Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Rapid Commun Mass Spectrom ; 26(2): 101-8, 2012 Jan 30.
Article in English | MEDLINE | ID: mdl-22173797

ABSTRACT

Apolipoprotein B100 (apoB100) and apolipoprotein A1 (apoA1) are the primary protein components of low density lipoprotein (LDL) and high density lipoprotein (HDL) particles, respectively, and plasma levels of these proteins are associated with risks of cardiovascular disease. Existing apoB100 quantitation methods for animal models have been limited to affinity capture techniques such as enzyme-linked immunosorbent assay (ELISA) and Western blot which require specialized reagents for each species and in many cases are not readily available. Here we demonstrate a single translatable ultra-performance liquid chromatography/tandem mass spectrometry (UPLC/MS/MS) assay that is fast and robust and can be used to measure apolipoprotein concentrations in plasma for six species. When possible, peptide sequences that are conserved across species were identified for this assay. The sample preparation is limited and can be carried out in 96-well microtiter plates and thus allows for multiplexed preparation of samples for analysis of large numbers of samples in a short time frame when combined with UPLC/MS/MS. Separation and quantitation of the tryptic peptides is carried out at 700 µL/min using a 1.7 µm core shell C18 column (2.1 × 50 mm). The chromatography is designed for the analysis of over 100 samples per day, and the UPLC run is less than 10 min. This assay is capable of supporting cardiovascular research by providing a single assay to measure critical biomarkers across multiple species without the need for antibodies, and does so in a high-throughput manner.


Subject(s)
Apolipoprotein B-100/blood , Apolipoprotein B-48/blood , Chromatography, High Pressure Liquid/methods , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Animals , Apolipoprotein A-I/blood , Apolipoprotein B-100/genetics , Apolipoprotein B-48/genetics , Cardiovascular Diseases/blood , Computer Simulation , Cricetinae , Dogs , Gene Knockdown Techniques , Humans , Linear Models , Macaca mulatta , Mice , Peptide Fragments/analysis , RNA, Small Interfering/genetics , Rats , Species Specificity , Trypsin/chemistry , Trypsin/metabolism
2.
Lipids ; 46(11): 991-1003, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21826528

ABSTRACT

The association between hypercholesterolemia and elevated serum apolipoprotein B (APOB) has generated interest in APOB as a therapeutic target for patients at risk of developing cardiovascular disease. In the clinic, mipomersen, an antisense oligonucleotide (ASO) APOB inhibitor, was associated with a trend toward increased hepatic triglycerides, and liver steatosis remains a concern. We found that siRNA-mediated knockdown of ApoB led to elevated hepatic triglycerides and liver steatosis in mice engineered to exhibit a human-like lipid profile. Many genes required for fatty acid synthesis were reduced, suggesting that the observed elevation in hepatic triglycerides is maintained by the cell through fatty acid uptake as opposed to fatty acid synthesis. Fatty acid transport protein 5 (Fatp5/Slc27a5) is required for long chain fatty acid (LCFA) uptake and bile acid reconjugation by the liver. Fatp5 knockout mice exhibited lower levels of hepatic triglycerides due to decreased fatty acid uptake, and shRNA-mediated knockdown of Fatp5 protected mice from diet-induced liver steatosis. Here, we evaluated if siRNA-mediated knockdown of Fatp5 was sufficient to alleviate ApoB knockdown-induced steatosis. We determined that, although Fatp5 siRNA treatment was sufficient to increase the proportion of unconjugated bile acids 100-fold, consistent with FATP5's role in bile acid reconjugation, Fatp5 knockdown failed to influence the degree, zonal distribution, or composition of the hepatic triglycerides that accumulated following ApoB siRNA treatment.


Subject(s)
Apolipoproteins B/genetics , Fatty Acid Transport Proteins/genetics , Fatty Liver/genetics , Animals , Apolipoproteins B/blood , Bile Acids and Salts/metabolism , Cholesterol/blood , Cholesterol, HDL/blood , Female , Gene Expression Profiling , Gene Knockdown Techniques , Lipid Metabolism/genetics , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , RNA, Small Interfering , Triglycerides/blood
3.
J Cardiovasc Transl Res ; 4(6): 801-10, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21822774

ABSTRACT

Lecithin:cholesterol acyltransferase (LCAT) is the key circulating enzyme responsible for high-density lipoprotein (HDL) cholesterol esterification, HDL maturation, and potentially reverse cholesterol transport. To further explore LCAT's mechanism of action on lipoprotein metabolism, we employed adeno-associated viral vector (AAV) serotype 8 to achieve long-term (32-week) high level expression of human LCAT in hCETP;Ldlr(+/-) mice, and characterized the lipid profiles in detail. The mice had a marked increase in HDL cholesterol, HDL particle size, and significant reduction in low-density lipoprotein (LDL) cholesterol, plasma triglycerides, and plasma apoB. Plasma LCAT activity significantly increased with humanized substrate specificity. HDL cholesteryl esters increased in a fashion that fits human LCAT specificity. HDL phosphatidylcholines trended toward decrease, with no change observed for HDL lysophosphatidylcholines. Triglycerides reduction appeared to reside in all lipoprotein particles (very low-density lipoprotein (VLDL), LDL, and HDL), with HDL triglycerides composition highly reflective of VLDL, suggesting that changes in HDL triglycerides were primarily driven by the altered triglycerides metabolism in VLDL. In summary, in this human-like model for lipoprotein metabolism, AAV8-mediated overexpression of human LCAT resulted in profound changes in plasma lipid profiles. Detailed lipid analyses in the lipoprotein particles suggest that LCAT's beneficial effect on lipid metabolism includes not only enhanced HDL cholesterol esterification but also improved metabolism of apoB-containing particles and triglycerides. Our findings thus shed new light on LCAT's mechanism of action and lend support to its therapeutic potential in treating dyslipidemia.


Subject(s)
Cholesterol Ester Transfer Proteins/metabolism , Dependovirus/genetics , Dyslipidemias/therapy , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Lipids/blood , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Receptors, LDL/deficiency , Animals , Cholesterol Ester Transfer Proteins/genetics , Disease Models, Animal , Dyslipidemias/enzymology , Dyslipidemias/genetics , Humans , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Particle Size , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Receptors, LDL/genetics , Time Factors
4.
J Cardiovasc Transl Res ; 4(3): 373-83, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21394531

ABSTRACT

Emerging evidence suggests apolipoprotein B (apoB) and apolipoprotein AI (apoAI) are strong risk predictors for atherosclerosis. Non-human primates (NHP), including rhesus monkeys, cynomolgus monkeys, and African green monkeys, are important preclinical species for studying dyslipidemia and atherosclerosis as they more closely resemble humans in lipid metabolism and disease physiology compared to lower species such as rodents. However, no commercial assays are currently available for measuring apoB and apoAI in NHP. We therefore evaluated analytical methods for routinely measuring apoB and apoAI in our NHP dyslipidemia and atherosclerosis research. Since NHP apoB and apoAI sequences are likely highly similar to human, we focused on the clinically validated and widely utilized human apoB and apoAI immunoturbidity assays. We carried out technical validation of these assays with NHP samples and leveraged orthogonal technical platforms including mass spectrometry, independent ELISA assay, and absolute quantitation via SDS-PAGE for further characterization. Analysis of purified lipoproteins demonstrated that the immunoturbidity assays detect NHP apoAI and apoB, with good dilution linearity and spike recovery from NHP plasma. Orthogonal studies showed apoAI correlated with protein concentration and apoB levels correlated with LC/MS and an independent ELISA. NHP samples from a drug treatment study were analyzed with the immunoturbidity assays and levels of apoB and apoAI fit our understanding of biology and expectations from literature. These studies serve as important technical and biological validation of the immunoturbidity assays for NHP samples, and demonstrate that these assays provide a high-throughput, fully automated analytical platform for NHP samples. Our studies pave the way for future translational research in NHP for developing therapies for treating dyslipidemia and atherosclerosis.


Subject(s)
Apolipoprotein A-I/blood , Apolipoproteins B/blood , Atherosclerosis/blood , Dyslipidemias/blood , Immunoassay , Nephelometry and Turbidimetry , Animals , Atherosclerosis/diagnosis , Atherosclerosis/drug therapy , Biomarkers/blood , Calibration , Disease Models, Animal , Dyslipidemias/diagnosis , Dyslipidemias/drug therapy , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Haplorhini , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Immunoassay/standards , Mass Spectrometry , Nephelometry and Turbidimetry/standards , Predictive Value of Tests , Reproducibility of Results , Simvastatin/pharmacology
5.
J Lipid Res ; 52(4): 679-87, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21262787

ABSTRACT

Reducing circulating LDL-cholesterol (LDL-c) reduces the risk of cardiovascular disease in people with hypercholesterolemia. Current approaches to reduce circulating LDL-c include statins, which inhibit cholesterol synthesis, and ezetimibe, which blocks cholesterol absorption. Both elevate serum PCSK9 protein levels in patients, which could attenuate their efficacy by reducing the amount of cholesterol cleared from circulation. To determine whether PCSK9 inhibition could enhance LDL-c lowering of both statins and ezetimibe, we utilized small interfering RNAs (siRNAs) to knock down Pcsk9, together with ezetimibe, rosuvastatin, and an ezetimibe/rosuvastatin combination in a mouse model with a human-like lipid profile. We found that ezetimibe, rosuvastatin, and ezetimibe/rosuvastatin combined lower serum cholesterol but induce the expression of Pcsk9 as well as the Srebp-2 hepatic cholesterol biosynthesis pathway. Pcsk9 knockdown in combination with either treatment led to greater reductions in serum non-HDL with a near-uniform reduction of all LDL-c subfractions. In addition to reducing serum cholesterol, the combined rosuvastatin/ezetimibe/Pcsk9 siRNA treatment exhibited a significant reduction in serum APOB protein and triglyceride levels. Taken together, these data provide evidence that PCSK9 inhibitors, in combination with current therapies, have the potential to achieve greater reductions in both serum cholesterol and triglycerides.


Subject(s)
Anticholesteremic Agents/therapeutic use , Azetidines/therapeutic use , Fluorobenzenes/therapeutic use , Pyrimidines/therapeutic use , Serine Endopeptidases/metabolism , Sulfonamides/therapeutic use , Animals , Apolipoproteins B/blood , Cholesterol/blood , Cholesterol, LDL/blood , Enzyme-Linked Immunosorbent Assay , Ezetimibe , Hypercholesterolemia/blood , Hypercholesterolemia/drug therapy , Hypercholesterolemia/therapy , Mice , Mice, Inbred C57BL , Proprotein Convertase 9 , Proprotein Convertases , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology , Reverse Transcriptase Polymerase Chain Reaction , Rosuvastatin Calcium , Serine Endopeptidases/genetics , Triglycerides/blood
6.
Obesity (Silver Spring) ; 16(6): 1178-85, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18388900

ABSTRACT

OBJECTIVE: Our main objective was to compare the regulation of cortisol production within omental (Om) and abdominal subcutaneous (Abd sc) human adipose tissue. METHODS AND PROCEDURES: Om and Abd sc adipose tissue were obtained at surgery from subjects with a wide range of BMI. Hydroxysteroid dehydrogenase (HSD) activity ((3)H-cortisone and (3)H-cortisol interconversion) and expression were measured before and after organ culture with insulin and/or dexamethasone. RESULTS: Type 1 HSD (HSD1) mRNA and reductase activity were mainly expressed within adipocytes and tightly correlated with adipocyte size within both depots. There was no depot difference in HSD1 expression or reductase activity, while cortisol inactivation and HSD2 mRNA expression (expressed in stromal cells) were higher in Om suggesting higher cortisol turnover in this depot. Culture with insulin decreased HSD reductase activity in both depots. Culture with dexamethasone plus insulin compared to insulin alone increased HSD reductase activity only in the Om depot. This depot-specific increase in reductase activity could not be explained by an alteration in HSD1 mRNA or protein, which was paradoxically decreased. However, in Om only, hexose-6-phosphate dehydrogenase (H6PDH) mRNA levels were increased by culture with dexamethasone plus insulin compared to insulin alone, suggesting that higher nicotinamide adenine dinucleotide phosphate-oxidase (NADPH) production within the endoplasmic reticulum (ER) contributed to the higher HSD reductase activity. DISCUSSION: We conclude that in the presence of insulin, glucocorticoids cause a depot-specific increase in the activation of cortisone within Om adipose tissue, and that this mechanism may contribute to adipocyte hypertrophy and visceral obesity.


Subject(s)
Cortisone/metabolism , Hydrocortisone/metabolism , Omentum/metabolism , Subcutaneous Fat/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Adult , Dexamethasone/pharmacology , Female , Glucocorticoids/pharmacology , Humans , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Insulin/pharmacology , Male , Middle Aged , Omentum/cytology , Oxidoreductases/metabolism , RNA, Messenger/metabolism , Subcutaneous Fat/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...