Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Indoor Air ; 19(1): 45-57, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19191927

ABSTRACT

UNLABELLED: The purpose of this study was to measure and evaluate the impact of the emissions of selected products of exotic wood on health. Ten products were screened for chemical compounds, and five of the most used products which emitted more than 800 microg/kg were selected for further quantitative analyses by climate chamber measurement (iroko, ramin, sheesham, merbau, and rubber tree). Samples of exotic wood (rubber tree and belalu) were further analyzed for emission of chemical compounds by migration into artificial saliva and for content of pesticides and allergenic natural rubber latex (NR latex) (rubber tree). The toxicological effects of all substances identified were evaluated and the lowest concentrations of interest (LCI) assessed. An R-value was calculated for each wood product (R-value below 1 is considered to be unproblematic as regards health). Emission from the evaluated exotic wood only took place to a very limited extent. None of the selected products, under the chosen rating system, is likely to cause adverse health effects. Products with surface treatment might pose a problem if used as kitchen utensils, as children's toys, or when they are in close contact with the skin for a long time. PRACTICAL IMPLICATIONS: The authors investigated the chemical emissions from selected products from exotic wood by climate chamber measurement. Quantitative chemical analyses of emissions from the five most used exotic products in Denmark were performed, and all chemical compounds found were evaluated toxicologically. Emission from the evaluated exotic wood was very limited. None of the products is likely, under our exposure conditions, to cause health problems in relation to indoor air.


Subject(s)
Pesticide Residues/analysis , Volatile Organic Compounds/toxicity , Wood/chemistry , Air Pollution, Indoor/analysis , Humans , Saliva, Artificial/chemistry , Volatile Organic Compounds/analysis
2.
J Appl Physiol (1985) ; 95(5): 1873-82, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12871965

ABSTRACT

Peripheral administration of a variety of inflammatory stimuli, such as endotoxin or cytokines, induces an orchestrated set of brain-mediated events referred to as the sickness response. The mechanism for how immune products signal the brain is not clear, but accumulating evidence supports the existence of neural as well as blood-borne pathways. Although endotoxin or cytokine administration results in sickness responses, few data exist regarding the role of circulating endotoxin or cytokines in the induction of sickness during a real bacterial infection. Thus the present studies examined whether subcutaneously administered Escherichia coli can activate sickness responses and whether circulating endotoxin and/or proinflammatory cytokines are a prerequisite for these responses. Male Sprague-Dawley rats were injected subcutaneously with one of three doses (2.5 x 10(7), 2.5 x 10(8), 2.5 x 10(9) colony-forming units) of replicating E. coli, a ubiquitous bacterial strain, or vehicle. Core body temperature (Tc) and activity were measured for 3 days after the injection. A second set of groups of animals were killed 3, 6, 12, 18, 24, and 48 h after the injection, and blood samples and brains were collected. Injections dose dependently and consistently increased Tc and decreased activity, with increases in Tc beginning 4 h after the injection. In addition, E. coli significantly increased serum interleukin (IL)-1beta, IL-6, and tumor necrosis factor-alpha and brain IL-1beta levels beginning at the 6-h time point. Corticosterone and endotoxin were first elevated in the circulation at 3 and 18 h after the injection, respectively. Because fever onset preceded brain cytokine induction, we also examined cytokine levels in the serum, brain, and inflammation site 2 and 4 h after injection. Cytokines were elevated at the inflammation site but were not detectable in the serum or brain at 2 and 4 h. We conclude that subcutaneous injection of replicating E. coli induces a consistent and naturalistic infection that includes features of the sickness response as well as increases in circulating, brain, and inflammation site tissue cytokines. In addition, injection of replicating E. coli produces a robust fever and corticosterone response at a time when there are no detectable increases in circulating cytokines or endotoxin. These results suggest that elevated levels of circulating cytokines and endotoxin are not necessary for the activation of the sickness or corticosterone response. Therefore, fever, activity reduction, and corticosterone elevation induced by E. coli infection may have been evoked by a neural, rather than a humoral, pathway from the periphery to the brain.


Subject(s)
Corticosterone/blood , Escherichia coli Infections/immunology , Interleukin-1/immunology , Neuroimmunomodulation/physiology , Tumor Necrosis Factor-alpha/immunology , Animals , Brain/immunology , Brain/microbiology , Escherichia coli Infections/blood , Fever/immunology , Fever/microbiology , Interleukin-1/metabolism , Lipopolysaccharides/blood , Male , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism
3.
J Vet Pharmacol Ther ; 26(2): 95-103, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12667178

ABSTRACT

Metomidate was administered to halibut (Hippoglossus hippoglossus) and turbot (Scophthalmus maximus) intravenously at a dose of 3 mg/kg bodyweight, as a bath treatment at a dose of 9 mg/L water for 5 min to study the disposition of metomidate, and as bath treatment (9 mg/L) for 10 min to study the absorption and effect of metomidate on respiration and balance/motor control. Additionally, turbot were given metomidate orally at a dose of 7 mg/kg. The studies were performed in seawater at a temperature of 10.3 +/- 0.4 degrees C (halibut) and 18.0 +/- 0.3 degrees C (turbot). Pharmacokinetic modeling of the data showed that metomidate had shorter elimination half-life and higher plasma concentrations in turbot compared with halibut, both species displaying a rapid uptake, distribution and excretion. Following intravenous administration, the volumes of distribution at steady state (Vd(ss)) were 0.21 L/kg (halibut) and 0.44 L/kg (turbot). Plasma clearances (Cl) were 0.099 L/h.kg in halibut and 0.26 L/h.kg in turbot and the elimination half-lives (t(1/2)lambdaz) were calculated to be 5.8 h and 2.2 h in halibut and turbot, respectively. Mean residence times (MRT) were 2.2 h in halibut and 1.7 h in turbot. Following oral administration, the t(1/2)lambdaz was 3.5 h in turbot. The maximum plasma concentration (Cmax) was 7.8 mg/L in turbot 1 h after administration. The oral bioavailability (F) was calculated to 100% in turbot. Following 5 min bath the maximum plasma concentrations (Cmax), which were observed immediately after end of the bath, were 9.5 mg/L and 13.3 mg/L in halibut and turbot, respectively. Metomidate rapidly immobilized the fish, with respiratory depression, reduced heart rate, and loss of balance/motor control within 1 min (mean). Recovery was slow, with resumed balance/motor control after 26.4 min. Opercular respiration movements were resumed more rapidly with a recorded mean of 1.7 min. Oral administration was demonstrated to be a way of immobilizing fish, for example in large aquariums, without exposing them to unwanted stress.


Subject(s)
Anesthetics/pharmacology , Anesthetics/pharmacokinetics , Etomidate/analogs & derivatives , Etomidate/pharmacology , Etomidate/pharmacokinetics , Flatfishes/metabolism , Administration, Oral , Anesthetics/administration & dosage , Anesthetics/blood , Animals , Biological Availability , Etomidate/administration & dosage , Etomidate/blood , Flounder/metabolism , Immersion , Injections, Intravenous/veterinary , Motor Activity/drug effects , Skin Absorption , Species Specificity
4.
Dis Aquat Organ ; 50(3): 161-9, 2002 Jul 29.
Article in English | MEDLINE | ID: mdl-12219972

ABSTRACT

This is the first description of a persistent subclinical nodavirus infection in the Atlantic halibut Hippoglossus hippoglossus. Juvenile fish (1 to 5 g) were sampled at 4, 5 and 8 mo of age at a fish farm in Norway during and after weaning. None showed clinical signs of viral encephalopathy and retinopathy (VER) or other disease. Pathological changes and/or nodavirus were detected by light microscopy, immunohistochemistry, reverse transcriptase polymerase chain reaction (RT-PCR) and transmission electron microscopy in all fish examined. High numbers of virus particles were found in macrophage-like cells in the central nervous system, including brain and retina (CNS). The virus particles displayed the icosahedral shape and size (approximately 25 nm) characteristic of nodaviruses. The virus-infected cells formed focal cell aggregates and were seen in all regions of the brain and all nuclear cell layers of the retina. The cytoplasm of the infected cells was filled with membrane-enclosed inclusions packed with virus particles. Some virus particles lay along membranes and formed membrane-bound necklace-like arrangements. The virus-infected cells of the retina also contained pigment granula located generally inside virus inclusions and sometimes forming a coating around the virus particles. All frontal parts with the eyes and brain and 50% of the mid-parts, which included the abdominal organs, were found positive for nodavirus with RT-PCR. Pathological changes in these persistently nodavirus-infected fish differ from earlier descriptions in Atlantic halibut during outbreaks of VER. Vertical transmission from infected spawners is believed to be a major route for nodavirus infection. Detection of nodavirus in subclinical infected fish and a better understanding of its pathogenesis are important in order to prevent the spread of nodavirus in the fish-farming industry.


Subject(s)
Fish Diseases/pathology , Flounder , Nodaviridae/isolation & purification , RNA Virus Infections/veterinary , Animals , Brain/pathology , Brain/ultrastructure , Brain/virology , Fish Diseases/transmission , Fish Diseases/virology , Fisheries , Immunohistochemistry/veterinary , Infectious Disease Transmission, Vertical/prevention & control , Infectious Disease Transmission, Vertical/veterinary , Microscopy, Electron/veterinary , Nodaviridae/genetics , Norway , RNA Virus Infections/pathology , RNA Virus Infections/transmission , RNA, Viral/analysis , Retina/pathology , Retina/ultrastructure , Retina/virology , Reverse Transcriptase Polymerase Chain Reaction/veterinary
5.
Am J Physiol Regul Integr Comp Physiol ; 280(5): R1434-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11294765

ABSTRACT

The paraventricular nucleus of the hypothalamus (PVH) occupies a pivotal point within the network of brain nuclei coordinating critical host-defense responses. In mice, T cell-dependent immune stimuli, including the bacterial superantigen staphylococcal enterotoxin B (SEB), can activate the PVH. To determine whether T cell-dependent immune stimuli activate the PVH in rats, we assessed plasma corticosterone (Cort) levels, fever responses, and c-Fos expression in the PVH in animals treated with intraperitoneal injections of SEB. In animals with previously implanted abdominal thermisters, intraperitoneal injection of 1 mg/kg SEB resulted in a significant rise in body temperature, with a latency of 3.5-4 h. In separate animals, intraperitoneal injection of 1 mg/kg SEB resulted in a significant elevation of plasma Cort and induced c-Fos expression in parvocellular neurons within the PVH. These results support the idea that T cell-dependent immune stimuli activate brain pathways mediating host-defense responses such as fever and neuroendocrine changes.


Subject(s)
Corticosterone/blood , Enterotoxins/pharmacology , Fever/physiopathology , Neurons/physiology , Paraventricular Hypothalamic Nucleus/physiology , Proto-Oncogene Proteins c-fos/metabolism , Animals , Body Temperature/drug effects , Body Temperature Regulation/drug effects , Enterotoxins/administration & dosage , Fever/chemically induced , Injections, Intraperitoneal , Male , Mice , Neurons/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/physiopathology , Rats , Rats, Sprague-Dawley , Staphylococcus aureus , Superantigens/pharmacology
6.
Am J Physiol Regul Integr Comp Physiol ; 280(4): R929-34, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11247812

ABSTRACT

It has been suggested that proinflammatory cytokines communicate to the brain via a neural pathway involving activation of vagal afferents by interleukin-1beta (IL-1beta), in addition to blood-borne routes. In support, subdiaphragmatic vagotomy blocks IL-1beta-induced, brain-mediated responses such as fever. However, vagotomy has also been reported to be ineffective. Neural signaling would be expected to be especially important at low doses of cytokine, when local actions could occur, but only very small quantities of cytokine would become systemic. Here, we examined core body temperature after intraperitoneal injections of three doses of recombinat human IL-1beta (rh-IL-1beta). Subdiaphragmatic vagotomy completely blocked the fever produced by 0.1 microg/kg, only partially blocked the fever produced by 0.5 microg/kg, and had no effect at all on the fever that followed 1.0 microg/kg rh-IL-1beta. Blood levels of rh-IL-1beta did not become greater than normal basal levels of endogenous rat IL-beta until the 0.5-microg/kg dose nor was IL-1beta induced in the pituitary until this dose. These results suggest that low doses of intraperitoneal IL-1beta induce fever via a vagal route and that dose may account for some of the discrepancies in the literature.


Subject(s)
Body Temperature Regulation/physiology , Brain/physiology , Fever/physiopathology , Interleukin-1/pharmacology , Vagus Nerve/physiology , Animals , Body Temperature/drug effects , Body Temperature Regulation/drug effects , Brain/drug effects , Brain/physiopathology , Diaphragm/innervation , Dose-Response Relationship, Drug , Fever/chemically induced , Humans , Male , Muscle Denervation , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Vagotomy , Vagus Nerve/drug effects , Vagus Nerve/physiopathology
7.
J Pain ; 2(6): 326-33, 2001 Dec.
Article in English | MEDLINE | ID: mdl-14622812

ABSTRACT

Intrathecal administration of the human immunodeficiency virus-1 envelope glycoprotein, gp120, activates astrocytes and microglia to release products that induce thermal hyperalgesia and mechanical allodynia. Both pain states are disrupted by intrathecal CNI-1493, a p38 mitogen-activated protein (MAP) kinase inhibitor. Whether CNI-1493, or any other p38 MAP kinase inhibitor, can cross the blood-brain barrier to affect spinal cord function is unknown. Given that several such drugs are in clinical trials, it is of interest to determine whether they may be potentially useful in treating centrally mediated pain. The aim of the present studies was to determine whether systemic CNI-1493 could block intrathecal gp120-induced thermal hyperalgesia and/or mechanical allodynia. Because p38 MAP kinase inhibition would be expected to prevent proinflammatory cytokine release and/or signal transduction, we sought to determine from the same animals the likely mechanism by which CNI-1493 blocks gp120-induced pain states. These studies show that systemic CNI-1493 blocks intrathecal gp120-induced thermal hyperalgesia and mechanical allodynia. Because CNI-1493 did not block proinflammatory cytokine release, this may suggest disruption at the level of signal transduction. These studies provide the first evidence that systemic p38 MAP kinase inhibitors can prevent centrally mediated exaggerated pain states. Thus, CNI-1493 may provide a novel therapeutic approach for the treatment of pain.

8.
Dis Aquat Organ ; 47(3): 183-91, 2001 Dec 05.
Article in English | MEDLINE | ID: mdl-11804417

ABSTRACT

The absorption, distribution and elimination of 14C-labelled flumequine were studied using whole body autoradiography and liquid scintillation counting. Flumequine was administered to eel Anguilla anguilla, turbot Scophthalmus maximus and halibut Hippoglossus hippoglossus intravenously and orally as a single dose of 5 mg kg(-1), corresponding to 0.1 mCi kg(-1). The turbot and halibut studies were performed in salt water (salinity of 32%) at temperatures of 16 +/- 1 degrees C (turbot) and 9.5 +/- 0.5 degrees C (halibut). The eel study was conducted in fresh water at 23 +/- 1 degrees C. In the intravenously administered groups flumequine was rapidly distributed to all major tissues and organs. After oral administration flumequine also appeared to have rapid and extensive absorption and distribution in all 3 species. After the distribution phase, the level of flumequine was higher in most organs and tissues than in the blood, except in muscle and brain. The most noticeable difference between the species was the slow elimination of flumequine from eel compared to turbot and halibut. In orally administered eels, substantial amounts of flumequine remained in all major organs/tissues for 7 d. At 28 d significant levels of flumequine were present in liver, kidney and skin (with traces in muscle), and at the last sampling point (56 d) in eye, bone, bile and posterior intestine. In orally administered turbot significant levels of flumequine were observed over 96 h in bile, urine, bone, skin, intestine and eye, and traces were detected over 28 d in bone and eye in addition to a significant level in bile. In orally administered halibut, significant levels of flumequine were observed in bile, skin, intestine and eye over 96 h. Traces were present in skin and eye over 7 d. The maximal flumequine concentrations in blood were calculated to be 2.5 mg equivalents l(-1) (eel at 12 h), 0.8 mg l(-1) (turbot at 6 h) and 0.6 mg l(-1) (halibut at 6 h) after oral administration.


Subject(s)
Anguilla/metabolism , Anti-Infective Agents/pharmacokinetics , Flatfishes/metabolism , Flounder/metabolism , Fluoroquinolones , Quinolizines/pharmacokinetics , Absorption , Administration, Oral , Animals , Anti-Infective Agents/administration & dosage , Autoradiography/veterinary , Carbon Isotopes , Injections, Intravenous/veterinary , Quinolizines/administration & dosage , Scintillation Counting/veterinary , Species Specificity , Tissue Distribution
9.
J Peripher Nerv Syst ; 6(3): 111-29, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11817330

ABSTRACT

We have recently developed a model of sciatic inflammatory neuritis (SIN) to assess how immune activation near peripheral nerves influences somatosensory processing. Administration of zymosan (yeast cell walls) around a single sciatic nerve produces dose-dependent low-threshold mechanical allodynia without thermal hyperalgesia. Low (4 microg) doses produce both territorial and extraterritorial allodynia restricted to the injected hindleg. In contrast, higher (40 microg) doses produce territorial and extraterritorial allodynias of both hindlegs, an effect not accounted for by systemic spread of the zymosan. The aim of these experiments was to determine whether these behavioral allodynias were correlated with immunological and/or anatomical changes in or around the sciatic nerve. These experiments reveal that zymosan-induced bilateral allodynia was associated with the following: (a) increased release of both interleukin-1beta and tumor necrosis factor-alpha from peri-sciatic immune cells; (b) increased release of reactive oxygen species from perisciatic immune cells; (c) no change in circulating levels of proinflammatory cytokine; (d) no apparent zymosan-induced influx of immune cells into the sciatic nerve from the endoneurial blood vessels; (e) mild edema of the sciatic, which was predominantly restricted to superficial regions closest to the peri-sciatic immune cells; and (f) no anatomic evidence of changes in either the ipsilateral saphenous nerve or contralateral sciatic nerve that could account for the appearance of extraterritorial or contralateral ("mirror") allodynia, respectively. No reliable differences were found when the low-dose zymosan was compared with vehicle controls. Taken together, these data suggest that substances released by peri-sciatic immune cells may induce changes in the sciatic nerve, leading to the appearance of bilateral allodynia.


Subject(s)
Cytokines/metabolism , Sciatic Neuropathy/immunology , Sciatic Neuropathy/metabolism , Superoxides/metabolism , Animals , Behavior, Animal , Disease Models, Animal , Flow Cytometry , Gelatin Sponge, Absorbable , Immunohistochemistry , Interleukin-1/metabolism , Interleukin-6/metabolism , Leukocytes/cytology , Leukocytes/drug effects , Luminescent Measurements , Lymphocyte Count , Male , Pain Measurement , Rats , Rats, Sprague-Dawley , Sciatic Nerve/immunology , Sciatic Nerve/pathology , Sciatic Neuropathy/chemically induced , Specific Pathogen-Free Organisms , Tumor Necrosis Factor-alpha/metabolism , Wallerian Degeneration/immunology , Zymosan/pharmacology
10.
Arch Environ Health ; 56(5): 419-32, 2001.
Article in English | MEDLINE | ID: mdl-11777023

ABSTRACT

In this study, the authors describe a method for evaluation of material emissions. The study was based on chemical analysis of emissions from 23 materials representing solid wood and wood-based materials commonly used in furniture, interior furnishings, and building products in Denmark in the 1990s. The authors used the emission chamber testing method to examine the selected materials with a qualitative screening and quantitative determination of volatile organic compounds. The authors evaluated the toxicological effects of all substances identified with chamber testing. Lowest concentration of interest and standard room concentrations were assessed, and the authors calculated an S-value for each wood and wood-based material. The authors identified 144 different chemical substances with the screening analyses, and a total of 84 individual substances were quantified with chamber measurements. The irritative effects dominated at low exposure levels; therefore, the lowest concentration of interest and the S-value were based predominantly on these effects. The S-values were very low for solid ash, oak, and beech. For solid spruce and pine, the determining substances for size of the S-value were delta3-carene, alpha-pinene, and limonene. For the surface-treated wood materials, the S-value reflected the emitted substances from the surface treatment.


Subject(s)
Air Pollution, Indoor/analysis , Organic Chemicals/analysis , Wood , Construction Materials , Humans , Interior Design and Furnishings , No-Observed-Adverse-Effect Level , Odorants , Volatilization
11.
J Vet Pharmacol Ther ; 23(3): 163-8, 2000 Jun.
Article in English | MEDLINE | ID: mdl-11110104

ABSTRACT

Knowledge of the pharmacokinetic properties of drugs to combat bacterial infections in cod (Gadus morhua) and wrasse (Ctenolabrus rupestris) is limited. One antimicrobial agent likely to be effective is flumequine. The aim of this study was to investigate the pharmacokinetic properties of flumequine in these two species. Flumequine was administered intravenously to cod (G. morhua) at a dose of 5 mg/kg bodyweight and wrasse (C. rupestris) at a dose of 10 mg/kg. Flumequine was also administered orally to both species at a dose of 10 mg/kg body weight, and as a bath treatment at a dose of 10 mg/L water for 2 h. Identical experimental designs were used otherwise. The study was performed in seawater with a salinity of 3.2% and a temperature of 8.0 +/- 0.2 degrees C (cod) and 14.5 +/- 0.4 degrees C (wrasse). Pharmacokinetic modelling of the data showed that flumequine had quite different pharmacokinetic properties in cod and wrasse. Following intravenous administration, the volumes of distribution at steady-state (Vss) were 2.41 L/kg (cod) and 2.15 L/kg (wrasse). Total body clearances (Cl) were 0.024 L/hxkg (cod) and 0.14 L/hxkg (wrasse) and the elimination half-lives (t1/2lambda z) were calculated to be 75 h (cod) and 31 h (wrasse). Mean residence times (MRT) were 99 h (cod) and 16 h (wrasse). Following oral administration, the t1/2 lambda z were 74 h (cod) and 41 h (wrasse). Maximal plasma concentrations (tmax) were 3.5 mg/L (cod) and 1.7 mg/L (wrasse), and were observed 24 h post-administration in cod and 1 h post-administration in wrasse. The oral bioavailabilities (F) were calculated to be 65% (cod) and 41% (wrasse). Following bath administration, maximal plasma concentrations were 0.13 mg/L (cod) and 0.09 mg/L (wrasse), and were observed immediately after the end of the bath.


Subject(s)
Anti-Infective Agents/pharmacokinetics , Fluoroquinolones , Quinolizines/pharmacokinetics , Administration, Oral , Animals , Anti-Infective Agents/blood , Area Under Curve , Biological Availability , Chromatography, High Pressure Liquid , Fishes , Half-Life , Injections, Intravenous , Metabolic Clearance Rate , Quinolizines/blood , Species Specificity
12.
J Vet Pharmacol Ther ; 23(3): 169-74, 2000 Jun.
Article in English | MEDLINE | ID: mdl-11110105

ABSTRACT

Knowledge of the pharmacokinetic properties of drugs to combat bacterial infections in the European eel (Anguilla anguilla) is limited. One antimicrobial agent likely to be effective is flumequine. The aim of this study was to investigate the pharmacokinetic properties of flumequine in European eels in fresh water. Flumequine was administered to eels (Anguilla anguilla) intravenously (i.v.) and orally (p.o.) at a dose of 10 mg/kg body weight, and as a bath treatment at a dose of 10 mg/L water for 2 h. The study was performed in fresh water with a temperature of 23 + 0.3 degrees C, pH 7.15. Identical experimental designs were used. Two additional bath treatments were also performed, one in which the pH in the water was lowered by approximately 1 unit to 6.07 (dose: 10 mg/L) and one at a dose of 40 mg/L for 2 h in a full-scale treatment. Following i.v. administration, the volume of distribution at steady state was 3.4 L/kg. Total body clearance was 0.012 L/h per kg and the elimination half-life (t1/2lambda z) was calculated to be 314 h. Mean residence time was 283 h. Following oral administration, the t1/2lambda z was 208 h. Maximal plasma concentration (Cmax) was 9.3 mg/L, at 7 h after administration (Cmax). The oral bioavailability (F) was calculated to be 85%. Following bath administration in 10 mg/L for 2 h, maximal plasma concentration was 2.1 mg/L, observed immediately after the end of the bath. The 'bioavailability' in eel following a 2-h bath treatment was 19.8%. Reducing the pH in the bath to 6.07 produced a maximal plasma concentration of 5.5 mg/L, observed immediately after the end of the bath. The 'bioavailability' was increased to 41% by the lowering of the pH. A similar effect was observed in a full-scale treatment (1 kg eels/L water). The CO2 produced by the eel lowered the pH and increased 'bioavailability' to 35%.


Subject(s)
Anti-Infective Agents/pharmacokinetics , Fluoroquinolones , Quinolizines/pharmacokinetics , Administration, Oral , Anguilla , Animals , Anti-Infective Agents/blood , Biological Availability , Chromatography, High Pressure Liquid , Fresh Water , Half-Life , Hydrogen-Ion Concentration , Injections, Intravenous , Metabolic Clearance Rate , Quinolizines/blood
13.
Brain Res ; 859(2): 193-201, 2000 Mar 24.
Article in English | MEDLINE | ID: mdl-10719064

ABSTRACT

Activation of peripheral immune cells leads to increases of interleukin-1beta (IL-1beta) mRNA, immunoreactivity, and protein levels in brain and pituitary. Furthermore, IL-1beta in brain plays a role in mediating many of the behavioral, physiological, and endocrine adjustments induced by immune activation. A similarity between the consequences of immune activation and exposure to stressors has often been noted, but the potential relationship between stress and brain IL-1beta has received very little attention. A prior report indicated that exposure to inescapable tailshocks (IS) raised levels of brain IL-1beta protein 2 h after IS, but only in adrenalectomized (and basal corticosterone replaced) subjects. The studies reported here explore this issue in more detail. A more careful examination revealed that IL-1beta protein levels in hypothalamus were elevated by IS in intact subjects, although adrenalectomy, ADX (with basal corticosterone replacement) exaggerated this effect. IL-1beta protein increases were already present immediately after the stress session, both in the hypothalamus and in other brain regions in adrenalectomized subjects, and no longer present 24 h later. Furthermore, IS elevated levels of IL-1beta protein in the pituitary, and did so in both intact and adrenalectomized subjects. IS also produced increased blood levels of IL-1beta, but only in adrenalectomized subjects. Finally, the administration of corticosterone in an amount that led to blood levels in adrenalectomized subjects that match those produced by IS, inhibited the IS-induced rise in IL-1beta in hypothalamus and pituitary, but not in other brain regions or blood.


Subject(s)
Brain/drug effects , Brain/metabolism , Corticosterone/metabolism , Corticosterone/pharmacology , Interleukin-1/blood , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Stress, Physiological/physiopathology , Time Factors , Adrenalectomy/adverse effects , Animals , Brain/cytology , Male , Pituitary Gland/cytology , Rats , Rats, Sprague-Dawley
14.
Am J Physiol Regul Integr Comp Physiol ; 278(2): R331-6, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10666132

ABSTRACT

The vagus nerve appears to play a role in communicating cytokine signals to the central nervous system, but the exact extent of its involvement in cytokine-to-brain communication remains controversial. Recently, subdiaphragmatic vagotomy was shown to increase bacterial translocation across the gut barrier and thus may cause endotoxin tolerance. The current experiment tested whether or not vagotomized animals have similar systemic responses to endotoxin challenge as do sham-operated animals. Subdiaphragmatically vagotomized and sham-operated animals were injected intraperitoneally with one of three doses (10, 50, 100 microg/kg) of lipopolysaccharide (LPS) or vehicle, and blood samples were taken at 15, 30, 60, 90, and 120 min after the injection. The intraperitoneal injection of LPS increased circulating LPS levels at all time points examined. In addition, all three doses of LPS significantly increased serum interleukin (IL)-1beta, IL-6, and corticosterone in both control and vagotomized rats. In conclusion, vagotomy itself has no marked effect on circulating endotoxin levels or the production of IL-1beta, IL-6, or corticosterone in blood after an intraperitoneal injection of LPS.


Subject(s)
Corticosterone/blood , Cytokines/blood , Endotoxins/blood , Lipopolysaccharides/pharmacology , Vagotomy , Animals , Injections, Intraperitoneal , Interleukin-1/analysis , Lipopolysaccharides/analysis , Liver/chemistry , Male , Peritoneum/chemistry , Rats , Rats, Sprague-Dawley , Therapeutic Irrigation
15.
Auton Neurosci ; 85(1-3): 119-26, 2000 Dec 20.
Article in English | MEDLINE | ID: mdl-11189018

ABSTRACT

The production of interleukin-1beta (IL-1beta) in brain is thought to be a critical step in the induction of central manifestations of the acute phase response, and the vagus nerve has been implicated in immune-to-brain communication. Thus, this study examined the effects of intraperitoneal (i.p.) injections of lipopolysaccharide (LPS) on brain IL-1beta protein levels in control and subdiaphragmatically vagotomized rats. In the first experiment, vagotomized and sham-operated male Sprague-Dawley rats were injected i.p. with one of three doses (10, 50, 100 microg/kg) of LPS or vehicle (sterile, pyrogen-free saline) and sacrificed 2 h after the injection. In the second experiment, vagotomized and sham-operated rats were injected i.p. with 100 microg/kg LPS or vehicle and sacrificed 1 h after the injection. The i.p. injection of LPS dose-dependently increased IL-1beta protein levels in the hypothalamus, hippocampus, dorsal vagal complex, cerebellum, posterior cortex, and pituitary 2 h after the injection. Brain and pituitary IL-1beta levels were also significantly increased 1 h after the injection of 100 microg/kg LPS. There were no significant differences in brain IL-1beta levels between sham-operated and vagotomized rats at either the 2 h or 1 h time points. The current data are consistent with previous studies showing increases in brain IL-1beta after peripheral injections of LPS, and support the notion that brain IL-1beta is a mediator in the illness-induction pathway. Furthermore, these data indicate that, at the doses and times tested, subdiaphragmatic vagal afferents are not crucial for LPS-induced brain IL-1beta protein.


Subject(s)
Brain Chemistry/immunology , Interleukin-1/metabolism , Vagotomy , Vagus Nerve/immunology , Animals , Cerebellum/cytology , Cerebellum/immunology , Cerebellum/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/immunology , Cerebral Cortex/metabolism , Cholecystokinin/pharmacology , Eating/drug effects , Hippocampus/cytology , Hippocampus/immunology , Hippocampus/metabolism , Hypothalamus/cytology , Hypothalamus/immunology , Hypothalamus/metabolism , Injections, Intraperitoneal , Interleukin-1/immunology , Lipopolysaccharides/blood , Lipopolysaccharides/pharmacokinetics , Male , Neuroimmunomodulation/physiology , Pituitary Gland/immunology , Pituitary Gland/metabolism , Rats , Rats, Sprague-Dawley , Vagus Nerve/cytology , Vagus Nerve/surgery
16.
Auton Neurosci ; 85(1-3): 49-59, 2000 Dec 20.
Article in English | MEDLINE | ID: mdl-11189026

ABSTRACT

The immune system operates as a diffuse sensory system, detecting the presence of specific chemical constituents associated with dangerous micro-organisms, and then signalling the brain. In this way, immunosensation constitutes a chemosensory system. Several submodalities of this sensory system function as pathways conveying immune-related information, and can be classified as either primarily brain barrier associated or neural. The vagus nerve provides the major neural pathway identified to date. The initial chemosensory transduction events occur in immune cells, which respond to specific chemical components expressed by dangerous micro-organisms. These immune chemosensory cells release mediators, such as cytokines, to activate neural elements, including primary afferent neurons of the vagal sensory ganglia. Primary afferent activation initiates local reflexes (e.g. cardiovascular and gastrointestinal) that support host defense. In addition, at least three parallel pathways of ascending immune-related information activate specific components of the illness response. In this way, immunosensory systems represent highly organized and coherent pathways for activating host defense against infection.


Subject(s)
Brain/cytology , Brain/immunology , Neuroimmunomodulation/physiology , Neurons, Afferent/immunology , Vagus Nerve/cytology , Vagus Nerve/immunology , Animals , Chemoreceptor Cells/immunology
17.
Auton Neurosci ; 85(1-3): 72-7, 2000 Dec 20.
Article in English | MEDLINE | ID: mdl-11189029

ABSTRACT

Peripheral interleukin-1beta has been implicated in the initiation of fever responses, yet the pathways by which it influences brain function are still unclear. Sectioning the abdominal vagus has been reported to inhibit fever after intraperitoneal administration of interleukin-1beta, suggesting that vagal afferents participate in signaling the brain to mount a fever response to interleukin-1beta. However, the inhibitory effect of subdiaphragmatic vagotomy could be due to alterations in pharmacokinetics such that the intraperitoneally injected cytokine does not reach the general circulation in sufficient quantities to activate the brain via blood-borne signaling. We measured both fever and plasma levels of interleukin-1beta in vagotomized and sham-operated rats after intraperitoneal administration of 1 microg/kg human recombinant interleukin-1beta to determine whether vagotomy reduces fever and levels of circulating interleukin-1beta after intraperitoneal injection. Plasma levels of human recombinant and endogenous rat interleukin-1beta were measured in separate enzyme-linked immunosorbent assays. While intraperitoneal administration of human recombinant interleukin-1beta elevated plasma levels of this cytokine similarly in vagotomized and sham-operated animals, only sham-operated rats responded with fever. Plasma levels of endogenous rat interleukin-1beta were unchanged by any treatment. These results demonstrate that the blockade of intraperitoneal interleukin-1beta-induced fever after subdiaphragmatic vagotomy cannot be accounted for by alterations of interleukin-1beta levels in the general circulation.


Subject(s)
Fever/immunology , Interleukin-1/blood , Interleukin-1/immunology , Vagotomy/methods , Vagus Nerve/immunology , Animals , Diaphragm , Fever/blood , Handling, Psychological , Humans , Injections, Intraperitoneal , Interleukin-1/pharmacology , Male , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Thermogenesis/immunology , Vagus Nerve/surgery
18.
Auton Neurosci ; 85(1-3): 83-7, 2000 Dec 20.
Article in English | MEDLINE | ID: mdl-11189031

ABSTRACT

Several recent findings, including the inability of subdiaphragmatic vagotomy to block lipopolysaccharide (LPS)-induced interleukin-1beta (IL-1beta) protein in brain, have made it necessary to reexamine the role of the subdiaphragmatic vagal afferents in immune-to-brain communication. In this study, we examined the effects of intraperitoneal (i.p.) injections of LPS on core body temperature in control and subdiaphragmatically vagotomized rats. Vagotomized and sham-operated male Sprague-Dawley rats were injected i.p. with vehicle (pyrogen-free saline) on the control day and LPS (1, 10 or 50 microg/kg) on the experimental day, and core body temperature was monitored by telemetry for 6 h after the injection. At this time, rats were sacrificed, and serum, liver, and pituitary samples were collected. The i.p. injection of LPS increased core body temperature in both sham-operated and vagotomized rats compared to the saline injection. In addition, LPS significantly increased IL-1beta levels in serum, liver, and pituitary compared to saline-injected controls. There were no significant differences in the magnitude of the fever or in the levels of IL-1beta in serum, liver, or pituitary between sham-operated and vagotomized rats. Thus, the current data indicate that, at the doses tested, subdiaphragmatic vagal afferents are not crucial for i.p. LPS-induced fever. Because several effects of vagotomy have been shown to be dependent on dose, we are currently investigating whether vagal afferents are involved in lower-dose i.p. LPS-induced fever.


Subject(s)
Fever/physiopathology , Neuroimmunomodulation/physiology , Vagotomy/methods , Vagus Nerve/immunology , Animals , Brain/cytology , Brain/immunology , Cholecystokinin/pharmacology , Diaphragm , Dose-Response Relationship, Drug , Eating/drug effects , Fever/chemically induced , Fever/immunology , Injections, Intraperitoneal , Interleukin-1/analysis , Interleukin-1/blood , Lipopolysaccharides/pharmacology , Liver/chemistry , Liver/immunology , Male , Pituitary Gland/chemistry , Pituitary Gland/immunology , Rats , Rats, Sprague-Dawley , Vagus Nerve/cytology , Vagus Nerve/surgery
19.
Sleep ; 22(6): 707-15, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10505816

ABSTRACT

The liver Kupffer cells constitute the largest population of fixed macrophages in the body and reside at a strategic position in liver sinusoids to interact with mediators from the gut. Previously, we showed that cafeteria feeding increases sleep by a subdiaphragmatic mechanism and increases interleukin-1beta (IL-1beta) mRNA expression in rat liver and brain. Thus, the aim of the present experiment was to test the hypothesis that macrophages, in particular liver Kupffer cells, contribute to the excess sleep observed in cafeteria diet fed rats. Sleep-wake activity and brain temperature (T(br)) were examined in rats injected with gadolinium chloride (GdCl3) alone and in rats fed a cafeteria diet with or without prior pretreatment with GdCl3. The intravenous administration of GdCl3 alone, using a dose that blocks phagocytosis and eliminates large Kupffer cells (7.5 mg/kg), increased sleep in the dark period of the light-dark cycle and decreased sleep in the light period. Sleep-wake activity returned to baseline levels 24 h after the injection. In control rats, cafeteria feeding increased non-rapid eye movement sleep (NREMS) and T(br), and decreased rapid eye movement sleep (REMS) and electroencephalographic slow-wave activity (SWA) during NREMS. GdCl3 pretreatment prevented the increase in NREMS, but did not significantly affect REMS, T(br), or SWA during NREMS compared with the control rats. These results suggest that liver Kupffer cells contribute to the excess NREMS that accompanies increased feeding possibly via their capacity to produce IL-1beta.


Subject(s)
Contrast Media/pharmacology , Diet , Gadolinium/pharmacology , Sleep, REM/drug effects , Animals , Body Temperature/drug effects , Electroencephalography , Electromyography , Energy Intake/drug effects , Injections, Intravenous , Interleukin-1/metabolism , Macrophages/drug effects , Male , RNA, Messenger/drug effects , Rats , Rats, Sprague-Dawley , Time Factors , Wakefulness/drug effects
20.
Life Sci ; 65(5): 449-81, 1999.
Article in English | MEDLINE | ID: mdl-10462074

ABSTRACT

Interleukin-1beta (IL-1beta) is a key mediator and modulator of a wide array of physiological responses important for survival. It is created by a variety of cell types, including immune cells, glia, and neurons. It is a very potent biological molecule, acting both at the periphery as well as within the central nervous system. The production and release of IL-1beta is tightly regulated by far more complex processes than previously thought. An appreciation of this complexity is necessary for proper interpretation of apparent contradictions in the literature where different aspects of IL-1beta expression are measured. Given that many researchers are not molecular biologists by training, yet need an appreciation of the controls that regulate the function of key proteins such as IL-1beta, this review is aimed at both: (a) clarifying the multiple levels at which IL-1beta production is modulated and (b) using IL-1beta regulation to explain the dynamics of gene regulation to non-molecular biologists. Three major topics will be discussed. First, regulation of IL-1beta production will be examined at every level from extracellular signals that trigger gene activation through release of active protein into the extracellular fluid. Second, regulation of IL-1beta bioavailability and bioactivity will be discussed. This section examines the fact that even after IL-1beta is released, it may or may not be able to exert a biological action due to multiple modulatory factors. Last is the introduction of the idea that IL-1beta regulation is, at times, beyond the direct control of host; that is, when IL-1beta production becomes dysregulated by pathogens.


Subject(s)
Interleukin-1/physiology , Receptors, Interleukin-1/physiology , Signal Transduction , Animals , Gene Expression Regulation/immunology , Humans , Inflammation , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...