Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-36690320

ABSTRACT

Alkylglycerol monooxygenase (AGMO) and plasmanylethanolamine desaturase (PEDS1) are enzymes involved in ether lipid metabolism. While AGMO degrades plasmanyl lipids by oxidative cleavage of the ether bond, PEDS1 exclusively synthesizes a specific subclass of ether lipids, the plasmalogens, by introducing a vinyl ether double bond into plasmanylethanolamine phospholipids. Ether lipids are characterized by an ether linkage at the sn-1 position of the glycerol backbone and they are found in membranes of different cell types. Decreased plasmalogen levels have been associated with neurological diseases like Alzheimer's disease. Agmo-deficient mice do not present an obvious phenotype under unchallenged conditions. In contrast, Peds1 knockout mice display a growth phenotype. To investigate the molecular consequences of Agmo and Peds1 deficiency on the mouse lipidome, five tissues from each mouse model were isolated and subjected to high resolution mass spectrometry allowing the characterization of up to 2013 lipid species from 42 lipid subclasses. Agmo knockout mice moderately accumulated plasmanyl and plasmenyl lipid species. Peds1-deficient mice manifested striking changes characterized by a strong reduction of plasmenyl lipids and a concomitant massive accumulation of plasmanyl lipids resulting in increased total ether lipid levels in the analyzed tissues except for the class of phosphatidylethanolamines where total levels remained remarkably constant also in Peds1 knockout mice. The rate-limiting enzyme in ether lipid metabolism, FAR1, was not upregulated in Peds1-deficient mice, indicating that the selective loss of plasmalogens is not sufficient to activate the feedback mechanism observed in total ether lipid deficiency.


Subject(s)
Lipid Metabolism , Plasmalogens , Animals , Mice , Plasmalogens/metabolism , Lipidomics , Ethers , Mice, Knockout
3.
Cell Mol Life Sci ; 79(11): 562, 2022 Oct 22.
Article in English | MEDLINE | ID: mdl-36271951

ABSTRACT

Multifunctional proteins are challenging as it can be difficult to confirm pathomechanisms associated with disease-causing genetic variants. The human 17ß-hydroxysteroid dehydrogenase 10 (HSD10) is a moonlighting enzyme with at least two structurally and catalytically unrelated functions. HSD10 disease was originally described as a disorder of isoleucine metabolism, but the clinical manifestations were subsequently shown to be linked to impaired mtDNA transcript processing due to deficient function of HSD10 in the mtRNase P complex. A surprisingly large number of other, mostly enzymatic and potentially clinically relevant functions have been attributed to HSD10. Recently, HSD10 was reported to exhibit phospholipase C-like activity towards cardiolipins (CL), important mitochondrial phospholipids. To assess the physiological role of the proposed CL-cleaving function, we studied CL architectures in living cells and patient fibroblasts in different genetic backgrounds and lipid environments using our well-established LC-MS/MS cardiolipidomic pipeline. These experiments revealed no measurable effect on CLs, indicating that HSD10 does not have a physiologically relevant function towards CL metabolism. Evolutionary constraints could explain the broad range of reported substrates for HSD10 in vitro. The combination of an essential structural with a non-essential enzymatic function in the same protein could direct the evolutionary trajectory towards improvement of the former, thereby increasing the flexibility of the binding pocket, which is consistent with the results presented here.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases , Hydroxysteroid Dehydrogenases , Humans , 3-Hydroxyacyl CoA Dehydrogenases/genetics , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Cardiolipins , Chromatography, Liquid , Tandem Mass Spectrometry , DNA, Mitochondrial , Type C Phospholipases
4.
J Lipid Res ; 63(6): 100222, 2022 06.
Article in English | MEDLINE | ID: mdl-35537527

ABSTRACT

Little is known about the physiological role of alkylglycerol monooxygenase (AGMO), the only enzyme capable of cleaving the 1-O-alkyl ether bond of ether lipids. Expression and enzymatic activity of this enzyme can be detected in a variety of tissues including adipose tissue. This labile lipolytic membrane-bound protein uses tetrahydrobiopterin as a cofactor, and mice with reduced tetrahydrobiopterin levels have alterations in body fat distribution and blood lipid concentrations. In addition, manipulation of AGMO in macrophages led to significant changes in the cellular lipidome, and alkylglycerolipids, the preferred substrates of AGMO, were shown to accumulate in mature adipocytes. Here, we investigated the roles of AGMO in lipid metabolism by studying 3T3-L1 adipogenesis. AGMO activity was induced over 11 days using an adipocyte differentiation protocol. We show that RNA interference-mediated knockdown of AGMO did not interfere with adipocyte differentiation or affect lipid droplet formation. Furthermore, lipidomics revealed that plasmalogen phospholipids were preferentially accumulated upon Agmo knockdown, and a significant shift toward longer and more polyunsaturated acyl side chains of diacylglycerols and triacylglycerols could be detected by mass spectrometry. Our results indicate that alkylglycerol catabolism has an influence not only on ether-linked species but also on the degree of unsaturation in the massive amounts of triacylglycerols formed during in vitro 3T3-L1 adipocyte differentiation.


Subject(s)
Ether , Lipidomics , 3T3-L1 Cells , Adipocytes/metabolism , Adipogenesis , Animals , Cell Differentiation , Ether/metabolism , Ethers , Lipid Metabolism/genetics , Mice , Phospholipids/metabolism , Triglycerides/metabolism
5.
Cell Mol Life Sci ; 79(4): 214, 2022 Mar 28.
Article in English | MEDLINE | ID: mdl-35347434

ABSTRACT

Plasmalogens are an abundant class of glycerophospholipids in the mammalian body, with special occurrence in the brain and in immune cell membranes. Plasmanylethanolamine desaturase (PEDS1) is the final enzyme of plasmalogen biosynthesis, which introduces the characteristic 1-O-alk-1'-enyl double bond. The recent sequence identification of PEDS1 as transmembrane protein 189 showed that its protein sequence is related to a special class of plant desaturases (FAD4), with whom it shares a motif of 8 conserved histidines, which are essential for the enzymatic activity. In the present work, we wanted to gain more insight into the sequence-function relationship of this enzyme and mutated to alanine additional 28 amino acid residues of murine plasmanylethanolamine desaturase including those 20 residues, which are also totally conserved-in addition to the eight-histidine-motif-among the animal PEDS1 and plant FAD4 plant desaturases. We measured the enzymatic activity by transient transfection of tagged murine PEDS1 expression clones to a PEDS1-deficient human HAP1 cell line by monitoring of labeled plasmalogens formed from supplemented 1-O-pyrenedecyl-sn-glycerol in relation to recombinant protein expression. Surprisingly, only a single mutation, namely aspartate 100, led to a total loss of PEDS1 activity. The second strongest impact on enzymatic activity had mutation of phenylalanine 118, leaving only 6% residual activity. A structural model obtained by homology modelling to available structures of stearoyl-CoA reductase predicted that this aspartate 100 residue interacts with histidine 96, and phenylalanine 118 interacts with histidine 187, both being essential histidines assumed to be involved in the coordination of the di-metal center of the enzyme.


Subject(s)
Aspartic Acid , Oxidoreductases , Amino Acid Sequence , Animals , Humans , Mammals/metabolism , Mice , Oxidoreductases/metabolism , Plants/metabolism
6.
Cell Biosci ; 11(1): 54, 2021 Mar 16.
Article in English | MEDLINE | ID: mdl-33726865

ABSTRACT

BACKGROUND: Genome editing in mice using either classical approaches like homologous recombination or CRISPR/Cas9 has been reported to harbor off target effects (insertion/deletion, frame shifts or gene segment duplications) that lead to mutations not only in close proximity to the target site but also outside. Only the genomes of few engineered mouse strains have been sequenced. Since the role of the ether-lipid cleaving enzyme alkylglycerol monooxygenase (AGMO) in physiology and pathophysiology remains enigmatic, we created a knockout mouse model for AGMO using EUCOMM stem cells but unforeseen genotyping issues that did not agree with Mendelian distribution and enzyme activity data prompted an in-depth genomic validation of the mouse model. RESULTS: We report a gene segment tandem duplication event that occurred during the generation of an Agmo knockout-first allele by homologous recombination. Only low homology was seen between the breakpoints. While a single copy of the recombinant 18 kb cassette was integrated correctly around exon 2 of the Agmo gene, whole genome nanopore sequencing revealed a 94 kb duplication in the Agmo locus that contains Agmo wild-type exons 1-3. The duplication fooled genotyping by routine PCR, but could be resolved using qPCR-based genotyping, targeted locus amplification sequencing and nanopore sequencing. Despite this event, this Agmo knockout mouse model lacks AGMO enzyme activity and can therefore be used to study its physiological role. CONCLUSIONS: A duplication event occurred at the exact locus of the homologous recombination and was not detected by conventional quality control filters such as FISH or long-range PCR over the recombination sites. Nanopore sequencing provides a cost convenient method to detect such underrated off-target effects, suggesting its use for additional quality assessment of gene editing in mice and also other model organisms.

7.
Life (Basel) ; 11(2)2021 Jan 26.
Article in English | MEDLINE | ID: mdl-33530536

ABSTRACT

The gene encoding alkylglycerol monooxygenase (AGMO) was assigned 10 years ago. So far, AGMO is the only known enzyme capable of catalysing the breakdown of alkylglycerols and lyso-alkylglycerophospholipids. With the knowledge of the genetic information, it was possible to relate a potential contribution for mutations in the AGMO locus to human diseases by genome-wide association studies. A possible role for AGMO was implicated by genetic analyses in a variety of human pathologies such as type 2 diabetes, neurodevelopmental disorders, cancer, and immune defence. Deficient catabolism of stored lipids carrying an alkyl bond by an absence of AGMO was shown to impact on the overall lipid composition also outside the ether lipid pool. This review focuses on the current evidence of AGMO in human diseases and summarises experimental evidence for its role in immunity, energy homeostasis, and development in humans and several model organisms. With the progress in lipidomics platform and genetic identification of enzymes involved in ether lipid metabolism such as AGMO, it is now possible to study the consequence of gene ablation on the global lipid pool and further on certain signalling cascades in a variety of model organisms in more detail.

8.
Anal Chem ; 92(16): 11268-11276, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32692545

ABSTRACT

Deficient ether lipid biosynthesis in rhizomelic chondrodysplasia punctata and other disorders is associated with a wide range of severe symptoms including small stature with proximal shortening of the limbs, contractures, facial dysmorphism, congenital cataracts, ichthyosis, spasticity, microcephaly, and mental disability. Mouse models are available but show less severe symptoms. In both humans and mice, it has remained elusive which of the symptoms can be attributed to lack of plasmanyl or plasmenyl ether lipids. The latter compounds, better known as plasmalogens, harbor a vinyl ether double bond conferring special chemical and physical properties. Discrimination between plasmanyl and plasmenyl ether lipids is a major analytical challenge, especially in complex lipid extracts with many isobaric species. Consequently, these lipids are often neglected also in recent lipidomic studies. Here, we present a comprehensive LC-MS/MS based approach that allows unequivocal distinction of these two lipid subclasses based on their chromatographic properties. The method was validated using a novel plasmalogen-deficient mouse model, which lacks plasmanylethanolamine desaturase and therefore cannot form plasmenyl ether lipids. We demonstrate that plasmanylethanolamine desaturase deficiency causes an accumulation of plasmanyl species, a too little studied but biologically important substance class.


Subject(s)
Ethers/analysis , Lipidomics/methods , Plasmalogens/analysis , Animals , Chromatography, Liquid , Ethers/chemistry , Female , Male , Mice, Knockout , Molecular Structure , Oxidoreductases/genetics , Plasmalogens/chemistry , Tandem Mass Spectrometry
9.
Proc Natl Acad Sci U S A ; 117(14): 7792-7798, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32209662

ABSTRACT

A significant fraction of the glycerophospholipids in the human body is composed of plasmalogens, particularly in the brain, cardiac, and immune cell membranes. A decline in these lipids has been observed in such diseases as Alzheimer's and chronic obstructive pulmonary disease. Plasmalogens contain a characteristic 1-O-alk-1'-enyl ether (vinyl ether) double bond that confers special biophysical, biochemical, and chemical properties to these lipids. However, the genetics of their biosynthesis is not fully understood, since no gene has been identified that encodes plasmanylethanolamine desaturase (E.C. 1.14.99.19), the enzyme introducing the crucial alk-1'-enyl ether double bond. The present work identifies this gene as transmembrane protein 189 (TMEM189). Inactivation of the TMEM189 gene in human HAP1 cells led to a total loss of plasmanylethanolamine desaturase activity, strongly decreased plasmalogen levels, and accumulation of plasmanylethanolamine substrates and resulted in an inability of these cells to form labeled plasmalogens from labeled alkylglycerols. Transient expression of TMEM189 protein, but not of other selected desaturases, recovered this deficit. TMEM189 proteins contain a conserved protein motif (pfam10520) with eight conserved histidines that is shared by an alternative type of plant desaturase but not by other mammalian proteins. Each of these histidines is essential for plasmanylethanolamine desaturase activity. Mice homozygous for an inactivated Tmem189 gene lacked plasmanylethanolamine desaturase activity and had dramatically lowered plasmalogen levels in their tissues. These results assign the TMEM189 gene to plasmanylethanolamine desaturase and suggest that the previously characterized phenotype of Tmem189-deficient mice may be caused by a lack of plasmalogens.


Subject(s)
Lipids/genetics , Oxidoreductases/genetics , Plasmalogens/genetics , Ubiquitin-Conjugating Enzymes/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Cell Line , Humans , Mice , Oxidation-Reduction , Oxidoreductases/metabolism , Phenotype , Plasmalogens/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Vinyl Compounds/metabolism
10.
Cell Rep ; 30(12): 4281-4291.e4, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32209484

ABSTRACT

Cardiolipin (CL) is a phospholipid specific for mitochondrial membranes and crucial for many core tasks of this organelle. Its acyl chain configurations are tissue specific, functionally important, and generated via post-biosynthetic remodeling. However, this process lacks the necessary specificity to explain CL diversity, which is especially evident for highly specific CL compositions in mammalian tissues. To investigate the so far elusive regulatory origin of CL homeostasis in mice, we combine lipidomics, integrative transcriptomics, and data-driven machine learning. We demonstrate that not transcriptional regulation, but cellular phospholipid compositions are closely linked to the tissue specificity of CL patterns allowing artificial neural networks to precisely predict cross-tissue CL compositions in a consistent mechanistic specificity rationale. This is especially relevant for the interpretation of disease-related perturbations of CL homeostasis, by allowing differentiation between specific aberrations in CL metabolism and changes caused by global alterations in cellular (phospho-)lipid metabolism.


Subject(s)
Cardiolipins/metabolism , Mitochondria/metabolism , Organ Specificity , Phospholipids/metabolism , Animals , Fatty Acids/metabolism , Mice, Inbred C57BL , Neural Networks, Computer , Transcription, Genetic
11.
Proc Natl Acad Sci U S A ; 115(16): 4158-4163, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29618609

ABSTRACT

Current strategies used to quantitatively describe the biological diversity of lipids by mass spectrometry are often limited in assessing the exact structural variability of individual molecular species in detail. A major challenge is represented by the extensive isobaric overlap present among lipids, hampering their accurate identification. This is especially true for cardiolipins, a mitochondria-specific class of phospholipids, which are functionally involved in many cellular functions, including energy metabolism, cristae structure, and apoptosis. Substituted with four fatty acyl side chains, cardiolipins offer a particularly high potential to achieve complex mixtures of molecular species. Here, we demonstrate how systematically generated high-performance liquid chromatography-mass spectral data can be utilized in a mathematical structural modeling approach, to comprehensively analyze and characterize the molecular diversity of mitochondrial cardiolipin compositions in cell culture and disease models, cardiolipin modulation experiments, and a broad variety of frequently studied model organisms.


Subject(s)
Cardiolipins/chemistry , Membrane Lipids/chemistry , Mitochondrial Membranes/chemistry , Animals , Bacteria/chemistry , Barth Syndrome/metabolism , Cardiolipins/isolation & purification , Cell Line , Chromatography, High Pressure Liquid , Fatty Acids/analysis , Fibroblasts/chemistry , Fungi/chemistry , Humans , Membrane Lipids/isolation & purification , Mice , Models, Molecular , Molecular Structure , Plants/chemistry , RAW 264.7 Cells , Tandem Mass Spectrometry , Vertebrates/metabolism
12.
J Lipid Res ; 59(5): 901-909, 2018 05.
Article in English | MEDLINE | ID: mdl-29540573

ABSTRACT

Plasmanylethanolamine desaturase (PEDS) (EC 1.14.99.19) introduces the 1-prime double bond into plasmalogens, one of the most abundant phospholipids in the human body. This labile membrane enzyme has not been purified and its coding sequence is unknown. Previous assays for this enzyme used radiolabeled substrates followed by multistep processing. We describe here a straight-forward method for the quantification of PEDS in enzyme incubation mixtures using pyrene-labeled substrates and reversed-phase HPLC with fluorescence detection. After stopping the reaction with hydrochloric acid in acetonitrile, the mixture was directly injected into the HPLC system without the need of lipid extraction. The substrate, 1-O-pyrenedecyl-2-acyl-sn-glycero-3-phosphoethanolamine, and the lyso-substrate, 1-O-pyrenedecyl-sn-glycero-3-phosphoethanolamine, were prepared from RAW-12 cells deficient in PEDS activity and were compared for their performance in the assay. Plasmalogen levels in mouse tissues and in cultured cells did not correlate with PEDS levels, indicating that the desaturase might not be the rate limiting step for plasmalogen biosynthesis. Among selected mouse organs, the highest activities were found in kidney and in spleen. Incubation of intact cultivated mammalian cells with 1-O-pyrenedecyl-sn-glycerol, extraction of lipids, and treatment with hydrochloric or acetic acid in acetonitrile allowed sensitive monitoring of PEDS activity in intact cells.


Subject(s)
Chromatography, High Pressure Liquid , Oxidoreductases/analysis , Plasmalogens/chemistry , Pyrenes/chemistry , Spectrometry, Fluorescence , Vinyl Compounds/chemistry , Animals , Cells, Cultured , Mice , Molecular Structure , Oxidoreductases/deficiency , Oxidoreductases/metabolism , Plasmalogens/biosynthesis , Pyrenes/metabolism , Substrate Specificity , Vinyl Compounds/metabolism
13.
Proc Natl Acad Sci U S A ; 112(8): 2431-6, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25675482

ABSTRACT

Tetrahydrobiopterin is a cofactor synthesized from GTP with well-known roles in enzymatic nitric oxide synthesis and aromatic amino acid hydroxylation. It is used to treat mild forms of phenylketonuria. Less is known about the role of tetrahydrobiopterin in lipid metabolism, although it is essential for irreversible ether lipid cleavage by alkylglycerol monooxygenase. Here we found intracellular alkylglycerol monooxygenase activity to be an important regulator of alkylglycerol metabolism in intact murine RAW264.7 macrophage-like cells. Alkylglycerol monooxygenase was expressed and active also in primary mouse bone marrow-derived monocytes and "alternatively activated" M2 macrophages obtained by interleukin 4 treatment, but almost missing in M1 macrophages obtained by IFN-γ and lipopolysaccharide treatment. The cellular lipidome of RAW264.7 was markedly changed in a parallel way by modulation of alkylglycerol monooxygenase expression and of tetrahydrobiopterin biosynthesis affecting not only various ether lipid species upstream of alkylglycerol monooxygenase but also other more complex lipids including glycosylated ceramides and cardiolipins, which have no direct connection to ether lipid pathways. Alkylglycerol monooxygenase activity manipulation modulated the IFN-γ/lipopolysaccharide-induced expression of inducible nitric oxide synthase, interleukin-1ß, and interleukin 1 receptor antagonist but not transforming growth factor ß1, suggesting that alkylglycerol monooxygenase activity affects IFN-γ/lipopolysaccharide signaling. Our results demonstrate a central role of tetrahydrobiopterin and alkylglycerol monooxygenase in ether lipid metabolism of murine macrophages and reveal that alteration of alkylglycerol monooxygenase activity has a profound impact on the lipidome also beyond the class of ether lipids.


Subject(s)
Biopterins/analogs & derivatives , Lipid Metabolism/drug effects , Macrophages/metabolism , Mixed Function Oxygenases/metabolism , Animals , Biopterins/pharmacology , Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Cell Line , Cells, Cultured , Cluster Analysis , GTP Cyclohydrolase/metabolism , Gene Knockdown Techniques , Interferon-gamma/pharmacology , Lentivirus/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/enzymology , Mice , Monocytes/cytology , Monocytes/drug effects , Monocytes/enzymology , Nitric Oxide Synthase Type II/metabolism
14.
Eur J Clin Invest ; 44(4): 395-401, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24467215

ABSTRACT

BACKGROUND: Cholesteryl ester transfer protein (CETP) has been considered as a possible target for treatment of cardiovascular disease. However, first clinical studies employing CETP inhibitors have failed to demonstrate clinical benefit. Additionally, we have previously shown that low endogenous plasma levels of CETP are associated with increased mortality in coronary artery disease (CAD) patients. We hypothesized that low CETP plasma levels are associated with decreased high-density lipoprotein (HDL) function. MATERIALS AND METHODS: Serum HDL efflux capacity was measured in 154 patients of the Ludwigshafen Risk and Cardiovascular Health (LURIC) study displaying extremely low (< 0·68 µg/mL, n = 77) or high (> 2·13 µg/mL, n = 77) CETP concentrations in their plasma, respectively. The LURIC study is a prospective observational study of patients referred to coronary angiography at baseline with a median follow-up time of 7·75 years. Primary and secondary endpoints were cardiovascular and all-cause mortality, respectively. RESULTS: High CETP patients showed a significant increase in the capacity of their plasma to mediate cholesterol efflux from cholesterol laden macrophages when compared to the efflux capacity observed in low CETP patients (+ 5·4%, P = 0·015). As shown by multiregression analysis, the impact of CETP on cholesterol efflux capacity was independent from classical risk and lifestyle factors, as well as from lipid parameters including HDL cholesterol, LDL cholesterol and triglycerides. CONCLUSIONS: Our findings indicate that low plasma concentrations of CETP might indeed lead to impaired HDL function within the reverse cholesterol transport pointing towards an atheroprotective role of CETP at least in patients with high risk of CAD.


Subject(s)
Cholesterol Ester Transfer Proteins/deficiency , Cholesterol/metabolism , Coronary Artery Disease/blood , Aged , Cells, Cultured , Humans , Life Style , Macrophages/metabolism , Male , Middle Aged , Prospective Studies , Risk Factors , Smoking/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...