Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 151
Filter
1.
Assay Drug Dev Technol ; 15(2): 64-76, 2017.
Article in English | MEDLINE | ID: mdl-27996302

ABSTRACT

Contamination of pharmaceutical products and medical devices with pyrogens such as endotoxins is the most common cause of systemic inflammation and, in worst cases, of septic shock. Thus, quantification of pyrogens is crucial. The limulus amebocyte lysate (LAL)-based assays are the reference tests for in vitro endotoxin detection, in association with the in vivo rabbit pyrogen test (RPT), according to European Pharmacopoeia (EP 2.6.14), and U.S. Pharmacopoeia (USP <85>). However, several substances interfere with LAL assay, while RPT is not accurate, not quantitative, and raises ethical limits. Biological assays, as monocyte activation tests, have been developed and included in European Pharmacopoeia (EP 7.0; 04/2010:20630) guidelines as an alternative to RPT and proved relevant to the febrile reaction in vivo. Because this reaction is carried out by endogenous mediators under the transcriptional control of nuclear factor-kappaB (NF-kappaB), we sought to determine whether a NF-kappaB reporter-gene assay, based on MonoMac-6 (MM6) cells, could reconcile the basic mechanism of innate immune response with the relevance of monocytoid cell lines to the organism reaction to endotoxins. This article describes both optimization and characterization of the reporter cells-based assay, which overall proved the linearity, accuracy, and precision of the test, and demonstrated the sensitivity of the assay to 0.24 EU/mL endotoxin, close to the pyrogenic threshold in humans. Moreover, the assay was experimentally compared to the LAL test in the evaluation of selected interfering samples. The good performance of the MM6 reporter test demonstrates the suitability of this assay to evaluate interfering or false-positive samples.


Subject(s)
Artifacts , Biological Assay/methods , Monocytes/drug effects , Monocytes/immunology , Pyrogens/administration & dosage , Pyrogens/analysis , Cell Line , Humans , Lipopolysaccharides , Reproducibility of Results , Sensitivity and Specificity
2.
Innate Immun ; 17(5): 486-95, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21088045

ABSTRACT

Although the rabbit pyrogen test is one of the crucial methods included in each pharmacopeia to evaluate the safety of parenteral medicine, the experimental procedures and pyrogen result judgment algorithms (PRJAs) are still greatly different from one another. In the first stage of testing, original data of 879 batches from a total of 2637 rabbits in our laboratory were judged by PRJAs in the Chinese Pharmacopoeia 2005 III, the Japanese Pharmacopoeia XIV, the Japanese Pharmacopoeia XV, the European Pharmacopeia 6.0, the United States Pharmacopoeia 32 NF27 and two theoretical models proposed by S. Hoffmann, respectively. The results were analyzed to evaluate the effects of various PRJAs. It was shown that: (i) the significant differences in the results judged by various pharmacopeias and Hoffmann's theoretical models were mainly due to the PRJAs and the great differences in PRJAs should be harmonized throughout the world based on balance of reducing animal use and guaranteeing the safety of medicines; (ii) it is better to use PRJAs that depend on the threshold of the sum of temperature rise of all tested rabbits than those that depend on the number of rabbits that are over the threshold of temperature rise of individual rabbit according to clinical proof and the experimental data; and (iii) the PRJA of the Japanese Pharmacopoeia XV has obvious advantages when the total suspicious rate of samples was less than 10%. Additionally, a new PRJA designed for reducing the additional experiment stages and animal consumption is promoted for evaluation.


Subject(s)
Drug Evaluation, Preclinical , Drug-Related Side Effects and Adverse Reactions , Fever/chemically induced , Fever/epidemiology , Pyrogens/adverse effects , Algorithms , Animals , Body Temperature/drug effects , China , Decision Support Techniques , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/standards , Europe , Fever/prevention & control , Humans , Infusions, Parenteral , Japan , Pharmaceutical Preparations/administration & dosage , Pharmacopoeias as Topic , Pyrogens/administration & dosage , Rabbits , Reference Standards , United States
3.
Arch Pharm Res ; 30(8): 1014-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17879756

ABSTRACT

The purpose of this study was to develop a nasal in situ gel system for Radix Bupleuri employing gellan gum as a polymer. Radix Bupleuri in situ gel containing 0.2 mL essential oil extracted from 450 g Radix Bupleuri, proper solubilizing agents and gellan gum (0.5% w/v) was prepared and characterized. The antipyretic effect produced by in situ gel formulation was investigated in fevered rabbits and compared to an intranasal solution. The resulting in situ gel was a clear and light-yellow liquid, with viscosity of 346 mPa x s and caproic acid content of 1.31 +/- 0.01 mg/mL. Intranasal administration of this preparation to fevered rabbits decreased body temperature markedly (1.1 degree C at the doses of oil from 1.5 g Bupleuri/body) and the effect could last for 20-30 h. The results suggest that Radix Bupleuri in situ gel can be greater effective than the solution in the treatment of fever.


Subject(s)
Medicine, Chinese Traditional , Plant Extracts/administration & dosage , Plant Oils/isolation & purification , Pyrogens/administration & dosage , Administration, Intranasal , Animals , Body Temperature , Bupleurum/chemistry , Drug Stability , Excipients/chemistry , Fever/drug therapy , Hydrogels , Male , Plant Extracts/chemistry , Plant Extracts/therapeutic use , Plant Oils/chemistry , Polysaccharides, Bacterial/chemistry , Pyrogens/chemistry , Pyrogens/therapeutic use , Rabbits , Viscosity
4.
Brain Res ; 1120(1): 124-30, 2006 Nov 20.
Article in English | MEDLINE | ID: mdl-17005162

ABSTRACT

We determined c-Fos immunoreactivity (Fos-IR) in selected hypothalamic nuclei, the organum vasculosum of the laminae terminals (OVLT) and somatosensory cortex of rats after hyperthermia induced by exogenous heat exposure, Gram-negative or Gram-positive pyrogen administration. The magnitude of Fos-IR was similar in thermoregulatory hypothalamic nuclei of rats after heat exposure or lipopolysaccharide (LPS) injection, despite the different origins of the hyperthermias. Heat-induced hyperthermia was associated with increased Fos-IR in the somatosensory cortex. LPS, but not heat exposure or injection of killed Staphylococcus aureus cells activated OVLT neurons. The OVLT may thus not be a port of entry for humoral mediators of Gram-positive bacterial fevers.


Subject(s)
Brain/drug effects , Brain/radiation effects , Hot Temperature , Lipopolysaccharides/administration & dosage , Proto-Oncogene Proteins c-fos/metabolism , Pyrogens/administration & dosage , Analysis of Variance , Animals , Brain/cytology , Brain/metabolism , Cell Count/methods , Immunohistochemistry/methods , In Vitro Techniques , Male , Rats , Rats, Sprague-Dawley , Time Factors
5.
Acta Physiol (Oxf) ; 187(3): 379-89, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16776663

ABSTRACT

AIMS: The aim of this study was to characterize the properties of synthetic double-stranded RNA to induce fever and circulating cytokines in guinea pigs with special emphasis on the route of administration and on a putative development of tolerance to this pyrogen. METHODS: Changes in abdominal temperature were recorded in unrestrained animals by use of intra-abdominally implanted radiotransmitters. Circulating concentrations of tumour necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) were measured by use of specific bioassays. RESULTS: The pyrogenic effect of double-stranded RNA at a dose of 500 microg kg(-1) depended on the route of its administration. Intra-arterial (i.a.) or intraperitoneal injections of double-stranded RNA induced pronounced fevers and strong elevations of circulating TNF-alpha and IL-6. Intramuscular injections of the synthetic pyrogen caused rather moderate febrile and cytokine responses. Administration of synthetic RNA into artificial subcutaneously implanted Teflon chambers had no pyrogenic and cytokine-inducing effects. I.a. injections of double-stranded RNA, repeated five times at intervals of 3 days, resulted in fevers of similar shape and duration and similar cytokine response patterns. However, the strength of fever and cytokine formation was significantly reduced, although not abolished, in response to the repeated injections compared with the first injection, indicating a partial development of tolerance. CONCLUSIONS: The modulation of the strength of RNA-induced fever, dependent on the route of administration, or the state of partial tolerance to this pyrogen, may thus be related to the formation of pyrogenic cytokines.


Subject(s)
Cytokines/blood , Fever/chemically induced , Pyrogens/adverse effects , RNA, Double-Stranded/adverse effects , Animals , Dose-Response Relationship, Drug , Guinea Pigs , Injections, Intra-Arterial , Injections, Intramuscular , Injections, Intraperitoneal , Injections, Subcutaneous , Interleukin-6/blood , Male , Pyrogens/administration & dosage , RNA, Double-Stranded/administration & dosage , Tumor Necrosis Factor-alpha/blood
6.
J Neurosci ; 26(4): 1190-8, 2006 Jan 25.
Article in English | MEDLINE | ID: mdl-16436606

ABSTRACT

Recent evidence suggests that neurons in the medullary raphe are critical to the activation of brown adipose tissue (BAT), the major source of nonshivering heat production in the rat. Yet it is unclear which medullary raphe cells participate in cold defense and how participating cells contribute to BAT activation. Therefore, we recorded extracellularly from raphe cells during three thermoregulatory challenges that evoked an increase in BAT temperature in anesthetized rats: central cold, ambient cold, or intracerebroventricular prostaglandin E2 (PGE2) injection. Physiologically identified serotonergic (p5HT) cell discharge increased in response to cold or PGE2 administration and was positively correlated with BAT temperature. However, none of the 147 physiologically identified non-serotonergic (non-p5HT) cells recorded responded to thermoregulatory challenges that evoked an increase in BAT temperature. To test for modulation of BAT activation by non-p5HT cells that are either excited (ON cells) or inhibited (OFF cells) by noxious cutaneous stimulation, noxious stimuli were applied during evoked BAT temperature increases. Noxious stimulation suppressed BAT activation, suggesting that cells inhibited by noxious stimulation facilitate spinal circuits controlling BAT. To test whether medullary OFF cells modulate BAT activity, the mu-opiate receptor agonist (d-Ala2, N-Me-Phe4, Gly-ol5)-enkephalin (DAMGO) was microinjected into the raphe magnus, a manipulation that selectively activates OFF cells. DAMGO microinjection blocked noxious stimulation-evoked suppression of PGE2-induced BAT temperature increases. Thus, both p5HT and non-p5HT OFF cells in the medullary raphe facilitate BAT activation in response to cold challenge or pyrogen.


Subject(s)
Adipose Tissue, Brown/physiology , Pain/physiopathology , Raphe Nuclei/physiology , Animals , Body Temperature , Cold Temperature , Dinoprostone/administration & dosage , Dinoprostone/toxicity , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Hot Temperature/adverse effects , Injections, Intraventricular , Male , Microinjections , Neurons/physiology , Nociceptors/physiology , Preoptic Area/physiology , Pressure/adverse effects , Pyrogens/administration & dosage , Pyrogens/toxicity , Raphe Nuclei/cytology , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/agonists , Serotonin/physiology , Thermogenesis/drug effects , Thermogenesis/physiology
7.
Physiol Behav ; 81(4): 615-21, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15178154

ABSTRACT

Cholecystokinin-octapeptide (CCK-8) has been shown to possess an acute thermogenic and hyperthermic action when given intracerebroventricularly in slightly restrained rats. To substantiate the febrile nature of that hyperthermia freely moving animals should be used and together with body core temperature, at least one behavioral parameter, such as general activity, should also be recorded. In the present studies, Wistar rats (N=34) exposed to thermoneutral (26-28 degrees C) or cold (4 degrees C) ambient temperature and to a 12:12-h light/darkness schedule were infused intracerebroventricularly with CCK-8 or prostaglandin E1 (PGE1) for several days using ALZET minipump and changes in body core temperature and general activity were recorded by biotelemetry (Minimitter). In rats exposed to a thermoneutral ambient temperature, low doses of CCK-8 induced slight but significant rises of day minima of circadian body temperature rhythm (CBTR) and with a high dose (1 microg/h) of the peptide--infused either at thermoneutrality or during cold exposure--an increase of acrometron could also be recorded. All of these changes were observed only during the first 2-4 days of 7-day-long infusions. Intracerebroventricular infusion of PGE1 administered at thermoneutrality in a dose of 1 microg/h for 7 days induced a marked rise in body core temperature with a disappearance of CBTR in some rats for 2-3 days or with rises of day minima/acrometron in others. General activity--running parallel with CBTR in periods without infusions--tended to be decreased when core temperature rose during the first couple of days of intracerebroventricular infusion of higher doses of CCK-8 or of PGE1. The decreased general activity--one component of sickness behavior--together with an increased body core temperature found in the present study, supports the view that they are components of a genuine fever induced by the central effect of the two mediators used.


Subject(s)
Alprostadil/physiology , Body Temperature Regulation/physiology , Circadian Rhythm/physiology , Fever/chemically induced , Hyperthermia, Induced/methods , Pyrogens/physiology , Sincalide/physiology , Alprostadil/administration & dosage , Animals , Body Temperature Regulation/drug effects , Circadian Rhythm/drug effects , Disease Models, Animal , Female , Fever/physiopathology , Infusion Pumps, Implantable , Injections, Intraventricular , Motor Activity/physiology , Pyrogens/administration & dosage , Rats , Rats, Wistar , Restraint, Physical , Sincalide/administration & dosage
8.
Fundam Clin Pharmacol ; 18(2): 163-9, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15066130

ABSTRACT

Macrophage inflammatory protein (MIP)-1beta and RANTES (regulated on activation, normal T-cells expressed and secreted) are members of the CC-family of chemokines. Although these two peptides are structurally and functionally related to one another, each exhibits distinct features, which allows it to independently regulate specific aspects of the host inflammatory response. They evoked intense and functionally different febrile responses when applied directly on pyrogen-sensitive cells located in the in the preoptic area of the anterior hypothalamus (POA). The present experiments were carried out to test the central role of CCR5, a functional receptor for MIP-1beta and RANTES, in the febrile responses induced by these chemokines when injected directly into the POA. The microinjection of an equimolecular dose (50 pg) of either MIP-1beta or RANTES into the POA induced a rapid onset; monophasic fever in rats that persisted for a long period. The microinjection of 2.0 microg specific neutralizing antibodies against CCR5 (anti-CCR5) into the POA fails to affect the effects on body temperature induced by MIP-1beta. However, pretreatment with the same dose of anti-CCR5 suppressed the febrile response induced by RANTES given at the same site. The microinjection of control IgG or anti-CCR5 does not affect basal temperature, when administered alone at the same hypothalamic site. The present experiments show that hypothalamic CCR5 are functionally involved in the febrile response induced by RANTES, but not by MIP-1beta. They also suggest the existence of functionally different components in the presumptive primary locus of the thermoregulatory controller, in which both chemotactic cytokines, together other mediators, could play a relevant role in the complex process of fever pathogenesis.


Subject(s)
Chemokines, CC/adverse effects , Fever/chemically induced , Pyrogens/adverse effects , Receptors, CCR5/administration & dosage , Animals , Antibodies/administration & dosage , Antibodies/physiology , Body Temperature/drug effects , Body Temperature/immunology , CCR5 Receptor Antagonists , Cerebrospinal Fluid/chemistry , Cerebrospinal Fluid/immunology , Chemokine CCL4 , Chemokine CCL5/administration & dosage , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/immunology , Chemokines, CC/administration & dosage , Chemokines, CC/immunology , Fever/immunology , Fever/physiopathology , Fever/prevention & control , Heating , Immunoglobulin G/administration & dosage , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Macrophage Inflammatory Proteins/adverse effects , Macrophage Inflammatory Proteins/antagonists & inhibitors , Macrophage Inflammatory Proteins/immunology , Male , Microinjections/methods , Preoptic Area/anatomy & histology , Preoptic Area/drug effects , Preoptic Area/physiopathology , Pyrogens/administration & dosage , Pyrogens/immunology , Rats , Rats, Wistar , Receptors, CCR5/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Stereotaxic Techniques , Time Factors
9.
J Pharmacol Sci ; 93(2): 155-62, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14578583

ABSTRACT

At first, we investigated whether both beta-endorphin release level in the hypothalamus and body temperature can be altered after intracerebroventricular (i.c.v.) injection of either lipopolysaccharide (LPS), interleukin-1beta (IL-1beta), or prostaglandin E(2) (PGE(2)) in rats. It was found that in the rat, i.c.v. administration of either LPS (0.5 microg in 10 microl), IL-1beta (10 ng in 10 microl), or PGE(2) (200 ng in 10 microl), in addition to producing fever, upregulated the immunoreactivity of beta-endorphin in the preoptic anterior hypothalamus of rat brain. Secondarily, we assessed whether the fever induced by either LPS, IL-1beta, or PGE(2) can be altered by pretreatment with buprenorphine (an opioid receptor antagonist). The results revealed that i.c.v. administration of buprenorphine (1 - 10 microg in 10 microl) alone had an insignificant effect on the body temperature. However, the fever induced by i.c.v. injection of either LPS, IL-1beta, or PGE(2) was significantly attenuated by pretreatment with i.c.v. injection of buprenorphine 1 h before the pyrogen injection in rats. The results suggest that pyrogens enhance beta-endorphin release in the hypothalamus and trigger fever which can be attenuated by buprenorphine, an opioid receptor antagonist.


Subject(s)
Buprenorphine/pharmacology , Fever/chemically induced , Fever/prevention & control , Hypothalamus/metabolism , Narcotic Antagonists/pharmacology , Pyrogens/pharmacology , beta-Endorphin/metabolism , Animals , Buprenorphine/administration & dosage , Dinoprostone/administration & dosage , Dinoprostone/pharmacology , Hypothalamus/drug effects , Immunohistochemistry , Injections, Intraventricular , Interleukin-1/administration & dosage , Interleukin-1/pharmacology , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/pharmacology , Male , Narcotic Antagonists/administration & dosage , Pyrogens/administration & dosage , Pyrogens/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
10.
Arch Pharm Res ; 26(5): 411-5, 2003 May.
Article in English | MEDLINE | ID: mdl-12785738

ABSTRACT

To investigate the mechanisms involved in hemin-induced febrile response, the rectal temperature of rats were measured after intracerebroventricular (i.c.v.) injections of hemin, with or without antagonists. Hemin (10 microg) elicited a significant febrile response, which lasted from 30 min, to more than 6 h, after its administration, but this was not the case with biliverdin (i.c.v.) and bilirubin (i.c.v.). The hemin-induced febrile response was significantly inhibited by pretreatment with an inhibitor of tyrosine kinase (genistein), but not by pretreatment with an inhibitor of protein kinase C (chelerythrine) and a scavenger of iron (deferoxamine). These results suggest that tyrosine kinase is involved in the hemin-induced febrile response.


Subject(s)
Fever/metabolism , Hemin/metabolism , Protein-Tyrosine Kinases/metabolism , Pyrogens/metabolism , Alkaloids , Animals , Benzophenanthridines , Body Temperature/drug effects , Deferoxamine/administration & dosage , Deferoxamine/pharmacology , Fever/chemically induced , Fever/enzymology , Genistein/administration & dosage , Genistein/pharmacology , Heme Oxygenase (Decyclizing)/metabolism , Hemin/administration & dosage , Injections, Intraventricular , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/pharmacology , Male , Phenanthridines/administration & dosage , Phenanthridines/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrogens/administration & dosage , Rats , Rats, Sprague-Dawley
11.
Brain Res Bull ; 59(3): 181-7, 2002 Nov 30.
Article in English | MEDLINE | ID: mdl-12431747

ABSTRACT

It has been proposed that prostaglandin (PG)E(2) production via a process catalyzed by the inducible isoform of cyclooxygenase (COX)-2 and activation of specific PGE(2) receptor subtypes within the preoptic/anterior hypothalamus (AH/POA) is the last step and unique pathway in the induction of a fever. However, many data support the existence of a PG-independent pathway. That is, other more rapid mechanisms, which involve the constitutive COX-1 isozyme, may be more critical for a PG-dependent fever. Thus, we examined the role of both COX isoforms in the AH/POA in fevers induced by macrophage inflammatory protein (MIP)-1beta, a PG-independent pyrogen, and RANTES (regulated on activation, normal T-cells expressed and secreted), a PG-dependent pyrogen. In freely moving rats, two independent polyclonal antibodies were used which neutralize COX-1 and COX-2. The microinjection of either MIP-1beta or RANTES into the pyrogen-sensitive region of the AH/POA induced an intense fever of rapid onset. Peripheral pretreatment with an antipyretic dose of dexamethasone which prevents COX-2 expression, or the microinjections into the AH/POA of either anti-COX-1 or anti-COX-2, blocked the febrile response induced by RANTES but not that induced by MIP-1beta. These results provide strong evidence for the existence of rapid mechanisms in the AH/POA which involve both COX isozymes during the fever induced by RANTES, and further support the existence of an alternative PG-independent pathway in the febrile response.


Subject(s)
Fever/chemically induced , Isoenzymes/antagonists & inhibitors , Pyrogens/metabolism , Animals , Antibodies/administration & dosage , Antibodies/immunology , Chemokine CCL4 , Chemokine CCL5/administration & dosage , Chemokine CCL5/metabolism , Cyclooxygenase 1 , Cyclooxygenase 2 , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Dinoprostone/metabolism , Isoenzymes/drug effects , Isoenzymes/immunology , Macrophage Inflammatory Proteins/administration & dosage , Macrophage Inflammatory Proteins/metabolism , Male , Membrane Proteins , Microinjections , Preoptic Area/metabolism , Prostaglandin-Endoperoxide Synthases/drug effects , Prostaglandin-Endoperoxide Synthases/immunology , Pyrogens/administration & dosage , Pyrogens/pharmacology , Rats , Rats, Wistar , Time Factors
12.
Exp Physiol ; 87(3): 391-9, 2002 May.
Article in English | MEDLINE | ID: mdl-12089607

ABSTRACT

We have investigated the febrile responses of New Zealand White rabbits to a Gram-negative pyrogen (bacterial lipopolysaccharide (LPS) from Salmonella typhosa), commonly associated with systemic infection, and a Gram-positive pyrogen (Staphylococcus aureus), more frequently associated with superficial soft tissue infection, each administered via one of four different routes (intravenous, intramuscular, subcutaneous or intraperitoneal) at each of three different doses (LPS: 0.1, 1 and 10 microg kg(-1); S. aureus: 1.5 x 10(7), 1.5 x 10(8) and 1.5 x 10(9) cell walls kg(-1)). Intravenous administration of LPS evoked rapid, dose-dependent biphasic fever. Injection of LPS by the other routes also evoked dose-dependent fever. However, these fevers were monophasic, had increased latency of onset, and were of lower amplitude. It is important to note that a dose of approximately 10 and 100 times that of the standard intravenous dose was required to produce a similar peak rise in temperature when administered subcutaneously and intraperitoneally, respectively. Intravenous injection of the highest dose of S. aureus evoked dose-dependent biphasic fever, with short latency of onset, which was very similar to that induced by intravenous LPS. At lower doses, intravenous S. aureus induced monophasic fever. No fever occurred when the same doses of S. aureus were administered by any other route. We conclude that any of the four routes may be used for the study of LPS-induced fever, provided that the doses are adjusted. However, studies of S. aureus-induced fever, and detection of contamination with either pyrogen, requires intravenous injection.


Subject(s)
Fever/chemically induced , Gram-Negative Bacteria/chemistry , Gram-Positive Bacteria/chemistry , Pyrogens/administration & dosage , Pyrogens/pharmacology , Animals , Body Temperature/drug effects , Dose-Response Relationship, Drug , Female , Fever/physiopathology , Injections, Intramuscular , Injections, Intraperitoneal , Injections, Intravenous , Injections, Subcutaneous , Male , Rabbits , Salmonella typhi/chemistry , Staphylococcus aureus/chemistry
13.
J Endotoxin Res ; 7(5): 359-64, 2001.
Article in English | MEDLINE | ID: mdl-11753204

ABSTRACT

There is a growing body of evidence suggesting that vagal afferents play a major role in peripheral-neural communication. This study was undertaken to determine whether a dose-dependent effect of lipopolysaccharide (LPS) on vagotomy-induced febrile unresponsiveness exists, and to examine the effect of vagotomy on LPS-induced increase in hypothalamic prostaglandin E2 (HT PGE2) production. Vagotomized and sham-operated rats were subjected to two experimental protocols. In the first, vagotomized and sham-operated rats were injected intraperitoneally with different doses of LPS (200, 500 and 1000 microg/kg) in order to examine the dose-dependent effect of LPS on the biphasic febrile response of the rats. In the second protocol, vagotomized and sham-operated rats were injected intraperitoneally with LPS (500 microg/kg). Two hours post injection, body temperature was measured, the rats were decapitated and blood was collected. Simultaneously, the rats' hypothalami were excised and incubated for 1 h in a Krebs-Henseleit buffer. Next, HT PGE2 was determined by radioimmunoassay. Vagotomy-induced gastric enlargement was then measured to examine the correlation between the magnitude of the enlargement and that of the vagotomy-related febrile unresponsiveness. It was found that vagotomized-induced febrile unresponsiveness is a dose-dependent effect. Subdiaphragmatic resection of the vagus prevented the biphasic febrile response caused by the lowest dose (200 microg/kg) of LPS, whereas the highest dose of LPS (1000 microg/kg) caused a similar biphasic febrile response in both vagotomized and sham-operated rats. Indeed, vagotomy attenuates LPS-induced increase in HT PGE2, and blocks the hypothermic phase of the febrile response. On the other hand, no correlation between gastric enlargement and febrile unresponsiveness was found. The results of the present study may cast further light on the crucial role of the vagus nerve as a peripheral-neural pathway in the mediation of the febrile response.


Subject(s)
Body Temperature/drug effects , Lipopolysaccharides/pharmacology , Pyrogens/pharmacology , Vagotomy , Animals , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Escherichia coli/immunology , Hypothalamus, Anterior/drug effects , Hypothalamus, Anterior/metabolism , Injections, Intravenous , Lipopolysaccharides/administration & dosage , Male , Microdialysis , Organ Size/drug effects , Pyrogens/administration & dosage , Rats , Rats, Sprague-Dawley , Stomach/drug effects , Stomach/pathology
14.
Neuropharmacology ; 41(7): 811-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11684145

ABSTRACT

Whether the glutamate release in the organum vasculosum laminae terminalis (OVLT) is attributable to genesis of a pyrogenic fever is unclear. The lack of information led us to evaluate the changes in glutamate concentrations of OVLT during the fever induced by staphylococcal enterotoxin A (SEA) in unanesthetized rabbits. Both the OVLT concentrations of glutamate and the colonic temperatures were simultaneously monitored during systemic injection of SEA, MK801 (an N-methyl-D-aspartate (NMDA) receptor channel blocker), ketamine (an NMDA receptor channel blocker), or normal saline. The extracellular dialysates in the brain were collected using a microdialysis probe previously placed in the OVLT region. The concentrations of glutamate in the microdialysates were measured by a high-pressure liquid chromatography in combination with a fluorescence detector. Systemic administration of SEA (30 ng x kg(-1) I.V.) increased both the concentrations of glutamate in the OVLT and the colonic temperatures. Glutamate appeared to rise slightly earlier than body temperature. Pretreatment or posttreatment with MK801 or ketamine significantly attenuated the SEA-induced augmenting glutamate release in the OVLT and fever in rabbits. The suppression of glutamate release appeared to start slightly earlier than temperature decline. In addition, the SEA-induced fever could be mimicked by direct injection of glutamate or SEA into the OVLT area. The fever induced by intra-OVLT injection of SEA or glutamate was significantly attenuated by pretreatment with an intra-OVLT dose of MK801 (5 microg) or ketamine (10 microg). The results suggest that glutamatergic pathways in the OVLT region are in pyrogenic fever genesis.


Subject(s)
Brain/metabolism , Fever/metabolism , Glutamic Acid/metabolism , Pyrogens/administration & dosage , Animals , Brain/physiology , Enterotoxins/administration & dosage , Fever/chemically induced , Glutamic Acid/physiology , Hypothalamus/metabolism , Injections, Intraventricular , Male , Rabbits
15.
J Neuroendocrinol ; 13(6): 490-5, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11412335

ABSTRACT

The cytokine interleukin-1 (IL-1), which mediates many responses to infection and injury, induces anorexia and fever through direct actions in the central nervous system. The melanocortin neuropeptides, such as alpha melanocyte-stimulating hormone (alpha-MSH), reportedly antagonize many actions of IL-1, including fever and anorexia. However, it is unknown whether endogenous melanocortins modulate anorexia induced by IL-1. The objective of the present study was to establish the effect of endogenous melanocortins on IL-1-induced anorexia and fever in the rat. Intracerebroventricular (i.c.v.) injection of IL-1beta caused a significant reduction in food intake and body weight gain, and a rise in core body temperature in conscious rats. Coadministration of the melanocortin-3/4 receptor (MC3/4-R) antagonist, SHU9119, reversed IL-1beta-induced reductions in food intake and body weight, but did not affect the febrile response to IL-1beta. These data suggest IL-1beta may elicit its effects on food intake through the melanocortin system, predominantly via the MC3-R or MC4-R. In contrast, IL-1beta-induced fever does not appear to be mediated or modulated by MC3-R or MC4-R activity.


Subject(s)
Appetite Depressants/pharmacology , Brain/drug effects , Interleukin-1/pharmacology , Pyrogens/pharmacology , Receptors, Corticotropin/physiology , Animals , Anorexia/chemically induced , Appetite Depressants/administration & dosage , Body Temperature/drug effects , Brain/physiology , Eating/drug effects , Fever/chemically induced , Injections, Intraventricular , Interleukin-1/administration & dosage , Kinetics , Male , Melanocyte-Stimulating Hormones/administration & dosage , Melanocyte-Stimulating Hormones/pharmacology , Pyrogens/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin/agonists , Receptors, Melanocortin , Weight Loss/drug effects , alpha-MSH/administration & dosage , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacology
16.
Ross Fiziol Zh Im I M Sechenova ; 87(10): 1450-6, 2001 Oct.
Article in Russian | MEDLINE | ID: mdl-11767459

ABSTRACT

Administration of Interleukin-1 beta (IL-1 beta) in pyrogenic and subpyrogenic doses induced a depression of social and exploratory behaviour in rats. A reduction in locomotor activity only occurred with pyrogenic doses of the IL-1 beta. The low dose induced the reduction whereas the high dose the increase of anxiety in elevated plus maze. The opposite effects of two doses of IL-1 beta were observed also in a test with saccharine.


Subject(s)
Depression/psychology , Interleukin-1 , Animals , Depression/chemically induced , Dose-Response Relationship, Drug , Exploratory Behavior , Interleukin-1/administration & dosage , Interleukin-1/physiology , Male , Maze Learning , Motor Activity , Pyrogens/administration & dosage , Rats , Rats, Wistar , Social Behavior
17.
Postepy Hig Med Dosw ; 54(4): 537-51, 2000.
Article in Polish | MEDLINE | ID: mdl-11016271

ABSTRACT

Pyrogenic tolerance has been recognized for many years in a variety of species although the mechanisms that are responsible for its development are not well understood. The development of pyrogenic tolerance is associated with the stepwise diminution of pathophysiological and behavioral responses induced by exogenous pyrogens, such as fever, reduction in food and water intake. Several studies either in vivo or ex vivo have indicated the role of various proinflammatory cytokines in the development of pyrogenic tolerance. Most of these studies have indicated that pyrogenic tolerance is associated with down-regulation of cytokine production as well as their biological activity. The mechanisms responsible for down-regulation of cytokine production during development of pyrogenic tolerance are unclear. Since glucocorticoids are required for induction of tolerance, it has been postulated that well known glucocorticoids-dependent negative feedback on the production and biological activity of cytokines may play an important role in development of pyrogenic tolerance. We can not, however, rule out possibility that other mechanisms may participate directly or indirectly in a suppression of cytokines response due to repeated exogenous pyrogen challenge. Either the enhanced uptake of exogenous pyrogens by the hepatic Kupffer cells or the desensitization to exogenous pyrogens by the loss of binding sites, have been proposed as an additional mechanisms which may participate in exogenous pyrogen hyporesponsiveness.


Subject(s)
Pyrogens/immunology , Animals , Antibody Formation , Cytokines/metabolism , Down-Regulation , Fever/immunology , Humans , Immune Tolerance , Lipopolysaccharides/immunology , Liver/metabolism , Pyrogens/administration & dosage
18.
Neuropharmacology ; 39(12): 2505-13, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10974335

ABSTRACT

Fever, a hallmark of disease, is a highly complex process initiated by the action of a number of endogenous pyrogens on the thermosensitive cells of the brain. We describe the activity of RANTES, a chemotactic cytokine, as intrinsically pyrogenic in the rat, when it is delivered directly to the thermosensitive region of the rat's anterior hypothalamic, pre-optic area (AH/POA). RANTES, microinjected into the AH/POA in a dose of 1, 5, 10, 15, 25 or 50 pg, produces an immediate and intense dose-related fever following injection. Increasing the dose to 100 pg did not result in a further increase in the febrile response. No significant change in body temperature was produced by heat-inactivated RANTES. The intrahypothalamic injection of antibodies against RANTES (2.0 microg, 15 min prior to RANTES) significantly blocked the fever induced by this chemokine. Pretreatment with ibuprofen blocked the fever induced by RANTES. In order of potency, the magnitude of the febrile response induced by RANTES was greater than that produced with equipotent doses of either macrophage inflammatory protein-1beta or interleukin-6. The results thus demonstrate that RANTES is the most potent endopyrogen discovered thus far and exerts its action directly on pyrogen-sensitive cells of the AH/POA through a prostaglandin-dependent pathway.


Subject(s)
Chemokine CCL5/pharmacology , Prostaglandins/physiology , Pyrogens/pharmacology , Animals , Body Temperature Regulation/drug effects , Chemokine CCL5/administration & dosage , Cyclooxygenase Inhibitors/pharmacology , Hypothalamus, Anterior/anatomy & histology , Hypothalamus, Anterior/physiology , Ibuprofen/pharmacology , Interleukin-6/pharmacology , Male , Microinjections , Preoptic Area/anatomy & histology , Preoptic Area/physiology , Pyrogens/administration & dosage , Rats , Rats, Wistar , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Transcription Factors/pharmacology
19.
J Immunol ; 165(4): 2306-12, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10925320

ABSTRACT

Streptococcal pyrogenic exotoxin C (SPE C) is a superantigen produced by many strains of Streptococcus pyogenes that (along with streptococcal pyrogenic exotoxin A) is highly associated with streptococcal toxic shock syndrome (STSS) and other invasive streptococcal diseases. Based on the three-dimensional structure of SPE C, solvent-exposed residues predicted to be important for binding to the TCR or the MHC class II molecule, or important for dimerization, were generated. Based on decreased mitogenic activity of various single-site mutants, the double-site mutant Y15A/N38D and the triple-site mutant Y15A/H35A/N38D were constructed and analyzed for superantigenicity, toxicity (lethality), immunogenicity, and the ability to protect against wild-type SPE C-induced STSS. The Y15A/N38D and Y15A/H35A/N38D mutants were nonmitogenic for rabbit splenocytes and human PBMCs and nonlethal in two rabbit models of STSS, yet both mutants were highly immunogenic. Animals vaccinated with the Y15A/N38D or Y15A/H35A/N38D toxoids were protected from challenge with wild-type SPE C. Collectively, these data indicate that the Y15A/N38D and Y15A/H35A/N38D mutants may be useful as toxoid vaccine candidates.


Subject(s)
Bacterial Proteins , Bacterial Vaccines/immunology , Exotoxins/immunology , Membrane Proteins , Pyrogens/immunology , Shock, Septic/immunology , Shock, Septic/prevention & control , Streptococcus pyogenes/immunology , Toxoids/immunology , Animals , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/chemical synthesis , Bacterial Vaccines/genetics , Cells, Cultured , Dimerization , Disease Models, Animal , Exotoxins/administration & dosage , Exotoxins/chemical synthesis , Exotoxins/genetics , Humans , Infusion Pumps, Implantable , Lymphocyte Activation , Models, Molecular , Mutagenesis, Site-Directed , Pyrogens/administration & dosage , Pyrogens/chemical synthesis , Pyrogens/genetics , Rabbits , Streptococcus pyogenes/genetics , Structure-Activity Relationship , Toxoids/administration & dosage , Toxoids/chemical synthesis , Toxoids/genetics , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...