Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Sci Rep ; 10(1): 1385, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31992800

ABSTRACT

CHF5633 is a novel synthetic clinical pulmonary surfactant preparation composed by two phospholipid species, dipalmitoyl phosphatidylcholine (DPPC) and palmitoyloleoyl phosphatidylglycerol (POPG), and synthetic analogues of the hydrophobic surfactant proteins SP-B and SP-C. In this study, the interfacial properties of CHF5633 in the absence and in the presence of inhibitory serum proteins have been assessed in comparison with a native surfactant purified from porcine lungs and with poractant alpha, a widely used clinical surfactant preparation. The study of the spreading properties of CHF5633 in a Wilhelmy balance, its ability to adsorb and accumulate at air-liquid interfaces as revealed by a multiwell fluorescence assay, and its dynamic behavior under breathing-like compression-expansion cycling in a Captive Bubble Surfactometer (CBS), all revealed that CHF5633 exhibits a good behavior to reduce and sustain surface tensions to values below 5 mN/m. CHF5633 shows somehow slower initial interfacial adsorption than native surfactant or poractant alpha, but a better resistance to inhibition by serum proteins than the animal-derived clinical surfactant, comparable to that of the full native surfactant complex. Interfacial CHF5633 films formed in a Langmuir-Blodgett balance coupled with epifluorescence microscopy revealed similar propensity to segregate condensed lipid domains under compression than films made by native porcine surfactant or poractant alpha. This ability of CHF5633 to segregate condensed lipid phases can be related with a marked thermotropic transition from ordered to disordered membrane phases as exhibited by differential scanning calorimetry (DSC) of CHF5633 suspensions, occurring at similar temperatures but with higher associated enthalpy than that shown by poractant alpha. The good interfacial behavior of CHF5633 tested under physiologically meaningful conditions in vitro and its higher resistance to inactivation by serum proteins, together with its standardized and well-defined composition, makes it a particularly useful therapeutic preparation to be applied in situations associated with lung inflammation and edema, alone or in combined strategies to exploit surfactant-facilitated drug delivery.


Subject(s)
Blood Proteins/chemistry , Drug Delivery Systems , Peptide Fragments , Phosphatidylcholines , Pulmonary Surfactant-Associated Protein B , Pulmonary Surfactant-Associated Protein C , Pulmonary Surfactants , Animals , Biological Products/chemistry , Humans , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Phosphatidylcholines/antagonists & inhibitors , Phosphatidylcholines/chemistry , Phosphatidylglycerols/chemistry , Phospholipids/chemistry , Pulmonary Surfactant-Associated Protein B/antagonists & inhibitors , Pulmonary Surfactant-Associated Protein B/chemistry , Pulmonary Surfactant-Associated Protein C/antagonists & inhibitors , Pulmonary Surfactant-Associated Protein C/chemistry , Pulmonary Surfactants/antagonists & inhibitors , Pulmonary Surfactants/chemistry , Structure-Activity Relationship , Surface Tension , Swine
2.
PLoS One ; 14(8): e0221032, 2019.
Article in English | MEDLINE | ID: mdl-31437171

ABSTRACT

INTRODUCTION: Plasmodium falciparum synthesizes phosphatidylcholine for the membrane development through serine decarboxylase-phosphoethanolamine methyltransferase pathway for growth in human host. Phosphoethanolamine-methyltransferase (PfPMT) is a crucial enzyme for the synthesis of phosphocholine which is a precursor for phosphatidylcholine synthesis and is considered as a pivotal drug target as it is absent in the host. The inhibition of PfPMT may kill malaria parasite and hence is being considered as potential target for rational antimalarial drug designing. METHODS: In this study, we have used computer aided drug designing (CADD) approaches to establish potential PfPMT inhibitors from Asinex compound library virtually screened for ADMET and the docking affinity. The selected compounds were tested for in-vitro schizonticidal, gametocidal and cytotoxicity activity. Nontoxic compounds were further studied for PfPMT enzyme specificity and antimalarial efficacy for P. berghei in albino mice model. RESULTS: Our results have identified two nontoxic PfPMT competitive inhibitors ASN.1 and ASN.3 with better schizonticidal and gametocidal activity which were found to inhibit PfPMT at IC50 1.49µM and 2.31µM respectively. The promising reduction in parasitaemia was found both in orally (50 & 10 mg/kg) and intravenous (IV) (5& 1 mg/kg) however, the better growth inhibition was found in intravenous groups. CONCLUSION: We report that the compounds containing Pyridinyl-Pyrimidine and Phenyl-Furan scaffolds as the potential inhibitors of PfPMT and thus may act as promising antimalarial inhibitor candidates which can be further optimized and used as leads for template based antimalarial drug development.


Subject(s)
Antimalarials/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Malaria/drug therapy , Methyltransferases/antagonists & inhibitors , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Protozoan Proteins/antagonists & inhibitors , Administration, Oral , Amino Acid Sequence , Animals , Antimalarials/pharmacology , Binding Sites , Drug Design , Enzyme Inhibitors/pharmacology , Furans/chemical synthesis , Furans/pharmacology , Injections, Intravenous , Malaria/parasitology , Male , Methyltransferases/chemistry , Methyltransferases/metabolism , Mice , Molecular Docking Simulation , Parasitic Sensitivity Tests , Phosphatidylcholines/antagonists & inhibitors , Phosphatidylcholines/biosynthesis , Plasmodium berghei/enzymology , Plasmodium berghei/growth & development , Plasmodium falciparum/enzymology , Plasmodium falciparum/growth & development , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Pyridines/chemical synthesis , Pyridines/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Sequence Alignment , Sequence Homology, Amino Acid , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
4.
Sci Rep ; 7(1): 5447, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28710476

ABSTRACT

Phospholipids occurring in cell membranes and lipoproteins are converted into oxidized phospholipids (OxPL) by oxidative stress promoting atherosclerotic plaque formation. Here, OxPL were characterized as novel targets in acute and chronic inflammatory pain. Oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (OxPAPC) and its derivatives were identified in inflamed tissue by mass spectrometry and binding assays. They elicited calcium influx, hyperalgesia and induced pro-nociceptive peptide release. Genetic, pharmacological and mass spectrometric evidence in vivo as well as in vitro confirmed the role of transient receptor potential channels (TRPA1 and TRPV1) as OxPAPC targets. Treatment with the monoclonal antibody E06 or with apolipoprotein A-I mimetic peptide D-4F, capturing OxPAPC in atherosclerosis, prevented inflammatory hyperalgesia, and in vitro TRPA1 activation. Administration of D-4F or E06 to rats profoundly ameliorated mechanical hyperalgesia and inflammation in collagen-induced arthritis. These data reveal a clinically relevant role for OxPAPC in inflammation offering therapy for acute and chronic inflammatory pain treatment by scavenging OxPAPC.


Subject(s)
Antibodies, Monoclonal/pharmacology , Apolipoprotein A-I/pharmacology , Arthritis, Experimental/drug therapy , Hyperalgesia/drug therapy , Pain/drug therapy , Phosphatidylcholines/antagonists & inhibitors , TRPA1 Cation Channel/genetics , TRPV Cation Channels/genetics , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Calcium/metabolism , Calcium Signaling/drug effects , Collagen Type II/administration & dosage , Female , Gene Expression , HEK293 Cells , Hindlimb , Humans , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Hyperalgesia/pathology , Male , Nociception/drug effects , Nociception/physiology , Pain/chemically induced , Pain/metabolism , Pain/pathology , Patch-Clamp Techniques , Phosphatidylcholines/metabolism , Phosphatidylcholines/pharmacology , Rats , Rats, Inbred Lew , Rats, Wistar , TRPA1 Cation Channel/antagonists & inhibitors , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism
5.
J Lipid Res ; 53(7): 1304-15, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22550136

ABSTRACT

Oxidation products of 1-palmitoyl-2-arachidonoyl-sn-glycerol-3-phosphatidylcholine (PAPC), referred to as OxPAPC, and an active component, 1-palmitoyl-2-(5,6-epoxyisoprostane E2)-sn-glycero-3-phosphatidylcholine (PEIPC), accumulate in atherosclerotic lesions and regulate over 1,000 genes in human aortic endothelial cells (HAEC). We previously demonstrated that OxPNB, a biotinylated analog of OxPAPC, covalently binds to a number of proteins in HAEC. The goal of these studies was to gain insight into the binding mechanism and determine whether binding regulates activity. In whole cells, N-acetylcysteine inhibited gene regulation by OxPAPC, and blocking cell cysteines with N-ethylmaleimide strongly inhibited the binding of OxPNB to HAEC proteins. Using MS, we demonstrate that most of the binding of OxPAPC to cysteine is mediated by PEIPC. We also show that OxPNB and PEIPE-NB, the analog of PEIPC, bound to a model protein, H-Ras, at cysteines previously shown to regulate activity in response to 15-deoxy-Δ12,14-prostaglandin J2 (15dPGJ2). This binding was observed with recombinant protein and in cells overexpressing H-Ras. OxPAPC and PEIPC compete with OxPNB for binding to H-Ras. 15dPGJ2 and OxPAPC increased H-Ras activity at comparable concentrations. Using microarray analysis, we demonstrate a considerable overlap of gene regulation by OxPAPC, PEIPC, and 15dPGJ2 in HAEC, suggesting that some effects attributed to 15dPGJ2 may also be regulated by PEIPC because both molecules accumulate in inflammatory sites. Overall, we provide evidence for the importance of OxPAPC-cysteine interactions in regulating HAEC function.


Subject(s)
Cysteine/metabolism , Endothelial Cells/metabolism , Phosphatidylcholines/metabolism , Binding Sites , Cells, Cultured , Cysteine/chemistry , Endothelial Cells/drug effects , Ethylmaleimide/pharmacology , Humans , Isoprostanes/chemistry , Isoprostanes/metabolism , Phosphatidylcholines/antagonists & inhibitors , Phosphatidylcholines/chemistry , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/chemistry , Prostaglandin D2/metabolism
6.
Biochimie ; 94(6): 1437-40, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22490726

ABSTRACT

The Human Secreted Group IID Phospholipase A(2) (hsPLA2GIID) may be involved in the human acute immune response. Here we have demonstrated that the hsPLA2GIID presents bactericidal and Ca(2+)-independent liposome membrane-damaging activities and we have compared these effects with the catalytic activity of active-site mutants of the protein. All mutants showed reduced hydrolytic activity against DOPC:DOPG liposome membranes, however bactericidal effects against Escherichia coli and Micrococcus luteus were less affected, with the D49K mutant retaining 30% killing of the Gram-negative bacteria at a concentration of 10µg/mL despite the absence of catalytic activity. The H48Q mutant maintained Ca(2+)-independent membrane-damaging activity whereas the G30S and D49K mutants were approximately 50% of the wild-type protein, demonstrating that phospholipid bilayer permeabilization by the hsPLA2GIID is independent of catalytic activity. We suggest that this Ca(2+)-independent damaging activity may play a role in the bactericidal function of the protein.


Subject(s)
Escherichia coli/drug effects , Group II Phospholipases A2/metabolism , Group II Phospholipases A2/pharmacology , Micrococcus luteus/drug effects , Anti-Bacterial Agents/pharmacology , Bacteria , Calcium/pharmacology , Cell Membrane/drug effects , Group II Phospholipases A2/genetics , Humans , Hydrolysis , Lipid Bilayers , Liposomes , Phosphatidylcholines/antagonists & inhibitors , Phosphatidylglycerols/antagonists & inhibitors
7.
Amino Acids ; 39(1): 135-44, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19946788

ABSTRACT

LyeTx I, an antimicrobial peptide isolated from the venom of Lycosa erythrognatha, known as wolf spider, has been synthesised and its structural profile studied by using the CD and NMR techniques. LyeTx I has shown to be active against bacteria (Escherichia coli and Staphylococcus aureus) and fungi (Candida krusei and Cryptococcus neoformans) and able to alter the permeabilisation of L: -alpha-phosphatidylcholine-liposomes (POPC) in a dose-dependent manner. In POPC containing cholesterol or ergosterol, permeabilisation has either decreased about five times or remained unchanged, respectively. These results, along with the observed low haemolytic activity, indicated that antimicrobial membranes, rather than vertebrate membranes seem to be the preferential targets. However, the complexity of biological membranes compared to liposomes must be taken in account. Besides, other membrane components, such as proteins and even specific lipids, cannot be discarded to be important to the preferential action of the LyeTx I to the tested microorganisms. The secondary structure of LyeTx I shows a small random-coil region at the N-terminus followed by an alpha-helix that reached the amidated C-terminus, which might favour the peptide-membrane interaction. The high activity against bacteria together with the moderate activity against fungi and the low haemolytic activity have indicated LyeTx I as a good prototype for developing new antibiotic peptides.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Spider Venoms/chemistry , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Antifungal Agents/chemistry , Antifungal Agents/isolation & purification , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/isolation & purification , Candida/drug effects , Cryptococcus neoformans/drug effects , Dose-Response Relationship, Drug , Escherichia coli/drug effects , Microbial Sensitivity Tests , Models, Molecular , Phosphatidylcholines/antagonists & inhibitors , Protein Structure, Secondary , Spiders , Staphylococcus aureus/drug effects
8.
Chem Biol Interact ; 179(2-3): 125-30, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19103186

ABSTRACT

Important biological activities have been described for mesoionic compounds. We previously reported that MI-D (4-phenyl-5-(4-nitro-cinnamoyl)-1,3,4-thiadiazolium-2-phenylamine chloride) inhibited the respiratory chain, collapsed the transmembrane potential, and stimulated ATPase activity in intact rat liver mitochondria. It is known that drugs that affect mitochondrial membrane potential may facilitate the induction of cell death by apoptosis. Mitochondria have also a central role in the generation of reactive oxygen species, therefore it would be important to investigate how MI-D could affect processes related to oxidative stress. In this work, we evaluated the effects of MI-D on the lipoperoxidation and its ability to scavenge free radicals. Interestingly, it was observed that MI-D promoted a strong inhibition of the lipoperoxidation induced by Fe(3+)-ADP/2-oxoglutarate in isolated mitochondria (95%+/-0.27 at the highest concentration of 80 nmol mg(-1) protein) in a dose-dependent manner. However, at the same concentration its effect was less intense (22%+/-3.46) when the lipoperoxidation was initiated by peroxyl radicals generated from the azocompound AAPH. Lipid peroxidation in both coupled and uncoupled submitochondrial particles initiated with Fe(2+)/NADH was also inhibited by MI-D. The inhibition was about four times greater in coupled particles (approximately 34% at 80 nmol mg(-1) protein) in relation to uncoupled. MI-D inhibited the soybean phosphatidylcholine liposomes lipoperoxidation in a dose-dependent manner (5-80 microM) regardless of the radical being generated in lipid or aqueous phase. The mesoionic compound showed ability of scavenging superoxide radical (7, 11 and 31% for 25, 38 and 80 microM, respectively). Our results strongly suggest that the inhibition of lipoperoxidation promoted by MI-D is due to its scavenger action and to its previously described uncoupling effect.


Subject(s)
Cinnamates/pharmacology , Free Radical Scavengers/pharmacology , Lipid Peroxidation/drug effects , Thiadiazoles/pharmacology , Amidines/pharmacology , Animals , Apoptosis/drug effects , Dose-Response Relationship, Drug , Liposomes/antagonists & inhibitors , Liposomes/metabolism , Male , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Oxidative Stress/drug effects , Phosphatidylcholines/antagonists & inhibitors , Phosphatidylcholines/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Time Factors
9.
Antimicrob Agents Chemother ; 50(10): 3381-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17005821

ABSTRACT

A leading bisthiazolium drug, T16, designed to mimic choline, was shown to exert potent antibabesial activity, with 50% inhibitory concentrations of 28 and 7 nM against Babesia divergens and B. canis, respectively. T16 accumulated inside Babesia-infected erythrocytes (cellular accumulation ratio, >60) by a saturable process with an apparent K(m) of 0.65 microM. Subcellular fractionation of Babesia parasites revealed the accumulation of T16 into a low-density fraction, while in malaria-infected erythrocytes a significant fraction of the drug was associated with heme malaria pigment. T16 exerts an early and specific inhibition of the de novo biosynthesis of phosphatidylcholine both in B. divergens- and Plasmodium falciparum-infected erythrocytes. Choline accumulation into isolated Babesia parasites was highly sensitive to inhibition by T16. These data are consistent with the hypothesis that bisthiazolium drugs target the de novo phosphatidylcholine biosynthesis of intraerythrocytic hematozoan parasites. In malaria parasites, which generate ferriprotoporphyrin IX during hemoglobin digestion, T16 binding to heme may enhance the accumulation and activity of the drug. The selectivity of accumulation and potent activity of this class of drug into parasite-infected erythrocytes offers unique advantages over more traditional antihematozoan drugs.


Subject(s)
Antiprotozoal Agents/pharmacology , Babesia/drug effects , Erythrocytes/parasitology , Phosphatidylcholines/antagonists & inhibitors , Plasmodium falciparum/drug effects , Thiazoles/pharmacology , Animals , Antimalarials/pharmacology , Babesia/metabolism , Babesiosis/parasitology , Erythrocytes/drug effects , Hemolysis , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Phosphatidylcholines/biosynthesis , Plasmodium falciparum/metabolism
10.
Biochim Biophys Acta ; 1686(1-2): 85-99, 2004 Nov 08.
Article in English | MEDLINE | ID: mdl-15522825

ABSTRACT

During entry into the cell cycle a phosphatidylcholine (PC) metabolic cycle is activated. We have examined the hypothesis that PC synthesis during the G(0) to G(1) transition is controlled by one or more lipid products of PC turnover acting directly on the rate-limiting enzyme in the synthesis pathway, CTP: phosphocholine cytidylyltransferase (CCT). The acceleration of PC synthesis was two- to threefold during the first hour after addition of serum to quiescent IIC9 fibroblasts. The rate increased to approximately 15-fold above the basal rate during the second hour. The production of arachidonic acid, diacylglycerol (DAG), and phosphatidic acid (PA) preceded the second, rapid phase of PC synthesis. However, an increase in the cellular content of these lipid mediators was detected only for DAG. CCT activation and translocation to membranes accompanied the second phase of the PC synthesis acceleration. Bromoenol lactone (BEL), an inhibitor of calcium-independent phospholipase A(2) and PA phosphatase, blocked production of fatty acids and DAG, inhibited both phases of the PC synthesis response to serum, and reduced CCT activity and membrane affinity. The effect of BEL on PC synthesis was partially reversed by in situ generation of DAG via exogenous PC-specific phospholipase C to generate approximately 2-fold elevation in PC-derived DAG. Exogenous arachidonic acid also partially reversed the inhibition by BEL, but only at a concentration that generated a supra-physiological cellular content of free fatty acid. 1-Butanol, which blocks PA production, had no effect on DAG generation, or on PC synthesis. We conclude that fatty acids and DAG could contribute to the initial slow phase of the PC synthesis response. DAG is the most likely lipid regulator of CCT activity and the rapid phase of PC synthesis. However, processes other than direct activation of CCT by lipid mediators likely contribute to the highly accelerated phase during entry into the cell cycle.


Subject(s)
Cell Cycle/physiology , Fatty Acids/metabolism , Phosphatidylcholines/biosynthesis , Second Messenger Systems/physiology , Animals , Arachidonic Acid/antagonists & inhibitors , Arachidonic Acid/chemistry , Arachidonic Acid/metabolism , Arachidonic Acid/pharmacology , Bridged-Ring Compounds/pharmacology , Butanols/pharmacology , Cattle , Cell Line , Choline/analogs & derivatives , Choline/metabolism , Choline-Phosphate Cytidylyltransferase/metabolism , Cricetinae , Cricetulus , Diglycerides/chemistry , Diglycerides/metabolism , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , G1 Phase/physiology , Glycerophospholipids/chemistry , Glycerophospholipids/metabolism , Group VI Phospholipases A2 , Humans , Naphthalenes/antagonists & inhibitors , Naphthalenes/pharmacology , Norbornanes , Phosphatidic Acids/chemistry , Phosphatidic Acids/metabolism , Phosphatidylcholines/antagonists & inhibitors , Phosphatidylcholines/metabolism , Phospholipases A/antagonists & inhibitors , Phospholipases A/metabolism , Pyrones/antagonists & inhibitors , Pyrones/pharmacology , Resting Phase, Cell Cycle/physiology , Serum Albumin, Bovine/pharmacology , Thiocarbamates , Thiones/pharmacology , Type C Phospholipases/pharmacology
11.
Biochim Biophys Acta ; 1636(2-3): 99-107, 2004 Mar 22.
Article in English | MEDLINE | ID: mdl-15164757

ABSTRACT

The anticancer drug hexadecylphosphocholine (HePC), an alkyl-lysophospholipid analog (ALP), has been shown to induce apoptosis and inhibit the synthesis of phosphatidylcholine (PC) in a number of cell lines. We investigated whether inhibition of PC synthesis plays a major causative role in the induction of apoptosis by HePC. We therefore directly compared the apoptosis caused by HePC in CHO cells to the apoptotic process in CHO-MT58 cells, which contain a genetic defect in PC synthesis. HePC-provoked apoptosis was found to differ substantially from the apoptosis observed in MT58 cells, since it was (i) not accompanied by a large decrease in the amount of PC and diacylglycerol (DAG), (ii) not preceded by induction of the pro-apoptotic protein GADD153/CHOP, and (iii) not dependent on the synthesis of new proteins. Furthermore, lysoPC as well as lysophosphatidylethanolamine (lysoPE) could antagonize the apoptosis induced by HePC, whereas only lysoPC was able to rescue MT58 cells. HePC also induced a rapid externalisation of phosphatidylserine (PS). These observations suggest that inhibition of PC synthesis is not the primary pathway in HePC-induced apoptosis.


Subject(s)
Apoptosis/drug effects , Phosphatidylcholines/biosynthesis , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/pharmacology , Animals , CHO Cells , Cricetinae , Phosphatidylcholines/antagonists & inhibitors
12.
Biochemistry ; 43(14): 4227-39, 2004 Apr 13.
Article in English | MEDLINE | ID: mdl-15065867

ABSTRACT

The human surfactant protein A (SP-A) locus consists of two functional genes, SP-A1 and SP-A2, with several alleles characterized for each gene. Functional variations between SP-A1 and SP-A2 variants either before or after ozone exposure have been observed. To understand the basis of these differences, we studied SP-A1 and SP-A2 variants by comparing coding sequences, oligomerization patterns under various conditions, composition of oligomers with regard to amino terminal sequence isoforms, biological activity (regulation of phosphatidylcholine (PC) secretion by alveolar type II cells), and the impact of ozone-induced oxidation. We found that (i) the SP-A1 (6A(4)) allele is the most divergent from all SP-A2 alleles, particularly from the SP-A2 (1A(1)). (ii) Differences exist in oligomerization among SP-A1, SP-A2, and coexpressed SP-A1/SP-A2, with higher order multimers (i.e., consisting of more subunits) observed for SP-A1 than for SP-A2 variants. Differences among SP-A1 or SP-A2 gene products are minimal. (iii) Amino acid variants in the amino terminal sequences are observed after signal peptide removal, including variants with an extra cysteine. (iv) Oxidation is observed after ozone exposure, involving several SP-A residues that include cysteine, methionine, and tryptophan. (v) The SP-A2 variant (1A(0)) and the coexpressed protein 1A(0)/6A(2) inhibit ATP-stimulated PC secretion from alveolar type II cells to a greater extent than SP-A1 (6A(2)), a biologic activity that was susceptible to ozone treatment.


Subject(s)
Pulmonary Surfactant-Associated Protein A/analogs & derivatives , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein A/physiology , Animals , CHO Cells , Cricetinae , Humans , Oxidation-Reduction/drug effects , Ozone/toxicity , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/physiology , Phosphatidylcholines/antagonists & inhibitors , Phosphatidylcholines/metabolism , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein A/metabolism , Rats , Rats, Sprague-Dawley , Sequence Analysis, Protein , Sequence Homology, Amino Acid , Transfection
13.
J Biol Chem ; 277(31): 27765-71, 2002 Aug 02.
Article in English | MEDLINE | ID: mdl-12021268

ABSTRACT

15-Deoxyspergualin (DSG) strongly inhibited growth of mouse EL-4 lymphoma cells in vitro and in vivo. It significantly prolonged the survival days of EL-4-transplanted mice. In vitro study revealed that its antiproliferative effect appeared only after 2 days of treatment. At that time, protein synthesis was significantly inhibited rather than DNA and RNA syntheses. Furthermore, DSG induced apoptosis without arresting the cell cycle. p70 S6 kinase (p70S6K), a key molecule in protein synthesis, was inhibited by 2 days of treatment of DSG. Akt, an upstream kinase of p70S6K, was also deactivated by 2 days of treatment of DSG. Hsp90 is reported to bind to and stabilize Akt kinase and also to bind to DSG. Yet DSG did not inhibit the binding of Hsp90 to Akt kinase. PI3-kinase, an activator of Akt, was not affected by DSG treatment. However, when we looked into phospholipid synthesis, we found that DSG inhibited phosphatidylcholine (PC) synthesis strongly rather than phosphatidylinositol even by 1 day of treatment. Moreover, DSG failed to inhibit Akt kinase activation and PC synthesis in DSG-less sensitive human K562 leukemia cells. These results demonstrate that DSG inhibits tumor cell growth through the inhibition of protein synthesis and induction of apoptosis, which is caused by the down-regulation of Akt kinase and p70S6K. It is also indicated that the down-regulation of Akt kinase by DSG should not depend on PI3-kinase and Hsp90. There might be possible involvement of PC in Akt kinase activity.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Guanidines/pharmacology , Lymphoma/drug therapy , Phosphatidylcholines/biosynthesis , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/antagonists & inhibitors , Animals , Antibiotics, Antineoplastic/therapeutic use , Cell Division/drug effects , Enzyme Activation/drug effects , Female , Guanidines/therapeutic use , Humans , K562 Cells , Kinetics , Mice , Mice, Inbred C57BL , Phosphatidylcholines/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-akt , Ribosomal Protein S6 Kinases/metabolism , Time Factors , Tumor Cells, Cultured
14.
Prostaglandins Other Lipid Mediat ; 67(3-4): 197-208, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12013527

ABSTRACT

This study was designed to investigate the effect of platelet-activating factor (PAF) in the secretory response of type II pneumocytes, that are involved in the synthesis and secretion of the pulmonary surfactant. PAF increased phosphatidylcholine secretion in a concentration-dependent manner in the 10(-5) - 10(-10) M range, with a maximum phosphatidylcholine secretion of up to 3.3 fold the basal values (3.4 +/- 0.3% phosphatidylcholine secreted). This effect was prevented by the synthetic PAF-receptor antagonist WEB 2086. A study of the mechanism through which PAF exerts its stimulatory effect was carried out adding different agents that are well known stimulants of phosphatidylcholine secretion. Thus, PAF increased the TPA- and terbutaline-stimulated phosphatidylcholine secretion, that are PKC and PKA activators respectively, suggesting the involvement of both protein kinases in the process. This involvement was further supported by the use of inhibitors of protein kinases and by the stimulation of cAMP production in type II pneumocytes incubated with PAF.


Subject(s)
Lung/metabolism , Phosphatidylcholines/metabolism , Platelet Activating Factor/pharmacology , Animals , Azepines/metabolism , Azepines/pharmacology , Cell Line , Cyclic AMP/analysis , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinase Type II , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , L-Lactate Dehydrogenase/metabolism , Lung/cytology , Lung/drug effects , Male , Phosphatidylcholines/antagonists & inhibitors , Phosphatidylcholines/biosynthesis , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rats , Rats, Wistar , Stimulation, Chemical , Time Factors , Triazoles/metabolism , Triazoles/pharmacology
15.
Am J Physiol Lung Cell Mol Physiol ; 281(1): L98-L107, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11404252

ABSTRACT

Tissue injury in inflammation involves the release of several cytokines that activate sphingomyelinases and generate ceramide. In the lung, the impaired metabolism of surfactant phosphatidylcholine (PC) accompanies this acute and chronic injury. These effects are long-lived and extend beyond the time frame over which tumor necrosis factor (TNF)-alpha and interleukin-1beta are elevated. In this paper, we demonstrate that in H441 lung cells these two processes, cytokine-induced metabolism of sphingomyelin and the inhibition of PC metabolism, are directly interrelated. First, metabolites of sphingomyelin hydrolysis themselves inhibit key enzymes necessary for restoring homeostasis between sphingomyelin and its metabolites. Ceramide stimulates sphingomyelinases as effectively as TNF-alpha, thereby amplifying the sphingomyelinase activation, and TNF-alpha, ceramide, and sphingosine all inhibit PC:ceramide phosphocholine transferase (sphingomyelin synthase), the enzyme that restores homeostasis between sphingomyelin and ceramide pools. Second, ceramide inhibits PC synthesis, probably because of its effects on CTP:phosphocholine cytidylyltransferase, the rate-limiting enzymatic step in de novo PC synthesis. The data presented here suggest that TNF-alpha may be an inhibitor of phospholipid metabolism in inflammatory tissue injury. These actions may be amplified because of the ability of metabolites of sphingomyelin to inhibit the pathways that should restore the normal ceramide-sphingomyelin homeostasis.


Subject(s)
Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Sphingomyelins/metabolism , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Ceramides/pharmacology , Diacylglycerol Cholinephosphotransferase/metabolism , Enzyme Inhibitors/pharmacology , Humans , Phosphatidylcholines/antagonists & inhibitors , Sphingolipids/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
16.
J Neurochem ; 77(1): 13-22, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11279257

ABSTRACT

Glutamate receptor overactivation induces excitotoxic neuronal death, but the contribution of glutamate receptor subtypes to this excitotoxicity is unclear. We have previously shown that excitotoxicity by NMDA receptor overactivation is associated with choline release and inhibition of phosphatidylcholine synthesis. We have now investigated whether the ability of non-NMDA ionotropic glutamate receptor subtypes to induce excitotoxicity is related to the ability to inhibit phosphatidylcholine synthesis. alpha-Amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA)-induced a concentration-dependent increase in extracellular choline and inhibited phosphatidylcholine synthesis when receptor desensitization was prevented. Kainate released choline and inhibited phosphatidylcholine synthesis by an action at AMPA receptors, because these effects of kainate were blocked by the AMPA receptor antagonist LY300164. Selective activation of kainate receptors failed to release choline, even when kainate receptor desensitization was prevented. The inhibition of phosphatidylcholine synthesis evoked by activation of non-desensitizing AMPA receptors was followed by neuronal death. In contrast, specific kainate receptor activation, which did not inhibit phosphatidylcholine synthesis, did not produce neuronal death. Choline release and inhibition of phosphatidylcholine synthesis were induced by AMPA at non-desensitizing AMPA receptors well before excitotoxicity. Furthermore, choline release by AMPA required the entry of Ca(2+) through the receptor channel. Our results show that AMPA, but not kainate, receptor overactivation induces excitotoxic cell death, and that this effect is directly related to the ability to inhibit phosphatidylcholine synthesis. Moreover, these results indicate that inhibition of phosphatidylcholine synthesis is an early event of the excitotoxic process, downstream of glutamate receptor-mediated Ca(2+) overload.


Subject(s)
Neurons/metabolism , Phosphatidylcholines/biosynthesis , Receptors, AMPA/metabolism , Receptors, Kainic Acid/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Benzothiadiazines/pharmacology , Calcium/metabolism , Cell Death/drug effects , Cells, Cultured , Choline/metabolism , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Neurons/cytology , Neurons/drug effects , Phosphatidylcholines/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Receptors, AMPA/agonists , Receptors, AMPA/antagonists & inhibitors , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
17.
Cancer Res ; 60(18): 5204-13, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-11016649

ABSTRACT

De novo production of phosphatidic acid (PA) in tumor cells is required for phospholipid biosynthesis and growth of tumor cells. In addition, PA production by phospholipase D has been cited among the effects of certain oncogenes and growth factors. In this report, it has been demonstrated that enhanced phospholipid metabolism through PA in tumor cells can be exploited pharmacologically for development of anticancer agents, such as CT-2584, a cancer chemotherapeutic drug candidate currently in Phase II clinical trials. By inhibiting CTP:choline-phosphate cytidylyltransferase (CT), CT-2584 caused de novo phospholipid biosynthesis via PA to be shunted away from phosphatidylcholine (PC) and into phosphatidylinositol (PI), the latter of which was doubled in a variety of CT-2584-treated tumor cell lines. In contrast, cytotoxic concentrations of cisplatin did not induce accumulation of PI, indicating that PI elevation by CT-2584 was not a general consequence of chemotherapy-induced cell death. Consistent with this mechanism of action, propranolol, an inhibitor of PA phosphohydrolase and phosphatidylcholine biosynthesis, was also cytotoxic to tumor cell lines, induced PI accumulation, and potentiated the activity of CT-2584 in cytotoxicity assays. As expected from biophysical properties of anionic phospholipids on cellular membranes, CT-2584 cytotoxicity was associated with disruption and swelling of endoplasmic reticulum and mitochondria. We conclude that CT-2584 effects a novel mechanism of cytotoxicity to cancer cells, involving a specific modulation of phospholipid metabolism.


Subject(s)
Antineoplastic Agents/toxicity , Phosphatidylcholines/biosynthesis , Phosphatidylinositols/metabolism , Xanthines/toxicity , Adrenergic beta-Antagonists/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Death/drug effects , Cell Death/physiology , Cell Survival/drug effects , Choline Kinase , Choline-Phosphate Cytidylyltransferase/metabolism , Cytidine Diphosphate Diglycerides/metabolism , Drug Synergism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Phosphatidic Acids/biosynthesis , Phosphatidic Acids/metabolism , Phosphatidylcholines/antagonists & inhibitors , Phosphatidylinositols/biosynthesis , Propranolol/pharmacology , Substrate Specificity , Tumor Cells, Cultured
18.
Biochemistry ; 39(19): 5838-44, 2000 May 16.
Article in English | MEDLINE | ID: mdl-10801334

ABSTRACT

Transmembrane phospholipid redistribution (scrambling), leading to exposure of phosphatidylserine on the cell surface, plays a physiological role to induce platelet procoagulant activity and clearance of injured or apoptotic cells. Scrambling is generally attributed to an increase in intracellular Ca(2+) and would be mediated by a protein (scramblase), whose activity could be modulated by cofactors. We reported previously that phosphatidylinositol 4,5-bisphosphate (PIP(2)) is a positive regulator of Ca(2+)-induced scrambling. We show here, using inside-out vesicles from erythrocyte membranes, that a pleckstrin homology (PH) domain, which interacts with high affinity with PIP(2), inhibited Ca(2+)-induced scrambling, confirming the role of PIP(2). As Ca(2+) is known to interact with PIP(2) and to promote the formation of lateral domains of acidic phospholipids in membranes, we investigated whether PIP(2) domain formation could be involved in scrambling. Spermine, polylysine, and MARCKS (151-175) peptide caused scrambling in parallel to their reported ability to form domains of acidic phospholipids, including PIP(2). Similarly, neomycine, another PIP(2)-interacting polycation, induced scrambling. A PIP(2) antibody was also found to induce scrambling, presumably by a similar mechanism, since phospholipid antibodies are known to promote phospholipid capping. In conclusion, Ca(2+) is not the sole inducer of scrambling, and formation of PIP(2) domains could play a critical role in this process.


Subject(s)
Erythrocyte Membrane/chemistry , Phosphatidylinositol 4,5-Diphosphate/biosynthesis , Phospholipids/biosynthesis , Phospholipids/chemistry , Antibodies, Monoclonal/pharmacology , Blood Platelets/chemistry , Blood Proteins/pharmacology , Calcium/antagonists & inhibitors , Calcium/pharmacology , Cations/pharmacology , Erythrocyte Membrane/metabolism , Humans , Hydrogen-Ion Concentration , Peptide Fragments/pharmacology , Phosphatidylcholines/antagonists & inhibitors , Phosphatidylcholines/metabolism , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/immunology , Phospholipids/metabolism , Phosphoproteins/pharmacology , Polyamines/pharmacology , Polyelectrolytes , Protein Structure, Tertiary , Sequence Homology, Amino Acid
19.
Phytother Res ; 13(6): 479-83, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10479757

ABSTRACT

The antioxidative effect of ganhuangenin (GHG), isolated from Scutellaria baicalensis Georgi, was examined by measuring its ability to suppress the formation of phospatidylcholine hydroperoxide (PCOOH). The results show that a pretreatment with GHG effectively suppressed PCOOH formation, which was initiated by the peroxyl-generating oxidant, AAPH (2,2'-azobis-2-aminopropane hydrochloride). The protective action of GHG against the formation of the PCOOH was observed in liver, lung, and kidney. When compared with other known antioxidants, we found the antioxidative potency of GHG to be greater than that of alpha-tocopherol. Our data strongly indicate that GHG is a powerful antioxidant against lipid peroxidation and is, therefore, responsible for this prophylactic effect.


Subject(s)
Antioxidants/pharmacology , Flavonoids/pharmacology , Lipid Peroxidation/drug effects , Microsomes, Liver/metabolism , Plant Extracts/chemistry , Animals , Male , Medicine, Chinese Traditional , Microsomes, Liver/drug effects , Phosphatidylcholines/antagonists & inhibitors , Plant Extracts/pharmacology , Plant Roots , Quercetin/pharmacology , Rats , Rats, Wistar , Vitamin E/pharmacology , beta Carotene/pharmacology
20.
Eur J Biochem ; 264(1): 152-60, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10447683

ABSTRACT

Phospholipids and sphingolipids are important precursors of lipid-derived second messengers such as diacylglycerol and ceramide, which participate in several signal transduction pathways and in that way mediate the effects of various agonists. The cross-talk between glycerophospholipid and sphingolipid metabolism was investigated by examining the effects of cell-permeable ceramides on phosphatidylcholine (PtdCho) and phosphatidylethanolamine (PtdEtn) synthesis in Rat-2 fibroblasts. Addition of short-chain C6-ceramide to the cells resulted in a dose- and time-dependent inhibition of the CDP-pathways for PtdCho and PtdEtn synthesis. Treatment of cells for 4 h with 50 microM C6-ceramide caused an 83% and a 56% decrease in incorporation of radiolabelled choline and ethanolamine into PtdCho and PtdEtn, respectively. Exposure of the cells for longer time-periods (>/= 16 h) to 50 microM C6-ceramide resulted in apoptosis. The structural analogue dihydro-C6-ceramide did not affect PtdCho and PtdEtn synthesis. In pulse-chase experiments, radioactive choline and ethanolamine accumulated in CDP-choline and CDP-ethanolamine under the influence of C6-ceramide, suggesting that synthesis of both PtdCho and PtdEtn were inhibited at the final step in the CDP-pathways. Indeed, cholinephosphotransferase and ethanolaminephosphotransferase activities in membrane fractions from C6-ceramide-treated cells were reduced by 64% and 43%, respectively, when compared with control cells. No changes in diacylglycerol mass levels or synthesis of diacylglycerol from radiolabelled palmitate were observed. It was concluded that C6-ceramide affected glycerophospholipid synthesis predominantly by inhibition of the step in the CDP-pathways catalysed by cholinephosphotransferase and ethanolaminephosphotransferase.


Subject(s)
Ceramides/pharmacology , Phosphatidylcholines/antagonists & inhibitors , Phosphatidylethanolamines/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line , Cell Membrane Permeability , Ceramides/metabolism , Choline/metabolism , Diglycerides/biosynthesis , Diglycerides/metabolism , Dose-Response Relationship, Drug , Ethanolamine/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Palmitic Acid/metabolism , Phosphatidylcholines/biosynthesis , Phosphatidylethanolamines/biosynthesis , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...