Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Marina Cuchel is active.

Publication


Featured researches published by Marina Cuchel.


The New England Journal of Medicine | 2011

Cholesterol Efflux Capacity, High-Density Lipoprotein Function, and Atherosclerosis

Amit Khera; Marina Cuchel; Margarita de la Llera-Moya; Amrith Rodrigues; Megan F. Burke; Kashif Jafri; Benjamin French; Julie A. Phillips; Megan Mucksavage; Robert L. Wilensky; Emile R. Mohler; George H. Rothblat; Daniel J. Rader

BACKGROUND High-density lipoprotein (HDL) may provide cardiovascular protection by promoting reverse cholesterol transport from macrophages. We hypothesized that the capacity of HDL to accept cholesterol from macrophages would serve as a predictor of atherosclerotic burden. METHODS We measured cholesterol efflux capacity in 203 healthy volunteers who underwent assessment of carotid artery intima-media thickness, 442 patients with angiographically confirmed coronary artery disease, and 351 patients without such angiographically confirmed disease. We quantified efflux capacity by using a validated ex vivo system that involved incubation of macrophages with apolipoprotein B-depleted serum from the study participants. RESULTS The levels of HDL cholesterol and apolipoprotein A-I were significant determinants of cholesterol efflux capacity but accounted for less than 40% of the observed variation. An inverse relationship was noted between efflux capacity and carotid intima-media thickness both before and after adjustment for the HDL cholesterol level. Furthermore, efflux capacity was a strong inverse predictor of coronary disease status (adjusted odds ratio for coronary disease per 1-SD increase in efflux capacity, 0.70; 95% confidence interval [CI], 0.59 to 0.83; P<0.001). This relationship was attenuated, but remained significant, after additional adjustment for the HDL cholesterol level (odds ratio per 1-SD increase, 0.75; 95% CI, 0.63 to 0.90; P=0.002) or apolipoprotein A-I level (odds ratio per 1-SD increase, 0.74; 95% CI, 0.61 to 0.89; P=0.002). Additional studies showed enhanced efflux capacity in patients with the metabolic syndrome and low HDL cholesterol levels who were treated with pioglitazone, but not in patients with hypercholesterolemia who were treated with statins. CONCLUSIONS Cholesterol efflux capacity from macrophages, a metric of HDL function, has a strong inverse association with both carotid intima-media thickness and the likelihood of angiographic coronary artery disease, independently of the HDL cholesterol level. (Funded by the National Heart, Lung, and Blood Institute and others.).


The Lancet | 2013

Efficacy and safety of a microsomal triglyceride transfer protein inhibitor in patients with homozygous familial hypercholesterolaemia: a single-arm, open-label, phase 3 study

Marina Cuchel; Emma A. Meagher; Hendrik du Toit Theron; Dirk Blom; A. David Marais; Robert A. Hegele; Maurizio Averna; Cesare R. Sirtori; Prediman K. Shah; Daniel Gaudet; Claudia Stefanutti; Giovanni Battista Vigna; Anna M.E. Du Plessis; Kathleen J. Propert; William J. Sasiela; LeAnne T. Bloedon; Daniel J. Rader

BACKGROUND Patients with homozygous familial hypercholesterolaemia respond inadequately to existing drugs. We aimed to assess the efficacy and safety of the microsomal triglyceride transfer protein inhibitor lomitapide in adults with this disease. METHODS We did a single-arm, open-label, phase 3 study of lomitapide for treatment of patients with homozygous familial hypercholesterolemia. Current lipid lowering therapy was maintained from 6 weeks before baseline through to at least week 26. Lomitapide dose was escalated on the basis of safety and tolerability from 5 mg to a maximum of 60 mg a day. The primary endpoint was mean percent change in levels of LDL cholesterol from baseline to week 26, after which patients remained on lomitapide through to week 78 for safety assessment. Percent change from baseline to week 26 was assessed with a mixed linear model. FINDINGS 29 men and women with homozygous familial hypercholesterolaemia, aged 18 years or older, were recruited from 11 centres in four countries (USA, Canada, South Africa, and Italy). 23 of 29 enrolled patients completed both the efficacy phase (26 weeks) and the full study (78 weeks). The median dose of lomitapide was 40 mg a day. LDL cholesterol was reduced by 50% (95% CI -62 to -39) from baseline (mean 8·7 mmol/L [SD 2·9]) to week 26 (4·3 mmol/L [2·5]; p<0·0001). Levels of LDL cholesterol were lower than 2·6 mmol/L in eight patients at 26 weeks. Concentrations of LDL cholesterol remained reduced by 44% (95% CI -57 to -31; p<0·0001) at week 56 and 38% (-52 to -24; p<0·0001) at week 78. Gastrointestinal symptoms were the most common adverse event. Four patients had aminotransaminase levels of more than five times the upper limit of normal, which resolved after dose reduction or temporary interruption of lomitapide. No patient permanently discontinued treatment because of liver abnormalities. INTERPRETATION Our study suggests that treatment with lomitapide could be a valuable drug in the management of homozygous familial hypercholesterolaemia. FUNDING FDA Office of the Orphan Product Development, Aegerion Pharmaceuticals.


Circulation | 2006

Macrophage Reverse Cholesterol Transport: Key to the Regression of Atherosclerosis?

Marina Cuchel; Daniel J. Rader

The concept of “reverse cholesterol transport” (RCT) was first introduced in 1968 by Glomset1 to describe the process by which extrahepatic (peripheral) cholesterol is returned to the liver for excretion in the bile and ultimately the feces. The physiological need for this process is clear, as nonhepatic cells acquire cholesterol through uptake of lipoproteins and de novo synthesis and yet (with the exception of steroidogenic tissues that convert cholesterol to steroid hormones) are unable to catabolize it. Excess unesterified cholesterol (UC) is toxic to cells, and therefore, cells have developed several ways to protect themselves against cholesterol toxicity. One key pathway is the efflux of cholesterol to extracellular “acceptors.” The return of this “peripheral” cholesterol to the liver is necessary to balance cholesterol intake and de novo synthesis and thus to maintain whole-body steady-state cholesterol metabolism. The relationship of RCT to atherosclerosis was first suggested by Ross and Glomset,2 who hypothesized that atherosclerotic lesions develop when an imbalance occurs between the deposition and removal of arterial cholesterol after endothelial injury. This concept was further developed by Miller and Miller,3 who suggested that on the basis of the inverse relation between HDL cholesterol (HDL-C) and cardiovascular disease, emphasis should be placed on increasing HDL as a way to increase clearance of cholesterol from the arterial wall to prevent cardiovascular disease. Despite 3 decades of work, the relationship of RCT to atherosclerosis remains more of a hypothesis than an established fact. Because the physiological process of RCT clearly occurs from all peripheral tissues, it has often been measured and discussed as a general peripheral process. However, in atherosclerotic lesions, the primary cell type that is overloaded with cholesterol is the macrophage, and therefore, it makes more sense to conceptualize and measure RCT as a macrophage-specific phenomenon when it comes …


European Heart Journal | 2014

Homozygous familial hypercholesterolaemia: new insights and guidance for clinicians to improve detection and clinical management. A position paper from the Consensus Panel on Familial Hypercholesterolaemia of the European Atherosclerosis Society

Marina Cuchel; Eric Bruckert; Henry N. Ginsberg; Raal Fj; Raul D. Santos; Robert A. Hegele; Jan Albert Kuivenhoven; Børge G. Nordestgaard; Olivier S. Descamps; Elisabeth Steinhagen-Thiessen; Anne Tybjærg-Hansen; Gerald F. Watts; Maurizio Averna; Catherine Boileau; Jan Borén; Alberico L. Catapano; Joep C. Defesche; G. Kees Hovingh; Steve E. Humphries; Petri T. Kovanen; Luis Masana; Päivi Pajukanta; Parhofer Kg; Kausik K. Ray; Anton F. H. Stalenhoef; Erik S. G. Stroes; Marja-Riitta Taskinen; Albert Wiegman; Olov Wiklund; M. John Chapman

Aims Homozygous familial hypercholesterolaemia (HoFH) is a rare life-threatening condition characterized by markedly elevated circulating levels of low-density lipoprotein cholesterol (LDL-C) and accelerated, premature atherosclerotic cardiovascular disease (ACVD). Given recent insights into the heterogeneity of genetic defects and clinical phenotype of HoFH, and the availability of new therapeutic options, this Consensus Panel on Familial Hypercholesterolaemia of the European Atherosclerosis Society (EAS) critically reviewed available data with the aim of providing clinical guidance for the recognition and management of HoFH. Methods and results Early diagnosis of HoFH and prompt initiation of diet and lipid-lowering therapy are critical. Genetic testing may provide a definitive diagnosis, but if unavailable, markedly elevated LDL-C levels together with cutaneous or tendon xanthomas before 10 years, or untreated elevated LDL-C levels consistent with heterozygous FH in both parents, are suggestive of HoFH. We recommend that patients with suspected HoFH are promptly referred to specialist centres for a comprehensive ACVD evaluation and clinical management. Lifestyle intervention and maximal statin therapy are the mainstays of treatment, ideally started in the first year of life or at an initial diagnosis, often with ezetimibe and other lipid-modifying therapy. As patients rarely achieve LDL-C targets, adjunctive lipoprotein apheresis is recommended where available, preferably started by age 5 and no later than 8 years. The number of therapeutic approaches has increased following approval of lomitapide and mipomersen for HoFH. Given the severity of ACVD, we recommend regular follow-up, including Doppler echocardiographic evaluation of the heart and aorta annually, stress testing and, if available, computed tomography coronary angiography every 5 years, or less if deemed necessary. Conclusion This EAS Consensus Panel highlights the need for early identification of HoFH patients, prompt referral to specialized centres, and early initiation of appropriate treatment. These recommendations offer guidance for a wide spectrum of clinicians who are often the first to identify patients with suspected HoFH.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2010

The Ability to Promote Efflux Via ABCA1 Determines the Capacity of Serum Specimens With Similar High-Density Lipoprotein Cholesterol to Remove Cholesterol From Macrophages

Margarita de la Llera-Moya; Denise Drazul-Schrader; Bela F. Asztalos; Marina Cuchel; Daniel J. Rader; George H. Rothblat

Objective—We measured efflux from macrophages to apolipoprotein B-depleted serum from 263 specimens and found instances in which serum having similar high-density lipoprotein cholesterol (HDL-C) differed in their efflux capacity. Thus, we wanted to elucidate why efflux capacity could be independent of total HDL-C or apolipoprotein A-I (apoA-I). Methods and Results—To understand why sera with similar HDL-C or apoA-I could differ in total efflux capacity, we assessed their ability to promote efflux via the pathways expressed in cAMP-treated J774 macrophages. Briefly, macrophages were preincubated with probucol to block ABCA1, with BLT-1 to block SR-BI, and with both inhibitors to measure residual efflux. ABCG1 efflux was measured with transfected BHK-1 cells. We used apolipoprotein B-depleted serum from specimens with similar HDL-C values at the 25th and 75th percentiles. Specimens in each group were classified as having high or low efflux based on total efflux being above or below the group average. We found that independently of HDL-C, sera with higher efflux capacity had a significant increase in ABCA1-mediated efflux, which was significantly correlated to the concentration of pre&bgr;-1 HDL. The same result was obtained when these sera were similarly analyzed based on similar apoA-I. Conclusion—Sera with similar HDL-C or apoA-I differ in their ability to promote macrophage efflux because of differences in the concentration of pre&bgr;-1 HDL.


European Heart Journal | 2015

Familial hypercholesterolaemia in children and adolescents: gaining decades of life by optimizing detection and treatment

Albert Wiegman; Samuel S. Gidding; Gerald F. Watts; M.J. Chapman; Henry N. Ginsberg; Marina Cuchel; Leiv Ose; Maurizio Averna; Catherine Boileau; Jan Borén; Eric Bruckert; A.L. Catapano; Joep C. Defesche; Olivier S. Descamps; Robert A. Hegele; G.K. Hovingh; S.E. Humphries; Petri T. Kovanen; Jan Albert Kuivenhoven; Luis Masana; Børge G. Nordestgaard; Päivi Pajukanta; Klaus G. Parhofer; Frederick J. Raal; Kausik K. Ray; Raul D. Santos; Anton F. H. Stalenhoef; Elisabeth Steinhagen-Thiessen; Erik S.G. Stroes; Marja-Riitta Taskinen

Familial hypercholesterolaemia (FH) is a common genetic cause of premature coronary heart disease (CHD). Globally, one baby is born with FH every minute. If diagnosed and treated early in childhood, individuals with FH can have normal life expectancy. This consensus paper aims to improve awareness of the need for early detection and management of FH children. Familial hypercholesterolaemia is diagnosed either on phenotypic criteria, i.e. an elevated low-density lipoprotein cholesterol (LDL-C) level plus a family history of elevated LDL-C, premature coronary artery disease and/or genetic diagnosis, or positive genetic testing. Childhood is the optimal period for discrimination between FH and non-FH using LDL-C screening. An LDL-C ≥5 mmol/L (190 mg/dL), or an LDL-C ≥4 mmol/L (160 mg/dL) with family history of premature CHD and/or high baseline cholesterol in one parent, make the phenotypic diagnosis. If a parent has a genetic defect, the LDL-C cut-off for the child is ≥3.5 mmol/L (130 mg/dL). We recommend cascade screening of families using a combined phenotypic and genotypic strategy. In children, testing is recommended from age 5 years, or earlier if homozygous FH is suspected. A healthy lifestyle and statin treatment (from age 8 to 10 years) are the cornerstones of management of heterozygous FH. Target LDL-C is <3.5 mmol/L (130 mg/dL) if >10 years, or ideally 50% reduction from baseline if 8–10 years, especially with very high LDL-C, elevated lipoprotein(a), a family history of premature CHD or other cardiovascular risk factors, balanced against the long-term risk of treatment side effects. Identifying FH early and optimally lowering LDL-C over the lifespan reduces cumulative LDL-C burden and offers health and socioeconomic benefits. To drive policy change for timely detection and management, we call for further studies in the young. Increased awareness, early identification, and optimal treatment from childhood are critical to adding decades of healthy life for children and adolescents with FH.


Science | 2016

Rare variant in scavenger receptor BI raises HDL cholesterol and increases risk of coronary heart disease

Paolo Zanoni; Sumeet A. Khetarpal; Daniel B. Larach; William F. Hancock-Cerutti; John S. Millar; Marina Cuchel; Anatol Kontush; Praveen Surendran; Danish Saleheen; Stella Trompet; J.W. Jukema; De Craen A; Panos Deloukas; Naveed Sattar; Ian Ford; Chris J. Packard; Majumder Aa; Dewan S. Alam; Di Angelantonio E; Gonçalo R. Abecasis; Rajiv Chowdhury; Jeanette Erdmann; Børge G. Nordestgaard; Sune F. Nielsen; Anne Tybjærg-Hansen; Schmidt Rf; Kari Kuulasmaa; Dajiang J. Liu; Markus Perola; Stefan Blankenberg

A scavenger that protects the heart Coronary heart disease is a tale of two forms of plasma cholesterol. In contrast to the well-established effects of “bad” cholesterol (LDL-C), the role of “good” cholesterol (HDL-C) is mysterious. Elevated HDL-C correlates with a lower risk of heart disease, yet drugs that raise HDL-C levels do not reduce risk. Zanoni et al. found that some people with exceptionally high levels of HDL-C carry a rare sequence variant in the gene encoding the major HDL-C receptor, scavenger receptor BI. This variant destroys the receptors ability to take up HDL-C. Interestingly, people with this variant have a higher risk of heart disease despite having high levels of HDL-C. Science, this issue p. 1166 A human genetics study sheds light on how HDL (good) cholesterol protects against cardiovascular disease. Scavenger receptor BI (SR-BI) is the major receptor for high-density lipoprotein (HDL) cholesterol (HDL-C). In humans, high amounts of HDL-C in plasma are associated with a lower risk of coronary heart disease (CHD). Mice that have depleted Scarb1 (SR-BI knockout mice) have markedly elevated HDL-C levels but, paradoxically, increased atherosclerosis. The impact of SR-BI on HDL metabolism and CHD risk in humans remains unclear. Through targeted sequencing of coding regions of lipid-modifying genes in 328 individuals with extremely high plasma HDL-C levels, we identified a homozygote for a loss-of-function variant, in which leucine replaces proline 376 (P376L), in SCARB1, the gene encoding SR-BI. The P376L variant impairs posttranslational processing of SR-BI and abrogates selective HDL cholesterol uptake in transfected cells, in hepatocyte-like cells derived from induced pluripotent stem cells from the homozygous subject, and in mice. Large population-based studies revealed that subjects who are heterozygous carriers of the P376L variant have significantly increased levels of plasma HDL-C. P376L carriers have a profound HDL-related phenotype and an increased risk of CHD (odds ratio = 1.79, which is statistically significant).


American Journal of Cardiology | 2008

Longitudinal evaluation and assessment of cardiovascular disease in patients with homozygous familial hypercholesterolemia.

Daniel M. Kolansky; Marina Cuchel; Bernard J. Clark; Steve Paridon; Brian W. McCrindle; Susan E. Wiegers; Luis Araujo; Yogesh Vohra; Joep C. Defesche; James M. Wilson; Daniel J. Rader

Homozygous familial hypercholesterolemia (hoFH) is caused by mutations in the low-density lipoprotein receptor gene and is characterized by severe hypercholesterolemia from birth and onset of premature cardiovascular disease (CVD) during childhood. The onset and progression of CVD using currently available testing methods in children with hoFH have not been fully characterized. A large cohort of patients with hoFH referred to our subspecialty clinic was studied. Thirty-nine patients (22 aged < or =16 years) underwent extensive cardiovascular, lipid, and genetic evaluation. Sixteen children < or =16 years without known CVD when first evaluated were followed up longitudinally for up to 8 years. CVD was clinically evident in 88% of subjects aged >16 years and 9% of those < or =16 years. Markers of atherosclerosis correlated significantly with age at which lipid-lowering treatment was initiated (abnormal coronary angiogram, abnormal aortic valve using echocardiography, and high calcium score using electron beam computed tomography; all p <0.01; abnormal carotid Doppler result; p = 0.03). Twenty of 22 children had no clinical evidence of coronary artery disease, yet 7 of these children had angiographically confirmed mild coronary artery disease (<50%) and 8 had mild to moderate aortic regurgitation using echocardiography. Of noninvasive tests, only evaluation of aortic valve regurgitation using echocardiography predicted the presence of angiographic coronary stenosis (p <0.001). During follow-up, 7 children developed progression of coronary and/or aortic valvular disease during their teenage years and 4 required surgical interventions. In conclusion, in these patients aggressive lipid-lowering treatment initiated in early childhood is warranted. Careful coronary and valvular surveillance strategies and coronary revascularization when appropriate are also warranted in this high-risk population.


Arteriosclerosis, Thrombosis, and Vascular Biology | 1997

Lovastatin Decreases De Novo Cholesterol Synthesis and LDL Apo B-100 Production Rates in Combined-Hyperlipidemic Males

Marina Cuchel; Ernst J. Schaefer; John S. Millar; Peter J. H. Jones; Gregory G. Dolnikowski; Carlo Vergani; Alice H. Lichtenstein

The effect of lovastatin, an inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity, on the kinetics of de novo cholesterol synthesis and apolipoprotein (apo) B in very-low-density lipoprotein (VLDL), intermediate-density lipoprotein (IDL), and low-density lipoprotein (LDL) was investigated in five male patients with combined hyperlipidemia. Subjects were counseled to follow a Step 2 diet and were treated with lovastatin and placebo in randomly assigned order for 6-week periods. At the end of each experimental period, subjects were given deuterium oxide orally and de novo cholesterol synthesis was assessed from deuterium incorporation into cholesterol and expressed as fractional synthesis rate (C-FSR) and production rate (C-PR). Simultaneously, the kinetics of VLDL, IDL, and LDL apo B-100 were studied in the fed state using a primed-constant infusion of deuterated leucine to measure fractional catabolic rates (FCR) and production rates (PR). Drug treatment resulted in significant decreases in total cholesterol (-29%), VLDL cholesterol (-40%), LDL cholesterol (-27%), and apo B (-16%) levels and increases in HDL cholesterol (+13%) and apolipoprotein (apo) A-I (+11%) levels. Associated with these plasma lipoprotein responses was a significant reduction in both de novo C-FSR (-40%; P = .04) and C-PR (-42%; P = .03). Treatment with lovastain in these patients had no significant effect on the FCR of apoB-100 in VLDL, IDL, or LDL, but resulted in a significant decrease in the PR of apoB-100 in IDL and LDL. Comparing the kinetic data of these patients with those of 10 normolipidemic control subjects indicates that lovastatin treatment normalized apoB-100 IDL and LDL PR. The results of these studies suggest that the declines in plasma lipid levels observed after treatment of combined hyperlipidemic patients with lovastatin are attributable to reductions in the C-FSR and C-PR of de novo cholesterol synthesis and the PR of apoB-100 containing lipoproteins. The decline in de novo cholesterol synthesis, rather than an increase in direct uptake of VLDL and IDL, may have contributed to the decline in the PR observed.


Journal of Clinical Investigation | 2010

Primary deficiency of microsomal triglyceride transfer protein in human abetalipoproteinemia is associated with loss of CD1 function

Sebastian Zeissig; Stephanie K. Dougan; Duarte C. Barral; Yvonne Junker; Zhangguo Chen; Arthur Kaser; Madelyn M. Ho; Hannah Mandel; Adam D. McIntyre; Susan M. Kennedy; Gavin F. Painter; Natacha Veerapen; Gurdyal S. Besra; Vincenzo Cerundolo; Simon Yue; Sarah Beladi; Samuel M. Behar; Xiuxu Chen; Jenny E. Gumperz; Karine Breckpot; Anna Raper; Amanda Baer; Mark A. Exley; Robert A. Hegele; Marina Cuchel; Daniel J. Rader; Nicholas O. Davidson; Richard S. Blumberg

Abetalipoproteinemia (ABL) is a rare Mendelian disorder of lipid metabolism due to genetic deficiency in microsomal triglyceride transfer protein (MTP). It is associated with defects in MTP-mediated lipid transfer onto apolipoprotein B (APOB) and impaired secretion of APOB-containing lipoproteins. Recently, MTP was shown to regulate the CD1 family of lipid antigen-presenting molecules, but little is known about immune function in ABL patients. Here, we have shown that ABL is characterized by immune defects affecting presentation of self and microbial lipid antigens by group 1 (CD1a, CD1b, CD1c) and group 2 (CD1d) CD1 molecules. In dendritic cells isolated from ABL patients, MTP deficiency was associated with increased proteasomal degradation of group 1 CD1 molecules. Although CD1d escaped degradation, it was unable to load antigens and exhibited functional defects similar to those affecting the group 1 CD1 molecules. The reduction in CD1 function resulted in impaired activation of CD1-restricted T and invariant natural killer T (iNKT) cells and reduced numbers and phenotypic alterations of iNKT cells consistent with central and peripheral CD1 defects in vivo. These data highlight MTP as a unique regulator of human metabolic and immune pathways and reveal that ABL is not only a disorder of lipid metabolism but also an immune disease involving CD1.

Collaboration


Dive into the Marina Cuchel's collaboration.

Top Co-Authors

Avatar

Daniel J. Rader

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

John S. Millar

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Robert A. Hegele

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

LeAnne T. Bloedon

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Prediman K. Shah

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dirk Blom

University of Cape Town

View shared research outputs
Top Co-Authors

Avatar

Emma A. Meagher

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Paolo Zanoni

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge