1
|
Johnson SM, Bao H, McMahon CE, Chen Y, Burr SD, Anderson AM, Madeyski-Bengtson K, Lindén D, Han X, Liu J. PNPLA3 is a triglyceride lipase that mobilizes polyunsaturated fatty acids to facilitate hepatic secretion of large-sized very low-density lipoprotein. Nat Commun 2024; 15:4847. [PMID: 38844467 PMCID: PMC11156938 DOI: 10.1038/s41467-024-49224-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 05/22/2024] [Indexed: 06/09/2024] Open
Abstract
The I148M variant of PNPLA3 is closely associated with hepatic steatosis. Recent evidence indicates that the I148M mutant functions as an inhibitor of PNPLA2/ATGL-mediated lipolysis, leaving the role of wild-type PNPLA3 undefined. Despite showing a triglyceride hydrolase activity in vitro, PNPLA3 has yet to be established as a lipase in vivo. Here, we show that PNPLA3 preferentially hydrolyzes polyunsaturated triglycerides, mobilizing polyunsaturated fatty acids for phospholipid desaturation and enhancing hepatic secretion of triglyceride-rich lipoproteins. Under lipogenic conditions, mice with liver-specific knockout or acute knockdown of PNPLA3 exhibit aggravated liver steatosis and reduced plasma VLDL-triglyceride levels. Similarly, I148M-knockin mice show decreased hepatic triglyceride secretion during lipogenic stimulation. Our results highlight a specific context whereby the wild-type PNPLA3 facilitates the balance between hepatic triglyceride storage and secretion, and suggest the potential contribution of a loss-of-function by the I148M variant to the development of fatty liver disease in humans.
Collapse
Affiliation(s)
- Scott M Johnson
- Department of Biochemistry and Molecular Biology; Mayo Clinic College of Medicine & Science, Rochester, MN, 55905, USA
- Mayo Clinic Graduate School of Biomedical Sciences; Mayo Clinic College of Medicine & Science, Rochester, MN, 55905, USA
- Department of Cell Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Hanmei Bao
- Barshop Institute for Longevity and Aging Studies and Department of Medicine, Division of Diabetes; University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Cailin E McMahon
- Molecular Biology and Genetics Department; Cornell College of Agriculture and Life Sciences, Ithaca, NY, 14853, USA
| | - Yongbin Chen
- Department of Biochemistry and Molecular Biology; Mayo Clinic College of Medicine & Science, Rochester, MN, 55905, USA
| | - Stephanie D Burr
- Department of Biochemistry and Molecular Biology; Mayo Clinic College of Medicine & Science, Rochester, MN, 55905, USA
| | - Aaron M Anderson
- Department of Developmental Biology; Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Katja Madeyski-Bengtson
- Translational Genomics, Discovery Sciences; BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Daniel Lindén
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM); BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Division of Endocrinology, Department of Neuroscience and Physiology; Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies and Department of Medicine, Division of Diabetes; University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Jun Liu
- Department of Biochemistry and Molecular Biology; Mayo Clinic College of Medicine & Science, Rochester, MN, 55905, USA.
- Division of Endocrinology, Diabetes, Metabolism and Nutrition; Mayo Clinic in Rochester, Rochester, MN, 55905, USA.
| |
Collapse
|
2
|
Wang J, Li J, Fu Y, Zhu Y, Lin L, Li Y. Research progress, challenges and perspectives of phospholipids metabolism in the LXR‑LPCAT3 signaling pathway and its relation to NAFLD (Review). Int J Mol Med 2024; 53:32. [PMID: 38362962 PMCID: PMC10903931 DOI: 10.3892/ijmm.2024.5356] [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: 09/22/2023] [Accepted: 01/16/2024] [Indexed: 02/17/2024] Open
Abstract
Phospholipids (PLs) are principle constituents of biofilms, with their fatty acyl chain composition significantly impacting the biophysical properties of membranes, thereby influencing biological processes. Recent studies have elucidated that fatty acyl chains, under the enzymatic action of lyso‑phosphatidyl‑choline acyltransferases (LPCATs), expedite incorporation into the sn‑2 site of phosphatidyl‑choline (PC), profoundly affecting pathophysiology. Accumulating evidence suggests that alterations in LPCAT activity are implicated in various diseases, including non‑alcoholic fatty liver disease (NAFLD), hepatitis C, atherosclerosis and cancer. Specifically, LPCAT3 is instrumental in maintaining systemic lipid homeostasis through its roles in hepatic lipogenesis, intestinal lipid absorption and lipoprotein secretion. The liver X receptor (LXR), pivotal in lipid homeostasis, modulates cholesterol, fatty acid (FA) and PL metabolism. LXR's capacity to modify PL composition in response to cellular sterol fluctuations is a vital mechanism for protecting biofilms against lipid stress. Concurrently, LXR activation enhances LPCAT3 expression on cell membranes and elevates polyunsaturated PL levels. This activation can ameliorate saturated free FA effects in vitro or endoplasmic reticulum stress in vivo due to lipid accumulation in hepatic cells. Pharmacological interventions targeting LXR, LPCAT and membrane PL components could offer novel therapeutic directions for NAFLD management. The present review primarily focused on recent advancements in understanding the LPCAT3 signaling pathway's role in lipid metabolism related to NAFLD, aiming to identify new treatment targets for the disease.
Collapse
Affiliation(s)
- Junmin Wang
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Jiacheng Li
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Yugang Fu
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Yingying Zhu
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Liubing Lin
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Yong Li
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| |
Collapse
|
3
|
Danielewski M, Rapak A, Kruszyńska A, Małodobra-Mazur M, Oleszkiewicz P, Dzimira S, Kucharska AZ, Słupski W, Matuszewska A, Nowak B, Szeląg A, Piórecki N, Zaleska-Dorobisz U, Sozański T. Cornelian Cherry ( Cornus mas L.) Fruit Extract Lowers SREBP-1c and C/EBPα in Liver and Alters Various PPAR-α, PPAR-γ, LXR-α Target Genes in Cholesterol-Rich Diet Rabbit Model. Int J Mol Sci 2024; 25:1199. [PMID: 38256272 PMCID: PMC10816641 DOI: 10.3390/ijms25021199] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
Cornelian cherry (Cornus mas L.) fruits, abundant in iridoids and anthocyanins, are natural products with proven beneficial impacts on the functions of the cardiovascular system and the liver. This study aims to assess and compare whether and to what extent two different doses of resin-purified cornelian cherry extract (10 mg/kg b.w. or 50 mg/kg b.w.) applied in a cholesterol-rich diet rabbit model affect the levels of sterol regulatory element-binding protein 1c (SREBP-1c) and CCAAT/enhancer binding protein α (C/EBPα), and various liver X receptor-α (LXR-α), peroxisome proliferator-activated receptor-α (PPAR-α), and peroxisome proliferator-activated receptor-γ (PPAR-γ) target genes. Moreover, the aim is to evaluate the resistive index (RI) of common carotid arteries (CCAs) and aortas, and histopathological changes in CCAs. For this purpose, the levels of SREBP-1c, C/EBPα, ATP-binding cassette transporter A1 (ABCA1), ATP-binding cassette transporter G1 (ABCG1), fatty acid synthase (FAS), endothelial lipase (LIPG), carnitine palmitoyltransferase 1A (CPT1A), and adiponectin receptor 2 (AdipoR2) in liver tissue were measured. Also, the levels of lipoprotein lipase (LPL), visceral adipose tissue-derived serine protease inhibitor (Vaspin), and retinol-binding protein 4 (RBP4) in visceral adipose tissue were measured. The RI of CCAs and aortas, and histopathological changes in CCAs, were indicated. The oral administration of the cornelian cherry extract decreased the SREBP-1c and C/EBPα in both doses. The dose of 10 mg/kg b.w. increased ABCA1 and decreased FAS, CPT1A, and RBP4, and the dose of 50 mg/kg b.w. enhanced ABCG1 and AdipoR2. Mitigations in atheromatous changes in rabbits' CCAs were also observed. The obtained outcomes were compared to the results of our previous works. The beneficial results confirm that cornelian cherry fruit extract may constitute a potentially effective product in the prevention and treatment of obesity-related disorders.
Collapse
Affiliation(s)
- Maciej Danielewski
- Department of Pharmacology, Wroclaw Medical University, J. Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (W.S.); (A.M.); (B.N.); (A.S.)
| | - Andrzej Rapak
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland; (A.R.); (A.K.)
| | - Angelika Kruszyńska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland; (A.R.); (A.K.)
| | - Małgorzata Małodobra-Mazur
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, M. Sklodowskiej-Curie 52, 50-369 Wroclaw, Poland;
| | - Paweł Oleszkiewicz
- Department of Radiology and Imaging Diagnostics II, Lower Silesian Center of Oncology, Pulmonology and Hematology, Grabiszynska 105, 53-439 Wroclaw, Poland;
| | - Stanisław Dzimira
- Department of Pathology, Wroclaw University of Environmental and Life Sciences, C. K. Norwida 31, 50-375 Wroclaw, Poland;
| | - Alicja Z. Kucharska
- Department of Fruit, Vegetable, and Plant Nutraceutical Technology, Wroclaw University of Environmental and Life Sciences, J. Chelmonskiego 37, 51-630 Wroclaw, Poland;
| | - Wojciech Słupski
- Department of Pharmacology, Wroclaw Medical University, J. Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (W.S.); (A.M.); (B.N.); (A.S.)
| | - Agnieszka Matuszewska
- Department of Pharmacology, Wroclaw Medical University, J. Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (W.S.); (A.M.); (B.N.); (A.S.)
| | - Beata Nowak
- Department of Pharmacology, Wroclaw Medical University, J. Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (W.S.); (A.M.); (B.N.); (A.S.)
| | - Adam Szeląg
- Department of Pharmacology, Wroclaw Medical University, J. Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (W.S.); (A.M.); (B.N.); (A.S.)
| | - Narcyz Piórecki
- Bolestraszyce Arboretum and Institute of Physiography, Bolestraszyce 130, 37-722 Wyszatyce, Poland;
- Institute of Physical Culture Sciences, Medical College, University of Rzeszow, Cicha 2A, 35-326 Rzeszow, Poland
| | - Urszula Zaleska-Dorobisz
- Department of General and Pediatric Radiology, Wroclaw Medical University, M. Sklodowskiej-Curie 50/52, 50-369 Wroclaw, Poland;
| | - Tomasz Sozański
- Department of Preclinical Sciences, Pharmacology and Medical Diagnostics, Faculty of Medicine, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland;
| |
Collapse
|
4
|
Wang X, Chen Y, Meng H, Meng F. SREBPs as the potential target for solving the polypharmacy dilemma. Front Physiol 2024; 14:1272540. [PMID: 38269061 PMCID: PMC10806128 DOI: 10.3389/fphys.2023.1272540] [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: 09/04/2023] [Accepted: 12/26/2023] [Indexed: 01/26/2024] Open
Abstract
The phenomenon of polypharmacy is a common occurrence among older people with multiple health conditions due to the rapid increase in population aging and the popularization of clinical guidelines. The prevalence of metabolic syndrome is growing quickly, representing a serious threat to both the public and the worldwide healthcare systems. In addition, it enhances the risk of cardiovascular disease as well as mortality and morbidity. Sterol regulatory element binding proteins (SREBPs) are basic helix-loop-helix leucine zipper transcription factors that transcriptionally modulate genes that regulate lipid biosynthesis and uptake, thereby serving an essential role in biological systems regulation. In this article, we have described the structure of SREBPs and explored their activation and regulation of signals. We also reveal that SREBPs are intricately involved in the modulation of metabolic diseases and thus have tremendous potential as the novel target for single-drug therapy for multiple diseases.
Collapse
Affiliation(s)
| | | | | | - Fanbo Meng
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis (Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute), Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Bae S, Moon YA. Deletion of Elovl5 leads to dyslipidemia and atherosclerosis in LDLR-deficient mice. Biochem Biophys Res Commun 2024; 690:149292. [PMID: 38000296 DOI: 10.1016/j.bbrc.2023.149292] [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: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease for which hepatic steatosis and atherogenic dyslipidemia are significant risk factors. We investigated the effects of endogenously generated very-long-chain polyunsaturated fatty acids (VL-PUFAs) on dyslipidemia and atherosclerosis development using mice that lack ELOVL5, a PUFA elongase that is required for the synthesis of arachidonic acid, EPA, and DHA from the essential fatty acids linoleic and linolenic acids, and the LDL receptor (LDLR). Elovl5-/-;Ldlr-/- mice manifest increased liver triglyceride and cholesterol concentrations due to the activation of sterol regulatory element binding protein-1, a transcription factor that activates enzymes required for de novo lipogenesis. Plasma levels of triglycerides and cholesterol in VLDL, IDL, and LDL were markedly elevated in Elovl5-/-;Ldlr-/- mice fed a chow and the mice exhibited marked aortic atherosclerotic plaques. Bone marrow-derived monocytes from wild-type (WT) and Elovl5-/- mice were polarized to M1 and M2 macrophages, and the effects of ELOVL5 on inflammatory activity were determined. There were no differences in most of the markers tested for M1 and M2 polarized cells between WT and Elovl5-/- cells, except for a slight increase in PGE2 secretion in Elovl5-/- cells, likely due to elevated Cox-2 expression. These results suggest that the deletion of Elovl5 leads to hepatic steatosis and dyslipidemia, which are the major factors in severe atherosclerosis in Elovl5-/-;Ldlr-/- mice.
Collapse
Affiliation(s)
- Sijeong Bae
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, South Korea
| | - Young-Ah Moon
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, South Korea.
| |
Collapse
|
6
|
Xia Q, Lu F, Chen Y, Li J, Huang Z, Fang K, Hu M, Guo Y, Dong H, Xu L, Gong J. 6-Gingerol regulates triglyceride and cholesterol biosynthesis to improve hepatic steatosis in MAFLD by activating the AMPK-SREBPs signaling pathway. Biomed Pharmacother 2024; 170:116060. [PMID: 38147735 DOI: 10.1016/j.biopha.2023.116060] [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: 11/01/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 12/28/2023] Open
Abstract
Excessive synthesis of triglycerides and cholesterol accelerates the progression of hepatic steatosis in metabolic-associated fatty liver disease (MAFLD). However, the precise mechanism by which 6-gingerol mitigates hepatic steatosis in MAFLD model mice has yet to be fully understood. The present study observed that 6-gingerol administration exhibited significant protective effects against obesity, insulin resistance, and hepatic steatosis in mice subjected to a high-fat diet (HFD), and mitigated lipid accumulation in HepG2 cells treated with palmitate (PA). Following the hepatic lipidomic analysis, we confirmed that the AMPK-SREBPs signaling pathway as the underlying molecular mechanism by which 6-gingerol inhibited triglyceride and cholesterol biosynthesis, both in vivo and in vitro, through Western blot and immunofluorescence assay. Additionally, the application of an AMPK agonist/inhibitor further validated that 6-gingerol promoted AMPK activation by increasing the phosphorylation level of AMPK in vitro. Notably, the inhibitory effect of 6-gingerol on cholesterol biosynthesis, rather than triglyceride biosynthesis, was significantly diminished after silencing SREBP2 using a lentiviral plasmid shRNA in HepG2 cells. Our study demonstrates that 6-gingerol mitigates hepatic triglyceride and cholesterol biosynthesis to alleviate hepatic steatosis by activating the AMPK-SREBPs signaling pathway, indicating that 6-gingerol may be a potential candidate in the therapy of MAFLD.
Collapse
Affiliation(s)
- Qingsong Xia
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China; Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Fuer Lu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Yu Chen
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Jingbin Li
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Zhaoyi Huang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Ke Fang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Meilin Hu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Yujin Guo
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Hui Dong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Lijun Xu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Jing Gong
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| |
Collapse
|
7
|
Kvasnička A, Friedecký D, Brumarová R, Pavlíková M, Pavelcová K, Mašínová J, Hasíková L, Závada J, Pavelka K, Ješina P, Stibůrková B. Alterations in lipidome profiles distinguish early-onset hyperuricemia, gout, and the effect of urate-lowering treatment. Arthritis Res Ther 2023; 25:234. [PMID: 38042879 PMCID: PMC10693150 DOI: 10.1186/s13075-023-03204-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 11/03/2023] [Indexed: 12/04/2023] Open
Abstract
BACKGROUND Currently, it is not possible to predict whether patients with hyperuricemia (HUA) will develop gout and how this progression may be affected by urate-lowering treatment (ULT). Our study aimed to evaluate differences in plasma lipidome between patients with asymptomatic HUA detected ≤ 40 years (HUA ≤ 40) and > 40 years, gout patients with disease onset ≤ 40 years (Gout ≤ 40) and > 40 years, and normouricemic healthy controls (HC). METHODS Plasma samples were collected from 94 asymptomatic HUA (77% HUA ≤ 40) subjects, 196 gout patients (59% Gout ≤ 40), and 53 HC. A comprehensive targeted lipidomic analysis was performed to semi-quantify 608 lipids in plasma. Univariate and multivariate statistics and advanced visualizations were applied. RESULTS Both HUA and gout patients showed alterations in lipid profiles with the most significant upregulation of phosphatidylethanolamines and downregulation of lysophosphatidylcholine plasmalogens/plasmanyls. More profound changes were observed in HUA ≤ 40 and Gout ≤ 40 without ULT. Multivariate statistics differentiated HUA ≤ 40 and Gout ≤ 40 groups from HC with an overall accuracy of > 95%. CONCLUSION Alterations in the lipidome of HUA and Gout patients show a significant impact on lipid metabolism. The most significant glycerophospholipid dysregulation was found in HUA ≤ 40 and Gout ≤ 40 patients, together with a correction of this imbalance with ULT.
Collapse
Affiliation(s)
- Aleš Kvasnička
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czech Republic
| | - David Friedecký
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czech Republic
| | - Radana Brumarová
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czech Republic
| | - Markéta Pavlíková
- Department of Probability and Mathematical Statistics, Faculty of Mathematics and Physics, Charles University, Prague, Czech Republic
| | - Kateřina Pavelcová
- Institute of Rheumatology, Na Slupi 4, 128 50 Prague 2, Prague, Czech Republic
| | - Jana Mašínová
- Institute of Rheumatology, Na Slupi 4, 128 50 Prague 2, Prague, Czech Republic
| | - Lenka Hasíková
- Institute of Rheumatology, Na Slupi 4, 128 50 Prague 2, Prague, Czech Republic
| | - Jakub Závada
- Institute of Rheumatology, Na Slupi 4, 128 50 Prague 2, Prague, Czech Republic
| | - Karel Pavelka
- Institute of Rheumatology, Na Slupi 4, 128 50 Prague 2, Prague, Czech Republic
| | - Pavel Ješina
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Blanka Stibůrková
- Institute of Rheumatology, Na Slupi 4, 128 50 Prague 2, Prague, Czech Republic.
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic.
| |
Collapse
|
8
|
Wu D, Wang L, Fan Z, Li J, Tang S, Zhao C, Zhang H, Zheng X. Comprehensive assessment of detoxification mechanisms of hydrolysis fish peptides in largemouth bass (Micropterus salmoides) under copper exposure: Tracing from bioaccumulation, oxidative stress, lipid deposition to metabolomics. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 264:115418. [PMID: 37651792 DOI: 10.1016/j.ecoenv.2023.115418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/23/2023] [Accepted: 08/26/2023] [Indexed: 09/02/2023]
Abstract
As a heavy metal, copper is toxic to aquatic organisms in water, causing oxidative stress and lipid deposition. However, there is currently no effective dietary strategy to prevent damage caused by copper exposure. Here, copper bioaccumulation, antioxidant enzymes, lipogenic enzymes, lipid metabolism-related gene expression levels and metabolic pathways were synthesized and evaluated in copper-exposed largemouth bass (Micropterus salmoides) after hydrolysis fish peptides (HFP) pretreatment. The results showed that supplementation with 1% (P < 0.05), 3% (P < 0.01) and 5% (P < 0.05) HFP significantly reduced the copper bioaccumulation in largemouth bass. Hydrolysis fish peptides supplementation significantly reduced the activities of total antioxidant capacity (P < 0.01) and catalase (P < 0.01) and the contents of glutathione (P < 0.01) and malondialdehyde (P < 0.05). Fatty acid synthetase concentration was significantly reduced in fish supplemented with 3% (P < 0.05) and 5% HFP (P < 0.05). Similarly, fish fed 3% (P < 0.05) and 5% (P < 0.01) HFP significantly reduced the glucose-6-phosphate dehydrogenase concentration. Serum metabolomics revealed that 85, 144 and 207 differential metabolites were obtained in fish supplemented with 1%, 3% and 5% HFP, respectively. The differential metabolites were mainly lipids and lipid-like molecules, which were associated with the lipid metabolism pathways. The expression levels of fatty acid synthase (P < 0.01), sterol regulatory element binding protein-1c (P < 0.05), liver X receptor (P < 0.001), peroxisome proliferator activated γ (P < 0.01), apolipoprotein B (P < 0.001) and fatty acid-binding protein 1 (P < 0.01) were significantly down-regulated and the expression levels of carnitine palmitoyltransferase 1α (P < 0.01), hormone-sensitive lipase (P < 0.001), apolipoprotein A 1 (P < 0.05) were significantly up-regulated in fish fed with 3% HFP. Additionally, supplementation with 3% (P < 0.01) and 5% (P < 0.001) HFP significantly up-regulated the expression level of B-cell lymphoma-2 with a dose-dependent effect. In conclusion, our study confirmed that HFP supplementation was closely associated with oxidative stress, enzymatic activities and related pathways of lipid metabolism, and apoptosis, and in general alleviated lipid deposition caused by copper exposure in largemouth bass.
Collapse
Affiliation(s)
- Di Wu
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China; Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Harbin 150070, China
| | - Liansheng Wang
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China; Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Harbin 150070, China.
| | - Ze Fan
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China; Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Harbin 150070, China
| | - Jinnan Li
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China; Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Harbin 150070, China
| | - Shizhan Tang
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China; Ministry of Agriculture and Rural Affairs Fishery Environment and Aquatic Product Quality Supervision, Inspection and Testing Center, Harbin 150070, China
| | - Chen Zhao
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China; Heilongjiang River Basin Fishery Ecological Environment Monitoring Center, Ministry of Agriculture and Rural Affairs, Harbin 150010, China
| | - Haitao Zhang
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture and Rural Affairs, Guangdong Evergreen Feed Industry Co., Ltd., Zhanjiang 524000, China
| | - Xianhu Zheng
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China; National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Harbin 150070, China
| |
Collapse
|
9
|
Johnson S, Bao H, McMahon C, Chen Y, Burr S, Anderson A, Madeyski-Bengtson K, Lindén D, Han X, Liu J. Substrate-Specific Function of PNPLA3 Facilitates Hepatic VLDL-Triglyceride Secretion During Stimulated Lipogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.30.553213. [PMID: 37693552 PMCID: PMC10491159 DOI: 10.1101/2023.08.30.553213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The I148M variant of PNPLA3 is strongly linked to hepatic steatosis. Evidence suggests a gain-of-function role for the I148M mutant as an ATGL inhibitor, leaving the physiological relevance of wild-type PNPLA3 undefined. Here we show that PNPLA3 selectively degrades triglycerides (TGs) enriched in polyunsaturated fatty acids (PUFAs) independently of ATGL in cultured cells and mice. Lipidomics and metabolite tracing analyses demonstrated that PNPLA3 mobilizes PUFAs from intracellular TGs for phospholipid desaturation, supporting hepatic secretion of TG-rich lipoproteins. Consequently, mice with liver-specific knockout or acute knockdown of PNPLA3 both exhibited aggravated liver steatosis and concomitant decreases in plasma VLDL-TG, phenotypes that manifest only under lipogenic conditions. I148M-knockin mice similarly displayed impaired hepatic TG secretion during lipogenic stimulation. Our results highlight a specific context whereby PNPLA3 facilitates the balance between hepatic TG storage and secretion and suggest the potential contributions of I148M variant loss-of-function to the development of hepatic steatosis in humans. Summary Statement We define the physiological role of wild type PNPLA3 in maintaining hepatic VLDL-TG secretion.
Collapse
|
10
|
Kim HL, Jung Y, Kim HI, Sung NY, Kim MJ, Han IJ, Kim G, Nho EY, Park SY, Han Y, Jung JH, Kim DS, Park J. Antidiabetic Effect of Fermented Mesembryanthemum crystallinum L. in db/ db Mice Involves Regulation of PI3K-Akt Pathway. Curr Issues Mol Biol 2023; 45:6415-6431. [PMID: 37623224 PMCID: PMC10453056 DOI: 10.3390/cimb45080405] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023] Open
Abstract
Type 2 diabetes (T2D) is a serious health issue with increasing incidences worldwide. However, current medications have limitations due to side effects such as decreased appetite, stomach pain, diarrhea, and extreme tiredness. Here, we report the effect of fermented ice plant (FMC) in the T2M mouse model of db/db mice. FMC showed a greater inhibition of lipid accumulation compared to unfermented ice plant extract. Two-week oral administration with FMC inhibited body weight gain, lowered fasting blood glucose, and improved glucose tolerance. Serum parameters related to T2D including insulin, glycosylated hemoglobin, adiponectin, and cholesterols were improved as well. Histological analysis confirmed the protective effect of FMC on pancreas and liver destruction. FMC treatment significantly increased the expression and phosphorylation of IRS-1, PI3K, and AKT. Additionally, AMP-activated protein kinase phosphorylation and nuclear factor erythroid 2-related factor 2 were also increased in the liver tissues of db/db mice treated with FMC. Overall, our results indicate the anti-diabetic effect of FMC; therefore, we suggest that FMC may be useful as a therapeutic agent for T2D.
Collapse
Affiliation(s)
- Hye-Lin Kim
- College of Korean Medicine, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Yunu Jung
- Division of Natural Product Research, Korea Prime Pharmacy Co., Ltd., Suwon 16229, Republic of Korea (E.Y.N.); (S.-Y.P.)
| | - Hyo In Kim
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Nak-Yun Sung
- Division of Natural Product Research, Korea Prime Pharmacy Co., Ltd., Suwon 16229, Republic of Korea (E.Y.N.); (S.-Y.P.)
| | - Min-Jee Kim
- Division of Natural Product Research, Korea Prime Pharmacy Co., Ltd., Suwon 16229, Republic of Korea (E.Y.N.); (S.-Y.P.)
| | - In-Jun Han
- Division of Natural Product Research, Korea Prime Pharmacy Co., Ltd., Suwon 16229, Republic of Korea (E.Y.N.); (S.-Y.P.)
| | - Geon Kim
- Division of Natural Product Research, Korea Prime Pharmacy Co., Ltd., Suwon 16229, Republic of Korea (E.Y.N.); (S.-Y.P.)
| | - Eun Yeong Nho
- Division of Natural Product Research, Korea Prime Pharmacy Co., Ltd., Suwon 16229, Republic of Korea (E.Y.N.); (S.-Y.P.)
| | - Sang-Yun Park
- Division of Natural Product Research, Korea Prime Pharmacy Co., Ltd., Suwon 16229, Republic of Korea (E.Y.N.); (S.-Y.P.)
| | - Yohan Han
- College of Korean Medicine, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Ji Hoon Jung
- College of Korean Medicine, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Dong-Sub Kim
- Division of Natural Product Research, Korea Prime Pharmacy Co., Ltd., Suwon 16229, Republic of Korea (E.Y.N.); (S.-Y.P.)
| | - Jinbong Park
- College of Korean Medicine, Kyung Hee University, Seoul 02453, Republic of Korea
| |
Collapse
|
11
|
Ma S, Pang X, Tian S, Sun J, Hu Q, Li X, Lu Y. The protective effects of sulforaphane on high-fat diet-induced metabolic associated fatty liver disease in mice via mediating the FXR/LXRα pathway. Food Funct 2022; 13:12966-12982. [PMID: 36448414 DOI: 10.1039/d2fo02341e] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Metabolic-associated fatty liver disease (MAFLD) is becoming the key factor in causing chronic liver disease all over the world. Sulforaphane (SFN) has been proven to be effective in alleviating many metabolic diseases, such as obesity and type 2 diabetes. In this study, C57BL/6 mice were fed a high-fat diet for 12 weeks to induce MAFLD and given SFN (10 mg per kg bw) daily. Our results showed that SFN not only improved the excessive accumulation of fat in the liver cells but also ameliorated liver and serum inflammatory and antioxidant levels. In addition, SFN can regulate bile-acid metabolism and fatty-acid synthesis by affecting their farnesoid X receptor (FXR)/liver X receptor alpha (LXRα) signaling pathway, ultimately alleviating MAFLD. Our study provides a theoretical basis for the mechanism by which SFN alleviates hepatic steatosis.
Collapse
Affiliation(s)
- Shaotong Ma
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Xinyi Pang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Shuhua Tian
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Jing Sun
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Qiaobin Hu
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Xiangfei Li
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Yingjian Lu
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| |
Collapse
|
12
|
Fujita S, Nishizawa H, Miyashita Y, Imada T, Yamaguchi T, Murano T, Bujo H, Asano Y, Kozawa J, Maeda N, Shimomura I. Genetic assessment using whole-exome sequencing for a young hypertriglyceridemic patient with repeated acute pancreatitis. Endocr J 2022; 69:1101-1108. [PMID: 35387941 DOI: 10.1507/endocrj.ej22-0024] [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] [Indexed: 11/23/2022] Open
Abstract
Hypertriglyceridemia is caused not only by environmental factors but also by genetic factors. Severe hypertriglyceridemia is prone to complications of acute pancreatitis. Here, we report a whole-exome sequencing (WES) analysis for a young hypertriglyceridemic patient with recurrent acute pancreatitis and the patient's mother. A 28-year-old hypertriglyceridemic female was admitted to our hospital. At 23 years old, a health checkup clarified her hypertriglyceridemia. At the age of 26 and 27, she had repeated acute pancreatitis with severe hypertriglyceridemia (serum triglyceride level were 3,888 mg/dL and 12,080 mg/dL, respectively). The patient's BMI was 29.0 kg/m2, and blood samples under fibrate medication showed triglyceride 451 mg/dL and HbA1c 7.2%. Type V dyslipidemia became more apparent at postprandial state. The WES analysis showed that the patients had two heterozygous variants in Apolipoprotein A5 (APOA5) gene (p.G185C and p.V153M), a heterozygous variant in Apolipoprotein E (APOE) gene (p.R176C), three heterozygous variants in Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) gene (p.T1220I, p.R1453W and p.V470M). On the other hand, her mother, who had moderate hypertriglyceridemia without acute pancreatitis, had a heterozygous variant in APOA5 gene (p.G185C) and two heterozygous variants in CFTR gene (p.T1220I and p.V470M). These results suggest that the more severe pathology of the patient than her mother might be due to the possible compound heterozygous APOA5 variants, the heterozygous APOE variant, and the possible compound heterozygous CFTR variants. In this case, WES analyses were useful to evaluate not only the causative genes of hypertriglyceridemia (APOA5 and APOE) but also the genes involved in the development of acute pancreatitis (CFTR) simultaneously.
Collapse
Affiliation(s)
- Shingo Fujita
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Hitoshi Nishizawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Yohei Miyashita
- Department of Legal Medicine, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Tasuku Imada
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Takashi Yamaguchi
- Center of Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, Chiba 285-8741, Japan
| | - Takeyoshi Murano
- Clinical Laboratory Program, Faculty of Science, Toho University, Chiba 274-8510, Japan
| | - Hideaki Bujo
- Department of Clinical-Laboratory and Experimental-Research Medicine, Toho University Sakura Medical Center, Chiba 285-8741, Japan
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Junji Kozawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
- Department of Diabetes Care Medicine, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Norikazu Maeda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
- Department of Metabolism and Atherosclerosis, Graduate School of Medicine Osaka University, Osaka 565-0871, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
13
|
Liu W, Shang J, Deng Y, Han X, Chen Y, Wang S, Yang R, Dong F, Shang H. Network pharmacology analysis on mechanism of Jian Pi Qing Gan Yin decoction ameliorating high fat diet-induced non-alcoholic fatty liver disease and validated in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2022; 295:115382. [PMID: 35577161 DOI: 10.1016/j.jep.2022.115382] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 04/24/2022] [Accepted: 05/10/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jian Pi Qing Gan Yin (JPQGY) has been used clinically to relieve non-alcoholic fatty liver disease (NAFLD) in China for decades; however, the underlying mechanisms of JPQGY remain unclear. AIM OF THE STUDY We evaluated the effects and mechanisms of JPQGY and hepatic steatosis caused by the middle stage of 13-week-high-fat-diet-induced NAFLD in mice. MATERIALS AND METHODS Different dosages of JPQGY (5.5, 11, and 22 g/kg/day) were administered to NAFLD mice simultaneously. Body weight, body mass index (BMI), and liver lipid- and inflammation-related serum indicators were measured enzymatically. Liver samples were stained with Oil Red O and hematoxylin and eosin (H&E). Next, we performed a network pharmacology analysis and verified eight target genes mapping to NAFLD-related lipid metabolism pathways. The mRNA/protein expression was analyzed by real-time polymerase chain reaction (PCR) and western blotting. RESULTS JPQGY significantly relieved histological damage (steatosis-inflammation-fibrosis), prevented the downregulation of AMPK and Pparα, and upregulated LXRα, Srebp-1c, F4/80, Nf-κb, and Cyp2e1 in the HFD-induced NAFLD mouse model. CONCLUSIONS The present results suggest that chronic treatment with JPQGY ameliorated HFD-induced NAFLD in mice by targeting the first and second phases of hepatic steatosis by stimulating the AMPK/PPARα pathway and inhibiting the LXRα/Srebp1/Nf-κb pathway. Our findings provide evidence that supports the clinical use of this formula for high-fat diet-induced fatty liver disease.
Collapse
Affiliation(s)
- Weiwei Liu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China; Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Jingyu Shang
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Yinxiang Deng
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China; Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Xiuzhen Han
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China; Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Yugen Chen
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China; Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Shuangshuang Wang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China; Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Ruwen Yang
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Fan Dong
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Hongtao Shang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China; Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China.
| |
Collapse
|
14
|
Huang YQ, Tang YX, Qiu BH, Talukder M, Li XN, Li JL. Di-2-ethylhexyl phthalate (DEHP) induced lipid metabolism disorder in liver via activating the LXR/SREBP-1c/PPARα/γ and NF-κB signaling pathway. Food Chem Toxicol 2022; 165:113119. [PMID: 35537648 DOI: 10.1016/j.fct.2022.113119] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 04/02/2022] [Accepted: 05/04/2022] [Indexed: 02/07/2023]
Affiliation(s)
- Yue-Qiang Huang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yi-Xi Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Bai-Hao Qiu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Milton Talukder
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Department of Physiology and Pharmacology, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal, 8210, Bangladesh
| | - Xue-Nan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
15
|
Zhang Q, Liu X, Piao C, Jiao Z, Ma Y, Wang Y, Liu T, Xu J, Wang H. Effect of conditioned medium from adipose derived mesenchymal stem cells on endoplasmic reticulum stress and lipid metabolism after hepatic ischemia reperfusion injury and hepatectomy in swine. Life Sci 2022; 289:120212. [PMID: 34896163 DOI: 10.1016/j.lfs.2021.120212] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/23/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023]
Abstract
AIMS Hepatic ischemia reperfusion injury (HIRI) is associated with liver failure after liver transplantation and hepatectomy. Thus, this study aims to explore the effect of conditioned medium from adipose derived stem cells (ADSC-CM) on endoplasmic reticulum stress (ERS) and lipid metabolism after HIRI combined with hepatectomy in miniature pigs. MAIN METHODS A model of HIRI combined with hepatectomy in miniature pigs was established. The expression of ERS-related proteins and lipid metabolism related genes, as well as triglyceride (TG), total cholesterol (TC), high density lipoprotein (HDL), very low density lipoprotein (VLDL) and acetyl-CoA carboxylase 1 (ACC1) level were measured in liver tissues. KEY FINDINGS Both ADSCs and ADSC-CM could improve the damage in the ultrastructure of hepatocytes. ADSC-CM significantly decreased the protein expression of GRP78, ATF6, XBP1, p-eIF2α, ATF4, p-JNK and CHOP. Oil red O staining revealed that the degree of hepatocyte steatosis was also significantly reduced after treatment with ADSC-CM. In addition, ADSC-CM remarkably decreased TG, TC, HDL and ACC1 level in liver tissues, while enhanced VLDL content. Finally, SREBP1, SCAP, FASN, ACC1, HMGCR and HMGCS1 mRNA expression was also markedly downregulated in liver tissues. SIGNIFICANCE Injection of ADSC-CM into the hepatic parenchymal could represent a novel cell-free therapeutic approach to improve HIRI combined with hepatectomy injury. The inhibition of ERS and the improvement of lipid metabolism in the hepatocytes might be a potential mechanism used by ADSC-CM to prevent liver injury from HIRI combined with hepatectomy.
Collapse
Affiliation(s)
- Qianzhen Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, PR China
| | - Xiaoning Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Chenxi Piao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Zhihui Jiao
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, PR China
| | - Yajun Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yue Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Tao Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jiayuan Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Hongbin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
16
|
Lands B. Lipid nutrition: "In silico" studies and undeveloped experiments. Prog Lipid Res 2021; 85:101142. [PMID: 34818526 DOI: 10.1016/j.plipres.2021.101142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 12/14/2022]
Abstract
This review examines lipids and lipid-binding sites on proteins in relation to cardiovascular disease. Lipid nutrition involves food energy from ingested fatty acids plus fatty acids formed from excess ingested carbohydrate and protein. Non-esterified fatty acids (NEFA) and lipoproteins have many detailed attributes not evident in their names. Recognizing attributes of lipid-protein interactions decreases unexpected outcomes. Details of double bond position and configuration interacting with protein binding sites have unexpected consequences in acyltransferase and cell replication events. Highly unsaturated fatty acids (HUFA) have n-3 and n-6 motifs with documented differences in intensity of destabilizing positive feedback loops amplifying pathophysiology. However, actions of NEFA have been neglected relative to cholesterol, which is co-produced from excess food. Native low-density lipoproteins (LDL) bind to a high-affinity cell surface receptor which poorly recognizes biologically modified LDLs. NEFA increase negative charge of LDL and decrease its processing by "normal" receptors while increasing processing by "scavenger" receptors. A positive feedback loop in the recruitment of monocytes and macrophages amplifies chronic inflammatory pathophysiology. Computer tools combine multiple components in lipid nutrition and predict balance of energy and n-3:n-6 HUFA. The tools help design and execute precise clinical nutrition monitoring that either supports or disproves expectations.
Collapse
Affiliation(s)
- Bill Lands
- Fellow ASN, AAAS, SFRBM, ISSFAL, College Park, MD, USA.
| |
Collapse
|
17
|
Okazaki H, Gotoda T, Ogura M, Ishibashi S, Inagaki K, Daida H, Hayashi T, Hori M, Masuda D, Matsuki K, Yokoyama S, Harada-Shiba M. Current Diagnosis and Management of Primary Chylomicronemia. J Atheroscler Thromb 2021; 28:883-904. [PMID: 33980761 PMCID: PMC8532063 DOI: 10.5551/jat.rv17054] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Primary chylomicronemia (PCM) is a rare and intractable disease characterized by marked accumulation of chylomicrons in plasma. The levels of plasma triglycerides (TGs) typically range from 1,000 - 15,000 mg/dL or higher.
PCM is caused by defects in the lipoprotein lipase (LPL) pathway due to genetic mutations, autoantibodies, or unidentified causes. The monogenic type is typically inherited as an autosomal recessive trait with loss-of-function mutations in LPL pathway genes (
LPL
,
LMF1
,
GPIHBP1
,
APOC2
, and
APOA5
). Secondary/environmental factors (diabetes, alcohol intake, pregnancy, etc.) often exacerbate hypertriglyceridemia (HTG).
The signs, symptoms, and complications of chylomicronemia include eruptive xanthomas, lipemia retinalis, hepatosplenomegaly, and acute pancreatitis with onset as early as in infancy. Acute pancreatitis can be fatal and recurrent episodes of abdominal pain may lead to dietary fat intolerance and failure to thrive. The main goal of treatment is to prevent acute pancreatitis by reducing plasma TG levels to at least less than 500-1,000 mg/dL. However, current TG-lowering medications are generally ineffective for PCM. The only other treatment options are modulation of secondary/environmental factors. Most patients need strict dietary fat restriction, which is often difficult to maintain and likely affects their quality of life. Timely diagnosis is critical for the best prognosis with currently available management, but PCM is often misdiagnosed and undertreated. The aim of this review is firstly to summarize the pathogenesis, signs, symptoms, diagnosis, and management of PCM, and secondly to propose simple diagnostic criteria that can be readily translated into general clinical practice to improve the diagnostic rate of PCM. In fact, these criteria are currently used to define eligibility to receive social support from the Japanese government for PCM as a rare and intractable disease. Nevertheless, further research to unravel the molecular pathogenesis and develop effective therapeutic modalities is warranted. Nationwide registry research on PCM is currently ongoing in Japan with the aim of better understanding the disease burden as well as the unmet needs of this life-threatening disease with poor therapeutic options.
Collapse
Affiliation(s)
- Hiroaki Okazaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Takanari Gotoda
- Department of Metabolic Biochemistry, Faculty of Medicine, Kyorin University
| | - Masatsune Ogura
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University
| | - Kyoko Inagaki
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Nippon Medical School
| | - Hiroyuki Daida
- Faculty of Health Science, Juntendo University, Juntendo University Graduate School of Medicine
| | - Toshio Hayashi
- School of Health Sciences, Nagoya University Graduate School of Medicine
| | - Mika Hori
- Department of Endocrinology, Research Institute of Environmental Medicine, Nagoya University
| | - Daisaku Masuda
- Department of Cardiology, Health Care Center, Rinku Innovation Center for Wellness Care and Activities (RICWA), Rinku General Medical Center
| | - Kota Matsuki
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine
| | | | - Mariko Harada-Shiba
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center Research Institute
| | | |
Collapse
|
18
|
IL-2Rβγ signalling in lymphocytes promotes systemic inflammation and reduces plasma cholesterol in atherosclerotic mice. Atherosclerosis 2021; 326:1-10. [PMID: 33945906 DOI: 10.1016/j.atherosclerosis.2021.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 04/08/2021] [Accepted: 04/21/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIMS The relationship between inflammation and lipid metabolism is complex and bidirectional. Lymphocyte-driven inflammation has been shown to modulate both atherosclerotic plaque development and cholesterol levels, but the mechanisms are incompletely understood. METHODS The cardiometabolic effects of IL-2Rβγ signalling in atherosclerotic Apoe-/- mice were investigated by treatment with an agonistic IL-2Rβγ-targeting IL-2/anti-IL-2 complex or a monoclonal anti-CD122 (IL-2Rβ) blocking antibody. RESULTS Administration of IL-2Rβγ agonistic IL-2/anti-IL-2 complexes to Apoe-/- mice augmented opposing arms of the adaptive immune system. Expansion of effector/memory T cells and increased levels of circulating pro-inflammatory cytokines were observed along with elevated levels of regulatory T cells and IL-10. Notably, IL-2/anti-IL-2 treatment did not affect plaque size but decreased levels of plasma cholesterol. The cholesterol lowering effect of IL-2Rβγ agonism was not affected by anti-CD8 or anti-NK1.1 depleting antibody treatment but was contingent on the presence of adaptive immunity. Expression of multiple liver X receptor (LXR)-related genes, including Pltp and Srebp1c in the liver, was decreased by IL-2/anti-IL-2 treatment. Although IL-2Rβγ agonism lowered cholesterol levels, blocking IL-2Rβγ signalling using an anti-CD122 monoclonal antibody did not impact cholesterol levels or plaque burden in Apoe-/- mice. CONCLUSIONS Elevated IL-2Rβγ signalling results in activation of both inflammatory and regulatory lymphocytes with a net zero effect on atherosclerosis and decreased plasma cholesterol levels. Changes in cholesterol levels were associated with reductions in hepatic LXR-related gene expression. Further studies are needed to investigate the clinical significance of IL-2 mediated modulation of hepatic LXR signalling in inflammatory disorders.
Collapse
|
19
|
Zhang K, Zheng J, Chen Y, Dong J, Li Z, Chiang YP, He M, Huang Q, Tang H, Jiang XC. Inducible phospholipid transfer protein deficiency ameliorates atherosclerosis. Atherosclerosis 2021; 324:9-17. [PMID: 33798923 DOI: 10.1016/j.atherosclerosis.2021.03.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/23/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Atherosclerosis progression and regression studies are related to its prevention and treatment. Although we have gained extensive knowledge on germline phospholipid transfer protein (PLTP) deficiency, the effect of inducible PLTP deficiency in atherosclerosis remains unexplored. METHODS We generated inducible PLTP (iPLTP)-knockout (KO) mice and measured their plasma lipid levels after feeding a normal chow or a Western-type diet. Adenovirus associated virus-proprotein convertase subtilisin/kexin type 9 (AAV-PCSK9) was used to induce hypercholesterolemia in the mice. Collars were placed around the common carotid arteries, and atherosclerosis progression and regression in the carotid arteries and aortic roots were evaluated. RESULTS On a normal chow diet, iPLTP-KO mice exhibited decreased cholesterol, phospholipid, apoA-I, and apoB levels compared with control mice. Furthermore, the overall amount of high-density lipoprotein (HDL) particles was reduced in these mice, but this effect was more profound for larger HDL particles. On a Western-type diet, iPLTP-KO mice again exhibited reduced levels of all tested lipids, even though the basal lipid levels were increased. Additionally, these mice displayed significantly reduced atherosclerotic plaque sizes with increased plaque stability. Importantly, inducible PLTP deficiency significantly ameliorated atherosclerosis by reducing the size of established plaques and the number of macrophages in the plaques without causing lipid accumulation in the liver. CONCLUSIONS Induced PLTP deficiency in adult mice reduces plasma total cholesterol and triglycerides, prevents atherosclerosis progression, and promotes atherosclerosis regression. Thus, PLTP inhibition is a promising therapeutic approach for atherosclerosis.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA; Department of Emergency, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jiao Zheng
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA; Beijing University of Chinese Medicine, Beijing, China
| | | | | | - Zhiqiang Li
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA; Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, Brooklyn, New York, USA
| | - Yeun-Po Chiang
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Mulin He
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | | | | | - Xian-Cheng Jiang
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA; Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, Brooklyn, New York, USA.
| |
Collapse
|
20
|
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: 21] [Impact Index Per Article: 7.0] [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
|
21
|
Berger JM, Moon YA. Increased Hepatic Lipogenesis Elevates Liver Cholesterol Content. Mol Cells 2021; 44:116-125. [PMID: 33658436 PMCID: PMC7941001 DOI: 10.14348/molcells.2021.2147] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/17/2020] [Accepted: 02/07/2021] [Indexed: 01/01/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the most common cause of death in patients with nonalcoholic fatty liver disease (NAFLD) and dyslipidemia is considered at least partially responsible for the increased CVD risk in NAFLD patients. The aim of the present study is to understand how hepatic de novo lipogenesis influences hepatic cholesterol content as well as its effects on the plasma lipid levels. Hepatic lipogenesis was induced in mice by feeding a fat-free/high-sucrose (FF/HS) diet and the metabolic pathways associated with cholesterol were then analyzed. Both liver triglyceride and cholesterol contents were significantly increased in mice fed an FF/HS diet. Activation of fatty acid synthesis driven by the activation of sterol regulatory element binding protein (SREBP)-1c resulted in the increased liver triglycerides. The augmented cholesterol content in the liver could not be explained by an increased cholesterol synthesis, which was decreased by the FF/HS diet. HMGCoA reductase protein level was decreased in mice fed an FF/HS diet. We found that the liver retained more cholesterol through a reduced excretion of bile acids, a reduced fecal cholesterol excretion, and an increased cholesterol uptake from plasma lipoproteins. Very low-density lipoproteintriglyceride and -cholesterol secretion were increased in mice fed an FF/HS diet, which led to hypertriglyceridemia and hypercholesterolemia in Ldlr-/- mice, a model that exhibits a more human like lipoprotein profile. These findings suggest that dietary cholesterol intake and cholesterol synthesis rates cannot only explain the hypercholesterolemia associated with NAFLD, and that the control of fatty acid synthesis should be considered for the management of dyslipidemia.
Collapse
Affiliation(s)
- Jean-Mathieu Berger
- Departments of Internal Medicine and Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Young-Ah Moon
- Department of Molecular Medicine, Inha University College of Medicine, Incheon 22212, Korea
| |
Collapse
|
22
|
Zhang Y, Gu Y, Chen Y, Huang Z, Li M, Jiang W, Chen J, Rao W, Luo S, Chen Y, Chen J, Li L, Jia Y, Liu M, Zhou F. Dingxin Recipe IV attenuates atherosclerosis by regulating lipid metabolism through LXR-α/SREBP1 pathway and modulating the gut microbiota in ApoE -/- mice fed with HFD. JOURNAL OF ETHNOPHARMACOLOGY 2021; 266:113436. [PMID: 33011372 DOI: 10.1016/j.jep.2020.113436] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/25/2020] [Accepted: 09/27/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dingxin Recipe (DXR) is a traditional Chinese medicine formula that has been reported to be effective and safe treatment for cardiovascular diseases, such as arrhythmias, coronary heart disease. Dingxin Recipe IV (DXR IV) was further improved from the DXR according to the traditional use. However, the mechanism of DXR IV in atherosclerosis is unclear. AIM OF THE STUDY This study aimed to illustrate whether DXR IV improve atherosclerosis through modulating the lipid metabolism and gut microbiota in atherosclerosis mice. MATERIALS AND METHODS 40 male ApoE-/- mice were fed on HFD for 12 weeks and were then treated with DXR IV (1.8, 0.9, or 0.45 g/kg/d) for another 12 weeks. The decroation of DXR IV contains four traditional Chinese medicines: the dried rhizome of Coptis chinensis Franch. (15.09%), the root of Salvia miltiorrhiza Bunge (28.30%), the seed of Ziziphus jujuba Mill. (37.74%) and the fruiting body of Ganoderma lucidum (Leyss.ex Fr.) Karst. (18.87%). 8 male c57BL/6 mice fed a normal diet served as control group. The atherosclerotic plaque was quantified by oil-red O staining and masson trichrome staining. Mice feces were collected. The gut micobiota were detected by 16S rRNA gene sequencing and fecal metabolites were analyzed by 1H NMR spectroscopy. The effect of DXR IV on blood lipids (TG, TC, LDL-C, HDL-C) was investigated. The lipid metabolism related genes were determined by RT-qPCR and western blotting respectively. RESULTS DXR IV exerted the anti-atherosclerosis effect by inhibiting the excessive cholesterol deposition in aorta and regulating the level of TG, TC, LDL-C and HDL-C. The composition of gut microbiota was changed. Interestingly, the relative abundance of Muribaculaceae and Ruminococcaceae increased after DXR IV administration, whereas the abundance of Erysipelotrichaceae decreased, which have been beneficial to lipid metabolism. Nine potential metabolic biomarkers, including acetate, butyrate, propionate, alanine, succinate, valerate, xylose, choline, glutamate, were identified, which were related to fatty acid metabolism. Further, the pathway of fatty acid was detected by the RT-qPCR and western blotting. Compared with model group, the level of LXR-α and SREBP1 decreased significantly in DXR IV group while LXR-β, SREBP2 showed no statistical significance. It indicated that DXR IV modulated lipid metabolism by LXR-α/SREBP1 but not LXRβ and SREBP2. CONCLUSIONS DXR IV exhibits potential anti-atherosclerosis effect, which is closely related to lipid metabolism and the gut microbiota. This study may provide novel insights into the mechanism of DXR IV on atherosclerosis and a basis for promising clinical usage.
Collapse
Affiliation(s)
- Yaxin Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Yuyan Gu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Yihao Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Zhiyong Huang
- The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Mei Li
- The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China.
| | - Weihao Jiang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Jiahui Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Wenting Rao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Shangfei Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yuyao Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Junqi Chen
- The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Lijun Li
- The Oncology Department, The 982 Hospital of PLA, Tangshan, Hebei Province, China.
| | - Yuhua Jia
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Menghua Liu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Fenghua Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
23
|
Jiang XC, Yu Y. The Role of Phospholipid Transfer Protein in the Development of Atherosclerosis. Curr Atheroscler Rep 2021; 23:9. [PMID: 33496859 DOI: 10.1007/s11883-021-00907-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Phospholipid transfer protein (PLTP), a member of lipid transfer protein family, is an important protein involved in lipid metabolism in the circulation. This article reviews recent PLTP research progresses, involving lipoprotein metabolism and atherogenesis. RECENT FINDINGS PLTP activity influences atherogenic and anti-atherogenic lipoprotein levels. Human serum PLTP activity is a risk factor for human cardiovascular disease and is an independent predictor of all-cause mortality. PLTP deficiency reduces VLDL and LDL levels and attenuates atherosclerosis in mouse models, while PLTP overexpression exerts an opposite effect. Both PLTP deficiency and overexpression result in reduction of HDL which has different size, inflammatory index, and lipid composition. Moreover, although both PLTP deficiency and overexpression reduce cholesterol efflux capacity, but this effect has no impact in macrophage reverse cholesterol transport in mice. Furthermore, PLTP activity is related with metabolic syndrome, thrombosis, and inflammation. PLTP could be target for the treatment of dyslipidemia and atherosclerosis, although some potential off-target effects should be noted.
Collapse
Affiliation(s)
- Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY, USA.
| | - Yang Yu
- Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, People's Republic of China
| |
Collapse
|
24
|
Kim HY, Hong MH, Kim KW, Yoon JJ, Lee JE, Kang DG, Lee HS. Improvement of Hypertriglyceridemia by Roasted Nelumbinis folium in High Fat/High Cholesterol Diet Rat Model. Nutrients 2020; 12:nu12123859. [PMID: 33348773 PMCID: PMC7766402 DOI: 10.3390/nu12123859] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 11/16/2022] Open
Abstract
Hypertriglyceridemia is a condition characterized by high triglyceride levels and is a major risk factor for the development of cardiovascular diseases. The present study was designed to investigate the inhibitory effect of roasted Nelumbinis folium (RN), which is a medicinal substance produced by heating lotus leaves, on lipid metabolism in high fat/cholesterol (HFC) diet-induced hypertriglyceridemia. Except for those in the control group, Sprague–Dawley rats were fed an HFC diet for four weeks to induce hypertriglyceridemia. During the next nine weeks, the control, regular diet; HFC, HFC diet, FLU, fluvastatin (3 mg/kg/day); RNL, RN (100 mg/kg/day); RNH, RN (200 mg/kg/day) were orally administered together with the diet, and the experiments were conducted for a total of 13 weeks. The weight of the epididymal adipose tissue, liver, and heart of rats in the HFC diet group significantly increased compared to those in the control group but improved in the RN-treated group. It was also confirmed that vascular function, which is damaged by an HFC diet, was improved after RN treatment. The levels of insulin, glucose, triglycerides, total cholesterol, and low-density lipoprotein increased in the HFC diet group compared to those in the control group, while the administration of RN attenuated these parameters. In addition, the administration of RN significantly reduced the gene expression of both LXR and SREBP-1, which indicated the inhibitory effect of the biosynthesis of triglycerides caused by RN. The results indicated that RN administration resulted in an improvement in the overall lipid metabolism and a decrease in the concentration of triglycerides in the HFC diet-induced rat model of hypertriglyceridemia. Therefore, our findings suggest that the RN can be a candidate material to provide a new direction for treating hypertriglyceridemia.
Collapse
Affiliation(s)
- Hye Yoom Kim
- Hanbang Cardio-Renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, Korea; (H.Y.K.); (M.H.H.); (K.W.K.); (J.J.Y.)
- Department of Physiology, College of Oriental Medicine, Wonkwang University, Iksan 54538, Korea
| | - Mi Hyeon Hong
- Hanbang Cardio-Renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, Korea; (H.Y.K.); (M.H.H.); (K.W.K.); (J.J.Y.)
- Department of Physiology, College of Oriental Medicine, Wonkwang University, Iksan 54538, Korea
| | - Kwan Woo Kim
- Hanbang Cardio-Renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, Korea; (H.Y.K.); (M.H.H.); (K.W.K.); (J.J.Y.)
| | - Jung Joo Yoon
- Hanbang Cardio-Renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, Korea; (H.Y.K.); (M.H.H.); (K.W.K.); (J.J.Y.)
- Department of Physiology, College of Oriental Medicine, Wonkwang University, Iksan 54538, Korea
| | - Jung Eun Lee
- Clinical Medicine Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea;
| | - Dae Gill Kang
- Hanbang Cardio-Renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, Korea; (H.Y.K.); (M.H.H.); (K.W.K.); (J.J.Y.)
- Department of Physiology, College of Oriental Medicine, Wonkwang University, Iksan 54538, Korea
- Correspondence: (D.G.K.); (H.S.L.); Tel.: +82-63-6447 (D.G.K. & H.S.L.); Fax: +82-63-850-7260 (D.G.K. & H.S.L.)
| | - Ho Sub Lee
- Hanbang Cardio-Renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, Korea; (H.Y.K.); (M.H.H.); (K.W.K.); (J.J.Y.)
- Department of Physiology, College of Oriental Medicine, Wonkwang University, Iksan 54538, Korea
- Correspondence: (D.G.K.); (H.S.L.); Tel.: +82-63-6447 (D.G.K. & H.S.L.); Fax: +82-63-850-7260 (D.G.K. & H.S.L.)
| |
Collapse
|
25
|
Ruhanen H, Haridas PAN, Jauhiainen M, Olkkonen VM. Angiopoietin-like protein 3, an emerging cardiometabolic therapy target with systemic and cell-autonomous functions. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158791. [PMID: 32777482 DOI: 10.1016/j.bbalip.2020.158791] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/23/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022]
Abstract
Angiopoietin like protein 3 (ANGPTL3) is best known for its function as an inhibitor of lipoprotein and endothelial lipases. Due to the capacity of genetic or pharmacologic ANGPTL3 suppression to markedly reduce circulating lipoproteins, and the documented cardioprotection upon such suppression, ANGPTL3 has become an emerging therapy target for which both antibody and antisense oligonucleotide (ASO) therapeutics are being clinically tested. While the antibody is relatively selective for circulating ANGPTL3, the ASO also depletes the intra-hepatocellular protein, and there is emerging evidence for cell-autonomous functions of ANGPTL3 in the liver. These include regulation of hepatocyte glucose and fatty acid uptake, insulin sensitivity, LDL/VLDL remnant uptake, VLDL assembly/secretion, polyunsaturated fatty acid (PUFA) and PUFA-derived lipid mediator content, and gene expression. In this review we elaborate on (i) why ANGPTL3 is considered one of the most promising new cardiometabolic therapy targets, and (ii) the present evidences for its intra-hepatocellular or cell-autonomous functions.
Collapse
Affiliation(s)
- Hanna Ruhanen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland; Molecular and Integrative Biosciences, University of Helsinki, Finland
| | | | - Matti Jauhiainen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland; Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland.
| |
Collapse
|
26
|
Kaufman A, Abuqayyas L, Denney WS, Tillman EJ, Rolph T. AKR-001, an Fc-FGF21 Analog, Showed Sustained Pharmacodynamic Effects on Insulin Sensitivity and Lipid Metabolism in Type 2 Diabetes Patients. Cell Rep Med 2020; 1:100057. [PMID: 33205064 PMCID: PMC7659583 DOI: 10.1016/j.xcrm.2020.100057] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 05/04/2020] [Accepted: 06/23/2020] [Indexed: 12/18/2022]
Abstract
Experimental fibroblast growth factor 21 (FGF21) analogs can improve lipid profiles in patients with metabolic diseases. However, their effects on markers of insulin sensitivity appear to be minimal, potentially because of insufficient exposure. Systemic drug levels vary from sub-pharmacological to demonstrating pharmacodynamic effects but with dose-limiting adverse events. Here we report results from a phase 1 multiple ascending dose study of AKR-001, an Fc-FGF21 fusion protein engineered for sustained systemic pharmacologic exposure, in individuals with type 2 diabetes. With a half-life of 3-3.5 days, the peak-to-trough ratio under steady-state conditions is approximately 2 following QW dosing. AKR-001 appears to demonstrate pharmacodynamic effects on serum markers of insulin sensitivity and acceptable tolerability up to and including 70 mg QW. Positive trends in lipoprotein profile, including triglycerides, non-high-density lipoprotein (non-HDL) cholesterol, HDL-C, and apolipoproteins B and C3 are consistent with other FGF21 analogs. AKR-001's clinical profile supports further evaluation as a treatment for metabolic diseases.
Collapse
Affiliation(s)
| | | | | | | | - Tim Rolph
- Akero Therapeutics, South San Francisco, CA 94080, USA
| |
Collapse
|
27
|
Mayneris-Perxachs J, Puig J, Burcelin R, Dumas ME, Barton RH, Hoyles L, Federici M, Fernández-Real JM. The APOA1bp-SREBF-NOTCH axis is associated with reduced atherosclerosis risk in morbidly obese patients. Clin Nutr 2020; 39:3408-3418. [PMID: 32199697 DOI: 10.1016/j.clnu.2020.02.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/23/2020] [Accepted: 02/24/2020] [Indexed: 01/21/2023]
Abstract
BACKGROUND & AIMS Atherosclerosis is characterized by an inflammatory disease linked to excessive lipid accumulation in the artery wall. The Notch signalling pathway has been shown to play a key regulatory role in the regulation of inflammation. Recently, in vitro and pre-clinical studies have shown that apolipoprotein A-I binding protein (AIBP) regulates cholesterol metabolism (SREBP) and NOTCH signalling (haematopoiesis) and may be protective against atherosclerosis, but the evidence in humans is scarce. METHODS We evaluated the APOA1bp-SREBF-NOTCH axis in association with atherosclerosis in two well-characterized cohorts of morbidly obese patients (n = 78) within the FLORINASH study, including liver transcriptomics, 1H NMR plasma metabolomics, high-resolution ultrasonography evaluating carotid intima-media thickness (cIMT), and haematological parameters. RESULTS The liver expression levels of APOA1bp were associated with lower cIMT and leukocyte counts, a better plasma lipid profile and higher circulating levels of metabolites associated with lower risk of atherosclerosis (glycine, histidine and asparagine). Conversely, liver SREBF and NOTCH mRNAs were positively associated with atherosclerosis, liver steatosis, an unfavourable lipid profile, higher leukocytes and increased levels of metabolites linked to inflammation and CVD such as branched-chain amino acids and glycoproteins. APOA1bp and NOTCH signalling also had a strong association, as revealed by the negative correlations among APOA1bp expression levels and those of all NOTCH receptors and jagged ligands. CONCLUSIONS We here provide the first evidence in human liver of the putative APOA1bp-SREBF-NOTCH axis signalling pathway and its association with atherosclerosis and inflammation.
Collapse
Affiliation(s)
- Jordi Mayneris-Perxachs
- Department of Endocrinology, Diabetes and Nutrition, Hospital of Girona "Dr Josep Trueta", Departament de Ciències Mèdiques, University of Girona, Girona Biomedical Research Institute (IdibGi), Girona, Spain; CIBERobn Pathophysiology of Obesity and Nutrition, Instituto de Salud Carlos III, Madrid, Spain
| | - Josep Puig
- Department of Radiology, Diagnostic Imaging Institute (IDI), Dr Josep Trueta University Hospital, IDIBGI, Girona, Spain
| | - Rémy Burcelin
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France; Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Team 2: 'Intestinal Risk Factors, Diabetes, Dyslipidemia, and Heart Failure', F-31432 Toulouse Cedex 4, France
| | - Marc-Emmanuel Dumas
- Section of Biomolecular Medicine, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, Exhibition Road, London SW7 2AZ, United Kingdom; Section of Genomic and Environmental Medicine, Respiratory Division, National Heart and Lung Institute, Imperial College London, Dovehouse St, London SW3 6KY, United Kingdom
| | - Richard H Barton
- Section of Biomolecular Medicine, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, Exhibition Road, London SW7 2AZ, United Kingdom
| | - Lesley Hoyles
- Department of Biosciences, Nottingham Trent University, Clifton Campus, Nottingham NG11 8NS, United Kingdom
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - José-Manuel Fernández-Real
- Department of Endocrinology, Diabetes and Nutrition, Hospital of Girona "Dr Josep Trueta", Departament de Ciències Mèdiques, University of Girona, Girona Biomedical Research Institute (IdibGi), Girona, Spain; CIBERobn Pathophysiology of Obesity and Nutrition, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
28
|
Takanashi M, Kimura T, Li C, Tanaka M, Matsuhashi A, Yoshida H, Noda A, Xu P, Takase S, Okazaki S, Iizuka Y, Kumagai H, Ikeda Y, Gotoda T, Takahashi M, Yagyu H, Ishibashi S, Yamauchi T, Kadowaki T, Liang G, Okazaki H. Critical Role of SREBP-1c Large-VLDL Pathway in Environment-Induced Hypertriglyceridemia of Apo AV Deficiency. Arterioscler Thromb Vasc Biol 2020; 39:373-386. [PMID: 30700132 DOI: 10.1161/atvbaha.118.311931] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective- APOA5 variants are strongly associated with hypertriglyceridemia, as well as increased risks of cardiovascular disease and acute pancreatitis. Hypertriglyceridemia in apo AV dysfunction often aggravates by environmental factors such as high-carbohydrate diets or aging. To date, the molecular mechanisms by which these environmental factors induce hypertriglyceridemia are poorly defined, leaving the high-risk hypertriglyceridemia condition undertreated. Previously, we reported that LXR (liver X receptor)-SREBP (sterol regulatory element-binding protein)-1c pathway regulates large-VLDL (very low-density lipoprotein) production induced by LXR agonist. However, the pathophysiological relevance of the finding remains unknown. Approach and Results- Here, we reconstitute the environment-induced hypertriglyceridemia phenotype of human APOA5 deficiency in Apoa5-/- mice and delineate the role of SREBP-1c in vivo by generating Apoa5-/- ;Srebp-1c-/- mice. The Apoa5-/- mice, which showed moderate hypertriglyceridemia on a chow diet, developed severe hypertriglyceridemia on high-carbohydrate feeding or aging as seen in patients with human apo AV deficiency. These responses were nearly completely abolished in the Apoa5-/- ;Srebp-1c-/- mice. Further mechanistic studies revealed that in response to these environmental factors, SREBP-1c was activated to increase triglyceride synthesis and to permit the incorporation of triglyceride into abnormally large-VLDL particles, which require apo AV for efficient clearance. Conclusions- Severe hypertriglyceridemia develops only when genetic factors (apo AV deficiency) and environmental effects (SREBP-1c activation) coexist. We demonstrate that the regulated production of large-sized VLDL particles via SREBP-1c determines plasma triglyceride levels in apo AV deficiency. Our findings explain the long-standing enigma of the late-onset hypertriglyceridemia phenotype of apo AV deficiency and suggest a new approach to treat hypertriglyceridemia by targeting genes that mediate environmental effects.
Collapse
Affiliation(s)
- Mikio Takanashi
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Takeshi Kimura
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Chengcheng Li
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Masaki Tanaka
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Ako Matsuhashi
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Hiroki Yoshida
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Akari Noda
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Pengfei Xu
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Satoru Takase
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Sachiko Okazaki
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Yoko Iizuka
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Hidetoshi Kumagai
- Department of Cardiovascular Medicine (H.K., Y. Ikeda), Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Japan
| | - Yuichi Ikeda
- Department of Cardiovascular Medicine (H.K., Y. Ikeda), Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Japan
| | - Takanari Gotoda
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Manabu Takahashi
- Division of Endocrinology and Metabolism, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan (Manabu Takahashi, S.I.)
| | - Hiroaki Yagyu
- Department of Endocrinology and Metabolism, Mito Medical Center, Tsukuba University Hospital, Mito, Ibaraki, Japan (H. Yagyu)
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan (Manabu Takahashi, S.I.)
| | - Toshimasa Yamauchi
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Takashi Kadowaki
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
| | - Guosheng Liang
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX (G.L., H.O.)
| | - Hiroaki Okazaki
- From the Department of Diabetes and Metabolic Diseases (Mikio Takanashi, T. Kimura, C.L., M. Tanaka, A.M., H. Yoshida, A.N., P.X., S.T., S.O., Y. Iizuka, T.G., T.Y., T. Kadowaki, H.O.)
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX (G.L., H.O.)
| |
Collapse
|
29
|
Impact of Phospholipid Transfer Protein in Lipid Metabolism and Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:1-13. [PMID: 32705590 DOI: 10.1007/978-981-15-6082-8_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PLTP plays an important role in lipoprotein metabolism and cardiovascular disease development in humans; however, the mechanisms are still not completely understood. In mouse models, PLTP deficiency reduces cardiovascular disease, while its overexpression induces it. Therefore, we used mouse models to investigate the involved mechanisms. In this chapter, the recent main progresses in the field of PLTP research are summarized, and our focus is on the relationship between PLTP and lipoprotein metabolism, as well as PLTP and cardiovascular diseases.
Collapse
|
30
|
Liver X Receptors and Male (In)fertility. Int J Mol Sci 2019; 20:ijms20215379. [PMID: 31671745 PMCID: PMC6862486 DOI: 10.3390/ijms20215379] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 12/19/2022] Open
Abstract
Liver X receptors (LXRs) are ligand-dependent transcription factors acting as ‘cholesterol sensors’ to regulate lipid homeostasis in cells. The two isoforms, LXRα (NR1H3) and LXRβ (NR1H2), are differentially expressed, with the former expressed predominantly in metabolically active tissues and the latter more ubiquitously. Both are activated by oxidised cholesterol metabolites, endogenously produced oxysterols. LXRs have important roles in lipid metabolism and inflammation, plus a number of newly emerging roles. They are implicated in regulating lipid balance in normal male reproductive function and may provide a link between male infertility and lipid disorders and/or obesity. Studies from Lxr knockout mouse models provide compelling evidence to support this. More recently published data suggest distinct and overlapping roles of the LXR isoforms in the testis and recent evidence of a role for LXRs in human male fertility. This review summarises the current literature and explores the likely link between LXR, lipid metabolism and male fertility as part of a special issue on Liver X receptors in International Journal of Molecular Sciences.
Collapse
|
31
|
Santana-Codina N, Marcé-Grau A, Muixí L, Nieva C, Marro M, Sebastián D, Muñoz JP, Zorzano A, Sierra A. GRP94 Is Involved in the Lipid Phenotype of Brain Metastatic Cells. Int J Mol Sci 2019; 20:ijms20163883. [PMID: 31395819 PMCID: PMC6720951 DOI: 10.3390/ijms20163883] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/06/2019] [Accepted: 08/06/2019] [Indexed: 12/18/2022] Open
Abstract
Metabolic adaptation may happen in response to the pressure exerted by the microenvironment and is a key step in survival of metastatic cells. Brain metastasis occurs as a consequence of the systemic dissemination of tumor cells, a fact that correlates with poor prognosis and high morbidity due to the difficulty in identifying biomarkers that allow a more targeted therapy. Previously, we performed transcriptomic analysis of human breast cancer patient samples and evaluated the differential expression of genes in brain metastasis (BrM) compared to lung, bone and liver metastasis. Our network approach identified upregulation of glucose-regulated protein 94 (GRP94) as well as proteins related to synthesis of fatty acids (FA) in BrM. Here we report that BrM cells show an increase in FA content and decreased saturation with regard to parental cells measured by Raman spectroscopy that differentiate BrM from other metastases. Moreover, BrM cells exerted a high ability to oxidize FA and compensate hypoglycemic stress due to an overexpression of proteins involved in FA synthesis and degradation (SREBP-1, LXRα, ACOT7). GRP94 ablation restored glucose dependence, down-regulated ACOT7 and SREBP-1 and decreased tumorigenicity in vivo. In conclusion, GRP94 is required for the metabolic stress survival of BrM cells, and it might act as a modulator of lipid metabolism to favor BrM progression.
Collapse
Affiliation(s)
- Naiara Santana-Codina
- Biological Clues of the Invasive and Metastatic Phenotype Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, E-08908 Barcelona, Spain.
- Universitat Autònoma de Barcelona (UAB), Campus Bellaterra, Cerdanyola del Vallés, E-08193 Barcelona, Spain.
| | - Anna Marcé-Grau
- Biological Clues of the Invasive and Metastatic Phenotype Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, E-08908 Barcelona, Spain
| | - Laia Muixí
- Biological Clues of the Invasive and Metastatic Phenotype Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, E-08908 Barcelona, Spain
| | - Claudia Nieva
- Biological Clues of the Invasive and Metastatic Phenotype Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, E-08908 Barcelona, Spain
- ICFO-Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Carl Friedrich Gauss 3, 08036 Barcelona, Spain
| | - Mónica Marro
- ICFO-Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Carl Friedrich Gauss 3, 08036 Barcelona, Spain
| | - David Sebastián
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08036 Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 08028 Barcelona, Spain
| | - Juan Pablo Muñoz
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08036 Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 08028 Barcelona, Spain
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08036 Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 08028 Barcelona, Spain
| | - Angels Sierra
- Laboratory of Molecular and Translational Oncology, Centre de Recerca Biomèdica CELLEX-CRBC-Institut d'Investigacions Biomèdiques August Pi i Sunyer-IDIBAPS, E-08036 Barcelona, Spain.
| |
Collapse
|
32
|
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: 17] [Impact Index Per Article: 3.4] [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
|
33
|
Hantani R, Takahashi Y, Sotani T, Hantani Y. Identification of Novel Phospholipid Transfer Protein Inhibitors by High-Throughput Screening. SLAS DISCOVERY 2019; 24:579-586. [PMID: 31017809 DOI: 10.1177/2472555219842210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Atherogenesis has been recognized as a risk factor for lethal cardiovascular diseases. Plasma low-density lipoprotein levels are correlated to the occurrence of atherosclerosis, and their control is critical for both the prevention and treatment of these diseases. Phospholipid transfer protein (PLTP) is one of the key regulators of lipoprotein metabolism; PLTP-deficient mice exhibit decreased apolipoprotein B (apoB)-containing lipoprotein secretion and atherosclerosis, indicating the validity of PLTP as a promising therapeutic target. Here, we demonstrate a high-throughput screening (HTS) method to identify a novel chemotype of PLTP inhibitors. Instead of using recombinant proteins, we used human plasma as a source of enzymes in the first screening, so as to efficiently exclude promiscuous inhibitors. The selected compounds were further confirmed to target PLTP both biochemically and biophysically and were shown to inhibit apoB secretion from hepatic cells with no apparent toxicity. We believe that our approach is suitable for filtering out nonspecific inhibitors at an earlier stage of screening campaigns and that these compounds should have potential to be developed into drugs to treat dyslipidemia.
Collapse
Affiliation(s)
- Rie Hantani
- 1 Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki, Osaka, Japan
| | - Yu Takahashi
- 1 Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki, Osaka, Japan
| | - Tomohiro Sotani
- 1 Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki, Osaka, Japan
| | - Yoshiji Hantani
- 1 Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki, Osaka, Japan
| |
Collapse
|
34
|
Xu HF, Luo J, Zhang XY, Li J, Bionaz M. Activation of liver X receptor promotes fatty acid synthesis in goat mammary epithelial cells via modulation of SREBP1 expression. J Dairy Sci 2019; 102:3544-3555. [PMID: 30738675 DOI: 10.3168/jds.2018-15538] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 12/12/2018] [Indexed: 01/04/2023]
Abstract
In bovine mammary tissue and cells, liver X receptor (LXR) regulates lipid synthesis mainly via transactivation of the transcription factor sterol regulatory element binding protein 1 (SREBP1). In the present work, we investigated the role of LXR in controlling lipid synthesis via transactivation of SREBP1 in goat primary mammary cells (GMEC). The GMEC were treated with a synthetic agonist of LXR, T0901317, and transactivation and transcription of SREBP1, expression of lipogenic genes, and fatty acid profiling and triacylglycerol (TAG) content of the cells were measured. A mild increase in the mRNA expression level of LXRα (NR1H3) was observed following treatment with different concentrations of T0901317, and a dose-dependent increase in mRNA and transactivation of SREBP1 was detected. Activation of LXR resulted in a significant increase in the mRNA expression of most of the measured genes related to de novo synthesis, desaturation, and transport of fatty acids; TAG synthesis; and transcription regulators. Compared with the control, total content of cellular TAG increased by more than 20% with T0901317 treatment. Furthermore, addition of T0901317 increased the proportion of unsaturated fatty acids (e.g., C16:1, C18:1, C20:1, and C22:1), and decreased the proportion of saturated fatty acids (e.g., C16:0, C18:0, C20:0, and C22:0). These results provide evidence that LXR regulates the expression and activity of SREBP1. Our results indicated that LXR participate in regulating the transcription of genes involved in milk fat synthesis in GMEC in an SREBP1-dependent fashion.
Collapse
Affiliation(s)
- H F Xu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, P. R. China
| | - J Luo
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China.
| | - X Y Zhang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - J Li
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, 450046, P. R. China
| | - M Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis 97331.
| |
Collapse
|
35
|
Kim KH, Choi JM, Li F, Dong B, Wooton-Kee CR, Arizpe A, Anakk S, Jung SY, Hartig SM, Moore DD. Constitutive Androstane Receptor Differentially Regulates Bile Acid Homeostasis in Mouse Models of Intrahepatic Cholestasis. Hepatol Commun 2018; 3:147-159. [PMID: 30620001 PMCID: PMC6312660 DOI: 10.1002/hep4.1274] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/03/2018] [Indexed: 12/14/2022] Open
Abstract
Bile acid (BA) homeostasis is tightly regulated by multiple transcription factors, including farnesoid X receptor (FXR) and small heterodimer partner (SHP). We previously reported that loss of the FXR/SHP axis causes severe intrahepatic cholestasis, similar to human progressive familial intrahepatic cholestasis type 5 (PFIC5). In this study, we found that constitutive androstane receptor (CAR) is endogenously activated in Fxr:Shp double knockout (DKO) mice. To test the hypothesis that CAR activation protects DKO mice from further liver damage, we generated Fxr;Shp;Car triple knockout (TKO) mice. In TKO mice, residual adenosine triphosphate (ATP) binding cassette, subfamily B member 11 (ABCB11; alias bile salt export pump [BSEP]) function and fecal BA excretion are completely impaired, resulting in severe hepatic and biliary damage due to excess BA overload. In addition, we discovered that pharmacologic CAR activation has different effects on intrahepatic cholestasis of different etiologies. In DKO mice, CAR agonist 1,4‐bis[2‐(3,5‐dichloropyridyloxy)]benzene (TCPOBOP; here on TC) treatment attenuated cholestatic liver injury, as expected. However, in the PFIC2 model Bsep knockout (BKO) mice, TC treatment exhibited opposite effects that reflect increased BA accumulation and liver injury. These contrasting results may be linked to differential regulation of systemic cholesterol homeostasis in DKO and BKO livers. TC treatment selectively up‐regulated hepatic cholesterol levels in BKO mice, supporting de novo BA synthesis. Conclusion: CAR activation in DKO mice is generally protective against cholestatic liver injury in these mice, which model PFIC5, but not in the PFIC2 model BKO mice. Our results emphasize the importance of the genetic and physiologic background when implementing targeted therapies to treat intrahepatic cholestasis.
Collapse
Affiliation(s)
- Kang Ho Kim
- Department of Molecular and Cellular Biology Baylor College of Medicine Houston TX
| | - Jong Min Choi
- Department of Molecular and Cellular Biology Baylor College of Medicine Houston TX
| | - Feng Li
- Department of Molecular and Cellular Biology Baylor College of Medicine Houston TX.,Center for Drug Discovery Baylor College of Medicine Houston TX
| | - Bingning Dong
- Department of Molecular and Cellular Biology Baylor College of Medicine Houston TX
| | | | - Armando Arizpe
- School of Natural Science University of Texas Austin Austin TX
| | - Sayeepriyadarshini Anakk
- Department of Molecular and Integrative Physiology University of Illinois at Urbana-Champaign Urbana IL
| | - Sung Yun Jung
- Department of Molecular and Cellular Biology Baylor College of Medicine Houston TX.,Verna and Marrs McLean Department of Biochemistry and Molecular Biology Baylor College of Medicine Houston TX
| | - Sean M Hartig
- Department of Molecular and Cellular Biology Baylor College of Medicine Houston TX.,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine Baylor College of Medicine Houston TX
| | - David D Moore
- Department of Molecular and Cellular Biology Baylor College of Medicine Houston TX
| |
Collapse
|
36
|
Abstract
Phospholipids are major constituents of biological membranes. The fatty acyl chain composition of phospholipids determines the biophysical properties of membranes and thereby affects their impact on biological processes. The composition of fatty acyl chains is also actively regulated through a deacylation and reacylation pathway called Lands' cycle. Recent studies of mouse genetic models have demonstrated that lysophosphatidylcholine acyltransferases (LPCATs), which catalyze the incorporation of fatty acyl chains into the sn-2 site of phosphatidylcholine, play important roles in pathophysiology. Two LPCAT family members, LPCAT1 and LPCAT3, have been particularly well studied. LPCAT1 is crucial for proper lung function due to its role in pulmonary surfactant biosynthesis. LPCAT3 maintains systemic lipid homeostasis by regulating lipid absorption in intestine, lipoprotein secretion, and de novo lipogenesis in liver. Mounting evidence also suggests that changes in LPCAT activity may be potentially involved in pathological conditions, including nonalcoholic fatty liver disease, atherosclerosis, viral infections, and cancer. Pharmacological manipulation of LPCAT activity and membrane phospholipid composition may provide new therapeutic options for these conditions.
Collapse
Affiliation(s)
- Bo Wang
- Department of Pathology and Laboratory Medicine, Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, California 90272, USA;
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, California 90272, USA;
| |
Collapse
|
37
|
Phospholipid transfer protein and alpha-1 antitrypsin regulate Hck kinase activity during neutrophil degranulation. Sci Rep 2018; 8:15394. [PMID: 30337619 PMCID: PMC6193999 DOI: 10.1038/s41598-018-33851-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/07/2018] [Indexed: 01/21/2023] Open
Abstract
Excessive neutrophil degranulation is a common feature of many inflammatory disorders, including alpha-1 antitrypsin (AAT) deficiency. Our group has demonstrated that phospholipid transfer protein (PLTP) prevents neutrophil degranulation but serine proteases, which AAT inhibits, cleave PLTP in diseased airways. We propose to identify if airway PLTP activity can be restored by AAT augmentation therapy and how PLTP subdues degranulation of neutrophils in AAT deficient subjects. Airway PLTP activity was lower in AAT deficient patients but elevated in the airways of patients on augmentation therapy. Functional AAT protein (from PiMM homozygotes) prevented PLTP cleavage unlike its mutated ZZ variant (PiZZ). PLTP lowered leukotriene B4 induced degranulation of primary, secondary and tertiary granules from neutrophils from both groups (n = 14/group). Neutrophils isolated from Pltp knockout mice have enhance neutrophil degranulation. Both AAT and PLTP reduced neutrophil degranulation and superoxide production, possibly though their inhibition of the Src tyrosine kinase, Hck. Src kinase inhibitors saracatinib and dasatinib reduced neutrophil degranulation and superoxide production. Therefore, AAT protects PLTP from proteolytic cleavage and both AAT and PLTP mediate degranulation, possibly via Hck tyrosine kinase inhibition. Deficiency of AAT could contribute to reduced lung PLTP activity and elevated neutrophil signaling associated with lung disease.
Collapse
|
38
|
Abstract
Liver X receptors α and β (LXRα and LXRβ) are nuclear receptors with pivotal roles in the transcriptional control of lipid metabolism. Transcriptional activity of LXRs is induced in response to elevated cellular levels of cholesterol. LXRs bind to and regulate the expression of genes that encode proteins involved in cholesterol absorption, transport, efflux, excretion and conversion to bile acids. The coordinated, tissue-specific actions of the LXR pathway maintain systemic cholesterol homeostasis and regulate immune and inflammatory responses. LXRs also regulate fatty acid metabolism by controlling the lipogenic transcription factor sterol regulatory element-binding protein 1c and regulate genes that encode proteins involved in fatty acid elongation and desaturation. LXRs exert important effects on the metabolism of phospholipids, which, along with cholesterol, are major constituents of cellular membranes. LXR activation preferentially drives the incorporation of polyunsaturated fatty acids into phospholipids by inducing transcription of the remodelling enzyme lysophosphatidylcholine acyltransferase 3. The ability of the LXR pathway to couple cellular sterol levels with the saturation of fatty acids in membrane phospholipids has implications for several physiological processes, including lipoprotein production, dietary lipid absorption and intestinal stem cell proliferation. Understanding how LXRs regulate membrane composition and function might provide new therapeutic insight into diseases associated with dysregulated lipid metabolism, including atherosclerosis, diabetes mellitus and cancer.
Collapse
Affiliation(s)
- Bo Wang
- Department of Pathology and Laboratory Medicine, Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
39
|
Yu Y, Lei X, Jiang H, Li Z, Creemers JWM, Zhang M, Qin S, Jin W, Jiang X. Prodomain of Furin Promotes Phospholipid Transfer Protein Proteasomal Degradation in Hepatocytes. J Am Heart Assoc 2018; 7:e008526. [PMID: 29680823 PMCID: PMC6015287 DOI: 10.1161/jaha.118.008526] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/22/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Phospholipid transfer protein (PLTP) is one of the major modulators of lipoprotein metabolism and atherosclerosis development; however, little is known about the regulation of PLTP. The effect of hepatic prodomain of furin (profurin) expression on PLTP processing and function is investigated. METHODS AND RESULTS We used adenovirus expressing profurin in mouse liver to evaluate PLTP activity, mass, and plasma lipid levels. We coexpressed PLTP and profurin in human hepatoma cell line cells and studied their interaction. We found profurin expression significantly reduced plasma lipids, plasma PLTP activity, and mass in all tested mouse models, compared with controls. Moreover, the expression of profurin dramatically reduced liver PLTP activity and protein level. We further explored the mechanism using in vivo and ex vivo approaches. We found that profurin can interact with intracellular PLTP and promote its ubiquitination and proteasomal degradation, resulting in less PLTP secretion from the hepatocytes. Furin does not cleave PLTP; instead, it forms a complex with PLTP, likely through its prodomain. CONCLUSIONS Our study reveals that hepatic PLTP protein is targeted for proteasomal degradation by profurin expression, which could be a novel posttranslational mechanism underlying PLTP regulation.
Collapse
Affiliation(s)
- Yang Yu
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of AtherosclerosisTaishan Medical UniversityTaianChina
| | - Xia Lei
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
| | - Hui Jiang
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
| | - Zhiqiang Li
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
| | - John W. M. Creemers
- Laboratory of Biochemical NeuroendocrinologyDepartment of Human GeneticsHerestraat 49 bus 6023000 LeuvenBelgium
| | - Ming Zhang
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
| | - Shucun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of AtherosclerosisTaishan Medical UniversityTaianChina
| | - Weijun Jin
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
| | - Xian‐Cheng Jiang
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
- Molecular and Cellular Cardiology ProgramVeterans Affair New York Harbor Healthcare SystemBrooklynNY
| |
Collapse
|
40
|
Sirwi A, Hussain MM. Lipid transfer proteins in the assembly of apoB-containing lipoproteins. J Lipid Res 2018; 59:1094-1102. [PMID: 29650752 DOI: 10.1194/jlr.r083451] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 04/12/2018] [Indexed: 11/20/2022] Open
Abstract
A better understanding of intracellular lipoprotein assembly may help identify proteins with important roles in lipid disorders. apoB-containing lipoproteins (B-lps) are macromolecular lipid and protein micelles that act as specialized transport vehicles for hydrophobic lipids. They are assembled predominantly in enterocytes and hepatocytes to transport dietary and endogenous fat, respectively, to different tissues. Assembly occurs in the endoplasmic reticulum (ER) and is dependent on lipid resynthesis in the ER and on a chaperone, namely, microsomal triglyceride transfer protein (MTTP). Precursors for lipid synthesis are obtained from extracellular sources and from cytoplasmic lipid droplets. MTTP is the major and essential lipid transfer protein that transfers phospholipids and triacylglycerols to nascent apoB for the assembly of lipoproteins. Assembly is aided by cell death-inducing DFF45-like effector B and by phospholipid transfer protein, which may facilitate additional deposition of triacylglycerols and phospholipids, respectively, to apoB. Here, we summarize the current understanding of the different steps in the assembly of B-lps and discuss the role of lipid transfer proteins in these steps to help identify new clinical targets for lipid-associated disorders, such as heart disease.
Collapse
Affiliation(s)
- Alaa Sirwi
- School of Graduate Studies, Molecular and Cell Biology Program, State University of New York Downstate Medical Center, Brooklyn, NY
| | - M Mahmood Hussain
- New York University Winthrop Hospital, Mineola, NY and Veterans Affairs New York Harbor Healthcare System, Brooklyn, NY
| |
Collapse
|
41
|
Jiang XC. Phospholipid transfer protein: its impact on lipoprotein homeostasis and atherosclerosis. J Lipid Res 2018; 59:764-771. [PMID: 29438986 DOI: 10.1194/jlr.r082503] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/10/2018] [Indexed: 12/25/2022] Open
Abstract
Phospholipid transfer protein (PLTP) is one of the major modulators of lipoprotein metabolism and atherosclerosis development in humans; however, we still do not quite understand the mechanisms. In mouse models, PLTP overexpression induces atherosclerosis, while its deficiency reduces it. Thus, mouse models were used to explore the mechanisms. In this review, I summarize the major progress made in the PLTP research field and emphasize its impact on lipoprotein metabolism and atherosclerosis, as well as its regulation.
Collapse
Affiliation(s)
- Xian-Cheng Jiang
- Department of Cell Biology, Downstate Medical Center, State University of New York, Brooklyn, NY
| |
Collapse
|
42
|
Abstract
Cellular lipid metabolism and homeostasis are controlled by sterol regulatory-element binding proteins (SREBPs). In addition to performing canonical functions in the transcriptional regulation of genes involved in the biosynthesis and uptake of lipids, genome-wide system analyses have revealed that these versatile transcription factors act as important nodes of convergence and divergence within biological signalling networks. Thus, they are involved in myriad physiological and pathophysiological processes, highlighting the importance of lipid metabolism in biology. Changes in cell metabolism and growth are reciprocally linked through SREBPs. Anabolic and growth signalling pathways branch off and connect to multiple steps of SREBP activation and form complex regulatory networks. In addition, SREBPs are implicated in numerous pathogenic processes such as endoplasmic reticulum stress, inflammation, autophagy and apoptosis, and in this way, they contribute to obesity, dyslipidaemia, diabetes mellitus, nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, chronic kidney disease, neurodegenerative diseases and cancers. This Review aims to provide a comprehensive understanding of the role of SREBPs in physiology and pathophysiology at the cell, organ and organism levels.
Collapse
Affiliation(s)
- Hitoshi Shimano
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty 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, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Ryuichiro Sato
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo 100-0004, Japan
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo 113-8657, Japan
| |
Collapse
|
43
|
Rong X, Wang B, Palladino EN, de Aguiar Vallim TQ, Ford DA, Tontonoz P. ER phospholipid composition modulates lipogenesis during feeding and in obesity. J Clin Invest 2017; 127:3640-3651. [PMID: 28846071 DOI: 10.1172/jci93616] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 07/11/2017] [Indexed: 12/31/2022] Open
Abstract
Sterol regulatory element-binding protein 1c (SREBP-1c) is a central regulator of lipogenesis whose activity is controlled by proteolytic cleavage. The metabolic factors that affect its processing are incompletely understood. Here, we show that dynamic changes in the acyl chain composition of ER phospholipids affect SREBP-1c maturation in physiology and disease. The abundance of polyunsaturated phosphatidylcholine in liver ER is selectively increased in response to feeding and in the setting of obesity-linked insulin resistance. Exogenous delivery of polyunsaturated phosphatidylcholine to ER accelerated SREBP-1c processing through a mechanism that required an intact SREBP cleavage-activating protein (SCAP) pathway. Furthermore, induction of the phospholipid-remodeling enzyme LPCAT3 in response to liver X receptor (LXR) activation promoted SREBP-1c processing by driving the incorporation of polyunsaturated fatty acids into ER. Conversely, LPCAT3 deficiency increased membrane saturation, reduced nuclear SREBP-1c abundance, and blunted the lipogenic response to feeding, LXR agonist treatment, or obesity-linked insulin resistance. Desaturation of the ER membrane may serve as an auxiliary signal of the fed state that promotes lipid synthesis in response to nutrient availability.
Collapse
Affiliation(s)
- Xin Rong
- Department of Pathology and Laboratory Medicine, Department of Medicine, UCLA, Los Angeles, California, USA
| | - Bo Wang
- Department of Pathology and Laboratory Medicine, Department of Medicine, UCLA, Los Angeles, California, USA
| | - Elisa Nd Palladino
- Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University, St. Louis, Missouri, USA
| | | | - David A Ford
- Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University, St. Louis, Missouri, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, Department of Medicine, UCLA, Los Angeles, California, USA.,Molecular Biology Institute, and.,Howard Hughes Medical Institute, UCLA, Los Angeles, California, USA
| |
Collapse
|
44
|
Liu M, Chung S, Shelness GS, Parks JS. Hepatic ABCA1 deficiency is associated with delayed apolipoprotein B secretory trafficking and augmented VLDL triglyceride secretion. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1035-1043. [PMID: 28694219 DOI: 10.1016/j.bbalip.2017.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/30/2017] [Accepted: 07/05/2017] [Indexed: 11/30/2022]
Abstract
ATP binding cassette transporter A1 (ABCA1) is a membrane transporter that facilitates nascent HDL formation. Tangier disease subjects with complete ABCA1 deficiency have <5% of normal levels of plasma HDL, elevated triglycerides (TGs), and defective vesicular trafficking in fibroblasts and macrophages. Hepatocyte-specific ABCA1 knockout mice (HSKO) have a similar lipid phenotype with 20% of normal plasma HDL levels and a two-fold elevation of plasma TGs due to hepatic overproduction of large, triglyceride-enriched VLDL. We hypothesized that enhanced VLDL TG secretion in the absence of hepatocyte ABCA1 is due to altered intracellular trafficking of apolipoprotein B (apoB), resulting in augmented TG addition to nascent VLDL. We found that trafficking of newly synthesized apoB through the secretory pathway was delayed in ABCA1-silenced rat hepatoma cells and HSKO primary hepatocytes, relative to controls. Endoglycosidase H treatment of cellular apoB revealed a likely delay in apoB trafficking in post-ER compartments. The reduced rate of protein trafficking was also observed for an adenoviral-expressed GPI-linked fluorescent fusion protein, but not albumin, suggesting a selective delay of secretory cargoes in the absence of hepatocyte ABCA1. Our results suggest an important role for hepatic ABCA1 in regulating secretory trafficking and modulating VLDL expansion during the TG accretion phase of hepatic lipoprotein particle assembly.
Collapse
Affiliation(s)
- Mingxia Liu
- Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| | - Soonkyu Chung
- Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Gregory S Shelness
- Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - John S Parks
- Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA; Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
45
|
Schulman IG. Liver X receptors link lipid metabolism and inflammation. FEBS Lett 2017; 591:2978-2991. [PMID: 28555747 DOI: 10.1002/1873-3468.12702] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/23/2017] [Indexed: 12/14/2022]
Abstract
The response of immune cells to pathogens is often associated with changes in the flux through basic metabolic pathways. Indeed, in many cases changes in metabolism appear to be necessary for a robust immune response. The Liver X receptors (LXRs) are members of the nuclear hormone receptor superfamily that regulate gene networks controlling cholesterol and lipid metabolism. In immune cells, particularly in macrophages, LXRs also inhibit proinflammatory gene expression. This Review will highlight recent studies that connect LXR-dependent control of lipid metabolism to regulation of the immune response.
Collapse
Affiliation(s)
- Ira G Schulman
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
46
|
Jianhua L, Xueqin M, Jifen H. Expression and clinical significance of LXRα and SREBP-1c in placentas of preeclampsia. Open Med (Wars) 2016; 11:292-296. [PMID: 28352810 PMCID: PMC5329844 DOI: 10.1515/med-2016-0057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 06/29/2016] [Indexed: 12/18/2022] Open
Abstract
Objective To evaluate the expression and correlations of liver X receptor alpha (LXRa) and its target gene sterol regulatory element-binding protein-1c (SREBP-1c) in placentas of preeclampsia (PE) and their significance in PE. Methods Pregnancies were divided into two groups, 60 cases (29 cases of mild and 31 cases of severe) of PE group and 56 cases of normal group. The level of mRNA and protein of LXRa and SREBP-1c were analyzed by reverse transcription-polymerase chain reaction (RTPCR) and immunohistochemistry (IHC) in the placentas. Results RT-PCR and IHC results showed that the mRNA and protein expression of both LXRa and SREBP-1c increased gradually with the extent of PE among normal pregnancy, mild PE and severe PE groups, and the differences were of statistically significance (P<0.01 or P<0.05). There were positive correlations between the expression of LXRa mRNA and SREBP-1c mRNA, also between LXRa mRNA and LXRa protein (r=0.521, P<0.01; r=0.422, P<0.01). The expression of SREBP-1c mRNA positively correlated with its protein level (r=0.598, P<0.01). There were positive correlations between the expression of LXRa protein and SREBP-1c protein (r=0.612, P<0.01). Conclusion The expression of LXRa is elevated significantly in placentas of PE patients, and might contribute for promoting the transcription and translation of its target gene SREBP1-c, which is related to the occurrence and development of PE.
Collapse
Affiliation(s)
- Li Jianhua
- Department of Gynaecology and Obstetrics, The 1st Affiliated Hospital of Fujian Medical University, Fuzhou 350004, China
| | - Miao Xueqin
- Departmento of Gynaecology and Obstetrics, the 2nd Affiliated Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350003, China
| | - Hu Jifen
- Department of Gynaecology and Obstetrics, The 1st Affiliated Hospital of Fujian Medical University, Fuzhou 350004, China
| |
Collapse
|
47
|
Smagris E, Gilyard S, BasuRay S, Cohen JC, Hobbs HH. Inactivation of Tm6sf2, a Gene Defective in Fatty Liver Disease, Impairs Lipidation but Not Secretion of Very Low Density Lipoproteins. J Biol Chem 2016; 291:10659-76. [PMID: 27013658 PMCID: PMC4865914 DOI: 10.1074/jbc.m116.719955] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Indexed: 12/18/2022] Open
Abstract
A missense mutation (E167K) in TM6SF2 (transmembrane 6 superfamily member 2), a polytopic protein of unknown function, is associated with the full spectrum of fatty liver disease. To investigate the role of TM6SF2 in hepatic triglyceride (TG) metabolism, we inactivated the gene in mice. Chronic inactivation of Tm6sf2 in mice is associated with hepatic steatosis, hypocholesterolemia, and transaminitis, thus recapitulating the phenotype observed in humans. No dietary challenge was required to elicit the phenotype. Immunocytochemical and cell fractionation studies revealed that TM6SF2 was present in the endoplasmic reticulum and Golgi complex, whereas the excess neutral lipids in the Tm6sf2(-/-) mice were located in lipid droplets. Plasma VLDL-TG levels were reduced in the KO animals due to a 3-fold decrease in VLDL-TG secretion rate without any associated reduction in hepatic apoB secretion. Both VLDL particle size and plasma cholesterol levels were significantly reduced in KO mice. Despite levels of TM6SF2 protein being 10-fold higher in the small intestine than in the liver, dietary lipid absorption was only modestly reduced in the KO mice. Our data, taken together, reveal that TM6SF2 is required to mobilize neutral lipids for VLDL assembly but is not required for secretion of apoB-containing lipoproteins. Despite TM6SF2 being located in the endoplasmic reticulum and Golgi complex, the lipids that accumulate in its absence reside in lipid droplets.
Collapse
Affiliation(s)
- Eriks Smagris
- From the Departments of Molecular Genetics and Internal Medicine and
| | - Shenise Gilyard
- From the Departments of Molecular Genetics and Internal Medicine and
| | | | | | - Helen H Hobbs
- From the Departments of Molecular Genetics and Internal Medicine and the Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
48
|
Sun X, Haas ME, Miao J, Mehta A, Graham MJ, Crooke RM, Pais de Barros JP, Wang JG, Aikawa M, Masson D, Biddinger SB. Insulin Dissociates the Effects of Liver X Receptor on Lipogenesis, Endoplasmic Reticulum Stress, and Inflammation. J Biol Chem 2015; 291:1115-22. [PMID: 26511317 DOI: 10.1074/jbc.m115.668269] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Indexed: 12/24/2022] Open
Abstract
Diabetes is characterized by increased lipogenesis as well as increased endoplasmic reticulum (ER) stress and inflammation. The nuclear hormone receptor liver X receptor (LXR) is induced by insulin and is a key regulator of lipid metabolism. It promotes lipogenesis and cholesterol efflux, but suppresses endoplasmic reticulum stress and inflammation. The goal of these studies was to dissect the effects of insulin on LXR action. We used antisense oligonucleotides to knock down Lxrα in mice with hepatocyte-specific deletion of the insulin receptor and their controls. We found, surprisingly, that knock-out of the insulin receptor and knockdown of Lxrα produced equivalent, non-additive effects on the lipogenic genes. Thus, insulin was unable to induce the lipogenic genes in the absence of Lxrα, and LXRα was unable to induce the lipogenic genes in the absence of insulin. However, insulin was not required for LXRα to modulate the phospholipid profile, or to suppress genes in the ER stress or inflammation pathways. These data show that insulin is required specifically for the lipogenic effects of LXRα and that manipulation of the insulin signaling pathway could dissociate the beneficial effects of LXR on cholesterol efflux, inflammation, and ER stress from the negative effects on lipogenesis.
Collapse
Affiliation(s)
- Xiaowei Sun
- From the Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Mary E Haas
- From the Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Ji Miao
- From the Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Abhiruchi Mehta
- From the Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | | | | | | | - Jian-Guo Wang
- the Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Masanori Aikawa
- the Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - David Masson
- the Centre de Recherche INSERM-UMR866, Université de Bourgogne, 21000 Dijon, France, and
| | - Sudha B Biddinger
- From the Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115,
| |
Collapse
|
49
|
Rong X, Wang B, Dunham MM, Hedde PN, Wong JS, Gratton E, Young SG, Ford DA, Tontonoz P. Lpcat3-dependent production of arachidonoyl phospholipids is a key determinant of triglyceride secretion. eLife 2015; 4. [PMID: 25806685 PMCID: PMC4400582 DOI: 10.7554/elife.06557] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 03/24/2015] [Indexed: 11/13/2022] Open
Abstract
The role of specific phospholipids (PLs) in lipid transport has been difficult to assess due to an inability to selectively manipulate membrane composition in vivo. Here we show that the phospholipid remodeling enzyme lysophosphatidylcholine acyltransferase 3 (Lpcat3) is a critical determinant of triglyceride (TG) secretion due to its unique ability to catalyze the incorporation of arachidonate into membranes. Mice lacking Lpcat3 in the intestine fail to thrive during weaning and exhibit enterocyte lipid accumulation and reduced plasma TGs. Mice lacking Lpcat3 in the liver show reduced plasma TGs, hepatosteatosis, and secrete lipid-poor very low-density lipoprotein (VLDL) lacking arachidonoyl PLs. Mechanistic studies indicate that Lpcat3 activity impacts membrane lipid mobility in living cells, suggesting a biophysical basis for the requirement of arachidonoyl PLs in lipidating lipoprotein particles. These data identify Lpcat3 as a key factor in lipoprotein production and illustrate how manipulation of membrane composition can be used as a regulatory mechanism to control metabolic pathways.
Collapse
Affiliation(s)
- Xin Rong
- Department of Pathology and Laboratory Medicine, Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, United States
| | - Bo Wang
- Department of Pathology and Laboratory Medicine, Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, United States
| | - Merlow M Dunham
- Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, United States
| | - Per Niklas Hedde
- Laboratory of Fluorescence Dynamics, Biomedical Engineering Department, University of California, Irvine, Irvine, United States
| | - Jinny S Wong
- Electron Microscopy Core, Gladstone Institute of Cardiovascular Disease, San Francisco, United States
| | - Enrico Gratton
- Laboratory of Fluorescence Dynamics, Biomedical Engineering Department, University of California, Irvine, Irvine, United States
| | - Stephen G Young
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - David A Ford
- Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, United States
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, United States
| |
Collapse
|
50
|
Manchekar M, Liu Y, Sun Z, Richardson PE, Dashti N. Phospholipid transfer protein plays a major role in the initiation of apolipoprotein B-containing lipoprotein assembly in mouse primary hepatocytes. J Biol Chem 2015; 290:8196-205. [PMID: 25638820 DOI: 10.1074/jbc.m114.602748] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In this study, we tested the hypothesis that phospholipid transfer protein (PLTP) is a plausible mediator of phospholipid (PL) transfer to the N-terminal 1000 residues of apoB (apoB:1000) leading to the initiation of apoB-containing lipoprotein assembly. To this end, primary hepatocytes from wild type (WT) and PLTP knock-out (KO) mice were transduced with adenovirus-apoB:1000 with or without co-transduction with adenovirus-PLTP, and the assembly and secretion of apoB:1000-containing lipoproteins were assessed. PLTP deficiency resulted in a 65 and 72% reduction in the protein and lipid content, respectively, of secreted apoB:1000-containing lipoproteins. Particles secreted by WT hepatocytes contained 69% PL, 9% diacylglycerol (DAG), and 23% triacylglycerol (TAG) with a stoichiometry of 46 PL, 6 DAG, and 15 TAG molecules per apoB:1000. PLTP absence drastically altered the lipid composition of apoB:1000 lipoproteins; these particles contained 46% PL, 13% DAG, and 41% TAG with a stoichiometry of 27 PL, 10 DAG, and 23 TAG molecules per apoB:1000. Reintroduction of Pltp gene into PLTP-KO hepatocytes stimulated the lipidation and secretion of apoB:1000-containing lipoproteins by ∼3-fold; the lipid composition and stoichiometry of these particles were identical to those secreted by WT hepatocytes. In contrast to the WT, apoB:1000 in PLTP-KO hepatocytes was susceptible to intracellular degradation predominantly in the post-endoplasmic reticulum, presecretory compartment. Reintroduction of Pltp gene into PLTP-KO hepatocytes restored the stability of apoB:1000. These results provide compelling evidence that in hepatocytes initial recruitment of PL by apoB:1000 leading to the formation of the PL-rich apoB-containing initiation complex is mediated to a large extent by PLTP.
Collapse
Affiliation(s)
- Medha Manchekar
- From the Department of Medicine, Division of Gerontology, Geriatrics, and Palliative Care, Basic Sciences Section, University of Alabama, Birmingham, Alabama 35294 and
| | - Yanwen Liu
- From the Department of Medicine, Division of Gerontology, Geriatrics, and Palliative Care, Basic Sciences Section, University of Alabama, Birmingham, Alabama 35294 and
| | - Zhihuan Sun
- From the Department of Medicine, Division of Gerontology, Geriatrics, and Palliative Care, Basic Sciences Section, University of Alabama, Birmingham, Alabama 35294 and
| | - Paul E Richardson
- the Department of Chemistry and Physics, Coastal Carolina University, Conway, South Carolina 29528
| | - Nassrin Dashti
- From the Department of Medicine, Division of Gerontology, Geriatrics, and Palliative Care, Basic Sciences Section, University of Alabama, Birmingham, Alabama 35294 and
| |
Collapse
|