Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Pharmacol Exp Ther ; 389(2): 136-149, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-37442620

RESUMEN

Traumatic brain injury (TBI) is a leading cause of death and disability in the United States, and survivors often experience mental and physical health consequences that reduce quality of life. We previously reported that blockade of the nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor reduced tissue damage markers produced by blast TBI. The goal of this study was to determine the extent to which N/OFQ and NOP receptor levels change following mild (mTBI) and moderate TBI (modTBI) and whether the absence of the NOP receptor attenuates TBI-induced sequelae. Male and female NOP receptor knockout (KO) or wild-type (WT) rats received craniotomy-only (sham) or craniotomy plus mTBI, or modTBI impact to the left cerebral hemisphere. Neurologic and vestibulomotor deficits and nociceptive hyperalgesia and allodynia found in WT male and female rats following mTBI and modTBI were greatly reduced or absent in NOP receptor KO rats. NOP receptor levels increased in brain tissue from injured males but remained unchanged in females. Neurofilament light chain (NF-L) and glial fibrillary acidic protein (GFAP) expression were reduced in NOP receptor KO rats compared with WT following TBI. Levels of N/OFQ in injured brain tissue correlated with neurobehavioral outcomes and GFAP in WT males, but not with KO male or WT and KO female rats. This study reveals a significant contribution of the N/OFQ-NOP receptor system to TBI-induced deficits and suggests that the NOP receptor should be regarded as a potential therapeutic target for TBI. SIGNIFICANCE STATEMENT: This study revealed that nociceptin/orphanin FQ peptide (NOP) receptor knockout animals experienced fewer traumatic brain injury (TBI)-induced deficits than their wild-type counterparts in a sex- and injury severity-dependent manner, suggesting that NOP receptor antagonists may be a potential therapy for TBI.

2.
Int J Mol Sci ; 25(3)2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38338936

RESUMEN

Traumatic brain injury (TBI) is a major cause of mortality and disability around the world, for which no treatment has been found. Nociceptin/Orphanin FQ (N/OFQ) and the nociceptin opioid peptide (NOP) receptor are rapidly increased in response to fluid percussion, stab injury, and controlled cortical impact (CCI) TBI. TBI-induced upregulation of N/OFQ contributes to cerebrovascular impairment, increased excitotoxicity, and neurobehavioral deficits. Our objective was to identify changes in N/OFQ and NOP receptor peptide, protein, and mRNA relative to the expression of injury markers and extracellular regulated kinase (ERK) 24 h following mild (mTBI) and moderate TBI (ModTBI) in wildtype (WT) and NOP receptor-knockout (KO) rats. N/OFQ was quantified by radioimmunoassay, mRNA expression was assessed using real-time PCR and protein levels were determined by immunoblot analysis. This study revealed increased N/OFQ mRNA and peptide levels in the CSF and ipsilateral tissue of WT, but not KO, rats 24 h post-TBI; NOP receptor mRNA increased after ModTBI. Cofilin-1 activation increased in the brain tissue of WT but not KO rats, ERK activation increased in all rats following ModTBI; no changes in injury marker levels were noted in brain tissue at this time. In conclusion, this study elucidates transcriptional and translational changes in the N/OFQ-NOP receptor system relative to TBI-induced neurological deficits and initiation of signaling cascades that support the investigation of the NOP receptor as a therapeutic target for TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Receptor de Nociceptina , Nociceptina , Animales , Ratas , Analgésicos Opioides , Lesiones Traumáticas del Encéfalo/genética , Péptidos Opioides/metabolismo , Receptores Opioides/metabolismo , ARN Mensajero/metabolismo
3.
Cell Mol Neurobiol ; 41(5): 1009-1018, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32930941

RESUMEN

Chronic headache pain is one of the most commonly reported comorbid pain conditions with post-traumatic stress disorder (PTSD) patients and resistant to effective treatment, yet no combined preclinical model of the two disorders has been reported. Here, we used a modified chronic headache pain model to investigate the contribution of single prolonged stress (SPS) model of PTSD with sodium nitroprusside (SNP)-induced hyperalgesia. Injection of SNP (2 mg/kg, i.p.) occurred every other day from day 7 to day 15 after initiation of SPS in rats. Paw withdrawal threshold (PWT) to von Frey stimuli and tail flick latencies (TFL) dramatically decreased as early as 7 days after SPS and lasted until at least day 21. Basal PWT and TFL also significantly decreased during the SNP treatment period. The lower nociceptive thresholds recovered in 6 days following the final SNP injection in SNP group, but not in SPS + SNP group. Elevated nociceptin/OFQ (N/OFQ) levels observed in cerebrospinal fluid of SPS rats were even higher in SPS + SNP group. Glial fibrillary acidic protein (GFAP) and N/OFQ peptide (NOP) receptor mRNA expression increased in dorsal root ganglia (DRG) 21 days after SPS exposure; mRNA increases in the SPS/SNP group was more pronounced than SPS or SNP alone. GFAP protein expression was upregulated in trigeminal ganglia by SPS. Our results indicate that traumatic stress exaggerated chronic SNP-induced nociceptive hypersensitivity, and that N/OFQ and activated satellite glia cells may play an important role in the interaction between both conditions.


Asunto(s)
Modelos Animales de Enfermedad , Cefalea/metabolismo , Dolor/metabolismo , Trastornos por Estrés Postraumático/metabolismo , Estrés Psicológico/metabolismo , Animales , Proteína Ácida Fibrilar de la Glía/metabolismo , Cefalea/inducido químicamente , Cefalea/psicología , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Hiperalgesia/psicología , Masculino , Nitroprusiato/toxicidad , Péptidos Opioides/metabolismo , Dolor/inducido químicamente , Dolor/psicología , Ratas , Ratas Sprague-Dawley , Trastornos por Estrés Postraumático/psicología , Estrés Psicológico/psicología
4.
Cell Mol Neurobiol ; 41(5): 827-834, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33978862

RESUMEN

This special issue is a tribute to our mentor, colleague and friend, Gavril W. Pasternak, MD, PhD. Homage to the breadth and depth of his work (~ 450 publications) over a 40 career in pharmacology and medicine cannot be captured fully in one special issue, but the 22 papers collected herein represent seven of the topics near and dear to Gav's heart, and the colleagues, friends and mentees who held him near to theirs. The seven themes include: (1) sites and mechanisms of opioid actions in vivo; (2) development of novel analgesic agents; (3) opioid tolerance, withdrawal and addiction: mechanisms and treatment; (4) opioid receptor splice variants; (5) novel research tools and approaches; (6) receptor signaling and crosstalk in vitro; and (7) mentorship. This introduction to the issue summarizes contributions and includes formal and personal remembrances of Gav that illustrate his personality, warmth, and dedication to making a difference in patient care and people's lives.


Asunto(s)
Analgesia/historia , Analgésicos Opioides/historia , Personal de Laboratorio/historia , Manejo del Dolor/historia , Dolor/historia , Médicos/historia , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Receptores Opioides/historia
5.
Mol Pharmacol ; 83(5): 907-18, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23395957

RESUMEN

The nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor is the fourth and most recently discovered member of the opioid receptor superfamily that also includes µ, δ, and κ opioid receptor subtypes (MOR, DOR, and KOR, respectively). The widespread anatomic distribution of the NOP receptor enables the modulation of several physiologic processes by its endogenous agonist, N/OFQ. Accordingly, the NOP receptor has gained a lot of attention as a potential target for the development of ligands with therapeutic use in several pathophysiological states. NOP receptor activation frequently results in effects opposing classic opioid receptor action; therefore, regulation of the NOP receptor and conditions affecting its modulatory tone are important to understand. Mounting evidence reveals a heterologous interaction of the NOP receptor with other G protein-coupled receptors, including MOR, DOR, and KOR, which may subsequently influence their function. Our focus in this review is to summarize and discuss the findings that delineate the cellular mechanisms of NOP receptor signaling and regulation and the regulation of other receptors by N/OFQ and the NOP receptor.


Asunto(s)
Péptidos Opioides/metabolismo , Receptores Opioides/metabolismo , Animales , Humanos , Ligandos , Receptor de Nociceptina , Nociceptina
6.
Front Pharmacol ; 14: 1272969, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37920208

RESUMEN

Traumatic brain injury (TBI) affects more than 2.5 million people in the U.S. each year and is the leading cause of death and disability in children and adults ages 1 to 44. Approximately 90% of TBI cases are classified as mild but may still lead to acute detrimental effects such as impaired cerebral blood flow (CBF) that result in prolonged impacts on brain function and quality of life in up to 15% of patients. We previously reported that nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor antagonism reversed mild blast TBI-induced vestibulomotor deficits and prevented hypoxia. To explore mechanisms by which the NOP receptor-N/OFQ pathway modulates hypoxia and other TBI sequelae, the ability of the NOP antagonist, SB-612111 (SB), to reverse TBI-induced CBF and associated injury marker changes were tested in this study. Male Wistar rats randomly received sham craniotomy or craniotomy + TBI via controlled cortical impact. Injury severity was assessed after 1 h (modified neurological severity score (mNSS). Changes in CBF were assessed 2 h post-injury above the exposed cortex using laser speckle contrast imaging in response to the direct application of increasing concentrations of vehicle or SB (1, 10, and 100 µM) to the brain surface. TBI increased mNSS scores compared to baseline and confirmed mild TBI (mTBI) severity. CBF was significantly impaired on the ipsilateral side of the brain following mTBI, compared to contralateral side and to sham rats. SB dose-dependently improved CBF on the ipsilateral side after mTBI compared to SB effects on the respective ipsilateral side of sham rats but had no effect on contralateral CBF or in uninjured rats. N/OFQ levels increased in the cerebral spinal fluid (CSF) following mTBI, which correlated with the percent decrease in ipsilateral CBF. TBI also activated ERK and cofilin within 3 h post-TBI; ERK activation correlated with increased CSF N/OFQ. In conclusion, this study reveals a significant contribution of the N/OFQ-NOP receptor system to TBI-induced dysregulation of cerebral vasculature and suggests that the NOP receptor should be considered as a potential therapeutic target for TBI.

7.
Mol Pain ; 8: 76, 2012 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-23082795

RESUMEN

BACKGROUND: Clinical studies indicate that post-traumatic stress disorder (PTSD) frequently shares co-morbidity with chronic pain. Although in animals acute stress-induced antinociception is well documented, the effect of PTSD-like stress on nociceptive sensitivity is unclear. Though a few studies measured nociceptive responses at a single time point, no studies have examined changes in nociceptive sensitivity over time following exposure to PTSD-like stress. Nociceptin/orphanin FQ (N/OFQ), an endogenous ligand for the N/OFQ peptide (NOP) receptor, modulates various biological functions in the central nervous system that are affected by PTSD, including nociceptive sensitivity, stress and anxiety, learning and memory. RESULTS: The present study examined thermal and mechanical nociceptive sensitivity in male Sprague Dawley rats between 7 and 28 days after single-prolonged stress (SPS), an established animal model for PTSD. Rat paw withdrawal thresholds (PWT) to von Frey and paw withdrawal latencies (PWL) to radiant heat stimuli, respectively, dramatically decreased as early as 7 days after initiation of SPS and lasted the length of the study, 28 days. In addition, N/OFQ levels increased in cerebrospinal fluid (CSF; on days 9, 14 and 28) and serum (day 28), while levels of circulating corticosterone (CORT) decreased 28 days after initiation of SPS. SPS exposure induced anxiety-like behavior and enhanced inhibition of the hypothalamo-pituitary-adrenal (HPA) axis, as previously reported for this model. CONCLUSIONS: Our results demonstrate that SPS induces the development of persistent mechanical allodynia and thermal hyperalgesia that is accompanied by increased N/OFQ content in the CSF, and eventually, in serum. These findings suggest a link between N/OFQ and the development of hyperalgesia and allodynia in a rat model of PTSD.


Asunto(s)
Péptidos Opioides/metabolismo , Trastornos por Estrés Postraumático/metabolismo , Animales , Hiperalgesia/metabolismo , Masculino , Dolor/metabolismo , Ratas , Ratas Sprague-Dawley , Nociceptina
8.
Pharmacol Ther ; 231: 107982, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34480968

RESUMEN

The nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor is a member of the opioid receptor superfamily with N/OFQ as its endogenous agonist. Wide expression of the NOP receptor and N/OFQ, both centrally and peripherally, and their ability to modulate several biological functions has led to development of NOP receptor modulators by pharmaceutical companies as therapeutics, based upon their efficacy in preclinical models of pain, anxiety, depression, Parkinson's disease, and substance abuse. Both posttraumatic stress disorder (PTSD) and traumatic brain injury (TBI) are debilitating conditions that significantly affect the quality of life of millions of people around the world. PTSD is often a consequence of TBI, and, especially for those deployed to, working and/or living in a war zone or are first responders, they are comorbid. PTSD and TBI share common symptoms, and negatively influence outcomes as comorbidities of the other. Unfortunately, a lack of effective therapies or therapeutic agents limits the long term quality of life for either TBI or PTSD patients. Ours, and other groups, demonstrated that PTSD and TBI preclinical models elicit changes in the N/OFQ-NOP receptor system, and that administration of NOP receptor ligands alleviated some of the neurobiological and behavioral changes induced by brain injury and/or traumatic stress exposure. Here we review the past and most recent progress on understanding the role of the N/OFQ-NOP receptor system in PTSD and TBI neurological and behavioral sequelae. There is still more to understand about this neuropeptide system in both PTSD and TBI, but current findings warrant further examination of the potential utility of NOP modulators as therapeutics for these disorders and their co-morbidities. We advocate the development of standards for common data elements (CDE) reporting for preclinical PTSD studies, similar to current preclinical TBI CDEs. That would provide for more standardized data collection and reporting to improve reproducibility, interpretation and data sharing across studies.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Calidad de Vida , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Humanos , Morbilidad , Péptidos Opioides/metabolismo , Péptidos Opioides/uso terapéutico , Reproducibilidad de los Resultados , Nociceptina
9.
Front Psychiatry ; 12: 721999, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34512420

RESUMEN

Post-Traumatic Stress Disorder (PTSD) is a debilitating mental health disorder that occurs after exposure to a traumatic event. Patients with comorbid chronic pain experience affective distress, worse quality of life, and poorer responses to treatments for pain or PTSD than those with either condition alone. FDA-approved PTSD treatments are often ineffective analgesics, requiring additional drugs to treat co-morbid symptoms. Therefore, development of new treatment strategies necessitate a better understanding of the pathophysiology of PTSD and comorbid pain. The single prolonged stress (SPS) model of PTSD induces the development of persistent mechanical allodynia and thermal hyperalgesia. Increased Nociceptin/Orphanin FQ (N/OFQ) levels in serum and CSF accompany these exaggerated nociceptive responses, as well as increased serum levels of the pro-inflammatory cytokine tumor necrosis factor (TNF-α). Therefore, the primary goal was to determine the role of TNF-α in the development of SPS-induced allodynia/hyperalgesia and elevated serum and CNS N/OFQ using two approaches: TNF-α synthesis inhibition, and blockade with anti-TNF-α antibody that acts primarily in the periphery. Administration of TNF-α synthesis blocker, thalidomide (THL), immediately after SPS prevented increased TNF-α and development of allodynia and hyperalgesia. The THL effect lasted at least 21 days, well after thalidomide treatment ended (day 5). THL also prevented SPS-induced increases in serum N/OFQ and reversed regional N/OFQ mRNA expression changes in the CNS. Serum TNF-α increases detected at 4 and 24 h post SPS were not accompanied by blood brain barrier disruption. A single injection of anti-TNF-α antibody to male and female rats during the SPS procedure prevented the development of allodynia, hyperalgesia, and elevated serum N/OFQ, and reduced SPS-induced anxiety-like behaviors in males. Anti-TNFα treatment also blocked development of SPS-induced allodynia in females, and blocked increased hypothalamic N/OFQ in males and females. This suggests that a peripheral TNF-α surge is necessary for the initiation of allodynia associated with SPS, as well as the altered central and peripheral N/OFQ that maintains nociceptive sensitivity. Therefore, early alleviation of TNF-α provides new therapeutic options for investigation as future PTSD and co-morbid pain treatments.

10.
Front Psychiatry ; 9: 731, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30670988

RESUMEN

Nociceptin/Orphanin FQ (N/OFQ) is a neuropeptide that modulates pain transmission, learning/memory, stress, anxiety, and fear responses via activation of the N/OFQ peptide (NOP or ORL1) receptor. Post-traumatic stress disorder (PTSD) is an anxiety disorder that may arise after exposure to a traumatic or fearful event, and often is co-morbid with chronic pain. Using an established animal model of PTSD, single-prolonged stress (SPS), we were the first to report that NOP receptor antagonist treatment reversed traumatic stress-induced allodynia, thermal hyperalgesia, and anxiety-like behaviors in male Sprague-Dawley rats. NOP antagonist treatment also reversed SPS-induced serum and CSF N/OFQ increase and circulating corticosterone decrease. The objective of this study was to examine the role of the NOP receptor in male and female rats subjected to traumatic stress using Wistar wild type (WT) and NOP receptor knockout (KO) rats. The severity of co-morbid allodynia was assessed as change in paw withdrawal threshold (PWT) to von Frey and paw withdrawal latency (PWL) to radiant heat stimuli, respectively. PWT and PWL decreased in male and female WT rats within 7 days after SPS, and remained decreased through day 28. Baseline sensitivity did not differ between genotypes. However, while male NOP receptor KO rats were protected from SPS-induced allodynia and thermal hypersensitivity, female NOP receptor KO rats exhibited tactile allodynia and thermal hypersensitivity to the same extent as WT rats. Male NOP receptor KO rats had a lower anxiety index (AI) than WT, but SPS did not increase AI in WT males. In contrast, SPS significantly increased AI in WT and NOP receptor KO female rats. SPS increased circulating N/OFQ levels in male WT, but not in male NOP receptor KO, or WT or KO female rats. These results indicate that the absence of the NOP receptor protects males from traumatic-stress-induced allodynia and hyperalgesia, consistent with our previous findings utilizing a NOP receptor antagonist. However, female NOP receptor KO rats experience allodynia, hyperalgesia and anxiety-like symptoms to the same extent as WT females following SPS. This suggests that endogenous N/OFQ-NOP receptor signaling plays an important, but distinct, role in males and females following exposure to traumatic stress.

11.
Behav Brain Res ; 340: 183-194, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-27793733

RESUMEN

Mild traumatic brain injury (mTBI) diagnoses have increased due to aggressive sports and blast-related injuries, but the cellular mechanisms and pathology underlying mTBI are not completely understood. Previous reports indicate that Nociceptin Orphanin/FQ (N/OFQ), an endogenous neuropeptide, contributes to post-injury ischemia following mechanical brain injury, yet its specific role in cerebral hypoxia, vestibulomotor function and injury marker expression following blast-induced TBI is not known. This study is the first to identify a direct association of N/OFQ and its N/OFQ peptide (NOP) receptor with TBI-induced changes following a single 80psi head blast exposure in male rats. N/OFQ and NOP receptor expression increased in brain tissue and plasma following TBI, concurrent with vestibular dysfunction but preceding hypoxia and appearance of injury markers compared to sham rats. A single post-blast treatment with the NOP receptor antagonist, SB-612111, transiently improved acute vestibulomotor performance. It also prevented increases in markers of TBI-induced hypoxia, pro-apoptotic proteins and injury seen 8-10days post-blast. This study reveals an apparent role for the N/OFQ-NOP receptor system in blast TBI and suggests potential therapeutic utility of NOP receptor antagonists for mTBI.


Asunto(s)
Traumatismos por Explosión/tratamiento farmacológico , Conmoción Encefálica/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Cicloheptanos/farmacología , Hipoxia Encefálica/prevención & control , Antagonistas de Narcóticos/farmacología , Piperidinas/farmacología , Animales , Traumatismos por Explosión/patología , Traumatismos por Explosión/fisiopatología , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Encéfalo/patología , Conmoción Encefálica/etiología , Conmoción Encefálica/patología , Conmoción Encefálica/fisiopatología , Hipoxia Encefálica/etiología , Hipoxia Encefálica/patología , Hipoxia Encefálica/fisiopatología , Masculino , Actividad Motora/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Proteoma/efectos de los fármacos , Ratas Sprague-Dawley , Receptores Opioides/metabolismo , Receptor de Nociceptina
12.
BMC Pharmacol ; 7: 16, 2007 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-18096057

RESUMEN

BACKGROUND: In addition to the regulation of blood pressure, alpha2- and beta-adrenoceptor (AR) subtypes play an important role in the modulation of noradrenergic neurotransmission in the human CNS and PNS. Several studies suggest that the alpha2-AR responsiveness in cells and tissues after chronic epinephrine (EPI) or norepinephrine (NE) exposure may vary, depending on the beta-AR activity present there. Recently, we reported that in BE(2)-C human neuroblastoma cells (endogenously expressing alpha2A- and beta2-AR), chronic EPI treatment (300 nM) produced a dramatic beta-adrenoceptor-dependent desensitization of the alpha2A-AR response. The aim of this study is to determine if stable addition of a beta2-AR to a second neuroblastoma cell line (SH-SY5Y), that normally expresses only alpha2A-ARs that are not sensitive to 300 nM EPI exposure, would suddenly render alpha2A-ARs in that cell line sensitive to treatment with the same EPI concentration. METHODS: These studies employed RT-PCR, receptor binding and inhibition of cAMP accumulation to confirm alpha2-AR subtype expression. Stable clones of SH-SY5Y cells transfected to stably express functional beta2-ARs (SHbeta2AR4) were selected to compare sensitivity of alpha2-AR to EPI in the presence or absence of beta2-ARs. RESULTS: A series of molecular, biochemical and pharmacological studies indicated that the difference between the cell lines could not be attributed to alpha2-AR heterogeneity. We now report that after transfection of functional beta2-AR into SH-SY5Y cells (SHbeta2AR4), chronic treatment with modest levels of EPI desensitizes the alpha2A-AR. This effect results from a beta2-AR dependent down-regulation of native alpha2A-ARs by EPI accompanied by enhanced translocation of GRK2 and GRK3 to the membrane (required for GRK-mediated phosphorylation of agonist-occupied receptors). CONCLUSION: This study further supports the hypothesis that the presence of the beta-AR renders the alpha2A-AR more susceptible to desensitization with physiological levels of EPI.


Asunto(s)
Epinefrina/farmacología , Receptores Adrenérgicos alfa 2/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Agonistas de Receptores Adrenérgicos alfa 2 , Agonistas alfa-Adrenérgicos/farmacología , Antagonistas de Receptores Adrenérgicos beta 2 , Tartrato de Brimonidina , Línea Celular Tumoral , Membrana Celular/metabolismo , AMP Cíclico/antagonistas & inhibidores , AMP Cíclico/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Quinasa 3 del Receptor Acoplado a Proteína-G/metabolismo , Humanos , Norepinefrina/farmacología , Propranolol/farmacología , Quinoxalinas/farmacología , ARN Mensajero/metabolismo , Receptores Adrenérgicos alfa 2/genética , Receptores Adrenérgicos beta 2/genética , Transfección
13.
J Pediatr Intensive Care ; 6(2): 83-90, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31073429

RESUMEN

Tolerance is a complication of fentanyl continuous infusions (CINs) in critically ill children, but the incidence and time of onset are lacking. The primary objective was to identify the incidence of tolerance. Secondary objectives were to determine the onset time and compare risk factors between children with tolerance versus no tolerance and between children with early (< 24 hours) versus late tolerance. Children aged 0 to 17 years, receiving fentanyl CIN > 3 days from May 1, 2012 to June 30, 2013 were included. Tolerance was defined as a doubling of the fentanyl CIN dose. Descriptive and inferential statistics were performed. A logistic regression model was used to assess the relationship between the development of tolerance and independent variables. A total of 59 CINs were included. Tolerance occurred in 46 CINs (78%), with median time to tolerance of 26 hours (range: 1-160 hours). Early tolerance was identified in 21 CINs (45.7%). Patients with tolerance had higher peak CIN doses (p < 0.001), final CIN doses (p = 0.031), and cumulative exposure (p = 0.017). No significant differences were noted between those with early versus late tolerance. The regression model noted factors associated with the odds of development of tolerance were lower initial fentanyl dose (p = 0.007; odds ratio [OR]: 0.011, 95% confidence interval [CI]: 0.0004-0.29) and higher cumulative exposure (p = 0.009; OR: 1.01, 95% CI: 1.001-1.01). Tolerance developed in 78% of children, and half developed it within 24 hours. Lower initial opioid dose and higher cumulative exposure were independently associated with tolerance.

14.
BMC Neurosci ; 7: 54, 2006 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-16839421

RESUMEN

BACKGROUND: Syntaxins 1 through 4 are SNAP receptor (SNARE) proteins that mediate vesicular trafficking to the plasma membrane. In retina, syntaxins 1 and 3 are expressed at conventional and ribbon synapses, respectively, suggesting that synaptic trafficking functions differ among syntaxin isoforms. To better understand syntaxins in synaptic signaling and trafficking, we further examined the cell- and synapse-specific expression of syntaxins 1 through 4 in the mouse retina by immunolabeling and confocal microscopy. RESULTS: Each isoform was expressed in the retina and showed a unique distribution in the synaptic layers of the retina, with little or no colocalization of isoforms. Syntaxin 1 was present in amacrine cell bodies and processes and conventional presynaptic terminals in the inner plexiform layer (IPL). Syntaxin 2 was present in amacrine cells and their processes in the IPL, but showed little colocalization with syntaxin 1 or other presynaptic markers. Syntaxin 3 was found in glutamatergic photoreceptor and bipolar cell ribbon synapses, but was absent from putative conventional glutamatergic amacrine cell synapses. Syntaxin 4 was localized to horizontal cell processes in the ribbon synaptic complexes of photoreceptor terminals and in puncta in the IPL that contacted dopaminergic and CD15-positive amacrine cells. Syntaxins 2 and 4 often were apposed to synaptic active zones labeled for bassoon. CONCLUSION: These results indicate that each syntaxin isoform has unique, non-redundant functions in synaptic signaling and trafficking. Syntaxins 1 and 3 mediate presynaptic transmitter release from conventional and ribbon synapses, respectively. Syntaxins 2 and 4 are not presynaptic and likely mediate post-synaptic trafficking.


Asunto(s)
Proteínas Qa-SNARE/metabolismo , Retina/citología , Sinapsis/metabolismo , Animales , Western Blotting/métodos , Expresión Génica/fisiología , Inmunohistoquímica/métodos , Ratones , Ratones Endogámicos C57BL , Isoformas de Proteínas/metabolismo , Sinapsis/clasificación , Sinapsis/ultraestructura
15.
Front Neurol ; 6: 132, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26136722

RESUMEN

Physiological alterations, anxiety, and cognitive disorders are strongly associated with blast-induced traumatic brain injury (blast TBI), and are common symptoms in service personnel exposed to blasts. Since 2006, 25,000-30,000 new TBI cases are diagnosed annually in U.S. Service members; increasing evidence confirms that primary blast exposure causes diffuse axonal injury and is often accompanied by altered behavioral outcomes. Behavioral and acute metabolic effects resulting from blast to the head in the absence of thoracic contributions from the periphery were examined, following a single blast wave directed to the head of male Sprague-Dawley rats protected by a lead shield over the torso. An 80 psi head blast produced cognitive deficits that were detected in working memory. Blast TBI rats displayed increased anxiety as determined by elevated plus maze at day 9 post-blast compared to sham rats; blast TBI rats spent significantly more time than the sham controls in the closed arms (p < 0.05; n = 8-11). Interestingly, anxiety symptoms were absent at days 22 and 48 post-blast. Instead, blast TBI rats displayed increased rearing behavior at day 48 post-blast compared to sham rats. Blast TBI rats also exhibited suppressed acoustic startle responses, but similar pre-pulse inhibition at day 15 post-blast compared to sham rats. Acute physiological alterations in cerebral glucose metabolism were determined by positron emission tomography 1 and 9 days post-blast using (18)F-fluorodeoxyglucose ((18)F-FDG). Global glucose uptake in blast TBI rat brains increased at day 1 post-blast (p < 0.05; n = 4-6) and returned to sham levels by day 9. Our results indicate a transient increase in cerebral metabolism following a blast injury. Markers for reactive astrogliosis and neuronal damage were noted by immunoblotting motor cortex tissue from day 10 post-blast in blast TBI rats compared to sham controls (p < 0.05; n = 5-6).

16.
J Comp Neurol ; 443(3): 275-88, 2002 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-11807837

RESUMEN

Regeneration of functionally normal synapses is required for functional recovery after degenerative central nervous system insults and requires proper expression and targeting of presynaptic proteins by regenerating neurons. The reconstitution of presynaptic terminals by regenerating adult neurons is poorly understood, however. We examined the intrinsic ability of regenerating adult retinal photoreceptors to reconstitute properly differentiated presynaptic terminals in the absence of target contact. The expression and localization of vesicle-associated membrane protein (VAMP), synaptic vesicle protein 2 (SV2), synaptophysin, synapsin I, and synaptosomal-associated protein of 25 kDa (SNAP-25) was assessed immunocytochemically. Photoreceptor terminals in the intact retina contain VAMP, SV2, synaptophysin, and SNAP-25, but not synapsin I. Isolated, regenerating adult photoreceptors intrinsically expressed the proper complement of synaptic vesicle proteins in the absence of target contact: VAMP, SV2, and synaptophysin were present at all stages of regenerative growth; synapsin I was never expressed. At early stages of regenerative growth, VAMP, SV2, and synaptophysin were diffusely localized in the cell, with prominent VAMP labeling distributed along the plasma membrane. SV2 and synaptophysin rapidly localized to regenerated terminals, but VAMP accumulated much more slowly, indicating that these proteins are trafficked independently. In contrast, labeling for SNAP-25, which is associated with the presynaptic plasma membrane, was undetectable in regenerating photoreceptors, suggesting that SNAP-25 expression is target-regulated. Thus, regenerating photoreceptors can intrinsically regulate the expression of the proper set of synaptic vesicle proteins. Proper expression of other presynaptic proteins, such as SNAP-25, and proper subcellular localization of synaptic proteins such as VAMP, however, may require extrinsic cues such as target contact.


Asunto(s)
Ambystoma/crecimiento & desarrollo , Diferenciación Celular/fisiología , Regeneración Nerviosa/fisiología , Proteínas del Tejido Nervioso/metabolismo , Células Fotorreceptoras/metabolismo , Terminales Presinápticos/metabolismo , Factores de Edad , Ambystoma/anatomía & histología , Ambystoma/metabolismo , Animales , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica/fisiología , Inmunohistoquímica , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/ultraestructura , Neuritas/metabolismo , Neuritas/ultraestructura , Células Fotorreceptoras/citología , Terminales Presinápticos/ultraestructura , Seudópodos/metabolismo , Seudópodos/ultraestructura , Proteínas R-SNARE , Sinapsinas/metabolismo , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestructura , Sinaptofisina/metabolismo , Proteína 25 Asociada a Sinaptosomas , Factores de Tiempo
17.
Pain ; 45(1): 87-94, 1991 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-1677751

RESUMEN

Central morphine analgesia is significantly greater in male than in female rats. Since mu and delta opioid receptor subtypes have been implicated in supraspinal analgesia, the present study evaluated whether gender or adult gonadectomy altered (a) analgesia on the tail-flick and jump tests following central administration of the mu-selective agonist, [D-Ala2, Me-Phe4, Gly(ol)5] enkephalin (DAMGO) and the delta-selective agonist, [D-Ser2,Leu5] enkephalin-Thr6 (DSLET) and (b) mu1, mu2 and delta opioid receptor binding. Sham-operated male rats displayed significantly greater magnitudes of peak and total analgesia than sham-operated females on the tail-flick test following DAMGO, but not DSLET. Gender differences were not observed for DAMGO and DSLET analgesia on the jump test. Gonadectomy failed to consistently affect either DAMGO or DSLET analgesia. Regression analyses failed to reflect significant shifts in the dose-response functions for either agonist on either measure. Gender differences were not observed for mu1, mu2, or delta binding in hypothalamus or cortex. These data are compared with analgesic responses sensitive to gender differences.


Asunto(s)
Analgésicos Opioides , Encéfalo/efectos de los fármacos , Narcóticos/farmacología , Caracteres Sexuales , Animales , Encéfalo/metabolismo , Encéfalo/fisiología , Encefalina Ala(2)-MeFe(4)-Gli(5) , Encefalina Leucina/análogos & derivados , Encefalina Leucina/farmacología , Encefalinas/farmacología , Femenino , Inyecciones Intraventriculares , Masculino , Morfina/farmacología , Ratas , Ratas Endogámicas , Tiempo de Reacción/efectos de los fármacos , Receptores Opioides/metabolismo , Receptores Opioides delta , Receptores Opioides mu , Vesículas Seminales/anatomía & histología , Umbral Sensorial/efectos de los fármacos , Útero/anatomía & histología
18.
Br J Pharmacol ; 138(5): 921-31, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12642394

RESUMEN

(1) Adrenaline (ADR) and noradrenaline (NA) can simultaneously activate inhibitory alpha(2)- and stimulatory beta-adrenoceptors (AR). However, ADR and NA differ significantly in that ADR is a potent beta(2)-AR agonist while NA is not. Only recently has the interaction resulting from the simultaneous activation of alpha(2)- and beta(2)-AR been examined at the cellular level to determine the mechanisms of alpha(2)-AR regulation following concomitant activation of both alpha(2)- and beta(2)-ARs by chronic ADR. (2) This study evaluates beta(2)-AR regulation of alpha(2A)-AR signalling following chronic ADR (300 nM) and NA (1 and 30 micro M) treatments of BE(2)-C human neuroblastoma cells that natively express both beta(2)- and alpha(2A)-ARs. (3) Chronic (24 h) treatment with ADR (300 nM) desensitized the response to the alpha(2A)-AR agonist, brimonidine, in BE(2)-C cells. Addition of the beta-AR antagonist, propranolol, blocked the ADR-induced alpha(2A)-AR desensitization. Unlike ADR, chronic NA (1 micro M) treatment had no effect on the alpha(2A)-AR response. However if NA was increased to 30 micro M for 24 h, alpha(2A)-AR desensitization was observed; this desensitization was partially reversed by propranolol. (4) Chronic ADR (300 nM) treatment reduced alpha(2A)-AR binding levels, contributing to the alpha(2A)-AR desensitization. This decrease was prevented by addition of propranolol during ADR treatment. Chronic NA (30 micro M), like ADR, treatment lowered specific binding, whereas 1 micro M NA treatment was without effect. (5) Chronic ADR treatment produced a significant increase in GRK3 levels and this was blocked by propranolol or GRK2/3 antisense DNA treatment. This antisense DNA, common to both GRK2 and GRK3, also blocked chronic ADR-induced alpha(2A)-AR desensitization and down-regulation. (6) Acute (1 h) ADR (300 nM) or NA treatment (1 micro M) produced alpha(2A)-AR desensitization. The desensitization produced by acute treatment was beta-AR independent, as it was not blocked by propranolol. (7) We conclude that chronic treatment with modest levels of ADR produces alpha(2A)-AR desensitization by mechanisms that involve up-regulation of GRK3 and down-regulation of alpha(2A)-AR levels through interactions with the beta(2)-AR.


Asunto(s)
Epinefrina/administración & dosificación , Proteínas Serina-Treonina Quinasas/biosíntesis , Receptores Adrenérgicos alfa 2/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/fisiología , Agonistas de Receptores Adrenérgicos alfa 2 , Agonistas de Receptores Adrenérgicos beta 2 , Relación Dosis-Respuesta a Droga , Quinasa 3 del Receptor Acoplado a Proteína-G , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/fisiología , Humanos , Transducción de Señal/fisiología , Células Tumorales Cultivadas , Regulación hacia Arriba/efectos de los fármacos
19.
Brain Res Mol Brain Res ; 105(1-2): 38-46, 2002 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-12399106

RESUMEN

The recently discovered endogenous peptide orphanin FQ/nociceptin (OFQ/N) activates the opioid receptor-like 1 (ORL1) receptor and produces diverse effects on pain perception. In addition to producing spinal analgesia, OFQ/N also exhibits an 'anti-opioid activity' against functional (supraspinal analgesia) and behavioral (conditioned place preference and withdrawal) properties of morphine. One manifestation of the behavioral changes resulting from chronic use of morphine is the upregulation of tyrosine hydroxylase (TH, the rate-limiting enzyme in catecholamine biosynthesis), which contributes to the dramatic increases in catecholamine release in the target regions of the locus coeruleus (LC) and the ventral tegmental area (VTA). The present study sought to determine the molecular mechanism(s) by which OFQ/N modulates the chronic actions of morphine by utilizing human neuroblastoma cell lines [BE(2)-C and SH-SY5Y] that endogenously express TH, and mu and ORL1 receptors. Activation of mu or ORL1 receptors in these cells in turn activates extracellular signal-regulated protein kinases (ERKs), ERK1 and ERK2. Chronic activation of mu, but not ORL1, receptors upregulated TH levels in these cells as previously reported in rat brain. Morphine-induced TH upregulation was blocked upon inclusion of a MEK-1 (mitogen-activated protein kinase kinase-1) inhibitor (PD98059), confirming the role for ERKs in this adaptive response to morphine. Inclusion of OFQ/N during chronic morphine exposure also blocked morphine-induced TH upregulation. Furthermore, chronic OFQ/N exposure increased levels of the TH gene repressor, Oct-2, irrespective of the presence or absence of morphine. This report suggests a potential role for Oct-2 in mediating the anti-opioid actions of OFQ/N against the behavioral manifestations resulting from chronic use of morphine.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Dependencia de Morfina/enzimología , Morfina/antagonistas & inhibidores , Péptidos Opioides/farmacología , Tirosina 3-Monooxigenasa/antagonistas & inhibidores , Regulación hacia Arriba/efectos de los fármacos , Encéfalo/fisiopatología , Catecolaminas/metabolismo , Enfermedad Crónica , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Morfina/metabolismo , Dependencia de Morfina/fisiopatología , Neuronas/efectos de los fármacos , Neuronas/enzimología , Factor 2 de Transcripción de Unión a Octámeros , Péptidos Opioides/metabolismo , Receptores Opioides/efectos de los fármacos , Receptores Opioides/metabolismo , Receptores Opioides mu/efectos de los fármacos , Receptores Opioides mu/metabolismo , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas , Tirosina 3-Monooxigenasa/metabolismo , Regulación hacia Arriba/fisiología , Receptor de Nociceptina , Nociceptina
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda