1
|
Jalil A, Pilot T, Bourgeois T, Laubriet A, Li X, Diedisheim M, Deckert V, Magnani C, Le Guern N, Pais de Barros JP, Nguyen M, Pallot G, Vouilloz A, Proukhnitzky L, Hermetet F, Aires V, Lesniewska E, Lagrost L, Auwerx J, Le Goff W, Venteclef N, Steinmetz E, Thomas C, Masson D. Plasmalogen remodeling modulates macrophage response to cytotoxic oxysterols and atherosclerotic plaque vulnerability. Cell Rep Med 2025; 6:102131. [PMID: 40345182 DOI: 10.1016/j.xcrm.2025.102131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 11/18/2024] [Accepted: 04/16/2025] [Indexed: 05/11/2025]
Abstract
Essential fatty acid metabolism in myeloid cells plays a critical but underexplored role in immune function. Here, we demonstrate that simultaneous inactivation of two key enzymes involved in macrophage polyunsaturated fatty acid (PUFA) metabolism-ELOVL5, which elongates long-chain PUFAs, and LPCAT3, which incorporates them into phospholipids-disrupts membrane organization by promoting the formation of cholesterol-enriched domains. This increases macrophage sensitivity to cytotoxic oxysterols and leads to more vulnerable atherosclerotic plaques with enlarged necrotic cores in a mouse model of atherosclerosis. In humans, analysis of 187 carotid plaques reveals a positive correlation between LPCAT3/ELOVL5-generated phospholipids-including arachidonate (C20:4 n-6)-containing ether lipids-and more stable plaque profiles. Additionally, Mendelian randomization analysis supports a causal relationship between LPCAT3 expression and reduced risk of ischemic stroke. Our findings uncover a regulatory circuit essential for PUFA-containing phospholipid generation in macrophages, positioning PUFA-containing ether lipids as promising biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Antoine Jalil
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Thomas Pilot
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Thibaut Bourgeois
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Aline Laubriet
- CHRU Dijon Bourgogne, Department of Cardiovascular Surgery, Dijon University Medical Center, 21000 Dijon, France
| | - Xiaoxu Li
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Marc Diedisheim
- Centre - Clinique Saint Gatien Alliance (NCT+), 37214 Saint-Cyr-sur-Loire, France; Institut Necker-Enfants Malades, INSERM UMR-S1151, Université Paris Cité, 75015 Paris, France
| | - Valérie Deckert
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Charlène Magnani
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Naig Le Guern
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Jean-Paul Pais de Barros
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France; Lipidomic Analytic Platform, UBFC, 21000 Dijon, France
| | - Maxime Nguyen
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France; CHRU Dijon Bourgogne, Department of Anesthesiology and Critical Care Medicine, Dijon University Medical Center, 21000 Dijon, France
| | - Gaëtan Pallot
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Adrien Vouilloz
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Lil Proukhnitzky
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - François Hermetet
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Virginie Aires
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Eric Lesniewska
- Université Bourgogne, UMR1231, 21000 Dijon, France; Laboratory of Physics, National Center for Scientific Research, URA 5027, UFR Sciences et techniques, 21000 Dijon, France
| | - Laurent Lagrost
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Wilfried Le Goff
- Sorbonne Université, Inserm, ICAN Institut, UMR_S1166, Hôpital de la Pitié, 75013 Paris, France
| | - Nicolas Venteclef
- Institut Necker-Enfants Malades, INSERM UMR-S1151, Université Paris Cité, 75015 Paris, France
| | - Eric Steinmetz
- CHRU Dijon Bourgogne, Department of Cardiovascular Surgery, Dijon University Medical Center, 21000 Dijon, France
| | - Charles Thomas
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - David Masson
- Université Bourgogne, UMR1231, 21000 Dijon, France; INSERM, UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France; CHRU Dijon Bourgogne, Laboratory of Clinical Chemistry, 21000 Dijon, France.
| |
Collapse
|
2
|
Koyama T, Osada K. Exogenous oxidized phytosterol may modulate linoleic acid metabolism through upregulation of fatty acid desaturase in rats. Lipids 2025. [PMID: 40204289 DOI: 10.1002/lipd.12444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/03/2025] [Accepted: 03/20/2025] [Indexed: 04/11/2025]
Abstract
Previous in vitro studies have indicated that oxidized phytosterol (OPS) exhibits some toxicity; however, the harmful effects of OPS on fatty acid metabolism are not completely understood yet. Therefore, this study examined the effects of exogenous phytosterol (PS) and OPS on growth parameters and lipid metabolism in rats. Rats were provided with AIN-76 basal diet, basal diet +0.5% PS, or basal diet +0.5% OPS. We found that the level of cholesterol and triacylglycerols in the liver was significantly lower in OPS-fed rats than in basal diet-fed rats. The ratio of Δ6 desaturation index (20:3(n-6) + 20:4(n-6))/18:2(n-6) in the plasma was significantly higher in the OPS-fed rats than in the PS-fed rats. Additionally, the proportion of arachidonic acid (20:4) in the liver was significantly higher in the OPS-fed group compared with the control group. The mRNA expression levels of Δ6 and Δ5 desaturases were significantly higher in OPS-fed rats than in basal diet-fed rats, but remained unchanged in PS-fed rats. Moreover, the protein level of Δ6 desaturase was significantly higher in both PS- and OPS-fed rats compared with basal diet-fed rats, while the protein level of Δ5 desaturase tended to be higher only in OPS-fed rats than in basal diet-fed rats. Thus, exogenous OPS, but not PS, altered fatty acid composition through the upregulation of mRNA and protein levels of fatty acid desaturation enzymes in the liver. This indicates that exogenous OPS, unlike PS, may modulate the production of eicosanoids from arachidonic acid, potentially promoting allergic reactions, inflammation, and atherosclerosis.
Collapse
Affiliation(s)
- Tomonari Koyama
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| | - Kyoichi Osada
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| |
Collapse
|
3
|
Vouilloz A, Bourgeois T, Diedisheim M, Pilot T, Jalil A, Le Guern N, Bergas V, Rohmer N, Castelli F, Leleu D, Varin A, de Barros JPP, Degrace P, Rialland M, Blériot C, Venteclef N, Thomas C, Masson D. Impaired unsaturated fatty acid elongation alters mitochondrial function and accelerates metabolic dysfunction-associated steatohepatitis progression. Metabolism 2025; 162:156051. [PMID: 39454822 DOI: 10.1016/j.metabol.2024.156051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/18/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND AND AIMS Although qualitative and quantitative alterations in liver Polyunsaturated Fatty Acids (PUFAs) are observed in MASH in humans, a causal relationship of PUFAs biosynthetic pathways is yet to be clarified. ELOVL5, an essential enzyme in PUFA elongation regulates hepatic triglyceride metabolism. Nonetheless, the long-term consequences of elongase disruption, particularly in murine models of MASH, have not been evaluated. APPROACH & RESULTS In humans, transcriptomic data indicated that PUFAs biosynthesis enzymes and notably ELOVL5 were induced during MASH progression. Moreover, gene module association determination revealed that ELOVL5 expression was associated with mitochondrial function in both humans and mice. WT and Elovl5-deficient mice were fed a high-fat, high-sucrose (HF/HS) diet for four months. Elovl5 deficiency led to limited systemic metabolic alterations but significant hepatic phenotype was observed in Elovl5-/- mice after the HF/HS diet, including hepatomegaly, pronounced macrovesicular and microvesicular steatosis, hepatocyte ballooning, immune cell infiltration, and fibrosis. Lipid analysis confirmed hepatic triglyceride accumulation and a reshaping of FA profile. Transcriptomic analysis indicated significant upregulation of genes involved in immune cell recruitment and fibrosis, and downregulation of genes involved in oxidative phosphorylation in Elovl5-/- mice. Alterations of FA oxidation and energy metabolism were confirmed by non-targeted metabolomic approach. Analysis of mitochondrial function in Elovl5-/- mice showed morphological alterations, qualitative cardiolipin changes with an enrichment in species containing shorter unsaturated FAs, and decreased activity of I and III respiratory chain complexes. CONCLUSION Enhanced susceptibility to diet-induced MASH and fibrosis in Elovl5-/- mice is intricately associated with disruptions in mitochondrial homeostasis, stemming from a profound reshaping of mitochondrial lipids, notably cardiolipins.
Collapse
Affiliation(s)
- Adrien Vouilloz
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Thibaut Bourgeois
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Marc Diedisheim
- Institut Necker-Enfants Malades, INSERM UMR-S1151, Université Paris Cité, 75015 Paris, France; Clinique Saint Gatien Alliance (NCT+), Saint-Cyr-sur-Loire, France
| | - Thomas Pilot
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Antoine Jalil
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Naig Le Guern
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Victoria Bergas
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; Lipidomic Analytical Facility, 21000 Dijon, France
| | - Noéline Rohmer
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour La Santé (DMTS), MetaboHUB, F-91191 Gif-sur-Yvette, France
| | - Florence Castelli
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour La Santé (DMTS), MetaboHUB, F-91191 Gif-sur-Yvette, France
| | - Damien Leleu
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France; CHRU Dijon Bourgogne, Laboratory of Clinical Chemistry, 21000 Dijon, France
| | - Alexis Varin
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France; Lipidomic Analytical Facility, 21000 Dijon, France
| | - Jean-Paul Pais de Barros
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France; Lipidomic Analytical Facility, 21000 Dijon, France
| | - Pascal Degrace
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Mickael Rialland
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Camille Blériot
- Institut Necker-Enfants Malades, INSERM UMR-S1151, Université Paris Cité, 75015 Paris, France
| | - Nicolas Venteclef
- Institut Necker-Enfants Malades, INSERM UMR-S1151, Université Paris Cité, 75015 Paris, France
| | - Charles Thomas
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - David Masson
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France; CHRU Dijon Bourgogne, Laboratory of Clinical Chemistry, 21000 Dijon, France.
| |
Collapse
|
4
|
Ge X, Slütter B, Lambooij JM, Zhou E, Ying Z, Agirman C, Heijink M, Rimbert A, Guigas B, Kuiper J, Müller C, Bracher F, Giera M, Kooijman S, Rensen PC, Wang Y, Schönke M. DHCR24 inhibitor SH42 increases desmosterol without preventing atherosclerosis development in mice. iScience 2024; 27:109830. [PMID: 38770137 PMCID: PMC11103367 DOI: 10.1016/j.isci.2024.109830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/29/2024] [Accepted: 04/24/2024] [Indexed: 05/22/2024] Open
Abstract
The liver X receptor (LXR) is considered a therapeutic target for atherosclerosis treatment, but synthetic LXR agonists generally also cause hepatic steatosis and hypertriglyceridemia. Desmosterol, a final intermediate in cholesterol biosynthesis, has been identified as a selective LXR ligand that suppresses inflammation without inducing lipogenesis. Δ24-Dehydrocholesterol reductase (DHCR24) converts desmosterol into cholesterol, and we previously showed that the DHCR24 inhibitor SH42 increases desmosterol to activate LXR and attenuate experimental peritonitis and metabolic dysfunction-associated steatotic liver disease. Here, we aimed to evaluate the effect of SH42 on atherosclerosis development in APOE∗3-Leiden.CETP mice and low-density lipoproteins (LDL) receptor knockout mice, models for lipid- and inflammation-driven atherosclerosis, respectively. In both models, SH42 increased desmosterol without affecting plasma lipids. While reducing liver lipids in APOE∗3-Leiden.CETP mice, and regulating populations of circulating monocytes in LDL receptor knockout mice, SH42 did not attenuate atherosclerosis in either model.
Collapse
Affiliation(s)
- Xiaoke Ge
- Department of Medicine, Div. of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Bram Slütter
- Div. of BioTherapeutics, Leiden Academic Center for Drug Research, Leiden University, Leiden 2333 AL, the Netherlands
| | - Joost M. Lambooij
- Department of Parasitology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Enchen Zhou
- Department of Medicine, Div. of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Zhixiong Ying
- Department of Medicine, Div. of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Ceren Agirman
- Department of Medicine, Div. of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Marieke Heijink
- The Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Antoine Rimbert
- Nantes Université, CNRS, INSERM, l’institut du thorax, F-44000 Nantes, France
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Johan Kuiper
- Div. of BioTherapeutics, Leiden Academic Center for Drug Research, Leiden University, Leiden 2333 AL, the Netherlands
| | - Christoph Müller
- Department of Pharmacy, Center for Drug Research, Ludwig Maximilians Universität München, 80539 Munich, Germany
| | - Franz Bracher
- Department of Pharmacy, Center for Drug Research, Ludwig Maximilians Universität München, 80539 Munich, Germany
| | - Martin Giera
- The Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Sander Kooijman
- Department of Medicine, Div. of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Patrick C.N. Rensen
- Department of Medicine, Div. of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Yanan Wang
- Department of Medicine, Div. of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
- Med-X institute, Center for Immunological and Metabolic Diseases, and Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an Jiaotong University, Xi’an 710061, China
| | - Milena Schönke
- Department of Medicine, Div. of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| |
Collapse
|
5
|
Bousquet D, Nader E, Connes P, Guillot N. Liver X receptor agonist upregulates LPCAT3 in human aortic endothelial cells. Front Physiol 2024; 15:1388404. [PMID: 38694208 PMCID: PMC11061552 DOI: 10.3389/fphys.2024.1388404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/08/2024] [Indexed: 05/04/2024] Open
Abstract
Objective Endothelial cells (ECs) play an important role in tissue homeostasis. Recently, EC lipid metabolism has emerged as a regulator of EC function. The liver X receptors (LXRs) are involved in the transcriptional regulation of genes involved in lipid metabolism and have been identified as a potential target in cardiovascular disease. We aimed to decipher the role of LXRs in the regulation of lipid metabolism in human aortic endothelial cells. Approach and Results Lipid composition analysis of endothelial cells treated with the LXR agonist T0901317 revealed that LXR activation increased the proportion of polyunsaturated fatty acids (PUFAs) and decreased the proportion of saturated fatty acids. The LXR agonist decreased the uptake of fatty acids (FAs) by ECs. This effect was abolished by LXRα silencing. LXR activation increased the activity and the expression of lysophosphatidylcholine acyltransferase, LPCAT3, which is involved in the turnover of FAs at the sn-2 position of phospholipids. Transcriptomic analysis also revealed that LXRs increased the expression of key genes involved in the synthesis of PUFAs, including FA desaturase one and 2, FA elongase 5 and fatty acid synthase. Subsequently, the LXR agonist increased PUFA synthesis and enhanced arachidonic acid, eicosapentaenoic acid, and docosahexaenoic acid content in the EC phospholipids. Modification of the FA composition of ECs by LXRs led to a decrease of arachidonate and linoleate derived prostaglandins synthesis and release. No change on markers of inflammation induced by plasma from sickle cell patient were observed in presence of LXR agonist. Conclusion These results identify LXR as a key regulator of lipid metabolism in human aortic endothelial cells and a direct effect of LXR agonist on lysophosphatidylacyl transferase (LPCAT3).
Collapse
Affiliation(s)
- Delphine Bousquet
- University Lyon, LIBM EA7424, Vascular Biology and Red Blood Cell Team, Universite Lyon 1, Villeurbanne, France
- Labex GR-Ex, PRES Sorbonne, Paris, France
| | - Elie Nader
- University Lyon, LIBM EA7424, Vascular Biology and Red Blood Cell Team, Universite Lyon 1, Villeurbanne, France
- Labex GR-Ex, PRES Sorbonne, Paris, France
| | - Philippe Connes
- University Lyon, LIBM EA7424, Vascular Biology and Red Blood Cell Team, Universite Lyon 1, Villeurbanne, France
- Labex GR-Ex, PRES Sorbonne, Paris, France
| | - Nicolas Guillot
- University Lyon, LIBM EA7424, Vascular Biology and Red Blood Cell Team, Universite Lyon 1, Villeurbanne, France
- Labex GR-Ex, PRES Sorbonne, Paris, France
- INSA Lyon, Villeurbanne, France
| |
Collapse
|
6
|
Ferreira AV, Alarcon-Barrera JC, Domínguez-Andrés J, Bulut Ö, Kilic G, Debisarun PA, Röring RJ, Özhan HN, Terschlüsen E, Ziogas A, Kostidis S, Mohammed Y, Matzaraki V, Renieris G, Giamarellos-Bourboulis EJ, Netea MG, Giera M. Fatty acid desaturation and lipoxygenase pathways support trained immunity. Nat Commun 2023; 14:7385. [PMID: 37968313 PMCID: PMC10651900 DOI: 10.1038/s41467-023-43315-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 11/06/2023] [Indexed: 11/17/2023] Open
Abstract
Infections and vaccines can induce enhanced long-term responses in innate immune cells, establishing an innate immunological memory termed trained immunity. Here, we show that monocytes with a trained immunity phenotype, due to exposure to the Bacillus Calmette-Guérin (BCG) vaccine, are characterized by an increased biosynthesis of different lipid mediators (LM) derived from long-chain polyunsaturated fatty acids (PUFA). Pharmacological and genetic approaches show that long-chain PUFA synthesis and lipoxygenase-derived LM are essential for the BCG-induced trained immunity responses of human monocytes. Furthermore, products of 12-lipoxygenase activity increase in monocytes of healthy individuals after BCG vaccination. Grasping the underscoring lipid metabolic pathways contributes to our understanding of trained immunity and may help to identify therapeutic tools and targets for the modulation of innate immune responses.
Collapse
Affiliation(s)
- Anaísa V Ferreira
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, 6500HB, Nijmegen, The Netherlands.
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-313, Porto, Portugal.
| | | | - Jorge Domínguez-Andrés
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, 6500HB, Nijmegen, The Netherlands
| | - Özlem Bulut
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, 6500HB, Nijmegen, The Netherlands
| | - Gizem Kilic
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, 6500HB, Nijmegen, The Netherlands
| | - Priya A Debisarun
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, 6500HB, Nijmegen, The Netherlands
| | - Rutger J Röring
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, 6500HB, Nijmegen, The Netherlands
| | - Hatice N Özhan
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, 6500HB, Nijmegen, The Netherlands
| | - Eva Terschlüsen
- Department of Medical Microbiology, Radboud University Medical Centre, 6500HB, Nijmegen, The Netherlands
| | - Athanasios Ziogas
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, 6500HB, Nijmegen, The Netherlands
| | - Sarantos Kostidis
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333ZA, Leiden, the Netherlands
| | - Yassene Mohammed
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333ZA, Leiden, the Netherlands
| | - Vasiliki Matzaraki
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, 6500HB, Nijmegen, The Netherlands
| | - George Renieris
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | | | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, 6500HB, Nijmegen, The Netherlands
- Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115, Bonn, Germany
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333ZA, Leiden, the Netherlands.
| |
Collapse
|
7
|
Han SI, Nakakuki M, Nakagawa Y, Wang Y, Araki M, Yamamoto Y, Tokiwa H, Takeda H, Mizunoe Y, Motomura K, Ohno H, Kainoh K, Murayama Y, Aita Y, Takeuchi Y, Osaki Y, Miyamoto T, Sekiya M, Matsuzaka T, Yahagi N, Sone H, Daitoku H, Sato R, Kawano H, Shimano H. Rhomboid protease RHBDL4/RHBDD1 cleaves SREBP-1c at endoplasmic reticulum monitoring and regulating fatty acids. PNAS NEXUS 2023; 2:pgad351. [PMID: 37954160 PMCID: PMC10637267 DOI: 10.1093/pnasnexus/pgad351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 10/02/2023] [Indexed: 11/14/2023]
Abstract
The endoplasmic reticulum (ER)-embedded transcription factors, sterol regulatory element-binding proteins (SREBPs), master regulators of lipid biosynthesis, are transported to the Golgi for proteolytic activation to tune cellular cholesterol levels and regulate lipogenesis. However, mechanisms by which the cell responds to the levels of saturated or unsaturated fatty acids remain underexplored. Here, we show that RHBDL4/RHBDD1, a rhomboid family protease, directly cleaves SREBP-1c at the ER. The p97/VCP, AAA-ATPase complex then acts as an auxiliary segregase to extract the remaining ER-embedded fragment of SREBP-1c. Importantly, the enzymatic activity of RHBDL4 is enhanced by saturated fatty acids (SFAs) but inhibited by polyunsaturated fatty acids (PUFAs). Genetic deletion of RHBDL4 in mice fed on a Western diet enriched in SFAs and cholesterol prevented SREBP-1c from inducing genes for lipogenesis, particularly for synthesis and incorporation of PUFAs, and secretion of lipoproteins. The RHBDL4-SREBP-1c pathway reveals a regulatory system for monitoring fatty acid composition and maintaining cellular lipid homeostasis.
Collapse
Affiliation(s)
- Song-Iee Han
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Masanori Nakakuki
- Pharmaceutical Research Center, Mochida Pharmaceutical Co., Ltd., Gotemba, Shizuoka 412-8524, Japan
| | - Yoshimi Nakagawa
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Division of Complex Biosystem Research, Department of Research and Development, Institute of Natural Medicine, University of Toyama, Toyama, Toyama 930-0194, Japan
| | - Yunong Wang
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Masaya Araki
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuta Yamamoto
- Department of Chemistry, Rikkyo University, Toshima-ku, Tokyo 171-8501, Japan
| | - Hiroaki Tokiwa
- Laboratory of Organic Chemistry, Gifu Pharmaceutical University, Daigaku-Nishi, Gifu 501-1196, Japan
| | - Hiroyuki Takeda
- Division of Proteo Drug Discovery Sciences, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Yuhei Mizunoe
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Kaori Motomura
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Hiroshi Ohno
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Kenta Kainoh
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuki Murayama
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuichi Aita
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yoshinori Takeuchi
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yoshinori Osaki
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Takafumi Miyamoto
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan
| | - Motohiro Sekiya
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Takashi Matsuzaka
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan
| | - Naoya Yahagi
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Hirohito Sone
- Department of Internal Medicine, Faculty of Medicine, Niigata University, Niigata, Niigata 951-8510, Japan
| | - Hiroaki Daitoku
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan
| | - Ryuichiro Sato
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, Nutri-Life Science Laboratory, The University of Tokyo, Tokyo 113-8657, Japan
| | - Hiroyuki Kawano
- Pharmaceutical Research Center, Mochida Pharmaceutical Co., Ltd., Gotemba, Shizuoka 412-8524, Japan
| | - Hitoshi Shimano
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan
- Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
8
|
Taskinen JH, Ruhanen H, Matysik S, Käkelä R, Olkkonen VM. Systemwide effects of ER-intracellular membrane contact site disturbance in primary endothelial cells. J Steroid Biochem Mol Biol 2023; 232:106349. [PMID: 37321512 DOI: 10.1016/j.jsbmb.2023.106349] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/09/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023]
Abstract
Membrane contact sites (MCS) make up a crucial route of inter-organelle non-vesicular transport within the cell. Multiple proteins are involved in this process, which includes the ER-resident proteins vesicle associated membrane protein associated protein A and -B (VAPA/B) that form MCS between the ER and other membrane compartments. Currently most functional data on VAP depleted phenotypes have shown alterations in lipid homeostasis, induction of ER stress, dysfunction of UPR and autophagy, as well as neurodegeneration. Literature on concurrent silencing of VAPA/B is still sparse; therefore, we investigated how it affects the macromolecule pools of primary endothelial cells. Our transcriptomics results showed significant upregulation in genes related to inflammation, ER and Golgi dysfunction, ER stress, cell adhesion, as well as Coat Protein Complex-I and -II (COP-I, COP-II) vesicle transport. Genes related to cellular division were downregulated, as well as key genes of lipid and sterol biosynthesis. Lipidomics analyses revealed reductions in cholesteryl esters, very long chain highly unsaturated and saturated lipids, whereas increases in free cholesterol and relatively short chain unsaturated lipids were evident. Furthermore, the knockdown resulted in an inhibition of angiogenesis in vitro. We speculate that ER MCS depletion has led to multifaceted outcomes, which include elevated ER free cholesterol content and ER stress, alterations in lipid metabolism, ER-Golgi function and vesicle transport, which have led to a reduction in angiogenesis. The silencing also induced an inflammatory response, consistent with upregulation of markers of early atherogenesis. To conclude, ER MCS mediated by VAPA/B play a crucial role in maintaining cholesterol traffic and sustain normal endothelial functions.
Collapse
Affiliation(s)
- Juuso H Taskinen
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290 Helsinki, Finland
| | - Hanna Ruhanen
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, Viikinkaari 1, PO BOX 65, 00014 University of Helsinki, Finland; Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, University of Helsinki, Viikinkaari 1, PO BOX 65, 00014 University of Helsinki, Finland
| | - Silke Matysik
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Reijo Käkelä
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, Viikinkaari 1, PO BOX 65, 00014 University of Helsinki, Finland; Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, University of Helsinki, Viikinkaari 1, PO BOX 65, 00014 University of Helsinki, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290 Helsinki, Finland; Department of Anatomy, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland.
| |
Collapse
|
9
|
Liu R, Scimeca M, Sun Q, Melino G, Mauriello A, Shao C, Shi Y, Piacentini M, Tisone G, Agostini M. Harnessing metabolism of hepatic macrophages to aid liver regeneration. Cell Death Dis 2023; 14:574. [PMID: 37644019 PMCID: PMC10465526 DOI: 10.1038/s41419-023-06066-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/31/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023]
Abstract
Liver regeneration is a dynamic and regulated process that involves inflammation, granulation, and tissue remodeling. Hepatic macrophages, abundantly distributed in the liver, are essential components that actively participate in each step to orchestrate liver regeneration. In the homeostatic liver, resident macrophages (Kupffer cells) acquire a tolerogenic phenotype and contribute to immunological tolerance. Following toxicity-induced damage or physical resection, Kupffer cells as well as monocyte-derived macrophages can be activated and promote an inflammatory process that supports the survival and activation of hepatic myofibroblasts and thus promotes scar tissue formation. Subsequently, these macrophages, in turn, exhibit the anti-inflammatory effects critical to extracellular matrix remodeling during the resolution stage. However, continuous damage-induced chronic inflammation generally leads to hepatic macrophage dysfunction, which exacerbates hepatocellular injury and triggers further liver fibrosis and even cirrhosis. Emerging macrophage-targeting strategies have shown efficacy in both preclinical and clinical studies. Increasing evidence indicates that metabolic rewiring provides substrates for epigenetic modification, which endows monocytes/macrophages with prolonged "innate immune memory". Therefore, it is reasonable to conceive novel therapeutic strategies for metabolically reprogramming macrophages and thus mediate a homeostatic or reparative process for hepatic inflammation management and liver regeneration.
Collapse
Affiliation(s)
- Rui Liu
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Manuel Scimeca
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Qiang Sun
- Institute of Biotechnology, Academy of Military Medical Science; Research Unit of Cell Death Mechanism, 2021RU008, Chinese Academy of Medical Science, 100071, Beijing, China
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Suzhou Medical College of Soochow University, 215123, Suzhou, Jiangsu, China
| | - Yufang Shi
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, 215123, Suzhou, China.
| | - Mauro Piacentini
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
| | - Giuseppe Tisone
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
| | - Massimiliano Agostini
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
| |
Collapse
|
10
|
Chen W, Li T, Du S, Chen H, Wang Q. Microalgal polyunsaturated fatty acids: Hotspots and production techniques. Front Bioeng Biotechnol 2023; 11:1146881. [PMID: 37064250 PMCID: PMC10102661 DOI: 10.3389/fbioe.2023.1146881] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023] Open
Abstract
Algae play a crucial role in the earth’s primary productivity by producing not only oxygen but also a variety of high-value nutrients. One such nutrient is polyunsaturated fatty acids (PUFAs), which are accumulated in many algae and can be consumed by animals through the food chain and eventually by humans. Omega-3 and omega-6 PUFAs are essential nutrients for human and animal health. However, compared with plants and aquatic sourced PUFA, the production of PUFA-rich oil from microalgae is still in the early stages of exploration. This study has collected recent reports on algae-based PUFA production and analyzed related research hotspots and directions, including algae cultivation, lipids extraction, lipids purification, and PUFA enrichment processes. The entire technological process for the extraction, purification and enrichment of PUFA oils from algae is systemically summarized in this review, providing important guidance and technical reference for scientific research and industrialization of algae-based PUFA production.
Collapse
Affiliation(s)
- Weixian Chen
- State Key Laboratory of Crop Stress Adaptation and Improvement, School of Life Sciences, Henan University, Kaifeng, China
| | - Tianpei Li
- State Key Laboratory of Crop Stress Adaptation and Improvement, School of Life Sciences, Henan University, Kaifeng, China
| | - Shuwen Du
- School of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Hui Chen
- State Key Laboratory of Crop Stress Adaptation and Improvement, School of Life Sciences, Henan University, Kaifeng, China
| | - Qiang Wang
- State Key Laboratory of Crop Stress Adaptation and Improvement, School of Life Sciences, Henan University, Kaifeng, China
- Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, China
- *Correspondence: Qiang Wang,
| |
Collapse
|
11
|
Zhang W, Zhang C, Luo J, Xu H, Liu J, Loor JJ, Shi H. The LXRB-SREBP1 network regulates lipogenic homeostasis by controlling the synthesis of polyunsaturated fatty acids in goat mammary epithelial cells. J Anim Sci Biotechnol 2022; 13:120. [PMID: 36336695 PMCID: PMC9639257 DOI: 10.1186/s40104-022-00774-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 09/05/2022] [Indexed: 11/09/2022] Open
Abstract
Background In rodents, research has revealed a role of liver X receptors (LXR) in controlling lipid homeostasis and regulating the synthesis of polyunsaturated fatty acids (PUFA). Recent data suggest that LXRB is the predominant LXR subtype in ruminant mammary cells, but its role in lipid metabolism is unknown. It was hypothesized that LXRB plays a role in lipid homeostasis via altering the synthesis of PUFA in the ruminant mammary gland. We used overexpression and knockdown of LXRB in goat primary mammary epithelial cells (GMEC) to evaluate abundance of lipogenic enzymes, fatty acid profiles, content of lipid stores and activity of the stearoyl-CoA desaturase (SCD1) promoter. Results Overexpression of LXRB markedly upregulated the protein abundance of LXRB while incubation with siRNA targeting LXRB markedly decreased abundance of LXRB protein. Overexpression of LXRB plus T0901317 (T09, a ligand for LXR) dramatically upregulated SCD1 and elongation of very long chain fatty acid-like fatty acid elongases 5–7 (ELOVL 5–7), which are related to PUFA synthesis. Compared with the control, cells overexpressing LXRB and stimulated with T09 had greater concentrations of C16:0, 16:1, 18:1n7,18:1n9 and C18:2 as well as desaturation and elongation indices of C16:0. Furthermore, LXRB-overexpressing cells incubated with T09 had greater levels of triacylglycerol and cholesterol. Knockdown of LXRB in cells incubated with T09 led to downregulation of genes encoding elongases and desaturases. Knockdown of LXRB attenuated the increase in triacylglycerol and cholesterol that was induced by T09. In cells treated with dimethylsulfoxide, knockdown of LXRB increased the concentration of C16:0 at the expense of C18:0, while a significant decrease in C18:2 was observed in cells incubated with both siLXRB and T09. The abundance of sterol regulatory element binding transcription factor 1 precursor (pSREBP1) and its mature fragment (nSREBP1) was upregulated by T09, but not LXRB overexpression. In the cells cultured with T09, knockdown of LXRB downregulated the abundance for pSREBP1 and nSREBP1. Luciferase reporter assays revealed that the activities of wild type SCD1 promoter or fragment with SREBP1 response element (SRE) mutation were decreased markedly when LXRB was knocked down. Activity of the SCD1 promoter that was induced by T09 was blocked when the SRE mutation was introduced. Conclusion The current study provides evidence of a physiological link between the LXRB and SREBP1 in the ruminant mammary cell. An important role was revealed for the LXRB-SREBP1 network in the synthesis of PUFA via the regulation of genes encoding elongases and desaturases. Thus, targeting this network might elicit broad effects on lipid homeostasis in ruminant mammary gland. Supplementary Information The online version contains supplementary material available at 10.1186/s40104-022-00774-4.
Collapse
Affiliation(s)
- Wenying Zhang
- grid.13402.340000 0004 1759 700XInstitute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058 China
| | - Changhui Zhang
- grid.144022.10000 0004 1760 4150College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 China
| | - Jun Luo
- grid.144022.10000 0004 1760 4150College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 China
| | - Huifen Xu
- grid.108266.b0000 0004 1803 0494College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046 China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, 310058, China
| | - Jianxin Liu
- grid.13402.340000 0004 1759 700XInstitute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058 China
| | - Juan J. Loor
- grid.35403.310000 0004 1936 9991Mammalian Nutrition Physiology Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801 USA
| | - Hengbo Shi
- grid.13402.340000 0004 1759 700XInstitute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058 China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, 310058, China
| |
Collapse
|
12
|
Saito S, Ohashi H, Nakamura K, Otagaki J, Nishioka K, Nishiuchi K, Nakamura A, Tsurukawa Y, Shibasaki H, Murakami H, Nagane M, Okada M, Kuramochi K, Watashi K, Kamisuki S. Cyclic Phthalate Esters as Liver X Receptor Antagonists with Anti-hepatitis C Virus and Anti-severe Acute Respiratory Syndrome Coronavirus 2 Properties. Chem Pharm Bull (Tokyo) 2022; 70:679-683. [PMID: 36184450 DOI: 10.1248/cpb.c22-00345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The liver X receptor is a nuclear hormone receptor that regulates lipid metabolism. Previously, we had demonstrated the antiviral properties of a liver X receptor antagonist associated with the hepatitis C virus and severe acute respiratory syndrome coronavirus 2. In this study, we screened a chemical library and identified two potential liver X receptor antagonists. Spectroscopic analysis revealed that the structures of both antagonists (compounds 1 and 2) were cyclic dimer and trimer of esters, respectively, that consisted of phthalate and 1,6-hexane diol. This study is the first to report the structure of the cyclic trimer of phthalate ester. Further experiments revealed that the compounds were impurities of solvents used for purification, although their source could not be traced. Both phthalate esters exhibited anti-hepatitis C virus activity, whereas the cyclic dimer showed anti-severe acute respiratory syndrome coronavirus 2 activity. Cyclic phthalate derivatives may constitute a novel class of liver X receptor antagonists and broad-spectrum antivirals.
Collapse
Affiliation(s)
- Shiki Saito
- School of Veterinary Medicine, Azabu University
| | - Hirofumi Ohashi
- Department of Applied Biological Science, Tokyo University of Science.,Department of Virology II, National Institute of Infectious Diseases.,Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases
| | | | | | - Kazane Nishioka
- Department of Applied Biological Science, Tokyo University of Science.,Department of Virology II, National Institute of Infectious Diseases
| | - Kota Nishiuchi
- Department of Applied Biological Science, Tokyo University of Science
| | | | | | | | - Hironobu Murakami
- School of Veterinary Medicine, Azabu University.,Center for Human and Animal Symbiosis Science, Azabu University
| | - Masaki Nagane
- School of Veterinary Medicine, Azabu University.,Center for Human and Animal Symbiosis Science, Azabu University
| | - Maiko Okada
- School of Bioscience and Biotechnology, Tokyo University of Technology
| | - Kouji Kuramochi
- Department of Applied Biological Science, Tokyo University of Science
| | - Koichi Watashi
- Department of Applied Biological Science, Tokyo University of Science.,Department of Virology II, National Institute of Infectious Diseases.,Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases
| | - Shinji Kamisuki
- School of Veterinary Medicine, Azabu University.,Center for Human and Animal Symbiosis Science, Azabu University
| |
Collapse
|
13
|
Zhu L, An J, Chinnarasu S, Luu T, Pettway YD, Fahey K, Litts B, Kim HYH, Flynn CR, Linton MF, Stafford JM. Expressing the Human Cholesteryl Ester Transfer Protein Minigene Improves Diet-Induced Fatty Liver and Insulin Resistance in Female Mice. Front Physiol 2022; 12:799096. [PMID: 35082691 PMCID: PMC8784660 DOI: 10.3389/fphys.2021.799096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/16/2021] [Indexed: 01/22/2023] Open
Abstract
Mounting evidence has shown that CETP has important physiological roles in adapting to chronic nutrient excess, specifically, to protect against diet-induced insulin resistance. However, the underlying mechanisms for the protective roles of CETP in metabolism are not yet clear. Mice naturally lack CETP expression. We used transgenic mice with a human CETP minigene (huCETP) controlled by its natural flanking region to further understand CETP-related physiology in response to obesity. Female huCETP mice and their wild-type littermates were fed a high-fat diet for 6 months. Blood lipid profile and liver lipid metabolism were studied. Insulin sensitivity was analyzed with euglycemic-hyperinsulinemic clamp studies combined with 3H-glucose tracer techniques. While high-fat diet feeding induced obesity for huCETP mice and their wild-type littermates lacking CETP expression, insulin sensitivity was higher for female huCETP mice than for their wild-type littermates. There was no difference in insulin sensitivity for male huCETP mice vs. littermates. The increased insulin sensitivity in females was largely caused by the better insulin-mediated suppression of hepatic glucose production. In huCETP females, CETP in the circulation decreased HDL-cholesterol content and increased liver cholesterol uptake and liver cholesterol and oxysterol contents, which was associated with the upregulation of LXR target genes in long-chain polyunsaturated fatty acid biosynthesis and PPARα target genes in fatty acid β-oxidation in the liver. The upregulated fatty acid β-oxidation may account for the improved fatty liver and liver insulin action in female huCETP mice. This study provides further evidence that CETP has beneficial physiological roles in the metabolic adaptation to nutrient excess by promoting liver fatty acid oxidation and hepatic insulin sensitivity, particularly for females.
Collapse
Affiliation(s)
- Lin Zhu
- VA Tennessee Valley Healthcare System, Nashville, TN, United States
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Julia An
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Sivaprakasam Chinnarasu
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Thao Luu
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Yasminye D. Pettway
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Kelly Fahey
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Bridget Litts
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Hye-Young H. Kim
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
| | - Charles R. Flynn
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - MacRae F. Linton
- Atherosclerosis Research Unit, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - John M. Stafford
- VA Tennessee Valley Healthcare System, Nashville, TN, United States
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University School of Medicine, Nashville, TN, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
14
|
Laval T, Pedró-Cos L, Malaga W, Guenin-Macé L, Pawlik A, Mayau V, Yahia-Cherbal H, Delos O, Frigui W, Bertrand-Michel J, Guilhot C, Demangel C. De novo synthesized polyunsaturated fatty acids operate as both host immunomodulators and nutrients for Mycobacterium tuberculosis. eLife 2021; 10:71946. [PMID: 34951591 PMCID: PMC8752091 DOI: 10.7554/elife.71946] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/24/2021] [Indexed: 11/30/2022] Open
Abstract
Successful control of Mycobacterium tuberculosis (Mtb) infection by macrophages relies on immunometabolic reprogramming, where the role of fatty acids (FAs) remains poorly understood. Recent studies unraveled Mtb’s capacity to acquire saturated and monounsaturated FAs via the Mce1 importer. However, upon activation, macrophages produce polyunsaturated fatty acids (PUFAs), mammal-specific FAs mediating the generation of immunomodulatory eicosanoids. Here, we asked how Mtb modulates de novo synthesis of PUFAs in primary mouse macrophages and whether this benefits host or pathogen. Quantitative lipidomics revealed that Mtb infection selectively activates the biosynthesis of ω6 PUFAs upstream of the eicosanoid precursor arachidonic acid (AA) via transcriptional activation of Fads2. Inhibiting FADS2 in infected macrophages impaired their inflammatory and antimicrobial responses but had no effect on Mtb growth in host cells nor mice. Using a click-chemistry approach, we found that Mtb efficiently imports ω6 PUFAs via Mce1 in axenic culture, including AA. Further, Mtb preferentially internalized AA over all other FAs within infected macrophages by mechanisms partially depending on Mce1 and supporting intracellular persistence. Notably, IFNγ repressed de novo synthesis of AA by infected mouse macrophages and restricted AA import by intracellular Mtb. Together, these findings identify AA as a major FA substrate for intracellular Mtb, whose mobilization by innate immune responses is opportunistically hijacked by the pathogen and downregulated by IFNγ.
Collapse
Affiliation(s)
- Thomas Laval
- Immunobiology of Infection Unit, Institut Pasteur, Paris, France
| | - Laura Pedró-Cos
- Immunobiology of Infection Unit, Institut Pasteur, Paris, France
| | - Wladimir Malaga
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS UMR5089, Toulouse, France
| | | | - Alexandre Pawlik
- Integrated Mycobacterial Pathogenomics Unit, Institut Pasteur, Paris, France
| | - Véronique Mayau
- Immunobiology of Infection Unit, Institut Pasteur, Paris, France
| | | | | | - Wafa Frigui
- Integrated Mycobacterial Pathogenomics Unit, Institut Pasteur, Paris, France
| | | | - Christophe Guilhot
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS UMR5089, Toulouse, France
| | | |
Collapse
|
15
|
Xie D, Chen C, Dong Y, You C, Wang S, Monroig Ó, Tocher DR, Li Y. Regulation of long-chain polyunsaturated fatty acid biosynthesis in teleost fish. Prog Lipid Res 2021; 82:101095. [PMID: 33741387 DOI: 10.1016/j.plipres.2021.101095] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/24/2021] [Accepted: 03/12/2021] [Indexed: 12/26/2022]
Abstract
Omega-3 (n-3) long-chain polyunsaturated fatty acids (LC-PUFA, C20-24), including eicosapentaenoic acid (EPA, 20:5n-3) and docosahexaenoic acid (DHA, 22:6n-3), are involved in numerous biological processes and have a range of health benefits. Fish have long been considered as the main source of n-3 LC-PUFA in human diets. However, the capacity for endogenous biosynthesis of LC-PUFA from C18 PUFA varies in fish species based on the presence, expression and activity of key enzymes including fatty acyl desaturases (Fads) and elongation of very long-chain fatty acids (Elovl) proteins. In this article, we review progress on the identified Fads and Elovl, as well as the regulatory mechanisms of LC-PUFA biosynthesis both at transcriptional and post-transcriptional levels in teleosts. The most comprehensive advances have been obtained in rabbitfish Siganus canaliculatus, a marine teleost demonstrated to have the entire pathway for LC-PUFA biosynthesis, including the roles of transcription factors hepatocyte nuclear factor 4α (Hnf4α), liver X receptor alpha (Lxrα), sterol regulatory element-binding protein 1 (Srebp-1), peroxisome proliferator-activated receptor gamma (Pparγ) and stimulatory protein 1 (Sp1), as well as post-transcriptional regulation by individual microRNA (miRNA) or clusters. This research has, for the first time, demonstrated the involvement of Hnf4α, Pparγ and miRNA in the regulation of LC-PUFA biosynthesis in vertebrates. The present review provides readers with a relatively comprehensive overview of the progress made into understanding LC-PUFA biosynthetic systems in teleosts, and some insights into improving endogenous LC-PUFA biosynthesis capacity aimed at reducing the dependence of aquafeeds on fish oil while maintaining or increasing flesh LC-PUFA content and the nutritional quality of farmed fish.
Collapse
Affiliation(s)
- Dizhi Xie
- College of Marine Sciences of South China Agricultural University & Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Cuiying Chen
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China
| | - Yewei Dong
- Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510642, China
| | - Cuihong You
- Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510642, China
| | - Shuqi Wang
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China.
| | - Óscar Monroig
- Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas (IATS-CSIC), 12595 Castellón, Spain.
| | - Douglas R Tocher
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China; Institute of Aquaculture, Faculty of Natural Sciences, University of Stirling, Stirling FK94LA, Scotland, United Kingdom
| | - Yuanyou Li
- College of Marine Sciences of South China Agricultural University & Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China.
| |
Collapse
|
16
|
Russo-Savage L, Schulman IG. Liver X receptors and liver physiology. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166121. [PMID: 33713792 DOI: 10.1016/j.bbadis.2021.166121] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/29/2022]
Abstract
The liver x receptors LXRα (NR1H3) and LXRβ (NR1H2) are members of the nuclear hormone receptor superfamily of ligand dependent transcription factors that regulate transcription in response to the direct binding of cholesterol derivatives. Studies using genetic knockouts and synthetic ligands have defined the LXRs as important modulators of lipid homeostasis throughout the body. This review focuses on the control of cholesterol and fatty acid metabolism by LXRs in the liver and how modifying LXR activity can influence the pathology of liver diseases.
Collapse
Affiliation(s)
- Lillian Russo-Savage
- Department of Pharmacology, University of Virginia, School of Medicine, United States of America
| | - Ira G Schulman
- Department of Pharmacology, University of Virginia, School of Medicine, United States of America.
| |
Collapse
|
17
|
Sun L, Yang X, Huang X, Yao Y, Wei X, Yang S, Zhou D, Zhang W, Long Z, Xu X, Zhu X, He S, Su X. 2-Hydroxylation of Fatty Acids Represses Colorectal Tumorigenesis and Metastasis via the YAP Transcriptional Axis. Cancer Res 2020; 81:289-302. [PMID: 33203703 DOI: 10.1158/0008-5472.can-20-1517] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/16/2020] [Accepted: 11/12/2020] [Indexed: 11/16/2022]
Abstract
Alteration in lipid composition is an important metabolic adaptation by cancer cells to support tumorigenesis and metastasis. Fatty acid 2-hydroxylase (FA2H) introduces a chiral hydroxyl group at the second carbon of fatty acid (FA) backbones and influences lipid structures and metabolic signaling. However, the underlying mechanisms through which FA 2-hydroxylation is coupled to metabolic adaptation and tumor growth remain elusive. Here, we show that FA2H regulates specific metabolic reprogramming and oncogenic signaling in the development of colorectal cancer. FA2H is highly expressed in normal colorectal tissues. Assessments through deciphering both published high-throughput data and curated human colorectal cancer samples revealed significant suppression of FA2H in tumors, which is correlated with unfavorable prognosis. Experiments with multiple models of genetic manipulation or treatment with an enzymatic product of FA2H, (R)-2-hydroxy palmitic acid, demonstrated that FA 2-hydroxylation inhibits colorectal cancer cell proliferation, migration, epithelial-to-mesenchymal transition progression, and tumor growth. Bioinformatics analysis suggested that FA2H functions through AMP-activated protein kinase/Yes-associated protein (AMPK/YAP) pathway, which was confirmed in colorectal cancer cells, as well as in tumors. Lipidomics analysis revealed an accumulation of polyunsaturated fatty acids in cells with FA2H overexpression, which may contribute to the observed nutrient deficiency and AMPK activation. Collectively, these data demonstrate that FA 2-hydroxylation initiates a metabolic signaling cascade to suppress colorectal tumor growth and metastasis via the YAP transcriptional axis and provides a strategy to improve colorectal cancer treatment. SIGNIFICANCE: These findings identify a novel metabolic mechanism regulating the tumor suppressor function of FA 2-hydroxylation in colorectal cancer.
Collapse
Affiliation(s)
- Liang Sun
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu, P.R. China.,Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Xiaoqin Yang
- Department of Bioinformatics, Soochow University Medical College, Suzhou, Jiangsu, P.R. China
| | - Xiaoheng Huang
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu, P.R. China
| | - Yizhou Yao
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu, P.R. China.,Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Xiangyu Wei
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu, P.R. China
| | - Shugao Yang
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu, P.R. China
| | - Diyuan Zhou
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu, P.R. China.,Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Wei Zhang
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri
| | - Zhimin Long
- Shanghai Sciex Analytical Instrument Trading Co., Shanghai, P.R. China
| | - Xiaoyan Xu
- Shanghai Sciex Analytical Instrument Trading Co., Shanghai, P.R. China
| | - Xinguo Zhu
- Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China.
| | - Songbing He
- Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Xiong Su
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu, P.R. China. .,Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
18
|
Snodgrass RG, Benatzy Y, Schmid T, Namgaladze D, Mainka M, Schebb NH, Lütjohann D, Brüne B. Efferocytosis potentiates the expression of arachidonate 15-lipoxygenase (ALOX15) in alternatively activated human macrophages through LXR activation. Cell Death Differ 2020; 28:1301-1316. [PMID: 33177619 PMCID: PMC8027700 DOI: 10.1038/s41418-020-00652-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 10/09/2020] [Accepted: 10/15/2020] [Indexed: 12/31/2022] Open
Abstract
Macrophages acquire anti-inflammatory and proresolving functions to facilitate resolution of inflammation and promote tissue repair. While alternatively activated macrophages (AAMs), also referred to as M2 macrophages, polarized by type 2 (Th2) cytokines IL-4 or IL-13 contribute to the suppression of inflammatory responses and play a pivotal role in wound healing, contemporaneous exposure to apoptotic cells (ACs) potentiates the expression of anti-inflammatory and tissue repair genes. Given that liver X receptors (LXRs), which coordinate sterol metabolism and immune cell function, play an essential role in the clearance of ACs, we investigated whether LXR activation following engulfment of ACs selectively potentiates the expression of Th2 cytokine-dependent genes in primary human AAMs. We show that AC uptake simultaneously upregulates LXR-dependent, but suppresses SREBP-2-dependent gene expression in macrophages, which are both prevented by inhibiting Niemann–Pick C1 (NPC1)-mediated sterol transport from lysosomes. Concurrently, macrophages accumulate sterol biosynthetic intermediates desmosterol, lathosterol, lanosterol, and dihydrolanosterol but not cholesterol-derived oxysterols. Using global transcriptome analysis, we identify anti-inflammatory and proresolving genes including interleukin-1 receptor antagonist (IL1RN) and arachidonate 15-lipoxygenase (ALOX15) whose expression are selectively potentiated in macrophages upon concomitant exposure to ACs or LXR agonist T0901317 (T09) and Th2 cytokines. We show priming macrophages via LXR activation enhances the cellular capacity to synthesize inflammation-suppressing specialized proresolving mediator (SPM) precursors 15-HETE and 17-HDHA as well as resolvin D5. Silencing LXRα and LXRβ in macrophages attenuates the potentiation of ALOX15 expression by concomitant stimulation of ACs or T09 and IL-13. Collectively, we identify a previously unrecognized mechanism of regulation whereby LXR integrates AC uptake to selectively shape Th2-dependent gene expression in AAMs.
Collapse
Affiliation(s)
- Ryan G Snodgrass
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Yvonne Benatzy
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Tobias Schmid
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Dmitry Namgaladze
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Malwina Mainka
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Nils Helge Schebb
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Dieter Lütjohann
- Institute for Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| | - Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany.
| |
Collapse
|
19
|
He P, Gelissen IC, Ammit AJ. Regulation of ATP binding cassette transporter A1 (ABCA1) expression: cholesterol-dependent and - independent signaling pathways with relevance to inflammatory lung disease. Respir Res 2020; 21:250. [PMID: 32977800 PMCID: PMC7519545 DOI: 10.1186/s12931-020-01515-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022] Open
Abstract
The role of the ATP binding cassette transporter A1 (ABCA1) in maintaining cellular lipid homeostasis in cardiovascular disease is well established. More recently, the important beneficial role played by ABCA1 in modulating pathogenic disease mechanisms, such as inflammation, in a broad range of chronic conditions has been realised. These studies position ABCA1 as a potential therapeutic target in a diverse range of diseases where inflammation is an underlying cause. Chronic respiratory conditions such as asthma and chronic obstructive pulmonary disease (COPD) are driven by inflammation, and as such, there is now a growing recognition that we need a greater understanding of the signaling pathways responsible for regulation of ABCA1 expression in this clinical context. While the signaling pathways responsible for cholesterol-mediated ABCA1 expression have been clearly delineated through decades of studies in the atherosclerosis field, and thus far appear to be translatable to the respiratory field, less is known about the cholesterol-independent signaling pathways that can modulate ABCA1 expression in inflammatory lung disease. This review will identify the various signaling pathways and ligands that are associated with the regulation of ABCA1 expression and may be exploited in future as therapeutic targets in the setting of chronic inflammatory lung diseases.
Collapse
Affiliation(s)
- Patrick He
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Ingrid C Gelissen
- Sydney Pharmacy School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| | - Alaina J Ammit
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia.
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
20
|
Li T, Yin J, Ji Y, Lin P, Li Y, Yang Z, Hu S, Wang J, Zhang B, Koshti S, Wang J, Ji C, Guo S. Setosphapyrone C and D accelerate macrophages cholesterol efflux by promoting LXRα/ABCA1 pathway. Arch Pharm Res 2020; 43:788-797. [PMID: 32779151 DOI: 10.1007/s12272-020-01255-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 07/22/2020] [Indexed: 02/08/2023]
Abstract
LXRα agonists have attracted significant attention due to their potential biological activities on promoting cholesterol efflux. This study was designed to investigate whether setosphapyrone C and D have potential lipid-lowering capacity and the underlying mechanisms in vitro. Our data showed that setosphapyrone C and D had weak cytotoxicity compared to the liver X receptor α (LXRα) agonist T0901317. In RAW 264.7 macrophages, setosphapyrone C and D significantly enhanced [3H]-cholesterol efflux by ~ 21.3% and 32.4%, respectively; furthermore, setosphapyrone C and D enhanced the protein levels of ATP-binding cassette transporter (ABC) A1 and LXRα by 58% and 69%, and 60% and 70% (8 µM), respectively; however, they had no effect on the protein levels of ABCG1 and scavenger receptor B type 1; additionally, they had minor effect on the mRNA expression of lipogenic genes. Of note, setosphapyrone C and D significantly enhanced LXRα/ABCA1pathway in mice primary macrophages. In BRL cells, setosphapyrone C and D significantly improved the protein levels of ABCA1 and ABCG1; setosphapyrone D significantly enhanced the protein expression of low-density lipoprotein. Collectively, setosphapyrone C and D with weak cytotoxicity exhibited effective lipid-lowering effect via enhancing LXRα/ABC pathways. Setosphapyrones possess potential application for the treatment of hyperlipidemic diseases.
Collapse
Affiliation(s)
- Ting Li
- College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, 150076, Harbin, China.,Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China
| | - Jiayu Yin
- College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, 150076, Harbin, China.,Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China
| | - Yubin Ji
- College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, 150076, Harbin, China
| | - Ping Lin
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China
| | - Yanjie Li
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China
| | - Zixun Yang
- College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, 150076, Harbin, China.,Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China
| | - Shumei Hu
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China
| | - Jin Wang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China
| | - Baihui Zhang
- College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, 150076, Harbin, China.,Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China
| | - Saloni Koshti
- Department of Physiology, University of Alberta, T6G2R3, Edmonton, Canada
| | - Junfeng Wang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica/RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510220, Guangzhou, China.
| | - Chenfeng Ji
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China.
| | - Shoudong Guo
- College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, 150076, Harbin, China. .,Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China.
| |
Collapse
|
21
|
Chen C, Wang S, Hu Y, Zhang M, He X, You C, Wen X, Monroig Ó, Tocher DR, Li Y. miR-26a mediates LC-PUFA biosynthesis by targeting the Lxrα-Srebp1 pathway in the marine teleost Siganus canaliculatus. J Biol Chem 2020; 295:13875-13886. [PMID: 32759307 DOI: 10.1074/jbc.ra120.014858] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/30/2020] [Indexed: 11/06/2022] Open
Abstract
MicroRNAs have been recently shown to be important regulators of lipid metabolism. However, the mechanisms of microRNA-mediated regulation of long-chain polyunsaturated fatty acid (LC-PUFA) biosynthesis in vertebrates remain largely unknown. Herein, we for the first time addressed the role of miR-26a in LC-PUFA biosynthesis in the marine rabbitfish Siganus canaliculatus The results showed that miR-26a was significantly down-regulated in liver of rabbitfish reared in brackish water and in S. canaliculatus hepatocyte line (SCHL) incubated with the LC-PUFA precursor α-linolenic acid, suggesting that miR-26a may be involved in LC-PUFA biosynthesis because of its abundance being regulated by factors affecting LC-PUFA biosynthesis. Opposite patterns were observed in the expression of liver X receptor α (lxrα) and sterol regulatory element-binding protein-1 (srebp1), as well as the LC-PUFA biosynthesis-related genes (Δ4 fads2, Δ6Δ5 fads2, and elovl5) in SCHL cells incubated with α-linolenic acid. Luciferase reporter assays revealed rabbitfish lxrα as a target of miR-26a, and overexpression of miR-26a in SCHL cells markedly reduced protein levels of Lxrα, Srebp1, and Δ6Δ5 Fads2 induced by the agonist T0901317. Moreover, increasing endogenous Lxrα by knockdown of miR-26a facilitated Srebp1 activation and concomitant increased expression of genes involved in LC-PUFA biosynthesis and consequently promoted LC-PUFA biosynthesis both in vitro and in vivo These results indicate a critical role of miR-26a in regulating LC-PUFA biosynthesis through targeting the Lxrα-Srebp1 pathway and provide new insights into the regulatory network controlling LC-PUFA biosynthesis and accumulation in vertebrates.
Collapse
Affiliation(s)
- Cuiying Chen
- Guangdong Provincial Key Laboratory of Marine Biotechnology and Research Center for Nutrition, Feed and Healthy Breeding of Aquatic Animals of Guangdong Province, Shantou University, Shantou, China
| | - Shuqi Wang
- Guangdong Provincial Key Laboratory of Marine Biotechnology and Research Center for Nutrition, Feed and Healthy Breeding of Aquatic Animals of Guangdong Province, Shantou University, Shantou, China
| | - Yu Hu
- Guangdong Provincial Key Laboratory of Marine Biotechnology and Research Center for Nutrition, Feed and Healthy Breeding of Aquatic Animals of Guangdong Province, Shantou University, Shantou, China
| | - Mei Zhang
- Guangdong Provincial Key Laboratory of Marine Biotechnology and Research Center for Nutrition, Feed and Healthy Breeding of Aquatic Animals of Guangdong Province, Shantou University, Shantou, China
| | - Xianda He
- Guangdong Provincial Key Laboratory of Marine Biotechnology and Research Center for Nutrition, Feed and Healthy Breeding of Aquatic Animals of Guangdong Province, Shantou University, Shantou, China
| | - Cuihong You
- Guangdong Provincial Key Laboratory of Marine Biotechnology and Research Center for Nutrition, Feed and Healthy Breeding of Aquatic Animals of Guangdong Province, Shantou University, Shantou, China
| | - Xiaobo Wen
- College of Marine Sciences of South China Agricultural University and Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Óscar Monroig
- Instituto de Acuicultura Torre de la Sal-Consejo Superior de Investigaciones Científicas, Castellón, Spain
| | - Douglas R Tocher
- Institute of Aquaculture, Faculty of Natural Sciences, University of Stirling, Stirling, Scotland, United Kingdom
| | - Yuanyou Li
- College of Marine Sciences of South China Agricultural University and Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China.
| |
Collapse
|
22
|
Dahik VD, Frisdal E, Le Goff W. Rewiring of Lipid Metabolism in Adipose Tissue Macrophages in Obesity: Impact on Insulin Resistance and Type 2 Diabetes. Int J Mol Sci 2020; 21:ijms21155505. [PMID: 32752107 PMCID: PMC7432680 DOI: 10.3390/ijms21155505] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022] Open
Abstract
Obesity and its two major comorbidities, insulin resistance and type 2 diabetes, represent worldwide health issues whose incidence is predicted to steadily rise in the coming years. Obesity is characterized by an accumulation of fat in metabolic tissues resulting in chronic inflammation. It is now largely accepted that adipose tissue inflammation underlies the etiology of these disorders. Adipose tissue macrophages (ATMs) represent the most enriched immune fraction in hypertrophic, chronically inflamed adipose tissue, and these cells play a key role in diet-induced type 2 diabetes and insulin resistance. ATMs are triggered by the continuous influx of dietary lipids, among other stimuli; however, how these lipids metabolically activate ATM depends on their nature, composition and localization. This review will discuss the fate and molecular programs elicited within obese ATMs by both exogenous and endogenous lipids, as they mediate the inflammatory response and promote or hamper the development of obesity-associated insulin resistance and type 2 diabetes.
Collapse
|
23
|
Li T, Hu S, Pang X, Wang J, Yin J, Li F, Wang J, Yang X, Xia B, Liu Y, Song W, Guo S. The marine-derived furanone reduces intracellular lipid accumulation in vitro by targeting LXRα and PPARα. J Cell Mol Med 2020; 24:3384-3398. [PMID: 31981312 PMCID: PMC7131916 DOI: 10.1111/jcmm.15012] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/30/2019] [Accepted: 01/10/2020] [Indexed: 12/22/2022] Open
Abstract
Recent studies have demonstrated that commercially available lipid-lowering drugs cause various side effects; therefore, searching for anti-hyperlipidaemic compounds with lower toxicity is a research hotspot. This study was designed to investigate whether the marine-derived compound, 5-hydroxy-3-methoxy-5-methyl-4-butylfuran-2(5H)-one, has an anti-hyperlipidaemic activity, and the potential underlying mechanism in vitro. Results showed that the furanone had weaker cytotoxicity compared to positive control drugs. In RAW 264.7 cells, the furanone significantly lowered ox-LDL-induced lipid accumulation (~50%), and its triglyceride (TG)-lowering effect was greater than that of liver X receptor (LXR) agonist T0901317. In addition, it significantly elevated the protein levels of peroxisome proliferator-activated receptors (PPARα) and ATP-binding cassette (ABC) transporters, which could be partially inhibited by LXR antagonists, GSK2033 and SR9243. In HepG2 cells, it significantly decreased oleic acid-induced lipid accumulation, enhanced the protein levels of low-density lipoprotein receptor (LDLR), ABCG5, ABCG8 and PPARα, and reduced the expression of sterol regulatory element-binding protein 2 (~32%). PPARα antagonists, GW6471 and MK886, could significantly inhibit the furanone-induced lipid-lowering effect. Furthermore, the furanone showed a significantly lower activity on the activation of the expression of lipogenic genes compared to T0901317. Taken together, the furanone exhibited a weak cytotoxicity but had powerful TC- and TG-lowering effects most likely through targeting LXRα and PPARα, respectively. These findings indicate that the furanone has a potential application for the treatment of dyslipidaemia.
Collapse
Affiliation(s)
- Ting Li
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Shu‐Mei Hu
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Xiao‐Yan Pang
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Jun‐feng Wang
- CAS Key Laboratory of Tropical Marine Bio‐resources and Ecology/Guangdong Key Laboratory of Marine Materia Medica/RNAM Center for Marine MicrobiologySouth China Sea Institute of OceanologyChinese Academy of SciencesGuangzhouChina
| | - Jia‐Yu Yin
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Fa‐Hui Li
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Jin Wang
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Xiao‐Qian Yang
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Bin Xia
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Yong‐Hong Liu
- CAS Key Laboratory of Tropical Marine Bio‐resources and Ecology/Guangdong Key Laboratory of Marine Materia Medica/RNAM Center for Marine MicrobiologySouth China Sea Institute of OceanologyChinese Academy of SciencesGuangzhouChina
| | - Wei‐Guo Song
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Shou‐Dong Guo
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| |
Collapse
|
24
|
Inflammation Triggers Liver X Receptor-Dependent Lipogenesis. Mol Cell Biol 2020; 40:MCB.00364-19. [PMID: 31658997 DOI: 10.1128/mcb.00364-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 10/21/2019] [Indexed: 12/29/2022] Open
Abstract
Immune cell function can be modulated by changes in lipid metabolism. Our studies indicate that cholesterol and fatty acid synthesis increases in macrophages between 12 and 18 h after the activation of Toll-like receptors with proinflammatory stimuli and that the upregulation of lipogenesis may contribute to the resolution of inflammation. The inflammation-dependent increase in lipogenesis requires the induction of the liver X receptors, members of the nuclear receptor superfamily of transcription factors, by type I interferons in response to inflammatory signals. Instead of the well-established role for liver X receptors in stimulating cholesterol efflux, we demonstrate that liver X receptors are necessary for the proper resumption of cholesterol synthesis in response to inflammatory signals. Thus, liver X receptors function as bidirectional regulators of cholesterol homeostasis, driving efflux when cholesterol levels are high and facilitating synthesis in response to inflammatory signals. Liver X receptor activity is also required for the proper shutdown of a subset of type I interferon-stimulated genes as inflammation subsides, placing the receptors in a negative-feedback loop that may contribute to the resolution of the inflammatory response.
Collapse
|
25
|
Hidalgo-Lanussa O, Baez-Jurado E, Echeverria V, Ashraf GM, Sahebkar A, Garcia-Segura LM, Melcangi RC, Barreto GE. Lipotoxicity, neuroinflammation, glial cells and oestrogenic compounds. J Neuroendocrinol 2020; 32:e12776. [PMID: 31334878 DOI: 10.1111/jne.12776] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023]
Abstract
The high concentrations of free fatty acids as a consequence of obesity and being overweight have become risk factors for the development of different diseases, including neurodegenerative ailments. Free fatty acids are strongly related to inflammatory events, causing cellular and tissue alterations in the brain, including cell death, deficits in neurogenesis and gliogenesis, and cognitive decline. It has been reported that people with a high body mass index have a higher risk of suffering from Alzheimer's disease. Hormones such as oestradiol not only have beneficial effects on brain tissue, but also exert some adverse effects on peripheral tissues, including the ovary and breast. For this reason, some studies have evaluated the protective effect of oestrogen receptor (ER) agonists with more specific tissue activities, such as the neuroactive steroid tibolone. Activation of ERs positively affects the expression of pro-survival factors and cell signalling pathways, thus promoting cell survival. This review aims to discuss the relationship between lipotoxicity and the development of neurodegenerative diseases. We also elaborate on the cellular and molecular mechanisms involved in neuroprotection induced by oestrogens.
Collapse
Affiliation(s)
- Oscar Hidalgo-Lanussa
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Eliana Baez-Jurado
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Valentina Echeverria
- Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile
- Bay Pines VA Healthcare System, Research and Development, Bay Pines, FL, USA
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Roberto C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| |
Collapse
|
26
|
Ménégaut L, Jalil A, Thomas C, Masson D. Macrophage fatty acid metabolism and atherosclerosis: The rise of PUFAs. Atherosclerosis 2019; 291:52-61. [PMID: 31693943 DOI: 10.1016/j.atherosclerosis.2019.10.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 09/26/2019] [Accepted: 10/09/2019] [Indexed: 01/24/2023]
Abstract
Among the pathways involved in the regulation of macrophage functions, the metabolism of unsaturated fatty acids is central. Indeed, unsaturated fatty acids act as precursors of bioactive molecules such as prostaglandins, leukotrienes, resolvins and related compounds. As components of phospholipids, they have a pivotal role in cell biology by regulating membrane fluidity and membrane-associated cellular processes. Finally, polyunsaturated fatty acids (PUFAs) are also endowed with ligand properties for numerous membrane or nuclear receptors. Although myeloid cells are dependent on the metabolic context for the uptake of essential FAs, recent studies showed that these cells autonomously handle the synthesis of n-3 and n-6 long chain PUFAs such as arachidonic acid and eicosapentaenoic acid. Moreover, targeting PUFA metabolism in macrophages influences pathological processes, including atherosclerosis, by modulating macrophage functions. Omics evidence also supports a role for macrophage PUFA metabolism in the development of cardiometabolic diseases in humans. Currently, there is a renewed interest in the role of n-3/n-6 PUFAs and their oxygenated derivatives in the onset of atherosclerosis and plaque rupture. Purified n-3 FA supplementation appears as a potential strategy in the treatment and prevention of cardiovascular diseases. In this context, the ability of immune cells to handle and to synthesize very long chain PUFA must absolutely be integrated and better understood.
Collapse
Affiliation(s)
- Louise Ménégaut
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - Antoine Jalil
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - Charles Thomas
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - David Masson
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France.
| |
Collapse
|
27
|
Inhibition of Δ24-dehydrocholesterol reductase activates pro-resolving lipid mediator biosynthesis and inflammation resolution. Proc Natl Acad Sci U S A 2019; 116:20623-20634. [PMID: 31548397 DOI: 10.1073/pnas.1911992116] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Targeting metabolism through bioactive key metabolites is an upcoming future therapeutic strategy. We questioned how modifying intracellular lipid metabolism could be a possible means for alleviating inflammation. Using a recently developed chemical probe (SH42), we inhibited distal cholesterol biosynthesis through selective inhibition of Δ24-dehydrocholesterol reductase (DHCR24). Inhibition of DHCR24 led to an antiinflammatory/proresolving phenotype in a murine peritonitis model. Subsequently, we investigated several omics layers in order to link our phenotypic observations with key metabolic alterations. Lipidomic analysis revealed a significant increase in endogenous polyunsaturated fatty acid (PUFA) biosynthesis. These data integrated with gene expression analysis, revealing increased expression of the desaturase Fads6 and the key proresolving enzyme Alox-12/15 Protein array analysis, as well as immune cell phenotype and functional analysis, substantiated these results confirming the antiinflammatory/proresolving phenotype. Ultimately, lipid mediator (LM) analysis revealed the increased production of bioactive lipids, channeling the observed metabolic alterations into a key class of metabolites known for their capacity to change the inflammatory phenotype.
Collapse
|
28
|
Jalil A, Bourgeois T, Ménégaut L, Lagrost L, Thomas C, Masson D. Revisiting the Role of LXRs in PUFA Metabolism and Phospholipid Homeostasis. Int J Mol Sci 2019; 20:ijms20153787. [PMID: 31382500 PMCID: PMC6696407 DOI: 10.3390/ijms20153787] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 01/19/2023] Open
Abstract
Liver X receptors (LXRs) play a pivotal role in fatty acid (FA) metabolism. So far, the lipogenic consequences of in vivo LXR activation, as characterized by a major hepatic steatosis, has constituted a limitation to the clinical development of pharmacological LXR agonists. However, recent studies provided a different perspective. Beyond the quantitative accumulation of FA, it appears that LXRs induce qualitative changes in the FA profile and in the distribution of FAs among cellular lipid species. Thus, LXRs activate the production of polyunsaturated fatty acids (PUFAs) and their distribution into phospholipids via the control of FA desaturases, FA elongases, lysophosphatidylcholine acyltransferase (LPCAT3), and phospholipid transfer protein (PLTP). Therefore, LXRs control, in a dynamic manner, the PUFA composition and the physicochemical properties of cell membranes as well as the release of PUFA-derived lipid mediators. Recent studies suggest that modulation of PUFA and phospholipid metabolism by LXRs are involved in the control of lipogenesis and lipoprotein secretion by the liver. In myeloid cells, the interplay between LXR and PUFA metabolism affects the inflammatory response. Revisiting the complex role of LXRs in FA metabolism may open new opportunities for the development of LXR modulators in the field of cardiometabolic diseases.
Collapse
Affiliation(s)
- Antoine Jalil
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France
- INSERM, LNC UMR 1231, F-21000 Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France
| | - Thibaut Bourgeois
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France
- INSERM, LNC UMR 1231, F-21000 Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France
| | - Louise Ménégaut
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France
- INSERM, LNC UMR 1231, F-21000 Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France
| | - Laurent Lagrost
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France
- INSERM, LNC UMR 1231, F-21000 Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France
| | - Charles Thomas
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France
- INSERM, LNC UMR 1231, F-21000 Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France
| | - David Masson
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000 Dijon, France.
- INSERM, LNC UMR 1231, F-21000 Dijon, France.
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000 Dijon, France.
| |
Collapse
|
29
|
Affiliation(s)
- Ziad Mallat
- From the Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, United Kingdom; and Institut National de la Santé et de la Recherche Médicale, Paris, France.
| |
Collapse
|
30
|
Ma X, Warnier M, Raynard C, Ferrand M, Kirsh O, Defossez PA, Martin N, Bernard D. The nuclear receptor RXRA controls cellular senescence by regulating calcium signaling. Aging Cell 2018; 17:e12831. [PMID: 30216632 PMCID: PMC6260923 DOI: 10.1111/acel.12831] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 07/10/2018] [Accepted: 07/21/2018] [Indexed: 12/18/2022] Open
Abstract
Calcium signaling is emerging as a key pathway controlling cellular senescence, a stable cell proliferation arrest playing a fundamental role in pathophysiological conditions, such as embryonic development, wound healing, cancer, and aging. However, how calcium signaling is regulated is still only partially understood. The inositol 1, 4, 5‐trisphosphate receptor type 2 (ITPR2), an endoplasmic reticulum calcium release channel, was recently shown to critically contribute to the implementation of senescence, but how ITPR2 expression is controlled is unclear. To gain insights into the regulation of ITPR2 expression, we performed an siRNA screen targeting 160 transcription factors and epigenetic regulators. Interestingly, we discovered that the retinoid X receptor alpha (RXRA), which belongs to the nuclear receptor family, represses ITPR2 expression and regulates calcium signaling though ITPR2 and the mitochondrial calcium uniporter (MCU). Knockdown of RXRA induces the production of reactive oxygen species (ROS) and DNA damage via the ITPR2‐MCU calcium signaling axis and consequently triggers cellular senescence by activating p53, whereas RXRA overexpression decreases DNA damage accumulation and then delays replicative senescence. Altogether, our work sheds light on a novel mechanism controlling calcium signaling and cellular senescence and provides new insights into the role of nuclear receptors.
Collapse
Affiliation(s)
- Xingjie Ma
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard; Université de Lyon; Lyon France
| | - Marine Warnier
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard; Université de Lyon; Lyon France
| | - Clotilde Raynard
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard; Université de Lyon; Lyon France
| | - Mylène Ferrand
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard; Université de Lyon; Lyon France
| | - Olivier Kirsh
- Epigenetics and Cell Fate, Sorbonne Paris Cité, CNRS UMR 7216; Université Paris Diderot; Paris France
| | - Pierre-Antoine Defossez
- Epigenetics and Cell Fate, Sorbonne Paris Cité, CNRS UMR 7216; Université Paris Diderot; Paris France
| | - Nadine Martin
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard; Université de Lyon; Lyon France
| | - David Bernard
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard; Université de Lyon; Lyon France
| |
Collapse
|
31
|
Tang CH, Lin CY, Tsai YL, Lee SH, Wang WH. Lipidomics as a diagnostic tool of the metabolic and physiological state of managed whales: A correlation study of systemic metabolism. Zoo Biol 2018; 37:440-451. [PMID: 30457161 DOI: 10.1002/zoo.21452] [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: 06/01/2018] [Revised: 09/09/2018] [Accepted: 10/09/2018] [Indexed: 11/06/2022]
Abstract
Integrating multifactor blood analysis is a key step toward a precise diagnosis of the health status of marine mammals. Variations in the circulating lipid profile reflect changes in the metabolism and physiology of an individual. To demonstrate the practicability of lipid profiling for physiological assessment, the phosphorylcholine-containing lipids in the plasma of long-term managed beluga whales (Delphinapterus leucas) were profiled using a lipidomics methodology. Using a multivariate analysis, the mean corpuscular volume, cholesterol, potassium, and γ-glutamyltranspeptidase levels were well modeled with the lipid profile of the female whales. In the models, the correlated lipids provided information about blood parameter-related metabolism and physiological regulation, in particular relating to cholesterol and inflammation. In the males, the levels of cholesterol, triglycerides, blood urea nitrogen, creatinine, plasma iron, and segmented neutrophil were well modeled with the lipid profile. In addition to providing information about the related metabolism and regulation, through a cross-linked analysis of the blood parameters, the correlated lipids indicated a parallel regulation involved in the energy metabolism of the male whales. Lipidomics as a method for revealing the context of physiological change shows practical potential for the health care of managed whales.
Collapse
Affiliation(s)
- Chuan-Ho Tang
- Department of Biology, National Museum of Marine Biology and Aquarium, Pingtung, Taiwan.,Institute of Marine Biology, National Dong Hwa University, Pingtung, Taiwan
| | - Ching-Yu Lin
- Institute of Environmental Health, National Taiwan University, Taipei City, Taiwan
| | - Yi-Lun Tsai
- Department of Veterinary Medicine and Animal Hospital, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Shu-Hui Lee
- Central of General Education, National Kaohsiung Marine University, Kaohsiung, Taiwan
| | - Wei-Hsien Wang
- Department of Biology, National Museum of Marine Biology and Aquarium, Pingtung, Taiwan.,Department of Marine Biotechnology and Resources and Asia-Pacific Ocean Research Center, National Sun Yat-Sen University, Kaohsiung, Taiwan
| |
Collapse
|
32
|
Qian X, Yang Z, Mao E, Chen E. Regulation of fatty acid synthesis in immune cells. Scand J Immunol 2018; 88:e12713. [PMID: 30176060 DOI: 10.1111/sji.12713] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 07/20/2018] [Accepted: 08/22/2018] [Indexed: 02/06/2023]
Abstract
Metabolic reprogramming plays a critical role in the important cellular metabolic alterations that occur during the activation of immune cells to enable them to adapt to the extracellular environment. Here, we review recent studies on how substrate availability and metabolites mediate the signalling pathways that regulate fatty acid synthesis (FAS) in different immune cells and how FAS determines cellular fate and function. The major regulators sterol regulatory element-binding proteins and liver X receptors, the key enzyme ATP citrate lyase and the PI3K-Akt-mTOR signalling axis play important roles in de novo FAS during a variety of biological events, including cellular proliferation and differentiation and the development of organelles and intracellular membrane components in immune cells. In addition, the regulation of FAS substantially contributes to the inflammatory response of immune cells. Post-transcriptional modifications in FAS are also closely associated with the functional processes of immune cells. Understanding and investigating the intrinsic regulatory mechanism of FAS is of great significance for developing novel therapies for inflammation-induced diseases.
Collapse
Affiliation(s)
- Xuchen Qian
- Department of Emergency and Critical Care Medicine, First People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhitao Yang
- Department of Emergency Intensive Care Unit, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Enqiang Mao
- Department of Emergency Intensive Care Unit, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Erzhen Chen
- Department of Emergency Intensive Care Unit, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW It is increasingly recognized that profound metabolic changes occur in activated myeloid cells, which shape their inflammatory phenotype and cellular functions. The purpose of this review is to summarize the accumulating evidence that major metabolic adaptations occur in monocytes and macrophages in the context of atherosclerosis ultimately modulating atherosclerotic plaque formation. RECENT FINDINGS Plaque macrophages show a profound metabolic reprogramming which is driven by atherogenic factors in the plaque microenvironment, such as damage associated molecular patterns, modified lipoproteins, and hypoxia. In addition, systemic atherogenic factors modulate metabolism of circulating monocytes and their bone marrow progenitors. Activation of glycolysis, the pentose phosphate pathway, and fatty acid synthesis, a reduction of fatty acid oxidation accompanied by complex changes in the lysosomal handling of lipids all appear to facilitate atherogenesis. These processes also drive the development of trained immunity, a phenomenon describing the persistent pro-inflammatory phenotype that develops after brief stimulation of monocytes with pro-atherogenic stimuli. SUMMARY A pro-atherosclerotic environment reprograms the metabolism of myeloid cells in the various developmental phases of atherosclerosis. Knowledge of these metabolic programs facilitates the development of novel drugs to prevent atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA, Nijmegen
| | - Rinke Stienstra
- Department of Internal Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA, Nijmegen
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
34
|
Zhang H, de Aguiar Vallim TQ, Martel C. Translational and Therapeutic Approaches to the Understanding and Treatment of Dyslipidemia. Arterioscler Thromb Vasc Biol 2018; 36:e56-61. [PMID: 27335468 DOI: 10.1161/atvbaha.116.307808] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Hanrui Zhang
- From the Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (H.Z.); Division of Cardiology, School of Medicine, University of California Los Angeles (T.Q. de A. V.); and Department of Medicine, Montreal Heart Institute Research Center, Université de Montréal, Montreal, Quebec, Canada (C.M.).
| | - Thomas Q de Aguiar Vallim
- From the Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (H.Z.); Division of Cardiology, School of Medicine, University of California Los Angeles (T.Q. de A. V.); and Department of Medicine, Montreal Heart Institute Research Center, Université de Montréal, Montreal, Quebec, Canada (C.M.).
| | - Catherine Martel
- From the Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (H.Z.); Division of Cardiology, School of Medicine, University of California Los Angeles (T.Q. de A. V.); and Department of Medicine, Montreal Heart Institute Research Center, Université de Montréal, Montreal, Quebec, Canada (C.M.).
| | | |
Collapse
|
35
|
El-Gendy BEDM, Goher SS, Hegazy LS, Arief MMH, Burris TP. Recent Advances in the Medicinal Chemistry of Liver X Receptors. J Med Chem 2018; 61:10935-10956. [DOI: 10.1021/acs.jmedchem.8b00045] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Bahaa El-Dien M. El-Gendy
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
- Chemistry Department, Faculty of Science, Benha University, Benha 13518, Egypt
| | - Shaimaa S. Goher
- Chemistry Department, Faculty of Science, Benha University, Benha 13518, Egypt
| | - Lamees S. Hegazy
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - Mohamed M. H. Arief
- Chemistry Department, Faculty of Science, Benha University, Benha 13518, Egypt
| | - Thomas P. Burris
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
| |
Collapse
|
36
|
Zhang C, Qin JJ, Gong FH, Tong JJ, Cheng WL, Wang H, Zhang Y, Zhu X, She ZG, Xia H, Zhu LH. Mindin deficiency in macrophages protects against foam cell formation and atherosclerosis by targeting LXR-β. Clin Sci (Lond) 2018; 132:1199-1213. [PMID: 29695588 DOI: 10.1042/cs20180033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/21/2018] [Accepted: 04/25/2018] [Indexed: 02/07/2023]
Abstract
Mindin, which is a highly conserved extracellular matrix protein, has been documented to play pivotal roles in regulating angiogenesis, inflammatory processes, and immune responses. The aim of the present study was to assess whether mindin contributes to the development of atherosclerosis. A significant up-regulation of Mindin expression was observed in the serum, arteries and atheromatous plaques of ApoE-/- mice after high-fat diet treatment. Mindin-/-ApoE-/- mice and macrophage-specific mindin overexpression in ApoE-/- mice (Lyz2-mindin-TG) were generated to evaluate the effect of mindin on the development of atherosclerosis. The Mindin-/-ApoE-/- mice exhibited significantly ameliorated atherosclerotic burdens in the entire aorta and aortic root and increased atherosclerotic plaque stability. Moreover, bone marrow transplantation further demonstrated that mindin deficiency in macrophages was largely responsible for the alleviated atherogenesis. The Lyz2-mindin-TG mice exhibited the opposite phenotype. Mindin deficiency enhanced foam cell formation by increasing the expression of cholesterol effectors, including ABCA1 and ABCG1. The mechanistic study indicated that mindin ablation promoted LXR-β expression via a direct interaction. Importantly, LXR-β inhibition largely reversed the ameliorating effect of mindin deficiency on foam cell formation and ABCA1 and ABCG1 expression. The present study demonstrated that mindin deficiency serves as a novel mediator that protects against foam cell formation and atherosclerosis by directly interacting with LXR-β.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Fu-Han Gong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Jing-Jing Tong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Wen-Lin Cheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Haiping Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Yan Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Xueyong Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Hao Xia
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Li-Hua Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| |
Collapse
|
37
|
Xi D, Zhao J, Zhao M, Fu W, Guo Z, Chen H. Identification of Gene Expression Changes in the Aorta of ApoE Null Mice Fed a High-Fat Diet. Genes (Basel) 2017; 8:genes8100289. [PMID: 29064389 PMCID: PMC5664139 DOI: 10.3390/genes8100289] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/08/2017] [Accepted: 10/16/2017] [Indexed: 12/19/2022] Open
Abstract
Atherosclerosis is a chronic multifactorial inflammatory disease with high worldwide prevalence, and has become the leading cause of death. In the present study, we analyzed global gene expression changes in the aorta of Apolipoprotein E (ApoE) null mice fed a high-fat diet by using RNA-seq. We identified a total of 280 differentially expressed genes, of which 163 genes were upregulated and 117 genes were downregulated by high-fat diet compared with normal diet. Functional clustering and gene network analysis revealed that fatty acid metabolic process is crucial for atherosclerosis. By examining of the promoter regions of differentially expressed genes, we identified four causal transcription factors. Additionally, through connectivity map (CMap) analysis, multiple compounds were identified to have anti-atherosclerotic effects due to their ability to reverse gene expression during atherosclerosis. Our study provides a valuable resource for in-depth understanding of the mechanism underlying atherosclerosis.
Collapse
Affiliation(s)
- Dan Xi
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China.
| | - Jinzhen Zhao
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China.
| | - Miao Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Weijun Fu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China.
| | - Zhigang Guo
- Department of Cardiology, Huqiao Medical Center, Nanfang Hospital, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China.
| | - Hui Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China.
| |
Collapse
|
38
|
Thomas C, Ménégaut L, Masson D. Fatty acids and macrophage functions: an update. Curr Opin Lipidol 2017; 28:443-444. [PMID: 28857859 DOI: 10.1097/mol.0000000000000448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Charles Thomas
- aLipSTIC LabEx, LNC UMR866, INSERM, LNC UMR1231, FCS Bourgogne-Franche Comté, University Bourgogne Franche-Comté bLaboratoire de Biochimie, CHU Dijon, France
| | | | | |
Collapse
|
39
|
Jack-Roberts C, Joselit Y, Nanobashvili K, Bretter R, Malysheva OV, Caudill MA, Saxena A, Axen K, Gomaa A, Jiang X. Choline Supplementation Normalizes Fetal Adiposity and Reduces Lipogenic Gene Expression in a Mouse Model of Maternal Obesity. Nutrients 2017; 9:nu9080899. [PMID: 28820499 PMCID: PMC5579692 DOI: 10.3390/nu9080899] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/11/2017] [Accepted: 08/15/2017] [Indexed: 01/06/2023] Open
Abstract
Maternal obesity increases fetal adiposity which may adversely affect metabolic health of the offspring. Choline regulates lipid metabolism and thus may influence adiposity. This study investigates the effect of maternal choline supplementation on fetal adiposity in a mouse model of maternal obesity. C57BL/6J mice were fed either a high-fat (HF) diet or a control (NF) diet and received either 25 mM choline supplemented (CS) or control untreated (CO) drinking water for 6 weeks before timed-mating and throughout gestation. At embryonic day 17.5, HF feeding led to higher (p < 0.05) percent total body fat in fetuses from the HFCO group, while the choline supplemented HFCS group did not show significant difference versus the NFCO group. Similarly, HF feeding led to higher (p < 0.05) hepatic triglyceride accumulation in the HFCO but not the HFCS fetuses. mRNA levels of lipogenic genes such as Acc1, Fads1, and Elovl5, as well as the transcription factor Srebp1c that favors lipogenesis were downregulated (p < 0.05) by maternal choline supplementation in the HFCS group, which may serve as a mechanism to reduce fat accumulation in the fetal liver during maternal HF feeding. In summary, maternal choline supplementation improves indices of fetal adiposity in obese dams at late gestation.
Collapse
Affiliation(s)
- Chauntelle Jack-Roberts
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA.
| | - Yaelle Joselit
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA.
| | - Khatia Nanobashvili
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA.
| | - Rachel Bretter
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA.
| | - Olga V Malysheva
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA.
| | - Marie A Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA.
| | - Anjana Saxena
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA.
| | - Kathleen Axen
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA.
| | - Ahmed Gomaa
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA.
| | - Xinyin Jiang
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA.
| |
Collapse
|
40
|
Shi H, Zhang C, Xu Z, Xu X, Lv Z, Luo J, Loor J. Nuclear receptor subfamily 1 group H member 2 (LXRB) is the predominant liver X receptor subtype regulating transcription of 2 major lipogenic genes in goat primary mammary epithelial cells. J Dairy Sci 2017. [DOI: 10.3168/jds.2016-12510] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
41
|
Connecting the Dots Between Fatty Acids, Mitochondrial Function, and DNA Methylation in Atherosclerosis. Curr Atheroscler Rep 2017; 19:36. [PMID: 28735349 DOI: 10.1007/s11883-017-0673-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The quest for factors and mechanisms responsible for aberrant DNA methylation in human disease-including atherosclerosis-is a promising area of research. This review focuses on the role of fatty acids (FAs) as modulators of DNA methylation-in particular the role of mitochondrial beta-oxidation in FA-induced changes in DNA methylation during the progression of atherosclerosis. RECENT FINDINGS Recent publications have advanced the knowledge in all areas touched by this review: the causal role of lipids in shaping the DNA methylome, the associations between chronic degenerative disease and mitochondrial function, the lipid composition of the atheroma, and the relevance of DNA hypermethylation in atherosclerosis. Evidence is beginning to emerge, linking the dynamics of FA type abundance, mitochondrial function, and DNA methylation in the atheroma and systemically. In particular, this review highlights mitochondrial beta-oxidation as an important regulator of DNA methylation in metabolic disease. Despite the many questions still unanswered, this area of research promises to identify mechanisms and molecular factors that establish a pathological gene expression pattern in atherosclerosis.
Collapse
|
42
|
Lukic A, Larssen P, Fauland A, Samuelsson B, Wheelock CE, Gabrielsson S, Radmark O. GM-CSF- and M-CSF-primed macrophages present similar resolving but distinct inflammatory lipid mediator signatures. FASEB J 2017. [PMID: 28637652 DOI: 10.1096/fj.201700319r] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
M1 and M2 activated macrophages (Mϕs) have different roles in inflammation. Because pathogens may first encounter resting cells, we investigated lipid mediator profiles prior to full activation. Human monocytes were differentiated with granulocyte Mϕ colony-stimulating factor (GM-CSF) or Mϕ colony-stimulating factor (M-CSF), which are known to prime toward M1 or M2 phenotypes, respectively. Lipid mediators released during resting conditions and produced in response to bacterial stimuli (LPS/N-formylmethionyl-leucyl-phenylalanine or peptidoglycan) were quantified by liquid chromatography-mass spectrometry. In resting conditions, both Mϕ phenotypes released primarily proresolving lipid mediators (prostaglandin E2 metabolite, lipoxin A4, and 18-hydroxyeicosapentaenoic acid). A striking shift toward proinflammatory eicosanoids was observed when the same cells were exposed (30 min) to bacterial stimuli: M-CSF Mϕs produced considerably more 5-lipoxygenase products, particularly leukotriene C4, potentially linked to M2 functions in asthma. Prostaglandins were formed by both Mϕ types. In the M-CSF cells, there was also an enhanced release of arachidonic acid and activation of cytosolic phospholipase A2 However, GM-CSF cells expressed higher levels of 5-lipoxygenase and 5-lipoxygenase-activating protein, and in ionophore incubations these cells also produced the highest levels of 5-hydroxyeicosatetraenoic acid. In summary, GM-CSF and M-CSF Mϕs displayed similar proresolving lipid mediator formation in resting conditions but shifted toward different proinflammatory eicosanoids upon bacterial stimuli. This demonstrates that preference for specific eicosanoid pathways is primed by CSFs before full M1/M2 activation.-Lukic, A., Larssen, P., Fauland, A., Samuelsson, B., Wheelock, C. E., Gabrielsson, S., Radmark, O. GM-CSF- and M-CSF-primed macrophages present similar resolving but distinct inflammatory lipid mediator signatures.
Collapse
Affiliation(s)
- Ana Lukic
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Pia Larssen
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Alexander Fauland
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Samuelsson
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Craig E Wheelock
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Gabrielsson
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Olof Radmark
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden;
| |
Collapse
|
43
|
Ménégaut L, Thomas C, Lagrost L, Masson D. Fatty acid metabolism in macrophages: a target in cardio-metabolic diseases. Curr Opin Lipidol 2017; 28:19-26. [PMID: 27870652 DOI: 10.1097/mol.0000000000000370] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Recent studies have highlighted that macrophages dynamically and autonomously handle all the facets of fatty acid (FA) metabolism including FA oxidation and FA synthesis as well as the synthesis of monounsaturated FAs and long chain n-3 and n-6 polyunsaturated FAs. RECENT FINDINGS Macrophage M2 polarization is associated with an increase of FA oxidation. However, whether increased FA oxidation simply correlates with or is required for M2 polarization needs to be further evaluated. Macrophage M1 polarization is associated with the activation of FA synthesis, which directly contributes to the inflammatory response and affects cholesterol homeostasis and neutral lipid accumulation. Finally, recent evidences suggest that macrophages are able to autonomously produce signaling monounsaturated FAs, such as palmitoleic acid (C16 : 1 n-7), and long chain n-3 and n-6 polyunsaturated FAs, such as arachidonic acid, eicosapentaenoic acid, and docosahexaenoic acid. This pathway is regulated by liver X receptors and has significant consequences on inflammation and on the FA composition of atheroma plaques. SUMMARY These studies shed new light on the tight relationship between FA metabolism, macrophage polarization, and M1/M2 macrophage functions. These processes may have major consequences for atherosclerosis pathogenesis as well as other metabolic disorders.
Collapse
Affiliation(s)
- Louise Ménégaut
- aUniversity Bourgogne Franche-Comté, LNC UMR866 bINSERM, LNC UMR866 cFCS Bourgogne-Franche Comté dCHU Dijon, laboratoire de Biochimie, Dijon, France
| | | | | | | |
Collapse
|
44
|
Smet M, Van Hoecke L, De Beuckelaer A, Vander Beken S, Naessens T, Vergote K, Willart M, Lambrecht BN, Gustafsson JÅ, Steffensen KR, Grooten J. Cholesterol-sensing liver X receptors stimulate Th2-driven allergic eosinophilic asthma in mice. IMMUNITY INFLAMMATION AND DISEASE 2016; 4:350-61. [PMID: 27621817 PMCID: PMC5004289 DOI: 10.1002/iid3.118] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/02/2016] [Accepted: 06/29/2016] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Liver X receptors (LXRs) are nuclear receptors that function as cholesterol sensors and regulate cholesterol homeostasis. High cholesterol has been recognized as a risk factor in asthma; however, the mechanism of this linkage is not known. METHODS To explore the importance of cholesterol homeostasis for asthma, we investigated the contribution of LXR activity in an ovalbumin- and a house dust mite-driven eosinophilic asthma mouse model. RESULTS In both models, airway inflammation, airway hyper-reactivity, and goblet cell hyperplasia were reduced in mice deficient for both LXRα and LXRβ isoforms (LXRα(-/-)β(-/-)) as compared to wild-type mice. Inversely, treatment with the LXR agonist GW3965 showed increased eosinophilic airway inflammation. LXR activity contributed to airway inflammation through promotion of type 2 cytokine production as LXRα(-/-)β(-/-) mice showed strongly reduced protein levels of IL-5 and IL-13 in the lungs as well as reduced expression of these cytokines by CD4(+) lung cells and lung-draining lymph node cells. In line herewith, LXR activation resulted in increased type 2 cytokine production by the lung-draining lymph node cells. CONCLUSIONS In conclusion, our study demonstrates that the cholesterol regulator LXR acts as a positive regulator of eosinophilic asthma in mice, contributing to airway inflammation through regulation of type 2 cytokine production.
Collapse
Affiliation(s)
- Muriel Smet
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
| | - Lien Van Hoecke
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium; Medical Biotechnology CenterFlanders Institute for BiotechnologyGhentBelgium
| | - Ans De Beuckelaer
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
| | - Seppe Vander Beken
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
| | - Thomas Naessens
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
| | - Karl Vergote
- Department of Respiratory MedicineGhent University HospitalGhentBelgium; Inflammation Research CenterFlanders Institute for BiotechnologyGhentBelgium
| | - Monique Willart
- Department of Respiratory MedicineGhent University HospitalGhentBelgium; Inflammation Research CenterFlanders Institute for BiotechnologyGhentBelgium
| | - Bart N Lambrecht
- Department of Respiratory MedicineGhent University HospitalGhentBelgium; Inflammation Research CenterFlanders Institute for BiotechnologyGhentBelgium
| | - Jan-Åke Gustafsson
- Department of Biosciences and Nutrition at NovumKarolinska InstitutetStockholmSweden; Department of Biology and BiochemistryUniversity of HoustonHoustonTexas
| | - Knut R Steffensen
- Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | - Johan Grooten
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
| |
Collapse
|
45
|
Yao DW, Luo J, He QY, Xu HF, Li J, Shi HB, Wang H, Chen Z, Loor JJ. Liver X receptor α promotes the synthesis of monounsaturated fatty acids in goat mammary epithelial cells via the control of stearoyl-coenzyme A desaturase 1 in an SREBP-1-dependent manner. J Dairy Sci 2016; 99:6391-6402. [PMID: 27209141 DOI: 10.3168/jds.2016-10990] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/31/2016] [Indexed: 12/30/2022]
Abstract
Stearoyl-coenzyme A desaturase 1 (SCD1) is a pivotal enzyme in the biosynthesis of monounsaturated fatty acids (MUFA). It is tightly regulated by transcription factors that control lipogenesis. In nonruminants, liver X receptor α (LXRα) is a nuclear receptor and transcription factor that acts as a key sensor of cholesterol and lipid homeostasis. However, the mechanism whereby LXRα regulates the expression and transcriptional activity of SCD1 in ruminant mammary cells remains unknown. In this study with goat mammary epithelial cells (GMEC), the LXRα agonist T 4506585 (T09) markedly enhanced the mRNA expression of SCD1 and sterol regulatory element binding factor 1 (SREBF1). The concentrations of C16:1 and C18:1 and their desaturation indices also were increased by LXRα activation. However, knockdown of LXRα did not alter the mRNA expression of SCD1. Although SCD1 was repressed by SREBF1 knockdown, T09 significantly increased SCD1 expression. Further analysis revealed that the SCD1 promoter activity was activated by LXRα overexpression. The goat SCD1 promoter contains 2 LXR response elements (LXRE), 1 sterol response element (SRE), and 1 nuclear factor Y (NF-Y) binding site. Site-directed mutagenesis of LXRE1, LXRE2, or SRE alone did not eliminate the upregulation of SCD1 when LXRα was overexpressed. In contrast, when NF-Y alone or in combination with SRE was mutated simultaneously, the basal transcriptional activity of the SCD1 promoter was markedly decreased and did not respond to LXRα overexpression. Furthermore, when SREBF1 was knocked down, overexpression of LXRα did not affect the promoter activity of SCD1. Together, these data suggest that LXRα regulates the expression of SCD1 through increasing SREBP-1 abundance to promote interaction with SRE and NF-Y binding sites. The present study provides evidence that LXRα is involved in the synthesis of MUFA in the goat mammary gland through an indirect mechanism.
Collapse
Affiliation(s)
- D W Yao
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China
| | - J Luo
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China.
| | - Q Y He
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China
| | - H F Xu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China
| | - J Li
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, P. R. China 450046
| | - H B Shi
- College of Animal Sciences, Zhejiang Sci-Tech University, Hangzhou, P. R. China 310058
| | - H Wang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China
| | - Z Chen
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China
| | - J J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801.
| |
Collapse
|
46
|
Jiang Z, Huang X, Huang S, Guo H, Wang L, Li X, Huang X, Wang T, Zhang L, Sun L. Sex-Related Differences of Lipid Metabolism Induced by Triptolide: The Possible Role of the LXRα/SREBP-1 Signaling Pathway. Front Pharmacol 2016; 7:87. [PMID: 27065871 PMCID: PMC4814849 DOI: 10.3389/fphar.2016.00087] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 03/16/2016] [Indexed: 12/13/2022] Open
Abstract
Triptolide, a diterpenoid isolated from the plant Tripterygium wilfordii Hook. f., exerts a unique bioactive spectrum of anti-inflammatory and anticancer activities. However, triptolide's clinical applications are limited due to its severe toxicities. Fatty liver toxicity occurs in response to triptolide, and this toxic response significantly differs between males and females. This report investigated the pathogenesis underlying the sex-related differences in the dyslipidosis induced by triptolide in rats. Wistar rats were administered 0, 150, 300, or 450 μg triptolide/kg/day by gavage for 28 days. Ultrastructural examination revealed that more lipid droplets were present in female triptolide-treated rats than in male triptolide-treated rats. Furthermore, liver triglyceride, total bile acid and free fatty acid levels were significantly increased in female rats in the 300 and 450 μg/kg dose groups. The expression of liver X receptor α (LXRα) and its target genes, cholesterol 7α-hydroxylase (CYP7A1) and Sterol regulatory element-binding transcription factor 1(SREBP-1), increased following triptolide treatment in both male and female rats; however, the female rats were more sensitive to triptolide than the male rats. In addition, the expression of acetyl-CoA carboxylase 1(ACC1), a target gene of SREBP-1, increased in the female rats treated with 450 μg triptolide/kg/day, and ACC1 expression contributed to the sex-related differences in the triptolide-induced dysfunction of lipid metabolism. Our results demonstrate that the sex-related differences in LXR/SREBP-1-mediated regulation of gene expression in rats are responsible for the sex-related differences in lipid metabolism induced by triptolide, which likely underlie the sex-related differences in triptolide hepatotoxicity. This study will be important for predicting sex-related effects on the pharmacokinetics and toxicity of triptolide and for improving its safety.
Collapse
Affiliation(s)
- Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug Screening and Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical UniversityNanjing, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical UniversityNanjing, China
| | - Xiao Huang
- Jiangsu Key Laboratory of Drug Screening and Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University Nanjing, China
| | - Shan Huang
- Jiangsu Key Laboratory of Drug Screening and Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University Nanjing, China
| | - Hongli Guo
- Jiangsu Key Laboratory of Drug Screening and Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University Nanjing, China
| | - Lu Wang
- Jiangsu Key Laboratory of Drug Screening and Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University Nanjing, China
| | - Xiaojiaoyang Li
- Jiangsu Key Laboratory of Drug Screening and Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University Nanjing, China
| | - Xin Huang
- Jiangsu Key Laboratory of Drug Screening and Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical UniversityNanjing, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical UniversityNanjing, China
| | - Tao Wang
- Jiangsu Key Laboratory of Drug Screening and Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University Nanjing, China
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug Screening and Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical UniversityNanjing, China; State Key Laboratory of Natural Medicines, China Pharmaceutical UniversityNanjing, China
| | - Lixin Sun
- Jiangsu Key Laboratory of Drug Screening and Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical UniversityNanjing, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical UniversityNanjing, China
| |
Collapse
|
47
|
Wang H, Luo J, Zhang T, Tian H, Ma Y, Xu H, Yao D, Loor JJ. MicroRNA-26a/b and their host genes synergistically regulate triacylglycerol synthesis by targeting the INSIG1 gene. RNA Biol 2016; 13:500-10. [PMID: 27002347 DOI: 10.1080/15476286.2016.1164365] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The microRNA-26 (miR-26) family is known to control adipogenesis in non-ruminants. The genomic loci of miR-26a and miR-26b have been localized in the introns of genes encoding for the proteins of the C-terminal domain RNA polymerase II polypeptide A small phosphatase (CTDSP) family. Insulin-induced gene 1 (INSIG1) encodes a protein with a key role in the regulation of lipogenesis in rodent liver. In the present study, we investigated the synergistic function of the miR-26 family and their host genes in goat mammary epithelial cells (GMEC). Downregulation of miR-26a/b and their host genes in GMEC decreased the expression of genes relate to fatty acid synthesis (PPARG, LXRA, SREBF1, FASN, ACACA, GPAM, LPIN1, DGAT1 and SCD1), triacylglycerol accumulation and unsaturated fatty acid synthesis. Luciferase reporter assays confirmed INSIG1 as a direct target of miR-26a/b. Furthermore, inhibition of the CTDSP family also downregulated the expression of INSIG1. Taken together, our findings highlight a functional association of miR-26a/b, their host genes and INSIG1, and provide new insights into the regulatory network controlling milk fat synthesis in GMEC. The data indicate that targeting this network via nutrition might be important for regulating milk fat synthesis in ruminants.
Collapse
Affiliation(s)
- Hui Wang
- a Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University , Yangling , Shaanxi , PR China
| | - Jun Luo
- a Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University , Yangling , Shaanxi , PR China
| | - Tianying Zhang
- a Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University , Yangling , Shaanxi , PR China
| | - Huibin Tian
- a Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University , Yangling , Shaanxi , PR China
| | - Yue Ma
- a Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University , Yangling , Shaanxi , PR China
| | - Huifen Xu
- a Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University , Yangling , Shaanxi , PR China
| | - Dawei Yao
- a Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University , Yangling , Shaanxi , PR China
| | - Juan J Loor
- b Mammalian NutriPhysioGenomics , Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois , Urbana , USA
| |
Collapse
|
48
|
Ballestri S, Nascimbeni F, Romagnoli D, Baldelli E, Lonardo A. The Role of Nuclear Receptors in the Pathophysiology, Natural Course, and Drug Treatment of NAFLD in Humans. Adv Ther 2016; 33:291-319. [PMID: 26921205 DOI: 10.1007/s12325-016-0306-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) describes steatosis, nonalcoholic steatohepatitis with or without fibrosis, and hepatocellular carcinoma, namely the entire alcohol-like spectrum of liver disease though observed in the nonalcoholic, dysmetabolic, individual free of competing causes of liver disease. NAFLD, which is a major public health issue, exhibits intrahepatic triglyceride storage giving rise to lipotoxicity. Nuclear receptors (NRs) are transcriptional factors which, activated by ligands, are master regulators of metabolism and also have intricate connections with circadian control accounting for cyclical patterns in the metabolic fate of nutrients. Several transcription factors, such as peroxisome proliferator-activated receptors, liver X receptors, farnesoid X receptors, and their molecular cascades, finely regulate energetic fluxes and metabolic pathways. Dysregulation of such pathways is heavily implicated in those metabolic derangements characterizing insulin resistance and metabolic syndrome and in the histogenesis of progressive NAFLD forms. We review the role of selected NRs in NAFLD pathogenesis. Secondly, we analyze the role of NRs in the natural history of human NAFLD. Next, we discuss the results observed in humans following administration of drug agonists or antagonists of the NRs pathogenically involved in NAFLD. Finally, general principles of treatment and lines of research in human NAFLD are briefly examined.
Collapse
Affiliation(s)
| | - Fabio Nascimbeni
- NOCSAE, Outpatient Liver Clinic and Operating Unit Internal Medicine, Azienda USL Modena, Modena, Italy
- University of Modena and Reggio Emilia, Modena, Italy
| | - Dante Romagnoli
- NOCSAE, Outpatient Liver Clinic and Operating Unit Internal Medicine, Azienda USL Modena, Modena, Italy
| | | | - Amedeo Lonardo
- NOCSAE, Outpatient Liver Clinic and Operating Unit Internal Medicine, Azienda USL Modena, Modena, Italy.
| |
Collapse
|
49
|
Zheng Z, Zheng F. Immune Cells and Inflammation in Diabetic Nephropathy. J Diabetes Res 2016; 2016:1841690. [PMID: 26824038 PMCID: PMC4707326 DOI: 10.1155/2016/1841690] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 10/21/2015] [Indexed: 12/20/2022] Open
Abstract
Diabetic nephropathy (DN) is a serious complication of diabetes. At its core, DN is a metabolic disorder which can also manifest itself in terms of local inflammation in the kidneys. Such inflammation can then drive the classical markers of fibrosis and structural remodeling. As a result, resolution of immune-mediated inflammation is critical towards achieving a cure for DN. Many immune cells play a part in DN, including key members of both the innate and adaptive immune systems. While these cells were classically understood to primarily function against pathogen insult, it has also become increasingly clear that they also serve a major role as internal sensors of damage. In fact, damage sensing may serve as the impetus for much of the inflammation that occurs in DN, in a vicious positive feedback cycle. Although direct targeting of these proinflammatory cells may be difficult, new approaches that focus on their metabolic profiles may be able to alleviate DN significantly, especially since dysregulation of the local metabolic environment may well be responsible for triggering inflammation to begin with. In this review, the authors consider the metabolic profile of several relevant immune types and discuss their respective roles.
Collapse
Affiliation(s)
- Zihan Zheng
- College of Arts and Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Feng Zheng
- Department of Nephrology, Advanced Institute for Medical Sciences, Second Hospital, Dalian Medical University, Dalian 116023, China
- Department of Nephrology and Basic Science Laboratory, Fujian Medical University, Fuzhou 350002, China
- *Feng Zheng:
| |
Collapse
|
50
|
Vallerie SN, Bornfeldt KE. Metabolic Flexibility and Dysfunction in Cardiovascular Cells. Arterioscler Thromb Vasc Biol 2015; 35:e37-42. [PMID: 26310811 DOI: 10.1161/atvbaha.115.306226] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Sara N Vallerie
- From the Division of Metabolism, Endocrinology and Nutrition, Departments of Medicine (S.N.V., K.E.B.) and Pathology (K.E.B.), Diabetes and Obesity Center of Excellence, University of Washington School of Medicine, Seattle
| | - Karin E Bornfeldt
- From the Division of Metabolism, Endocrinology and Nutrition, Departments of Medicine (S.N.V., K.E.B.) and Pathology (K.E.B.), Diabetes and Obesity Center of Excellence, University of Washington School of Medicine, Seattle.
| |
Collapse
|