Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.926
Filter
1.
Pak J Pharm Sci ; 37(3): 705-713, 2024 May.
Article in English | MEDLINE | ID: mdl-39340861

ABSTRACT

The investigation's aim was to explore the medical usefulness and mechanism of GBE in the management of elderly ischemic cerebrovascular disease (ICVD).120 cases of elderly patients with ICVD admitted to our hospital from August 2022 to August 2023 were chosen as participants for this research and the sufferer were allocated to the conventional (60 cases) and GBE group (60 cases) using the method of randomized number. NIHSS score, Barthel index, hemodynamic indexes, serum inflammatory factor levels, platelet-activating factor (PAF) and clinical efficacy were recorded before (T0) and after treatment(T1),and recorded the adverse reactions of the two groups during the treatment. At T1, NIHSS score, WBLSV, PV, HCT, TNF-α, IL-6 and PAF in the two groups, which were all notably reduced compared to T0 and the Barthel index demonstrated a significant increase compared to its T0 value (P<0.05). At T1, GBE group exhibited notably reductions in NIHSS score, WBLSV, PV, HCT, TNF-α, IL-6 and PAF compared to conventional group, whereas Barthel index and the total effective rate were considerably elevated (P<0.05). Incidence of adverse reactions were similar in both groups (P>0.05). GBE has good therapeutic benefits in managing elderly ICVD, effectively facilitate the recuperation of patients' neurological function, has obvious anti-inflammatory effect, improves patients' cerebral circulation and hemorheology indexes and makes the patients' daily life ability significantly improved, which has a good clinical application value.


Subject(s)
Ginkgo biloba , Plant Extracts , Humans , Ginkgo biloba/chemistry , Aged , Male , Female , Plant Extracts/therapeutic use , Plant Extracts/adverse effects , Treatment Outcome , Brain Ischemia/drug therapy , Brain Ischemia/blood , Platelet Activating Factor/metabolism , Aged, 80 and over , Tumor Necrosis Factor-alpha/blood , Hemodynamics/drug effects , Interleukin-6/blood , Ginkgo Extract
2.
Biol Pharm Bull ; 47(9): 1467-1476, 2024.
Article in English | MEDLINE | ID: mdl-39218668

ABSTRACT

Since its first discovery as a bioactive phospholipid inducing potent platelet aggregation, platelet-activating factor (PAF) has been shown to be involved in a wide variety of inflammatory and allergic disease states. Many pharmacological studies in the 1980s and 1990s also showed that PAF induces endothelium-dependent vascular relaxation and contraction of various smooth muscles (SMs), including those in the airway, gastrointestinal organs, and uterus. However, since the late 1990s, there have been few reports on the SM contractions induced by PAF. The lower urinary tract (LUT), particularly the urinary bladder (UB) has attracted recent attention in SM pharmacology research because patients with LUT dysfunctions including overactive bladder are increasing as the population ages. In addition, recent clinical studies have implicated the substantial role of PAF in the inflammatory state in LUT because its production increases with smoking and with cancer. However, the effects of PAF on mechanical activities of LUT SMs including UBSM have not been investigated to date. Recently, we found that PAF very strongly increased mechanical activities of UBSM in guinea pigs and mice, and partly elucidated the possible mechanisms underlying these actions of PAF. In this review, we describe the effects of PAF on LUT SMs by introducing our recent findings obtained in isolated UBSMs and discuss the physiological and pathophysiological significance. We also introduce our data showing the effects of PAF on the SM mechanical activities of genital tissues (prostate and vas deferens).


Subject(s)
Muscle Contraction , Muscle, Smooth , Platelet Activating Factor , Platelet Activating Factor/pharmacology , Platelet Activating Factor/metabolism , Animals , Humans , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Muscle, Smooth/metabolism , Muscle Contraction/drug effects , Urinary Bladder/drug effects , Urinary Bladder/metabolism , Urinary Bladder/physiology , Male , Female
3.
Sheng Li Xue Bao ; 76(4): 537-546, 2024 Aug 25.
Article in Chinese | MEDLINE | ID: mdl-39192787

ABSTRACT

This paper aimed to investigate the effects of exercise on hepatic platelet-activating factor (PAF) metabolism in rats fed a high-fat diet. Thirty-two male Sprague-Dawley (SD) rats were divided into control group (C), high-fat diet group (H), exercise group (EC), and high-fat diet+exercise group (EH). Serum lipids, glucose, insulin and markers of hepatic injury after a 16-week dietary and/or exercise intervention (60 min/day, 6 times/week) were measured by biochemical analysis; liver lipidomic profiles were analyzed by liquid chromatograph-mass spectrometer (LC-MS). Gene and protein expression of enzymes related to PAF metabolism were determined by qPCR and Western blot respectively. The results showed that high-fat diet feeding significantly increased the levels of low-density lipoprotein-cholesterol (LDL-C) and liver injury markers including purine nucleoside phosphorylase (PNP) and malondialdehyde (MDA) in rats, which were decreased by exercise. Furthermore, high-fat diet feeding significantly increased the hepatic PAF content, which was also attenuated by exercise. In addition, although high-fat diet treatment resulted in an increase in the expression of both PAF synthetase (PAF-CPT and PLA2) and hydrolase (Lp-PLA2 and PAF-AH(II)), induction of PAF synthetase was much greater than that of PAF hydrolase. While exercise increased the expression of Lp-PLA2 and PAF-AH(II) and decreased the expression of PAF-CPT and PLA2, key PAF synthesizing enzymes. In conclusion, high-fat diet-induced increase in hepatic PAF content is mainly due to the increase of its pathological synthesis at the translational level. Exercise reduces hepatic PAF content in high-fat fed rats by increasing PAF hydrolysis and decreasing its synthesis.


Subject(s)
Diet, High-Fat , Liver , Physical Conditioning, Animal , Platelet Activating Factor , Animals , Male , Rats , Diet, High-Fat/adverse effects , Liver/metabolism , Physical Conditioning, Animal/physiology , Platelet Activating Factor/metabolism
4.
Proc Natl Acad Sci U S A ; 121(35): e2406748121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39178229

ABSTRACT

Chronic inflammatory milieu in the tumor microenvironment (TME) leads to the recruitment and differentiation of myeloid-derived suppressor cells (MDSCs). Polymorphonuclear (PMN)-MDSCs, which are phenotypically and morphologically defined as a subset of neutrophils, cause major immune suppression in the TME, posing a significant challenge in the development of effective immunotherapies. Despite recent advances in our understanding of PMN-MDSC functions, the mechanism that gives rise to immunosuppressive neutrophils within the TME remains elusive. Both in vivo and in vitro, newly recruited neutrophils into the tumor sites remained activated and highly motile for several days and developed immunosuppressive phenotypes, as indicated by increased arginase 1 (Arg1) and dcTrail-R1 expression and suppressed anticancer CD8 T cell cytotoxicity. The strong suppressive function was successfully recapitulated by incubating naive neutrophils with cancer cell culture supernatant in vitro. Cancer metabolite secretome analyses of the culture supernatant revealed that both murine and human cancers released lipid mediators to induce the differentiation of immunosuppressive neutrophils. Liquid chromatography-mass spectrometry (LC-MS) lipidomic analysis identified platelet-activation factor (PAF; 1-O-alkyl-2-acetyl-sn-glycero-3-phosphocholine) as a common tumor-derived lipid mediator that induces neutrophil differentiation. Lysophosphatidylcholine acyltransferase 2 (LPCAT2), the PAF biosynthetic enzyme, is up-regulated in human pancreatic ductal adenocarcinoma (PDAC) and shows an unfavorable correlation with patient survival across multiple cancer types. Our study identifies PAF as a lipid-driven mechanism of MDSC differentiation in the TME, providing a potential target for cancer immunotherapy.


Subject(s)
Cell Differentiation , Myeloid-Derived Suppressor Cells , Neutrophils , Platelet Activating Factor , Tumor Microenvironment , Neutrophils/immunology , Neutrophils/metabolism , Humans , Animals , Mice , Tumor Microenvironment/immunology , Platelet Activating Factor/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/immunology , Cell Line, Tumor , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mice, Inbred C57BL
5.
Reproduction ; 168(4)2024 10 01.
Article in English | MEDLINE | ID: mdl-39056485

ABSTRACT

In Brief: In many mammals, the lipid platelet-activating factor (PAF) has important functions in female reproduction and fertility. This study shows that PAF is present in the reproductive tissues of mares and is involved in processes related to ovulation and early pregnancy. Abstract: Platelet-activating factor (PAF) has been implicated in a number of reproductive processes ranging from ovulation to embryo motility but has not been widely explored in the mare. To identify the presence and examine the role of PAF in the equine periconception processes, targeted mass spectrometry coupled with chromatographic separation was performed on equine follicular fluid (FF), and PAF was quantitatively detected. Subsequently, untargeted high-resolution mass spectrometry-based lipidomic analysis was carried out to quantify PAF in different-sized pre-ovulatory follicles, whereby different molecular species of PAF, PAF (14:0) and PAF (16:1), were both seen to be increasing with follicle diameter. These findings suggest that PAF within FF is increasing as preovulatory follicles approach ovulation. Additionally, immunofluorescence staining identified the PAF receptor in the luminal pericellular, apical, and basal aspect of equine oviductal epithelial cells. Lastly, an equine oviductal epithelial organoid model was generated and showed that the addition of PAF significantly increased the ciliary beat frequency (CBF) (Hz), an action consistent with a role for PAF in embryo migration. It is proposed that the local action of PAF on the ciliated cells of the oviduct propels both the oocyte and the conceptus towards the uterus. In the mare, it appears that PAF is a contributor during the periconception period, potentially being a mediator in the mechanisms of ovulation and in the dialogue of very early pregnancy.


Subject(s)
Ovulation , Platelet Activating Factor , Animals , Horses/physiology , Female , Platelet Activating Factor/metabolism , Platelet Activating Factor/pharmacology , Pregnancy , Ovulation/physiology , Ovarian Follicle/metabolism , Ovarian Follicle/physiology , Follicular Fluid/metabolism , Fertilization/physiology
6.
Commun Biol ; 7(1): 782, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951147

ABSTRACT

Acute immune responses with excess production of cytokines, lipid/chemical mediators, or coagulation factors, often result in lethal damage. In addition, the innate immune system utilizes multiple types of receptors that recognize neurotransmitters as well as pathogen-associated molecular patterns, making immune responses complex and clinically unpredictable. We here report an innate immune and adrenergic link inducing lethal levels of platelet-activating factor. Injecting mice with toll-like receptor (TLR) 4 ligand lipopolysaccharide (LPS), cell wall N-glycans of Candida albicans, and the α2-adrenergic receptor (α2-AR) agonist medetomidine induces lethal damage. Knocking out the C-type lectin Dectin-2 prevents the lethal damage. In spleen, large amounts of platelet-activating factor (PAF) are detected, and knocking out lysophospholipid acyltransferase 9 (LPLAT9/LPCAT2), which encodes an enzyme that converts inactive lyso-PAF to active PAF, protects mice from the lethal damage. These results reveal a linkage/crosstalk between the nervous and the immune system, possibly inducing lethal levels of PAF.


Subject(s)
Platelet Activating Factor , Animals , Platelet Activating Factor/metabolism , Mice , Mice, Knockout , Mice, Inbred C57BL , Lipopolysaccharides , Candida albicans , Immunity, Innate , Male , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Adrenergic alpha-2 Receptor Agonists/pharmacology
8.
Cell Rep ; 43(7): 114422, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38943642

ABSTRACT

Platelet-activating factor (PAF) is a potent phospholipid mediator crucial in multiple inflammatory and immune responses through binding and activating the PAF receptor (PAFR). However, drug development targeting the PAFR has been limited, partly due to an incomplete understanding of its activation mechanism. Here, we present a 2.9-Å structure of the PAF-bound PAFR-Gi complex. Structural and mutagenesis analyses unveil a specific binding mode of PAF, with the choline head forming cation-π interactions within PAFR hydrophobic pocket, while the alkyl tail penetrates deeply into an aromatic cleft between TM4 and TM5. Binding of PAF modulates conformational changes in key motifs of PAFR, triggering the outward movement of TM6, TM7, and helix 8 for G protein coupling. Molecular dynamics simulation suggests a membrane-side pathway for PAF entry into PAFR via the TM4-TM5 cavity. By providing molecular insights into PAFR signaling, this work contributes a foundation for developing therapeutic interventions targeting PAF signal axis.


Subject(s)
Platelet Activating Factor , Platelet Membrane Glycoproteins , Receptors, G-Protein-Coupled , Platelet Membrane Glycoproteins/metabolism , Platelet Membrane Glycoproteins/chemistry , Platelet Activating Factor/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/chemistry , Humans , Molecular Dynamics Simulation , Protein Binding , Binding Sites , HEK293 Cells , Signal Transduction
9.
Life Sci ; 347: 122617, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38608835

ABSTRACT

BACKGROUND: Acute myocardial infarction (AMI) is one of the main causes of death. It is quite obvious that there is an urgent need to develop new approaches for treatment of AMI. OBJECTIVE: This review analyzes data on the role of platelets in the regulation of cardiac tolerance to ischemia/reperfusion (I/R). METHODS: It was performed a search of topical articles using PubMed databases. FINDINGS: Platelets activated by a cholesterol-enriched diet, thrombin, and myocardial ischemia exacerbate I/R injury of the heart. The P2Y12 receptor antagonists, remote ischemic postconditioning and conditioning alter the properties of platelets. Platelets acquire the ability to increase cardiac tolerance to I/R. Platelet-derived growth factors (PDGFs) increase tolerance of cardiomyocytes and endothelial cells to I/R. PDGF receptors (PDGFRs) were found in cardiomyocytes and endothelial cells. PDGFs decrease infarct size and partially abrogate adverse postinfarction remodeling. Protein kinase C, phosphoinositide 3-kinase, and Akt involved in the cytoprotective effect of PDGFs. Vascular endothelial growth factor increased cardiac tolerance to I/R and alleviated adverse postinfarction remodeling. The platelet-activating factor (PAF) receptor inhibitors increase cardiac tolerance to I/R in vivo. PAF enhances cardiac tolerance to I/R in vitro. It is possible that PAF receptor inhibitors could protect the heart by blocking PAF receptor localized outside the heart. PAF protects the heart through activation of PAF receptor localized in cardiomyocytes or endothelial cells. Reactive oxygen species and kinases are involved in the cardioprotective effect of PAF. CONCLUSION: Platelets play an important role in the regulation of cardiac tolerance to I/R.


Subject(s)
Blood Platelets , Myocardial Reperfusion Injury , Platelet Activating Factor , Platelet-Derived Growth Factor , Vascular Endothelial Growth Factor A , Humans , Animals , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Blood Platelets/metabolism , Platelet Activating Factor/metabolism , Platelet-Derived Growth Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/prevention & control , Myocardial Infarction/pathology
10.
Front Biosci (Landmark Ed) ; 29(4): 159, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38682205

ABSTRACT

OBJECTIVE: The effect of the daily consumption of a low-fat yogurt (150 g) enriched with Platelet-Activating Factor receptor (PAF-R) antagonists, or the plain one, on gut microbiota and faecal metabolites was investigated in healthy overweight subjects. METHODS: A randomized, three-arm, double-blind, placebo-controlled, parallel-group study was performed that lasted 8 weeks. Blood and stools were collected and analyzed before and after the intervention. RESULTS: Our findings revealed that the intake of the enriched yogurt resulted in a significant increase in the levels of Bifidobacterium spp., Clostridium perfringens group and Firmicutes-to-Bacteroidetes (F/B) ratio. On the other hand, a significant increase in the levels of Lactobacillus and C. perfringens group was detected after the intake of the plain yogurt. The increase in the levels of C. perfringens group was inversely associated with the plasma catabolic enzyme of PAF, namely LpPLA2 (lipoprotein-associated phospholipase A2), a cardiovascular risk marker that has been linked with inflammation and atherosclerosis. Moreover, in the enriched with PAF-R antagonists yogurt group, the increased levels of C. perfringens group were also associated with lower PAF action assessed as ex vivo human platelet-rich plasma (PRP) aggregation. Additionally, a higher % increase in molar ratio of Branched Short Chain Fatty Acids (BSCFAs) was detected for both yogurt groups after the 8 week-intervention compared to control. The consumption of the enriched yogurt also resulted in a significant drop in faecal caproic levels and a trend for lower ratio of butyrate to total volatile fatty acids (VFAs) compared to baseline levels. CONCLUSION: Yogurt consumption seems to favorably affect gut microbiota while its enrichment with PAF-R antagonists from olive oil by-products, may provide further benefits in healthy overweight subjects. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov (NCT02259205).


Subject(s)
Feces , Gastrointestinal Microbiome , Olive Oil , Overweight , Platelet Activating Factor , Yogurt , Humans , Yogurt/microbiology , Gastrointestinal Microbiome/drug effects , Overweight/metabolism , Overweight/microbiology , Overweight/diet therapy , Feces/microbiology , Feces/chemistry , Male , Female , Adult , Double-Blind Method , Middle Aged , Platelet Activating Factor/metabolism , Receptors, G-Protein-Coupled/metabolism , Platelet Membrane Glycoproteins/metabolism , Platelet Membrane Glycoproteins/antagonists & inhibitors
11.
J Leukoc Biol ; 116(4): 766-778, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38531065

ABSTRACT

Thermal burn injuries can result in significant morbidity and mortality. The combination of ethanol intoxication with thermal burn injury results in increased morbidity through an exaggerated inflammatory response involving many organs. Recent studies have linked involvement of the lipid mediator platelet-activating factor (PAF) in the pathology associated with intoxicated thermal burn injury (ITBI). The present studies tested the roles of PAF and the elevated levels of subcellular microvesicle particles (MVP) generated in response to ITBI in the subsequent multiorgan toxicity. First, thermal burn injury of HaCaT keratinocytes preincubated with ethanol resulted in augmented MVP release, which was blocked by inhibiting the PAF-generating enzyme cytosolic phospholipase A2 and the PAF receptor (PAFR). Second, ITBI of mice resulted in increased proinflammatory cytokine production and neutrophilic inflammation in multiple organs, which were not present in mice deficient in PAFRs or the MVP-generating enzyme acid sphingomyelinase (aSMase). Moreover, the increased bacterial translocation from the gut to mesenteric lymph nodes previously reported in murine ITBI was also dependent on PAFR and aSMase. MVP released from ITBI-treated keratinocytes contained high levels of PAFR agonistic activity. Finally, use of topical aSMase inhibitor imipramine following ITBI attenuated the widespread organ inflammatory response of ITBI, suggesting a potential therapeutic for this condition. These studies provide evidence for PAF-enriched MVP generated in skin, which then act on the gut PAFR, resulting in bacterial translocation as the mechanism for the multiorgan dysfunction associated with ITBI. Inasmuch as aSMase inhibitors are widely available, these studies could result in effective treatments for ITBI.


Subject(s)
Burns , Cell-Derived Microparticles , Keratinocytes , Platelet Activating Factor , Sphingomyelin Phosphodiesterase , Animals , Burns/pathology , Burns/complications , Keratinocytes/metabolism , Keratinocytes/pathology , Platelet Activating Factor/metabolism , Humans , Cell-Derived Microparticles/metabolism , Mice , Sphingomyelin Phosphodiesterase/metabolism , Receptors, G-Protein-Coupled/metabolism , Platelet Membrane Glycoproteins/metabolism , Ethanol/toxicity , Mice, Inbred C57BL , Bacterial Translocation/drug effects , Cytokines/metabolism , Male , Mice, Knockout
12.
Am J Respir Cell Mol Biol ; 70(6): 482-492, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38377392

ABSTRACT

Cigarette smoking is known to be the leading cause of chronic obstructive pulmonary disease (COPD). However, the detailed mechanisms have not been elucidated. PAF (platelet-activating factor), a potent inflammatory mediator, is involved in the pathogenesis of various respiratory diseases such as bronchial asthma and COPD. We focused on LPLAT9 (lysophospholipid acyltransferase 9), a biosynthetic enzyme of PAF, in the pathogenesis of COPD. LPLAT9 gene expression was observed in excised COPD lungs and single-cell RNA sequencing data of alveolar macrophages (AMs). LPLAT9 was predominant and upregulated in AMs, particularly monocyte-derived AMs, in patients with COPD. To identify the function of LPLAT9/PAF in AMs in the pathogenesis of COPD, we exposed systemic LPLAT9-knockout (LPALT9-/-) mice to cigarette smoke (CS). CS increased the number of AMs, especially the monocyte-derived fraction, which secreted MMP12 (matrix metalloprotease 12). Also, CS augmented LPLAT9 phosphorylation/activation on macrophages and, subsequently, PAF synthesis in the lung. The LPLAT9-/- mouse lung showed reduced PAF production after CS exposure. Intratracheal PAF administration accumulated AMs by increasing MCP1 (monocyte chemoattractant protein-1). After CS exposure, AM accumulation and subsequent pulmonary emphysema, a primary pathologic change of COPD, were reduced in LPALT9-/- mice compared with LPLAT9+/+ mice. Notably, these phenotypes were again worsened by LPLAT9+/+ bone marrow transplantation in LPALT9-/- mice. Thus, CS-induced LPLAT9 activation in monocyte-derived AMs aggravated pulmonary emphysema via PAF-induced further accumulation of AMs. These results suggest that PAF synthesized by LPLAT9 has an important role in the pathogenesis of COPD.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase , Macrophages, Alveolar , Mice, Knockout , Platelet Activating Factor , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Animals , Female , Humans , Male , Mice , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Cigarette Smoking/adverse effects , Cigarette Smoking/metabolism , Lung/metabolism , Lung/pathology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Matrix Metalloproteinase 12/metabolism , Matrix Metalloproteinase 12/genetics , Mice, Inbred C57BL , Platelet Activating Factor/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Pulmonary Emphysema/genetics
13.
Prostaglandins Other Lipid Mediat ; 172: 106818, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38340978

ABSTRACT

Platelet-activating factor (PAF) plays a significant role in several leucocyte functions, including platelet aggregation and inflammation. Additionally, PAF has a role in the behavioral and physiological changes in mammals. However, the effect of PAF has not been well studied in birds. Therefore, the study aimed to determine if PAF affects feeding behavior, voluntary activity, cloacal temperature, and feed passage through the digestive tract in chicks (Gallus gallus). We also studied the involvement of PAF in the innate immune system induced by lipopolysaccharide (LPS), a cell wall component of gram-negative bacteria. Both intraperitoneal (IP) and intracerebroventricular (ICV) injections of PAF significantly decreased food intake. IP injection of PAF significantly decreased voluntary activity and slowed the feed passage from the crop, whereas ICV injection had no effect. Conversely, ICV injection of PAF significantly increased the cloacal temperature, but IP injection had no effect. The IP injection of LPS significantly reduced the mRNA expression of lysophosphatidylcholine acyltransferase 2, an enzyme responsible for PAF production in the heart and pancreas. On the other hand, LPS significantly increased the mRNA expression of the PAF receptor in the peripheral organs. The present study shows that PAF influences behavioral and physiological responses and is related to the response against bacterial infections in chicks.


Subject(s)
Body Temperature , Chickens , Cloaca , Crop, Avian , Eating , Platelet Activating Factor , Animals , Male , Body Temperature/drug effects , Cloaca/drug effects , Cloaca/physiology , Crop, Avian/drug effects , Crop, Avian/metabolism , Eating/drug effects , Feeding Behavior/drug effects , Lipopolysaccharides/pharmacology , Platelet Activating Factor/pharmacology , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics
14.
Am J Pathol ; 194(6): 862-878, 2024 06.
Article in English | MEDLINE | ID: mdl-38403163

ABSTRACT

Nearly 70% of preterm deliveries occur spontaneously, and the clinical pathways involved include preterm labor and preterm premature rupture of membranes. Prediction of preterm delivery is considered crucial due to the significant effects of preterm birth on health and the economy at both the personal and community levels. Although similar inflammatory processes occur in both term and preterm delivery, the premature activation of these processes or exaggerated inflammatory response triggered by infection or sterile factors leads to preterm delivery. Platelet activating factor (PAF) is a phosphoglycerylether lipid mediator of inflammation that is implicated in infections, cancers, and various chronic diseases and disorders including cardiovascular, renal, cerebrovascular, and central nervous system diseases. In gestational tissues, PAF mediates the inflammatory pathways that stimulate the effector mechanisms of labor, including myometrial contraction, cervical dilation, and fetal membrane rupture. Women with preterm labor and preterm premature rupture of membranes have increased levels of PAF in their amniotic fluid. In mice, the intrauterine or intraperitoneal administration of carbamyl PAF activates inflammation in gestational tissues, thereby eliciting preterm delivery. This review summarizes recent research on PAF as an important inflammatory mediator in preterm delivery and in other inflammatory disorders, highlighting its potential value for prediction, intervention, and prevention of these diseases.


Subject(s)
Inflammation , Platelet Activating Factor , Premature Birth , Humans , Platelet Activating Factor/metabolism , Female , Pregnancy , Animals , Inflammation/metabolism , Inflammation/pathology , Premature Birth/metabolism , Fetal Membranes, Premature Rupture/metabolism , Obstetric Labor, Premature/metabolism
15.
Int J Mol Sci ; 25(4)2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38396790

ABSTRACT

Platelet-activating factor (PAF) is a phospholipid-derived inflammatory mediator that triggers various inflammatory conditions, including eosinophil activation and recruitment. This study aimed to evaluate the expressions of PAF-metabolism-associated genes, namely genes coding the enzymes involved in PAF synthesis (LPCAT1, LPCAT2, LPCAT3, and LPCAT4), PAF degradation (PAFAH1B2, PAFAH1B3, and PAFAH2), and the gene for the PAF receptor (PTAFR) in subtypes of CRSwNP classified by clinical- or hierarchal-analysis-based classifications. Transcriptomic analysis using bulk RNA barcoding and sequencing (BRB-seq) was performed with CRSwNP, including eosinophilic CRS (ECRS) (n = 9), nonECRS (n = 8), ECRS with aspirin-exacerbated respiratory disease (Asp) (n = 3), and controls with a normal uncinate process mucosa (n = 6). PTAFR was only upregulated in ECRS and nonECRS. In the hierarchical cluster analysis with clusters 1 and 2 reflecting patients with low-to-moderate and high levels of type 2 inflammation, respectively, cluster 1 exhibited a significant downregulation of LPCAT2 and an upregulation of PTAFR expression, while cluster 2 showed an upregulation of LPCAT1, PAFAH1B2, and PTAFR and downregulation of PAFAH2 expression. Understanding this strong PAF-associated pathophysiology in the severe type 2 inflammation group could provide valuable insights into the treatment and management of CRSwNP.


Subject(s)
Nasal Polyps , Rhinitis , Rhinosinusitis , Sinusitis , Humans , Rhinitis/pathology , Platelet Activating Factor/genetics , Platelet Activating Factor/metabolism , Nasal Mucosa/metabolism , RNA/metabolism , Nasal Polyps/pathology , Sinusitis/metabolism , Inflammation/metabolism , Chronic Disease , Cluster Analysis , Eosinophils/metabolism
16.
Photochem Photobiol ; 100(5): 1457-1466, 2024.
Article in English | MEDLINE | ID: mdl-38287748

ABSTRACT

Photosensitivity can be due to numerous causes. The photosensitivity associated with deficiency of xeroderma pigmentosum type A (XPA) has been previously shown to be associated with excess levels of the lipid mediator platelet-activating factor (PAF) generated by the keratinocyte. As PAF has been reported to trigger the production of subcellular microvesicle particles (MVP) due to the enzyme acid sphingomyelinase (aSMase), the goal of these studies was to discern if PAF and aSMase could serve as therapeutic targets for the XPA deficiency photosensitivity. HaCaT keratinocytes lacking XPA generated greater levels of MVP in comparison to control cells. Mice deficient in XPA also generated enhanced MVP levels in skin and in plasma in response to UV radiation. Use of a genetic strategy with mice deficient in both XPA and PAF receptors revealed that these mice generated less MVP release as well as decreased skin erythema and cytokine release compared to XPA knockout mice alone. Finally, the aSMase inhibitor imipramine blocked UV-induced MVP release in HaCaT keratinocytes, as well as XPA knockout mice. These studies support the concept that the photosensitivity associated with XPA involves PAF- and aSMase-mediated MVP release and provides a potential pharmacologic target in treating this form of photosensitivity.


Subject(s)
Keratinocytes , Mice, Knockout , Ultraviolet Rays , Xeroderma Pigmentosum , Keratinocytes/radiation effects , Keratinocytes/metabolism , Animals , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/metabolism , Mice , Humans , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/deficiency , Xeroderma Pigmentosum Group A Protein/metabolism , Xeroderma Pigmentosum Group A Protein/genetics , Photosensitivity Disorders , Platelet Activating Factor/metabolism , Cell-Derived Microparticles/metabolism , Imipramine/pharmacology
17.
Nat Chem Biol ; 20(7): 835-846, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38287154

ABSTRACT

Synchronized ferroptosis contributes to nephron loss in acute kidney injury (AKI). However, the propagation signals and the underlying mechanisms of the synchronized ferroptosis for renal tubular injury remain unresolved. Here we report that platelet-activating factor (PAF) and PAF-like phospholipids (PAF-LPLs) mediated synchronized ferroptosis and contributed to AKI. The emergence of PAF and PAF-LPLs in ferroptosis caused the instability of biomembranes and signaled the cell death of neighboring cells. This cascade could be suppressed by PAF-acetylhydrolase (II) (PAFAH2) or by addition of antibodies against PAF. Genetic knockout or pharmacological inhibition of PAFAH2 increased PAF production, augmented synchronized ferroptosis and exacerbated ischemia/reperfusion (I/R)-induced AKI. Notably, intravenous administration of wild-type PAFAH2 protein, but not its enzymatically inactive mutants, prevented synchronized tubular cell death, nephron loss and AKI. Our findings offer an insight into the mechanisms of synchronized ferroptosis and suggest a possibility for the preventive intervention of AKI.


Subject(s)
Acute Kidney Injury , Ferroptosis , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/drug therapy , Ferroptosis/drug effects , Animals , Mice , Mice, Inbred C57BL , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Platelet Activating Factor/metabolism , Mice, Knockout , Humans , Male
18.
Sci Rep ; 13(1): 21637, 2023 12 07.
Article in English | MEDLINE | ID: mdl-38062077

ABSTRACT

Although it is well established that platelet-activated receptor (PAF) and protease-activated receptor 2 (PAR2) play a pivotal role in the pathophysiology of lung and airway inflammatory diseases, a role for a PAR2-PAFR cooperation in lung inflammation has not been investigated. Here, we investigated the role of PAR2 in PAF-induced lung inflammation and neutrophil recruitment in lungs of BALB/c mice. Mice were pretreated with the PAR2 antagonist ENMD1068, PAF receptor (PAFR) antagonist WEB2086, or aprotinin prior to intranasal instillation of carbamyl-PAF (C-PAF) or the PAR2 agonist peptide SLIGRL-NH2 (PAR2-AP). Leukocyte infiltration in bronchoalveolar lavage fluid (BALF), C-X-C motif ligand 1 (CXCL)1 and CXCL2 chemokines, myeloperoxidase (MPO), and N-acetyl-glycosaminidase (NAG) levels in BALF, or lung inflammation were evaluated. Intracellular calcium signaling, PAFR/PAR2 physical interaction, and the expression of PAR2 and nuclear factor-kappa B (NF-КB, p65) transcription factor were investigated in RAW 264.7 cells stimulated with C-PAF in the presence or absence of ENMD1068. C-PAF- or PAR2-AP-induced neutrophil recruitment into lungs was inhibited in mice pretreated with ENMD1068 and aprotinin or WEB2086, respectively. PAR2 blockade impaired C-PAF-induced neutrophil rolling and adhesion, lung inflammation, and production of MPO, NAG, CXCL1, and CXCL2 production in lungs of mice. PAFR activation reduced PAR2 expression and physical interaction of PAR2 and PAFR; co-activation is required for PAFR/PAR2 physical interaction. PAR2 blockade impaired C-PAF-induced calcium signal and NF-κB p65 translocation in RAW 264.7 murine macrophages. This study provides the first evidence for a cooperation between PAFR and PAR2 mediating neutrophil recruitment, lung inflammation, and macrophage activation.


Subject(s)
NF-kappa B , Pneumonia , Mice , Animals , NF-kappa B/metabolism , Platelet Activating Factor/metabolism , Receptor, PAR-2/genetics , Receptor, PAR-2/metabolism , Receptors, G-Protein-Coupled/metabolism , Aprotinin/metabolism , Neutrophil Infiltration , Transcriptional Activation , Pneumonia/chemically induced
19.
Molecules ; 28(19)2023 Oct 01.
Article in English | MEDLINE | ID: mdl-37836742

ABSTRACT

Inflammatory mediators constitute a recently coined term in the field of metal-based complexes with antiplatelet activities. Our strategy targets Platelet-Activating Factor (PAF) and its receptor, which is the most potent lipid mediator of inflammation. Thus, the antiplatelet (anti-PAF) potency of any substance could be exerted by inhibiting the PAF-induced aggregation in washed rabbit platelets (WRPs), which internationally is a well-accepted methodology. Herein, a series of mononuclear (mer-[Cr(pqx)Cl3(H2O]) (1), [Co(pqx)Cl2(DMF)] (2) (DMF = N,N'-dimethyl formamide), [Cu(pqx)Cl2(DMSO)] (3) (DMSO = dimethyl sulfoxide), [Zn(pqx)Cl2] (4)) and dinuclear complexes ([Mn(pqx)(H2O)2Cl2]2 (5), [Fe(pqx)Cl2]2 (6) and [Ni(pqx)Cl2]2 (7)) incorporating the 2-(2'-pyridyl)quinoxaline ligand (pqx), were biologically evaluated as inhibitors of the PAF- and thrombin-induced aggregation in washed rabbit platelets (WRPs). The molecular structure of the five-co-ordinate analog (3) has been elucidated by single-crystal X-ray diffraction revealing a trigonal bipyramidal geometry. All complexes are potent inhibitors of the PAF-induced aggregation in WRPs in the micromolar range. Complex (6) displayed a remarkable in vitro dual inhibition against PAF and thrombin, with IC50 values of 1.79 µM and 0.46 µM, respectively. Within the series, complex (5) was less effective (IC50 = 39 µM) while complex (1) was almost 12-fold more potent against PAF, as opposed to thrombin-induced aggregation. The biological behavior of complexes 1, 6 and 7 on PAF's basic metabolic enzymatic pathways reveals that they affect key biosynthetic and catabolic enzymes of PAF underlying the anti-inflammatory properties of the relevant complexes. The in vitro cytotoxic activities of all complexes in HEK293T (human embryonic kidney cells) and HeLa cells (cervical cancer cells) are described via the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. The results reveal that complex 3 is the most potent within the series.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Transition Elements , Animals , Humans , Rabbits , Platelet Aggregation , Platelet Activating Factor/pharmacology , Platelet Activating Factor/metabolism , Blood Platelets/metabolism , Thrombin/metabolism , Coordination Complexes/pharmacology , Coordination Complexes/metabolism , Ligands , Inflammation Mediators/metabolism , Dimethyl Sulfoxide/pharmacology , Quinoxalines/pharmacology , HEK293 Cells , HeLa Cells , Antineoplastic Agents/pharmacology , Transition Elements/metabolism
20.
Molecules ; 28(15)2023 Jul 27.
Article in English | MEDLINE | ID: mdl-37570665

ABSTRACT

Lidocaine, a local anesthetic, is known to possess anti-inflammatory properties. However, its clinical use is limited by inconveniences, such as its local synesthetic effects. This study evaluated lidocaine analogs designed and synthesized to overcome the disadvantages of lidocaine, having anti-inflammatory properties. Interleukin 5 (IL-5)-induced eosinophil activation and survival were evaluated using 36 lidocaine analogs with modified lidocaine structure on the aromatic or the acyl moiety or both. Eosinophil survival was evaluated using a CellTiter 96® aqueous cell proliferation assay kit. Superoxide production was determined using the superoxide dismutase-inhibitable reduction of cytochrome C method. Eosinophil cationic protein (ECP), IL-8, and transcription factor expression were determined using enzyme-linked immunosorbent assay. The platelet-activating factor (PAF)-induced migration assay was performed using a Transwell insert system. Compounds EI137 and EI341 inhibited IL-5-induced eosinophil survival and superoxide and ECP production in a concentration-dependent manner. These compounds also significantly reduced IL-8 production. Although compounds EI137 and EI341 significantly reduced phosphorylated ERK 1/2 expression, they did not influence other total and phosphorylated transcription factors. Moreover, 1000 µM of compound EI341 only inhibited PAF-induced migration of eosinophils. Lidocaine analogs EI137 and EI341 inhibited IL-5-mediated activation and survival of eosinophils. These compounds could be new therapeutic agents to treat eosinophilic inflammatory diseases.


Subject(s)
Eosinophils , Superoxides , Superoxides/metabolism , Lidocaine/pharmacology , Interleukin-5/metabolism , Interleukin-5/pharmacology , Interleukin-8/metabolism , Platelet Activating Factor/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL