Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
JAMA ; 327(17): 1679-1687, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35368052

ABSTRACT

Importance: Lipoprotein(a) (Lp[a]) is an important risk factor for atherothrombotic cardiovascular disease and aortic stenosis, for which there are no treatments approved by regulatory authorities. Objectives: To assess adverse events and tolerability of a short interfering RNA (siRNA) designed to reduce hepatic production of apolipoprotein(a) and to assess associated changes in plasma concentrations of Lp(a) at different doses. Design, Setting, and Participants: A single ascending dose study of SLN360, an siRNA targeting apolipoprotein(a) synthesis conducted at 5 clinical research unit sites located in the US, United Kingdom, and Australia. The study enrolled adults with Lp(a) plasma concentrations of 150 nmol/L or greater at screening and no known clinically overt cardiovascular disease. Participants were enrolled between November 18, 2020, and July 21, 2021, with last follow-up on December 29, 2021. Interventions: Participants were randomized to receive placebo (n = 8) or single doses of SLN360 at 30 mg (n = 6), 100 mg (n = 6), 300 mg (n = 6), or 600 mg (n = 6), administered subcutaneously. Main Outcomes and Measures: The primary outcome was evaluation of safety and tolerability. Secondary outcomes included change in plasma concentrations of Lp(a) to a maximum follow-up of 150 days. Results: Among 32 participants who were randomized and received the study intervention (mean age, 50 [SD, 13.5] years; 17 women [53%]), 32 (100%) completed the trial. One participant experienced 2 serious adverse event episodes: admission to the hospital for headache following SARS-CoV-2 vaccination and later for complications of cholecystitis, both of which were judged to be unrelated to study drug. Median baseline Lp(a) concentrations were as follows: placebo, 238 (IQR, 203-308) nmol/L; 30-mg SLN360, 171 (IQR, 142-219) nmol/L; 100-mg SLN360, 217 (IQR, 202-274) nmol/L; 300-mg SLN360, 285 (IQR, 195-338) nmol/L; and 600-mg SLN360, 231 (IQR, 179-276) nmol/L. Maximal median changes in Lp(a) were -20 (IQR, -61 to 3) nmol/L, -89 (IQR, -119 to -61) nmol/L, -185 (IQR, -226 to -163) nmol/L, -268 (IQR, -292 to -189) nmol/L, and -227 (IQR, -270 to -174) nmol/L, with maximal median percentage changes of -10% (IQR, -16% to 1%), -46% (IQR, -64% to -40%), -86% (IQR, -92% to -82%), -96% (IQR, -98% to -89%), and -98% (IQR, -98% to -97%), for the placebo group and the 30-mg, 100-mg, 300-mg, and 600-mg SLN360 groups, respectively. The duration of Lp(a) lowering was dose dependent, persisting for at least 150 days after administration. Conclusions and Relevance: In this phase 1 study of 32 participants with elevated Lp(a) levels and no known cardiovascular disease, the siRNA SLN360 was well tolerated, and a dose-dependent lowering of plasma Lp(a) concentrations was observed. The findings support further study to determine the safety and efficacy of this siRNA. Trial Registration: ClinicalTrials.gov Identifier: NCT04606602; EudraCT Identifier: 2020-002471-35.


Subject(s)
Apoprotein(a) , Hyperlipoproteinemias , RNA, Small Interfering , Adult , Apoprotein(a)/adverse effects , Apoprotein(a)/biosynthesis , Apoprotein(a)/blood , Cardiovascular Diseases/etiology , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Hyperlipoproteinemias/blood , Hyperlipoproteinemias/genetics , Hyperlipoproteinemias/metabolism , Hyperlipoproteinemias/therapy , Injections, Subcutaneous , Lipoprotein(a)/adverse effects , Lipoprotein(a)/biosynthesis , Lipoprotein(a)/blood , Male , Middle Aged , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/adverse effects , RNA, Small Interfering/therapeutic use , Treatment Outcome
2.
Arterioscler Thromb Vasc Biol ; 40(3): 819-829, 2020 03.
Article in English | MEDLINE | ID: mdl-32078365

ABSTRACT

OBJECTIVE: To clarify the association between PCSK9 (proprotein convertase subtilisin/kexin type 9) and Lp(a) (lipoprotein [a]), we studied Lp(a) kinetics in patients with loss-of-function and gain-of-function PCSK9 mutations and in patients in whom extended-release niacin reduced Lp(a) and PCSK9 concentrations. Approach and Results: Six healthy controls, 9 heterozygous patients with familial hypercholesterolemia (5 with low-density lipoprotein receptor [LDLR] mutations and 4 with PCSK9 gain-of-function mutations) and 3 patients with heterozygous dominant-negative PCSK9 loss-of-function mutations were included in the preliminary study. Eight patients were enrolled in a second study assessing the effects of 2 g/day extended-release niacin. Apolipoprotein kinetics in VLDL (very-low-density lipoprotein), LDL (low-density lipoprotein), and Lp(a) were studied using stable isotope techniques. Plasma Lp(a) concentrations were increased in PCSK9-gain-of-function and familial hypercholesterolemia-LDLR groups compared with controls and PCSK9-loss-of-function groups (14±12 versus 5±4 mg/dL; P=0.04), but no change was observed in Lp(a) fractional catabolic rate. Subjects with PCSK9-loss-of-function mutations displayed reduced apoE (apolipoprotein E) concentrations associated with a VLDL-apoE absolute production rate reduction. Lp(a) and VLDL-apoE absolute production rates were correlated (r=0.50; P<0.05). ApoE-to-apolipoprotein (a) molar ratios in Lp(a) increased with plasma Lp(a) (r=0.96; P<0.001) but not with PCSK9 levels. Extended-release niacin-induced reductions in Lp(a) and VLDL-apoE absolute production rate were correlated (r=0.83; P=0.015). In contrast, PCSK9 reduction (-35%; P=0.008) was only correlated with that of VLDL-apoE absolute production rate (r=0.79; P=0.028). CONCLUSIONS: VLDL-apoE production could determine Lp(a) production and/or assembly. As PCSK9 inhibitors reduce plasma apoE and Lp(a) concentrations, apoE could be the link between PCSK9 and Lp(a).


Subject(s)
Apolipoproteins E/blood , Hyperlipoproteinemia Type II/blood , Lipoprotein(a)/blood , Lipoproteins, VLDL/blood , Adolescent , Adult , Anticholesteremic Agents/therapeutic use , Biomarkers/blood , Case-Control Studies , Delayed-Action Preparations , Female , Genetic Predisposition to Disease , Heterozygote , Humans , Hyperlipoproteinemia Type II/diagnosis , Hyperlipoproteinemia Type II/drug therapy , Hyperlipoproteinemia Type II/genetics , Kinetics , Lipoprotein(a)/biosynthesis , Male , Middle Aged , Mutation , Niacin/therapeutic use , Phenotype , Proprotein Convertase 9/genetics , Randomized Controlled Trials as Topic , Receptors, LDL/genetics , Treatment Outcome , Young Adult
3.
Curr Mol Med ; 19(8): 570-578, 2019.
Article in English | MEDLINE | ID: mdl-31210105

ABSTRACT

BACKGROUND: This study was conducted to determine the effects of vitamin D supplementation on some of the gene expressions related to insulin and lipid metabolism in diabetic hemodialysis (HD) patients. METHODS: A double-blind, randomized, placebo-controlled clinical trial was carried out in 55 patients with diabetic HD. The current project used two groups in which each subject received vitamin D supplements (50,000 IU, n=28) or placebo (50,000 IU, n=27) every 2 weeks for 12 weeks. Gene expression analyses (RT-PCR) were included to obtain the rate of gene expression of the related insulin and lipid metabolism genes in peripheral blood mononuclear cells (PBMCs) of patients with diabetic HD. RESULTS: Our data revealed that consumption of vitamin D supplementation enables to overexpress the peroxisome proliferation-activated receptor gamma (PPAR-γ) (P=0.001), AKT (P=0.04), PI3K (P=0.02), insulin receptor substrate-1 (IRS1) (P0.008) and glucose transporter type 4 (GLUT-4) (P=0.01) and downregulate the expression of protein kinase C (PKC) (P=0.001) in patients with diabetic HD than control group following the 12-week intervention. In addition, vitamin D supplementation downregulated low-density lipoprotein receptor (LDLR) (P=0.03) expression in the subjects with diabetic HD than the control group. Vitamin D supplementation did not show any effects on the expression of pyruvate dehydrogenase kinase 1 (PDK1) (P=0.37), IRS2 (P=0.90) and lipoprotein (a) [Lp(a)] (P=0.05). CONCLUSION: Our findings confirmed that diabetic HD subjects who received the vitamin D supplementation (for 12 weeks), showed a significant overexpression in the PPAR-γ, AKT, PI3K, IRS1 and GLUT4 genes, and also showed a significant downregulation in the PKC and LDLR genes. Moreover, no effects on PDK1, IRS2 and Lp(a) expression were observed.


Subject(s)
Diabetic Nephropathies/therapy , Gene Expression Regulation/drug effects , Insulin/metabolism , Lipid Metabolism/drug effects , Renal Dialysis , Vitamin D/therapeutic use , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Diabetic Nephropathies/genetics , Dietary Supplements , Double-Blind Method , Enzyme Induction/drug effects , Female , Glucose Transporter Type 4/biosynthesis , Glucose Transporter Type 4/genetics , Humans , Insulin Receptor Substrate Proteins/biosynthesis , Insulin Receptor Substrate Proteins/genetics , Lipoprotein(a)/biosynthesis , Lipoprotein(a)/genetics , Male , Middle Aged , PPAR gamma/biosynthesis , PPAR gamma/genetics , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphatidylinositol 3-Kinases/genetics , Protein Kinase C/biosynthesis , Protein Kinase C/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/biosynthesis , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , Signal Transduction , Vitamin D/administration & dosage , Vitamin D/pharmacology , Young Adult
4.
Metabolism ; 65(4): 381-90, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26975530

ABSTRACT

OBJECTIVES: Lipoprotein(a) [Lp(a)] is mainly similar in composition to LDL, but differs in having apolipoprotein (apo) (a) covalently linked to apoB-100. Our purpose was to examine the individual metabolism of apo(a) and apoB-100 within plasma Lp(a). MATERIALS AND METHODS: The kinetics of apo(a) and apoB-100 in plasma Lp(a) were assessed in four men with dyslipidemia [Lp(a) concentration: 8.9-124.7nmol/L]. All subjects received a primed constant infusion of [5,5,5-(2)H3] L-leucine while in the constantly fed state. Lp(a) was immunoprecipitated directly from whole plasma; apo(a) and apoB-100 were separated by gel electrophoresis; and isotopic enrichment was determined by gas chromatography/mass spectrometry. RESULTS: Multicompartmental modeling analysis indicated that the median fractional catabolic rates of apo(a) and apoB-100 within Lp(a) were significantly different at 0.104 and 0.263 pools/day, respectively (P=0.04). The median Lp(a) apo(a) production rate at 0.248nmol/kg·day(-1) was significantly lower than that of Lp(a) apoB-100 at 0.514nmol/kg·day(-1) (P=0.03). CONCLUSION: Our data indicate that apo(a) has a plasma residence time (11days) that is more than twice as long as that of apoB-100 (4days) within Lp(a), supporting the concept that apo(a) and apoB-100 within plasma Lp(a) are not catabolized from the bloodstream as a unit in humans in the fed state.


Subject(s)
Apolipoprotein B-100/metabolism , Apolipoproteins A/metabolism , Lipoprotein(a)/metabolism , Apolipoprotein B-100/biosynthesis , Apolipoprotein B-100/blood , Apolipoproteins A/biosynthesis , Dyslipidemias/blood , Humans , Hypertriglyceridemia/metabolism , Kinetics , Leucine/metabolism , Lipids/blood , Lipoprotein(a)/biosynthesis , Male , Middle Aged
5.
Blood Purif ; 41(4): 270-6, 2016.
Article in English | MEDLINE | ID: mdl-26789774

ABSTRACT

Increased concentrations of low-density-lipoprotein (LDL)-cholesterol (LDL-C) and lipoprotein a (Lp(a)) are scientifically accepted, independent risk factors for the development of atherosclerosis. The complications of atherosclerosis occur early and more frequently. They are strongly linked with lifestyle factors and an increase of LDL-C concentrations in industrialized countries. A new therapeutic approach seems to be the modulation of the proprotein convertase subtilisin/kexin type 9 (PCSK9), which reduces the number of LDL-receptors at the cell membrane of the liver cells and thus increases the concentration of LDL-C in the blood. Results of current studies show, that in particular, a combination of PCSK9-AB and statins, independent of the dosage of the statins, is suitable to increase a reduction of LDL-C and Lp(a). This article gives an overview of the pathophysiology, the current study and research situation as well as the possible different approaches to the therapeutic influence of PCSK9 in the future.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Atherosclerosis/therapy , Hypercholesterolemia/therapy , PCSK9 Inhibitors , Antibodies, Monoclonal, Humanized , Atherosclerosis/complications , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Blood Component Removal , Cholesterol, LDL/antagonists & inhibitors , Cholesterol, LDL/biosynthesis , Clinical Trials, Phase III as Topic , Gene Expression , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/complications , Hypercholesterolemia/metabolism , Hypercholesterolemia/physiopathology , Lipoprotein(a)/antagonists & inhibitors , Lipoprotein(a)/biosynthesis , Liver/drug effects , Liver/metabolism , Proprotein Convertase 9/genetics , Proprotein Convertase 9/metabolism
7.
Circulation ; 132(8): 677-90, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26224810

ABSTRACT

BACKGROUND: Mendelian randomization studies have highlighted that lipoprotein(a) [Lp(a)] was associated with calcific aortic valve disease (CAVD). Lp(a) transports oxidized phospholipids with a high content in lysophosphatidylcholine. Autotaxin (ATX) transforms lysophosphatidylcholine into lysophosphatidic acid. We hypothesized that ATX-lysophosphatidic acid could promote inflammation/mineralization of the aortic valve. METHODS AND RESULTS: We have documented the expression of ATX in control and mineralized aortic valves. By using different approaches, we have also investigated the role of ATX-lysophosphatidic acid in the mineralization of isolated valve interstitial cells and in a mouse model of CAVD. Enzyme-specific ATX activity was elevated by 60% in mineralized aortic valves in comparison with control valves. Immunohistochemistry studies showed a high level of ATX in mineralized aortic valves, which colocalized with oxidized phospholipids and apolipoprotein(a). We detected a high level of ATX activity in the Lp(a) fraction in circulation. Interaction between ATX and Lp(a) was confirmed by in situ proximity ligation assay. Moreover, we documented that valve interstitial cells also expressed ATX in CAVD. We showed that ATX-lysophosphatidic acid promotes the mineralization of the aortic valve through a nuclear factor κB/interleukin 6/bone morphogenetic protein pathway. In LDLR(-/-)/ApoB(100/100)/IGFII mice, ATX is overexpressed and lysophosphatidic acid promotes a strong deposition of hydroxyapatite of calcium in aortic valve leaflets and accelerates the development of CAVD. CONCLUSIONS: ATX is transported in the aortic valve by Lp(a) and is also secreted by valve interstitial cells. ATX-lysophosphatidic acid promotes inflammation and mineralization of the aortic valve and thus could represent a novel therapeutic target in CAVD.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/metabolism , Lipoprotein(a)/biosynthesis , Phosphoric Diester Hydrolases/biosynthesis , Aged , Animals , Aortic Valve/pathology , Aortic Valve Stenosis/pathology , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Lipoprotein(a)/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged
8.
Vaccine ; 33(1): 141-8, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25444801

ABSTRACT

Contagious bovine pleuropneumonia (CBPP), caused by Mycoplasma mycoides subsp. mycoides small colony type (MmmSC), is a devastating respiratory disease of cattle. In sub-Saharan Africa, where CBPP is enzootic, live attenuated vaccines are deployed but afford only short-lived protection. In cattle, recovery from experimental MmmSC infection has been associated with the presence of CD4(+) T lymphocytes that secrete interferon gamma in response to MmmSC, and in particular to the lipoprotein A (LppA) antigen. In an effort to develop a better vaccine against CBPP, a viral vector (Ad5-LppA) that expressed LppA was generated from human adenovirus type 5. The LppA-specific immune responses elicited by the Ad5-LppA vector were evaluated in mice, and compared to those elicited by recombinant LppA formulated with a potent adjuvant. Notably, a single administration of Ad5-LppA, but not recombinant protein, sufficed to elicit a robust LppA-specific humoral response. After a booster administration, both vector and recombinant protein elicited strong LppA-specific humoral and cell-mediated responses. Ex vivo stimulation of splenocytes induced extensive proliferation of CD4(+) T cells for mice immunized with vector or protein, and secretion of T helper 1-associated and proinflammatory cytokines for mice immunized with Ad5-LppA. Our study - by demonstrating the potential of a viral-vectored prototypic vaccine to elicit prompt and robust immune responses against a major antigen of MmmSC - represents a first step in developing a recombinant vaccine against CBPP.


Subject(s)
Adenoviruses, Human/genetics , Bacterial Vaccines/immunology , Drug Carriers , Genetic Vectors , Lipoprotein(a)/immunology , Mycoplasma mycoides/immunology , Pleuropneumonia, Contagious/prevention & control , Animals , Antibodies, Bacterial/blood , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Cytokines/metabolism , Female , Humans , Lipoprotein(a)/biosynthesis , Lipoprotein(a)/genetics , Mice, Inbred BALB C , Mycoplasma mycoides/genetics
9.
Metabolism ; 62(4): 479-91, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23040268

ABSTRACT

Lipoprotein(a) [Lp(a)] resembles low-density lipoprotein (LDL), with an LDL lipid core and apolipoprotein B (apoB), but contains a unique apolipoprotein, apo(a). Elevated Lp(a) is an independent risk factor for coronary and peripheral vascular diseases. The size and concentration of plasma Lp(a) are related to the synthetic rate, not the catabolic rate, and are highly variable with small isoforms associated with high concentrations and pathogenic risk. Apo(a) is synthesized in the liver, although assembly of apo(a) and LDL may occur in the hepatocytes or plasma. While the uptake and clearance site of Lp(a) is poorly delineated, the kidney is the site of apo(a) fragment excretion. The structure of apo(a) has high homology to plasminogen, the zymogen for plasmin and the primary clot lysis enzyme. Apo(a) interferes with plasminogen binding to C-terminal lysines of cell surface and extracellular matrix proteins. Lp(a) and apo(a) inhibit fibrinolysis and accumulate in the vascular wall in atherosclerotic lesions. The pathogenic role of Lp(a) is not known. Small isoforms and high concentrations of Lp(a) are found in healthy octogenarians that suggest Lp(a) may also have a physiological role. Studies of Lp(a) function have been limited since it is not found in commonly studied small mammals. An important aspect of Lp(a) metabolism is the modification of circulating Lp(a), which has the potential to alter the functions of Lp(a). There are no therapeutic drugs that selectively target elevated Lp(a), but a number of possible agents are being considered. Recently, new modifiers of apo(a) synthesis have been identified. This review reports the regulation of Lp(a) metabolism and potential sites for therapeutic targets.


Subject(s)
Lipoprotein(a)/drug effects , Lipoprotein(a)/metabolism , Animals , Apolipoproteins A/metabolism , Gene Expression Regulation/physiology , Humans , Lipoprotein(a)/biosynthesis , Lipoprotein(a)/physiology , Receptors, LDL/metabolism , Tissue Distribution
10.
Blood Coagul Fibrinolysis ; 22(6): 549-51, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21670664

ABSTRACT

Cerebral infarction associated with ulcerative colitis is particularly rare, but clinically important because of its high morbidity and mortality. The pathogenesis of thrombosis in ulcerative colitis remains unclear even if platelet and coagulation cascade abnormalities, systemic inflammation, and the presence of other well recognized vascular risk factors are likely to play a key role in clot formation. We report a case of a young woman with recurrent ischemic stroke secondary to the interaction between ulcerative colitis-related dysbalanced hemostasis and high levels of lipoprotein (a).


Subject(s)
Carotid Stenosis/blood , Colitis, Ulcerative/complications , Lipoprotein(a) , Stroke/blood , Thrombosis/blood , Anticholesteremic Agents/administration & dosage , Anticoagulants/administration & dosage , Aspirin/administration & dosage , Atorvastatin , Blood Coagulation Tests , Carotid Stenosis/diagnostic imaging , Carotid Stenosis/etiology , Clopidogrel , Colitis, Ulcerative/blood , Female , Heparin, Low-Molecular-Weight/administration & dosage , Heptanoic Acids/administration & dosage , Humans , Lipoprotein(a)/adverse effects , Lipoprotein(a)/biosynthesis , Magnetic Resonance Imaging , Middle Aged , Pyrroles/administration & dosage , Risk Factors , Stroke/diagnostic imaging , Stroke/etiology , Thrombosis/etiology , Ticlopidine/administration & dosage , Ticlopidine/analogs & derivatives , Ultrasonography
11.
Atherosclerosis ; 215(2): 392-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21300353

ABSTRACT

OBJECTIVE: Lipoprotein(a) [Lp(a)] is a risk factor for stroke, as has recently been further confirmed by meta analysis, and consists of low-density lipoprotein and apolipoprotein(a), which shares a significant amino acid homology with plasminogen. Oxidized Lp(a) [Ox-Lp(a)] is a more pathogenic species of Lp(a). A monoclonal antibody, specific to Ox-Lp(a), distinguishes immunolocalization of Ox-Lp(a) from that of Lp(a) and that of plasminogen which carries highly homologous domains. It is worth examining their possibly differential immunolocalizations around atherosclerotic lesions in human carotid and cerebral arteries. METHODS AND RESULTS: Stage-related differences in immunolocalization of Ox-Lp(a), native Lp(a), and plasminogen were investigated in various atherosclerotic lesions of the human carotid (obtained from 13 patients undergoing carotid endarterectomy) and cerebral arteries (from 11 cadavers). Native Lp(a) was seen in the fibrous cap, extracellular matrix, endothelial cells, and subendothelial layer. Unorganized mural thrombi were positive for plasminogen, but not Lp(a). In contrast, fibrin deposits in thickened intima were positive for Lp(a), but not plasminogen. Ox-Lp(a)-like immunoreactivity was seen in endothelial cells in the early stage of atherosclerosis. Ox-Lp(a) deposition was more abundant in synthetic phase vascular smooth muscle cells (VSMC) than in contractile phase VSMC. CONCLUSION: We demonstrated differential immunoexpression patterns between native Lp(a) and plasminogen, and suggested that Lp(a)-plasminogen interaction may play a part in differential mechanisms in all atherosclerotic lesions of human carotid and cerebral arteries. The preferential presence of Ox-Lp(a) seen in endothelial cells suggests initial dysfunction of endothelial cells in atherosclerosis. The relative abundance of Ox-Lp(a) in synthetic phase VSMC is associated with their phenotypic changes during the progression of atherosclerosis.


Subject(s)
Carotid Arteries/metabolism , Cerebral Arteries/metabolism , Lipoprotein(a)/biosynthesis , Plaque, Atherosclerotic/metabolism , Plasminogen/biosynthesis , Aged , Aged, 80 and over , Humans , Lipoprotein(a)/immunology , Middle Aged , Oxidation-Reduction
12.
Expert Opin Pharmacother ; 11(15): 2569-72, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20707601

ABSTRACT

Mipomersen, an antisense oligonucleotide directed against apolipoprotein B-100 (apoB), was investigated for its safety and efficacy in reducing low-density lipoprotein (LDL) cholesterol (C) as adjunctive treatment for patients with homozygous familial hypercholesterolemia (HoFH) in a Phase III, double-blind, randomized, controlled trial. HoFH patients are very rare in the general population (∼ 1:1,000,000) and have very high risk for cardiovascular events. HoFH patients respond poorly to statins and most other existing lipid-lowering therapies. Mipomersen (200 or 160 mg) administered subcutaneously to 34 HoFH patients for 26 weeks significantly reduced LDL-C by 24.7% from baseline. In addition, mipomersen lowered plasma lipoprotein (a). In most patients, mipomersen administration was most associated with injection-site reactions; influenza-like symptoms were also more common in mipomersen-treated patients. Four patients had elevated serum alanine aminotransferase (ALT) concentrations, one of whom also had a significant increase in intrahepatic triglyceride content. Another patient met the stopping rules for increased ALT concentrations. No patient developed steatohepatitis during the study. Thus, so far short-term data indicate that mipomersen is safe and effective as an adjunctive drug for lowering LDL-C. Despite these promising results, the longer-term safety and efficacy of mipomersen still needs to be determined.


Subject(s)
Cholesterol, LDL/antagonists & inhibitors , Clinical Trials, Phase II as Topic/standards , Clinical Trials, Phase III as Topic/standards , Oligonucleotides/therapeutic use , Animals , Anticholesteremic Agents/therapeutic use , Cholesterol, LDL/blood , Clinical Trials, Phase II as Topic/methods , Clinical Trials, Phase III as Topic/methods , Double-Blind Method , Humans , Hyperlipoproteinemia Type II/blood , Hyperlipoproteinemia Type II/drug therapy , Lipoprotein(a)/antagonists & inhibitors , Lipoprotein(a)/biosynthesis , Lipoprotein(a)/blood , Oligonucleotides/pharmacokinetics
13.
Eur J Vasc Endovasc Surg ; 38(1): 104-12, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19233690

ABSTRACT

OBJECT: Abdominal aortic aneurysm (AAA) pathogenesis remains poorly understood. This study investigated the gene expression profile of peripheral blood from patients with AAA using microarray technology. METHODS AND RESULTS: We determined gene expression profiles in pooled RNA from 10 AAA patients and 10 matched controls with arrays representing 14,000 transcripts. Microarray data for selected genes were confirmed by real-time PCR in two different AAA (n=36) and control (n=36) populations and integrated with biochemical data. We identified 91 genes which were differentially expressed in AAA patients. Gene Ontology analysis indicated a significant alteration of oxygen transport (increased hemoglobin gene expression) and lipid metabolism [including monoglyceride lipase and low density lipoprotein receptor-related protein 5 (LRP5) gene]. LRP5 expression was associated inversely with serum lipoprotein(a) [Lp(a)] concentration. CONCLUSIONS: Increased expression of hemoglobin chain genes as well as of genes involved in erythrocyte mechanical stability were observed in the AAA RNA pools. The association between low levels of LRP5 gene expression and increased levels of Lp(a) in AAA patients suggests a potential role of LRP5 in Lp(a) catabolism. Our data underline the power of microarrays in identifying further molecular perturbations associated with AAA.


Subject(s)
Aortic Aneurysm, Abdominal/genetics , DNA/genetics , Gene Expression Profiling/methods , Gene Expression Regulation , LDL-Receptor Related Proteins/genetics , Lipoprotein(a)/genetics , Monoacylglycerol Lipases/genetics , Aged , Aged, 80 and over , Aortic Aneurysm, Abdominal/blood , Female , Humans , LDL-Receptor Related Proteins/biosynthesis , Lipoprotein(a)/biosynthesis , Low Density Lipoprotein Receptor-Related Protein-5 , Male , Middle Aged , Monoacylglycerol Lipases/biosynthesis , Polymerase Chain Reaction , Prognosis
14.
Biochemistry ; 46(21): 6299-307, 2007 May 29.
Article in English | MEDLINE | ID: mdl-17474718

ABSTRACT

Small approximately 8.5 nm disks formed spontaneously when dimyristoylphosphatidylcholine (DMPC) large unilamellar vesicles (LUVs) were incubated with apolipoprotein A-I (apoA-I) (100:1 molar ratio). However, in a time course study, the transient production of approximately 11 nm large disks was detected and isolated by gel filtration. The intermediate large disks contained three apoA-I molecules and were stable over time; however, when additional apoA-I was added, they formed small disks containing two molecules of apoA-I. The reaction kinetics of apoA-I with DMPC LUVs was monitored by fluorescence resonance energy transfer, and two phases were observed, supporting the presence of the intermediate in the formation of small disks. The lipid dynamics of LUVs and disks were assayed, revealing the presence of sequestered lipid-protein domains upon apoA-I binding to DMPC LUVs. In addition, the lipids in the intermediate large disks were more constrained than those in the small disks. We propose that apoA-I binds with DMPC LUVs to form small lipid-protein domains on the LUV; then the domains are released to form large disks, which can mature in the presence of additional apoA-I to form small disks. Thus, the formation of small apoA-I lipid disks proceeds through the formation of a large disk intermediate.


Subject(s)
Apolipoprotein A-I/metabolism , Dimyristoylphosphatidylcholine/metabolism , Lipoprotein(a)/biosynthesis , Fluorescence Resonance Energy Transfer , Humans , Kinetics , Unilamellar Liposomes
15.
Thromb Haemost ; 93(5): 867-71, 2005 May.
Article in English | MEDLINE | ID: mdl-15886801

ABSTRACT

From 1998 to 2003, 133 Caucasian women aged 17-40 years (median 29 years) suffering from unexplained recurrent miscarriage (uRM) were consecutively enrolled. In patients and 133 age-matched healthy controls prothrombotic risk factors (factor V (FV) G1691A, factor II (FII) G20210A, MTHFR T677T, 4G/5G plasminogen activator inhibitor (PAI)-1, lipoprotein (Lp) (a), protein C (PC), protein S (PS), antithrombin (AT), antiphospholipid/anticardiolipin (APA/ACA) antibodies) as well as associated environmental conditions (smoking and obesity) were investigated. 70 (52.6%) of the patients had at least one prothrombotic risk factor compared with 26 control women (19.5%; p<0.0001). Body mass index (BMI; p=0.78) and smoking habits (p=0.44) did not differ significantly between the groups investigated. Upon univariate analysis the heterozygous FV mutation, Lp(a) > 30 mg/dL, increased APA/ACA and BMI > 25 kg/m(2) in combination with a prothrombotic risk factor were found to be significantly associated with uRM. In multivariate analysis, increased Lp(a) (odds ratio (OR): 4.7/95% confidence interval (CI): 2.0-10.7), the FV mutation (OR:3.8/CI:1.4-10.7), and increased APA/ACA (OR: 4.5/CI: 1.1-17.7) had independent associations with uRM.


Subject(s)
Abortion, Habitual/blood , Lipoprotein(a)/chemistry , Thrombosis/blood , Abortion, Habitual/diagnosis , Adolescent , Adult , Antibodies, Anticardiolipin/biosynthesis , Antibodies, Antiphospholipid/blood , Anticoagulants/pharmacology , Antithrombins/biosynthesis , Body Mass Index , Case-Control Studies , Factor V/biosynthesis , Female , Follow-Up Studies , Heterozygote , Humans , Lipoprotein(a)/biosynthesis , Logistic Models , Methylenetetrahydrofolate Dehydrogenase (NAD+)/biosynthesis , Multivariate Analysis , Mutation , Odds Ratio , Plasminogen Activator Inhibitor 1/biosynthesis , Protein C/biosynthesis , Protein S/biosynthesis , Prothrombin/biosynthesis , Risk Factors , Thrombosis/diagnosis
16.
J Thromb Haemost ; 3(5): 929-34, 2005 May.
Article in English | MEDLINE | ID: mdl-15869586

ABSTRACT

BACKGROUND: In recent years there has been a significant increase in the diagnosis of sudden sensorineural hearing loss (SSHL) in western, countries with an incidence of 20 of 100,000 people affected every year. No clear causes for this disease have been found thus far, but cochlear ischemia has been hypothesized in patients in whom an infectious episode or acoustic neurinoma have been excluded. OBJECTIVES: The aim of this case-control study was to investigate a number of acquired and inherited thrombophilic risk factors [antithrombin, protein C and S; factor V (FV) Leiden, FII polymorphism; lupus anticoagulant (LA); anticardiolipin (aCL) antibodies; fasting homocysteine (Hcy); lipoprotein(a) (Lp(a)); plasminogen activator inhibitor-1 (PAI-1)] in addition to cardiovascular risk factors in patients with idiopathic SSHL (ISSHL). PATIENTS AND METHODS: We investigated 155 patients (67 male/88 female; age: 55 (range 19-79 years) with a diagnosis of ISSHL within 30 days from the onset of symptoms, and 155 controls (67 male/88 female; age 54 (range 19-78 years). Fasting Hcy levels were significantly higher in patients than in controls [11.6 (6.7-60) micromol/L vs. 8.7 (5.0-24) micromol/L] as well as PAI-1 levels [19 (2-95) mg/dL vs. 14.5 (4.0-87) mg/dL]. Lupus anticoagulant was present in 13 of 155 (8.4%) patients; 20 patients (12.9%) had positivity of aCL (four IgM and 16 IgG). In no patient was a deficiency of physiological clotting inhibitors antithrombin, protein C and protein S found. No significant differences between patients and controls were observed for Lp(a) plasma levels [111 (1-1146) mg/L vs. 103 (11-695) mg/L] and for the presence of FV Leiden (4.5% vs. 4.5%) and FII variant G20210A (3.8% vs. 3.2%). RESULTS AND CONCLUSIONS: Independent risk factors for ISSHL at the multivariate analysis (adjusted for age, sex and the traditional cardiovascular risk factors) were the positivity of aCL: OR 5.6 (95% CI 2.0-15.3); cholesterol levels within the second and third tertiles (with respect to the first tertile): T2 = OR 4.8 (95% CI 1.9-12.6)/T3 = OR 19 (95% CI 7-50.1); PAI-1 and Hcy levels within the third tertile (with respect to the first tertile): OR 20 (95% CI 7.8-78) and OR 4.0 (95% CI 2.0-8.1), respectively. These preliminary data suggest that hypercholesterolemia, hyperhomocysteinemia, elevated PAI-1 levels and anticardiolipin antibodies are associated with ISSHL, so indirectly supporting the hypothesis of a vascular occlusion in the pathogenesis of the disease.


Subject(s)
Hearing Loss, Sensorineural/diagnosis , Thrombophilia/diagnosis , Adult , Aged , Antibodies, Anticardiolipin/biosynthesis , Antithrombins/biosynthesis , Case-Control Studies , Factor V/genetics , Female , Hearing Loss, Sensorineural/complications , Homocysteine/biosynthesis , Humans , Hypercholesterolemia/complications , Hyperhomocysteinemia/complications , Lipoprotein(a)/biosynthesis , Lupus Coagulation Inhibitor/biosynthesis , Male , Middle Aged , Multivariate Analysis , Odds Ratio , Plasminogen Activator Inhibitor 1/biosynthesis , Protein C/biosynthesis , Protein S/biosynthesis , Risk Factors , Time Factors
17.
Biochemistry ; 44(2): 555-64, 2005 Jan 18.
Article in English | MEDLINE | ID: mdl-15641780

ABSTRACT

Human apolipoprotein(a) kringle IV type 10 [apo(a) KIV(10)] contains a strong lysine-binding site (LBS) that mediates the interaction of Lp(a) with biological substrates such as fibrin. Mutations in the KIV(10) LBS have been reported in both the rhesus monkey and chimpanzee, and have been proposed to explain the lack of ability of the corresponding Lp(a) species to bind to lysine and fibrin. To further the comparative analyses with other primate species, we sequenced a segment of baboon liver apo(a) cDNA spanning KIV(9) through the protease domain. Like rhesus monkey apo(a), baboon apo(a) lacks a kringle V (KV)-like domain. Interestingly, we found that the baboon apo(a) KIV(10) sequence contains all of the canonical LBS residues. We sequenced the apo(a) KIV(10) sequence from an additional 10 unrelated baboons; 17 of 20 alleles encoded Trp at position 70, whereas only two alleles encoded Arg at this position and thus a defective LBS. Despite the apparent presence of a functional KIV(10) LBS in most of the baboons, none of the Lp(a) in the plasma of the corresponding baboons bound specifically to lysine-Sepharose (agarose) even upon partial purification. Moreover, baboon Lp(a) bound very poorly to plasmin-modified fibrinogen. Expression of baboon and human KIV(10) in bacteria allowed us to verify that these domains bind comparably to lysine and lysine analogues. We conclude that presentation of KIV(10) in the context of apo(a) lacking KV may interfere with the ability of KIV(10) to bind to substrates such as fibrin; this paradigm may also be present in other non-human primates.


Subject(s)
Apolipoproteins/metabolism , Fibrin/metabolism , Kringles , Lipoprotein(a)/metabolism , Lysine/metabolism , Sepharose/metabolism , Amino Acid Sequence , Animals , Apolipoproteins/genetics , Apolipoproteins/isolation & purification , Apoprotein(a) , Arginine/genetics , Base Sequence , Binding Sites/genetics , Gene Expression Regulation , Humans , Kringles/genetics , Lipoprotein(a)/biosynthesis , Lipoprotein(a)/genetics , Lipoprotein(a)/isolation & purification , Liver/chemistry , Lysine/genetics , Molecular Sequence Data , Papio , Polymorphism, Genetic , Protein Binding/genetics , Recombinant Proteins/metabolism , Tryptophan/genetics
18.
Expert Opin Investig Drugs ; 13(11): 1405-18, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15500389

ABSTRACT

Lipid lowering is established as a proven intervention to reduce atherosclerosis and its complications. This article summarises novel developments in the lipid-altering therapies under development, including combination therapies, squalene synthase inhibitors, microsomal transfer protein inhibitors, acyl-cholesterol acyl transferase inhibitors, cholesterol ester transfer protein antagonists, peroxisome proliferator-activated receptor agonists, high-density lipoprotein-derived peptides and inflammation inhibitors, which have at least reached trials in animal models. Lipid-altering drugs are likely to to be a fast-developing area for novel treatments as possible synergies exist between new and established compounds for the treatment of atherosclerosis. New agents will have to show significant advantage in tolerability or efficacy over existing agents and have the potential to be used in combination therapy as is well established for bile acid sequestrants, nicotinic acid or fibrates and statins. Any new drugs will also have to be assessed in clinical end-point trials against current compounds with proven outcome benefits.


Subject(s)
Clinical Trials as Topic , Drug Design , Lipids/blood , Cholesterol, LDL/blood , Cholesterol, LDL/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Lipid Metabolism , Lipoprotein(a)/biosynthesis
19.
Cancer Res ; 64(19): 7092-8, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15466205

ABSTRACT

The formation of hepatic metastases in colorectal cancer is the main cause of patient death. Current therapies directed at hepatic metastasis of colorectal cancer have had minimal impact on outcome. Therefore, alternative treatment strategies for liver metastasis require development. The present study was performed to evaluate the application of cDNA of LK68 encoding apolipoprotein(a) kringles IV-9, IV-10, and V as possible candidates for gene therapy treatment of this life-threatening disease. The murine colorectal cancer cell line CT26 was transduced ex vivo with LK68 cDNA via retroviral gene transfer, and an experimental model of hepatic metastasis was established by injecting LK68-expressing and control cells into the spleens of BALB/c mice. Expression of LK68 did not affect the growth characteristics and viability of transduced CT26 cells in vitro. LK68 produced from CT26 cells substantially inhibited the migration of endothelial cells in vitro. In vivo, substantial suppression of liver metastasis and prolonged survival were observed in mice bearing LK68-expressing CT26 cells, compared with controls. LK68-expressing liver metastases were restricted to smaller sizes and displayed decreased microvessel density and increased tumor cell apoptosis. Our data collectively indicate that LK68 suppresses angiogenesis-dependent progression of prevascular micrometastases to macroscopic tumors and their growth, which are clinically accessible and biologically relevant therapeutic targets. We propose that antiangiogenic gene therapy with LK68 is a promising strategy for the treatment of colorectal cancer liver metastasis.


Subject(s)
Apolipoproteins/genetics , Colorectal Neoplasms/therapy , Genetic Therapy/methods , Kringles/genetics , Lipoprotein(a)/genetics , Liver Neoplasms, Experimental/prevention & control , Liver Neoplasms, Experimental/secondary , Animals , Apolipoproteins/biosynthesis , Apoprotein(a) , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA, Complementary/administration & dosage , DNA, Complementary/genetics , Disease Models, Animal , Endothelium, Vascular/pathology , Lipoprotein(a)/biosynthesis , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Male , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/therapy
20.
Curr Opin Lipidol ; 15(2): 167-74, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15017359

ABSTRACT

PURPOSE OF REVIEW: Lipoprotein(a) is a structurally and functionally unique lipoprotein consisting of the glycoprotein apolipoprotein(a) covalently linked to LDL. Lipoprotein(a) is assembled extracellularly by a two-step mechanism, still incompletely understood, in which initial non-covalent interactions between apolipoprotein(a) and apolipoprotein B precede specific disulfide bond formation. Elevated concentrations of plasma lipoprotein(a) are a risk factor for a variety of vascular diseases, including coronary heart disease, ischaemic stroke and venous thrombosis. Whereas many pathogenic mechanisms have been proposed for lipoprotein(a), it remains to be conclusively demonstrated which mechanisms are relevant to human disease. RECENT FINDINGS: Structural and functional studies have verified that apolipoprotein(a) kringle 4 types 6-8 contain lysine binding sites of a weaker affinity for lysine analogues than kringle 4 type 10. Recent evidence has conclusively shown a role for kringle 4 types 7 and 8 in lipoprotein(a) assembly. Moreover, apolipoprotein(a) has been shown to undergo a conformational change, from a closed to an open form, which accelerates the rate of covalent lipoprotein(a) assembly. Functional studies in vitro have identified the domains in apolipoprotein(a) that mediate its inhibitory effects on fibrin clot lysis, binding to fibrin and other biological substrates, and pro-inflammatory and anti-angiogenic properties. SUMMARY: Extensive structure-function studies of apolipoprotein(a) have begun to yield important insights into the domains in apolipoprotein(a) that mediate lipoprotein(a) assembly and the pathogenic effects of this lipoprotein. Continued investigations of these relationships will contribute critically to unravelling the many outstanding questions about lipoprotein(a) metabolism and pathophysiology.


Subject(s)
Apolipoproteins/chemistry , Apolipoproteins/metabolism , Cardiovascular Diseases/etiology , Lipoprotein(a)/chemistry , Lipoprotein(a)/metabolism , Apolipoproteins/biosynthesis , Apoprotein(a) , Humans , Kringles , Lipoprotein(a)/biosynthesis , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...