Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Atherosclerosis ; 243(1): 328-33, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26433113

ABSTRACT

BACKGROUND AND AIMS: There is a paucity of data concerning the influence of lipid-lowering therapy on cardiovascular (CV) outcomes in patients with homozygous familial hypercholesterolaemia (FH). To redress this a retrospective analysis was undertaken of the demographic features, lipid levels, low density lipoprotein receptor and Autosomal Recessive Hypercholesterolaemia gene mutations, CV outcomes and vital status of 44 FH homozygotes referred to a single centre in the UK between 1964 and 2014. METHODS: Data were obtained from past publications, case records and death certificates. Differences in categorical and continuous variables between living and dead patients were analysed using Fisher's exact test and an independent t-test respectively. RESULTS: During the 50 years covered by this survey 13 patients have died, 30 are still alive and 1 was lost to follow up. The mean age of Alive patients was 32.6 ± 11.5 versus 28.3 ± 14.9 years in Dead ones (P = 0.31) and they were born 18 years later (P = 0.0001). Pre-treatment serum total cholesterol (TC) was similar in Alive and Dead (20.2 ± 5.1 v 21.3 ± 4.4 mmol/l, P = 0.52) but on-treatment TC was lower in Alive than Dead (8.1 ± 2.8 v 14.5 ± 6.0 mmol/l, P = 0.0001) and CV adverse events were far less frequent (eg aortic stenosis, 33% v 77%, P = 0.02). CONCLUSIONS: The lower on-treatment TC and fewer CV adverse events in FH homozygotes still living reflect advances in apheresis and drug therapy since the 1990s. Further improvements in prognosis can be expected with the impending introduction of novel lipid-lowering agents.


Subject(s)
Cardiovascular Diseases/therapy , Hyperlipoproteinemia Type II/complications , Hyperlipoproteinemia Type II/genetics , Adolescent , Adult , Anticholesteremic Agents/therapeutic use , Aortic Valve Stenosis/drug therapy , Blood Component Removal , Cardiovascular Diseases/complications , Cardiovascular Diseases/genetics , Cholesterol, LDL/blood , Cohort Studies , Female , Homozygote , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hyperlipoproteinemia Type II/therapy , Male , Middle Aged , Models, Statistical , Mutation , Receptors, LDL/blood , Receptors, LDL/genetics , Retrospective Studies , United Kingdom , Young Adult
2.
J Lipid Res ; 54(12): 3491-505, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24103848

ABSTRACT

The purpose of this study was to determine the core biological processes perturbed in the subcutaneous adipose tissue of familial combined hyperlipidemia (FCHL) patients. Annotation of FCHL and control microarray datasets revealed a distinctive FCHL transcriptome, characterized by gene expression changes regulating five overlapping systems: the cytoskeleton, cell adhesion and extracellular matrix; vesicular trafficking; lipid homeostasis; and cell cycle and apoptosis. Expression values for the cell-cycle inhibitor CDKN2B were increased, replicating data from an independent FCHL cohort. In 3T3-L1 cells, CDKN2B knockdown induced C/EBPα expression and lipid accumulation. The minor allele at SNP site rs1063192 (C) was predicted to create a perfect seed for the human miRNA-323b-5p. A miR-323b-5p mimic significantly reduced endogenous CDKN2B protein levels and the activity of a CDKN2B 3'UTR luciferase reporter carrying the rs1063192 C allele. Although the allele displayed suggestive evidence of association with reduced CDKN2B mRNA in the MuTHER adipose tissue dataset, family studies suggest the association between increased CDKN2B expression and FCHL-lipid abnormalities is driven by factors external to this gene locus. In conclusion, from a comparative annotation analysis of two separate FCHL adipose tissue transcriptomes and a subsequent focus on CDKN2B, we propose that dysfunctional adipogenesis forms an integral part of FCHL pathogenesis.


Subject(s)
Adipose Tissue/metabolism , Cyclin-Dependent Kinase Inhibitor p15/genetics , Gene Expression Regulation , Hyperlipidemia, Familial Combined/genetics , 3T3-L1 Cells , Adipogenesis/genetics , Adipose Tissue/pathology , Animals , Cell Cycle/genetics , HEK293 Cells , Haplotypes , Humans , Hyperlipidemia, Familial Combined/pathology , Male , Mice , Middle Aged
3.
Genome Res ; 22(6): 1144-53, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22399572

ABSTRACT

Genomic inversions are an increasingly recognized source of genetic variation. However, a lack of reliable high-throughput genotyping assays for these structures has precluded a full understanding of an inversion's phylogenetic, phenotypic, and population genetic properties. We characterize these properties for one of the largest polymorphic inversions in man (the ∼4.5-Mb 8p23.1 inversion), a structure that encompasses numerous signals of natural selection and disease association. We developed and validated a flexible bioinformatics tool that utilizes SNP data to enable accurate, high-throughput genotyping of the 8p23.1 inversion. This tool was applied retrospectively to diverse genome-wide data sets, revealing significant population stratification that largely follows a clinal "serial founder effect" distribution model. Phylogenetic analyses establish the inversion's ancestral origin within the Homo lineage, indicating that 8p23.1 inversion has occurred independently in the Pan lineage. The human inversion breakpoint was localized to an inverted pair of human endogenous retrovirus elements within the large, flanking low-copy repeats; experimental validation of this breakpoint confirmed these elements as the likely intermediary substrates that sponsored inversion formation. In five data sets, mRNA levels of disease-associated genes were robustly associated with inversion genotype. Moreover, a haplotype associated with systemic lupus erythematosus was restricted to the derived inversion state. We conclude that the 8p23.1 inversion is an evolutionarily dynamic structure that can now be accommodated into the understanding of human genetic and phenotypic diversity.


Subject(s)
Chromosome Inversion , Chromosomes, Human, Pair 8 , Polymorphism, Genetic , Animals , Base Sequence , Evolution, Molecular , Gene Expression , Genetics, Population , Haplotypes/genetics , High-Throughput Nucleotide Sequencing , Humans , Lupus Erythematosus, Systemic/genetics , Molecular Sequence Data , Pan troglodytes/genetics , Phylogeny , Polymorphism, Single Nucleotide , RNA, Messenger , Segmental Duplications, Genomic
4.
J Vasc Interv Radiol ; 19(10): 1403-8, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18693045

ABSTRACT

PURPOSE: To compare the aortic plaque burden in patients with heterozygous familial hypercholesterolemia on long-term statin treatment with that of matched control subjects. MATERIALS AND METHODS: The authors studied 11 heterozygous, nonsmoking, nondiabetic, and nonhypertensive patients with familial hypercholesterolemia (mean age, 44 years +/- 10) who had been receiving cholesterol-lowering management for a mean of 12 years +/- 5, including 8.25 years +/- 4.24 with the highest tolerable doses of a statin (or a statin plus ezetimibe), and 26 age- and sex-matched control subjects with 3T magnetic resonance (MR) imaging of the descending thoracic aorta by using an axial T2-weighted turbo spin-echo sequence. RESULTS: Quantitative analysis demonstrated that the aortic vessel wall area was significantly larger in patients with familial hypercholesterolemia than in control subjects (123 mm(2) +/- 23 vs 102 mm(2) +/- 18, respectively; P < .007), as was vessel wall thickness (1.63 mm +/- 0.28 vs 1.37 mm +/- 0.16, respectively; P < .001). No significant difference was found between mean values of routine serum lipid and lipoprotein parameters. CONCLUSIONS: The results of this preliminary study show that patients with heterozygous familial hypercholesterolemia have a higher aortic atherosclerotic plaque burden than control subjects at quantitative MR imaging despite long-term lipid-lowering therapy. This information may help design future studies evaluating plaque burden and cardiovascular risk.


Subject(s)
Aorta, Thoracic/pathology , Aortic Valve Stenosis/diagnosis , Aortic Valve Stenosis/prevention & control , Hyperlipoproteinemia Type II/diagnosis , Hyperlipoproteinemia Type II/drug therapy , Hypolipidemic Agents/administration & dosage , Magnetic Resonance Imaging/methods , Adult , Aged , Aorta, Thoracic/drug effects , Female , Humans , Male , Middle Aged , Pilot Projects , Treatment Outcome
5.
Radiology ; 247(2): 550-7, 2008 May.
Article in English | MEDLINE | ID: mdl-18349314

ABSTRACT

Research ethics committee approval was obtained for this study, and written informed consent was obtained from all participants. The purpose was to prospectively evaluate the feasibility of breath-hold multiecho in- and out-of-phase magnetic resonance (MR) imaging for simultaneous lipid quantification and T2* measurement. A spoiled gradient-echo sequence with seven echo times alternately in phase and out of phase was used at 3.0 T. Imaging was performed in a lipid phantom, in five healthy volunteers (all men; mean age, 37 years), and in five obese individuals with hyperlipidemia or diabetes (four men, one woman; mean age, 53 years). A biexponential curve-fitting model was used to derive the relative signal contributions from fat and water, and these results were compared with results of liver proton MR spectroscopy, the reference standard. There was a significant correlation between multiecho and spectroscopic measurements of hepatic lipid concentration (r(2) = 0.99, P < .001). In vivo, the T2* of water was consistently longer than that of fat and reliably enabled the signal components to be correctly assigned. In the lipid phantom, the multiecho method could be used to determine the fat-to-water ratio and the T2* values of fat and water throughout the entire range of fat concentrations. Multiecho imaging shows promise as a method of simultaneous fat and T2* quantification.


Subject(s)
Fatty Liver/diagnosis , Magnetic Resonance Imaging/methods , Adult , Aged , Feasibility Studies , Female , Humans , Image Interpretation, Computer-Assisted , Least-Squares Analysis , Male , Middle Aged , Phantoms, Imaging , Prospective Studies , Sensitivity and Specificity
6.
Eur Radiol ; 18(4): 800-5, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18034347

ABSTRACT

This study was designed to assess whether breath-hold cardiac multiecho imaging at 3.0 T is achievable without significant image artefacts and if fat/water phase interference modulates the exponential T2* signal decay. Twelve healthy volunteers (mean age 39) were imaged on a Philips Intera 3.0 T MRI scanner. Multiecho imaging was performed with a breath-hold spoiled gradient echo sequence with a seven echo readout (echo times 1.15-8.05 ms, repetition time 11 ms) using a black-blood prepulse and volume shimming. T2* values were calculated with both mono- and biexpoential fits from the mean signal intensity of the interventricular septum. The global mean T2* was 27.3 ms +/- 6.4. The mean signal-to-noise ratio (SNR) of the septum was 22.8 +/- 9.9, and the contrast-to-noise ratio (CNR) of the septum to the left ventricular cavity 20.3 +/- 9.4. A better fit was obtained with a biexponential model and the mean fat fraction derived was 3.7%. Cardiac functional parameters were in the normal range and showed no correlation with T2*. Cardiac T2* estimation with gradient multiecho imaging at 3.0 T can be achieved with minimal artefact and modelling the signal decay with a biexponential function allows estimation of myocardial lipid content as well as T2* decay.


Subject(s)
Heart/anatomy & histology , Magnetic Resonance Imaging/methods , Adipose Tissue/anatomy & histology , Adult , Artifacts , Female , Humans , Iron Overload/diagnosis , Male , Middle Aged , Pilot Projects
7.
J Am Coll Cardiol ; 50(21): 2051-8, 2007 Nov 20.
Article in English | MEDLINE | ID: mdl-18021872

ABSTRACT

OBJECTIVES: This study's aim was to examine whether treatment with pioglitazone, added to conventional lipid-lowering therapy, would improve myocardial glucose utilization (MGU) and blood flow (MBF) in nondiabetic patients with familial combined hyperlipidemia (FCHL). BACKGROUND: Thiazolidinediones were found to improve insulin sensitivity and MGU in type 2 diabetes and MBF in Mexican Americans with insulin resistance. Familial combined hyperlipidemia is a complex genetic disorder conferring a high risk of premature coronary artery disease, characterized by high serum cholesterol and/or triglyceride, low high-density lipoprotein (HDL) cholesterol, and insulin resistance. METHODS: We undertook a randomized, double-blind, placebo-controlled study in 26 patients with FCHL, treated with pioglitazone or matching placebo 30 mg daily for 4 weeks, followed by 45 mg daily for 12 weeks. Positron emission tomography was used to measure MBF at rest and during adenosine-induced hyperemia and MGU during euglycemic hyperinsulinemic clamp at baseline and after treatment. RESULTS: Whereas no change was observed in the placebo group after treatment, patients receiving pioglitazone showed a significant increase in whole body glucose disposal (3.93 +/- 1.59 mg/kg/min to 5.24 +/- 1.65 mg/kg/min; p = 0.004) and MGU (0.62 +/- 0.26 micromol/g/min to 0.81 +/- 0.14 micromol/g/min; p = 0.0007), accompanied by a significant improvement in resting MBF (1.11 +/- 0.20 ml/min/g to 1.25 +/- 0.21 ml/min/g; p = 0.008). Furthermore, in the pioglitazone group HDL cholesterol (+28%; p = 0.003) and adiponectin (+156.2%; p = 0.0001) were increased and plasma insulin (-35%; p = 0.017) was reduced. CONCLUSIONS: In patients with FCHL treated with conventional lipid-lowering therapy, the addition of pioglitazone led to significant improvements in MGU and MBF, with a favorable effect on blood lipid and metabolic parameters. (A study to investigate the effect of pioglitazone on whole body and myocardial glucose uptake and myocardial blood flow/coronary vasodilator reserve in patients with familial combined hyperlipidaemia; http://www.controlled-trials.com/mrct/trial/230761/ISRCTN78563659; ISRCTN78563659).


Subject(s)
Coronary Circulation/drug effects , Hyperlipidemia, Familial Combined/drug therapy , Hypoglycemic Agents/pharmacology , Thiazolidinediones/pharmacology , Blood Glucose/analysis , Double-Blind Method , Drug Therapy, Combination , Female , Glucose/analysis , Glucose/metabolism , Heart/drug effects , Humans , Hypoglycemic Agents/therapeutic use , Hypolipidemic Agents/therapeutic use , Male , Middle Aged , Pioglitazone , Positron-Emission Tomography , Thiazolidinediones/therapeutic use , Treatment Outcome
8.
Neuroradiology ; 49(11): 927-31, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17643240

ABSTRACT

INTRODUCTION: Homozygous familial hypercholesterolemia (FH) is considered a model disease for excessive plasma cholesterol levels. Patients with untreated homozygous FH have a markedly increased risk for premature atherosclerosis. The frequency and extent of ischemic brain damage detectable by high-field magnetic resonance imaging (MRI) after long-term intensive treatment are unknown. METHODS: In a case control study, five patients with homozygous FH (one male and four females; mean age: 23.6 +/- 9.2, range: 12-36 years; mean pre-treatment serum total cholesterol level: 26.9 +/- 3.24 mmol/L; all patients with documented atherosclerotic plaques in the carotid arteries) and five age- and sex-matched healthy controls were studied. All patients had been on maximal lipid-lowering medication since early childhood, and four of them were also on treatment with low-density lipoprotein (LDL) apheresis at bi-weekly intervals. Brain MRI was performed at 3 Tesla field strength with fluid-attenuated T2-weighted inversion recovery and T1-weighted spin-echo MR pulse sequences and subsequently evaluated by two independent readers. RESULTS: The maximal lipid-lowering treatment reduced the total serum cholesterol by more than 50% in the patients, but their serum concentrations were still 3.6-fold higher than those found in the controls (11.9 +/- 4.2 vs. 4.5 +/- 0.5 mmol/L; p < 0.0047). No brain abnormality was observed in any of the patients with homozygous FH. CONCLUSION: Homozygous FH patients on intensive cholesterol-lowering therapy have no evidence of ischemic brain damage at 3 Tesla MRI despite the remaining high cholesterol levels.


Subject(s)
Brain/pathology , Carotid Artery Diseases/complications , Hyperlipoproteinemia Type II/pathology , Hyperlipoproteinemia Type II/therapy , Adolescent , Adult , Anticholesteremic Agents/therapeutic use , Blood Component Removal , Brain Ischemia/etiology , Brain Ischemia/pathology , Brain Ischemia/prevention & control , Carotid Artery Diseases/pathology , Carotid Artery Diseases/therapy , Case-Control Studies , Child , Cohort Studies , Female , Humans , Hyperlipoproteinemia Type II/complications , Magnetic Resonance Imaging , Male
9.
Atherosclerosis ; 195(1): e181-90, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17482623

ABSTRACT

Familial combined hyperlipidaemia (FCHL) is a complex genetic disorder conferring high risk of premature atherosclerosis, characterized by high cholesterol and/or triglyceride, low high density lipoprotein (HDL) cholesterol and insulin resistance. We examined whether pioglitazone, added to conventional lipid-lowering therapy, would favourably affect metabolic parameters and alter body fat content. We undertook a randomized, double blind, placebo-controlled study in 22 male patients with FCHL treated with pioglitazone or matching placebo 30 mg daily for 4 weeks, increasing to 45 mg for 12 weeks. Magnetic resonance imaging and proton magnetic resonance spectroscopy were performed to measure adipose tissue (AT) body content as well as intrahepatocellular lipids (IHCL) and intramyocellular lipids (IMCL) at baseline and after treatment. Significantly improved in the pioglitazone group were: triglyceride/HDL (atherogenic index of plasma) -32.3% (p=0.002), plasma glucose -4.4% (p=0.03), alanine-aminotransferase (ALT) -7.7% (p=0.005) and adiponectin 130.1% (p=0.001). Pioglitazone treatment resulted in a significant increase in total (5.3%, p=0.02) and subcutaneous (7.1%, p=0.003) adipose tissue as well as in soleus-IMCL levels (47.4%, p=0.02) without alteration in intra-abdominal AT or IHCL. Changes in ALT and AST and IHCL were strongly correlated (r=0.72, p<0.01; r=.0.86, p<0.01, respectively). In patients with FCHL on conventional lipid-lowering therapy, the addition of pioglitazone acts favourably on several metabolic parameters.


Subject(s)
Adipose Tissue/metabolism , Hyperlipidemias/drug therapy , Lipids/chemistry , Liver/metabolism , Muscles/metabolism , Thiazolidinediones/pharmacology , Adult , Double-Blind Method , Humans , Hypoglycemic Agents/pharmacology , Magnetic Resonance Imaging/methods , Male , Middle Aged , Pioglitazone , Placebos
10.
Ultrasound Med Biol ; 33(7): 1029-38, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17448589

ABSTRACT

Our aim was to assess the predictive value of a measurement of intima-medial layer (IML) reflectivity in the differentiation of pathological from physiological increases in intima-medial thickness (IMT). Both common carotid arteries (CCA) of familial hypercholesterolemia (FH) patients and age- and sex-matched controls (no cardiovascular risk factors) were imaged using a 10- to 15-MHz linear array transducer (n = 30). Images of the CCA far wall were analyzed in the IMT "plug-in" of "HDI Lab." The IML reflectivity, averaged over an 8- to 12-mm length of arterial wall, was expressed as a ratio of reflectivity at a point 0.21-mm deep to the intima-medial interface divided by the reflectivity at the intima-medial interface, termed the intima-medial reflectivity index (IMRI). The risk of atherosclerosis was assessed in terms of IMT alone and IMT coupled with IMRI. Defining high risk of atherosclerosis in FH, in terms of both IMT alone and IMT coupled with IMRI, produced an appropriate, when compared with cholesterol-years score, statistically significant stratification (p < 0.01 and p < 0.005). Analysis of the low-risk subjects revealed a tendency to define a subject as "high risk" based on a physiological increase in IMT, but when IMRI is included in the assessment, all controls are correctly identified as low risk. This method of quantifying the reflectivity of the IML improved the discriminatory performance of IMT increase as an indicator of atherosclerotic risk by enabling a smaller, therefore earlier, increase in IMT to be considered pathologic when accompanied by an increase in IMRI.


Subject(s)
Atherosclerosis/diagnostic imaging , Carotid Artery, Common/diagnostic imaging , Hyperlipoproteinemia Type II/diagnostic imaging , Tunica Intima/diagnostic imaging , Adult , Aged , Cholesterol/blood , Endothelium, Vascular/diagnostic imaging , Female , Humans , Image Processing, Computer-Assisted/methods , Male , Middle Aged , Reproducibility of Results , Risk Factors , Sensitivity and Specificity , Ultrasonography
11.
Nat Clin Pract Cardiovasc Med ; 4(4): 214-25, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17380167

ABSTRACT

Familial hypercholesterolemia (FH) is characterized by raised serum LDL cholesterol levels, which result in excess deposition of cholesterol in tissues, leading to accelerated atherosclerosis and increased risk of premature coronary heart disease. FH results from defects in the hepatic uptake and degradation of LDL via the LDL-receptor pathway, commonly caused by a loss-of-function mutation in the LDL-receptor gene (LDLR) or by a mutation in the gene encoding apolipoprotein B (APOB). FH is primarily an autosomal dominant disorder with a gene-dosage effect. An autosomal recessive form of FH caused by loss-of-function mutations in LDLRAP1, which encodes a protein required for clathrin-mediated internalization of the LDL receptor by liver cells, has also been documented. The most recent addition to the database of genes in which defects cause FH is one encoding a member of the proprotein convertase family, PCSK9. Rare dominant gain-of-function mutations in PCSK9 cosegregate with hypercholesterolemia, and one mutation is associated with a particularly severe FH phenotype. Expression of PCSK9 normally downregulates the LDL-receptor pathway by indirectly causing degradation of LDL-receptor protein, and loss-of-function mutations in PCSK9 result in low plasma LDL levels. Thus, PCSK9 is an attractive target for new drugs aimed at lowering serum LDL cholesterol, which should have additive lipid-lowering effects to the statins currently used.


Subject(s)
Apolipoprotein B-100/genetics , Genetic Predisposition to Disease , Hyperlipoproteinemia Type II/genetics , Lipoproteins, LDL/blood , Receptors, LDL/genetics , Adult , Age Factors , Aged , Female , Gene Expression Regulation , Humans , Hyperlipoproteinemia Type II/epidemiology , Hyperlipoproteinemia Type II/physiopathology , Incidence , Lipoproteins, LDL/analysis , Male , Middle Aged , Mutation, Missense , Prognosis , Risk Assessment , Severity of Illness Index , Sex Factors
12.
Atherosclerosis ; 194(1): 102-11, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17094996

ABSTRACT

Familial hypercholesterolaemia (FH) results from defective catabolism of low density lipoproteins (LDL), leading to premature atherosclerosis and early coronary heart disease. It is commonly caused by mutations in LDLR, encoding the LDL receptor that mediates hepatic uptake of LDL, or in APOB, encoding its major ligand. More rarely, dominant mutations in PCSK9 or recessive mutations in LDLRAP1 (ARH) cause FH, gene defects that also affect the LDL-receptor pathway. We have used multiplex ligation-dependent probe amplification (MLPA) to identify deletions and rearrangements in LDLR, some not detectable by Southern blotting, thus completing our screening for mutations causing FH in a group of FH patients referred to a Lipid Clinic in London. To summarise, mutations in LDLR were found in 153 unrelated heterozygous FH patients and 24 homozygotes/compound heterozygotes, and in over 200 relatives of 80 index patients. LDLR mutations included 85 different point mutations (7 not previously described) and 13 different large rearrangements. The APOB R3500Q mutation was present in 14 heterozygous patients and a mutation in PCSK9 in another 4; LDLRAP1 mutations were found in 4 "homozygous" FH patients. Our data confirm that DNA-based diagnosis provides information that is important for management of FH in a considerable number of families.


Subject(s)
Gene Deletion , Gene Duplication , Hyperlipoproteinemia Type II/genetics , Receptors, LDL/genetics , Apolipoproteins B/genetics , Exons/genetics , Gene Rearrangement , Genetic Testing/methods , Heterozygote , Homozygote , Humans , Nucleic Acid Amplification Techniques , Point Mutation , Proprotein Convertase 9 , Proprotein Convertases , Serine Endopeptidases/genetics
13.
Nat Clin Pract Cardiovasc Med ; 2(5): 252-60, 2005 May.
Article in English | MEDLINE | ID: mdl-16265509

ABSTRACT

Insulin-resistance syndromes are of pandemic proportions; 150 million people worldwide and an estimated 43 million people in the US are currently affected by type 2 diabetes mellitus or metabolic syndrome respectively. Treatment of heart disease in the context of type 2 diabetes requires multifactorial risk-factor management, including lifestyle modification and drug treatment for comorbidities. Management of coronary risk extends beyond simple cholesterol lowering. Early use of cardiac imaging and, where appropriate, revascularization should be considered in high-risk or symptomatic patients. Traditionally, patients with type 2 diabetes and coronary arterial disease have been treated surgically, but percutaneous revascularization of these patients is increasingly common. Indeed, revascularization by use of drug-eluting coronary stents combined with administration of novel antiplatelet agents has revolutionized percutaneous coronary intervention in patients with type 2 diabetes. Despite these advances, there is no consensus of opinion regarding revascularization strategies or risk-factor management in insulin-resistant patients with symptomatic or prognostically important coronary arterial disease. Furthermore, specific therapies and preventative strategies for diabetic cardiomyopathy and heart failure in patients with type 2 diabetes remain elusive. The identification of optimized approaches for the prevention and treatment of the metabolic syndrome and heart disease in insulin-resistant, nondiabetic patients remains a major global challenge.


Subject(s)
Coronary Artery Disease/prevention & control , Diabetes Mellitus, Type 2/complications , Heart Failure/therapy , Insulin Resistance/physiology , Cardiomyopathies/etiology , Cardiomyopathies/therapy , Coronary Artery Disease/complications , Coronary Artery Disease/therapy , Heart Failure/etiology , Humans , Metabolic Syndrome/complications , Risk Factors
14.
Arterioscler Thromb Vasc Biol ; 25(12): 2654-60, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16224054

ABSTRACT

OBJECTIVE: Analysis of long-term (30 years) clinical history and response to treatment of 13 patients with the D374Y mutation of PCSK9 (PCSK9 patients) from 4 unrelated white British families compared with 36 white British patients with heterozygous familial hypercholesterolemia attributable to 3 specific mutations in the low-density lipoprotein (LDL) receptor gene (LDLR) known to cause severe phenotype. METHODS AND RESULTS: The PCSK9 patients, when compared with the LDLR patients, were younger at presentation (20.8+/-14.7 versus 30.2+/-15.7 years; P=0.003), had higher pretreatment serum cholesterol levels (13.6+/-2.9 versus 9.6+/-1.6 mmol/L; P=0.004) that remained higher during treatment with simvastatin (10.1+/-3.0 versus 6.5+/-0.9 mmol/L; P=0.006), atorvastatin (9.6+/-2.9 versus 6.4+/-1.0 mmol/L; P=0.006), or current lipid-lowering therapy, including LDL apheresis and partial ileal bypass in 2 PCSK9 patients (7.0+/-1.6 versus 5.4+/-1.0 mmol/L; P=0.001), and were affected >10 years earlier by premature coronary artery disease (35.2+/-4.8 versus 46.8+/-8.9 years; P=0.002). LDL from PCSK9 patients competed significantly less well for binding to fibroblast LDL receptors than LDL from either controls or LDLR patients. CONCLUSIONS: These British PCSK9 patients with the D374Y mutation have an unpredictably severe clinical phenotype, which may be a unique feature for this cohort, and requires early and aggressive lipid-lowering management to prevent cardiovascular complications.


Subject(s)
Hypercholesterolemia/genetics , Hypercholesterolemia/therapy , Point Mutation , Serine Endopeptidases/genetics , Severity of Illness Index , Adolescent , Adult , Atherosclerosis/genetics , Atherosclerosis/therapy , Child , Cholesterol, LDL/chemistry , Cholesterol, LDL/metabolism , Family Health , Female , Fibroblasts/metabolism , Follow-Up Studies , Humans , Male , Middle Aged , Particle Size , Proprotein Convertase 9 , Proprotein Convertases , Receptors, LDL/genetics , Receptors, LDL/metabolism , Retrospective Studies , Skin/cytology , United Kingdom , White People
15.
Blood ; 106(2): 542-9, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15790791

ABSTRACT

Scott syndrome (SS) is a bleeding disorder characterized by a failure to expose phosphatidylserine (PS) to the outer leaflet of the platelet plasma membrane. Because the adenosine triphosphate (ATP)-binding cassette transporter A1 (ABCA1) is implicated in the exofacial translocation of PS, we assessed its role in the pathophysiology of a patient with SS. Substantially reduced levels of ABCA1 mRNA were found in the patient's leukocytes, compared with controls. The SS patient was heterozygous for a novel missense mutation c.6064G>A (ABCA1 R1925Q), absent from unaffected family members and controls. Both mutant and wild-type alleles were reduced in mRNA expression, and no causative mutation for this phenomenon was identified in the ABCA1 gene or its proximal promoter, suggesting a putative second mutation in a trans-acting regulatory gene may also be involved in the disorder in this patient. In vitro expression studies showed impaired trafficking of ABCA1 R1925Q to the plasma membrane. Overexpression of wild-type ABCA1 in SS lymphocytes complemented the Ca2+-dependent PS exposure at the cell surface. These data identify a mutation in ABCA1 that contributes to the defective PS translocation phenotype in our patient with SS.


Subject(s)
ATP-Binding Cassette Transporters/blood , ATP-Binding Cassette Transporters/genetics , Blood Coagulation Disorders, Inherited/blood , Blood Coagulation Disorders, Inherited/genetics , Mutation, Missense , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Biological Transport, Active/drug effects , Blood Coagulation Disorders, Inherited/metabolism , Calcium/pharmacology , Cell Line , DNA/genetics , Female , Gene Expression , Genetic Complementation Test , Humans , In Vitro Techniques , Leukocytes/drug effects , Leukocytes/metabolism , Male , Middle Aged , Molecular Sequence Data , Pedigree , Phosphatidylserines/blood , Phosphatidylserines/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid
16.
Hum Mol Genet ; 14(9): 1161-9, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15772090

ABSTRACT

Typically, autosomal dominant familial hypercholesterolaemia (FH) is caused by mutations in the low density lipoprotein (LDL) receptor or apolipoprotein B genes that result in defective clearance of plasma LDL by the liver, but a third gene (PCSK9), encoding a putative proprotein convertase, has recently been implicated. Two independent microarray studies support a role for PCSK9 in sterol metabolism and adenoviral-mediated over-expression of PCSK9 in mouse liver depletes hepatic LDL-receptor protein, but the mechanism by which dominant mutations cause human FH is unclear. We have identified the D374Y mutant of PCSK9 in three FH families of English origin; all 12 affected individuals have unusually severe hypercholesterolaemia and require more stringent treatment than typical FH patients, who are heterozygous for defects in the LDL receptor. We have stably expressed wild-type (WT) and variant PCSK9 in McArdle-7777 rat hepatoma cells and shown by confocal microscopy that all forms of PCSK9 co-localize with protein disulphide isomerase in the ER whether or not they can be autocleaved. Expression of the proposed pathogenic variants, but not of WT, S386A or F216L PCSK9, increases secretion of apolipoprotein B100-containing lipoproteins from the cells by 2-4-fold probably by reducing the degradation of nascent protein; no differences in LDL-receptor content were observed in cells expressing WT, S386A or F216L PCSK9 and only a small reduction in cells expressing the D374Y or S127R mutants. This suggests that the variants of PCSK9 found in FH influence the secretion of apoB-containing lipoproteins, providing an explanation for the marked increase in circulating LDL in heterozygous carriers.


Subject(s)
Apolipoproteins B/metabolism , Genes, Dominant , Hypercholesterolemia/etiology , Mutation , Serine Endopeptidases/genetics , Adult , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Transformed , Cell Line, Tumor , Cell Transformation, Viral , Endoplasmic Reticulum/metabolism , Female , Genetic Variation , Heterozygote , Humans , Hypercholesterolemia/genetics , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , Male , Microscopy, Confocal , Pedigree , Proprotein Convertase 9 , Proprotein Convertases , Protein Disulfide-Isomerases/metabolism , Rats , Serine Endopeptidases/metabolism , Severity of Illness Index , Transfection
17.
Trends Mol Med ; 10(8): 362-5, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15310455

ABSTRACT

The upstream stimulatory factor (USF) proteins are ubiquitously expressed and, as such, represent unusual candidates for involvement in disorders of carbohydrate and lipid metabolism. Nonetheless, a recent study has reported an association between specific alleles of USF1 and familial combined hyperlipidaemia, a common disorder that substantially increases the risk of premature atherosclerotic cardiovascular disease. USF1 might, therefore, also contribute to the metabolic syndrome. The use of chromatin immunoprecipitation methodologies combined with promoter microarray assays will help to define the transcriptional networks that underlie whole-body glucose and lipid homeostasis.


Subject(s)
DNA-Binding Proteins/physiology , Hyperlipidemia, Familial Combined/etiology , Metabolic Syndrome/etiology , Transcription Factors/physiology , Female , Humans , Hyperlipidemia, Familial Combined/metabolism , Leucine Zippers , Lipid Metabolism , Male , Metabolic Syndrome/metabolism , Pedigree , Upstream Stimulatory Factors
18.
Curr Opin Lipidol ; 15(4): 413-22, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15243214

ABSTRACT

PURPOSE OF REVIEW: This review focuses on recent advances in the management of patients with homozygous familial hypercholesterolaemia, autosomal recessive hypercholesterolaemia and familial defective apolipoprotein B. RECENT FINDINGS: Autosomal recessive hypercholesterolaemia has been described as a 'phenocopy' of homozygous familial hypercholesterolaemia. Although the clinical phenotypes are similar, autosomal recessive hypercholesterolaemia seems to be less severe, more variable within a single family, and more responsive to lipid-lowering drug therapy. The cardiovascular complications of premature atherosclerosis are delayed in some individuals and involvement of the aortic root and valve is less common than in homozygous familial hypercholesterolaemia. Apheresis is still the treatment of choice in homozygous familial hypercholesterolaemia and in autosomal recessive hypercholesterolaemia patients in whom maximal drug therapy does not achieve adequate control. In addition to the profound cholesterol-lowering effects of apheresis, other potentially beneficial phenomena have been documented: improved vascular endothelial function and haemorheology, reduction in lipoprotein (a) and procoagulatory status, and a decrease in adhesion molecules and C-reactive protein. SUMMARY: Patients with severe homozygous hypercholesterolaemia illustrate the natural history of atherosclerosis within a condensed timeframe. Effective cholesterol-lowering treatment started in early childhood is essential to prevent onset of life-threatening atherosclerotic involvement of the aortic root and valve, and the coronary arteries. Noninvasive methods for regular monitoring of the major sites involved in the atherosclerotic process are necessary in patients with no symptoms or signs of ischaemia. Management of patients with severe homozygous hypercholesterolaemia continues to be a major challenge.


Subject(s)
Hypercholesterolemia/genetics , Hypercholesterolemia/therapy , Adolescent , Adult , Apolipoproteins B/genetics , Azetidines/therapeutic use , Blood Component Removal , Child , Cholesterol/metabolism , Coronary Disease/therapy , Ezetimibe , Female , Genes, Recessive , Genetic Therapy , Homozygote , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Lipid Metabolism , Liver Transplantation , Male , Models, Genetic , Mutation , Pregnancy , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...