Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Cell Mol Neurobiol ; 42(3): 597-620, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33095392

RESUMEN

Sigma-1 receptor (Sig-1R) is a protein present in several organs such as brain, lung, and heart. In a cell, Sig-1R is mainly located across the membranes of the endoplasmic reticulum and more specifically at the mitochondria-associated membranes. Despite numerous studies showing that Sig-1R could be targeted to rescue several cellular mechanisms in different pathological conditions, less is known about its fundamental relevance. In this review, we report results from various studies and focus on the importance of Sig-1R in physiological conditions by comparing Sig-1R KO mice to wild-type mice in order to investigate the fundamental functions of Sig-1R. We note that the Sig-1R deletion induces cognitive, psychiatric, and motor dysfunctions, but also alters metabolism of heart. Finally, taken together, observations from different experiments demonstrate that those dysfunctions are correlated to poor regulation of ER and mitochondria metabolism altered by stress, which could occur with aging.


Asunto(s)
Receptores sigma , Animales , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico , Ratones , Mitocondrias/metabolismo , Receptores sigma/genética , Receptores sigma/metabolismo , Receptor Sigma-1
2.
J Neurosci ; 35(42): 14086-102, 2015 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-26490852

RESUMEN

The sensation of touch is initiated when fast conducting low-threshold mechanoreceptors (Aß-LTMRs) generate impulses at their terminals in the skin. Plasticity in this system is evident in the process of adaption, in which a period of diminished sensitivity follows prior stimulation. CaMKII is an ideal candidate for mediating activity-dependent plasticity in touch because it shifts into an enhanced activation state after neuronal depolarizations and can thereby reflect past firing history. Here we show that sensory neuron CaMKII autophosphorylation encodes the level of Aß-LTMR activity in rat models of sensory deprivation (whisker clipping, tail suspension, casting). Blockade of CaMKII signaling limits normal adaptation of action potential generation in Aß-LTMRs in excised skin. CaMKII activity is also required for natural filtering of impulse trains as they travel through the sensory neuron T-junction in the DRG. Blockade of CaMKII selectively in presynaptic Aß-LTMRs removes dorsal horn inhibition that otherwise prevents Aß-LTMR input from activating nociceptive lamina I neurons. Together, these consequences of reduced CaMKII function in Aß-LTMRs cause low-intensity mechanical stimulation to produce pain behavior. We conclude that, without normal sensory activity to maintain adequate levels of CaMKII function, the touch pathway shifts into a pain system. In the clinical setting, sensory disuse may be a critical factor that enhances and prolongs chronic pain initiated by other conditions. SIGNIFICANCE STATEMENT: The sensation of touch is served by specialized sensory neurons termed low-threshold mechanoreceptors (LTMRs). We examined the role of CaMKII in regulating the function of these neurons. Loss of CaMKII function, such as occurred in rats during sensory deprivation, elevated the generation and propagation of impulses by LTMRs, and altered the spinal cord circuitry in such a way that low-threshold mechanical stimuli produced pain behavior. Because limbs are protected from use during a painful condition, this sensitization of LTMRs may perpetuate pain and prevent functional rehabilitation.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Mecanorreceptores/fisiología , Nociceptores/fisiología , Umbral del Dolor/fisiología , Dolor/fisiopatología , Tacto/genética , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Dependovirus/genética , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Ganglios Espinales/citología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hiperalgesia/fisiopatología , Masculino , Mecanorreceptores/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Actividad Motora/genética , Proteínas del Tejido Nervioso/metabolismo , Dolor/etiología , Enfermedades del Sistema Nervioso Periférico/complicaciones , Ratas , Ratas Sprague-Dawley , Privación Sensorial/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Piel/inervación
3.
J Transl Med ; 14: 18, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26792056

RESUMEN

BACKGROUND: Heart failure with ejection fraction (HFpEF) is a syndrome resulting from several co-morbidities in which specific mediators are unknown. The platelet proteome responds to disease processes. We hypothesize that the platelet proteome will change composition in patients with HFpEF and may uncover mediators of the syndrome. METHODS AND RESULTS: Proteomic changes were assessed in platelets from hospitalized subjects with symptoms of HFpEF (n = 9), the same subjects several weeks later without symptoms (n = 7) and control subjects (n = 8). Mass spectrometry identified 6102 proteins with five scans with peptide probabilities of ≥0.85. Of the 6102 proteins, 165 were present only in symptomatic subjects, 78 were only found in outpatient subjects and 157 proteins were unique to the control group. The S100A8 protein was identified consistently in HFpEF samples when compared with controls. We validated the fining that plasma S100A8 levels are increased in subjects with HFpEF (654 ± 391) compared to controls (352 ± 204) in an external cohort (p = 0.002). Recombinant S100A8 had direct effects on the electrophysiological and calcium handling profile in human induced pluripotent stem cell-derived cardiomyocytes. CONCLUSIONS: Platelets may harbor proteins associated with HFpEF. S100A8 is present in the platelets of subjects with HFpEF and increased in the plasma of the same subjects. We further established a bedside-to-bench translational system that can be utilized as a secondary screen to ascertain whether the biomarkers may be an associated finding or causal to the disease process. S100A8 has been linked with other cardiovascular disease such as atherosclerosis and risk for myocardial infarction, stroke, or death. This is the first report on association of S100A8 with HFpEF.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteómica/métodos , Volumen Sistólico , Anciano , Secuencia de Aminoácidos , Calgranulina A/sangre , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática , Femenino , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/patología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Miocitos Cardíacos/efectos de los fármacos , Péptidos/química , Fenotipo , Proteoma/metabolismo , Proteínas Recombinantes/farmacología , Reproducibilidad de los Resultados , Volumen Sistólico/efectos de los fármacos , Espectrometría de Masas en Tándem , Ultrasonografía
4.
Mol Pain ; 11: 5, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25888914

RESUMEN

BACKGROUND: Cell-based therapy may hold promise for treatment of chronic pain. Mesenchymal stem cells (MSCs) are readily available and robust, and their secretion of therapeutic peptides can be enhanced by genetically engineering. We explored the analgesic potential of transplanting bone marrow-derived MSCs that have been transduced with lentivectors. To optimize efficacy and safety, primary sensory neurons were targeted by MSC injection into the dorsal root ganglia (DRGs). RESULTS: MSCs were transduced using lentivectors to express enhanced green fluorescent protein (EGFP) or to co-express the analgesic peptide glial cell line-derived neurotrophic factor (GDNF) and EGFP by a viral 2A bicistronic transgene cassette. Engineered MSCs were injected into the 4(th) lumbar (L4) and L5 DRGs of adult allogeneic rats to evaluate survival in the DRGs. MSCs were detected by immunofluorescence staining up to 2-3 weeks after injection, distributed in the extracellular matrix space without disrupting satellite glial cell apposition to sensory neurons, suggesting well-tolerated integration of engrafted MSCs into DRG tissue. To examine their potential for inhibiting development of neuropathic pain, MSCs were injected into the L4 and L5 DRGs ipsilateral to a spinal nerve ligation injury. Animals injected with GDNF-engineered MSCs showed moderate but significant reduction in mechanical allodynia and hyperalgesia compared to controls implanted with MSCs expressing EGFP alone. We also observed diminished long-term survival of allografted MSCs at 3 weeks, and the development of a highly-proliferating population of MSCs in 12% of DRGs after transplantation. CONCLUSIONS: These data indicate that genetically modified MSCs secreting analgesic peptides could potentially be developed as a novel DRG-targeted cell therapy for treating neuropathic pain. However, further work is needed to address the challenges of MSC survival and excess proliferation, possibly with trials of autologous MSCs, evaluation of clonally selected populations of MSCs, and investigation of regulation of MSC proliferation.


Asunto(s)
Analgesia , Ganglios Espinales/trasplante , Células Madre Mesenquimatosas/citología , Neuralgia/terapia , Neuronas Aferentes/citología , Analgesia/métodos , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Ganglios Espinales/metabolismo , Masculino , Trasplante de Células Madre Mesenquimatosas , Neuralgia/genética , Neuralgia/metabolismo , Manejo del Dolor/métodos , Ratas Sprague-Dawley , Nervios Espinales/metabolismo
5.
Mol Cell Neurosci ; 62: 10-8, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25064143

RESUMEN

Calcium/calmodulin-dependent protein kinase II (CaMKII) is recognized as a key element in encoding depolarization activity of excitable cells into facilitated voltage-gated Ca(2+) channel (VGCC) function. Less is known about the participation of CaMKII in regulating VGCCs in resting cells. We examined constitutive CaMKII control of Ca(2+) currents in peripheral sensory neurons acutely isolated from dorsal root ganglia (DRGs) of adult rats. The small molecule CaMKII inhibitor KN-93 (1.0µM) reduced depolarization-induced ICa by 16-30% in excess of the effects produced by the inactive homolog KN-92. The specificity of CaMKII inhibition on VGCC function was shown by the efficacy of the selective CaMKII blocking peptide autocamtide-2-related inhibitory peptide in a membrane-permeable myristoylated form, which also reduced VGCC current in resting neurons. Loss of VGCC currents is primarily due to reduced N-type current, as application of mAIP selectively reduced N-type current by approximately 30%, and prior N-type current inhibition eliminated the effect of mAIP on VGCCs, while prior block of L-type channels did not reduce the effect of mAIP on total ICa. T-type currents were not affected by mAIP in resting DRG neurons. Transduction of sensory neurons in vivo by DRG injection of an adeno-associated virus expressing AIP also resulted in a loss of N-type currents. Together, these findings reveal a novel molecular adaptation whereby sensory neurons retain CaMKII support of VGCCs despite remaining quiescent.


Asunto(s)
Canales de Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calcio/metabolismo , Ganglios Espinales/citología , Células Receptoras Sensoriales/metabolismo , Animales , Bloqueadores de los Canales de Calcio/farmacología , Masculino , Potenciales de la Membrana/fisiología , Neuronas Aferentes/metabolismo , Ratas Sprague-Dawley , Células Receptoras Sensoriales/efectos de los fármacos
6.
J Pharmacol Exp Ther ; 350(2): 290-300, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24891452

RESUMEN

Sigma-1 receptor (σ1R), an endoplasmic reticulum-chaperone protein, can modulate painful response after peripheral nerve injury. We have demonstrated that voltage-gated calcium current is inhibited in axotomized sensory neurons. We examined whether σ1R contributes to the sensory dysfunction of voltage-gated calcium channel (VGCC) after peripheral nerve injury through electrophysiological approach in dissociated rat dorsal root ganglion (DRG) neurons. Animals received either skin incision (Control) or spinal nerve ligation (SNL). Both σ1R agonists, (+)pentazocine (PTZ) and DTG [1,3-di-(2-tolyl)guanidine], dose dependently inhibited calcium current (ICa) with Ba(2+) as charge carrier in control sensory neurons. The inhibitory effect of σ1R agonists on ICa was blocked by σ1R antagonist, BD1063 (1-[2-(3,4-dichlorophenyl)ethyl]-4-m​ethylpiperazine dihydrochloride) or BD1047 (N-[2-(3,4-dichlorophenyl)ethyl]-N-m​ethyl-2-(dimethylamino)ethylamine dihydrobromide). PTZ and DTG showed similar effect on ICa in axotomized fifth DRG neurons (SNL L5). Both PTZ and DTG shifted the voltage-dependent activation and steady-state inactivation of VGCC to the left and accelerated VGCC inactivation rate in both Control and axotomized L5 SNL DRG neurons. The σ1R antagonist, BD1063 (10 µM), increases ICa in SNL L5 neurons but had no effect on Control and noninjured fourth lumbar neurons in SNL rats. Together, the findings suggest that activation of σR1 decreases ICa in sensory neurons and may play a pivotal role in pain generation.


Asunto(s)
Canales de Calcio/fisiología , Traumatismos de los Nervios Periféricos/tratamiento farmacológico , Canales de Potasio Calcio-Activados/fisiología , Receptores sigma/antagonistas & inhibidores , Células Receptoras Sensoriales/fisiología , Animales , Etilenodiaminas/farmacología , Ganglios Espinales/fisiología , Masculino , Traumatismos de los Nervios Periféricos/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptores sigma/fisiología , Receptor Sigma-1
7.
Mol Neurobiol ; 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38180612

RESUMEN

Expansion of the GGGGCC-RNA repeat is a known cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), which currently have no cure. Recent studies have indicated the activation of Sigma-1 receptor plays an important role in providing neuroprotection, especially in ALS and Alzheimer's disease. Nevertheless, the mechanisms underlying Sigma-1R activation and its effect on (G4C2)n-RNA-induced cell death remain unclear. In this study, we demonstrated that fluvoxamine is a Sigma-1R agonist that can increase chaperone activity and stabilize the protein expression of Pom121 in (G4C2)31-RNA-expressing NSC34 cells, leading to increased colocalization at the nuclear envelope. Interestingly, fluvoxamine treatment increased Pom121 protein expression without affecting transcription. In C9orf72-ALS, the nuclear translocation of TFEB autophagy factor decreased owing to nucleocytoplasmic transport defects. Our results showed that pretreatment of NSC34 cells with fluvoxamine promoted the shuttling of TFEB into the nucleus and elevated the expression of LC3-II compared to the overexpression of (G4C2)31-RNA alone. Additionally, even when used alone, fluvoxamine increases Pom121 expression and TFEB translocation. To summarize, fluvoxamine may act as a promising repurposed medicine for patients with C9orf72-ALS, as it stabilizes the nucleoporin Pom121 and promotes the translocation of TFEB in (G4C2)31-RNA-expressing NSC34 cells.

8.
J Neurosci ; 32(34): 11737-49, 2012 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-22915116

RESUMEN

Currents through voltage-gated Ca²âº channels (I(Ca)) may be regulated by cytoplasmic Ca²âº levels ([Ca²âº](c)), producing Ca²âº-dependent inactivation (CDI) or facilitation (CDF). Since I(Ca) regulates sensory neuron excitability, altered CDI or CDF could contribute to pain generation after peripheral nerve injury. We explored this by manipulating [Ca²âº](c) while recording I(Ca) in rat sensory neurons. In uninjured neurons, elevating [Ca²âº](c) with a conditioning prepulse (-15 mV, 2 s) inactivated I(Ca) measured during subsequent test pulses (-15 mV, 5 ms). This inactivation was Ca²âº-dependent (CDI), since it was decreased with elimination of Ca²âº influx by depolarization to above the I(Ca) reversal potential, with high intracellular Ca²âº buffering (EGTA 10 mm or BAPTA 20 mm), and with substitution of Ba²âº for extracellular Ca²âº, revealing a residual voltage-dependent inactivation. At longer latencies after conditioning (>6 s), I(Ca) recovered beyond baseline. This facilitation also proved to be Ca²âº-dependent (CDF) using the protocols limiting cytoplasmic Ca²âº elevation. Ca²âº/calmodulin-dependent protein kinase II (CaMKII) blockers applied by bath (KN-93, myristoyl-AIP) or expressed selectively in the sensory neurons (AIP) reduced CDF, unlike their inactive analogues. Protein kinase C inhibition (chelerythrine) had no effect. Selective blockade of N-type Ca²âº channels eliminated CDF, whereas L-type channel blockade had no effect. Following nerve injury, CDI was unaffected, but CDF was eliminated in axotomized neurons. Excitability of sensory neurons in intact ganglia from control animals was diminished after a similar conditioning pulse, but this regulation was eliminated by injury. These findings indicate that I(Ca) in sensory neurons is subject to both CDI and CDF, and that hyperexcitability following injury-induced loss of CDF may result from diminished CaMKII activity.


Asunto(s)
Fenómenos Biofísicos/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calcio/metabolismo , Neuronas Aferentes/fisiología , Traumatismos de los Nervios Periféricos/patología , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Fenómenos Biofísicos/efectos de los fármacos , Biofisica , Bloqueadores de los Canales de Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Quelantes/farmacología , Dantroleno/farmacología , Interacciones Farmacológicas , Ácido Egtácico/análogos & derivados , Estimulación Eléctrica , Inhibidores Enzimáticos/farmacología , Ganglios Espinales/citología , Vectores Genéticos/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hiperalgesia/etiología , Hiperalgesia/metabolismo , Laminectomía , Masculino , Potenciales de la Membrana/efectos de los fármacos , Neuronas Aferentes/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Técnicas de Placa-Clamp , Traumatismos de los Nervios Periféricos/complicaciones , Traumatismos de los Nervios Periféricos/enzimología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
9.
Mol Pain ; 9: 47, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-24015960

RESUMEN

BACKGROUND: The sigma-1 receptor (σ1R), an endoplasmic reticulum chaperone protein, is widely distributed and regulates numerous intracellular processes in neurons. Nerve injury alters the structure and function of axotomized dorsal root ganglion (DRG) neurons, contributing to the development of pain. The σ1R is enriched in the spinal cord and modulates pain after peripheral nerve injury. However, σ1R expression in the DRG has not been studied. We therefore characterized σ1R expression in DRGs at baseline and following spinal nerve ligation (SNL) in rats. RESULTS: Immunohistochemical (IHC) studies in DRG sections show σ1R in both neuronal somata and satellite glial cells. The punctate distribution of σ1R in the neuronal cytoplasm suggests expression in the endoplasmic reticulum. When classified by neuronal size, large neurons (>1300 µm) showed higher levels of σ1R staining than other groups (700-1300 µm, <700 µm). Comparing σ1R expression in neuronal groups characterized by expression of calcitonin gene-related peptide (CGRP), isolectin-B4 (IB4) and neurofilament-200 (NF-200), we found σ1R expression in all three neuronal subpopulations, with highest levels of σ1R expression in the NF-200 group. After SNL, lysates from L5 DRGs that contains axotomized neurons showed decreased σ1R protein but unaffected transcript level, compared with Control DRGs. IHC images also showed decreased σ1R protein expression, in SNL L5 DRGs, and to a lesser extent in the neighboring SNL L4 DRGs. Neurons labeled by CGRP and NF-200 showed decreased σ1R expression in L5 and, to a lesser extent, L4 DRGs. In IB4-labeled neurons, σ1R expression decreased only in axotomized L5 DRGs. Satellite cells also showed decreased σ1R expression in L5 DRGs after SNL. CONCLUSIONS: Our data show that σ1R is present in both sensory neurons and satellite cells in rat DRGs. Expression of σ1R is down-regulated in axotomized neurons as well as in their accompanying satellite glial cells, while neighboring uninjured neurons show a lesser down-regulation. Therefore, elevated σ1R expression in neuropathic pain is not an explanation for pain relief after σ1R blockade. This implies that increased levels of endogenous σ1R agonists may play a role, and diminished neuroprotection from loss of glial σ1R may be a contributing factor.


Asunto(s)
Regulación de la Expresión Génica , Traumatismos de los Nervios Periféricos/metabolismo , Receptores sigma/genética , Receptores sigma/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Retículo Endoplásmico/metabolismo , Ganglios Espinales/metabolismo , Masculino , Proteínas de Neurofilamentos/metabolismo , Ratas , Ratas Sprague-Dawley , Células Satélites Perineuronales/metabolismo , Receptor Sigma-1
10.
Autophagy ; 19(1): 126-151, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35507432

RESUMEN

Macroautophagy/autophagy is an essential process for cellular survival and is implicated in many diseases. A critical step in autophagy is the transport of the transcription factor TFEB from the cytosol into the nucleus, through the nuclear pore (NP) by KPNB1/importinß1. In the C9orf72 subtype of amyotrophic lateral sclerosis-frontotemporal lobar degeneration (ALS-FTD), the hexanucleotide (G4C2)RNA expansion (HRE) disrupts the nucleocytoplasmic transport of TFEB, compromising autophagy. Here we show that a molecular chaperone, the SIGMAR1/Sigma-1 receptor (sigma non-opioid intracellular receptor 1), facilitates TFEB transport into the nucleus by chaperoning the NP protein (i.e., nucleoporin) POM121 which recruits KPNB1. In NSC34 cells, HRE reduces TFEB transport by interfering with the association between SIGMAR1 and POM121, resulting in reduced nuclear levels of TFEB, KPNB1, and the autophagy marker LC3-II. Overexpression of SIGMAR1 or POM121, or treatment with the highly selective and potent SIGMAR1 agonist pridopidine, currently in phase 2/3 clinical trials for ALS and Huntington disease, rescues all of these deficits. Our results implicate nucleoporin POM121 not merely as a structural nucleoporin, but also as a chaperone-operated signaling molecule enabling TFEB-mediated autophagy. Our data suggest the use of SIGMAR1 agonists, such as pridopidine, for therapeutic development of diseases in which autophagy is impaired.Abbreviations: ALS-FTD, amyotrophic lateral sclerosis-frontotemporal dementiaC9ALS-FTD, C9orf72 subtype of amyotrophic lateral sclerosis-frontotemporal dementiaCS, citrate synthaseER, endoplasmic reticulumGSS, glutathione synthetaseHRE, hexanucleotide repeat expansionHSPA5/BiP, heat shock protein 5LAMP1, lysosomal-associated membrane protein 1MAM, mitochondria-associated endoplasmic reticulum membraneMAP1LC3/LC3, microtubule-associated protein 1 light chain 3NP, nuclear poreNSC34, mouse motor neuron-like hybrid cell lineNUPs, nucleoporinsPOM121, nuclear pore membrane protein 121SIGMAR1/Sigma-1R, sigma non-opioid intracellular receptor 1TFEB, transcription factor EBTMEM97/Sigma-2R, transmembrane protein 97.


Asunto(s)
Esclerosis Amiotrófica Lateral , Autofagia , Demencia Frontotemporal , Proteínas de la Membrana , Receptores sigma , Animales , Ratones , Esclerosis Amiotrófica Lateral/metabolismo , Autofagia/genética , Proteína C9orf72/genética , Demencia Frontotemporal/genética , Proteínas de Choque Térmico/metabolismo , Proteínas de Complejo Poro Nuclear , Factores de Transcripción/metabolismo , Proteínas de la Membrana/genética , Receptores sigma/metabolismo , Receptor Sigma-1
11.
Mol Neurobiol ; 60(4): 2200-2208, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36633805

RESUMEN

Astroglial-fibrotic scars resulted from spinal cord injury affect motor and sensory function, leading to paralysis. In particular, the fibrotic scar is a main barrier that disrupts neuronal regeneration after spinal cord injury. However, the association between astrocytes and fibrotic scar formation is not yet understood. We have previously demonstrated that the transcriptional factor Cebpd contributes to astrogliosis, which promotes glial scar formation after spinal cord injury. Herein, we show that fibrotic scar formation was decreased in the epicenter region in Cebpd-/- mice after contusive spinal cord injury and astrocytic Cebpd promoted fibroblast migration through secretion of Ptx3. Furthermore, the expression of Mmp3 was increased under recombinant protein Ptx3 treatment in fibroblasts by observing microarray data, resulting in fibroblast migration. In addition, regulation of Mmp3 occurs through the NFκB signaling pathway by using an irreversible inhibitor of IκBα phosphorylation in pretreated fibroblasts. Of note, we used the synthetic peptide RI37, which blocks fibroblast migration and decreases fibroblast Mmp3 expression in IL-1ß-treated astrocyte conditioned media. Collectively, our data suggest that fibroblast migration can be affected by astrocytic Cebpd through the Ptx3/NFκB/Mmp3 axis pathway and that the RI37 peptide may act as a therapeutic medicine to inhibit fibrotic scar formation after spinal cord injury.


Asunto(s)
Cicatriz , Traumatismos de la Médula Espinal , Ratones , Animales , Cicatriz/patología , Astrocitos/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Traumatismos de la Médula Espinal/patología , Fibrosis , Gliosis/patología , Médula Espinal/patología
12.
J Neurosci ; 31(10): 3536-49, 2011 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-21389210

RESUMEN

Painful nerve injury disrupts levels of cytoplasmic and stored Ca(2+) in sensory neurons. Since influx of Ca(2+) may occur through store-operated Ca(2+) entry (SOCE) as well as voltage- and ligand-activated pathways, we sought confirmation of SOCE in sensory neurons from adult rats and examined whether dysfunction of SOCE is a possible pathogenic mechanism. Dorsal root ganglion neurons displayed a fall in resting cytoplasmic Ca(2+) concentration when bath Ca(2+) was withdrawn, and a subsequent elevation of cytoplasmic Ca(2+) concentration (40 ± 5 nm) when Ca(2+) was reintroduced, which was amplified by store depletion with thapsigargin (1 µm), and was significantly reduced by blockers of SOCE, but was unaffected by antagonists of voltage-gated membrane Ca(2+) channels. We identified the underlying inwardly rectifying Ca(2+)-dependent I(CRAC) (Ca(2+) release activated current), as well as a large thapsigargin-sensitive inward current activated by withdrawal of bath divalent cations, representing SOCE. Molecular components of SOCE, specifically STIM1 and Orai1, were confirmed in sensory neurons at both the transcript and protein levels. Axonal injury by spinal nerve ligation (SNL) elevated SOCE and I(CRAC). However, SOCE was comparable in injured and control neurons when stores were maximally depleted by thapsigargin, and STIM1 and Orai1 levels were not altered by SNL, showing that upregulation of SOCE after SNL is driven by store depletion. Blockade of SOCE increased neuronal excitability in control and injured neurons, whereas injured neurons showed particular dependence on SOCE for maintaining levels of cytoplasmic and stored Ca(2+), which indicates a compensatory role for SOCE after injury.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Calcio/metabolismo , Ganglios Espinales/metabolismo , Hiperalgesia/metabolismo , Células Receptoras Sensoriales/metabolismo , Nervios Espinales/lesiones , Análisis de Varianza , Animales , Western Blotting , Células Cultivadas , Ganglios Espinales/citología , Hiperalgesia/fisiopatología , Inmunohistoquímica , Masculino , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Receptoras Sensoriales/citología , Nervios Espinales/metabolismo
13.
Mol Pain ; 8: 46, 2012 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-22713297

RESUMEN

BACKGROUND: The plasma membrane Ca2+-ATPase (PMCA) is the principal means by which sensory neurons expel Ca2+ and thereby regulate the concentration of cytoplasmic Ca2+ and the processes controlled by this critical second messenger. We have previously found that painful nerve injury decreases resting cytoplasmic Ca2+ levels and activity-induced cytoplasmic Ca2+ accumulation in axotomized sensory neurons. Here we examine the contribution of PMCA after nerve injury in a rat model of neuropathic pain. RESULTS: PMCA function was isolated in dissociated sensory neurons by blocking intracellular Ca2+ sequestration with thapsigargin, and cytoplasmic Ca2+ concentration was recorded with Fura-2 fluorometry. Compared to control neurons, the rate at which depolarization-induced Ca2+ transients resolved was increased in axotomized neurons after spinal nerve ligation, indicating accelerated PMCA function. Electrophysiological recordings showed that blockade of PMCA by vanadate prolonged the action potential afterhyperpolarization, and also decreased the rate at which neurons could fire repetitively. CONCLUSION: We found that PMCA function is elevated in axotomized sensory neurons, which contributes to neuronal hyperexcitability. Accelerated PMCA function in the primary sensory neuron may contribute to the generation of neuropathic pain, and thus its modulation could provide a new pathway for peripheral treatment of post-traumatic neuropathic pain.


Asunto(s)
Axotomía , Membrana Celular/enzimología , Neuralgia/enzimología , Neuralgia/patología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Células Receptoras Sensoriales/enzimología , Nervios Espinales/patología , Potenciales de Acción/efectos de los fármacos , Animales , Calcio/metabolismo , Membrana Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neuralgia/fisiopatología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/patología , Intercambiador de Sodio-Calcio/metabolismo , Nervios Espinales/efectos de los fármacos , Nervios Espinales/enzimología , Nervios Espinales/fisiopatología , Tapsigargina/farmacología
14.
Proc Natl Acad Sci U S A ; 106(21): 8725-30, 2009 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-19439665

RESUMEN

Painful axotomy decreases K(ATP) channel current (IK(ATP)) in primary afferent neurons. Because cytosolic Ca(2+) signaling is depressed in injured dorsal root ganglia (DRG) neurons, we investigated whether Ca(2+)-calmodulin (CaM)-Ca(2+)/CaM-dependent kinase II (CaMKII) regulates IK(ATP) in large DRG neurons. Immunohistochemistry identified the presence of K(ATP) channel subunits SUR1, SUR2, and Kir6.2 but not Kir6.1, and pCaMKII in neurofilament 200-positive DRG somata. Single-channel recordings from cell-attached patches revealed that basal and evoked IK(ATP) by ionomycin, a Ca(2+) ionophore, is activated by CaMKII. In axotomized neurons from rats made hyperalgesic by spinal nerve ligation (SNL), basal K(ATP) channel activity was decreased, and sensitivity to ionomycin was abolished. Basal and Ca(2+)-evoked K(ATP) channel activity correlated inversely with the degree of hyperalgesia induced by SNL in the rats from which the neurons were isolated. Inhibition of IK(ATP) by glybenclamide, a selective K(ATP) channel inhibitor, depolarized resting membrane potential (RMP) recorded in perforated whole-cell patches and enhanced neurotransmitter release measured by amperometry. The selective K(ATP) channel opener diazoxide hyperpolarized the RMP and attenuated neurotransmitter release. Axotomized neurons from rats made hyperalgesic by SNL lost sensitivity to the myristoylated form of autocamtide-2-related inhibitory peptide (AIPm), a pseudosubstrate blocker of CaMKII, whereas axotomized neurons from SNL animals that failed to develop hyperalgesia showed normal IK(ATP) inhibition by AIPm. AIPm also depolarized RMP in control neurons via K(ATP) channel inhibition. Unitary current conductance and sensitivity of K(ATP) channels to cytosolic ATP and ligands were preserved even after painful nerve injury, thus providing opportunities for selective therapeutic targeting against neuropathic pain.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calcio/metabolismo , Calmodulina/metabolismo , Hiperalgesia/metabolismo , Canales KATP/metabolismo , Neuronas Aferentes/metabolismo , Animales , Axotomía , Sistema Libre de Células , Fenómenos Electrofisiológicos , Ganglios Espinales/metabolismo , Ionomicina/farmacología , Masculino , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley
15.
Mol Neurobiol ; 58(6): 2523-2541, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33459966

RESUMEN

Sigma-1 receptors (Sig-1Rs) are endoplasmic reticulum (ER) chaperones implicated in neuropathic pain. Here we examine if the Sig-1R may relate to neuropathic pain at the level of dorsal root ganglia (DRG). We focus on the neuronal excitability of DRG in a "spare nerve injury" (SNI) model of neuropathic pain in rats and find that Sig-1Rs likely contribute to the genesis of DRG neuronal excitability by decreasing the protein level of voltage-gated Cav2.2 as a translational inhibitor of mRNA. Specifically, during SNI, Sig-1Rs translocate from ER to the nuclear envelope via a trafficking protein Sec61ß. At the nucleus, the Sig-1R interacts with cFos and binds to the promoter of 4E-BP1, leading to an upregulation of 4E-BP1 that binds and prevents eIF4E from initiating the mRNA translation for Cav2.2. Interestingly, in Sig-1R knockout HEK cells, Cav2.2 is upregulated. In accordance with those findings, we find that intra-DRG injection of Sig-1R agonist (+)pentazocine increases frequency of action potentials via regulation of voltage-gated Ca2+ channels. Conversely, intra-DRG injection of Sig-1R antagonist BD1047 attenuates neuropathic pain. Hence, we discover that the Sig-1R chaperone causes neuropathic pain indirectly as a translational inhibitor.


Asunto(s)
Genoma , Neuralgia/genética , Receptores sigma/metabolismo , Animales , Canales de Calcio Tipo N/genética , Canales de Calcio Tipo N/metabolismo , Retículo Endoplásmico/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Ganglios Espinales/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Tejido Nervioso/lesiones , Tejido Nervioso/patología , Membrana Nuclear/metabolismo , Regiones Promotoras Genéticas/genética , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas c-fos/metabolismo , Caperuzas de ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Receptores sigma/agonistas , Receptores sigma/genética , Canales de Translocación SEC/metabolismo , Transcripción Genética , Receptor Sigma-1
16.
Am J Physiol Heart Circ Physiol ; 298(6): H2201-7, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20400686

RESUMEN

We previously demonstrated that several epoxyeicosatrienoic acids (EETs) produce reductions in myocardial infarct size in rats and dogs. Since a recent study demonstrated the release of opioids in mediating the antinociceptive effect of 14,15-EET, we hypothesized that endogenous opioids may also be involved in mediating the cardioprotective effect of the EETs. To test this hypothesis, we used an in vivo rat model of infarction and a rat Langendorff model. In the infarct model, hearts were subjected to 30 min occlusion of the left coronary artery and 2 h reperfusion. Animals were treated with 11,12-EET or 14,15-EET (2.5 mg/kg) alone 15 min before occlusion or with opioid antagonists [naloxone, naltrindole, nor-binaltorphimine (nor-BNI), and d-Phe-Cys-Tyr-d-Trp-Om-Thr-Pen-Thr-NH(2) (CTOP), a nonselective, a selective delta, a selective kappa, and a selective mu receptor antagonist, respectively] 10 min before EET administration. In four separate groups, antiserum to Met- and Leu-enkephalin and dynorphin-A-(1-17) was administered 50 min before the 11,12-EET administration. Infarct size expressed as a percent of the area at risk (IS/AAR) was 63.5 + or - 1.2, 45.3 + or - 1.0, and 40.9 + or - 1.2% for control, 11,12-EET, and 14,15-EET, respectively. The protective effects of 11,12-EET were abolished by pretreatment with either naloxone (60.5 + or - 1.8%), naltrindole (60.8 + or - 1.0%), nor-BNI (62.3 + or - 2.8%), or Met-enkephalin antiserum (63.2 + or - 1.7%) but not CTOP (42.0 + or - 3.0%). In isolated heart experiments, 11,12-EET was administered to the perfusate 15 min before 20 min global ischemia followed by 45 min reperfusion in control hearts or in those pretreated with pertussis toxin (48 h). 11,12-EET increased the recovery of left ventricular developed pressure from 33 + or - 1 to 45 + or - 6% (P < 0.05) and reduced IS/AAR from 37 + or - 4 to 20 + or - 3% (P < 0.05). Both pertussis toxin and naloxone abolished these beneficial effects of 11,12-EET. Taken together, these results suggest that the major cardioprotective effects of the EETs depend on activation of a G(i/o) protein-coupled delta- and/or kappa-opioid receptor.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Analgésicos Opioides , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Ácido 8,11,14-Eicosatrienoico/uso terapéutico , Analgésicos Opioides/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Masculino , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/fisiopatología , Naloxona/farmacología , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Opioides/fisiología , Somatostatina/análogos & derivados , Somatostatina/farmacología
17.
Nat Commun ; 11(1): 5580, 2020 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-33149115

RESUMEN

In a subgroup of patients with amyotrophic lateral sclerosis (ALS)/Frontotemporal dementia (FTD), the (G4C2)-RNA repeat expansion from C9orf72 chromosome binds to the Ran-activating protein (RanGAP) at the nuclear pore, resulting in nucleocytoplasmic transport deficit and accumulation of Ran in the cytosol. Here, we found that the sigma-1 receptor (Sig-1R), a molecular chaperone, reverses the pathological effects of (G4C2)-RNA repeats in cell lines and in Drosophila. The Sig-1R colocalizes with RanGAP and nuclear pore proteins (Nups) and stabilizes the latter. Interestingly, Sig-1Rs directly bind (G4C2)-RNA repeats. Overexpression of Sig-1Rs rescues, whereas the Sig-1R knockout exacerbates, the (G4C2)-RNA repeats-induced aberrant cytoplasmic accumulation of Ran. In Drosophila, Sig-1R (but not the Sig-1R-E102Q mutant) overexpression reverses eye necrosis, climbing deficit, and firing discharge caused by (G4C2)-RNA repeats. These results on a molecular chaperone at the nuclear pore suggest that Sig-1Rs may benefit patients with C9orf72 ALS/FTD by chaperoning the nuclear pore assembly and sponging away deleterious (G4C2)-RNA repeats.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Drosophila/metabolismo , Demencia Frontotemporal/metabolismo , Neuronas Motoras/metabolismo , Poro Nuclear/metabolismo , Receptores sigma/metabolismo , Proteína de Unión al GTP ran/metabolismo , Transporte Activo de Núcleo Celular/genética , Esclerosis Amiotrófica Lateral/genética , Animales , Citosol/metabolismo , Modelos Animales de Enfermedad , Drosophila/genética , Drosophila/fisiología , Demencia Frontotemporal/genética , Técnicas de Inactivación de Genes , Células HeLa , Humanos , Poro Nuclear/genética , Unión Proteica , ARN Interferente Pequeño , Receptores sigma/genética , Proteína de Unión al GTP ran/genética , Receptor Sigma-1
18.
Mol Pain ; 5: 12, 2009 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-19284878

RESUMEN

BACKGROUND: ATP-sensitive potassium (KATP) channels in neurons regulate excitability, neurotransmitter release and mediate protection from cell-death. Furthermore, activation of KATP channels is suppressed in DRG neurons after painful-like nerve injury. NO-dependent mechanisms modulate both KATP channels and participate in the pathophysiology and pharmacology of neuropathic pain. Therefore, we investigated NO modulation of KATP channels in control and axotomized DRG neurons. RESULTS: Cell-attached and cell-free recordings of KATP currents in large DRG neurons from control rats (sham surgery, SS) revealed activation of KATP channels by NO exogenously released by the NO donor SNAP, through decreased sensitivity to [ATP]i. This NO-induced KATP channel activation was not altered in ganglia from animals that demonstrated sustained hyperalgesia-type response to nociceptive stimulation following spinal nerve ligation. However, baseline opening of KATP channels and their activation induced by metabolic inhibition was suppressed by axotomy. Failure to block the NO-mediated amplification of KATP currents with specific inhibitors of sGC and PKG indicated that the classical sGC/cGMP/PKG signaling pathway was not involved in the activation by SNAP. NO-induced activation of KATP channels remained intact in cell-free patches, was reversed by DTT, a thiol-reducing agent, and prevented by NEM, a thiol-alkylating agent. Other findings indicated that the mechanisms by which NO activates KATP channels involve direct S-nitrosylation of cysteine residues in the SUR1 subunit. Specifically, current through recombinant wild-type SUR1/Kir6.2 channels expressed in COS7 cells was activated by NO, but channels formed only from truncated isoform Kir6.2 subunits without SUR1 subunits were insensitive to NO. Further, mutagenesis of SUR1 indicated that NO-induced KATP channel activation involves interaction of NO with residues in the NBD1 of the SUR1 subunit. CONCLUSION: NO activates KATP channels in large DRG neurons via direct S-nitrosylation of cysteine residues in the SUR1 subunit. The capacity of NO to activate KATP channels via this mechanism remains intact even after spinal nerve ligation, thus providing opportunities for selective pharmacological enhancement of KATP current even after decrease of this current by painful-like nerve injury.


Asunto(s)
Activación del Canal Iónico/efectos de los fármacos , Canales KATP/metabolismo , Mamíferos/metabolismo , Óxido Nítrico/farmacología , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Células COS , Chlorocebus aethiops , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Cisteína/genética , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Masculino , Mutación/genética , Óxido Nítrico/metabolismo , Nitrosación/efectos de los fármacos , Técnicas de Placa-Clamp , Canales de Potasio de Rectificación Interna/química , Canales de Potasio de Rectificación Interna/metabolismo , Estructura Terciaria de Proteína , Ratas , Receptores de Droga/química , Receptores de Droga/metabolismo , Proteínas Recombinantes/metabolismo , S-Nitroso-N-Acetilpenicilamina/farmacología , Células Receptoras Sensoriales/enzimología , Receptores de Sulfonilureas
19.
Anesthesiology ; 111(2): 381-92, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19602958

RESUMEN

BACKGROUND: The cellular mechanisms of neuropathic pain are inadequately understood. Previous investigations have revealed disrupted Ca signaling in primary sensory neurons after injury. The authors examined the effect of injury on intracellular Ca stores of the endoplasmic reticulum, which critically regulate the Ca signal and neuronal function. METHODS: Intracellular Ca levels were measured with Fura-2 or mag-Fura-2 microfluorometry in axotomized fifth lumbar (L5) dorsal root ganglion neurons and adjacent L4 neurons isolated from hyperalgesic rats after L5 spinal nerve ligation, compared to neurons from control animals. RESULTS: Endoplasmic reticulum Ca stores released by the ryanodine-receptor agonist caffeine decreased by 46% in axotomized small neurons. This effect persisted in Ca-free bath solution, which removes the contribution of store-operated membrane Ca channels, and after blockade of the mitochondrial, sarco-endoplasmic Ca-ATPase and the plasma membrane Ca ATPase pathways. Ca released by the sarco-endoplasmic Ca-ATPase blocker thapsigargin and by the Ca-ionophore ionomycin was also diminished by 25% and 41%, respectively. In contrast to control neurons, Ca stores in axotomized neurons were not expanded by neuronal activation by K depolarization, and the proportionate rate of refilling by sarco-endoplasmic Ca-ATPase was normal. Luminal Ca concentration was also reduced by 38% in axotomized neurons in permeabilized neurons. The adjacent neurons of the L4 dorsal root ganglia showed modest and inconsistent changes after L5 spinal nerve ligation. CONCLUSIONS: Painful nerve injury leads to diminished releasable endoplasmic reticulum Ca stores and a reduced luminal Ca concentration. Depletion of Ca stores may contribute to the pathogenesis of neuropathic pain.


Asunto(s)
Axotomía , Señalización del Calcio/fisiología , Calcio/metabolismo , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología , Animales , Axones/patología , Cafeína/farmacología , Capsaicina/farmacología , Células Cultivadas , Estimulantes del Sistema Nervioso Central/farmacología , Retículo Endoplásmico/fisiología , Hiperalgesia/patología , Ionomicina/farmacología , Ligadura , Masculino , Degeneración Nerviosa/patología , Dimensión del Dolor , Ratas , Ratas Sprague-Dawley , Canal Liberador de Calcio Receptor de Rianodina/efectos de los fármacos , Nervio Ciático/lesiones , Nervios Espinales/lesiones , Tapsigargina/farmacología
20.
J Pharmacol Exp Ther ; 326(2): 614-22, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18492947

RESUMEN

Cytochrome P450 genes catalyze formation of epoxyeicosatrienoic acids (EETs) from arachidonic acid. The effects of 5,6-EET, 8,9-EET, 11,12-EET, and 14,15-EET microinjected into the ventrolateral periaqueductal gray (vlPAG) on the thermally produced tail-flick response were studied in male Sprague-Dawley rats. 14,15-EET microinjected into vlPAG (3-156 pmol) dose-dependently inhibited the tail-flick response (ED50 = 32.5 pmol). In contrast, 5,6-EET, 8,9-EET, and 11,12-EET at a dose of 156 pmol were not active when injected into the vlPAG. 14,15-EET failed to displace the radiobinding of [3H][D-Ala2,NHPe4, Gly-ol5]enkephalin (mu-opioid receptor ligand) or [3H]naltrindole (delta-opioid receptor ligand) in crude membrane fractions of rat brain. Tail-flick inhibition produced by 14,15-EET from vlPAG was blocked by intra-vlPAG pretreatment with antiserum against beta-endorphin or Met-enkephalin or the mu-opioid receptor antagonist D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP) or the delta-opioid receptor antagonist naltrindole but not with dynorphin A[1-17] antiserum or the kappa-opioid receptor antagonist nor-binaltorphimine. In addition, tail-flick inhibition produced by 14,15-EET treatment was blocked by intrathecal pretreatment with Met-enkephalin antiserum, naltrindole, or CTOP but not with beta-endorphin antiserum. It is concluded that 1) 14,15-EET itself does not have any affinity for mu- or delta-opioid receptors and 2) 14,15-EET activates beta-endorphin and Met-enkephalin, which subsequently act on mu- and delta-opioid receptors to produce antinociception.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Analgésicos/farmacología , Encefalina Metionina/metabolismo , Sustancia Gris Periacueductal/efectos de los fármacos , betaendorfina/metabolismo , Ácido 8,11,14-Eicosatrienoico/farmacología , Animales , Relación Dosis-Respuesta a Droga , Masculino , Microinyecciones , Dimensión del Dolor/efectos de los fármacos , Sustancia Gris Periacueductal/metabolismo , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA