Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Exp Cell Res ; 407(2): 112753, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34499887

ABSTRACT

Peroxisome proliferator-activated receptor α (PPARα) activation has been reported to exert protective effects on podocytes, whereas angiopoietin-like 3 (ANGPTL3) has been shown to exert significant pathogenic effects on these cells. This study aimed to investigate the link between the protective effects of PPARα activation and the pathogenic effects of ANGPTL3 in podocytes. Both PPARα and ANGPTL3 were expressed in cultured podocytes. PPARα mRNA and protein levels decreased whereas ANGPTL3 mRNA and protein levels increased in a time-dependent manner in podocytes treated with puromycin aminonucleoside (PAN). Gemfibrozil, a pharmacological agonist of PPARα, increased PPARα levels and activity in podocytes. The drug also decreased ANGPTL3 levels by potentially weakening ANGPTL3 promoter activity in both normal and PAN-treated podocytes. Furthermore, gemfibrozil significantly decreased PAN-induced apoptosis and F-actin rearrangement. Primary podocytes from Angptl3-knockout mice were cultured. There was no significant difference between Angptl3-/- podocytes treated with or without gemfibrozil in the lamellipodia numbers after PAN treatment. The results suggested that the protective effects of gemfibrozil on podocytes were not exerted following knockout of the Angptl3 gene. This study identified a novel mechanism of the PPARα agonist gemfibrozil that exerts its protective effects by inhibiting PAN-induced apoptosis and cytoskeleton rearrangements through inhibition of ANGPTL3 expression.


Subject(s)
Actin Cytoskeleton/drug effects , Angiopoietin-like Proteins/physiology , Gemfibrozil/pharmacology , PPAR alpha/agonists , Podocytes/drug effects , Pseudopodia/drug effects , Puromycin Aminonucleoside/pharmacology , Angiopoietin-Like Protein 3 , Animals , Apoptosis , Hypolipidemic Agents/pharmacology , Mice , Mice, Knockout , Podocytes/metabolism , Podocytes/pathology , Protective Factors , Pseudopodia/metabolism
2.
Endocrinology ; 162(3)2021 03 01.
Article in English | MEDLINE | ID: mdl-33508120

ABSTRACT

COP9 signalosome subunit 5 (CSN5) plays a key role in carcinogenesis of multiple cancers and contributes to the stabilization of target proteins through deubiquitylation. However, the underlying role of CSN5 in thyroid carcinoma has not been reported. In this research, our data showed that CSN5 was overexpressed in thyroid carcinoma tissues compared with paracancerous tissues. Furthermore, a series of gain/loss functional assays were performed to demonstrate the role of CSN5 in facilitating thyroid carcinoma cell proliferation and metastasis. Additionally, we found there was a positive correlation between CSN5 and angiopoietin-like protein 2 (ANGPTL2) protein levels in thyroid carcinoma tissues and that CSN5 promoted thyroid carcinoma cell proliferation and metastasis through ANGPTL2. We also identified the underlying mechanism that CSN5 elevated ANGPTL2 protein level by directly binding it, decreasing its ubiquitination and degradation. Overall, our results highlight the significance of CSN5 in promoting thyroid carcinoma carcinogenesis and implicate CSN5 as a promising candidate for thyroid carcinoma treatment.


Subject(s)
Angiopoietin-like Proteins/physiology , COP9 Signalosome Complex/physiology , Carcinogenesis/genetics , Intracellular Signaling Peptides and Proteins/physiology , Peptide Hydrolases/physiology , Thyroid Neoplasms/genetics , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins/metabolism , Animals , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Processing, Post-Translational/genetics , Proteolysis , Signal Transduction/genetics , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Ubiquitination/genetics
3.
Prog Lipid Res ; 80: 101067, 2020 11.
Article in English | MEDLINE | ID: mdl-33011191

ABSTRACT

Angiopoietin-like protein (ANGPTL) family members, mainly ANGPTL3, ANGPTL4 and ANGPTL8, are physiological inhibitors of lipoprotein lipase (LPL), and play a critical role in lipoprotein and triglyceride metabolism in response to nutritional cues. ANGPTL8 has been described by different names in various studies and has been ascribed various functions at the systemic and cellular levels. Circulating ANGPTL8 originates mainly from the liver and to a smaller extent from adipose tissues. In the blood, ANGPTL8 forms a complex with ANGPTL3 or ANGPTL4 to inhibit LPL in fed or fasted conditions, respectively. Evidence is emerging for additional intracellular and receptor-mediated functions of ANGPTL8, with implications in NFκB mediated inflammation, autophagy, adipogenesis, intra-cellular lipolysis and regulation of circadian clock. Elevated levels of plasma ANGPTL8 are associated with metabolic syndrome, type 2 diabetes, atherosclerosis, hypertension and NAFLD/NASH, even though the precise relationship is not known. Whether ANGPTL8 has direct pathogenic role in these diseases, remains to be explored. In this review, we develop a balanced view on the proposed association of this protein in the regulation of several pathophysiological processes. We also discuss the well-established functions of ANGPTL8 in lipoprotein metabolism in conjunction with the emerging novel extracellular and intracellular roles of ANGPTL8 and the implicated metabolic and signalling pathways. Understanding the diverse functions of ANGPTL8 in various tissues and metabolic states should unveil new opportunities of therapeutic intervention for cardiometabolic disorders.


Subject(s)
Angiopoietin-like Proteins/physiology , Cardiovascular Diseases/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism , Peptide Hormones/physiology , Angiopoietin-Like Protein 3 , Angiopoietin-Like Protein 8 , Angiopoietin-like Proteins/blood , Angiopoietin-like Proteins/genetics , Circadian Rhythm , Gene Expression Regulation , Humans , Inflammation/metabolism , Insulin/metabolism , Lipid Metabolism , Peptide Hormones/blood
4.
Sci Rep ; 10(1): 12859, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32732946

ABSTRACT

ANGPTL8, an important regulator of glucose and lipid metabolism, is associated with diabetes, but the role of ANGPTL8 in the outcomes of novel subgroups of diabetes remains unclear. To assess the circulating ANGPTL8 levels in novel subgroups of diabetes and their association with health outcomes, we performed a data-driven cluster analysis (k-means) of patients with newly diagnosed diabetes (741 patients enrolled from 2011 through 2016) from the Risk Evaluation of Cancers in Chinese Diabetic Individuals: a longitudinal (REACTION) study. The primary outcomes were mortality from all causes and cardiovascular diseases (CVD), and the secondary outcome was any cardiovascular event. Comparisons among groups were performed using the Kruskal-Wallis test, and the correlations between variables were assessed using the Pearson correlation test. Logistic regression was used to detect associations between the risk of outcomes and the ANGPTL8 levels. We identified four replicable clusters of patients with diabetes that exhibited significantly different patient characteristics and risks of all-cause mortality. The serum ANGPTL8 levels in the cluster of mild age-related diabetes (MARD), severe insulin-resistant diabetes (SIRD), and severe insulin-deficient diabetes (SIDD) were significantly higher than those in the mild obesity-related diabetes (MOD) cluster (685.01 ± 24.50 vs. 533.5 ± 18.39, p < 0.001; 649.69 ± 55.83 vs. 533.5 ± 18.39, = 0.040; 643.29 ± 30.89 vs. 533.5 ± 18.39, p = 0.001). High circulating ANGPTL8 levels were more highly associated with a greater hazard of all-cause mortality (quartile 4 vs 1: risk ratio [RR] 3.23, 95% CI 1.13-9.22; per unit increase in the Z score: RR 1.53, 95% CI 1.17-2.01) than low circulating ANGPTL8 levels. In conclusion, this 5-year follow-up REACTION study revealed that the circulating ANGPTL8 levels show differences among novel subgroups of adult patients with diabetes and are associated with all-cause mortality in the subsequent 5 years.


Subject(s)
Angiopoietin-like Proteins/blood , Diabetes Mellitus/genetics , Diabetes Mellitus/mortality , Genetic Association Studies , Peptide Hormones/blood , Adult , Angiopoietin-Like Protein 8 , Angiopoietin-like Proteins/physiology , Biomarkers/blood , Cardiovascular Diseases/etiology , Cardiovascular Diseases/mortality , Diabetes Complications/etiology , Diabetes Complications/mortality , Diabetes Mellitus/diagnosis , Female , Follow-Up Studies , Humans , Insulin Resistance , Male , Peptide Hormones/physiology , Severity of Illness Index , Time Factors
5.
FASEB J ; 34(10): 13548-13560, 2020 10.
Article in English | MEDLINE | ID: mdl-32786125

ABSTRACT

Angptl7 is a secreted and circulating cytokine that belongs to Angiopoietin-like family. The current knowledge about the function of Angptl7 is still limited, and its biological role is only marginally known, such as in the promotion of angiogenesis and inflammation. Here, we demonstrated that Angptl7 promotes insulin resistance and type 2 diabetes mellitus (T2DM). We found that the circulating Angptl7 levels in T2DM patient and mouse models were significantly elevated. Artificial overexpression of Angptl7 in hepatic cells inhibited glucose uptake and impaired insulin signaling pathway. Furthermore, in vivo overexpression of Angptl7 in experimental healthy mice also caused insulin resistance-like characteristics. Mechanistic studies revealed that Angptl7 can upregulate SOCS3 expression, leading to the IRS1 degradation in proteasome. Furthermore, over-expressed Angptl7 inhibited the phosphorylation of Akt and promoted the phosphorylation of ERK1/2, which was known to be associated with insulin resistance. Taken together, our study provided strong evidence that Angptl7 promotes insulin resistance and T2DM by multiple mechanisms, which made Angptl7 a new potential therapeutic target for treatment of insulin resistance and T2DM.


Subject(s)
Angiopoietin-like Proteins , Diabetes Mellitus, Type 2/metabolism , Hepatocytes , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , Suppressor of Cytokine Signaling 3 Protein/metabolism , Aged , Angiopoietin-Like Protein 7 , Angiopoietin-like Proteins/blood , Angiopoietin-like Proteins/physiology , Animals , Case-Control Studies , Cohort Studies , Female , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged
6.
JCI Insight ; 5(17)2020 09 03.
Article in English | MEDLINE | ID: mdl-32730227

ABSTRACT

The angiopoietin-like protein ANGPTL8 (A8) is one of 3 ANGPTLs (A8, A3, A4) that coordinate changes in triglyceride (TG) delivery to tissues by inhibiting lipoprotein lipase (LPL), an enzyme that hydrolyzes TG. Previously we showed that A8, which is expressed in liver and adipose tissue, is required to redirect dietary TG from oxidative to storage tissues following food intake. Here we show that A8 from liver and adipose tissue have different roles in this process. Mice lacking hepatic A8 have no circulating A8, high intravascular LPL activity, low plasma TG levels, and evidence of decreased delivery of dietary lipids to adipose tissue. In contrast, mice lacking A8 in adipose tissue have higher postprandial TG levels and similar intravascular LPL activity and plasma A8 levels and higher levels of plasma TG. Expression of A8, together with A4, in cultured cells reduced A4 secretion and A4-mediated LPL inhibition. Thus, hepatic A8 (with A3) acts in an endocrine fashion to inhibit intravascular LPL in oxidative tissues, whereas A8 in adipose tissue enhances LPL activity by autocrine/paracrine inhibition of A4. These combined actions of A8 ensure that TG stores are rapidly replenished and sufficient energy is available until the next meal.


Subject(s)
Adipose Tissue/metabolism , Angiopoietin-like Proteins/physiology , Autocrine Communication , Dietary Fats/metabolism , Lipoprotein Lipase/metabolism , Liver/metabolism , Triglycerides/metabolism , Adipose Tissue/cytology , Angiopoietin-Like Protein 8 , Animals , Female , Liver/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Paracrine Communication
7.
Clin Chim Acta ; 503: 19-34, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31923423

ABSTRACT

Hypertriglyceridemia is a risk factor for a series of diseases, such as cardiovascular disease (CVD), diabetes and nonalcoholic fatty liver disease (NAFLD). Angiopoietin-like proteins (ANGPTLs) family, especially ANGPTL3, ANGPTL4 and ANGPTL8, which regulate lipoprotein lipase (LPL) activity, play pivotal roles in triglyceride (TG) metabolism and related diseases/complications. There are many transcriptional and post-transcriptional factors that participate in physiological and pathological regulation of ANGPTLs to affect triglyceride metabolism. This review is intended to focus on the similarity and difference in the expression, structural features, regulation profile of the three ANGPTLs and inhibitory models for LPL. Description of the regulatory factors of ANGPTLs and the properties in regulating the lipid metabolism involved in the underlying mechanisms in pathological effects on diseases will provide potential therapeutic approaches for the treatment of dyslipidemia related diseases.


Subject(s)
Angiopoietin-like Proteins/physiology , Lipoprotein Lipase/antagonists & inhibitors , Triglycerides/metabolism , Angiopoietin-Like Protein 3 , Angiopoietin-Like Protein 4 , Angiopoietin-Like Protein 8 , Animals , Humans , Lipoprotein Lipase/metabolism , Peptide Hormones
8.
Endocr J ; 66(12): 1039-1046, 2019 Dec 25.
Article in English | MEDLINE | ID: mdl-31631098

ABSTRACT

Type 2 diabetes mellitus (T2DM) is a chronic metabolic disease with increasing prevalence worldwide. Angiopoietin-like protein 8 (ANGPTL8), a member of the angiopoietin-like protein family, is involved in glucose metabolism, lipid metabolism, and energy homeostasis and believed to be associated with T2DM. Expression levels of ANGPTL8 are often significantly altered in metabolic diseases, such as non-alcoholic fatty liver disease (NAFLD) and diabetes mellitus. Studies have shown that ANGPTL8, together with other members of this protein family, such as angiopoietin-like protein 3 (ANGPTL3) and angiopoietin-like protein 4 (ANGPTL4), regulates the activity of lipoprotein lipase (LPL), thereby participating in the regulation of triglyceride related lipoproteins (TRLs). In addition, members of the angiopoietin-like protein family are varyingly expressed among different tissues and respond differently under diverse nutritional and metabolic status. These findings may provide new options for the diagnosis and treatment of diabetes, metabolic syndromes and other diseases. In this review, the interaction between ANGPTL8 and ANGPTL3 or ANGPTL4, and the differential expression of ANGPTL8 responding to different nutritional and metabolic status during the regulation of LPL activity were reviewed.


Subject(s)
Angiopoietin-like Proteins/genetics , Angiopoietin-like Proteins/physiology , Gene Expression Regulation/physiology , Metabolic Diseases/metabolism , Nutritional Status/physiology , Peptide Hormones/genetics , Angiopoietin-Like Protein 3 , Angiopoietin-Like Protein 4/physiology , Angiopoietin-Like Protein 8 , Animals , Diabetes Mellitus, Type 2 , Glucagon-Like Peptide 1/physiology , Glucose/metabolism , Homeostasis , Humans , Insulin/physiology , Lipid Metabolism , Lipoprotein Lipase , Peptide Hormones/physiology
9.
Invest Ophthalmol Vis Sci ; 59(10): 4210-4217, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30128492

ABSTRACT

Purpose: Fenofibrate has been demonstrated to exert a promising therapeutic effect against diabetic retinopathy. Angiopoietin-like 3 (ANGPTL3) has been shown to exert significant pathogenic effects on vascular endothelial cells, which are critically involved in the pathogenesis of diabetic retinopathy. The present study aimed to investigate the link between the therapeutic effects of fenofibrate and the pathogenic effects of ANGPTL3 in diabetic retinopathy. Methods: Diabetic and control rats were randomly assigned to the following treatments: intravitreal injection with ANGPTL3 small interfering RNA (siRNA), recombinant human (rh)ANGPTL3, fed with normal feeds, or fenofibrate-containing feeds for 8 weeks. Human retinal microvascular endothelial cells (HRMECs) were exposed to normal glucose or high glucose levels with ANGPTL3 siRNA, ANGPTL3 RNA overexpression, blank vector, cilengitide, or fenofibrate treatment. Expression levels of ANGPTL3, IL-1, IL-6, Bax, P53, VEGF, and integrin αVß3 in the retinas of rats and HRMECs were examined by Western blotting and real-time PCR. The apoptosis rates of HRMECs were examined using a TUNEL apoptosis assay kit. Results: Expression levels of ANGPTL3, IL-1ß, IL-6, Bax, P53, VEGF, and integrin αVß3 were found to be upregulated after high-glucose stimulation or ANGPTL3 overexpression in HRMECs or diabetic retinal tissue. However, expression levels of the above markers were downregulated following fenofibrate intervention, blockage of integrin αVß3 receptor, or ANGPTL3 siRNA interference. Conclusions: We identified fenofibrate exerts its protective effects by inhibiting ANGPTL3-induced apoptosis and inflammation in diabetic retinopathy, which is a novel mechanism.


Subject(s)
Angiopoietin-like Proteins/antagonists & inhibitors , Diabetes Mellitus, Experimental/drug therapy , Fenofibrate/therapeutic use , Hypolipidemic Agents/therapeutic use , Angiopoietin-like Proteins/physiology , Animals , Apoptosis/drug effects , Cells, Cultured , Cytokines/metabolism , Diabetes Mellitus, Experimental/metabolism , Endothelial Cells , Glucose/pharmacology , Intravitreal Injections , Male , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Random Allocation , Rats , Retina/cytology
10.
Proc Natl Acad Sci U S A ; 115(6): E1249-E1258, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29358393

ABSTRACT

Dietary triglyceride (TG) is the most efficient energy substrate. It is processed and stored at substantially lower metabolic cost than is protein or carbohydrate. In fed animals, circulating TGs are preferentially routed for storage to white adipose tissue (WAT) by angiopoietin-like proteins 3 (A3) and 8 (A8). Here, we show that mice lacking A3 and A8 (A3-/-A8-/- mice) have decreased fat mass and a striking increase in temperature (+1 °C) in the fed (but not fasted) state, without alterations in food intake or physical activity. Subcutaneous WAT (WAT-SQ) from these animals had morphologic and metabolic changes characteristic of beiging. O2 consumption rates (OCRs) and expression of genes involved in both fatty acid synthesis and fatty acid oxidation were increased in WAT-SQ of A3-/-A8-/- mice, but not in their epididymal or brown adipose tissue (BAT). The hyperthermic response to feeding was blocked by maintaining A3-/-A8-/- mice at thermoneutrality or by treating with a ß3-adrenergic receptor (AR) antagonist. To determine if sympathetic stimulation was sufficient to increase body temperature in A3-/-A8-/- mice, WT and A3-/-A8-/- animals were maintained at thermoneutrality and then treated with a ß3-AR agonist; treatment induced hyperthermia in A3-/-A8-/- , but not WT, mice. Antibody-mediated inactivation of both circulating A3 and A8 induced hyperthermia in WT mice. Together, these data indicate that A3 and A8 are essential for efficient storage of dietary TG and that disruption of these genes increases feeding-induced thermogenesis and energy utilization.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Angiopoietin-like Proteins/physiology , Thermogenesis/physiology , Triglycerides/metabolism , Angiopoietin-Like Protein 3 , Angiopoietin-Like Protein 8 , Animals , Dietary Fats , Female , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen Consumption , Receptors, Adrenergic, beta-3/metabolism
11.
Osteoarthritis Cartilage ; 26(1): 108-117, 2018 01.
Article in English | MEDLINE | ID: mdl-29074299

ABSTRACT

OBJECTIVE: Chondrocyte differentiation is crucial for long bone growth. Many cartilage extracellular matrix (ECM) proteins reportedly contribute to chondrocyte differentiation, indicating that mechanisms underlying chondrocyte differentiation are likely more complex than previously appreciated. Angiopoietin-like protein 2 (ANGPTL2) is a secreted factor normally abundantly produced in mesenchymal lineage cells such as adipocytes and fibroblasts, but its loss contributes to the pathogenesis of lifestyle- or aging-related diseases. However, the function of ANGPTL2 in chondrocytes, which are also differentiated from mesenchymal stem cells, remains unclear. Here, we investigate whether ANGPTL2 is expressed in or functions in chondrocytes. METHODS: First, we evaluated Angptl2 expression during chondrocyte differentiation using chondrogenic ATDC5 cells and wild-type epiphyseal cartilage of newborn mice. We next assessed ANGPTL2 function in chondrogenic differentiation and associated signaling using Angptl2 knockdown ATDC5 cells and Angptl2 knockout mice. RESULTS: ANGPTL2 is expressed in chondrocytes, particularly those located in resting and proliferative zones, and accumulates in ECM surrounding chondrocytes. Interestingly, long bone growth was retarded in Angptl2 knockout mice from neonatal to adult stages via attenuation of chondrocyte differentiation. Both in vivo and in vitro experiments show that changes in ANGPTL2 expression can also alter p38 mitogen-activated protein kinase (MAPK) activity mediated by integrin α5ß1. CONCLUSION: ANGPTL2 contributes to chondrocyte differentiation and subsequent endochondral ossification through α5ß1 integrin and p38 MAPK signaling during bone growth. Our findings provide insight into molecular mechanisms governing communication between chondrocytes and surrounding ECM components in bone growth activities.


Subject(s)
Angiopoietin-like Proteins/physiology , Bone Development/physiology , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins/metabolism , Animals , Animals, Newborn , Cell Differentiation/physiology , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis/physiology , Enzyme Inhibitors/pharmacokinetics , Femur/growth & development , Imidazoles/pharmacokinetics , MAP Kinase Signaling System/physiology , Matrilin Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Pyridines/pharmacokinetics , Tibia/growth & development
12.
Nat Rev Endocrinol ; 13(12): 731-739, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28984319

ABSTRACT

Triglycerides and cholesterol circulate in the bloodstream as part of various lipoprotein particles. Three members of the angiopoietin-like (ANGPTL) protein family - ANGPTL3, ANGPTL4 and ANGPTL8 - have emerged as important regulators of plasma lipoprotein levels by inhibiting the enzyme lipoprotein lipase. Here, I review the role of ANGPTL3 in lipoprotein metabolism. In contrast to ANGPTL4 and ANGPTL8, ANGPTL3 is exclusively produced in the liver and can therefore be classified as a true hepatokine. ANGPTL3 cooperates with ANGPTL8 to inhibit lipoprotein lipase and is mostly active after feeding, whereas ANGPTL4 is mostly active after fasting. Inactivation of ANGPTL3 in mice reduces plasma triglyceride and free fatty acid levels and suppresses atherosclerosis. In humans, homozygous loss-of-function mutations in ANGPTL3 lead to low plasma levels of low-density lipoproteins, high-density lipoproteins and triglycerides, a condition referred to as familial combined hypolipidaemia. Heterozygous carriers of loss-of-function mutations in ANGPTL3 have a lower risk of coronary artery disease than non-carriers. At present, researchers are investigating antisense oligonucleotide and monoclonal antibody-based inactivation of ANGPTL3 in human clinical trials for the therapeutic management of dyslipidaemia and atherosclerosis. Thus, ANGPTL3 is an important liver-derived regulator of lipoprotein metabolism that holds considerable promise as a target for atherosclerosis.


Subject(s)
Angiopoietin-like Proteins/physiology , Lipoproteins/metabolism , Angiopoietin-Like Protein 3 , Angiopoietin-like Proteins/antagonists & inhibitors , Angiopoietin-like Proteins/genetics , Animals , Antibodies, Monoclonal/therapeutic use , Atherosclerosis/therapy , Dyslipidemias/therapy , Fasting , Fatty Acids, Nonesterified/blood , Food , Humans , Lipoprotein Lipase/antagonists & inhibitors , Liver/metabolism , Mice , Mutation , Oligonucleotides, Antisense/therapeutic use , Triglycerides/blood
13.
Diabetes Res Clin Pract ; 133: 168-177, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28965028

ABSTRACT

Insulin resistance is prevalent worldwide and is associated with many metabolic diseases, in particular, type 2 diabetes mellitus (T2DM), obesity, nonalcoholic fatty liver disease (NAFLD), polycystic ovary syndrome (PCOS) and metabolic syndrome (MetS). Angiopoietin-like protein 8 (ANGPTL8), a newly-identified secreted protein composing of 198 amino acids, is enriched in the liver of human. Considering its promising potential for ß-cell proliferation and therapeutic prospect for diabetes, ANGPTL8 has aroused extensive interests. However, a recent collaborative study confirmed that ANGPTL8 didn't stimulate dramatic ß-cell regeneration. At present, a controversial scientific discussion on whether and how ANGPTL8 regulate insulin resistance has been ongoing. Interestingly, several in vitro and in vivo studies have suggested the complex roles of ANGPTL8 in insulin resistance. Data resulting from cross-sectional and longitudinal researches in human individuals involving the influence of ANGPTL8 on the development of insulin resistance were controversial. We therefore summarize currently clinical literature to exploit whether this exciting hormone could be applied for clinical application asa potential clinical biomarker to predict insulin resistance and related disorders.


Subject(s)
Angiopoietin-like Proteins/physiology , Diabetes Mellitus, Type 2/blood , Insulin Resistance , Peptide Hormones/physiology , Angiopoietin-Like Protein 8 , Animals , Biomarkers/blood , Diabetes Mellitus, Type 2/diagnosis , Humans , Insulin-Secreting Cells/metabolism , Non-alcoholic Fatty Liver Disease/blood , Obesity/blood
14.
Circ J ; 81(10): 1379-1385, 2017 Sep 25.
Article in English | MEDLINE | ID: mdl-28867689

ABSTRACT

In parallel with the increase in the number of elderly people worldwide, the number of patients with heart disease is also rapidly increasing. Of the heart diseases, cardiovascular disease (CVD) and heart failure (HF) are strongly associated with adverse health outcomes that decrease productivity in later years. Recently, ANGPTL2, a secreted glycoprotein and member of the angiopoietin-like protein family, has received attention as a causal player in the development of CVD and HF. Prolonged ANGPTL2 autocrine/paracrine signaling in vascular tissue leads to chronic inflammation and pathologic tissue remodeling, accelerating CVD development. Excess ANGPTL2 autocrine/paracrine signaling induced in the pathologically stressed heart accelerates cardiac dysfunction by decreasing myocardial energy metabolism. Conversely, ANGPTL2 inactivation in vascular tissue and the heart delays development or progression of CVD and HF, respectively. Moreover, there is increased evidence for an association between elevated circulating ANGPTL2 levels and CVD and HF. Interestingly, ANGPTL2 expression is also associated with cellular senescence, which may promote premature aging and development of aging-associated diseases, including CVD and HF. Overall, ANGPTL2 autocrine/paracrine signaling is a new factor in accelerating heart disease development in the aging. Here, we focus on current topics relevant to ANGPTL2 function in heart disease.


Subject(s)
Angiopoietin-like Proteins/physiology , Heart Diseases/etiology , Aged , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins/blood , Autocrine Communication , Cardiovascular Diseases/etiology , Cardiovascular Diseases/pathology , Cellular Senescence , Heart Diseases/pathology , Heart Failure/etiology , Heart Failure/pathology , Humans , Inflammation , Paracrine Communication
SELECTION OF CITATIONS
SEARCH DETAIL
...