151
|
Szendroedi J, Roden M. Perilipin 5: from fatty liver to hepatic lipodystrophy? Hepatology 2015; 61:751-3. [PMID: 25420909 DOI: 10.1002/hep.27618] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 11/19/2014] [Indexed: 01/29/2023]
Affiliation(s)
- Julia Szendroedi
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich-Heine University, Düsseldorf, Germany; German Center of Diabetes Research (DZD e.V.), Partner Site, Düsseldorf, Germany; Department of Endocrinology and Diabetology, Heinrich-Heine University, Düsseldorf, Germany
| | | |
Collapse
|
152
|
Amrutkar M, Cansby E, Nuñez-Durán E, Pirazzi C, Ståhlman M, Stenfeldt E, Smith U, Borén J, Mahlapuu M. Protein kinase STK25 regulates hepatic lipid partitioning and progression of liver steatosis and NASH. FASEB J 2015; 29:1564-76. [PMID: 25609431 DOI: 10.1096/fj.14-264937] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/08/2014] [Indexed: 12/21/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common form of liver disease, and 10% to 20% of NAFLD patients progress to nonalcoholic steatohepatitis (NASH). The molecular pathways controlling progression to NAFLD/NASH remain poorly understood. We recently identified serine/threonine protein kinase 25 (STK25) as a regulator of whole-body insulin and glucose homeostasis. This study investigates the role of STK25 in liver lipid accumulation and NASH. Stk25 transgenic mice challenged with a high-fat diet displayed a dramatic increase in liver steatosis and hepatic insulin resistance compared to wild-type siblings. Focal fibrosis, hepatocellular damage, and inflammation were readily seen in transgenic but not wild-type livers. Transgenic livers displayed reduced β-oxidation and triacylglycerol secretion, while lipid uptake and synthesis remained unchanged. STK25 was associated with lipid droplets, colocalizing with the main hepatic lipid droplet-coating protein adipose differentiation-related protein, the level of which was increased 3.8 ± 0.7-fold in transgenic livers (P < 0.01), while a key hepatic lipase, adipose triacylglycerol lipase, was translocated from the lipid droplets surface to the cytoplasm, providing the likely mechanism underlying the effect of STK25. In summary, STK25 is a lipid droplet-associated protein that promotes NAFLD through control of lipid release from the droplets for β-oxidation and triacylglycerol secretion. STK25 also drives pathogenesis of NASH.
Collapse
Affiliation(s)
- Manoj Amrutkar
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Emmelie Cansby
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Esther Nuñez-Durán
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Carlo Pirazzi
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Marcus Ståhlman
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Elin Stenfeldt
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Ulf Smith
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jan Borén
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Margit Mahlapuu
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
153
|
Pollak NM, Jaeger D, Kolleritsch S, Zimmermann R, Zechner R, Lass A, Haemmerle G. The interplay of protein kinase A and perilipin 5 regulates cardiac lipolysis. J Biol Chem 2015; 290:1295-306. [PMID: 25418045 PMCID: PMC4340377 DOI: 10.1074/jbc.m114.604744] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 11/21/2014] [Indexed: 12/21/2022] Open
Abstract
Defective lipolysis in mice lacking adipose triglyceride lipase provokes severe cardiac steatosis and heart dysfunction, markedly shortening life span. Similarly, cardiac muscle (CM)-specific Plin5 overexpression (CM-Plin5) leads to severe triglyceride (TG) accumulation in cardiomyocytes via impairing TG breakdown. Interestingly, cardiac steatosis due to overexpression of Plin5 is compatible with normal heart function and life span indicating a more moderate impact of Plin5 overexpression on cardiac lipolysis and energy metabolism. We hypothesized that cardiac Plin5 overexpression does not constantly impair cardiac lipolysis. In line with this assumption, TG levels decreased in CM of fasted compared with nonfasted CM-Plin5 mice indicating that fasting may lead to a diminished barrier function of Plin5. Recent studies demonstrated that Plin5 is phosphorylated, and activation of adenylyl cyclase leads to phosphorylation of Plin5, suggesting that Plin5 is a substrate for PKA. Furthermore, any significance of Plin5 phosphorylation by PKA in the regulation of TG mobilization from lipid droplets (LDs) is unknown. Here, we show that the lipolytic barrier of Plin5-enriched LDs, either prepared from cardiac tissue of CM-Plin5 mice or Plin5-transfected cells, is abrogated by incubation with PKA. Notably, PKA-induced lipolysis of LDs enriched with Plin5 carrying a single mutation at serine 155 (PlinS155A) of the putative PKA phosphorylation site was substantially impaired revealing a critical role for PKA in Plin5-regulated lipolysis. The strong increase in protein levels of phosphorylated PKA in CM of Plin5 transgenic mice may partially restore fatty acid release from Plin5-enriched LDs, rendering these hearts compatible with normal heart function despite massive steatosis.
Collapse
Affiliation(s)
- Nina M Pollak
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Doris Jaeger
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | | | - Robert Zimmermann
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Rudolf Zechner
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Achim Lass
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Guenter Haemmerle
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| |
Collapse
|
154
|
Liu S, Geng B, Zou L, Wei S, Wang W, Deng J, Xu C, Zhao X, Lyu Y, Su X, Xu G. Development of hypertrophic cardiomyopathy in perilipin-1 null mice with adipose tissue dysfunction. Cardiovasc Res 2014; 105:20-30. [DOI: 10.1093/cvr/cvu214] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
|
155
|
Asimakopoulou A, Borkham-Kamphorst E, Henning M, Yagmur E, Gassler N, Liedtke C, Berger T, Mak TW, Weiskirchen R. Lipocalin-2 (LCN2) regulates PLIN5 expression and intracellular lipid droplet formation in the liver. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1842:1513-1524. [PMID: 25086218 DOI: 10.1016/j.bbalip.2014.07.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 07/17/2014] [Accepted: 07/22/2014] [Indexed: 01/14/2023]
Abstract
Lipocalin-2 (LCN2) belongs to the superfamily of lipocalins and plays critical roles in the control of cellular homeostasis during inflammation and in responses to cellular stress or injury. In the liver, LCN2 triggers protective effects following acute or chronic injury, and its expression is a reliable indicator of liver damage. However, little is known about LCN2's functions in the homeostasis and metabolism of hepatic lipids or in the development of steatosis. In this study, we fed wild type (WT) and LCN2-deficient (Lcn2(-/-)) mice a methionine- and choline-deficient (MCD) diet as a nutritional model of non-alcoholic steatohepatitis, and compared intrahepatic lipid accumulation, lipid droplet formation, mitochondrial content, and expression of the Perilipin proteins that regulate cellular lipid metabolism. We found that Lcn2(-/-) mice fed an MCD diet accumulated more lipids in the liver than WT controls, and that the basal expression of the lipid droplet coat protein Perilipin 5 (PLIN5, also known as OXPAT) was significantly reduced in these animals. Similarly, the overexpression of LCN2 and PLIN5 were also found in animals that were fed with a high fat diet. Furthermore, the loss of LCN2 and/or PLIN5 in hepatocytes prevented normal intracellular lipid droplet formation both in vitro and in vivo. Restoration of LCN2 in Lcn2(-/-) primary hepatocytes by either transfection or adenoviral vector infection induced PLIN5 expression and restored proper lipid droplet formation. Our data indicate that LCN2 is a key modulator of hepatic lipid homeostasis that controls the formation of intracellular lipid droplets by regulating PLIN5 expression. LCN2 may therefore represent a novel therapeutic drug target for the treatment of liver diseases associated with elevated fat accumulation and steatosis.
Collapse
Affiliation(s)
- Anastasia Asimakopoulou
- Institute of Clinical Chemistry and Pathobiochemistry, RWTH University Hospital Aachen, Aachen, Germany
| | - Erawan Borkham-Kamphorst
- Institute of Clinical Chemistry and Pathobiochemistry, RWTH University Hospital Aachen, Aachen, Germany
| | - Marc Henning
- Institute of Clinical Chemistry and Pathobiochemistry, RWTH University Hospital Aachen, Aachen, Germany
| | - Eray Yagmur
- MVZ Medical Laboratory Center, Dr. Stein and Partner, Mönchengladbach, Germany
| | - Nikolaus Gassler
- Institute of Pathology, RWTH University Hospital Aachen, Aachen, Germany
| | - Christian Liedtke
- Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| | - Thorsten Berger
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, ON, Canada
| | - Tak W Mak
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, ON, Canada; Ontario Cancer Institute, University Health Network, Toronto, ON, Canada
| | - Ralf Weiskirchen
- Institute of Clinical Chemistry and Pathobiochemistry, RWTH University Hospital Aachen, Aachen, Germany.
| |
Collapse
|
156
|
Soufi N, Hall AM, Chen Z, Yoshino J, Collier SL, Mathews JC, Brunt EM, Albert CJ, Graham MJ, Ford DA, Finck BN. Inhibiting monoacylglycerol acyltransferase 1 ameliorates hepatic metabolic abnormalities but not inflammation and injury in mice. J Biol Chem 2014; 289:30177-88. [PMID: 25213859 DOI: 10.1074/jbc.m114.595850] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Abnormalities in hepatic lipid metabolism and insulin action are believed to play a critical role in the etiology of nonalcoholic steatohepatitis. Monoacylglycerol acyltransferase (MGAT) enzymes convert monoacylglycerol to diacylglycerol, which is the penultimate step in one pathway for triacylglycerol synthesis. Hepatic expression of Mogat1, which encodes an MGAT enzyme, is increased in the livers of mice with hepatic steatosis, and knocking down Mogat1 improves glucose metabolism and hepatic insulin signaling, but whether increased MGAT activity plays a role in the etiology of nonalcoholic steatohepatitis is unclear. To examine this issue, mice were placed on a diet containing high levels of trans fatty acids, fructose, and cholesterol (HTF-C diet) or a low fat control diet for 4 weeks. Mice were injected with antisense oligonucleotides (ASOs) to knockdown Mogat1 or a scrambled ASO control for 12 weeks while remaining on diet. The HTF-C diet caused glucose intolerance, hepatic steatosis, and induced hepatic gene expression markers of inflammation, macrophage infiltration, and stellate cell activation. Mogat1 ASO treatment, which suppressed Mogat1 expression in liver and adipose tissue, attenuated weight gain, improved glucose tolerance, improved hepatic insulin signaling, and decreased hepatic triacylglycerol content compared with control ASO-treated mice on HTF-C chow. However, Mogat1 ASO treatment did not reduce hepatic diacylglycerol, cholesterol, or free fatty acid content; improve histologic measures of liver injury; or reduce expression of markers of stellate cell activation, liver inflammation, and injury. In conclusion, inhibition of hepatic Mogat1 in HTF-C diet-fed mice improves hepatic metabolic abnormalities without attenuating liver inflammation and injury.
Collapse
Affiliation(s)
| | | | | | | | | | - James C Mathews
- Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri 63110
| | - Elizabeth M Brunt
- Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri 63110
| | - Carolyn J Albert
- the Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, Missouri 63104, and
| | - Mark J Graham
- ISIS Pharmaceuticals Inc., Carlsbad, California 92008
| | - David A Ford
- the Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, Missouri 63104, and
| | | |
Collapse
|
157
|
Mason RR, Mokhtar R, Matzaris M, Selathurai A, Kowalski GM, Mokbel N, Meikle PJ, Bruce CR, Watt MJ. PLIN5 deletion remodels intracellular lipid composition and causes insulin resistance in muscle. Mol Metab 2014; 3:652-63. [PMID: 25161888 PMCID: PMC4142393 DOI: 10.1016/j.molmet.2014.06.002] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 05/30/2014] [Accepted: 06/05/2014] [Indexed: 01/28/2023] Open
Abstract
Defective control of lipid metabolism leading to lipotoxicity causes insulin resistance in skeletal muscle, a major factor leading to diabetes. Here, we demonstrate that perilipin (PLIN) 5 is required to couple intramyocellular triacylglycerol lipolysis with the metabolic demand for fatty acids. PLIN5 ablation depleted triacylglycerol stores but increased sphingolipids including ceramide, hydroxylceramides and sphingomyelin. We generated perilipin 5 (Plin5)(-/-) mice to determine the functional significance of PLIN5 in metabolic control and insulin action. Loss of PLIN5 had no effect on body weight, feeding or adiposity but increased whole-body carbohydrate oxidation. Plin5 (-/-) mice developed skeletal muscle insulin resistance, which was associated with ceramide accumulation. Liver insulin sensitivity was improved in Plin5 (-/-) mice, indicating tissue-specific effects of PLIN5 on insulin action. We conclude that PLIN5 plays a critical role in coordinating skeletal muscle triacylglycerol metabolism, which impacts sphingolipid metabolism, and is requisite for the maintenance of skeletal muscle insulin action.
Collapse
Affiliation(s)
- Rachael R. Mason
- Biology of Lipid Metabolism Laboratory, Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Ruzaidi Mokhtar
- Biology of Lipid Metabolism Laboratory, Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Maria Matzaris
- Biology of Lipid Metabolism Laboratory, Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Ahrathy Selathurai
- Biology of Lipid Metabolism Laboratory, Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Greg M. Kowalski
- Biology of Lipid Metabolism Laboratory, Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Nancy Mokbel
- Garvan Institute of Medical Research, Darlinghurst., New South Wales, 2006, Australia
| | - Peter J. Meikle
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia
| | - Clinton R. Bruce
- Biology of Lipid Metabolism Laboratory, Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Matthew J. Watt
- Biology of Lipid Metabolism Laboratory, Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| |
Collapse
|
158
|
Renaud HJ, Cui YJ, Lu H, Zhong XB, Klaassen CD. Ontogeny of hepatic energy metabolism genes in mice as revealed by RNA-sequencing. PLoS One 2014; 9:e104560. [PMID: 25102070 PMCID: PMC4125194 DOI: 10.1371/journal.pone.0104560] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 07/14/2014] [Indexed: 12/18/2022] Open
Abstract
The liver plays a central role in metabolic homeostasis by coordinating synthesis, storage, breakdown, and redistribution of nutrients. Hepatic energy metabolism is dynamically regulated throughout different life stages due to different demands for energy during growth and development. However, changes in gene expression patterns throughout ontogeny for factors important in hepatic energy metabolism are not well understood. We performed detailed transcript analysis of energy metabolism genes during various stages of liver development in mice. Livers from male C57BL/6J mice were collected at twelve ages, including perinatal and postnatal time points (n = 3/age). The mRNA was quantified by RNA-Sequencing, with transcript abundance estimated by Cufflinks. One thousand sixty energy metabolism genes were examined; 794 were above detection, of which 627 were significantly changed during at least one developmental age compared to adult liver. Two-way hierarchical clustering revealed three major clusters dependent on age: GD17.5–Day 5 (perinatal-enriched), Day 10–Day 20 (pre-weaning-enriched), and Day 25–Day 60 (adolescence/adulthood-enriched). Clustering analysis of cumulative mRNA expression values for individual pathways of energy metabolism revealed three patterns of enrichment: glycolysis, ketogenesis, and glycogenesis were all perinatally-enriched; glycogenolysis was the only pathway enriched during pre-weaning ages; whereas lipid droplet metabolism, cholesterol and bile acid metabolism, gluconeogenesis, and lipid metabolism were all enriched in adolescence/adulthood. This study reveals novel findings such as the divergent expression of the fatty acid β-oxidation enzymes Acyl-CoA oxidase 1 and Carnitine palmitoyltransferase 1a, indicating a switch from mitochondrial to peroxisomal β-oxidation after weaning; as well as the dynamic ontogeny of genes implicated in obesity such as Stearoyl-CoA desaturase 1 and Elongation of very long chain fatty acids-like 3. These data shed new light on the ontogeny of homeostatic regulation of hepatic energy metabolism, which could ultimately provide new therapeutic targets for metabolic diseases.
Collapse
Affiliation(s)
- Helen J. Renaud
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Yue Julia Cui
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, New York, United States of America
| | - Xiao-bo Zhong
- Department of Pharmaceutical Sciences, University of Connecticut School of Pharmacy, Storrs, Connecticut, United States of America
| | - Curtis D. Klaassen
- College of Medicine, University of Kansas, Kansas City, Kansas, United States of America
- * E-mail:
| |
Collapse
|
159
|
Church RJ, Wu H, Mosedale M, Sumner SJ, Pathmasiri W, Kurtz CL, Pletcher MT, Eaddy JS, Pandher K, Singer M, Batheja A, Watkins PB, Adkins K, Harrill AH. A systems biology approach utilizing a mouse diversity panel identifies genetic differences influencing isoniazid-induced microvesicular steatosis. Toxicol Sci 2014; 140:481-92. [PMID: 24848797 DOI: 10.1093/toxsci/kfu094] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Isoniazid (INH), the mainstay therapeutic for tuberculosis infection, has been associated with rare but serious hepatotoxicity in the clinic. However, the mechanisms underlying inter-individual variability in the response to this drug have remained elusive. A genetically diverse mouse population model in combination with a systems biology approach was utilized to identify transcriptional changes, INH-responsive metabolites, and gene variants that contribute to the liver response in genetically sensitive individuals. Sensitive mouse strains developed severe microvesicular steatosis compared with corresponding vehicle control mice following 3 days of oral treatment with INH. Genes involved in mitochondrial dysfunction were enriched among liver transcripts altered with INH treatment. Those associated with INH treatment and susceptibility to INH-induced steatosis in the liver included apolipoprotein A-IV, lysosomal-associated membrane protein 1, and choline phosphotransferase 1. These alterations were accompanied by metabolomic changes including reduced levels of glutathione and the choline metabolites betaine and phosphocholine, suggesting that oxidative stress and reduced lipid export may additionally contribute to INH-induced steatosis. Finally, genome-wide association mapping revealed that polymorphisms in perilipin 2 were linked to increased triglyceride levels following INH treatment, implicating a role for inter-individual differences in lipid packaging in the susceptibility to INH-induced steatosis. Taken together, our data suggest that INH-induced steatosis is caused by not one, but multiple events involving lipid retention in the livers of genetically sensitive individuals. This work also highlights the value of using a mouse diversity panel to investigate drug-induced responses across a diverse population.
Collapse
Affiliation(s)
- Rachel J Church
- Hamner-University of North Carolina Institute for Drug Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709
| | - Hong Wu
- Drug Safety Research and Development, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Merrie Mosedale
- Hamner-University of North Carolina Institute for Drug Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709
| | - Susan J Sumner
- Discovery Sciences, RTI International, Research Triangle Park, North Carolina 27709
| | - Wimal Pathmasiri
- Discovery Sciences, RTI International, Research Triangle Park, North Carolina 27709
| | - Catherine L Kurtz
- Hamner-University of North Carolina Institute for Drug Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709
| | - Mathew T Pletcher
- Drug Safety Research and Development, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - John S Eaddy
- Hamner-University of North Carolina Institute for Drug Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709
| | - Karamjeet Pandher
- Drug Safety Research and Development, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Monica Singer
- Janssen Research and Development, Drug Safety Sciences, Raritan, New Jersey 08869
| | - Ameesha Batheja
- Janssen Research and Development, Drug Safety Sciences, Raritan, New Jersey 08869
| | - Paul B Watkins
- Hamner-University of North Carolina Institute for Drug Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709 Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599 School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Karissa Adkins
- Drug Safety Research and Development, Pfizer Global Research and Development, Groton, Connecticut 06340
| | - Alison H Harrill
- Hamner-University of North Carolina Institute for Drug Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709 Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599 The University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| |
Collapse
|
160
|
Zelenchuk LV, Hedge AM, Rowe PSN. PHEX mimetic (SPR4-peptide) corrects and improves HYP and wild type mice energy-metabolism. PLoS One 2014; 9:e97326. [PMID: 24839967 PMCID: PMC4026222 DOI: 10.1371/journal.pone.0097326] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 04/17/2014] [Indexed: 12/19/2022] Open
Abstract
CONTEXT PHEX or DMP1 mutations cause hypophosphatemic-rickets and altered energy metabolism. PHEX binds to DMP1-ASARM-motif to form a complex with α5β3 integrin that suppresses FGF23 expression. ASARM-peptides increase FGF23 by disrupting the PHEX-DMP1-Integrin complex. We used a 4.2 kDa peptide (SPR4) that binds to ASARM-peptide/motif to study the DMP1-PHEX interaction and to assess SPR4 for the treatment of energy metabolism defects in HYP and potentially other bone-mineral disorders. DESIGN Subcutaneously transplanted osmotic pumps were used to infuse SPR4-peptide or vehicle (VE) into wild-type mice (WT) and HYP-mice (PHEX mutation) for 4 weeks. RESULTS SPR4 partially corrected HYP mice hypophosphatemia and increased serum 1.25(OH)2D3. Serum FGF23 remained high and PTH was unaffected. WT-SPR4 mice developed hypophosphatemia and hypercalcemia with increased PTH, FGF23 and 1.25(OH)2D3. SPR4 increased GAPDH HYP-bone expression 60× and corrected HYP-mice hyperglycemia and hypoinsulinemia. HYP-VE serum uric-acid (UA) levels were reduced and SPR4 infusion suppressed UA levels in WT-mice but not HYP-mice. SPR4 altered leptin, adiponectin, and sympathetic-tone and increased the fat mass/weight ratio for HYP and WT mice. Expression of perlipin-2 a gene involved in obesity was reduced in HYP-VE and WT-SPR4 mice but increased in HYP-SPR4 mice. Also, increased expression of two genes that inhibit insulin-signaling, ENPP1 and ESP, occurred with HYP-VE mice. In contrast, SPR4 reduced expression of both ENPP1 and ESP in WT mice and suppressed ENPP1 in HYP mice. Increased expression of FAM20C and sclerostin occurred with HYP-VE mice. SPR4 suppressed expression of FAM20C and sclerostin in HYP and WT mice. CONCLUSIONS ASARM peptides and motifs are physiological substrates for PHEX and modulate osteocyte PHEX-DMP1-α5β3-integrin interactions and thereby FGF23 expression. These interactions also provide a nexus that regulates bone and energy metabolism. SPR4 suppression of sclerostin and/or sequestration of ASARM-peptides improves energy metabolism and may have utility for treating familial rickets, osteoporosis, obesity and diabetes.
Collapse
Affiliation(s)
- Lesya V. Zelenchuk
- Internal Medicine, The Kidney Institute, Kansas University Medical Center (KUMC), Kansas City, Kansas, United States of America
| | - Anne-Marie Hedge
- Internal Medicine, The Kidney Institute, Kansas University Medical Center (KUMC), Kansas City, Kansas, United States of America
| | - Peter S. N. Rowe
- Internal Medicine, The Kidney Institute, Kansas University Medical Center (KUMC), Kansas City, Kansas, United States of America
| |
Collapse
|
161
|
Carr RM, Peralta G, Yin X, Ahima RS. Absence of perilipin 2 prevents hepatic steatosis, glucose intolerance and ceramide accumulation in alcohol-fed mice. PLoS One 2014; 9:e97118. [PMID: 24831094 PMCID: PMC4022498 DOI: 10.1371/journal.pone.0097118] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 04/14/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Perilipin 2 (Plin2) is a lipid droplet protein that has roles in both lipid and glucose homeostasis. An increase in Plin2 in liver is associated with the development of steatosis, glucose intolerance, and ceramide accumulation in alcoholic liver disease. We investigated the role of Plin2 on energy balance and glucose and lipid homeostasis in wildtype and Plin2 knockout (Plin2KO) mice chronically fed a Lieber-DeCarli liquid ethanol or control diet for six weeks. METHODS We performed in vivo measurements of energy intake and expenditure; body composition; and glucose tolerance. After sacrifice, liver was dissected for histology and lipid analysis. RESULTS We found that neither genotype nor diet had a significant effect on final weight, body composition, or energy intake between WT and Plin2KO mice fed alcohol or control diets. Additionally, alcohol feeding did not affect oxygen consumption or carbon dioxide production in Plin2KO mice. We performed glucose tolerance testing and observed that alcohol feeding failed to impair glucose tolerance in Plin2KO mice. Most notably, absence of Plin2 prevented hepatic steatosis and ceramide accumulation in alcohol-fed mice. These changes were related to downregulation of genes involved in lipogenesis and triglyceride synthesis. CONCLUSIONS Plin2KO mice chronically fed alcohol are protected from hepatic steatosis, glucose intolerance, and hepatic ceramide accumulation, suggesting a critical pathogenic role of Plin2 in experimental alcoholic liver disease.
Collapse
Affiliation(s)
- Rotonya M. Carr
- University of Pennsylvania, Perelman School of Medicine, Department of Medicine, Gastroenterology Division, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| | - Giselle Peralta
- University of Pennsylvania, Perelman School of Medicine, Institute for Diabetes, Obesity and Metabolism, Philadelphia, Pennsylvania, United States of America
| | - Xiaoyan Yin
- University of Pennsylvania, Perelman School of Medicine, Institute for Diabetes, Obesity and Metabolism, Philadelphia, Pennsylvania, United States of America
| | - Rexford S. Ahima
- University of Pennsylvania, Perelman School of Medicine, Institute for Diabetes, Obesity and Metabolism, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
162
|
Targeting Hepatic Glycerolipid Synthesis and Turnover to Treat Fatty Liver Disease. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/498369] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) encompasses a spectrum of metabolic abnormalities ranging from simple hepatic steatosis (accumulation of neutral lipid) to development of steatotic lesions, steatohepatitis, and cirrhosis. NAFLD is extremely prevalent in obese individuals and with the epidemic of obesity; nonalcoholic steatohepatitis (NASH) has become the most common cause of liver disease in the developed world. NASH is rapidly emerging as a prominent cause of liver failure and transplantation. Moreover, hepatic steatosis is tightly linked to risk of developing insulin resistance, diabetes, and cardiovascular disease. Abnormalities in hepatic lipid metabolism are part and parcel of the development of NAFLD and human genetic studies and work conducted in experimentally tractable systems have identified a number of enzymes involved in fat synthesis and degradation that are linked to NAFLD susceptibility as well as progression to NASH. The goal of this review is to summarize the current state of our knowledge on these pathways and focus on how they contribute to etiology of NAFLD and related metabolic diseases.
Collapse
|
163
|
Wan J, Benkdane M, Alons E, Lotersztajn S, Pavoine C. M2 kupffer cells promote hepatocyte senescence: an IL-6-dependent protective mechanism against alcoholic liver disease. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1763-72. [PMID: 24713392 DOI: 10.1016/j.ajpath.2014.02.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 01/08/2014] [Accepted: 02/19/2014] [Indexed: 12/22/2022]
Abstract
Alcoholic liver disease is a predominant cause of liver-related mortality in Western countries. The early steps of alcohol-induced steatosis and liver injury involve several mechanisms, including inflammation and oxidative stress. The inflammatory process is initiated by polarization of Kupffer cells toward a proinflammatory M1 phenotype, and we recently found that promoting anti-inflammatory M2 Kupffer cell polarization protects against alcohol-induced hepatocyte steatosis and apoptosis. Alcohol-induced oxidative stress is a potential trigger of senescence, and senescent cells exhibit characteristic functional resistance to apoptosis. We sought to evaluate induction of hepatocyte senescence as an early protective mechanism against alcoholic liver disease. Combining in vivo and in vitro studies, we show that M2 macrophages trigger hepatocyte senescence and enhance alcohol-induced hepatocyte senescence, as indicated by increased β-galactosidase activity, elevated CDKN1A mRNA expression, and induction of nuclear p21. We identify IL-6 as the mediator of M2-induced hepatocyte senescence. Senescent hepatocytes display characteristic resistance to apoptosis but also to steatosis, thus arguing for an early protective effect against alcoholic liver disease. These findings further suggest that pharmacologic interventions targeting M2 polarization during the early stages of alcoholic liver disease may represent an attractive strategy for the limitation of inflammation, hepatocyte apoptosis, and steatosis.
Collapse
Affiliation(s)
- Jinghong Wan
- INSERM, U955, Team 17, Créteil, France; UMR_S955, University of Paris-Est. Créteil, Créteil, France
| | - Merieme Benkdane
- INSERM, U955, Team 17, Créteil, France; UMR_S955, University of Paris-Est. Créteil, Créteil, France
| | - Elizabeth Alons
- INSERM, U955, Team 17, Créteil, France; UMR_S955, University of Paris-Est. Créteil, Créteil, France
| | - Sophie Lotersztajn
- INSERM, U955, Team 17, Créteil, France; UMR_S955, University of Paris-Est. Créteil, Créteil, France; Department of Hepatology and Gastroenterology, AP-HP, Hôpital H. Mondor-A. Chenevier, Créteil, France
| | - Catherine Pavoine
- INSERM, U955, Team 17, Créteil, France; UMR_S955, University of Paris-Est. Créteil, Créteil, France.
| |
Collapse
|
164
|
Zierler KA, Zechner R, Haemmerle G. Comparative gene identification-58/α/β hydrolase domain 5: more than just an adipose triglyceride lipase activator? Curr Opin Lipidol 2014; 25:102-9. [PMID: 24565921 PMCID: PMC4170181 DOI: 10.1097/mol.0000000000000058] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Comparative gene identification-58 (CGI-58) is a lipid droplet-associated protein that controls intracellular triglyceride levels by its ability to activate adipose triglyceride lipase (ATGL). Additionally, CGI-58 was described to exhibit lysophosphatidic acid acyl transferase (LPAAT) activity. This review focuses on the significance of CGI-58 in energy metabolism in adipose and nonadipose tissue. RECENT FINDINGS Recent studies with transgenic and CGI-58-deficient mouse strains underscored the importance of CGI-58 as a regulator of intracellular energy homeostasis by modulating ATGL-driven triglyceride hydrolysis. In accordance with this function, mice and humans that lack CGI-58 accumulate triglyceride in multiple tissues. Additionally, CGI-58-deficient mice develop an ATGL-independent severe skin barrier defect and die soon after birth. Although the premature death prevented a phenotypical characterization of adult global CGI-58 knockout mice, the characterization of mice with tissue-specific CGI-58 deficiency revealed new insights into its role in neutral lipid and energy metabolism. Concerning the ATGL-independent function of CGI-58, a recently identified LPAAT activity for CGI-58 was shown to be involved in the generation of signaling molecules regulating inflammatory processes and insulin action. SUMMARY Although the function of CGI-58 in the catabolism of cellular triglyceride depots via ATGL is well established, further studies are required to consolidate the function of CGI-58 as LPAAT and to clarify the involvement of CGI-58 in the metabolism of skin lipids.
Collapse
Affiliation(s)
- Kathrin A Zierler
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | | |
Collapse
|
165
|
Abstract
PURPOSE OF REVIEW We summarize recent mechanistic and physiological studies related to the role of perilipin 5 (Plin5) in regulating lipid droplet accumulation and protection to fatty acids in tissues with high lipid oxidative metabolism. RECENT FINDINGS Plin5 is a lipid droplet targeting protein that promotes association of lipid droplets with mitochondria and is most highly expressed in oxidative tissues, including cardiac and skeletal muscle. Recent in-vivo and in-vitro data indicate an important role for Plin5 in the regulation of cardiac lipid storage and function. Targeted overexpression of Plin5 in heart causes steatosis, mild mitochondria dysfunction, and hypertrophy in cardiac tissue, but without affecting cardiac function. In contrast, whole body ablation of Plin5 (Plin5 mice) reduces cardiac lipid droplet formation, increases cardiac fatty acid oxidation, and promotes cardiac dysfunction; cardiac defects can be prevented with antioxidative therapy. These data suggest a cytoprotective role for Plin5 to promote lipid storage but to limit fatty acid toxicity, parameters critical for tissues with high lipid oxidative metabolism. SUMMARY In-vivo and in-vitro data suggest that Plin5 is part of a cell-adaptive response to high lipid oxidative metabolism to protect lipid droplet storage against neutral lipases and, so, limit fatty acid accumulation. Although the specific mechanisms that underlie Plin5 lipid droplet storage protection in oxidative tissues remain to be fully elucidated, Plin5 provides a basis for the novel cytoprotective nature of lipid droplets.
Collapse
Affiliation(s)
- Alan R. Kimmel
- Laboratory of Cellular and Developmental Biology (50/3351), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, United States. ; Tel: 301-496-3016
| | - Carole Sztalryd
- Division of Endocrinology, Department of Medicine, School of Medicine, University of Maryland, Baltimore, Maryland 21201, United States
- The Geriatric Research, Education and Clinical Center, Baltimore Veterans Affairs Health Care Center, Division of Endocrinology, Department of Medicine, School of Medicine, University of Maryland, Baltimore, Maryland 21201, United States. ; Tel: 410-706-4047
| |
Collapse
|
166
|
Modulation of adipose tissue lipolysis and body weight by high-density lipoproteins in mice. Nutr Diabetes 2014; 4:e108. [PMID: 24567123 PMCID: PMC3940828 DOI: 10.1038/nutd.2014.4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 01/05/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Obesity is associated with reduced levels of circulating high-density lipoproteins (HDLs) and its major protein, apolipoprotein (apo) A-I. As a result of the role of HDL and apoA-I in cellular lipid transport, low HDL and apoA-I may contribute directly to establishing or maintaining the obese condition. METHODS To test this, male C57BL/6 wild-type (WT), apoA-I deficient (apoA-I(-/-)) and apoA-I transgenic (apoA-I(tg/tg)) mice were fed obesogenic diets (ODs) and monitored for several clinical parameters. We also performed cell culture studies. RESULTS ApoA-I(-/-) mice gained significantly more body weight and body fat than WT mice over 20 weeks despite their reduced food intake. During a caloric restriction regime imposed on OD-fed mice, apoA-I deficiency significantly inhibited the loss of body fat as compared with WT mice. Reduced body fat loss with caloric restriction in apoA-I(-/-) mice was associated with blunted stimulated adipose tissue lipolysis as verified by decreased levels of phosphorylated hormone-sensitive lipase (p-HSL) and lipolytic enzyme mRNA. In contrast to apoA-I(-/-) mice, apoA-I(tg/tg) mice gained relatively less weight than WT mice, consistent with other reports. ApoA-I(tg/tg) mice showed increased adipose tissue lipolysis, verified by increased levels of p-HSL and lipolytic enzyme mRNA. In cell culture studies, HDL and apoA-I specifically increased catecholamine-induced lipolysis possibly through modulating the adipocyte plasma membrane cholesterol content. CONCLUSIONS Thus, apoA-I and HDL contribute to modulating body fat content by controlling the extent of lipolysis. ApoA-I and HDL are key components of lipid metabolism in adipose tissue and constitute new therapeutic targets in obesity.
Collapse
|
167
|
Trent CM, Yu S, Hu Y, Skoller N, Huggins LA, Homma S, Goldberg IJ. Lipoprotein lipase activity is required for cardiac lipid droplet production. J Lipid Res 2014; 55:645-58. [PMID: 24493834 PMCID: PMC3966699 DOI: 10.1194/jlr.m043471] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The rodent heart accumulates TGs and lipid droplets during fasting. The sources of heart lipids could be either FFAs liberated from adipose tissue or FAs from lipoprotein-associated TGs via the action of lipoprotein lipase (LpL). Because circulating levels of FFAs increase during fasting, it has been assumed that albumin transported FFAs are the source of lipids within heart lipid droplets. We studied mice with three genetic mutations: peroxisomal proliferator-activated receptor α deficiency, cluster of differentiation 36 (CD36) deficiency, and heart-specific LpL deletion. All three genetically altered groups of mice had defective accumulation of lipid droplet TGs. Moreover, hearts from mice treated with poloxamer 407, an inhibitor of lipoprotein TG lipolysis, also failed to accumulate TGs, despite increased uptake of FFAs. TG storage did not impair maximal cardiac function as measured by stress echocardiography. Thus, LpL hydrolysis of circulating lipoproteins is required for the accumulation of lipids in the heart of fasting mice.
Collapse
Affiliation(s)
- Chad M Trent
- Division of Preventive Medicine and Nutrition, Columbia University College of Physicians and Surgeons, New York, NY 10032
| | | | | | | | | | | | | |
Collapse
|
168
|
Abstract
PURPOSE OF REVIEW With the realization that lipid droplets are not merely inert fat storage organelles, but highly dynamic and actively involved in cellular lipid homeostasis, there has been an increased interest in lipid droplet biology. Recent studies have begun to unravel the roles that lipid dropletss play in cellular physiology and provide insights into the mechanisms by which lipid droplets contribute to cellular homeostasis. This review provides a summary of these recent publications on lipid droplet metabolism. RECENT FINDINGS Perilipins have different preferences for associating with triacylglycerol (TAG) or cholesteryl esters, different tissue distributions, and each contributes to lipid metabolism in its unique way. Cell death-inducing DFF45-like effector proteins are not only involved in lipid droplet expansion, but also in the cellular response to stress and lipid secretion. Lipid droplets undergo an active cycle of lipolysis and re-esterification to form microlipid droplets. TAG synthesis for lipid droplet formation and expansion occurs in the endoplasmic reticulum and on lipid droplets, and TAG transfers between lipid droplets during lipid droplet fusion. Lipid droplets interact with the endoplasmic reticulum and mitochondria to facilitate lipid transfer, lipid droplet expansion, and metabolism. SUMMARY Lipid droplets are dynamically active, responding to changes in cellular physiology, as well as interacting with cytosolic proteins and other organelles to control lipid homeostasis.
Collapse
Affiliation(s)
- Victor K Khor
- aVeterans Affairs Palo Alto Healthcare System, Palo Alto bDivision of Endocrinology, Stanford University, Stanford, California, USA
| | | | | |
Collapse
|
169
|
Perilipins: lipid droplet coat proteins adapted for tissue-specific energy storage and utilization, and lipid cytoprotection. Biochimie 2013; 96:96-101. [PMID: 24036367 DOI: 10.1016/j.biochi.2013.08.026] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 08/28/2013] [Indexed: 12/18/2022]
Abstract
Cytosolic lipid storage droplets are primary functional organelles that regulate cellular lipid metabolism and homeostasis. Paradoxically, excess lipid stores are linked to both adaptive (fasting and chronic exercise) and mal-adaptive (obesity and related health complications) conditions. Thus, collective metabolic and physiological processes must balance lipid storage and utilization with prevention of lipocytotoxicity and compounding tissue dysfunctions, urging the need to further define the connection of mammalian lipid droplet function and lipid homeostasis. The perilipins are a multi-protein family that targets lipid droplet surfaces and regulates lipid storage and hydrolysis. Study of perilipin functions has provided insight into the physiological roles of cytosolic lipid droplets and their relationship with obesity-related pathologies. Here, we review the current knowledge of the multiple perilipin proteins in regulating tissue-specific lipid droplets and associations with tissue and systemic energetics.
Collapse
|
170
|
Senthivinayagam S, McIntosh AL, Moon KC, Atshaves BP. Plin2 inhibits cellular glucose uptake through interactions with SNAP23, a SNARE complex protein. PLoS One 2013; 8:e73696. [PMID: 24040030 PMCID: PMC3765312 DOI: 10.1371/journal.pone.0073696] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 07/25/2013] [Indexed: 11/19/2022] Open
Abstract
Although a link between excess lipid storage and aberrant glucose metabolism has been recognized for many years, little is known what role lipid storage droplets and associated proteins such as Plin2 play in managing cellular glucose levels. To address this issue, the influence of Plin2 on glucose uptake was examined using 2-NBD-Glucose and [(3)H]-2-deoxyglucose to show that insulin-mediated glucose uptake was decreased 1.7- and 1.8-fold, respectively in L cell fibroblasts overexpressing Plin2. Conversely, suppression of Plin2 levels by RNAi-mediated knockdown increased 2-NBD-Glucose uptake several fold in transfected L cells and differentiated 3T3-L1 cells. The effect of Plin2 expression on proteins involved in glucose uptake and transport was also examined. Expression of the SNARE protein SNAP23 was increased 1.6-fold while levels of syntaxin-5 were decreased 1.7-fold in Plin2 overexpression cells with no significant changes observed in lipid droplet associated proteins Plin1 or FSP27 or with the insulin receptor, GLUT1, or VAMP4. FRET experiments revealed a close proximity of Plin2 to SNAP23 on lipid droplets to within an intramolecular distance of 51 Å. The extent of targeting of SNAP23 to lipid droplets was determined by co-localization and co-immunoprecipitation experiments to show increased partitioning of SNAP23 to lipid droplets when Plin2 was overexpressed. Taken together, these results suggest that Plin2 inhibits glucose uptake by interacting with, and regulating cellular targeting of SNAP23 to lipid droplets. In summary, the current study for the first time provides direct evidence for the role of Plin2 in mediating cellular glucose uptake.
Collapse
Affiliation(s)
- Subramanian Senthivinayagam
- Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, United States of America
| | - Avery L. McIntosh
- Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas, United States of America
| | - Kenneth C. Moon
- Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, United States of America
| | - Barbara P. Atshaves
- Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, United States of America
- * E-mail:
| |
Collapse
|
171
|
Crunk AE, Monks J, Murakami A, Jackman M, MacLean PS, Ladinsky M, Bales ES, Cain S, Orlicky DJ, McManaman JL. Dynamic regulation of hepatic lipid droplet properties by diet. PLoS One 2013; 8:e67631. [PMID: 23874434 PMCID: PMC3708958 DOI: 10.1371/journal.pone.0067631] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 05/20/2013] [Indexed: 12/21/2022] Open
Abstract
Cytoplasmic lipid droplets (CLD) are organelle-like structures that function in neutral lipid storage, transport and metabolism through the actions of specific surface-associated proteins. Although diet and metabolism influence hepatic CLD levels, how they affect CLD protein composition is largely unknown. We used non-biased, shotgun, proteomics in combination with metabolic analysis, quantitative immunoblotting, electron microscopy and confocal imaging to define the effects of low- and high-fat diets on CLD properties in fasted-refed mice. We found that the hepatic CLD proteome is distinct from that of CLD from other mammalian tissues, containing enzymes from multiple metabolic pathways. The hepatic CLD proteome is also differentially affected by dietary fat content and hepatic metabolic status. High fat feeding markedly increased the CLD surface density of perilipin-2, a critical regulator of hepatic neutral lipid storage, whereas it reduced CLD levels of betaine-homocysteine S-methyltransferase, an enzyme regulator of homocysteine levels linked to fatty liver disease and hepatocellular carcinoma. Collectively our data demonstrate that the hepatic CLD proteome is enriched in metabolic enzymes, and that it is qualitatively and quantitatively regulated by diet and metabolism. These findings implicate CLD in the regulation of hepatic metabolic processes, and suggest that their properties undergo reorganization in response to hepatic metabolic demands.
Collapse
Affiliation(s)
- Amanda E. Crunk
- Graduate Program of Molecular Biology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- Division of Basic Reproductive Sciences, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Jenifer Monks
- Division of Basic Reproductive Sciences, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Aya Murakami
- Graduate Program of Molecular Biology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Matthew Jackman
- Division of Endocrinology and Metabolism, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- The Center for Human Nutrition, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- The Colorado Obesity Research Initiative, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Paul S. MacLean
- Division of Endocrinology and Metabolism, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- The Center for Human Nutrition, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- The Colorado Obesity Research Initiative, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Mark Ladinsky
- The Boulder Laboratory for 3D Electron Microscopy, University of Colorado Boulder, Boulder Colorado, United States of America
| | - Elise S. Bales
- Division of Basic Reproductive Sciences, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Shannon Cain
- The Colorado Obesity Research Initiative, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - David J. Orlicky
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - James L. McManaman
- Graduate Program of Molecular Biology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- Division of Basic Reproductive Sciences, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- The Center for Human Nutrition, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- The Colorado Obesity Research Initiative, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|