Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Anesthesiology ; 133(3): 628-644, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32568844

RESUMEN

BACKGROUND: Increased descending pain facilitation accounts for opioid-induced hyperalgesia, but the underlying mechanisms remain elusive. Given the role of µ-opioid receptors in opioid-induced hyperalgesia in animals, the authors hypothesized that the dorsal reticular nucleus, a medullary pain facilitatory area, is involved in opioid-induced hyperalgesia through altered µ-opioid receptor signaling. METHODS: The authors used male Wistar rats (n = 5 to 8 per group), chronically infused with morphine, to evaluate in the dorsal reticular nucleus the expressions of the µ-opioid receptor and phosphorylated cAMP response element-binding, a downstream marker of excitatory µ-opioid receptor signaling. The authors used pharmacologic and gene-mediated approaches. Nociceptive behaviors were evaluated by the von Frey and hot-plates tests. RESULTS: Lidocaine fully reversed mechanical and thermal hypersensitivity induced by chronic morphine. Morphine-infusion increased µ-opioid receptor, without concomitant messenger RNA changes, and phosphorylated cAMP response element-binding levels at the dorsal reticular nucleus. µ-opioid receptor knockdown in morphine-infused animals attenuated the decrease of mechanical thresholds and heat-evoked withdrawal latencies compared with the control vector (von Frey [mean ± SD]: -17 ± 8% vs. -40 ± 9.0%; P < 0.001; hot-plate: -10 ± 5% vs. -32 ± 10%; P = 0.001). µ-opioid receptor knockdown in control animals induced the opposite (von Frey: -31 ± 8% vs. -17 ± 8%; P = 0.053; hotplate: -24 ± 6% vs. -3 ± 10%; P = 0.001). The µ-opioid receptor agonist (D-ALA2,N-ME-PHE4,GLY5-OL)-enkephalin acetate (DAMGO) decreased mechanical thresholds and did not affect heat-evoked withdrawal latencies in morphine-infused animals. In control animals, DAMGO increased both mechanical thresholds and heat-evoked withdrawal latencies. Ultra-low-dose naloxone, which prevents the excitatory signaling of the µ-opioid receptor, administered alone, attenuated mechanical and thermal hypersensitivities, and coadministered with DAMGO, restored DAMGO analgesic effects and decreased phosphorylated cAMP response element-binding levels. CONCLUSIONS: Chronic morphine shifted µ-opioid receptor signaling from inhibitory to excitatory at the dorsal reticular nucleus, likely enhancing descending facilitation during opioid-induced hyperalgesia in the rat.


Asunto(s)
Analgésicos Opioides/farmacología , Hiperalgesia/inducido químicamente , Bulbo Raquídeo/efectos de los fármacos , Morfina/farmacología , Receptores Opioides mu/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos
2.
Hippocampus ; 28(12): 900-912, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30098276

RESUMEN

Diabetes increases adrenal steroids in humans and animal models, but potential interactions with psychological stress remain poorly understood. Diabetic rodents exhibit anxiety and reductions in hippocampal brain-derived neurotrophic factor (BDNF) expression, and these studies investigated whether loss of BDNF-driven hippocampal activity promotes anxiety and disinhibits the HPA axis. Mice with genetic obesity and diabetes (db/db) received intrahippocampal injections of lentivirus for BDNF overexpression (db/db-BDNFOE), and Wt mice received lentiviral constructs for BDNF knockdown (Wt-BDNFKD). Behavioral anxiety and glucocorticoid responses to acute restraint were compared with mice that received a fluorescent reporter (Wt-GFP, db/db-GFP). These experiments revealed that changes in hippocampal BDNF were necessary and sufficient for behavioral anxiety and HPA axis disinhibition. To examine patterns of stress-induced regional activity, we used algorithmic detection of cFos and automated segmentation of forebrain regions to generate maps of functional covariance, which were subsequently aligned with anatomical connectivity weights from the Brain Architecture Management database. db/db-GFP mice exhibited reduced activation of the hippocampal ventral subiculum (vSub) and anterior bed nucleus of stria terminalis (aBNST), and increases in the paraventricular hypothalamus (PVH), relative to Wt-GFP. BDNFKD recapitulated this pattern in Wt mice, and BDNFOE normalized activation of the vSub > aBNST > PVH pathway in db/db mice. Analysis of forebrain activation revealed largely overlapping patterns of network disruption in db/db-GFP and Wt-BDNFKD mice, implicating BDNF-driven hippocampal activity as a determinant of stress vulnerability in both the intact and diabetic brain.


Asunto(s)
Mapeo Encefálico , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Estrés Psicológico/metabolismo , Análisis de Varianza , Animales , Ansiedad/metabolismo , Conducta Animal , Corticosterona/sangre , Retroalimentación Fisiológica , Genes Inmediatos-Precoces , Genes fos , Hipocampo/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Núcleos Septales/fisiopatología
3.
Anesthesiology ; 128(5): 967-983, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29334500

RESUMEN

BACKGROUND: The current study used recombinant herpes simplex virus type I to increase expression of µ-opiate receptors and the opioid ligand preproenkephalin in peripheral nerve fibers in a mouse model of neuropathic pain. It was predicted that viral vector delivery of a combination of genes encoding the µ-opioid receptor and preproenkephalin would attenuate neuropathic pain and enhance opioid analgesia. The behavioral effects would be paralleled by changes in response properties of primary afferent neurons. METHODS: Recombinant herpes simplex virus type 1 containing cDNA sequences of the µ-opioid receptor, human preproenkephalin, a combination, or Escherichia coli lacZ gene marker (as a control) was used to investigate the role of peripheral opioids in neuropathic pain behaviors. RESULTS: Inoculation with the µ-opioid receptor viral vector (n = 13) reversed mechanical allodynia and thermal hyperalgesia and produced leftward shifts in loperamide (ED50 = 0.6 ± 0.2 mg/kg vs. ED50 = 0.9 ± 0.2 mg/kg for control group, n = 8, means ± SD) and morphine dose-response curves (ED50 = 0.3 ± 0.5 mg/kg vs. ED50 = 1.1 ± 0.1 mg/kg for control group). In µ-opioid receptor viral vector inoculated C-fibers, heat-evoked responses (n = 12) and ongoing spontaneous activity (n = 18) were decreased after morphine application. Inoculation with both µ-opioid receptor and preproenkephalin viral vectors did not alter mechanical and thermal responses. CONCLUSIONS: Increasing primary afferent expression of opioid receptors can decrease neuropathic pain-associated behaviors and increase systemic opioid analgesia through inhibition of peripheral afferent fiber activity.


Asunto(s)
Analgésicos Opioides/farmacología , Encefalinas/fisiología , Neuralgia/prevención & control , Neuronas Aferentes/fisiología , Receptores Opioides mu/fisiología , Analgesia , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Herpesvirus Humano 1/genética , Masculino , Ratones , Morfina/farmacología , Proteínas Proto-Oncogénicas c-fos/análisis , Receptores Opioides mu/análisis
4.
Med Ref Serv Q ; 37(2): 153-167, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29558324

RESUMEN

The goal of this content analysis was to identify commonly used content and design features of academic health sciences library home pages. After developing a checklist, data were collected from 135 academic health sciences library home pages. The core components of these library home pages included a contact phone number, a contact email address, an Ask-a-Librarian feature, the physical address listed, a feedback/suggestions link, subject guides, a discovery tool or database-specific search box, multimedia, social media, a site search option, a responsive web design, and a copyright year or update date.


Asunto(s)
Almacenamiento y Recuperación de la Información/métodos , Bibliotecas Médicas/organización & administración , Servicios de Biblioteca/organización & administración , Medios de Comunicación Sociales , Humanos
5.
Epilepsy Behav ; 71(Pt B): 238-242, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27993512

RESUMEN

Herpes virus technology involving manipulation of GAD65 was used to study effects on audiogenic seizures (AGS). Audiogenic seizure behaviors were examined following injections of replication-defective herpes simplex virus (HSV-1) vectors incorporating sense or antisense toward GAD65 along with 10% lac-Z into the central nucleus of inferior colliculus (CNIC) of Long-Evans rats. In seizure-sensitive animals developmentally primed by intense sound exposure, injection of GAD65 in the sense orientation increased wild running latencies and reduced incidence of clonus compared with lac-Z only, unoperated, and vehicle seizure groups. In contrast, infection of CNIC with GAD65 antisense virus resulted in 100% incidence of wild running and clonus behaviors in AGS animals. Unprimed animals not operated continued to show uniform absence of seizure activity. Administration of GAD65 antisense virus into CNIC produced novel wild running and clonus behaviors in some unprimed animals. Staining for ß-galactosidase in all vector animals revealed no differences in pattern or numbers of immunoreactive cells at injection sites. Qualitatively, typical small and medium multipolar/stellate and medium fusiform neurons appeared in the CNIC of vector animals. These results demonstrate that HSV-1 vector constructs implanted into the CNIC can predictably influence incidence and severity of AGS and suggest that viral vectors can be useful in studying GABA mechanisms with potential for therapeutic application in epilepsy. This article is part of a Special Issue entitled "Genetic and Reflex Epilepsies, Audiogenic Seizures and Strains: From Experimental Models to the Clinic".


Asunto(s)
Estimulación Acústica/efectos adversos , Epilepsia Refleja/inducido químicamente , Glutamato Descarboxilasa/toxicidad , Herpesvirus Humano 1 , Colículos Inferiores/efectos de los fármacos , Convulsiones/inducido químicamente , Animales , Epilepsia Refleja/patología , Epilepsia Refleja/fisiopatología , Femenino , Glutamato Descarboxilasa/administración & dosificación , Colículos Inferiores/patología , Colículos Inferiores/fisiopatología , Masculino , Neuronas/efectos de los fármacos , Neuronas/patología , Ratas , Ratas Long-Evans , Convulsiones/fisiopatología
6.
Anesthesiology ; 123(3): 642-53, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26146901

RESUMEN

BACKGROUND: Noradrenaline reuptake inhibitors are known to produce analgesia through a spinal action but they also act in the brain. However, the action of noradrenaline on supraspinal pain control regions is understudied. The authors addressed the noradrenergic modulation of the dorsal reticular nucleus (DRt), a medullary pronociceptive area, in the spared nerve injury (SNI) model of neuropathic pain. METHODS: The expression of the phosphorylated cAMP response element-binding protein (pCREB), a marker of neuronal activation, was evaluated in the locus coeruleus and A5 noradrenergic neurons (n = 6 rats/group). pCREB was studied in noradrenergic DRt-projecting neurons retrogradely labeled in SNI animals (n = 3). In vivo microdialysis was used to measure noradrenaline release in the DRt on nociceptive stimulation or after DRt infusion of clonidine (n = 5 to 6 per group). Pharmacology, immunohistochemistry, and western blot were used to study α-adrenoreceptors in the DRt (n = 4 to 6 per group). RESULTS: pCREB expression significantly increased in the locus coeruleus and A5 of SNI animals, and most noradrenergic DRt-projecting neurons expressed pCREB. In SNI animals, noradrenaline levels significantly increased on pinprick (mean ± SD, 126 ± 14%; P = 0.025 vs. baseline) and acetone stimulation (mean ± SD, 151 ± 12%; P < 0.001 vs. baseline), and clonidine infusion showed decreased α2-mediated inhibitory function. α1-adrenoreceptor blockade decreased nociceptive behavioral responses in SNI animals. α2-adrenoreceptor expression was not altered. CONCLUSIONS: Chronic pain induces brainstem noradrenergic activation that enhances descending facilitation from the DRt. This suggests that antidepressants inhibiting noradrenaline reuptake may enhance pain facilitation from the brain, counteracting their analgesic effects at the spinal cord.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Dolor Crónico/metabolismo , Locus Coeruleus/metabolismo , Bulbo Raquídeo/metabolismo , Neuralgia/metabolismo , Norepinefrina/metabolismo , Neuronas Adrenérgicas/patología , Animales , Dolor Crónico/patología , Locus Coeruleus/patología , Masculino , Bulbo Raquídeo/patología , Vías Nerviosas/metabolismo , Vías Nerviosas/patología , Neuralgia/patología , Dimensión del Dolor/métodos , Ratas , Ratas Wistar , Transmisión Sináptica/fisiología
7.
J Neurosci ; 31(1): 172-83, 2011 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-21209202

RESUMEN

Phosphodiesterase-4 (PDE4) plays an important role in mediating memory via the control of intracellular cAMP signaling; inhibition of PDE4 enhances memory. However, development of PDE4 inhibitors as memory enhancers has been hampered by their major side effect of emesis. PDE4 has four subtypes (PDE4A-D) consisting of 25 splice variants. Mice deficient in PDE4D displayed memory enhancement in radial arm maze, water maze, and object recognition tests. These effects were mimicked by repeated treatment with rolipram in wild-type mice. In addition, similarly as rolipram-treated wild-type mice, PDE4D-deficient mice also displayed increased hippocampal neurogenesis and phosphorylated cAMP response element-binding protein (pCREB). Furthermore, microinfusion of lentiviral vectors that contained microRNAs (miRNAs) targeting long-form PDE4D isoforms into bilateral dentate gyri of the mouse hippocampus downregulated PDE4D4 and PDE4D5, enhanced memory, and increased hippocampal neurogenesis and pCREB. Finally, while rolipram and PDE4D deficiency shortened α2 adrenergic receptor-mediated anesthesia, a surrogate measure of emesis, miRNA-mediated PDE4D knock-down in the hippocampus did not. The present results suggest that PDE4D, in particular long-form PDE4D, plays a critical role in the mediation of memory and hippocampal neurogenesis, which are mediated by cAMP/CREB signaling; reduced expression of PDE4D, or at least PDE4D4 and PDE4D5, in the hippocampus enhances memory but appears not to cause emesis. These novel findings will aid in the development of PDE4 subtype- or variant-selective inhibitors for treatment of disorders involving impaired cognition, including Alzheimer's disease.


Asunto(s)
AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/deficiencia , Hipocampo/citología , Memoria/fisiología , Neurogénesis/fisiología , Interferencia de ARN/fisiología , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Reacción de Prevención/efectos de los fármacos , Reacción de Prevención/fisiología , Bromodesoxiuridina/metabolismo , Proteína de Unión a CREB/metabolismo , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , MicroARNs/farmacología , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Pruebas Neuropsicológicas , Inhibidores de Fosfodiesterasa 4/farmacología , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología , Rolipram/farmacología , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo
8.
J Neurosci ; 31(42): 15009-15, 2011 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-22016534

RESUMEN

This study elucidates the neural circuits by which circulating angiotensin II (ANGII) acts in the brain to influence humoral and behavioral responses to psychological stressors. To test the hypothesis that systemic ANGII mediates stress responding via the subfornical organ (SFO), we first found that the timing of increased systemic ANGII in response to 60 min restraint coincides with increased c-fos mRNA expression in the SFO. Next, we administered an anterograde neuronal tract tracer into the SFO and found that fibers originating there make appositions onto neurons in the paraventricular nucleus of the hypothalamus that are also c-fos positive following restraint stress. To determine whether circulating ANGII stimulates the release of stress hormones via activation of angiotensin type 1 receptors (AT1R) within the SFO, we delivered lentivirus to knockdown AT1R expression locally in the SFO. Inhibition of AT1R specifically within the SFO blunted the release of adrenocorticotrophin-releasing hormone and corticosterone in response to restraint stress and caused rats to spend more time in the open arms of an elevated-plus maze than controls, indicating that inhibition of AT1R within the SFO is anxiolytic. Collectively, these results suggest that circulating ANGII acts on AT1R in the SFO to influence responding to psychological stressors.


Asunto(s)
Angiotensina II/farmacología , Conducta Animal/efectos de los fármacos , Sistema Endocrino/efectos de los fármacos , Estrés Psicológico , Órgano Subfornical/efectos de los fármacos , Análisis de Varianza , Angiotensina II/metabolismo , Animales , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hormonas/sangre , Masculino , Fitohemaglutininas/farmacología , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Estrés Psicológico/sangre , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/patología , Órgano Subfornical/metabolismo , Factores de Tiempo , Transducción Genética
9.
PLoS Biol ; 6(1): e13, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18232734

RESUMEN

In all mammals, tissue inflammation leads to pain and behavioral sensitization to thermal and mechanical stimuli called hyperalgesia. We studied pain mechanisms in the African naked mole-rat, an unusual rodent species that lacks pain-related neuropeptides (e.g., substance P) in cutaneous sensory fibers. Naked mole-rats show a unique and remarkable lack of pain-related behaviors to two potent algogens, acid and capsaicin. Furthermore, when exposed to inflammatory insults or known mediators, naked mole-rats do not display thermal hyperalgesia. In contrast, naked mole-rats do display nocifensive behaviors in the formalin test and show mechanical hyperalgesia after inflammation. Using electrophysiology, we showed that primary afferent nociceptors in naked mole-rats are insensitive to acid stimuli, consistent with the animal's lack of acid-induced behavior. Acid transduction by sensory neurons is observed in birds, amphibians, and fish, which suggests that this tranduction mechanism has been selectively disabled in the naked mole-rat in the course of its evolution. In contrast, nociceptors do respond vigorously to capsaicin, and we also show that sensory neurons express a transient receptor potential vanilloid channel-1 ion channel that is capsaicin sensitive. Nevertheless, the activation of capsaicin-sensitive sensory neurons in naked mole-rats does not produce pain-related behavior. We show that capsaicin-sensitive nociceptors in the naked mole-rat are functionally connected to superficial dorsal horn neurons as in mice. However, the same nociceptors are also functionally connected to deep dorsal horn neurons, a connectivity that is rare in mice. The pain biology of the naked mole-rat is unique among mammals, thus the study of pain mechanisms in this unusual species can provide major insights into what constitutes "normal" mammalian nociception.


Asunto(s)
Hiperalgesia/inducido químicamente , Ratas Topo , Nociceptores/efectos de los fármacos , Umbral del Dolor/fisiología , Dolor/fisiopatología , Ácidos/farmacología , Animales , Capsaicina/farmacología , Inflamación , Neuronas Aferentes , Dolor/psicología , Dimensión del Dolor , Células del Asta Posterior
10.
Neurobiol Stress ; 13: 100274, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33344727

RESUMEN

Exposure to prolonged stress is a major risk-factor for psychiatric disorders such as generalized anxiety and major depressive disorder. Human imaging studies have identified structural and functional abnormalities in the prefrontal cortex of subjects with depression and anxiety disorders, particularly Brodmann's area 25 (BA25). Further, deep brain stimulation of BA25 reduces symptoms of treatment-resistant depression. The rat homolog of BA25 is the infralimbic cortex (IL), which is critical for cognitive appraisal, executive function, and physiological stress reactivity. Previous studies indicate that the IL undergoes stress-induced changes in excitatory/inhibitory balance culminating in reduced activity of glutamate output neurons. However, the regulatory role of IL glutamate output in mood-related behaviors after chronic variable stress (CVS) is unknown. Here, we utilized a lentiviral-packaged small-interfering RNA to reduce translation of vesicular glutamate transporter 1 (vGluT1 siRNA), thereby constraining IL glutamate output. This viral-mediated gene transfer was used in conjunction with a quantitative anatomical analysis of cells expressing the stable immediate-early gene product FosB/ΔFosB, which accumulates in response to repeated neural activation. Through assessment of FosB/ΔFosB-expressing neurons across the frontal lobe in adult male rats, we mapped regions altered by chronic stress and determined the coordinating role of the IL in frontal cortical plasticity. Specifically, CVS-exposed rats had increased density of FosB/ΔFosB-expressing cells in the IL and decreased density in the insula. The latter effect was dependent on IL glutamate output. Next, we examined the interaction of CVS and reduced IL glutamate output in behavioral assays examining coping, anxiety-like behavior, associative learning, and nociception. IL glutamate knockdown decreased immobility during the forced swim test compared to GFP controls, both in rats exposed to CVS as well as rats without previous stress exposure. Further, vGluT1 siRNA prevented CVS-induced avoidance behaviors, while also reducing risk aversion and passive coping. Ultimately, this study identifies the necessity of IL glutamatergic output for regulating frontal cortical neural activity and behavior following chronic stress. These findings also highlight how disruption of excitatory/inhibitory balance within specific frontal cortical cell populations may impact neurobehavioral adaptation and lead to stress-related disorders.

11.
Mol Cell Neurosci ; 39(4): 508-18, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18725300

RESUMEN

During chronic pain, the supraspinal pain modulatory system undergoes plastic changes with enhancement of facilitation transmission at the spinal cord. The changes induced by chronic pain at descending modulation often affect opioidergic modulation, and were never described for a key facilitatory component of the system, the dorsal reticular nucleus (DRt). Neurochemical characterization of the DRt-spinal pathway showed that delta-opioid receptors are positioned as to indirectly modulate the activity of non-projecting DRt neurons, whereas neurons expressing mu-opioid receptors project to the spinal dorsal horn or act as interneurons, the latter of which co-expressing GABA(B) receptors. In monoarthritic rats, the expression of mu-opioid receptors decreased in the DRt whereas the levels of endogenous enkephalin remained unaltered. To increase the opioidergic inhibition of the DRt, we locally injected selective agonists of delta- and mu-opioid receptors or a viral vector containing the human preproenkephalin transgene. Injection of the Herpes Simplex viral vector encoding preproenkephalin induced thermal hypoalgesia in non-inflamed animals and hyperalgesia in monoarthritic rats. The opioid agonists [D-Ala(2), Glu(4)]-deltorphin (DELT) and [D-Ala(2), NMePhe(4)Gly-ol(5)]-enkephalin (DAMGO) induced thermal hyperalgesia in both non-inflamed and monoarthritic rats, but with lower doses in the latter group. The present study shows that opioidergic neurons at the DRt are modulated by GABAergic cells herein controlling the descending facilitation of pain transmission. The DRt exhibits plastic changes during chronic inflammatory pain, with decrease opioid receptor expression which may account for increased descending facilitation during chronic pain.


Asunto(s)
Analgésicos Opioides/metabolismo , Dolor/metabolismo , Formación Reticular/metabolismo , Animales , Artritis/inducido químicamente , Artritis/patología , Conducta Animal/fisiología , Vías Eferentes/anatomía & histología , Vías Eferentes/metabolismo , Encefalinas/metabolismo , Humanos , Masculino , Neuronas/citología , Neuronas/metabolismo , Dimensión del Dolor , Ratas , Ratas Wistar , Receptores de GABA-B/metabolismo , Receptores Opioides/metabolismo , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Formación Reticular/anatomía & histología , Ácido gamma-Aminobutírico/metabolismo
12.
Comp Med ; 59(1): 83-90, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19295058

RESUMEN

Herpes simplex virus type 1 (HSV1) is widely used as a gene delivery vector in a variety of laboratory animals. In a recent study, a thymidine-kinase-inactive (replication-conditional) HSV1 used as a delivery vector was lethal in naked mole rats, whereas mice infected with the identical virus showed no adverse effects. This result prompted us to undertake a controlled comparative histologic study of the effect of HSV1 infection on naked mole rats and mice. Replication-competent and replication-conditional HSV1 caused widespread inflammation and necrosis in multiple organ systems of naked mole rats but not mice; naked mole rats infected with replication-defective virus showed no adverse effects. We conclude that the lethality of HSV1 for naked mole rats is likely the result of overwhelming infection, possibly in part due to this species' natural lack of proinflammatory neuropeptides at the initial site of infection.


Asunto(s)
Susceptibilidad a Enfermedades , Herpes Simple/veterinaria , Herpesvirus Humano 1/patogenicidad , Enfermedades de los Roedores/virología , Animales , Antígenos Virales/inmunología , Femenino , Vectores Genéticos , Herpes Simple/patología , Herpes Simple/transmisión , Herpesvirus Humano 1/fisiología , Inflamación/patología , Inflamación/virología , Hígado/patología , Hígado/virología , Ganglios Linfáticos/patología , Ganglios Linfáticos/virología , Masculino , Ratones , Ratas Topo , Necrosis/patología , Necrosis/virología , Enfermedades de los Roedores/transmisión , Especificidad de la Especie , Bazo/patología , Bazo/virología , Timo/patología , Timo/virología , Replicación Viral
13.
Med Ref Serv Q ; 28(1): 10-21, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19197740

RESUMEN

The goal of this study was to determine whether a passive or an interactive tutorial design improves understanding of key concepts, as measured by pre- and post-test data. The authors also collected data regarding the participants' preference for taking an interactive versus a passive tutorial. The interactive tutorial group improved statistically significantly from pre-test to post-test for all three learning questions. While the passive tutorial group improved from pre-test to post-test on all three questions, the improvement was statistically significant for just two of the three questions. The majority of the participants preferred interactive tutorials (78%) to passive tutorials (22%).


Asunto(s)
Instrucción por Computador , Bibliotecas Médicas , Interfaz Usuario-Computador , Humanos , Almacenamiento y Recuperación de la Información , Evaluación de Programas y Proyectos de Salud , Encuestas y Cuestionarios
14.
Front Cell Neurosci ; 13: 287, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31316354

RESUMEN

Opioids play a major role at descending pain modulation but the effects of neuropathic pain on the brain opioidergic system remain understudied. Since descending facilitation is enhanced during neuropathic pain, we studied the opioidergic modulation of the dorsal reticular nucleus (DRt), a medullary pain facilitatory area, in the spared nerve injury (SNI) model of neuropathic pain. We first performed a series of behavioral experiments in naïve-animals to establish the role of µ-opioid receptor (MOR) in the effects of endogenous and exogenous opioids at the DRt. Specifically, we showed that lentiviral-mediated MOR-knockdown at the DRt increased sensitivity to thermal and mechanical stimuli while the MOR agonist DAMGO induced the opposite effects. Additionally, we showed that MOR-knockdown and the pharmacological blockade of MOR by CTAP at the DRt decreased and inhibited, respectively, the analgesic effects of systemic morphine. Then, we performed in vivo microdialysis to measure enkephalin peptides in the DRt and evaluated MOR expression in the DRt at mRNA, protein and phosphorylated form levels by quantitative real-time PCR and immunohistochemistry, respectively. SNI-animals, compared to sham control, showed higher levels of enkephalin peptides, lower MOR-labeled cells without alterations in MOR mRNA levels, and higher phosphorylated MOR-labeled cells. Finally, we performed behavioral studies in SNI animals to determine the potency of systemic morphine and the effects of the pharmacologic and genetic manipulation of MOR at the DRt. We showed a reduced potency of the antiallodynic effects of systemic morphine in SNI-animals compared to the antinociceptive effects in sham animals. Increasing MOR-cells at the DRt of SNI-animals by lentiviral-mediated MOR-overexpression produced no effects on mechanical allodynia. DAMGO induced anti-allodynia only after MOR-overexpression. These results show that MOR inhibits DRt pain facilitatory actions and that this action contributes to the analgesic effects of systemic opioids. We further show that the inhibitory function of MOR is impaired during neuropathic pain. This is likely due to desensitization and degradation of MOR which are adaptations of the receptor that can be triggered by MOR phosphorylation. Skipping counter-regulatory pathways involved in MOR adaptations might restore the opioidergic inhibition at pain facilitatory areas.

15.
Brain Struct Funct ; 224(1): 73-97, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30269223

RESUMEN

The medial prefrontal cortex is critical for contextual appraisal, executive function, and goal-directed behavior. Additionally, the infralimbic (IL) subregion of the prefrontal cortex has been implicated in stress responding, mood, and fear memory. However, the specific circuit mechanisms that mediate these effects are largely unknown. To date, IL output to the limbic forebrain has been examined largely qualitatively or within a restricted number of sites. To quantify IL presynaptic input to structures throughout the forebrain, we utilized a lentiviral construct expressing synaptophysin-mCherry. Thus, allowing quantification of IL efferents that are specifically synaptic, as opposed to fibers of passage. Additionally, this approach permitted the determination of IL innervation on a sub-structural level within the multiple heterogeneous limbic nuclei. To examine the functional output of the IL, optogenetic activation of IL projections was followed by quantification of neuronal activation throughout the limbic forebrain via fos-related antigen (Fra). Quantification of synaptophysin-mCherry indicated that the IL provides robust synaptic input to a number of regions within the thalamus, hypothalamus, amygdala, and bed nucleus of the stria terminalis, with limited input to the hippocampus and nucleus accumbens. Furthermore, there was high concordance between structural connectivity and functional activation. Interestingly, some regions receiving substantial synaptic input did not exhibit significant increases in Fra-immunoreactivity. Collectively, these studies represent a step toward a comprehensive and quantitative analysis of output circuits. This large-scale efferent quantification or 'projectome' also opens the door for data-driven analyses of the downstream synaptic mechanisms that mediate the integrative aspects of cortico-limbic interactions.


Asunto(s)
Lentivirus/genética , Sistema Límbico/fisiología , Optogenética , Corteza Prefrontal/fisiología , Prosencéfalo/fisiología , Transmisión Sináptica , Animales , Genes Reporteros , Lentivirus/metabolismo , Sistema Límbico/citología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Microscopía Fluorescente , Vías Nerviosas/fisiología , Corteza Prefrontal/citología , Corteza Prefrontal/metabolismo , Terminales Presinápticos/fisiología , Prosencéfalo/citología , Prosencéfalo/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Sinaptofisina/genética , Sinaptofisina/metabolismo , Proteína Fluorescente Roja
16.
J Am Heart Assoc ; 8(24): e014451, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31838941

RESUMEN

Background The medial prefrontal cortex is necessary for appropriate appraisal of stressful information, as well as coordinating visceral and behavioral processes. However, prolonged stress impairs medial prefrontal cortex function and prefrontal-dependent behaviors. Additionally, chronic stress induces sympathetic predominance, contributing to health detriments associated with autonomic imbalance. Previous studies identified a subregion of rodent prefrontal cortex, infralimbic cortex (IL), as a key regulator of neuroendocrine-autonomic integration after chronic stress, suggesting that IL output may prevent chronic stress-induced autonomic imbalance. In the current study, we tested the hypothesis that the IL regulates hemodynamic, vascular, and cardiac responses to chronic stress. Methods and Results A viral-packaged small interfering RNA construct was used to knockdown vesicular glutamate transporter 1 (vGluT1) and reduce glutamate packaging and release from IL projection neurons. Male rats were injected with a vGluT1 small interfering RNA-expressing construct or GFP (green fluorescent protein) control into the IL and then remained as unstressed controls or were exposed to chronic variable stress. IL vGluT1 knockdown increased heart rate and mean arterial pressure reactivity, while chronic variable stress increased chronic mean arterial pressure only in small interfering RNA-treated rats. In another cohort, chronic variable stress and vGluT1 knockdown interacted to impair both endothelial-dependent and endothelial-independent vasoreactivity ex vivo. Furthermore, vGluT1 knockdown and chronic variable stress increased histological markers of fibrosis and hypertrophy. Conclusions Knockdown of glutamate release from IL projection neurons indicates that these cells are necessary to prevent the enhanced physiological responses to stress that promote susceptibility to cardiovascular pathophysiology. Ultimately, these findings provide evidence for a neurobiological mechanism mediating the relationship between stress and poor cardiovascular health outcomes.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Corteza Prefrontal/fisiopatología , Estrés Psicológico/complicaciones , Animales , Enfermedad Crónica , Susceptibilidad a Enfermedades , Masculino , Ratas , Ratas Sprague-Dawley
17.
Mol Pain ; 4: 8, 2008 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-18307791

RESUMEN

BACKGROUND: A chronic pancreatitis model was developed in young male Lewis rats fed a high-fat and alcohol liquid diet beginning at three weeks. The model was used to assess time course and efficacy of a replication defective herpes simplex virus type 1 vector construct delivering human cDNA encoding preproenkephalin (HSV-ENK). RESULTS: Most surprising was the relative lack of inflammation and tissue disruption after HSV-ENK treatment compared to the histopathology consistent with pancreatitis (inflammatory cell infiltration, edema, acinar cell hypertrophy, fibrosis) present as a result of the high-fat and alcohol diet in controls. The HSV-ENK vector delivered to the pancreatic surface at week 3 reversed pancreatitis-associated hotplate hypersensitive responses for 4-6 weeks, while control virus encoding beta-galactosidase cDNA (HSV-beta-gal) had no effect. Increased Fos expression seen bilaterally in pain processing regions in control animals with pancreatitis was absent in HSV-ENK-treated animals. Increased met-enkephalin staining was evident in pancreas and lower thoracic spinal cord laminae I-II in the HSV-ENK-treated rats. CONCLUSION: Thus, clear evidence is provided that site specific HSV-mediated transgene delivery of human cDNA encoding preproenkephalin ameliorates pancreatic inflammation and significantly reduces hypersensitive hotplate responses for an extended time consistent with HSV mediated overexpression, without tolerance or evidence of other opiate related side effects.


Asunto(s)
Encefalinas/genética , Herpesvirus Humano 1/genética , Pancreatitis Crónica/terapia , Precursores de Proteínas/genética , Animales , Conducta Animal , ADN Complementario/metabolismo , Vectores Genéticos , Inflamación/metabolismo , Inflamación/terapia , Masculino , Modelos Animales , Ratas , Ratas Endogámicas Lew , Receptores Opioides mu/metabolismo , Médula Espinal/metabolismo , Factores de Tiempo
18.
Eur J Neurosci ; 27(5): 1153-65, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18364035

RESUMEN

This study assessed enkephalin expression induced by intra-articular application of recombinant, enkephalin-encoding herpes virus (HSV-1) and the impact of expression on nociceptive behaviours and synovial lining inflammation in arthritic rats. Replication-conditional HSV-1 recombinant vectors with cDNA encoding preproenkephalin (HSV-ENK), or control transgene beta-galactosidase cDNA (HSV-beta-gal; control) were injected into knee joints with complete Freund's adjuvant (CFA). Joint temperatures, circumferences and nociceptive behaviours were monitored on days 0, 7, 14 and 21 post CFA and vector treatments. Lumbar (L4-6) dorsal root ganglia (DRG) and spinal cords were immunostained for met-enkephalin (met-ENK), beta-gal, HSV-1 proteins and Fos. Joint tissues were immunostained for met-ENK, HSV-1 proteins, and inflammatory mediators Regulated on Activation, Normal T-cell Expressed and Secreted (RANTES) and cyclo-oxygenase-2, or stained with haematoxylin and eosin for histopathology. Compared to exuberant synovial hypertrophy and inflammatory cell infiltration seen in arthritic rats treated with CFA only or CFA and HSV-beta-gal, the CFA- and HSV-ENK-treated arthritic rats had: (i) striking preservation of synovial membrane cytoarchitecture with minimal inflammatory cell infiltrates; (ii) significantly improved nociceptive behavioural responses to mechanical and thermal stimuli; (iii) normalized Fos staining in lumbar dorsal horn; and (iv) significantly increased met-ENK staining in ipsilateral synovial tissue, lumbar DRG and spinal cord. The HSV-1 and transgene product expression were confined to ipsilateral lumbar DRG (HSV-1, met-ENK, beta-gal). Only transgene product (met-ENK and beta-gal) was seen in lumbar spinal cord sections. Targeted delivery of enkephalin-encoding HSV-1 vector generated safe, sustained opioid-induced analgesia with protective anti-inflammatory blunting in rat inflammatory arthritis.


Asunto(s)
Artritis Experimental/patología , Encefalinas/administración & dosificación , Vectores Genéticos/administración & dosificación , Herpesvirus Humano 1 , Mediadores de Inflamación/administración & dosificación , Mediadores de Inflamación/fisiología , Cápsula Articular/patología , Dolor/patología , Animales , Artritis Experimental/fisiopatología , Artritis Experimental/prevención & control , Conducta Animal/fisiología , Encefalinas/genética , Adyuvante de Freund/administración & dosificación , Marcación de Gen/métodos , Vectores Genéticos/genética , Herpesvirus Humano 1/genética , Inflamación/patología , Inflamación/prevención & control , Cápsula Articular/efectos de los fármacos , Masculino , Dolor/prevención & control , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Ratas , Ratas Sprague-Dawley
19.
Anesthesiology ; 108(2): 305-13, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18212576

RESUMEN

BACKGROUND: The use of opioids to treat pain is often limited by side effects mediated through the central nervous system. The current study used a recombinant herpes simplex virus type 1 to increase expression of the mu-opioid receptor (muOR) in primary afferent neurons. The goal of this strategy was to enhance peripheral opioid analgesia. METHODS: Cutaneous inoculation with herpes simplex virus containing muOR complementary DNA (cDNA) in antisense (SGAMOR) or sense (SGMOR) orientation relative to a constitutive promoter, or complementary DNA for Escherichia coli lac Z gene as a control virus (SGZ) was used to modify the levels of muOR in primary afferents. The effects of altered muOR levels on peripheral analgesia were then examined. RESULTS: At 4 weeks after SGAMOR and SGMOR infection, decreased and increased muOR immunoreactivity was observed in ipsilateral dorsal hind paw skin, lumbar dorsal root ganglion cells, and superficial dorsal horns, respectively, compared with SGZ. This change in muOR expression in mice by SGAMOR and SGMOR was accompanied at the behavioral level with a rightward and leftward shift in the loperamide dose-response curve, respectively, compared with SGZ. CONCLUSIONS: This gene therapy approach may provide an innovative strategy to enhance peripheral opioid analgesia for the treatment of pain in humans, thereby minimizing centrally mediated opioid side effects such as sedation and addiction.


Asunto(s)
Analgesia/métodos , Loperamida/farmacología , Receptores Opioides mu/genética , Anestesia Raquidea , Animales , ADN Complementario/genética , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiología , Técnicas de Transferencia de Gen , Herpesviridae/genética , Ratones , Terminaciones Nerviosas/efectos de los fármacos , Terminaciones Nerviosas/fisiología , Piel/inervación , Médula Espinal/efectos de los fármacos , Médula Espinal/fisiología
20.
Med Ref Serv Q ; 27(4): 406-18, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19042720

RESUMEN

In a 2007 study, librarians at the University of South Carolina School of Medicine Library examined freely available online tutorials on academic medical library Web sites. The team identified tutorial topics, determined common design features, and assessed elements of active learning in library-created tutorials; the team also generated a list of third-party tutorials to which medical libraries link. This article updates the earlier study, describing changes and trends in tutorial content and design on medical libraries' Web sites; the project team plans to continue to track trends in tutorial development by repeating this study annually.


Asunto(s)
Instrucción por Computador/estadística & datos numéricos , Internet/estadística & datos numéricos , Bibliotecas Médicas/estadística & datos numéricos , Sistemas en Línea/estadística & datos numéricos , Facultades de Medicina , DC-I , Instrucción por Computador/métodos , Instrucción por Computador/provisión & distribución , Educación a Distancia/métodos , Educación a Distancia/estadística & datos numéricos , Humanos , Encuestas de Bibliotecas , Sistemas en Línea/provisión & distribución , PubMed , Edición/estadística & datos numéricos , Programas Informáticos , South Carolina , Encuestas y Cuestionarios , Estados Unidos , Interfaz Usuario-Computador
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA