Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Inflamm Res ; 73(4): 669-691, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38483556

RESUMEN

OBJECTIVE AND DESIGN: Our aim was to determine an age-dependent role of Nav1.8 and ASIC3 in dorsal root ganglion (DRG) neurons in a rat pre-clinical model of long-term inflammatory pain. METHODS: We compared 6 and 24 months-old female Wistar rats after cutaneous inflammation. We used behavioral pain assessments over time, qPCR, quantitative immunohistochemistry, selective pharmacological manipulation, ELISA and in vitro treatment with cytokines. RESULTS: Older rats exhibited delayed recovery from mechanical allodynia and earlier onset of spontaneous pain than younger rats after inflammation. Moreover, the expression patterns of Nav1.8 and ASIC3 were time and age-dependent and ASIC3 levels remained elevated only in aged rats. In vivo, selective blockade of Nav1.8 with A803467 or of ASIC3 with APETx2 alleviated mechanical and cold allodynia and also spontaneous pain in both age groups with slightly different potency. Furthermore, in vitro IL-1ß up-regulated Nav1.8 expression in DRG neurons cultured from young but not old rats. We also found that while TNF-α up-regulated ASIC3 expression in both age groups, IL-6 and IL-1ß had this effect only on young and aged neurons, respectively. CONCLUSION: Inflammation-associated mechanical allodynia and spontaneous pain in the elderly can be more effectively treated by inhibiting ASIC3 than Nav1.8.


Asunto(s)
Canales Iónicos Sensibles al Ácido , Hiperalgesia , Canal de Sodio Activado por Voltaje NAV1.8 , Dolor , Animales , Femenino , Ratas , Canales Iónicos Sensibles al Ácido/genética , Canales Iónicos Sensibles al Ácido/metabolismo , Canales Iónicos Sensibles al Ácido/farmacología , Analgésicos/uso terapéutico , Ganglios Espinales , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Inflamación/metabolismo , Dolor/tratamiento farmacológico , Dolor/metabolismo , Ratas Sprague-Dawley , Ratas Wistar , Células Receptoras Sensoriales/metabolismo , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo
2.
Nature ; 628(8008): 604-611, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38538784

RESUMEN

The immune system has a critical role in orchestrating tissue healing. As a result, regenerative strategies that control immune components have proved effective1,2. This is particularly relevant when immune dysregulation that results from conditions such as diabetes or advanced age impairs tissue healing following injury2,3. Nociceptive sensory neurons have a crucial role as immunoregulators and exert both protective and harmful effects depending on the context4-12. However, how neuro-immune interactions affect tissue repair and regeneration following acute injury is unclear. Here we show that ablation of the NaV1.8 nociceptor impairs skin wound repair and muscle regeneration after acute tissue injury. Nociceptor endings grow into injured skin and muscle tissues and signal to immune cells through the neuropeptide calcitonin gene-related peptide (CGRP) during the healing process. CGRP acts via receptor activity-modifying protein 1 (RAMP1) on neutrophils, monocytes and macrophages to inhibit recruitment, accelerate death, enhance efferocytosis and polarize macrophages towards a pro-repair phenotype. The effects of CGRP on neutrophils and macrophages are mediated via thrombospondin-1 release and its subsequent autocrine and/or paracrine effects. In mice without nociceptors and diabetic mice with peripheral neuropathies, delivery of an engineered version of CGRP accelerated wound healing and promoted muscle regeneration. Harnessing neuro-immune interactions has potential to treat non-healing tissues in which dysregulated neuro-immune interactions impair tissue healing.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina , Macrófagos , Neutrófilos , Nociceptores , Cicatrización de Heridas , Animales , Ratones , Comunicación Autocrina , Péptido Relacionado con Gen de Calcitonina/metabolismo , Péptido Relacionado con Gen de Calcitonina/farmacología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , 60574 , Macrófagos/citología , Macrófagos/metabolismo , Monocitos/citología , Monocitos/metabolismo , Músculo Esquelético , Canal de Sodio Activado por Voltaje NAV1.8/deficiencia , Canal de Sodio Activado por Voltaje NAV1.8/genética , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Neutrófilos/citología , Neutrófilos/metabolismo , Nociceptores/metabolismo , Comunicación Paracrina , Enfermedades del Sistema Nervioso Periférico/complicaciones , Proteína 1 Modificadora de la Actividad de Receptores/metabolismo , Regeneración/efectos de los fármacos , Piel , Trombospondina 1/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/inmunología , Humanos , Masculino , Femenino
3.
Int J Mol Sci ; 24(12)2023 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-37373335

RESUMEN

In heart failure and atrial fibrillation, a persistent Na+ current (INaL) exerts detrimental effects on cellular electrophysiology and can induce arrhythmias. We have recently shown that NaV1.8 contributes to arrhythmogenesis by inducing a INaL. Genome-wide association studies indicate that mutations in the SCN10A gene (NaV1.8) are associated with increased risk for arrhythmias, Brugada syndrome, and sudden cardiac death. However, the mediation of these NaV1.8-related effects, whether through cardiac ganglia or cardiomyocytes, is still a subject of controversial discussion. We used CRISPR/Cas9 technology to generate homozygous atrial SCN10A-KO-iPSC-CMs. Ruptured-patch whole-cell patch-clamp was used to measure the INaL and action potential duration. Ca2+ measurements (Fluo 4-AM) were performed to analyze proarrhythmogenic diastolic SR Ca2+ leak. The INaL was significantly reduced in atrial SCN10A KO CMs as well as after specific pharmacological inhibition of NaV1.8. No effects on atrial APD90 were detected in any groups. Both SCN10A KO and specific blockers of NaV1.8 led to decreased Ca2+ spark frequency and a significant reduction of arrhythmogenic Ca2+ waves. Our experiments demonstrate that NaV1.8 contributes to INaL formation in human atrial CMs and that NaV1.8 inhibition modulates proarrhythmogenic triggers in human atrial CMs and therefore NaV1.8 could be a new target for antiarrhythmic strategies.


Asunto(s)
Fibrilación Atrial , Insuficiencia Cardíaca , Humanos , Fibrilación Atrial/genética , Fibrilación Atrial/metabolismo , Estudio de Asociación del Genoma Completo , Antiarrítmicos/farmacología , Miocitos Cardíacos/metabolismo , Insuficiencia Cardíaca/metabolismo , Potenciales de Acción , Canal de Sodio Activado por Voltaje NAV1.8/genética , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo
4.
Philos Trans R Soc Lond B Biol Sci ; 378(1879): 20220175, 2023 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-37122207

RESUMEN

We illustrate use of induced pluripotent stem cells (iPSCs) as platforms for investigating cardiomyocyte phenotypes in a human family pedigree exemplified by novel heterozygous RYR2-A1855D and SCN10A-Q1362H variants occurring alone and in combination. The proband, a four-month-old boy, presented with polymorphic ventricular tachycardia. Genetic tests revealed double novel heterozygous RYR2-A1855D and SCN10A-Q1362H variants inherited from his father (F) and mother (M), respectively. His father showed ventricular premature beats; his mother was asymptomatic. Molecular biological characterizations demonstrated greater TNNT2 messenger RNA (mRNA) expression in the iPSCs-induced cardiomyocytes (iPS-CMs) than in the iPSCs. Cardiac troponin Ts became progressively organized but cytoplasmic RYR2 and SCN10A aggregations occurred in the iPS-CMs. Proband-specific iPS-CMs showed decreased RYR2 and SCN10A mRNA expression. The RYR2-A1855D variant resulted in premature spontaneous sarcoplasmic reticular Ca2+ transients, Ca2+ oscillations and increased action potential durations. SCN10A-Q1362H did not confer any specific phenotype. However, the combined heterozygous RYR2-A1855D and SCN10A-Q1362H variants in the proband iPS-CMs resulted in accentuated Ca2+ homeostasis disorders, action potential prolongation and susceptibility to early afterdepolarizations at high stimulus frequencies. These findings attribute the clinical phenotype in the proband to effects of the heterozygous RYR2 variant exacerbated by heterozygous SCN10A modification. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.


Asunto(s)
Células Madre Pluripotentes Inducidas , Taquicardia Ventricular , Humanos , Lactante , Masculino , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Calcio/metabolismo , Homeostasis , Mutación , Canal de Sodio Activado por Voltaje NAV1.8/genética , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/farmacología , Taquicardia Ventricular/genética , Taquicardia Ventricular/metabolismo
5.
Mol Pain ; 19: 17448069231170072, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37002193

RESUMEN

BACKGROUND: Irritable bowel syndrome (IBS) is a common functional gastrointestinal disorder, and its specific pathogenesis is still unclear. We have previously reported that TTX-resistant (TTX-R) sodium channels in colon-specific dorsal root ganglion (DRG) neurons were sensitized in a rat model of visceral hypersensitivity induced by neonatal colonic inflammation (NCI). However, the detailed molecular mechanism for activation of sodium channels remains unknown. This study was designed to examine roles for melatonin (MT) in sensitization of sodium channels in NCI rats. METHODS: Colorectal distention (CRD) in adult male rats as a measure of visceral hypersensitivity. Colon-specific dorsal root ganglion (DRG) neurons were labeled with DiI and acutely dissociated for measuring excitability and sodium channel current under whole-cell patch clamp configurations. Western blot and Immunofluorescence were employed to detect changes in expression of Nav1.8 and MT2. RESULTS: The results showed that rats exhibited visceral hypersensitivity after NCI treatment. Intrathecal application of melatonin significantly increased the threshold of CRD in NCI rats with a dose-dependent manner, but has no role in the control group. Whole-cell patch clamp recording showed that melatonin remarkably decreased the excitability and the density of TTX-R sodium channel in DRG neurons from NCI rats. The expression of MT2 receptor at the protein level was markedly lower in NCI rats. 8MP, an agonist of MT2 receptor, enhanced the distention threshold in NCI rats. Application of 8MP reversed the enhanced hypersensitivity of DRG neurons from NCI rats. 8MP also reduced TTX-R sodium current density and modulated dynamics of TTX-R sodium current activation. CONCLUSIONS: These data suggest that sensitization of sodium channels of colon DRG neurons in NCI rats is most likely mediated by MT2 receptor, thus identifying a potential target for treatment for chronic visceral pain in patients with IBS.


Asunto(s)
Síndrome del Colon Irritable , Melatonina , Dolor Visceral , Ratas , Animales , Masculino , Síndrome del Colon Irritable/complicaciones , Síndrome del Colon Irritable/tratamiento farmacológico , Síndrome del Colon Irritable/metabolismo , Ratas Sprague-Dawley , Melatonina/farmacología , Melatonina/uso terapéutico , Melatonina/metabolismo , Dolor Visceral/metabolismo , Nocicepción , Receptor de Melatonina MT2/metabolismo , Ganglios Espinales/metabolismo , Tetrodotoxina , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo
6.
Nature ; 615(7952): 472-481, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36859544

RESUMEN

The meninges are densely innervated by nociceptive sensory neurons that mediate pain and headache1,2. Bacterial meningitis causes life-threatening infections of the meninges and central nervous system, affecting more than 2.5 million people a year3-5. How pain and neuroimmune interactions impact meningeal antibacterial host defences are unclear. Here we show that Nav1.8+ nociceptors signal to immune cells in the meninges through the neuropeptide calcitonin gene-related peptide (CGRP) during infection. This neuroimmune axis inhibits host defences and exacerbates bacterial meningitis. Nociceptor neuron ablation reduced meningeal and brain invasion by two bacterial pathogens: Streptococcus pneumoniae and Streptococcus agalactiae. S. pneumoniae activated nociceptors through its pore-forming toxin pneumolysin to release CGRP from nerve terminals. CGRP acted through receptor activity modifying protein 1 (RAMP1) on meningeal macrophages to polarize their transcriptional responses, suppressing macrophage chemokine expression, neutrophil recruitment and dural antimicrobial defences. Macrophage-specific RAMP1 deficiency or pharmacological blockade of RAMP1 enhanced immune responses and bacterial clearance in the meninges and brain. Therefore, bacteria hijack CGRP-RAMP1 signalling in meningeal macrophages to facilitate brain invasion. Targeting this neuroimmune axis in the meninges can enhance host defences and potentially produce treatments for bacterial meningitis.


Asunto(s)
Encéfalo , Meninges , Meningitis Bacterianas , Neuroinmunomodulación , Humanos , Encéfalo/inmunología , Encéfalo/microbiología , Péptido Relacionado con Gen de Calcitonina/metabolismo , Meninges/inmunología , Meninges/microbiología , Meninges/fisiopatología , Dolor/etiología , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Meningitis Bacterianas/complicaciones , Meningitis Bacterianas/inmunología , Meningitis Bacterianas/microbiología , Meningitis Bacterianas/patología , Streptococcus agalactiae/inmunología , Streptococcus agalactiae/patogenicidad , Streptococcus pneumoniae/inmunología , Streptococcus pneumoniae/patogenicidad , Nociceptores/metabolismo , Proteína 1 Modificadora de la Actividad de Receptores/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo
7.
Mol Psychiatry ; 28(1): 76-82, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36224259

RESUMEN

Pitt Hopkins Syndrome (PTHS) is a rare syndromic form of autism spectrum disorder (ASD) caused by autosomal dominant mutations in the Transcription Factor 4 (TCF4) gene. TCF4 is a basic helix-loop-helix transcription factor that is critical for neurodevelopment and brain function through its binding to cis-regulatory elements of target genes. One potential therapeutic strategy for PTHS is to identify dysregulated target genes and normalize their dysfunction. Here, we propose that SCN10A is an important target gene of TCF4 that is an applicable therapeutic approach for PTHS. Scn10a encodes the voltage-gated sodium channel Nav1.8 and is consistently shown to be upregulated in PTHS mouse models. In this perspective, we review prior literature and present novel data that suggests inhibiting Nav1.8 in PTHS mouse models is effective at normalizing neuron function, brain circuit activity and behavioral abnormalities and posit this therapeutic approach as a treatment for PTHS.


Asunto(s)
Discapacidad Intelectual , Canal de Sodio Activado por Voltaje NAV1.8 , Animales , Ratones , Trastorno del Espectro Autista/tratamiento farmacológico , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Facies , Hiperventilación/genética , Discapacidad Intelectual/tratamiento farmacológico , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Factor de Transcripción 4/genética , Canal de Sodio Activado por Voltaje NAV1.8/química , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo
8.
Mol Pain ; 19: 17448069221150138, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36550597

RESUMEN

Polysorbate 80 is a non-ionic detergent derived from polyethoxylated sorbitan and oleic acid. It is widely used in pharmaceuticals, foods, and cosmetics as an emulsifier. Nav1.7 is a peripheral sodium channel that is highly expressed in sympathetic and sensory neurons, and it plays a critical role in determining the threshold of action potentials (APs). We found that 10 µg/mL polysorbate 80 either abolished APs or increased the threshold of the APs of dorsal root ganglions. We thus investigated whether polysorbate 80 inhibits Nav1.7 sodium current using a whole-cell patch-clamp recording technique. Polysorbate 80 decreased the Nav1.7 current in a concentration-dependent manner with a half-maximal inhibitory concentration (IC50) of 250.4 µg/mL at a holding potential of -120 mV. However, the IC50 was 1.1 µg/mL at a holding potential of -90 mV and was estimated to be 0.9 µg/mL at the resting potentials of neurons, where most channels are inactivated. The activation rate and the voltage dependency of activation of Nav1.7 were not changed by polysorbate 80. However, polysorbate 80 caused hyperpolarizing shifts in the voltage dependency of the steady-state fast inactivation curve. The blocking of Nav1.7 currents by polysorbate 80 was not reversible at a holding potential of -90 mV but was completely reversible at -120 mV, where the channels were mostly in the closed state. Polysorbate 80 also slowed recovery from inactivation and induced robust use-dependent inhibition, indicating that it is likely to bind to and stabilize the inactivated state. Our results indicate that polysorbate 80 inhibits Nav1.7 current in concentration-, state-, and use-dependent manners when used even below commercial concentrations. This suggests that polysorbate 80 may be helpful in pain medicine as an excipient. In addition, in vitro experiments using polysorbate 80 with neurons should be conducted with caution.


Asunto(s)
Neuronas , Polisorbatos , Polisorbatos/farmacología , Polisorbatos/metabolismo , Neuronas/metabolismo , Canales de Sodio/metabolismo , Potenciales de la Membrana/fisiología , Potenciales de Acción , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo
9.
eNeuro ; 9(2)2022.
Artículo en Inglés | MEDLINE | ID: mdl-35131865

RESUMEN

The antidiabetic drug metformin has been shown to reduce pain hypersensitivity in preclinical models of chronic pain and in neuropathic pain in humans. Multiple intracellular pathways have been described as metformin targets. Among them, metformin is an activator of the adenosine 5'-monophosphate protein kinase that can in turn modulate the activity of the E3 ubiquitin ligase NEDD4-2 and thus post-translational expression of voltage-gated sodium channels (NaVs). In this study, we found that the bulk of the effect of metformin on Na1.7 is dependent on NEDD4-2. In HEK cells, the expression of NaV1.7 at the membrane fraction, obtained by a biotinylation approach, is only reduced by metformin when cotransfected with NEDD4-2. Similarly, in voltage-clamp recordings, metformin significantly reduced NaV1.7 current density when cotransfected with NEDD4-2. In mouse dorsal root ganglion (DRG) neurons, without changing the biophysical properties of NaV1.7, metformin significantly decreased NaV1.7 current densities, but not in Nedd4L knock-out mice (SNS-Nedd4L-/-). In addition, metformin induced a significant reduction in NEDD4-2 phosphorylation at the serine-328 residue in DRG neurons, an inhibitory phosphorylation site of NEDD4-2. In current-clamp recordings, metformin reduced the number of action potentials elicited by DRG neurons from Nedd4Lfl/fl , with a partial decrease also present in SNS-Nedd4L-/- mice, suggesting that metformin can also change neuronal excitability in an NEDD4-2-independent manner. We suggest that NEDD4-2 is a critical player for the effect of metformin on the excitability of nociceptive neurons; this action may contribute to the relief of neuropathic pain.


Asunto(s)
Metformina , Canales de Sodio Activados por Voltaje , Animales , Ganglios Espinales/metabolismo , Hipoglucemiantes/farmacología , Metformina/metabolismo , Metformina/farmacología , Ratones , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Ubiquitina/metabolismo , Ubiquitina/farmacología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Canales de Sodio Activados por Voltaje/metabolismo
10.
Clin Exp Pharmacol Physiol ; 49(3): 350-359, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34750860

RESUMEN

It has been repeatedly proved that Nav1.8 tetrodotoxin (TTX)-resistant sodium currents are expressed in peripheral sensory neurons where they play important role in nociception. There are very few publications that show the presence of TTX-resistant sodium currents in central neurons. The aim of this study was to assess if functional Nav1.8 TTX-resistant sodium currents are expressed in prefrontal cortex pyramidal neurons. All recordings were performed in the presence of TTX in the extracellular solution to block TTX-sensitive sodium currents. The TTX-resistant sodium current recorded in this study was mainly carried by the Nav1.8 sodium channel isoform because the Nav1.9 current was inhibited by the -65 mV holding potential that we used throughout the study. Moreover, the sodium current that we recorded was inhibited by treatment with the selective Nav1.8 inhibitor A-803467. Confocal microscopy experiments confirmed the presence of the Nav1.8 α subunit in prefrontal cortex pyramidal neurons. Activation and steady state inactivation properties of TTX-resistant sodium currents were also assessed in this study and they were similar to activation and inactivation properties of TTX-resistant sodium currents expressed in dorsal root ganglia (DRG) neurons. Moreover, this study showed that carbamazepine (60 µM) inhibited the maximal amplitude of the TTX-resistant sodium current. Furthermore, we found that carbamazepine shifts steady state inactivation curve of TTX-resistant sodium currents toward hyperpolarization. This study suggests that the Nav1.8 TTX-resistant sodium channel is expressed not only in DRG neurons, but also in cortical neurons and may be molecular target for antiepileptic drugs such as carbamazepine.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Corteza Prefrontal/citología , Células Piramidales/fisiología , Sodio/metabolismo , Tetrodotoxina/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Anticonvulsivantes/farmacología , Carbamazepina/farmacología , Activación del Canal Iónico/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.8/genética , Ratas , Ratas Wistar
11.
Nat Commun ; 12(1): 6586, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34782600

RESUMEN

An interplay between Ca2+/calmodulin-dependent protein kinase IIδc (CaMKIIδc) and late Na+ current (INaL) is known to induce arrhythmias in the failing heart. Here, we elucidate the role of the sodium channel isoform NaV1.8 for CaMKIIδc-dependent proarrhythmia. In a CRISPR-Cas9-generated human iPSC-cardiomyocyte homozygous knock-out of NaV1.8, we demonstrate that NaV1.8 contributes to INaL formation. In addition, we reveal a direct interaction between NaV1.8 and CaMKIIδc in cardiomyocytes isolated from patients with heart failure (HF). Using specific blockers of NaV1.8 and CaMKIIδc, we show that NaV1.8-driven INaL is CaMKIIδc-dependent and that NaV1.8-inhibtion reduces diastolic SR-Ca2+ leak in human failing cardiomyocytes. Moreover, increased mortality of CaMKIIδc-overexpressing HF mice is reduced when a NaV1.8 knock-out is introduced. Cellular and in vivo experiments reveal reduced ventricular arrhythmias without changes in HF progression. Our work therefore identifies a proarrhythmic CaMKIIδc downstream target which may constitute a prognostic and antiarrhythmic strategy.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Insuficiencia Cardíaca/metabolismo , Homeostasis/genética , Canal de Sodio Activado por Voltaje NAV1.8/genética , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Animales , Arritmias Cardíacas/metabolismo , Sistemas CRISPR-Cas , Insuficiencia Cardíaca/patología , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Medicina Molecular , Miocitos Cardíacos
12.
Invest Ophthalmol Vis Sci ; 62(14): 15, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34787642

RESUMEN

Purpose: The aim of the present study was to investigate the contribution of Nav1.8 expressing corneal afferent neurons to the presence of ongoing pain in lacrimal gland excision (LGE)-induced dry eye. Methods: The proton pump archaerhodopsin-3/eGFP (ArchT/eGFP) was conditionally expressed in corneal afferents using Nav1.8-cre mice. Dry eye was produced by unilateral LGE. Real time place preference was assessed using a three-chamber apparatus. A neutral, unlit center chamber was flanked by one illuminated with a control light and one illuminated with an ArchT activating light. For real-time preference, animals were placed in the neutral chamber and tracked over five 10-minute sessions, with the lights turned on during the second and fourth sessions. In other studies, movement was tracked over three 10-minute sessions (the lights turned on only during the second session), with animals tested once per day over the course of 4 days. A local anesthetic was used to examine the role of ongoing corneal afferent activity in producing place preference. Results: The corneal afferent nerves and trigeminal ganglion cell bodies showed a robust eGFP signal in Nav1.8-cre;ArchT/eGFP mice. After LGE, Nav1.8-cre;ArchT/eGFP mice demonstrated a preference for the ArchT activating light paired chamber. Preference was prevented with pre-application to the cornea of a local anesthetic. Nav1.8-cre;ArchT/eGFP mice with sham surgery and LGE wild-type control mice did not develop preference. Conclusions: Results indicate LGE-induced persistent, ongoing pain, driven by Nav1.8 expressing corneal afferents. Inhibition of these neurons represents a potential strategy for treating ongoing dry eye-induced pain.


Asunto(s)
Córnea/inervación , Síndromes de Ojo Seco/prevención & control , Dolor Ocular/prevención & control , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Neuronas Aferentes/metabolismo , Nervio Oftálmico/metabolismo , Optogenética/métodos , Analgesia/métodos , Animales , Proteína Coatómero/metabolismo , Modelos Animales de Enfermedad , Síndromes de Ojo Seco/metabolismo , Síndromes de Ojo Seco/fisiopatología , Dolor Ocular/metabolismo , Dolor Ocular/fisiopatología , Femenino , Fluoresceína/metabolismo , Colorantes Fluorescentes/metabolismo , Aparato Lagrimal/cirugía , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
13.
Nat Commun ; 12(1): 5962, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34645823

RESUMEN

Pitt-Hopkins syndrome (PTHS) is a rare autism spectrum-like disorder characterized by intellectual disability, developmental delays, and breathing problems involving episodes of hyperventilation followed by apnea. PTHS is caused by functional haploinsufficiency of the gene encoding transcription factor 4 (Tcf4). Despite the severity of this disease, mechanisms contributing to PTHS behavioral abnormalities are not well understood. Here, we show that a Tcf4 truncation (Tcf4tr/+) mouse model of PTHS exhibits breathing problems similar to PTHS patients. This behavioral deficit is associated with selective loss of putative expiratory parafacial neurons and compromised function of neurons in the retrotrapezoid nucleus that regulate breathing in response to tissue CO2/H+. We also show that central Nav1.8 channels can be targeted pharmacologically to improve respiratory function at the cellular and behavioral levels in Tcf4tr/+ mice, thus establishing Nav1.8 as a high priority target with therapeutic potential in PTHS.


Asunto(s)
Haploinsuficiencia , Proteínas de Homeodominio/genética , Hiperventilación/genética , Discapacidad Intelectual/genética , Canal de Sodio Activado por Voltaje NAV1.8/genética , Neuronas/metabolismo , Factor de Transcripción 4/genética , Factores de Transcripción/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Bencimidazoles/farmacología , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/metabolismo , Tronco Encefálico/patología , Dióxido de Carbono/metabolismo , Dióxido de Carbono/farmacología , Modelos Animales de Enfermedad , Facies , Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Hiperventilación/tratamiento farmacológico , Hiperventilación/metabolismo , Hiperventilación/patología , Discapacidad Intelectual/tratamiento farmacológico , Discapacidad Intelectual/metabolismo , Discapacidad Intelectual/patología , Masculino , Ratones , Ratones Noqueados , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Desempeño Psicomotor/efectos de los fármacos , Desempeño Psicomotor/fisiología , Pirazoles/farmacología , Respiración/efectos de los fármacos , Factor de Transcripción 4/deficiencia , Factores de Transcripción/metabolismo
14.
Toxins (Basel) ; 13(7)2021 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-34357973

RESUMEN

The voltage-gated sodium channel Nav1.8 is linked to neuropathic and inflammatory pain, highlighting the potential to serve as a drug target. However, the biophysical mechanisms that regulate Nav1.8 activation and inactivation gating are not completely understood. Progress has been hindered by a lack of biochemical tools for examining Nav1.8 gating mechanisms. Arizona bark scorpion (Centruroides sculpturatus) venom proteins inhibit Nav1.8 and block pain in grasshopper mice (Onychomys torridus). These proteins provide tools for examining Nav1.8 structure-activity relationships. To identify proteins that inhibit Nav1.8 activity, venom samples were fractioned using liquid chromatography (reversed-phase and ion exchange). A recombinant Nav1.8 clone expressed in ND7/23 cells was used to identify subfractions that inhibited Nav1.8 Na+ current. Mass-spectrometry-based bottom-up proteomic analyses identified unique peptides from inhibitory subfractions. A search of the peptides against the AZ bark scorpion venom gland transcriptome revealed four novel proteins between 40 and 60% conserved with venom proteins from scorpions in four genera (Centruroides, Parabuthus, Androctonus, and Tityus). Ranging from 63 to 82 amino acids, each primary structure includes eight cysteines and a "CXCE" motif, where X = an aromatic residue (tryptophan, tyrosine, or phenylalanine). Electrophysiology data demonstrated that the inhibitory effects of bioactive subfractions can be removed by hyperpolarizing the channels, suggesting that proteins may function as gating modifiers as opposed to pore blockers.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Venenos de Escorpión/farmacología , Escorpiones , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio Activados por Voltaje/metabolismo , Animales , Arizona , Ratones , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Dolor , Péptidos , Corteza de la Planta , Proteómica , Escorpiones/metabolismo
15.
Toxins (Basel) ; 13(5)2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-34067828

RESUMEN

The primary studies have shown that scorpion analgesic peptide N58A has a significant effect on voltage-gated sodium channels (VGSCs) and plays an important role in neuropathic pain. The purpose of this study was to investigate the analgesic effect of N58A on trigeminal neuralgia (TN) and its possible mechanism. The results showed that N58A could significantly increase the threshold of mechanical pain and thermal pain and inhibit the spontaneous asymmetric scratching behavior of rats. Western blotting results showed that N58A could significantly reduce the protein phosphorylation level of ERK1/2, P38, JNK, and ERK5/CREB pathways and the expression of Nav1.8 and Nav1.9 proteins in a dose-dependent manner. The changes in current and kinetic characteristics of Nav1.8 and Nav1.9 channels in TG neurons were detected by the whole-cell patch clamp technique. The results showed that N58A significantly decreased the current density of Nav1.8 and Nav1.9 in model rats, and shifted the activation curve to hyperpolarization and the inactivation curve to depolarization. In conclusion, the analgesic effect of N58A on the chronic constriction injury of the infraorbital (IoN-CCI) model rats may be closely related to the regulation of the MAPK pathway and Nav1.8 and Nav1.9 sodium channels.


Asunto(s)
Analgésicos/farmacología , Péptidos/farmacología , Venenos de Escorpión/química , Neuralgia del Trigémino/tratamiento farmacológico , Analgésicos/administración & dosificación , Analgésicos/aislamiento & purificación , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.8/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Canal de Sodio Activado por Voltaje NAV1.9/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.9/metabolismo , Dolor/tratamiento farmacológico , Técnicas de Placa-Clamp , Péptidos/administración & dosificación , Péptidos/aislamiento & purificación , Ratas , Ratas Sprague-Dawley , Escorpiones , Tetrodotoxina/farmacología
16.
J Neurosci ; 41(30): 6371-6387, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34131037

RESUMEN

The nonpsychoactive phytocannabinoid cannabidiol (CBD) has been shown to have analgesic effects in animal studies but little is known about its mechanism of action. We examined the effects of CBD on intrinsic excitability of primary pain-sensing neurons. Studying acutely dissociated capsaicin-sensitive mouse DRG neurons at 37°C, we found that CBD effectively inhibited repetitive action potential firing, from 15-20 action potentials evoked by 1 s current injections in control to 1-3 action potentials with 2 µm CBD. Reduction of repetitive firing was accompanied by a reduction of action potential height, widening of action potentials, reduction of the afterhyperpolarization, and increased propensity to enter depolarization block. Voltage-clamp experiments showed that CBD inhibited both TTX-sensitive and TTX-resistant (TTX-R) sodium currents in a use-dependent manner. CBD showed strong state-dependent inhibition of TTX-R channels, with fast binding to inactivated channels during depolarizations and slow unbinding on repolarization. CBD alteration of channel availability at various voltages suggested that CBD binds especially tightly [Kd (dissociation constant), ∼150 nm] to the slow inactivated state of TTX-R channels, which can be substantially occupied at voltages as negative as -40 mV. Remarkably, CBD was more potent in inhibiting TTX-R channels and inhibiting action potential firing than the local anesthetic bupivacaine. We conclude that CBD might produce some of its analgesic effects by direct effects on neuronal excitability, with tight binding to the slow inactivated state of Nav1.8 channels contributing to effective inhibition of repetitive firing by modest depolarizations.SIGNIFICANCE STATEMENT Cannabidiol (CBD) has been shown to inhibit pain in various rodent models, but the mechanism of this effect is unknown. We describe the ability of CBD to inhibit repetitive action potential firing in primary nociceptive neurons from mouse dorsal root ganglia and analyze the effects on voltage-dependent sodium channels. We find that CBD interacts with TTX-resistant sodium channels in a state-dependent manner suggesting particularly tight binding to slow inactivated states of Nav1.8 channels, which dominate the overall inactivation of Nav1.8 channels for small maintained depolarizations from the resting potential. The results suggest that CBD can exert analgesic effects in part by directly inhibiting repetitive firing of primary nociceptors and suggest a strategy of identifying compounds that bind selectively to slow inactivated states of Nav1.8 channels for developing effective analgesics.


Asunto(s)
Analgésicos/farmacología , Cannabidiol/farmacología , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Nociceptores/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Células Cultivadas , Femenino , Ganglios Espinales , Masculino , Ratones , Canal de Sodio Activado por Voltaje NAV1.8/efectos de los fármacos , Nociceptores/metabolismo
17.
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
18.
Molecules ; 26(6)2021 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-33808667

RESUMEN

Novel α-aminoamide derivatives containing different benzoheterocyclics moiety were synthesized and evaluated as voltage-gated sodium ion channels blocks the treatment of pain. Compounds 6a, 6e, and 6f containing the benzofuran group displayed more potent in vivo analgesic activity than ralfinamide in both the formalin test and the writhing assay. Interestingly, they also exhibited potent in vitro anti-Nav1.7 and anti-Nav1.8 activity in the patch-clamp electrophysiology assay. Therefore, compounds 6a, 6e, and 6f, which have inhibitory potency for two pain-related Nav targets, could serve as new leads for the development of analgesic medicines.


Asunto(s)
Amidas , Analgésicos , Dolor/tratamiento farmacológico , Bloqueadores de los Canales de Sodio , Amidas/síntesis química , Amidas/química , Amidas/farmacología , Analgésicos/síntesis química , Analgésicos/química , Analgésicos/farmacología , Animales , Evaluación de Medicamentos , Masculino , Ratones , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Dolor/inducido químicamente , Dolor/metabolismo , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/farmacología
19.
Neurosci Lett ; 753: 135844, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33775738

RESUMEN

The two voltage gated sodium channels Nav1.7 and Nav1.8 are expressed in the peripheral nervous system and involved in various pain conditions including inflammatory and neuropathic pain. Rodent models bearing deletions or mutations of the corresponding genes, Scn9a and Scn10a, were created in order to understand the role of these channels in the pathophysiological mechanism underlying pain symptoms. This review summarizes the pain behavior profiles reported in Scn9a and Scn10a rodent models. The complete loss-of-function or knockout (KO) of Scn9a or Scn10a and the conditional KO (cKO) of Scn9a in specific cell populations were shown to decrease sensitivity to various pain stimuli. The Possum mutant mice bearing a dominant hypermorphic mutation in Scn10a revealed higher sensitivity to noxious stimuli. Several gain-of-function mutations were identified in patients with painful small fiber neuropathy. Future knowledge obtained from preclinical models bearing these mutations will allow understanding how these mutations affect pain. In addition, the review gives perspectives for creating models that better mimic patients' pain symptoms in view to developing novel analgesic strategies.


Asunto(s)
Analgésicos/farmacología , Canal de Sodio Activado por Voltaje NAV1.7/genética , Canal de Sodio Activado por Voltaje NAV1.8/genética , Dolor/genética , Neuropatía de Fibras Pequeñas/complicaciones , Analgésicos/uso terapéutico , Animales , Modelos Animales de Enfermedad , Mutación con Ganancia de Función , Humanos , Mutación con Pérdida de Función , Ratones , Ratones Noqueados , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Nocicepción/efectos de los fármacos , Dolor/tratamiento farmacológico , Ratas , Ratas Transgénicas , Neuropatía de Fibras Pequeñas/tratamiento farmacológico , Neuropatía de Fibras Pequeñas/genética , Agonistas del Canal de Sodio Activado por Voltaje/farmacología , Agonistas del Canal de Sodio Activado por Voltaje/uso terapéutico , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Bloqueadores del Canal de Sodio Activado por Voltaje/uso terapéutico
20.
Inflammation ; 44(4): 1405-1415, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33515125

RESUMEN

The aim of the present study was to observe the changes of TTX-R, Nav1.8, and Nav1.9 Na+ currents in MSU-induced gouty arthritis mice, and to explore the possibility of Nav1.8 and Nav1.9 channels as potential targets for gout pain treatment. Acute gouty arthritis was induced by monosodium urate (MSU) in mice. Swelling degree was evaluated by measuring the circumference of the ankle joint. Mechanical allodynia was assessed by applying the electronic von Frey. Na+ currents were recorded by patch-clamp techniques in acute isolated dorsal root ganglion (DRG) neurons. MSU treatment significantly increased the swelling degree of ankle joint and decreased the mechanical pain threshold. The amplitude of TTX-R Na+ current was significantly increased and reached its peak on the 4th day after injection of MSU. For TTX-R Na+ channel subunits, Nav1.8 current density was significantly increased, but Nav1.9 current density was markedly decreased after MSU treatment. MSU treatment shifted the steady-state activation curves of TTX-R Na+ channel, Nav1.8 and Nav1.9 channels, and the inactivation curves of TTX-R Na+ channel and Nav1.8 channels to the depolarizing direction, but did not affect the inactivation curve of Nav1.9 channel. Compared with the normal group, the recovery of Nav1.8 channel was faster, while that of Nav1.9 channel was slower. The recovery of TTX-R Na+ channel remained unchanged after MSU treatment. Additionally, MSU treatment increased DRG neurons excitability by reducing action potential threshold. Nav1.8 channel, not Nav1.9 channel, may be involved in MSU-induced gout pain by increasing nerve excitability.


Asunto(s)
Artritis Gotosa/inducido químicamente , Artritis Gotosa/metabolismo , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Canal de Sodio Activado por Voltaje NAV1.9/metabolismo , Ácido Úrico/toxicidad , Animales , Artritis Gotosa/patología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Hiperalgesia/patología , Masculino , Ratones , Ratones Endogámicos ICR , Bloqueadores de los Canales de Sodio/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA