1
|
McPhee M, Lee J, Salsman J, Pinelli M, Di Cara F, Rosen K, Dellaire G, Ridgway ND. Nuclear lipid droplets in Caco2 cells originate from nascent precursors and in situ at the nuclear envelope. J Lipid Res 2024; 65:100540. [PMID: 38570093 PMCID: PMC11077042 DOI: 10.1016/j.jlr.2024.100540] [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: 07/11/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/05/2024] Open
Abstract
Intestinal epithelial cells convert excess fatty acids into triglyceride (TAG) for storage in cytoplasmic lipid droplets and secretion in chylomicrons. Nuclear lipid droplets (nLDs) are present in intestinal cells but their origin and relationship to cytoplasmic TAG synthesis and secretion is unknown. nLDs and related lipid-associated promyelocytic leukemia structures (LAPS) were abundant in oleate-treated Caco2 but less frequent in other human colorectal cancer cell lines and mouse intestinal organoids. nLDs and LAPS in undifferentiated oleate-treated Caco2 cells harbored the phosphatidate phosphatase Lipin1, its product diacylglycerol, and CTP:phosphocholine cytidylyltransferase (CCT)α. CCTα knockout Caco2 cells had fewer but larger nLDs, indicating a reliance on de novo PC synthesis for assembly. Differentiation of Caco2 cells caused large nLDs and LAPS to form regardless of oleate treatment or CCTα expression. nLDs and LAPS in Caco2 cells did not associate with apoCIII and apoAI and formed dependently of microsomal triglyceride transfer protein expression and activity, indicating they are not derived from endoplasmic reticulum luminal LDs precursors. Instead, undifferentiated Caco2 cells harbored a constitutive pool of nLDs and LAPS in proximity to the nuclear envelope that expanded in size and number with oleate treatment. Inhibition of TAG synthesis did affect the number of nascent nLDs and LAPS but prevented their association with promyelocytic leukemia protein, Lipin1α, and diacylglycerol, which instead accumulated on the nuclear membranes. Thus, nLD and LAPS biogenesis in Caco2 cells is not linked to lipoprotein secretion but involves biogenesis and/or expansion of nascent nLDs by de novo lipid synthesis.
Collapse
Affiliation(s)
- Michael McPhee
- Depts of Pediatrics and Biochemistry & Molecular Biology, Atlantic Research Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jonghwa Lee
- Depts of Pediatrics and Biochemistry & Molecular Biology, Atlantic Research Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jayme Salsman
- Depts of Pathology and Biochemistry & Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Marinella Pinelli
- Dept of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Francesca Di Cara
- Dept of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kirill Rosen
- Depts of Pediatrics and Biochemistry & Molecular Biology, Atlantic Research Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Graham Dellaire
- Depts of Pathology and Biochemistry & Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada.
| | - Neale D Ridgway
- Depts of Pediatrics and Biochemistry & Molecular Biology, Atlantic Research Centre, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
2
|
Slane EG, Tambrini SJ, Cummings BS. Therapeutic potential of lipin inhibitors for the treatment of cancer. Biochem Pharmacol 2024; 222:116106. [PMID: 38442792 DOI: 10.1016/j.bcp.2024.116106] [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: 12/18/2023] [Revised: 01/28/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
Lipins are phosphatidic acid phosphatases (PAP) that catalyze the conversion of phosphatidic acid (PA) to diacylglycerol (DAG). Three lipin isoforms have been identified: lipin-1, -2 and -3. In addition to their PAP activity, lipin-1 and -2 act as transcriptional coactivators and corepressors. Lipins have been intensely studied for their role in regulation of lipid metabolism and adipogenesis; however, lipins are hypothesized to mediate several pathologies, such as those involving metabolic diseases, neuropathy and even cognitive impairment. Recently, an emerging role for lipins have been proposed in cancer. The study of lipins in cancer has been hampered by lack of inhibitors that have selectivity for lipins, that differentiate between lipin family members, or that are suitable for in vivo studies. Such inhibitors have the potential to be extremely useful as both molecular tools and therapeutics. This review describes the expression and function of lipins in various tissues and their roles in several diseases, but with an emphasis on their possible role in cancer. The mechanisms by which lipins mediate cancer cell growth are discussed and the potential usefulness of selective lipin inhibitors is hypothesized. Finally, recent studies reporting the crystallization of lipin-1 are discussed to facilitate rational design of novel lipin inhibitors.
Collapse
Affiliation(s)
- Elizabeth G Slane
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Samantha J Tambrini
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Brian S Cummings
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
3
|
Foster J, McPhee M, Yue L, Dellaire G, Pelech S, Ridgway ND. Lipid- and phospho-regulation of CTP:Phosphocholine Cytidylyltransferase α association with nuclear lipid droplets. Mol Biol Cell 2024; 35:ar33. [PMID: 38170618 PMCID: PMC10916874 DOI: 10.1091/mbc.e23-09-0354] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Fatty acids stored in triacylglycerol-rich lipid droplets are assembled with a surface monolayer composed primarily of phosphatidylcholine (PC). Fatty acids stimulate PC synthesis by translocating CTP:phosphocholine cytidylyltransferase (CCT) α to the inner nuclear membrane, nuclear lipid droplets (nLD) and lipid associated promyelocytic leukemia (PML) structures (LAPS). Huh7 cells were used to identify how CCTα translocation onto these nuclear structures are regulated by fatty acids and phosphorylation of its serine-rich P-domain. Oleate treatment of Huh7 cells increased nLDs and LAPS that became progressively enriched in CCTα. In cells expressing the phosphatidic acid phosphatase Lipin1α or 1β, the expanded pool of nLDs and LAPS had a proportional increase in associated CCTα. In contrast, palmitate induced few nLDs and LAPS and inhibited the oleate-dependent translocation of CCTα without affecting total nLDs. Phospho-memetic or phospho-null mutations in the P-domain revealed that a 70% phosphorylation threshold, rather than site-specific phosphorylation, regulated CCTα association with nLDs and LAPS. In vitro candidate kinase and inhibitor studies in Huh7 cells identified cyclin-dependent kinase (CDK) 1 and 2 as putative P-domain kinases. In conclusion, CCTα translocation onto nLDs and LAPS is dependent on available surface area and fatty acid composition, as well as threshold phosphorylation of the P-domain potentially involving CDKs.
Collapse
Affiliation(s)
- Jason Foster
- Departments of Pediatrics and Biochemistry & Molecular Biology, Atlantic Research Centre, and
| | - Michael McPhee
- Departments of Pediatrics and Biochemistry & Molecular Biology, Atlantic Research Centre, and
| | - Lambert Yue
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada V6T 2B5
| | - Graham Dellaire
- Departments of Pathology and Biochemistry & Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada B3H4R2
| | - Steven Pelech
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada V6T 2B5
- Kinexus Bioinformatics Corporation, Vancouver, BC, Canada V6P 6T3
| | - Neale D. Ridgway
- Departments of Pediatrics and Biochemistry & Molecular Biology, Atlantic Research Centre, and
| |
Collapse
|
4
|
Shi Y, Pizzini J, Wang H, Das F, Abdul Azees PA, Ghosh Choudhury G, Barnes JL, Zang M, Weintraub ST, Yeh CK, Katz MS, Kamat A. β2-Adrenergic receptor agonist induced hepatic steatosis in mice: modeling nonalcoholic fatty liver disease in hyperadrenergic states. Am J Physiol Endocrinol Metab 2021; 321:E90-E104. [PMID: 34029162 PMCID: PMC8321826 DOI: 10.1152/ajpendo.00651.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/23/2021] [Accepted: 05/08/2021] [Indexed: 12/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a spectrum of disorders ranging from hepatic steatosis [excessive accumulation of triglycerides (TG)] to nonalcoholic steatohepatitis, which can progress to cirrhosis and hepatocellular carcinoma. The molecular pathogenesis of steatosis and progression to more severe NAFLD remains unclear. Obesity and aging, two principal risk factors for NAFLD, are associated with a hyperadrenergic state. β-Adrenergic responsiveness in liver increases in animal models of obesity and aging, and in both is linked to increased hepatic expression of β2-adrenergic receptors (β2-ARs). We previously showed that in aging rodents intracellular signaling from elevated hepatic levels of β2-ARs may contribute to liver steatosis. In this study we demonstrate that injection of formoterol, a highly selective β2-AR agonist, to mice acutely results in hepatic TG accumulation. Further, we have sought to define the intrahepatic mechanisms underlying β2-AR mediated steatosis by investigating changes in hepatic expression and cellular localization of enzymes, transcription factors, and coactivators involved in processes of lipid accrual and disposition-and also functional aspects thereof-in livers of formoterol-treated animals. Our results suggest that β2-AR activation by formoterol leads to increased hepatic TG synthesis and de novo lipogenesis, increased but incomplete β-oxidation of fatty acids with accumulation of potentially toxic long-chain acylcarnitine intermediates, and reduced TG secretion-all previously invoked as contributors to fatty liver disease. Experiments are ongoing to determine whether sustained activation of hepatic β2-AR signaling by formoterol might be utilized to model fatty liver changes occurring in hyperadrenergic states of obesity and aging, and thereby identify novel molecular targets for the prevention or treatment of NAFLD.NEW & NOTEWORTHY Results of our study suggest that β2-adrenergic receptor (β2-AR) activation by agonist formoterol leads to increased hepatic TG synthesis and de novo lipogenesis, incomplete β-oxidation of fatty acids with accumulation of long-chain acylcarnitine intermediates, and reduced TG secretion. These findings may, for the first time, implicate a role for β2-AR responsive dysregulation of hepatic lipid metabolism in the pathogenetic processes underlying NAFLD in hyperadrenergic states such as obesity and aging.
Collapse
Affiliation(s)
- Yun Shi
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Jason Pizzini
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Hanzhou Wang
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Falguni Das
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Parveez Ahamed Abdul Azees
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Goutam Ghosh Choudhury
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Jeffrey L Barnes
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Mengwei Zang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, Texas
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Geriatric Research, Education and Clinical Center, Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, Texas
| | - Susan T Weintraub
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Chih-Ko Yeh
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Geriatric Research, Education and Clinical Center, Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, Texas
| | - Michael S Katz
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Geriatric Research, Education and Clinical Center, Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, Texas
| | - Amrita Kamat
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Geriatric Research, Education and Clinical Center, Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, Texas
| |
Collapse
|
5
|
Sołtysik K, Ohsaki Y, Tatematsu T, Cheng J, Maeda A, Morita SY, Fujimoto T. Nuclear lipid droplets form in the inner nuclear membrane in a seipin-independent manner. J Cell Biol 2021; 220:211592. [PMID: 33315072 PMCID: PMC7737703 DOI: 10.1083/jcb.202005026] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/25/2020] [Accepted: 11/11/2020] [Indexed: 01/09/2023] Open
Abstract
Nuclear lipid droplets (LDs) in hepatocytes are derived from precursors of very-low-density lipoprotein in the ER lumen, but it is not known how cells lacking the lipoprotein secretory function form nuclear LDs. Here, we show that the inner nuclear membrane (INM) of U2OS cells harbors triglyceride synthesis enzymes, including ACSL3, AGPAT2, GPAT3/GPAT4, and DGAT1/DGAT2, and generates nuclear LDs in situ. mTOR inhibition increases nuclear LDs by inducing the nuclear translocation of lipin-1 phosphatidic acid (PA) phosphatase. Seipin, a protein essential for normal cytoplasmic LD formation in the ER, is absent in the INM. Knockdown of seipin increases nuclear LDs and PA in the nucleus, whereas seipin overexpression decreases these. Seipin knockdown also up-regulates lipin-1β expression, and lipin-1 knockdown decreases the effect of seipin knockdown on nuclear LDs without affecting PA redistribution. These results indicate that seipin is not directly involved in nuclear LD formation but instead restrains it by affecting lipin-1 expression and intracellular PA distribution.
Collapse
Affiliation(s)
- Kamil Sołtysik
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuki Ohsaki
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tsuyako Tatematsu
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jinglei Cheng
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Asami Maeda
- Laboratory of Molecular Cell Biology, Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shin-Ya Morita
- Department of Pharmacy, Shiga University of Medical Science Hospital, Otsu City, Shiga, Japan
| | - Toyoshi Fujimoto
- Laboratory of Molecular Cell Biology, Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Lu S, Lyu Z, Wang Z, Kou Y, Liu C, Li S, Hu M, Zhu H, Wang W, Zhang C, Kuan YS, Liu YW, Chen J, Tian J. Lipin 1 deficiency causes adult-onset myasthenia with motor neuron dysfunction in humans and neuromuscular junction defects in zebrafish. Theranostics 2021; 11:2788-2805. [PMID: 33456573 PMCID: PMC7806489 DOI: 10.7150/thno.53330] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/12/2020] [Indexed: 12/03/2022] Open
Abstract
Lipin 1 is an intracellular protein acting as a phosphatidic acid phosphohydrolase enzyme controlling lipid metabolism. Human recessive mutations in LPIN1 cause recurrent, early-onset myoglobinuria, a condition normally associated with muscle pain and weakness. Whether and how lipin 1 deficiency in humans leads to peripheral neuropathy is yet unclear. Herein, two novel compound heterozygous mutations in LPIN1 with neurological disorders, but no myoglobinuria were identified in an adult-onset syndromic myasthenia family. The present study sought to explore the pathogenic mechanism of LPIN1 in muscular and neural development. Methods: The clinical diagnosis of the proband was compared to the known 48 cases of LPIN1 recessive homozygous mutations. Whole-exome sequencing was carried out on the syndromic myasthenia family to identify the causative gene. The pathogenesis of lipin 1 deficiency during somitogenesis and neurogenesis was investigated using the zebrafish model. Whole-mount in situ hybridization, immunohistochemistry, birefringence analysis, touch-evoke escape response and locomotion assays were performed to observe in vivo the changes in muscles and neurons. The conservatism of the molecular pathways regulated by lipin 1 was evaluated in human primary glioblastoma and mouse myoblast cells by siRNA knockdown, drug treatment, qRT-PCR and Western blotting analysis. Results: The patient exhibited adult-onset myasthenia accompanied by muscle fiber atrophy and nerve demyelination without myoglobinuria. Two novel heterozygous mutations, c.2047A>C (p.I683L) and c.2201G>A (p.R734Q) in LPIN1, were identified in the family and predicted to alter the tertiary structure of LPIN1 protein. Lipin 1 deficiency in zebrafish embryos generated by lpin1 morpholino knockdown or human LPIN1 mutant mRNA injections reproduced the myotomes defects, a reduction both in primary motor neurons and secondary motor neurons projections, morphological changes of post-synaptic clusters of acetylcholine receptors, and myelination defects, which led to reduced touch-evoked response and abnormalities of swimming behaviors. Loss of lipin 1 function in zebrafish and mammalian cells also exhibited altered expression levels of muscle and neuron markers, as well as abnormally enhanced Notch signaling, which was partially rescued by the specific Notch pathway inhibitor DAPT. Conclusions: These findings pointed out that the compound heterozygous mutations in human LPIN1 caused adult-onset syndromic myasthenia with peripheral neuropathy. Moreover, zebrafish could be used to model the neuromuscular phenotypes due to the lipin 1 deficiency, where a novel pathological role of over-activated Notch signaling was discovered and further confirmed in mammalian cell lines.
Collapse
|
7
|
Lee J, Salsman J, Foster J, Dellaire G, Ridgway ND. Lipid-associated PML structures assemble nuclear lipid droplets containing CCTα and Lipin1. Life Sci Alliance 2020; 3:3/8/e202000751. [PMID: 32461215 PMCID: PMC7266991 DOI: 10.26508/lsa.202000751] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 11/24/2022] Open
Abstract
PML proteins assemble into noncanonical lipid-associated PML structures (LAPS) on nuclear lipid droplets, which recruit CCTα and Lipin1 for the synthesis of phosphatidylcholine and triacylglycerol. Nuclear lipid droplets (nLDs) form on the inner nuclear membrane by a mechanism involving promyelocytic leukemia (PML), the protein scaffold of PML nuclear bodies. We report that PML structures on nLDs in oleate-treated U2OS cells, referred to as lipid-associated PML structures (LAPS), differ from canonical PML nuclear bodies by the relative absence of SUMO1, SP100, and DAXX. These nLDs were also enriched in CTP:phosphocholine cytidylyltransferase α (CCTα), the phosphatidic acid phosphatase Lipin1, and DAG. Translocation of CCTα onto nLDs was mediated by its α-helical M-domain but was not correlated with its activator DAG. High-resolution imaging revealed that CCTα and LAPS occupied distinct polarized regions on nLDs. PML knockout U2OS (PML KO) cells lacking LAPS had a 40–50% reduction in nLDs with associated CCTα, and residual nLDs were almost devoid of Lipin1 and DAG. As a result, phosphatidylcholine and triacylglycerol synthesis was inhibited in PML KO cells. We conclude that in response to excess exogenous fatty acids, LAPS are required to assemble nLDs that are competent to recruit CCTα and Lipin1.
Collapse
Affiliation(s)
- Jonghwa Lee
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada
| | - Jayme Salsman
- Department of Pathology, Dalhousie University, Halifax, Canada
| | - Jason Foster
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada
| | - Graham Dellaire
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada .,Department of Pathology, Dalhousie University, Halifax, Canada
| | - Neale D Ridgway
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada .,Department of Pediatrics, Dalhousie University, Halifax, Canada
| |
Collapse
|
8
|
Lee J, Ridgway ND. Substrate channeling in the glycerol-3-phosphate pathway regulates the synthesis, storage and secretion of glycerolipids. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1865:158438. [PMID: 30959116 DOI: 10.1016/j.bbalip.2019.03.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 01/16/2023]
Abstract
The successive acylation of glycerol-3-phosphate (G3P) by glycerol-3-phosphate acyltransferases and acylglycerol-3-phosphate acyltransferases produces phosphatidic acid (PA), a precursor for CDP-diacylglycerol-dependent phospholipid synthesis. PA is further dephosphorylated by LIPINs to produce diacylglycerol (DG), a substrate for the synthesis of triglyceride (TG) by DG acyltransferases and a precursor for phospholipid synthesis via the CDP-choline and CDP-ethanolamine (Kennedy) pathways. The channeling of fatty acids into TG for storage in lipid droplets and secretion in lipoproteins or phospholipids for membrane biogenesis is dependent on isoform expression, activity and localization of G3P pathway enzymes, as well as dietary and hormonal and tissue-specific factors. Here, we review the mechanisms that control partitioning of substrates into lipid products of the G3P pathway.
Collapse
Affiliation(s)
- Jonghwa Lee
- Atlantic Research Center, Depts. of Pediatrics and Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Neale D Ridgway
- Atlantic Research Center, Depts. of Pediatrics and Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
9
|
You M, Jogasuria A, Lee K, Wu J, Zhang Y, Lee YK, Sadana P. Signal Transduction Mechanisms of Alcoholic Fatty Liver Disease: Emer ging Role of Lipin-1. Curr Mol Pharmacol 2019; 10:226-236. [PMID: 26278388 DOI: 10.2174/1874467208666150817112109] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 08/07/2015] [Accepted: 08/07/2015] [Indexed: 02/06/2023]
Abstract
Lipin-1, a mammalian phosphatidic acid phosphatase (PAP), is a bi-functional molecule involved in various signaling pathways via its function as a PAP enzyme in the triglyceride synthesis pathway and in the nucleus as a transcriptional co-regulator. In the liver, lipin-1 is known to play a vital role in controlling the lipid metabolism and inflammation process at multiple regulatory levels. Alcoholic fatty liver disease (AFLD) is one of the earliest forms of liver injury and approximately 8-20% of patients with simple steatosis can develop into more severe forms of liver injury, including steatohepatitis, fibrosis/ cirrhosis, and eventually hepatocellular carcinoma (HCC). The signal transduction mechanisms for alcohol-induced detrimental effects in liver involves alteration of complex and multiple signaling pathways largely governed by a central and upstream signaling system, namely, sirtuin 1 (SIRT1)-AMP activated kinase (AMPK) axis. Emerging evidence suggests a pivotal role of lipin-1 as a crucial downstream regulator of SIRT1-AMPK signaling system that is likely to be ultimately responsible for development and progression of AFLD. Several lines of evidence demonstrate that ethanol exposure significantly induces lipin-1 gene and protein expression levels in cultured hepatocytes and in the livers of rodents, induces lipin-1-PAP activity, impairs the functional activity of nuclear lipin-1, disrupts lipin-1 mRNA alternative splicing and induces lipin-1 nucleocytoplasmic shuttling. Such impairment in response to ethanol leads to derangement of hepatic lipid metabolism, and excessive production of inflammatory cytokines in the livers of the rodents and human alcoholics. This review summarizes current knowledge about the role of lipin-1 in the pathogenesis of AFLD and its potential signal transduction mechanisms.
Collapse
Affiliation(s)
- Min You
- 4209 State Route 44, Rootstown OH 44272. United States
| | | | | | - Jiashin Wu
- Department of Pharmaceutical Sciences. 0
| | - Yanqiao Zhang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, College of Pharmacy and College of Medicine, Rootstown OH 44272. United States
| | - Yoon Kwang Lee
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, College of Pharmacy and College of Medicine, Rootstown OH 44272. United States
| | | |
Collapse
|
10
|
Yu J, Peng J, Luan Z, Zheng F, Su W. MicroRNAs as a Novel Tool in the Diagnosis of Liver Lipid Dysregulation and Fatty Liver Disease. Molecules 2019; 24:molecules24020230. [PMID: 30634538 PMCID: PMC6358728 DOI: 10.3390/molecules24020230] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/23/2018] [Accepted: 12/24/2018] [Indexed: 02/07/2023] Open
Abstract
In recent years, metabolic disorder, especially fatty liver disease, has been considered a major challenge to global health. The attention of researchers focused on expanding knowledge of the regulation mechanism behind these diseases and towards the new diagnostics tools and treatments. The pathophysiology of the fatty liver disease is undoubtedly complex. Abnormal hepatic lipid accumulation is a major symptom of most metabolic diseases. Therefore, the identification of novel regulation factors of lipid metabolism is important and meaningful. As a new diagnostic tool, the function of microRNAs during fatty liver disease has recently come into notice in biological research. Accumulating evidence supports the influence of miRNAs in lipid metabolism. In this review, we discuss the potential role of miRNAs in liver lipid metabolism and the pathogenesis of fatty liver disease.
Collapse
Affiliation(s)
- Jingwei Yu
- Shenzhen University Medical Center, Shenzhen University Health Science Center, Shenzhen 518060, China.
- Department of Biology, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Jun Peng
- Shenzhen University Medical Center, Shenzhen University Health Science Center, Shenzhen 518060, China.
| | - Zhilin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Feng Zheng
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Wen Su
- Shenzhen University Medical Center, Shenzhen University Health Science Center, Shenzhen 518060, China.
| |
Collapse
|
11
|
Sellers RS, Mahmood SR, Perumal GS, Macaluso FP, Kurland IJ. Phenotypic Modulation of Skeletal Muscle Fibers in LPIN1-Deficient Lipodystrophic ( fld) Mice. Vet Pathol 2018; 56:322-331. [PMID: 30381013 DOI: 10.1177/0300985818809126] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lipin-1 ( Lpin1)-deficient lipodystrophic mice have scant and immature adipocytes and develop transient fatty liver early in life. Unlike normal mice, these mice cannot rely on stored triglycerides to generate adenosine triphosphate (ATP) from the β-oxidation of fatty acids during periods of fasting. To compensate, these mice store much higher amounts of glycogen in skeletal muscle and liver than wild-type mice in order to support energy needs during periods of fasting. Our studies demonstrated that there are phenotypic changes in skeletal muscle fibers that reflect an adaptation to this unique metabolic situation. The phenotype of skeletal muscle (soleus, gastrocnemius, plantaris, and extensor digitorum longus [EDL]) from Lpin1-/- was evaluated using various methods including immunohistochemistry for myosin heavy chains (Myh) 1, 2, 2a, 2b, and 2x; enzyme histochemistry for myosin ATPase, cytochrome-c oxidase (COX), and succinyl dehydrogenase (SDH); periodic acid-Schiff; and transmission electron microscopy. Fiber-type changes in the soleus muscle of Lpin1-/- mice were prominent and included decreased Myh1 expression with concomitant increases in Myh2 expression and myosin-ATPase activity; this change was associated with an increase in the presence of Myh1/2a or Myh1/2x hybrid fibers. Alterations in mitochondrial enzyme activity (COX and SDH) were apparent in the myofibers in the soleus, gastrocnemius, plantaris, and EDL muscles. Electron microscopy revealed increases in the subsarcolemmal mitochondrial mass in the muscles of Lpin1-/- mice. These data demonstrate that lipin-1 deficiency results in phenotypic fiber-specific modulation of skeletal muscle necessary for compensatory fuel utilization adaptations in lipodystrophy.
Collapse
Affiliation(s)
- Rani S Sellers
- 1 Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA.,Current address: Drug Safety and Research Development, Pfizer, Inc, Pearl River, NY, USA
| | - S Radma Mahmood
- 1 Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Geoffrey S Perumal
- 2 Analytical Imaging Facility, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Frank P Macaluso
- 2 Analytical Imaging Facility, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Irwin J Kurland
- 3 Department of Medicine (Endocrinology), Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
12
|
Wang J, Wang S, Yan C, Deng Y, Huang Z, Shi P. Mass spectrometry-based proteomic analysis reveals the interacting partners of lipin1. IUBMB Life 2018; 70:753-762. [PMID: 30092116 DOI: 10.1002/iub.1873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 04/25/2018] [Indexed: 12/13/2022]
Abstract
As a lipin family founding member, lipin1 exerts dual functions as a phosphatidate phosphatase enzyme and/or a co-transcriptional regulator in lipid metabolism. In fact, it is also involved in many other cell processes. In this study, we utilized pull down assay coupled with mass spectrometry (MS) to unravel protein-protein interaction networks of lipin1 in 293T human embryonic kidney cells. Pull-down assay on the Ni2+ -chelating column was used to isolate lipin1 complexes from 293T cells transfected with 6-His tagged lipin1. The lipin1 complexes were analyzed on Q Exactive mass spectrometer. A total of 30 proteins were identified from label free quantitation of the MS data by Proteome Discoverer platform. The physical interaction between lipin1 and eEF1A1 was further affirmed in 293T cells transfected with 6-His tagged lipin1 and hepatocyte SMMC7721 cells by protein immunoprecipitation and immunofluorescence microscopy. Lipin1 also interacted with HIST1H2BK, which was confirmed in SMMC7721 cells by protein immunoprecipitation. Our proteomic analysis implicated lipin1 in novel roles in various cellular processes. © 2018 IUBMB Life, 70(8):753-762, 2018.
Collapse
Affiliation(s)
- Jian Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Song Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Chongjia Yan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Yunxia Deng
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, 2999 Renmin Road, Shanghai, 201620, China
| | - Zhiwei Huang
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, 2999 Renmin Road, Shanghai, 201620, China
| | - Ping Shi
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| |
Collapse
|
13
|
Lee J, Hong SW, Kwon H, Park SE, Rhee EJ, Park CY, Oh KW, Park SW, Lee WY. Exendin-4 improves ER stress-induced lipid accumulation and regulates lipin-1 signaling in HepG2 cells. Cell Stress Chaperones 2018; 23:629-638. [PMID: 29934713 PMCID: PMC6045528 DOI: 10.1007/s12192-017-0872-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 12/20/2017] [Accepted: 12/25/2017] [Indexed: 12/12/2022] Open
Abstract
Lipin-1 performs dual function during lipid metabolism, i.e., it functions as a transcriptional coactivator and as a phosphatidate phosphatase during triglyceride biosynthesis. We investigated whether exendin-4 prevented endoplasmic reticulum (ER) stress-induced hepatic steatosis and whether the protective effects of exendin-4 were associated with lipin-1 signaling. Tunicamycin and thapsigargin, ER stress inducers, increased triglycerides (TG) content and expression of genes encoding lipid droplet surface proteins. Exendin-4 decreased the expression of ER stress markers phosphorylated PKR like ER kinase (PERK), phosphorylated inositol-requiring enzyme 1 alpha (IRE1α), and glucose-regulated protein 78 kDa (GRP78) proteins and spliced X-box binding protein 1 (XBP-1s) mRNA and increased the expression of genes encoding lipolytic enzymes hormone-sensitive lipase (HSL) and monoacylglycerol lipase (MGL) and VLDL assembly-associated proteins microsomal triglyceride transfer protein (MTP) and apolipoprotein B (APOB) in tunicamycin-pretreated cells. Moreover, exendin-4 significantly decreased lipin-1β/α ratio by increasing SFRP10 and increased lipin-1 nuclear localization. The decrease in lipin-1β/α ratio was also observed in SIRT1 and AMPK agonist-treated cells. These data suggest that exendin-4 improves ER stress-induced hepatic lipid accumulation by increasing lipolysis and VLDL assembly, which is partially mediated by the regulation of lipin-1 signaling.
Collapse
Affiliation(s)
- Jinmi Lee
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Seok-Woo Hong
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Hyemi Kwon
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Se Eun Park
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Eun-Jung Rhee
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Cheol-Young Park
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Ki-Won Oh
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Sung-Woo Park
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Won-Young Lee
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea.
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea.
| |
Collapse
|
14
|
Abstract
Alcoholic liver disease (ALD) is a definition encompassing a spectrum of disorders ranging from simple steatosis to cirrhosis and hepatocellular carcinoma. Excessive alcohol consumption triggers a series of metabolic reactions that affect the liver by inducing lipogenesis, increasing oxidative stress, and causing abnormal inflammatory responses. The metabolic pathways regulating lipids, reactive oxygen species (ROS), and immune system are closely related and in some cases cross-regulate each other. Therefore, it must be taken into account that major genetic and epigenetic abnormalities affecting enzymes involved in one of such pathways can play a pivotal role in ALD pathogenesis. However, recent studies have pointed out how a significant predisposition can also be determined by minor variants, such as relatively common polymorphisms, epigenetic modifications, and microRNA abnormalities. Genetic and epigenetic factors can also affect the progression of liver diseases, promoting fibrogenesis, cirrhosis, and ultimately hepatocellular carcinoma. It is noteworthy that some of these factors, such as some of the cytokines involved in the abnormal inflammatory responses, are shared with non-alcoholic liver disease, while other factors are unique to ALD. The study of the genetic and epigenetic components involved in the liver damages caused by alcohol is crucial to identify individuals with high risk of developing ALD, design personalized protocols for prevention and/or treatment, and select the best molecular targets for new therapies.
Collapse
Affiliation(s)
- Luigi Boccuto
- Greenwood Genetic Center, Greenwood, SC, USA. Clemson University School of Health Research, Clemson, SC, USA
| | - Ludovico Abenavoli
- Department of Health Sciences, University "Magna Græcia", Catanzaro, Italy
| |
Collapse
|
15
|
Ren L, Sun Y, Lu H, Ye D, Han L, Wang N, Daugherty A, Li F, Wang M, Su F, Tao W, Sun J, Zelcer N, Mullick AE, Danser AHJ, Jiang Y, He Y, Ruan X, Lu X. (Pro)renin Receptor Inhibition Reprograms Hepatic Lipid Metabolism and Protects Mice From Diet-Induced Obesity and Hepatosteatosis. Circ Res 2018; 122:730-741. [PMID: 29301853 DOI: 10.1161/circresaha.117.312422] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 11/18/2017] [Accepted: 12/29/2017] [Indexed: 01/12/2023]
Abstract
RATIONALE An elevated level of plasma LDL (low-density lipoprotein) is an established risk factor for cardiovascular disease. Recently, we reported that the (pro)renin receptor ([P]RR) regulates LDL metabolism in vitro via the LDLR (LDL receptor) and SORT1 (sortilin-1), independently of the renin-angiotensin system. OBJECTIVES To investigate the physiological role of (P)RR in lipid metabolism in vivo. METHODS AND RESULTS We used N-acetylgalactosamine modified antisense oligonucleotides to specifically inhibit hepatic (P)RR expression in C57BL/6 mice and studied the consequences this has on lipid metabolism. In line with our earlier report, hepatic (P)RR silencing increased plasma LDL-C (LDL cholesterol). Unexpectedly, this also resulted in markedly reduced plasma triglycerides in a SORT1-independent manner in C57BL/6 mice fed a normal- or high-fat diet. In LDLR-deficient mice, hepatic (P)RR inhibition reduced both plasma cholesterol and triglycerides, in a diet-independent manner. Mechanistically, we found that (P)RR inhibition decreased protein abundance of ACC (acetyl-CoA carboxylase) and PDH (pyruvate dehydrogenase). This alteration reprograms hepatic metabolism, leading to reduced lipid synthesis and increased fatty acid oxidation. As a result, hepatic (P)RR inhibition attenuated diet-induced obesity and hepatosteatosis. CONCLUSIONS Collectively, our study suggests that (P)RR plays a key role in energy homeostasis and regulation of plasma lipids by integrating hepatic glucose and lipid metabolism.
Collapse
Affiliation(s)
- Liwei Ren
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.)
| | - Yuan Sun
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.)
| | - Hong Lu
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.)
| | - Dien Ye
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.)
| | - Lijuan Han
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.)
| | - Na Wang
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.)
| | - Alan Daugherty
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.)
| | - Furong Li
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.)
| | - Miaomiao Wang
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.)
| | - Fengting Su
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.)
| | - Wenjun Tao
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.)
| | - Jie Sun
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.)
| | - Noam Zelcer
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.)
| | - Adam E Mullick
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.)
| | - A H Jan Danser
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.)
| | - Yizhou Jiang
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.)
| | - Yongcheng He
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.)
| | - Xiongzhong Ruan
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.).
| | - Xifeng Lu
- From the AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (L.R., Y.S., D.Y., L.H., N.W., M.W., F.S., W.T., J.S., X.R., X.L.); Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (L.R., Y.S., F.L., X.L.); Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam University, The Netherlands (L.R., Y.S., A.H.J.D.); Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.L., A.D.); Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (N.Z.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M.); Institute for Advanced Study, Shenzhen University, China (Y.J.); The First Affiliated Hospital of Shenzhen University, China (Y.H.); and John Moorhead Laboratory, Center for Nephrology, University College London, United Kingdom (X.R.).
| |
Collapse
|
16
|
Seo K, Shin SM. Induction of Lipin1 by ROS-Dependent SREBP-2 Activation. Toxicol Res 2017; 33:219-224. [PMID: 28744353 PMCID: PMC5523558 DOI: 10.5487/tr.2017.33.3.219] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/30/2017] [Accepted: 06/12/2017] [Indexed: 01/26/2023] Open
Abstract
Lipin1 was identified as a phosphatidate phosphatase enzyme, and it plays a key role in lipid metabolism. Since free radicals contribute to metabolic diseases in the liver, this study investigated the effects of free radicals on the regulation of Lipin1 expression in Huh7 and AML12 cells. Hydrogen peroxide induced mRNA and protein expression of Lipin1 in Huh7 cells, which was assayed by quantitative RT-PCR and immunoblotting, respectively. Induction of Lipin1 by hydrogen peroxide was confirmed in AML12 cells. Hydrogen peroxide treatment significantly increased expression of sterol regulatory element-binding protein (SREBP)-2, but not SREBP-1. Moreover, nuclear translocation of SREBP-2 was detected after hydrogen peroxide treatment. Hydrogen peroxide-induced Lipin1 or SREBP-2 expression was significantly reduced by N-acetyl-l-cysteine treatment, indicating that reactive oxygen species (ROS) were implicated in Lipin1 expression. Next, we investigated whether the hypoxic environments that cause endogenous ROS production in mitochondria in metabolic diseases affect the expression of Lipin1. Exposure to hypoxia also increased Lipin1 expression. In contrast, pretreatment with antioxidants attenuated hypoxia-induced Lipin1 expression. Collectively, our results show that ROS activate SREBP-2, which induces Lipin1 expression.
Collapse
Affiliation(s)
- Kyuhwa Seo
- College of Pharmacy, Chosun University, Gwangju, Korea
| | - Sang Mi Shin
- College of Pharmacy, Chosun University, Gwangju, Korea
| |
Collapse
|
17
|
Sigruener A, Wolfrum C, Boettcher A, Kopf T, Liebisch G, Orsó E, Schmitz G. Lipidomic and metabolic changes in the P4-type ATPase ATP10D deficient C57BL/6J wild type mice upon rescue of ATP10D function. PLoS One 2017; 12:e0178368. [PMID: 28542499 PMCID: PMC5444826 DOI: 10.1371/journal.pone.0178368] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 05/11/2017] [Indexed: 01/01/2023] Open
Abstract
Background Sequence variants near the human gene for P4-type ATPase, class V, type 10D (ATP10D) were shown to significantly associate with circulating hexosylceramide d18:1/16:0 and d18:1/24:1 levels, obesity, insulin resistance, plasma high density lipoprotein (HDL), coronary stenotic index and intracranial atherosclerotic index. In mice Atp10d is associated with HDL modulation and C57BL/6 mice expressing a truncated, non-functional form of ATP10D easily develop obesity and insulin resistance on high-fat diet. Results We analyzed metabolic differences of ATP10D deficient C57BL/6J wild type and ATP10D transgenic C57BL/6J BAC129 mice. ATP10D transgenic mice gain 25% less weight on high-fat diet concomitant with a reduced increase in fat cell mass but independent of adipocyte size change. ATP10D transgenic mice also had 26% lower triacylglycerol levels with approximately 76% bound to very low density lipoprotein while in ATP10D deficient wild type mice 57% are bound to low density lipoprotein. Furthermore increased oxygen consumption and CO2 production, 38% lower glucose and 69% lower insulin levels and better insulin sensitivity were observed in ATP10D transgenic mice. Besides decreased hexosylceramide species levels were detected. Part of these effects may be due to reduced hepatic stearoyl-CoA desaturase 1 (SCD1) expression in ATP10D transgenic mice, which was reflected by altered fatty acid and lipid species patterns. There was a significant decrease in the hepatic 18:1 to 18:0 free fatty acid ratio in transgenic mice. The ratio of 16:1 to 16:0 was not significantly different. Interestingly both ratios were significantly reduced in plasma total fatty acids. Summary In summary we found that ATP10D reduces high-fat diet induced obesity and improves insulin sensitivity. ATP10D transgenic mice showed altered hepatic expression of lipid-metabolism associated genes, including Scd1, along with changes in hepatic and plasma lipid species and plasma lipoprotein pattern.
Collapse
Affiliation(s)
- Alexander Sigruener
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Medical Center, Regensburg, Germany
- * E-mail:
| | - Christian Wolfrum
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Alfred Boettcher
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Medical Center, Regensburg, Germany
| | - Thomas Kopf
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Medical Center, Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Medical Center, Regensburg, Germany
| | - Evelyn Orsó
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Medical Center, Regensburg, Germany
| | - Gerd Schmitz
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Medical Center, Regensburg, Germany
| |
Collapse
|
18
|
Kajimoto K, Suemitsu E, Sato Y, Sakurai Y, Harashima H. Liver-Specific Silencing of Lipin1 Reduces Fat Mass as Well as Hepatic Triglyceride Biosynthesis in Mice. Biol Pharm Bull 2017; 39:1653-1661. [PMID: 27725442 DOI: 10.1248/bpb.b16-00353] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Lipin1, a bifunctional protein, regulates fatty acid utilization in the triglyceride biosynthesis pathway. In the current study, using a liver-specific in vivo short interfering RNA (siRNA) delivery system, we examined the pathological and physiological roles of hepatic Lipin1 in the development of insulin resistance and the maintenance of systemic energy homeostasis. Liver-specific silencing of Lipin1 expression was achieved by the systemic administration of siRNA against Lpin1 mRNA (siLpin1)-loaded lipid nanoparticles (LNPs) to wild type mice at 3-4 d intervals for 25 d. The siLpin1-treated mice showed normal blood glucose levels and insulin sensitivity, however, triglyceride (TG) levels were reduced in liver and peripheral blood of them. The knockdown of hepatic Lipin1 in mice led to marked decrease in adipose tissue mass and adipocyte diameters in epididymal and inguinal fat depots without the undesired silencing of Lipin1 in adipose tissue. In summary, we report for the first time that the down-regulation of hepatic Lipin1 expression leads to less adiposity as well as a decrease in TG level in the liver and blood circulation, without any alterations in the glucose tolerance and blood glucose levels. Our findings may provide new insights into the physiological roles of hepatic Lipin1 in systemic energy homeostasis.
Collapse
|
19
|
Jiao XL, Jing JJ, Qiao LY, Liu JH, Li LA, Zhang J, Jia XL, Liu WZ. Ontogenetic Expression of Lpin2 and Lpin3 Genes and Their Associations with Traits in Two Breeds of Chinese Fat-tailed Sheep. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2016; 29:333-42. [PMID: 26950863 PMCID: PMC4811783 DOI: 10.5713/ajas.15.0467] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/17/2015] [Accepted: 09/10/2015] [Indexed: 12/14/2022]
Abstract
Lipins play dual function in lipid metabolism by serving as phosphatidate phosphatase and transcriptional co-regulators of gene expression. Mammalian lipin proteins consist of lipin1, lipin2, and lipin3 and are encoded by their respective genes Lpin1, Lpin2, and Lpin3. To date, most studies are concerned with Lpin1, only a few have addressed Lpin2 and Lpin3. Ontogenetic expression of Lpin2 and Lpin3 and their associations with traits would help to explore their molecular and physiological functions in sheep. In this study, 48 animals with an equal number of males and females each for both breeds of fat-tailed sheep such as Guangling Large Tailed (GLT) and Small Tailed Han (STH) were chosen to evaluate the ontogenetic expression of Lpin2 and Lpin3 from eight different tissues and months of age by quantitative real-time polymerase chain reaction (PCR). Associations between gene expression and slaughter and tail traits were also analyzed. The results showed that Lpin2 mRNA was highly expressed in perirenal and tail fats, and was also substantially expressed in liver, kidney, reproductive organs (testis and ovary), with the lowest levels in small intestine and femoral biceps. Lpin3 mRNA was prominently expressed in liver and small intestine, and was also expressed at high levels in kidney, perirenal and tail fats as well as reproductive organs (testis and ovary), with the lowest level in femoral biceps. Global expression of Lpin2 and Lpin3 in GLT both were significantly higher than those in STH. Spatiotemporal expression showed that the highest levels of Lpin2 expression occurred at 10 months of age in two breeds of sheep, with the lowest expression at 2 months of age in STH and at 8 months of age in GLT. The greatest levels of Lpin3 expression occurred at 4 months of age in STH and at 10 months of age in GLT, with the lowest expression at 12 months of age in STH and at 8 months of age in GLT. Breed and age significantly influenced the tissue expression patterns of Lpin2 and Lpin3, respectively, and sex significantly influenced the spatiotemporal expression patterns of Lpin3. Meanwhile, Lpin2 and Lpin3 mRNA expression both showed significant correlations with slaughter and tail traits, and the associations appear to be related with the ontogenetic expression as well as the potential functions of lipin2 and lipin3 in sheep.
Collapse
Affiliation(s)
- Xiao-Li Jiao
- College of Animal Science and Technology, Shanxi Agricultural University, Taigu 030-801, China ; College of Animal Science and Veterinary medicine, Tianjin Agricultural University, Tianjin 300-384, China
| | - Jiong-Jie Jing
- College of Animal Science and Technology, Shanxi Agricultural University, Taigu 030-801, China
| | - Li-Ying Qiao
- College of Animal Science and Technology, Shanxi Agricultural University, Taigu 030-801, China
| | - Jian-Hua Liu
- College of Animal Science and Technology, Shanxi Agricultural University, Taigu 030-801, China
| | - Liu-An Li
- College of Animal Science and Veterinary medicine, Tianjin Agricultural University, Tianjin 300-384, China
| | - Jing Zhang
- College of Animal Science and Technology, Shanxi Agricultural University, Taigu 030-801, China
| | - Xia-Li Jia
- College of Animal Science and Technology, Shanxi Agricultural University, Taigu 030-801, China
| | - Wen-Zhong Liu
- College of Animal Science and Technology, Shanxi Agricultural University, Taigu 030-801, China
| |
Collapse
|
20
|
El Kebbaj R, Andreoletti P, El Hajj HI, El Kharrassi Y, Vamecq J, Mandard S, Saih FE, Latruffe N, El Kebbaj MS, Lizard G, Nasser B, Cherkaoui-Malki M. Argan oil prevents down-regulation induced by endotoxin on liver fatty acid oxidation and gluconeogenesis and on peroxisome proliferator-activated receptor gamma coactivator-1α, (PGC-1α), peroxisome proliferator-activated receptor α (PPARα) and estrogen related receptor α (ERRα). BIOCHIMIE OPEN 2015; 1:51-59. [PMID: 29632829 PMCID: PMC5889474 DOI: 10.1016/j.biopen.2015.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 10/20/2015] [Indexed: 01/04/2023]
Abstract
In patients with sepsis, liver metabolism and its capacity to provide other organs with energetic substrates are impaired. This and many other pathophysiological changes seen in human patients are reproduced in mice injected with purified endotoxin (lipopolysaccharide, LPS). In the present study, down-regulation of genes involved in hepatic fatty acid oxidation (FAOx) and gluconeogenesis in mice exposed to LPS was challenged by nutritional intervention with Argan oil. Mice given a standard chow supplemented or not with either 6% (w/w) Argan oil (AO) or 6% (w/w) olive oil (OO) prior to exposure to LPS were explored for liver gene expressions assessed by mRNA transcript levels and/or enzyme activities. AO (or OO) food supplementation reveals that, in LPS-treated mice, hepatic expression of genes involved in FAOx and gluconeogenesis was preserved. This preventive protection might be related to the recovery of the gene expressions of nuclear receptors peroxisome proliferator-activated receptor α (PPARα) and estrogen related receptor α (ERRα) and their coactivator peroxisome proliferator-activated receptor gamma coactivator-1α, (PGC-1α). These preventive mechanisms conveyed by AO against LPS-induced metabolic dysregulation might add new therapeutic potentialities in the management of human sepsis. Argan oil prevents LPS-treated mice from liver dysregulation of FAOx and gluconeogenesis. Argan oil improves hepatic expression of PPARα and ERRα, and their coactivators PGC-1α and Lipin-1. New preventive mechanisms conveyed by Argan oil against LPS-induced metabolic dysregulation.
Collapse
Key Words
- ACADL, acyl CoA dehydrogenase long-chain
- ACADM, acyl CoA dehydrogenase medium-chain
- ACADS, acyl CoA dehydrogenase short-chain
- ACOX1, acyl-CoA oxidase 1
- AO, Argan oil
- Argan oil
- Beta-oxidation
- Coactivator
- ERRα, estrogen related receptor α
- G6PH, glucose-6-phosphatase
- Gluconeogenesis
- Glut2, glucose transporter 2
- Glut4, glucose transporter 4
- HNF-4α, hepatic nuclear factor-4α
- LPS, lipopolysaccharide
- Nuclear receptor
- OO, olive oil
- PEPCK, phospoenolpyruvate carboxykinase
- PGC-1α, peroxisome proliferator-activated receptor γ coactivator-1α
- PPARα, peroxisome proliferator-activated receptor α
Collapse
Affiliation(s)
- Riad El Kebbaj
- Univ. Bourgogne-Franche Comté, Laboratoire BioPeroxIL (Biochimie du Peroxysome, Inflammation et Métabolisme Lipidique), EA 7270, 21000 Dijon, France.,Laboratoir de Biochimie et Neurosciences, Faculté des Sciences et Techniques, Université Hassan I, BP 577, 26 000 Settat, Morocco.,Laboratoire des Sciences et Technologies de la Santé, Institut supérieur des sciences de la santé Université Hassan I, Route de Casablanca. 14 BP 539, 26 000 Settat, Morocco
| | - Pierre Andreoletti
- Univ. Bourgogne-Franche Comté, Laboratoire BioPeroxIL (Biochimie du Peroxysome, Inflammation et Métabolisme Lipidique), EA 7270, 21000 Dijon, France
| | - Hammam I El Hajj
- Univ. Bourgogne-Franche Comté, Laboratoire BioPeroxIL (Biochimie du Peroxysome, Inflammation et Métabolisme Lipidique), EA 7270, 21000 Dijon, France
| | - Youssef El Kharrassi
- Univ. Bourgogne-Franche Comté, Laboratoire BioPeroxIL (Biochimie du Peroxysome, Inflammation et Métabolisme Lipidique), EA 7270, 21000 Dijon, France.,Laboratoir de Biochimie et Neurosciences, Faculté des Sciences et Techniques, Université Hassan I, BP 577, 26 000 Settat, Morocco
| | - Joseph Vamecq
- INSERM and HMNO, CBP, CHRU Lille, 59037 Lille and RADEME EA 7364, Faculté de Médecine, Université de Lille 2, 59045 Lille, France
| | - Stéphane Mandard
- Lipness Team, INSERM, Research Center UMR866 and LabEx LipSTIC, Université de Bourgogne-Franche Comté, Dijon, France
| | - Fatima-Ezzahra Saih
- Univ. Bourgogne-Franche Comté, Laboratoire BioPeroxIL (Biochimie du Peroxysome, Inflammation et Métabolisme Lipidique), EA 7270, 21000 Dijon, France.,Laboratoir de Biochimie et Neurosciences, Faculté des Sciences et Techniques, Université Hassan I, BP 577, 26 000 Settat, Morocco
| | - Norbert Latruffe
- Univ. Bourgogne-Franche Comté, Laboratoire BioPeroxIL (Biochimie du Peroxysome, Inflammation et Métabolisme Lipidique), EA 7270, 21000 Dijon, France
| | - M'Hammed Saïd El Kebbaj
- Laboratoire de recherche sur les lipoprotéines et l'Athérosclérose, Faculté des Sciences Ben M'sik, Avenue Cdt Driss El Harti, BP 7955, Université Hassan II-Mohammedia-Casablanca, Morocco
| | - Gérard Lizard
- Univ. Bourgogne-Franche Comté, Laboratoire BioPeroxIL (Biochimie du Peroxysome, Inflammation et Métabolisme Lipidique), EA 7270, 21000 Dijon, France
| | - Boubker Nasser
- Laboratoir de Biochimie et Neurosciences, Faculté des Sciences et Techniques, Université Hassan I, BP 577, 26 000 Settat, Morocco
| | - Mustapha Cherkaoui-Malki
- Univ. Bourgogne-Franche Comté, Laboratoire BioPeroxIL (Biochimie du Peroxysome, Inflammation et Métabolisme Lipidique), EA 7270, 21000 Dijon, France
| |
Collapse
|
21
|
Cardozo Gizzi AM, Prucca CG, Gaveglio VL, Renner ML, Pasquaré SJ, Caputto BL. The Catalytic Efficiency of Lipin 1β Increases by Physically Interacting with the Proto-oncoprotein c-Fos. J Biol Chem 2015; 290:29578-92. [PMID: 26475860 DOI: 10.1074/jbc.m115.678821] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Indexed: 01/19/2023] Open
Abstract
Phosphatidic acid (PA) is a central precursor for membrane phospholipid biosynthesis. The lipin family is a magnesium-dependent type I PA phosphatase involved in de novo synthesis of neutral lipids and phospholipids. The regulation of lipin activity may govern the pathways by which these lipids are synthesized and control the cellular levels of important signaling lipids. Moreover, the proto-oncoprotein c-Fos has an emerging role in glycerolipid synthesis regulation; by interacting with key synthesizing enzymes it is able to increase overall phospho- and glycolipid synthesis. We studied the lipin 1β enzyme activity in a cell-free system using PA/Triton X-100 mixed micelles as substrate, analyzing it in the presence/absence of c-Fos. We found that lipin 1β kcat value increases around 40% in the presence of c-Fos, with no change in the lipin 1β affinity for the PA/Triton X-100 mixed micelles. We also probed a physical interaction between both proteins. Although the c-Fos domain involved in lipin activation is its basic domain, the interaction domain is mapped to the N-terminal c-Fos. In conclusion, we provide evidence for a novel positive regulator of lipin 1β PA phosphatase activity that is not achieved via altering its subcellular localization or affinity for membranes but rather through directly increasing its catalytic efficiency.
Collapse
Affiliation(s)
- Andres M Cardozo Gizzi
- From the Centro de Investigaciones en Química Biológica de Córdoba (Consejo Nacional de Investigaciones Científicas y Técnicas), Departamento de Química Biológica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Ciudad Universitaria, X5000HUA Córdoba and
| | - Cesar G Prucca
- From the Centro de Investigaciones en Química Biológica de Córdoba (Consejo Nacional de Investigaciones Científicas y Técnicas), Departamento de Química Biológica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Ciudad Universitaria, X5000HUA Córdoba and
| | - Virginia L Gaveglio
- the Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Edificio El Camino La Carrindanga Km 7, 8000 Bahía Blanca, Argentina
| | - Marianne L Renner
- From the Centro de Investigaciones en Química Biológica de Córdoba (Consejo Nacional de Investigaciones Científicas y Técnicas), Departamento de Química Biológica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Ciudad Universitaria, X5000HUA Córdoba and
| | - Susana J Pasquaré
- the Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Edificio El Camino La Carrindanga Km 7, 8000 Bahía Blanca, Argentina
| | - Beatriz L Caputto
- From the Centro de Investigaciones en Química Biológica de Córdoba (Consejo Nacional de Investigaciones Científicas y Técnicas), Departamento de Química Biológica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Ciudad Universitaria, X5000HUA Córdoba and
| |
Collapse
|
22
|
Seo MJ, Lee YJ, Hwang JH, Kim KJ, Lee BY. The inhibitory effects of quercetin on obesity and obesity-induced inflammation by regulation of MAPK signaling. J Nutr Biochem 2015; 26:1308-16. [PMID: 26277481 DOI: 10.1016/j.jnutbio.2015.06.005] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 04/27/2015] [Accepted: 06/13/2015] [Indexed: 12/20/2022]
Abstract
Quercetin is a flavonoid found in fruits, vegetables, leaves and grains. It has inhibitory, antiviral, antiasthma, anticancer and antiinflammatory effects. Research has suggested that obesity is linked to metabolic disorders. In this study, we examined the inhibitory effect of quercetin on lipid accumulation and obesity-induced inflammation using 3T3-L1, RAW264.7, zebrafish and mouse models. Quercetin suppressed protein levels of the key adipogenic factors C/EBPβ, C/EBPα, PPARγ and FABP4 and the TG-synthesis enzymes lipin1, DGAT1 and LPAATθ. Activation of m-TOR and p70S6K, which are related to insulin and adipogenesis, was down-regulated during adipogenesis in 3T3-L1 cells. Recent research suggested that MAPK signaling factors were involved in adipogenesis and inflammation and that the adipokines MCP-1 and TNF-α attracted macrophages into adipose tissue. Our data showed that quercetin inhibited the MAPK signaling factors ERK1/2, JNK and p38MAPK and MCP-1 and TNF-α in adipocytes and macrophages. Quercetin also inhibited secretion of the inflammatory cytokines IL-1β and IL-6 and stimulated that of IL-10, an antiinflammatory cytokine. In this study, we confirmed the inhibitory effects of quercetin in adipogenesis and inflammation using a mouse model. In mice, quercetin reduced body weight (almost 40%) and suppressed expression of adipogenic, lipogenic and inflammation-related cytokines. Our data demonstrated that quercetin inhibits lipid accumulation and obesity-induced inflammation in the cell and animal models. Our study suggested that quercetin may represent a potential therapeutic agent for other metabolic disorders by regulating obesity and obesity-induced inflammation.
Collapse
Affiliation(s)
- Min-Jung Seo
- Department of Food Science and Biotechnology, CHA University, Seongnam, Kyonggi 463-400, South Korea
| | - Yeon-Joo Lee
- Department of Food Science and Biotechnology, CHA University, Seongnam, Kyonggi 463-400, South Korea
| | - Ji-Hyun Hwang
- Department of Food Science and Biotechnology, CHA University, Seongnam, Kyonggi 463-400, South Korea
| | - Kui-Jin Kim
- Department of Medicine, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Boo-Yong Lee
- Department of Food Science and Biotechnology, CHA University, Seongnam, Kyonggi 463-400, South Korea.
| |
Collapse
|
23
|
Ramasamy I. Recent advances in physiological lipoprotein metabolism. Clin Chem Lab Med 2015; 52:1695-727. [PMID: 23940067 DOI: 10.1515/cclm-2013-0358] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/08/2013] [Indexed: 01/21/2023]
Abstract
Research into lipoprotein metabolism has developed because understanding lipoprotein metabolism has important clinical indications. Lipoproteins are risk factors for cardiovascular disease. Recent advances include the identification of factors in the synthesis and secretion of triglyceride rich lipoproteins, chylomicrons (CM) and very low density lipoproteins (VLDL). These included the identification of microsomal transfer protein, the cotranslational targeting of apoproteinB (apoB) for degradation regulated by the availability of lipids, and the characterization of transport vesicles transporting primordial apoB containing particles to the Golgi. The lipase maturation factor 1, glycosylphosphatidylinositol-anchored high density lipoprotein binding protein 1 and an angiopoietin-like protein play a role in lipoprotein lipase (LPL)-mediated hydrolysis of secreted CMs and VLDL so that the right amount of fatty acid is delivered to the right tissue at the right time. Expression of the low density lipoprotein (LDL) receptor is regulated at both transcriptional and post-transcriptional level. Proprotein convertase subtilisin/kexin type 9 (PCSK9) has a pivotal role in the degradation of LDL receptor. Plasma remnant lipoproteins bind to specific receptors in the liver, the LDL receptor, VLDL receptor and LDL receptor-like proteins prior to removal from the plasma. Reverse cholesterol transport occurs when lipid free apoAI recruits cholesterol and phospholipid to assemble high density lipoprotein (HDL) particles. The discovery of ABC transporters (ABCA1 and ABCG1) and scavenger receptor class B type I (SR-BI) provided further information on the biogenesis of HDL. In humans HDL-cholesterol can be returned to the liver either by direct uptake by SR-BI or through cholesteryl ester transfer protein exchange of cholesteryl ester for triglycerides in apoB lipoproteins, followed by hepatic uptake of apoB containing particles. Cholesterol content in cells is regulated by several transcription factors, including the liver X receptor and sterol regulatory element binding protein. This review summarizes recent advances in knowledge of the molecular mechanisms regulating lipoprotein metabolism.
Collapse
|
24
|
Rojas JM, Bruinstroop E, Printz RL, Alijagic-Boers A, Foppen E, Turney MK, George L, Beck-Sickinger AG, Kalsbeek A, Niswender KD. Central nervous system neuropeptide Y regulates mediators of hepatic phospholipid remodeling and very low-density lipoprotein triglyceride secretion via sympathetic innervation. Mol Metab 2015; 4:210-21. [PMID: 25737956 PMCID: PMC4338317 DOI: 10.1016/j.molmet.2015.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 12/29/2014] [Accepted: 01/09/2015] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE Elevated very low-density lipoprotein (VLDL)-triglyceride (TG) secretion from the liver contributes to an atherogenic dyslipidemia that is associated with obesity, diabetes and the metabolic syndrome. Numerous models of obesity and diabetes are characterized by increased central nervous system (CNS) neuropeptide Y (NPY); in fact, a single intracerebroventricular (icv) administration of NPY in lean fasted rats elevates hepatic VLDL-TG secretion and does so, in large part, via signaling through the CNS NPY Y1 receptor. Thus, our overarching hypothesis is that elevated CNS NPY action contributes to dyslipidemia by activating central circuits that modulate liver lipid metabolism. METHODS Chow-fed Zucker fatty (ZF) rats were pair-fed by matching their caloric intake to that of lean controls and effects on body weight, plasma TG, and liver content of TG and phospholipid (PL) were compared to ad-libitum (ad-lib) fed ZF rats. Additionally, lean 4-h fasted rats with intact or disrupted hepatic sympathetic innervation were treated with icv NPY or NPY Y1 receptor agonist to identify novel hepatic mechanisms by which NPY promotes VLDL particle maturation and secretion. RESULTS Manipulation of plasma TG levels in obese ZF rats, through pair-feeding had no effect on liver TG content; however, hepatic PL content was substantially reduced and was tightly correlated with plasma TG levels. Treatment with icv NPY or a selective NPY Y1 receptor agonist in lean fasted rats robustly activated key hepatic regulatory proteins, stearoyl-CoA desaturase-1 (SCD-1), ADP-ribosylation factor-1 (ARF-1), and lipin-1, known to be involved in remodeling liver PL into TG for VLDL maturation and secretion. Lastly, we show that the effects of CNS NPY on key liporegulatory proteins are attenuated by hepatic sympathetic denervation. CONCLUSIONS These data support a model in which CNS NPY modulates mediators of hepatic PL remodeling and VLDL maturation to stimulate VLDL-TG secretion that is dependent on the Y1 receptor and sympathetic signaling to the liver.
Collapse
Key Words
- AGPAT, 1-acyl-glycerol-3-phosphate acyltransferase
- ARF-1, ADP-ribosylation factor-1
- ApoB, apolipoprotein B
- CNS, central nervous system
- Cyto, cytoplasmic
- DAG, diacylglycerol
- DGAT, diacylglycerol acyltransferase
- ER, endoplasmic reticulum
- FFA(s), free fatty acid(s)
- GAPDH, glyceraldehyde 3-phosphate dehydrogenase
- HDAC-1, histone deacetylase-1
- Lipin-1
- NE, norepinephrine
- NPY Y1 receptor
- NPY, neuropeptide Y
- Nuc, nuclear
- PA, phosphatidic acid
- PAP-1, phosphatidic acid phosphatase-1
- PF, pair-fed
- PL, phospholipid
- PLD, phospholipase D
- POMC, proopiomelanocortin
- Phospholipid
- RPL13A, ribosomal protein L13a
- RT-PCR, real-time PCR
- SCD-1, stearoyl-CoA desaturase-1
- SNS, sympathetic nervous system
- Sham, sham-denervation
- Sx, sympathetic denervation
- Sympathetic denervation
- TG, triglyceride
- Triglyceride
- VLDL
- VLDL, very low-density lipoprotein
- Veh, vehicle
- ZF, Zucker fatty
- ad-lib, ad-libitum
- icv, intracerebroventricular
Collapse
Affiliation(s)
- Jennifer M. Rojas
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Eveline Bruinstroop
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Richard L. Printz
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Aldijana Alijagic-Boers
- Department of Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Science, Amsterdam, The Netherlands
| | - Ewout Foppen
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Maxine K. Turney
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Leena George
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Annette G. Beck-Sickinger
- Institute of Biochemistry, Faculty of Bioscience, Pharmacy and Psychology, Leipzig University, Leipzig, Germany
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Science, Amsterdam, The Netherlands
| | - Kevin D. Niswender
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| |
Collapse
|
25
|
Bi L, Jiang Z, Zhou J. The role of lipin-1 in the pathogenesis of alcoholic fatty liver. Alcohol Alcohol 2015; 50:146-51. [PMID: 25595739 DOI: 10.1093/alcalc/agu102] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
AIMS The aim of this review was to focus on the knowledge of the role of lipin-1 in the pathogenesis of alcoholic fatty liver. METHODS Systematic review of animal clinical and cell level studies related to the function of lipin-1 on alcoholic fatty liver, alcoholic hepatitis and alcoholic liver cirrhosis disease. RESULT Ethanol could increase the expression of lipin-1 through the AMPK-SREBP-1 signaling and dramatically increase the ratio of Lpin1β to Lpin1α by SIRT1-SFRS10-Lpin1β/α axis in the liver. Moreover, research has shown that over-expression of lipin-1 could also remarkably suppress very low density lipoprotein-triacylglyceride secretion. Last, lipin-1 has potent anti-inflammatory property. CONCLUSION In conclusion, lipin-1 has dual functions in lipid metabolism. In the cytoplasm, lipin-1β functions as a Mg(2+)-dependent phosphatidic acid phosphohydrolase (PAP) enzyme in triglyceride synthesis pathways. In the nucleus, lipin-1α acts as a transcriptional co-regulator to regulate the capacity of the liver for fatty acid oxidation and activity of the lipogenic enzyme. In hepatocytes of alcoholic fatty liver disease (AFLD), ethanol increases the expression of lipin-1 through the AMPK-SREBP-1 signaling and the Lpin1β/α ratio by SIRT1-SFRS10- Lpin1β/α axis. Of course, in addition to that, ethanol could also produce the PAP activity and interrupt the nucleus function of lipin-1. Furthermore, over-expression of lipin-1 could remarkably suppress very low-density lipoprotein-triacylglyceride (VLDL-TAG) secretion. In the end, endogenous lipin-1 has potent anti-inflammatory property. Increased synthesis of TAG, decreased fatty acid oxidation, impaired VLDL-TAG secretion and activated inflammatory factors act together to exacerbate the development of AFLD.
Collapse
Affiliation(s)
- Lijuan Bi
- Department of Infectious Disease, Third Hospital, Hebei Medical University, 139 ZiQiang Road, Shijiazhuang 050051, China
| | - Zhian Jiang
- Department of Infectious Disease, Third Hospital, Hebei Medical University, 139 ZiQiang Road, Shijiazhuang 050051, China
| | - Junying Zhou
- Department of Infectious Disease, Third Hospital, Hebei Medical University, 139 ZiQiang Road, Shijiazhuang 050051, China
| |
Collapse
|
26
|
Chen Y, Rui BB, Tang LY, Hu CM. Lipin Family Proteins - Key Regulators in Lipid Metabolism. ANNALS OF NUTRITION AND METABOLISM 2014; 66:10-8. [DOI: 10.1159/000368661] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 09/19/2014] [Indexed: 11/19/2022]
Abstract
Background: Proteins in the lipin family play a key role in lipid synthesis due to their phosphatidate phosphatase activity, and they also act as transcriptional coactivators to regulate the expression of genes involved in lipid metabolism. The lipin family includes three members, lipin1, lipin2, and lipin3, which exhibit tissue-specific expression, indicating that they may have distinct roles in mediating disease. To date, most studies have focused on lipin1, whereas the roles of lipin2 and lipin3 are less understood. Summary: This review introduces the structural characteristics, physiological functions, relationship to lipid metabolism, and patterns of expression of the lipin family proteins, highlighting their roles in lipid metabolic homeostasis. © 2014 S. Karger AG, Basel
Collapse
|
27
|
Troxerutin improves hepatic lipid homeostasis by restoring NAD(+)-depletion-mediated dysfunction of lipin 1 signaling in high-fat diet-treated mice. Biochem Pharmacol 2014; 91:74-86. [PMID: 25026599 DOI: 10.1016/j.bcp.2014.07.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 06/22/2014] [Accepted: 07/01/2014] [Indexed: 12/11/2022]
Abstract
Recent evidences suggest that NAD(+) depletion leads to abnormal hepatic lipid metabolism in high-fat diet (HFD)-induced nonalcoholic fatty liver disease (NAFLD); however, the contributing mechanism is not well understood. Our previous study showed that troxerutin, a trihydroxyethylated derivative of natural bioflavonoid rutin, effectively inhibited obesity, and normalized hyperglycemia and hyperlipidemia in high-cholesterol diet-induced diabetic mice. Here we investigated whether troxerutin improved hepatic lipid metabolism via preventing NAD(+) depletion in HFD-induced NAFLD mouse model and the mechanisms underlying these effects. Our results showed that troxerutin markedly prevented obesity, liver steatosis and injury in HFD-fed mice. Troxerutin largely suppressed oxidative stress-mediated NAD(+)-depletion by increasing nicotinamide phosphoribosyltransferase (NAMPT) protein expression and decreasing poly (ADP-ribose) polymerase-1 (PARP1) protein expression and activity in HFD-treated mouse livers. Consequently, troxerutin remarkably restored Silent mating type information regulation 2 homolog1 (SirT1) protein expression and activity in HFD-treated mouse livers. Therefore, troxerutin promoted SirT1-mediated AMP-activated protein kinase (AMPK) activation to inhibit mammalian target of rapamycin complex 1 (mTORC1) signaling, which enhanced nuclear lipin 1 localization, lowered cytoplasmic lipin 1 localization and the ratio of hepatic Lpin 1β/α. Ultimately, troxerutin improved lipid homeostasis by enhancing fatty acid oxidation and triglyceride secretion, and suppressing lipogenesis in HFD-fed mouse livers. In conclusion, troxerutin displayed beneficial effects on hepatic lipid homeostasis in HFD-induced NAFLD by blocking oxidative stress to restore NAD(+)-depletion-mediated dysfunction of lipin 1 signaling. This study provides novel mechanistic insights into NAFLD pathogenesis and indicates that troxerutin is a candidate for pharmacological intervention of NAFLD via restoring NAD(+) levels.
Collapse
|
28
|
Yin H, Hu M, Liang X, Ajmo JM, Li X, Bataller R, Odena G, Stevens SM, You M. Deletion of SIRT1 from hepatocytes in mice disrupts lipin-1 signaling and aggravates alcoholic fatty liver. Gastroenterology 2014; 146:801-11. [PMID: 24262277 PMCID: PMC3943758 DOI: 10.1053/j.gastro.2013.11.008] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 11/08/2013] [Accepted: 11/12/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Sirtuin (SIRT1) is a nicotinamide adenine dinucleotide-dependent protein deacetylase that regulates hepatic lipid metabolism by modifying histones and transcription factors. Ethanol exposure disrupts SIRT1 activity and contributes to alcoholic liver disease in rodents, but the exact pathogenic mechanism is not clear. We compared mice with liver-specific deletion of Sirt1 (Sirt1LKO) mice with their LOX littermates (controls). METHODS We induced alcoholic liver injury in male Sirt1LKO and control mice, placing them on Lieber-DeCarli ethanol-containing diets for 10 days and then administering a single dose of ethanol (5 g/kg body weight) via gavage. Liver and serum samples were collected. We also measured messenger RNA levels of SIRT1, SFRS10, and lipin-1β and lipin-1α in liver samples from patients with alcoholic hepatitis and individuals without alcoholic hepatitis (controls). RESULTS On the ethanol-containing diet, livers of Sirt1LKO mice accumulated larger amounts of hepatic lipid and expressed higher levels of inflammatory cytokines than control mice; serum of Sirt1LKO mice had increased levels of alanine aminotransferase and aspartate aminotransferase. Hepatic deletion of SIRT1 exacerbated ethanol-mediated defects in lipid metabolism, mainly by altering the function of lipin-1, a transcriptional regulator of lipid metabolism. In cultured mouse AML-12 hepatocytes, transgenic expression of SIRT1 prevented fat accumulation in response to ethanol exposure, largely by reversing the aberrations in lipin-1 signaling induced by ethanol. Liver samples from patients with alcoholic hepatitis had reduced levels of SIRT1 and a higher ratio of Lpin1β/α messenger RNAs than controls. CONCLUSIONS In mice, hepatic deletion of Sirt1 promotes steatosis, inflammation, and fibrosis in response to ethanol challenge. Ethanol-mediated impairment of hepatic SIRT1 signaling via lipin-1 contributes to development of alcoholic steatosis and inflammation. Reagents designed to increase SIRT1 regulation of lipin-1 can be developed to treat patients with alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Huquan Yin
- Department of Molecular Pharmacology and Physiology, University of South Florida Health Sciences Center, 12901 Bruce B. Downs Blvd. Tampa, FL 33612 , USA
| | - Ming Hu
- Department of Molecular Pharmacology and Physiology, University of South Florida Health Sciences Center, 12901 Bruce B. Downs Blvd. Tampa, FL 33612 , USA
| | - Xiaomei Liang
- Department of Molecular Pharmacology and Physiology, University of South Florida Health Sciences Center, 12901 Bruce B. Downs Blvd. Tampa, FL 33612 , USA
| | - Joanne M. Ajmo
- Department of Molecular Pharmacology and Physiology, University of South Florida Health Sciences Center, 12901 Bruce B. Downs Blvd. Tampa, FL 33612 , USA
| | - Xiaoling Li
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Ramon Bataller
- Departments of Medicine and Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Gemma Odena
- Departments of Medicine and Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Stanley M. Stevens
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 E. Flowler Ave. Tampa FL, 33620, USA
| | - Min You
- Department of Molecular Pharmacology and Physiology, University of South Florida Health Sciences Center, Tampa, Florida.
| |
Collapse
|
29
|
Hu M, Yin H, Mitra MS, Liang X, Ajmo JM, Nadra K, Chrast R, Finck BN, You M. Hepatic-specific lipin-1 deficiency exacerbates experimental alcohol-induced steatohepatitis in mice. Hepatology 2013; 58:1953-63. [PMID: 23787969 PMCID: PMC3835749 DOI: 10.1002/hep.26589] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 06/12/2013] [Indexed: 12/22/2022]
Abstract
UNLABELLED Lipin-1 regulates lipid metabolism by way of its function as an enzyme in the triglyceride synthesis pathway and as a transcriptional coregulatory protein and is highly up-regulated in alcoholic fatty liver disease. In the present study, using a liver-specific lipin-1-deficient (lipin-1LKO) mouse model, we aimed to investigate the functional role of lipin-1 in the development of alcoholic steatohepatitis and explore the underlying mechanisms. Alcoholic liver injury was achieved by pair feeding wild-type and lipin-1LKO mice with modified Lieber-DeCarli ethanol-containing low-fat diets for 4 weeks. Surprisingly, chronically ethanol-fed lipin-1LKO mice showed markedly greater hepatic triglyceride and cholesterol accumulation, and augmented elevation of serum liver enzymes accompanied by increased hepatic proinflammatory cytokine expression. Our studies further revealed that hepatic removal of lipin-1 in mice augmented ethanol-induced impairment of hepatic fatty acid oxidation and lipoprotein production, likely by way of deactivation of peroxisome proliferator-activated receptor γ coactivator-1 alpha, a prominent transcriptional regulator of lipid metabolism. CONCLUSIONS Liver-specific lipin-1 deficiency in mice exacerbates the development and progression of experimental alcohol-induced steatohepatitis. Pharmacological or nutritional modulation of hepatic lipin-1 may be beneficial for the prevention or treatment of human alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Ming Hu
- Department of Molecular Pharmacology and Physiology, University of South Florida Health Sciences Center, 12901 Bruce B. Downs Blvd. Tampa, FL 33612, USA
| | - Huquan Yin
- Department of Molecular Pharmacology and Physiology, University of South Florida Health Sciences Center, 12901 Bruce B. Downs Blvd. Tampa, FL 33612, USA
| | - Mayurranjan S. Mitra
- Department of Medicine, Washington University School of Medicine, St.Louis, MO 63110, USA
| | - Xiaomei Liang
- Department of Molecular Pharmacology and Physiology, University of South Florida Health Sciences Center, 12901 Bruce B. Downs Blvd. Tampa, FL 33612, USA
| | - Joanne M. Ajmo
- Department of Molecular Pharmacology and Physiology, University of South Florida Health Sciences Center, 12901 Bruce B. Downs Blvd. Tampa, FL 33612, USA
| | - Karim Nadra
- Department of Medical Genetics, University of Lausanne, 1005 Lausanne, Switzerland
| | - Roman Chrast
- Department of Medical Genetics, University of Lausanne, 1005 Lausanne, Switzerland
| | - Brian N. Finck
- Department of Medicine, Washington University School of Medicine, St.Louis, MO 63110, USA
| | - Min You
- Department of Molecular Pharmacology and Physiology, University of South Florida Health Sciences Center, 12901 Bruce B. Downs Blvd. Tampa, FL 33612, USA
| |
Collapse
|
30
|
Sembongi H, Miranda M, Han GS, Fakas S, Grimsey N, Vendrell J, Carman GM, Siniossoglou S. Distinct roles of the phosphatidate phosphatases lipin 1 and 2 during adipogenesis and lipid droplet biogenesis in 3T3-L1 cells. J Biol Chem 2013; 288:34502-13. [PMID: 24133206 PMCID: PMC3843065 DOI: 10.1074/jbc.m113.488445] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 10/04/2013] [Indexed: 11/06/2022] Open
Abstract
Lipins are evolutionarily conserved Mg(2+)-dependent phosphatidate phosphatase (PAP) enzymes with essential roles in lipid biosynthesis. Mammals express three paralogues: lipins 1, 2, and 3. Loss of lipin 1 in mice inhibits adipogenesis at an early stage of differentiation and results in a lipodystrophic phenotype. The role of lipins at later stages of adipogenesis, when cells initiate the formation of lipid droplets, is less well characterized. We found that depletion of lipin 1, after the initiation of differentiation in 3T3-L1 cells but before the loading of lipid droplets with triacylglycerol, results in a reciprocal increase of lipin 2, but not lipin 3. We generated 3T3-L1 cells where total lipin protein and PAP activity levels are down-regulated by the combined depletion of lipins 1 and 2 at day 4 of differentiation. These cells still accumulated triacylglycerol but displayed a striking fragmentation of lipid droplets without significantly affecting their total volume per cell. This was due to the lack of the PAP activity of lipin 1 in adipocytes after day 4 of differentiation, whereas depletion of lipin 2 led to an increase of lipid droplet volume per cell. We propose that in addition to their roles during early adipogenesis, lipins also have a role in lipid droplet biogenesis.
Collapse
Affiliation(s)
- Hiroshi Sembongi
- From the Cambridge Institute for Medical Research, University of Cambridge, CB2 0XY Cambridge, United Kingdom
| | - Merce Miranda
- the Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Institut d'Investigació Pere Virgili, Universitat Rovira i Virgili, Hospital Universitari Joan XXIII, Tarragona, Spain, and
| | - Gil-Soo Han
- the Department of Food Science, Rutgers Center for Lipid Research, and New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, New Jersey 08901
| | - Stylianos Fakas
- the Department of Food Science, Rutgers Center for Lipid Research, and New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, New Jersey 08901
| | - Neil Grimsey
- From the Cambridge Institute for Medical Research, University of Cambridge, CB2 0XY Cambridge, United Kingdom
| | - Joan Vendrell
- the Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Institut d'Investigació Pere Virgili, Universitat Rovira i Virgili, Hospital Universitari Joan XXIII, Tarragona, Spain, and
| | - George M. Carman
- the Department of Food Science, Rutgers Center for Lipid Research, and New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, New Jersey 08901
| | - Symeon Siniossoglou
- From the Cambridge Institute for Medical Research, University of Cambridge, CB2 0XY Cambridge, United Kingdom
| |
Collapse
|
31
|
Vasiljević A, Veličković N, Bursać B, Djordjevic A, Milutinović DV, Nestorović N, Matić G. Enhanced prereceptor glucocorticoid metabolism and lipogenesis impair insulin signaling in the liver of fructose-fed rats. J Nutr Biochem 2013; 24:1790-7. [DOI: 10.1016/j.jnutbio.2013.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 02/18/2013] [Accepted: 04/01/2013] [Indexed: 12/19/2022]
|
32
|
Krahmer N, Farese RV, Walther TC. Balancing the fat: lipid droplets and human disease. EMBO Mol Med 2013; 5:973-83. [PMID: 23740690 PMCID: PMC3721468 DOI: 10.1002/emmm.201100671] [Citation(s) in RCA: 348] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 04/30/2013] [Accepted: 05/02/2013] [Indexed: 01/04/2023] Open
Abstract
Lipid droplets (LDs) are dynamic, cytosolic lipid-storage organelles found in nearly all cell types. Too many or too few LDs during excess or deficient fat storage lead to many different human diseases. Recent insights into LD biology and LD protein functions shed new light on mechanisms underlying those metabolic pathologies. These findings will likely provide opportunities for treatment of diseases associated with too much or too little fat.
Collapse
Affiliation(s)
- Natalie Krahmer
- Department of Cell Biology, Yale School of MedicineNew Haven, CT, USA
| | - Robert V Farese
- Gladstone Institutes, Departments of Medicine and Biochemistry & Biophysics, University of CaliforniaSan Francisco, CA, USA
| | - Tobias C Walther
- Department of Cell Biology, Yale School of MedicineNew Haven, CT, USA
| |
Collapse
|
33
|
Senmaru T, Fukui M, Okada H, Mineoka Y, Yamazaki M, Tsujikawa M, Hasegawa G, Kitawaki J, Obayashi H, Nakamura N. Testosterone deficiency induces markedly decreased serum triglycerides, increased small dense LDL, and hepatic steatosis mediated by dysregulation of lipid assembly and secretion in mice fed a high-fat diet. Metabolism 2013; 62:851-60. [PMID: 23332447 DOI: 10.1016/j.metabol.2012.12.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 12/03/2012] [Accepted: 12/10/2012] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Although low serum testosterone (T) is associated with metabolic disorders, the mechanism of this association is unclear. The objective of the present study was to investigate the combined effects of T deficiency and a high-fat diet (HFD) on hepatic lipid homeostasis in mice. MATERIALS/METHODS Orchiectomized (ORX) mice and sham-operated (SHAM) mice were randomly divided into five groups: SHAM mice fed a standard diet (SD), SHAM mice fed HFD, ORX mice fed SD, ORX mice fed HFD, and ORX mice fed HFD with T supplementation. After 4weeks of treatment, we investigated the synthesis and secretion of lipids in the liver and detailed serum lipoprotein profiles in each group. RESULTS ORX mice fed HFD showed increased hepatic steatosis, markedly decreased serum triglyceride (TG) and TG-VLDL content, and increased serum very small-LDL content. Gene expression analysis revealed that ORX mice fed HFD showed significantly decreased expression of microsomal triglyceride transfer protein, lipin-1, peroxisome proliferator-activated receptor (PPAR)-α and PPAR-γ coactivator 1-α, and significantly increased sterol regulatory element-binding protein-1, diacylglycerol acyltransferase-2 and fatty acid synthase. Reduction of hepatic AMPK phosphorylation was observed in ORX mice fed HFD. These perturbations in ORX mice fed HFD were normalized to the levels of SHAM mice fed HFD by T supplementation. CONCLUSION T deficiency is associated with failure of lipid homeostasis mediated by altered expression of genes involved in hepatic assembly and secretion of lipids.
Collapse
Affiliation(s)
- Takafumi Senmaru
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
He J, Gao J, Xu M, Ren S, Stefanovic-Racic M, O'Doherty RM, Xie W. PXR ablation alleviates diet-induced and genetic obesity and insulin resistance in mice. Diabetes 2013; 62:1876-87. [PMID: 23349477 PMCID: PMC3661619 DOI: 10.2337/db12-1039] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The pregnane X receptor (PXR), along with its sister receptor constitutive androstane receptor (CAR), was initially characterized as a xenobiotic receptor that regulates drug metabolism. In this study, we have uncovered an unexpected endobiotic role of PXR in obesity and type 2 diabetes. PXR ablation inhibited high-fat diet (HFD)-induced obesity, hepatic steatosis, and insulin resistance, which were accounted for by increased oxygen consumption, increased mitochondrial β-oxidation, inhibition of hepatic lipogenesis and inflammation, and sensitization of insulin signaling. In an independent model, introducing the PXR(-/-) allele into the ob/ob background also improved body composition and relieved the diabetic phenotype. The ob/ob mice deficient of PXR showed increased oxygen consumption and energy expenditure, as well as inhibition of gluconeogenesis and increased rate of glucose disposal during euglycemic clamp. Mechanistically, the metabolic benefits of PXR ablation were associated with the inhibition of c-Jun NH2-terminal kinase activation and downregulation of lipin-1, a novel PXR target gene. The metabolic benefit of PXR ablation was opposite to the reported prodiabetic effect of CAR ablation. Our results may help to establish PXR as a novel therapeutic target, and PXR antagonists may be used for the prevention and treatment of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Jinhan He
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jie Gao
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Meishu Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Songrong Ren
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Maja Stefanovic-Racic
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert Martin O'Doherty
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Corresponding author: Wen Xie,
| |
Collapse
|
35
|
Cardozo Gizzi AM, Caputto BL. Mechanistic insights into the nongenomic regulation of phospholipid synthesizing enzymes. IUBMB Life 2013; 65:584-92. [PMID: 23712998 DOI: 10.1002/iub.1173] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 03/21/2013] [Indexed: 11/06/2022]
Abstract
Lipid synthesis is a complex process regulated at multiple levels. Here, we will discuss nongenomic regulatory mechanisms, particularly the activation and/or recruitment of key enzymes to membranes. The phospholipid synthesis enzymes Lipin and CTP:phosphocholine cytidylyltransferase are taken as examples of these mechanisms that are mediated by posttranslational modifications or by an intrinsic property of the enzyme that senses lipid composition. In addition, special emphasis will be put on another relevant non genomic lipid synthesis regulation mechanism that is dependent on c-Fos, a protein that has deserved less attention so far. This latter regulatory mechanism is emerging as an important determinant for processes that require high rates of lipid synthesis such as those of growth and proliferation.
Collapse
Affiliation(s)
- Andrés M Cardozo Gizzi
- Centro de Investigaciones en Química Biológica de Córdoba, CIQUIBIC (UNC-CONICET), Departamento de Química Biológica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Ciudad Universitaria, Córdoba, Argentina
| | | |
Collapse
|
36
|
Lagace TA, Ridgway ND. The role of phospholipids in the biological activity and structure of the endoplasmic reticulum. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:2499-510. [PMID: 23711956 DOI: 10.1016/j.bbamcr.2013.05.018] [Citation(s) in RCA: 170] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/09/2013] [Accepted: 05/15/2013] [Indexed: 01/22/2023]
Abstract
The endoplasmic reticulum (ER) is an interconnected network of tubular and planar membranes that supports the synthesis and export of proteins, carbohydrates and lipids. Phospholipids, in particular phosphatidylcholine (PC), are synthesized in the ER where they have essential functions including provision of membranes required for protein synthesis and export, cholesterol homeostasis, and triacylglycerol storage and secretion. Coordination of these biological processes is essential, as highlighted by findings that link phospholipid metabolism in the ER with perturbations in lipid storage/secretion and stress responses, ultimately contributing to obesity/diabetes, atherosclerosis and neurological disorders. Phospholipid synthesis is not uniformly distributed in the ER but is localized at membrane interfaces or contact zones with other organelles, and in dynamic, proliferating ER membranes. The topology of phospholipid synthesis is an important consideration when establishing the etiology of diseases that arise from ER dysfunction. This review will highlight our current understanding of the contribution of phospholipid synthesis to proper ER function, and how alterations contribute to aberrant stress responses and disease. This article is part of a Special Issue entitled: Functional and structural diversity of endoplasmic reticulum.
Collapse
Affiliation(s)
- Thomas A Lagace
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada.
| | | |
Collapse
|
37
|
Lipins, lipinopathies, and the modulation of cellular lipid storage and signaling. Prog Lipid Res 2013; 52:305-16. [PMID: 23603613 DOI: 10.1016/j.plipres.2013.04.001] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 03/29/2013] [Accepted: 04/04/2013] [Indexed: 01/13/2023]
Abstract
Members of the lipin protein family are phosphatidate phosphatase (PAP) enzymes, which catalyze the dephosphorylation of phosphatidic acid to diacylglycerol, the penultimate step in TAG synthesis. Lipins are unique among the glycerolipid biosynthetic enzymes in that they also promote fatty acid oxidation through their activity as co-regulators of gene expression by DNA-bound transcription factors. Lipin function has been evolutionarily conserved from a single ortholog in yeast to the mammalian family of three lipin proteins-lipin-1, lipin-2, and lipin-3. In mice and humans, the levels of lipin activity are a determinant of TAG storage in diverse cell types, and humans with deficiency in lipin-1 or lipin-2 have severe metabolic diseases. Recent work has highlighted the complex physiological interactions between members of the lipin protein family, which exhibit both overlapping and unique functions in specific tissues. The analysis of "lipinopathies" in mouse models and in humans has revealed an important role for lipin activity in the regulation of lipid intermediates (phosphatidate and diacylglycerol), which influence fundamental cellular processes including adipocyte and nerve cell differentiation, adipocyte lipolysis, and hepatic insulin signaling. The elucidation of lipin molecular and physiological functions could lead to novel approaches to modulate cellular lipid storage and metabolic disease.
Collapse
|
38
|
Kok BPC, Dyck JRB, Harris TE, Brindley DN. Differential regulation of the expressions of the PGC-1α splice variants, lipins, and PPARα in heart compared to liver. J Lipid Res 2013; 54:1662-1677. [PMID: 23505321 DOI: 10.1194/jlr.m036624] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Peroxisome proliferator-activated receptor α (PPARα) and PPARγ coactivator 1α (PGC-1α) are crucial transcriptional regulators for genes involved in FA oxidation. Lipin-1 is essential for this increased capacity for β-oxidation in fasted livers, and it is also a phosphatidate phosphatase involved in triacylglycerol and phospholipid synthesis. Little is known about the regulation of these proteins in the heart during fasting, where there is increased FA esterification and oxidation. Lipin-1, lipin-2, lipin-3, carnitine palmitoyltransferase-1b (Cpt1b), and PGC-1α-b mRNA were increased by glucocorticoids and cAMP in neonatal rat cardiomyocytes. However, Cpt1b upregulation was caused by increased PPARα activation rather than expression. By contrast, the effects of PPARα in fasted livers are mediated through increased expression. During fasting, the expressions of PGC-1α-b and PGC-1α-c are increased in mouse hearts, and this is explained by increased cAMP-dependent signaling. By contrast, PGC-1α-a expression is increased in liver. Contrary to our expectations, lipin-1 expression was decreased and lipin-2 remained unchanged in hearts compared with increases in fasted livers. Our results identify novel differences in the regulation of lipins, PPARα, and PGC-1α splice variants during fasting in heart versus liver, even though the ultimate outcome in both tissues is to increase FA turnover and oxidation.
Collapse
Affiliation(s)
- Bernard P C Kok
- Signal Transduction Research Group, University of Alberta, Edmonton, Alberta, Canada
| | - Jason R B Dyck
- Department of Biochemistry, and Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada; Department of Pediatrics, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Thurl E Harris
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - David N Brindley
- Signal Transduction Research Group, University of Alberta, Edmonton, Alberta, Canada; Department of Pediatrics, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
39
|
Siniossoglou S. Phospholipid metabolism and nuclear function: Roles of the lipin family of phosphatidic acid phosphatases. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:575-81. [DOI: 10.1016/j.bbalip.2012.09.014] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 09/19/2012] [Accepted: 09/24/2012] [Indexed: 01/22/2023]
|
40
|
[PPARβ/δ Activation prevents hypertriglyceridemia caused by a high fat diet. Involvement of AMPK and PGC-1α-Lipin1-PPARα pathway]. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2013; 25:63-73. [PMID: 23849213 DOI: 10.1016/j.arteri.2013.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 01/10/2013] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Excessive consume of hypercaloric and high in saturated fat food causes an atherogenic dyslipidemia. In this study we analyzed the effects of PPARβ/δ activator GW501516 on the hypertriglyceridemia induced by a high-fat diet. METHODS Male mice were randomized in three groups: control (standard chow), high fat diet (HFD, 35% fat by weight, 58% Kcal from fat) and high fat diet plus GW501516 (3mg/Kg/day). Treatment duration was three weeks. RESULTS HFD-induced hypertriglyceridemia was accompanied by a reduction in hepatic levels of phospho-AMPK and in PGC-1α and Lipin1 mRNA levels. All these effects were reversed by GW501516 treatment. The lack of changes in phospho-AMPK levels after GW501516 treatment in HFD-fed animals could be the result of an increase in the AMP/ATP ratio. GW501516 treatment also increased Lipin1 protein levels in the nucleus, led to the amplification of the PGC-1α-PPARα pathway and increased PPARα DNA-binding activity, as well as the expression of PPARα-target genes involved in fatty acid oxidation. GW501516 also increased β-hydroxibutirate plasmatic levels, a hepatic β-oxidation end product. Finally, GW501516 increased the hepatic levels of the PPARα endogenous ligand 16:0/18:1-PC and the expression of the VLDL receptor. CONCLUSION These data indicate that the hypotriglyceridemic effect of GW501516 in mice subjected to HFD-fed mice is accompanied by an increase in phospho-AMPK levels and the amplification of the PGC-1α-Lipin1-PPARα pathway.
Collapse
|
41
|
Li S, Chen W, Kang X, Han R, Sun G, Huang Y. Distinct tissue expression profiles of chicken Lpin1-α/β isoforms and the effect of the variation on muscle fiber traits. Gene 2013; 515:281-90. [PMID: 23266642 DOI: 10.1016/j.gene.2012.11.075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 10/14/2012] [Accepted: 11/27/2012] [Indexed: 11/17/2022]
Affiliation(s)
- Suya Li
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, Henan, China
| | | | | | | | | | | |
Collapse
|
42
|
Tang SQ, Jiang QY, Yang CF, Zou XT, Dong XY. [Research and development of Lipin family.]. YI CHUAN = HEREDITAS 2012; 32:981-93. [PMID: 20943485 DOI: 10.3724/sp.j.1005.2010.00981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Lipin family including at least three members Lipin 1, Lipin 2, and Lipin 3 is a critical regulatory enzyme identified recently, which plays dual roles in lipid metabolisms. Lipin family has physiological effects not only on regulating lipid metabolism, but also on maintaining normal peripheral nervous functions, liver lipoprotein secretion, cell morphous, reproductive functions, and energy homeostasis. Since mutations in Lipin gene express may be associated with AIDS, insulin resistance, obesity, diabetes mellitus, and the other diseases of metabolic syndrome, Lipin may be a new useful target in treatment of above-mentioned clinical-related diseases. In this article, we focused on discovery, construction features, expression, regulatory mechanism, and biological functions of Lipin, as well as its correlation research with clinical-related diseases.
Collapse
|
43
|
Michot C, Hubert L, Romero NB, Gouda A, Mamoune A, Mathew S, Kirk E, Viollet L, Rahman S, Bekri S, Peters H, McGill J, Glamuzina E, Farrar M, von der Hagen M, Alexander IE, Kirmse B, Barth M, Laforet P, Benlian P, Munnich A, JeanPierre M, Elpeleg O, Pines O, Delahodde A, de Keyzer Y, de Lonlay P. Study of LPIN1, LPIN2 and LPIN3 in rhabdomyolysis and exercise-induced myalgia. J Inherit Metab Dis 2012; 35:1119-28. [PMID: 22481384 DOI: 10.1007/s10545-012-9461-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 01/19/2012] [Accepted: 01/30/2012] [Indexed: 01/21/2023]
Abstract
BACKGROUND Recessive LPIN1 mutations were identified as a cause of severe rhabdomyolysis in pediatric patients. The human lipin family includes two other closely related members, lipin-2 and 3, which share strong homology and similar activity. The study aimed to determine the involvement of the LPIN family genes in a cohort of pediatric and adult patients (n = 171) presenting with muscular symptoms, ranging from severe (CK >10 000 UI/L) or moderate (CK <10 000 UI/L) rhabdomyolysis (n = 141) to exercise-induced myalgia (n = 30), and to report the clinical findings in patients harboring mutations. METHODS Coding regions of LPIN1, LPIN2 and LPIN3 genes were sequenced using genomic or complementary DNAs. RESULTS Eighteen patients harbored two LPIN1 mutations, including a frequent intragenic deletion. All presented with severe episodes of rhabdomyolysis, starting before age 6 years except two (8 and 42 years). Few patients also suffered from permanent muscle symptoms, including the eldest ones (≥ 40 years). Around 3/4 of muscle biopsies showed accumulation of lipid droplets. At least 40% of heterozygous relatives presented muscular myalgia. Nine heterozygous SNPs in LPIN family genes were identified in milder phenotypes (mild rhabdomyolysis or myalgia). These variants were non-functional in yeast complementation assay based on respiratory activity, except the LPIN3-P24L variant. CONCLUSION LPIN1-related myolysis constitutes a major cause of early-onset rhabdomyolysis and occasionally in adults. Heterozygous LPIN1 mutations may cause mild muscular symptoms. No major defects of LPIN2 or LPIN3 genes were associated with muscular manifestations.
Collapse
Affiliation(s)
- Caroline Michot
- Paris Descartes University, INSERM U781 and Reference Center of Metabolic Diseases, Necker Hospital, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mouse lipin-1 and lipin-2 cooperate to maintain glycerolipid homeostasis in liver and aging cerebellum. Proc Natl Acad Sci U S A 2012; 109:E2486-95. [PMID: 22908270 DOI: 10.1073/pnas.1205221109] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The three lipin phosphatidate phosphatase (PAP) enzymes catalyze a step in glycerolipid biosynthesis, the conversion of phosphatidate to diacylglycerol. Lipin-1 is critical for lipid synthesis and homeostasis in adipose tissue, liver, muscle, and peripheral nerves. Little is known about the physiological role of lipin-2, the predominant lipin protein present in liver and the deficient gene product in the rare disorder Majeed syndrome. By using lipin-2-deficient mice, we uncovered a functional relationship between lipin-1 and lipin-2 that operates in a tissue-specific and age-dependent manner. In liver, lipin-2 deficiency led to a compensatory increase in hepatic lipin-1 protein and elevated PAP activity, which maintained lipid homeostasis under basal conditions, but led to diet-induced hepatic triglyceride accumulation. As lipin-2-deficient mice aged, they developed ataxia and impaired balance. This was associated with the combination of lipin-2 deficiency and an age-dependent reduction in cerebellar lipin-1 levels, resulting in altered cerebellar phospholipid composition. Similar to patients with Majeed syndrome, lipin-2-deficient mice developed anemia, but did not show evidence of osteomyelitis, suggesting that additional environmental or genetic components contribute to the bone abnormalities observed in patients. Combined lipin-1 and lipin-2 deficiency caused embryonic lethality. Our results reveal functional interactions between members of the lipin family in vivo, and a unique role for lipin-2 in central nervous system biology that may be particularly important with advancing age. Additionally, as has been observed in mice and humans with lipin-1 deficiency, the pathophysiology in lipin-2 deficiency is associated with dysregulation of lipid intermediates.
Collapse
|
45
|
|
46
|
Sparks JD, Sparks CE, Adeli K. Selective hepatic insulin resistance, VLDL overproduction, and hypertriglyceridemia. Arterioscler Thromb Vasc Biol 2012; 32:2104-12. [PMID: 22796579 DOI: 10.1161/atvbaha.111.241463] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Insulin plays a central role in regulating energy metabolism, including hepatic transport of very low-density lipoprotein (VLDL)-associated triglyceride. Hepatic hypersecretion of VLDL and consequent hypertriglyceridemia leads to lower circulating high-density lipoprotein levels and generation of small dense low-density lipoproteins characteristic of the dyslipidemia commonly observed in metabolic syndrome and type 2 diabetes mellitus. Physiological fluctuations of insulin modulate VLDL secretion, and insulin inhibition of VLDL secretion upon feeding may be the first pathway to become resistant in obesity that leads to VLDL hypersecretion. This review summarizes the role of insulin-related signaling pathways that determine hepatic VLDL production. Disruption in signaling pathways that reduce generation of the second messenger phosphatidylinositide (3,4,5) triphosphate downstream of activated phosphatidylinositide 3-kinase underlies the development of VLDL hypersecretion. As insulin resistance progresses, a number of pathways are altered that further augment VLDL hypersecretion, including hepatic inflammatory pathways. Insulin plays a complex role in regulating glucose metabolism, and it is not surprising that the role of insulin in VLDL and lipid metabolism will prove equally complex.
Collapse
Affiliation(s)
- Janet D Sparks
- University of Rochester Medical Center, Department of Pathology and Laboratory Medicine, Rochester, NY, USA
| | | | | |
Collapse
|
47
|
Kok BPC, Venkatraman G, Capatos D, Brindley DN. Unlike two peas in a pod: lipid phosphate phosphatases and phosphatidate phosphatases. Chem Rev 2012; 112:5121-46. [PMID: 22742522 DOI: 10.1021/cr200433m] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Bernard P C Kok
- Signal Transduction Research Group, Department of Biochemistry, School of Translational Medicine, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | | | | | | |
Collapse
|
48
|
Liu M, Chung S, Shelness GS, Parks JS. Hepatic ABCA1 and VLDL triglyceride production. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1821:770-7. [PMID: 22001232 PMCID: PMC3272310 DOI: 10.1016/j.bbalip.2011.09.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 09/23/2011] [Accepted: 09/26/2011] [Indexed: 02/04/2023]
Abstract
Elevated plasma triglyceride (TG) and reduced high density lipoprotein (HDL) concentrations are prominent features of metabolic syndrome (MS) and type 2 diabetes (T2D). Individuals with Tangier disease also have elevated plasma TG concentrations and a near absence of HDL, resulting from mutations in ATP binding cassette transporter A1 (ABCA1), which facilitates the efflux of cellular phospholipid and free cholesterol to assemble with apolipoprotein A-I (apoA-I), forming nascent HDL particles. In this review, we summarize studies focused on the regulation of hepatic very low density lipoprotein (VLDL) TG production, with particular attention on recent evidence connecting hepatic ABCA1 expression to VLDL, LDL, and HDL metabolism. Silencing ABCA1 in McArdle rat hepatoma cells results in diminished assembly of large (>10nm) nascent HDL particles, diminished PI3 kinase activation, and increased secretion of large, TG-enriched VLDL1 particles. Hepatocyte-specific ABCA1 knockout (HSKO) mice have a similar plasma lipid phenotype as Tangier disease subjects, with a two-fold elevation of plasma VLDL TG, 50% lower LDL, and 80% reduction in HDL concentrations. This lipid phenotype arises from increased hepatic secretion of VLDL1 particles, increased hepatic uptake of plasma LDL by the LDL receptor, elimination of nascent HDL particle assembly by the liver, and hypercatabolism of apoA-I by the kidney. These studies highlight a novel role for hepatic ABCA1 in the metabolism of all three major classes of plasma lipoproteins and provide a metabolic link between elevated TG and reduced HDL levels that are a common feature of Tangier disease, MS, and T2D. This article is part of a Special Issue entitled: Triglyceride Metabolism and Disease.
Collapse
Affiliation(s)
- Mingxia Liu
- Department of Pathology/Section on Lipid Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | | | | |
Collapse
|
49
|
Yin H, Hu M, Zhang R, Shen Z, Flatow L, You M. MicroRNA-217 promotes ethanol-induced fat accumulation in hepatocytes by down-regulating SIRT1. J Biol Chem 2012; 287:9817-9826. [PMID: 22308024 DOI: 10.1074/jbc.m111.333534] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Ethanol-mediated inhibition of hepatic sirtuin 1 (SIRT1) plays a crucial role in the pathogenesis of alcoholic fatty liver disease. Here, we investigated the underlying mechanisms of this inhibition by identifying a new hepatic target of ethanol action, microRNA-217 (miR-217). The role of miR-217 in the regulation of the effects of ethanol was investigated in cultured mouse AML-12 hepatocytes and in the livers of chronically ethanol-fed mice. In AML-12 hepatocytes and in mouse livers, chronic ethanol exposure drastically and specifically induced miR-217 levels and caused excess fat accumulation. Further studies revealed that overexpression of miR-217 in AML-12 cells promoted ethanol-mediated impairments of SIRT1 and SIRT1-regulated genes encoding lipogenic or fatty acid oxidation enzymes. More importantly, miR-217 impairs functions of lipin-1, a vital lipid regulator, in hepatocytes. Taken together, our novel findings suggest that miR-217 is a specific target of ethanol action in the liver and may present as a potential therapeutic target for treating human alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Huquan Yin
- Departments of Molecular Pharmacology and Physiology, University of South Florida Health Sciences Center, Tampa, Florida 33612
| | - Ming Hu
- Departments of Molecular Pharmacology and Physiology, University of South Florida Health Sciences Center, Tampa, Florida 33612
| | - Ray Zhang
- Departments of Molecular Pharmacology and Physiology, University of South Florida Health Sciences Center, Tampa, Florida 33612
| | - Zheng Shen
- Departments of Molecular Pharmacology and Physiology, University of South Florida Health Sciences Center, Tampa, Florida 33612
| | - Laura Flatow
- Departments of Molecular Pharmacology and Physiology, University of South Florida Health Sciences Center, Tampa, Florida 33612
| | - Min You
- Departments of Molecular Pharmacology and Physiology, University of South Florida Health Sciences Center, Tampa, Florida 33612.
| |
Collapse
|
50
|
Salvadó L, Serrano-Marco L, Barroso E, Palomer X, Vázquez-Carrera M. Targeting PPARβ/δ for the treatment of type 2 diabetes mellitus. Expert Opin Ther Targets 2012; 16:209-23. [DOI: 10.1517/14728222.2012.658370] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|