Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Osteoarthritis Cartilage ; 26(12): 1609-1618, 2018 12.
Article in English | MEDLINE | ID: mdl-30240937

ABSTRACT

OBJECTIVE: This study tested whether galcanezumab, a humanized monoclonal antibody with efficacy against migraine, was superior to placebo for the treatment of mild or moderate osteoarthritis (OA) knee pain. METHOD: In a multicenter, double-blind, placebo- and celecoxib-controlled trial, patients with moderate to severe OA pain were randomized to placebo; celecoxib 200 mg daily for 16 weeks; or galcanezumab 5, 50, 120, and 300 mg subcutaneously every 4 weeks, twice. The primary outcome was change from baseline at Week 8 in Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) pain subscore measured by 100 mm visual analog scale (VAS). The trial was considered positive if ≥1 dose of galcanezumab demonstrated ≥95% Bayesian posterior probability of superiority to placebo and ≥50% posterior probability of superiority to placebo by ≥9 mm. A planned interim analysis allowed termination of the study if posterior probability of superiority to placebo by ≥9 mm was ≤5%. Secondary endpoints included WOMAC function subscore and Patient Global Assessment (PGA) of OA. Safety and tolerability were also assessed. RESULTS: The study was terminated after interim analysis suggested inadequate efficacy. Celecoxib significantly reduced WOMAC pain subscore compared with placebo [-12.0 mm; 95% confidence interval (CI) -23 to -2 mm]. None of the galcanezumab arms demonstrated clinically meaningful improvement (range: 1.5 to -5.0 mm) or met the prespecified success criteria. No improvement in any secondary objective was observed. Galcanezumab was well tolerated by OA patients. CONCLUSIONS: This study failed to demonstrate sufficient statistical evidence that galcanezumab was efficacious for treating OA knee pain. STUDY IDENTIFICATION: NCT02192190.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Calcitonin Gene-Related Peptide/antagonists & inhibitors , Osteoarthritis, Knee/drug therapy , Adult , Aged , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Celecoxib/therapeutic use , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Humans , Injections, Subcutaneous , Male , Middle Aged , Osteoarthritis, Knee/complications , Pain/drug therapy , Pain/etiology , Pain Management/methods , Pain Measurement/methods , Severity of Illness Index , Treatment Outcome
2.
Diabetes Obes Metab ; 18(2): 159-68, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26434665

ABSTRACT

AIMS: To compare the immunogenicity profiles and the potential effects on clinical outcomes of LY2963016 insulin glargine (LY IGlar) and Lantus® insulin glargine (IGlar), products with identical primary amino acid sequences, in patients with type 1 or type 2 diabetes mellitus (T1DM or T2DM). METHODS: To assess immunogenicity, anti-insulin glargine antibodies (measured as percent binding) were compared between treatments in 52-week (open-label) and 24-week (double-blind) randomized studies in total study populations of patients with T1DM (N = 535) and T2DM (N = 756), respectively, and two subgroups of patients with T2DM: insulin-naïve patients and those reporting prestudy IGlar treatment (prior IGlar). Relationships between insulin antibody levels and clinical outcomes were assessed using analysis of covariance and partial correlations. Insulin antibody levels were assessed using Wilcoxon rank sum. Treatment comparisons for treatment-emergent antibody response (TEAR) and incidence of detectable antibodies were analysed using Fisher's exact test. RESULTS: No significant treatment differences were observed for insulin antibody levels, incidence of detectable anti-insulin glargine antibodies, or incidence of TEAR [overall and endpoint, by last-observation-carried-forward (LOCF)] in patients with T1DM or patients with T2DM, including the insulin-naïve subgroup. A statistically significant difference was noted in the overall incidence of detectable antibodies but not at endpoint (LOCF) nor in TEAR for the prior IGlar subgroup of patients with T2DM. Insulin antibody levels were low (<5%) in both treatment groups. Insulin antibody levels or developing TEAR was not associated with clinical outcomes. CONCLUSIONS: LY IGlar and IGlar have similar immunogenicity profiles; anti-insulin glargine antibody levels were low for both treatments, with no observed effect on efficacy and safety outcomes.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Drug Hypersensitivity/etiology , Hypoglycemic Agents/adverse effects , Insulin Antibodies/analysis , Insulin Glargine/analogs & derivatives , Insulin Glargine/adverse effects , Asymptomatic Diseases/epidemiology , Biosimilar Pharmaceuticals/adverse effects , Biosimilar Pharmaceuticals/therapeutic use , Cross Reactions , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/immunology , Double-Blind Method , Drug Hypersensitivity/complications , Drug Hypersensitivity/epidemiology , Drug Hypersensitivity/immunology , Humans , Hyperglycemia/prevention & control , Hypoglycemia/chemically induced , Hypoglycemia/prevention & control , Hypoglycemic Agents/therapeutic use , Immunogenetic Phenomena/drug effects , Incidence , Insulin Glargine/therapeutic use , Insulin, Regular, Human/adverse effects , Insulin, Regular, Human/analogs & derivatives , Insulin, Regular, Human/genetics , Insulin, Regular, Human/therapeutic use , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use
3.
Diabetes Obes Metab ; 17(4): 414-22, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25656305

ABSTRACT

AIM: To describe the clinical effects of single and multiple doses of a potent, selective, orally administered, small-molecule antagonist of the human glucagon receptor, LY2409021, in healthy subjects and in patients with type 2 diabetes. METHODS: LY2409021 was administered in dose-escalation studies to healthy subjects (n = 23) and patients with type 2 diabetes (n = 9) as single doses (Study 1) and daily to patients with type 2 diabetes (n = 47) for 28 days (Study 2). Safety, tolerability, pharmacokinetic (PK) and pharmacodynamic (PD) assessments were made after single doses and in patients receiving once-daily doses of LY2409021 (5, 30, 60 or 90 mg) for 28 days. RESULTS: LY2409021 was well tolerated at all dose levels in both studies. Fasting and postprandial glucose were reduced and glucagon levels increased after single and multiple dosing, with reductions in fasting serum glucose of up to ∼1.25 mmol/l on day 28. Serum aminotransferases increased in a dose-dependent manner with multiple dosing and reversed after cessation of dosing. Significant glucose-lowering was observed with LY2409021 at dose levels associated with only minor aminotransferase increases. CONCLUSION: Blockade of glucagon signalling in patients with type 2 diabetes is well tolerated and results in substantial reduction of fasting and postprandial glucose with minimal hypoglycaemia, but with reversible increases in aminotransferases. Inhibition of glucagon signalling by LY2409021 is a promising potential treatment for patients with type 2 diabetes and should be evaluated in longer clinical trials to better evaluate benefits and risks.


Subject(s)
Biphenyl Compounds/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Hyperglycemia/prevention & control , Hypoglycemia/prevention & control , Hypoglycemic Agents/therapeutic use , Molecular Targeted Therapy , Receptors, Glucagon/antagonists & inhibitors , Adult , Aged , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/blood , Biphenyl Compounds/pharmacokinetics , Cohort Studies , Cross-Over Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Female , Glucagon/agonists , Glucagon/blood , Glucagon/metabolism , Glycated Hemoglobin/analysis , Half-Life , Humans , Hypoglycemia/chemically induced , Hypoglycemia/epidemiology , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/blood , Hypoglycemic Agents/pharmacokinetics , Male , Middle Aged , Molecular Targeted Therapy/adverse effects , Risk , Single-Blind Method
4.
J Pathol ; 203(4): 961-7, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15259000

ABSTRACT

Numerous apolipoproteins associate with amyloid plaques. A minor high-density lipoprotein-associated protein, glycosylphosphatidylinositol-specific phospholipase D (GPI-PLD), has recently been described by the authors and others. Since GPI-PLD is synthesized by, and secreted from, pancreatic islet beta cells, the present study examined the hypothesis that GPI-PLD associates with islet amyloid. GPI-PLD immunoreactivity was examined in pancreatic tissues from type 2 diabetic and non-diabetic humans. GPI-PLD binding to heparan sulphate proteoglycan was determined in the absence or presence of heparan sulphate or heparin. Fibril formation from human islet amyloid polypeptide was determined in the absence or presence of GPI-PLD. In non-diabetics, GPI-PLD immunoreactivity was present and co-localized with insulin, as opposed to co-localizing with amyloid in diabetics. No immunoreactivity for apolipoprotein A-I was present in islet cells or islet amyloid. Heparan sulphate proteoglycan, which is commonly present in most amyloid, bound GPI-PLD in vitro. GPI-PLD inhibited the formation of amyloid fibrils from synthetic islet amyloid polypeptide in vitro. GPI-PLD is therefore present in islet amyloid and appears to derive from local production from islets. This localization likely derives from interaction between GPI-PLD and heparan sulphate proteoglycan. Since GPI-PLD also inhibited islet amyloid polypeptide fibril formation in vitro, it is concluded that GPI-PLD may play a role in islet amyloid formation in type 2 diabetes.


Subject(s)
Amyloid/metabolism , Diabetes Mellitus, Type 2/enzymology , Islets of Langerhans/enzymology , Phospholipase D/metabolism , Amyloid/biosynthesis , Amyloid/drug effects , Heparan Sulfate Proteoglycans/metabolism , Humans , Islet Amyloid Polypeptide , Microscopy, Confocal , Phospholipase D/pharmacology
5.
Metabolism ; 50(12): 1489-92, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11735099

ABSTRACT

Insulin resistance is associated with a compensatory islet hyperactivity to sustain adequate insulin biosynthesis and secretion to maintain near euglycemia. Both glucose and insulin are involved in regulating proteins required for insulin synthesis and secretion within the islet and islet hypertrophy. We have determined that glycosylphosphatidylinositol-specific phospholipase D (GPI-PLD) is present within the secretory granules of islet beta cells. To determine if GPI-PLD is regulated in islet beta cells, we examined the effect of glucose and insulin on GPI-PLD expression in rat islets and murine insulinoma cell lines. Glucose (16.7 mmol/L) increased cellular GPI-PLD activity and mRNA levels 2- to 7-fold in isolated rat islets and betaTC3 and betaTC6-F7 cells. Insulin (10(-7) mol/L) also increased GPI-PLD mRNA levels in rat islets and betaTC6-F7 cells 2- to 4-fold commensurate with an increase in GPI-PLD biosynthesis. To determine if islet GPI-PLD expression is increased in vivo under conditions of islet hyperactivity, we compared GPI-PLD mRNA levels in islets and liver from ob/ob mice and their lean littermates. Islet GPI-PLD mRNA was increased 5-fold while liver mRNA and serum GPI-PLD levels were reduced 30% in ob/ob mice compared with lean littermate controls. These results suggest that glucose and insulin regulate GPI-PLD mRNA levels in isolated islets and beta-cell lines. These regulators may also account for the increased expression of GPI-PLD mRNA in islets from ob/ob mice, a model of insulin resistance and islet hyperactivity.


Subject(s)
Gene Expression/drug effects , Glucose/pharmacology , Insulin/pharmacology , Phospholipase D/genetics , Animals , Insulinoma , Islets of Langerhans/drug effects , Islets of Langerhans/enzymology , Liver/enzymology , Male , Mice , Mice, Obese , Obesity/enzymology , Pancreatic Neoplasms , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured
6.
Am J Physiol Endocrinol Metab ; 281(1): E147-54, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11404232

ABSTRACT

Glycosylphosphatidylinositol-specific phospholipase D (GPI-PLD) is a high-density lipoprotein-associated protein. However, the tissue source(s) for circulating GPI-PLD and whether serum levels are regulated are unknown. Because the diabetic state alters lipoprotein metabolism, and liver and pancreatic islets are possible sources of GPI-PLD, we hypothesized that GPI-PLD levels would be altered in diabetes. GPI-PLD serum activity and liver mRNA were examined in two mouse models of type 1 diabetes, a nonobese diabetic (NOD) mouse model and low-dose streptozotocin-induced diabetes in CD-1 mice. With the onset of hyperglycemia (2- to 5-fold increase over nondiabetic levels), GPI-PLD serum activity and liver mRNA increased 2- to 4-fold in both models. Conversely, islet expression of GPI-PLD was absent as determined by immunofluorescence. Insulin may regulate GPI-PLD expression, because insulin treatment of diabetic NOD mice corrected the hyperglycemia along with reducing serum GPI-PLD activity and liver mRNA. Our data demonstrate that serum GPI-PLD levels are altered in the diabetic state and are consistent with liver as a contributor to circulating GPI-PLD.


Subject(s)
Diabetes Mellitus, Type 1/enzymology , Gene Expression Regulation, Enzymologic/genetics , Phospholipase D/biosynthesis , Animals , Chromatography, High Pressure Liquid , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/pathology , Fluorescent Antibody Technique , Liver/enzymology , Male , Mice , Mice, Inbred NOD , Nutritional Status , Pancreas/pathology , Phenotype , RNA, Messenger/biosynthesis
7.
J Lipid Res ; 42(3): 442-51, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11254757

ABSTRACT

Glycosylphosphatidylinositol-specific phospholipase D (GPI-PLD) is abundant in serum and associates with high density lipoproteins (HDL). We have characterized the distribution of GPI-PLD among lipoproteins in human plasma. Apolipoprotein (apo)-specific lipoproteins containing apoB (Lp[B]), apoA-I and A-II (Lp[A-I, A-II]), or apoA-I only (Lp[A-I]) were isolated using dextran sulfate and immunoaffinity chromatography. In six human plasma samples with HDL cholesterol ranging from 39 to 129 mg/dl, 79 +/- 14% (mean +/- SD) of the total plasma GPI-PLD activity was associated with Lp[A-I], 9 +/- 12% with Lp[A-I, A-II], and 1 +/- 1% with Lp[B]; and 11 +/- 10% was present in plasma devoid of these lipoproteins. Further characterization of the GPI-PLD-containing lipoproteins by gel-filtration chromatography and nondenaturing polyacrylamide and agarose gel electrophoresis revealed that these apoA-I-containing particles/complexes were small (8 nm) and migrated with pre-beta particles on agarose electrophoresis. Immunoprecipitation of GPI-PLD with a monoclonal antibody to GPI-PLD co-precipitated apoA-I and apoA-IV but little or no apoA-II, apoC-II, apoC-III, apoD, or apoE. In vitro, apoA-I but not apoA-IV or bovine serum albumin interacted directly with GPI-PLD, but did not stimulate GPI-PLD-mediated cleavage of a cell surface GPI-anchored protein. Thus, the majority of plasma GPI-PLD appears to be specifically associated with a small, discrete, and minor fraction of lipoproteins containing apoA-I and apoA-IV. -- Deeg, M. A., E. L. Bierman, and M. C. Cheung. GPI-specific phospholipase D associates with an apoA-I- and apoA-IV-containing complex. J. Lipid Res. 2001. 42: 442--451.


Subject(s)
Apolipoprotein A-I/blood , Apolipoproteins A/blood , Phospholipase D/blood , Adult , Antibodies, Monoclonal , Apolipoprotein A-I/pharmacology , Cholesterol, HDL/blood , Chromatography, Gel , Electrophoresis, Agar Gel , Electrophoresis, Polyacrylamide Gel , Female , Glycosylphosphatidylinositols/metabolism , Humans , Immunosorbent Techniques , Male , Middle Aged , Particle Size
9.
Arch Biochem Biophys ; 370(2): 278-84, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10510287

ABSTRACT

Limited information is known regarding the regulation, structural features, and functional domains of glycosylphosphatidylinositol-specific phospholipase D (GPI-PLD, EC 3. 1.4.50). Previous studies demonstrated that trypsin cleavage of GPI-PLD at or near Arg325 and/or Arg589 in bovine serum GPI-PLD was associated with an increase in enzymatic activity. Since the Arg325 is predicted to be in a region between the catalytic domain and predicted beta-propeller structure in the C-terminal portion of GPI-PLD (T. A. Springer, Proc. Natl. Acad. Sci. USA 94, 65-72, 1997), we hypothesized that this connecting region is important for catalytic activity. Trypsin cleavage of human serum GPI-PLD, which has an Arg325 but lacks the Arg589 present in bovine serum GPI-PLD, also increased GPI-PLD activity. Peptide-specific antibodies to residues 275-296 (anti-GPI-PLD(275)) and a monoclonal antibody, 191, with an epitope encompassing Arg325, also stimulated GPI-PLD activity. Pretreating human GPI-PLD with trypsin demonstrated that anti-GPI-PLD(275) only stimulated the activity of intact GPI-PLD. These results suggest that trypsin activation and anti-GPI-PPLD(275) may have similar effects on GPI-PLD. Consistent with this is the observation that both manipulations decreased the affinity of GPI-PLD for mixed micelle substrates. These results indicate that the midportion region of GPI-PLD is important in regulating enzymatic activity.


Subject(s)
Phospholipase D/immunology , Phospholipase D/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , Catalytic Domain , Cattle , Enzyme Activation/drug effects , Epitopes/chemistry , Humans , Immunochemistry , In Vitro Techniques , Kinetics , Mice , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/immunology , Phospholipase D/chemistry , Rabbits , Substrate Specificity , Trypsin/pharmacology
10.
Circulation ; 99(22): 2876-82, 1999 Jun 08.
Article in English | MEDLINE | ID: mdl-10359731

ABSTRACT

BACKGROUND: Glycosylphosphatidylinositol-specific phospholipase D (GPI-PLD) may play an important role in inflammation, because it can hydrolyze the GPI anchors of several inflammatory membrane proteins (eg, CD106, CD55, and CD59) and its hydrolytic products upregulate macrophage cytokine expression (eg, interleukin-1 and tumor necrosis factor-alpha). Because of its potential regulatory role in inflammatory reactions, we hypothesized that GPI-PLD might be expressed in atherosclerosis. METHODS AND RESULTS: Immunohistochemistry using human GPI-PLD-specific rabbit polyclonal antiserum was performed on a total of 83 nonatherosclerotic and atherosclerotic human coronary arteries from 23 patients. Macrophages, smooth muscle cells, apoA-I, and oxidation epitopes also were identified immunohistochemically. Cell-associated GPI-PLD was detected in 95% of atherosclerotic segments, primarily on a subset of macrophages. Extracellular GPI-PLD was present in only 30% of atherosclerotic segments and localized to regions with extracellular apoA-I. In contrast, GPI-PLD was not detected in nonatherosclerotic segments. Expression of GPI-PLD mRNA by human macrophages was confirmed in vitro by reverse transcription/polymerase chain reaction. Further studies demonstrated that GPI-PLD-positive plaque macrophages contained oxidation epitopes, suggesting a link between oxidant stress and GPI-PLD expression. This possibility was supported by studies in which exposure of a macrophage cell line to H2O2 led to a 50+/-3% increase in steady-state GPI-PLD mRNA levels. CONCLUSIONS: Collectively, these results suggest that oxidative processes may regulate GPI-PLD expression and suggest a role for GPI-PLD in inflammation and in the pathogenesis of atherosclerosis.


Subject(s)
Arteriosclerosis/enzymology , Glycosylphosphatidylinositols/metabolism , Macrophages/enzymology , Phospholipase D/metabolism , Adult , Arteries/enzymology , Cell Line , Coronary Vessels/enzymology , Epitopes , Homeostasis , Humans , Hydrogen Peroxide/pharmacology , Middle Aged , Monocytes/cytology , Oxidation-Reduction , Phospholipase D/genetics , RNA, Messenger/metabolism , Reference Values , Substrate Specificity , Tissue Distribution/physiology
12.
Mamm Genome ; 9(9): 710-4, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9716655

ABSTRACT

Glycosylphosphatidylinositol-specific phospholipase D (GPI-PLD) is an 110-kDa monomeric protein found in the circulation that is capable of degrading the GPI anchor utilized by dozens of cell-surface proteins in the presence of detergent. This protein is relatively abundant (5-10 microgram/ml in human serum), yet its sites of synthesis, gene structure, and overall function are unclear. It is our purpose to use the mouse system to determine its putative roles in lipid transport, pathogen control, and diabetes. We have isolated murine full-length cDNA for GPI-PLD from a pancreatic alpha cell library. The deduced amino acid sequence shows 74% homology to bovine and human GPI-PLD. There is a single structural gene (Gpld1) mapping to mouse Chromosome (Chr) 13, and among nine tissues, liver showed the greatest abundance of GPI-PLD mRNA. Genetic differences in serum GPI-PLD activity were seen among four mouse strains, and no correlation was seen between GPI-PLD activity and circulating levels of high density lipoproteins in these mice. This is the first report of map position and genetic regulation for Gpld1. This information will enable us to further study the expression and function of GPI-PLD in normal and pathological conditions.


Subject(s)
Phospholipase D/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Cattle , Chromosome Mapping , Cloning, Molecular , DNA, Complementary/analysis , Diet, Atherogenic , Female , Humans , Lipoproteins, HDL/blood , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Molecular Sequence Data , Pancreas/chemistry , Pancreas/cytology , Phospholipase D/blood , Protein Structure, Secondary , Sequence Alignment , Sequence Analysis, DNA
13.
Arterioscler Thromb Vasc Biol ; 17(11): 2698-706, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9409245

ABSTRACT

Interaction of HDL with cells activates protein kinase C (PKC), a process that may be important in stimulating efflux of excess cellular cholesterol. Here we report that HDL treatment of cholesterol-loaded fibroblasts increases 32P labeling of three acidic phosphoproteins. These phosphoproteins, called pp80, pp27, and pp18 based on apparent M(r) in kD, were also phosphorylated by acute treatment of cells with phorbol myristate acetate, suggesting that they are regulated in response to PKC activation. The HDL-stimulated phosphorylation of pp80 and pp18 was significant after only 30 seconds and was sustained for at least 30 and 120 minutes, respectively, while increased phosphorylation of pp27 was transient, reaching a maximum at 10 minutes. Both pp27 and pp18 were phosphorylated on serine/threonine residues, whereas pp80 was phosphorylated on serine/threonine and tyrosine residues. Immunoprecipitation studies suggested that pp80 is the myristoylated alanine-rich C kinase substrate protein, but the identities of pp27 and pp18 are unknown. HDL and trypsin-digested HDL stimulated phosphorylation of pp80 and pp27, while purified apoA-I, apoA-II, or apoE had no stimulatory effects, indicating that the active component in HDL was trypsin resistant and unlikely to be an apolipoprotein. Conversely, HDL, apoA-I, apoA-II, and apoE all stimulated pp18 phosphorylation, while trypsin-digested HDL had less effect, consistent with pp18's being responsive to HDL apolipoproteins. Treatment of cholesterol-depleted cells with apoA-I also stimulated phosphorylation of pp18, but only transiently. These results suggest that HDL interaction with cells activates diverse PKC-mediated pathways that target different phosphoproteins. Of these three phosphoproteins, only pp18 has a phosphorylation response consistent with its being involved in apolipoprotein-mediated lipid transport.


Subject(s)
Carrier Proteins , Cholesterol/metabolism , Fibroblasts/drug effects , Intracellular Signaling Peptides and Proteins , Lipoproteins, HDL/pharmacology , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Protein Processing, Post-Translational/drug effects , RNA-Binding Proteins , Receptors, Lipoprotein/drug effects , Apolipoprotein A-I/pharmacology , Apolipoprotein A-II/pharmacology , Apolipoproteins E/pharmacology , Biological Transport , Cells, Cultured , Fibroblasts/metabolism , Humans , Male , Myristoylated Alanine-Rich C Kinase Substrate , Phosphorylation/drug effects , Phosphoserine/analysis , Phosphothreonine/analysis , Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , Receptors, Lipoprotein/physiology , Signal Transduction/drug effects , Skin/cytology
14.
Arterioscler Thromb Vasc Biol ; 17(9): 1667-74, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9327761

ABSTRACT

Protein kinase C (PKC) seems to play an important role in many of HDL effects on cells, including removal of excess cholesterol. HDL removes cholesterol by at least two mechanisms. One mechanism involves desorption/diffusion of cholesterol from the plasma membrane onto the acceptor particle, whereas the second is mediated by apolipoproteins and may involve intracellular translocation of cholesterol to the plasma membrane for subsequent efflux. In this report, we examined the possibility that mitogen-activated protein (MAP) kinase is one of the downstream events from HDL activation of PKC. Using a gel kinase assay with myelin basic protein incorporated into the gel, HDL (50 micrograms protein/mL) stimulated multiple kinases of 42, 50, 52, 58, and 60 kDa. The 42-kDa protein kinase, corresponding to the unresolved MAP kinases ERK1 and ERK2 based on immunoblotting, was activated over 2-fold by HDL. HDL activated all identified kinases in a concentration- and time-dependent manner, which became maximal within 5 to 10 minutes and remained activated for at least 60 minutes. HDL activation of MAP kinase seems to be partially mediated by PKC, because down-regulation of PKC and known PKC inhibitors inhibited the HDL effect by 40 to 50%. Free apolipoproteins A-I (10 micrograms/mL) and A-II (10 micrograms/mL) had no significant effect on MAP kinase activation. Moreover, modifying HDL with trypsin or tetranitromethane, which abolishes apolipoprotein-mediated cholesterol efflux, had no effect on HDL activation of MAP kinase. These results suggest that HDL activates MAP kinase via multiple signal transduction pathways that are likely involved in an HDL effect unrelated to apolipoprotein-mediated cholesterol translocation and efflux.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Lipoproteins, HDL/pharmacology , Skin/enzymology , Apolipoproteins/physiology , Cholesterol/metabolism , Enzyme Activation/drug effects , Fibroblasts/enzymology , Fibroblasts/metabolism , Glycogen Synthase Kinase 3 , Humans , Osmolar Concentration , Protein Kinase C/physiology , Skin/cytology , Time Factors
15.
Endocrinology ; 138(2): 819-26, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9003020

ABSTRACT

Glycosylphosphatidylinositol-specific phospholipase D (GPI-PLD) is abundant in mammalian serum, but the source of the circulating enzyme is unknown. Pancreatic islets have been reported to contain and secrete GPI-PLD. In this report we examined the regulation of GPI-PLD secretion from beta TC3 cells, a mouse insulinoma cell line. In the absence of glucose, phorbol myristic acid (0.1 microM) stimulated insulin secretion by 2.5-fold and GPI-PLD secretion by 2-fold. Carbachol (5 microM), glucagon-like peptide I-(7-36) amide (0.1 microM), and isobutylmethylxanthine (0.1 mM) had no significant effect on insulin or GPI-PLD secretion in the absence of glucose. Glucose (16.7 mM) stimulated both GPI-PLD and insulin secretion from beta TC3 cells by 55% and 235%, respectively. In addition, glucose potentiated the secretagogue effect of isobutylmethylxanthine, phorbol myristic acid, and glucagon-like peptide I on both insulin and GPI-PLD secretion. By immunohistochemistry and confocal microscopy, beta TC3 cells contain both insulin and GPI-PLD, which generally colocalized intracellularly. However, GPI-PLD secretion differed from insulin secretion by a higher rate of basal release (2.8% vs. 0.23%/h), a lower magnitude of response to secretagogues, and a more prolonged period of increased secretion. These results demonstrate that beta TC3 cells secrete GPI-PLD in response to insulin secretagogues and suggest that GPI-PLD may be secreted via the regulated pathway in these cells.


Subject(s)
Insulinoma/enzymology , Islets of Langerhans/enzymology , Pancreatic Neoplasms/enzymology , Phospholipase D/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Carbachol/pharmacology , Cycloheximide/pharmacology , Glucagon , Glucagon-Like Peptide 1 , Glucagon-Like Peptides , Glucose/pharmacology , Immunohistochemistry , Insulin/analysis , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Kinetics , Mice , Peptide Fragments/pharmacology , Phospholipase D/analysis , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured
16.
Curr Opin Lipidol ; 8(1): 7-11, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9127704

ABSTRACT

A protective effect of habitual fish consumption on the development of impaired glucose tolerance and diabetes is once again suggested in a recent trial. In nondiabetic individuals with essential hypertension, a group known to be insulin resistant, fish oil was not associated with a negative impact on glycemic control, insulin secretion or peripheral insulin sensitivity, even in a subgroup who had impaired glucose tolerance. Furthermore, more recent, long term, placebo-controlled trials in type II patients have failed to demonstrate a significant impact of fish oil supplementation on glycemic control. Additional information is available regarding qualitative changes in VLDL- and LDL-lipoproteins in type II diabetes patients in response to dietary fish oil supplementation. The impact of fish oil on LDL oxidation is the focus of two recent trials. Vitamin E supplementation may mitigate much of the enhanced oxidation of LDL that is potentially seen with dietary fish oil supplementation.


Subject(s)
Diabetes Mellitus, Type 2/diet therapy , Fatty Acids, Omega-3/therapeutic use , Glucose Tolerance Test , Hyperlipidemias/diet therapy , Animals , Blood Glucose/analysis , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diet , Fish Oils/administration & dosage , Fishes , Hyperlipidemias/blood , Hyperlipidemias/complications
19.
Braz J Med Biol Res ; 27(2): 375-81, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8081252

ABSTRACT

Glycosylphosphatidylinositol-specific phospholipase D (GPI-PLD) has recently been shown to be associated with high-density lipoproteins (HDL) in bovine serum. To determine the distribution of GPI-PLD among lipoproteins and characterize the GPI-PLD-containing lipoproteins in human plasma, we used dextran sulfate and immunoaffinity chromatography to isolate apolipoprotein-specific lipoproteins. This procedure allowed fractionation of lipoprotein particles into those containing apolipoprotein B (Lp B), apolipoproteins AI and AII (Lp AI/AII), or apolipoprotein AI only (Lp AI). In five plasma samples with HDL cholesterol ranging from 40 to 129 mg/dl, 75 +/- 12% (mean +/- SD) of the GPI-PLD activity was associated with Lp AI, 11 +/- 13% with Lp AI/AII, while only 13 +/- 9% was present in plasma devoid of these lipoproteins, suggesting that most of the GPI-PLD in human plasma is associated with apolipoprotein AI. No GPI-PLD activity was detected in Lp B. Further characterization of the GPI-PLD-containing lipoproteins by gel filtration chromatography, nondenaturing poly-acrylamide and agarose gel electrophoresis revealed that GPI-PLD was restricted to an apolipoprotein AI-containing particle or complex that was small (apparent mean Mw of 140 kDa) and distinct from the bulk of HDL. Thus, the majority of plasma GPI-PLD appears to be specifically associated with a small, minor fraction of apolipoprotein AI.


Subject(s)
Apolipoprotein A-II/analysis , Apolipoprotein A-I/analysis , Apolipoproteins B/analysis , Phospholipase D/analysis , Animals , Glycosylphosphatidylinositols/metabolism , Humans , Lipoproteins, HDL/blood , Substrate Specificity
20.
Braz. j. med. biol. res ; 27(2): 375-81, Feb. 1994. ilus
Article in English | LILACS | ID: lil-140278

ABSTRACT

Glycosylphosphatidylinositol-specific phospholipase D (GPI-PLD) has recently been shown to be associated with high-density lipoproteins (HDL) in bovine serum. To determine the distribution of GPI-PLD among lipoproteins and characterize the GPI-PLD-containing lipoproteins in human plasma, we used dextram sulfate and immunoaffinity chromatography to isolate apolipoprotein-specific lipoproteins. This procedure allowed fractionation of lipoprotein particles into those containing apolipoprotein B (Lp B), apolipoproteins AI and AII (Lp AI/AII), or apolipoprotein AI only (Lp AI). In five plasma samples with HDL cholesterol ranging from 40 to 129 mg/dl, 75 ñ 12 percent (mean ñ SD) of the GPI-PLD activity was associated with LpAI, 11 ñ 13 percent with Lp AI/AII, while only 13 ñ 9 percent was present in plasma devoid of these lipoproteins, suggesting that most of the GPI-PLD in human plasma is associated with apolipoprotein AI. No GPI-PLD activity was detected in Lp B. Further characterization of the GPI-PLD-containing lipoproteins by gel filtration chromatography, nondenaturing poly-acrylamide and agarose gel electrophoresis revealed that GPI-PLD was restricted to an apolipoprotein AI-containing particle or complex that was small (apparent mean Mw of 140 KDa) and distinct from the bulk of HDL. Thus, the majority of plasma GPI-PLD appears to be specifically associated with a small, minor fraction of apoloprotein AI


Subject(s)
Humans , Cattle , Apolipoprotein A-I , Dextran Sulfate , Phosphatidylinositols/chemistry , Glycolipids/chemistry , Lipoproteins, HDL , Phospholipase D/metabolism , Plasma/enzymology , Chromatography, Affinity , Chromatography, Gel , Electrophoresis, Agar Gel/methods , Phosphatidylinositols/metabolism , Glycolipids/metabolism , Ultracentrifugation/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...