Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Eur J Trauma Emerg Surg ; 44(1): 137-141, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28493013

ABSTRACT

BACKGROUND: Hurricane Sandy was a particularly unusual storm with regard to both size and location of landfall. The storm landed in New Jersey, which is unusual for a tropical storm of such scale, and created hazardous conditions which caused injury to residents during the storm and in the months following. This study aims to describe differences in trauma center admissions and patterns of injury during this time period when compared to a period with no such storm. METHODS: Data were collected for this study from patients who were admitted to the trauma center at Morristown Medical Center during Hurricane Sandy or the ensuing cleanup efforts (patients admitted between 29 October 2012 and 27 December 2012) as well as a control group consisting of all patients admitted to the trauma center between 29 October 2013 and 27 December 2013. Patient information was collected to compare the admissions of the trauma center during the period of the storm and cleanup to the control period. RESULTS: A total of 419 cases were identified in the storm and cleanup period. 427 were identified for the control. Striking injuries were more common in the storm and cleanup group by 266.7% (p = 0.0107); cuts were more common by 650.8% (p = 0.0044). Medical records indicate that many of these injuries were caused by Hurricane Sandy. Self-inflicted injuries were more common by 301.3% (p = 0.0294). There were no significant differences in the total number of patients, mortality, or injury severity score between the two cohorts. CONCLUSION: The data we have collected show that the conditions caused by Hurricane Sandy and the following cleanup had a significant effect on injury patterns, with more patients having been injured by being struck by falling or thrown objects, cut while using tools, or causing self-inflicted injuries. These changes, particularly during the cleanup period, are indicative of environmental changes following the storm which increase these risks of injury.


Subject(s)
Cyclonic Storms , Hospitalization/statistics & numerical data , Trauma Centers , Wounds and Injuries/epidemiology , Female , Humans , Male , Middle Aged , New Jersey/epidemiology , Retrospective Studies
2.
Purinergic Signal ; 14(1): 27-36, 2018 03.
Article in English | MEDLINE | ID: mdl-29116551

ABSTRACT

Adenosine represents a powerful modulating factor, which has been shown to orchestrate the scope, duration, and remission of the inflammatory response through the activation of four specific receptors, classified as A1, A2A, A2B, and A3, all being widely expressed in a variety of immune cells. Several selective A2A receptor agonists have displayed anti-inflammatory effects, through the suppression of IL-12, TNF, and IFN-γ production by monocytes and lymphocytes, in the setting of chronic intestinal inflammation. However, the therapeutic application of A2A receptor agonists remains hindered by the risk of serious cardiovascular adverse effects arising from the wide systemic distribution of A2A receptors. The present study focused on evaluating the anti-inflammatory effects of the novel poorly absorbed A2A receptor agonist PSB-0777 in a rat model of oxazolone-induced colitis as well as to evaluate its cardiovascular adverse effects, paying particular attention to the onset of hypotension, one of the main adverse effects associated with the systemic pharmacological activation of A2A receptors. Colitis was associated with decreased body weight, an enhanced microscopic damage score and increased levels of colonic myeloperoxidase (MPO). PSB-0777, but not dexamethasone, improved body weight. PSB-0777 and dexamethasone ameliorated microscopic indexes of inflammation and reduced MPO levels. The beneficial effects of PSB-0777 on inflammatory parameters were prevented by the pharmacological blockade of A2A receptors. No adverse cardiovascular events were observed upon PSB-0777 administration. The novel A2A receptor agonist PSB-0777 could represent the base for the development of innovative pharmacological entities able to act in an event-specific and site-specific manner.


Subject(s)
Adenosine A2 Receptor Agonists/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colitis/pathology , Colon/drug effects , Furans/pharmacology , Adjuvants, Immunologic/toxicity , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Colitis/chemically induced , Disease Models, Animal , Furans/administration & dosage , Furans/chemistry , Male , Oxazolone/toxicity , Rats , Rats, Sprague-Dawley
3.
Acta Physiol Hung ; 96(1): 19-28, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19264039

ABSTRACT

The transcription factor HIF-1 is one of the principal mediators of homeostasis in human tissues exposed to hypoxia. It is implicated in virtually every process of rapid gene expression in response to low oxygen levels. The most common causes of tissue hypoxia are inflammation and/or insufficient circulation or a combination of both. Inflamed tissues and the areas surrounding malignant tumors are characterized by hypoxia and low concentrations of glucose. Serious and generalized inflammation can lead to sepsis and circulatory collapse resulting in acute or chronic tissue hypoxia in various vital organs which induces a rapid homeostatic process in all nucleated cells of affected organs in the human body. Under hypoxic conditions the alpha and beta subunits of HIF-1 make an active heterodimer and drive the transcription of over 60 genes important for cell survival, adaptation, anaerobic metabolism, immune reaction, cytokine production, vascularization and general tissue homeostasis. In addition, HIF-1 plays a key role in the development of physiological systems in fetal and postnatal life. It is also a critical mediator of cancer, lung and cardiovascular diseases. The better understanding of the functions of HIF-1 and the pharmacological modulation of its activity could mean a successful therapeutic approach to these diseases.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Hypoxia/metabolism , Oxygen/metabolism , Signal Transduction , Animals , Apoptosis , Communicable Diseases/metabolism , Embryonic Development , Energy Metabolism , Humans , Hypoxia/pathology , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1/chemistry , Inflammation/metabolism , Neoplasms/metabolism , Neovascularization, Physiologic , Protein Conformation , Protein Structure, Tertiary , Reperfusion Injury/metabolism
4.
J Endocrinol ; 171(2): 355-61, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11691656

ABSTRACT

It is increasingly apparent that there is a bidirectional interaction between the maternal immune system and the reproductive system during pregnancy. Pregnancy is associated with a suppression of maternal specific immune responses, which process underlies the protection of fetal tissues expressing paternally inherited alloantigens. However, recent evidence indicates that the suppression of specific, lymphocyte-mediated immune responses during pregnancy is accompanied by activation of the non-specific arm of the maternal immune response. In the present study, we have investigated the effect of pregnancy on the non-specific immune response induced by bacterial lipopolysaccharide (LPS, endotoxin) in mice. Pregnancy enhanced the LPS-induced production of proinflammatory cytokines, including tumor necrosis factor-alpha, interleukin (IL)-6, and interferon-gamma. On the other hand, LPS-induced levels of the anti-inflammatory cytokine IL-10 were suppressed in pregnant mice. These alterations in cytokine production correlated with an increased susceptibility for endotoxemic mortality in the pregnant mice. Although adrenergic receptors are important regulators of cytokine production in non-pregnant mice, the alpha(2)- and the beta-adrenoceptor-mediated modulation of cytokine production ceases to operate during pregnancy associated with severe endotoxemia. These data may explain how excessive activation of the non-specific immune responses during pregnancy can contribute to the increased severity of some maternal diseases, including septic shock, and can be an important pathophysiological factor in disseminated intravascular coagulation or preeclampsia.


Subject(s)
Berberine/analogs & derivatives , Interleukin-10/blood , Pregnancy Complications, Infectious/immunology , Shock, Septic/immunology , Tumor Necrosis Factor-alpha/analysis , Adrenergic alpha-Antagonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Berberine/pharmacology , Female , Gestational Age , Interferon-gamma/blood , Interleukin-1/blood , Interleukin-6/blood , Lipopolysaccharides , Mice , Mice, Inbred Strains , Models, Animal , Pregnancy , Propranolol/pharmacology
5.
Int J Mol Med ; 8(6): 617-21, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11712075

ABSTRACT

Inosine is an endogenous purine, which has been recently shown to exert immunomodulatory, anti-inflammatory and anti-shock effects in rodent experimental systems. Some of these actions may be related to partial adenosine receptor agonistic effects. It has not been investigated previously whether inosine exerts similar immunomodulatory or anti-inflammatory effects in human cells or enzymes. Here we investigated the effects of inosine on the activation of human monocytes, neutrophils and epithelial cells in vitro. Furthermore, using a human inosine-5'-monophosphate dehydrogenase (IMPDH) enzyme, we examined the potential effects of inosine on the activity of IMPDH, an enzyme involved in the regulation of certain inflammatory/immune processes. Tumor necrosis factor alpha (TNF-alpha) production of bacterial lipopolysaccharide (LPS) stimulated whole blood was used as an indicator of human monocyte activation. The response was dose-dependently, partially suppressed in the presence of inosine. Inosine exerted a dose-dependent and, at the highest dose (3 mM), complete inhibition of the ability of human neutrophils activated with N-formyl-methionyl-leucyl-phenylalanine (fMLP) to induce cytochrome C reduction in vitro. In the human colon cancer cell line HT-29, inosine dose-dependently attenuated the production of IL-8. Inosine failed to affect the activity of IMPDH. Taken together, we conclude that inosine exerts anti-inflammatory effects in many human cell types. Further studies need to establish whether inosine supplementation exerts anti-inflammatory effects in human beings.


Subject(s)
Anti-Inflammatory Agents/pharmacology , HT29 Cells/drug effects , Inosine/pharmacology , Monocytes/drug effects , Neutrophils/drug effects , Blood/drug effects , Blood/metabolism , Cytochrome c Group/drug effects , Cytochrome c Group/metabolism , Dose-Response Relationship, Drug , HT29 Cells/metabolism , Humans , IMP Dehydrogenase/drug effects , IMP Dehydrogenase/metabolism , Interleukin-1/pharmacology , Interleukin-8/biosynthesis , Lipopolysaccharides/pharmacology , Monocytes/metabolism , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/metabolism , Oxidation-Reduction/drug effects , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/drug effects
6.
Immunology ; 103(4): 473-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11529938

ABSTRACT

The anti-inflammatory agent sulphasalazine is an important component of several treatment regimens in the therapy of ulcerative colitis, Crohn's disease and rheumatoid arthritis. Sulphasalazine has many immunomodulatory actions, including modulation of the function of a variety of cell types, such as lymphocytes, natural killer cells, epithelial cells and mast cells. However, the effect of this agent on macrophage (M phi) function has not been characterized in detail. In the present study, we investigated the effect of sulphasalazine and two related compounds - sulphapyridine and 5-aminosalicylic acid - on M phi activation induced by bacterial lipopolysaccharide (LPS) and interferon-gamma (IFN-gamma). In J774 M phi stimulated with LPS (10 microg/ml) and IFN-gamma (100 U/ml), sulphasalazine (50-500 microM) suppressed nitric oxide (NO) production in a concentration-dependent manner. The expression of the inducible NO synthase (iNOS) was suppressed by sulphasalazine at 500 microM. Sulphasalazine inhibited the LPS/IFN-gamma-induced production of both interleukin-12 (IL-12) p40 and p70. The suppression of both NO and IL-12 production by sulphasalazine was superior to that by either sulphapyridine or 5-aminosalicylic acid. Although the combination of LPS and IFN-gamma induced a rapid expression of the active forms of p38 and p42/44 mitogen-activated protein kinases and c-Jun terminal kinase, sulphasalazine failed to interfere with the activation of any of these kinases. Finally, sulphasalazine suppressed the IFN-gamma-induced expression of major histocompatibility complex class II. These results demonstrate that the M phi is an important target of the immunosuppressive effect of sulphasalazine.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Macrophage Activation/drug effects , Sulfasalazine/pharmacology , Animals , Cell Culture Techniques , Histocompatibility Antigens Class II/metabolism , Interferon-gamma/immunology , Interleukin-12/biosynthesis , JNK Mitogen-Activated Protein Kinases , Lipopolysaccharides/immunology , Macrophages, Peritoneal/immunology , Mesalamine/pharmacology , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/metabolism , Phosphorylation/drug effects , Sulfapyridine/pharmacology , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases
7.
Biochim Biophys Acta ; 1539(3): 233-42, 2001 Jun 20.
Article in English | MEDLINE | ID: mdl-11420121

ABSTRACT

The amiloride-inhibitable Na(+)/H(+) antiporter plays an important role in macrophage activation. The intracellular pathways leading to interleukin (IL)-12 p40 production by activated macrophages are incompletely understood. In the present study, we examined the contribution of the Na(+)/H(+) antiporter to the production of IL-12 p40. Amiloride or its analogs decreased the production of IL-12 p40 in macrophages stimulated with bacterial lipopolysaccharide and interferon-gamma. The order of potency of amiloride analogs was consistent with the proposition that the effect of amiloride is mediated by the inhibition of the Na(+)/H(+) antiporter. The effect of amiloride was post-transcriptional, as IL-12 p40 mRNA levels induced by lipopolysaccharide and interferon-gamma were not affected by this inhibitor. Furthermore, the inhibitory effect of amiloride on IL-12 p40 production was not a result of interference with the activation of the p38 and p42/44 mitogen-activated protein kinases or c-Jun kinase. In summary, the production of IL-12 p40 requires a functional Na(+)/H(+) antiporter.


Subject(s)
Amiloride/pharmacology , Interleukin-12/metabolism , Macrophages, Peritoneal/drug effects , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Amiloride/analogs & derivatives , Animals , Chemokine CCL4 , Chemokine CXCL2 , Chemokines/metabolism , Diuretics/pharmacology , Drug Interactions , Enzyme Activation/drug effects , Gene Silencing , JNK Mitogen-Activated Protein Kinases , Lipopolysaccharides/pharmacology , Macrophage Inflammatory Proteins/metabolism , Macrophages, Peritoneal/metabolism , Male , Methacrylates/pharmacology , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Sodium-Hydrogen Exchangers/metabolism , p38 Mitogen-Activated Protein Kinases
8.
Shock ; 14(2): 144-9, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10947158

ABSTRACT

Polyamines are endogenous immunomodulatory molecules. Recent studies revealed that polyamines suppress the production of proinflammatory cytokines and nitric oxide. In the present study, we investigated the effect of the polyamines spermine, spermidine, and putrescine on the production of interleukin (IL)-12 p40, IL-10, and interferon (IFN-gamma) in mouse peritoneal macrophages and spleen cell suspensions. Spermine, but not spermidine or putrescine, suppressed, in a concentration-dependent manner, the production of IL-12 p40 by lipopolysaccharide (LPS)-stimulated macrophages. The effect of spermine was post-transcriptional, because steady-state levels of messenger ribonucleic acid (mRNAs) for IL-12 (p35 and p40) were not affected. In contrast to its inhibitory effect on IL-12 p40, spermine (0.3-3 microM) augmented IL-10 production. The down-regulation of IL-12 p40 by spermine was independent of enhancement of IL-10 by this agent, for spermine retained its ability to suppress IL-12 production in peritoneal macrophages obtained from IL-10-deficient mice. The alterations in cytokine production by spermine did not involve an effect on early intracellular pathways of LPS signal transduction, including the p38 or p42/44 mitogen-activated protein kinases, or the c-jun terminal kinase. In spleen cell suspensions, spermine suppressed the release of IFN-gamma induced either by LPS or anti-CD3 antibody. In summary, spermine exerts anti-inflammatory effects by suppressing IL-12 and IFN-gamma and by augmenting the production of IL-10.


Subject(s)
Gene Expression Regulation/drug effects , Interferon-gamma/metabolism , Interleukin-10/biosynthesis , Interleukin-12/biosynthesis , Spermine/pharmacology , Th1 Cells/drug effects , Animals , Inflammation , Interleukin-10/genetics , Interleukin-12/genetics , JNK Mitogen-Activated Protein Kinases , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Subunits , Putrescine/pharmacology , RNA, Messenger/biosynthesis , Spermidine/pharmacology , Spleen/drug effects , Spleen/metabolism , Th1 Cells/metabolism , Transcription, Genetic/drug effects , p38 Mitogen-Activated Protein Kinases
9.
J Immunol ; 164(2): 1013-9, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10623851

ABSTRACT

Extracellular purines, including adenosine and ATP, are potent endogenous immunomodulatory molecules. Inosine, a degradation product of these purines, can reach high concentrations in the extracellular space under conditions associated with cellular metabolic stress such as inflammation or ischemia. In the present study, we investigated whether extracellular inosine can affect inflammatory/immune processes. In immunostimulated macrophages and spleen cells, inosine potently inhibited the production of the proinflammatory cytokines TNF-alpha, IL-1, IL-12, macrophage-inflammatory protein-1alpha, and IFN-gamma, but failed to alter the production of the anti-inflammatory cytokine IL-10. The effect of inosine did not require cellular uptake by nucleoside transporters and was partially reversed by blockade of adenosine A1 and A2 receptors. Inosine inhibited cytokine production by a posttranscriptional mechanism. The activity of inosine was independent of activation of the p38 and p42/p44 mitogen-activated protein kinases, the phosphorylation of the c-Jun terminal kinase, the degradation of inhibitory factor kappaB, and elevation of intracellular cAMP. Inosine suppressed proinflammatory cytokine production and mortality in a mouse endotoxemic model. Taken together, inosine has multiple anti-inflammatory effects. These findings, coupled with the fact that inosine has very low toxicity, suggest that this agent may be useful in the treatment of inflammatory/ischemic diseases.


Subject(s)
Cytokines/antagonists & inhibitors , Immunosuppressive Agents/pharmacology , Inflammation Mediators/antagonists & inhibitors , Inosine/pharmacology , Lipopolysaccharides/toxicity , Protein Processing, Post-Translational/drug effects , Shock, Septic/immunology , Shock, Septic/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chemokines/antagonists & inhibitors , Chemokines/biosynthesis , Cytokines/biosynthesis , Enzyme Activation/drug effects , Enzyme Activation/immunology , I-kappa B Proteins/metabolism , Inflammation Mediators/metabolism , Injections, Intraperitoneal , Inosine/administration & dosage , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , JNK Mitogen-Activated Protein Kinases , Macrophage Activation/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Protein Processing, Post-Translational/immunology , Purinergic P1 Receptor Agonists , Receptors, Purinergic P1/physiology , Shock, Septic/etiology , Shock, Septic/pathology , Th1 Cells/drug effects , Th1 Cells/metabolism
10.
Br J Pharmacol ; 125(6): 1297-303, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9863660

ABSTRACT

Noradrenaline (NA) and adrenaline (Ad) are modulators of cytokine production. Here we investigated the role of these neurotransmitters in the regulation of macrophage inflammatory protein (MIP)-1alpha expression. Pretreatment of RAW 264.7 macrophages with NA or Ad decreased, in a concentration-dependent manner (1 nM-100 microM), MIP-1alpha release induced by bacterial lipopolysaccharide (LPS 10 ng ml(-1) LPS). The effect of NA was reversed by the selective beta-adrenoceptor antagonist propranolol (10 microM), but not by the alpha-adrenoceptor antagonist phentolamine (10 microM). In the concentration range of 10 nM-10 microM, isoproterenol, a beta-adrenoceptor agonist, but not phenylephrine (a selective alpha1-adrenoceptor agonist) or UK-14304 (a selective alpha2-adrenoceptor agonist) mimicked the inhibitory effects of catecholamines on MIP-1alpha production. Increases in intracellular cyclic adenosine monophosphate, elicited either by the selective type IV phosphodiesterase inhibitor rolipram (0.1 - 10 microM), or by prostaglandin E2, (10 nM-10 microM) decreased MIP-1alpha release, suggesting that increased cyclic AMP may contribute to the suppression of MIP-1alpha release by beta-adrenoceptor stimulation. Northern blot analysis demonstrated that NA (100 nM-10 microM), Ad, isoproterenol, as well as rolipram (100 nM-10 microM) decreased LPS-induced MIP-1alpha mRNA accumulation. NA and Ad (1-100 microM) also decreased MIP-1alpha production in thioglycollate-elicited murine peritoneal macrophages. Pretreatment of mice with either isoproterenol (10 mg kg(-1), i.p.) or rolipram (25 mg kg(-1), i.p.) decreased LPS-induced plasma levels of MIP-1alpha, while propranolol (10 mg kg(-1), i.p.) augmented the production of this chemokine, confirming the role of a beta-adrenoceptor mediated endogenous catecholamine action in the regulation of MIP-1alpha production in vivo. Thus, based on our data we conclude that catecholamines are important endogenous regulators of MIP-1alpha expression in inflammation.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Adrenergic beta-Agonists/pharmacology , Epinephrine/pharmacology , Epinephrine/physiology , Macrophage Inflammatory Proteins/biosynthesis , Norepinephrine/pharmacology , Norepinephrine/physiology , Receptors, Adrenergic, beta/physiology , Animals , Antibody Formation , Cell Adhesion/physiology , Cells, Cultured , Chemokine CCL3 , Chemokine CCL4 , Cyclic AMP/metabolism , Cyclic AMP/physiology , Isoproterenol/pharmacology , Lipopolysaccharides/pharmacology , Macrophage Inflammatory Proteins/blood , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/physiology , Male , Mice , Mice, Inbred BALB C , RNA, Messenger/metabolism , Receptors, Adrenergic, beta/drug effects , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiology , Thioglycolates/pharmacology
11.
Eur J Pharmacol ; 358(3): 261-8, 1998 Oct 09.
Article in English | MEDLINE | ID: mdl-9822893

ABSTRACT

We have recently observed that the selective adenosine A3 receptor agonist N6-(3-iodobenzyl)-adenosine-5'-N-methyluronamide (IB-MECA) augments interleukin-10 and inhibits tumor necrosis factor-alpha production in endotoxemic mice. In the present study, we extended our investigations into the effect of this compound on the bacterial lipopolysaccharide (endotoxin)-induced inflammatory response in the BALB/c, as well as in the C57BL/6 interleukin-10+/+ and the interleukin-10 deficient C57BL/6 interleukin-10(0)/0 mice strains. In the BALB/c mice, i.p. pre-treatment with IB-MECA (0.2 and 0.5 mg/kg) decreased lipopolysaccharide (60 mg/kg i.p.)-induced plasma levels of interleukin-12 (p40 and p70), interferon-gamma, and nitrite/nitrate (breakdown products of nitric oxide (NO)). On the other hand, pre-treatment with this compound failed to influence lipopolysaccharide-induced plasma interleukin-1 alpha, interleukin-6, and corticosterone concentrations. Similar to its effect in BALB/c mice, IB-MECA enhanced the release of interleukin-10 in the C57BL/6 interleukin-10+/+ mice. Furthermore, IB-MECA inhibited the production of interleukin-12, interferon-gamma, and NO in both the C57BL/6 interleukin-10+/+ and C57BL/6 interleukin-10(0)/0 mice, suggesting that the inhibition of pro-inflammatory cytokine production by this compound is independent of the increased release of interleukin-10. Finally, pre-treatment with this compound protected mice against lipopolysaccharide (60 mg/kg i.p.)-induced lethality. These results indicate that stimulation of adenosine A3 receptors has potent anti-inflammatory effects and may represent a potential strategy in the treatment of septic shock and other inflammatory diseases.


Subject(s)
Adenosine/analogs & derivatives , Endotoxemia/prevention & control , Interferon-gamma/drug effects , Interleukin-12/blood , Purinergic P1 Receptor Agonists , Adenosine/pharmacology , Adenosine/therapeutic use , Animals , Corticosterone/blood , Endotoxemia/blood , Endotoxemia/mortality , Endotoxins/pharmacology , Interferon-gamma/blood , Interleukin-1/blood , Interleukin-10/blood , Interleukin-10/genetics , Interleukin-6/blood , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitrates/blood , Nitrites/blood , Polysaccharides, Bacterial/pharmacology , Receptor, Adenosine A3 , Species Specificity , Survival Analysis
12.
Shock ; 10(1): 49-53, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9688091

ABSTRACT

During endotoxemia, immune cells activated by lipopolysaccharide (LPS) produce various inflammatory mediators, including cytokines and nitric oxide (NO). The genes of several mediators are activated in part by the rapid binding of the transcription factor nuclear factor-kappa B (NF-kappaB) to its promoter. The induction of this transcription factor can be blocked by a wide range of antioxidants, including pyrrolidine dithiocarbamate (PDTC). Here we investigated in mice the effect of this compound on the plasma tumor necrosis factor-alpha (TNF-alpha), interferon-gamma (IFN-gamma), interleukin-1alpha (IL-1alpha), interleukin-6 (IL-6), interleukin-10 (IL-10), interleukin-12 (IL-12), macrophage inflammatory protein-1alpha (MIP-1alpha), and nitric oxide (NO) response to intraperitoneal (i.p.) injection of LPS. Pretreatment of animals with PDTC (10-100 mg/kg) 30 min prior to LPS challenge (4 mg/kg, i.p.) decreased plasma TNF-alpha, IL-12, MIP-1alpha, and nitrite/nitrate (breakdown products of NO) concentrations, but enhanced plasma levels of IL-10. Moreover, pretreatment of mice with PDTC (10-100 mg/kg, i.p.) did not alter LPS-induced (4 mg/kg) production of IL-1alpha, IL-6, and IFN-gamma. Finally, PDTC (100 mg/kg) protected the mice against LPS (100 mg/kg)-induced lethality. These results indicate that blockade of the NF-kappaB pathway by PDTC has potent anti-inflammatory action in systemic inflammatory processes.


Subject(s)
Endotoxemia/drug therapy , Interleukin-10/metabolism , Nitric Oxide/metabolism , Pyrrolidines/pharmacology , Thiocarbamates/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Animals , Antioxidants/pharmacology , Chemokine CCL3 , Chemokine CCL4 , Endotoxemia/mortality , Interferon-gamma/metabolism , Interleukin-1/metabolism , Interleukin-12/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/toxicity , Macrophage Inflammatory Proteins/metabolism , Male , Mice , Nitrates/blood , Nitrites/blood , Survival Rate , Tumor Necrosis Factor-alpha/drug effects
13.
J Neuroimmunol ; 88(1-2): 57-61, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9688324

ABSTRACT

Stimulation of beta-adrenoceptors has been shown to regulate the production of various inflammatory mediators. In the present study, we investigated in mice whether ligation of beta-adrenoceptors, modulates lipopolysaccharide (LPS)-induced plasma levels of interleukin (IL)-12, interferon-gamma (IFN-gamma), and IL-10. In BALB/c mice, isoproterenol (1-10 mg kg(-1) , i.p.), a selective agonist of beta-adrenoceptors and also dexamethasone (10 mg kg(-1), i.p.) pretreatment 30 min before the administration of LPS suppressed plasma IL-12 (p40 and p70) concentrations as determined at various time points after the LPS challenge. The inhibition of IL-12 release by isoproterenol was prevented by the beta-adrenoceptor antagonist propranolol confirming the involvement of beta-adrenoceptors in the effect of isoproterenol. Furthermore, pretreatment of the animals with propranolol alone enhanced LPS-induced plasma IL-12, suggesting that endogenous catecholamines inhibit IL-12 production via the beta-adrenoceptors. In IL-10 deficient C57BL/6 IL-10(-/-) mice, plasma levels of IL-12 and IFN-gamma were significantly higher than in their counterparts, with more than 70-fold increase in IL-12. Furthermore, while augmenting the IL-10 response in C57BL/6 IL-10(+/+), isoproterenol inhibited the production of IL-12 in both the C57BL/6 IL-10(+/+) and C57BL/6 IL-10(-/-) mice, suggesting that the inhibition of IL-12 production by this compound is independent of the increased release of IL-10. Our results demonstrate, for the first time, that stimulation of beta-adrenoceptors by isoproterenol or endogenous catecholamines suppresses IL-12 production in vivo.


Subject(s)
Endotoxins/pharmacology , Interleukin-10/deficiency , Interleukin-12/biosynthesis , Receptors, Adrenergic, beta/physiology , Adrenergic beta-Agonists/pharmacology , Animals , Interleukin-12/blood , Isoproterenol/pharmacology , Male , Mice , Mice, Inbred BALB C , Reference Values
14.
Br J Pharmacol ; 124(6): 1099-106, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9720779

ABSTRACT

1. Intracellular calcium has been suggested to be an important mediator of the cellular response in endotoxaemia and shock. Dantrolene is an agent that interferes with intracellular calcium fluxes resulting in a decreased availability of calcium in the cytoplasm. Here we have investigated the effect of dantrolene on lipopolysaccharide (LPS)-induced production of interleukin-10 (IL-10), tumour necrosis factor-alpha (TNF-alpha), and nitric oxide (NO) in mice and in cultured RAW 264.7 macrophages in vitro. 2. In BALB/c mice, LPS-induced plasma IL-10 levels were significantly enhanced by pretreatment with dantrolene (20 mg kg(-1), i.p.) (P < 0.005 at the 90 min time-point). On the other hand, dantrolene pretreatment suppressed circulating TNF-alpha and nitrite/nitrate (breakdown products of NO) concentrations. However, dantrolene had no effect on LPS-induced plasma interleukin-6 (IL-6) levels (67.22+/-5.51 ng ml(-1) in vehicle-pretreated mice and 62.22+/-3.66 ng ml(-1) in dantrolene-pretreated mice, n = 9). 3. Dantrolene inhibited TNF-alpha and NO production in C57BL/6 IL-10+/+ mice, as well as in their IL-10 deficient counterparts (C57BL/6 IL-10(0/0)). 4. In RAW 264.7 macrophages, dantrolene (10-300 microM) reduced IL-10, TNF-alpha, and nitrite (breakdown product of NO) production elicited by LPS (10 microg ml(-1)). Dantrolene (300 microM) did not affect the LPS-induced nuclear translocation of transcription factor nuclear factor kappaB in these cells. 5. Although LPS failed to alter the intracellullar concentration of calcium in single macrophages loaded with Fura-2, dantrolene caused a significant decrease of the basal calcium level as determined 30 min after dantrolene treatment (P < 0.005). ATP (1 mM) caused a rapid rise in intracellular calcium levels in both dantrolene-pretreated and vehicle-pretreated cells. 6. These results indicate that unlike the secretion of TNF-alpha and NO, IL-10 production is differentially regulated in vitro and in vivo. The decrease of plasma levels of the pro-inflammatory mediators TNF-alpha and NO, and increase in circulating IL-10 concentrations by dantrolene suggest that this drug might offer a new therapeutic approach in inflammatory diseases and septic shock.


Subject(s)
Dantrolene/pharmacology , Interleukin-10/biosynthesis , Lipopolysaccharides/pharmacology , Nitric Oxide/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Biological Transport , Calcium/metabolism , Cell Line , Cell Nucleus/metabolism , Interleukin-10/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitrates/blood , Nitrites/blood
15.
Brain Res Bull ; 46(3): 257-61, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9667821

ABSTRACT

Recent studies suggested that transmitters released from the sympathetic nerve terminals can modulate various inflammatory responses by occupation of receptors on immune cells. These neurotransmitters act via alteration of intracellular concentration of second messengers. For instance, intracellular calcium as a second messenger plays an important role in the regulation of immune responses. Endotoxemia has been shown to be associated with an increase in cytosolic free calcium concentration ([Ca2+]i). Previously we have demonstrated that the calcium channel blockers verapamil and diltiazem, as well as dantrolene, an agent that blocks the release of calcium from its cytoplasmic stores, inhibits tumor necrosis factor-a (TNF-alpha) and augments interleukin-10 (IL-10) plasma levels in endotoxemic BALB/c mice. Here we investigated the effects of verapamil, diltiazem, and dantrolene on lipopolysaccharide (LPS)-evoked production of interleukin-12 (IL-12) and interferon-gamma (IFN-gamma) in BALB/c, C57BL/6 IL-10+/+, and the IL-10 deficient C57BL/6 IL-10(0/0) mice. Intraperitoneal (i.p.) pretreatment with dantrolene (20 mg/kg), but not verapamil (10 mg/kg, i.p.) or diltiazem (20 mg/kg, i.p.) suppressed the LPS-induced (80 mg/kg, i.p.) plasma levels of IL-12 and IFN-gamma in BALB/c mice. Similarly to the BALB/c mice, dantrolene increased IL-10 plasma levels in C57BL/6 IL-10+/+ mice. On the other hand, dantrolene suppressed IL-12 and IFN-gamma production in both the C57BL/6 IL-10+/+ and C57BL/6 IL-10(0/0) mice. These data show that calcium entry blockers and dantrolene differentially regulate IL-12 and IFN-gamma production. Furthermore, dantrolene inhibits the IL-12 and IFN-gamma response independently of the increased release of IL-10.


Subject(s)
Calcium Channel Blockers/pharmacology , Dantrolene/pharmacology , Endotoxemia/metabolism , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Animals , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred BALB C
16.
Eur J Immunol ; 28(2): 468-72, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9521054

ABSTRACT

Phosphodiesterase (PDE) inhibitors are potent regulators of various immune processes. Immune cells contain type IV and type III PDE. Here we studied in mice the effects of rolipram, a selective PDE IV inhibitor, and amrinone, a selective PDE III blocker, on plasma levels of IL-12 (p70), IFN-gamma, IL-1, TNF-alpha, and nitric oxide (NO) induced by intraperitoneal injection of Escherichia coli lipopolysaccharide (LPS) (80 mg/kg). Pretreatment of BALB/c mice with both rolipram (1-25 mg/kg) and amrinone (10-100 mg/kg) decreased plasma IL-12 levels in a dose-dependent manner. Similarly, LPS-elicited plasma IFN-gamma concentrations were suppressed by both rolipram and amrinone. However, LPS-induced plasma IL-1alpha levels were not affected by either of these compounds. In addition, rolipram inhibited IL-12, IFN-gamma, TNF-alpha and nitrite/nitrate (breakdown products of NO) production in C57BL/6 IL-10(+/+) mice as well as in their IL-10-deficient counterparts (C57BL/6 IL-10(-/-)). Our results suggest that rolipram and amrinone decrease the immune activation in endotoxemia through inhibition of the production of pro-inflammatory mediators IL-12, IFN-gamma, TNF-alpha and NO. These effects are not the consequences of the increase in IL-10 production by PDE inhibition.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Endotoxemia/enzymology , Endotoxemia/immunology , Interleukin-10/physiology , Interleukin-12/antagonists & inhibitors , Interleukin-12/biosynthesis , Amrinone/administration & dosage , Animals , Cyclic Nucleotide Phosphodiesterases, Type 4 , Injections, Intraperitoneal , Interferon-gamma/biosynthesis , Interferon-gamma/blood , Interleukin-10/biosynthesis , Interleukin-10/deficiency , Interleukin-12/blood , Lipopolysaccharides/administration & dosage , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/antagonists & inhibitors , Pyrrolidinones/administration & dosage , Rolipram , Tumor Necrosis Factor-alpha/antagonists & inhibitors
17.
Brain Res Bull ; 45(2): 183-7, 1998.
Article in English | MEDLINE | ID: mdl-9443838

ABSTRACT

In a previous study we have demonstrated in conscious endotoxemic mice that isoproterenol, a nonselective agonist of beta-adrenergic receptors, decreased the production of proinflammatory mediators tumor necrosis factor-alpha (TNF-alpha) and nitric oxide (NO), and enhanced the formation of the anti-inflammatory cytokine interleukin-10 (IL-10). In the present study we investigated the effect of isoproterenol on the bacterial lipopolysaccharide (endotoxin, LPS; 10 microg/ml)-induced inflammatory response in RAW 264.7 macrophages in vitro. Pretreatment of cells with isoproterenol (10-300 microM) resulted in an inhibition of TNF-alpha, NO (reflected as its stable breakdown product nitrite), as well as IL-10 production that was paralleled with a restoration of the LPS-induced suppression of mitochondrial respiration. In addition, isoproterenol elevated cAMP accumulation in these cells. Finally, isoproterenol (300 microM) did not influence the nuclear translocation of nuclear factor kappaB. These data demonstrate that isoproterenol potently downregulates the LPS-induced inflammatory response and further support the notion that stimulation of beta-adrenoreceptors can be an effective strategy in the treatment of inflammatory diseases.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Interleukin-10/biosynthesis , Isoproterenol/pharmacology , Macrophages/metabolism , Nitric Oxide/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Blotting, Western , Cell Line , Cyclic AMP/metabolism , Depression, Chemical , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , NF-kappa B/metabolism
18.
Shock ; 7(5): 371-5, 1997 May.
Article in English | MEDLINE | ID: mdl-9165673

ABSTRACT

Intracellular cyclic nucleotide levels play an important role in the regulation of several immunological processes. Since elevation of intracellular cyclic adenosine monophosphate and/or cyclic guanosine monophosphate concentration by inhibition of phosphodiesterase (PDE) is known to modulate the inflammatory response, we compared the effect of amrinone, an inhibitor of the PDE III isoenzyme, and of theophylline, a nonspecific PDE inhibitor, on the plasma tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), interleukin-10 (IL-10), and nitric oxide response in mice to intraperitoneal injection of bacterial lipopolysaccharide (LPS). Intraperitoneal treatment of animals with amrinone (100 mg/kg) 30 min before LPS administration decreased both plasma IL-6 and IL-10 concentrations in the first phase of the response, but enhanced plasma levels of these cytokines in the second part. In contrast, pretreatment of the animals with theophylline (100 mg/kg) enhanced LPS-induced plasma IL-6 and IL-10 levels during the whole response. However, pretreatment with both PDE inhibitors resulted in a marked inhibition of LPS-evoked plasma concentrations of TNF-alpha and nitrite/nitrate (breakdown products of nitric oxide) throughout the response. This study demonstrates for the first time that amrinone and theophylline possess differential, but primarily anti-inflammatory, properties during LPS-induced systemic inflammation in the mouse.


Subject(s)
Amrinone/therapeutic use , Cytokines/biosynthesis , Endotoxemia/drug therapy , Nitric Oxide/biosynthesis , Phosphodiesterase Inhibitors/therapeutic use , Theophylline/therapeutic use , Animals , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Endotoxemia/metabolism , Interleukin-10/biosynthesis , Interleukin-6/biosynthesis , Lipopolysaccharides , Male , Mice , Tumor Necrosis Factor-alpha/biosynthesis
19.
Shock ; 7(4): 304-7, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9110418

ABSTRACT

Intracellular calcium is an important mediator of the cellular response in endotoxemia and shock. Here we investigated the effects of verapamil and diltiazem, two calcium entry blockers, on endotoxin (bacterial lipopolysaccharide, LPS)-induced production of pro- and anti-inflammatory cytokines and of nitric oxide in mice. LPS-induced interleukin-10 plasma levels were significantly enhanced, and circulating tumor necrosis factor-alpha concentrations were significantly suppressed in animals pretreated intraperitoneally with verapamil (10 mg/kg) or diltiazem (20 mg/kg). However, LPS-induced interleukin-6 levels were unaffected by the calcium antagonists. Similarly, LPS-induced production of nitrite/nitrate (breakdown products of nitric oxide) was not affected by verapamil and diltiazem. We conclude that calcium entry blockers; selectively modulate the production of some pro- and anti-inflammatory mediators in endotoxemia. These effects may contribute to the cytoprotective and anti-inflammatory effects of calcium entry blockers in shock and trauma.


Subject(s)
Calcium Channel Blockers/pharmacology , Endotoxemia/metabolism , Interleukin-10/metabolism , Animals , Diltiazem/pharmacology , Endotoxemia/chemically induced , Endotoxemia/drug therapy , Interleukin-10/blood , Interleukin-6/blood , Interleukin-6/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred BALB C , Nitric Oxide/metabolism , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/metabolism , Verapamil/pharmacology
20.
Immunology ; 90(1): 95-100, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9038718

ABSTRACT

Pro-inflammatory cytokines, such as tumour necrosis factor (TNF) and free radicals, such as nitric oxide (NO), are mediators of endotoxaemia. Catecholamines are in clinical use to treat the haemodynamic consequences of severe septic shock. Beta-adrenergic agonists exert many of their effects by elevation of intracellular cyclic AMP (cAMP) concentration. Cyclic AMP can modulate endotoxin-induced cytokine and NO production. Here we investigate the effect of isoproterenol pretreatment on the cytokine and NO production induced by bacterial lipopolysaccharide (LPS, 4-10 mg/kg). Pretreatment with isoproterenol (10 mg/kg) blunted the LPS-induced TNF response, increased the LPS-induced formation of interleukin-10 and interleukin-6 and reduced the LPS-induced production of NO in conscious mice. In anaesthetized rats, pretreatment with isoproterenol prevented the LPS-induced suppression of vascular contractility to norepinephrine in the thoracic aorta ex vivo. The hyporeactivity is due to expression of the inducible isoform of NO synthase (iNOS) and was restored by in vitro administration of NG-methyl-L-arginine (L-NMA), an inhibitor of NO synthase. However, L-NMA did not alter vascular contractility in control vessels or in rings taken from the LPS-treated rats pretreated with isoproterenol. Our findings suggest that, in addition to its haemodynamic actions, isoproterenol may also exert beneficial effects by modulating the endotoxin-induced inflammatory response.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Cytokines/drug effects , Endotoxemia/immunology , Isoproterenol/pharmacology , Nitric Oxide/biosynthesis , Animals , Endotoxemia/physiopathology , Interleukin-10/biosynthesis , Interleukin-6/biosynthesis , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/biosynthesis , Vasoconstriction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...