Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 22(15)2021 Jul 26.
Article in English | MEDLINE | ID: mdl-34360721

ABSTRACT

Host lipid metabolism reprogramming is essential for hepatitis C virus (HCV) infection and progression to severe liver disease. Direct-acting antivirals (DAAs) achieve a sustained virological response (SVR) in most patients, but virus eradication does not always protect against hepatocellular carcinoma (HCC). Angiopoietin-like protein-3 (ANGPTL-3) and angiopoietin-like protein-4 (ANGPTL-4) regulate the clearance of plasma lipids by inhibiting cellular lipase activity and possess emerging roles in tumourigenesis. We used ELISA and RT-qPCR to investigate ANGPTL-3 and ANGPTL-4 expression in HCV patients with characterised fibrosis throughout the natural history of hepatitis C and in long-term HCV infection in vitro, before and after DAA treatment. ANGPTL-3 was decreased in patients with advanced fibrosis compared to other disease stages, while ANGPTL-4 was progressively increased from acute infection to cirrhosis and HCC, peaking at the advanced fibrosis stage. Only ANGPTL-3 mRNA was down-regulated during early infection in vitro, although both ANGPTLs were increased later. DAA treatment did not alter ANGPTL-3 levels in advanced fibrosis/cirrhosis and in HCV infection in vitro, in contrast to ANGPTL-4. The association between ANGPTLs and fibrosis in HCV infection was underlined by an inverse correlation between the levels of ANGPTLs and serum transforming growth factor- ß (TGF-ß). Collectively, we demonstrate the pivotal role of advanced fibrosis in defining the expression fate of ANGPTLs in HCV infection and after treatment and propose a role for ANGPTL-3 as a contributor to post-treatment deregulation of lipid metabolism that could predispose certain individuals to HCC development.


Subject(s)
Angiopoietin-Like Protein 4/biosynthesis , Angiopoietin-like Proteins/biosynthesis , Antiviral Agents/administration & dosage , Gene Expression Regulation/drug effects , Hepacivirus/metabolism , Hepatitis C, Chronic , Liver Cirrhosis , Angiopoietin-Like Protein 3 , Cell Line, Tumor , Female , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/metabolism , Humans , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Male
2.
Inflammation ; 43(4): 1397-1410, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32170602

ABSTRACT

The present study aimed to investigate the role of Forkhead box protein C2 (Foxc2) in oxidized low-density lipoprotein (ox-LDL)-induced macrophages and identify the potential mechanisms. RAW264.7 cells, the murine macrophage cell line, were stimulated by ox-LDL, and cell proliferation was examined. The levels of inflammation- and oxidative stress-related markers were detected using kits after induction with ox-LDL. Subsequently, the expression of Foxc2 was measured using Western blotting. After transfection with Foxc2 pcDNA3.1, intracellular lipid droplets were examined using oil red O staining. The levels of total cholesterol (TC), free cholesterol (FC), inflammatory cytokines, and oxidative stress markers were determined. Moreover, apoptosis of RAW264.7 cells was detected using flow cytometry, and apoptosis-related proteins were measured using Western blotting. Angiopoietin-like protein 2 (Angptl2) was predicted as a target gene of Foxc2. Therefore, the expression of Angptl2 was examined after Foxc2 overexpression in ox-LDL-induced RAW264.7 cells. Then, the changes of intracellular lipid droplets, TC, FC, inflammatory cytokines, oxidative stress factors, and cell apoptosis were detected after Angptl2 overexpression or co-transfection with Foxc2 and Angptl2 pcDNA3.1. The results revealed that ox-LDL induction inhibited proliferation of RAW264.7 cells and promoted the release of inflammatory factors. Importantly, the expression of Foxc2 was obviously decreased after stimulation by ox-LDL. Foxc2 overexpression suppressed lipid accumulation, TC, FC levels, inflammation, oxidative stress, and apoptosis induced by ox-LDL, whereas these inhibitory effects were relieved after co-transfection with Angptl2 pcDNA3.1. These findings demonstrated that Foxc2 can alleviate ox-LDL-induced lipid accumulation, inflammation, and apoptosis of macrophage via regulating the expression of Angptl2.


Subject(s)
Angiopoietin-like Proteins/biosynthesis , Apoptosis/physiology , Forkhead Transcription Factors/biosynthesis , Lipid Metabolism/physiology , Lipoproteins, LDL/toxicity , Macrophages/metabolism , Angiopoietin-Like Protein 2 , Animals , Apoptosis/drug effects , Gene Expression , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/prevention & control , Lipid Metabolism/drug effects , Macrophages/drug effects , Mice , RAW 264.7 Cells
3.
Med Sci Monit ; 25: 9524-9530, 2019 Dec 13.
Article in English | MEDLINE | ID: mdl-31835268

ABSTRACT

BACKGROUND Angiopoietin-like proteins (ANGPTL) are a family of secretory glycoproteins that are involved in many pathophysiological processes. ANGPTL7 is a newly-discovered member of the ANGPTL family and plays a role in corneal morphogenesis, angiogenesis, glaucoma, and cancer. To date, whether ANGPTL7 is involved in osteoporosis is unknown. Therefore, to discover the effects of ANGPTL7 on osteoporosis, we explored the expression of ANGPTL7 in preosteoblasts and assessed the mechanism underlying its effects on proliferation and differentiation abilities of preosteoblasts. MATERIAL AND METHODS Mouse MC3T3-E1 cells were cultured in osteogenic medium for osteogenic differentiation. The expression levels of ANGPTL7 were detected by RT-qPCR and Western blot assays. Moreover, the overexpressed plasmid of ANGPTL7 pMSCV-ANGPTL7 was transfected into MC3T3-E1 cells. CCK-8 was used to evaluate cell proliferation. ALP activity detection and alizarin red staining were performed to measure the effect of ANGPTL7 on osteogenic differentiation. The expression levels bone morphogenetic proteins (BMPs) and osteogenic markers ALP, runt-related transcription factor 2 (Runx2), osteocalcin (OCN), and collagen I (Col I) were analyzed by Western blot. RESULTS When MC3T3-E1 cells were exposed to osteogenic medium, there was a significant increase in ANGPTL7, and overexpression of ANGPTL7 markedly promoted cell proliferation, ALP activity, and mineralization. Moreover, ANGPTL7 upregulated the levels of BMPs, especially BMP2/7, and the osteogenic markers ALP, Runx2, OCN, and Col I. CONCLUSIONS The results suggest that by regulating the expression of BMPs, ANGPTL7 directly promotes proliferation, differentiation, and mineralization of osteoblasts.


Subject(s)
Angiopoietin-like Proteins/metabolism , 3T3 Cells , Angiopoietin-Like Protein 7 , Angiopoietin-like Proteins/biosynthesis , Angiopoietins/metabolism , Animals , Biomarkers/metabolism , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/physiology , Cell Line , Cell Proliferation/physiology , Collagen Type I/metabolism , Mice , Osteoblasts/metabolism , Osteocalcin/metabolism , Osteogenesis/physiology , Osteoporosis/metabolism
4.
J Lipid Res ; 60(4): 783-793, 2019 04.
Article in English | MEDLINE | ID: mdl-30686789

ABSTRACT

Angiopoietin-like (ANGPTL) 8 is a secreted inhibitor of LPL, a key enzyme in plasma triglyceride metabolism. It was previously reported that ANGPTL8 requires another member of the ANGPTL family, ANGPTL3, to act on LPL. ANGPTL3, much like ANGPTL4, is a physiologically relevant regulator of LPL activity, which causes irreversible inactivation of the enzyme. Here, we show that ANGPTL8 can form complexes with either ANGPTL3 or ANGPTL4 when the proteins are refolded together from their denatured states. In contrast to the augmented inhibitory effect of the ANGPTL3/ANGPTL8 complex on LPL activity, the ANGPTL4/ANGPTL8 complex is less active compared with ANGPTL4 alone. In our experiments, all three members of the ANGPTL family use the same mechanism to inactivate LPL, which involves dissociation of active dimeric LPL to monomers. This inactivation can be counteracted by the presence of glycosylphosphatidylinositol-anchored HDL binding protein 1, the endothelial LPL transport protein previously known to protect LPL from spontaneous and ANGPTL4-catalyzed inactivation. Our data demonstrate that ANGPTL8 may function as an important metabolic switch, by forming complexes with ANGPTL3, or with ANGPTL4, in order to direct the flow of energy from triglycerides in blood according to the needs of the body.


Subject(s)
Angiopoietin-like Proteins/biosynthesis , Lipoprotein Lipase/metabolism , Peptide Hormones/biosynthesis , Angiopoietin-Like Protein 8 , Angiopoietin-like Proteins/genetics , Angiopoietin-like Proteins/isolation & purification , Humans , Peptide Hormones/genetics , Peptide Hormones/isolation & purification , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
5.
Exp Hematol ; 57: 21-29, 2018 01.
Article in English | MEDLINE | ID: mdl-28911908

ABSTRACT

Integration-deficient lentiviruses (IdLVs) deliver genes effectively to tissues but are lost rapidly from dividing cells. This property can be harnessed to express transgenes transiently to manipulate cell biology. Here, we demonstrate the utility of short-term gene expression to improve functional potency of hematopoietic stem and progenitor cells (HSPCs) during transplantation by delivering HOXB4 and Angptl3 using IdLVs to enhance the engraftment of HSPCs. Constitutive overexpression of either of these genes is likely to be undesirable, but the transient nature of IdLVs reduces this risk and those associated with unsolicited gene expression in daughter cells. Transient expression led to increased multilineage hematopoietic engraftment in in vivo competitive repopulation assays without the side effects reported in constitutive overexpression models. Adult stem cell fate has not been programmed previously using IdLVs, but we demonstrate that these transient gene expression tools can produce clinically relevant alterations or be applied to investigate basic biology.


Subject(s)
Genetic Vectors/genetics , Hematopoietic Stem Cells/physiology , Lentivirus/genetics , Transduction, Genetic , Angiopoietin-Like Protein 3 , Angiopoietin-like Proteins/biosynthesis , Angiopoietin-like Proteins/genetics , Animals , Cell Lineage , Gene Expression Regulation , Genes, Reporter , Graft Survival , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , K562 Cells , Mice , Mice, Inbred C57BL , Radiation Chimera , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transgenes
6.
J Biol Chem ; 293(5): 1596-1609, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29191837

ABSTRACT

Skeletal muscle atrophy, or sarcopenia, is commonly observed in older individuals and in those with chronic disease and is associated with decreased quality of life. There is recent medical and broad concern that sarcopenia is rapidly increasing worldwide as populations age. At present, strength training is the only effective intervention for preventing sarcopenia development, but it is not known how this exercise regimen counteracts this condition. Here, we report that expression of the inflammatory mediator angiopoietin-like protein 2 (ANGPTL2) increases in skeletal muscle of aging mice. Moreover, in addition to exhibiting increased inflammation and accumulation of reactive oxygen species (ROS), denervated atrophic skeletal muscles in a mouse model of denervation-induced muscle atrophy had increased ANGPTL2 expression. Interestingly, mice with a skeletal myocyte-specific Angptl2 knockout had attenuated inflammation and ROS accumulation in denervated skeletal muscle, accompanied by increased satellite cell activity and inhibition of muscular atrophy compared with mice harboring wildtype Angptl2 Moreover, consistent with these phenotypes, wildtype mice undergoing exercise training displayed decreased ANGPTL2 expression in skeletal muscle. In conclusion, ANGPTL2 up-regulation in skeletal myocytes accelerates muscle atrophy, and exercise-induced attenuation of ANGPTL2 expression in those tissues may partially explain how exercise training prevents sarcopenia.


Subject(s)
Aging/metabolism , Angiopoietin-like Proteins/biosynthesis , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Sarcopenia/metabolism , Up-Regulation , Aging/genetics , Aging/pathology , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins/genetics , Animals , Female , Male , Mice , Mice, Knockout , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Physical Conditioning, Animal , Sarcopenia/genetics , Sarcopenia/pathology , Sarcopenia/prevention & control
7.
Graefes Arch Clin Exp Ophthalmol ; 255(8): 1515-1523, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28456825

ABSTRACT

PURPOSE: To investigate the vitreous and serum levels of angiopoietin-like protein 8 (ANGPTL-8) and vascular endothelial growth factor (VEGF) in patients with proliferative diabetic retinopathy (PDR). The serum levels of these factors were also analyzed in patients with diabetes and no diabetic retinopathy (NDR) and with non-proliferative diabetic retinopathy (NPDR), to detect the possible correlation between the ANGPTL-8 levels and hyperlipidemia. METHODS: Vitreous samples were obtained from 28 patients with PDR and from 12 patients without diabetes and with idiopathic macular hole (IMH). Serum samples were also obtained from 26 patients with NDR and 22 patients with NPDR. ANGPTL-8 levels and other factors were determined using an enzyme-linked immunosorbent assay. RESULTS: The ANGPTL-8 and VEGF levels in the vitreous and serum of the patients with PDR were higher than those in the patients with IMH, and were significantly correlated. The vitreous and serum ANGPTL-8 levels were more correlated with the triglyceride and low-density lipoprotein cholesterol levels than with the high-density lipoprotein cholesterol or total cholesterol levels in the patients with PDR. CONCLUSIONS: The vitreous and serum ANGPTL-8 levels were both upregulated in patients with PDR. There was an association between the elevation in the ANGPTL-8 levels and angiogenic and hyperlipidemic factors in the patients with PDR. These results suggest that ANGPTL-8 is a potential new diagnostic marker and therapeutic target for PDR treatment.


Subject(s)
Angiopoietin-like Proteins/biosynthesis , Diabetic Retinopathy/metabolism , Peptide Hormones/biosynthesis , Retinal Neovascularization/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vitreous Body/metabolism , Angiopoietin-Like Protein 8 , Biomarkers/metabolism , Diabetic Retinopathy/complications , Diabetic Retinopathy/diagnosis , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Retinal Neovascularization/complications , Retinal Neovascularization/diagnosis
8.
Oncol Res ; 25(8): 1349-1355, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28247845

ABSTRACT

Angiopoietin-like protein 2 (ANGPTL2), a member of the glycoprotein family, is mainly secreted by adipose tissues under normal conditions. Recently, ANGPTL2 has been found to be upregulated in some types of cancers and is considered to be a tumor promoter. However, the functional significance of ANGPTL2 in glioma has not yet been elucidated. In this study, we investigated the specific role of ANGPTL2 in glioma. The results showed that ANGPTL2 was highly expressed in glioma tissues and cell lines. Knockdown of ANGPTL2 reduced the proliferative and invasive abilities of glioma cells. Moreover, the tumorigenesis assay showed that ANGPTL2 knockdown inhibited glioma tumor growth in vivo. We also found that ANGPTL2 knockdown decreased the protein levels of p-ERK1/2 in glioma cells and thus blocked the activity of the ERK/MAPK signaling pathway. Taken together, our study provided the first evidence that ANGPTL2 played an oncogenic role in glioma development and might be considered as a new therapeutic target for glioma treatment.


Subject(s)
Angiopoietin-like Proteins/metabolism , Brain Neoplasms/metabolism , Glioma/metabolism , MAP Kinase Signaling System , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins/biosynthesis , Angiopoietin-like Proteins/deficiency , Angiopoietin-like Proteins/genetics , Animals , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Female , Gene Knockdown Techniques , Glioma/enzymology , Glioma/genetics , Glioma/pathology , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness
9.
Oncotarget ; 7(49): 80391-80403, 2016 Dec 06.
Article in English | MEDLINE | ID: mdl-27823982

ABSTRACT

Established adriamycin cardiomyopathy is a lethal disease. When congestive heart failure develops, mortality is approximately 50% in a year. It has been known that ANGPTLs has various functions in lipid metabolism, inflammation, cancer cell invasion, hematopoietic stem activity and diabetes. We hypothesized that ANGPTL8 is capable of maintaining heart function by stimulating adult cardiac progenitor cells to initiate myocardial regeneration. We employed UTMD to deliver piggybac transposon plasmids with the human ANGPTL8 gene to the liver of rats with adriamycin cardiomyopathy. After ANGPTL8 gene liver delivery, overexpression of transgenic human ANGPTL8 was found in rat liver cells and blood. UTMD- ANGPTL8 gene therapy restored LV mass, fractional shortening index, and LV posterior wall diameter to nearly normal. Our results also showed that ANGPTL8 reversed established ADM cardiomyopathy. This was associated with activation of ISL-1 positive cardiac progenitor cells in the epicardium. A time-course experiment shown that ISL-1 cardiac progenitor cells proliferated and formed a niche in the epicardial layer and then migrated into sub-epicardium. The observed myocardial regeneration accompanying reversal of adriamycin cardiomyopathy was associated with upregulation of PirB expression on the cell membrane of cardiac muscle cells or progenitor cells stimulated by ANGPTL8.


Subject(s)
Angiopoietin-like Proteins/biosynthesis , Cardiomyopathies/therapy , Doxorubicin , Genetic Therapy/methods , Liver/metabolism , Myocytes, Cardiac/metabolism , Peptide Hormones/biosynthesis , Stem Cells/metabolism , Angiopoietin-Like Protein 8 , Angiopoietin-like Proteins/blood , Angiopoietin-like Proteins/genetics , Animals , Cardiomyopathies/chemically induced , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiotoxicity , Cell Line , Cell Movement , Cell Proliferation , Disease Models, Animal , Gene Transfer Techniques , Humans , LIM-Homeodomain Proteins/metabolism , Male , Microbubbles , Myocardial Contraction , Myocytes, Cardiac/pathology , Peptide Hormones/blood , Peptide Hormones/genetics , Rats, Sprague-Dawley , Receptors, Immunologic/metabolism , Recovery of Function , Regeneration , Stem Cell Niche , Stem Cells/pathology , Time Factors , Transcription Factors/metabolism , Ultrasonics , Ventricular Function, Left , Ventricular Remodeling
SELECTION OF CITATIONS
SEARCH DETAIL
...