1
|
Hirabayashi T, Kawaguchi M, Harada S, Mouri M, Takamiya R, Miki Y, Sato H, Taketomi Y, Yokoyama K, Kobayashi T, Tokuoka SM, Kita Y, Yoda E, Hara S, Mikami K, Nishito Y, Kikuchi N, Nakata R, Kaneko M, Kiyonari H, Kasahara K, Aiba T, Ikeda K, Soga T, Kurano M, Yatomi Y, Murakami M. Hepatic phosphatidylcholine catabolism driven by PNPLA7 and PNPLA8 supplies endogenous choline to replenish the methionine cycle with methyl groups. Cell Rep 2023; 42:111940. [PMID: 36719796 DOI: 10.1016/j.celrep.2022.111940] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/10/2022] [Accepted: 12/19/2022] [Indexed: 01/31/2023] Open
Abstract
Choline supplies methyl groups for regeneration of methionine and the methyl donor S-adenosylmethionine in the liver. Here, we report that the catabolism of membrane phosphatidylcholine (PC) into water-soluble glycerophosphocholine (GPC) by the phospholipase/lysophospholipase PNPLA8-PNPLA7 axis enables endogenous choline stored in hepatic PC to be utilized in methyl metabolism. PNPLA7-deficient mice show marked decreases in hepatic GPC, choline, and several metabolites related to the methionine cycle, accompanied by various signs of methionine insufficiency, including growth retardation, hypoglycemia, hypolipidemia, increased energy consumption, reduced adiposity, increased fibroblast growth factor 21 (FGF21), and an altered histone/DNA methylation landscape. Moreover, PNPLA8-deficient mice recapitulate most of these phenotypes. In contrast to wild-type mice fed a methionine/choline-deficient diet, both knockout strains display decreased hepatic triglyceride, likely via reductions of lipogenesis and GPC-derived glycerol flux. Collectively, our findings highlight the biological importance of phospholipid catabolism driven by PNPLA8/PNPLA7 in methyl group flux and triglyceride synthesis in the liver.
Collapse
Affiliation(s)
- Tetsuya Hirabayashi
- Laboratory of Biomembrane, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; Lipid Metabolism Project, Department of Advanced Science for Biomolecules, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan.
| | - Mai Kawaguchi
- Laboratory of Biomembrane, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; Laboratory of Microenvironmental Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Sayaka Harada
- Laboratory of Microenvironmental Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Misa Mouri
- Lipid Metabolism Project, Department of Advanced Science for Biomolecules, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; Department of Biology, Faculty of Science, Ochanomizu University, Tokyo 112-8610, Japan
| | - Rina Takamiya
- Laboratory of Microenvironmental Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yoshimi Miki
- Lipid Metabolism Project, Department of Advanced Science for Biomolecules, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; Laboratory of Microenvironmental Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Hiroyasu Sato
- Lipid Metabolism Project, Department of Advanced Science for Biomolecules, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; Laboratory of Microenvironmental Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yoshitaka Taketomi
- Lipid Metabolism Project, Department of Advanced Science for Biomolecules, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; Laboratory of Microenvironmental Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Kohei Yokoyama
- Laboratory of Biomembrane, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; Lipid Metabolism Project, Department of Advanced Science for Biomolecules, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Tetsuyuki Kobayashi
- Department of Biology, Faculty of Science, Ochanomizu University, Tokyo 112-8610, Japan
| | - Suzumi M Tokuoka
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yoshihiro Kita
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; Life Sciences Core Facility, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Emiko Yoda
- Division of Health Chemistry, Department of Healthcare and Regulatory Sciences, School of Pharmacy, Showa University, Tokyo 142-8555, Japan
| | - Shuntaro Hara
- Division of Health Chemistry, Department of Healthcare and Regulatory Sciences, School of Pharmacy, Showa University, Tokyo 142-8555, Japan
| | - Kyohei Mikami
- Center for Basic Technology Research, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Yasumasa Nishito
- Center for Basic Technology Research, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Norihito Kikuchi
- Laboratory of Biomembrane, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Rieko Nakata
- Department of Food Science and Nutrition, Nara Women's University, Nara, 630-8506, Japan
| | - Mari Kaneko
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
| | - Kohji Kasahara
- Laboratory of Biomembrane, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Toshiki Aiba
- Laboratory of Microenvironmental Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Kazutaka Ikeda
- Kazusa DNA Research Institute, Kisarazu, Chiba 292-0818, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Makoto Murakami
- Lipid Metabolism Project, Department of Advanced Science for Biomolecules, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; Laboratory of Microenvironmental Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan.
| |
Collapse
|
2
|
Sailer S, Lackner K, Pras-Raves ML, Wever EJ, van Klinken JB, Dane AD, Geley S, Koch J, Golderer G, Werner-Felmayer G, Keller MA, Zwerschke W, Vaz FM, Werner ER, Watschinger K. Adaptations of the 3T3-L1 adipocyte lipidome to defective ether lipid catabolism upon alkylglycerol monooxygenase knockdown. J Lipid Res 2022; 63:100222. [PMID: 35537527 PMCID: PMC9192799 DOI: 10.1016/j.jlr.2022.100222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 10/24/2022] Open
|
3
|
Smith T, Knudsen KJ, Ritchie SA. Pharmacokinetics, Mass Balance, Excretion, and Tissue Distribution of Plasmalogen Precursor PPI-1011. Front Cell Dev Biol 2022; 10:867138. [PMID: 35547803 PMCID: PMC9081329 DOI: 10.3389/fcell.2022.867138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/03/2022] [Indexed: 11/19/2022] Open
Abstract
PPI-1011 is a synthetic plasmalogen precursor in development as a treatment for multiple plasmalogen-deficiency disorders. Previous work has demonstrated the ability of PPI-1011 to augment plasmalogens and its effects in vitro and in vivo, however, the precise uptake and distribution across tissues in vivo has not been investigated. The purpose of this study was to evaluate the pharmacokinetics, mass balance, and excretion of [14C]PPI-1011 following a single oral administration at 100 mg/kg in Sprague-Dawley rats. Further tissue distribution was examined using quantitative whole-body autoradiography after both single and repeat daily doses at 100 mg/kg/day. Non-compartmental analysis showed that following a single dose, PPI-1011 exhibited peak levels between 6 and 12 h but also a long half-life with mean t1/2 of 40 h. Mass balance showed that over 50% of the compound-associated radioactivity was absorbed by the body, while approximately 40% was excreted in the feces, 2.5% in the urine, and 10% in expired air within the first 24 h. Quantitative whole-body autoradiography following a single dose showed uptake to nearly all tissues, with the greatest initial uptake in the intestines, liver, and adipose tissue, which decreased time-dependently throughout 168 h post-dose. Following 15 consecutive daily doses, uptake was significantly higher across the entire body at 24 h compared to single dose and remained high out to 96 h where 75% of the initially-absorbed compound-associated radioactivity was still present. The adipose tissue remained particularly high, suggesting a possible reserve of either plasmalogens or alkyl diacylglycerols that the body can pull from for plasmalogen biosynthesis. Uptake to the brain was also definitively confirmed, proving PPI-1011’s ability to cross the blood-brain barrier. In conclusion, our results suggest that oral administration of PPI-1011 results in high uptake across the body, and that repeated dosing over time represents a viable therapeutic strategy for treating plasmalogen deficiencies.
Collapse
|
4
|
Villaume WA. Marginal BH4 Deficiencies, iNOS, and Self-perpetuating Oxidative Stress in Post-acute Sequelae of Covid-19. Med Hypotheses 2022; 163:110842. [PMID: 35431403 PMCID: PMC9006446 DOI: 10.1016/j.mehy.2022.110842] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 03/01/2022] [Accepted: 03/04/2022] [Indexed: 11/30/2022]
Abstract
The treatment of post-acute sequelae of Covid-19 (PASC) has been informed primarily by symptomatic parallels with other chronic inflammatory syndromes. This manuscript takes a more systemic approach by examining how a marginal deficiency of tetrahydrobiopterin (BH4) resulting from mutations of the GCH1 (GTP cyclohydrolase 1) gene may result in the uncoupling of inducible Nitric Oxide Synthase (iNOS) early in the initial response of the innate immune system to SARS-CoV-2. The resulting production of superoxide instead of nitric oxide leads to a self-perpetuating cycle of oxidative stress with the potential to impair numerous metabolic processes and damage multiple organ systems. This marginal deficiency of BH4 may be exhibited by 30% or more of the patient population that have heterozygous or homozygous mutations of GCH1. As the cycle of oxidative stress continues, there is less BH4 available for other metabolic needs such as 1) resisting increased ferroptosis with its damage to organs, and 2) regulating the deactivation of the hyperinflammatory state. Finally, possible steps are proposed for clinical treatment of the hypothesized oxidative stress involved with PASC.
Collapse
|
5
|
AGMO Inhibitor Reduces 3T3-L1 Adipogenesis. Cells 2021; 10:cells10051081. [PMID: 34062826 PMCID: PMC8147360 DOI: 10.3390/cells10051081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 12/20/2022] Open
Abstract
Alkylglycerol monooxygenase (AGMO) is a tetrahydrobiopterin (BH4)-dependent enzyme with major expression in the liver and white adipose tissue that cleaves alkyl ether glycerolipids. The present study describes the disclosure and biological characterization of a candidate compound (Cp6), which inhibits AGMO with an IC50 of 30–100 µM and 5–20-fold preference of AGMO relative to other BH4-dependent enzymes, i.e., phenylalanine-hydroxylase and nitric oxide synthase. The viability and metabolic activity of mouse 3T3-L1 fibroblasts, HepG2 human hepatocytes and mouse RAW264.7 macrophages were not affected up to 10-fold of the IC50. However, Cp6 reversibly inhibited the differentiation of 3T3-L1 cells towards adipocytes, in which AGMO expression was upregulated upon differentiation. Cp6 reduced the accumulation of lipid droplets in adipocytes upon differentiation and in HepG2 cells exposed to free fatty acids. Cp6 also inhibited IL-4-driven differentiation of RAW264.7 macrophages towards M2-like macrophages, which serve as adipocyte progenitors in adipose tissue. Collectively, the data suggest that pharmacologic AGMO inhibition may affect lipid storage.
Collapse
|
6
|
Sailer S, Coassin S, Lackner K, Fischer C, McNeill E, Streiter G, Kremser C, Maglione M, Green CM, Moralli D, Moschen AR, Keller MA, Golderer G, Werner-Felmayer G, Tegeder I, Channon KM, Davies B, Werner ER, Watschinger K. When the genome bluffs: a tandem duplication event during generation of a novel Agmo knockout mouse model fools routine genotyping. Cell Biosci 2021; 11:54. [PMID: 33726865 PMCID: PMC7962373 DOI: 10.1186/s13578-021-00566-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
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.
Collapse
Affiliation(s)
- Sabrina Sailer
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Coassin
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Katharina Lackner
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Caroline Fischer
- Institute of Clinical Pharmacology of the Medical Faculty, Goethe-University, Frankfurt (Main), Germany
| | - Eileen McNeill
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.,Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Gertraud Streiter
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Kremser
- Department of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Manuel Maglione
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Catherine M Green
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Daniela Moralli
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Alexander R Moschen
- Department of Internal Medicine I, Gastroenterology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus A Keller
- Institute of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Georg Golderer
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Gabriele Werner-Felmayer
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology of the Medical Faculty, Goethe-University, Frankfurt (Main), Germany
| | - Keith M Channon
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.,Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Benjamin Davies
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Ernst R Werner
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Katrin Watschinger
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria. .,Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria.
| |
Collapse
|
7
|
Sailer S, Keller MA, Werner ER, Watschinger K. The Emerging Physiological Role of AGMO 10 Years after Its Gene Identification. Life (Basel) 2021; 11:life11020088. [PMID: 33530536 PMCID: PMC7911779 DOI: 10.3390/life11020088] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/21/2021] [Accepted: 01/21/2021] [Indexed: 02/07/2023] Open
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.
Collapse
Affiliation(s)
- Sabrina Sailer
- Biocenter, Institute of Biological Chemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.R.W.)
| | - Markus A. Keller
- Institute of Human Genetics, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Ernst R. Werner
- Biocenter, Institute of Biological Chemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.R.W.)
| | - Katrin Watschinger
- Biocenter, Institute of Biological Chemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.R.W.)
- Correspondence: ; Tel.: +43-512-9003-70344
| |
Collapse
|
8
|
Marathe GK, Chaithra VH, Ke LY, Chen CH. Effect of acyl and alkyl analogs of platelet-activating factor on inflammatory signaling. Prostaglandins Other Lipid Mediat 2020; 151:106478. [PMID: 32711129 DOI: 10.1016/j.prostaglandins.2020.106478] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 07/20/2020] [Accepted: 07/20/2020] [Indexed: 01/11/2023]
Abstract
Platelet-activating factor (PAF), a bioactive ether phospholipid with significant pro-inflammatory properties, was identified almost half a century ago. Despite extensive study of this autocoid, therapeutic strategies for targeting its signaling components have not been successful, including the recent clinical trials with darapladib, a drug that targets plasma PAF-acetylhydrolase (PAF-AH). We recently provided experimental evidence that the previously unrecognized acyl analog of PAF, which is concomitantly produced along with PAF during biosynthesis, dampens PAF signaling by acting both as a sacrificial substrate for PAF-AH and probably as an endogenous PAF-receptor antagonist/partial agonist. If this is the scenario in vivo, PAF-AH needs to catalyze the selective hydrolysis of alkyl-PAF and not acyl-PAF. Accordingly, different approaches are needed for treating inflammatory diseases in which PAF signaling is implicated. The interplay between acyl-PAF, alkyl-PAF, PAF-AH, and PAF-R is complex, and the outcome of this interplay has not been previously appreciated. In this review, we discuss this interaction based on our recent findings. It is very likely that the relative abundance of acyl and alkyl-PAF and their interactions with PAF-R in the presence of their hydrolyzing enzyme PAF-AH may exert a modulatory effect on PAF signaling during inflammation.
Collapse
Affiliation(s)
- Gopal Kedihithlu Marathe
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, 570006, Karnataka, India; Department of Studies in Molecular Biology, University of Mysore, Manasagangothri, Mysuru, 570006, Karnataka, India.
| | | | - Liang-Yin Ke
- College of Health Sciences, Kaohsiung Medical University, Vascular and Medicinal Research, Kaohsiung, 80708, Taiwan.
| | - Chu-Huang Chen
- Vascular and Medicinal Research, Texas Heart Institute, Houston, Texas 77030, USA.
| |
Collapse
|
9
|
Dorninger F, Forss-Petter S, Wimmer I, Berger J. Plasmalogens, platelet-activating factor and beyond - Ether lipids in signaling and neurodegeneration. Neurobiol Dis 2020; 145:105061. [PMID: 32861763 PMCID: PMC7116601 DOI: 10.1016/j.nbd.2020.105061] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/12/2022] Open
Abstract
Glycerol-based ether lipids including ether phospholipids form a specialized branch of lipids that in mammals require peroxisomes for their biosynthesis. They are major components of biological membranes and one particular subgroup, the plasmalogens, is widely regarded as a cellular antioxidant. Their vast potential to influence signal transduction pathways is less well known. Here, we summarize the literature showing associations with essential signaling cascades for a wide variety of ether lipids, including platelet-activating factor, alkylglycerols, ether-linked lysophosphatidic acid and plasmalogen-derived polyunsaturated fatty acids. The available experimental evidence demonstrates links to several common players like protein kinase C, peroxisome proliferator-activated receptors or mitogen-activated protein kinases. Furthermore, ether lipid levels have repeatedly been connected to some of the most abundant neurological diseases, particularly Alzheimer's disease and more recently also neurodevelopmental disorders like autism. Thus, we critically discuss the potential role of these compounds in the etiology and pathophysiology of these diseases with an emphasis on signaling processes. Finally, we review the emerging interest in plasmalogens as treatment target in neurological diseases, assessing available data and highlighting future perspectives. Although many aspects of ether lipid involvement in cellular signaling identified in vitro still have to be confirmed in vivo, the compiled data show many intriguing properties and contributions of these lipids to health and disease that will trigger further research.
Collapse
Affiliation(s)
- Fabian Dorninger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, Vienna 1090, Austria.
| | - Sonja Forss-Petter
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, Vienna 1090, Austria
| | - Isabella Wimmer
- Department of Neurology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna 1090, Austria
| | - Johannes Berger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, Vienna 1090, Austria.
| |
Collapse
|
10
|
Watschinger K, Keller MA, Golderer G, Coassin S, Zschocke J, Werner ER. Biochemical Characterization of AGMO Variants Implicated in Relapses in Visceral Leishmaniasis. J Infect Dis 2019; 217:1846-1847. [PMID: 29741738 PMCID: PMC5946819 DOI: 10.1093/infdis/jiy090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 02/15/2018] [Indexed: 12/20/2022] Open
Affiliation(s)
- Katrin Watschinger
- Division of Biological Chemistry, Biocenter, Medical University of Innsbruck, Austria
| | - Markus A Keller
- Division of Human Genetics, Medical University of Innsbruck, Austria
| | - Georg Golderer
- Division of Biological Chemistry, Biocenter, Medical University of Innsbruck, Austria
| | - Stefan Coassin
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Austria
| | - Johannes Zschocke
- Division of Human Genetics, Medical University of Innsbruck, Austria
| | - Ernst R Werner
- Division of Biological Chemistry, Biocenter, Medical University of Innsbruck, Austria
| |
Collapse
|
11
|
Yu H, Dilbaz S, Coßmann J, Hoang AC, Diedrich V, Herwig A, Harauma A, Hoshi Y, Moriguchi T, Landgraf K, Körner A, Lucas C, Brodesser S, Balogh L, Thuróczy J, Karemore G, Kuefner MS, Park EA, Rapp C, Travers JB, Röszer T. Breast milk alkylglycerols sustain beige adipocytes through adipose tissue macrophages. J Clin Invest 2019; 129:2485-2499. [PMID: 31081799 PMCID: PMC6546455 DOI: 10.1172/jci125646] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 03/12/2019] [Indexed: 12/26/2022] Open
Abstract
Prevalence of obesity among infants and children below 5 years of age is rising dramatically, and early childhood obesity is a forerunner of obesity and obesity-associated diseases in adulthood. Childhood obesity is hence one of the most serious public health challenges today. Here, we have identified a mother-to-child lipid signaling that protects from obesity. We have found that breast milk-specific lipid species, so-called alkylglycerol-type (AKG-type) ether lipids, which are absent from infant formula and adult-type diets, maintain beige adipose tissue (BeAT) in the infant and impede the transformation of BeAT into lipid-storing white adipose tissue (WAT). Breast milk AKGs are metabolized by adipose tissue macrophages (ATMs) to platelet-activating factor (PAF), which ultimately activates IL-6/STAT3 signaling in adipocytes and triggers BeAT development in the infant. Accordingly, lack of AKG intake in infancy leads to a premature loss of BeAT and increases fat accumulation. AKG signaling is specific for infants and is inactivated in adulthood. However, in obese adipose tissue, ATMs regain their ability to metabolize AKGs, which reduces obesity. In summary, AKGs are specific lipid signals of breast milk that are essential for healthy adipose tissue development.
Collapse
Affiliation(s)
| | - Sedat Dilbaz
- Institute of Neurobiology, and
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | | | | | | | | | - Akiko Harauma
- Department of Food and Life Science, Azabu University, Sagamihara, Kanagawa, Japan
| | - Yukino Hoshi
- Department of Food and Life Science, Azabu University, Sagamihara, Kanagawa, Japan
| | - Toru Moriguchi
- Department of Food and Life Science, Azabu University, Sagamihara, Kanagawa, Japan
| | - Kathrin Landgraf
- Center for Pediatric Research, University Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
| | - Antje Körner
- Center for Pediatric Research, University Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
| | - Christina Lucas
- Lipidomics Facility, CECAD Research Center, University of Cologne, Cologne, Germany
| | - Susanne Brodesser
- Lipidomics Facility, CECAD Research Center, University of Cologne, Cologne, Germany
| | - Lajos Balogh
- Department of Nuclear Medicine, National Public Health Center (NPHC), Budapest, Hungary
| | - Julianna Thuróczy
- Department of Nuclear Medicine, National Public Health Center (NPHC), Budapest, Hungary
| | - Gopal Karemore
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Michael Scott Kuefner
- Veterans Affairs Medical Center and the Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Edwards A. Park
- Veterans Affairs Medical Center and the Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Christine Rapp
- Department of Pharmacology and Toxicology, Wright State University, Dayton, Ohio, USA
| | | | | |
Collapse
|
12
|
Marquet S, Bucheton B, Reymond C, Argiro L, El-Safi SH, Kheir MM, Desvignes JP, Béroud C, Mergani A, Hammad A, Dessein AJ. Exome Sequencing Identifies Two Variants of the Alkylglycerol Monooxygenase Gene as a Cause of Relapses in Visceral Leishmaniasis in Children, in Sudan. J Infect Dis 2017; 216:22-28. [PMID: 28586473 DOI: 10.1093/infdis/jix277] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 06/02/2017] [Indexed: 01/01/2023] Open
Abstract
Background Visceral leishmaniasis (kala-azar, KA) is the most severe form of leishmaniasis, characterized by fever, weight loss, hepatosplenomegaly, and lymphadenopathy. During an outbreak of KA in Babar El Fugara (Sudan), 5.7% of cured patients displayed relapses, with familial clustering in half the cases. Methods We performed whole-exome sequencing on 10 relapsing individuals and 11 controls from 5 nuclear families. Results Rare homozygous and compound-heterozygous nonsense (c.1213C > T, rs139309795, p.Arg405*) and missense (c.701A > G, rs143439626, p.Lys234Arg) mutations of the alkylglycerol monooxygenase (AGMO) gene were associated with KA relapse in 3 families. Sequencing in additional family members confirmed the segregation of these mutations with relapse and revealed an autosomal dominant mode of transmission. These mutations were detected heterozygous in 2 subjects among 100 unrelated individuals with KA who never relapsed after cure, suggesting incomplete penetrance of AGMO deficiency. AGMO is expressed in hematopoietic cells, and is strongly expressed in the liver. AGMO modulates PAF production by mouse macrophages, suggesting that it may act through the PAF/PAF receptor pathway previously shown to have anti-Leishmania activity. Conclusions This is the first demonstration that relapses after a first episode of KA are due to differences in human genetic susceptibility and not to modifications of parasite pathogenicity.
Collapse
Affiliation(s)
- Sandrine Marquet
- INSERM UMR906, GIMP, Labex ParaFrap, Aix-Marseille University, Marseille
| | - Bruno Bucheton
- INSERM UMR906, GIMP, Labex ParaFrap, Aix-Marseille University, Marseille.,Institut de Recherche pour le Développement, Unité Mixte de Recherche IRD-CIRAD 177, Campus International de Baillarguet, Montpellier, France
| | - Camille Reymond
- INSERM UMR906, GIMP, Labex ParaFrap, Aix-Marseille University, Marseille
| | - Laurent Argiro
- INSERM UMR906, GIMP, Labex ParaFrap, Aix-Marseille University, Marseille
| | - Sayda Hassan El-Safi
- Department of Microbiology and Parasitology, Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Musa Mohamed Kheir
- Department of Microbiology and Parasitology, Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | | | - Christophe Béroud
- INSERM UMR910, GMGF, Aix-Marseille University.,AP-HM, Département de Génétique Médicale, Hôpital Timone Enfants, Marseille, France
| | - Adil Mergani
- College of Applied Medical Sciences, Taif University, Turabah, Saudi Arabia
| | - Awad Hammad
- Department of Microbiology and Parasitology, Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Alain J Dessein
- INSERM UMR906, GIMP, Labex ParaFrap, Aix-Marseille University, Marseille
| |
Collapse
|
13
|
Orlova M, Schurr E. Human Genomics of Mycobacterium tuberculosis Infection and Disease. CURRENT GENETIC MEDICINE REPORTS 2017; 5:125-131. [PMID: 29201558 DOI: 10.1007/s40142-017-0124-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Purpose of review The study of the genetic basis of tuberculosis pathogenesis has benefited from powerful technological innovations, a more structured definition of latent and clinical manifestations of the disease, and the application of functional genomics approaches. This short review aims to summarize recent advances and to provide a link with results of previous human genetic studies of tuberculosis susceptibility. Recent findings Transcriptomics has been shown to be a useful tool to predict progression from latency to clinical disease while functional genomics has traced the molecular events that link pathogen-triggered gene expression and host genetics. Resistance to infection with Mycobacterium tuberculosis has been revealed to be strongly impacted by host genetics. Host genomics of clinical disease has been shown to be most powerful when focusing on carefully selected clinical entities and possibly by considering host pathogen combinations. Summary Future studies need to build on the latest molecular findings to define disease subtypes to successfully elucidate the human genetic component in tuberculosis pathogenesis.
Collapse
Affiliation(s)
- Marianna Orlova
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,McGill International TB Centre, McGill University, Montreal, Quebec, Canada.,Departments of Medicine and Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Erwin Schurr
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,McGill International TB Centre, McGill University, Montreal, Quebec, Canada.,Departments of Medicine and Human Genetics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
14
|
Waqas SFH, Hoang AC, Lin YT, Ampem G, Azegrouz H, Balogh L, Thuróczy J, Chen JC, Gerling IC, Nam S, Lim JS, Martinez-Ibañez J, Real JT, Paschke S, Quillet R, Ayachi S, Simonin F, Schneider EM, Brinkman JA, Lamming DW, Seroogy CM, Röszer T. Neuropeptide FF increases M2 activation and self-renewal of adipose tissue macrophages. J Clin Invest 2017; 127:2842-2854. [PMID: 28581443 DOI: 10.1172/jci90152] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 04/06/2017] [Indexed: 12/30/2022] Open
Abstract
The quantity and activation state of adipose tissue macrophages (ATMs) impact the development of obesity-induced metabolic diseases. Appetite-controlling hormones play key roles in obesity; however, our understanding of their effects on ATMs is limited. Here, we have shown that human and mouse ATMs express NPFFR2, a receptor for the appetite-reducing neuropeptide FF (NPFF), and that NPFFR2 expression is upregulated by IL-4, an M2-polarizing cytokine. Plasma levels of NPFF decreased in obese patients and high-fat diet-fed mice and increased following caloric restriction. NPFF promoted M2 activation and increased the proliferation of murine and human ATMs. Both M2 activation and increased ATM proliferation were abolished in NPFFR2-deficient ATMs. Mechanistically, the effects of NPFF involved the suppression of E3 ubiquitin ligase RNF128 expression, resulting in enhanced stability of phosphorylated STAT6 and increased transcription of the M2 macrophage-associated genes IL-4 receptor α (Il4ra), arginase 1 (Arg1), IL-10 (Il10), and alkylglycerol monooxygenase (Agmo). NPFF induced ATM proliferation concomitantly with the increase in N-Myc downstream-regulated gene 2 (Ndrg2) expression and suppressed the transcription of Ifi200 cell-cycle inhibitor family members and MAF bZIP transcription factor B (Mafb), a negative regulator of macrophage proliferation. NPFF thus plays an important role in supporting healthy adipose tissue via the maintenance of metabolically beneficial ATMs.
Collapse
Affiliation(s)
| | - Anh Cuong Hoang
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Ya-Tin Lin
- Department of Physiology and Pharmacology and Graduate Institute of Biomedical Sciences, Chang Gung University; Neuroscience Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Grace Ampem
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Hind Azegrouz
- Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Lajos Balogh
- National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
| | | | - Jin-Chung Chen
- Department of Physiology and Pharmacology and Graduate Institute of Biomedical Sciences, Chang Gung University; Neuroscience Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ivan C Gerling
- Department of Medicine, University of Tennessee, Memphis, Tennessee, USA
| | - Sorim Nam
- Department of Biological Science, Sookmyung Women's University, Seoul, South Korea
| | - Jong-Seok Lim
- Department of Biological Science, Sookmyung Women's University, Seoul, South Korea
| | - Juncal Martinez-Ibañez
- Department of Medicine, Hospital Clínico Universitario de València, Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Valencia, Spain
| | - José T Real
- Department of Medicine, Hospital Clínico Universitario de València, Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Valencia, Spain
| | - Stephan Paschke
- Department of General and Visceral Surgery, University Hospital Ulm, Ulm, Germany
| | - Raphaëlle Quillet
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de Recherche Scientifique (CNRS), Université de Strasbourg, Illkirch, France
| | - Safia Ayachi
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de Recherche Scientifique (CNRS), Université de Strasbourg, Illkirch, France
| | - Frédéric Simonin
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de Recherche Scientifique (CNRS), Université de Strasbourg, Illkirch, France
| | - E Marion Schneider
- Division of Experimental Anesthesiology, University Hospital Ulm, Ulm, Germany
| | - Jacqueline A Brinkman
- University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, USA.,William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Dudley W Lamming
- University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, USA.,William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Christine M Seroogy
- University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Tamás Röszer
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| |
Collapse
|
15
|
Zschiebsch K, Fischer C, Pickert G, Häussler A, Radeke H, Grösch S, Ferreirós N, Geisslinger G, Werner ER, Tegeder I. Tetrahydrobiopterin Attenuates DSS-evoked Colitis in Mice by Rebalancing Redox and Lipid Signalling. J Crohns Colitis 2016; 10:965-78. [PMID: 26928964 DOI: 10.1093/ecco-jcc/jjw056] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 02/01/2016] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND AIMS Guanosine triphosphate cyclohydrolase [GCH1] governs the production of the enzyme cofactor tetrahydrobiopterin [BH4] which is essential for biogenic amine synthesis, lipid metabolism via alkylglycerol monooxygenase [AGMO], and redox coupling of nitric oxide synthases [NOSs]. Inflammation-evoked unequal regulation of GCH1 and NOS or AGMO may cause redox stress and lipid imbalances. METHODS The present study assessed potential therapeutic effects of rebalancing these systems with BH4 in experimental colitis in mice. RESULTS Oral treatment with BH4 as a suspension of crushed tablets attenuated colitis, whereas inhibition of its production had opposite effects: aggravated weight loss, epithelial haemorrhages and ulcers, neutrophil infiltrates, production of reactive oxygen species, and unfavourable profile changes of endocannabinoids, ceramides, and lysophosphatidic acids. Conversely, oral BH4 normalised biopterin, reduced in vivo activity of oxidases and peroxidases in the inflamed gut, favoured nitric oxide over hydrogen peroxide, and maintained normal levels of lipid signalling molecules. BH4 favoured thereby resident CD3+CD8+ and regulatory CD3+CD25+ intraepithelial T cells that are important for epithelial integrity. CONCLUSIONS BH4 protected against colitis in mice via two major pathways: [i] by reduction of oxidative stress; and [ii] by re-orchestration of alkyl- and acylglycerolipid signalling via AGMO. Oral treatment with BH4 is a safe approved supplementary therapy for genetic BH4 deficiency and did not excessively increase systemic BH4 levels. Therefore, one may consider repurposing of oral BH4 as an adjunctive treatment for colitis.
Collapse
Affiliation(s)
- Katja Zschiebsch
- Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Germany
| | - Caroline Fischer
- Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Germany
| | - Geethanjali Pickert
- Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Germany
| | - Annett Häussler
- Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Germany
| | - Heinfried Radeke
- Department of Experimental Pharmacology, Goethe-University Hospital Frankfurt, Germany
| | - Sabine Grösch
- Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Germany
| | - Nerea Ferreirós
- Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Germany
| | - Gerd Geisslinger
- Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Germany
| | - Ernst R Werner
- Division of Biological Chemistry, Medical University of Innsbruck, Austria
| | - Irmgard Tegeder
- Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Germany
| |
Collapse
|
16
|
Grant AV, Sabri A, Abid A, Abderrahmani Rhorfi I, Benkirane M, Souhi H, Naji Amrani H, Alaoui-Tahiri K, Gharbaoui Y, Lazrak F, Sentissi I, Manessouri M, Belkheiri S, Zaid S, Bouraqadi A, El Amraoui N, Hakam M, Belkadi A, Orlova M, Boland A, Deswarte C, Amar L, Bustamante J, Boisson-Dupuis S, Casanova JL, Schurr E, El Baghdadi J, Abel L. A genome-wide association study of pulmonary tuberculosis in Morocco. Hum Genet 2016; 135:299-307. [PMID: 26767831 DOI: 10.1007/s00439-016-1633-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 01/04/2016] [Indexed: 01/04/2023]
Abstract
Although epidemiological evidence suggests a human genetic basis of pulmonary tuberculosis (PTB) susceptibility, the identification of specific genes and alleles influencing PTB risk has proven to be difficult. Previous genome-wide association (GWA) studies have identified only three novel loci with modest effect sizes in sub-Saharan African and Russian populations. We performed a GWA study of 550,352 autosomal SNPs in a family-based discovery Moroccan sample (on the full population and on the subset with PTB diagnosis at <25 years), which identified 143 SNPs with p < 1 × 10(-4). The replication study in an independent case/control sample identified four SNPs displaying a p < 0.01 implicating the same risk allele. In the combined sample including 556 PTB subjects and 650 controls these four SNPs showed suggestive association (2 × 10(-6) < p < 4 × 10(-5)): rs358793 and rs17590261 were intergenic, while rs6786408 and rs916943 were located in introns of FOXP1 and AGMO, respectively. Both genes are involved in the function of macrophages, which are the site of latency and reactivation of Mycobacterium tuberculosis. The most significant finding (p = 2 × 10(-6)) was obtained for the AGMO SNP in an early (<25 years) age-at-onset subset, confirming the importance of considering age-at-onset to decipher the genetic basis of PTB. Although only suggestive, these findings highlight several avenues for future research in the human genetics of PTB.
Collapse
Affiliation(s)
- A V Grant
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, 75015, Paris, France, EU
- Paris Descartes University, Imagine Institute, 75015, Paris, France, EU
| | - A Sabri
- Genetics Unit, Military Hospital Mohammed V, Hay Riad, 10100, Rabat, Morocco
- Faculty of Sciences-Kenitra, Ibn Tofail University, Kenitra, Morocco
| | - A Abid
- Department of Pneumology, Military Hospital Mohammed V, Hay Riad, 10100, Rabat, Morocco
| | - I Abderrahmani Rhorfi
- Department of Pneumology, Military Hospital Mohammed V, Hay Riad, 10100, Rabat, Morocco
| | - M Benkirane
- Blood Transfusion Center, Military Hospital Mohammed V, Hay Riad, 10100, Rabat, Morocco
| | - H Souhi
- Department of Pneumology, Military Hospital Mohammed V, Hay Riad, 10100, Rabat, Morocco
| | - H Naji Amrani
- Department of Pneumology, Military Hospital Mohammed V, Hay Riad, 10100, Rabat, Morocco
| | - K Alaoui-Tahiri
- Department of Pneumology, Military Hospital Mohammed V, Hay Riad, 10100, Rabat, Morocco
| | - Y Gharbaoui
- Department of Pneumology, Military Hospital Mohammed V, Hay Riad, 10100, Rabat, Morocco
| | - F Lazrak
- Centre de Diagnostic de la tuberculose et des Maladies Respiratoires [CDTMR], Salé, Morocco
| | - I Sentissi
- Centre de Diagnostic de la tuberculose et des Maladies Respiratoires [CDTMR], Salé, Morocco
| | - M Manessouri
- Centre de Diagnostic de la tuberculose et des Maladies Respiratoires [CDTMR], Salé, Morocco
| | - S Belkheiri
- Centre de Diagnostic de la tuberculose et des Maladies Respiratoires [CDTMR], Salé, Morocco
| | - S Zaid
- Centre de Diagnostic de la tuberculose et des Maladies Respiratoires [CDTMR], Salé, Morocco
| | - A Bouraqadi
- Centre de Diagnostic de la tuberculose et des Maladies Respiratoires [CDTMR], Salé, Morocco
| | - N El Amraoui
- National Blood Transfusion Center, Rabat, Morocco
| | - M Hakam
- National Blood Transfusion Center, Rabat, Morocco
| | - A Belkadi
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, 75015, Paris, France, EU
- Paris Descartes University, Imagine Institute, 75015, Paris, France, EU
| | - M Orlova
- McGill Centre for the Study of Host Resistance, The Research Institute of the McGill University Health Centre, Montreal, PQ H3G 1A4, Canada
| | - A Boland
- CEA, Institut de Génomique, Centre National de Génotypage, 91000, Evry, France, EU
| | - C Deswarte
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, 75015, Paris, France, EU
- Paris Descartes University, Imagine Institute, 75015, Paris, France, EU
| | - L Amar
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, 75015, Paris, France, EU
- Paris Descartes University, Imagine Institute, 75015, Paris, France, EU
| | - J Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, 75015, Paris, France, EU
- Paris Descartes University, Imagine Institute, 75015, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, 10065, New York, NY, USA
| | - S Boisson-Dupuis
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, 75015, Paris, France, EU
- Paris Descartes University, Imagine Institute, 75015, Paris, France, EU
- Center for the Study of Primary Immunodeficiencies, AP-HP, Necker hospital, 75015, Paris, France, EU
| | - J L Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, 75015, Paris, France, EU
- Paris Descartes University, Imagine Institute, 75015, Paris, France, EU
- Center for the Study of Primary Immunodeficiencies, AP-HP, Necker hospital, 75015, Paris, France, EU
- Howard Hughes Medical Institute, New York, NY, USA
- Pediatric Hematology-Immunology Unit, AP-HP, Necker Hospital, 75015, Paris, France, EU
| | - E Schurr
- McGill Centre for the Study of Host Resistance, The Research Institute of the McGill University Health Centre, Montreal, PQ H3G 1A4, Canada
| | - J El Baghdadi
- Genetics Unit, Military Hospital Mohammed V, Hay Riad, 10100, Rabat, Morocco.
| | - L Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, 75015, Paris, France, EU.
- Paris Descartes University, Imagine Institute, 75015, Paris, France, EU.
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, 10065, New York, NY, USA.
- Human Genetics of Infectious Diseases, INSERM, Université Paris Descartes, Unit 1163, Imagine Institute, 24 Bd du Montparnasse, 75105, Paris, France.
| |
Collapse
|
17
|
Watschinger K, Keller MA, McNeill E, Alam MT, Lai S, Sailer S, Rauch V, Patel J, Hermetter A, Golderer G, Geley S, Werner-Felmayer G, Plumb RS, Astarita G, Ralser M, Channon KM, Werner ER. Tetrahydrobiopterin and alkylglycerol monooxygenase substantially alter the murine macrophage lipidome. Proc Natl Acad Sci U S A 2015; 112:2431-6. [PMID: 25675482 PMCID: PMC4345615 DOI: 10.1073/pnas.1414887112] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
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-1β, and interleukin 1 receptor antagonist but not transforming growth factor β1, 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.
Collapse
Affiliation(s)
- Katrin Watschinger
- Division of Biological Chemistry, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Markus A Keller
- Division of Biological Chemistry, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria; Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge CB2 1GA, United Kingdom
| | - Eileen McNeill
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Mohammad T Alam
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge CB2 1GA, United Kingdom
| | | | - Sabrina Sailer
- Division of Biological Chemistry, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Veronika Rauch
- Division of Molecular Pathophysiology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Jyoti Patel
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Albin Hermetter
- Institute of Biochemistry, Graz University of Technology, 8010 Graz, Austria
| | - Georg Golderer
- Division of Biological Chemistry, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Stephan Geley
- Division of Molecular Pathophysiology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Gabriele Werner-Felmayer
- Division of Biological Chemistry, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Robert S Plumb
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, South Kensington, London SW7 2AZ, United Kingdom
| | - Giuseppe Astarita
- Waters Corporation, Milford, MA 01757; Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057-1468; and
| | - Markus Ralser
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge CB2 1GA, United Kingdom; Medical Research Council National Institute for Medical Research, London NW7 1AA, United Kingdom
| | - Keith M Channon
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Ernst R Werner
- Division of Biological Chemistry, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| |
Collapse
|
18
|
Bendall JK, Douglas G, McNeill E, Channon KM, Crabtree MJ. Tetrahydrobiopterin in cardiovascular health and disease. Antioxid Redox Signal 2014; 20:3040-77. [PMID: 24294830 PMCID: PMC4038990 DOI: 10.1089/ars.2013.5566] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 11/01/2013] [Accepted: 12/02/2013] [Indexed: 01/03/2023]
Abstract
Tetrahydrobiopterin (BH4) functions as a cofactor for several important enzyme systems, and considerable evidence implicates BH4 as a key regulator of endothelial nitric oxide synthase (eNOS) in the setting of cardiovascular health and disease. BH4 bioavailability is determined by a balance of enzymatic de novo synthesis and recycling, versus degradation in the setting of oxidative stress. Augmenting vascular BH4 levels by pharmacological supplementation has been shown in experimental studies to enhance NO bioavailability. However, it has become more apparent that the role of BH4 in other enzymatic pathways, including other NOS isoforms and the aromatic amino acid hydroxylases, may have a bearing on important aspects of vascular homeostasis, inflammation, and cardiac function. This article reviews the role of BH4 in cardiovascular development and homeostasis, as well as in pathophysiological processes such as endothelial and vascular dysfunction, atherosclerosis, inflammation, and cardiac hypertrophy. We discuss the therapeutic potential of BH4 in cardiovascular disease states and attempt to address how this modulator of intracellular NO-redox balance may ultimately provide a powerful new treatment for many cardiovascular diseases.
Collapse
Affiliation(s)
- Jennifer K Bendall
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford , John Radcliffe Hospital, Oxford, United Kingdom
| | | | | | | | | |
Collapse
|
19
|
Tarui M, Shindou H, Kumagai K, Morimoto R, Harayama T, Hashidate T, Kojima H, Okabe T, Nagano T, Nagase T, Shimizu T. Selective inhibitors of a PAF biosynthetic enzyme lysophosphatidylcholine acyltransferase 2. J Lipid Res 2014; 55:1386-96. [PMID: 24850807 PMCID: PMC4076079 DOI: 10.1194/jlr.m049205] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Indexed: 01/09/2023] Open
Abstract
Platelet-activating factor (PAF) is a potent pro-inflammatory phospholipid mediator. In response to extracellular stimuli, PAF is rapidly biosynthesized by lyso-PAF acetyltransferase (lyso-PAFAT). Previously, we identified two types of lyso-PAFATs: lysophosphatidylcholine acyltransferase (LPCAT)1, mostly expressed in the lungs where it produces PAF and dipalmitoyl-phosphatidylcholine essential for respiration, and LPCAT2, which biosynthesizes PAF and phosphatidylcholine (PC) in the inflammatory cells. Under inflammatory conditions, LPCAT2, but not LPCAT1, is activated and upregulated to produce PAF. Thus, it is important to develop inhibitors specific for LPCAT2 in order to ameliorate PAF-related inflammatory diseases. Here, we report the first identification of LPCAT2-specific inhibitors, N-phenylmaleimide derivatives, selected from a 174,000-compound library using fluorescence-based high-throughput screening followed by the evaluation of the effects on LPCAT1 and LPCAT2 activities, cell viability, and cellular PAF production. Selected compounds competed with acetyl-CoA for the inhibition of LPCAT2 lyso-PAFAT activity and suppressed PAF biosynthesis in mouse peritoneal macrophages stimulated with a calcium ionophore. These compounds had low inhibitory effects on LPCAT1 activity, indicating that adverse effects on respiratory functions may be avoided. The identified compounds and their derivatives will contribute to the development of novel drugs for PAF-related diseases and facilitate the analysis of LPCAT2 functions in phospholipid metabolism in vivo.
Collapse
Affiliation(s)
- Megumi Tarui
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan Department of Respiratory Medicine, Faculty of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hideo Shindou
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
| | - Kazuo Kumagai
- Open Innovation Center for Drug Discovery, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Ryo Morimoto
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takeshi Harayama
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Tomomi Hashidate
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Hirotatsu Kojima
- Open Innovation Center for Drug Discovery, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takayoshi Okabe
- Open Innovation Center for Drug Discovery, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tetsuo Nagano
- Open Innovation Center for Drug Discovery, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takahide Nagase
- Department of Respiratory Medicine, Faculty of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takao Shimizu
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
20
|
Morimoto R, Shindou H, Tarui M, Shimizu T. Rapid production of platelet-activating factor is induced by protein kinase Cα-mediated phosphorylation of lysophosphatidylcholine acyltransferase 2 protein. J Biol Chem 2014; 289:15566-76. [PMID: 24742674 PMCID: PMC4140912 DOI: 10.1074/jbc.m114.558874] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Platelet-activating factor (PAF), a potent proinflammatory lipid mediator, is synthesized rapidly in response to extracellular stimuli by the activation of acetyl-CoA:lyso-PAF acetyltransferase (lyso-PAFAT). We have reported previously that lyso-PAFAT activity is enhanced in three distinct ways in mouse macrophages: rapid activation (30 s) after PAF stimulation and minutes to hours after LPS stimulation. Lysophosphatidylcholine acyltransferase 2 (LPCAT2) was later identified as a Ca(2+)-dependent lyso-PAFAT. However, the mechanism of rapid lyso-PAFAT activation within 30 s has not been elucidated. Here we show a new signaling pathway for rapid biosynthesis of PAF that is mediated by phosphorylation of LPCAT2 at Ser-34. Stimulation by either PAF or ATP resulted in PKCα-mediated phosphorylation of LPCAT2 to enhance lyso-PAFAT activity and rapid PAF production. Biochemical analyses showed that the phosphorylation of Ser-34 resulted in augmentation of Vmax with minimal Km change. Our results offer an answer for the previously unknown mechanism of rapid PAF production.
Collapse
Affiliation(s)
- Ryo Morimoto
- From the Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan, the Department of Biochemistry and Molecular Biology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, and
| | - Hideo Shindou
- From the Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan, the Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Megumi Tarui
- From the Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Takao Shimizu
- From the Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan, the Department of Biochemistry and Molecular Biology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, and
| |
Collapse
|