Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Cell Mol Med ; 24(18): 10730-10743, 2020 09.
Article in English | MEDLINE | ID: mdl-32745373

ABSTRACT

Insufficient endometrial angiogenesis during peri-implantation impairs endometrial receptivity (ER), which contributes to recurrent implantation failure (RIF) during in vitro fertilization and embryo transfer (IVF-ET). Angiopoietin-like protein 4 (ANGPTL4) acts as a multifunctional secretory protein and is involved in the regulation of lipid metabolism and angiogenesis in various tissues including the endometrium. Herein, we found decreased ANGPTL4 expression in endometrial tissue and serum during peri-implantation period in 18 RIF-affected women with elevated uterine arterial impedance (UAI) compared with the pregnancy controls. ANGPTL4 and peroxisome proliferator-activated receptor gamma (PPARγ) expression were up-regulated upon decidualization on human endometrial stromal cells (HESCs). Rosiglitazone promoted the expression of ANGPTL4 in HESCs and human umbilical vein endothelial cells (HUVECs) via PPARγ. ANGPTL4 promoted the proliferation, migration and angiogenesis of HUVECs in vitro. Our results suggest that decreased abundance of ANGPTL4 in endometrial tissues impairs the endometrial receptivity via restraining endometrial angiogenesis during decidualization; while rosiglitazone-induced ANGPTL4 up-regulation in hESCs and HUVECs through PPARγ. Therefore, ANGPTL4 could be a potential therapeutic approach for some RIF-affected women with elevated UAI.


Subject(s)
Angiopoietin-Like Protein 4/physiology , Embryo Implantation/physiology , Embryo Loss/physiopathology , Endometrium/blood supply , Neovascularization, Physiologic/physiology , Adult , Angiopoietin-Like Protein 4/biosynthesis , Angiopoietin-Like Protein 4/deficiency , Angiopoietin-Like Protein 4/genetics , Cell Line , Decidua/pathology , Electric Impedance , Embryo Loss/blood , Embryo Loss/pathology , Embryo Transfer , Endometrium/metabolism , Endometrium/pathology , Female , Fertilization in Vitro , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells , Humans , PPAR gamma/agonists , PPAR gamma/biosynthesis , PPAR gamma/genetics , Pregnancy , Recurrence , Rosiglitazone/pharmacology , Sperm Injections, Intracytoplasmic , Stromal Cells/metabolism , Uterine Artery/physiopathology , Young Adult
2.
Artif Cells Nanomed Biotechnol ; 47(1): 3368-3373, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31387395

ABSTRACT

Diabetic nephropathy (DN) is one of the major diabetic complications that lead to end-stage renal failure. Angiopoietin-like protein-4 (ANGPTL-4) has been reported to be dysregulated in diabetes mellitus and diabetic complications. However, the role of ANGPTL-4 in glomerular mesangial cells (MCs) during DN remains unclear. In the present study, we evaluated the role of ANGPTL-4 in MCs in response to high glucose (HG) condition and the potential mechanism. The results proved that ANGPTL-4 expression is significantly increased in HG-stimulated MCs. Knockdown of ANGPTL-4 suppressed HG-induced cell proliferation of MCs. The production of pro-inflammatory cytokines including TNF-α, IL-1ß, IL-6 were decreased in ANGPTL-4 knocked down MCs. Inhibition of ANGPTL-4 markedly suppressed the expressions of extracellular matrix (ECM) proteins, collagen IV (Col IV) and fibronectin (FN), in HG-stimulated MCs. Furthermore, ANGPTL-4 knockdown inhibited the HG-induced activation of NF-κB signaling pathway in MCs. Collectively, knockdown of ANGPTL-4 suppressed HG-induced cell proliferation, inflammatory response, and ECM accumulation inhibiting NF-κB signaling pathway in MCs. These findings suggested that ANGPTL-4 might be a therapeutic target for the prevention and treatment of DN.


Subject(s)
Angiopoietin-Like Protein 4/deficiency , Angiopoietin-Like Protein 4/genetics , Extracellular Matrix/metabolism , Gene Knockdown Techniques , Glucose/pharmacology , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Animals , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/genetics , Collagen Type IV/biosynthesis , Dose-Response Relationship, Drug , Extracellular Matrix/drug effects , Fibronectins/biosynthesis , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mesangial Cells/pathology , NF-kappa B/metabolism , Rats , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation/drug effects
3.
J Lipid Res ; 60(10): 1741-1754, 2019 10.
Article in English | MEDLINE | ID: mdl-31409739

ABSTRACT

Angiopoietin-like protein (ANGPTL)4 regulates plasma lipids, making it an attractive target for correcting dyslipidemia. However, ANGPTL4 inactivation in mice fed a high fat diet causes chylous ascites, an acute-phase response, and mesenteric lymphadenopathy. Here, we studied the role of ANGPTL4 in lipid uptake in macrophages and in the above-mentioned pathologies using Angptl4-hypomorphic and Angptl4-/- mice. Angptl4 expression in peritoneal and bone marrow-derived macrophages was highly induced by lipids. Recombinant ANGPTL4 decreased lipid uptake in macrophages, whereas deficiency of ANGPTL4 increased lipid uptake, upregulated lipid-induced genes, and increased respiration. ANGPTL4 deficiency did not alter LPL protein levels in macrophages. Angptl4-hypomorphic mice with partial expression of a truncated N-terminal ANGPTL4 exhibited reduced fasting plasma triglyceride, cholesterol, and NEFAs, strongly resembling Angptl4-/- mice. However, during high fat feeding, Angptl4-hypomorphic mice showed markedly delayed and attenuated elevation in plasma serum amyloid A and much milder chylous ascites than Angptl4-/- mice, despite similar abundance of lipid-laden giant cells in mesenteric lymph nodes. In conclusion, ANGPTL4 deficiency increases lipid uptake and respiration in macrophages without affecting LPL protein levels. Compared with the absence of ANGPTL4, low levels of N-terminal ANGPTL4 mitigate the development of chylous ascites and an acute-phase response in mice.


Subject(s)
Adipocytes/metabolism , Angiopoietin-Like Protein 4/deficiency , Angiopoietin-Like Protein 4/genetics , Gene Knockout Techniques , Macrophages/metabolism , Animals , Cell Respiration , Chylous Ascites/genetics , Chylous Ascites/pathology , Exons/genetics , Gene Expression Regulation , Lipoprotein Lipase/metabolism , Lymphadenopathy/genetics , Lymphadenopathy/pathology , Mice , Mice, Inbred C57BL , Triglycerides/blood
4.
J Biol Chem ; 294(23): 9213-9224, 2019 06 07.
Article in English | MEDLINE | ID: mdl-31053639

ABSTRACT

Chronic or excess glucocorticoid exposure causes lipid disorders such as hypertriglyceridemia and hepatic steatosis. Angptl4 (angiopoietin-like 4), a primary target gene of the glucocorticoid receptor in hepatocytes and adipocytes, is required for hypertriglyceridemia and hepatic steatosis induced by the synthetic glucocorticoid dexamethasone. Angptl4 has also been shown to be required for dexamethasone-induced hepatic ceramide production. Here, we further examined the role of ceramide-mediated signaling in hepatic dyslipidemia caused by chronic glucocorticoid exposure. Using a stable isotope-labeling technique, we found that dexamethasone treatment induced the rate of hepatic de novo lipogenesis and triglyceride synthesis. These dexamethasone responses were compromised in Angptl4-null mice (Angptl4-/-). Treating mice with myriocin, an inhibitor of the rate-controlling enzyme of de novo ceramide synthesis, serine palmitoyltransferase long-chain base subunit 1 (SPTLC1)/SPTLC2, decreased dexamethasone-induced plasma and liver triglyceride levels in WT but not Angptl4-/- mice. We noted similar results in mice infected with adeno-associated virus-expressing small hairpin RNAs targeting Sptlc2. Protein phosphatase 2 phosphatase activator (PP2A) and protein kinase Cζ (PKCζ) are two known downstream effectors of ceramides. We found here that mice treated with an inhibitor of PKCζ, 2-acetyl-1,3-cyclopentanedione (ACPD), had lower levels of dexamethasone-induced triglyceride accumulation in plasma and liver. However, small hairpin RNA-mediated targeting of the catalytic PP2A subunit (Ppp2ca) had no effect on dexamethasone responses on plasma and liver triglyceride levels. Overall, our results indicate that chronic dexamethasone treatment induces an ANGPTL4-ceramide-PKCζ axis that activates hepatic de novo lipogenesis and triglyceride synthesis, resulting in lipid disorders.


Subject(s)
Angiopoietin-Like Protein 4/metabolism , Ceramides/metabolism , Dexamethasone/toxicity , Liver/drug effects , Protein Kinase C/metabolism , Angiopoietin-Like Protein 4/deficiency , Angiopoietin-Like Protein 4/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Fatty Acids, Monounsaturated/pharmacology , Fatty Liver/etiology , Fatty Liver/metabolism , Hypertriglyceridemia/etiology , Hypertriglyceridemia/metabolism , Lipogenesis/drug effects , Liver/metabolism , Male , Mice , Mice, Knockout , Protein Kinase C/antagonists & inhibitors , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Serine C-Palmitoyltransferase/antagonists & inhibitors , Serine C-Palmitoyltransferase/genetics , Serine C-Palmitoyltransferase/metabolism , Triglycerides/blood , Triglycerides/metabolism
5.
Eur Rev Med Pharmacol Sci ; 22(16): 5056-5062, 2018 08.
Article in English | MEDLINE | ID: mdl-30178822

ABSTRACT

OBJECTIVE: To explore ANGPTL4 expressions in patients with gestational diabetes mellitus (GDM) and its underlying mechanism. PATIENTS AND METHODS: We first detected serum expressions of ANGPTL4 in GDM patients and healthy pregnancies. Subsequently, effects of ANGPTL4 knockdown on apoptosis, proliferation, and cell cycle in 3T3-L1 cells were determined, respectively. Effects of ANGPTL4 on glucose uptake and adipocyte differentiation were also evaluated, respectively. The cytokine secretion in adipocytes transfected with sh-ANGPTL4 was detected by quantitative Reverse Transcriptase-Polymerase Chain Reaction (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA). Furthermore, effects of ANGPTL4 knockdown on NF-kB and Akt pathway were detected by Western blot. RESULTS: ANGPTL4 was down-regulated in serum of GDM patients. In vitro experiments suggested that down-regulated ANGPTL4 inhibited apoptosis and promoted proliferation of 3T3-L1 cells. Meanwhile, down-regulated ANGPTL4 significantly inhibited glucose uptake and Akt pathway. However, ANGPTL4 expression did not affect cell cycle and adipocyte differentiation. Detection of inflammatory cytokines suggested that down-regulated ANGPTL4 resulted in increased expressions of inflammatory cytokines and activation of NF-kB pathway. CONCLUSIONS: ANGPTL4 is down-regulated in GDM and may participate in the GDM development by promoting insulin resistance and secretion of inflammatory cytokines.


Subject(s)
Angiopoietin-Like Protein 4/metabolism , Diabetes, Gestational/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , 3T3-L1 Cells , Adipocytes/metabolism , Adult , Angiopoietin-Like Protein 4/deficiency , Angiopoietin-Like Protein 4/genetics , Animals , Diabetes, Gestational/genetics , Female , Humans , Inflammation Mediators/metabolism , Insulin Resistance/physiology , Male , Mice , Pregnancy , Proto-Oncogene Proteins c-akt/genetics
6.
Nat Commun ; 9(1): 2252, 2018 06 13.
Article in English | MEDLINE | ID: mdl-29899519

ABSTRACT

Angiopoietin-like 4 (ANGPTL4) is an endogenous inhibitor of lipoprotein lipase that modulates lipid levels, coronary atherosclerosis risk, and nutrient partitioning. We hypothesize that loss of ANGPTL4 function might improve glucose homeostasis and decrease risk of type 2 diabetes (T2D). We investigate protein-altering variants in ANGPTL4 among 58,124 participants in the DiscovEHR human genetics study, with follow-up studies in 82,766 T2D cases and 498,761 controls. Carriers of p.E40K, a variant that abolishes ANGPTL4 ability to inhibit lipoprotein lipase, have lower odds of T2D (odds ratio 0.89, 95% confidence interval 0.85-0.92, p = 6.3 × 10-10), lower fasting glucose, and greater insulin sensitivity. Predicted loss-of-function variants are associated with lower odds of T2D among 32,015 cases and 84,006 controls (odds ratio 0.71, 95% confidence interval 0.49-0.99, p = 0.041). Functional studies in Angptl4-deficient mice confirm improved insulin sensitivity and glucose homeostasis. In conclusion, genetic inactivation of ANGPTL4 is associated with improved glucose homeostasis and reduced risk of T2D.


Subject(s)
Angiopoietin-Like Protein 4/deficiency , Angiopoietin-Like Protein 4/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Amino Acid Substitution , Angiopoietin-Like Protein 4/metabolism , Animals , Blood Glucose/metabolism , Case-Control Studies , Diabetes Mellitus, Type 2/etiology , Female , Gene Silencing , Genetic Association Studies , Genetic Variation , Heterozygote , Homeostasis , Humans , Insulin Resistance/genetics , Lipoprotein Lipase/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Risk Factors , Exome Sequencing
7.
J Lipid Res ; 59(7): 1230-1243, 2018 07.
Article in English | MEDLINE | ID: mdl-29739862

ABSTRACT

Mice lacking glycosylphosphatidylinositol-anchored HDL-binding protein 1 (GPIHBP1) are unable to traffic LPL to the vascular lumen. Thus, triglyceride (TG) clearance is severely blunted, and mice are extremely hypertriglyceridemic. Paradoxically, mice lacking both GPIHBP1 and the LPL regulator, angiopoietin-like 4 (ANGPTL4), are far less hypertriglyceridemic. We sought to determine the mechanism by which Angptl4-/-Gpihbp1-/- double-knockout mice clear plasma TGs. We confirmed that, on a normal chow diet, plasma TG levels were lower in Angptl4-/-Gpihbp1-/- mice than in Gpihbp1-/- mice; however, the difference disappeared with administration of a high-fat diet. Although LPL remained mislocalized in double-knockout mice, plasma TG clearance in brown adipose tissue (BAT) increased compared with Gpihbp1-/- mice. Whole lipoprotein uptake was observed in the BAT of both Gpihbp1-/- and Angptl4-/-Gpihbp1-/- mice, but BAT lipase activity was significantly higher in the double-knockout mice. We conclude that Angptl4-/-Gpihbp1-/- mice clear plasma TGs primarily through a slow and noncanonical pathway that includes the uptake of whole lipoprotein particles.


Subject(s)
Angiopoietin-Like Protein 4/deficiency , Receptors, Lipoprotein/deficiency , Triglycerides/blood , Adipose Tissue, Brown/metabolism , Angiopoietin-Like Protein 4/genetics , Animals , Gene Expression Regulation, Enzymologic , Gene Knockout Techniques , Lipoprotein Lipase/metabolism , Mice , Protein Transport , Receptors, Lipoprotein/genetics , Receptors, Lipoprotein/metabolism
8.
J Am Heart Assoc ; 6(6)2017 Jun 23.
Article in English | MEDLINE | ID: mdl-28645936

ABSTRACT

BACKGROUND: ANGPTL4 (angiopoietin-like protein 4) is a LPL (lipoprotein lipase) inhibitor and is present in high-density lipoprotein (HDL). However, it is not defined whether ANGPTL4 in HDLs could affect HDL metabolism and function in type 2 diabetes mellitus (T2DM). METHODS AND RESULTS: ANGPTL4 levels in the circulation and HDLs were quantified in nondiabetic participants (n=201, 68.7% females) and T2DM patients (n=185, 66.5% females). HDLs were isolated from nondiabetic controls and T2DM patients to assess cholesterol efflux or subjected to endothelial lipase (EL)-overexpressed HEK293 cells for EL hydrolysis in vitro. The association between ANGPTL4 in HDLs and HDL components and function was analyzed in nondiabetic participants or diabetic patients, respectively. Plasma or HDLs of ANGPTL4+/+ and ANGPTL4-/- mice was subjected for cholesterol efflux or EL hydrolysis, respectively. ANGPTL4 levels in the plasma and HDLs were 1.7- and 2.0-fold higher in T2DM patients than nondiabetic controls, respectively (P<0.0001). Multivariable analysis demonstrated that per 1 doubling increase of ANGPTL4 levels in HDLs, the changes amounted to +0.27% cholesterol efflux (P=0.03), +0.06 µg/mL apolipoprotein A-I (P=0.09) and -9.41 µg/L serum amyloid A (P=0.02) in nondiabetic controls. In T2DM patients, the corresponding estimates were -0.06% cholesterol efflux (P=0.10), -0.06 µg/mL apolipoprotein A-I (P=0.38), and +3.64 µg/L serum amyloid A (P=0.72). HDLs isolated from ANGPTL4-/- mice showed accelerated hydrolysis by EL and reduced cholesterol efflux compared with ANGPTL4+/+ littermates. CONCLUSIONS: Physically, ANGPTL4 in HDLs protected HDLs from hydrolysis. Resulting from increased circulating ANGPTL4 levels in T2DM, ANGPTL4 levels in HDLs were elevated but with compromised inhibitory effect on EL, leading to increased HDL hydrolysis and dysfunction.


Subject(s)
Angiopoietin-Like Protein 4/blood , Diabetes Mellitus, Type 2/blood , Lipoproteins, HDL/blood , Angiopoietin-Like Protein 4/deficiency , Angiopoietin-Like Protein 4/genetics , Animals , Biomarkers/blood , Case-Control Studies , Cholesterol/blood , Female , HEK293 Cells , Humans , Hydrolysis , Lipase/genetics , Lipase/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Transfection
9.
J Lipid Res ; 58(6): 1100-1113, 2017 06.
Article in English | MEDLINE | ID: mdl-28412693

ABSTRACT

Angiopoietin-like 4 (ANGPTL4) regulates plasma triglyceride levels by inhibiting LPL. Inactivation of ANGPTL4 decreases plasma triglycerides and reduces the risk of coronary artery disease. Unfortunately, targeting ANGPTL4 for the therapeutic management of dyslipidemia and atherosclerosis is hampered by the observation that mice and monkeys in which ANGPTL4 is inactivated exhibit lipid accumulation in the mesenteric lymph nodes (MLNs). In mice these pathological events exclusively unfold upon feeding a high saturated FA diet and are followed by an ultimately lethal pro-inflammatory response and chylous ascites. Here, we show that Angptl4-/- mice fed a diet rich in trans FAs develop numerous lipid-filled giant cells in their MLNs, yet do not have elevated serum amyloid and haptoglobin, do not exhibit ascites, and survive, unlike Angptl4-/- mice fed a saturated FA-rich diet. In RAW264.7 macrophages, the saturated FA, palmitate, markedly increased markers of inflammation and the unfolded protein response, whereas the trans-unsaturated elaidate and the cis-unsaturated oleate had the opposite effect. In conclusion, trans and saturated FAs have very distinct biological effects in macrophages. Furthermore, lipid accumulation in MLNs is uncoupled from activation of an acute-phase response and chylous ascites, suggesting that ANGPTL4 should not be fully dismissed as target for dyslipidemia.


Subject(s)
Angiopoietin-Like Protein 4/deficiency , Chylous Ascites/chemically induced , Dietary Fats/adverse effects , Foam Cells/drug effects , Lymph Nodes/drug effects , Mesentery , Trans Fatty Acids/adverse effects , Acute-Phase Proteins/metabolism , Animals , Chylous Ascites/metabolism , Chylous Ascites/pathology , Foam Cells/cytology , Foam Cells/pathology , Giant Cells/drug effects , Giant Cells/pathology , Liver/drug effects , Liver/metabolism , Lymph Nodes/cytology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Peritonitis/chemically induced , Peritonitis/metabolism , Peritonitis/pathology , RAW 264.7 Cells
10.
Nat Commun ; 7: 12313, 2016 07 27.
Article in English | MEDLINE | ID: mdl-27460411

ABSTRACT

Lipid accumulation in macrophages has profound effects on macrophage gene expression and contributes to the development of atherosclerosis. Here, we report that angiopoietin-like protein 4 (ANGPTL4) is the most highly upregulated gene in foamy macrophages and it's absence in haematopoietic cells results in larger atherosclerotic plaques, characterized by bigger necrotic core areas and increased macrophage apoptosis. Furthermore, hyperlipidemic mice deficient in haematopoietic ANGPTL4 have higher blood leukocyte counts, which is associated with an increase in the common myeloid progenitor (CMP) population. ANGPTL4-deficient CMPs have higher lipid raft content, are more proliferative and less apoptotic compared with the wild-type (WT) CMPs. Finally, we observe that ANGPTL4 deficiency in macrophages promotes foam cell formation by enhancing CD36 expression and reducing ABCA1 localization in the cell surface. Altogether, these findings demonstrate that haematopoietic ANGPTL4 deficiency increases atherogenesis through regulating myeloid progenitor cell expansion and differentiation, foam cell formation and vascular inflammation.


Subject(s)
Angiopoietin-Like Protein 4/deficiency , Atherosclerosis/metabolism , Atherosclerosis/pathology , Disease Progression , Hematopoietic Stem Cells/metabolism , Monocytes/metabolism , Angiopoietin-Like Protein 4/metabolism , Animals , Apoptosis , Atherosclerosis/complications , Bone Marrow Transplantation , Cell Proliferation , Cell Survival , Foam Cells/metabolism , Humans , Inflammation/complications , Inflammation/pathology , Leukocytosis/complications , Leukocytosis/pathology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Biological , Myeloid Progenitor Cells/metabolism , Plaque, Atherosclerotic/complications , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...