1
|
Griseti E, Bello AA, Bieth E, Sabbagh B, Iacovoni JS, Bigay J, Laurell H, Čopič A. Molecular mechanisms of perilipin protein function in lipid droplet metabolism. FEBS Lett 2024; 598:1170-1198. [PMID: 38140813 DOI: 10.1002/1873-3468.14792] [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: 10/21/2023] [Revised: 11/27/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
Perilipins are abundant lipid droplet (LD) proteins present in all metazoans and also in Amoebozoa and fungi. Humans express five perilipins, which share a similar domain organization: an amino-terminal PAT domain and an 11-mer repeat region, which can fold into amphipathic helices that interact with LDs, followed by a structured carboxy-terminal domain. Variations of this organization that arose during vertebrate evolution allow for functional specialization between perilipins in relation to the metabolic needs of different tissues. We discuss how different features of perilipins influence their interaction with LDs and their cellular targeting. PLIN1 and PLIN5 play a direct role in lipolysis by regulating the recruitment of lipases to LDs and LD interaction with mitochondria. Other perilipins, particularly PLIN2, appear to protect LDs from lipolysis, but the molecular mechanism is not clear. PLIN4 stands out with its long repetitive region, whereas PLIN3 is most widely expressed and is used as a nascent LD marker. Finally, we discuss the genetic variability in perilipins in connection with metabolic disease, prominent for PLIN1 and PLIN4, underlying the importance of understanding the molecular function of perilipins.
Collapse
Affiliation(s)
- Elena Griseti
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, Université de Toulouse, Inserm, Université Toulouse III - Paul Sabatier (UPS), France
| | - Abdoul Akim Bello
- Institut de Pharmacologie Moléculaire et Cellulaire - IPMC, Université Côte d'Azur, CNRS, Valbonne, France
| | - Eric Bieth
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, Université de Toulouse, Inserm, Université Toulouse III - Paul Sabatier (UPS), France
- Departement de Génétique Médicale, Centre Hospitalier Universitaire de Toulouse, France
| | - Bayane Sabbagh
- Centre de Recherche en Biologie Cellulaire de Montpellier - CRBM, Université de Montpellier, CNRS, France
| | - Jason S Iacovoni
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, Université de Toulouse, Inserm, Université Toulouse III - Paul Sabatier (UPS), France
| | - Joëlle Bigay
- Institut de Pharmacologie Moléculaire et Cellulaire - IPMC, Université Côte d'Azur, CNRS, Valbonne, France
| | - Henrik Laurell
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, Université de Toulouse, Inserm, Université Toulouse III - Paul Sabatier (UPS), France
| | - Alenka Čopič
- Centre de Recherche en Biologie Cellulaire de Montpellier - CRBM, Université de Montpellier, CNRS, France
| |
Collapse
|
2
|
Engin AB. Mechanism of Obesity-Related Lipotoxicity and Clinical Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:131-166. [PMID: 39287851 DOI: 10.1007/978-3-031-63657-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The link between cellular exposure to fatty acid species and toxicity phenotypes remains poorly understood. However, structural characterization and functional profiling of human plasma free fatty acids (FFAs) analysis has revealed that FFAs are located either in the toxic cluster or in the cluster that is transcriptionally responsive to lipotoxic stress and creates genetic risk factors. Genome-wide short hairpin RNA screen has identified more than 350 genes modulating lipotoxicity. Hypertrophic adipocytes in obese adipose are both unable to expand further to store excess lipids in the diet and are resistant to the antilipolytic action of insulin. In addition to lipolysis, the inability of packaging the excess lipids into lipid droplets causes circulating fatty acids to reach toxic levels in non-adipose tissues. Deleterious effects of accumulated lipid in non-adipose tissues are known as lipotoxicity. Although triglycerides serve a storage function for long-chain non-esterified fatty acid and their products such as ceramide and diacylglycerols (DAGs), overloading of palmitic acid fraction of saturated fatty acids (SFAs) raises ceramide levels. The excess DAG and ceramide load create harmful effects on multiple organs and systems, inducing chronic inflammation in obesity. Thus, lipotoxic inflammation results in β cells death and pancreatic islets dysfunction. Endoplasmic reticulum stress stimuli induce lipolysis by activating cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) and extracellular signal-regulated kinase (Erk) 1/2 signaling in adipocytes. However, palmitic acid-induced endoplasmic reticulum stress-c-Jun N-terminal kinase (JNK)-autophagy axis in hypertrophic adipocytes is a pro-survival mechanism against endoplasmic reticulum stress and cell death induced by SFAs. Endoplasmic reticulum-localized acyl-coenzyme A (CoA): glycerol-3-phosphate acyltransferase (GPAT) enzymes are mediators of lipotoxicity, and inhibiting these enzymes has therapeutic potential for lipotoxicity. Lipotoxicity increases the number of autophagosomes, which engulf palmitic acid, and thus suppress the autophagic turnover. Fatty acid desaturation promotes palmitate detoxification and storages into triglycerides. As therapeutic targets of glucolipotoxicity, in addition to caloric restriction and exercise, there are four different pharmacological approaches, which consist of metformin, glucagon-like peptide 1 (GLP-1) receptor agonists, peroxisome proliferator-activated receptor-gamma (PPARγ) ligands thiazolidinediones, and chaperones are still used in clinical practice. Furthermore, induction of the brown fat-like phenotype with the mixture of eicosapentanoic acid and docosahexaenoic acid appears as a potential therapeutic application for treatment of lipotoxicity.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| |
Collapse
|
3
|
Janilkarn-Urena I, Idrissova A, Zhang M, VanDreal M, Sanghavi N, Skinner SG, Cheng S, Zhang Z, Watanabe J, Asatryan L, Cadenas E, Davies DL. Dihydromyricetin supplementation improves ethanol-induced lipid accumulation and inflammation. Front Nutr 2023; 10:1201007. [PMID: 37680900 PMCID: PMC10481966 DOI: 10.3389/fnut.2023.1201007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 08/01/2023] [Indexed: 09/09/2023] Open
Abstract
Introduction Excessive alcohol consumption leads to a myriad of detrimental health effects, including alcohol-associated liver disease (ALD). Unfortunately, no available treatments exist to combat the progression of ALD beyond corticosteroid administration and/or liver transplants. Dihydromyricetin (DHM) is a bioactive polyphenol and flavonoid that has traditionally been used in Chinese herbal medicine for its robust antioxidant and anti-inflammatory properties. It is derived from many plants, including Hovenia dulcis and is found as the active ingredient in a variety of popular hangover remedies. Investigations utilizing DHM have demonstrated its ability to alleviate ethanol-induced disruptions in mitochondrial and lipid metabolism, while demonstrating hepatoprotective activity. Methods Female c57BL/6J mice (n = 12/group) were treated using the Lieber DeCarli forced-drinking and ethanol (EtOH) containing liquid diet, for 5 weeks. Mice were randomly divided into three groups: (1) No-EtOH, (2) EtOH [5% (v/v)], and (3) EtOH [5% (v/v)] + DHM (6 mg/mL). Mice were exposed to ethanol for 2 weeks to ensure the development of ALD pathology prior to receiving dihydromyricetin supplementation. Statistical analysis included one-way ANOVA along with Bonferroni multiple comparison tests, where p ≤ 0.05 was considered statistically significant. Results Dihydromyricetin administration significantly improved aminotransferase levels (AST/ALT) and reduced levels of circulating lipids including LDL/VLDL, total cholesterol (free cholesterol), and triglycerides. DHM demonstrated enhanced lipid clearance by way of increased lipophagy activity, shown as the increased interaction and colocalization of p62/SQSTM-1, LC3B, and PLIN-1 proteins. DHM-fed mice had increased hepatocyte-to-hepatocyte lipid droplet (LD) heterogeneity, suggesting increased neutralization and sequestration of free lipids into LDs. DHM administration significantly reduced prominent pro-inflammatory cytokines commonly associated with ALD pathology such as TNF-α, IL-6, and IL-17. Discussion Dihydromyricetin is commercially available as a dietary supplement. The results of this proof-of-concept study demonstrate its potential utility and functionality as a cost-effective and safe candidate to combat inflammation and the progression of ALD pathology.
Collapse
Affiliation(s)
- Isis Janilkarn-Urena
- Titus Family Department of Clinical Pharmacy, University of Southern California Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, United States
| | - Alina Idrissova
- Titus Family Department of Clinical Pharmacy, University of Southern California Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, United States
| | - Mindy Zhang
- Titus Family Department of Clinical Pharmacy, University of Southern California Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, United States
| | - Masha VanDreal
- Titus Family Department of Clinical Pharmacy, University of Southern California Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, United States
| | - Neysa Sanghavi
- Titus Family Department of Clinical Pharmacy, University of Southern California Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, United States
| | - Samantha G. Skinner
- Titus Family Department of Clinical Pharmacy, University of Southern California Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, United States
| | - Sydney Cheng
- Titus Family Department of Clinical Pharmacy, University of Southern California Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, United States
| | - Zeyu Zhang
- Titus Family Department of Clinical Pharmacy, University of Southern California Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, United States
- Translational Research Lab, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, United States
| | - Junji Watanabe
- Translational Research Lab, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, United States
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Liana Asatryan
- Titus Family Department of Clinical Pharmacy, University of Southern California Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, United States
| | - Enrique Cadenas
- Titus Family Department of Clinical Pharmacy, University of Southern California Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, United States
| | - Daryl L. Davies
- Titus Family Department of Clinical Pharmacy, University of Southern California Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, United States
| |
Collapse
|
4
|
Bombarda-Rocha V, Silva D, Badr-Eddine A, Nogueira P, Gonçalves J, Fresco P. Challenges in Pharmacological Intervention in Perilipins (PLINs) to Modulate Lipid Droplet Dynamics in Obesity and Cancer. Cancers (Basel) 2023; 15:4013. [PMID: 37568828 PMCID: PMC10417315 DOI: 10.3390/cancers15154013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Perilipins (PLINs) are the most abundant proteins in lipid droplets (LD). These LD-associated proteins are responsible for upgrading LD from inert lipid storage structures to fully functional organelles, fundamentally integrated in the lipid metabolism. There are five distinct perilipins (PLIN1-5), each with specific expression patterns and metabolic activation, but all capable of regulating the activity of lipases on LD. This plurality creates a complex orchestrated mechanism that is directly related to the healthy balance between lipogenesis and lipolysis. Given the essential role of PLINs in the modulation of the lipid metabolism, these proteins can become interesting targets for the treatment of lipid-associated diseases. Since reprogrammed lipid metabolism is a recognized cancer hallmark, and obesity is a known risk factor for cancer and other comorbidities, the modulation of PLINs could either improve existing treatments or create new opportunities for the treatment of these diseases. Even though PLINs have not been, so far, directly considered for pharmacological interventions, there are many established drugs that can modulate PLINs activity. Therefore, the aim of this study is to assess the involvement of PLINs in diseases related to lipid metabolism dysregulation and whether PLINs can be viewed as potential therapeutic targets for cancer and obesity.
Collapse
Affiliation(s)
- Victória Bombarda-Rocha
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Dany Silva
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Allal Badr-Eddine
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
| | - Patrícia Nogueira
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Jorge Gonçalves
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Paula Fresco
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
5
|
Hammoudeh N, Soukkarieh C, Murphy DJ, Hanano A. Mammalian lipid droplets: structural, pathological, immunological and anti-toxicological roles. Prog Lipid Res 2023; 91:101233. [PMID: 37156444 DOI: 10.1016/j.plipres.2023.101233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
Mammalian lipid droplets (LDs) are specialized cytosolic organelles consisting of a neutral lipid core surrounded by a membrane made up of a phospholipid monolayer and a specific population of proteins that varies according to the location and function of each LD. Over the past decade, there have been significant advances in the understanding of LD biogenesis and functions. LDs are now recognized as dynamic organelles that participate in many aspects of cellular homeostasis plus other vital functions. LD biogenesis is a complex, highly-regulated process with assembly occurring on the endoplasmic reticulum although aspects of the underpinning molecular mechanisms remain elusive. For example, it is unclear how many enzymes participate in the biosynthesis of the neutral lipid components of LDs and how this process is coordinated in response to different metabolic cues to promote or suppress LD formation and turnover. In addition to enzymes involved in the biosynthesis of neutral lipids, various scaffolding proteins play roles in coordinating LD formation. Despite their lack of ultrastructural diversity, LDs in different mammalian cell types are involved in a wide range of biological functions. These include roles in membrane homeostasis, regulation of hypoxia, neoplastic inflammatory responses, cellular oxidative status, lipid peroxidation, and protection against potentially toxic intracellular fatty acids and lipophilic xenobiotics. Herein, the roles of mammalian LDs and their associated proteins are reviewed with a particular focus on their roles in pathological, immunological and anti-toxicological processes.
Collapse
Affiliation(s)
- Nour Hammoudeh
- Department of Animal Biology, Faculty of Sciences, University of Damascus, Damascus, Syria
| | - Chadi Soukkarieh
- Department of Animal Biology, Faculty of Sciences, University of Damascus, Damascus, Syria
| | - Denis J Murphy
- School of Applied Sciences, University of South Wales, Pontypridd, CF37 1DL, Wales, United Kingdom..
| | - Abdulsamie Hanano
- Department of Molecular Biology and Biotechnology, Atomic Energy Commission of Syria (AECS), P.O. Box 6091, Damascus, Syria..
| |
Collapse
|
6
|
Abstract
SQSTM1/p62 (sequestosome 1) is a well-established indicator of macroautophagic/autophagic flux. It was initially characterized as the ubiquitin-binding autophagic receptor in aggrephagy, the selective autophagy of ubiquitinated protein aggregates. Recently, several studies correlated its levels with the abundance of intracellular lipid droplets (LDs). In the absence of a bona fide receptor for the selective autophagy of LDs (lipophagy), a few studies demonstrated the role of SQSTM1 in lipophagy. Our analysis of these studies shows that SQSTM1 colocalizes with LDs, bridges them with phagophores, is co-degraded with them in the lysosomes, and affects LD abundance in a variety of cells and under diverse experimental conditions. Although only one study reported all these functions together, the overwhelming and complementary evidence from other studies suggests that the role of SQSTM1 in lipophagy via tagging, movement, aggregation/clustering and sequestration of LDs is rather a common phenomenon in mammalian cells. As ubiquitination of the LD-associated proteins under stress conditions is increasingly recognized as another common phenomenon, some other ubiquitin-binding autophagic receptors, such as NBR1 and OPTN, might soon join SQSTM1 on a list of the non-exclusive lipophagy receptors.Abbreviations: LD: lipid droplet; LIR: LC3-interacting region; PAT: Perilipin, ADRP and TIP47 domain; SAR: selective autophagy receptor.
Collapse
Affiliation(s)
- Ankit Shroff
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Taras Y. Nazarko
- Department of Biology, Georgia State University, Atlanta, GA, USA,CONTACT Taras Y. Nazarko Department of Biology, Georgia State University, P.O. Box 4010, Atlanta, GA30303-4010, USA
| |
Collapse
|
7
|
Yang S, Liu T, Hu C, Li W, Meng Y, Li H, Song C, He C, Zhou Y, Fan Y. Ginsenoside Compound K Protects against Obesity through Pharmacological Targeting of Glucocorticoid Receptor to Activate Lipophagy and Lipid Metabolism. Pharmaceutics 2022; 14:pharmaceutics14061192. [PMID: 35745765 PMCID: PMC9231161 DOI: 10.3390/pharmaceutics14061192] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 12/10/2022] Open
Abstract
(1) Background: The glucocorticoid receptor (GR) plays a key role in lipid metabolism, but investigations of GR activation as a potential therapeutic approach have been hampered by a lack of selective agonists. Ginsenoside compound K (CK) is natural small molecule with a steroid-like structure that offers a variety of therapeutic benefits. Our study validates CK as a novel GR agonist for the treatment of obesity. (2) Methods: By using pulldown and RNA interference, we determined that CK binds to GR. The anti-obesity potential effects of CK were investigated in obese mice, including through whole-body energy homeostasis, glucose and insulin tolerance, and biochemical and proteomic analysis. Using chromatin immunoprecipitation, we identified GR binding sites upstream of lipase ATGL. (3) Results: We demonstrated that CK reduced the weight and blood lipids of mice more significantly than the drug Orlistat. Proteomics data showed that CK up-regulated autophagy regulatory proteins, enhanced fatty acid oxidation proteins, and decreased fatty acid synthesis proteins. CK induced lipophagy with the initial formation of the phagophore via AMPK/ULK1 activation. However, a blockade of autophagy did not disturb the increase in CK on lipase expression, suggesting that autophagy and lipase are independent pathways in the function of CK. The pulldown and siRNA experiments showed that GR is the critical target. After binding to GR, CK not only activated lipophagy, but also promoted the binding of GR to the ATGL promoter. (4) Conclusions: Our findings indicate that CK is a natural food candidate for reducing fat content and weight.
Collapse
Affiliation(s)
- Siwen Yang
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
| | - Ting Liu
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
| | - Chenxing Hu
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
| | - Weili Li
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
| | - Yuhan Meng
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
| | - Haiyang Li
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
| | - Chengcheng Song
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
| | - Congcong He
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Yifa Zhou
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
- Correspondence: (Y.Z.); (Y.F.)
| | - Yuying Fan
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
- Correspondence: (Y.Z.); (Y.F.)
| |
Collapse
|
8
|
Xu Z, Huang J, Kong D, Yang Y, Guo L, Jia X, Zhong G, Liu Z. Potent half-sandwich Ru(Ⅱ) N^N (aryl-BIAN) complexes: Lysosome-mediated apoptosis, in vitro and in vivo anticancer activities. Eur J Med Chem 2020; 207:112763. [PMID: 32882612 DOI: 10.1016/j.ejmech.2020.112763] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/09/2020] [Accepted: 08/15/2020] [Indexed: 12/22/2022]
Abstract
Herein a new series of organometallic half-sandwich Ru(Ⅱ) complexes bearing aryl-BIAN chelating ligands with various electron-withdrawing and electron-donating substituents have been developed as theranostic agents. All the complexes display much higher anti-proliferative potency than the clinical chemotherapeutic drug cisplatin towards seven cancer cell lines. The anti-proliferative efficacy of these complexes is correlated to their electron-withdrawing ability. Interestingly, complex Ru1 also potently suppresses cancer cell migration in vitro and effectively inhibit tumor growth in vivo in a CT26 colon cancer mouse xenograft model. Mechanisms of action studies display that Ru1 can favorably accumulate in lysosome and exerts anti-cancer potency by inducing a series of events related to lysosomal dysfunction in CT26 cells. Interestingly, inhibition of lysosomal enzymes leads to suppression of cytotoxicity and apoptosis induced by Ru1. Our results elucidate that complex Ru1 can elicit cytotoxicity through lysosome-mediated apoptosis in vitro and suppress tumor growth in vivo.
Collapse
Affiliation(s)
- Zhishan Xu
- College of Chemistry, Chemistry Engineering and Materials Science, Shandong Normal University, Jinan, 250014, China; Institute of Anticancer Agents Development and Theranostic Application, The Key Laboratory of Life-Organic Analysis and Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine, Department of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, China
| | - Jie Huang
- Qingdao University of Science and Technology, Qingdao, 266061, China.
| | - Deliang Kong
- Institute of Anticancer Agents Development and Theranostic Application, The Key Laboratory of Life-Organic Analysis and Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine, Department of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, China
| | - Yuliang Yang
- Institute of Anticancer Agents Development and Theranostic Application, The Key Laboratory of Life-Organic Analysis and Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine, Department of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, China
| | - Lihua Guo
- Institute of Anticancer Agents Development and Theranostic Application, The Key Laboratory of Life-Organic Analysis and Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine, Department of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, China
| | - Xianglei Jia
- Henan Key Laboratory of Neural Regeneration, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, China
| | - Genshen Zhong
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Zhe Liu
- Institute of Anticancer Agents Development and Theranostic Application, The Key Laboratory of Life-Organic Analysis and Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine, Department of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, China.
| |
Collapse
|
9
|
Yadati T, Houben T, Bitorina A, Shiri-Sverdlov R. The Ins and Outs of Cathepsins: Physiological Function and Role in Disease Management. Cells 2020; 9:cells9071679. [PMID: 32668602 PMCID: PMC7407943 DOI: 10.3390/cells9071679] [Citation(s) in RCA: 262] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 12/14/2022] Open
Abstract
Cathepsins are the most abundant lysosomal proteases that are mainly found in acidic endo/lysosomal compartments where they play a vital role in intracellular protein degradation, energy metabolism, and immune responses among a host of other functions. The discovery that cathepsins are secreted and remain functionally active outside of the lysosome has caused a paradigm shift. Contemporary research has unraveled many versatile functions of cathepsins in extralysosomal locations including cytosol and extracellular space. Nevertheless, extracellular cathepsins are majorly upregulated in pathological states and are implicated in a wide range of diseases including cancer and cardiovascular diseases. Taking advantage of the differential expression of the cathepsins during pathological conditions, much research is focused on using cathepsins as diagnostic markers and therapeutic targets. A tailored therapeutic approach using selective cathepsin inhibitors is constantly emerging to be safe and efficient. Moreover, recent development of proteomic-based approaches for the identification of novel physiological substrates offers a major opportunity to understand the mechanism of cathepsin action. In this review, we summarize the available evidence regarding the role of cathepsins in health and disease, discuss their potential as biomarkers of disease progression, and shed light on the potential of extracellular cathepsin inhibitors as safe therapeutic tools.
Collapse
|
10
|
Gopal T, Kumar N, Perriotte-Olson C, Casey CA, Donohue TM, Harris EN, Talmon G, Kabanov AV, Saraswathi V. Nanoformulated SOD1 ameliorates the combined NASH and alcohol-associated liver disease partly via regulating CYP2E1 expression in adipose tissue and liver. Am J Physiol Gastrointest Liver Physiol 2020; 318:G428-G438. [PMID: 31928222 PMCID: PMC7099493 DOI: 10.1152/ajpgi.00217.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Enhanced free fatty acid (FFA) flux from adipose tissue (AT) to liver plays an important role in the development of nonalcoholic steatohepatitis (NASH) and alcohol-associated liver disease (AALD). We determined the effectiveness of nanoformulated superoxide dismutase 1 (Nano) in attenuating liver injury in a mouse model exhibiting a combination of NASH and AALD. Male C57BL6/J mice were fed a chow diet (CD) or a high-fat diet (HF) for 10 wk followed by pair feeding of the Lieber-DeCarli control (control) or ethanol (ET) diet for 4 wk. Nano was administered once every other day for the last 2 wk of ET feeding. Mice were divided into 1) CD + control diet (CD + Cont), 2) high-fat diet (HF) + control diet (HF + Cont), 3) HF + Cont + Nano, 4) HF + ET diet (HF + ET), and 5) HF + ET + Nano. The total fat mass, visceral AT mass (VAT), and VAT perilipin 1 content were significantly lower only in HF + ET-fed mice but not in HF + ET + Nano-treated mice compared with controls. The HF + ET-fed mice showed an upregulation of VAT CYP2E1 protein, and Nano abrogated this effect. We noted a significant rise in plasma FFAs, ALT, and monocyte chemoattractant protein-1 in HF + ET-fed mice, which was blunted in HF + ET + Nano-treated mice. HF + ET-induced increases in hepatic steatosis and inflammatory markers were attenuated upon Nano treatment. Nano reduced hepatic CYP2E1 and enhanced catalase levels in HF + ET-fed mice with a concomitant increase in SOD1 protein and activity in liver. Nano was effective in attenuating AT and liver injury in mice exhibiting a combination of NASH and AALD, partly via reduced CYP2E1-mediated ET metabolism in these organs.NEW & NOTEWORTHY Increased free fatty acid flux from adipose tissue (AT) to liver accompanied by oxidative stress promotes nonalcoholic steatohepatitis (NASH) and alcohol-associated liver injury (AALD). Obesity increases the severity of AALD. Using a two-hit model involving a high-fat diet and chronic ethanol feeding to mice, and treating them with nanoformulated superoxide dismutase (nanoSOD), we have shown that nanoSOD improves AT lipid storage, reduces CYP2E1 in AT and liver, and attenuates the combined NASH/AALD in mice.
Collapse
Affiliation(s)
- Thiyagarajan Gopal
- 1Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Narendra Kumar
- 1Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Curtis Perriotte-Olson
- 1Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Carol A. Casey
- 2Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska,3Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Terrence M. Donohue
- 2Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska,3Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Edward N. Harris
- 4Department of Biochemistry, University of Nebraska Lincoln, Lincoln, Nebraska
| | - Geoffrey Talmon
- 5Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Alexander V. Kabanov
- 6Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina
| | - Viswanathan Saraswathi
- 1Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska,3Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| |
Collapse
|
11
|
Sun X, Feng X, Wu X, Lu Y, Chen K, Ye Y. Fat Wasting Is Damaging: Role of Adipose Tissue in Cancer-Associated Cachexia. Front Cell Dev Biol 2020; 8:33. [PMID: 32117967 PMCID: PMC7028686 DOI: 10.3389/fcell.2020.00033] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/15/2020] [Indexed: 12/11/2022] Open
Abstract
Loss of body weight, especially loss of adipose tissue and skeletal muscle weight, characterizes cancer-associated cachexia (CAC). Clinically, therapeutic options for CAC are limited due to the complicated signaling between cancer and other organs. Recent research advances show that adipose tissues play a critical role during thermogenesis, glucose homeostasis, insulin sensitivity, and lipid metabolism. Understanding the adipocyte lipolysis, the formation of beige adipocytes, and the activation of brown adipocytes is vital for novel therapies for metabolic syndromes like CAC. The system-level crosstalk between adipose tissue and other organs involves adipocyte lipolysis, white adipose tissue browning, and secreted factors and metabolites. Novel CAC animal models and accumulating molecular signaling knowledge have provided mechanisms that may ultimately be translated into future therapeutic possibilities that benefit CAC patients. This mini review discusses the role of adipose tissue in CAC development, mechanism, and therapy.
Collapse
Affiliation(s)
- Xiaoting Sun
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaogang Feng
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Xiaojing Wu
- Department of Cardiology, Shenzhen University General Hospital, Shenzhen, China
| | - Yongtian Lu
- Department of ENT, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Kaihong Chen
- Department of Cardiology, The Affiliated Longyan First Hospital of Fujian Medical University, Longyan, China
| | - Ying Ye
- Department of Oral Implantology, School and Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| |
Collapse
|
12
|
Cathepsin B overexpression induces degradation of perilipin 1 to cause lipid metabolism dysfunction in adipocytes. Sci Rep 2020; 10:634. [PMID: 31959889 PMCID: PMC6971249 DOI: 10.1038/s41598-020-57428-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 12/24/2019] [Indexed: 02/06/2023] Open
Abstract
Obesity, caused by the dysfunction of white adipose tissue (WAT), is reportedly accompanied by exacerbation of lipolysis. Perilipin 1 (PLIN1), which forms a coat around lipid droplets, interacts with several lipolysis proteins to regulate lipolysis. While it is known that perilipin family proteins are degraded in lysosomes, the underlying molecular mechanisms related to the downregulated expression of PLIN1 in obese WAT remain unknown. Recently, we found that lysosomal dysfunction originating from an abnormality of cathepsin B (CTSB), a lysosomal representative protease, occurs in obese WAT. Therefore, we investigated the effect of CTSB alterations on PLIN1 expression in obese WAT. PLIN1 protein disappeared and CTSB protein appeared in the cytoplasm of adipocytes in the early stage of obese WAT. Overexpression of CTSB reduced PLIN1 protein in 3T3L1 adipocytes, and treatment with a CTSB inhibitor significantly recovered this reduction. In addition, CTSB overexpression induced the dysfunction of lipolysis in 3T3L1 adipocytes. Therefore, we concluded that upregulation of CTSB induced the reduction of PLIN1 protein in obese WAT, resulting in lipolysis dysfunction. This suggests a novel pathology of lipid metabolism involving PLIN1 in adipocytes and that CTSB might be a therapeutic candidate molecule for obese WAT.
Collapse
|
13
|
Nlrp1b1 negatively modulates obesity-induced inflammation by promoting IL-18 production. Sci Rep 2019; 9:13815. [PMID: 31554824 PMCID: PMC6761090 DOI: 10.1038/s41598-019-49546-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 08/22/2019] [Indexed: 02/08/2023] Open
Abstract
Obesity-induced inflammation, triggered by lipid-mediated activation of the Nlrp3 inflammasome, results in glucose metabolism alterations and type 2 diabetes. This knowledge has been generated using animals deficient for any of the different components of this inflammasome (Caspase-1, Asc or Nlrp3) in the C57BL/6 background. Unlike C57BL/6 mice, which carry allele 2 of the Nlrp1b gene (Nlrp1b2), Balb/c mice that carry allele 1 (Nlrp1b1) are less prone to develop alterations in the glucose metabolism when fed with a high fat diet. However, the molecular bases for these metabolic differences are unknown. Here we show that the Nlrp1b1 allele down regulates the adipose tissue inflammatory response attenuating glucose intolerance and insulin resistance in obese C57BL/mice. Our results indicate that the positive effects of the Nlrp1b1 inflammasome on glucose tolerance and insulin sensitivity involve IL-18-mediated effects on lipolysis, pointing out that differential expression of allelic variants of genes coding for inflammasome components might control susceptibility or resistance to develop diabetes in obese individuals.
Collapse
|
14
|
Varghese M, Kimler VA, Ghazi FR, Rathore GK, Perkins GA, Ellisman MH, Granneman JG. Adipocyte lipolysis affects Perilipin 5 and cristae organization at the cardiac lipid droplet-mitochondrial interface. Sci Rep 2019; 9:4734. [PMID: 30894648 PMCID: PMC6426865 DOI: 10.1038/s41598-019-41329-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 03/04/2019] [Indexed: 12/14/2022] Open
Abstract
This study investigated the effects of elevated fatty acid (FA) supply from adipose tissue on the ultrastructure of cardiac lipid droplets (LDs) and the expression and organization of LD scaffold proteins perilipin-2 (PLIN2) and perilipin-5 (PLIN5). Stimulation of adipocyte lipolysis by fasting (24 h) or β3-adrenergic receptor activation by CL316, 243 (CL) increased cardiac triacylglycerol (TAG) levels and LD size, whereas CL treatment also increased LD number. LDs were tightly associated with mitochondria, which was maintained during LD expansion. Electron tomography (ET) studies revealed continuity of LD and smooth endoplasmic reticulum (SER), suggesting interconnections among LDs. Under fed ad libitum conditions, the cristae of mitochondria that apposed LD were mostly organized perpendicularly to the tangent of the LD surface. Fasting significantly reduced, whereas CL treatment greatly increased, the perpendicular alignment of mitochondrial cristae. Fasting and CL treatment strongly upregulated PLIN5 protein and PLIN2 to a lesser extent. Immunofluorescence and immuno-electron microscopy demonstrated strong targeting of PLIN5 to the cardiac LD-mitochondrial interface, but not to the mitochondrial matrix. CL treatment augmented PLIN5 targeting to the LD-mitochondrial interface, whereas PLIN2 was not significantly affected. Together, our results support the concept that the interface between LD and cardiac mitochondria represents an organized and dynamic "metabolic synapse" that is highly responsive to FA trafficking.
Collapse
Affiliation(s)
- Mita Varghese
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Victoria A Kimler
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Fariha R Ghazi
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Gurnoor K Rathore
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Guy A Perkins
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, 92093, USA
| | - James G Granneman
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| |
Collapse
|
15
|
Ju L, Han J, Zhang X, Deng Y, Yan H, Wang C, Li X, Chen S, Alimujiang M, Li X, Fang Q, Yang Y, Jia W. Obesity-associated inflammation triggers an autophagy-lysosomal response in adipocytes and causes degradation of perilipin 1. Cell Death Dis 2019; 10:121. [PMID: 30741926 PMCID: PMC6370809 DOI: 10.1038/s41419-019-1393-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 01/19/2019] [Accepted: 01/25/2019] [Indexed: 12/17/2022]
Abstract
In obesity, adipocytes exhibit high metabolic activity accompanied by an increase in lipid mobilization. Recent findings indicate that autophagy plays an important role in metabolic homeostasis. However, the role of this process in adipocytes remains controversial. Therefore, we performed an overall analysis of the expression profiles of 322 lysosomal/autophagic genes in the omental adipose tissue of lean and obese individuals, and found that among 35 significantly differentially expressed genes, 34 genes were upregulated. A large number of lysosomal/autophagic genes also were upregulated in murine 3T3-L1 adipocytes challenged with tumor necrosis factor α (TNFα) (within 24 h), which is in accordance with increased autophagy flux in adipocytes. SQSTM1/p62, a selective autophagy receptor that recognizes and binds specifically to ubiquitinated proteins, is transcriptionally upregulated upon TNFα stimulation as well. Perilipin 1 (PLIN1), a crucial lipid droplet protein, can be ubiquitinated and interacts with SQSTM1 directly. Thus, TNFα-induced autophagy is a more selective process that signals through SQSTM1 and can selectively degrade PLIN1. Our study indicates that local proinflammatory cytokines in obese adipose tissue impair triglyceride storage via autophagy induction.
Collapse
Affiliation(s)
- Liping Ju
- Shanghai Key Laboratory of Diabetes, Shanghai Institute for Diabetes, Shanghai Clinical Medical Centre of Diabetes, Shanghai Key Clinical Centre of Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai JiaoTong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Junfeng Han
- Shanghai Key Laboratory of Diabetes, Shanghai Institute for Diabetes, Shanghai Clinical Medical Centre of Diabetes, Shanghai Key Clinical Centre of Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai JiaoTong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Xiaoyan Zhang
- Department of Endocrine and Metabolic Diseases, Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.,Department of Endocrinology and Metabolism, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, China
| | - Yujie Deng
- Department of Endocrine and Metabolic Diseases, Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.,Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Han Yan
- Shanghai Key Laboratory of Diabetes, Shanghai Institute for Diabetes, Shanghai Clinical Medical Centre of Diabetes, Shanghai Key Clinical Centre of Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai JiaoTong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Congrong Wang
- Shanghai Key Laboratory of Diabetes, Shanghai Institute for Diabetes, Shanghai Clinical Medical Centre of Diabetes, Shanghai Key Clinical Centre of Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai JiaoTong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Xiaohua Li
- Department of Endocrinology, Seventh People's Hospital of Shanghai University of TCM, Shanghai, 200137, China
| | - Shuqin Chen
- Shanghai Key Laboratory of Diabetes, Shanghai Institute for Diabetes, Shanghai Clinical Medical Centre of Diabetes, Shanghai Key Clinical Centre of Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai JiaoTong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Miriayi Alimujiang
- Shanghai Key Laboratory of Diabetes, Shanghai Institute for Diabetes, Shanghai Clinical Medical Centre of Diabetes, Shanghai Key Clinical Centre of Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai JiaoTong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Xu Li
- Shanghai Key Laboratory of Diabetes, Shanghai Institute for Diabetes, Shanghai Clinical Medical Centre of Diabetes, Shanghai Key Clinical Centre of Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai JiaoTong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Qichen Fang
- Shanghai Key Laboratory of Diabetes, Shanghai Institute for Diabetes, Shanghai Clinical Medical Centre of Diabetes, Shanghai Key Clinical Centre of Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai JiaoTong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Ying Yang
- Shanghai Key Laboratory of Diabetes, Shanghai Institute for Diabetes, Shanghai Clinical Medical Centre of Diabetes, Shanghai Key Clinical Centre of Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai JiaoTong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| | - Weiping Jia
- Shanghai Key Laboratory of Diabetes, Shanghai Institute for Diabetes, Shanghai Clinical Medical Centre of Diabetes, Shanghai Key Clinical Centre of Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai JiaoTong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
16
|
Ju L, Chen S, Alimujiang M, Bai N, Yan H, Fang Q, Han J, Ma X, Yang Y, Jia W. A novel role for Bcl2l13 in promoting beige adipocyte biogenesis. Biochem Biophys Res Commun 2018; 506:485-491. [DOI: 10.1016/j.bbrc.2018.10.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 10/05/2018] [Indexed: 01/27/2023]
|
17
|
Fan J, Wang S, Zhang X, Chen W, Li Y, Yang P, Cao Z, Wang Y, Lu W, Ju D. Quantum Dots Elicit Hepatotoxicity through Lysosome-Dependent Autophagy Activation and Reactive Oxygen Species Production. ACS Biomater Sci Eng 2018; 4:1418-1427. [PMID: 33418671 DOI: 10.1021/acsbiomaterials.7b00824] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Jiajun Fan
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Shaofei Wang
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Xuyao Zhang
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Wei Chen
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Yubin Li
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Ping Yang
- Instrumental Analysis Center, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Zhonglian Cao
- Instrumental Analysis Center, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Yichen Wang
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Weiyue Lu
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Dianwen Ju
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| |
Collapse
|
18
|
HDAC6 Suppresses Age-Dependent Ectopic Fat Accumulation by Maintaining the Proteostasis of PLIN2 in Drosophila. Dev Cell 2017; 43:99-111.e5. [DOI: 10.1016/j.devcel.2017.09.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 05/01/2017] [Accepted: 08/31/2017] [Indexed: 01/05/2023]
|
19
|
Abstract
Enlarged fat cells in obese adipose tissue diminish capacity to store fat and are resistant to the anti-lipolytic effect of insulin. Insulin resistance (IR)-associated S-nitrosylation of insulin-signaling proteins increases in obesity. In accordance with the inhibition of insulin-mediated anti-lipolytic action, plasma free fatty acid (FFA) levels increase. Additionally, endoplasmic reticulum stress stimuli induce lipolysis by activating cyclic adenosine monophosphate/Protein kinase A (cAMP/PKA) and extracellular signal-regulated kinase ½ (ERK1/2) signaling in adipocytes. Failure of packaging of excess lipid into lipid droplets causes chronic elevation of circulating fatty acids, which can reach to toxic levels within non-adipose tissues. Deleterious effects of lipid accumulation in non-adipose tissues are known as lipotoxicity. In fact, triglycerides may also serve a storage function for long-chain non-esterified fatty acids and their products such as ceramides and diacylglycerols (DAGs). Thus, excess DAG, ceramide and saturated fatty acids in obesity can induce chronic inflammation and have harmful effect on multiple organs and systems. In this context, chronic adipose tissue inflammation, mitochondrial dysfunction and IR have been discussed within the scope of lipotoxicity.
Collapse
|
20
|
Sztalryd C, Brasaemle DL. The perilipin family of lipid droplet proteins: Gatekeepers of intracellular lipolysis. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1221-1232. [PMID: 28754637 DOI: 10.1016/j.bbalip.2017.07.009] [Citation(s) in RCA: 397] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 07/18/2017] [Accepted: 07/19/2017] [Indexed: 12/21/2022]
Abstract
Lipid droplets in chordates are decorated by two or more members of the perilipin family of lipid droplet surface proteins. The perilipins sequester lipids by protecting lipid droplets from lipase action. Their relative expression and protective nature is adapted to the balance of lipid storage and utilization in specific cells. Most cells of the body have tiny lipid droplets with perilipins 2 and 3 at the surfaces, whereas specialized fat-storing cells with larger lipid droplets also express perilipins 1, 4, and/or 5. Perilipins 1, 2, and 5 modulate lipolysis by controlling the access of lipases and co-factors of lipases to substrate lipids stored within lipid droplets. Although perilipin 2 is relatively permissive to lipolysis, perilipins 1 and 5 have distinct control mechanisms that are altered by phosphorylation. Here we evaluate recent progress toward understanding functions of the perilipins with a focus on their role in regulating lipolysis and autophagy. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.
Collapse
Affiliation(s)
- Carole Sztalryd
- Department of Medicine, Division of Endocrinology, School of Medicine, University of Maryland, Baltimore, MD, USA; Geriatric Research, Education, and Clinical Center, Baltimore Veterans Affairs Health Care Center, Baltimore, MD, USA.
| | - Dawn L Brasaemle
- Department of Nutritional Sciences and Center for Lipid Research, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
21
|
Kimmel AR, Sztalryd C. The Perilipins: Major Cytosolic Lipid Droplet-Associated Proteins and Their Roles in Cellular Lipid Storage, Mobilization, and Systemic Homeostasis. Annu Rev Nutr 2017; 36:471-509. [PMID: 27431369 DOI: 10.1146/annurev-nutr-071813-105410] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The discovery by Dr. Constantine Londos of perilipin 1, the major scaffold protein at the surface of cytosolic lipid droplets in adipocytes, marked a fundamental conceptual change in the understanding of lipolytic regulation. Focus then shifted from the enzymatic activation of lipases to substrate accessibility, mediated by perilipin-dependent protein sequestration and recruitment. Consequently, the lipid droplet became recognized as a unique, metabolically active cellular organelle and its surface as the active site for novel protein-protein interactions. A new area of investigation emerged, centered on lipid droplets' biology and their role in energy homeostasis. The perilipin family is of ancient origin and has expanded to include five mammalian genes and a growing list of evolutionarily conserved members. Universally, the perilipins modulate cellular lipid storage. This review provides a summary that connects the perilipins to both cellular and whole-body homeostasis.
Collapse
Affiliation(s)
- Alan R Kimmel
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, The National Institutes of Health, Bethesda, Maryland 20892;
| | - Carole Sztalryd
- The Geriatric Research Education and Clinical Center, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland 21201.,Division of Endocrinology, Department of Medicine, School of Medicine, University of Maryland, Baltimore, Maryland 21201;
| |
Collapse
|
22
|
Natori Y, Nasui M, Kihara-Negishi F. Neu1 sialidase interacts with perilipin 1 on lipid droplets and inhibits lipolysis in 3T3-L1 adipocytes. Genes Cells 2017; 22:485-492. [PMID: 28429532 DOI: 10.1111/gtc.12490] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 03/13/2017] [Indexed: 01/08/2023]
Abstract
Fatty acids are stored within adipocytes in lipid droplets (LDs) as triacylglycerol (TG), which is converted to free fatty acid (FFA) and glycerol via lipolysis. Increased plasma FFA levels in obesity are associated with several clinical conditions. We previously found that Neu1 activity is aberrant in the epididymal fat and liver of obese and diabetic mice. Here, we examined involvement of Neu1 in lipolysis in 3T3-L1 adipocytes. Small interfering RNA against Neu1 was introduced into adipocytes, and glycerol concentrations were measured in the culture medium. We then assessed the effects of Neu1 knockdown on lipolytic protein expression and phosphorylation, as well as interactions between perilipin 1 (Plin1) and hormone-sensitive lipase (HSL) after isoproterenol (IS) stimulation. Interactions between Neu1 and Plin1 were analyzed by immunoprecipitation and immunofluorescent imaging using adipocytes transfected with pCMV6-mNeu1-myc-DYKDDDDK (mNeu1DDK). Neu1 knockdown increased glycerol concentrations in culture media and Plin1 phosphorylation in whole lysates of IS-stimulated cells. Neu1 knockdown increased interaction between Plin1 and HSL after IS stimulation whereas that between Neu1 and Plin1 on LD observed under basal conditions was lost. These results suggest that Neu1 inhibits lipolysis induced by β-adrenergic stimulation in adipocytes via interactions with Plin1 on LD.
Collapse
Affiliation(s)
- Yujin Natori
- Department of Life and Health Sciences, School of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Miwako Nasui
- Department of Life and Health Sciences, School of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Fumiko Kihara-Negishi
- Department of Life and Health Sciences, School of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| |
Collapse
|
23
|
Evans TD, Sergin I, Zhang X, Razani B. Target acquired: Selective autophagy in cardiometabolic disease. Sci Signal 2017; 10:eaag2298. [PMID: 28246200 PMCID: PMC5451512 DOI: 10.1126/scisignal.aag2298] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The accumulation of damaged or excess proteins and organelles is a defining feature of metabolic disease in nearly every tissue. Thus, a central challenge in maintaining metabolic homeostasis is the identification, sequestration, and degradation of these cellular components, including protein aggregates, mitochondria, peroxisomes, inflammasomes, and lipid droplets. A primary route through which this challenge is met is selective autophagy, the targeting of specific cellular cargo for autophagic compartmentalization and lysosomal degradation. In addition to its roles in degradation, selective autophagy is emerging as an integral component of inflammatory and metabolic signaling cascades. In this Review, we focus on emerging evidence and key questions about the role of selective autophagy in the cell biology and pathophysiology of metabolic diseases such as obesity, diabetes, atherosclerosis, and steatohepatitis. Essential players in these processes are the selective autophagy receptors, defined broadly as adapter proteins that both recognize cargo and target it to the autophagosome. Additional domains within these receptors may allow integration of information about autophagic flux with critical regulators of cellular metabolism and inflammation. Details regarding the precise receptors involved, such as p62 and NBR1, and their predominant interacting partners are just beginning to be defined. Overall, we anticipate that the continued study of selective autophagy will prove to be informative in understanding the pathogenesis of metabolic diseases and to provide previously unrecognized therapeutic targets.
Collapse
Affiliation(s)
- Trent D Evans
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ismail Sergin
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xiangyu Zhang
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Babak Razani
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
24
|
Amber Light (590 nm) Induces the Breakdown of Lipid Droplets through Autophagy-Related Lysosomal Degradation in Differentiated Adipocytes. Sci Rep 2016; 6:28476. [PMID: 27346059 PMCID: PMC4921916 DOI: 10.1038/srep28476] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/03/2016] [Indexed: 11/08/2022] Open
Abstract
Lipolysis in the adipocytes provides free fatty acids for other tissues in response to the energy demand. With the rapid increase in obesity-related diseases, finding novel stimuli or mechanisms that regulate lipid metabolism becomes important. We examined the effects of visible light (410, 457, 505, 530, 590, and 660 nm) irradiation on lipolysis regulation in adipocytes differentiated from human adipose-derived stem cells (ADSCs). Interestingly, 590 nm (amber) light irradiation significantly reduced the concentration of lipid droplets (LDs). We further investigated the lipolytic signaling pathways that are involved in 590 nm light irradiation-induced breakdown of LDs. Immunoblot analysis revealed that 590 nm light irradiation-induced phosphorylation of hormone-sensitive lipase (HSL) was insufficient to promote reduction of LDs. We observed that 590 nm light irradiation decreased the expression of perilipin 1. We found that 590 nm light irradiation, but not 505 nm, induced conversion of LC3 I to LC3 II, a representative autophagic marker. We further demonstrated that the lysosomal inhibitors leupeptin/NH4Cl inhibited 590 nm light irradiation-induced reduction of LDs in differentiated adipocytes. Our data suggest that 590 nm light irradiation-induced LD breakdown is partially mediated by autophagy-related lysosomal degradation, and can be applied in clinical settings to reduce obesity.
Collapse
|
25
|
Kourjian G, Rucevic M, Berberich MJ, Dinter J, Wambua D, Boucau J, Le Gall S. HIV Protease Inhibitor-Induced Cathepsin Modulation Alters Antigen Processing and Cross-Presentation. THE JOURNAL OF IMMUNOLOGY 2016; 196:3595-607. [PMID: 27009491 DOI: 10.4049/jimmunol.1600055] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/01/2016] [Indexed: 02/06/2023]
Abstract
Immune recognition by T cells relies on the presentation of pathogen-derived peptides by infected cells, but the persistence of chronic infections calls for new approaches to modulate immune recognition. Ag cross-presentation, the process by which pathogen Ags are internalized, degraded, and presented by MHC class I, is crucial to prime CD8 T cell responses. The original degradation of Ags is performed by pH-dependent endolysosomal cathepsins. In this article, we show that HIV protease inhibitors (PIs) prescribed to HIV-infected persons variably modulate cathepsin activities in human APCs, dendritic cells and macrophages, and CD4 T cells, three cell subsets infected by HIV. Two HIV PIs acted in two complementary ways on cathepsin hydrolytic activities: directly on cathepsins and indirectly on their regulators by inhibiting Akt kinase activities, reducing NADPH oxidase 2 activation, and lowering phagolysosomal reactive oxygen species production and pH, which led to enhanced cathepsin activities. HIV PIs modified endolysosomal degradation and epitope production of proteins from HIV and other pathogens in a sequence-dependent manner. They altered cross-presentation of Ags by dendritic cells to epitope-specific T cells and T cell-mediated killing. HIV PI-induced modulation of Ag processing partly changed the MHC self-peptidome displayed by primary human cells. This first identification, to our knowledge, of prescription drugs modifying the regulation of cathepsin activities and the MHC-peptidome may provide an alternate therapeutic approach to modulate immune recognition in immune disease beyond HIV.
Collapse
Affiliation(s)
| | | | | | - Jens Dinter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139
| | - Daniel Wambua
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139
| | - Julie Boucau
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139
| | - Sylvie Le Gall
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139
| |
Collapse
|
26
|
Liu Y, Takahashi Y, Desai N, Zhang J, Serfass JM, Shi YG, Lynch CJ, Wang HG. Bif-1 deficiency impairs lipid homeostasis and causes obesity accompanied by insulin resistance. Sci Rep 2016; 6:20453. [PMID: 26857140 PMCID: PMC4746598 DOI: 10.1038/srep20453] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/04/2016] [Indexed: 12/27/2022] Open
Abstract
Bif-1 is a membrane-curvature inducing protein that is implicated in the regulation of autophagy and tumorigenesis. Here, we report that Bif-1 plays a critical role in regulating lipid catabolism to control the size of lipid droplets and prevent the development of obesity and insulin resistance upon aging or dietary challenge. Our data show that Bif-1 deficiency promotes the expansion of adipose tissue mass without altering food intake or physical activities. While Bif-1 is dispensable for adipose tissue development, its deficiency reduces the basal rate of adipose tissue lipolysis and results in adipocyte hypertrophy upon aging. The importance of Bif-1 in lipid turnover is not limited to adipose tissue since fasting and refeeding-induced lipid droplet clearance is also attenuated by Bif-1 loss in the liver. Interestingly, obesity induced by a high fat-diet or Bif-1 deficiency downregulates the expression of proteins involved in the autophagy-lysosomal pathway, including Atg9a and Lamp1 in the adipose tissue. These findings thus identify Bif-1 as a novel regulator of lipid homeostasis to prevent the pathogenesis of obesity and its associated metabolic complications.
Collapse
Affiliation(s)
- Ying Liu
- Department of Pharmacology, Penn State University College of Medicine, Hershey, PA 17033, USA
| | - Yoshinori Takahashi
- Department of Pediatrics, Penn State University College of Medicine, Hershey, PA 17033, USA
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA 17033, USA
| | - Neelam Desai
- Department of Pediatrics, Penn State University College of Medicine, Hershey, PA 17033, USA
| | - Jun Zhang
- Department of Molecular Physiology, Penn State University College of Medicine, Hershey, PA 17033, USA
| | - Jacob M. Serfass
- Department of Pharmacology, Penn State University College of Medicine, Hershey, PA 17033, USA
| | - Yu-Guang Shi
- Department of Molecular Physiology, Penn State University College of Medicine, Hershey, PA 17033, USA
| | - Christopher J. Lynch
- Department of Molecular Physiology, Penn State University College of Medicine, Hershey, PA 17033, USA
| | - Hong-Gang Wang
- Department of Pharmacology, Penn State University College of Medicine, Hershey, PA 17033, USA
- Department of Pediatrics, Penn State University College of Medicine, Hershey, PA 17033, USA
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
27
|
Lopategi A, López-Vicario C, Alcaraz-Quiles J, García-Alonso V, Rius B, Titos E, Clària J. Role of bioactive lipid mediators in obese adipose tissue inflammation and endocrine dysfunction. Mol Cell Endocrinol 2016; 419:44-59. [PMID: 26433072 DOI: 10.1016/j.mce.2015.09.033] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 09/18/2015] [Accepted: 09/28/2015] [Indexed: 12/14/2022]
Abstract
White adipose tissue is recognized as an active endocrine organ implicated in the maintenance of metabolic homeostasis. However, adipose tissue function, which has a crucial role in the development of obesity-related comorbidities including insulin resistance and non-alcoholic fatty liver disease, is dysregulated in obese individuals. This review explores the physiological functions and molecular actions of bioactive lipids biosynthesized in adipose tissue including sphingolipids and phospholipids, and in particular fatty acids derived from phospholipids of the cell membrane. Special emphasis is given to polyunsaturated fatty acids of the omega-6 and omega-3 families and their conversion to bioactive lipid mediators through the cyclooxygenase and lipoxygenase pathways. The participation of omega-3-derived lipid autacoids in the resolution of adipose tissue inflammation and in the prevention of obesity-associated hepatic complications is also thoroughly discussed.
Collapse
Affiliation(s)
- Aritz Lopategi
- Department of Biochemistry and Molecular Genetics, Hospital Clínic, IDIBAPS, Barcelona 08036, Spain.
| | - Cristina López-Vicario
- Department of Biochemistry and Molecular Genetics, Hospital Clínic, IDIBAPS, Barcelona 08036, Spain
| | - José Alcaraz-Quiles
- Department of Biochemistry and Molecular Genetics, Hospital Clínic, IDIBAPS, Barcelona 08036, Spain
| | - Verónica García-Alonso
- Department of Biochemistry and Molecular Genetics, Hospital Clínic, IDIBAPS, Barcelona 08036, Spain
| | - Bibiana Rius
- Department of Biochemistry and Molecular Genetics, Hospital Clínic, IDIBAPS, Barcelona 08036, Spain
| | - Esther Titos
- Department of Biochemistry and Molecular Genetics, Hospital Clínic, IDIBAPS, Barcelona 08036, Spain; CIBERehd, University of Barcelona, Barcelona 08036, Spain
| | - Joan Clària
- Department of Biochemistry and Molecular Genetics, Hospital Clínic, IDIBAPS, Barcelona 08036, Spain; CIBERehd, University of Barcelona, Barcelona 08036, Spain; Department of Physiological Sciences I, University of Barcelona, Barcelona 08036, Spain.
| |
Collapse
|
28
|
Zhang Y, Han Q, You S, Cao Y, Zhang X, Liu H, Hu L, Liu CF. Rapamycin Promotes the Autophagic Degradation of Oxidized Low-Density Lipoprotein in Human Umbilical Vein Endothelial Cells. J Vasc Res 2015; 52:210-9. [PMID: 26623657 DOI: 10.1159/000441143] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 08/16/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Oxidized low-density lipoprotein (ox-LDL) has been extensively implicated in the initiation of atherosclerosis. Our previous studies reported that ox-LDL could activate autophagy in human umbilical vein endothelial cells (HUVECs). Because of this, subsequent studies were designed to elucidate the possible role of the autophagic inducer, rapamycin, on ox-LDL degradation in endothelial cells. METHODS Intracellular cholesterol content was measured using a tissue total cholesterol assay kit. ox-LDL trafficking within endothelial cells was analyzed by flow cytometry. Levels of proteins involved in the autophagic process, microtubule-associated protein 1 light chain 3 (MAP1-LC3), lysosome-associated membrane protein 1 (LAMP1), Beclin 1 and p62, were assessed by Western blot analysis. RESULTS We discovered that rapamycin could decrease the ox-LDL content in HUVECs at the 3-hour time point. Rapamycin also mediated an obvious increase in Dil-labeled ox-LDL (Dil-ox-LDL)/LC3 and Dil-ox-LDL/LAMP1 co-localization, which was inhibited by 3-methyladenine (3-MA), an autophagic inhibitor. In addition, significant co-localization of LC3 and LAMP1 occurred in cells pretreated with rapamycin. In the presence of rapamycin, p62 levels were reduced, and autophagic flux was enhanced. CONCLUSION These data demonstrate that the activation of the autophagy-lysosome pathway by rapamycin may accelerate ox-LDL degradation.
Collapse
Affiliation(s)
- Yanlin Zhang
- Department of Neurology, Second Affiliated Hospital of Soochow University, Suzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Kaushik S, Cuervo AM. Degradation of lipid droplet-associated proteins by chaperone-mediated autophagy facilitates lipolysis. Nat Cell Biol 2015; 17:759-70. [PMID: 25961502 PMCID: PMC4449813 DOI: 10.1038/ncb3166] [Citation(s) in RCA: 526] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/23/2015] [Indexed: 12/28/2022]
Abstract
Chaperone-mediated autophagy (CMA) selectively degrades a subset of cytosolic proteins in lysosomes. A potent physiological activator of CMA is nutrient deprivation, a condition in which intracellular triglyceride stores or lipid droplets (LDs) also undergo hydrolysis (lipolysis) to generate free fatty acids for energetic purposes. Here we report that the LD-associated proteins perilipin 2 (PLIN2) and perilipin 3 (PLIN3) are CMA substrates and their degradation through CMA precedes lipolysis. In vivo studies revealed that CMA degradation of PLIN2 and PLIN3 was enhanced during starvation, concurrent with elevated levels of cytosolic adipose triglyceride lipase (ATGL) and macroautophagy proteins on LDs. CMA blockage both in cultured cells and mouse liver or expression of CMA-resistant PLINs leads to reduced association of ATGL and macrolipophagy-related proteins with LDs and the subsequent decrease in lipid oxidation and accumulation of LDs. We propose a role for CMA in LD biology and in the maintenance of lipid homeostasis.
Collapse
Affiliation(s)
- Susmita Kaushik
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| |
Collapse
|
30
|
Abstract
Autophagy is a conserved quality-control pathway that degrades cytoplasmic contents in lysosomes. Autophagy degrades lipid droplets through a process termed lipophagy. Starvation and an acute lipid stimulus increase autophagic sequestration of lipid droplets and their degradation in lysosomes. Accordingly, liver-specific deletion of the autophagy gene Atg7 increases hepatic fat content, mimicking the human condition termed nonalcoholic fatty liver disease. In this review, we provide insights into the molecular regulation of lipophagy, discuss fundamental questions related to the mechanisms by which autophagosomes recognize lipid droplets and how ATG proteins regulate membrane curvature for lipid droplet sequestration, and comment on the possibility of cross talk between lipophagy and cytosolic lipases in lipid mobilization. Finally, we discuss the contribution of lipophagy to the pathophysiology of human fatty liver disease. Understanding how lipophagy clears hepatocellular lipid droplets could provide new ways to prevent fatty liver disease, a major epidemic in developed nations.
Collapse
Affiliation(s)
- Nuria Martinez-Lopez
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Rajat Singh
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461
- Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York 10461
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
31
|
Tsai CY, Peh MT, Feng W, Dymock BW, Moore PK. Hydrogen sulfide promotes adipogenesis in 3T3L1 cells. PLoS One 2015; 10:e0119511. [PMID: 25822632 PMCID: PMC4378953 DOI: 10.1371/journal.pone.0119511] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 01/14/2015] [Indexed: 12/30/2022] Open
Abstract
The effect of hydrogen sulfide (H2S) on differentiation of 3T3L1-derived adipocytes was examined. Endogenous H2S was increased after 3T3L1 differentiation. The expression of the H2S-synthesising enzymes, cystathionine γ-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST), was increased in a time-dependent manner during 3T3L1 differentiation. Expression of genes associated with adipogenesis related genes including fatty acid binding protein 4 (FABP4/aP2), a key regulator of this process, was increased by GYY4137 (a slow-releasing H2S donor compound) and sodium hydrosulfide (NaHS, a classical H2S donor) but not by ZYJ1122 or time-expired NaHS. Furthermore expression of these genes were reduced by aminooxyacetic acid (AOAA, CBS inhibitor), DL-propargylglycine (PAG, CSE inhibitor) as well as by CSE small interference RNA (siCSE) and siCBS. The size and number of lipid droplets in mature adipocytes was significantly increased by both GYY4137 and NaHS, which also impaired the ability of CL316,243 (β3-agonist) to promote lipolysis in these cells. In contrast, AOAA and PAG had the opposite effect. Taken together, we show that the H2S-synthesising enzymes CBS, CSE and 3-MST are endogenously expressed during adipogenesis and that both endogenous and exogenous H2S modulate adipogenesis and adipocyte maturation.
Collapse
Affiliation(s)
- Chin-Yi Tsai
- Neurobiology Group, Life Science Institute, Department of Pharmacology, Yong Loo Lin School of Medicine, Singapore, Singapore
- * E-mail:
| | - Meng Teng Peh
- Neurobiology Group, Life Science Institute, Department of Pharmacology, Yong Loo Lin School of Medicine, Singapore, Singapore
| | - Wei Feng
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Brian William Dymock
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Philip Keith Moore
- Neurobiology Group, Life Science Institute, Department of Pharmacology, Yong Loo Lin School of Medicine, Singapore, Singapore
| |
Collapse
|
32
|
Kozusko K, Tsang V, Bottomley W, Cho YH, Gandotra S, Mimmack ML, Lim K, Isaac I, Patel S, Saudek V, O'Rahilly S, Srinivasan S, Greenfield JR, Barroso I, Campbell LV, Savage DB. Clinical and molecular characterization of a novel PLIN1 frameshift mutation identified in patients with familial partial lipodystrophy. Diabetes 2015; 64:299-310. [PMID: 25114292 PMCID: PMC4361744 DOI: 10.2337/db14-0104] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Perilipin 1 is a lipid droplet coat protein predominantly expressed in adipocytes, where it inhibits basal and facilitates stimulated lipolysis. Loss-of-function mutations in the PLIN1 gene were recently reported in patients with a novel subtype of familial partial lipodystrophy, designated as FPLD4. We now report the identification and characterization of a novel heterozygous frameshift mutation affecting the carboxy-terminus (439fs) of perilipin 1 in two unrelated families. The mutation cosegregated with a similar phenotype including partial lipodystrophy, severe insulin resistance and type 2 diabetes, extreme hypertriglyceridemia, and nonalcoholic fatty liver disease in both families. Poor metabolic control despite maximal medical therapy prompted two patients to undergo bariatric surgery, with remarkably beneficial consequences. Functional studies indicated that expression levels of the mutant protein were lower than wild-type protein, and in stably transfected preadipocytes the mutant protein was associated with smaller lipid droplets. Interestingly, unlike the previously reported 398 and 404 frameshift mutants, this variant binds and stabilizes ABHD5 expression but still fails to inhibit basal lipolysis as effectively as wild-type perilipin 1. Collectively, these findings highlight the physiological need for exquisite regulation of neutral lipid storage within adipocyte lipid droplets, as well as the possible metabolic benefits of bariatric surgery in this serious disease.
Collapse
Affiliation(s)
- K Kozusko
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, UK
| | - Vhm Tsang
- CSIR-IGIB, Sukhdev Vihar, Mathura Road, New Delhi, India
| | - W Bottomley
- Wellcome Trust Sanger Institute, Hinxton, University of Cambridge, UK
| | - Y H Cho
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Australia
- Discipline of Paediatrics and Child Health, University of Sydney, Australia
| | - S Gandotra
- CSIR-IGIB, Sukhdev Vihar, Mathura Road, New Delhi, India
| | - M L Mimmack
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, UK
| | - K Lim
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, UK
| | - I Isaac
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, UK
| | - Satish Patel
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, UK
| | - V Saudek
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, UK
| | - S O'Rahilly
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, UK
| | - S Srinivasan
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Australia
| | - J R Greenfield
- Diabetes and Obesity Research Program, Garvan Institute of Medical Research, Sydney, Australia
- Diabetes Centre and Department of Endocrinology, St Vincent's Hospital, Sydney, Australia
| | - I Barroso
- Wellcome Trust Sanger Institute, Hinxton, University of Cambridge, UK
| | - L V Campbell
- Diabetes and Obesity Research Program, Garvan Institute of Medical Research, Sydney, Australia
- Diabetes Centre and Department of Endocrinology, St Vincent's Hospital, Sydney, Australia
| | - D B Savage
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, UK
| |
Collapse
|
33
|
Siraj FM, Natarajan S, Huq MA, Kim YJ, Yang DC. Structural investigation of ginsenoside Rf with PPARγ major transcriptional factor of adipogenesis and its impact on adipocyte. J Ginseng Res 2014; 39:141-7. [PMID: 26045687 PMCID: PMC4452529 DOI: 10.1016/j.jgr.2014.10.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 10/14/2014] [Accepted: 10/16/2014] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Adipocytes, which are the main cellular component of adipose tissue, are the building blocks of obesity. The nuclear hormone receptor PPARγ is a major regulator of adipocyte differentiation and development. Obesity, which is one of the most dangerous yet silent diseases of all time, is fast becoming a critical area of research focus. METHODS In this study, we initially aimed to investigate whether the ginsenoside Rf, a compound that is only present in Panax ginseng Meyer, interacts with PPARγ by molecular docking simulations. After we performed the docking simulation the result has been analyzed with several different software programs, including Discovery Studio, Pymol, Chimera, Ligplus, and Pose View. All of the programs identified the same mechanism of interaction between PPARγ and Rf, at the same active site. To determine the drug-like and biological activities of Rf, we calculate its absorption, distribution, metabolism, excretion, and toxic (ADMET) and prediction of activity spectra for substances (PASS) properties. Considering the results obtained from the computational investigations, the focus was on the in vitro experiments. RESULTS Because the docking simulations predicted the formation of structural bonds between Rf and PPARγ, we also investigated whether any evidence for these bonds could be observed at the cellular level. These experiments revealed that Rf treatment of 3T3-L1 adipocytes downregulated the expression levels of PPARγ and perilipin, and also decreased the amount of lipid accumulated at different doses. CONCLUSION The ginsenoside Rf appears to be promising compound that could prove useful in antiobesity treatments.
Collapse
Affiliation(s)
- Fayeza Md Siraj
- Ginseng Genetic Resource Bank, Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Korea
| | - Sathishkumar Natarajan
- Ginseng Genetic Resource Bank, Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Korea
| | - Md Amdadul Huq
- Ginseng Genetic Resource Bank, Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Korea
| | - Yeon Ju Kim
- Ginseng Genetic Resource Bank, Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Korea
| | - Deok Chun Yang
- Ginseng Genetic Resource Bank, Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Korea
| |
Collapse
|
34
|
Siraj FM, Natarajan S, Kim YJ, Chun Yang D. In silicoscreening of ginsenoside Rh1 with PPARγ andin vitroanalysis on 3T3-L1 cell line. MOLECULAR SIMULATION 2014. [DOI: 10.1080/08927022.2014.970188] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
35
|
Abstract
In adipocytes the hydrolysis of TAG to produce fatty acids and glycerol under fasting conditions or times of elevated energy demands is tightly regulated by neuroendocrine signals, resulting in the activation of lipolytic enzymes. Among the classic regulators of lipolysis, adrenergic stimulation and the insulin-mediated control of lipid mobilisation are the best known. Initially, hormone-sensitive lipase (HSL) was thought to be the rate-limiting enzyme of the first lipolytic step, while we now know that adipocyte TAG lipase is the key enzyme for lipolysis initiation. Pivotal, previously unsuspected components have also been identified at the protective interface of the lipid droplet surface and in the signalling pathways that control lipolysis. Perilipin, comparative gene identification-58 (CGI-58) and other proteins of the lipid droplet surface are currently known to be key regulators of the lipolytic machinery, protecting or exposing the TAG core of the droplet to lipases. The neuroendocrine control of lipolysis is prototypically exerted by catecholaminergic stimulation and insulin-induced suppression, both of which affect cyclic AMP levels and hence the protein kinase A-mediated phosphorylation of HSL and perilipin. Interestingly, in recent decades adipose tissue has been shown to secrete a large number of adipokines, which exert direct effects on lipolysis, while adipocytes reportedly express a wide range of receptors for signals involved in lipid mobilisation. Recently recognised mediators of lipolysis include some adipokines, structural membrane proteins, atrial natriuretic peptides, AMP-activated protein kinase and mitogen-activated protein kinase. Lipolysis needs to be reanalysed from the broader perspective of its specific physiological or pathological context since basal or stimulated lipolytic rates occur under diverse conditions and by different mechanisms.
Collapse
|
36
|
Siraj FM, SathishKumar N, Kim YJ, Kim SY, Yang DC. Ginsenoside F2 possesses anti-obesity activity via binding with PPARγ and inhibiting adipocyte differentiation in the 3T3-L1 cell line. J Enzyme Inhib Med Chem 2014; 30:9-14. [DOI: 10.3109/14756366.2013.871006] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
37
|
Abstract
HIV-associated lipodystrophy syndrome (HALS), comprising metabolic and morphological alterations, is a known side effect of highly active antiretroviral therapy (HAART). Evidence for the role of nutrition in the management of the systemic parameters of HALS is currently limited. In the present paper we review the current knowledge base surrounding HALS, focusing particularly on the role of nutrition in mitigating the systemic parameters of the syndrome. Reported prevalence of HALS was found to vary from 9 to 83 % due to lack of a standardised definition, as well as variations in assessment methods and in the study population used. HALS is associated with both morphological (lipoatrophy, lipohypertrophy) and metabolic (dyslipidaemia, glucose intolerance, diabetes, hypertension, endothelial dysfunction and atherosclerosis) alterations, which may occur singly or in combination, and are associated with an increased risk of CVD. HAART-induced adipocyte inflammation, oxidative stress and macrophage infiltration, as well as altered adipocyte function and mitochondrial toxicity, have been shown to be central to the development of HALS. The adipocyte, therefore, represents a plausible target for treatment. Pharmacological and surgical treatment interventions have shown effect. However, their use is associated with numerous adverse effects and complications. Targeted lifestyle interventions may provide a useful alternative for managing HALS owing to their safety and tolerability. A Mediterranean-style diet has been found to be effective in improving the systemic parameters of HALS. Furthermore, the effects of n-3 PUFA supplementation are encouraging and future randomised controlled trials investigating the beneficial effects of n-3 PUFA in HALS are justified.
Collapse
|
38
|
Wolak T, Sion-Vardi N, Novack V, Greenberg G, Szendro G, Tarnovscki T, Nov O, Shelef I, Paran E, Rudich A. N-terminal rather than full-length osteopontin or its C-terminal fragment is associated with carotid-plaque inflammation in hypertensive patients. Am J Hypertens 2013; 26:326-33. [PMID: 23382482 DOI: 10.1093/ajh/hps043] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Hypertensive patients develop carotid atherosclerotic plaques with enhanced inflammation. Full-length osteopontin (OPN-FL), a multifunctional protein whose levels are elevated in association with atherosclerosis, is cleaved by thrombin and matrix metalloproteinases to form a C-terminal and a putatively biologically active N-terminal fragment (OPN-C, OPN-N, respectively). We conducted a study to examine whether plaque inflammation in hypertensive patients corresponds to the expression of OPN or of its cleaved forms or both. METHODS We collected 42 carotid plaques from 41 consecutive hypertensive patients during carotid endarterectomy. Plaque tissue was used to measure matrix metalloproteinase-12 (MMP-12) and OPN proteins, and for the classification of plaques as showing low- or high-degree inflammation through histological and immunohistochemical evaluation. RESULTS Fifteen highly inflamed plaques and 27 plaques with characteristics of low-grade inflammation were collected. Moderate to heavy staining for OPN characterized 87% of the plaques with high-degree inflammation but only 44% of those with low-degree inflammation, corresponding to the percentages of plaques that were heavily stained for the macrophage marker CD68 (93% versus 26%, respectively, P < 0.01). Western blot analysis showed that the abundance of OPN-FL and OPN-C was comparable in the two groups. However, the abundance of OPN-N was significantly greater in the highly inflamed plaques (median, 3.8 (range, 0.8-7.3) vs. median, 0.9 (range, 0.2-1.5); P = 0.017, respectively). The abundance of MMP-12 was significantly greater in the high- than in the low-degree plaque inflammation group (4.8 (range 1.9-8.8) vs. 1.1 (range 0.3-1.4), respectively; P = 0.03). CONCLUSIONS The N-terminal fragment of osteopontin, rather than OPN-FL or OPN-C, is associated with carotid plaque inflammation in hypertensive patients. Future studies should assess whether targeting OPN cleavage could present a new approach to preventing high-risk carotid plaques.
Collapse
Affiliation(s)
- Talya Wolak
- Hypertension Unit, Soroka University Medical Center and Faculty of Health Sciences, Ben-Gurion University of the Negev, Israel.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Caveolin-1 deficiency leads to increased susceptibility to cell death and fibrosis in white adipose tissue: characterization of a lipodystrophic model. PLoS One 2012. [PMID: 23049990 DOI: 10.1371/journal.pone0046242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Caveolin-1 (CAV1) is an important regulator of adipose tissue homeostasis. In the present study we examined the impact of CAV1 deficiency on the properties of mouse adipose tissue both in vivo and in explant cultures during conditions of metabolic stress. In CAV1(-/-) mice fasting caused loss of adipose tissue mass despite a lack of hormone-sensitive lipase (HSL) phosphorylation. In addition, fasting resulted in increased macrophage infiltration, enhanced deposition of collagen, and a reduction in the level of the lipid droplet protein perilipin A (PLIN1a). Explant cultures of CAV1(-/-) adipose tissue also showed a loss of PLIN1a during culture, enhanced secretion of IL-6, increased release of lactate dehydrogenase, and demonstrated increased susceptibility to cell death upon collagenase treatment. Attenuated PKA-mediated signaling to HSL, loss of PLIN1a and increased secretion of IL-6 were also observed in adipose tissue explants of CAV1(+/+) mice with diet-induced obesity. Together these results suggest that while alterations in adipocyte lipid droplet biology support adipose tissue metabolism in the absence of PKA-mediated pro-lipolytic signaling in CAV1(-/-) mice, the tissue is intrinsically unstable resulting in increased susceptibility to cell death, which we suggest underlies the development of fibrosis and inflammation during periods of metabolic stress.
Collapse
|
40
|
Caveolin-1 deficiency leads to increased susceptibility to cell death and fibrosis in white adipose tissue: characterization of a lipodystrophic model. PLoS One 2012; 7:e46242. [PMID: 23049990 PMCID: PMC3458842 DOI: 10.1371/journal.pone.0046242] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 08/28/2012] [Indexed: 12/31/2022] Open
Abstract
Caveolin-1 (CAV1) is an important regulator of adipose tissue homeostasis. In the present study we examined the impact of CAV1 deficiency on the properties of mouse adipose tissue both in vivo and in explant cultures during conditions of metabolic stress. In CAV1−/− mice fasting caused loss of adipose tissue mass despite a lack of hormone-sensitive lipase (HSL) phosphorylation. In addition, fasting resulted in increased macrophage infiltration, enhanced deposition of collagen, and a reduction in the level of the lipid droplet protein perilipin A (PLIN1a). Explant cultures of CAV1−/− adipose tissue also showed a loss of PLIN1a during culture, enhanced secretion of IL-6, increased release of lactate dehydrogenase, and demonstrated increased susceptibility to cell death upon collagenase treatment. Attenuated PKA-mediated signaling to HSL, loss of PLIN1a and increased secretion of IL-6 were also observed in adipose tissue explants of CAV1+/+ mice with diet-induced obesity. Together these results suggest that while alterations in adipocyte lipid droplet biology support adipose tissue metabolism in the absence of PKA-mediated pro-lipolytic signaling in CAV1−/− mice, the tissue is intrinsically unstable resulting in increased susceptibility to cell death, which we suggest underlies the development of fibrosis and inflammation during periods of metabolic stress.
Collapse
|
41
|
Ogasawara J, Kitadate K, Nishioka H, Fujii H, Sakurai T, Kizaki T, Izawa T, Ishida H, Ohno H. Oligonol-induced Degradation of Perilipin 1 is Regulated through Lysosomal Degradation Machinery. Nat Prod Commun 2012. [DOI: 10.1177/1934578x1200700923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The results obtained from our previous study showed that the addition of a lychee fruit-derived low molecular form of polyphenol, Oligonol, provoked higher levels of lipolytic activity via the degradation of perilipin 1 in primary rat adipocytes. In the current study, we investigated the possible mechanisms by which Oligonol could promote the degradation of perilipin 1 protein. The addition of Oligonol caused the degradation of GFP-tagged perilipin 1 in a time-dependent manner. Meanwhile, the co-addition of Oligonol and NH4Cl, a lysosome inhibitor, failed to promote the degradation of perilipin 1, while the co-addition of Oligonol and MG132, a proteasome inhibitor, induced a reduction in the levels of perilipin 1. These results suggest that the Oligonol-induced degradation of perilipin 1 is regulated via a lysosome-dependent mechanism.
Collapse
Affiliation(s)
- Junetsu Ogasawara
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University, School of Medicine, Tokyo 181-8611, Japan
| | | | | | - Hajime Fujii
- Amino Up Chemical Co., Ltd., Hokkaido 004-0839, Japan
| | - Takuya Sakurai
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University, School of Medicine, Tokyo 181-8611, Japan
| | - Takako Kizaki
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University, School of Medicine, Tokyo 181-8611, Japan
| | - Tetsuya Izawa
- Graduate School of Health and Sports Science, Doshisha University, Kyoto 610-0394, Japan
| | - Hitoshi Ishida
- Third Department of Internal Medicine, Kyorin University, School of Medicine, Tokyo 181-8611, Japan
| | - Hideki Ohno
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University, School of Medicine, Tokyo 181-8611, Japan
| |
Collapse
|
42
|
Nielsen TS, Kampmann U, Nielsen RR, Jessen N, Orskov L, Pedersen SB, Jørgensen JO, Lund S, Møller N. Reduced mRNA and protein expression of perilipin A and G0/G1 switch gene 2 (G0S2) in human adipose tissue in poorly controlled type 2 diabetes. J Clin Endocrinol Metab 2012; 97:E1348-52. [PMID: 22535977 DOI: 10.1210/jc.2012-1159] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
CONTEXT Increased lipolysis and free fatty acid (FFA) levels contribute significantly to the pathogenesis of chronic and acute insulin resistance in type 2 diabetes, but the underlying mechanisms are uncertain. OBJECTIVE Our objective was to test whether increased lipolysis and FFA levels induced by insulin withdrawal are accompanied by increased adipose tissue (AT) contents of adipose triglyceride lipase (ATGL) and/or altered intracellular ATGL regulation. DESIGN AND PARTICIPANTS Nine patients with type 2 diabetes were examined twice in a randomized crossover design after 16 h of 1) hyperglycemia/insulin withdrawal and 2) euglycemia/insulin infusion. Blood samples were drawn and a sc abdominal AT biopsy was obtained. SETTING The study was conducted at a university hospital research unit. RESULTS Circulating glucose (7.2 ± 0.3 vs. 11.2 ± 0.8 mmol/liter) and FFA (0.51 ± 0.05 vs. 0.65 ± 0.04 mmol/liter) were increased and insulin levels decreased after insulin withdrawal. AT ATGL protein tended to be increased (P = 0.075) after insulin withdrawal; by contrast, AT protein and mRNA content of perilipin A (Plin) and G(0)/G(1) switch gene 2 (G0S2), known negative regulators of ATGL activity, were decreased by 20-30% (all P values <0.03). All measured parameters related to hormone-sensitive lipase remained unaffected. CONCLUSIONS We found reduced mRNA and protein content of Plin and G0S2 and borderline increased ATGL protein in sc AT from poorly controlled type 2 diabetic subjects. This suggests that increased ATGL activity may contribute to the elevated lipolysis and circulating FFA levels in acute insulin withdrawal and metabolic dysregulation in type 2 diabetic patients and that this mechanism may be modifiable.
Collapse
Affiliation(s)
- Thomas S Nielsen
- Medical Research Laboratories, Aarhus University Hospital, 8000 Aarhus C, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ovadia H, Haim Y, Nov O, Almog O, Kovsan J, Bashan N, Benhar M, Rudich A. Increased adipocyte S-nitrosylation targets anti-lipolytic action of insulin: relevance to adipose tissue dysfunction in obesity. J Biol Chem 2011; 286:30433-30443. [PMID: 21724851 DOI: 10.1074/jbc.m111.235945] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Protein S-nitrosylation is a reversible protein modification implicated in both physiological and pathophysiological regulation of protein function. In obesity, skeletal muscle insulin resistance is associated with increased S-nitrosylation of insulin-signaling proteins. However, whether adipose tissue is similarly affected in obesity and, if so, what are the causes and functional consequences of increased S-nitrosylation in this tissue are unknown. Total protein S-nitrosylation was increased in intra-abdominal adipose tissue of obese humans and in high fat-fed or leptin-deficient ob/ob mice. Both the insulin receptor β-subunit and Akt were S-nitrosylated, correlating with body weight. Elevated protein and mRNA expression of inducible NO synthase and decreased protein levels of thioredoxin reductase were associated with increased adipose tissue S-nitrosylation. Cultured differentiated pre-adipocyte cell lines exposed to the NO donors S-nitrosoglutathione (GSNO) or S-nitroso-N-acetylpenicillamine exhibited diminished insulin-stimulated phosphorylation of Akt but not of GSK3 nor of insulin-stimulated glucose uptake. Yet the anti-lipolytic action of insulin was markedly impaired in both cultured adipocytes and in mice injected with GSNO prior to administration of insulin. In cells, impaired ability of insulin to diminish phosphorylated PKA substrates in response to isoproterenol suggested impaired insulin-induced activation of PDE3B. Consistently, increased S-nitrosylation of PDE3B was detected in adipose tissue of high fat-fed obese mice. Site-directed mutagenesis revealed that Cys-768 and Cys-1040, two putative sites for S-nitrosylation adjacent to the substrate-binding site of PDE3B, accounted for ∼50% of its GSNO-induced S-nitrosylation. Collectively, PDE3B and the anti-lipolytic action of insulin may constitute novel targets for increased S-nitrosylation of adipose tissue in obesity.
Collapse
Affiliation(s)
- Hilla Ovadia
- Department of Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84103
| | - Yulia Haim
- Department of Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84103
| | - Ori Nov
- Department of Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84103
| | - Orna Almog
- Department of Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84103
| | - Julia Kovsan
- Department of Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84103
| | - Nava Bashan
- Department of Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84103
| | - Moran Benhar
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096
| | - Assaf Rudich
- Department of Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84103; National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84103, Israel.
| |
Collapse
|
44
|
Wang H, Sztalryd C. Oxidative tissue: perilipin 5 links storage with the furnace. Trends Endocrinol Metab 2011; 22:197-203. [PMID: 21632259 PMCID: PMC3122074 DOI: 10.1016/j.tem.2011.03.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 03/25/2011] [Accepted: 03/30/2011] [Indexed: 10/18/2022]
Abstract
Cellular energy homeostasis is a crucial function of oxidative tissues and is altered in obesity, a continuously rising health problem. Lipid droplets (LD) are thought to play a central role in lipid homeostasis by mediating the transient storage of fatty acids in the form of triglyceride, while preventing high levels of toxic lipid intermediates or oxidized lipids that mediate cellular lipotoxicity. Members of the perilipin protein family coating LD surfaces have been found to serve important regulatory and structural functions crucial to the regulation of lipid stores. This review examines the results of studies on one of the newest members of the perilipin family, perilipin 5, which has emerged as a putative key player in LD function in oxidative tissues.
Collapse
Affiliation(s)
- Hong Wang
- The Geriatric Research, Education and Clinical Center, Baltimore Veterans Affairs Health Care Center, and Division of Endocrinology, Department of Medicine, School of Medicine, University of Maryland, Baltimore, Maryland 21201, USA
| | | |
Collapse
|
45
|
Dong H, Czaja MJ. Regulation of lipid droplets by autophagy. Trends Endocrinol Metab 2011; 22:234-40. [PMID: 21419642 PMCID: PMC3118855 DOI: 10.1016/j.tem.2011.02.003] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 02/07/2011] [Accepted: 02/13/2011] [Indexed: 12/11/2022]
Abstract
Autophagy is a lysosomal pathway by which intracellular organelles and proteins are degraded to supply the cell with energy and to maintain cellular homeostasis. Recently, lipid droplets (LDs) have been identified as a substrate for macroautophagy. In addition to the classic pathway of lipid metabolism by cytosolic lipases, LDs are sequestered in autophagosomes that fuse with lysosomes for the breakdown of LD components by lysosomal enzymes. The ability of autophagy to respond to changes in nutrient supply allows the cell to alter LD metabolism to meet the cell's energy demands. Pathophysiological changes in autophagic function can alter cellular lipid metabolism and promote disease states. Autophagy therefore represents a new cellular target for abnormalities in lipid metabolism and accumulation.
Collapse
Affiliation(s)
- Hanqing Dong
- Department of Medicine, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | |
Collapse
|
46
|
Greenberg AS, Coleman RA, Kraemer FB, McManaman JL, Obin MS, Puri V, Yan QW, Miyoshi H, Mashek DG. The role of lipid droplets in metabolic disease in rodents and humans. J Clin Invest 2011; 121:2102-10. [PMID: 21633178 DOI: 10.1172/jci46069] [Citation(s) in RCA: 498] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Lipid droplets (LDs) are intracellular organelles that store neutral lipids within cells. Over the last two decades there has been a dramatic growth in our understanding of LD biology and, in parallel, our understanding of the role of LDs in health and disease. In its simplest form, the LD regulates the storage and hydrolysis of neutral lipids, including triacylglycerol and/or cholesterol esters. It is becoming increasingly evident that alterations in the regulation of LD physiology and metabolism influence the risk of developing metabolic diseases such as diabetes. In this review we provide an update on the role of LD-associated proteins and LDs in metabolic disease.
Collapse
Affiliation(s)
- Andrew S Greenberg
- Obesity and Metabolism Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts 02111, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lampidonis AD, Rogdakis E, Voutsinas GE, Stravopodis DJ. The resurgence of Hormone-Sensitive Lipase (HSL) in mammalian lipolysis. Gene 2011; 477:1-11. [PMID: 21241784 DOI: 10.1016/j.gene.2011.01.007] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 12/22/2010] [Accepted: 01/07/2011] [Indexed: 12/13/2022]
Abstract
The ability to store energy in the form of energy-dense triacylglycerol and to mobilize these stores rapidly during periods of low carbohydrate availability or throughout the strong metabolic demand is a highly conserved process, absolutely essential for survival. In the industrialized world the regulation of this pathway is viewed as an important therapeutic target for disease prevention. Adipose tissue lipolysis is a catabolic process leading to the breakdown of triacylglycerols stored in fat cells, and release of fatty acids and glycerol. Mobilization of adipose tissue fat is mediated by the MGL, HSL and ATGL, similarly functioning enzymes. ATGL initiates lipolysis followed by the actions of HSL on diacylglycerol, and MGL on monoacylglycerol. HSL is regulated by reversible phosphorylation on five critical residues. Phosphorylation alone, however, is not enough to activate HSL. Probably, conformational alterations and a translocation from the cytoplasm to lipid droplets are also involved. In accordance, Perilipin functions as a master regulator of lipolysis, protecting or exposing the triacylglycerol core of a lipid droplet to lipases. The prototype processes of hormonal lipolytic control are the β-adrenergic stimulation and suppression by insulin, both of which affect cytoplasmic cyclic AMP levels. Lipolysis in adipocytes is an important process in the management of body energy reserves. Its deregulation may contribute to the symptoms of type 2 diabetes mellitus and other pathological situations. We, herein, discuss the metabolic regulation and function of lipases mediating mammalian lipolysis with a focus on HSL, quoting newly identified members of the lipolytic proteome.
Collapse
Affiliation(s)
- Antonis D Lampidonis
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Zografou, 157 84 Athens, Greece
| | | | | | | |
Collapse
|
48
|
Hůlková H, Elleder M. Adipocytes participate in storage in α-galactosidase deficiency (Fabry disease). J Inherit Metab Dis 2010; 33 Suppl 3:S297-300. [PMID: 20628902 PMCID: PMC3757258 DOI: 10.1007/s10545-010-9160-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Revised: 06/18/2010] [Accepted: 06/22/2010] [Indexed: 11/06/2022]
Abstract
Ultrastructural and histochemical studies of bioptic and postmortem tissue samples from ten Fabry hemizygotes showed lysosomal storage in adipocytes as a constant feature of the classic phenotype of α-galactosidase (GLA) deficiency. The storage was represented by a crescent-shaped line of storage lysosomes of varying thicknesses restricted to the perinuclear subplasmalemmal area. The ultrastructure of the storage lysosomes was dominated by concentric lipid membranes modified by simultaneous deposition of autofluorescent ceroid. Storage was widely expressed in adipose tissue. The number of storage lysosomes was increased, and the lysosomes were more clustered in adipocytes with less voluminous lipid content. The findings should attract interest to studies of adipose tissue biology in Fabry disease, a topic that has not been studied so far. In terms of cell biology, the observations represent indirect evidence of significant lysosomal turnover of α-galactose lipid conjugates in adipocytes demasked by GLA deficiency. The results extend the thus far limited information on the adipocyte lysosomal system and its participation in lysosomal storage disorders.
Collapse
Affiliation(s)
- Helena Hůlková
- Institute of Inherited Metabolic Disorders, Charles University, 1st Faculty of Medicine and Teaching Hospital, Prague, Czech Republic
| | - Milan Elleder
- Institute of Inherited Metabolic Disorders, Charles University, 1st Faculty of Medicine and Teaching Hospital, Prague, Czech Republic
- Institute of Inherited Metabolic Disorders, Division B, Bldg.D, Ke Karlovu 2, 128 08 Prague, Czech Republic
| |
Collapse
|
49
|
Leroyer S, Vatier C, Kadiri S, Quette J, Chapron C, Capeau J, Antoine B. Glyceroneogenesis is inhibited through HIV protease inhibitor-induced inflammation in human subcutaneous but not visceral adipose tissue. J Lipid Res 2010; 52:207-20. [PMID: 21068005 DOI: 10.1194/jlr.m000869] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Glyceroneogenesis, a metabolic pathway that participates during lipolysis in the recycling of free fatty acids to triglycerides into adipocytes, contributes to the lipid-buffering function of adipose tissue. We investigated whether glyceroneogenesis could be affected by human immunodeficiency virus (HIV) protease inhibitors (PIs) responsible or not for dyslipidemia in HIV-infected patients. We treated explants obtained from subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) depots from lean individuals. We observed that the dyslipidemic PIs nelfinavir, lopinavir and ritonavir, but not the lipid-neutral PI atazanavir, increased lipolysis and decreased glyceroneogenesis, leading to an increased release of fatty acids from SAT but not from VAT. At the same time, dyslipidemic PIs decreased the amount of perilipin and increased interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) secretion in SAT but not in VAT. Parthenolide, an inhibitor of the NFκB pathway, counteracted PI-induced increased inflammation and decreased glyceroneogenesis. IL-6 (100 ng) inhibited the activity of phosphoenolpyruvate carboxykinase, the key enzyme of glyceroneogenesis, in SAT but not in VAT. Our data show that dyslipidemic but not lipid-neutral PIs decreased glyceroneogenesis as a consequence of PI-induced increased inflammation in SAT that could have an affect on adipocytes and/or macrophages. These results add a new link between fat inflammation and increased fatty acids release and suggest a greater sensitivity of SAT than VAT to PI-induced inflammation.
Collapse
|
50
|
PERILIPIN-dependent control of lipid droplet structure and fat storage in Drosophila. Cell Metab 2010; 12:521-32. [PMID: 21035762 DOI: 10.1016/j.cmet.2010.10.001] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2010] [Revised: 08/03/2010] [Accepted: 08/23/2010] [Indexed: 12/31/2022]
Abstract
Lipid droplets are intracellular organelles enriched in adipose tissue that govern the body fat stores of animals. In mammals, members of the evolutionarily conserved PERILIPIN protein family are associated with the lipid droplet surface and participate in lipid homeostasis. Here, we show that Drosophila mutants lacking the PERILIPIN PLIN1 are hyperphagic and suffer from adult-onset obesity. PLIN1 is a central and Janus-faced component of fat metabolism. It provides barrier function to storage lipid breakdown and acts as a key factor of stimulated lipolysis by modulating the access of proteins to the lipid droplet surface. It also shapes lipid droplet structure, transforming unilocular into multilocular fat cells. We generated flies devoid of all PERILIPIN family members and show that they exhibit impaired yet functional body fat regulation. Our data reveal the existence of a basal and possibly ancient lipid homeostasis system.
Collapse
|