Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Obes Res Clin Pract ; 12(Suppl 2): 71-79, 2018.
Article in English | MEDLINE | ID: mdl-28320596

ABSTRACT

OBJECTIVE: Previous investigations have shown that the peroxisome proliferator activated receptor beta/delta (PPAR/)-angiopoietin-like protein 4 (ANGPTL4) pathways may be a new pharmacologic target for treatment of obesity. The present study was conducted to test the effect of phillyrin, a glucoside, on obesity in mice. METHOD: Fifty mice were randomly divided into 5 groups (n=10): control group (C57BL/6J mice), obese mice group, two groups of obese mice treated with phillyrin (15 or 45mg/kg/day), one group of obese mice treated with PPAR/ agonist GW0742 (3mg/kg/day). Twelve weeks after treatment, body weight, liver weight, fat weight, lipid levels in the liver, serum levels of tumour necrosis factor-(TNF-), leptin, and insulin, expression of PPAR/, ANGPTL4, and AMP-activated protein kinase (AMPK) were determined. RESULTS: Treatment with phillyrin (15 or 45mg/kg) significantly decreased body weight, liver weight, fat weight, hepatic total cholesterol, free fatty acid, and triglyceride concentrations, serum levels of TNF-, leptin, and insulin concomitantly with up-regulated expression of PPAR/, ANGPTL4, and p-AMPK-. In addition, GW0742 has similar effect of phillyrin. CONCLUSIONS: The present results suggest that phillyrin could regulate the PPAR/-ANGPTL 4 pathway to lower body weight in obese C57BL/6J mice.


Subject(s)
Angiopoietins/biosynthesis , Glucosides/pharmacology , Obesity/drug therapy , PPAR alpha/biosynthesis , Signal Transduction/drug effects , Angiopoietins/genetics , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , PPAR alpha/genetics
2.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(5): 533-540, 2017 May.
Article in English | MEDLINE | ID: mdl-28215713

ABSTRACT

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.


Subject(s)
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
3.
EMBO J ; 36(4): 409-424, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28043948

ABSTRACT

The intestinal epithelium continually self-renews and can rapidly regenerate after damage. Dysregulation of intestinal epithelial homeostasis leads to severe inflammatory bowel disease. Additionally, aberrant signaling by the secreted protein angiopoietin-like protein 2 (ANGPTL2) causes chronic inflammation in a variety of diseases. However, little is known about the physiologic role of ANGPTL2 in normal tissue homeostasis and during wound repair following injury. Here, we assessed ANGPTL2 function in intestinal physiology and disease in vivo Although intestinal development proceeded normally in Angptl2-deficient mice, expression levels of the intestinal stem cell (ISC) marker gene Lgr5 decreased, which was associated with decreased transcriptional activity of ß-catenin in Angptl2-deficient mice. Epithelial regeneration after injury was significantly impaired in Angptl2-deficient relative to wild-type mice. ANGPTL2 was expressed and functioned within the mesenchymal compartment cells known as intestinal subepithelial myofibroblasts (ISEMFs). ANGPTL2 derived from ISEMFs maintained the intestinal stem cell niche by modulating levels of competing signaling between bone morphogenetic protein (BMP) and ß-catenin. These results support the importance of ANGPTL2 in the stem cell niche in regulating stemness and epithelial wound healing in the intestine.


Subject(s)
Angiopoietins/biosynthesis , Gene Expression Regulation , Homeostasis , Intestinal Mucosa/injuries , Intestinal Mucosa/physiology , Regeneration , Stem Cell Niche , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins , Angiopoietins/deficiency , Animals , Disease Models, Animal , Mice , Mice, Knockout , Receptors, G-Protein-Coupled/analysis , Wound Healing , beta Catenin/analysis
4.
Clin Exp Pharmacol Physiol ; 44(1): 123-131, 2017 01.
Article in English | MEDLINE | ID: mdl-27712004

ABSTRACT

Increased pulmonary vascular resistance is a critical complication in sepsis. Toll-like receptor (TLR) as well as angiopoietin (ANG) signalling both contribute to the emergence of pulmonary arterial hypertension. We hypothesized that TLR stimulation by bacterial ligands directly affects expression and secretion of ligands and receptors of the angiopoietin/TIE axis. Microvascular endothelial (HPMEC) and smooth muscle cells (SMC) of pulmonary origin were incubated with thrombin and with ligands for TLR2, -4, -5, and -9. Expression and secretion of ANG1, -2, TIE2 and IL-8 were determined using quantitative real-time PCR and ELISA. TLR stimulation had no impact either on the expression of ANG2 and TIE2 in HPMEC or on that of ANG1 in SMC. However, overall levels of both released ANG1 and -2 were halved upon stimulation with the TLR9 ligand CpG, and ANG2 release was significantly enhanced by TLR4 activation when initially provoked by sequentially performed stimulation. Furthermore, enhanced ANG2 activity increased endothelial permeability, as demonstrated in an in vitro transwell assay. We conclude that sole TLR stimulation by bacterial ligands plays no significant role for altered expression and secretion of ANG1, -2 and TIE2 in human pulmonary vascular cells. The interplay between various stimuli is required to induce imbalances between ANG1 and -2.


Subject(s)
Angiopoietin-1/biosynthesis , Angiopoietin-2/biosynthesis , Pulmonary Artery/metabolism , Receptor, TIE-2/biosynthesis , Toll-Like Receptors/biosynthesis , Angiopoietins/biosynthesis , Cells, Cultured , Flagellin/toxicity , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation Mediators/metabolism , Microvessels/drug effects , Microvessels/metabolism , Pneumonia/chemically induced , Pneumonia/metabolism , Pulmonary Artery/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
5.
PLoS One ; 11(7): e0159747, 2016.
Article in English | MEDLINE | ID: mdl-27438462

ABSTRACT

Adipose tissue inflammation is believed to play a pivotal role in the development obesity-related morbidities such as insulin resistance. However, it is not known how this (low-grade) inflammatory state develops. It has been proposed that the leakage of lipopolysaccharides (LPS), originating from the gut microbiota, through the gut epithelium could drive initiation of inflammation. To get a better understanding of which proteins and intracellular pathways are affected by LPS in adipocytes, we performed SILAC proteomic analysis and identified proteins that were altered in expression. Furthermore, we tested the anti-inflammatory compound resveratrol. A total of 927 proteins were quantified by the SILAC method and of these 57- and 64 were significantly up- and downregulated by LPS, respectively. Bioinformatic analysis (GO analysis) revealed that the upregulated proteins were especially involved in the pathways of respiratory electron transport chain and inflammation. The downregulated proteins were especially involved in protein glycosylation. One of the latter proteins, GALNT2, has previously been described to regulate the expression of liver lipases such as ANGPTL3 and apoC-III affecting lipid metabolism. Furthermore, LPS treatment reduced the protein levels of the insulin sensitizing adipokine, adiponectin, and proteins participating in the final steps of triglyceride- and cholesterol synthesis. Generally, resveratrol opposed the effect induced by LPS and, as such, functioning as an ameliorating factor in disease state. Using an unbiased proteomic approach, we present novel insight of how the proteome is altered in adipocytes in response to LPS as seen in obesity. We suggest that LPS partly exerts its detrimental effects by altering glycosylation processes of the cell, which is starting to emerge as important posttranscriptional regulators of protein expression. Furthermore, resveratrol could be a prime candidate in ameliorating dysfunctioning adipose tissue induced by inflammatory stimulation.


Subject(s)
Inflammation/drug therapy , Insulin Resistance/genetics , Lipopolysaccharides/metabolism , Obesity/genetics , Stilbenes/administration & dosage , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Angiopoietin-Like Protein 3 , Angiopoietin-like Proteins , Angiopoietins/biosynthesis , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Gene Expression Regulation/drug effects , Glycosylation/drug effects , Humans , Inflammation/genetics , Inflammation/pathology , Insulin/metabolism , Lipid Metabolism , Lipogenesis/drug effects , N-Acetylgalactosaminyltransferases/biosynthesis , Obesity/drug therapy , Obesity/pathology , Proteome/genetics , Proteomics , Resveratrol , Polypeptide N-acetylgalactosaminyltransferase
6.
Clin Exp Metastasis ; 33(5): 417-29, 2016 06.
Article in English | MEDLINE | ID: mdl-26957434

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive malignant brain tumor. Since differentiation can attenuate or halt the growth of tumor cells, an image-based phenotypic screening was performed to find out drugs inducing morphological differentiation of GBMs. Bexarotene, a selective retinoid X receptor agonist, showed strong inhibition of neurospheroidal colony formation and migration of cultured primary GBM cells. Bexarotene treatment reduced nestin expression, while significantly increasing glial fibrillary acidic protein (GFAP) expression. The effect of bexarotene on gene expression profile was compared with the activity of all-trans retinoic acid (ATRA), a well-known differentiation inducer. Both drugs largely altered the gene expression pattern into a tumor-ameliorating direction. These drugs increased the gene expression levels of Krüppel-like factor 9 (KLF9), regulator of G-protein signaling 4 (RGS4), growth differentiation factor 15 (GDF15), angiopoietin-like protein 4 (ANGPTL4), and lowered the level of chemokine receptor type 4 (CXCR4). However, transglutaminase 2 (TG2) induction, an adverse effect of ATRA, was much weaker in bexarotene treated primary GBM cells. Consistently, the TG2 enzymatic activity was negligibly affected by bexarotene treatment. It is important to control TG2 overexpression since its upregulation is correlated with tumor transformation and drug resistance. Bexarotene also showed in vivo tumoricidal effects in a GBM xenograft mouse model. Therefore, we suggest bexarotene as a more beneficial differentiation agent than ATRA for GBM.


Subject(s)
Cell Differentiation/drug effects , GTP-Binding Proteins/genetics , Glioblastoma/drug therapy , Tetrahydronaphthalenes/administration & dosage , Transglutaminases/genetics , Angiopoietin-Like Protein 4 , Angiopoietins/biosynthesis , Animals , Bexarotene , Cell Line, Tumor , Cell Movement/drug effects , GTP-Binding Proteins/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/diagnostic imaging , Glioblastoma/genetics , Glioblastoma/pathology , Growth Differentiation Factor 15/biosynthesis , Humans , Kruppel-Like Transcription Factors/biosynthesis , Mice , Protein Glutamine gamma Glutamyltransferase 2 , RGS Proteins/biosynthesis , Receptors, CXCR4/biosynthesis , Retinoid X Receptors/agonists , Signal Transduction/drug effects , Transglutaminases/biosynthesis , Tretinoin/administration & dosage , Xenograft Model Antitumor Assays
7.
J Biol Chem ; 291(14): 7373-85, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26839315

ABSTRACT

Choroidal neovascularization (CNV) is a pathogenic process of age-related macular degeneration, a vision-threatening disease. The retinal pigment epithelium and macrophages both influence CNV development. However, the underlying mechanisms remain obscure. Here, we focus on Angptl2 (angiopoietin-like protein 2), a cytokine involved in age-related systemic diseases. Angptl2 was originally identified as an adipocytokine and is also expressed in the eye. Using a laser-induced CNV model, we found thatAngptl2KO mice exhibited suppressed CNV development with reduced macrophage recruitment and inflammatory mediator induction. The mediators monocyte chemotactic protein-1, interleukin-1ß (Il-1ß),Il-6, matrix metalloprotease-9 (Mmp-9), and transforming growth factor-ß1 (Tgf-ß1) that were up-regulated during CNV development were all suppressed in the retinal pigment epithelium-choroid of CNV models generated in theAngptl2KO mice. Bone marrow transplantation using wild-type and KO mice suggested that both bone marrow-derived and host-derived Angptl2 were responsible for macrophage recruitment and CNV development. Peritoneal macrophages derived fromAngptl2KO mice expressed lower levels of the inflammatory mediators. In the wild-type peritoneal macrophages and RAW264.7 cells, Angptl2 induced the mediators via integrins α4 and ß2, followed by the downstream activation of NF-κB and ERK. The activation of NF-κB and ERK by Angptl2 also promoted macrophage migration. Therefore, Angptl2 from focal tissue might trigger macrophage recruitment, and that from recruited macrophages might promote expression of inflammatory mediators including Angptl2 in an autocrine and/or paracrine fashion to facilitate CNV development. Angptl2 might therefore represent a multistep regulator of CNV pathogenesis and serve as a new therapeutic target for age-related macular degeneration.


Subject(s)
Angiopoietins/biosynthesis , Choroidal Neovascularization/metabolism , Macrophages/metabolism , Macular Degeneration/metabolism , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins , Angiopoietins/genetics , Animals , CD18 Antigens/genetics , CD18 Antigens/metabolism , Cell Line , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Choroidal Neovascularization/genetics , Choroidal Neovascularization/pathology , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Integrin alpha4/genetics , Integrin alpha4/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Macrophages/pathology , Macular Degeneration/genetics , Macular Degeneration/pathology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
8.
Eur Rev Med Pharmacol Sci ; 20(1): 44-53, 2016.
Article in English | MEDLINE | ID: mdl-26813452

ABSTRACT

OBJECTIVE: Chronic obstructive pulmonary disease (COPD) is both a pulmonary and systematic disease, which will cause abnormal expression of some circulating factors. Angiopoietin-like protein 4 (ANGPTL4) has been reported to play important role in inflammatory responses and several diseases. However, whether it contributes to COPD is an open question. The aim of this study is to explore the potential relationship between ANGPTL4 and COPD. PATIENTS AND METHODS: In this study, circulating levels of ANGPTL4, C-reactive protein (CRP), adiponectin, tumor necrosis factor (TNF)-α, matrix metalloproteinase (MMP)-9 and monocyte chemotactic protein (MCP)-1 in 73 COPD patients and 40 healthy volunteers were investigated using multiplex enzyme-linked immunosorbent assay Kits. Then, we analyzed the correlations between ANGPTL4 with other inflammatory mediators and pulmonary function. RESULTS: Serum ANGPTL4 levels were significantly elevated in COPD patients compared with healthy controls (122.86 ± 38.59 ng/mL versus 99.03 ± 31.84 ng/mL, p = 0.001). Besides, serum ANGTPL4 levels were much higher in ever-smokers with COPD than in never-smokers with COPD (131.71 ± 32.92 ng/mL versus 113.25 ± 42.34 ng/mL, p = 0.03). More importantly, the concentrations of circulating ANGPLT4 correlated inversely with forced expiratory volume in 1 second (FEV1) % predicted, an index of lung function in COPD (r = -0.450, p < 0.001) and in all participants (r = -0.369, p < 0.001), while correlated positively with CRP (r = 0.312, p = 0.007 for COPD; r = 0.404, p < 0.001 for total subjects), adiponectin (r = 0.266, p = 0.004 for total subjects), and MMP-9 (r = 0.254, p = 0.03 for COPD). CONCLUSIONS: Our results suggest that circulating ANGPTL4 levels are up-regulated in COPD patients, and have correlations with pulmonary function and systematic inflammation in COPD, which provides a novel idea to further dig the pathogenic mechanisms of COPD, and justifies more studies to determine how ANGPTL4 contributes to COPD.


Subject(s)
Angiopoietins/biosynthesis , Pulmonary Disease, Chronic Obstructive/metabolism , Angiopoietin-Like Protein 4 , Angiopoietins/blood , Biomarkers/blood , Humans , Inflammation/blood , Lung/metabolism , Lung/physiopathology , Pulmonary Disease, Chronic Obstructive/blood , Pulmonary Disease, Chronic Obstructive/physiopathology , Respiratory Function Tests
9.
J Physiol ; 594(11): 2971-83, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26670924

ABSTRACT

KEY POINTS: Angiopoietin-like 4 (ANGPTL4) modulates tendon neovascularization. Cyclic loading stimulates the activity of transforming growth factor-ß and hypoxia-inducible factor 1α and thereby increases the expression and release of ANGPTL4 from human tendon cells. Targeting ANGPTL4 and its regulatory pathways is a potential avenue for regulating tendon vascularization to improve tendon healing or adaptation. ABSTRACT: The mechanisms that regulate angiogenic activity in injured or mechanically loaded tendons are poorly understood. The present study examined the potential role of angiopoietin-like 4 (ANGPTL4) in the angiogenic response of tendons subjected to repetitive mechanical loading or injury. Cyclic stretching of human tendon fibroblasts stimulated the expression and release of ANGPTL4 protein via transforming growth factor-ß (TGF-ß) and hypoxia-inducible factor 1α (HIF-1α) signalling, and the released ANGPTL4 was pro-angiogenic. Angiogenic activity was increased following ANGPTL4 injection into mouse patellar tendons, whereas the patellar tendons of ANGPTL4 knockout mice displayed reduced angiogenesis following injury. In human rotator cuff tendons, the expression of ANGPTL4 was correlated with the density of tendon endothelial cells. To our knowledge, this is the first study characterizing a role of ANGPTL4 in the tendon. ANGPTL4 may assist in the regulation of vascularity in the injured or mechanically loaded tendon. TGF-ß and HIF-1α comprise two signalling pathways that modulate the expression of ANGPTL4 by mechanically stimulated tendon fibroblasts and, in the future, these could be manipulated to influence tendon healing or adaptation.


Subject(s)
Angiopoietins/biosynthesis , Fibroblasts/metabolism , Neovascularization, Physiologic/physiology , Tendons/metabolism , Weight-Bearing/physiology , Amino Acids, Dicarboxylic/pharmacology , Angiopoietin-Like Protein 4 , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Fibroblasts/drug effects , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic/drug effects , Tendons/drug effects
10.
Sci Signal ; 8(407): ra127, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26671148

ABSTRACT

A common variant of the FTO (fat mass- and obesity-associated) gene is a risk factor for obesity. We found that mice with an adipocyte-specific deletion of FTO gained more weight than control mice on a high-fat diet. Analysis of mice lacking FTO in adipocytes fed a normal diet or adipocytes from these mice revealed alterations in triglyceride metabolism that would be expected to favor increased fatty acid storage by adipose tissue. Mice lacking FTO in adipocytes showed increased serum triglyceride breakdown and clearance, which was associated with lower serum triglyceride concentrations. In addition, lipolysis in response to ß-adrenergic stimulation was decreased in adipocytes and ex vivo adipose explants from the mutant mice. FTO is a nucleic acid demethylase that removes N(6)-methyladenosine (m(6)A) from mRNAs. We found that FTO bound to Angptl4, which encodes an adipokine that stimulates intracellular lipolysis in adipocytes. Unexpectedly, the adipose tissue of fasted or fed mice lacking FTO in adipocytes had greater Angptl4 mRNA abundance. However, after high-fat feeding, the mutant mice had less Angptl4 protein and more m(6)A-modified Angptl4 than control mice, suggesting that lack of FTO prevented the translation of Angptl4. Injection of Angptl4-encoding adenovirus into mice lacking FTO in adipocytes restored serum triglyceride concentrations and lipolysis to values similar to those in control mice and abolished excessive weight gain from a high-fat diet. These results reveal that FTO regulates fatty acid mobilization in adipocytes and thus body weight in part through posttranscriptional regulation of Angptl4.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Angiopoietins/biosynthesis , Mixed Function Oxygenases/metabolism , Oxo-Acid-Lyases/metabolism , Protein Biosynthesis , Triglycerides/metabolism , 3T3-L1 Cells , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Angiopoietin-Like Protein 4 , Animals , Humans , Mice , Mice, Knockout , Mixed Function Oxygenases/genetics , Oxo-Acid-Lyases/genetics , Triglycerides/genetics
11.
PLoS One ; 10(9): e0137049, 2015.
Article in English | MEDLINE | ID: mdl-26352670

ABSTRACT

Podocyte injury plays central roles in proteinuria and kidney dysfunction, therefore, identifying specific biomarker to evaluate earlier podocyte injury is highly desirable. Podocyte-secreted angiopoietin-like-4 (Angptl4) mediates proteinuria in different types of podocytopathy. In the present study, we established an experimental minimal change disease (MCD) rat model, induced by adriamycin (ADR) and resulted in definite podocyte injury, to identify the dynamic changes in Angptl4 expression. We also investigated the direct effects of tacrolimus on Angptl4 and podocyte repair. We determined that the glomerular Angptl4 expression was rapidly upregulated and reached a peak earlier than desmin, an injured podocyte marker, in the ADR rats. Furthermore, this upregulation occurred prior to heavy proteinuria and was accompanied by increased urinary Angptl4. We observed that the Angptl4 upregulation occurred only when podocyte was mainly damaged since we didn't observe little Angptl4 upregulation in MsPGN patients. In addition, we observed the glomerular Angptl4 mainly located in injured podocytes rather than normal podocytes. Moreover, we found that tacrolimus treatment significantly promoted podocyte repair and reduced glomerular and urinary Angptl4 expression at an earlier stage with a significant serum Angptl4 upregulation. And similar results were confirmed in MCD patients. In conclusion, this study represents the first investigation to demonstrate that Angptl4 can predict podocyte injury at earlier stages in MCD and the identification of earlier podocyte injury biomarkers could facilitate the prompt diagnosis and treatment of patients with podocytopathy, as well as determination of the prognosis and treatment efficacy in these diseases.


Subject(s)
Angiopoietins/biosynthesis , Nephrosis, Lipoid/genetics , Podocytes/metabolism , Tacrolimus/administration & dosage , Angiopoietin-Like Protein 4 , Angiopoietins/blood , Angiopoietins/genetics , Animals , Disease Models, Animal , Doxorubicin/toxicity , Humans , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Nephrosis, Lipoid/chemically induced , Nephrosis, Lipoid/drug therapy , Nephrosis, Lipoid/pathology , Podocytes/drug effects , Podocytes/pathology , Proteinuria , Rats
12.
Exp Clin Endocrinol Diabetes ; 123(8): 445-60, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26069075

ABSTRACT

Angiopoietin-like protein 4 (Angptl4), also known as fasting-induced adiopogenic factor (FIAF), has recently been reported to influence bone metabolism. However, there have been few studies on regulatory factors other than hypoxia for Angptl4 in bone, and particularly in osteoblasts. Expression of interleukin-1ß (IL-1ß), a proinflammatory cytokine, is increased in serum or bone microenvironments in inflammatory bone diseases or estrogen deficient-conditions. The present study was conducted to determine whether Angptl4 expression in osteoblasts is affected by IL-1ß and investigate its involvement in MAP kinase signaling pathways. Angptl4 RNA levels were increased by IL-1ß treatment in murine MC3T3-E1 osteoblastic cells. Western blotting and immunofluorescent staining showed a corresponding increase in Angptl4 protein. IL-1ß treatment of osteoblasts induced phosphorylation of mitogen-activated protein kinases (MAPKs) including extracellular regulated kinases (ERKs), p38, and c-Jun N-terminal kinase (JNK). Furthermore, SP600125, an inhibitor of JNK, significantly blocked the upregulation of Angptl4 by IL-1ß. In contrast, treatment with an inhibitor of p38 MAP kinase (SB203580) or an ERK inhibitor (PD98059) produced responses similar to those seen with the DMSO control. Taken together, these results suggest that IL-1ß increases Angptl4 expression through a mechanism dependent on the JNK-MAPK signaling pathway in MC3T3-E1 cells.


Subject(s)
Angiopoietins/biosynthesis , Gene Expression Regulation/physiology , Interleukin-1beta/metabolism , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System/physiology , Osteoblasts/metabolism , Angiopoietin-Like Protein 4 , Animals , Cell Line , Gene Expression Regulation/drug effects , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Mice , Osteoblasts/cytology , Protein Kinase Inhibitors/pharmacology
13.
PLoS One ; 10(5): e0125371, 2015.
Article in English | MEDLINE | ID: mdl-25946041

ABSTRACT

The impact of commensal bacteria in eukaryotic transcriptional regulation has increasingly been demonstrated over the last decades. A multitude of studies have shown direct effects of commensal bacteria from local transcriptional activity to systemic impact. The commensal bacterium Streptococcus salivarius is one of the early bacteria colonizing the oral and gut mucosal surfaces. It has been shown to down-regulate nuclear transcription factor (NF-кB) in human intestinal cells, a central regulator of the host mucosal immune system response to the microbiota. In order to evaluate its impact on a further important transcription factor shown to link metabolism and inflammation in the intestine, namely PPARγ (peroxisome proliferator-activated receptor), we used human intestinal epithelial cell-lines engineered to monitor PPARγ transcriptional activity in response to a wide range of S. salivarius strains. We demonstrated that different strains from this bacterial group share the property to inhibit PPARγ activation independently of the ligand used. First attempts to identify the nature of the active compounds showed that it is a low-molecular-weight, DNase-, proteases- and heat-resistant metabolite secreted by S. salivarius strains. Among PPARγ-targeted metabolic genes, I-FABP and Angptl4 expression levels were dramatically reduced in intestinal epithelial cells exposed to S. salivarius supernatant. Both gene products modulate lipid accumulation in cells and down-regulating their expression might consequently affect host health. Our study shows that species belonging to the salivarius group of streptococci impact both host inflammatory and metabolic regulation suggesting a possible role in the host homeostasis and health.


Subject(s)
Intestinal Mucosa/metabolism , PPAR gamma/biosynthesis , Streptococcus/metabolism , Transcription, Genetic/genetics , Transcriptional Activation/genetics , Angiopoietin-Like Protein 4 , Angiopoietins/biosynthesis , Angiopoietins/genetics , Caco-2 Cells , Cell Line, Tumor , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fatty Acid-Binding Proteins/biosynthesis , Fatty Acid-Binding Proteins/genetics , Gene Expression Regulation , HT29 Cells , Humans , Immunity, Mucosal/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Microbiota/immunology , PPAR gamma/genetics , Streptococcus/immunology , Symbiosis
14.
Diabetologia ; 58(7): 1523-31, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25917759

ABSTRACT

AIMS/HYPOTHESIS: The identification of novel targets that stimulate endogenous regeneration of beta cells would represent a significant advance in the treatment of patients with diabetes. The betatrophin hypothesis suggests that increased expression of angiopoietin-like protein 8 (ANGPTL8) induces dramatic and specific beta cell proliferation and subsequent beta cell mass expansion with improved glucose tolerance. In light of recent controversy, we further investigated the effects of ANGPTL8 overexpression on beta cell proliferation. METHODS: We performed hydrodynamic tail vein injections of green fluorescent protein (GFP) or Angptl8 (also known as Gm6484) DNA in multiple cohorts of mice of different ages. We employed state-of-the-art methods to comprehensively quantify beta cell mass and proliferation, controlling for mouse age, genetic strain, source of DNA injected, Angptl8 gene expression and proliferation markers. RESULTS: In two young and two aged cohorts of B6.129 mice, no substantial change in beta cell replication, mass or glucose homeostasis was observed following ANGPTL8 overexpression. Even in mice with extremely elevated Angptl8 expression (26-fold increase), beta cell replication was not significantly altered. Finally, we considered mice on the ICR background exactly as studied by Melton and colleagues, and still no beta cell mitogenic effect was detected following ANGPTL8 overexpression. CONCLUSION/INTERPRETATION: ANGPTL8 does not stimulate beta cell replication in young or old mice.


Subject(s)
Angiopoietins/biosynthesis , Insulin-Secreting Cells/physiology , Peptide Hormones/biosynthesis , Aging/metabolism , Angiopoietin-Like Protein 8 , Angiopoietin-like Proteins , Angiopoietins/genetics , Animals , Cell Proliferation , DNA/genetics , Glucose/metabolism , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Male , Mice , Mice, Inbred ICR , Pancreatectomy , Peptide Hormones/genetics
15.
Expert Rev Mol Med ; 16: e17, 2014 Nov 24.
Article in English | MEDLINE | ID: mdl-25417860

ABSTRACT

Angptl2 is a multifaceted protein, displaying both physiological and pathological functions, in which scientific and clinical interest is growing exponentially within the past few years. Its physiological functions are not well understood, but angptl2 was first acknowledged for its pro-angiogenic and antiapoptotic capacities. In addition, angptl2 can be considered a growth factor, since it increases survival and expansion of hematopoietic stem cells and may promote vasculogenesis. Finally, angptl2 has an important, but largely unrecognised, physiological role: in the cytosol, angptl2 binds to type 1A angiotensin II receptors and induces their recycling, with recovery of the receptor signal functions. Despite these important physiological properties, angptl2 is better acknowledged for its deleterious pro-inflammatory properties and its contribution in multiple chronic diseases such as cancer, diabetes, atherosclerosis, metabolic disorders and many other chronic diseases. This review aims at presenting an updated description of both the beneficial and deleterious biological properties of angptl2, in addition to its molecular signalling pathways and transcriptional regulation. The multiplicity of diseases in which angptl2 contributes makes it a new highly relevant clinical therapeutic target.


Subject(s)
Angiopoietins/genetics , Neovascularization, Physiologic/genetics , Receptor, Angiotensin, Type 1/metabolism , Transcription, Genetic , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins , Angiopoietins/biosynthesis , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Humans , Molecular Targeted Therapy , Promoter Regions, Genetic , Signal Transduction/genetics
16.
J Leukoc Biol ; 96(3): 365-75, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24626957

ABSTRACT

On a molecular level, cells sense changes in oxygen availability through the PHDs, which regulate the protein stability of the α-subunit of the transcription factor HIF. Especially, PHD3 has been additionally associated with apoptotic cell death. We hypothesized that PHD3 plays a role in cell-fate decisions in macrophages. Therefore, myeloid-specific PHD3(-/-) mice were created and analyzed. PHD3(-/-) BMDM showed no altered HIF-1α or HIF-2α stabilization or increased HIF target gene expression in normoxia or hypoxia. Macrophage M1 and M2 polarization was unchanged likewise. Compared with macrophages from WT littermates, PHD3(-/-) BMDM exhibited a significant reduction in TUNEL-positive cells after serum withdrawal or treatment with stauro and SNAP. Under the same conditions, PHD3(-/-) BMDM also showed less Annexin V staining, which is representative for membrane disruption, and indicated a reduced early apoptosis. In an unbiased transcriptome screen, we found that Angptl2 expression was reduced in PHD3(-/-) BMDM under stress conditions. Addition of rAngptl2 rescued the antiapoptotic phenotype, demonstrating that it is involved in the PHD3-mediated response toward apoptotic stimuli in macrophages.


Subject(s)
Macrophages/cytology , Procollagen-Proline Dioxygenase/physiology , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins , Angiopoietins/biosynthesis , Angiopoietins/genetics , Angiopoietins/pharmacology , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/physiology , Bone Marrow Cells/cytology , Cell Hypoxia , Cells, Cultured , Gene Expression Regulation , Hydroxylation , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Macrophages/drug effects , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/enzymology , NF-kappa B/metabolism , Procollagen-Proline Dioxygenase/deficiency , Procollagen-Proline Dioxygenase/genetics , Protein Processing, Post-Translational , Recombinant Fusion Proteins/pharmacology , S-Nitroso-N-Acetylpenicillamine/pharmacology , Staurosporine/pharmacology , Transcription, Genetic , Transcriptome
17.
Int J Low Extrem Wounds ; 13(1): 58-63, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24659626

ABSTRACT

Diabetes mellitus (DM) is one of the severe metabolic disorders of carbohydrate metabolism worldwide. Developing countries are at higher risk of DM, and there is significant evidence that it is epidemic in many economically developing and newly industrialized countries. Among all other complications associated with DM, delayed wound healing is a major concern in diabetic patients. Wound healing is a natural healing process that starts immediately after injury. This involves interaction of a complex cascade of cellular events that generates resurfacing, reconstitution, and restoration of the tensile strength of injured skin. There are multiple factors responsible for delayed wound healing among which the contribution of DM has been well documented. The wound healing process is also delayed by the metabolic, vascular, neurological, and inflammatory alterations, which are well known in both type 1 and type 2 diabetes. Keratinocytes are crucial for wound re-epithelialization, and defects in directed migration of keratinocytes due to DM are associated with the delayed wound healing process. Many factors responsible for re-epithelialization have been identified, characterized, and well described; however, the genes responsible for the healing process have only partially been illustrated. This article will therefore focus on the efficacy of ANGPTL4 (angiopoietin-like 4) gene, which plays a novel role in keratinocyte migration during wound healing.


Subject(s)
Angiopoietins/genetics , Diabetes Mellitus/genetics , Gene Expression Regulation , RNA/genetics , Skin/injuries , Wound Healing/genetics , Wounds and Injuries/genetics , Angiopoietin-Like Protein 4 , Angiopoietins/biosynthesis , Diabetes Mellitus/metabolism , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Wounds and Injuries/complications , Wounds and Injuries/metabolism
18.
Angiogenesis ; 17(1): 261-74, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24114199

ABSTRACT

Semaphorin 4D (SEMA4D) is a member of a family of transmembrane and secreted proteins that have been shown to act through its receptor Plexin-B1 to regulate axon growth cone guidance, lymphocyte activation, and bone density. SEMA4D is also overexpressed by some malignancies and plays a role in tumor-induced angiogenesis similar to vascular endothelial growth factor (VEGF), a protein that has been targeted as part of some cancer therapies. In an attempt to examine the different effects on tumor growth and vascularity for these two pro-angiogenic factors, we previously noted that while inhibition of both VEGF and SEMA4D restricted tumor vascularity and size, vessels forming under conditions of VEGF blockade retained their association with pericytes while those arising in a background of SEMA4D/Plexin-B1 deficiency did not, an intriguing finding considering that alteration in pericyte association with endothelial cells is an emerging aspect of anti-angiogenic intervention in the treatment of cancer. Here we show through array analysis, immunoblots, migration and co-culture assays and VE-cadherin immunohistochemistry that SEMA4D production by head and neck carcinoma tumor cells induces expression of platelet-derived growth factor-B and angiopoietin-like protein 4 from endothelial cells in a Plexin-B1/Rho-dependent manner, thereby influencing proliferation and differentiation of pericytes and vascular permeability, whereas VEGF lacks these effects. These results partly explain the differences observed between SEMA4D and VEGF in pathological angiogenesis and suggest that targeting SEMA4D function along with VEGF could represent a novel anti-angiogenic therapeutic strategy for the treatment of solid tumors.


Subject(s)
Angiopoietins/biosynthesis , Antigens, CD/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Nerve Tissue Proteins/metabolism , Pericytes/metabolism , Proto-Oncogene Proteins c-sis/biosynthesis , Receptors, Cell Surface/metabolism , Semaphorins/metabolism , Signal Transduction , rhoA GTP-Binding Protein/metabolism , Angiopoietin-Like Protein 4 , Angiopoietins/genetics , Antigens, CD/genetics , Humans , Neoplasms/blood supply , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/therapy , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins c-sis/genetics , Receptors, Cell Surface/genetics , Semaphorins/genetics , rhoA GTP-Binding Protein/genetics
19.
Histochem Cell Biol ; 141(4): 383-91, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24141811

ABSTRACT

Gut hormones produced by entero-endocrine cells (EEC) located throughout the gastrointestinal tract play a major role in the regulation of glucose and energy homeostasis. Angiopoietin-like 4 (ANGPTL4, also referred to as fasting induced adipose factor) is a secreted factor involved in regulation of lipid homeostasis and has been proposed as circulating mediator between the gut microbiota and fat storage in adipose tissue, although discordant data exist. Currently, little is known about the site and regulation of ANGPTL4 production in the intestine. Here, we show using immunohistochemistry and immunofluorescence that cells positive for ANGPTL4 are scattered along the epithelial layer in the human small and large intestine. ANGPTL4-positive cells exhibit typical features of EEC characterized by large ANGPTL4-positive secretory granules directed towards the basolateral side. In support, extensive overlap was observed between ANGPTL4-positive cells and cells positive for the entero-endocrine marker chromogranin A. Higher resolution images revealed that ANGPTL4 and chromogranin A are partially present in distinct intracellular vesicles. Using entero-endocrine HuTu-80 cells, ANGPTL4 secretion was shown to be induced by short chain fatty acids and reduced by bile acids. Finally, levels of ANGPTL4 in human plasma were significantly decreased following meal consumption. In conclusion, ANGPTL4 is produced by EEC in human intestine and expression may be regulated by short chain fatty acids and bile acids.


Subject(s)
Angiopoietins/biosynthesis , Enteroendocrine Cells/metabolism , Gastrointestinal Tract/metabolism , Adult , Angiopoietin-Like Protein 4 , Angiopoietins/genetics , Cells, Cultured , Enteroendocrine Cells/cytology , Enzyme-Linked Immunosorbent Assay , Gastrointestinal Tract/cytology , Humans , Immunohistochemistry , Male , Real-Time Polymerase Chain Reaction
20.
Mod Pathol ; 26(8): 1032-40, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23558570

ABSTRACT

Angiosarcoma is an aggressive malignancy of endothelial differentiation. Potential roles of the endothelial angiopoietin-tunica interna endothelial cell kinase (ANGPT-TIE) system in angiosarcoma diagnosis, pathogenesis, prognosis and treatment are undefined. To examine the expression and prognostic significance of angiopoietin-1, angiopoietin-2, TIE1 and TEK (TIE2) proteins in angiosarcoma, we immunohistochemically evaluated clinically annotated human angiosarcoma samples. Correlations of protein expression with overall survival and pathological features were explored. The cohort included 51 patients diagnosed with angiosarcoma at the age of 30-86 years (median 67). The 5-year overall survival was 45% with a median of 26 months. Moderate to strong expression of angiopoietin-1, TIE1 and TEK (TIE2) was identified in the majority of angiosarcomas and moderate to strong expression of angiopoietin-2 was observed in 42% of angiosarcomas. Increased angiopoietin-1 expression correlated with improved survival. Non-significant trends toward longer survival were also observed with increased TIE1 and TEK (TIE2) expression. Increased expression of angiopoietin-2, TIE1 and TEK (TIE2) was associated with vasoformative architecture. No differences in expression of these proteins were observed when patients were segregated by age, gender, presence or absence of metastases at diagnosis, primary tumor location, radiation association or the presence of necrosis. We conclude that components of the ANGPT-TIE system are commonly expressed in angiosarcomas. Reduced expression of these proteins is associated with non-vasoformative and clinically more aggressive lesions.


Subject(s)
Angiopoietins/biosynthesis , Biomarkers, Tumor/analysis , Hemangiosarcoma/metabolism , Receptors, TIE/biosynthesis , Soft Tissue Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Angiopoietins/analysis , Female , Hemangiosarcoma/mortality , Hemangiosarcoma/pathology , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Receptors, TIE/analysis , Soft Tissue Neoplasms/mortality , Soft Tissue Neoplasms/pathology , Tissue Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...