Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Am J Physiol Regul Integr Comp Physiol ; 325(5): R446-R455, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37602385

ABSTRACT

Hypothermia develops during systemic anaphylaxis in rodents. The aim of this study was to elucidate the mechanism for the hypothermia by assessing the roles of locomotor activity, tail heat dissipation, heat production in the brown adipose tissue (BAT) activity, and chemical mediators during ovalbumin-induced anaphylactic hypotension in awake rats. We measured the core body temperature (Tcore) and mean blood pressure (MBP), along with the surface temperature of the interscapular region (TiScap), an indirect measure of BAT activity, and the tail (Ttail). During anaphylaxis, MBP decreased to the nadir of 53 ± 2 mmHg at 8 min with recovery toward baseline. Tcore began to decrease at 7.5 min with the nadir of 36.1 ± 0.2°C at 30 min from the baseline of 38.0 ± 0.1°C. TiScap also significantly decreased, but its onset was preceded by that of Tcore. Ttail decreased after antigen, suggesting the absence of increased heat dissipation from the tail. The physical activity, as evaluated by moved distances, did not decrease until 20 min after antigen, followed by a progressive decrease. Reduced movement using a restraint maneuver not only reduced Tcore in nonsensitized rats but also augmented the anaphylactic hypothermia in the early phase (1.5-18 min) in sensitized rats. Combined antagonism against platelet-activating factor (PAF) and histamine H1 receptors abolished antigen-induced hypotension but only attenuated hypothermia. In conclusion, decreased locomotor activity, but not tail heat dissipation or decreased BAT activity, may at least in part contribute to this hypothermia. PAF and histamine are involved mainly in hypotension but only partly in hypothermia during rat anaphylaxis.NEW & NOTEWORTHY Anaphylactic shock is a life-threatening systemic hypotension. Hypothermia is observed during systemic anaphylaxis of rats. We determined the mechanism as follows: decreased locomotor activity, but not tail heat dissipation or decreased BAT activity, may at least in part contribute to this hypothermia. PAF and histamine are involved mainly in hypotension, but only partly in hypothermia during rat anaphylaxis.


Subject(s)
Anaphylaxis , Hypotension , Hypothermia , Rats , Animals , Anaphylaxis/chemically induced , Histamine , Hypothermia/complications , Wakefulness , Hypotension/etiology , Platelet Activating Factor/adverse effects
2.
J Immunother Cancer ; 10(12)2022 12.
Article in English | MEDLINE | ID: mdl-36543377

ABSTRACT

BACKGROUND: With the increased use of immune checkpoint inhibitors (ICIs), side effects and toxicity are a great concern. Anaphylaxis has been identified as a potential adverse event induced by ICIs. Anaphylaxis is a life-threatening medical emergency. However, the mechanisms and factors that can potentially influence the incidence and severity of anaphylaxis in patients with cancer remain unclear. METHODS: Healthy, murine colon 26, CT26, breast 4T1, EMT6, and renal RENCA tumor-bearing mice were treated with an anti-PD-L1 antibody (clone 10F.9G2). Symptoms of anaphylaxis were evaluated along with body temperature and mortality. The amounts of antidrug antibody and platelet-activating factor (PAF) in the blood were quantified via ELISA and liquid chromatography-mass spectrometry (LC-MS/MS). Immune cells were analyzed and isolated using a flow cytometer and magnetic-activated cell sorting, respectively. RESULTS: Repeated administration of the anti-PD-L1 antibody 10F.9G2 to tumor-bearing mice caused fatal anaphylaxis, depending on the type of tumor model. After administration, antidrug immunoglobulin G (IgG), but not IgE antibodies, were produced, and PAF was released as a chemical mediator during anaphylaxis, indicating that anaphylaxis was caused by an IgG-dependent pathway. Anaphylaxis induced by 10F.9G2 was treated with a PAF receptor antagonist. We identified that neutrophils and macrophages were PAF-producing effector cells during anaphylaxis, and the tumor-bearing models with increased numbers of neutrophils and macrophages showed lethal anaphylaxis after treatment with 10F.9G2. Depletion of both neutrophils and macrophages using clodronate liposomes prevented anaphylaxis in tumor-bearing mice. CONCLUSIONS: Thus, increased numbers of neutrophils and macrophages associated with cancer progression may be risk factors for anaphylaxis. These findings may provide useful insights into the mechanism of anaphylaxis following the administration of immune checkpoint inhibitors in human subjects.


Subject(s)
Anaphylaxis , Neoplasms , Mice , Humans , Animals , Immunoglobulin G , Anaphylaxis/chemically induced , Anaphylaxis/pathology , Immune Checkpoint Inhibitors/adverse effects , Neutrophils/metabolism , Chromatography, Liquid , Tandem Mass Spectrometry , Macrophages , Platelet Activating Factor/adverse effects , Platelet Activating Factor/metabolism , Neoplasms/metabolism
3.
J Pept Sci ; 28(10): e3412, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35437860

ABSTRACT

Platelet-activating factor (PAF) is an important lipid mediator of anaphylaxis and therefore can be an anti-anaphylactic agent target. Recently, we reported that several synthetic biotinylated peptides containing a Tyr-Lys-Asp-Gly sequence markedly inhibited the bioactivities of PAF in vitro and in vivo; it also inhibited anaphylactic reactions such as hypothermia, hypotension, and vascular permeability in vivo. Here, we report the anti-anaphylactic effects of three biotinylated heptapeptides (peptide 1: H-Lys(biotinyl)-Trp-Tyr-Lys-Asp-Gly-Asp-OH, peptide 2: H-D -Lys(biotinyl)-Trp-Tyr-Lys-Asp-Gly-Asp-OH, and peptide 3: H-D -Lys(biotinyl)-Trp-Tyr-Lys-Asp-Gly-D -Asp-OH). The experiment using tryptophan fluorescence spectroscopy showed that the interaction of peptides 2 and 3 with PAF was larger than that of peptide 1. Experiments using a rat model of hind paw edema showed that peptides 1, 3, and 2 inhibited PAF-induced edema by 67.9%, 69.3%, and 79.3%, respectively. In a mouse model of anaphylaxis, both peptides 2 and 3 showed inhibitory effects on anaphylactic hypothermia, whereas peptide 1 did not. Furthermore, experiments involving in vitro rat plasma stability of peptides showed that both peptides 3 and 2 were more stable in plasma compared to peptide 1 (84.0%, 51.8%, and 0%, remained after 6 h, respectively). Our results suggest that both peptides 2 and 3 may show systemic and local inhibitory effects as anti-anaphylactic agents targeting PAF.


Subject(s)
Anaphylaxis , Hypothermia , Amino Acids , Anaphylaxis/drug therapy , Animals , Edema/chemically induced , Edema/drug therapy , Mice , Peptides/chemistry , Peptides/pharmacology , Platelet Activating Factor/adverse effects , Rats
4.
Peptides ; 73: 60-6, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26348270

ABSTRACT

Angiotensin (Ang)--a peptide that is part of the renin-angiotensin system-induces vasoconstriction and a subsequent increase in blood pressure; Ang peptides, especially AngII, can also act as potent pro-inflammatory mediators. Platelet-activating factor (PAF) is a potent phospholipid mediator that is implicated in many inflammatory diseases. In this study, we investigated the effects of Ang peptides (AngII, AngIII, and AngIV) on PAF-induced inflammatory activity. In experiments using a rat hind-paw oedema model, AngII markedly and dose-dependently attenuated the paw oedema induced by PAF. The inhibitory effects of AngIII and AngIV on PAF-induced paw oedema were lower than that of AngII. Two Ang receptors, the AT1 and AT2 receptors, did not affect the AngII-mediated attenuation of PAF-induced paw oedema. Moreover, intrinsic tyrosine fluorescence studies demonstrated that AngII, AngIII, and AngIV interact with PAF, and that their affinities were closely correlated with their inhibitory effects on PAF-induced rat paw oedema. Also, AngII interacted with metabolite/precursor of PAF (lyso-PAF), and an oxidized phospholipid, 1-palmitoyl-2-(5'-oxo-valeroyl)-sn-glycero-3-phosphocholine (POVPC), which bears a marked structural resemblance to PAF. Furthermore, POVPC dose-dependently inhibited AngII-mediated attenuation of PAF-induced paw oedema. These results suggest that Ang peptides can attenuate PAF-induced inflammatory activity through binding to PAF and lyso-PAF in rats. Therefore, Ang peptides may be closely involved in the regulation of many inflammatory diseases caused by PAF.


Subject(s)
Angiotensins/pharmacology , Edema , Peptides/pharmacology , Platelet Activating Factor/adverse effects , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Animals , Edema/chemically induced , Edema/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Male , Platelet Activating Factor/pharmacology , Rats , Rats, Wistar
5.
PLoS One ; 10(3): e0120802, 2015.
Article in English | MEDLINE | ID: mdl-25793535

ABSTRACT

Intestinal circulatory disturbances, atony, edema and swelling are of great clinical relevance, but the related mechanisms and possible therapeutic options are poorly characterized, in part because of the difficulties to comprehensively analyze these conditions. To overcome these limitations we have developed a model of the isolated perfused rat small intestine where all of these symptoms can be studied simultaneously. Here we used this model to study the role of eicosanoids, steroids and quinidine in platelet-activating factor (PAF)-induced intestinal disorders. A vascular bolus of PAF (0.5 nmol) triggered release of thromboxane and peptidoleukotrienes into the vascular bed (peak concentration 35 nM and 0.8 nM) and reproduced all symptoms of intestinal failure: mesenteric vasoconstriction, translocation of fluid and macromolecules from the vasculature to the lumen and lymphatics, intestinal edema formation, loss of intestinal peristalsis and decreased galactose uptake. All effects of PAF were abolished by the PAF-receptor antagonist ABT491 (2.5 µM). The COX and LOX inhibitors ASA and AA861 (500 µM, 10 µM) did not exhibit barrier-protective effects and the eicosanoid antagonists SQ29548 and MK571 (10 µM, each) only moderately attenuated the loss of vascular fluid, the redistribution to the lumen and the transfer of FITC dextran to the lumen. The steroid dexamethasone (10 µM) showed no barrier-protective properties and failed to prevent edema formation. Quinidine (100 µM) inhibited the increase in arterial pressure, stabilized all the intestinal barriers, and reduced lymph production and the transfer of FITC dextran to the lymph. While quinidine by itself reduced peristalsis, it also obviated paralysis, preserved intestinal functions and prevented edema formation. We conclude that quinidine exerts multiple protective effects against vasoconstriction, edema formation and paralysis in the intestine. The therapeutic use of quinidine for intestinal ailments deserves further study.


Subject(s)
Edema/prevention & control , Gastrointestinal Diseases/prevention & control , Paralysis/prevention & control , Platelet Activating Factor/adverse effects , Protective Agents/pharmacology , Quinidine/pharmacology , Vasoconstriction/drug effects , Animals , Dexamethasone/pharmacology , Disease Models, Animal , Edema/chemically induced , Edema/pathology , Eicosanoids/pharmacology , Female , Gastrointestinal Diseases/chemically induced , Gastrointestinal Diseases/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Paralysis/chemically induced , Paralysis/pathology , Platelet Activating Factor/administration & dosage , Rats
6.
Eur J Pharmacol ; 714(1-3): 142-7, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23792041

ABSTRACT

Platelet-activating factor (PAF), a potent proinflammatory mediator, is involved in many inflammatory diseases. We recently reported that synthetic biotinylated peptides having a Tyr-Lys-Asp-Gly sequence inhibit PAF-induced inflammation by directly binding to PAF. In this study, we investigated the effect of two synthetic biotinylated peptides, both of which have a sequence similar to Tyr-Lys-Asp-Gly-an endothelin-3 (ET-3)-related biotinylated pentapeptide (Tyr-Lys-Asp-Lys-Glu, BPET3) and a scavenger receptor CD36-related biotinylated tetrapeptide (Tyr-Lys-Gly-Lys, BPCD36)-on PAF-induced inflammation by using a rat model of hind paw oedema. BPET3 markedly inhibited PAF-induced oedema in a dose-dependent manner, and the dose that caused 50% inhibition was estimated to be approximately 2.64 nmol/paw. The inhibitory effect of BPCD36 on PAF-induced paw oedema was less than that of BPET3, while a synthetic biotinylated pentapeptide (Lys-Lys-Tyr-Asp-Glu) shuffling amino acid sequence of BPET3, an ET-1-related synthetic biotinylated pentapeptide (Leu-Met-Asp-Lys-Glu), or an ET-2-related synthetic biotinylated pentapeptide (Trp-Leu-Asp-Lys-Glu) did not inhibit PAF-induced paw oedema. Furthermore, intrinsic tryptophan fluorescence studies demonstrated that ET-3 specifically interacted with both PAF and its metabolite/precursor lyso-PAF. These results provide evidence that the Tyr-Lys-Asp region in both ET-3 and BPET3 is essential for marked inhibition of the peptide on PAF-induced inflammation, and strongly suggest that BPET3 may be useful as a novel anti-inflammatory drug targeting PAF.


Subject(s)
Biotinylation , Endothelin-3/chemistry , Oligopeptides/chemistry , Oligopeptides/pharmacology , Platelet Activating Factor/antagonists & inhibitors , Amino Acid Sequence , Animals , CD36 Antigens/chemistry , Dose-Response Relationship, Drug , Edema/chemically induced , Edema/drug therapy , Male , Oligopeptides/chemical synthesis , Oligopeptides/therapeutic use , Platelet Activating Factor/adverse effects , Platelet Activating Factor/analogs & derivatives , Rats , Rats, Wistar , Structure-Activity Relationship
7.
Am J Rhinol Allergy ; 27(2): e48-52, 2013.
Article in English | MEDLINE | ID: mdl-23562190

ABSTRACT

BACKGROUND: Platelet-activating factor (PAF) is a lipid mediator produced by most inflammatory cells. Clinical and experimental findings suggest that PAF participates in allergic rhinitis (AR) pathogenesis. The aim was to assess the PAF ability to induce clinical response in nasal airway after local stimulation. METHOD: Ten nonatopic healthy volunteers (HVs) and 10 AR patients out of pollen season were enrolled. PAF increasing concentrations (100, 200, and 400 nM) were instilled into both nasal cavities (0, 30, and 60 minutes, respectively). Nasal symptoms (congestion, rhinorrhea, sneezing, itching, and total 4 symptom score and nasal volume between the 2nd and 5th cm (Vol(2-5)) using acoustic rhinometry (AcR), were assessed at -30, 0, 30, 60, 90, 120, and 240 minutes. RESULT: PAF increased individual and total nasal symptom score in both HVs and seasonal AR (SAR) patients from 30 to 120 minutes (maximum score at 120', p < 0.05). Nasal obstruction was the most relevant and lasting nasal symptom. PAF also induced a significant reduction of Vol(2-5) at 90' (27%), 120' (38.7%), and 240' (36.4%). No differences in the response to PAF nasal challenge were observed between HVs and SAR subjects in either clinical symptoms or AcR. CONCLUSION: This is the first description of PAF effects on human nasal mucosa using a cumulative dose schedule and evaluated by both nasal symptoms and AcR. Nasal provocation with PAF showed long-lasting effects on nasal symptoms and nasal obstruction in HVs and in patients with SAR. Nasal challenge may be a useful tool to investigate the role of PAF in AR and the potential role of anti-PAF drugs.


Subject(s)
Nasal Cavity/drug effects , Platelet Activating Factor/administration & dosage , Rhinitis, Allergic, Seasonal/immunology , Administration, Intranasal , Adult , Allergens/immunology , Antigens, Plant/immunology , Disease Progression , Feasibility Studies , Female , Humans , Immunization , Male , Nasal Cavity/pathology , Nasal Cavity/surgery , Nasal Obstruction/etiology , Nasal Obstruction/prevention & control , Platelet Activating Factor/adverse effects , Pollen/adverse effects , Pollen/immunology , Rhinitis, Allergic, Seasonal/complications , Rhinitis, Allergic, Seasonal/diagnosis , Rhinometry, Acoustic , Treatment Outcome
8.
Allergy ; 68(2): 256-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23320562

ABSTRACT

BACKGROUND: Platelet-activating factor (PAF) causes wheal and flare responses which are abrogated by H1-antihistamines giving rise to the hypothesis that PAF-induced wheal development is secondary to histamine release from dermal mast cells. But is this hypothesis correct? METHODS: Wheal and flare responses were induced by intradermal injection of PAF, codeine and histamine in 14 healthy volunteers. Dermal histamine and PGD2 contractions were measured using microdialysis. RESULTS: PAF, unlike histamine and codeine, did not cause a statistically significant rise in mean histamine levels with ten persons showing negligible histamine release. Codeine caused a significant but variable histamine release, ranging from 29 to 282 ng/ml. Codeine, but not PAF or histamine, caused a small but statistically significant release of PGD2. CONCLUSION: Wheal and flare reactions in human skin induced by PAF are not associated with histamine release and, therefore, appear to be independent of mast cell degranulation.


Subject(s)
Codeine/pharmacology , Histamine Release/drug effects , Histamine/pharmacology , Platelet Activating Factor/pharmacology , Urticaria/chemically induced , Adult , Cell Degranulation/drug effects , Cell Degranulation/immunology , Codeine/adverse effects , Female , Germany , Histamine/adverse effects , Humans , Injections, Intradermal , Male , Mast Cells/drug effects , Mast Cells/immunology , Microdialysis , Platelet Activating Factor/adverse effects , Reference Values , Sampling Studies , Skin/drug effects , Skin Tests/methods , Urticaria/immunology , Young Adult
9.
PLoS One ; 7(12): e51955, 2012.
Article in English | MEDLINE | ID: mdl-23272193

ABSTRACT

The intestinal microflora is critical for normal development, with aberrant colonization increasing the risk for necrotizing enterocolitis (NEC). In contrast, probiotic bacteria have been shown to decrease its incidence. Multiple pro- and anti-inflammatory cytokines have been identified as markers of intestinal inflammation, both in human patients with NEC and in models of immature intestine. Specifically, IL-10 signaling attenuates intestinal responses to gut dysbiosis, and disruption of this pathway exacerbates inflammation in murine models of NEC. However, the effects of probiotics on IL-10 and its signaling pathway, remain poorly defined. Real-time PCR profiling revealed developmental regulation of MIP-2, TNF-α, IL-12, IL-10 and the IL-10R2 subunit of the IL-10 receptor in immature murine colon, while the expression of IL-6 and IL-18 was independent of postnatal age. Enteral administration of the probiotic Lactobacillus rhamnosus GG (LGG) down-regulated the expression of TNF-α and MIP-2 and yet failed to alter IL-10 mRNA and protein expression. LGG did however induce mRNA expression of the IL-10R2 subunit of the IL-10 receptor. IL-10 receptor activation has been associated with signal transducer and activator of transcription (STAT) 3-dependent induction of members of the suppressors of cytokine signaling (SOCS) family. In 2 week-old mice, LGG also induced STAT3 phosphorylation, increased colonic expression of SOCS-3, and attenuated colonic production of MIP-2 and TNF-α. These LGG-dependent changes in phosphoSTAT3, SOCS3, MIP-2 and TNF-α were all inhibited by antibody-mediated blockade of the IL-10 receptor. Thus LGG decreased baseline proinflammatory cytokine expression in the developing colon through upregulation of IL-10 receptor-mediated signaling, most likely due to the combined induction of phospho-STAT3 and SOCS3. Furthermore, LGG-dependent increases in IL-10R2 were associated with reductions in TNF-α, MIP-2 and disease severity in a murine model of intestinal injury in the immature colon.


Subject(s)
Colon/metabolism , Colon/microbiology , Interleukin-10 Receptor beta Subunit/metabolism , Interleukin-10/metabolism , Lacticaseibacillus rhamnosus/metabolism , Signal Transduction , Animals , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Cytokines/genetics , Cytokines/metabolism , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/microbiology , Gene Expression Regulation, Developmental , Humans , Inflammation Mediators/metabolism , Interleukin-10/genetics , Interleukin-10 Receptor beta Subunit/genetics , Intestinal Mucosa/metabolism , Intestines/immunology , Intestines/microbiology , Lipopolysaccharides/immunology , Mice , Phosphorylation , Platelet Activating Factor/adverse effects , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Weaning
10.
Br J Pharmacol ; 164(2b): 781-93, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21506958

ABSTRACT

BACKGROUND AND PURPOSE: 5-Lipoxygenase (5-LO) is the key enzyme in the biosynthesis of pro-inflammatory leukotrienes (LTs) representing a potential target for pharmacological intervention with inflammation and allergic disorders. Although many LT synthesis inhibitors are effective in simple in vitro test systems, they frequently fail in vivo due to lack of efficacy. Here, we attempted to assess the pharmacological potential of the previously identified 5-LO inhibitor 2-(4-(biphenyl-4-ylamino)-6-chloropyrimidin-2-ylthio)octanoic acid (HZ52). EXPERIMENTAL APPROACH: We evaluated the efficacy of HZ52 in vivo using carrageenan-induced pleurisy in rats and platelet-activating factor (PAF)-induced lethal shock in mice. We also characterized 5-LO inhibition by HZ52 at the cellular and molecular level in comparison with other types of 5-LO inhibitor, that is, BWA4C, ZM230487 and hyperforin. KEY RESULTS: HZ52, 1.5 mg·kg⁻¹ i.p., prevented carrageenan-induced pleurisy accompanied by reduced LTB(4) levels and protected mice (10 mg·kg⁻¹, i.p.) against PAF-induced shock. Detailed analysis in cell-based and cell-free assays revealed that inhibition of 5-LO by HZ52 (i) does not depend on radical scavenging properties and is reversible; (ii) is not impaired by an increased peroxide tone or by elevated substrate concentrations; and (iii) is little affected by the cell stimulus or by phospholipids, glycerides, membranes or Ca²âº. CONCLUSIONS AND IMPLICATIONS: HZ52 is a promising new type of 5-LO inhibitor with efficacy in vivo and with a favourable pharmacological profile. It possesses a unique 5-LO inhibitory mechanism different from classical 5-LO inhibitors and seemingly lacks the typical disadvantages of former classes of LT synthesis blockers.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arachidonate 5-Lipoxygenase/metabolism , Caprylates/pharmacology , Lipoxygenase Inhibitors/pharmacology , Pyrimidines/pharmacology , Animals , Arachidonate 5-Lipoxygenase/biosynthesis , Carrageenan , Leukotriene B4/antagonists & inhibitors , Leukotriene B4/metabolism , Male , Mice , Neutrophils/drug effects , Neutrophils/metabolism , Nuclear Envelope/drug effects , Nuclear Envelope/metabolism , Platelet Activating Factor/adverse effects , Platelet Activating Factor/pharmacology , Pleurisy/chemically induced , Pleurisy/metabolism , Protein Transport/drug effects , Rats , Rats, Wistar , Shock/chemically induced , Shock/metabolism
11.
Curr Eye Res ; 35(12): 1063-71, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20961214

ABSTRACT

PURPOSE: Although the exact mechanisms underlying corneal neovascularization remain unclear, cytokines and growth factors play an important role in their development. We have shown previously that the inflammatory mediator platelet-activating factor (PAF) is a potent inducer of corneal neovascularization in vivo. In this study, we investigate the role of stromal myofibroblasts in neovascularization and the effect of PAF on this process. METHODS: Myofibroblasts were obtained from rabbit corneal keratocytes and identified with anti-α-SMA antibody. Cells were treated with PAF (100 nM) for 24 hr. In some experiments, cells were pre-treated with the PAF antagonist LAU-0901 (150 nM). Expression of vascular endothelial growth factor (VEGF) and thrombospondin-1 (TSP-1) was examined by immunofluorescence and immunoblotting. To study the effect of myofibroblasts on vessel formation in vitro, Vybrant(®) CM-DiI labeled human umbilical vein endothelial cells (HUVECs) were cultured on myofibroblasts in a thin layer of collagen gel. CD31 was used as the cell marker of HUVEC. RESULTS: VEGF and TSP-1 were not detectable in keratocytes, but they were positively stained in myofibroblasts. PAF induced a significant increase in VEGF expression and a decrease in TSP-1 expression. These changes were inhibited in the presence of LAU-0901. HUVECs co-cultured with corneal myofibroblasts formed a typical structure of vessel-like tubes within 1 week. The addition of PAF to the medium increased HUVEC-induced vessel-like tube formation, which was abolished by LAU-0901. Addition of anti-VEGF antibody to the medium completely prevented the formation of vessel-like tubes. CONCLUSION: We provide evidence for the role of stromal myofibroblasts in the corneal neovascularization process. By enhancing VEGF production and decreasing TSP-1 production in myofibroblasts, PAF augments the angiogenic response. The PAF antagonist LAU-0901 could represent a new therapeutic venue for inhibiting corneal neovascularization.


Subject(s)
Cornea/cytology , Corneal Neovascularization/chemically induced , Myofibroblasts/drug effects , Platelet Activating Factor/adverse effects , Animals , Cells, Cultured , Coculture Techniques , Dihydropyridines/pharmacology , Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , Humans , Platelet Activating Factor/antagonists & inhibitors , Rabbits , Thrombospondin 1/metabolism , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A/metabolism
12.
Br J Pharmacol ; 160(1): 36-47, 2010 May.
Article in English | MEDLINE | ID: mdl-20331609

ABSTRACT

Cell expansion and metastasis are considered hallmarks of tumour progression. Therefore, efforts have been made to develop novel anti-cancer drugs that inhibit both the proliferation and the motility of tumour cells. Synthetic alkylphospholipids, compounds with aliphatic side chains that are ether linked to a glycerol backbone, are structurally derived from platelet-activating factor and represent a new class of drugs with anti-proliferative properties in tumour cells. These compounds do not interfere with the DNA or mitotic spindle apparatus of the cell. Instead, they are incorporated into cell membranes, where they accumulate and interfere with lipid metabolism and lipid-dependent signalling pathways. Recently, it has been shown that the most commonly studied alkylphospholipids inhibit proliferation by inducing apoptosis in malignant cells while leaving normal cells unaffected. This review focuses on a novel group of synthetic alkylphospholipids, the glycosidated phospholipids, which contain carbohydrates or carbohydrate-related molecules at the sn-2 position of the glycerol backbone. Members of this subfamily also exhibit anti-proliferative capacity and modulate the cell adhesion, differentiation, and migration of tumour cells. Among this group, Ino-C2-PAF shows the highest efficacy and low cytotoxicity. Apart from its anti-proliferative effect, Ino-C2-PAF strongly reduces cell motility via its inhibitory effect on the phosphorylation of the cytosolic tyrosine kinases FAK and Src. Signalling pathways under the control of the FAK/Src complex are normally required for both migration and proliferation and play a prominent role in tumour progression. We intend to highlight the potential of glycosidated phospholipids, especially Ino-C2-PAF, as a promising new group of drugs for the treatment of hyperproliferative and migration-based skin diseases.


Subject(s)
Antineoplastic Agents/pharmacology , Glycosides/pharmacology , Phospholipids/pharmacology , Animals , Antineoplastic Agents/adverse effects , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Glycosides/adverse effects , Humans , Inositol/adverse effects , Inositol/analogs & derivatives , Inositol/pharmacology , Molecular Structure , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/pathology , Phospholipids/adverse effects , Platelet Activating Factor/adverse effects , Platelet Activating Factor/analogs & derivatives , Platelet Activating Factor/pharmacology , Signal Transduction
13.
Biochem Biophys Res Commun ; 317(1): 205-10, 2004 Apr 23.
Article in English | MEDLINE | ID: mdl-15047169

ABSTRACT

Approximately 4% of the Japanese population genetically lack plasma platelet activating factor acetylhydrolase (PAF-AH) and show a higher prevalence of thromboembolic disease, but whether they are susceptible to another PAF-related disease, asthma, remains controversial. To determine the role of plasma PAF-AH in airway physiology, we performed PAF bronchoprovocation tests in 8 plasma PAF-AH-deficient subjects and 16 control subjects. Serial inhalation of PAF (1-1000 microg/ml) concentration-dependently induced acute bronchoconstriction, but there was no significant difference between PAF-AH-deficient and control subjects (11.7 +/- 4.6% vs. 9.6 +/- 2.8% decrease in forced expiratory volume in 1 s). Transient neutropenia after single inhalation of PAF (1000 microg/ml) showed no significant difference between the groups either in its magnitude (72 +/- 11% vs. 65 +/- 9% decrease) or duration (4.1 +/- 1.0 vs. 3.3 +/- 0.8 min). In conclusion, a lack of plasma PAF-AH activity alone does not augment physiological responses to PAF in the airway.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/deficiency , Bronchoconstriction/drug effects , Bronchoconstriction/physiology , Platelet Activating Factor/adverse effects , 1-Alkyl-2-acetylglycerophosphocholine Esterase/blood , 1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Administration, Inhalation , Adult , Alleles , Asthma/enzymology , Asthma/epidemiology , Female , Heterozygote , Homozygote , Humans , Japan/epidemiology , Kinetics , Lymphocytes/drug effects , Lymphocytes/metabolism , Male , Neutropenia/chemically induced , Neutropenia/metabolism , Platelet Activating Factor/administration & dosage , Respiratory Function Tests/methods
14.
Eur Respir J ; 23(1): 71-5, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14738234

ABSTRACT

Platelet-activating factor (PAF) is an inflammatory mediator that provokes neutropaenia, bronchoconstriction and gas exchange defects due to exudation of bulk plasma within the airways. While the inhibitory effects of short-acting beta2-agonists on PAF-induced disturbances have been consistently shown, those of long-acting beta2-agonists are less convincing. To further explore the mechanisms involved in PAF challenge in asthma, 12 patients (forced expiratory volume in one second, 90 +/- 4% predicted) were investigated 2 h after inhaled formoterol (18 microg), in a double-blind, placebo-controlled, crossover design following PAF (18 microg) inhalation. Compared with the placebo, at 5 min, premedication with formoterol reduced PAF-induced cough and dyspnoea, and attenuated increased respiratory system resistance (by 67%) and arterial deoxygenation (by 50%). Likewise, ventilation-perfusion (V'A/Q') inequality improved, as reflected by the dispersion of pulmonary blood flow (by 63%) and an overall index of V'A/Q' heterogeneity (by 71%). In contrast, PAF-induced facial flushing, neutropaenia and subsequent rebound neutrophilia remained unchanged. The improvement in gas exchange abnormalities shown after platelet-activating factor in patients with asthma pretreated with formoterol at the recommended clinical dose may reflect, in addition to its class effects, an anti-exudative effect of formoterol in the airways.


Subject(s)
Adrenergic beta-Agonists/therapeutic use , Asthma/physiopathology , Ethanolamines/therapeutic use , Platelet Activating Factor/adverse effects , Administration, Inhalation , Adrenergic beta-Agonists/administration & dosage , Adult , Airway Resistance/drug effects , Asthma/drug therapy , Cough/prevention & control , Cross-Over Studies , Double-Blind Method , Dyspnea/prevention & control , Ethanolamines/administration & dosage , Female , Flushing/prevention & control , Formoterol Fumarate , Humans , Male , Neutropenia/prevention & control , Pulmonary Circulation/drug effects , Pulmonary Gas Exchange/drug effects , Ventilation-Perfusion Ratio/drug effects
15.
J Ocul Pharmacol Ther ; 19(4): 315-24, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12964956

ABSTRACT

Platelet-activating factor (PAF) may be an important mediator in allergic conjunctivitis. In this study, apafant, a potent PAF antagonist, was evaluated for topical ocular anti-PAF activity in PAF and antigen stimulated conjunctivitis models. PAF, when injected into parpebral conjunctiva, provoked an acute increase, measured as dye leakage, in conjunctival vascular permeability. Apafant inhibited this response in a dose-related manner, and the inhibitory action of 0.1% apafant lasted for at least 6 hours duration. PAF, when instilled into the conjunctival sac, induced itch-scratching behavior and clinical symptoms, such as conjunctival redness and edema. These were inhibited by pretreatment with apafant ophthalmic solution. In a passive conjunctival anaphylaxis model in guinea pigs, significant inhibition of the allergic response was observed following topical ocular administration of apafant 5 and 15 minutes prior to the antigen challenge. We have demonstrated that PAF plays an important role in the development of allergic conjunctivitis. These results clearly indicate that apafant has potential as a topical ocular anti-PAF for treating allergic conjunctivitis.


Subject(s)
Azepines/pharmacokinetics , Capillary Permeability/drug effects , Conjunctivitis, Allergic/chemically induced , Conjunctivitis, Allergic/prevention & control , Platelet Activating Factor/adverse effects , Triazoles/pharmacokinetics , Animals , Antigens/adverse effects , Antigens/immunology , Azepines/administration & dosage , Capillary Permeability/immunology , Conjunctivitis, Allergic/immunology , Disease Models, Animal , Dose-Response Relationship, Drug , Dye Dilution Technique , Guinea Pigs , Humans , Immunization, Passive , Ketotifen/administration & dosage , Ketotifen/pharmacokinetics , Male , Ophthalmic Solutions , Pruritus/chemically induced , Pruritus/immunology , Pruritus/prevention & control , Time Factors , Triazoles/administration & dosage
16.
Br J Pharmacol ; 129(7): 1522-9, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10742310

ABSTRACT

1. Platelet-activating factor (PAF), an inflammatory mediator, plays an important role in mediating intestinal injury. However, it remains unclear whether PAF has a function in the intestine. The production of PAF by normal intestine and by unstimulated intestinal epithelial cell lines suggests that PAF may have a regulatory function in the normal bowel. 2. In this study we investigated the role of PAF in modulating intestinal mucosal permeability in rats. Lumen-to-blood transit of FD-4 (dextran 4400), (an index of intestinal permeability), was assessed in sham-operated rats and rats injected with PAF (1.25 microg kg(-1), i.v., a dose insufficient to induce intestinal injury). 3. PAF-induced villus cytoskeletal changes were examined by staining the intestine for F-actin. The effect of PAF on tyrosine phosphorylation of the junctional protein E-cadherin was examined by immunoprecipitation. Some rats were pretreated with AG1288 (a tyrosine kinase inhibitor) before PAF injection, and mucosal permeability change was assessed. 4. To investigate the role of endogenous PAF upon mucosal permeability, we studied the effect of PAF antagonists on (intraluminal) glucose-induced increase in mucosal permeability. 5. We found that low dose PAF: (a) alters the cytoskeletal structure of intestinal epithelium, (b) causes the influx of FD4 from intestinal lumen to systemic circulation, (c) induces tyrosine phosphorylation of E-cadherin and cadherin-associated proteins. Glucose-induced mucosal permeability increase is abolished by using two structurally different PAF antagonists. 6. These results suggest that endogenous PAF modulates macromolecular movement across the intestinal mucosal barrier, probably via tyrosine phosphorylation of E-cadherin and cytoskeletal alteration of enterocytes.


Subject(s)
Cadherins/metabolism , Intestinal Mucosa/drug effects , Platelet Activating Factor/pharmacology , Animals , Azepines/pharmacology , Cadherins/chemistry , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Dextrans/blood , Dextrans/pharmacokinetics , Enzyme Inhibitors/pharmacology , Epithelium/drug effects , Epithelium/metabolism , Glucose/pharmacology , Hypotension/chemically induced , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiology , Intestines/drug effects , Leukocytosis/chemically induced , Male , Permeability/drug effects , Phosphorylation , Phosphotyrosine , Platelet Activating Factor/adverse effects , Platelet Activating Factor/physiology , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinolinium Compounds/pharmacology , Rats , Rats, Sprague-Dawley , Triazoles/pharmacology , Tyrosine/metabolism , Tyrphostins/pharmacology
17.
J Vet Pharmacol Ther ; 22(5): 327-32, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10597537

ABSTRACT

The effects of intravenous (i.v.) infusion of platelet-activating factor (PAF), 100 ng/kg/min for 10 min, with and without pretreatment with a selective PAF-antagonist on gastrointestinal electrical activity, arterial pressure and clinical and haematological parameters were studied. Conscious miniature pigs with electrodes implanted in the wall of the antrum pylori and small and large intestine were used. Platelet-activating factor induced restlessness or depression, shivering, tachypnoea and coughing, retching and vomiting, hypotension and a delayed and sustained increase in leucocyte count with an increase in percentage of segmented neutrophils. The PAF-antagonist, SAH 63-675, administered at 10 mg/kg intravenously, inhibited these effects. Platelet-activating factor resulted in a decrease in electrical activity in the antrum and large intestine, whereas small intestinal activity was not significantly influenced. Pretreatment with the antagonist suppressed these inhibitory effects.


Subject(s)
Furans/pharmacology , Intestines/drug effects , Intestines/physiology , Platelet Activating Factor/adverse effects , Platelet Activating Factor/antagonists & inhibitors , Animals , Electrodes , Electrophysiology , Female , Hemodynamics/drug effects , Hypotension/chemically induced , Infusions, Intravenous , Leukocyte Count/drug effects , Pyloric Antrum/drug effects , Shivering , Swine , Swine, Miniature , Vomiting/chemically induced
19.
Arch Surg ; 131(8): 855-9; discussion 859-60, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8712910

ABSTRACT

OBJECTIVE: To investigate whether inhaled nitric oxide (INO) can prevent platelet activating factor (PAF)-induced pulmonary hypertension and shock. DESIGN: Randomized controlled animal trial. SETTING: Laboratory. SUBJECTS: Yorkshire swine. INTERVENTIONS: Animals received general anesthesia and invasive hemodynamic monitoring, then PAF only, 2.5 micrograms/kg intravenously over 45 minutes (PAF group, n = 9) or PAF in addition to INO, 20 ppm (PAF-INO group, n = 6). MAIN OUTCOME: Vascular pressures (mean arterial and mean pulmonary), vascular resistance indexes (systemic and pulmonary), cardiac indexes, and oxygen delivery and oxygen consumption. RESULTS: Mean arterial pressures, cardiac indexes, and oxygen delivery and consumption were significantly higher in the PAF-INO group. Mean pulmonary arterial pressures and systemic and pulmonary vascular resistance indexes were significantly lower in the PAF-INO group. There were 4 deaths (44%) in the PAF group vs none (0%) in the PAF-INO group (P = 10). CONCLUSIONS: The use of INO prevents pulmonary hypertension, circulatory failure, and death during PAF-induced shock.


Subject(s)
Hypertension, Pulmonary/drug therapy , Nitric Oxide/therapeutic use , Platelet Activating Factor/adverse effects , Shock, Septic/drug therapy , Administration, Inhalation , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Hemodynamics/drug effects , Hypertension, Pulmonary/chemically induced , Random Allocation , Shock, Septic/chemically induced , Swine
20.
J Cardiovasc Electrophysiol ; 7(2): 120-33, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8853022

ABSTRACT

INTRODUCTION: Both ischemia and reperfusion are associated with ventricular arrhythmias. In both instances, neutrophils migrate into the ischemic zone, are activated by locally released factors, and bind to myocytes. The activated neutrophils liberate platelet activating factor (PAF). We have studied the arrhythmogenic actions of PAF on transmembrane potentials of isolated canine cardiac myocytes. METHODS AND RESULTS: Cardiac myocytes were prepared from normal canine hearts by standard methods and studied in vitro by recording transmembrane potentials under control conditions and during exposure to graded doses of PAF, usually 0.25 to 1.25 micrograms (0.25 to 1.2 microM). Myocytes were superfused with Tyrode's solution (2.0 mL/min), paced at a cycle length of 1000 msec, and maintained at a temperature between 36 degrees and 38 degrees C. PAF caused a consistent and dose-dependent set of alterations in the transmembrane potential, including increased action potential duration, runs of early afterdepolarizations (EADs), and transient arrest of repolarization (PA). In addition, in some myocytes PAF caused intermittent small depolarizations both at the plateau voltage and resting potential. The effects of PAF were transient: only some residual action potential prolongation was noted after Tyrode's washout for 5 minutes. Effects of PAF were blocked in a dose-dependent manner by the PAF receptor antagonist, CV-6209. Both tetrodotoxin (1.2 x 10(-6) M) and xylocaine (5 x 10(-5) M) antagonized the ability of PAF to cause EADs and PA. CONCLUSIONS: PAF consistently exerts arrhythmogenic effects on the membrane of ventricular myocytes. Since PAF is liberated by activated neutrophils and since activated neutrophils migrate into ischemic myocardium on reperfusion, we judge that PAF liberated by such neutrophils is an important arrhythmogenic factor for reperfusion arrhythmias. The same mechanism may be a cause of arrhythmias during the evolution of infarction.


Subject(s)
Arrhythmias, Cardiac/metabolism , Platelet Activating Factor/adverse effects , Platelet Membrane Glycoproteins/antagonists & inhibitors , Pyridinium Compounds/pharmacology , Receptors, Cell Surface , Receptors, G-Protein-Coupled , Animals , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/chemically induced , Dogs , Dose-Response Relationship, Drug , Female , Lidocaine/pharmacology , Male , Membrane Potentials/drug effects , Patch-Clamp Techniques , Tetrodotoxin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...