Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 2982, 2022 05 27.
Article in English | MEDLINE | ID: mdl-35624087

ABSTRACT

Cytotoxic stress activates stress-activated kinases, initiates adaptive mechanisms, including the unfolded protein response (UPR) and autophagy, and induces programmed cell death. Fatty acid unsaturation, controlled by stearoyl-CoA desaturase (SCD)1, prevents cytotoxic stress but the mechanisms are diffuse. Here, we show that 1,2-dioleoyl-sn-glycero-3-phospho-(1'-myo-inositol) [PI(18:1/18:1)] is a SCD1-derived signaling lipid, which inhibits p38 mitogen-activated protein kinase activation, counteracts UPR, endoplasmic reticulum-associated protein degradation, and apoptosis, regulates autophagy, and maintains cell morphology and proliferation. SCD1 expression and the cellular PI(18:1/18:1) proportion decrease during the onset of cell death, thereby repressing protein phosphatase 2 A and enhancing stress signaling. This counter-regulation applies to mechanistically diverse death-inducing conditions and is found in multiple human and mouse cell lines and tissues of Scd1-defective mice. PI(18:1/18:1) ratios reflect stress tolerance in tumorigenesis, chemoresistance, infection, high-fat diet, and immune aging. Together, PI(18:1/18:1) is a lipokine that links fatty acid unsaturation with stress responses, and its depletion evokes stress signaling.


Subject(s)
Signal Transduction , Stearoyl-CoA Desaturase , Animals , Apoptosis , Fatty Acids , Mice , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Unfolded Protein Response
2.
FASEB J ; 35(5): e21558, 2021 05.
Article in English | MEDLINE | ID: mdl-33855766

ABSTRACT

Aging is accompanied by chronic, low-grade systemic inflammation, termed inflammaging, a main driver of age-associated diseases. Such sterile inflammation is typically characterized by elevated levels of pro-inflammatory mediators, such as cytokines, chemokines and reactive oxygen species causing organ damage. Lipid mediators play important roles in the fine-tuning of both the promotion and the resolution of inflammation. Yet, it remains unclear how lipid mediators fit within the concept of inflammaging and how their biosynthesis and function is affected by aging. Here, we provide comprehensive signature profiles of inflammatory markers in organs afflicted with inflammation of young and old C57BL/6 mice. We reveal an organ-specific footprint of inflammation-related cytokines, chemokines and lipid mediators, which are distinctively affected by aging. While some organs are characterized by a pronounced pro-inflammatory microenvironment and impaired resolution during aging, others display elevated levels of pro-resolving mediators or an overall decrease in inflammatory signaling. Our results demonstrate that it proves difficult to establish a unifying concept for alterations of immunomodulatory mediators as consequence of aging and that organ specificity needs to be considered. Moreover, our data imply that inclusion of lipid mediators into the concept of inflammaging provides a comprehensive tool to characterize the inflammatory microenvironment during aging on a broader and yet, more detailed scope.


Subject(s)
Aging/pathology , Biomarkers/metabolism , Cellular Microenvironment , Immunomodulation , Inflammation Mediators/metabolism , Inflammation/immunology , Animals , Cytokines/metabolism , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Organ Specificity
3.
Front Pharmacol ; 12: 818544, 2021.
Article in English | MEDLINE | ID: mdl-35046831

ABSTRACT

Introduction: Sex differences in inflammation are obvious and contribute to divergences in the incidence and severity of inflammation-related diseases that frequently preponderate in women. Lipid mediators (LMs), mainly produced by lipoxygenase (LOX) and cyclooxygenase (COX) pathways from polyunsaturated fatty acids (PUFAs), regulate all stages of inflammation. Experimental and clinical studies revealed sex divergences for selected LM pathways without covering the entire LM spectrum, and only few studies have addressed the respective role of sex hormones. Here, we performed the comprehensive LM profile analysis with inflammatory peritoneal exudates and plasma from male and female mice in zymosan-induced peritonitis to identify the potential sex differences in LM biosynthesis during the inflammatory response. We also addressed the impact of sex hormones by employing gonadectomy. Methods: Adult male and female CD1 mice received intraperitoneal injection of zymosan to induce peritonitis, a well-established experimental model of acute, self-resolving inflammation. Mice were gonadectomized 5 weeks prior to peritonitis induction. Peritoneal exudates and plasma were taken at 4 (peak of inflammation) and 24 h (onset of resolution) post zymosan and subjected to UPLC-MS-MS-based LM signature profiling; exudates were analyzed for LM biosynthetic proteins by Western blot; and plasma was analyzed for cytokines by ELISA. Results: Pro-inflammatory COX and 5-LOX products predominated in the peritoneum of males at 4 and 24 h post-zymosan, respectively, with slightly higher 12/15-LOX products in males after 24 h. Amounts of COX-2, 5-LOX/FLAP, and 15-LOX-1 were similar in exudates of males and females. In plasma of males, only moderate elevation of these LMs was apparent. At 4 h post-zymosan, gonadectomy strongly elevated 12/15-LOX products in the exudates of males, while in females, free PUFA and LOX products were rather impaired. In plasma, gonadectomy impaired most LMs in both sexes at 4 h with rather up-regulatory effects at 24 h. Finally, elevated 15-LOX-1 protein was evident in exudates of males at 24 h which was impaired by orchiectomy without the striking impact of gonadectomy on other enzymes in both sexes. Conclusions: Our results reveal obvious sex differences and roles of sex hormones in LM biosynthetic networks in acute self-resolving inflammation in mice, with several preponderances in males that appear under the control of androgens.

4.
Front Immunol ; 11: 2106, 2020.
Article in English | MEDLINE | ID: mdl-33013887

ABSTRACT

The same mechanisms that enable host defense against helminths also drive allergic inflammation. This suggests that pathomechanisms of allergic diseases represent evolutionary old responses against helminth parasites and that studying antihelminth immunity may provide insights into pathomechanisms of asthma. However, helminths have developed an intricate array of immunoregulatory mechanisms to modulate type 2 immune mechanisms. This has led to the hypothesis that the lack of helminth infection may contribute to the rise in allergic sensitization in modern societies. Indeed, the anti-inflammatory potential of helminth (worm) parasites and their products in allergy and asthma has been recognized for decades. As helminth infections bring about multiple undesired effects including an increased susceptibility to other infections, intended helminth infection is not a feasible approach to broadly prevent or treat allergic asthma. Thus, the development of new helminth-based biopharmaceutics may represent a safer approach of harnessing type 2-suppressive effects of helminths. However, progress regarding the mechanisms and molecules that are employed by helminths to modulate allergic inflammation has been relatively recent. The scavenging of alarmins and the modulation of lipid mediator pathways and macrophage function by helminth proteins have been identified as important immunoregulatory mechanisms targeting innate immunity in asthma and allergy. In addition, by regulating the activation of dendritic cells and by promoting regulatory T-cell responses, helminth proteins can counterregulate the adaptive T helper 2 cell response that drives allergic inflammation. Despite these insights, important open questions remain to be addressed before helminth molecules can be used for the prevention and treatment of asthma and other allergic diseases.


Subject(s)
Asthma/immunology , Helminthiasis/immunology , Host-Parasite Interactions/immunology , Hypersensitivity/immunology , Models, Immunological , Adaptive Immunity , Alarmins/metabolism , Animals , Asthma/epidemiology , Asthma/therapy , Biological Evolution , Comorbidity , Dendritic Cells/immunology , Helminth Proteins/administration & dosage , Helminth Proteins/physiology , Helminth Proteins/therapeutic use , Helminthiasis/epidemiology , Helminthiasis/parasitology , Helminths/physiology , Humans , Hypersensitivity/epidemiology , Hypersensitivity/therapy , Immunity, Cellular , Immunity, Innate , Immunomodulation , Inflammation , Macrophage Activation , Mice , Models, Animal , Rats , T-Lymphocyte Subsets/immunology , Therapy with Helminths
5.
J Immunol ; 203(4): 1031-1043, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31300512

ABSTRACT

Alternative (M2)-polarized macrophages possess high capacities to produce specialized proresolving mediators (SPM; i.e., resolvins, protectins, and maresins) that play key roles in resolution of inflammation and tissue regeneration. Vacuolar (H+)-ATPase (V-ATPase) is fundamental in inflammatory cytokine trafficking and secretion and was implicated in macrophage polarization toward the M2 phenotype, but its role in SPM production and lipid mediator biosynthesis in general is elusive. In this study, we show that V-ATPase activity is required for the induction of SPM-biosynthetic pathways in human M2-like monocyte-derived macrophages (MDM) and consequently for resolution of inflammation. Blockade of V-ATPase by archazolid during IL-4-induced human M2 polarization abrogated 15-lipoxygenase-1 expression and prevented the related biosynthesis of SPM in response to pathogenic Escherichia coli, assessed by targeted liquid chromatography-tandem mass spectrometry-based metabololipidomics. In classically activated proinflammatory M1-like MDM, however, the biosynthetic machinery for lipid mediator formation was independent of V-ATPase activity. Targeting V-ATPase in M2 influenced neither IL-4-triggered JAK/STAT6 nor the mTOR complex 1 signaling but strongly suppressed the ERK-1/2 pathway. Accordingly, the ERK-1/2 pathway contributes to 15-lipoxygenase-1 expression and SPM formation in M2-like MDM. Targeting V-ATPase in vivo delayed resolution of zymosan-induced murine peritonitis accompanied by decreased SPM levels without affecting proinflammatory leukotrienes or PGs. Together, our data propose that V-ATPase regulates 15-lipoxygenase-1 expression and consequent SPM biosynthesis involving ERK-1/2 during M2 polarization, implying a crucial role for V-ATPase in the resolution of inflammation.


Subject(s)
Inflammation Mediators/immunology , Macrophage Activation/immunology , Macrophages/immunology , Vacuolar Proton-Translocating ATPases/immunology , Animals , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/metabolism , Macrophages/metabolism , Male , Mice , Signal Transduction/immunology , Vacuolar Proton-Translocating ATPases/metabolism
6.
Biochem Pharmacol ; 165: 263-274, 2019 07.
Article in English | MEDLINE | ID: mdl-30836057

ABSTRACT

Lipid mediators (LM) encompass pro-inflammatory prostaglandins (PG) and leukotrienes (LT) but also specialized pro-resolving mediators (SPM) which display pivotal bioactivities in health and disease. Pharmacological intervention with inflammatory disorders such as osteoarthritis and rheumatoid arthritis commonly employs anti-inflammatory drugs that can suppress PG and LT formation, which however, possess limited effectiveness and side effects. Here, we report on the discovery and characterization of the two novel benzoxanthene lignans 1 and 2 that modulate select LM biosynthetic enzymes enabling the switch from pro-inflammatory LT to SPM biosynthesis as potential pharmacological strategy to intervene with inflammation. In cell-free assays, compound 1 and 2 inhibit microsomal prostaglandin E2 synthase-1 and leukotriene C4 synthase (IC50 ∼ 0.6-3.4 µM) and potently interfere with 5-lipoxygenase (5-LOX), the key enzyme in LT biosynthesis (IC50 = 0.04 and 0.09 µM). In human neutrophils, monocytes and M1 and M2 macrophages, compound 1 and 2 efficiently suppress LT biosynthesis (IC50 < 1 µM), accompanied by elevation of 15-LOX-derived LM including SPM. In zymosan-induced murine peritonitis, compound 1 and 2 ameliorated self-limited inflammation along with suppression of early LT formation and elevation of subsequent SPM biosynthesis in vivo. Together, these novel benzoxanthene lignans promote the LM class switch from pro-inflammatory towards pro-resolving LM to terminate inflammation, suggesting their suitability as novel leads for pharmacotherapy of arthritis and related inflammatory disorders.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Leukotrienes/biosynthesis , Lignans/pharmacology , Adult , Animals , Arachidonate 5-Lipoxygenase/physiology , Arthritis, Rheumatoid/drug therapy , HEK293 Cells , Humans , Leukocytes/metabolism , Macrophages/metabolism , Mice , Prostaglandin-E Synthases/antagonists & inhibitors
7.
Cell Chem Biol ; 26(4): 524-534.e5, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30745237

ABSTRACT

The epidithiodioxopiperazine gliotoxin is a virulence factor of Aspergillus fumigatus, the most important airborne fungal pathogen of humans. Gliotoxin suppresses innate immunity in invasive aspergillosis, particularly by compromising neutrophils, but the underlying molecular mechanisms remain elusive. Neutrophils are the first responders among innate immune cells recruited to sites of infection by the chemoattractant leukotriene (LT)B4 that is biosynthesized by 5-lipoxygenase and LTA4 hydrolase (LTA4H). Here, we identified gliotoxin as inhibitor of LTA4H that selectively abrogates LTB4 formation in human leukocytes and in distinct animal models. Gliotoxin failed to inhibit the formation of other eicosanoids and the aminopeptidase activity of the bifunctional LTA4H. Suppression of LTB4 formation by gliotoxin required the cellular environment and/or reducing conditions, and only the reduced form of gliotoxin inhibited LTA4H activity. Conclusively, gliotoxin suppresses the biosynthesis of the potent neutrophil chemoattractant LTB4 by direct interference with LTA4H thereby impairing neutrophil functions in invasive aspergillosis.


Subject(s)
Aspergillosis/immunology , Aspergillus fumigatus/immunology , Epoxide Hydrolases/immunology , Gliotoxin/immunology , Leukotriene B4/immunology , Animals , Aspergillosis/microbiology , Cell Line , Female , Humans , Immunity, Innate , Leukocytes/immunology , Leukocytes/microbiology , Male , Mice , Neutrophils/immunology , Neutrophils/microbiology , Rats, Wistar
8.
Pharmacol Res ; 139: 182-190, 2019 01.
Article in English | MEDLINE | ID: mdl-30468889

ABSTRACT

The incidence and severity of asthma preponderate in women versus men. Leukotrienes (LTs) are lipid mediators involved in asthma pathogenesis, and sex disparities in LT biosynthesis and anti-LT pharmacology in inflammation have recently emerged. Here, we report on sex dimorphism in LT production during allergen sensitization and its correlation to lung function. While high plasma levels of IgE, as sensitization index, were elevated in both sexes, LT levels increased only in lungs of female ovalbumin-sensitized BALB/c mice. Sex-dependent elevated LT levels strictly correlated to an enhanced airway hyperreactivity, pulmonary inflammation and mast cell infiltration/activation in female mice. Importantly, this sex bias was coupled to superior therapeutic efficacy of different types of clinically used LT modifiers like zileuton, MK886 and montelukast in female animals. Our findings reveal sex-dependent LT production as a basic mechanism of sex dimorphism in allergic asthma, and suggest that women might benefit more from anti-LT asthma therapy.


Subject(s)
Asthma/immunology , Leukotrienes/physiology , Sex Characteristics , Allergens/immunology , Animals , Asthma/pathology , Asthma/physiopathology , Female , Immunoglobulin E/blood , Lung/pathology , Lung/physiopathology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/immunology
9.
Nat Commun ; 9(1): 3834, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30237488

ABSTRACT

Systemic vitamin E metabolites have been proposed as signaling molecules, but their physiological role is unknown. Here we show, by library screening of potential human vitamin E metabolites, that long-chain ω-carboxylates are potent allosteric inhibitors of 5-lipoxygenase, a key enzyme in the biosynthesis of chemoattractant and vasoactive leukotrienes. 13-((2R)-6-hydroxy-2,5,7,8-tetramethylchroman-2-yl)-2,6,10-trimethyltridecanoic acid (α-T-13'-COOH) can be synthesized from α-tocopherol in a human liver-on-chip, and is detected in human and mouse plasma at concentrations (8-49 nM) that inhibit 5-lipoxygenase in human leukocytes. α-T-13'-COOH accumulates in immune cells and inflamed murine exudates, selectively inhibits the biosynthesis of 5-lipoxygenase-derived lipid mediators in vitro and in vivo, and efficiently suppresses inflammation and bronchial hyper-reactivity in mouse models of peritonitis and asthma. Together, our data suggest that the immune regulatory and anti-inflammatory functions of α-tocopherol depend on its endogenous metabolite α-T-13'-COOH, potentially through inhibiting 5-lipoxygenase in immune cells.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Inflammation/pathology , Vitamin E/metabolism , Adolescent , Adult , Aged , Animals , Arachidonate 5-Lipoxygenase/chemistry , Bronchial Hyperreactivity/pathology , Cell Survival/drug effects , Cell-Free System , Humans , Inhibitory Concentration 50 , Leukocytes/metabolism , Lipoxygenase Inhibitors/pharmacology , Liver/drug effects , Liver/metabolism , Metabolome , Mice , Middle Aged , Peritonitis/pathology , Recombinant Proteins/metabolism , Vitamin E/chemistry , Young Adult
10.
Eur J Med Chem ; 156: 815-830, 2018 Aug 05.
Article in English | MEDLINE | ID: mdl-30053720

ABSTRACT

Leukotrienes (LTs) and prostaglandin (PG)E2, produced by 5-lipoxygenase (5-LO) and microsomal prostaglandin E2 synthase-1 (mPGES-1), respectively, are key players in inflammation, and pharmacological suppression of these lipid mediators (LM) represents a strategy to intervene with inflammatory disorders. Previous studies revealed that the benzenesulfonamide scaffold displays efficient 5-LO-inhibitory properties. Here, we structurally optimized benzenesulfonamides which led to an N-phenylbenzenesulfonamide derivative (compound 47) with potent inhibitory activities (IC50 = 2.3 and 0.4 µM for isolated 5-LO and 5-LO in intact cells, respectively). Compound 47 prevented the interaction of 5-LO with its activating protein (FLAP) at the nuclear envelope in transfected HEK293 cells as shown by in situ proximity ligation assay. Comprehensive assessment of the LM profile produced by human macrophages revealed the ability of 47 to selectively down-regulate pro-inflammatory LMs (i.e. LTs and PGE2) in M1 but to enhance the formation of pro-resolving LMs (i.e. resolvins and maresins) in M2 macrophages. Moreover, 47 strongly inhibited LT formation and cell infiltration in two in vivo models of acute inflammation (i.e., peritonitis and air pouch sterile inflammation in mice). Together, 47 represents a novel LT biosynthesis inhibitor with an attractive pharmacological profile as anti-inflammatory drug that also promotes the biosynthesis of pro-resolving LM.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arachidonate 5-Lipoxygenase/metabolism , Lipoxygenase Inhibitors/pharmacology , Prostaglandin-E Synthases/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Cells, Cultured , HEK293 Cells , Humans , Inflammation/drug therapy , Inflammation/enzymology , Inflammation/metabolism , Lipoxygenase Inhibitors/chemistry , Macrophages/drug effects , Macrophages/enzymology , Macrophages/metabolism , Male , Mice , Molecular Docking Simulation , Prostaglandin-E Synthases/metabolism , Sulfonamides/chemistry , Benzenesulfonamides
11.
J Clin Invest ; 127(8): 3167-3176, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28737505

ABSTRACT

Proinflammatory leukotrienes (LTs) are produced by 5-lipoxygenase (5-LO) aided by 5-LO-activating protein (FLAP). LT biosynthesis inhibitors are currently under clinical investigation as treatments for respiratory and cardiovascular diseases. Here, we have revealed a sex bias in the efficiency of clinically relevant LT biosynthesis inhibitors, showing that their effects are superior in females. We found that androgens cause these sex differences by impeding the LT-biosynthetic 5-LO/FLAP complex assembly. Lower doses of the FLAP inhibitor MK886 were required to reduce LTB4 levels in exudates of female versus male mice and rats. Following platelet-activating factor-induced shock, MK886 increased survival exclusively in female mice, and this effect was abolished by testosterone administration. FLAP inhibitors and the novel-type 5-LO inhibitors licofelone and sulindac sulfide exhibited higher potencies in human blood from females, and bioactive 5-LO/FLAP complexes were formed in female, but not male, human and murine leukocytes. Supplementation of female blood or leukocytes with 5α-dihydrotestosterone abolished the observed sex differences. Our data suggest that females may benefit from anti-LT therapy to a greater extent than males, prompting consideration of sex issues in LT modifier development.


Subject(s)
Androgens/metabolism , Leukotrienes/biosynthesis , Sex Factors , Testosterone/administration & dosage , 5-Lipoxygenase-Activating Proteins/metabolism , Animals , Arachidonate 5-Lipoxygenase/metabolism , Dihydrotestosterone/metabolism , Female , Humans , Hydroxyurea/analogs & derivatives , Hydroxyurea/pharmacology , Leukocytes/metabolism , Lipoxygenase Inhibitors/pharmacology , Male , Mice , Pyrroles/administration & dosage , Rats , Rats, Wistar , Sulindac/administration & dosage , Sulindac/analogs & derivatives , Testosterone/metabolism
12.
Sci Rep ; 7(1): 3759, 2017 06 19.
Article in English | MEDLINE | ID: mdl-28630405

ABSTRACT

The severity and course of inflammatory processes differ between women and men, but the biochemical mechanisms underlying these sex differences are elusive. Prostaglandins (PG) and leukotrienes (LT) are lipid mediators linked to inflammation. We demonstrated superior LT biosynthesis in human neutrophils and monocytes, and in mouse macrophages from females, and we confirmed these sex differences in vivo where female mice produced more LTs during zymosan-induced peritonitis versus males. Here, we report sex differences in PG production in neutrophils during acute inflammation. In the late phase (4-8 hrs) of mouse zymosan-induced peritonitis and rat carrageenan-induced pleurisy, PG levels in males were higher versus females, seemingly due to higher PG production in infiltrated neutrophils. Accordingly, human neutrophils from males produced more PGE2 than cells from females. Increased PG biosynthesis in males was accompanied by elevated cyclooxygenase (COX)-2 expression connected to increased nuclear factor-kappa B activation, and was abolished when LT synthesis was pharmacologically blocked, suggesting that elevated PG production in males might be caused by increased COX-2 expression and by shunting phenomena due to suppressed LT formation. Conclusively, our data reveal that the biosynthesis of pro-inflammatory PGs and LTs is conversely regulated by sex with consequences for the inflammatory response.


Subject(s)
Neutrophils/metabolism , Peritonitis/metabolism , Prostaglandins/biosynthesis , Sex Characteristics , Acute Disease , Animals , Cyclooxygenase 2/biosynthesis , Female , Gene Expression Regulation, Enzymologic/drug effects , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Neutrophils/pathology , Peritonitis/chemically induced , Peritonitis/pathology , Zymosan/toxicity
13.
Pharmacol Res ; 106: 64-71, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26859523

ABSTRACT

Leukotrienes (LTs) are lipid mediators derived from arachidonic acid (AA) involved in a number of autoimmune/inflammatory disorders including asthma, allergic rhinitis and cardiovascular diseases. Salvinorin A (SA), a diterpene isolated from the hallucinogenic plant Salvia divinorum, is a well-established analgesic compound, but its anti-inflammatory properties are under-researched and its effects on LT production is unknown to date. Here, we studied the possible effect of SA on LT production and verified its actions on experimental models of inflammation in which LTs play a prominent role. Peritoneal macrophages (PM) stimulated by calcium ionophore A23187 were chosen as in vitro system to evaluate the effect of SA on LT production. Zymosan-induced peritonitis in mice and carrageenan-induced pleurisy in rats were selected as LT-related models to evaluate the effect of SA on inflammation as well as on LT biosynthesis. SA inhibited, in a concentration-dependent manner, A23187-induced LTB4 biosynthesis in isolated PM. In zymosan-induced peritonitis, SA inhibited cell infiltration, myeloperoxidase activity, vascular permeability and LTC4 production in the peritoneal cavity without decreasing the production of prostaglandin E2. In carrageenan-induced pleurisy in rats, a more sophisticated model of acute inflammation related to LTs, SA significantly inhibited LTB4 production in the inflammatory exudates, along with reducing the phlogistic process in the lung. In conclusion, SA inhibited LT production and it was effective in experimental models of inflammation in which LTs play a pivotal role. SA might be considered as a lead compound for the development of drugs useful in LTs-related diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Diterpenes, Clerodane/pharmacology , Diterpenes/pharmacology , Hallucinogens/pharmacology , Inflammation/drug therapy , Leukotriene Antagonists/pharmacology , Leukotriene B4/biosynthesis , Animals , Arachidonic Acid/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Leukotriene B4/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Models, Theoretical , Rats , Rats, Wistar , Zymosan/pharmacology
14.
Clin Ophthalmol ; 7: 927-32, 2013.
Article in English | MEDLINE | ID: mdl-23723684

ABSTRACT

BACKGROUND: This randomized double-blind study examined the use of a new anesthetic agent, levobupivacaine 0.5%, which is the S(-)-enantiomer of a racemic mixture of bupivacaine, for peribulbar anesthesia and compared it with racemic bupivacaine 0.5% alone or in combination with hyaluronidase 10 IU/mL. METHODS: A total of 160 patients undergoing ophthalmic surgery were randomized into four groups (n = 40 each) to receive inferotemporal peribulbar injection of levobupivacaine 0.5% (group L), racemic bupivacaine 0.5% (group B), levobupivacaine + hyaluronidase 10 IU/mL (group LH), or racemic bupivacaine + hyaluronidase 10 IU/mL (group BH) by two anesthetists and two ophthalmologists in a ratio of 25% each. Ocular akinesia and orbicularis oculi function were evaluated using a three-point scale; a value < 5 points was considered as requiring surgery, and movements were re-evaluated the day following surgery to confirm regression of the block. RESULTS: The time to onset (12 ± 2.6 minutes versus 13 ± 2.8 minutes) and duration of anesthesia (185 ± 33.2 minutes versus 188 ± 35.7 minutes) were similar between groups L and B. Complete akinesia (score 0) was obtained more frequently when hyaluronidase was used in addition to the anesthetic, with occurrences of 72.5% versus 57.5% in group LH versus L, respectively, and 67.5% versus 45% in group BH versus B. Moderate hypotension (<30% of baseline) was observed in four patients (10%) in group L, two (5.0%) in group B, one (2.5%) in group LH, and three (7.5%) in group BH. The time to onset was significantly different between groups L and BH, B and BH, and LH and BH, and the duration of anesthesia differed significantly between groups B and LH, B and BH, and L and LH. The akinesia score differed significantly between groups L and LH and between groups B and LH (P = 0.043 and P = 0.018, respectively), and the number of patients with a score of 0 differed significantly between groups B and LH and between groups B and BH (P = 0.004 and P = 0.017, respectively). CONCLUSION: Levobupivacaine is a long-lasting local anesthetic with limited cardiotoxicity and neurotoxicity, and may be considered the landmark for vitreoretinal surgery in elderly patients.

15.
J Pain ; 14(6): 604-12, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23725677

ABSTRACT

UNLABELLED: Repeated injections of the antibiotic ceftriaxone cause analgesia in rodents by upregulating the glutamate transporter, GLT-1. No evidence is available in humans. We studied the effect of a single intravenous administration of ceftriaxone in patients undergoing decompressive surgery of the median or ulnar nerves. Forty-five patients were randomized to receive saline, ceftriaxone (2 g), or cefazolin (2 g), 1 hour before surgery. Cefazolin, which is structurally related to ceftriaxone, was used as a negative control. Pain thresholds were measured 10 minutes before drug injections and then 4 to 6 hours after surgery. Ceftriaxone caused analgesia in all patients, whereas cefazolin was inactive. We also performed animal studies to examine whether a single dose of ceftriaxone was sufficient to induce analgesia. A single intraperitoneal injection of ceftriaxone (200 mg/kg), but not cefazoline (200 mg/kg), caused analgesia in mouse models of inflammatory or postsurgical pain, and upregulated GLT-1 in the spinal cord. Ceftriaxone-induced analgesia was additive to that produced by blockade of mGlu5 receptors, which are activated by extrasynaptic glutamate. These data indicate that a single dose of ceftriaxone causes analgesia in humans and mice and suggest that ceftriaxone should be used for preoperative antimicrobial prophylaxis when a fast relief of pain is desired. PERSPECTIVE: The study reports for the first time that a single preoperative dose of ceftriaxone causes analgesia in humans. A single dose of ceftriaxone could also relieve inflammatory and postsurgical pain and upregulate GLT-1 expression in mice. Ceftriaxone should be preferred to other antibiotics for antimicrobial prophylaxis to reduce postoperative pain.


Subject(s)
Analgesics/therapeutic use , Ceftriaxone/therapeutic use , Pain, Postoperative/drug therapy , Pain, Postoperative/prevention & control , Adult , Aged , Aged, 80 and over , Animals , Brain/drug effects , Brain/metabolism , Cefazolin/therapeutic use , Decompression, Surgical/adverse effects , Disease Models, Animal , Double-Blind Method , Excitatory Amino Acid Transporter 2/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Motor Activity/drug effects , Pain Measurement , Pain, Postoperative/etiology , Peripheral Nervous System Diseases/surgery , Rotarod Performance Test , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...