Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters

Publication year range
1.
Br J Anaesth ; 115(3): 457-62, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26205902

ABSTRACT

BACKGROUND: Peripheral nerve catheters (PNCs) are used with increasing frequency in children. Although adult studies have demonstrated safety with this technique, there have been few safety studies in children. The main objective of the current investigation was to examine the incidence of PNC complications in children undergoing surgery. METHODS: This is an observational, multi-institutional study using the Pediatric Regional Anesthesia Network (PRAN) database. Data pertaining to PNCs were entered prospectively into a secure, online database by each participating centre. Patient characteristics, anatomic location, localization techniques, medications used, and complications were recorded for each catheter. All complications and any sequelae were followed until resolution. RESULTS: There were 2074 PNCs included in the study. 251 adverse events and complications were recorded, resulting in an overall incidence (95% CI) of complications of 12.1% (10.7-13.5%). The most common complications were catheter malfunction, block failure, infection, and vascular puncture. There were no reports of persistent neurologic problems, serious infection, or local anaesthetic systemic toxicity, resulting in an estimated incidence (95% CI) of 0.04% (0.001-0.2%). Patients who developed an infection had used the catheters for a greater number of days, median (IQR) of 4.5 (3-7) days compared with 3 (1-3) days in the patients who did not develop an infection, P<0.0001. CONCLUSIONS: Our data support the safety of placing PNCs in children, with adverse event rates similar to adult studies. Catheter problems are common, yet minor, in severity.


Subject(s)
Anesthesia, Conduction/adverse effects , Anesthesia, Conduction/statistics & numerical data , Nerve Block/adverse effects , Nerve Block/statistics & numerical data , Peripheral Nerves , Practice Patterns, Physicians'/statistics & numerical data , Adolescent , Bacterial Infections/epidemiology , Catheters/adverse effects , Child , Databases, Factual , Equipment Failure , Female , Humans , Male , Prospective Studies , Time Factors
2.
J Minim Invasive Gynecol ; 22(6S): S117-S118, 2015.
Article in English | MEDLINE | ID: mdl-27678608
4.
Science ; 228(4703): 1106-8, 1985 May 31.
Article in English | MEDLINE | ID: mdl-2986292

ABSTRACT

Cerebrospinal fluid taken from rats subjected to electroshock-induced seizures and injected into the cerebral ventricles of rats that had not been shocked increased the seizure threshold of the recipients. The anticonvulsant activity of the donor cerebrospinal fluid was antagonized by opioid antagonists and enhanced by peptidase inhibitors. These results suggest the existence of an endogenous anticonvulsant substance in rat cerebrospinal fluid, possibly opioid in nature, which is activated as a consequence of a seizure and which may play a critical role in postseizure inhibition.


Subject(s)
Anticonvulsants/cerebrospinal fluid , Seizures/cerebrospinal fluid , Animals , Electroshock , Enkephalin, Leucine/analogs & derivatives , Enkephalin, Leucine/pharmacology , Male , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Peptide Hydrolases , Rats , Rats, Inbred Strains , Receptors, Opioid/drug effects , Receptors, Opioid, delta
5.
Science ; 227(4694): 1580-3, 1985 Mar 29.
Article in English | MEDLINE | ID: mdl-3975627

ABSTRACT

The blood-brain barrier restricts the passage of molecules from the blood to the brain. The permeability of the barrier to iodine-125-labeled bovine serum albumin was examined in rats that had undergone adrenalectomy, adrenal demedullation, and corticosterone replacement. Adrenalectomy, but not adrenal demedullation, increased the permeability of brain tissue to the isotopically labeled macromolecule; corticosterone replacement reversed this effect. These results indicate that the blood-brain barrier may be hormonally regulated; that is, the pituitary-adrenal axis may physiologically modulate the permeability of the brain microvasculature to macromolecules.


Subject(s)
Adrenal Cortex/physiology , Blood-Brain Barrier , Adrenal Medulla/physiology , Adrenalectomy , Animals , Blood Pressure , Central Nervous System/physiology , Corticosterone/blood , Corticosterone/physiology , Male , Pituitary-Adrenal System/physiology , Rats , Rats, Inbred Strains , Serum Albumin, Bovine/metabolism
6.
Neuron ; 9(3): 465-78, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1524827

ABSTRACT

Nerve growth factor (NGF) appears to act as a neurotrophic factor for basal forebrain and caudate-putamen cholinergic neurons. The mechanism by which NGF transduces its signal in these neurons is yet to be defined. Recent data indicate that the product of the trk gene, p140trk, is a critical component of the NGF receptor. Herein, we show that p140trk mRNA is highly restricted in its distribution in the adult rat forebrain, that it is present in cholinergic neurons, and that most if not all cholinergic neurons contain p140trk mRNA. Furthermore, induction of trk expression by NGF suggests that neurotrophin-mediated up-regulation of their receptor tyrosine kinases is an important feature of their actions and that neurotrophins may regulate the activity of responsive neurons through increasing the level of their receptors.


Subject(s)
Gene Expression Regulation , Nerve Growth Factors/physiology , Neurons/physiology , Prosencephalon/physiology , Proto-Oncogene Proteins/genetics , RNA, Messenger/metabolism , Animals , Caudate Nucleus/metabolism , Choline O-Acetyltransferase/genetics , Gene Expression Regulation/drug effects , Nerve Growth Factors/pharmacology , Neurons/metabolism , Putamen/metabolism , Receptor, trkA , Tissue Distribution
7.
J Hosp Infect ; 60(1): 19-26, 2005 May.
Article in English | MEDLINE | ID: mdl-15823652

ABSTRACT

A total of 85 Pseudomonas aeruginosa isolates were obtained from October 1999 to April 2000 in a tertiary care hospital in Rio de Janeiro, Brazil. The imipenem susceptibility was evaluated by disk diffusion and agar dilution methods, and the clonal relationship among 67 isolates was examined by macrorestriction profile analysis following pulsed-field gel electrophoresis. Imipenem resistance was observed in 52 (61.2%) isolates. Imipenem-resistant P. aeruginosa isolates were separated into 10 genotypes, 73% of which belonged to genotype A. Identification of a single P. aeruginosa clone with a high rate of imipenem resistance emphasizes the need to control the transmission of this organism among patients.


Subject(s)
Cross Infection/microbiology , Drug Resistance, Bacterial , Imipenem , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa , Brazil/epidemiology , Colony Count, Microbial , Cross Infection/epidemiology , Cross Infection/prevention & control , Cross Infection/transmission , DNA, Bacterial/analysis , DNA, Bacterial/genetics , Drug Resistance, Bacterial/genetics , Electrophoresis, Gel, Pulsed-Field , Genetic Variation/genetics , Genotype , Health Services Needs and Demand , Hospitals, Military , Hospitals, University , Humans , Immunodiffusion , Infection Control/methods , Microbial Sensitivity Tests , Molecular Epidemiology , Phylogeny , Pseudomonas Infections/epidemiology , Pseudomonas Infections/prevention & control , Pseudomonas Infections/transmission , Pseudomonas aeruginosa/classification , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/isolation & purification , Restriction Mapping , Time Factors
8.
Neuropharmacology ; 27(9): 965-74, 1988 Sep.
Article in English | MEDLINE | ID: mdl-2847072

ABSTRACT

Previous studies have demonstrated that chronic administration of morphine up-regulated the lower affinity binding site for [3H][D-ala2,D-leu5]enkephalin, without producing a detectable alteration in the higher affinity binding site for [3H][D-ala2,D-leu5]enkephalin (Rothman et al., Eur. J. Pharmac. 124: 113-119, 1986). The experiments reported in this paper tested the hypothesis that chronic administration of morphine and naltrexone up-regulated the binding sites for [3H][D-ala2,D-leu5]enkephalin by different mechanisms. Rats were given either morphine or naltrexone chronically. Chronic administration of morphine up-regulated the lower affinity site, while chronic administration of naltrexone up-regulated both the higher and lower affinity binding sites for [3H][D-ala2,D-leu5]enkephalin. Unlike the lower affinity binding site for [3H][D-ala2,D-leu5]enkephalin present in membranes prepared from rats treated with placebo pellets, the lower affinity binding sites which were up-regulated by naltrexone and morphine were partially (naltrexone) or completely (morphine) labile to preincubation for 60 min at 25 degrees C in 50 mM Tris-HCl, pH 7.4, containing 0.4 M NaCl. These data suggest that chronic administration of morphine and naltrexone up-regulate binding sites for [3H][D-ala2,D-leu5]enkephalin through different mechanisms, and that the lower affinity binding sites for [3H][D-ala2, D-leu5]enkephalin which are up-regulated by chronic administration of morphine and naltrexone might differ biochemically from the lower affinity binding sites present in membranes treated with placebo.


Subject(s)
Morphine/pharmacology , Naltrexone/pharmacology , Receptors, Opioid/drug effects , Animals , Drug Tolerance , Male , Morphine/administration & dosage , Morphine Dependence , Naltrexone/administration & dosage , Rats , Rats, Inbred Strains , Sodium Chloride/pharmacology
9.
Neuropharmacology ; 28(12): 1349-56, 1989 Dec.
Article in English | MEDLINE | ID: mdl-2559348

ABSTRACT

The series of experiments reported in this paper examined the spectrum of subtypes of opioid receptors alkylated in vitro by N-cyclopropylmethyl-7 alpha-methylfumaramido-6,14- endoethenotetrahydronororipavine (NIH10236) and four optical isomers of the methylfumaramidophenethyl derivatives of 3-methylfentanyl. Pretreatment of membranes with NIH10236 resulted in a wash-resistant inhibition of the binding of [3H]6 beta-fluoro-6-desoxyoxymorphone (mu binding sites), the binding of [3H][D-ala2,D-leu5]-enkephalin (both the higher and lower affinity delta binding sites) and was without effect on kappa binding sites labelled with [3H]bremazocine. All four potential alkylating derivatives of 3-methylfentanyl were inactive. Pretreatment of membranes with 1 microM of the reversible ligands, (+)-cis-3-methylfentanyl, but not its enantiomer, inhibited the binding of [3H]6 beta-fluoro-6-desoxyoxymorphone and the binding of [3H][D-ala2,D-leu5]enkephalin to the lower affinity binding sites by over 90%. This phenomenon is termed "pseudo-irreversible inhibition." Incubation of pretreated membranes for 60 min at 37 degrees C, in the presence of 200 mM NaCl and 50 microM GppNHp, only partially reversed the masking of opioid receptors by (+)-cis-3-methylfentanyl. For in vivo experiments, membranes were prepared 18-24 hr after the intracerebroventricular administration of 80 and 50 micrograms of NIH10236. This resulted in decreased labelling of mu binding sites, lower affinity [3H][D-ala2,D-leu5]enkephalin binding sites, as well as kappa binding sites, labelled by [3H]U69,593 and [3H]bremazocine. There was no apparent alteration in the higher affinity [3H][D-ala2,D-leu5]enkephalin binding site.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Alkylating Agents/metabolism , Receptors, Opioid/metabolism , Thebaine/analogs & derivatives , Animals , In Vitro Techniques , Ligands , Male , Membranes/metabolism , Rats , Rats, Inbred Strains , Receptors, Opioid/classification , Thebaine/metabolism
10.
Neuropharmacology ; 29(9): 805-10, 1990 Sep.
Article in English | MEDLINE | ID: mdl-1963479

ABSTRACT

This study examined the effect of beta-funaltrexamine (beta-FNA), an irreversible mu-receptor antagonist, on naltrexone-induced upregulation of mu-(mu cx + mu nex) and delta nex-opioid receptors. [The subscripts 'cx' and 'nex' denote binding sites 'in' (cx) and 'not in' (nex) the opioid receptor complex.] Rats were treated according to the following protocol. Two naltrexone or two placebo pellets were implanted subcutaneously in a nylon mesh on day 1. and were removed intact on day 8. Rats were given either saline or 20 nmol of beta-FNA in 10 microliters of saline (i.c.v.) on days 1, 3, 5 and 6, 60 min prior to implantation of the pellet. On day 9 frozen lysed-P2 membranes were prepared for assay of mu binding sites. In other experiments, membranes were depleted of mu-receptors by pretreatment with the site-directed acylating agent 2-(4-ethoxybenzyl)-l-diethylaminoethyl-5-isothiocyanatobenzimid azole.HCl (BIT) for assay of delta nex binding sites, using [3H] [D-ala2, D-leu5]enkephalin. The results demonstrated that beta-FNA did not upregulate the mu binding sites and also did not prevent naltrexone-induced upregulation of mu binding sites. Both beta-FNA and naltrexone increased the Bmax of delta nex binding sites and their effects were additive. These data suggest that the mechanism(s) responsible for antagonist-induced upregulation of opioid receptors are more complex than previously appreciated.


Subject(s)
Isothiocyanates , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Receptors, Opioid/metabolism , Up-Regulation/drug effects , Animals , Brain/drug effects , Brain/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Enkephalin, Leucine-2-Alanine/pharmacology , Enkephalins/pharmacology , In Vitro Techniques , Kinetics , Oxymorphone/analogs & derivatives , Oxymorphone/pharmacology , Rats , Rats, Inbred Strains , Receptors, Opioid, mu , Thiocyanates
11.
J Med Chem ; 32(6): 1392-8, 1989 Jun.
Article in English | MEDLINE | ID: mdl-2542560

ABSTRACT

Recently we reported the synthesis of the first enantiomeric pair of irreversible opioid ligands [(3S,4R)-(-)- and (3R,4S)-(+)-cis-4, SUPERFIT] and specific interaction of the latter with the delta receptor. Here we report another enantiomeric pair of irreversible opioid ligands, (+)-trans- and (-)-trans-3-methylfentanyl isothiocyanates [(3S,4S)-(+)-trans- and (3R,4R)-(-)-trans-4]. A single-crystal X-ray analysis of the 2,4,6-trinitrobenzenesulfonic acid salt of (+)-trans-3-methyl-N-phenyl-4-piperidinamine [(+)-trans-8] revealed it (and, therefore, 4) to have the trans configuration and the absolute configuration of (+)-trans-8 to be 3S,4S. The (+)-trans enantiomer of 4 was shown to be highly potent and about 10-fold more selective as an acylating agent than (-)-trans-4 for the higher affinity [3H]DADL (delta) binding site in rat brain membranes. In that assay, (+)-trans-4 and (+)-cis-4 were essentially equipotent as affinity ligands, and the levo enantiomers were considerably less potent. (+)-trans-4 was, thus, a potent, subtype-selective acylating agent for the delta opioid receptor in vitro. With membranes from NG108-15 neuroblastoma x glioma hybrid cells, containing only delta receptors, (+)-cis-4 was found to be a little more potent than (+)-trans-4. Similarly, (+)-cis-4 is the most effective inhibitor of adenylate cyclase in these membranes, (+)-trans-4 has weak activity, and the levo enantiomers are inactive. Only (+)-cis-4 was found to have antinociceptive activity in vivo.


Subject(s)
Fentanyl/analogs & derivatives , Receptors, Opioid/metabolism , Acylation , Adenylyl Cyclase Inhibitors , Analgesia , Animals , Brain/metabolism , Cell Membrane/metabolism , Chemical Phenomena , Chemistry , Enkephalin, Leucine/analogs & derivatives , Enkephalin, Leucine/metabolism , Enkephalin, Leucine/pharmacology , Enkephalin, Leucine-2-Alanine , Enkephalins/metabolism , Fentanyl/chemical synthesis , Fentanyl/metabolism , Fentanyl/pharmacology , Glioma/metabolism , Hybrid Cells , Indicators and Reagents , Ligands , Male , Molecular Structure , Neuroblastoma/metabolism , Rats , Rats, Inbred Strains , Receptors, Opioid, delta , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured
12.
J Neurotrauma ; 13(3): 149-62, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8965324

ABSTRACT

Laser-Doppler flowmetry (LDF) was used to record subcortical cerebral blood flow in hippocampus and striatum immediately following parasaggital fluid percussion brain injuries of mild to moderate severity (2.58 +/- 0.09 atm, 10-11 msec duration) in spontaneously breathing anesthetized rats. At 5 min postinjury, mean blood flow decreased bilaterally by 20-30% in both brain structures, and remained significantly reduced during the remainder of the 60 min postinjury recording interval. Blood flow did not change in the sham-injured rats. Subsequent beam-walk, beam-balance, and rope-hang assessments revealed significant neurological impairments in the injured rats but not in the sham controls. The magnitude of the blood flow changes and the severity of the ensuing neurological impairment were significantly correlated. Histopathological assessments revealed hemorrhagic contusions within ipsilateral cortical regions, occasional neuronal necrosis within underlying thalamus and CA3 and CA4 sectors of the hippocampus, and neuronal cell loss in the hilus of the dentate gyrus. In a second series of experiments, radiolabeled microspheres were used to validate the LDF blood flow measurements. The microsphere measurements revealed that the preinjury baseline and postinjury right hippocampal blood flow changes were not significantly altered by the intrahippocampal presence of an LDF probe, verifying that the LDF probe was not by itself an unacceptably disruptive influence on local cerebrovascular reactivity. Moreover, when right hippocampal blood flow was simultaneously evaluated in injured rats by both techniques, the relative blood flow changes were significantly correlated. These results indicate that laser-Doppler flowmetry provides a potentially useful means to appreciate acute regional cerebrovascular changes relative to other measures of outcome after brain trauma.


Subject(s)
Brain Injuries/physiopathology , Animals , Blood Flow Velocity , Blood Pressure/physiology , Brain Injuries/diagnostic imaging , Cerebrovascular Circulation/physiology , Laser-Doppler Flowmetry , Male , Rats , Rats, Sprague-Dawley , Time Factors , Ultrasonography
13.
J Neurotrauma ; 17(8): 679-93, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10972244

ABSTRACT

The purpose of this study was to determine the impact of secondary hypoxemia on visual discrimination accuracy after parasagittal fluid percussion injury (FPI). Rats lived singly in test cages, where they were trained to repeatedly execute a flicker-frequency visual discrimination for food. After learning was complete, all rats were surgically prepared and then retested over the following 4-5 days to ensure recovery to presurgery levels of performance. Rats were then assigned to one of three groups [FPI + Hypoxia (IH), FPI + Normoxia (IN), or Sham Injury + Hypoxia (SH)] and were anesthetized with halothane delivered by compressed air. Immediately after injury or sham injury, rats in groups IH and SH were switched to a 13% O2 source to continue halothane anesthesia for 30 min before being returned to their test cages. Anesthesia for rats in group IN was maintained using compressed air for 30 min after injury. FPI significantly reduced visual discrimination accuracy and food intake, and increased incorrect choices. Thirty minutes of immediate posttraumatic hypoxemia significantly (1) exacerbated the FPI-induced reductions of visual discrimination accuracy and food intake, (2) further increased numbers of incorrect choices, and (3) delayed the progressive recovery of visual discrimination accuracy. Thionine stains of midbrain coronal sections revealed that, in addition to the loss of neurons seen in several thalamic nuclei following FPI, cell loss in the ipsilateral dorsal lateral geniculate nucleus (dLG) was significantly greater after FPI and hypoxemia than after FPI alone. In contrast, neuropathological changes were not evident following hypoxemia alone. These results show that, although hypoxemia alone was without effect, posttraumatic hypoxemia exacerbates FPI-induced reductions in visual discrimination accuracy and secondary hypoxemia interferes with control of the rat's choices by flicker frequency, perhaps in part as a result of neuronal loss and fiber degeneration in the dLG. These results additionally confirm the utility of this visual discrimination procedure as a sensitive, noninvasive means of assessing behavioral function after experimental traumatic brain injury.


Subject(s)
Brain Injuries/physiopathology , Discrimination Learning , Geniculate Bodies/pathology , Hypoxia/physiopathology , Neurons/pathology , Visual Perception , Animals , Brain Injuries/psychology , Cell Count , Darkness , Discrimination Learning/physiology , Light , Male , Rats , Rats, Sprague-Dawley , Visual Perception/physiology
14.
Neuroreport ; 8(2): 395-8, 1997 Jan 20.
Article in English | MEDLINE | ID: mdl-9080414

ABSTRACT

Halothane-anesthetized male rats were subjected to either moderately severe parasagittal fluid percussion-induced traumatic brain injury (TBI) or sham injury, and for 30 min immediately after injury hypoxia was induced in half the rats from each group by substituting a 13% O2 source to deliver halothane for continued anesthesia. At 60 min post-TBI, Northern blot analysis showed a significant increase in c-fos mRNA levels, by 60-100% above sham control levels in the frontal cortex, cerebellum and hippocampus. Although hypoxia in sham-injured rats did not by itself alter c-fos mRNA levels, it did significantly potentiate the TBI-induced changes in c-fos mRNA in all three brain regions. These findings show that hypoxia is an important factor influencing genomic responses to TBI.


Subject(s)
Brain Injuries/metabolism , Hypoxia/physiopathology , Proto-Oncogene Proteins c-fos/metabolism , Animals , Disease Models, Animal , Male , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
15.
Arch Surg ; 123(6): 700-4, 1988 Jun.
Article in English | MEDLINE | ID: mdl-2967070

ABSTRACT

This study evaluated the effects of naloxone hydrochloride in the treatment of Escherichia coli-induced shock in baboons. The baboons were studied for 12 hours and monitored for survival times. All baboons were intravenously infused for two hours with E coli and treated as follows: group 1, E coli (control); group 2, E coli plus naloxone hydrochloride, 0.5 mg/kg bolus plus 0.5 mg/kg/h for 9.5 hours; and group 3, E coli plus naloxone hydrochloride, 2.0 mg/kg bolus plus 2.0 mg/kg/h for 3.8 hours. Naloxone was administered after arterial pressure had reached the nadir (more than two hours following initiation of E coli infusion). Mean arterial pressure was supported by the lower dose of naloxone; however, sustained leukopenia and neutropenia were not reversed by its infusion. Naloxone prevented the increase in plasma beta-endorphin level and blunted the increase in plasma cortisol level. Despite these effects, naloxone did not prevent multiple-organ disease and did not decrease mortality.


Subject(s)
Escherichia coli Infections/drug therapy , Naloxone/therapeutic use , Papio , Sepsis/drug therapy , Shock, Septic/drug therapy , Animals , Blood Pressure/drug effects , Blood Urea Nitrogen , Creatinine/blood , Disease Models, Animal , Drug Evaluation, Preclinical , Escherichia coli Infections/blood , Escherichia coli Infections/mortality , Female , Heart Rate/drug effects , Hydrocortisone/blood , Injections, Intravenous , Male , Monitoring, Physiologic , Naloxone/administration & dosage , Naloxone/pharmacology , Sepsis/blood , Sepsis/mortality , Shock, Septic/blood , Shock, Septic/mortality , Time Factors , beta-Endorphin/blood
16.
Peptides ; 14(6): 1271-7, 1993.
Article in English | MEDLINE | ID: mdl-8134310

ABSTRACT

Phe-Leu-Phe-Gln-Pro-Gln-Arg-Phe-NH2 (NPFF), an endogenous mammalian antiopioid peptide, has been shown by other laboratories to attenuate the acute antinociceptive effects of morphine, the development of morphine tolerance, and naloxone-induced withdrawal in morphine-dependent rats. The present study determined the effect of chronic NPFF on mu opioid receptors and mRNA for the endogenous opioids dynorphin and enkephalin. Rats received ICV infusions of either saline or NPFF (5 micrograms/h) for 13 days via Alzet 2002 osmotic minipumps. Homogenate binding studies, which used whole brain membranes, demonstrated that NPFF decreased the Bmax of mu binding sites (labeled by [3H][D-Ala2-MePhe4,Gly-ol5]enkephalin) from 262 +/- 12 to 192 +/- 12 fmolmg protein, and increased the Kd from 1.1 to 2.3 nM. Quantitative receptor autoradiography and in situ hybridization experiments were conducted with sections collected at the level of the striatum. The density of mu opioid binding sites labeled by [3H][D-Ala2-MePhe4,Gly-ol5]enkephalin was decreased in all brain areas measured except the corpus callosum, and there was no change in dynorphin mRNA or enkephalin mRNA in the caudate, the nucleus accumbens, or the ventral pallidum. Rats chronically administered ICV morphine sulfate (20 micrograms/h) for 14 days developed tolerance to morphine and a low degree of dependence, as measured by naloxone-precipitated withdrawal. Chronic administration of NPFF concurrently with morphine sulfate did not significantly alter naloxone-induced withdrawal signs or the development of morphine tolerance.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Oligopeptides/pharmacology , Receptors, Opioid, mu/drug effects , Amino Acid Sequence , Animals , Cerebral Ventricles , Down-Regulation , Drug Tolerance , Endorphins/genetics , Infusions, Parenteral , Male , Molecular Sequence Data , Morphine/pharmacology , Morphine Dependence/physiopathology , Oligopeptides/administration & dosage , RNA, Messenger/drug effects , Rats , Rats, Sprague-Dawley , Time Factors
17.
Peptides ; 9(6): 1335-44, 1988.
Article in English | MEDLINE | ID: mdl-3247252

ABSTRACT

Intrathecal (IT) injection of arginine vasopressin (AVP) in rats caused a transient (less than 30 min), dose-related paralysis of the hindlimbs, loss of hindlimb and tail nociceptive responsiveness, and increased mean arterial pressure. Motor dysfunction was produced with comparable potency by lysine vasopressin (LVP) and arginine vasotocin (AVT); oxytocin (OXY) was approximately 1000 times less potent. Paralysis induced by these peptides was selectively blocked following IT pretreatment with 0.5 nmoles of the vasopressin V1 receptor antagonist [1-(beta-mercapto-beta,beta-cyclopentamethylene propionic acid), 2-(O-methyl)tyrosine] Arg8-vasopressin (d(CH2)5[Tyr(Me2)]AVP). Pressor and antinociceptive responses to AVP were also blocked by this compound. However, at higher doses (2-5 nmoles, IT), d(CH2)5[Tyr(Me2)]AVP produced hindlimb paralysis, antinociception, and pressor responses by itself. In contrast to the fiber degeneration, cell loss, and necrosis found in lumbosacral cords of rats persistently paralyzed by other peptides (dynorphin A, somatostatin, and ICI 174864), neuropathological changes were not evident in spinal cords of rats transiently paralyzed by IT AVP. These results indicate that AVP-related peptides affected diverse spinal cord functions through interactions with a V1-like receptor. The similar pattern of cardiovascular and antinociceptive responses to other peptides (dynorphin A, somatostatin, and ICI 174864), which also caused hindlimb paralysis, suggests that the former responses may actually reflect the nonselective consequences of a peptide-induced disruption of spinal cord function, rather than specific shared pharmacological effects.


Subject(s)
Arginine Vasopressin/toxicity , Motor Activity/drug effects , Paralysis/chemically induced , Spinal Cord/physiology , Animals , Arginine Vasopressin/administration & dosage , Arginine Vasopressin/pharmacology , Hindlimb , Injections, Spinal , Lypressin/pharmacology , Male , Oxytocin/pharmacology , Pain/physiopathology , Rats , Rats, Inbred Strains , Reference Values , Spinal Cord/drug effects , Structure-Activity Relationship , Subarachnoid Space , Vasotocin/pharmacology
18.
Peptides ; 8(6): 1089-92, 1987.
Article in English | MEDLINE | ID: mdl-2894646

ABSTRACT

The short-term cardiovascular effects of dynorphin A (1-13), as well as its effects upon morphine bradycardia were investigated. In unanesthetized, unrestrained rats, intracerebroventricular (ICV) dynorphin A (1-13) injections (10-20 micrograms) produced a dose-related pressor effect, whereas intravenous (IV) dynorphin A (1-13) (1.0 mg/kg) produced a depressor effect; these responses persisted less than five min. Heart rate was not significantly altered by these doses or routes of administration. Dynorphin A (1-13) also produced behavioral effects in the unanesthetized animals, such as wet dog shakes in response to IV administration and wet dog shakes accompanied by barrel rolling in response to ICV administration. To evaluate the effects of dynorphin A (1-13) pretreatment on the bradycardic response to IV morphine, rats were pretreated with 10 micrograms dynorphin A (1-13) ICV four, six or eight hours prior to challenge with morphine sulfate (0.1 mg/kg IV). Four hour pretreatment with dynorphin A (1-13) (tested at 14:00 hr) resulted in a potention of morphine bradycardia, with six hours pretreatment (tested at 16:00 hr) no effect was observed, and eight hours following dynorphin A (1-13) pretreatment (tested at 18:00 hr) morphine bradycardia was attenuated. Additionally, the bradycardic response to IV morphine alone became more exaggerated as rats approached their nocturnal activity cycle. These data further establish that dynorphin A (1-13) exerts a potent, long lasting modulatory effect on morphine bradycardia and emphasize the importance of circadian variables in altering the magnitude of cardiovascular responses to opioid agonists.


Subject(s)
Blood Pressure/drug effects , Dynorphins/pharmacology , Heart Rate/drug effects , Morphine/pharmacology , Peptide Fragments/pharmacology , Analysis of Variance , Animals , Circadian Rhythm , Male , Rats , Rats, Inbred Strains , Receptors, Opioid/drug effects
19.
Peptides ; 22(12): 2091-7, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11786195

ABSTRACT

EEP is a tripeptide structurally similar to thyrotropin releasing hormone (TRH) and, like TRH, it is found in the mammalian brain. TRH has been found to increase in brain regions after seizures and to be neuroprotective. EEP has also been shown to increase in brain regions following seizure activity. We therefore sought to determine whether the similarities between these two peptides might be extended to include neuroprotection. Both TRH and EEP were found to be neuroprotective in vitro against an excitotoxic insult. Interestingly, the two tripeptides appeared to have different mechanisms of action. Even though EEP was as much as four times more neuroprotective than TRH, its ability to reduce glutamate-stimulated increases in intraneuronal Ca(2+) was about half that of TRH.


Subject(s)
Glutamates/toxicity , Neuroprotective Agents/pharmacology , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/pharmacology , Animals , Pyrrolidonecarboxylic Acid/analogs & derivatives , Rats
20.
Peptides ; 12(1): 151-60, 1991.
Article in English | MEDLINE | ID: mdl-1646998

ABSTRACT

Studies conducted after the development of the rapid filtration assay for opiate receptors, and before the recognition of multiple opioid receptors, failed to detect changes in opioid receptors induced by chronic morphine. Recent experiments conducted in our laboratories were designed to examine the hypothesis that only one of several opioid receptor types might be altered by chronic morphine. Using binding surface analysis and irreversible ligands to increase the "resolving power" of the ligand binding assay, the results indicated that chronic morphine increased both the Bmax and Kd of the opioid receptor complex, labeled with either [3H][D-Ala2,D-Leu5]enkephalin, [3H][D-Ala2-MePhe4,Gly-ol5]enkephalin or [3H]6-desoxy-6 beta-fluoronaltreone. In the present study rats were pretreated with drugs known to attenuate the development of tolerance and dependence [the irreversible mu-receptor antagonist, beta-funaltrexamine (beta-FNA), and the inhibitor of tryptophan hydroxylase, para-chlorophenylalanine], prior to subcutaneous implantation of morphine pellets. The results demonstrated that 1) unlike chronic naltrexone, beta-FNA failed to upregulate opioid receptors and 2) both beta-funaltrexamine and PCPA pretreatment attenuated the chronic morphine-induced increase in the Bmax, but not the Kd, of the opioid receptor complex. These results provide evidence that naltrex-one-induced upregulation of the opioid receptor complex might occur indirectly as a consequence of interactions at beta-funaltrexamine-insensitive opioid receptors and that morphine-induced upregulation (increased Bmax) of the opioid receptor complex is a relevant in vitro marker related to the development of tolerance and dependence. These data collectively support the hypothesis that endogenous antiopiate peptides play an important role in the development of tolerance and dependence to morphine.


Subject(s)
Morphine Dependence/metabolism , Morphine/pharmacology , Receptors, Opioid/drug effects , Amino Acid Sequence , Animals , Biomarkers/chemistry , Drug Tolerance/physiology , Male , Molecular Sequence Data , Morphine Dependence/prevention & control , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Radioligand Assay , Rats , Rats, Inbred Strains , Receptors, Opioid, delta , Receptors, Opioid, mu , Time Factors , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL