Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nat Commun ; 11(1): 4480, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32900992

ABSTRACT

Macroautophagy initiates by formation of isolation membranes, but the source of phospholipids for the membrane biogenesis remains elusive. Here, we show that autophagic membranes incorporate newly synthesized phosphatidylcholine, and that CTP:phosphocholine cytidylyltransferase ß3 (CCTß3), an isoform of the rate-limiting enzyme in the Kennedy pathway, plays an essential role. In starved mouse embryo fibroblasts, CCTß3 is initially recruited to autophagic membranes, but upon prolonged starvation, it concentrates on lipid droplets that are generated from autophagic degradation products. Omegasomes and isolation membranes emanate from around those lipid droplets. Autophagy in prolonged starvation is suppressed by knockdown of CCTß3 and is enhanced by its overexpression. This CCTß3-dependent mechanism is also present in U2OS, an osteosarcoma cell line, and autophagy and cell survival in starvation are decreased by CCTß3 depletion. The results demonstrate that phosphatidylcholine synthesis through CCTß3 activation on lipid droplets is crucial for sustaining autophagy and long-term cell survival.


Subject(s)
Autophagy/physiology , Choline-Phosphate Cytidylyltransferase/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Animals , Autophagosomes/metabolism , Cell Line, Tumor , Cell Survival , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Choline-Phosphate Cytidylyltransferase/genetics , Culture Media , Enzyme Activation , Gene Knockdown Techniques , Humans , Lipid Droplets/metabolism , Mice , Models, Biological , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Osteosarcoma/metabolism , Osteosarcoma/pathology , Phosphatidylcholines/metabolism
2.
J Biol Chem ; 294(43): 15862-15874, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31439667

ABSTRACT

De novo phosphatidylcholine (PC) biosynthesis via the Kennedy pathway involves highly endergonic biochemical reactions that must be fine-tuned with energy homeostasis. Previous studies have shown that CTP:phosphocholine cytidylyltransferase (CCT) is an important regulatory enzyme in this pathway and that its activity can be controlled at both transcriptional and posttranslational levels. Here we identified an important additional mechanism regulating plant CCT1 activity. Comparative analysis revealed that Arabidopsis CCT1 (AtCCT1) contains catalytic and membrane-binding domains that are homologous to those of rat CCT1. In contrast, the C-terminal phosphorylation domain important for stringent regulation of rat CCT1 was apparently missing in AtCCT1. Instead, we found that AtCCT1 contains a putative consensus site (Ser-187) for modification by sucrose nonfermenting 1-related protein kinase 1 (SnRK1 or KIN10/SnRK1.1), involved in energy homeostasis. Phos-tag SDS-PAGE coupled with MS analysis disclosed that SnRK1 indeed phosphorylates AtCCT1 at Ser-187, and we found that AtCCT1 phosphorylation substantially reduces its activity by as much as 70%. An S187A variant exhibited decreased activity, indicating the importance of Ser-187 in catalysis, and this variant was less susceptible to SnRK1-mediated inhibition. Protein truncation and liposome binding studies indicated that SnRK1-mediated AtCCT1 phosphorylation directly affects the catalytic domain rather than interfering with phosphatidate-mediated AtCCT1 activation. Overexpression of the AtCCT1 catalytic domain in Nicotiana benthamiana leaves increased PC content, and SnRK1 co-expression reduced this effect. Taken together, our results suggest that SnRK1 mediates the phosphorylation and concomitant inhibition of AtCCT1, revealing an additional mode of regulation for this key enzyme in plant PC biosynthesis.


Subject(s)
Arabidopsis Proteins/antagonists & inhibitors , Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Choline-Phosphate Cytidylyltransferase/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Arabidopsis Proteins/chemistry , Catalytic Domain , Choline-Phosphate Cytidylyltransferase/chemistry , Conserved Sequence , Evolution, Molecular , Kinetics , Models, Biological , Phosphorylation , Phosphorylcholine/metabolism , Phosphoserine/metabolism , Plant Leaves/genetics , Protein Domains , Rats , Structural Homology, Protein , Nicotiana/genetics
3.
Sci Rep ; 8(1): 8932, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29895950

ABSTRACT

The plasmodial CTP:phosphocholine cytidylyltransferase (PfCCT) is a promising antimalarial target, which can be inhibited to exploit the need for increased lipid biosynthesis during the erythrocytic life stage of Plasmodium falciparum. Notable structural and regulatory differences of plasmodial and mammalian CCTs offer the possibility to develop species-specific inhibitors. The aim of this study was to use CHO-MT58 cells expressing a temperature-sensitive mutant CCT for the functional characterization of PfCCT. We show that heterologous expression of wild type PfCCT restores the viability of CHO-MT58 cells at non-permissive (40 °C) temperatures, whereas catalytically perturbed or structurally destabilized PfCCT variants fail to provide rescue. Detailed in vitro characterization indicates that the H630N mutation diminishes the catalytic rate constant of PfCCT. The flow cytometry-based rescue assay provides a quantitative readout of the PfCCT function opening the possibility for the functional analysis of PfCCT and the high throughput screening of antimalarial compounds targeting plasmodial CCT.


Subject(s)
Choline-Phosphate Cytidylyltransferase/metabolism , Phosphatidylcholines/biosynthesis , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Animals , Antimalarials/pharmacology , Biocatalysis/drug effects , Biosynthetic Pathways/drug effects , CHO Cells , Cell Survival/drug effects , Cell Survival/genetics , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Choline-Phosphate Cytidylyltransferase/genetics , Cricetinae , Cricetulus , Gene Expression Regulation, Enzymologic/drug effects , Mutation , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Temperature
4.
J Biol Chem ; 293(18): 7070-7084, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29519816

ABSTRACT

The activity of CTP:phosphocholine cytidylyltransferase (CCT), a key enzyme in phosphatidylcholine synthesis, is regulated by reversible interactions of a lipid-inducible amphipathic helix (domain M) with membrane phospholipids. When dissociated from membranes, a portion of the M domain functions as an auto-inhibitory (AI) element to suppress catalysis. The AI helix from each subunit binds to a pair of α helices (αE) that extend from the base of the catalytic dimer to create a four-helix bundle. The bound AI helices make intimate contact with loop L2, housing a key catalytic residue, Lys122 The impacts of the AI helix on active-site dynamics and positioning of Lys122 are unknown. Extensive MD simulations with and without the AI helix revealed that backbone carbonyl oxygens at the point of contact between the AI helix and loop L2 can entrap the Lys122 side chain, effectively competing with the substrate, CTP. In silico, removal of the AI helices dramatically increased αE dynamics at a predicted break in the middle of these helices, enabling them to splay apart and forge new contacts with loop L2. In vitro cross-linking confirmed the reorganization of the αE element upon membrane binding of the AI helix. Moreover, when αE bending was prevented by disulfide engineering, CCT activation by membrane binding was thwarted. These findings suggest a novel two-part auto-inhibitory mechanism for CCT involving capture of Lys122 and restraint of the pliable αE helices. We propose that membrane binding enables bending of the αE helices, bringing the active site closer to the membrane surface.


Subject(s)
Choline-Phosphate Cytidylyltransferase/chemistry , Animals , Binding Sites , Binding, Competitive , Catalysis , Catalytic Domain , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Glycine/chemistry , Hydrogen Bonding , Lysine/chemistry , Molecular Dynamics Simulation , Protein Conformation , Protein Domains , Protein Multimerization , Rats
5.
FEBS Lett ; 589(9): 992-1000, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25771858

ABSTRACT

Phosphatidylcholine is the major lipid component of the malaria parasite membranes and is required for parasite multiplication in human erythrocytes. Plasmodium falciparum CTP:phosphocholine cytidylyltransferase (PfCCT) is the rate-limiting enzyme of the phosphatidylcholine biosynthesis pathway and thus considered as a potential antimalarial target. In contrast to its mammalian orthologs, PfCCT contains a duplicated catalytic domain. Here, we show that both domains are catalytically active with similar kinetic parameters. A virtual screening strategy allowed the identification of a drug-size molecule competitively inhibiting the enzyme. This compound also prevented phosphatidylcholine biosynthesis in parasites and exerted an antimalarial effect. This study constitutes the first step towards a rationalized design of future new antimalarial agents targeting PfCCT.


Subject(s)
Catalytic Domain , Choline-Phosphate Cytidylyltransferase/metabolism , Cytidine Diphosphate Choline/analogs & derivatives , Plasmodium falciparum/enzymology , Protozoan Proteins/metabolism , Amino Acid Sequence , Antimalarials/chemistry , Antimalarials/pharmacology , Biosynthetic Pathways/genetics , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Choline-Phosphate Cytidylyltransferase/genetics , Cytidine Diphosphate Choline/chemistry , Cytidine Diphosphate Choline/pharmacology , Humans , Immunoblotting , Kinetics , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Molecular Structure , Phosphatidylcholines/biosynthesis , Phosphatidylcholines/chemistry , Plasmodium falciparum/genetics , Protein Binding , Protein Structure, Tertiary , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Sequence Homology, Amino Acid
6.
Hypertension ; 65(2): 430-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25452470

ABSTRACT

C-reactive protein (CRP), an innate immune mediator, is elevated in the circulation before symptoms in patients with preeclampsia, a severe hypertensive pregnancy disorder with high mortality and morbidity. However, the specific sources underlying increased CRP and the role of elevated CRP in preeclampsia are undefined. Here, we report that circulating CRP levels are significantly increased in a large cohort of normotensive pregnant individuals when compared with nulligravid women and is further increased in patients with preeclampsia. These findings led us to discover further that placental syncytiotrophoblasts are previously unrecognized cellular sources of CRP and underlie elevated CRP in normotensive pregnant women and the additional increase in patients with preeclampsia. Next, we demonstrated that injection of CRP induces preeclampsia features, including hypertension (157 mm Hg CRP treated versus 119 mm Hg control), proteinuria (35.0 mg/µg CRP treated versus 14.1 mg/µg control), kidney, and placental damage and increased levels of sFlt-1 in pregnant mice but not in nonpregnant mice. Our study implicates that phosphocholine transferase, a placental-specific enzyme post-translationally modifying neurokinin B, is essential for the pathogenic role of CRP in preeclampsia through activation of the neurokinin 3 receptor. Overall, our studies have provided significant new insight on the pathogenic role of CRP in preeclampsia and highlighted innovative therapeutic strategies.


Subject(s)
C-Reactive Protein/physiology , Choline-Phosphate Cytidylyltransferase/physiology , Neurokinin B/metabolism , Pre-Eclampsia/etiology , Receptors, Neurokinin-3/physiology , Animals , Biomarkers , C-Reactive Protein/analysis , C-Reactive Protein/toxicity , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Disease Models, Animal , Double-Blind Method , Female , Humans , Kidney/pathology , Mice , Mice, Inbred C57BL , Phosphorylation , Phosphorylcholine/metabolism , Placenta/pathology , Pre-Eclampsia/chemically induced , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Pregnancy , Protein Binding , Protein Processing, Post-Translational , Quinolines/pharmacology , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Receptors, Neurokinin-3/antagonists & inhibitors , Receptors, Neurokinin-3/metabolism , Recombinant Proteins/toxicity , Single-Blind Method , Vascular Endothelial Growth Factor Receptor-1/blood
7.
Vet Parasitol ; 204(3-4): 104-10, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-24938827

ABSTRACT

Miltefosine, a membrane-active synthetic ether-lipid analogue, has antiproliferative and antiparasitic effects. In this study, the inhibitory effects of miltefosine were evaluated against three Babesia species and Theileria equi in vitro and against Babesia microti in mice. The drug showed significant growth inhibition from an initial parasitemia of 1% for Babesia bovis, Babesia bigemina, Babesia caballi, and T. equi with IC50 values of 25, 10.2, 10.4, and 99 µM, respectively. Complete inhibition was observed at 200 µM of miltefosine on the third day of culture for the three Babesia species and 400 µM on the fourth day for T. equi. Reverse-transcription PCR (RT-PCR) showed that miltefosine inhibited the transcription of choline-phosphate cytidylyltransferase in B. bovis. Miltefosine at a dose rate of 30 mg/kg resulted in a 71.7% inhibition of B. microti growth in BALB/c mice. Miltefosine might be used for drug therapy in babesiosis.


Subject(s)
Antiparasitic Agents/therapeutic use , Babesia/drug effects , Babesiosis/drug therapy , Phosphorylcholine/analogs & derivatives , Theileria/drug effects , Theileriasis/drug therapy , Animals , Babesia/growth & development , Babesiosis/parasitology , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Choline-Phosphate Cytidylyltransferase/metabolism , Female , Mice , Mice, Inbred BALB C , Parasitemia/veterinary , Phosphorylcholine/therapeutic use , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/metabolism , Real-Time Polymerase Chain Reaction/veterinary , Theileria/growth & development , Theileriasis/parasitology
8.
Biochim Biophys Acta ; 1841(9): 1264-71, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24832487

ABSTRACT

In eukaryotic cells, phospholipids are synthesized exclusively in the defined organelles specific for each phospholipid species. To explain the reason for this compartmental specificity in the case of phosphatidylcholine (PC) synthesis, we constructed and characterized a Saccharomyces cerevisiae strain that lacked endogenous phosphatidylethanolamine (PE) methyltransferases but had a recombinant PE methyltransferase from Acetobacter aceti, which was fused with a mitochondrial targeting signal from yeast Pet100p and a 3×HA epitope tag. This fusion protein, which we named as mitopmt, was determined to be localized to the mitochondria by fluorescence microscopy and subcellular fractionation. The expression of mitopmt suppressed the choline auxotrophy of a double deletion mutant of PEM1 and PEM2 (pem1Δpem2Δ) and enabled it to synthesize PC in the absence of choline. This growth suppression was observed even if the Kennedy pathway was inactivated by the repression of PCT1 encoding CTP:phosphocholine cytidylyltransferase, suggesting that PC synthesized in the mitochondria is distributed to other organelles without going through the salvage pathway. The pem1Δpem2Δ strain deleted for PSD1 encoding the mitochondrial phosphatidylserine decarboxylase was able to grow because of the expression of mitopmt in the presence of ethanolamine, implying that PE from other organelles, probably from the ER, was converted to PC by mitopmt. These results suggest that PC could move out of the mitochondria, and raise the possibility that its movement is not under strict directional limitations.


Subject(s)
Gene Expression Regulation, Fungal , Mitochondria/genetics , Phosphatidylcholines/biosynthesis , Phosphatidylethanolamine N-Methyltransferase/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Acetobacter/chemistry , Acetobacter/enzymology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carboxy-Lyases/deficiency , Carboxy-Lyases/genetics , Choline , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Choline-Phosphate Cytidylyltransferase/genetics , Choline-Phosphate Cytidylyltransferase/metabolism , Ethanolamine/metabolism , Genetic Complementation Test , Isoenzymes/deficiency , Isoenzymes/genetics , Mitochondria/enzymology , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Phosphatidylethanolamine N-Methyltransferase/deficiency , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Signal Transduction , Transgenes
9.
J Mol Biol ; 425(9): 1546-64, 2013 May 13.
Article in English | MEDLINE | ID: mdl-23238251

ABSTRACT

CTP:phosphocholine cytidylyltransferase (CCT), the regulatory enzyme in the synthesis of phosphatidylcholine, is activated by binding membranes using a lipid-induced amphipathic helix (domain M). Domain M functions to silence catalysis when CCT is not membrane engaged. The silencing mechanism is unknown. We used photo-cross-linking and mass spectrometry to identify contacts between domain M and other CCT domains in its soluble form. Each of four sites in domain M forged cross-links to the same set of peptides that flank the active site and overlap at helix αE at the base of the active site. These cross-links were broken in the presence of activating lipid vesicles. Mutagenesis of domain M revealed that multiple hydrophobic residues within a putative auto-inhibitory (AI) motif contribute to the contact with helix αE and silencing. Helix αE was confirmed as the docking site for domain M by deuterium exchange analysis. We compared the dynamics and fold stability of CCT domains by site-directed fluorescence anisotropy and urea denaturation. The results suggest a bipartite structure for domain M: a disordered N-terminal portion and an ordered C-terminal AI motif with an unfolding transition identical with that of helix αE. Reduction in hydrophobicity of the AI motif decreased its order and fold stability, as did deletion of the catalytic domain. These results support a model in which catalytic silencing is mediated by the docking of an amphipathic AI motif onto the amphipathic helices αE. An unstructured leash linking αE with the AI motif may facilitate both the silencing contact and its membrane-triggered disruption.


Subject(s)
Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Choline-Phosphate Cytidylyltransferase/chemistry , Membrane Proteins/chemistry , Membrane Proteins/physiology , Allosteric Regulation/genetics , Allosteric Site/genetics , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Choline-Phosphate Cytidylyltransferase/genetics , Membrane Proteins/antagonists & inhibitors , Models, Molecular , Molecular Sequence Data , Protein Interaction Domains and Motifs/genetics , Protein Structure, Secondary/genetics , Rats
10.
Cell Metab ; 14(4): 504-15, 2011 Oct 05.
Article in English | MEDLINE | ID: mdl-21982710

ABSTRACT

Lipid droplets (LDs) are cellular storage organelles for neutral lipids that vary in size and abundance according to cellular needs. Physiological conditions that promote lipid storage rapidly and markedly increase LD volume and surface. How the need for surface phospholipids is sensed and balanced during this process is unknown. Here, we show that phosphatidylcholine (PC) acts as a surfactant to prevent LD coalescence, which otherwise yields large, lipolysis-resistant LDs and triglyceride (TG) accumulation. The need for additional PC to coat the enlarging surface during LD expansion is provided by the Kennedy pathway, which is activated by reversible targeting of the rate-limiting enzyme, CTP:phosphocholine cytidylyltransferase (CCT), to growing LD surfaces. The requirement, targeting, and activation of CCT to growing LDs were similar in cells of Drosophila and mice. Our results reveal a mechanism to maintain PC homeostasis at the expanding LD monolayer through targeted activation of a key PC synthesis enzyme.


Subject(s)
Choline-Phosphate Cytidylyltransferase/metabolism , Lipid Metabolism/physiology , Phosphatidylcholines/physiology , Animals , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Choline-Phosphate Cytidylyltransferase/genetics , Drosophila , Lipolysis , Mice , Oleic Acid/metabolism , Phosphatidylcholines/biosynthesis , RNA Interference , Triglycerides/metabolism
11.
J Biol Chem ; 286(33): 28940-28947, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21705805

ABSTRACT

In the human malaria parasite Plasmodium falciparum, the synthesis of the major and essential membrane phospholipid, phosphatidylcholine, occurs via the CDP-choline and the serine decarboxylase phosphoethanolamine methylation (SDPM) pathways, which are fueled by host choline, serine, and fatty acids. Both pathways share the final two steps catalyzed by two essential enzymes, P. falciparum CTP:phosphocholine cytidylyltransferase (PfCCT) and choline-phosphate transferase (PfCEPT). We identified a novel class of phospholipid mimetics, which inhibit the growth of P. falciparum as well as Leishmania and Trypanosoma species. Metabolic analyses showed that one of these compounds, PG12, specifically blocks phosphatidylcholine biosynthesis from both the CDP-choline and SDPM pathways via inhibition of PfCCT. In vitro studies using recombinant PfCCT showed a dose-dependent inhibition of the enzyme by PG12. The potent antimalarial of this compound, its low cytotoxicity profile, and its established mode of action make it an excellent lead to advance for further drug development and efficacy in vivo.


Subject(s)
Antimalarials/pharmacology , Biomimetic Materials/pharmacology , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Malaria, Falciparum/drug therapy , Phospholipids/pharmacology , Plasmodium falciparum/enzymology , Protozoan Proteins/antagonists & inhibitors , Antimalarials/chemistry , Biomimetic Materials/chemistry , Choline-Phosphate Cytidylyltransferase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Humans , Malaria, Falciparum/enzymology , Phospholipids/chemistry , Protozoan Proteins/metabolism
12.
J Cell Sci ; 124(Pt 24): 4253-66, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22223883

ABSTRACT

Farnesylated prelamin A accumulates when the final endoproteolytic maturation of the protein fails to occur and causes a dysmorphic nuclear phenotype; however, the morphology and mechanisms of biogenesis of these changes remain unclear. We show here that acute prelamin A accumulation after reduction in the activity of the ZMPSTE24 endoprotease by short interfering RNA knockdown, results in the generation of a complex nucleoplasmic reticulum that depends for its formation on the enzyme CTP:phosphocholine-cytidylyltransferase-α (CCT-α, also known as choline-phosphate cytidylyltransferase A). This structure can form during interphase, confirming that it is independent of mitosis and therefore not a consequence of disordered nuclear envelope assembly. Serial-section dual-axis electron tomography reveals that these invaginations can take two forms: one in which the inner nuclear membrane infolds alone with an inter membrane space interior, and the other in which an invagination of both nuclear membranes occurs, enclosing a cytoplasmic core. Both types of invagination can co-exist in one nucleus and both are frequently studded with nuclear pore complexes (NPC), which reduces NPC abundance on the nuclear surface.


Subject(s)
Choline-Phosphate Cytidylyltransferase/physiology , Nuclear Envelope/ultrastructure , Nuclear Proteins/metabolism , Protein Precursors/metabolism , Animals , Cell Nucleus/ultrastructure , Cells, Cultured , Choline-Phosphate Cytidylyltransferase/analysis , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Lamin Type A , Lamin Type B/analysis , Membrane Proteins/antagonists & inhibitors , Metalloendopeptidases/antagonists & inhibitors , Mice , Mitosis , Nuclear Envelope/chemistry , Nuclear Envelope/enzymology , Nuclear Pore/ultrastructure , Nuclear Proteins/analysis , Nuclear Proteins/chemistry , Prenylation , Protein Precursors/analysis , Protein Precursors/chemistry
13.
J R Soc Interface ; 5(28): 1371-86, 2008 Nov 06.
Article in English | MEDLINE | ID: mdl-18426775

ABSTRACT

The alkyllysophospholipid (ALP) analogues Mitelfosine and Edelfosine are anticancer drugs whose mode of action is still the subject of debate. It is agreed that the primary interaction of these compounds is with cellular membranes. Furthermore, the membrane-associated protein CTP: phosphocholine cytidylyltransferase (CCT) has been proposed as the critical target. We present the evaluation of our hypothesis that ALP analogues disrupt membrane curvature elastic stress and inhibit membrane-associated protein activity (e.g. CCT), ultimately resulting in apoptosis. This hypothesis was tested by evaluating structure-activity relationships of ALPs from the literature. In addition we characterized the lipid typology, cytotoxicity and critical micelle concentration of novel ALP analogues that we synthesized. Overall we find the literature data and our experimental data provide excellent support for the hypothesis, which predicts that the most potent ALP analogues will be type I lipids.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Membrane/drug effects , Choline-Phosphate Cytidylyltransferase/metabolism , Neoplasms/drug therapy , Phospholipid Ethers/pharmacology , Antineoplastic Agents/metabolism , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Elasticity , HL-60 Cells , Humans , Micelles , Molecular Structure , Phospholipid Ethers/metabolism , Stress, Mechanical , Structure-Activity Relationship , Tetrazolium Salts , Thiazoles
14.
Cancer Res ; 67(21): 10368-78, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17974980

ABSTRACT

The endoplasmic reticulum (ER) has been posited as a potential anticancer target. The synthetic antitumor alkyl-lysophospholipid analogue edelfosine accumulates in the ER of solid tumor cells. This ER accumulation of the drug leads to the inhibition of phosphatidylcholine and protein synthesis, G(2)-M arrest, depletion of ER-stored Ca(2+), Bax up-regulation and activation, transcriptional factor growth arrest and DNA damage-inducible gene 153 up-regulation, caspase-4 and caspase-8 activation, and eventually to apoptosis. Edelfosine prompted ER stress apoptotic signaling, but not the survival unfolded protein response. Edelfosine also induced persistent c-Jun NH(2)-terminal kinase (JNK) activation. Gene transfer-mediated overexpression of apoptosis signal-regulating kinase 1, which plays a crucial role in ER stress, enhanced edelfosine-induced JNK activation and apoptosis. Inhibition of JNK, caspase-4, or caspase-8 activation diminished edelfosine-induced apoptosis. Edelfosine treatment led to the generation of the p20 caspase-8 cleavage fragment of BAP31, directing proapoptotic signals between the ER and the mitochondria. bax(-/-)bak(-/-) double-knockout cells fail to undergo edelfosine-induced ER-stored Ca(2+) release and apoptosis. Wild-type and bax(-/-)bak(-/-) cells showed similar patterns of phosphatidylcholine and protein synthesis inhibition, despite their differences in drug sensitivity. Thus, edelfosine-induced apoptosis is dependent on Bax/Bak-mediated ER-stored Ca(2+) release, but phosphatidylcholine and protein synthesis inhibition is not critical. Transfection-enforced expression of Bcl-X(L), which localizes specifically in mitochondria, prevented apoptosis without inhibiting ER-stored Ca(2+) release. These data reveal that edelfosine induces an ER stress response in solid tumor cells, providing novel insights into the edelfosine-mediated antitumor activity. Our data also indicate that mitochondria are indispensable for this edelfosine-induced cell death initiated by ER stress.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Endoplasmic Reticulum/drug effects , Neoplasms/drug therapy , Phospholipid Ethers/pharmacology , Calcium/metabolism , Caspases/physiology , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinase 5/physiology , Neoplasms/metabolism , Neoplasms/pathology , Phosphatidylcholines/biosynthesis , Protein Synthesis Inhibitors/pharmacology , bcl-2 Homologous Antagonist-Killer Protein/physiology , bcl-2-Associated X Protein/physiology , bcl-X Protein/physiology
15.
Regul Pept ; 140(3): 117-24, 2007 May 03.
Article in English | MEDLINE | ID: mdl-17218027

ABSTRACT

We previously reported that vasoactive intestinal peptide (VIP) promoted synthesis of phosphatidylcholine (PC) in alveolar type II (ATII) cells. But the intracellular mechanism for this effect was unknown. In this work, we investigated the intracellular signal transduction pathway for VIP promoted synthesis of PC, the major lipid component of pulmonary surfactant (PS), by using an antagonist of VIP receptors, inhibitor of protein kinase C (PKC) and antisense oligonucleotides (AS-ODN) for c-fos oncogene. Our results showed that: 1 in circle [D-P-Cl-Phe(6)-Leu(17)]-VIP (10(-6) mol/l), an antagonist of VIP receptors, could decrease the quantity of [(3)H] choline incorporation, microsomal choline-phosphate cytidylyltransferase (CCT) mRNA expression and CCT activity induced by VIP (10(-8) mol/l) in cultured lung explants to the control levels; 2 in circle VIP (10(-8) mol/l) upregulated c-Fos protein expression in ATII cells. AS-ODN for c-fos oncogene (9x10(-6) mol/l) could block the elevation of [(3)H] choline incorporation, microsomal CCT mRNA expression and CCT activity induced by VIP in cultured lung explants and in ATII cells; 3 in circle H7 (10(-5) mol/l), a PKC inhibitor could also reduce VIP induced [(3)H] choline incorporation, microsomal CCT mRNA expression and CCT activity in cultured lung explants and in ATII cells. These results demonstrated that VIP receptors, PKC and c-Fos protein played important roles in the signaling pathway through which VIP promoted the synthesis of PC.


Subject(s)
Genes, fos/physiology , Phosphatidylcholines/metabolism , Protein Kinase C/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Surfactants/metabolism , Vasoactive Intestinal Peptide/pharmacology , Animals , Cells, Cultured , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Choline-Phosphate Cytidylyltransferase/metabolism , Enzyme Inhibitors/pharmacology , Female , Genes, fos/drug effects , Immunohistochemistry , Male , Oligonucleotides, Antisense/pharmacology , Organ Culture Techniques , Protein Kinase C/antagonists & inhibitors , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , Rats , Rats, Wistar , Receptors, Vasoactive Intestinal Peptide/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/genetics
16.
Vascul Pharmacol ; 45(1): 65-71, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16750656

ABSTRACT

Given the heterogeneous nature of metabolic dysfunctions associated with insulin resistance and type 2 diabetes (T2D), a single pharmaceutical cannot be expected to provide complication-free therapy in all patients. Thiazolidinediones (TZD) increase insulin sensitivity, reduce blood glucose and improve cardiovascular parameters. However, in addition to increasing fat mass, TZD have the potential in certain individuals to exacerbate underlying hepatosteatosis and diabetic cardiomyopathy. Pharmacogenetics should allow patient selection to maximize therapy and minimize risk. To this end, we have combined two genetically diverse inbred strains, NON/Lt and NZO/Lt, to produce a "negative heterosis" increasing the frequency of T2D in F1 males. As in humans with T2D, treatment of diabetic and hyperlipemic F1 males with rosiglitazone (Rosi), an agonist of peroxisome proliferator-activated gamma receptor (PPARgamma), reverses these disease phenotypes. However, the hybrid genome perturbed both major pathways for phosphatidylcholine (PC) biosynthesis in the liver, and effected remarkable alterations in the composition of cardiolipin in heart mitochondria. These metabolic defects severely exacerbated an underlying hepatosteatosis and increased levels of the adipokine, plasminogen activator inhibitor-1 (PAI-1), a risk factor for cardiovascular events. This model system demonstrates how the power of mouse genetics can be used to identify the metabolic signatures of individuals who may be prone to drug side effects.


Subject(s)
Cardiovascular System/drug effects , Diabetes Mellitus, Type 2/metabolism , Hypoglycemic Agents/adverse effects , Liver/drug effects , Phosphatidylcholines/metabolism , Thiazolidinediones/adverse effects , Animals , Cardiolipins/metabolism , Cardiovascular System/metabolism , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Choline-Phosphate Cytidylyltransferase/metabolism , Crosses, Genetic , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Fatty Liver/chemically induced , Fatty Liver/metabolism , Gene Expression Regulation/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred NOD , Mice, Obese , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , PPAR gamma/agonists , PPAR gamma/metabolism , Phosphatidylethanolamine N-Methyltransferase/antagonists & inhibitors , Phosphatidylethanolamine N-Methyltransferase/metabolism , Rosiglitazone
17.
J Biol Chem ; 281(10): 6718-25, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16380371

ABSTRACT

Phosphatidylcholine (PtdCho) is a major membrane phospholipid, and its loss is sufficient in itself to induce cell death. PtdCho homeostasis is regulated by the balance between hydrolysis and synthesis. PtdCho is hydrolyzed by phospholipase A2 (PLA2), PtdChospecific phospholipase C (PtdCho-PLC), and phospholipase D (PLD). PtdCho synthesis is rate-limited by CTP:phosphocholine cytidylyltransferase (CCT), which makes CDP-choline. The final step of PtdCho synthesis is catalyzed by CDP-choline:1,2-diacylglycerol cholinephosphotransferase. PtdCho synthesis in the brain is predominantly through the CDP-choline pathway. Transient middle cerebral artery occlusion (tMCAO) significantly increased PLA2 activity, secretory PLA2 (sPLA2)-IIA mRNA and protein levels, PtdCho-PLC activity, and PLD2 protein expression following reperfusion. CDP-choline treatment significantly attenuated PLA2 activity, sPLA2-IIA mRNA and protein levels, and PtdCho-PLC activity, but did not affect PLD2 protein expression. tMCAO also resulted in loss of CCT activity and CCTalpha protein, which were partially restored by CDP-choline. No changes were observed in cytosolic PLA2 or calcium-independent PLA2 tMCAO. protein levels after Up-regulation of PLA2, PtdCho-PLC, and PLD and regulation of CCT collectively down-resulted in loss of PtdCho, which was significantly restored by CDP-choline treatment. CDP-choline treatment significantly attenuated the infarction volume by 55 +/- 5% after 1 h of tMCAO and 1 day of reperfusion. Taken together, these results suggest that CDP-choline significantly restores Ptd-Cho levels by differentially affecting sPLA2-IIA, PtdCho-PLC, and CCTalpha after transient focal cerebral ischemia. A hypothetical scheme is proposed integrating results from this study and from other reports in the literature.


Subject(s)
Brain Ischemia/enzymology , Choline-Phosphate Cytidylyltransferase/metabolism , Cytidine Diphosphate Choline/pharmacology , Phosphatidylcholines/metabolism , Phospholipases A/metabolism , Animals , Brain Ischemia/metabolism , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Gene Expression Regulation/physiology , Male , Phospholipase D/biosynthesis , Phospholipase D/genetics , Phospholipases A/genetics , Phospholipases A2 , RNA, Messenger/biosynthesis , Rats , Rats, Inbred Dahl , Rats, Sprague-Dawley
19.
Mol Pharmacol ; 62(5): 1068-75, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12391269

ABSTRACT

Apoptosis induced by antitumor phospholipid analogs takes place after the inhibition of the CTP:phosphocholine cytidylyltransferase (CCT; EC 2.7.7.15) catalyzed step of phosphatidylcholine (PtdCho) biosynthesis. Exposure of cells to synthetic short-chain ceramide analogs also triggers apoptosis concomitant with decreased PtdCho biosynthesis, and the present study was undertaken to ascertain whether C(2)-ceramide inhibition of PtdCho synthesis is direct or secondary to other ceramide-mediated cellular responses. The exposure of COS-7 cells to either C(2)-ceramide, ET-18-OCH(3), or farnesol resulted in time- and dose-dependent apoptotic cell death. Cells treated with C(2)-ceramide or ET-18-OCH(3) selectively and immediately accumulated phosphocholine, whereas CDP-choline increased with farnesol treatment. In vitro assays of CCT activity demonstrated that C(2)-ceramide directly inhibited CCT. Comparison of different N-linked sphingosine derivatives suggests an inverse relationship between the length of the N-linked carbon chain and the derivatives ability to trigger apoptosis and inhibit CCT. Taken together, our results suggest CCT as a primary target for C(2)-ceramide inhibition that accounts for its cytotoxic effects.


Subject(s)
Apoptosis , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Animals , COS Cells , Haplorhini , Microscopy, Electron
20.
FASEB J ; 16(13): 1814-6, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12223447

ABSTRACT

Glucosylceramide (GlcCer) accumulates in the inherited metabolic disorder, Gaucher disease, because of the defective activity of lysosomal glucocerebrosidase. We previously demonstrated that upon GlcCer accumulation, cultured hippocampal neurons exhibit modified growth patterns, altered endoplasmic reticulum density, and altered calcium release from intracellular stores. We here examined the relationship between GlcCer accumulation and phospholipid synthesis. After treatment of neurons with an active site-directed inhibitor of glucocerebrosidase, or in neurons obtained from a mouse model of Gaucher disease, [14C]methyl choline incorporation into [14C]phosphatidylcholine ([14C]PC) and [14C]sphingomyelin was elevated, as were [14C]CDP-choline levels, suggesting that CTP:phosphocholine cytidylyltransferase (CCT) is activated. Indeed, CCT activity was elevated in neurons that had accumulated GlcCer. GlcCer, but not galactosylceramide (GalCer), stimulated CCT activity in rat brain homogenates, and significantly higher levels of CCT were membrane associated in cortical homogenates from a mouse model of Gaucher disease compared with wild-type mice. Because CCT mRNA and protein levels were unaltered in either neurons or brain tissue that had accumulated GlcCer, it appeared likely that GlcCer activates CCT by a post-translational mechanism. This was verified by examination of the effect of GlcCer on CCT purified about 1200-fold from rat brain. GlcCer stimulated CCT activity, with stimulation observed at levels as low as 2.5 mol% and with maximal activation reached at 10 mol%. In contrast, GalCer had no effect. Together, these data demonstrate that GlcCer directly activates CCT, which results in elevated PC synthesis, which may account for some of the changes in growth rates observed upon neuronal GlcCer accumulation.


Subject(s)
Choline-Phosphate Cytidylyltransferase/metabolism , Choline/analogs & derivatives , Gaucher Disease/metabolism , Glucosylceramides/metabolism , Inositol/analogs & derivatives , Neurons/metabolism , Phosphatidylcholines/biosynthesis , Animals , Axons/metabolism , Brain/drug effects , Brain/enzymology , Carbon Radioisotopes , Cell Division , Choline/metabolism , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Galactosylceramides/pharmacology , Gaucher Disease/genetics , Gaucher Disease/pathology , Genotype , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Glucosylceramides/pharmacology , Inositol/pharmacology , Lipid Metabolism , Mice , Mutation , Rats
SELECTION OF CITATIONS
SEARCH DETAIL