Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 274
1.
Lipids Health Dis ; 20(1): 117, 2021 Sep 26.
Article En | MEDLINE | ID: mdl-34565390

BACKGROUND: Angiopoietin-like proteins (ANGPTL), primarily 3, 4, and 8, play a major role in maintaining energy homeostasis by regulating triglyceride metabolism. This study evaluated the level of ANGPTL3, 4, and 8 in the liver, brown adipose tissue (BAT), and subcutaneous white adipose tissue (SAT) of mice maintained under acute and chronic cold conditions. METHODS: C57BL/6J mice were exposed to cold temperature (4 °C) for 10 days with food provided ad libitum. Animal tissues were harvested at Day 0 (Control group, n = 5) and Days 1, 3, 5, and 10 (cold treatment groups, n = 10 per group). The expression levels of various genes were measured in the liver, SAT, and BAT. ANGPTL3, 4, and 8 expressions were measured in the liver. ANGPTL4, 8, and genes involved in browning and lipid metabolism [uncoupling protein 1 (UCP1), lipoprotein lipase (LPL), and adipose triglyceride lipase (ATGL)] were measured in SAT and BAT. Western blotting (WB) analysis and immunohistochemistry (IHC) were performed to confirm ANGPTL8 expression in these tissues. RESULTS: The expressions of ANGPTL3 and 8 mRNA were significantly reduced in mouse liver tissues after cold treatment (P < 0.05); however, the expression of ANGPTL4 was not significantly altered. In BAT, ANGPTL8 expression was unchanged after cold treatment, whereas ANGPTL4 expression was significantly reduced (P < 0.05). ANGPTL4 levels were also significantly reduced in SAT, whereas ANGPTL8 gene expression exhibited over a 5-fold increase. Similarly, UCP1 gene expression was also significantly increased in SAT. The mRNA levels of LPL and ATGL showed an initial increase followed by a gradual decrease with an increase in the days of cold exposure. ANGPTL8 protein overexpression was further confirmed by WB and IHC. CONCLUSIONS: This study shows that exposure to acute and chronic cold treatment results in the differential expression of ANGPTL proteins in the liver and adipose tissues (SAT and BAT). The results show a significant reduction in ANGPTL4 in BAT, which is linked to improved thermogenesis in response to acute cold exposure. ANGPTL8 was activated under acute and chronic cold conditions in SAT, suggesting that it is involved in regulating lipolysis and enhancing SAT browning.


Adipose Tissue, White/metabolism , Angiopoietin-Like Protein 8/biosynthesis , Cold Temperature , Gene Expression Regulation , Acyltransferases/biosynthesis , Adipose Tissue , Animals , Gene Expression Profiling , Homeostasis , Immunohistochemistry , Lipolysis , Lipoprotein Lipase/biosynthesis , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Rats , Temperature , Uncoupling Protein 1/biosynthesis
2.
Br J Haematol ; 182(4): 521-525, 2018 08.
Article En | MEDLINE | ID: mdl-29953583

Lipoprotein lipase (LPL) mRNA expression in chronic lymphocytic leukaemia (CLL) is associated with an unmutated immunoglobulin profile and poor clinical outcome. We evaluated the subcellular localization of LPL protein in CLL cells that did or did not express LPL mRNA. Our results show that LPL protein is differently located in CLL cells depending on whether it is incorporated from the extracellular medium in mutated CLL or generated de novo by leukaemic cells of unmutated patients. The specific quantification of endogenous LPL protein correlates with mRNA expression levels and mutational IGHV status, suggesting LPL protein as a possible reliable prognostic marker in CLL.


Biomarkers, Tumor/biosynthesis , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Leukemic , Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Lipoprotein Lipase/biosynthesis , Neoplasm Proteins/biosynthesis , Aged , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Prognosis , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis
3.
Neurotox Res ; 31(3): 421-435, 2017 04.
Article En | MEDLINE | ID: mdl-28247204

A growing body of evidence implicates the endocannabinoid (eCB) system in the pathophysiology of depression. The aim of this study was to investigate the influence of changes in the eCB system, such as levels of neuromodulators, eCB synthesizing and degrading enzymes, and cannabinoid (CB) receptors, in different brain structures in animal models of depression using behavioral and biochemical analyses. Both models used, i.e., bulbectomized (OBX) and Wistar Kyoto (WKY) rats, were characterized at the behavioral level by increased immobility time. In the OBX rats, anandamide (AEA) levels were decreased in the prefrontal cortex, hippocampus, and striatum and increased in the nucleus accumbens, while 2-arachidonoylglycerol (2-AG) levels were increased in the prefrontal cortex and decreased in the nucleus accumbens with parallel changes in the expression of eCB metabolizing enzymes in several structures. It was also observed that CB1 receptor expression decreased in the hippocampus, dorsal striatum, and nucleus accumbens, and CB2 receptor expression decreased in the prefrontal cortex and hippocampus. In WKY rats, the levels of eCBs were reduced in the prefrontal cortex (2-AG) and dorsal striatum (AEA) and increased in the prefrontal cortex (AEA) with different changes in the expression of eCB metabolizing enzymes, while the CB1 receptor density was increased in several brain regions. These findings suggest that dysregulation in the eCB system is implicated in the pathogenesis of depression, although neurochemical changes were linked to the particular brain structure and the factor inducing depression (surgical removal of the olfactory bulbs vs. genetic modulation).


Arachidonic Acids/metabolism , Brain/metabolism , Depression/metabolism , Endocannabinoids/metabolism , Glycerides/metabolism , Polyunsaturated Alkamides/metabolism , Receptor, Cannabinoid, CB1/biosynthesis , Receptor, Cannabinoid, CB2/biosynthesis , Amidohydrolases/biosynthesis , Animals , Disease Models, Animal , Immobility Response, Tonic , Lipoprotein Lipase/biosynthesis , Male , Monoacylglycerol Lipases/biosynthesis , Olfactory Bulb/surgery , Phospholipase D/biosynthesis , Rats , Rats, Inbred WKY
4.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(5): 533-540, 2017 May.
Article En | MEDLINE | ID: mdl-28215713

Lipoprotein lipase (LPL) hydrolyzes lipids in plasma lipoproteins so that the fatty acids can be taken up and used by cells. The activity of LPL changes rapidly in response to changes in nutrition, physical activity and other conditions. Angiopoietin-like protein 4 (ANGPTL4) is an important controller of LPL activity. Both LPL and ANGPTL4 are produced and secreted by adipocytes. When the transcription blocker Actinomycin D was added to cultures of 3T3-L1 adipocytes, LPL activity in the medium increased several-fold. LPL mRNA decreased moderately during 5h, while ANGPTL4 mRNA and protein declined rapidly, explaining that LPL activity was increased. TNF-α is known to reduce LPL activity in adipose tissue. We have shown that TNF-α increased ANGPTL4 both at the mRNA and protein level. Expression of ANGPTL4 is known to be under control of Foxo1. Use of the Foxo1-specific inhibitor AS1842856, or knockdown of ANGPTL4 by RNAi, resulted in increased LPL activity in the medium. Both with ActD and with the Foxo1 inhibitor the cells became unresponsive to TNF-α. This study shows that TNF-α, by a Foxo1 dependent pathway, increases the transcription of ANGPTL4 which is secreted by the cells and causes inactivation of LPL.


Adipocytes/metabolism , Angiopoietins/biosynthesis , Forkhead Box Protein O1/metabolism , Lipoprotein Lipase/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Angiopoietin-Like Protein 4 , Angiopoietins/genetics , Animals , Dactinomycin/administration & dosage , Forkhead Box Protein O1/antagonists & inhibitors , Gene Expression Regulation/drug effects , Lipoprotein Lipase/genetics , Mice , Quinolones/administration & dosage , RNA Interference , RNA, Messenger/biosynthesis , Transcriptional Activation/drug effects , Tumor Necrosis Factor-alpha/administration & dosage
5.
Biochim Biophys Acta Gene Regul Mech ; 1860(3): 327-336, 2017 Mar.
Article En | MEDLINE | ID: mdl-28126606

Lipoprotein lipase (LPL) plays a major role in the hydrolysis of triglycerides (TG) from circulating TG-rich lipoproteins. The role of LPL in the liver has been controversial but recent studies in mice with liver LPL overexpression or deficiency have revealed important new roles of the enzyme in glucose and lipid metabolism. The objective of this study was to identify regulatory elements and factors that control the transcription of the human LPL gene in hepatocytes. Deletion analysis of the human LPL promoter revealed that the proximal region which harbors a binding site for the forkhead box transcription factor FOXA2/HNF-3ß at position -47/-40 is important for its hepatic cell activity. Silencing of FOXA2 in HepG2 cells reduced the LPL mRNA and protein levels. Direct binding of FOXA2 to the novel binding site was established in vitro and ex vivo. Mutagenesis of the FOXA2 site reduced the basal activity and abolished the FOXA2-mediated transactivation of the LPL promoter. Ιnsulin decreased LPL mRNA levels in HepG2 cells and this was associated with phosphorylation of AKT and nuclear export of FOXA2. In summary, the data of the present study combined with previous findings on the role of FOXA2 in HDL metabolism and gluconeogenesis, suggest that FOXA2 is a key regulator of lipid and glucose homeostasis in the adult liver. Understanding the mechanisms by which FOXA2 exerts its functions in hepatocytes may open the way to novel therapeutic strategies for patients with metabolic diseases such as dyslipidemia, diabetes and the metabolic syndrome.


Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocytes/metabolism , Lipoprotein Lipase/biosynthesis , Liver/metabolism , Response Elements , Transcriptional Activation , Animals , Hep G2 Cells , Hepatocyte Nuclear Factor 3-beta/genetics , Humans , Lipoprotein Lipase/genetics , Mice , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
6.
Exp Clin Endocrinol Diabetes ; 125(1): 33-41, 2017 Jan.
Article En | MEDLINE | ID: mdl-27657995

Introduction: Accumulated evidence indicates that there are intrinsic differences between adipose tissue-derived stem cells (ASCs) obtained from different body fat depots. Here, we compared the proliferation and multipotency of subcutaneous ASCs (SC-ASCs) and epididymal ASCs (ED-ASCs) before and after induction of diabetes by streptozotocin. Methods: The adipogenic and osteogenic abilities of rat SC-ASCs and ED-ASCs were evaluated using Oil Red O and Alizarin Red staining, respectively. The expression of adipocyte (PPAR-γ, LPL) and osteoblast (ALP, SPP1) specific mRNAs was evaluated by quantitative real-time PCR. MTT test was used for determination of cell proliferation capacity. Results: The proliferation of SC-ASCs was higher than ED-ASCs, both before and after diabetes induction (P<0.05). Diabetes increased the proliferative capability of SC-ASCs (P<0.05) but not ED-ASCs. Before diabetes, both adipogenic and osteogenic differentiation of SC-ASCs were higher than ED-ASCs (P<0.05). After diabetes, both SC-ASCs and ED-ASCs were able to differentiate into adipocyte and osteoblast, but the levels of differentiation were higher in SC-ASCs than in ED-ASCs (P<0.05). Diabetes decreased the expression of PPAR-γ and LPL, but increased the SPP1 and ALP expression in both SC-ASCs and ED-ASCs. Conclusion: Our data suggested that diabetes increases the proliferation of ASCs but decreases their adipogenic differentiation. Also, SC-ASCs have higher proliferation and differentiation abilities than ED-ASCs in normal and diabetic conditions so can be more preferable for cell therapy.


Cell Differentiation , Cell Proliferation , Diabetes Mellitus, Experimental/metabolism , Intra-Abdominal Fat/metabolism , Mesenchymal Stem Cells/metabolism , Subcutaneous Fat/metabolism , Alkaline Phosphatase/biosynthesis , Animals , Diabetes Mellitus, Experimental/pathology , Gene Expression Regulation , Intra-Abdominal Fat/pathology , Lipoprotein Lipase/biosynthesis , Male , Mesenchymal Stem Cells/pathology , Osteopontin/biosynthesis , PPAR gamma/biosynthesis , Rats , Rats, Wistar , Subcutaneous Fat/pathology
7.
J Clin Lipidol ; 10(4): 816-823, 2016.
Article En | MEDLINE | ID: mdl-27578112

BACKGROUND: Type 1 hyperlipoproteinemia is a rare autosomal recessive disorder most often caused by mutations in the lipoprotein lipase (LPL) gene resulting in severe hypertriglyceridemia and pancreatitis. OBJECTIVES: The aim of this study was to identify novel mutations in the LPL gene causing type 1 hyperlipoproteinemia and to understand the molecular mechanisms underlying the severe hypertriglyceridemia. METHODS: Three patients presenting classical features of type 1 hyperlipoproteinemia were recruited for DNA sequencing of the LPL gene. Pre-heparin and post-heparin plasma of patients were used for protein detection analysis and functional test. Furthermore, in vitro experiments were performed in HEK293 cells. Protein synthesis and secretion were analyzed in lysate and medium fraction, respectively, whereas medium fraction was used for functional assay. RESULTS: We identified two novel mutations in the LPL gene causing type 1 hyperlipoproteinemia: a two base pair deletion (c.765_766delAG) resulting in a frameshift at position 256 of the protein (p.G256TfsX26) and a nucleotide substitution (c.1211 T > G) resulting in a methionine to arginine substitution (p.M404 R). LPL protein and activity were not detected in pre-heparin or post-heparin plasma of the patient with p.G256TfsX26 mutation or in the medium of HEK293 cells over-expressing recombinant p.G256TfsX26 LPL. A relatively small amount of LPL p.M404 R was detected in both pre-heparin and post-heparin plasma and in the medium of the cells, whereas no LPL activity was detected. CONCLUSIONS: We conclude that these two novel mutations cause type 1 hyperlipoproteinemia by inducing a loss or reduction in LPL secretion accompanied by a loss of LPL enzymatic activity.


Hyperlipoproteinemia Type I/enzymology , Hyperlipoproteinemia Type I/genetics , Lipoprotein Lipase/genetics , Adult , DNA Mutational Analysis , Female , HEK293 Cells , Humans , Hyperlipoproteinemia Type I/blood , Hyperlipoproteinemia Type I/metabolism , Lipoprotein Lipase/biosynthesis , Lipoprotein Lipase/blood , Lipoprotein Lipase/metabolism , Male , Mutation , Young Adult
8.
Neuron ; 91(3): 644-51, 2016 Aug 03.
Article En | MEDLINE | ID: mdl-27497223

Cannabinoid receptor type 1 (CB1R)-expressing CCK interneurons are key regulators of cortical circuits. Here we report that retrograde endocannabinoid signaling and CB1R-mediated regulation of inhibitory synaptic transmission onto basal amygdala principal neurons strongly depend on principal neuron projection target. Projection-specific asymmetries in the regulation of local inhibitory micro-circuits may contribute to the selective activation of distinct amygdala output pathways during behavioral changes.


Amygdala/cytology , Amygdala/physiology , Interneurons/physiology , Neural Inhibition/physiology , Amygdala/metabolism , Animals , Cholecystokinin/genetics , Cholecystokinin/physiology , Lipoprotein Lipase/biosynthesis , Male , Mice , Mice, Transgenic , Monoacylglycerol Lipases/biosynthesis , Neuronal Plasticity/physiology , Receptor, Cannabinoid, CB1/physiology , Synaptic Transmission
9.
Mol Med Rep ; 13(5): 4487-93, 2016 May.
Article En | MEDLINE | ID: mdl-27035287

Small for gestational age (SGA) at birth increases the risk of developing metabolic syndrome, which encompasses various symptoms including hypertriglyceridemia. The aim of the present study was to determine whether maternal undernutrition during pregnancy may lead to alterations in hepatic triglyceride content and the gene expression levels of hepatic lipoprotein lipase (LPL) in SGA male offspring. The present study focused on the male offspring in order to prevent confounding factors, such as estrus cycle and hormone profile. Female Sprague Dawley rats were arbitrarily assigned to receive an ad libitum chow diet or 50% food restricted diet from pregnancy day 1 until parturition. Reverse transcription quantitative polymerase chain reaction and western blot analysis were used to measure the gene expression levels of hepatic LPL at day 1 and upon completion of the third week of age. Chromatin immunoprecipitation quantified the binding activity of liver X receptor­α (LXR­α) gene to the LXR response elements (LXRE) on LPL promoter and LPL epigenetic characteristics. At 3 weeks of age, SGA male offspring exhibited significantly elevated levels of hepatic triglycerides, which was concomitant with increased expression levels of LPL. Since LPL is regulated by LXR­α, the expression levels of LXR­α were detected in appropriate for gestational age and SGA male offspring. Maternal undernutrition during pregnancy led to an increase in the hepatic expression levels of LXR­α, and enriched binding to the putative LXR response elements in the LPL promoter regions in 3­week­old male offspring. In addition, enhanced acetylation of histone H3 [H3 lysine (K)9 and H3K14] was detected surrounding the LPL promoter. The results of the present study indicated that maternal undernutrition during pregnancy may lead to an increase in hepatic triglycerides, via alterations in the transcriptional and epigenetic regulation of the LPL gene.


Epigenesis, Genetic , Gene Expression Regulation, Enzymologic , Lipoprotein Lipase/biosynthesis , Liver/metabolism , Malnutrition/metabolism , Maternal Exposure , Prenatal Exposure Delayed Effects/metabolism , Triglycerides/biosynthesis , Animals , Female , Liver/pathology , Male , Malnutrition/pathology , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Rats , Rats, Sprague-Dawley
10.
Biofactors ; 42(3): 307-15, 2016 May.
Article En | MEDLINE | ID: mdl-27095633

Considering the biological function of α-tocopherol (α-Toc) as a potent protective factor against oxidative stress, this antioxidant is in the focus of aging research. To understand the role of α-Toc during aging we investigated α-Toc concentrations in young and aged primary human fibroblasts after supplementation with RRR-α-Toc. Additionally, α-Toc contents were determined in brain, kidney, and liver tissue of 10 week-, 18 month-, and 24 month-old mice, which were fed a standard diet containing 100 mg/kg dl-α-tocopheryl acetate. α-Toc concentrations in isolated lysosomes and the expression of the α-Toc transport proteins Niemann Pick C1 (NPC1), Niemann Pick C2 (NPC2), and lipoprotein lipase were also analyzed. Obtained data show a significant age-related increase of α-Toc in murine liver, kidney, and brain tissue as well as in human dermal fibroblasts. Also liver and kidney lysosomes are marked by elevated α-Toc contents with aging. NPC1 and NPC2 protein amounts are significantly decreased in adult and aged murine kidney tissue. Also aged human dermal fibroblasts show decreased NPC1 amounts. Supplementation of young and aged fibroblasts led also to decreased NPC1 amounts, suggesting a direct role of this protein in α-Toc distribution. Our results indicate an age-dependent increase of α-Toc in different murine tissues as well as in human fibroblasts. Furthermore saturation and intracellular distribution of α-Toc seem to be strongly dependent on the availability of this vitamin as well as on the presence of the lysosomal protein NPC1. © 2016 BioFactors, 42(3):307-315, 2016.


Aging/metabolism , Carrier Proteins/biosynthesis , Fibroblasts/metabolism , Lysosomes/metabolism , Membrane Glycoproteins/biosynthesis , alpha-Tocopherol/metabolism , Adult , Aging/genetics , Aging/pathology , Animals , Antioxidants/administration & dosage , Antioxidants/metabolism , Brain/metabolism , Brain/pathology , Carrier Proteins/genetics , Fibroblasts/pathology , Gene Expression Regulation/drug effects , Glycoproteins/biosynthesis , Humans , Intracellular Signaling Peptides and Proteins , Kidney/metabolism , Kidney/pathology , Lipoprotein Lipase/biosynthesis , Liver/metabolism , Liver/pathology , Lysosomes/pathology , Male , Membrane Glycoproteins/genetics , Mice , Niemann-Pick C1 Protein , Oxidative Stress/drug effects , Vesicular Transport Proteins , alpha-Tocopherol/administration & dosage
11.
Br J Cancer ; 114(6): 704-12, 2016 Mar 15.
Article En | MEDLINE | ID: mdl-26924423

BACKGROUND: The B-cell receptor (BCR) has a key role in the cross-talk between chronic lymphocytic leukaemia (CLL) cells and the tissue microenvironment, which favours disease progression by promoting proliferation and drug resistance. In vitro studies on downstream signalling and functional effects of CLL BCR ligation often report contradictory results, in part owing to the lack of a standardised stimulation protocol. Our aim was to define a biologically relevant and robust in vitro stimulation method with regard to cellular phenotypic and transcriptional responses. METHODS: We evaluated mRNA (FOS, MYC, LPL) and protein (CD54, CD19, CD62L, CD184) expression of genes modulated by BCR triggering in immunoglobulin heavy-chain variable region genes (IGHV)-mutated and -unmutated CLL cells, after stimulation using soluble or immobilised anti-IgM antibodies from different suppliers. RESULTS: The effect of BCR stimulation on gene and protein expression was comparable in all CLL patients, irrespective of IGHV mutation status. However, immobilised anti-IgM stimulation elicited clear and robust changes in gene and protein expression, whereas the response to soluble anti-IgM was far less obvious. CONCLUSIONS: These data indicate that the method of BCR stimulation is of major importance regarding responsiveness of CLL cells in the context of the tumour microenvironment, whereas genetic differences in the BCR pathway are less critical.


Leukemia, Lymphocytic, Chronic, B-Cell/blood , Receptors, Antigen, B-Cell/blood , Tumor Microenvironment , Adult , Aged , Antibodies, Anti-Idiotypic/immunology , Antibodies, Anti-Idiotypic/pharmacology , Antigens, CD/biosynthesis , Antigens, CD/genetics , Female , Flow Cytometry , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lipoprotein Lipase/biosynthesis , Lipoprotein Lipase/genetics , Male , Middle Aged , Oncogene Proteins v-fos/biosynthesis , Oncogene Proteins v-fos/genetics , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology
12.
J Biochem ; 159(6): 585-97, 2016 Jun.
Article En | MEDLINE | ID: mdl-26790472

Diacylglycerol (DG) lipase, which hydrolyses 1-stearoyl-2-arachidonyl-sn-glycerol to produce an endocannabinoid, 2-arachidonoylglycerol, was purified from the soluble fraction of rat brain lysates. DG lipase was purified about 1,200-fold by a sequential column chromatographic procedure. Among proteins identified by mass spectrometry analysis in the partially purified DG lipase sample, only DDHD domain containing two (DDHD2), which was formerly regarded as a phospholipase A1, exhibited significant DG lipase activity. Rat DDHD2 expressed in Chinese hamster ovary cells showed similar enzymatic properties to partially purified DG lipase from rat brain. The source of DG lipase activity in rat brain was immunoprecipitated using anti-DDHD2 antibody. Thus, we concluded that the DG lipase activity in the soluble fraction of rat brain is derived from DDHD2. DDHD2 is distributed widely in the rat brain. Immunohistochemical analysis revealed that DDHD2 is expressed in hippocampal neurons, but not in glia.


Gene Expression Regulation, Enzymologic/physiology , Hippocampus/enzymology , Lipoprotein Lipase , Nerve Tissue Proteins , Neurons/enzymology , Animals , Arachidonic Acids/genetics , Arachidonic Acids/metabolism , CHO Cells , Cloning, Molecular , Cricetinae , Cricetulus , Endocannabinoids/genetics , Endocannabinoids/metabolism , Glycerides/genetics , Glycerides/metabolism , Lipoprotein Lipase/biosynthesis , Lipoprotein Lipase/chemistry , Lipoprotein Lipase/genetics , Lipoprotein Lipase/isolation & purification , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/isolation & purification , Neuroglia/enzymology , Protein Domains , Rats
13.
Sci Rep ; 5: 18502, 2015 Dec 21.
Article En | MEDLINE | ID: mdl-26687026

Lipasin/Angptl8 is a feeding-induced hepatokine that regulates triglyceride (TAG) metabolism; its therapeutical potential, mechanism of action, and relation to the lipoprotein lipase (LPL), however, remain elusive. We generated five monoclonal lipasin antibodies, among which one lowered the serum TAG level when injected into mice, and the epitope was determined to be EIQVEE. Lipasin-deficient mice exhibited elevated postprandial activity of LPL in the heart and skeletal muscle, but not in white adipose tissue (WAT), suggesting that lipasin suppresses the activity of LPL specifically in cardiac and skeletal muscles. Consistently, mice injected with the effective antibody or with lipasin deficiency had increased postprandial cardiac LPL activity and lower TAG levels only in the fed state. These results suggest that lipasin acts, at least in part, in an endocrine manner. We propose the following model: feeding induces lipasin, activating the lipasin-Angptl3 pathway, which inhibits LPL in cardiac and skeletal muscles to direct circulating TAG to WAT for storage; conversely, fasting induces Angptl4, which inhibits LPL in WAT to direct circulating TAG to cardiac and skeletal muscles for oxidation. This model suggests a general mechanism by which TAG trafficking is coordinated by lipasin, Angptl3 and Angptl4 at different nutritional statuses.


Angiopoietins/metabolism , Antibodies, Monoclonal/administration & dosage , Lipoprotein Lipase/biosynthesis , Peptide Hormones/immunology , Adipose Tissue/metabolism , Angiopoietin-Like Protein 3 , Angiopoietin-Like Protein 4 , Angiopoietin-Like Protein 8 , Angiopoietin-like Proteins , Animals , Antibodies, Monoclonal/immunology , Humans , Lipoprotein Lipase/immunology , Lipoprotein Lipase/metabolism , Mice , Muscle, Skeletal/metabolism , Myocardium/metabolism , Peptide Hormones/deficiency , Peptide Hormones/therapeutic use , Postprandial Period , Triglycerides/blood
14.
Reprod Biol Endocrinol ; 13: 100, 2015 Sep 04.
Article En | MEDLINE | ID: mdl-26336959

BACKGROUND: Preeclampsia (PE) is associated with maternal and neonatal morbidity and mortality. In PE, the physiological hyperlipidaemia of pregnancy is exaggerated. The purpose of this study was to examine the expression of adipose triglyceride lipase (ATGL), hormone sensitive lipase (HSL), lipoprotein lipase (LPL) and endothelial lipase (EL) in pregnancies complicated by PE. METHODS: Placentae were collected from 16 women with PE and 20 women with uncomplicated pregnancies matched for maternal prepregnancy BMI and gestational age of delivery. Gene and protein expression of the placental lipases were measured by Q-PCR and Western blot. DNA methylation of the promoter of LPL was assessed by bisulfite sequencing. Lipase localisation and activity were analysed. RESULTS: Gene expression of all lipases was significantly reduced, as was HSL protein level in women with PE. All lipases were localised to trophoblasts and endothelial cells in PE and control placentae. There was no difference in methylation of the LPL promoter between PE and control placentae. Lipase activity was not altered in placentae from women with PE. CONCLUSION: These results suggest that the decreased placental lipase gene but not protein expression or lipase activity, which is associated with late-onset PE is not a major contributor to the abnormal lipids seen in PE.


Lipase/biosynthesis , Lipoprotein Lipase/biosynthesis , Placenta/enzymology , Pre-Eclampsia/enzymology , Sterol Esterase/biosynthesis , Adult , Biomarkers/metabolism , Case-Control Studies , Female , Gene Expression Regulation, Developmental , Humans , Lipase/genetics , Lipoprotein Lipase/genetics , Placenta/pathology , Pre-Eclampsia/diagnosis , Pre-Eclampsia/genetics , Pregnancy , Sterol Esterase/genetics
15.
Circulation ; 131(9): 815-26, 2015 Mar 03.
Article En | MEDLINE | ID: mdl-25561514

BACKGROUND: Microvascular endothelium in different organs is specialized to fulfill the particular needs of parenchymal cells. However, specific information about heart capillary endothelial cells (ECs) is lacking. METHODS AND RESULTS: Using microarray profiling on freshly isolated ECs from heart, brain, and liver, we revealed a genetic signature for microvascular heart ECs and identified Meox2/Tcf15 heterodimers as novel transcriptional determinants. This signature was largely shared with skeletal muscle and adipose tissue endothelium and was enriched in genes encoding fatty acid (FA) transport-related proteins. Using gain- and loss-of-function approaches, we showed that Meox2/Tcf15 mediate FA uptake in heart ECs, in part, by driving endothelial CD36 and lipoprotein lipase expression and facilitate FA transport across heart ECs. Combined Meox2 and Tcf15 haplodeficiency impaired FA uptake in heart ECs and reduced FA transfer to cardiomyocytes. In the long term, this combined haplodeficiency resulted in impaired cardiac contractility. CONCLUSIONS: Our findings highlight a regulatory role for ECs in FA transfer to the heart parenchyma and unveil 2 of its intrinsic regulators. Our insights could be used to develop new strategies based on endothelial Meox2/Tcf15 targeting to modulate FA transfer to the heart and remedy cardiac dysfunction resulting from altered energy substrate usage.


Basic Helix-Loop-Helix Transcription Factors/physiology , Endothelial Cells/metabolism , Fatty Acid-Binding Proteins/biosynthesis , Fatty Acids/metabolism , Homeodomain Proteins/physiology , Myocardium/metabolism , Adipose Tissue/blood supply , Animals , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , CD36 Antigens/biosynthesis , CD36 Antigens/genetics , Cardiac Output, Low/etiology , Cardiac Output, Low/genetics , Cardiac Output, Low/metabolism , Cells, Cultured , Coronary Vessels/cytology , Fatty Acid-Binding Proteins/genetics , Glucose/metabolism , Heterozygote , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Humans , Lipoprotein Lipase/biosynthesis , Lipoprotein Lipase/genetics , Lipoproteins, VLDL/metabolism , Mice , Mice, Inbred C57BL , Protein Interaction Mapping , RNA, Small Interfering/pharmacology , Tissue Array Analysis , Transcriptome
16.
Hepatology ; 61(1): 141-52, 2015 Jan.
Article En | MEDLINE | ID: mdl-25131933

UNLABELLED: Identification of microRNAs (miRNAs) that regulate lipid metabolism is important to advance the understanding and treatment of some of the most common human diseases. In the liver, a few key miRNAs have been reported that regulate lipid metabolism, but since many genes contribute to hepatic lipid metabolism, we hypothesized that other such miRNAs exist. To identify genes repressed by miRNAs in mature hepatocytes in vivo, we injected adult mice carrying floxed Dicer1 alleles with an adenoassociated viral vector expressing Cre recombinase specifically in hepatocytes. By inactivating Dicer in adult quiescent hepatocytes we avoided the hepatocyte injury and regeneration observed in previous mouse models of global miRNA deficiency in hepatocytes. Next, we combined gene and miRNA expression profiling to identify candidate gene/miRNA interactions involved in hepatic lipid metabolism and validated their function in vivo using antisense oligonucleotides. A candidate gene that emerged from our screen was lipoprotein lipase (Lpl), which encodes an enzyme that facilitates cellular uptake of lipids from the circulation. Unlike in energy-dependent cells like myocytes, LPL is normally repressed in adult hepatocytes. We identified miR-29a as the miRNA responsible for repressing LPL in hepatocytes, and found that decreasing hepatic miR-29a levels causes lipids to accumulate in mouse livers. CONCLUSION: Our screen suggests several new miRNAs are regulators of hepatic lipid metabolism. We show that one of these, miR-29a, contributes to physiological lipid distribution away from the liver and protects hepatocytes from steatosis. Our results, together with miR-29a's known antifibrotic effect, suggest miR-29a is a therapeutic target in fatty liver disease.


Lipid Metabolism , Lipoprotein Lipase/biosynthesis , Liver/metabolism , MicroRNAs/metabolism , Animals , Enzyme Repression , Fatty Liver/etiology , Hepatocytes/metabolism , Male , Mice, Inbred C57BL
17.
Cell Rep ; 9(1): 336-348, 2014 Oct 09.
Article En | MEDLINE | ID: mdl-25284780

Organisms need to assess their nutritional state and adapt their digestive capacity to the demands for various nutrients. Modulation of digestive enzyme production represents a rational step to regulate nutriment uptake. However, the role of digestion in nutrient homeostasis has been largely neglected. In this study, we analyzed the mechanism underlying glucose repression of digestive enzymes in the adult Drosophila midgut. We demonstrate that glucose represses the expression of many carbohydrases and lipases. Our data reveal that the consumption of nutritious sugars stimulates the secretion of the transforming growth factor ß (TGF-ß) ligand, Dawdle, from the fat body. Dawdle then acts via circulation to activate TGF-ß/Activin signaling in the midgut, culminating in the repression of digestive enzymes that are highly expressed during starvation. Thus, our study not only identifies a mechanism that couples sugar sensing with digestive enzyme expression but points to an important role of TGF-ß/Activin signaling in sugar metabolism.


Activins/metabolism , Glycoside Hydrolases/biosynthesis , Lipoprotein Lipase/biosynthesis , Transforming Growth Factor beta/metabolism , Animals , Drosophila , Glucose/metabolism , Glycoside Hydrolases/metabolism , Lipoprotein Lipase/metabolism , Signal Transduction
18.
Eur Rev Med Pharmacol Sci ; 18(11): 1610-7, 2014 Jun.
Article En | MEDLINE | ID: mdl-24943971

OBJECTIVE: This work aims to investigate the effects of parathyroid hormone-related peptide (PTHrP) (1-86) on osteogenic and adipogenic differentiation of human mesenchymal stem cells (hMSCs) and the related mechanisms. MATERIALS AND METHODS: hMSCs were isolated and cultured in vitro. They were divided into control group, osteogenesis group, adipogenesis group, osteogenesis+PTHrP group and adipogenesis+PTHrP group. The cell proliferation and differentiation, and expression levels of osteopontin (OPN) and lipoprotein lipase (LPL) mRNA were observed. RESULTS: The proliferation rates of hMSCs in osteogenesis+PTHrP and adipogenesis+PTHrP group were significantly higher than that in control group, respectively (p < 0.01). The alkaline phosphatase (ALP)-positive osteoblasts firstly appeared in osteogenesis+PTHrP group, and Sudan IV-positive adipocytes firstly appeared in adipogenesis group. The expression level of OPN mRNA in osteogenesis+PTHrP group was significantly higher than that in osteogenesis group (p < 0.05), and that in adipogenesis+PTHrP group was also higher than adipogenesis group (p < 0.05). The expression level of LPL mRNA in osteogenesis+PTHrP group was significantly lower than that in osteogenesis group, and that in adipogenesis+PTHrP group was also lower than adipogenesis group (p < 0.05). CONCLUSIONS: The osteogenesis and adipogenesis are related to each other during the induced differentiation of hMSCs. PTHrP (1-86) can promote the osteogenic differentiation and inhibits the adipogenic differentiation for hMSCs.


Adipocytes/cytology , Adipocytes/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Parathyroid Hormone-Related Protein/pharmacology , Peptide Fragments/pharmacology , Adipocytes/metabolism , Adipogenesis , Adult , Cell Differentiation/drug effects , Cells, Cultured , Female , Humans , Lipoprotein Lipase/biosynthesis , Lipoprotein Lipase/genetics , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , Osteoblasts/metabolism , Osteogenesis , Osteopontin/biosynthesis , Osteopontin/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
19.
Am J Physiol Endocrinol Metab ; 306(11): E1274-83, 2014 Jun 01.
Article En | MEDLINE | ID: mdl-24735886

In diabetes, when glucose uptake and oxidation are impaired, the heart is compelled to use fatty acid (FA) almost exclusively for ATP. The vascular content of lipoprotein lipase (LPL), the rate-limiting enzyme that determines circulating triglyceride clearance, is largely responsible for this FA delivery and increases following diabetes. Glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein [GPIHBP1; a protein expressed abundantly in the heart in endothelial cells (EC)] collects LPL from the interstitial space and transfers it across ECs onto the luminal binding sites of these cells, where the enzyme is functional. We tested whether ECs respond to hyperglycemia by increasing GPIHBP1. Streptozotocin diabetes increased cardiac LPL activity and GPIHBP1 gene and protein expression. The increased LPL and GPIHBP1 were located at the capillary lumen. In vitro, passaging EC caused a loss of GPIHBP1, which could be induced on exposure to increasing concentrations of glucose. The high-glucose-induced GPIHBP1 increased LPL shuttling across EC monolayers. GPIHBP1 expression was linked to the EC content of heparanase. Moreover, active heparanase increased GPIHBP1 gene and protein expression. Both ECs and myocyte heparan sulfate proteoglycan-bound platelet-derived growth factor (PDGF) released by heparanase caused augmentation of GPIHBP1. Overall, our data suggest that this protein "ensemble" (heparanase-PDGF-GPIHBP1) cooperates in the diabetic heart to regulate FA delivery and utilization by the cardiomyocytes. Interrupting this axis may be a novel therapeutic strategy to restore metabolic equilibrium, curb lipotoxicity, and help prevent or delay heart dysfunction that is characteristic of diabetes.


Endothelial Cells/metabolism , Hyperglycemia/metabolism , Lipoprotein Lipase/biosynthesis , Receptors, Lipoprotein/biosynthesis , Animals , Biological Transport, Active/physiology , Blotting, Western , Cattle , Coculture Techniques , Cytokines/biosynthesis , Diabetes Mellitus, Experimental/metabolism , Fluorescent Antibody Technique , Glucose/pharmacology , Glucuronidase/metabolism , Lipolysis/physiology , Male , Monocytes/metabolism , Myocytes, Cardiac/metabolism , Platelet-Derived Growth Factor/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
20.
Biochem Biophys Res Commun ; 443(2): 428-34, 2014 Jan 10.
Article En | MEDLINE | ID: mdl-24309104

Atherosclerosis is a lipid disorder disease characterized by chronic blood vessel wall inflammation driven by the subendothelial accumulation of macrophages. Studies have shown that lipoprotein lipase (LPL) participates in lipid metabolism, but it is not yet known whether post-transcriptional regulation of LPL gene expression by microRNAs (miRNAs) occurs in vivo. Here, we tested that miR-467b provides protection against atherosclerosis by regulating the target gene LPL which leads to reductions in LPL expression, lipid accumulation, progression of atherosclerosis and production of inflammatory cytokines in apolipoprotein E knockout (apoE(-/-)) mice. Treatment of apoE(-/-) mice with intra-peritoneal injection of miR-467b agomir led to decreased blood plasma levels of total cholesterol (TC), triglyceride (TG), low density lipoprotein cholesterol (LDL-C), high density lipoprotein cholesterol (HDL-C), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), IL-1ß and monocyte chemotactic protein-1 (MCP-1). Using Western blots and real time PCR, we determined that LPL expression in aorta and abdominal cavity macrophages were significantly down-regulated in the miR-467b agomir group. Furthermore, systemic treatment with miR-467b antagomir accelerated the progression of atherosclerosis in the aorta of apoE(-/-) mice. The present study showed that miR-467b protects apoE(-/-) mice from atherosclerosis by reducing lipid accumulation and inflammatory cytokine secretion via downregulation of LPL expression. Therefore, targeting miR-467b may offer a promising strategy to treat atherosclerotic vascular disease.


Apolipoproteins E/genetics , Atherosclerosis/immunology , Cytokines/immunology , Inflammation/immunology , Lipid Metabolism/immunology , Lipoprotein Lipase/immunology , MicroRNAs/pharmacology , Animals , Atherosclerosis/prevention & control , Gene Expression Regulation, Enzymologic/drug effects , Inflammation/prevention & control , Lipid Metabolism/drug effects , Lipoprotein Lipase/biosynthesis , Male , Mice , Mice, Knockout , Treatment Outcome
...