Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Biol Chem ; 298(9): 102347, 2022 09.
Article in English | MEDLINE | ID: mdl-35963433

ABSTRACT

Cell death-inducing DNA fragmentation factor-like effector C (CIDEC) expression in adipose tissue positively correlates with insulin sensitivity in obese humans. Further, E186X, a single-nucleotide CIDEC variant is associated with lipodystrophy, hypertriglyceridemia, and insulin resistance. To establish the unknown mechanistic link between CIDEC and maintenance of systemic glucose homeostasis, we generated transgenic mouse models expressing CIDEC (Ad-CIDECtg) and CIDEC E186X variant (Ad-CIDECmut) transgene specifically in the adipose tissue. We found that Ad-CIDECtg but not Ad-CIDECmut mice were protected against high-fat diet-induced glucose intolerance. Furthermore, we revealed the role of CIDEC in lipid metabolism using transcriptomics and lipidomics. Serum triglycerides, cholesterol, and low-density lipoproteins were lower in high-fat diet-fed Ad-CIDECtg mice compared to their littermate controls. Mechanistically, we demonstrated that CIDEC regulates the enzymatic activity of adipose triglyceride lipase via interacting with its activator, CGI-58, to reduce free fatty acid release and lipotoxicity. In addition, we confirmed that CIDEC is indeed a vital regulator of lipolysis in adipose tissue of obese humans, and treatment with recombinant CIDEC decreased triglyceride breakdown in visceral human adipose tissue. Our study unravels a central pathway whereby adipocyte-specific CIDEC plays a pivotal role in regulating adipose lipid metabolism and whole-body glucose homeostasis. In summary, our findings identify human CIDEC as a potential 'drug' or a 'druggable' target to reverse obesity-induced lipotoxicity and glucose intolerance.


Subject(s)
Glucose Intolerance , Insulin Resistance , Animals , Cholesterol , Diet, High-Fat/adverse effects , Fatty Acids, Nonesterified , Glucose , Glucose Intolerance/genetics , Glucose Intolerance/prevention & control , Humans , Insulin Resistance/genetics , Lipase/genetics , Lipid Metabolism , Lipoproteins, LDL/metabolism , Mice , Nucleotides/metabolism , Obesity/genetics , Proteins/metabolism , Transgenes , Triglycerides
2.
Int J Mol Sci ; 23(16)2022 Aug 11.
Article in English | MEDLINE | ID: mdl-36012215

ABSTRACT

The underlying pathological mechanisms of diabetes are complicated and varied in diabetic patients, which may lead to the current medications often failing to maintain glycemic control in the long term. Thus, the discovery of diverse new compounds for developing medicines to treat diabetes and its complications are urgently needed. Polyphenols are metabolites of plants and have been employed in the prevention and treatment of a variety of diseases. Caffeic acid phenethyl ester (CAPE) is a category of compounds structurally similar to polyphenols. In this study, we aimed to investigate the antidiabetic activity and potential molecular mechanisms of a novel synthetic CAPE derivative N-octyl caffeamide (36M) using high-fat (HF) diet induced obese mouse models. Our results demonstrate that 36M prevented the progression of diabetes in the HF diet fed obese mice via increasing phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) and inhibiting expression of protein tyrosine phosphatase 1B (PTP1B). We also found that 36M could prevent hepatic lipid storage in the HF diet fed mice via inhibition of fatty acid synthase and lipid droplet proteins, including perilipins and Fsp27. In conclusion, 36M is a potential candidate compound that can be developed as AMPK inhibitor and PTP1B inhibitor for treating diabetes and hepatic steatosis.


Subject(s)
Diabetes Mellitus , Fatty Liver , AMP-Activated Protein Kinases/metabolism , Amides/metabolism , Amides/pharmacology , Animals , Caffeic Acids , Diabetes Mellitus/metabolism , Diet, High-Fat , Fatty Liver/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Polyphenols/metabolism , Polyphenols/pharmacology , Polyphenols/therapeutic use
3.
J Biol Chem ; 294(45): 16740-16755, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31548309

ABSTRACT

Lipid droplets (LDs) are evolutionarily conserved organelles that play critical roles in mammalian lipid storage and metabolism. However, the molecular mechanisms governing the biogenesis and growth of LDs remain poorly understood. Phosphatidic acid (PA) is a precursor of phospholipids and triacylglycerols and substrate of CDP-diacylglycerol (CDP-DAG) synthase 1 (CDS1) and CDS2, which catalyze the formation of CDP-DAG. Here, using siRNA-based gene knockdowns and CRISPR/Cas9-mediated gene knockouts, along with immunological, molecular, and fluorescence microscopy approaches, we examined the role of CDS1 and CDS2 in LD biogenesis and growth. Knockdown of either CDS1 or CDS2 expression resulted in the formation of giant or supersized LDs in cultured mammalian cells. Interestingly, down-regulation of cell death-inducing DFF45-like effector C (CIDEC), encoding a prominent regulator of LD growth in adipocytes, restored LD size in CDS1- but not in CDS2-deficient cells. On the other hand, reducing expression of two enzymes responsible for triacylglycerol synthesis, diacylglycerol O-acyltransferase 2 (DGAT2) and glycerol-3-phosphate acyltransferase 4 (GPAT4), rescued the LD phenotype in CDS2-deficient, but not CDS1-deficient, cells. Moreover, CDS2 deficiency, but not CDS1 deficiency, promoted the LD association of DGAT2 and GPAT4 and impaired initial LD maturation. Finally, although both CDS1 and CDS2 appeared to regulate PA levels on the LD surface, CDS2 had a stronger effect. We conclude that CDS1 and CDS2 regulate LD dynamics through distinct mechanisms.


Subject(s)
Diacylglycerol Cholinephosphotransferase/metabolism , Lipid Droplets/metabolism , Cell Line , Diacylglycerol Cholinephosphotransferase/deficiency , Diacylglycerol Cholinephosphotransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Gene Knockdown Techniques , Humans , Phosphatidic Acids/metabolism
4.
Am J Physiol Endocrinol Metab ; 318(2): E249-E261, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31846369

ABSTRACT

Hepatic fat-specific protein 27 [cell death-inducing DNA fragmentation effector protein C (Cidec)/Fsp27] mRNA levels have been associated with hepatic lipid droplet extent under certain circumstances. To address its hepatic expression under different dietary conditions and in both sexes, apolipoprotein E (Apoe)-deficient mice were subjected to different experimental conditions for 11 wk to test the influence of cholesterol, Western diet, squalene, oleanolic acid, sex, and surgical castration on Cidec/Fsp27 mRNA expression. Dietary cholesterol increased hepatic Cidec/Fsp27ß expression, an effect that was suppressed when cholesterol was combined with saturated fat as represented by Western diet feeding. Using the latter diet, neither oleanolic acid nor squalene modified its expression. Females showed lower levels of hepatic Cidec/Fsp27ß expression than males when they were fed Western diets, a result that was translated into a lesser amount of CIDEC/FSP27 protein in lipid droplets and microsomes. This was also confirmed in low-density lipoprotein receptor (Ldlr)-deficient mice. Incubation with estradiol resulted in decreased Cidec/Fsp27ß expression in AML12 cells. Whereas male surgical castration did not modify the expression, ovariectomized females did show increased levels compared with control females. Females also showed increased expression of peroxisome proliferator-activated receptor-γ coactivator 1-α (Pgc1a), suppressed by ovariectomy, and the values were significantly and inversely associated with those of Cidec/Fsp27ß. When Pgc1a-deficient mice were used, the sex differences in Cidec/Fsp27ß expression disappeared. Therefore, hepatic Cidec/Fsp27ß expression has a complex regulation influenced by diet and sex hormonal milieu. The mRNA sex differences are controlled by Pgc1a.


Subject(s)
Diet, Western/adverse effects , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Proteins/genetics , Animals , Cell Line , Cholesterol, Dietary/pharmacology , Female , Lipid Droplets/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Orchiectomy , Ovariectomy , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , RNA, Messenger/biosynthesis , Receptors, LDL/genetics , Receptors, LDL/metabolism , Sex Characteristics
5.
Biochem Biophys Res Commun ; 530(2): 432-439, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32553626

ABSTRACT

The CIDE (cell death-inducing DFF45-like effector) family composed of CIDEA, CIDEB, CIDEC/FSP27 (fat-specific protein 27), has a critical role in growth of lipid droplets. Of these, CIDEB and CIDEC2/FSP27B are abundant in the liver, and the steatotic livers, respectively. Hepatocyte nuclear factor 4α (HNF4α) has an important role in lipid homeostasis because liver-specific HNF4α-null mice (Hnf4aΔHep mice) exhibit hepatosteatosis. We investigated whether HNF4α directly regulates expression of CIDE family genes. Expression of Cideb and Fsp27b was largely decreased in Hnf4aΔHep mice, while expression of Cidea was increased. Similar results were observed only in CIDEC2, the human orthologue of the Fsp27b, in human hepatoma cell lines in which HNF4α expression was knocked down. Conversely, overexpression of HNF4α strongly induced CIDEC2 expression in hepatoma cell lines. Furthermore, HNF4α transactivated Fsp27b by direct binding to an HNF4α response element in the Fsp27b promoter. In addition, Fsp27b is known to be transactivated by CREBH that is regulated by HNF4α, and expression of CREBH was induced by HNF4α in human hepatoma cells. Co-transfection of HNF4α and CREBH resulted in synergistic transactivation and induction of Fsp27b compared to that of HNF4α or CREBH alone. These results suggest that HNF4α, in conjunction with CREBH, plays an important role in regulation of Fsp27b expression.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation , Hepatocyte Nuclear Factor 4/metabolism , Liver/metabolism , Proteins/genetics , Animals , Fatty Liver/genetics , Fatty Liver/metabolism , Hep G2 Cells , Humans , Mice , Transcriptional Activation
6.
J Sci Food Agric ; 100(5): 2244-2251, 2020 Mar 30.
Article in English | MEDLINE | ID: mdl-31919850

ABSTRACT

BACKGROUND: Sea cucumber is a rich source of eicosapentaenoic acid in the form of eicosapentaenoic acid-enriched phospholipids (EPA-PL). It is known to be efficacious in preventing obesity. However, few studies have focused on the role of EPA-PL in inhibiting lipid accumulation by lipid droplets (LDs). This study first investigated the effect of EPA-PL from sea cucumber on the formation of LDs and the underlying mechanism in C57BL/6J mice. The mice were randomly divided into two groups and treated for 8 weeks or 3, 7, and 14 days with either (i) a high-sucrose diet (model group), (ii) a high-sucrose diet plus 2% EPA-PL (EPA-PL group). RESULTS: Eight-week EPA-PL supplementation significantly reduced lipid accumulation and LD size in liver and white adipose tissue (WAT), which was accompanied by the decreased expression of LDs-associated protein FSP27. A 3-day EPA-PL treatment suppressed the mRNA expression of Fsp27. The mRNA level of Fsp27 reached its 'normal level' after withdrawing EPA-PL for 7 days, suggesting that EPA-PL might serve as a rapid regulator of FSP27. Furthermore, EPA-PL increased the expression of lipolysis genes Hsl and Atgl accompanied by the regulation of Pparγ in WAT. CONCLUSIONS: Dietary EPA-PL from sea cucumber (Cucumaria frondosa) protected against lipid accumulation by regulating LDs-associated protein FSP27, which might provide novel evidence for the anti-obesity action of EPA-PL. © 2020 Society of Chemical Industry.


Subject(s)
Anti-Obesity Agents/pharmacology , Eicosapentaenoic Acid/pharmacology , Lipid Droplets/metabolism , Phospholipids/metabolism , Adipose Tissue, White/metabolism , Animals , Cholesterol/blood , Eicosapentaenoic Acid/analogs & derivatives , Fatty Acids, Nonesterified/blood , Gene Expression Regulation , Lipid Droplets/drug effects , Lipolysis/drug effects , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/metabolism , Proteins/genetics , Proteins/metabolism , RNA, Messenger/metabolism , Triglycerides/blood
7.
Pediatr Endocrinol Rev ; 17(1): 4-16, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31599132

ABSTRACT

Growth hormone (GH) is a pleiotropic hormone that coordinates an array of physiological processes including growth and metabolism. GH promotes anabolic action in all tissues except adipose, where it catabolizes stored fat to release energy for the promotion of growth in other tissues. However, chronic stimulation of lipolysis by GH results in an increased flux of free fatty acids (FFAs) into systemic circulation. Hence, a sustained release of high levels of GH contributes significantly to the development of insulin resistance by antagonizing the anti-lipolytic action of insulin. The molecular pathways associated with the lipolytic effect of GH in adipose tissue however, remain elusive. Recent studies have provided molecular insights into GH-induced lipolysis and impairment of insulin signaling. This review discusses the physiological and metabolic actions of GH on adipose tissue as well as GH-mediated deregulation of the FSP27-PPARγ axis which alters adipose tissue homeostasis and contributes to the development of insulin resistance and Type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Human Growth Hormone , Insulin Resistance , Lipolysis , Adipose Tissue/drug effects , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/physiopathology , Human Growth Hormone/metabolism , Human Growth Hormone/pharmacology , Humans , Lipolysis/drug effects
8.
J Biol Chem ; 292(26): 10824-10834, 2017 06 30.
Article in English | MEDLINE | ID: mdl-28490632

ABSTRACT

Adipose tissue stores neutral lipids and is a major metabolic organ involved in regulating whole-body energy homeostasis. Triacylglycerol is stored as unilocular large lipid droplets (LDs) in white adipocytes and as multilocular small LDs in brown adipocytes. Proteins of the cell death-inducing DNA fragmentation factor A-like effector (Cide) family include CideA, CideB, and fat-specific protein of 27 (FSP27). Of these, FSP27 has been shown to play a crucial role in the formation of unilocular large LDs in white adipocytes. However, the mechanisms by which brown adipocytes store small and multilocular LDs remain unclear. An FSP27 isoform, FSP27ß, was recently identified. We herein report that CideA and FSP27ß are mainly expressed in brown adipose tissue and that FSP27ß overexpression inhibits CideA-induced LD enlargements in a dose-dependent manner in COS cells. Furthermore, RNAi-mediated FSP27ß depletion resulted in enlarged LDs in HB2 adipocytes, which possess the characteristics of brown adipocytes. Brown adipocytes in FSP27-knock-out mice that express CideA, but not FSP27ß, had larger and fewer LDs. Moreover, we confirmed that FSP27ß and CideA form a complex in brown adipose tissue. Our results suggest that FSP27ß negatively regulates CideA-promoted enlargement of LD size in brown adipocytes. FSP27ß appears to be responsible for the formation of small and multilocular LDs in brown adipose tissue, a morphology facilitating free fatty acid transport to mitochondria adjacent to LDs for oxidation in brown adipocytes.


Subject(s)
Adipocytes, Brown/metabolism , Apoptosis Regulatory Proteins/metabolism , Lipid Droplets/metabolism , Multiprotein Complexes/metabolism , Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , COS Cells , Chlorocebus aethiops , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Proteins/genetics
9.
FASEB J ; 31(9): 4023-4036, 2017 09.
Article in English | MEDLINE | ID: mdl-28533326

ABSTRACT

The purpose of this study was to investigate whether genetic ablation of prostaglandin E receptor subtype 4 (EP4) affects white adipose tissue (WAT) remodeling mediated by ß3-adrenergic stimulation. The selective ß3-adrenergic agonist, CL316243 (1 mg/kg/d, i.p.) caused a greater increase in metabolic rate in EP4-knockout mice. CL316243 fragmented the unilocular lipid droplet into multilocular lipid vacuoles and increased mitochondrial biogenesis and its activity. These changes were amplified in mice with EP4 deficiency and were selectively seen in subcutaneous WAT. The expression of fat-specific protein (FSP)-27, a protein that promotes fusion of triglycerides and formation of unilocular lipid droplets were diminished, whereas the expression of phosphorylated AMPK, the upstream regulator of FSP27, was enhanced in EP4-deficient mice. The present study showed that EP4 acts as a negative regulator of WAT remodeling, it tightly coordinates rates of triglyceride storage in lipid droplets and mitochondrial respiratory function in subcutaneous white adipocytes through the phosphorylated AMPK-FSP27 signaling axis. Thus, deletion of EP4 increases mitochondrial biogenesis and oxidative capacity in WAT, and fat mass loss ensues in mice.-Ying, F., Cai, Y., Cai, Y., Wang, Y., Tang, E. H. C. Prostaglandin E receptor subtype 4 regulates lipid droplet size and mitochondrial activity in murine subcutaneous white adipose tissue.


Subject(s)
Adipose Tissue, White/metabolism , Gene Expression Regulation/physiology , Lipids/chemistry , Mitochondria/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Subcutaneous Fat/metabolism , Animals , Dioxoles/pharmacology , Energy Metabolism/drug effects , Energy Metabolism/physiology , Mice , Mice, Knockout , Oxidation-Reduction , Receptors, Prostaglandin E, EP4 Subtype/genetics , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
10.
Biol Pharm Bull ; 40(6): 888-893, 2017.
Article in English | MEDLINE | ID: mdl-28566630

ABSTRACT

The fat-specific protein 27 (Fsp27) gene belongs to the cell death-inducing DNA fragmentation factor 45-like effector family. Fsp27 is highly expressed in adipose tissue as well as the fatty liver of ob/ob mice. Fsp27 is directly regulated by the peroxisome proliferator-activated receptor γ (PPARγ) in livers of genetically obese leptin deficient ob/ob mice. In the present study, Fsp27 was markedly induced by 24 h fasting in genetically normal mouse livers and repressed by refeeding a high sucrose diet. In contrast with the liver, Fsp27 expression was decreased in adipose tissue by fasting and increased by refeeding. Interestingly, fasting-induced Fsp27 liver expression was independent of PPARγ. Moreover, Fsp27 expression was induced in the insulin-depleted livers of streptozotocin-treated mice. Finally, Fsp27 expression was repressed by direct injection of glucose or insulin in fasting mice. These results suggest that insulin represses Fsp27 expression in the fasting liver.


Subject(s)
Fasting/metabolism , Insulin/pharmacology , Liver/metabolism , Proteins/genetics , Adipose Tissue, White/metabolism , Animals , Male , Mice, Knockout , Mice, Obese , PPAR gamma/genetics , Streptozocin
11.
Biochem Biophys Res Commun ; 439(4): 564-9, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-24025675

ABSTRACT

FSP27 (CIDE-3 in humans) plays critical roles in lipid metabolism and apoptosis and is known to be involved in regulation of lipid droplet (LD) size and lipid storage and apoptotic DNA fragmentation. Given that CIDE-containing proteins including FSP27 are associated with many human diseases including cancer, aging, diabetes, and obesity, studies of FSP27 and other CIDE-containing proteins are of great biological importance. As a first step toward elucidating the molecular mechanisms of FSP27-mediated lipid droplet growth and apoptosis, we report the crystal structure of the CIDE-N domain of FSP27 at a resolution of 2.0 Å. The structure revealed a possible biologically important homo-dimeric interface similar to that formed by the hetero-dimeric complex, CAD/ICAD. Comparison with other structural homologues revealed that the PB1 domain of BEM1P, ubiquitin-like domain of BAG6 and ubiquitin are structurally similar proteins. Our homo-dimeric structure of the CIDE-N domain of FSP27 will provide important information that will enable better understanding of the function of FSP27.


Subject(s)
Proteins/chemistry , Amino Acid Sequence , Apoptosis Regulatory Proteins , Binding Sites , Dimerization , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Proteins/metabolism
12.
Mol Nutr Food Res ; 65(1): e2000771, 2021 01.
Article in English | MEDLINE | ID: mdl-32997872

ABSTRACT

The endoplasmic reticulum (ER)-resident basic leucine zipper (bZIP) transcription factor c-AMP responsive element binding protein H (CREBH/CREB3L3) is exclusively expressed in the liver and intestine. Physiologically, CREBH is intrinsically linked to nutritional homeostasis via its regulation on fatty acid ß-oxidation, lipid droplet process, very low-density lipoprotein metabolism, gluconeogenesis, and iron metabolism. Pathologically, CREBH enhances hepatic acute-phase response gene expression (e.g., C-reactive protein and serum amyloid P-component) and mediates nutrient-surplus induced metabolic inflammation. Hyperactivation of CREBH in metabolic inflammation further contributes to the development of hyperlipidemia, lipotoxicity, non-alcoholic fatty liver disease, and potentially non-alcoholic steatohepatitis. This review highlights recent findings that delineate the interactions between CREBH and peroxisome proliferator activated receptor α (PPARα), fibroblast growth factor 21 (FGF21), fat-specific protein 27 (FSP27), and lipoprotein metabolism with a focus on the molecular and biochemical mechanisms that underlie the development of metabolic inflammation, non-alcoholic fatty liver disease and inflammatory associated bone disease.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Inflammation/metabolism , Metabolic Diseases/metabolism , Acute-Phase Reaction/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/chemistry , Cyclic AMP Response Element-Binding Protein/genetics , Cytokines/metabolism , Energy Metabolism , Fasting , Gluconeogenesis , Humans , Lipid Metabolism , Lipoproteins, LDL/metabolism , Mice , Non-alcoholic Fatty Liver Disease/metabolism
13.
JHEP Rep ; 3(4): 100300, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34159305

ABSTRACT

Gene therapy is becoming an increasingly valuable tool to treat many genetic diseases with no or limited treatment options. This is the case for hundreds of monogenic metabolic disorders of hepatic origin, for which liver transplantation remains the only cure. Furthermore, the liver contains 10-15% of the body's total blood volume, making it ideal for use as a factory to secrete proteins into the circulation. In recent decades, an expanding toolbox has become available for liver-directed gene delivery. Although viral vectors have long been the preferred approach to target hepatocytes, an increasing number of non-viral vectors are emerging as highly efficient vehicles for the delivery of genetic material. Herein, we review advances in gene delivery vectors targeting the liver and more specifically hepatocytes, covering strategies based on gene addition and gene editing, as well as the exciting results obtained with the use of RNA as a therapeutic molecule. Moreover, we will briefly summarise some of the limitations of current liver-directed gene therapy approaches and potential ways of overcoming them.

14.
Mol Cell Endocrinol ; 518: 111038, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32966863

ABSTRACT

Growth hormone (GH) is a pleiotropic hormone that coordinates an array of physiological processes, including effects on bone, muscle, and fat, ultimately resulting in growth. Metabolically, GH promotes anabolic action in most tissues except adipose, where its catabolic action causes the breakdown of stored triglycerides into free fatty acids (FFA). GH antagonizes insulin action via various molecular pathways. Chronic GH secretion suppresses the anti-lipolytic action of insulin and increases FFA flux into the systemic circulation; thus, promoting lipotoxicity, which causes pathophysiological problems, including insulin resistance. In this review, we will provide an update on GH-stimulated adipose lipolysis and its consequences on insulin signaling in liver, skeletal muscle, and adipose tissue. Furthermore, we will discuss the mechanisms that contribute to the diabetogenic action of GH.


Subject(s)
Growth Hormone/pharmacology , Insulin/metabolism , Adipose Tissue/metabolism , Animals , Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Growth Hormone/metabolism , Human Growth Hormone/metabolism , Human Growth Hormone/pharmacology , Humans , Insulin Resistance/physiology , Lipolysis/drug effects , Signal Transduction/drug effects
15.
Biochimie ; 169: 41-53, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31400447

ABSTRACT

Fat-Specific Protein 27 (FSP27) belongs to a small group of vertebrate proteins containing a Cell-death Inducing DNA fragmentation factor-α-like Effector (CIDE)-C domain and is involved in lipid droplet (LD) accumulation and energy homeostasis. FSP27 is predominantly expressed in white and brown adipose tissues, as well as liver, and plays a key role in mediating LD-LD fusion. No orthologs have been identified in invertebrates or plants. In this study, we tested the function of mouse FSP27 in stably-transformed Arabidopsis thaliana leaves and seeds, as well as through transient expression in Nicotiana tabacum suspension-cultured cells and N. benthamiana leaves. Confocal microscopic analysis of plant cells revealed that, similar to ectopic expression in mammalian cells, FSP27 produced in plants 1) correctly localized to LDs, 2) accumulated at LD-LD contact sites, and 3) induced an increase in the number and size of LDs and also promoted LD clustering and fusion. Furthermore, FSP27 increased oil content in transgenic A. thaliana seeds. Given that plant oils have uses in human and animal nutrition as well as industrial uses such as biofuels and bioplastics, our results suggest that ectopic expression of FSP27 in plants represents a potential strategy for increasing oil content and energy density in bioenergy or oilseed crops.


Subject(s)
Arabidopsis/genetics , Diacylglycerol O-Acyltransferase/genetics , Lipid Droplets/metabolism , Lipid Metabolism/genetics , Nicotiana/genetics , Proteins/genetics , Animals , Arabidopsis/metabolism , Cloning, Molecular , Diacylglycerol O-Acyltransferase/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Lipid Droplets/ultrastructure , Membrane Fusion , Mice , Organelle Size , Plant Cells/metabolism , Plant Leaves/genetics , Plant Leaves/metabolism , Plants, Genetically Modified , Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Seeds/genetics , Seeds/metabolism , Nicotiana/metabolism
16.
Cells ; 8(3)2019 03 13.
Article in English | MEDLINE | ID: mdl-30871156

ABSTRACT

Cell death-Inducing DNA Fragmentation Factor Alpha (DFFA)-like Effector (CIDE) proteins have emerged as lipid droplet-associated proteins that regulate fat metabolism. There are three members in the CIDE protein family-CIDEA, CIDEB, and CIDEC (also known as fat-specific protein 27 (FSP27)). CIDEA and FSP27 are primarily expressed in adipose tissue, while CIDEB is expressed in the liver. Originally, based upon their homology with DNA fragmentation factors, these proteins were identified as apoptotic proteins. However, recent studies have changed the perception of these proteins, redefining them as regulators of lipid droplet dynamics and fat metabolism, which contribute to a healthy metabolic phenotype in humans. Despite various studies in humans and gene-targeting studies in mice, the physiological roles of CIDE proteins remains elusive. This review will summarize the known physiological role and metabolic pathways regulated by the CIDE proteins in human health and disease.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Disease , Health , Animals , Apoptosis , Humans , Lipid Droplets/metabolism , Metabolism , Mice
17.
Mol Metab ; 29: 65-75, 2019 11.
Article in English | MEDLINE | ID: mdl-31668393

ABSTRACT

OBJECTIVE: Growth hormone (GH) stimulates lipolysis, but the underlying mechanisms remain incompletely understood. We examined the effect of GH on the expression of lipolytic regulators in adipose tissue (AT). METHODS: In a randomized, placebo-controlled, cross-over study, nine men were examined after injection of 1) a GH bolus and 2) a GH-receptor antagonist (pegvisomant) followed by four AT biopsies. In a second study, eight men were examined in a 2 × 2 factorial design including GH infusion and 36-h fasting with AT biopsies obtained during a basal period and a hyperinsulinemic-euglycemic clamp. Expression of GH-signaling intermediates and lipolytic regulators were studied by PCR and western blotting. In addition, mechanistic experiments in mouse models and 3T3-L1 adipocytes were performed. RESULTS: The GH bolus increased circulating free fatty acids (p < 0.0001) together with phosphorylation of signal transducer and activator of transcription 5 (STAT5) (p < 0.0001) and mRNA expression of the STAT5-dependent genes cytokine-inducible SH2-containing protein (CISH) and IGF-1 in AT. This was accompanied by suppressed mRNA expression of G0/G1 switch gene 2 (G0S2) (p = 0.007) and fat specific protein 27 (FSP27) (p = 0.002) and upregulation of phosphatase and tensin homolog (PTEN) mRNA expression (p = 0.03). Suppression of G0S2 was also observed in humans after GH infusion and fasting, as well as in GH transgene mice, and in vitro studies suggested MEK-PPARγ signaling to be involved. CONCLUSIONS: GH-induced lipolysis in human subjects in vivo is linked to downregulation of G0S2 and FSP27 and upregulation of PTEN in AT. Mechanistically, in vitro data suggest that GH acts via MEK to suppress PPARγ-dependent transcription of G0S2. ClinicalTrials.govNCT02782221 and NCT01209429.


Subject(s)
Adipose Tissue/metabolism , Human Growth Hormone/analogs & derivatives , Human Growth Hormone/administration & dosage , Adipose Tissue/pathology , Adult , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Biomarkers/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cross-Over Studies , Down-Regulation/drug effects , Fatty Acids, Nonesterified/blood , Human Growth Hormone/pharmacology , Humans , Lipolysis , Male , Mice , Mice, Transgenic , PPAR gamma/metabolism , Placebo Effect , Signal Transduction , Young Adult
18.
Atherosclerosis ; 275: 43-49, 2018 08.
Article in English | MEDLINE | ID: mdl-29859472

ABSTRACT

BACKGROUND AND AIMS: Obesity, hepatosteatosis, and hypertriglyceridemia are components of the metabolic syndrome and independent risk factors for cardiovascular disease. The lipid droplet-associated protein CIDEC (cell death-inducing DFFA-like effector C), known in mice as FSP27 (fat-specific protein 27), plays a key role in maintaining triacylglyceride (TAG) homeostasis in adipose tissue and liver, and controls circulating TAG levels in mice. Importantly, mutations and SNPs in CIDEC are associated with dyslipidemia and altered metabolic function in humans. Here we tested whether systemic silencing of Fsp27 using antisense oligonucleotides (ASOs) was atheroprotective in LDL receptor knock-out (Ldlr-/-) mice. METHODS: Atheroprone Ldlr-/- mice were fed a high-fat, high-cholesterol diet for 12 weeks while simultaneously dosed with saline, ASO-ctrl, or ASO-Fsp27. RESULTS: Data show that, compared to control treatments, silencing Fsp27 significantly reduced body weight gain and visceral adiposity, prevented diet-induced hypertriglyceridemia, and reduced atherosclerotic lesion size both in en face aortas and in the aortic root. CONCLUSIONS: Our findings suggest that therapeutic silencing of Fsp27 with ASOs may be beneficial in the prevention and management of atherogenic disease in patients with metabolic syndrome.


Subject(s)
Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Gene Silencing , Genetic Therapy/methods , Oligonucleotides, Antisense/genetics , Proteins/genetics , Receptors, LDL/deficiency , Adiposity , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cholesterol, Dietary , Diet, High-Fat , Disease Models, Animal , Disease Progression , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/physiopathology , Male , Mice, Knockout , Oligonucleotides, Antisense/metabolism , Plaque, Atherosclerotic , Proteins/metabolism , Receptors, LDL/genetics , Triglycerides/blood , Weight Gain
19.
Curr Protein Pept Sci ; 19(2): 221-233, 2018.
Article in English | MEDLINE | ID: mdl-28925902

ABSTRACT

Adipose triglyceride lipase (ATGL) is the key-enzyme for the release of fatty acids (FAs) from triacylglycerol (TG) stores during intracellular lipolysis producing FAs used for energy production. There is growing evidence that the products and intermediates from lipolytic breakdown during the FA mobilization process also have fundamental regulatory functions affecting cell signaling, gene expression, metabolism, cell growth, cell death, and lipotoxicity. Regulation of ATGL is therefore vital for maintaining a defined balance between lipid storage and mobilization. This review addresses the regulation of ATGL activity at the post-translational level with special emphasis on protein-mediated interaction at the site of hydrolytic action, namely to the lipid droplet.


Subject(s)
Fatty Acids/metabolism , Lipase/chemistry , Lipase/metabolism , Lipolysis/physiology , Triglycerides/metabolism , Animals , Humans , Lipid Metabolism , Lipids/chemistry , Molecular Structure , Protein Conformation , Signal Transduction
20.
J Atheroscler Thromb ; 24(10): 989-998, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28883211

ABSTRACT

White adipose tissue (WAT) stores energy as triacylglycerol in preparation for fasting state. In contrast, brown adipose tissue (BAT) consumes energy and produces heat in a cold environment. One of the major differences between these two adipose tissues is the morphology of the intracellular lipid droplet (LD), which is large and unilocular in WAT and small and multilocular in BAT. Although the fat-specific protein 27 alpha (FSP27α), belonging to the cell death-inducing DNA fragmentation factor A (DFFA)-like effector (Cide) family, was known to be indispensable for large unilocular LD formation in WAT, the mechanism that regulated small multilocular LD formation in BAT remained unknown. We recently uncovered that FSP27ß, a novel isoform of FSP27 abundantly expressed in BAT, plays a crucial role in small multilocular LD formation by inhibiting the homodimerization of CideA in BAT. We speculate that unilocular LD formation is ideal for efficient lipid storage in WAT because lipolysis from the LD surface is restricted due to the minimum LD surface area. In addition, hydrolyzed free fatty acid (FFA) and glycerol can efficiently flow out into the circulation from the cell surface. In contrast, small multilocular LD formation is ideal for efficient intracellular lipolysis from the LD surface and the subsequent facilitation of FFA transport to mitochondria that are adjacent to LDs for ß-oxidation in BAT. Thus, intracellular LD morphology is closely related to the functions and characteristics of adipose tissues. Given that the browning of adipose tissue leads to enhanced energy expenditure and the prevention of obesity, clarification of the mechanism with respect to intracellular LD formation is very meaningful.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Apoptosis Regulatory Proteins/metabolism , Energy Metabolism , Lipid Droplets , Animals , DNA Fragmentation , Disease Models, Animal , Fatty Acids, Nonesterified/chemistry , Glycerol/chemistry , Homeostasis , Humans , Hydrolysis , Lipid Metabolism , Lipolysis , Mice , Mitochondria/metabolism , Obesity/metabolism , Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL