Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Aging Neurosci ; 10: 261, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30237764

RESUMEN

Background: Early-onset Alzheimer's disease (EOAD) presents a different clinical profile than late-onset Alzheimer's disease (LOAD). Neuroimaging studies have demonstrated that patients with EOAD present more atrophy and functional disconnection than LOAD patients. However, it remains unknown whether the interhemispheric functional disconnection or its underlying structural impairment contributes to the different clinical profiles of EOAD and LOAD. Methods: According to the arbitrary cut-off age of 65, we included 22 EOAD patients, 27 LOAD patients and 38 healthy controls (further divided into 21 relatively young and 17 old controls). Participants underwent resting-state functional MRI, diffusion tensor imaging (DTI) and comprehensive neuropsychological assessments. We used voxel-mirrored homotopic connectivity (VMHC) to examine interhemispheric functional connectivity. Then, we calculated the diffusion index based on tract-based spatial statistics (TBSS). Two-sample t-tests were used to assess the interhemispheric connectivity differences between each patient group and its corresponding control group. Results: We found that the EOAD patients had lower VMHC in the hippocampus, parahippocampal gyrus (PHG), superior temporal gyrus (STG) and inferior parietal cortex (IPC) than did controls. Consistently, the EOAD patients exhibited white matter (WM) tract impairment in the posterior regions. On the other hand, the LOAD patients displayed increased VMHC and impaired WM tracts in the frontal region. Correlation analyses showed that VMHC in the IPC was related to executive function in the EOAD patients (r = -0.67, P < 0.05). Conclusion: In contrast to the LOAD patients, patients with EOAD exhibited more widely disrupted interhemispheric functional and structural connectivity, which overlapped well across brain regions. In addition, for the EOAD patients, decreased interhemispheric connectivity related to executive deficits. Our study suggested that different interhemispheric connectivity damage patterns may contribute to the distinct clinical profiles in EOAD and LOAD.

2.
Undersea Hyperb Med ; 45(3): 351-362, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30028921

RESUMEN

Nuclear factor kappa B (NF-κB) is the critical transcriptional factor in the pathogenesis of acute lung injury (ALI). NF-κB regulates the expression changes of inflammatory factors such as tumor necrosis factor alpha (TNF-α), interleukin-1ß (IL-1ß) and interleukin 6 (IL-6). In a previous study we showed that decompression sickness (DCS) caused by simulated unsafe fast buoyancy ascent escape (FBAE) could result in ALI, which was characterized by expression changes of inflammatory factors in rat lung tissue. The purpose of the present work was to study the roles of NF-κB and TNF-α in the process of DCS-induced rat lung injury caused by simulated unsafe FBAE. The research methods aimed to detect the rat lung tissue messenger ribonucleic acid (mRNA) and protein level variations of NF-κB, inhibitory ×B (I×B), TNF-α, IL-1ß, IL-6, IL-10 and IL-13 by using pretreatment of the NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC) and TNF-α antibody (Ab). Our experimental results demonstrated that PDTC could improve the survival rate of the rats with DCS caused by unsafe FBAE more effectively than TNF-α Ab. However, the inhibition of TNF-α Ab on the nuclear translocated protein expression of NF-κB was more effective than PDTC. Both PDTC and TNF-α Ab can abrogate the increment of the rat lung tissue mRNA levels of TNF-α, IL-1ß, IL-6 and protein levels of NF-κB, TNF-α, IL-1ß effectively and increase the rat lung tissue content of I×B significantly. In conclusion, TNF-α-mediated NF-κB signaling may be one of the critical signaling pathways in the pathogenesis of DCS-induced rat lung injury caused by simulated unsafe FBAE. PDTC may ameliorate this type of injury partly through inhibiting the NF-κB pathway.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Antioxidantes/farmacología , Enfermedad de Descompresión/complicaciones , Interleucinas/metabolismo , FN-kappa B/metabolismo , Pirrolidinas/farmacología , Tiocarbamatos/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/patología , Animales , Interleucina-10/metabolismo , Interleucina-13/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Pulmón/metabolismo , Pulmón/patología , Masculino , FN-kappa B/antagonistas & inhibidores , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Tasa de Supervivencia , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
4.
Cell Res ; 26(1): 83-102, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26691752

RESUMEN

Sensory neurons are distinguished by distinct signaling networks and receptive characteristics. Thus, sensory neuron types can be defined by linking transcriptome-based neuron typing with the sensory phenotypes. Here we classify somatosensory neurons of the mouse dorsal root ganglion (DRG) by high-coverage single-cell RNA-sequencing (10 950 ± 1 218 genes per neuron) and neuron size-based hierarchical clustering. Moreover, single DRG neurons responding to cutaneous stimuli are recorded using an in vivo whole-cell patch clamp technique and classified by neuron-type genetic markers. Small diameter DRG neurons are classified into one type of low-threshold mechanoreceptor and five types of mechanoheat nociceptors (MHNs). Each of the MHN types is further categorized into two subtypes. Large DRG neurons are categorized into four types, including neurexophilin 1-expressing MHNs and mechanical nociceptors (MNs) expressing BAI1-associated protein 2-like 1 (Baiap2l1). Mechanoreceptors expressing trafficking protein particle complex 3-like and Baiap2l1-marked MNs are subdivided into two subtypes each. These results provide a new system for cataloging somatosensory neurons and their transcriptome databases.


Asunto(s)
Ganglios Espinales/citología , Redes Reguladoras de Genes , Células Receptoras Sensoriales/citología , Transcriptoma , Animales , Secuencia de Bases , Células Cultivadas , Ganglios Espinales/metabolismo , Masculino , Mecanorreceptores/citología , Mecanorreceptores/metabolismo , Ratones , Ratones Endogámicos C57BL , Familia de Multigenes , Nociceptores/citología , Nociceptores/metabolismo , Técnicas de Placa-Clamp , Células Receptoras Sensoriales/metabolismo , Análisis de Secuencia de ARN
5.
Mol Brain ; 8: 53, 2015 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-26382033

RESUMEN

BACKGROUND: Radial glial cells (RGCs), the instructive scaffolds for neuronal migration, are well characterized by their unique morphology and polarization; these cells extend elongated basal processes to the pial basement membrane (BM) and parallel to one another. However, little is known about the mechanisms that underlie the developmental regulation and maintenance of this unique morphology. RESULTS: Here, by crossing Fstl1 (fl/fl) mice with an EIIa-Cre line, we identified a new role for the secreted glycoprotein Follistatin like-1 (FSTL1). The ablation of Fstl1 in both of its cortical expression domains, the ventricular zone (VZ) and the pia mater, resulted in RGC morphologic disruption; basal processes were not parallel to each other, and endfeet exhibited greater density and branching. However, Fstl1 deletion in only the VZ in the Emx1 (IREScre); Fstl1 (fl/fl) line did not affect RGC morphology, indicating that FSTL1 derived from the pia mater might be more important for RGC morphology. In addition, upper-layer projection neurons, not deeper-layer projection neurons, failed to reach their appropriate positions. We also found that BMP, AKT/PKB, Cdc42, GSK3ß, integrin and reelin signals, which have previously been reported to regulate RGC development, were unchanged, indicating that Fstl1 may function through a unique mechanism. CONCLUSIONS: In the present study, we identified a new role for FSTL1 in the development of radial glial scaffolds and the neuronal migration of upper-layer projection neurons. Our findings will improve understanding of the regulation of RGC development and neuronal migration.


Asunto(s)
Proteínas Relacionadas con la Folistatina/metabolismo , Neuroglía/metabolismo , Animales , Membrana Basal/metabolismo , Polaridad Celular , Forma de la Célula , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/metabolismo , Proteínas Relacionadas con la Folistatina/deficiencia , Eliminación de Gen , Ratones , Neuroglía/citología , Piamadre/metabolismo , Proteína Reelina
6.
Undersea Hyperb Med ; 42(1): 15-22, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26094300

RESUMEN

Fast buoyancy ascent escape is one of the major naval submarine escape maneuvers. Decompression sickness (DCS) is the major bottleneck to increase the depth of fast buoyancy ascent escape. Rapid decompression induces the release of inflammatory mediators and results in tissue inflammation cascades and a protective anti-inflammatory response. In our previous study, we found that DCS caused by simulated fast buoyancy ascent escape could induce acute lung injury (ALI) and the expression changes of the proinflammatory cytokines: tumor necrosis factor alpha (TNF-α), interleukin (IL)-1ß and IL-6 in rat lung tissue. In order to study the expression change characteristics of TNF-α, IL-1ß, IL-6, IL-10 and IL-13 in the rat lung of DCS caused by simulated fast buoyancy ascent escape, we detected the rat lung mRNA and protein levels of TNF-α, IL-1ß, IL-6, IL-10 and IL-13 at 0.5 hour after DCS caused by simulated fast buoyancy ascent escape (fast escape group), compared with the normal control group (control group) and diving DCS (decompression group). We observed that DCS caused by simulated fast buoyancy ascent escape could increase the mRNA levels of TNF-α, IL-1ß, IL-6, IL-10, and the protein levels of TNF-α, IL-10 in rat lung tissue. At the same time, we found that the protein level of IL-13 was also downregulated in rat lung tissue. TNF-α, IL-10 and IL-13 may be involved in the process of the rat lung injury of DCS caused by simulated fast buoyancy ascent escape. In conclusion, the expression changes of inflammatory factors in the rat lung of DCS caused by simulated fast buoyancy ascent escape were probably different from that in the rat lung of diving DCS, which indicated that the pathological mechanism of DCS caused by simulated fast buoyancy ascent escape might be different from that of diving DCS.


Asunto(s)
Enfermedad de Descompresión/metabolismo , Interleucinas/metabolismo , Pulmón/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Animales , Enfermedad de Descompresión/etiología , Enfermedad de Descompresión/mortalidad , Enfermedad de Descompresión/patología , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-13/genética , Interleucina-13/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucinas/genética , Pulmón/patología , Masculino , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Medicina Submarina , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética
7.
Undersea Hyperb Med ; 42(1): 23-31, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26094301

RESUMEN

Fast buoyancy ascent escape is the general submarine escape manner adopted by the majority of naval forces all over the world. However, if hyperbaric exposure time exceeds the time limit, fast buoyancy ascent escape has a high risk to result in decompression sickness (DCS). Tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß) and IL-6 have been all implicated in the process of inflammation associated with acute lung injury (ALI). Our work demonstrated that DCS caused by simulated fast buoyancy ascent escape could induce ALß in the rat model. The purpose of the present work was to study the expression changes of TNF-α, IL-1ß and IL-6 in the rat lung affected by DCS caused by simulated fast buoyancy ascent escape. The lung tissue mRNA levels of TNF-α, Il-1ß and Il-6 were significantly increased at 0.5 hour after DCS caused by simulated fast buoyancy ascent escape. The lung contents of TNF-α, IL-1ß and IL-6 were at an expression peak at 0.5 hour, although showing no statistical difference when compared with the normal control group. In conclusion, the rat lung expression variations of TNF-α, IL-1ß and IL-6 are the most obvious at 0.5 hour within 24 hours after the lung injury by DCS caused by simulated fast buoyancy ascent escape.


Asunto(s)
Enfermedad de Descompresión/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Pulmón/metabolismo , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Animales , Enfermedad de Descompresión/patología , Interleucina-1beta/genética , Interleucina-6/genética , Pulmón/patología , Masculino , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Medicina Submarina , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética
8.
J Mol Cell Biol ; 7(5): 466-75, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25782913

RESUMEN

Fibroblast growth factor (FGF) 7, a member of FGF family, is initially found to be secreted from mesenchymal cells to repair epithelial tissues. However, its functions in the nervous system are largely unknown. The present study showed that FGF7 was a neuromodulator localized in the large dense-core vesicles (LDCVs) in nociceptive neurons. FGF7 was mainly expressed in small-diameter neurons of the dorsal root ganglion and could be transported to the dorsal spinal cord. Interestingly, FGF7 was mostly stored in LDCVs that did not contain neuropeptide substance P. Electrophysiological recordings in the spinal cord slice showed that buffer-applied FGF7 increased the amplitude of excitatory post-synaptic current evoked by stimulating the sensory afferent fibers. Behavior tests showed that intrathecally applied FGF7 potentiated the formalin-induced acute nociceptive response. Moreover, both acute and inflammatory nociceptive responses were significantly reduced in Fgf7-deficient mice. These results suggest that FGF7 exerts an excitatory modulation of nociceptive afferent transmission.


Asunto(s)
Factor 7 de Crecimiento de Fibroblastos/metabolismo , Ganglios Espinales/metabolismo , Nociceptores/metabolismo , Dolor/metabolismo , Animales , Ratones
9.
Cell Res ; 25(3): 318-34, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25633594

RESUMEN

Na⁺, K⁺-ATPase (NKA) is required to generate the resting membrane potential in neurons. Nociceptive afferent neurons express not only the α and ß subunits of NKA but also the γ subunit FXYD2. However, the neural function of FXYD2 is unknown. The present study shows that FXYD2 in nociceptive neurons is necessary for maintaining the mechanical allodynia induced by peripheral inflammation. FXYD2 interacted with α1NKA and negatively regulated the NKA activity, depolarizing the membrane potential of nociceptive neurons. Mechanical allodynia initiated in FXYD2-deficient mice was abolished 4 days after inflammation, whereas it persisted for at least 3 weeks in wild-type mice. Importantly, the FXYD2/α1NKA interaction gradually increased after inflammation and peaked on day 4 post inflammation, resulting in reduction of NKA activity, depolarization of neuron membrane and facilitation of excitatory afferent neurotransmission. Thus, the increased FXYD2 activity may be a fundamental mechanism underlying the persistent hypersensitivity to pain induced by inflammation.


Asunto(s)
Hiperalgesia/fisiopatología , Inflamación/fisiopatología , Nociceptores/fisiología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Masculino , Potenciales de la Membrana/fisiología , Ratones , Ratones Noqueados , Nociceptores/metabolismo , Dolor/fisiopatología , ARN Mensajero/genética , ATPasa Intercambiadora de Sodio-Potasio/genética , Transmisión Sináptica/fisiología
10.
Magn Reson Imaging ; 33(3): 270-5, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25461305

RESUMEN

PURPOSE: The study was aimed to determine and optimize the parameters for the MR fast imaging employing steady-state acquisition (FIESTA) sequence, which was to obtain an acceptable image to evaluate the value of the movement of the temporomandibular joint (TMJ). METHODS: In this investigation, 20 volunteers were examined to determine and optimize the parameters of the FIESTA sequence. Then, 160 TMJs from 80 patients with temporomandibular joint disorders (TMD) of clinical suspicion were consecutively performed by both static MRI and dynamic FIESTA MRI on the oblique sagittal position. The FIESTA MR images of TMJs were obtained from a slow, consecutive, free and open-closed movement. Based on the cycles of TMJ movements during the process of FIESTA MRI (90seconds), we classified all TMJs into 2 groups: cycles of open-closed mouths less than or equal to 3 (group 1) and more than 3 (group 2). Each image was marked level 1-3 by its quality. Meanwhile, the location of articular disc, mandibular condyle, motive artifact, "jumping sign" and the joint effusion in each TMJ were assessed respectively. RESULTS: By dynamic FIESTA MRI among 160 TMJs, 92 TMJs (57.50%) were in group 1, and 68 TMJs were (42.50%) in group 2. There were statistically significant differences between group 1 and group 2(p<0.05). It was shown that the number of "level 3" in group 1 was greater than group 2, and the number of "level 1" in group 1 was less than group 2. The phenomenon of motion artifact and "jumping sign" were much significantly higher in group 2 than those in group 1 (p<0.01). Furthermore, in all of the "jumping sign" cases, the phenomenon of "jumping sign" was significantly higher in group ADDwR than in group ADDw/oR (p<0.01). There was a statistically significant correlation between disc-condyle complex in "jumping sign" phenomenon and group ADDwR (r=0.621, p<0.05). The data with the false matching rate of 31.52% showed that the maximum motion range on the dynamic imaging was greater than the static imaging. Among 160 TMJs, joint effusions of 37 TMJs (23.13%) were identified by dynamic FIESTA-MRI. Among 79 TMJs with ADDw/oR(anterior disc displacement without reduction), 42 sides were operated with Maxillofacial arthroscopy surgery. The surgical result was in agreement with the MR result. CONCLUSION: Most TMJs images with a slow free open-close movement (cycles≦3) could be successfully obtained by the dynamic FIESTA MRI. The FIESTA MRI might be considered as an additional method to evaluate the movement of the articular disk and the mandibular condyle.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Imagen por Resonancia Magnética/métodos , Trastornos de la Articulación Temporomandibular/patología , Articulación Temporomandibular/patología , Adulto , Femenino , Humanos , Masculino , Mandíbula/patología , Cóndilo Mandibular/patología , Trastornos de la Articulación Temporomandibular/diagnóstico , Adulto Joven
12.
Undersea Hyperb Med ; 40(4): 313-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23957201

RESUMEN

Prolonged exposure to hyperbaric oxygen can cause pulmonary and nerve system toxicity. Although hyperbaric oxygen treatment has been used for a broad spectrum of ailments, the mechanisms of prolonged hyperbaric oxygen-induced lung injury are not fully understood. The purpose of the present work was to investigate the roles of ERK, p38, and caspase-3 in rat lung tissue exposed to hyperbaric oxygen at 2.3 atmospheres absolute (atm abs) for two, six and 10 hours. The results showed that the ERK and p38 were phosphorylated at two hours and reached a peak at six hours into exposure to hyperbaric oxygen. While the phosphorylation level of ERK decreased, p38 remained at a high level of activation at 10 hours. The activation of ERK and p38 was down-regulated when rats were exposed to normoxic hyperbaric nitrogen for 10 hours. However, caspase-3 was activated at six hours and 10 hours into exposure to hyperbaric oxygen. These results demonstrated different changes of activation of ERK and p38 during lung injury induced by prolonged exposure to hyperbaric oxygen. The time course changes of activated caspase-3 were similar to the process of p38 activation upon exposure to hyperbaric oxygen. In this way, activation of p38, not ERK, seems to be a mechanism associated with prolonged hyperbaric oxygen-induced lung injury.


Asunto(s)
Caspasa 3/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Oxigenoterapia Hiperbárica/efectos adversos , Lesión Pulmonar/enzimología , Oxígeno/toxicidad , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Análisis de Varianza , Animales , Apoptosis , Activación Enzimática , Pulmón/patología , Lesión Pulmonar/etiología , Lesión Pulmonar/patología , Masculino , Nitrógeno , Fosforilación , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
13.
Neuroimmunomodulation ; 19(4): 235-40, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22441540

RESUMEN

OBJECTIVE: It has been shown that interferon-α (IFN-α) is synthesized and secreted by macrophages, monocytes, T lymphocytes, glial cells and neurons. IFN-α has been shown to have an antinociceptive effect at the supraspinal level in the nerve system. However, it is unclear how IFN-α is involved in the modulation of nociceptive transmission in the spinal cord. METHODS: In the present study, IFN-α was used to test the potential functional roles in the nociceptive transmission. Using the whole-cell patch-clamp technique, we examined the effects of IFN-α on substantia gelatinosa (SG) neurons in the dorsal root-attached spinal cord slice prepared from adult rats. RESULTS: We found that IFN-α increased glutamatergic excitatory postsynaptic currents evoked by the stimulation of either Aδ or C afferent fibers. Further studies showed that IFN-α treatment dose-dependently increased spontaneous excitatory postsynaptic current frequency in SG neurons, while not affecting the amplitude. Moreover, intrathecal antibody of IFN-α could reduce nociceptive responses in formalin test. CONCLUSIONS: These results suggest that IFN-α presynaptically facilitates the excitatory synaptic transmission to SG neurons. The nociceptive responses could be inhibited by IFN-α antibody in the formalin test. Thus, IFN-α enhances the nociceptive transmission, which contributes to the behavioral nociceptive responses.


Asunto(s)
Potenciales Postsinápticos Excitadores/fisiología , Interferón-alfa/fisiología , Nocicepción/fisiología , Sustancia Gelatinosa/fisiología , Vías Aferentes/fisiología , Animales , Ácido Glutámico/fisiología , Masculino , Fibras Nerviosas Mielínicas/fisiología , Fibras Nerviosas Amielínicas/fisiología , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley
14.
Brain ; 135(Pt 2): 391-403, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22275428

RESUMEN

Emerging evidence suggests that the suppressive modulators released from nociceptive afferent neurons contribute to pain regulation. However, the suppressive modulators expressed in small-diameter neurons of the dorsal root ganglion remain to be further identified. The present study shows that the activin C expressed in small dorsal root ganglion neurons is required for suppressing inflammation-induced nociceptive responses. The expression of activin C in small dorsal root ganglion neurons of rats was markedly downregulated during the early days of peripheral inflammation induced by intraplantar injection of the complete Freund's adjuvant. Intrathecal treatment with the small interfering RNA targeting activin ßC or the antibodies against activin C could enhance the formalin-induced nociceptive responses, and impair the recovery from the complete Freund's adjuvant-induced thermal hyperalgesia. Intrathecally applied activin C could reduce nociceptive responses induced by formalin or complete Freund's adjuvant. Moreover, activin C was found to inhibit the inflammation-induced phosphorylation of extracellular signal-regulated kinase in the dorsal root ganglia and the dorsal spinal cord. Thus, activin C functions as an endogenous suppressor of inflammatory nociceptive transmission and may have a therapeutic potential for treatment of inflammatory pain.


Asunto(s)
Activinas/metabolismo , Ganglios Espinales/metabolismo , Hiperalgesia/metabolismo , Inflamación/metabolismo , Subunidades beta de Inhibinas/metabolismo , Nociceptores/metabolismo , Animales , Conducta Animal , Recuento de Células , Dolor Crónico/inducido químicamente , Dolor Crónico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hiperalgesia/inducido químicamente , Inflamación/inducido químicamente , Fosforilación , Ratas , Ratas Sprague-Dawley
15.
Undersea Hyperb Med ; 38(2): 149-53, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21510275

RESUMEN

Prolonged hyperbaric oxygen exposure causes pulmonary and nervous system toxicity, although hyperbaric oxygen treatment has been used to treat a broad spectrum of ailments. In the current study, animals have been exposed to 100% oxygen at a pressure of 2.3 atmospheres absolute (ATA) for two, six and 10 hours or 0.23 MPa normoxic hyperbaric nitrogen (N2-O2 mixture, oxygen partial pressure = 21 kPa) for 10 hours. Then we investigated whether ERK1/2 and p38 had been activated in the dorsal root ganglion (DRG) by hyperbaric conditions. Using Western blot analysis, we found that the phosphorylation levels of ERK1/2 (phospho-ERK1/2) increased significantly (p < 0.05, n = 3 for each group) in the six-hour treatment of 100% oxygen at a pressure of 2.3 ATA. The phosphorylation levels of p38 (phospho-p38) increased significantly (p < 0.05, n = 3 for each group) in the 10-hour treatment of 100% oxygen at a pressure of 2.3 ATA--which was consistent with time course changes of an apoptosis marker, cleavage caspase-3--while the phospho-p38 decreased in the 10 hours of N2-O2 mixture. These results demonstrate that the ERK1/2 and p38 have been differently activated in the DRG by prolonged hyperbaric oxygen exposure.


Asunto(s)
Ganglios Espinales/enzimología , Oxigenoterapia Hiperbárica , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Apoptosis , Biomarcadores/metabolismo , Western Blotting , Caspasa 3/metabolismo , Activación Enzimática/fisiología , Ganglios Espinales/efectos de los fármacos , Oxigenoterapia Hiperbárica/efectos adversos , Masculino , Oxígeno/toxicidad , Fosforilación/fisiología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
16.
Neuron ; 69(5): 974-87, 2011 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-21382556

RESUMEN

Excitatory synaptic transmission is modulated by inhibitory neurotransmitters and neuromodulators. We found that the synaptic transmission of somatic sensory afferents can be rapidly regulated by a presynaptically secreted protein, follistatin-like 1 (FSTL1), which serves as a direct activator of Na(+),K(+)-ATPase (NKA). The FSTL1 protein is highly expressed in small-diameter neurons of the dorsal root ganglion (DRG). It is transported to axon terminals via small translucent vesicles and secreted in both spontaneous and depolarization-induced manners. Biochemical assays showed that FSTL1 binds to the α1 subunit of NKA and elevates NKA activity. Extracellular FSTL1 induced membrane hyperpolarization in cultured cells and inhibited afferent synaptic transmission in spinal cord slices by activating NKA. Genetic deletion of FSTL1 in small DRG neurons of mice resulted in enhanced afferent synaptic transmission and sensory hypersensitivity, which could be reduced by intrathecally applied FSTL1 protein. Thus, FSTL1-dependent activation of NKA regulates the threshold of somatic sensation.


Asunto(s)
Proteínas Relacionadas con la Folistatina/metabolismo , Células Receptoras Sensoriales/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Transmisión Sináptica/fisiología , Análisis de Varianza , Animales , Northern Blotting , Western Blotting , Células COS , Células Cultivadas , Chlorocebus aethiops , Proteínas Relacionadas con la Folistatina/genética , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Inmunohistoquímica , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp , Terminales Presinápticos/metabolismo , Ratas
18.
Mol Pain ; 6: 92, 2010 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-21143988

RESUMEN

BACKGROUND: It has been shown that estrogen is synthesized in the spinal dorsal horn and plays a role in modulating pain transmission. One of the estrogen receptor (ER) subtypes, estrogen receptor alpha (ERα), is expressed in the spinal laminae I-V, including substantia gelatinosa (SG, lamina II). However, it is unclear how ERs are involved in the modulation of nociceptive transmission. RESULTS: In the present study, a selective ERα antagonist, methyl-piperidino-pyrazole (MPP), was used to test the potential functional roles of spinal ERα in the nociceptive transmission. Using the whole-cell patch-clamp technique, we examined the effects of MPP on SG neurons in the dorsal root-attached spinal cord slice prepared from adult rats. We found that MPP increased glutamatergic excitatory postsynaptic currents (EPSCs) evoked by the stimulation of either Aδ- or C-afferent fibers. Further studies showed that MPP treatment dose-dependently increased spontaneous EPSCs frequency in SG neurons, while not affecting the amplitude. In addition, the PKC was involved in the MPP-induced enhancement of synaptic transmission. CONCLUSIONS: These results suggest that the selective ERα antagonist MPP pre-synaptically facilitates the excitatory synaptic transmission to SG neurons. The nociceptive transmission evoked by Aδ- and C-fiber stimulation could be potentiated by blocking ERα in the spinal neurons. Thus, the spinal estrogen may negatively regulate the nociceptive transmission through the activation of ERα.


Asunto(s)
Receptor alfa de Estrógeno/antagonistas & inhibidores , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Nociceptores/fisiología , Sustancia Gelatinosa/citología , Animales , Potenciales Postsinápticos Excitadores/fisiología , Masculino , Fibras Nerviosas Mielínicas , Fibras Nerviosas Amielínicas , Nociceptores/efectos de los fármacos , Técnicas de Placa-Clamp , Piperidinas/farmacología , Pirazoles/farmacología , Ratas , Médula Espinal/fisiología , Transmisión Sináptica/efectos de los fármacos
19.
J Neurosci ; 30(32): 10927-38, 2010 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-20702721

RESUMEN

B-type natriuretic peptide (BNP) has been known to be secreted from cardiac myocytes and activate its receptor, natriuretic peptide receptor-A (NPR-A), to reduce ventricular fibrosis. However, the function of BNP/NPR-A pathway in the somatic sensory system has been unknown. In the present study, we report a novel function of BNP in pain modulation. Using microarray and immunoblot analyses, we found that BNP and NPR-A were expressed in the dorsal root ganglion (DRG) of rats and upregulated after intraplantar injection of complete Freund's adjuvant (CFA). Immunohistochemistry showed that BNP was expressed in calcitonin gene-related peptide (CGRP)-containing small neurons and IB4 (isolectin B4)-positive neurons, whereas NPR-A was present in CGRP-containing neurons. Application of BNP reduced the firing frequency of small DRG neurons in the presence of glutamate through opening large-conductance Ca2+-activated K+ channels (BKCa channels). Furthermore, intrathecal injection of BNP yielded inhibitory effects on formalin-induced flinching behavior and CFA-induced thermal hyperalgesia in rats. Blockade of BNP signaling by BNP antibodies or cGMP-dependent protein kinase (PKG) inhibitor KT5823 [(9S,10R,12R)-2,3,9,10,11,12-hexahydro-10-methoxy-2,9-dimethyl-1-oxo-9,12-epoxy-1H-diindolo[1,2,3-fg:3',2',1'-kl]pyrrolo[3,4-i][1,6]benzodiazocine-10-carboxylic acid methyl ester] impaired the recovery from CFA-induced thermal hyperalgesia. Thus, BNP negatively regulates nociceptive transmission through presynaptic receptor NPR-A, and activation of the BNP/NPR-A/PKG/BKCa channel pathway in nociceptive afferent neurons could be a potential strategy for inflammatory pain therapy.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Péptido Natriurético Encefálico/metabolismo , Dolor/metabolismo , Células Receptoras Sensoriales/metabolismo , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Anticuerpos/farmacología , Anticuerpos/uso terapéutico , Fenómenos Biofísicos/efectos de los fármacos , Fenómenos Biofísicos/fisiología , Péptido Relacionado con Gen de Calcitonina/metabolismo , Carbazoles/farmacología , Carbazoles/uso terapéutico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Adyuvante de Freund , Ganglios Espinales/patología , Regulación de la Expresión Génica/efectos de los fármacos , Ácido Glutámico/farmacología , Hiperalgesia/complicaciones , Hiperalgesia/tratamiento farmacológico , Inflamación/inducido químicamente , Inflamación/complicaciones , Lectinas/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Péptido Natriurético Encefálico/inmunología , Dolor/tratamiento farmacológico , Dolor/etiología , Dimensión del Dolor/métodos , Técnicas de Placa-Clamp/métodos , Péptidos/farmacología , Ratas , Ratas Sprague-Dawley , Receptores del Factor Natriurético Atrial/metabolismo , Células Receptoras Sensoriales/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
20.
Proc Natl Acad Sci U S A ; 107(29): 13117-22, 2010 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-20615975

RESUMEN

Morphine-induced analgesia and antinociceptive tolerance are known to be modulated by interaction between delta-opioid receptors (DORs) and mu-opioid receptors (MORs) in the pain pathway. However, evidence for expression of DORs in nociceptive small-diameter neurons in dorsal root ganglia (DRG) and for coexistence of DORs with MORs and neuropeptides has recently been challenged. We now report, using in situ hybridization, single-cell PCR, and immunostaining, that DORs are widely expressed not only in large DRG neurons but in small ones and coexist with MORs in peptidergic small DRG neurons, with protachykinin-dependent localization in large dense-core vesicles. Importantly, both DOR and MOR agonists reduce depolarization-induced Ca(2+) currents in single small DRG neurons and inhibit afferent C-fiber synaptic transmission in the dorsal spinal cord. Thus, coexistence of DORs and MORs in small DRG neurons is a basis for direct interaction of opioid receptors in modulation of nociceptive afferent transmission and opioid analgesia.


Asunto(s)
Nociceptores/metabolismo , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Animales , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Nociceptores/citología , Nociceptores/efectos de los fármacos , Péptidos/metabolismo , Precursores de Proteínas/farmacología , Transporte de Proteínas/efectos de los fármacos , Ratas , Receptores Opioides delta/genética , Receptores Opioides mu/genética , Vesículas Secretoras/efectos de los fármacos , Vesículas Secretoras/metabolismo , Taquicininas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...