of mitochondria (mitophagy). Concomitantly, transcriptional upregulation of mitochondrial proteins, cytochrome c oxidase subunit IV, Mic60/Mitofilin, and voltage-dependent anion channel 1 was observed, which implied induction of mitochondrial biogenesis to compensate for the loss of mitochondria. Collectively, although trigeminal ganglion neurons transiently exhibit mitochondrial damage and TRPV1 dysfunction following moderate capsaicin exposure, they appear to be resilient to such a challenge. Our in vitro data show a dose-response relationship in capsaicin-mediated mitochondrial toxicity. We postulate that induction of mitophagy and mitochondrial biogenesis in response to capsaicin stimulation play important roles in repairing the damaged mitochondrial system.


Asunto(s)
Capsaicina/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neuronas/efectos de los fármacos , Canales Catiónicos TRPV/metabolismo , Ganglio del Trigémino/efectos de los fármacos , Animales , Capsaicina/toxicidad , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Calor , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Mitocondrias/enzimología , Mitocondrias/ultraestructura , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Mitofagia/efectos de los fármacos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Neuronas/metabolismo , Neuronas/ultraestructura , Células PC12 , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Canales Catiónicos TRPV/genética , Ganglio del Trigémino/citología , Ganglio del Trigémino/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/genética , Canal Aniónico 1 Dependiente del Voltaje/metabolismo
11.
Int J Mol Sci ; 21(17)2020 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-32825453

RESUMEN

Activation of the trigeminal system causes the release of various neuropeptides, cytokines, and other immune mediators. Calcitonin gene-related peptide (CGRP), which is a potent algogenic mediator, is expressed in the peripheral sensory neurons of trigeminal ganglion (TG). It affects the inflammatory responses and pain sensitivity by modulating the activity of glial cells. The primary aim of this study was to use array analysis to investigate the effect of CGRP on the glial cells of TG in regulating nuclear factor kappa B (NF-κB) signaling genes and to further check if CGRP in the TG can affect neuron-glia activation in the spinal trigeminal nucleus caudalis. The glial cells of TG were stimulated with CGRP or Minocycline (Min) + CGRP. The effect on various genes involved in NF-κB signaling pathway was analyzed compared to no treatment control condition using a PCR array analysis. CGRP, Min + CGRP or saline was directly injected inside the TG and the effect on gene expression of Egr1, Myd88 and Akt1 and protein expression of cleaved Caspase3 (cleav Casp3) in the TG, and c-Fos and glial fibrillary acidic protein (GFAP) in the spinal section containing trigeminal nucleus caudalis was analyzed. Results showed that CGRP stimulation resulted in the modulation of several genes involved in the interleukin 1 signaling pathway and some genes of the tumor necrosis factor pathway. Minocycline pre-treatment resulted in the modulation of several genes in the glial cells, including anti-inflammatory genes, and neuronal activation markers. A mild increase in cleav Casp3 expression in TG and c-Fos and GFAP in the spinal trigeminal nucleus of CGRP injected animals was observed. These data provide evidence that glial cells can participate in neuroimmune interaction due to CGRP in the TG via NF-κB signaling pathway.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/farmacología , FN-kappa B/metabolismo , Neuroglía/metabolismo , Ganglio del Trigémino/citología , Animales , Péptido Relacionado con Gen de Calcitonina/fisiología , Caspasa 3/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Minociclina/farmacología , Factor 88 de Diferenciación Mieloide/genética , FN-kappa B/genética , Neuroglía/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética , Ratas Sprague-Dawley , Transducción de Señal/genética , Núcleo Caudal del Trigémino/metabolismo , Ganglio del Trigémino/efectos de los fármacos , Ganglio del Trigémino/metabolismo
12.
J Photochem Photobiol B ; 210: 111959, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32739664

RESUMEN

Trigeminal ganglion (TG) neurons play an essential role in the sensory nerves of the face. Damaged TG neurons resulting from the accidental and non-intentional nerve lesions, commonly identified as neuropathic pain, which is known to cause intense pain and sensory abnormalities. For the treatment, surgical methods are conducted when the pharmacological treatment fails to provide satisfactory recovery. However, the process of surgery or drug intake can burden the patient or cause side effects. One of the logical choices of study becomes photobiomodulation (PBM) referred to as therapeutic approaches based on the interactions of visible or near-infrared (NIR) photons with biomolecules inside cells or tissues. In this study, we constructed a PBM illumination setup to stimulate the cultured primary TG neurons and compared the growth morphology between the non-irradiated control group and irradiation group with NIR laser of 808 nm wavelength. In addition, we applied various radiant exposures of 1, 2, and 10 J/cm2 with different pulse frequencies of 1, 10, and 100 Hz. We found that PBM could promote neurite growth of TG neurons, and it works at relatively low energy densities at 1 and 2 J/cm2. The irradiation group in the pulsed wave mode with the frequency of 10 Hz was found to be the most effective when compared to other frequencies. Thus, PBM on TG neurons facilitated neuronal growth in vitro in a dose and frequency-dependent fashion. PBM may provide a potential therapeutic approach to treat damaged peripheral nerves.


Asunto(s)
Rayos Infrarrojos , Animales , Células Cultivadas , Ratones , Neuritas/fisiología , Neuronas/citología , Neuronas/metabolismo , Neuronas/efectos de la radiación , Imagen Óptica , Ganglio del Trigémino/citología
13.
Sci Rep ; 10(1): 8632, 2020 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-32451393

RESUMEN

Pain evoked by visceral inflammation is often 'referred' to the somatic level. Transient receptor potential ankyrin 1 (TRPA1) has been reported to contribute to visceral pain-like behavior in dextran sulfate sodium (DSS)-evoked colitis. However, the role of TRPA1 in somatic component of hypersensitivity due to visceral inflammation is unknown. The present study investigated the role of TRPA1 in colitis-evoked mechanical hypersensitivity at the somatic level. Colitis was induced in mice by adding DSS to drinking water for one week. Control and DSS-treated mice were tested for various parameters of colitis as well as mechanical pain sensitivity in abdominal and facial regions. DSS treatment caused mechanical hypersensitivity in the abdominal and facial skin. Pharmacological blockade or genetic deletion of TRPA1 prevented the colitis-associated mechanical hypersensitivity in the abdominal and facial skin areas although the severity of colitis remained unaltered. DSS treatment increased expression of TRPA1 mRNA in cultured dorsal root ganglion (DRG) neurons, but not trigeminal ganglion neurons, and selectively enhanced currents evoked by the TRPA1 agonist, allyl isothiocyanate, in cultured DRG neurons. Our findings indicate that the TRPA1 channel contributes to colitis-associated mechanical hypersensitivity in somatic tissues, an effect associated with upregulation of TRPA1 expression and responsiveness in DRG nociceptors.


Asunto(s)
Colitis/patología , Dolor Nociceptivo/patología , Canal Catiónico TRPA1/metabolismo , Acetanilidas/farmacología , Animales , Colitis/inducido químicamente , Sulfato de Dextran/toxicidad , Potenciales Evocados/efectos de los fármacos , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Isotiocianatos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Purinas/farmacología , Estrés Mecánico , Canal Catiónico TRPA1/antagonistas & inhibidores , Canal Catiónico TRPA1/genética , Ganglio del Trigémino/citología , Ganglio del Trigémino/metabolismo
14.
PLoS One ; 15(3): e0230870, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32226020

RESUMEN

When herpes simplex virus 1 (HSV-1) infection is initiated in the ocular, nasal, or oral cavity, sensory neurons within trigeminal ganglia (TG) become infected. Following a burst of viral transcription in TG neurons, lytic cycle viral genes are suppressed and latency is established. The latency-associated transcript (LAT) is the only viral gene abundantly expressed during latency, and LAT expression is important for the latency-reactivation cycle. Reactivation from latency is required for virus transmission and recurrent disease, including encephalitis. The Wnt/ß-catenin signaling pathway is differentially expressed in TG during the bovine herpesvirus 1 latency-reactivation cycle. Hence, we hypothesized HSV-1 regulates the Wnt/ß-catenin pathway and promotes maintenance of latency because this pathway enhances neuronal survival and axonal repair. New studies revealed ß-catenin was expressed in significantly more TG neurons during latency compared to TG from uninfected mice or mice latently infected with a LAT-/- mutant virus. When TG explants were incubated with media containing dexamethasone to stimulate reactivation, significantly fewer ß-catenin+ TG neurons were detected. Conversely, TG explants from uninfected mice or mice latently infected with a LAT-/- mutant increased the number of ß-catenin+ TG neurons in the presence of DEX relative to samples not treated with DEX. Impairing Wnt signaling with small molecule antagonists reduced virus shedding during explant-induced reactivation. These studies suggested ß-catenin was differentially expressed during the latency-reactivation cycle, in part due to LAT expression.


Asunto(s)
Regulación de la Expresión Génica , Herpesvirus Humano 1/fisiología , Neuronas/metabolismo , Neuronas/virología , Ganglio del Trigémino/citología , Activación Viral , beta Catenina/metabolismo , Animales , Femenino , Ratones , Vía de Señalización Wnt
15.
Toxicol Lett ; 319: 74-84, 2020 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-31707104

RESUMEN

Benzalkonium chloride (BAK), a quaternary ammonium compound widely used as disinfecting agent as well as preservative in eye drops is known to induce toxic effects on the ocular surface with inflammation and corneal nerve damage leading to dry eye disease (DED) in the medium-to-long term. The aim of this study was to evaluate in vitro the toxicity of a conditioned medium produced by corneal epithelial cells previously exposed to BAK (BAK-CM) on trigeminal neuronal cells. A human corneal epithelial (HCE) cell line was exposed to 5.10-3% BAK (i.e. 0.005% BAK) for 15 min and let recover for 5 h to prepare a BAK-CM. This BAK concentration is the lowest one found in eye drops. After this recovery period, BAK effect on HCE cells displayed cytotoxicity, morphological alteration, apoptosis, oxidative stress, ATP release, CCL2 and IL6 gene induction, as well as an increase in CCL2, IL-6 and MIF release. Next, a mouse trigeminal ganglion primary culture was exposed to the BAK-CM for 2 h, 4 h or 24 h. Whereas BAK-CM did not alter neuronal cell morphology, or induced neuronal cytotoxicity or oxidative stress, BAK-CM induced gene expression of Fos (neuronal activation marker), Atf3 (neuronal injury marker), Ccl2 and Il6 (inflammatory markers). Two and 4 h BAK-CM exposure promoted a neuronal damage (ATF-3, phospho-p38 increases; phospho-Stat3 decreases) while 24 h-BAK-CM exposure initiated a prosurvival pathway activation (phospho-p44/42, phospho-Akt increases; ATF-3, GADD153, active Caspase-3 decreases). In conclusion, this in vitro model, simulating paracrine mechanisms, represents an interesting tool to highlight the indirect toxic effects of BAK or any other xenobiotic on corneal trigeminal neurons and may help to better understand the cellular mechanisms that occur during DED pathophysiology.


Asunto(s)
Apoptosis/efectos de los fármacos , Compuestos de Benzalconio/toxicidad , Células Epiteliales/efectos de los fármacos , Epitelio Corneal/efectos de los fármacos , Inflamación/inducido químicamente , Neuronas/efectos de los fármacos , Conservadores Farmacéuticos/toxicidad , Ganglio del Trigémino/efectos de los fármacos , Factor de Transcripción Activador 3/biosíntesis , Factor de Transcripción Activador 3/efectos de los fármacos , Animales , Línea Celular , Síndromes de Ojo Seco/inducido químicamente , Síndromes de Ojo Seco/patología , Epitelio Corneal/citología , Expresión Génica/efectos de los fármacos , Humanos , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Ganglio del Trigémino/citología
16.
Int J Biol Sci ; 15(12): 2676-2691, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31754339

RESUMEN

It is well known that the cornea plays an important role in providing protection to the eye, but it is fragile and vulnerable. To clarify the biological effects and molecular mechanisms of the pituitary adenylate cyclase activating polypeptide (PACAP)-derived peptide MPAPO (named MPAPO) to promote corneal wound healing, we applied a mechanical method to establish a corneal injury model and analyzed the repair effects of MPAPO on corneal injury. MPAPO significantly promoted corneal wound repair in C57BL/6 mice. In addition, we established injury models of epithelial cells and trigeminal ganglion cells with H2O2. The results show that when the concentration of MPAPO is 1 µM, it can significantly promote the repair of injured corneal epithelial cells and the regeneration of trigeminal ganglion cell axons. MPAPO repairs epithelial cells through the promotion of GSK3ß phosphorylation by binding to PAC1 and activating AKT. ß-catenin escapes the phosphorylation of GSK3ß and enters the nucleus to promote the expression of cyclin D1, accelerate cell cycle progression and promote cell proliferation. MPAPO promotes axonal regeneration by binding to the PAC1 receptor and activating adenylate cyclase activity, followed by the cAMP activation of protein kinase A activity and the promotion of CREB phosphorylation. Phosphorylated CREB promotes Bcl2 expression and axonal regeneration. In conclusion, our data support the role of MPAPO to facilitate corneal wound healing by promoting corneal epithelial cell proliferation and trigeminal ganglion cell axon regeneration.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Lesiones de la Cornea/tratamiento farmacológico , Células Epiteliales/efectos de los fármacos , Péptidos/farmacología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Ganglio del Trigémino/citología , Animales , Axones/efectos de los fármacos , Axones/fisiología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Córnea , Masculino , Ratones , Ratones Endogámicos C57BL , Regeneración Nerviosa/efectos de los fármacos , Péptidos/química , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/química , Distribución Aleatoria , Ganglio del Trigémino/efectos de los fármacos
17.
Nitric Oxide ; 93: 90-101, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31604145

RESUMEN

The mechanisms underlying temporomandibular disorders following orofacial pain remain unclear. Hydrogen sulfide (H2S), a newly identified gasotransmitter, has been reported to modulate inflammation. Cystathionine γ-lyase (CSE) is responsible for the systemical production of H2S, which exerts both pro- and antinociceptive effects through inflammation. In the current study, we investigated whether the endogenous H2S production pathway contributes to arousal and maintenance of orofacial inflammatory pain, through the investigation of the effects of a CSE inhibitor, propargyglycine (PAG), in a rat CFA (Complete Freund Adjuvant)-induced temporomandibular inflammation model to mimic persistent pain in the orofacial region. For this, rats received either CFA or saline in the temporomandibular joints (TMJs), and after 3 or 14 days, they received a single injection of PAG or saline and were evaluated for nociception with the von Frey and formalin test. Also, pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α), and interleukin-1ß (IL-1ß) were analyzed in TMJs and trigeminal ganglion (TG). In this last one, glial cells reactivity was also verified. Endogenous H2S production rate were measured in both, TMJ and TG. Our results indicated decreased allodynia and hyperalgesic responses in rats submitted to CFA after injection of PAG. Moreover, PAG inhibited leucocyte migration to temporomandibular synovial fluid after 3 and 14 days of inflammation. PAG was able to reduce levels of CBS, CSE, TNF-α, and IL-1ß in the TMJ and TG, after 13 days of CFA injection. The observed increased activation of glial cells in the trigeminal ganglia on the 14th day of inflammation can be prevented by the highest dose of PAG. Finally, CBS and CSE expression, and endogenous H2S production rate in the TMJ and TG was found higher in rats with persistent temporomandibular inflammation compared to rats injected with saline and PAG was able to prevent this elevation. Our results elucidated the molecular mechanisms by which H2S exerts its pro-inflammatory and pro-nociceptive role in the orofacial region by alterations in both local tissue and TG.


Asunto(s)
Alquinos/uso terapéutico , Glicina/análogos & derivados , Sulfuro de Hidrógeno/metabolismo , Hiperalgesia/tratamiento farmacológico , Inflamación/metabolismo , Dolor/tratamiento farmacológico , Articulación Temporomandibular/metabolismo , Animales , Cistationina gamma-Liasa/antagonistas & inhibidores , Inhibidores Enzimáticos/uso terapéutico , Glicina/uso terapéutico , Interleucina-1beta/metabolismo , Masculino , Neuroglía/efectos de los fármacos , Ratas Wistar , Ganglio del Trigémino/citología , Ganglio del Trigémino/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
18.
Sci Signal ; 12(600)2019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31551295

RESUMEN

Although brain-derived neurotrophic factor (BDNF) is implicated in the nociceptive signaling of peripheral sensory neurons, the underlying mechanisms remain largely unknown. Here, we elucidated the effects of BDNF on the neuronal excitability of trigeminal ganglion (TG) neurons and the pain sensitivity of rats mediated by T-type Ca2+ channels. BDNF reversibly and dose-dependently enhanced T-type channel currents through the activation of tropomyosin receptor kinase B (TrkB). Antagonism of phosphatidylinositol 3-kinase (PI3K) but not of its downstream target, the kinase AKT, abolished the BDNF-induced T-type channel response. BDNF application activated p38 mitogen-activated protein kinase (MAPK), and this effect was prevented by inhibition of PI3K but not of protein kinase A (PKA). Antagonism of either PI3K or p38 MAPK prevented the BDNF-induced stimulation of PKA activity, whereas PKA inhibition blocked the BDNF-mediated increase in T-type currents. BDNF increased the rate of action potential firing in TG neurons and enhanced the pain sensitivity of rats to mechanical stimuli. Moreover, inhibition of TrkB signaling abolished the increased mechanical sensitivity in a rat model of chronic inflammatory pain, and this effect was attenuated by either T-type channel blockade or knockdown of the channel Cav3.2. Together, our findings indicate that BDNF enhances T-type currents through the stimulation of TrkB coupled to PI3K-p38-PKA signaling, thereby inducing neuronal hyperexcitability of TG neurons and pain hypersensitivity in rats.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Canales de Calcio Tipo T/efectos de los fármacos , Dolor/fisiopatología , Células Receptoras Sensoriales/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Canales de Calcio Tipo T/metabolismo , Canales de Calcio Tipo T/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas Sprague-Dawley , Receptor trkB/metabolismo , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología , Transducción de Señal/efectos de los fármacos , Ganglio del Trigémino/citología , Ganglio del Trigémino/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
J Headache Pain ; 20(1): 87, 2019 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-31375062

RESUMEN

BACKGROUND: Migraine is a debilitating neurological disorder involving abnormal trigeminovascular activation and sensitization. However, the underlying cellular and molecular mechanisms remain unclear. METHODS: A rat model of conscious migraine was established through the electrical stimulation (ES) of the dural mater surrounding the superior sagittal sinus. Using patch clamp recording, immunofluorescent labelling, enzyme-linked immunosorbent assays and western blot analysis, we studied the effects of ES on sensory neuronal excitability and elucidated the underlying mechanisms mediated by voltage-gated ion channels. RESULTS: The calcitonin gene-related peptide (CGRP) level in the jugular vein blood and the number of CGRP-positive neurons in the trigeminal ganglia (TGs) were significantly increased in rats with ES-induced migraine. The application of ES increased actional potential firing in both small-sized IB4-negative (IB4-) and IB4+ TG neurons. No significant changes in voltage-gated Na+ currents were observed in the ES-treated groups. ES robustly suppressed the transient outward K+ current (IA) in both types of TG neurons, while the delayed rectifier K+ current remained unchanged. Immunoblot analysis revealed that the protein expression of Kv4.3 was significantly decreased in the ES-treated groups, while Kv1.4 remained unaffected. Interestingly, ES increased the P/Q-type and T-type Ca2+ currents in small-sized IB4- TG neurons, while there were no significant changes in the IB4+ subpopulation of neurons. CONCLUSION: These results suggest that ES decreases the IA in small-sized TG neurons and increases P/Q- and T-type Ca2+ currents in the IB4- subpopulation of TG neurons, which might contribute to neuronal hyperexcitability in a rat model of ES-induced migraine.


Asunto(s)
Estimulación Eléctrica/métodos , Seno Sagital Superior/metabolismo , Ganglio del Trigémino/metabolismo , Potenciales de Acción , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Masculino , Neuronas Aferentes/fisiología , Ratas , Ratas Sprague-Dawley , Seno Sagital Superior/citología , Ganglio del Trigémino/citología
20.
J Neurophysiol ; 122(4): 1606-1622, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31411931

RESUMEN

Rats use their whiskers to extract sensory information from their environment. While exploring, they analyze peripheral stimuli distributed over several whiskers. Previous studies have reported cross-whisker integration of information at several levels of the neuronal pathways from whisker follicles to the somatosensory cortex. In the present study, we investigated the possible coupling between whiskers at a preneuronal level, transmitted by the skin and muscles between follicles. First, we quantified the movement induced on one whisker by deflecting another whisker. Our results show significant mechanical coupling, predominantly when a given whisker's caudal neighbor in the same row is deflected. The magnitude of the effect was correlated with the diameter of the deflected whisker. In addition to changes in whisker angle, we observed curvature changes when the whisker shaft was constrained distally from the base. Second, we found that trigeminal ganglion neurons innervating a given whisker follicle fire action potentials in response to high-magnitude deflections of an adjacent whisker. This functional coupling also shows a bias toward the caudal neighbor located in the same row. Finally, we designed a two-whisker biomechanical model to investigate transmission of forces across follicles. Analysis of the whisker-follicle contact forces suggests that activation of mechanoreceptors in the ring sinus region could account for our electrophysiological results. The model can fully explain the observed caudal bias by the gradient in whisker diameter, with possible contribution of the intrinsic muscles connecting follicles. Overall, our study demonstrates the functional relevance of mechanical coupling on early information processing in the whisker system.NEW & NOTEWORTHY Rodents explore their environment actively by touching objects with their whiskers. A major challenge is to understand how sensory inputs from different whiskers are merged together to form a coherent tactile percept. We demonstrate that external sensory events on one whisker can influence the position of another whisker and, importantly, that they can trigger the activity of mechanoreceptors at its base. This cross-whisker interaction occurs pre-neuronally, through mechanical transmission of forces in the skin.


Asunto(s)
Mecanorreceptores/fisiología , Movimiento , Percepción del Tacto , Vibrisas/fisiología , Potenciales de Acción , Animales , Masculino , Ratas , Ratas Wistar , Ganglio del Trigémino/citología , Ganglio del Trigémino/fisiología , Vibrisas/inervación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...