Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Int J Mol Sci ; 22(12)2021 Jun 21.
Article in English | MEDLINE | ID: mdl-34205674

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is strongly associated to the features of metabolic syndrome which can progress to cirrhosis, liver failure and hepatocellular carcinoma. However, the most common cause of mortality in people with NAFLD is not liver-related but stems from atherosclerotic cardiovascular disease (CVD). The prevalence of NAFLD is on the rise, mainly as a consequence of its close association with two major worldwide epidemics, obesity and type 2 diabetes (T2D). The exact pathogenesis of NAFLD and especially the mechanisms leading to disease progression and CVD have not been completely elucidated. Human fetuin-A (alpha-2-Heremans Schmid glycoprotein), a glycoprotein produced by the liver and abundantly secreted into the circulation appears to play a role in insulin resistance, metabolic syndrome and inflammation. This review discusses the links between NAFLD and CVD by specifically focusing on fetuin-A's function in the pathogenesis of NAFLD and atherosclerotic CVD.


Subject(s)
Cardiovascular Diseases/etiology , Non-alcoholic Fatty Liver Disease/etiology , alpha-2-HS-Glycoprotein/physiology , Animals , Fibrosis , Humans , Liver/pathology
2.
J Pediatr Endocrinol Metab ; 34(7): 911-916, 2021 Jul 27.
Article in English | MEDLINE | ID: mdl-33887129

ABSTRACT

OBJECTIVES: Polycystic ovary syndrome (PCOS) is an endocrinopathy, in which hyperandrogenism and hyperinsulinism have both occurred. Fetuin-A, a natural inhibitor of tyrosine kinase, leads to insulin resistance. The aim was to evaluate the relationship between fetuin-A and hyperandrogenism and hyperinsulinism and the role of fetuin-A in the pathophysiology of PCOS. METHODS: Thirty-eight cases with PCOS and 40 healthy adolescents were included in the study. PCOS and controls were divided into obese/non-obese subgroups. LH, FSH, total and free testosterone (TT, FT), SHBG, androstenedione, DHEAS were measured in patients with PCOS. Fasting glucose, insulin, lipid profile, AST, ALT, HsCRP, and fetuin levels of PCOS patients and healthy controls were also measured. RESULTS: Fetuin-A levels were higher in PCOS patients than in controls. In the obese-PCOS group, when compared to non-obese PCOS patients; the levels of SHBG and HDL were low while cholesterol, LDL, triglyceride, HOMA-IR, FT, FAI, and HSCRP levels were high, but Fetuin-A levels were similar. In the obese-PCOS group, fetuin-A levels were higher than in obese-controls. HOMA-IR and fetuin-A levels were higher in non-obese PCOS patients than in non-obese controls. In the PCOS group, fetuin-A was positively correlated with TT, FT, FAI and androstenedione and negatively correlated with SHBG. Regression analysis demonstrated that FT, SHBG, and androstenedione significantly predicted fetuin-A levels (R2=54%). In non-obese PCOS patients and controls, fetuin-A was positively correlated with insulin and HOMA-IR. CONCLUSIONS: These results suggest a relationship between androgen levels and fetuin-A in PCOS cases, independent of insulin resistance, and may shed light on further studies.


Subject(s)
Polycystic Ovary Syndrome/etiology , alpha-2-HS-Glycoprotein/physiology , Adolescent , Female , Humans , Insulin/blood , Insulin Resistance , Lipids/blood , Polycystic Ovary Syndrome/blood , Sex Hormone-Binding Globulin/analysis , Testosterone/blood , alpha-2-HS-Glycoprotein/analysis
3.
PLoS One ; 15(2): e0228938, 2020.
Article in English | MEDLINE | ID: mdl-32074140

ABSTRACT

Calcifications can disrupt organ function in the cardiovascular system and the kidney, and are particularly common in patients with chronic kidney disease (CKD). Fetuin-A deficient mice maintained against the genetic background DBA/2 exhibit particularly severe soft tissue calcifications, while fetuin-A deficient C57BL/6 mice remain healthy. We employed molecular genetic analysis to identify risk factors of calcification in fetuin-A deficient mice. We sought to identify pharmaceutical therapeutic targets that could be influenced by dietary of parenteral supplementation. We studied the progeny of an intercross of fetuin-A deficient DBA/2 and C57BL/6 mice to identify candidate risk genes involved in calcification. We determined that a hypomorphic mutation of the Abcc6 gene, a liver ATP transporter supplying systemic pyrophosphate, and failure to regulate the Trpm6 magnesium transporter in kidney were associated with severity of calcification. Calcification prone fetuin-A deficient mice were alternatively treated with parenteral administration of fetuin-A dietary magnesium supplementation, phosphate restriction, or by or parenteral pyrophosphate. All treatments markedly reduced soft tissue calcification, demonstrated by computed tomography, histology and tissue calcium measurement. We show that pathological ectopic calcification in fetuin-A deficient DBA/2 mice is caused by a compound deficiency of three major extracellular and systemic inhibitors of calcification, namely fetuin-A, magnesium, and pyrophosphate. All three of these are individually known to contribute to stabilize protein-mineral complexes and thus inhibit mineral precipitation from extracellular fluid. We show for the first time a compound triple deficiency that can be treated by simple dietary or parenteral supplementation. This is of special importance in patients with advanced CKD, who commonly exhibit reduced serum fetuin-A, magnesium and pyrophosphate levels.


Subject(s)
Calcinosis/pathology , Microvessels/physiology , alpha-2-HS-Glycoprotein/metabolism , Animals , Calcinosis/genetics , Diphosphates/metabolism , Disease Models, Animal , Female , Kidney/pathology , Liver/pathology , Magnesium/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Microvessels/metabolism , Minerals , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Renal Insufficiency, Chronic/complications , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , alpha-2-HS-Glycoprotein/physiology , alpha-Fetoproteins
4.
Atherosclerosis ; 292: 75-83, 2020 01.
Article in English | MEDLINE | ID: mdl-31783201

ABSTRACT

BACKGROUND & AIMS: The role of epicardial adipose tissue (EAT) in the pathophysiology of late stage-coronary artery disease (CAD) has not been investigated. We explored the association of EAT volume and its proteome with advanced coronary atherosclerosis. METHODS: The EPICHEART Study prospectively enrolled 574 severe aortic stenosis patients referred to cardiac surgery. Before surgery, EAT volume was quantified by computed tomography (CT). During surgery, epicardial, mediastinal (MAT) and subcutaneous (SAT) adipose tissue samples were collected to explore fat phenotype by analyzing the proteomic profile using SWATH-mass spectrometry; pericardial fluid and peripheral venous blood were also collected. CAD presence was defined as coronary artery stenosis ≥50% in invasive angiography and by CT-derived Agatston coronary calcium score (CCS). RESULTS: EAT volume adjusted for body fat was associated with higher CCS, but not with the presence of coronary stenosis. In comparison with mediastinal and subcutaneous fat depots, EAT exhibited a pro-calcifying proteomic profile in patients with CAD characterized by upregulation of annexin-A2 and downregulation of fetuin-A; annexin-A2 protein levels in EAT samples were also positively correlated with CCS. We confirmed that the annexin-A2 gene was overexpressed in EAT samples of CAD patients and positively correlated with CCS. Fetuin-A gene was not detected in EAT samples, but systemic fetuin-A was higher in CAD than in non-CAD patients, suggesting that fetuin-A was locally downregulated. CONCLUSIONS: In an elderly cohort of stable patients, CCS was associated with EAT volume and annexin-A2/fetuin-A signaling, suggesting that EAT might orchestrate pro-calcifying conditions in the late phases of CAD.


Subject(s)
Adipose Tissue/anatomy & histology , Adipose Tissue/diagnostic imaging , Annexin A2/analysis , Annexin A2/physiology , Coronary Artery Disease/diagnostic imaging , Pericardium/anatomy & histology , Pericardium/diagnostic imaging , Signal Transduction , Tomography, X-Ray Computed , Vascular Calcification/diagnostic imaging , alpha-2-HS-Glycoprotein/analysis , alpha-2-HS-Glycoprotein/physiology , Adipose Tissue/chemistry , Aged , Aged, 80 and over , Biomarkers/analysis , Coronary Artery Disease/blood , Coronary Artery Disease/etiology , Female , Humans , Male , Organ Size , Pericardium/chemistry , Prospective Studies , Proteomics , Severity of Illness Index , Vascular Calcification/blood , Vascular Calcification/etiology
5.
Nat Rev Endocrinol ; 13(9): 509-520, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28621339

ABSTRACT

Hepatic steatosis is an underlying feature of nonalcoholic fatty liver disease (NAFLD), which is the most common form of liver disease and is present in up to ∼70% of individuals who are overweight. NAFLD is also associated with hypertriglyceridaemia and low levels of HDL, glucose intolerance, insulin resistance and type 2 diabetes mellitus. Hepatic steatosis is a strong predictor of the development of insulin resistance and often precedes the onset of other known mediators of insulin resistance. This sequence of events suggests that hepatic steatosis has a causal role in the development of insulin resistance in other tissues, such as skeletal muscle. Hepatokines are proteins that are secreted by hepatocytes, and many hepatokines have been linked to the induction of metabolic dysfunction, including fetuin A, fetuin B, retinol-binding protein 4 (RBP4) and selenoprotein P. In this Review, we describe the factors that influence the development of hepatic steatosis, provide evidence of strong links between hepatic steatosis and insulin resistance in non-hepatic tissues, and discuss recent advances in our understanding of how steatosis alters hepatokine secretion to influence metabolic phenotypes through inter-organ communication.


Subject(s)
Blood Proteins/physiology , Insulin Resistance , Liver/physiopathology , Non-alcoholic Fatty Liver Disease/physiopathology , Animals , Diabetes Mellitus, Type 2 , Diet, High-Fat , Fetuin-B/physiology , Humans , Hypertriglyceridemia , Lipid Metabolism/physiology , Obesity , Overweight , Retinol-Binding Proteins, Plasma/physiology , Selenoprotein P/physiology , alpha-2-HS-Glycoprotein/physiology
6.
Acta Biochim Pol ; 63(3): 459-67, 2016.
Article in English | MEDLINE | ID: mdl-27262842

ABSTRACT

Nowadays non-alcoholic fatty liver disease (NAFLD) is becoming the most common chronic liver pathology both in adults and children. NAFLD manifestation ranges from a simple liver steatosis to steatohepatitis (nonalcoholic steatohepatitis - NASH), which may progress to advanced fibrosis, cirrhosis and end-stage liver disease. Due to the coexistence of visceral obesity, insulin resistance and dyslipidemia, NAFLD is considered to be the hepatic manifestation of metabolic syndrome. In recent years, in the pathogenesis of metabolic syndrome, type 2 diabetes mellitus, cardiovascular disease and also NAFLD, more and more attention has been paid to the so-called organokines, proteins with both paracrine or/and endocrine activities. These include most known adipokines (mainly produced by adipose tissue), myokines (mainly produced by skeletal muscles) and hepatokines exclusively or predominantly produced by the liver. It was shown that the liver may affect the lipids and glucose metabolism by hepatokines released into the blood and NAFLD seems to be associated with altered hepatokines production. Fetuin-A, fibroblast growth factor-21 (FGF-21), selenoprotein P, sex hormone-binding globulin (SHBG), angiopoietin-related growth factor (also known as angiopoietin-related protein 6) and leukocyte derived chemotaxin 2 (LECT2) are considered as the most important hepatokines. In this review, we provide an overview of the main hepatokines and we summarize the association of liver-derived proteins with the development and progression of NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease/metabolism , Animals , Fibroblast Growth Factors/physiology , Humans , Intercellular Signaling Peptides and Proteins/physiology , Liver/metabolism , Liver/pathology , Selenoprotein P/physiology , Sex Hormone-Binding Globulin/physiology , Signal Transduction , alpha-2-HS-Glycoprotein/physiology
7.
Ann Biol Clin (Paris) ; 73(3): 315-22, 2015.
Article in French | MEDLINE | ID: mdl-26069068

ABSTRACT

Vascular calcification is a marker of cardiovascular risk increase. Age and specific disease such as diabetes or chronic kidney disease are important factors for calcification genesis. Vascular calcification process is a complex phenomenon, involving several activators and inhibitors factors. Indeed, recent works related to in vitro and in vivo experimental studies have led to a better understanding of calcification process and identification of molecules able to modulate this system. This revue will summarize some of these molecules with a particular interest of those with therapeutic relevance. We will present: i) calcium sensing receptor and its modulation by cinacalcet; ii) pyrophosphate supplementation; iii) fetuin A and overall propensity serum test for calcification and finally; iv) matrix-Gla-protein and the use of vitamin K to prevent vascular calcification progression.


Subject(s)
Cardiovascular Diseases/prevention & control , Vascular Calcification/prevention & control , Calcium-Binding Proteins/physiology , Cardiovascular Diseases/etiology , Diphosphates/therapeutic use , Extracellular Matrix Proteins/physiology , Humans , Vascular Calcification/etiology , alpha-2-HS-Glycoprotein/physiology , Matrix Gla Protein
8.
Mol Cell Endocrinol ; 412: 1-11, 2015 Sep 05.
Article in English | MEDLINE | ID: mdl-25986658

ABSTRACT

OBJECTIVE: Fetuin A (FetA), a secreted glycoprotein, is known to affect inflammation and insulin resistance (IR) in obese humans and animals. Lipotoxicity from chronic hyperlipidemia damages pancreatic ß cells, hastening the onset of diabetes. We sought to determine whether FetA promotes lipotoxicity through modulation of the toll-like receptor 4 (TLR4) inflammatory signaling pathway as well as the protective effect of pioglitazone(PIO) on lipotoxicity. METHODS: ßTC6, a glucose-sensitive mouse pancreatic ß cell line, and Sprague-Dawley rats with diet-induced obesity, were used to investigate FetA-mediated lipotoxicity. Protein expression/activation were measured by Western blotting. Small interfering (si)RNAs for TLR4 were used. Cell apoptosis was quantified by TUNEL analysis or flow cytometry, respectively. Insulin release was assessed with an insulin ELISA. RESULTS: FetA dose-dependently aggravated palmitic acid (PA)-induced ßTC6 cell apoptosis, insulin secretion impairment, and inhibition of the expression of G-protein-coupled receptor 40 (GPR40) and pancreatic duodenal homeobox-1(PDX-1). Combined FetA + PA induced TLR4 expression, and subsequent inhibition of TLR4 signaling or expression was shown to prevent the strengthening effect of FetA on PA-induced lipotoxicity in ßTC6 cells. FetA + PA induced p-JNK and nuclear factor-κB (NF-κB) subunit P65 expression, and inhibition of this activity reduced PA+ FetA lipotoxicity in ßTC6 cells. PIO could ameliorate PA+ FetA-induced damage to ßTC6 cells. Similarly, PIO improved insulin secretion disorder, reduced apoptosis, decreased FetA, TLR4, p-JNK, NF-κB subunit P65 and cleaved caspase 3 expression, and increased GPR40 and PDX-1 expression in islet ß cells of diet-induced obese rats. The correlative bivariate analysis showed that increases in Fetuin A were directly proportional to the development of ß cell injury. CONCLUSIONS: FetA can promote lipotoxicity in ß cells through the TLR4-JNK-NF-κB signaling pathway. The protective effects of PIO on lipotoxicity in ß cells may involve the inhibition of the activation of the FetA and TLR4 signaling pathway.


Subject(s)
Insulin-Secreting Cells/metabolism , Thiazolidinediones/pharmacology , Toll-Like Receptor 4/metabolism , alpha-2-HS-Glycoprotein/physiology , Animals , Apoptosis , Cell Line , Diet, High-Fat/adverse effects , Drug Evaluation, Preclinical , Homeodomain Proteins/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/immunology , Lipid Metabolism , Lipopolysaccharides/pharmacology , Male , Neoplasm Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Obesity/etiology , Obesity/metabolism , Obesity/pathology , PPAR gamma/agonists , Palmitic Acid/pharmacology , Pioglitazone , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Trans-Activators/metabolism
11.
Osteoporos Int ; 25(5): 1555-62, 2014 May.
Article in English | MEDLINE | ID: mdl-24570294

ABSTRACT

UNLABELLED: Using mediation analysis, a causal relationship between the AHSG gene and bone mineral density (BMD) through fetuin-A and body mass index (BMI) mediators was suggested. INTRODUCTION: Fetuin-A, a multifunctional protein of hepatic origin, is associated with bone mineral density. It is unclear if this association is causal. This study aimed at clarification of this issue. METHODS: A cross-sectional study was conducted among 1,741 healthy workers from the Electricity Generating Authority of Thailand (EGAT) cohort. The alpha-2-Heremans-Schmid glycoprotein (AHSG) rs2248690 gene was genotyped. Three mediation models were constructed using seemingly unrelated regression analysis. First, the ln[fetuin-A] group was regressed on the AHSG gene. Second, the BMI group was regressed on the AHSG gene and the ln[fetuin-A] group. Finally, the BMD model was constructed by fitting BMD on two mediators (ln[fetuin-A] and BMI) and the independent AHSG variable. All three analyses were adjusted for confounders. RESULTS: The prevalence of the minor T allele for the AHSG locus was 15.2%. The AHSG locus was highly related to serum fetuin-A levels (P < 0.001). Multiple mediation analyses showed that AHSG was significantly associated with BMD through the ln[fetuin-A] and BMI pathway, with beta coefficients of 0.0060 (95% CI 0.0038, 0.0083) and 0.0030 (95% CI 0.0020, 0.0045) at the total hip and lumbar spine, respectively. About 27.3 and 26.0% of total genetic effects on hip and spine BMD, respectively, were explained by the mediation effects of fetuin-A and BMI. CONCLUSIONS: Our study suggested evidence of a causal relationship between the AHSG gene and BMD through fetuin-A and BMI mediators.


Subject(s)
Body Mass Index , Bone Density/genetics , alpha-2-HS-Glycoprotein/genetics , alpha-2-HS-Glycoprotein/physiology , Absorptiometry, Photon/methods , Adult , Bone Density/physiology , Cross-Sectional Studies , Female , Genetic Association Studies , Genotype , Hip Joint/physiology , Humans , Lumbar Vertebrae/physiology , Male , Middle Aged , Motor Activity/physiology , alpha-2-HS-Glycoprotein/analysis
12.
Diabetologia ; 57(5): 1057-66, 2014 May.
Article in English | MEDLINE | ID: mdl-24493202

ABSTRACT

AIMS/HYPOTHESIS: Fetuin-A (alpha2-Heremans-Schmid glycoprotein), a liver-derived circulating glycoprotein, contributes to lipid disorders, diabetes and cardiovascular diseases. In a previous study we found that perivascular fat cells (PVFCs) have a higher angiogenic potential than other fat cell types. The aim was to examine whether fetuin-A influences PVFC and vascular cell growth and the expression and secretion of proinflammatory and angiogenic proteins, and whether TLR4-independent pathways are involved. METHODS: Mono- and co-cultures of human PVFCs and endothelial cells were treated with fetuin-A and/or palmitate for 6-72 h. Proteins were quantified by ELISA and Luminex, mRNA expression by real-time PCR, and cell growth by BrDU-ELISA. Some PVFCs were preincubated with a nuclear factor κB NFκBp65 inhibitor, or the toll-like receptor 4 (TLR4) inhibitor CLI-095, or phosphoinositide 3-kinase (PI3K)/Akt inhibitors and/or stimulated with insulin. Intracellular forkhead box protein O1 (FoxO1), NFκBp65 and inhibitor of κB kinase ß (IKKß) localisation was visualised by immunostaining. RESULTS: PVFCs expressed and secreted IL-6, IL-8, plasminogen activator inhibitor 1 (PAI-1), basic fibroblast growth factor (bFGF), platelet-derived growth factor (PDGF)-BB, monocyte chemotactic protein-1 (MCP-1), vascular endothelial growth factor (VEGF), placental growth factor (PLGF) and hepatocyte growth factor (HGF). Fetuin-A upregulated IL-6 and IL-8, and this was potentiated by palmitate and blocked by CLI-095. Immunostaining and electrophoretic mobility shift assay (EMSA) showed partial NFκBp65 activation. MCP-1 was upregulated and blocked by CLI-095, but not by palmitate. However, HGF was downregulated, which was slightly potentiated by palmitate. This effect persisted after TLR4 pathway blockade. Stimulation of insulin-PI3K-Akt signalling by insulin resulted in nuclear FoxO1 extrusion and HGF upregulation. Fetuin-A counteracted these insulin effects. CONCLUSIONS/INTERPRETATION: Fetuin-A and/or palmitate influence the expression of proinflammatory and angiogenic proteins only partially via TLR4 signalling. HGF downregulation seems to be mediated by interference with the insulin-dependent receptor tyrosine kinase pathway. Fetuin-A may also influence angiogenic and proinflammatory proteins involved in atherosclerosis.


Subject(s)
Adipose Tissue/cytology , Angiogenic Proteins/metabolism , Blood Vessels/cytology , Inflammation , alpha-2-HS-Glycoprotein/physiology , Adipose Tissue/metabolism , Atherosclerosis/metabolism , Blood Vessels/metabolism , Cell Proliferation , Coculture Techniques , Glycoproteins/metabolism , Hepatocyte Growth Factor/metabolism , Humans , Lipopolysaccharides/chemistry , Neovascularization, Pathologic , Palmitates/chemistry , Signal Transduction , Toll-Like Receptor 4/metabolism
13.
Exp Cell Res ; 321(2): 123-32, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24332981

ABSTRACT

This study was performed to identify the potential role of Alpha-2 Heremans Schmid Glycoprotein (AHSG) in Head and Neck Squamous Cell Carcinoma (HNSCC) tumorigenesis using an HNSCC cell line model. HNSCC cell lines are unique among cancer cell lines, in that they produce endogenous AHSG and do not rely, solely, on AHSG derived from serum. To produce our model, we performed a stable transfection to down-regulate AHSG in the HNSCC cell line SQ20B, resulting in three SQ20B sublines, AH50 with 50% AHSG production, AH20 with 20% AHSG production and EV which is the empty vector control expressing wild-type levels of AHSG. Utilizing these sublines, we examined the effect of AHSG depletion on cellular adhesion, proliferation, migration and invasion in a serum-free environment. We demonstrated that sublines EV and AH50 adhered to plastic and laminin significantly faster than the AH20 cell line, supporting the previously reported role of exogenous AHSG in cell adhesion. As for proliferative potential, EV had the greatest amount of proliferation with AH50 proliferation significantly diminished. AH20 cells did not proliferate at all. Depletion of AHSG also diminished cellular migration and invasion. TGF-ß was examined to determine whether levels of the TGF-ß binding AHSG influenced the effect of TGF-ß on cell signaling and proliferation. Whereas higher levels of AHSG blunted TGF-ß influenced SMAD and ERK signaling, it did not clearly affect proliferation, suggesting that AHSG influences on adhesion, proliferation, invasion and migration are primarily due to its role in adhesion and cell spreading. The previously reported role of AHSG in potentiating metastasis via protecting MMP-9 from autolysis was also supported in this cell line based model system of endogenous AHSG production in HNSCC. Together, these data show that endogenously produced AHSG in an HNSCC cell line, promotes in vitro cellular properties identified as having a role in tumorigenesis.


Subject(s)
Carcinoma, Squamous Cell/genetics , Head and Neck Neoplasms/genetics , alpha-2-HS-Glycoprotein/physiology , Binding, Competitive/drug effects , Binding, Competitive/genetics , Carcinogenesis/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Matrix Metalloproteinase 9/metabolism , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Squamous Cell Carcinoma of Head and Neck , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/metabolism , alpha-2-HS-Glycoprotein/antagonists & inhibitors
15.
Nat Rev Endocrinol ; 9(3): 144-52, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23337953

ABSTRACT

The liver is known to be involved in the natural history of the ongoing epidemics of type 2 diabetes mellitus and cardiovascular disease. In particular, the liver has a role in increased glucose production and dysregulated lipoprotein metabolism, conditions that are often found in patients with nonalcoholic fatty liver disease. Additionally, several proteins that are exclusively or predominantly secreted from the liver are now known to directly affect glucose and lipid metabolism. In analogy to the functional proteins released from adipose tissue and skeletal muscle-adipokines and myokines-these liver-derived proteins are known as hepatokines. The first hepatokine that has been proven to have a major pathogenetic role in metabolic diseases is α2-HS-glycoprotein (fetuin-A). Production of this glycoprotein is increased in steatotic and inflamed liver, but not in expanded and dysregulated adipose tissue. Thus, research into this molecule and other hepatokines is expected to aid in differentiating between the contribution of liver and those of skeletal muscle and adipose tissue, to the pathogenesis of type 2 diabetes mellitus and cardiovascular disease.


Subject(s)
Adipokines/physiology , Liver/metabolism , Muscle Proteins/physiology , Muscle, Skeletal/metabolism , Adipose Tissue/metabolism , Cardiovascular Diseases/etiology , Diabetes Mellitus, Type 2/etiology , Fatty Liver/metabolism , Female , Glucose/metabolism , Humans , Lipid Metabolism/physiology , Male , Non-alcoholic Fatty Liver Disease , alpha-2-HS-Glycoprotein/physiology
16.
Dev Neurobiol ; 73(5): 354-69, 2013 May.
Article in English | MEDLINE | ID: mdl-23109215

ABSTRACT

The serum protein fetuin-A is essential for mineral homeostasis and shows immunomodulatory functions, for example by binding to TGF superfamily proteins. It proved neuroprotective in a rat stroke model and reduced lethality after systemic lipopolysaccharide challenge in mice. Serum fetuin-A concentrations are highest during intrauterine life. Different species show intrauterine cerebral fetuin-A immunoreactivity, suggesting a contribution to brain development. We therefore aimed at specifying fetuin-A immunoreactivity in brains of newborn rats (age P0-P28) and human neonates (20-40 weeks of gestation). In humans and rats, fetuin-A was found in cortex, white matter, subplate, hippocampus, subventricular zone, and ependymal cells which supports a global role for brain function. In rats, overall fetuin-A immunoreactivity decreased with age. At P0 fetuin-A immunoreactivity affected most brain structures. Thereafter, it became increasingly restricted to distinct cells of the hippocampus, cingular gyrus, periventricular stem cell layer, and ependyma. In ependymal cells the staining pattern complied with active transependymal transport from cerebrospinal fluid. Double immunofluorescence studies revealed colocalization with NeuN (mature neurons), beta III tubulin (immature neurons), GFAP (astrocytes), and CD68 (activated microglia). This points to a role of fetuin-A in different brain functional systems. In human neonatal autopsy cases, frequently affected from severe neurological and non-neurological diseases, fetuin-A immunoreactivity was heterogeneous and much less associated with age than in healthy tissues studied earlier, suggesting an impact of exogeneous noxious factors on fetuin-A regulation. Further research on the role of fetuin-A in the neonatal brain during physiological and pathological conditions is recommended.


Subject(s)
Brain Chemistry , Brain/embryology , Nerve Tissue Proteins/analysis , alpha-2-HS-Glycoprotein/analysis , Animals , Animals, Newborn , Brain/growth & development , Female , Gestational Age , Humans , Immunoenzyme Techniques , Male , Microscopy, Fluorescence , Nerve Tissue Proteins/physiology , Organ Specificity , Rats , Rats, Wistar , alpha-2-HS-Glycoprotein/physiology
17.
FEBS Lett ; 586(19): 3458-63, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22980907

ABSTRACT

Our goal in this study was to define the mechanisms by which fetuin-A mediates the adhesion of tumor cells. The data show that in the absence of fetuin-A, detached tumor cells secrete exosomes that contain most of the known exosomal associated proteins but lack the capacity to mediate cellular adhesion. In the presence of fetuin-A, the cells secrete exosomes, which contain, in addition to the other exosomal proteins, fetuin-A, plasminogen and histones. These exosomes mediate adhesion and cell spreading. Plasminogen is a participant in this novel adhesion mechanism. The data suggest that these exosomes play a role in tumor progression.


Subject(s)
Exosomes/physiology , Neoplasms/pathology , Neoplasms/physiopathology , alpha-2-HS-Glycoprotein/physiology , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/physiology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/physiopathology , Female , Humans , Models, Biological , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Neoplasm Proteins/physiology , Plasminogen/physiology
19.
Nat Med ; 18(8): 1279-85, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22842477

ABSTRACT

Toll-like receptor 4 (TLR4) has a key role in innate immunity by activating an inflammatory signaling pathway. Free fatty acids (FFAs) stimulate adipose tissue inflammation through the TLR4 pathway, resulting in insulin resistance. However, current evidence suggests that FFAs do not directly bind to TLR4, but an endogenous ligand for TLR4 remains to be identified. Here we show that fetuin-A (FetA) could be this endogenous ligand and that it has a crucial role in regulating insulin sensitivity via Tlr4 signaling in mice. FetA (officially known as Ahsg) knockdown in mice with insulin resistance caused by a high-fat diet (HFD) resulted in downregulation of Tlr4-mediated inflammatory signaling in adipose tissue, whereas selective administration of FetA induced inflammatory signaling and insulin resistance. FFA-induced proinflammatory cytokine expression in adipocytes occurred only in the presence of both FetA and Tlr4; removing either of them prevented FFA-induced insulin resistance. We further found that FetA, through its terminal galactoside moiety, directly binds the residues of Leu100-Gly123 and Thr493-Thr516 in Tlr4. FFAs did not produce insulin resistance in adipocytes with mutated Tlr4 or galactoside-cleaved FetA. Taken together, our results suggest that FetA fulfills the requirement of an endogenous ligand for TLR4 through which lipids induce insulin resistance. This may position FetA as a new therapeutic target for managing insulin resistance and type 2 diabetes.


Subject(s)
Fatty Acids, Nonesterified/pharmacology , Insulin Resistance/physiology , Toll-Like Receptor 4/physiology , alpha-2-HS-Glycoprotein/physiology , 3T3-L1 Cells , Adipocytes/chemistry , Adipocytes/drug effects , Adipocytes/physiology , Animals , Base Sequence , Binding Sites , Blood Glucose/analysis , Chronic Disease , Cytokines/biosynthesis , Cytokines/genetics , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/physiopathology , Dietary Fats/toxicity , Galactosides/metabolism , Gene Expression Regulation/drug effects , Humans , Inflammation/chemically induced , Inflammation/physiopathology , Ligands , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Obese , Molecular Sequence Data , NF-kappa B/metabolism , Obesity/blood , Obesity/physiopathology , Receptors, Leptin/deficiency , Signal Transduction/drug effects , Signal Transduction/physiology , Toll-Like Receptor 4/analysis , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/chemistry , alpha-2-HS-Glycoprotein/antagonists & inhibitors , alpha-2-HS-Glycoprotein/chemistry , alpha-2-HS-Glycoprotein/toxicity
20.
Endocrine ; 42(3): 496-505, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22820893

ABSTRACT

Although type 2 diabetes mellitus (DM) is a chronic metabolic disorder with multiple etiologies, obesity has been constantly linked with insulin resistance and manifestation of type 2 DM. In addition, obesity is associated with hypertension, dyslipidemia, and fatty liver disease and is regarded as a subclinical inflammatory condition characterized by release of pro-inflammatory mediators such as cytokines from adipose tissue. Both, type 2 DM and obesity are considered as major risks for developing micro- and macrovascular diseases. Recent studies showed that impaired circulating levels of fetuin-A, which is involved in propagating insulin resistance as well as circulating levels of angiopoietins, which are growth factors promoting angiogenesis, were observed in patients with obesity, metabolic syndrome, and type 2 DM. However, independent of type 2 DM and obesity, defective regulation of fetuin-A and angiopoietin are playing a critical role in predisposing to coronary and peripheral vascular diseases. Therefore, mechanisms linking type 2 DM and obesity with fetuin-A and angiopoietins seem to be complex and are in need of further exploration. In this review, we aimed to present a summary concerning associations of type 2 diabetes, obesity, and vascular diseases with circulating levels of angiopoietins and fetuin-A. Furthermore, we aimed to focus on roles of fetuin-A and angiopoietins and to highlight the most plausible mechanisms that might explain their associations with type 2 DM and obesity.


Subject(s)
Angiopoietins/physiology , Diabetes Mellitus, Type 2/metabolism , alpha-2-HS-Glycoprotein/physiology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Angiopoietins/metabolism , Diabetes Mellitus, Type 2/pathology , Humans , Neovascularization, Physiologic/physiology , Obesity/metabolism , Obesity/pathology , alpha-2-HS-Glycoprotein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...