Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 313
Filter
1.
Cell Commun Signal ; 22(1): 189, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38519981

ABSTRACT

The proinflammatory cytokines and arachidonic acid (AA)-derived eicosanoids play a key role in cartilage degeneration in osteoarthritis (OA). The lysophosphatidylcholine acyltransferase 3 (LPCAT3) preferentially incorporates AA into the membranes. Our recent studies showed that MALT1 [mucosa-associated lymphoid tissue lymphoma translocation protein 1]) plays a crucial role in propagating inflammatory signaling triggered by IL-1ß and other inflammatory mediators in endothelial cells. The present study shows that LPCAT3 expression was up-regulated in both human and mice articular cartilage of OA, and correlated with severity of OA. The IL-1ß-induces cell death via upregulation of LPCAT3, MMP3, ADAMTS5, and eicosanoids via MALT1. Gene silencing or pharmacological inhibition of LPCAT3 or MALT1 in chondrocytes and human cartilage explants notably suppressed the IL-1ß-induced cartilage catabolism through inhibition of expression of MMP3, ADAMTS5, and also secretion of cytokines and eicosanoids. Mechanistically, overexpression of MALT1 in chondrocytes significantly upregulated the expression of LPCAT3 along with MMP3 and ADAMTS5 via c-Myc. Inhibition of c-Myc suppressed the IL-1ß-MALT1-dependent upregulation of LPCAT3, MMP3 and ADAMTS5. Consistent with the in vitro data, pharmacological inhibition of MALT1 or gene silencing of LPCAT3 using siRNA-lipid nanoparticles suppressed the synovial articular cartilage erosion, pro-inflammatory cytokines, and eicosanoids such as PGE2, LTB4, and attenuated osteoarthritis induced by the destabilization of the medial meniscus in mice. Overall, our data reveal a previously unrecognized role of the MALT1-LPCAT3 axis in osteoarthritis. Targeting the MALT1-LPCAT3 pathway with MALT1 inhibitors or siRNA-liposomes of LPCAT3 may become an effective strategy to treat OA by suppressing eicosanoids, matrix-degrading enzymes, and proinflammatory cytokines.


Subject(s)
Cartilage, Articular , Osteoarthritis , Animals , Humans , Mice , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism , 1-Acylglycerophosphocholine O-Acyltransferase/pharmacology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/metabolism , Cytokines/metabolism , Eicosanoids/metabolism , Eicosanoids/pharmacology , Eicosanoids/therapeutic use , Endothelial Cells/metabolism , Interleukin-1beta/metabolism , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 3/pharmacology , Matrix Metalloproteinase 3/therapeutic use , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Osteoarthritis/metabolism , RNA, Small Interfering/metabolism
2.
Redox Biol ; 63: 102765, 2023 07.
Article in English | MEDLINE | ID: mdl-37269686

ABSTRACT

Alveolar epithelial cell (AEC) senescence is a key driver of a variety of chronic lung diseases. It remains a challenge how to alleviate AEC senescence and mitigate disease progression. Our study identified a critical role of epoxyeicosatrienoic acids (EETs), downstream metabolites of arachidonic acid (ARA) by cytochrome p450 (CYP), in alleviating AEC senescence. In vitro, we found that 14,15-EET content was significantly decreased in senescent AECs. Exogenous EETs supplementation, overexpression of CYP2J2, or inhibition of EETs degrading enzyme soluble epoxide hydrolase (sEH) to increase EETs alleviated AECs' senescence. Mechanistically, 14,15-EET promoted the expression of Trim25 to ubiquitinate and degrade Keap1 and promoted Nrf2 to enter the nucleus to exert an anti-oxidant effect, thereby inhibiting endoplasmic reticulum stress (ERS) and alleviating AEC senescence. Furthermore, in D-galactose (D-gal)-induced premature aging mouse model, inhibiting the degradation of EETs by Trifluoromethoxyphenyl propionylpiperidin urea (TPPU, an inhibitor of sEH) significantly inhibited the protein expression of p16, p21, and γH2AX. Meanwhile, TPPU reduced the degree of age-related pulmonary fibrosis in mice. Our study has confirmed that EETs are novel anti-senescence substances for AECs, providing new targets for the treatment of chronic lung diseases.


Subject(s)
Alveolar Epithelial Cells , Cellular Senescence , Eicosanoids , Endoplasmic Reticulum Stress , NF-E2-Related Factor 2 , Animals , Mice , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/physiology , Eicosanoids/pharmacology , Endoplasmic Reticulum Stress/drug effects , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2/genetics , Pulmonary Fibrosis , Cellular Senescence/drug effects
3.
Cannabis Cannabinoid Res ; 8(3): 434-444, 2023 06.
Article in English | MEDLINE | ID: mdl-37074668

ABSTRACT

Introduction: The endogenous cannabinoid (endocannabinoid) system is an emerging target for the treatment of chronic inflammatory disease with the potential to advance treatment for many respiratory illnesses. The varied effects of endocannabinoids across tissue types makes it imperative that we explore their physiologic impact within unique tissue targets. The aim of this scoping review is to explore the impact of endocannabinoid activity on eicosanoid production as a measure of human airway inflammation. Methods: A scoping literature review was conducted according to PRISMA-ScR (Preferred Reporting Items for Systematic reviews and Meta-Analyses extension for Scoping Reviews) guidelines. Search strategies using MeSH terms related to cannabinoids, eicosanoids, cyclooxygenase (COX), and the respiratory system were used to query Medline, Embase, Cochrane, CINAHL, Web of Science, and Biosis Previews in December 2021. Only studies that investigated the relationship between endocannabinoids and the eicosanoid system in mammalian respiratory tissue after 1992 were included. Results: Sixteen studies were incorporated in the final qualitative review. Endocannabinoid activation increases COX-2 expression, potentially through ceramide-dependent or p38 and p42/44 Mitogen-Activated Protein Kinase pathways and is associated with a concentration-dependent increase in prostaglandin (PG)E2. Inhibitors of endocannabinoid hydrolysis found either an increase or no change in levels of PGE2 and PGD2 and decreased levels of leukotriene (LT)B4, PGI2, and thromboxane A2 (TXA2). Endocannabinoids increase bronchial epithelial cell permeability and have vasorelaxant effects in human pulmonary arteries and cause contraction of bronchi and decreased gas trapping in guinea pigs. Inhibitors of endocannabinoid hydrolysis were found to have anti-inflammatory effects on pulmonary tissue and are primarily mediated by COX-2 and activation of eicosanoid receptors. Direct agonism of endocannabinoid receptors appears to play a minor role. Conclusion: The endocannabinoid system has diverse effects on the mammalian airway. While endocannabinoid-derived PGs can have anti-inflammatory effects, endocannabinoids also produce proinflammatory conditions, such as increased epithelial permeability and bronchial contraction. These conflicting findings suggest that endocannabinoids produce a variety of effects depending on their local metabolism and receptor agonism. Elucidation of the complex interplay between the endocannabinoid and eicosanoid pathways is key to leveraging the endocannabinoid system as a potential therapeutic target for human airway disease.


Subject(s)
Cannabinoids , Endocannabinoids , Animals , Guinea Pigs , Humans , Anti-Inflammatory Agents , Cannabinoids/pharmacology , Cannabinoids/therapeutic use , Cyclooxygenase 2 , Dinoprostone , Eicosanoids/metabolism , Eicosanoids/pharmacology , Eicosanoids/therapeutic use , Endocannabinoids/metabolism , Mammals/metabolism , Respiratory System/metabolism
4.
Biomolecules ; 12(10)2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36291611

ABSTRACT

We previously identified that NO derived from neuronal cells acts on glial cells and causes vasodilation in the healthy rat retina via the release of epoxyeicosatrienoic acids (EETs) and prostaglandins (PGs) by activation of the arachidonic acid cascade. However, it is not clear which PG types are involved in these responses. The aim of the present study was to identify prostanoid receptors involved in glial cell-derived vasodilation induced by NO in rat retina. Male Wistar rats were used to examine the effects of intravitreal pretreatment with indomethacin, a cyclooxygenase inhibitor; PF-04418948, a prostanoid EP2 receptor antagonist; and CAY10441, a prostanoid IP receptor antagonist, on the changes in the retinal arteriolar diameter induced by intravitreal administration of NOR3, an NO donor. Retinal arteriolar diameters were measured using ocular fundus images captured with a high-resolution digital camera in vivo. The increase in the retinal arteriolar diameter induced by intravitreal injection of NOR3 was significantly suppressed by intravitreal pretreatment with indomethacin and PF-04418948, but not by CAY10441. The dose of PF-04418948 and CAY10441 injected intravitreally in the present study significantly reduced the increase in the retinal arteriolar diameter induced by prostaglandin E2 (PGE2) and prostaglandin I2 (PGI2), respectively. These results suggest that activation of the arachidonic acid cascade and subsequent stimulation of prostanoid EP2 receptors are involved in rat retinal vasodilatory responses evoked by NO-induced glial cell stimulation. Therefore, glial cell-derived PGE2, similar to EETs, may play an important role in retinal vasodilatory mechanisms.


Subject(s)
Prostaglandins , Vasodilation , Animals , Rats , Male , Nitric Oxide/pharmacology , Epoprostenol/pharmacology , Rats, Wistar , Neuroglia , Retina , Dinoprostone , Cyclooxygenase Inhibitors/pharmacology , Indomethacin , Eicosanoids/pharmacology , Arachidonic Acids/pharmacology
5.
Int J Mol Sci ; 23(9)2022 Apr 27.
Article in English | MEDLINE | ID: mdl-35563200

ABSTRACT

Atherosclerosis is one of the most important problems of modern medicine as it is the leading cause of hospitalizations, disability, and mortality. The key role in the development and progression of atherosclerosis is the imbalance between the activation of inflammation in the vascular wall and the mechanisms of its control. The resolution of inflammation is the most important physiological mechanism that is impaired in atherosclerosis. The resolution of inflammation has complex, not fully known mechanisms, in which lipid mediators derived from polyunsaturated fatty acids (PUFAs) play an important role. Specialized pro-resolving mediators (SPMs) represent a group of substances that carry out inflammation resolution and may play an important role in the pathogenesis of atherosclerosis. SPMs include lipoxins, resolvins, maresins, and protectins, which are formed from PUFAs and regulate many processes related to the active resolution of inflammation. Given the physiological importance of these substances, studies examining the possibility of pharmacological effects on inflammation resolution are of interest.


Subject(s)
Atherosclerosis , Lipoxins , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Eicosanoids/pharmacology , Fatty Acids, Unsaturated/pharmacology , Fatty Acids, Unsaturated/therapeutic use , Humans , Inflammation/drug therapy , Inflammation/pathology , Inflammation Mediators
6.
Am J Physiol Cell Physiol ; 322(5): C1011-C1021, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35385329

ABSTRACT

Arachidonic acid metabolites epoxyeicosatrienoates (EETs) and hydroxyeicosatetraenoates (HETEs) are important regulators of myocardial blood flow and coronary vascular resistance (CVR), but their mechanisms of action are not fully understood. We applied a chemoproteomics strategy using a clickable photoaffinity probe to identify G protein-coupled receptor 39 (GPR39) as a microvascular smooth muscle cell (mVSMC) receptor selective for two endogenous eicosanoids, 15-HETE and 14,15-EET, which act on the receptor to oppose each other's activity. The former increases mVSMC intracellular calcium via GPR39 and augments coronary microvascular resistance, and the latter inhibits these actions. Furthermore, we find that the efficacy of both ligands is potentiated by zinc acting as an allosteric modulator. Measurements of coronary perfusion pressure (CPP) in GPR39-null hearts using the Langendorff preparation indicate the receptor senses these eicosanoids to regulate microvascular tone. These results implicate GPR39 as an eicosanoid receptor and key regulator of myocardial tissue perfusion. Our findings will have a major impact on understanding the roles of eicosanoids in cardiovascular physiology and disease and provide an opportunity for the development of novel GPR39-targeting therapies for cardiovascular disease.


Subject(s)
Cytochrome P-450 Enzyme System , Eicosanoids , Arachidonic Acid/metabolism , Coronary Vessels/metabolism , Cytochrome P-450 Enzyme System/metabolism , Eicosanoids/analysis , Eicosanoids/metabolism , Eicosanoids/pharmacology , Vascular Resistance
7.
Int J Mol Sci ; 23(7)2022 Mar 25.
Article in English | MEDLINE | ID: mdl-35408952

ABSTRACT

Vascular remodeling is a typical feature of vascular diseases, such as atherosclerosis, aneurysms or restenosis. Excessive inflammation is a key mechanism underlying vascular remodeling via the modulation of vascular fibrosis, phenotype and function. Recent evidence suggests that not only augmented inflammation but unresolved inflammation might also contribute to different aspects of vascular diseases. Resolution of inflammation is mediated by a family of specialized pro-resolving mediators (SPMs) that limit immune cell infiltration and initiate tissue repair mechanisms. SPMs (lipoxins, resolvins, protectins, maresins) are generated from essential polyunsaturated fatty acids. Synthases and receptors for SPMs were initially described in immune cells, but they are also present in endothelial cells (ECs) and vascular smooth muscle cells (VSMCs), where they regulate processes important for vascular physiology, such as EC activation and VSMC phenotype. Evidence from genetic models targeting SPM pathways and pharmacological supplementation with SPMs have demonstrated that these mediators may play a protective role against the development of vascular remodeling in atherosclerosis, aneurysms and restenosis. This review focuses on the latest advances in understanding the role of SPMs in vascular cells and their therapeutic effects in the vascular remodeling associated with different cardiovascular diseases.


Subject(s)
Atherosclerosis , Inflammation Mediators , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Eicosanoids/pharmacology , Endothelial Cells/metabolism , Humans , Inflammation/drug therapy , Inflammation Mediators/metabolism , Vascular Remodeling
8.
Lasers Med Sci ; 37(3): 1953-1962, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34731332

ABSTRACT

Allergic rhinitis (AR) is an inflammatory disorder of the nasal mucosa, and is a worldwide health problem with a significant impact on the quality of life. The main goal of AR treatment is to relieve symptoms. However, standard treatments have considerable side effects or are not effective. Photobiomodulation (PBM) therapy has emerged as an alternative treatment. Here, we evaluated the effects of transcutaneous systemic (tail) or local (skin over nostrils) PBM using a 660-nm light-emitting diode (LED) array. Adult rats were assigned into 4 groups: basal, as non-manipulated animals; Sham, as rats sensitized with 7 intradermal injections of ovalbumin (OVA) plus alum followed by intranasal instillation with OVA (2%) daily for 7 days; and the LPBM and SPBM groups, in which the animals were treated with PBM (local or systemic) immediately after the last instillation of OVA (1%) daily for 3 days. Our results showed that local PBM treatment reduced mast cell degranulation in the nasopharynx and nostrils; levels of leukotriene B4, thromboxane A2, and interleukin 4 (IL-4) in the nasopharynx; and gene expression of IL-4. Moreover, we showed higher levels and gene expression of IL-10 after local PBM treatment. Systemic PBM treatment did not change any of the evaluated parameters. In conclusion, our data showed that local (but not systemic) treatment with PBM could improve parameters related to AR in an animal model, and should be tested clinically.


Subject(s)
Cytokines , Rhinitis, Allergic , Animals , Cell Degranulation , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Eicosanoids/pharmacology , Eicosanoids/therapeutic use , Mice , Mice, Inbred BALB C , Ovalbumin/pharmacology , Ovalbumin/therapeutic use , Quality of Life , Rats , Rhinitis, Allergic/drug therapy , Rhinitis, Allergic/radiotherapy
9.
Platelets ; 33(5): 709-718, 2022 Jul 04.
Article in English | MEDLINE | ID: mdl-34697991

ABSTRACT

Sigma-1 receptor (S1R) is detected in different cell types and can regulate intracellular signaling pathways. S1R plays a role in the pathomechanism of diseases and the regulation of neurotransmitters. Fluvoxamine can bind to S1R and reduce the serotonin uptake of neurons and platelets. We therefore hypothesized that platelets express S1R, which can modify platelet function. The expression of the SIGMAR1 gene in rat platelets was examined with a reverse transcription polymerase chain reaction and a quantitative polymerase chain reaction. The receptor was also visualized by immunostaining and confocal laser scanning microscopy. The effect of S1R agonist PRE-084 on the eicosanoid synthesis of isolated rat platelets and ADP- and AA-induced platelet aggregation was examined. S1R was detected in rat platelets both at gene and protein levels. Pretreatment with PRE-084 of resting platelets induced elevation of eicosanoid synthesis. The rate of elevation in thromboxane B2 and prostaglandin D2 synthesis was similar, but the production of prostaglandin E2 was higher. The concentration-response curve showed a sigmoidal form. The most effective concentration of the agonist was 2 µM. PRE-084 increased the quantity of cyclooxygenase-1 as detected by ELISA. PRE-084 also elevated the ADP- and AA-induced platelet aggregation. S1R of platelets might regulate physiological or pathological functions.


Subject(s)
Blood Platelets , Platelet Aggregation , Adenosine Diphosphate/pharmacology , Animals , Blood Platelets/metabolism , Eicosanoids/metabolism , Eicosanoids/pharmacology , Humans , Prostaglandins/metabolism , Prostaglandins/pharmacology , Rats
10.
Front Immunol ; 12: 768133, 2021.
Article in English | MEDLINE | ID: mdl-34868025

ABSTRACT

Both the initiation and the resolution of inflammatory responses are governed by the sequential activation, migration, and control/suppression of immune cells at the site of injury. Bioactive lipids play a major role in the fine-tuning of this dynamic process in a timely manner. During inflammation and its resolution, polymorphonuclear cells (PMNs) and macrophages switch from producing pro-inflammatory prostaglandins and leukotrienes to specialized pro-resolving lipid mediators (SPMs), namely, lipoxins, resolvins, protectins, and maresins, which are operative at the local level to limit further inflammation and tissue injury and restore homeostasis. Accumulating evidences expand now the role and actions of these lipid mediators from innate to adaptive immunity. In particular, SPMs have been shown to contribute to the control of chronic inflammation, and alterations in their production and/or function have been associated with the persistence of several pathological conditions, including autoimmunity, in human and experimental models. In this review, we focus on the impact of pro-resolving lipids on T cells through their ability to modulate T-cell responses. In particular, the effects of the different families of SPMs to restrain effector T-cell functions while promoting regulatory T cells will be reviewed, along with the underlying mechanisms. Furthermore, the emerging concept of SPMs as new biological markers for disease diagnostic and progression and as putative therapeutic tools to regulate the development and magnitude of inflammatory and autoimmune diseases is discussed.


Subject(s)
Eicosanoids/pharmacology , Immunomodulating Agents/pharmacology , Inflammation Mediators/pharmacology , Lipoxins/pharmacology , T-Lymphocytes/drug effects , Cellular Reprogramming , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/pharmacology , Humans , Inflammation Mediators/therapeutic use , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
11.
Infect Immun ; 89(8): e0009521, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34031130

ABSTRACT

Eicosanoids are lipid-based signaling molecules that play a unique role in innate immune responses. The multiple types of eicosanoids, such as prostaglandins (PGs) and leukotrienes (LTs), allow the innate immune cells to respond rapidly to bacterial invaders. Bacterial pathogens alter cyclooxygenase (COX)-derived prostaglandins (PGs) in macrophages, such as PGE2 15d-PGJ2, and lipoxygenase (LOX)-derived leukotriene LTB4, which has chemotactic functions. The PG synthesis and secretion are regulated by substrate availability of arachidonic acid and by the COX-2 enzyme, and the expression of this protein is regulated at multiple levels, both transcriptionally and posttranscriptionally. Bacterial pathogens use virulence strategies such as type three secretion systems (T3SSs) to deliver virulence factors altering the expression of eicosanoid-specific biosynthetic enzymes, thereby modulating the host response to bacterial lipopolysaccharides (LPS). Recent advances have identified a novel role of eicosanoids in inflammasome activation during intracellular infection with bacterial pathogens. Specifically, PGE2 was found to enhance inflammasome activation, driving the formation of pore-induced intracellular traps (PITs), thus trapping bacteria from escaping the dying cell. Finally, eicosanoids and IL-1ß released from macrophages are implicated in the efferocytosis of neighboring neutrophils. Neutrophils play an essential role in phagocytosing and degrading PITs and associated bacteria to restore homeostasis. This review focuses on the novel functions of host-derived eicosanoids in the host-pathogen interactions.


Subject(s)
Bacterial Infections/immunology , Bacterial Infections/microbiology , Chemotaxis, Leukocyte/immunology , Eicosanoids/metabolism , Host-Pathogen Interactions/immunology , Immunity, Innate , Neutrophils/immunology , Neutrophils/metabolism , Bacterial Infections/metabolism , Biomarkers , Dinoprostone/metabolism , Eicosanoids/pharmacology , Inflammation Mediators , Lipid Metabolism , Neutrophils/pathology , Type III Secretion Systems
12.
Sci Rep ; 11(1): 3739, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33580125

ABSTRACT

This study addressed the hypothesis that epoxyeicosatrienoic acids (EETs) synthesized by CYP450 and catabolized by soluble epoxide hydrolase (sEH) are involved in the maintenance of renal allograft function, either directly or through modulation of cardiovascular function. The impact of single nucleotide polymorphisms (SNPs) in the sEH gene EPHX2 and CYP450 on renal and vascular function, plasma levels of EETs and peripheral blood monuclear cell sEH activity was assessed in 79 kidney transplant recipients explored at least one year after transplantation. Additional experiments in a mouse model mimicking the ischemia-reperfusion (I/R) injury suffered by the transplanted kidney evaluated the cardiovascular and renal effects of the sEH inhibitor t-AUCB administered in drinking water (10 mg/l) during 28 days after surgery. There was a long-term protective effect of the sEH SNP rs6558004, which increased EET plasma levels, on renal allograft function and a deleterious effect of K55R, which increased sEH activity. Surprisingly, the loss-of-function CYP2C9*3 was associated with a better renal function without affecting EET levels. R287Q SNP, which decreased sEH activity, was protective against vascular dysfunction while CYP2C8*3 and 2C9*2 loss-of-function SNP, altered endothelial function by reducing flow-induced EET release. In I/R mice, sEH inhibition reduced kidney lesions, prevented cardiac fibrosis and dysfunction as well as preserved endothelial function. The preservation of EET bioavailability may prevent allograft dysfunction and improve cardiovascular disease in kidney transplant recipients. Inhibition of sEH appears thus as a novel therapeutic option but its impact on other epoxyfatty acids should be carefully evaluated.


Subject(s)
Eicosanoids/metabolism , Eicosanoids/pharmacology , Kidney Transplantation/methods , Adult , Aged , Allografts/physiology , Animals , Biological Availability , Cytochrome P-450 Enzyme System/metabolism , Disease Models, Animal , Epoxide Hydrolases/metabolism , Epoxy Compounds/pharmacology , Female , Humans , Kidney/cytology , Male , Mice , Mice, 129 Strain , Middle Aged , Reperfusion Injury/prevention & control
13.
Int J Mol Sci ; 22(2)2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33467433

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor superfamily that regulate the expression of genes related to lipid and glucose metabolism and inflammation. There are three members: PPARα, PPARß or PPARγ. PPARγ have several ligands. The natural agonists are omega 9, curcumin, eicosanoids and others. Among the synthetic ligands, we highlight the thiazolidinediones, clinically used as an antidiabetic. Many of these studies involve natural or synthetic products in different pathologies. The mechanisms that regulate PPARγ involve post-translational modifications, such as phosphorylation, sumoylation and ubiquitination, among others. It is known that anti-inflammatory mechanisms involve the inhibition of other transcription factors, such as nuclear factor kB(NFκB), signal transducer and activator of transcription (STAT) or activator protein 1 (AP-1), or intracellular signaling proteins such as mitogen-activated protein (MAP) kinases. PPARγ transrepresses other transcription factors and consequently inhibits gene expression of inflammatory mediators, known as biomarkers for morbidity and mortality, leading to control of the exacerbated inflammation that occurs, for instance, in lung injury/acute respiratory distress. Many studies have shown the therapeutic potentials of PPARγ on pulmonary diseases. Herein, we describe activities of the PPARγ as a modulator of inflammation, focusing on lung injury and including definition and mechanisms of regulation, biological effects and molecular targets, and its role in lung diseases caused by inflammatory stimuli, bacteria and virus, and molecular-based therapy.


Subject(s)
Inflammation/metabolism , Lung Diseases/metabolism , PPAR gamma/metabolism , Signal Transduction/physiology , Animals , Curcumin/metabolism , Curcumin/pharmacology , Eicosanoids/metabolism , Eicosanoids/pharmacology , Humans , Ligands , Lung Diseases/drug therapy , PPAR gamma/agonists , Protein Processing, Post-Translational/drug effects , Signal Transduction/drug effects
14.
Mol Nutr Food Res ; 65(5): e1901269, 2021 03.
Article in English | MEDLINE | ID: mdl-32780927

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by chronic relapsing inflammation in the intestine. Given their role in regulation of inflammation, long-chain n-3 polyunsaturated fatty acids (PUFAs) represent a potential supplementary therapeutic approach to current drug regimens used for IBD. Mechanistically, there is ample evidence for an anti-inflammatory and pro-resolution effect of long-chain n-3 PUFAs after they incorporate into cell membrane phospholipids. They disrupt membrane rafts and when released from the membrane suppress inflammatory signaling by activating PPAR-γ and free fatty acid receptor 4; furthermore, they shift the lipid mediator profile from pro-inflammatory eicosanoids to specialized pro-resolving mediators. The allocation of long-chain n-3 PUFAs also leads to a higher microbiome diversity in the gut, increases short-chain fatty acid-producing bacteria, and improves intestinal barrier function by sealing epithelial tight junctions. In line with these mechanistic studies, most epidemiological studies support a beneficial effect of long-chain n-3 PUFAs intake on reducing the incidence of IBD. However, the results from intervention trials on the prevention of relapse in IBD patients show no or only a marginal effect of long-chain n-3 PUFAs supplementation. In light of the current literature, international recommendations are supported that adequate diet-derived n-3 PUFAs might be beneficial in maintaining remission in IBD patients.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Gastritis/diet therapy , Gastrointestinal Microbiome/drug effects , Inflammatory Bowel Diseases/diet therapy , Animals , Diet, Western/adverse effects , Dietary Supplements , Eicosanoids/pharmacology , Fatty Acids, Omega-3/chemistry , Fatty Acids, Omega-3/physiology , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastritis/etiology , Gastrointestinal Microbiome/physiology , Humans
15.
Cell Rep ; 33(13): 108547, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33378679

ABSTRACT

Mycobacterium tuberculosis (Mtb) regulates the macrophage metabolic state to thrive in the host, yet the responsible mechanisms remain elusive. Macrophage activation toward the microbicidal (M1) program depends on the HIF-1α-mediated metabolic shift from oxidative phosphorylation (OXPHOS) toward glycolysis. Here, we ask whether a tuberculosis (TB) microenvironment changes the M1 macrophage metabolic state. We expose M1 macrophages to the acellular fraction of tuberculous pleural effusions (TB-PEs) and find lower glycolytic activity, accompanied by elevated levels of OXPHOS and bacillary load, compared to controls. The eicosanoid fraction of TB-PE drives these metabolic alterations. HIF-1α stabilization reverts the effect of TB-PE by restoring M1 metabolism. Furthermore, Mtb-infected mice with stabilized HIF-1α display lower bacillary loads and a pronounced M1-like metabolic profile in alveolar macrophages (AMs). Collectively, we demonstrate that lipids from a TB-associated microenvironment alter the M1 macrophage metabolic reprogramming by hampering HIF-1α functions, thereby impairing control of Mtb infection.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lipids/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mycobacterium tuberculosis/metabolism , Tuberculosis, Pleural/metabolism , Animals , Bacterial Load , Eicosanoids/pharmacology , Female , Glycolysis/drug effects , Host-Pathogen Interactions , Humans , Macrophage Activation , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Pleural Effusion , Tuberculosis, Pleural/microbiology
16.
Biomolecules ; 10(10)2020 10 02.
Article in English | MEDLINE | ID: mdl-33023184

ABSTRACT

As a regulator of cellular inflammation and proliferation, cytosolic phospholipase A2 α (cPLA2α) is a promising therapeutic target for psoriasis; indeed, the cPLA2α inhibitor AVX001 has shown efficacy against plaque psoriasis in a phase I/IIa clinical trial. To improve our understanding of the anti-psoriatic properties of AVX001, we sought to determine how the compound modulates inflammation and keratinocyte hyperproliferation, key characteristics of the psoriatic epidermis. We measured eicosanoid release from human peripheral blood mononuclear cells (PBMC) and immortalized keratinocytes (HaCaT) and studied proliferation in HaCaT grown as monolayers and stratified cultures. We demonstrated that inhibition of cPLA2α using AVX001 produced a balanced reduction of prostaglandins and leukotrienes; significantly limited prostaglandin E2 (PGE2) release from both PBMC and HaCaT in response to pro-inflammatory stimuli; attenuated growth factor-induced arachidonic acid and PGE2 release from HaCaT; and inhibited keratinocyte proliferation in the absence and presence of exogenous growth factors, as well as in stratified cultures. These data suggest that the anti-psoriatic properties of AVX001 could result from a combination of anti-inflammatory and anti-proliferative effects, probably due to reduced local eicosanoid availability.


Subject(s)
Dinoprostone/genetics , Group IV Phospholipases A2/genetics , Inflammation/drug therapy , Psoriasis/drug therapy , Celecoxib/pharmacology , Cell Proliferation/drug effects , Eicosanoids/pharmacology , Fatty Acids, Omega-3/genetics , Fatty Acids, Omega-3/pharmacology , Group IV Phospholipases A2/antagonists & inhibitors , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/pathology , Keratinocytes/drug effects , Leukocytes, Mononuclear/drug effects , Lipopolysaccharides/toxicity , Naproxen/pharmacology , Psoriasis/genetics , Psoriasis/pathology
17.
Am J Pathol ; 190(9): 1782-1788, 2020 09.
Article in English | MEDLINE | ID: mdl-32650004

ABSTRACT

Severe coronavirus disease 2019 (COVID-19) symptoms, including systemic inflammatory response and multisystem organ failure, are now affecting thousands of infected patients and causing widespread mortality. Coronavirus infection causes tissue damage, which triggers the endoplasmic reticulum stress response and subsequent eicosanoid and cytokine storms. Although proinflammatory eicosanoids, including prostaglandins, thromboxanes, and leukotrienes, are critical mediators of physiological processes, such as inflammation, fever, allergy, and pain, their roles in COVID-19 are poorly characterized. Arachidonic acid-derived epoxyeicosatrienoic acids could alleviate the systemic hyperinflammatory response in COVID-19 infection by modulating endoplasmic reticulum stress and stimulating the resolution of inflammation. Soluble epoxide hydrolase (sEH) inhibitors, which increase endogenous epoxyeicosatrienoic acid levels, exhibit potent anti-inflammatory activity and inhibit various pathologic processes in preclinical disease models, including pulmonary fibrosis, thrombosis, and acute respiratory distress syndrome. Therefore, targeting eicosanoids and sEH could be a novel therapeutic approach in combating COVID-19. In this review, we discuss the predominant role of eicosanoids in regulating the inflammatory cascade and propose the potential application of sEH inhibitors in alleviating COVID-19 symptoms. The host-protective action of omega-3 fatty acid-derived epoxyeicosanoids and specialized proresolving mediators in regulating anti-inflammation and antiviral response is also discussed. Future studies determining the eicosanoid profile in COVID-19 patients or preclinical models are pivotal in providing novel insights into coronavirus-host interaction and inflammation modulation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Animals , Betacoronavirus/pathogenicity , COVID-19 , Eicosanoids/pharmacology , Eicosanoids/therapeutic use , Endoplasmic Reticulum Stress/drug effects , Humans , Pandemics , SARS-CoV-2
18.
J Hypertens ; 38(9): 1802-1810, 2020 09.
Article in English | MEDLINE | ID: mdl-32384390

ABSTRACT

OBJECTIVES: The global morbidity and mortality related to hypertension and associated disorders increases continuously and novel therapeutic strategies are still in high demand. Increasing evidence suggests the important role in blood pressure regulation of cytochrome P-450-dependent metabolites of arachidonic acid. Epoxyeicosatrienoic acids (EETs) induce vasodilation and natriuresis, and have renoprotective and anti-inflammatory properties. 20-HETE is an arachidonic acid metabolite with both prohypertensive and antihypertensive activities. To explore the pathophysiological role of arachidonic acid metabolites in more detail, we examined the antihypertensive efficiency of EET-A, a stable analog of 14,15-EET, and of AAA, a novel antagonist of the 20-HETE receptors. METHODS: Male spontaneously hypertensive rats (SHR) were treated for 5 weeks with EET-A, AAA or the combination; age-matched untreated SHR and normotensive Wistar-Kyoto rats served as controls. EET-A and AAA were administered in drinking water at 10 mg/kg/day each. SBP was measured by telemetry and urine, blood, and tissue samples were collected for relevant analyses. RESULTS: EET-A or AAA given alone had no significant effect on SHR blood pressure. In contrast, combined treatment with AAA and EET-A was significantly antihypertensive, causing a decrease in SBP from 180 ±â€Š3 to 160 ±â€Š5 mmHg (P < 0.05). Additionally, the combined treatment attenuated cardiac hypertrophy, decreased kidney ANG II level, increased natriuresis, and increased the excretion of nitric oxide metabolites. CONCLUSION: Considering the beneficial impact of the combined treatment with EET-A and AAA on SHR blood pressure and cardiovascular and renal function, we suggest that the treatment is a promising therapeutic strategy for human hypertension.


Subject(s)
Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Eicosanoids/pharmacology , Hydroxyeicosatetraenoic Acids/antagonists & inhibitors , Hypertension/metabolism , Animals , Male , Rats , Rats, Inbred SHR
19.
Curr Opin Clin Nutr Metab Care ; 23(2): 76-81, 2020 03.
Article in English | MEDLINE | ID: mdl-31904604

ABSTRACT

PURPOSE OF REVIEW: Sepsis is a life-threatening condition caused by a dysregulated host response to infection that remains a huge clinical challenge. Recent evidence indicates that bioactive lipid mediators derived from polyunsaturated fatty acids termed specialized pro-resolving mediators (SPMs) are promising new candidates for treating critical illness. RECENT FINDINGS: We highlight herein the protective actions of SPMs in experimental sepsis, cardiac dysfunction, and also lung and cerebral injury, and discuss their mechanisms of action. We also emphasize that failed resolution responses and dysregulated SPM pathways may provide an explanation for the ongoing chronic inflammation in many diseases including chronic heart failure. SUMMARY: Importantly, monitoring plasma SPM profiles can predict patient outcomes in sepsis indicating their utility as new early biomarkers that may help stratify patients upon ICU admission.


Subject(s)
Docosahexaenoic Acids/analogs & derivatives , Eicosanoids/pharmacology , Eicosapentaenoic Acid/analogs & derivatives , Sepsis/drug therapy , Signal Transduction/drug effects , Biomarkers/blood , Critical Illness/therapy , Docosahexaenoic Acids/blood , Eicosanoids/blood , Eicosapentaenoic Acid/blood , Humans , Sepsis/blood
20.
J Cell Mol Med ; 24(1): 862-874, 2020 01.
Article in English | MEDLINE | ID: mdl-31749335

ABSTRACT

Growing evidence has well established the protective effects of CYP2J2/EET on the cardiovascular system. The aim of the present study was to determine whether CYP2J2/EET has a preventive effect on atrial fibrillation (AF) and to investigate the underlying mechanisms. Wild-type mice were injected with or without AAV9-CYP2J2 before abdominal aortic constriction (AAC) operation. After 8 weeks, compared with wild-type mice, AAC mice display higher AF inducibility and longer AF durations, which were remarkably attenuated with AAV9-CYP2J2. Also, AAV9-CYP2J2 reduced atrial fibrosis area and the deposit of collagen-I/III in AAC mice, accompanied by the blockade of TGF-ß/Smad-2/3 signalling pathways, as well as the recovery in Smad-7 expression. In vitro, isolated atrial fibroblasts were administrated with TGF-ß1, EET, EEZE, GW9662, SiRNA Smad-7 and pre-MiR-21, and EET was demonstrated to restrain the differentiation of atrial fibroblasts largely dependent on Smad-7, due to the inhibition of EET on MiR-21. In addition, increased inflammatory cytokines, as well as activated NF-κB pathways induced by AAC surgery, were also significantly blunted by AAV9-CYP2J2 treatment. These effects of CYP2J2/EET were partially blocked by GW9662, the antagonist of PPAR-γ. In conclusion, this study revealed that CYP2J2/EET ameliorates atrial fibrosis through modulating atrial fibroblasts activation by disinhibition of MiR-21 on Smad-7, and attenuates atrial inflammatory response by repressing NF-κB pathways, reducing the vulnerability to AF, and CYP2J2/EET exerts its role at least partially through PPAR-γ activation. Our findings might provide a novel upstream therapeutic strategy for AF.


Subject(s)
Aorta, Abdominal/pathology , Arterial Pressure , Atrial Fibrillation/prevention & control , Constriction, Pathologic/complications , Cytochrome P-450 Enzyme System/administration & dosage , Eicosanoids/pharmacology , Protective Agents/pharmacology , Animals , Atrial Fibrillation/etiology , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Cytochrome P-450 CYP2J2 , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Cytokines/metabolism , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...