Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Metab ; 36(7): 1534-1549.e7, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38878772

ABSTRACT

Tirzepatide, a glucose-dependent insulinotropic polypeptide/glucagon-like peptide 1 receptor (GIPR/GLP-1R) agonist, has, in clinical trials, demonstrated greater reductions in glucose, body weight, and triglyceride levels compared with selective GLP-1R agonists in people with type 2 diabetes (T2D). However, cellular mechanisms by which GIPR agonism may contribute to these improved efficacy outcomes have not been fully defined. Using human adipocyte and mouse models, we investigated how long-acting GIPR agonists regulate fasted and fed adipocyte functions. In functional assays, GIPR agonism enhanced insulin signaling, augmented glucose uptake, and increased the conversion of glucose to glycerol in a cooperative manner with insulin; however, in the absence of insulin, GIPR agonists increased lipolysis. In diet-induced obese mice treated with a long-acting GIPR agonist, circulating triglyceride levels were reduced during oral lipid challenge, and lipoprotein-derived fatty acid uptake into adipose tissue was increased. Our findings support a model for long-acting GIPR agonists to modulate both fasted and fed adipose tissue function differentially by cooperating with insulin to augment glucose and lipid clearance in the fed state while enhancing lipid release when insulin levels are reduced in the fasted state.


Subject(s)
Adipocytes , Mice, Inbred C57BL , Receptors, Gastrointestinal Hormone , Animals , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Gastrointestinal Hormone/agonists , Adipocytes/metabolism , Adipocytes/drug effects , Humans , Mice , Male , Insulin/metabolism , Glucose/metabolism , Lipolysis/drug effects , Triglycerides/metabolism , Gastric Inhibitory Polypeptide/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Obesity/metabolism , Obesity/drug therapy , Nutrients/metabolism , Signal Transduction/drug effects , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-2 Receptor
3.
STAR Protoc ; 4(2): 102304, 2023 May 12.
Article in English | MEDLINE | ID: mdl-37178114

ABSTRACT

Human subcutaneous adipocytes are an attractive therapeutic target in regulating overall physiological homeostasis. However, the differentiation of primary human adipose-derived models remains challenging. Here, we present a protocol to differentiate primary subcutaneous adipose-derived preadipocytes from human subcutaneous adipocytes and to measure lipolytic activity. We describe steps for seeding of subcutaneous preadipocytes, removal of growth factors, induction and maturation of adipocytes, removal of serum/phenol red in media, and treatment of mature adipocytes. We then detail glycerol measurement in conditioned media and its interpolation. For complete details on the use and execution of this protocol, please refer to Coskun et al.1.

4.
Nat Commun ; 12(1): 6486, 2021 11 10.
Article in English | MEDLINE | ID: mdl-34759311

ABSTRACT

The hepatokine follistatin is elevated in patients with type 2 diabetes (T2D) and promotes hyperglycemia in mice. Here we explore the relationship of plasma follistatin levels with incident T2D and mechanisms involved. Adjusted hazard ratio (HR) per standard deviation (SD) increase in follistatin levels for T2D is 1.24 (CI: 1.04-1.47, p < 0.05) during 19-year follow-up (n = 4060, Sweden); and 1.31 (CI: 1.09-1.58, p < 0.01) during 4-year follow-up (n = 883, Finland). High circulating follistatin associates with adipose tissue insulin resistance and non-alcoholic fatty liver disease (n = 210, Germany). In human adipocytes, follistatin dose-dependently increases free fatty acid release. In genome-wide association study (GWAS), variation in the glucokinase regulatory protein gene (GCKR) associates with plasma follistatin levels (n = 4239, Sweden; n = 885, UK, Italy and Sweden) and GCKR regulates follistatin secretion in hepatocytes in vitro. Our findings suggest that GCKR regulates follistatin secretion and that elevated circulating follistatin associates with an increased risk of T2D by inducing adipose tissue insulin resistance.


Subject(s)
Diabetes Mellitus, Type 2/blood , Follistatin/blood , Adaptor Proteins, Signal Transducing/blood , Adipose Tissue/metabolism , Genome-Wide Association Study , Hepatocytes/metabolism , Humans , Insulin Resistance/physiology , Middle Aged , Non-alcoholic Fatty Liver Disease/blood
5.
J Lipid Res ; 61(8): 1203-1220, 2020 08.
Article in English | MEDLINE | ID: mdl-32487544

ABSTRACT

Angiopoietin-like protein (ANGPTL)8 has been implicated in metabolic syndrome and reported to regulate adipose FA uptake through unknown mechanisms. Here, we studied how complex formation of ANGPTL8 with ANGPTL3 or ANGPTL4 varies with feeding to regulate LPL. In human serum, ANGPTL3/8 and ANGPTL4/8 complexes both increased postprandially, correlated negatively with HDL, and correlated positively with all other metabolic syndrome markers. ANGPTL3/8 also correlated positively with LDL-C and blocked LPL-facilitated hepatocyte VLDL-C uptake. LPL-inhibitory activity of ANGPTL3/8 was >100-fold more potent than that of ANGPTL3, and LPL-inhibitory activity of ANGPTL4/8 was >100-fold less potent than that of ANGPTL4. Quantitative analyses of inhibitory activities and competition experiments among the complexes suggested a model in which localized ANGPTL4/8 blocks the LPL-inhibitory activity of both circulating ANGPTL3/8 and localized ANGPTL4, allowing lipid sequestration into fat rather than muscle during the fed state. Supporting this model, insulin increased ANGPTL3/8 secretion from hepatocytes and ANGPTL4/8 secretion from adipocytes. These results suggest that low ANGPTL8 levels during fasting enable ANGPTL4-mediated LPL inhibition in fat tissue to minimize adipose FA uptake. During feeding, increased ANGPTL8 increases ANGPTL3 inhibition of LPL in muscle via circulating ANGPTL3/8, while decreasing ANGPTL4 inhibition of LPL in adipose tissue through localized ANGPTL4/8, thereby increasing FA uptake into adipose tissue. Excessive caloric intake may shift this system toward the latter conditions, possibly predisposing to metabolic syndrome.


Subject(s)
Angiopoietin-Like Protein 4/metabolism , Angiopoietin-like Proteins/metabolism , Fatty Acids/metabolism , Peptide Hormones/metabolism , Postprandial Period , Angiopoietin-Like Protein 3 , Angiopoietin-Like Protein 8 , Biomarkers/metabolism , Humans
6.
Basic Res Cardiol ; 114(3): 25, 2019 04 19.
Article in English | MEDLINE | ID: mdl-31004234

ABSTRACT

The goal of the present study was to evaluate the effects of SGLT2i on cardiac contractile function, substrate utilization, and efficiency before and during regional myocardial ischemia/reperfusion injury in normal, metabolically healthy swine. Lean swine received placebo or canagliflozin (300 mg PO) 24 h prior to and the morning of an invasive physiologic study protocol. Hemodynamic and cardiac function measurements were obtained at baseline, during a 30-min complete occlusion of the circumflex coronary artery, and during a 2-h reperfusion period. Blood pressure, heart rate, coronary flow, and myocardial oxygen consumption were unaffected by canagliflozin treatment. Ventricular volumes remained unchanged in controls throughout the protocol. At the onset of ischemia, canagliflozin produced acute large increases in left ventricular end-diastolic and systolic volumes which returned to baseline with reperfusion. Canagliflozin-mediated increases in end-diastolic volume were directly associated with increases in stroke volume and stroke work relative to controls during ischemia. Canagliflozin also increased cardiac work efficiency during ischemia relative to control swine. No differences in myocardial uptake of glucose, lactate, free fatty acids or ketones, were noted between treatment groups at any time. In separate experiments using a longer 60 min coronary occlusion followed by 2 h of reperfusion, canagliflozin increased end-diastolic volume and stroke volume and significantly diminished myocardial infarct size relative to control swine. These data demonstrate that SGLT2i with canagliflozin preserves cardiac contractile function and efficiency during regional myocardial ischemia and provides ischemia protection independent of alterations in myocardial substrate utilization.


Subject(s)
Canagliflozin/pharmacology , Energy Metabolism/drug effects , Myocardial Contraction/drug effects , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/drug therapy , Myocardium/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Ventricular Function, Left/drug effects , Animals , Disease Models, Animal , Male , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Sus scrofa
7.
Bone ; 36(2): 284-91, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15780954

ABSTRACT

Suramin is a naphthalene trisulfonic acid derivative that inhibits osteoclast differentiation and bone resorption in vitro and in vivo; however, the mechanisms underlying this activity have not been studied. Receptor activator of NF-kB (RANK) ligand (RANKL) is a key regulator of osteoclast differentiation and function and this study evaluated the ability of suramin, which has been shown to disrupt protein-protein interactions, to interfere with RANKL functional activity and binding to RANK. Suramin inhibited osteoclastic bone resorption in a calvarial model and inhibited osteoclast differentiation in RANKL-stimulated murine spleen cells and RAW264.7 cells. RANKL-induced second messenger signaling (AKT and p38 MAP Kinase phosphorylation) was completely blocked by 100 microM suramin. The ability of RANKL to bind to recombinant human RANK-Fc (rhRANK-Fc) was reduced 50% by suramin in an in vitro binding assay. Surface plasmon resonance technology and nuclear magnetic resonance (NMR) were used to evaluate the ability of suramin to bind to rhRANK-Fc. Suramin was found to selectively interact with immobilized rhRANK-Fc chimera in a concentration-dependent manner by Biacore 3000 analysis. Similar results were obtained using saturation transfer difference NMR spectroscopy to demonstrate that suramin binds to rhRANK-Fc, but not IgG1Fc or sRANKL. In summary, these findings demonstrate that suramin inhibits sRANKL-induced osteoclast differentiation and suggest that these effects are mediated by suramin binding to RANK and blocking the ability of sRANKL to induce second messenger signaling.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Carrier Proteins/physiology , Cell Differentiation/physiology , Glycoproteins/metabolism , Growth Inhibitors/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/physiology , Osteoclasts/cytology , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Suramin/metabolism , Suramin/pharmacology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Growth Inhibitors/pharmacology , Mice , Mice, Inbred ICR , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoprotegerin , Protein Binding/drug effects , Protein Binding/physiology , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B
SELECTION OF CITATIONS
SEARCH DETAIL
...