101
|
Fischer AW, Albers K, Krott LM, Hoffzimmer B, Heine M, Schmale H, Scheja L, Gordts PLSM, Heeren J. The adaptor protein PID1 regulates receptor-dependent endocytosis of postprandial triglyceride-rich lipoproteins. Mol Metab 2018; 16:88-99. [PMID: 30100244 PMCID: PMC6158030 DOI: 10.1016/j.molmet.2018.07.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/20/2018] [Accepted: 07/25/2018] [Indexed: 01/19/2023] Open
Abstract
Objective Insulin resistance is associated with impaired receptor dependent hepatic uptake of triglyceride-rich lipoproteins (TRL), promoting hypertriglyceridemia and atherosclerosis. Next to low-density lipoprotein (LDL) receptor (LDLR) and syndecan-1, the LDLR-related protein 1 (LRP1) stimulated by insulin action contributes to the rapid clearance of TRL in the postprandial state. Here, we investigated the hypothesis that the adaptor protein phosphotyrosine interacting domain-containing protein 1 (PID1) regulates LRP1 function, thereby controlling hepatic endocytosis of postprandial lipoproteins. Methods Localization and interaction of PID1 and LRP1 in cultured hepatocytes was studied by confocal microscopy of fluorescent tagged proteins, by indirect immunohistochemistry of endogenous proteins, by GST-based pull down and by immunoprecipitation experiments. The in vivo relevance of PID1 was assessed using whole body as well as liver-specific Pid1-deficient mice on a wild type or Ldlr-deficient (Ldlr−/−) background. Intravital microscopy was used to study LRP1 translocation in the liver. Lipoprotein metabolism was investigated by lipoprotein profiling, gene and protein expression as well as organ-specific uptake of radiolabelled TRL. Results PID1 co-localized in perinuclear endosomes and was found associated with LRP1 under fasting conditions. We identified the distal NPxY motif of the intracellular C-terminal domain (ICD) of LRP1 as the site critical for the interaction with PID1. Insulin-mediated NPxY-phosphorylation caused the dissociation of PID1 from the ICD, causing LRP1 translocation to the plasma membrane. PID1 deletion resulted in higher LRP1 abundance at the cell surface, higher LDLR protein levels and, paradoxically, reduced total LRP1. The latter can be explained by higher receptor shedding, which we observed in cultured Pid1-deficient hepatocytes. Consistently, PID1 deficiency alone led to increased LDLR-dependent endocytosis of postprandial lipoproteins and lower plasma triglycerides. In contrast, hepatic PID1 deletion on an Ldlr−/− background reduced lipoprotein uptake into liver and caused plasma TRL accumulation. Conclusions By acting as an insulin-dependent retention adaptor, PID1 serves as a regulator of LRP1 function controlling the disposal of postprandial lipoproteins. PID1 inhibition provides a novel approach to lower plasma levels of pro-atherogenic TRL remnants by stimulating endocytic function of both LRP1 and LDLR in the liver. PID1 is a retention adaptor protein that regulates activity of the endocytic receptor LDL receptor-related protein 1 (LRP1). PID1 regulates the insulin-dependent LRP1-mediated endocytosis of lipoproteins in vivo. PID1 deficiency in liver reduces LRP1 levels via cell surface shedding, and paradoxically increases LDL receptor activity. PID1 antagonism has therapeutic potential to reduce pro-atherogenic lipoproteins in hyperlipidemic and diabetic patients.
Collapse
Affiliation(s)
- Alexander W Fischer
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Kirstin Albers
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Lucia M Krott
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Britta Hoffzimmer
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Hartwig Schmale
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Philip L S M Gordts
- Department of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| |
Collapse
|
102
|
Mytilinaiou M, Kyrou I, Khan M, Grammatopoulos DK, Randeva HS. Familial Hypercholesterolemia: New Horizons for Diagnosis and Effective Management. Front Pharmacol 2018; 9:707. [PMID: 30050433 PMCID: PMC6052892 DOI: 10.3389/fphar.2018.00707] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 06/12/2018] [Indexed: 12/11/2022] Open
Abstract
Familial hypercholesterolemia (FH) is a common genetic cause of premature cardiovascular disease (CVD). The reported prevalence rates for both heterozygous FH (HeFH) and homozygous FH (HoFH) vary significantly, and this can be attributed, at least in part, to the variable diagnostic criteria used across different populations. Due to lack of consistent data, new global registries and unified guidelines are being formed, which are expected to advance current knowledge and improve the care of FH patients. This review presents a comprehensive overview of the pathophysiology, epidemiology, manifestations, and pharmacological treatment of FH, whilst summarizing the up-to-date relevant recommendations and guidelines. Ongoing research in FH seems promising and novel therapies are expected to be introduced in clinical practice in order to compliment or even substitute current treatment options, aiming for better lipid-lowering effects, fewer side effects, and improved clinical outcomes.
Collapse
Affiliation(s)
- Maria Mytilinaiou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom.,Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, United Kingdom.,Division of Translational and Experimental Medicine, Warwick Medical School, University of Warwick, Coventry, United Kingdom.,Centre of Applied Biological and Exercise Sciences, Faculty of Health and Life Sciences, Coventry University, Coventry, United Kingdom
| | - Mike Khan
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom
| | - Dimitris K Grammatopoulos
- Division of Translational and Experimental Medicine, Warwick Medical School, University of Warwick, Coventry, United Kingdom.,Institute of Precision Diagnostics and Translational Medicine, Coventry and Warwickshire Pathology Service, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom
| | - Harpal S Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom.,Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, United Kingdom.,Division of Translational and Experimental Medicine, Warwick Medical School, University of Warwick, Coventry, United Kingdom.,Centre of Applied Biological and Exercise Sciences, Faculty of Health and Life Sciences, Coventry University, Coventry, United Kingdom.,Institute of Precision Diagnostics and Translational Medicine, Coventry and Warwickshire Pathology Service, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom
| |
Collapse
|
103
|
Zhang YY, Fu ZY, Wei J, Qi W, Baituola G, Luo J, Meng YJ, Guo SY, Yin H, Jiang SY, Li YF, Miao HH, Liu Y, Wang Y, Li BL, Ma YT, Song BL. A LIMA1 variant promotes low plasma LDL cholesterol and decreases intestinal cholesterol absorption. Science 2018; 360:1087-1092. [DOI: 10.1126/science.aao6575] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 02/07/2018] [Accepted: 04/17/2018] [Indexed: 12/19/2022]
Abstract
A high concentration of low-density lipoprotein cholesterol (LDL-C) is a major risk factor for cardiovascular disease. Although LDL-C levels vary among humans and are heritable, the genetic factors affecting LDL-C are not fully characterized. We identified a rare frameshift variant in the LIMA1 (also known as EPLIN or SREBP3) gene from a Chinese family of Kazakh ethnicity with inherited low LDL-C and reduced cholesterol absorption. In a mouse model, LIMA1 was mainly expressed in the small intestine and localized on the brush border membrane. LIMA1 bridged NPC1L1, an essential protein for cholesterol absorption, to a transportation complex containing myosin Vb and facilitated cholesterol uptake. Similar to the human phenotype, Lima1-deficient mice displayed reduced cholesterol absorption and were resistant to diet-induced hypercholesterolemia. Through our study of both mice and humans, we identify LIMA1 as a key protein regulating intestinal cholesterol absorption.
Collapse
|
104
|
Saeed A, Virani SS, Jones PH, Ballantyne CM, Nambi V. Case reports of proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibition nonresponse. J Clin Lipidol 2018; 12:1141-1145. [PMID: 30318064 DOI: 10.1016/j.jacl.2018.05.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 05/14/2018] [Accepted: 05/22/2018] [Indexed: 10/14/2022]
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, a novel class of monoclonal antibodies, reduces low-density lipoprotein cholesterol levels and improves cardiovascular outcomes. Given the short time frame, these agents have been available for use; reports of nonresponse to the PCSK9 inhibitor therapy are scarce in literature. We describe 2 cases with substantially lesser than expected low-density lipoprotein cholesterol lowering on PCSK9 therapy. Nonresponse to PCSK9 inhibition was attributed to autosomal recessive hypercholesterolemia (secondary to low-density lipoprotein receptor adaptor protein 1 mutation) and plasmapheresis after PCSK9 inhibitor drug injections. Additional PCSK9 inhibitor nonresponders are likely to emerge as the use of these agents increases overtime.
Collapse
Affiliation(s)
- Anum Saeed
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston, TX, USA
| | - Salim S Virani
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston, TX, USA; Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA; Health Policy, Quality & Informatics Program, Michael E. DeBakey Veterans Affairs Medical Center Health Services Research and Development Center for Innovations, Houston, TX, USA; Section of Health Services Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Peter H Jones
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Christie M Ballantyne
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston, TX, USA; Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Vijay Nambi
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston, TX, USA; Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA; Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
105
|
Veljkovic N, Zaric B, Djuric I, Obradovic M, Sudar-Milovanovic E, Radak D, Isenovic ER. Genetic Markers for Coronary Artery Disease. MEDICINA (KAUNAS, LITHUANIA) 2018; 54:E36. [PMID: 30344267 PMCID: PMC6122104 DOI: 10.3390/medicina54030036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 12/22/2022]
Abstract
Coronary artery disease (CAD) and myocardial infarction (MI) are recognized as leading causes of mortality in developed countries. Although typically associated with behavioral risk factors, such as smoking, sedentary lifestyle, and poor dietary habits, such vascular phenotypes have also long been recognized as being related to genetic background. We review the currently available data concerning genetic markers for CAD in English and non-English articles with English abstracts published between 2003 and 2018. As genetic testing is increasingly available, it may be possible to identify adequate genetic markers representing the risk profile and to use them in a clinical setting.
Collapse
Affiliation(s)
- Nevena Veljkovic
- Centre for Multidisciplinary Research and Engineering, Institute of Nuclear Science Vinca, University of Belgrade, 11000 Belgrade, Serbia.
| | - Bozidarka Zaric
- Laboratory of Radiobiology and Molecular Genetics, Institute of Nuclear Science Vinca, University of Belgrade, 11000 Belgrade, Serbia.
| | - Ilona Djuric
- Department for Endocrinology and Immunoradiology 11080 Zemun, Institute for the Application of Nuclear Energy-INEP, University of Belgrade, 11000 Belgrade, Serbia.
| | - Milan Obradovic
- Laboratory of Radiobiology and Molecular Genetics, Institute of Nuclear Science Vinca, University of Belgrade, 11000 Belgrade, Serbia.
| | - Emina Sudar-Milovanovic
- Laboratory of Radiobiology and Molecular Genetics, Institute of Nuclear Science Vinca, University of Belgrade, 11000 Belgrade, Serbia.
| | - Djordje Radak
- School of Medicine, Dedinje Cardiovascular Institute, University of Belgrade, 11000 Belgrade, Serbia.
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia.
- Serbian Academy of Sciences and Arts, 11000 Belgrade, Serbia.
| | - Esma R Isenovic
- Laboratory of Radiobiology and Molecular Genetics, Institute of Nuclear Science Vinca, University of Belgrade, 11000 Belgrade, Serbia.
| |
Collapse
|
106
|
Jordan DM, Do R. Using Full Genomic Information to Predict Disease: Breaking Down the Barriers Between Complex and Mendelian Diseases. Annu Rev Genomics Hum Genet 2018; 19:289-301. [PMID: 29641912 DOI: 10.1146/annurev-genom-083117-021136] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
While sequence-based genetic tests have long been available for specific loci, especially for Mendelian disease, the rapidly falling costs of genome-wide genotyping arrays, whole-exome sequencing, and whole-genome sequencing are moving us toward a future where full genomic information might inform the prognosis and treatment of a variety of diseases, including complex disease. Similarly, the availability of large populations with full genomic information has enabled new insights about the etiology and genetic architecture of complex disease. Insights from the latest generation of genomic studies suggest that our categorization of diseases as complex may conceal a wide spectrum of genetic architectures and causal mechanisms that ranges from Mendelian forms of complex disease to complex regulatory structures underlying Mendelian disease. Here, we review these insights, along with advances in the prediction of disease risk and outcomes from full genomic information.
Collapse
Affiliation(s)
- Daniel M Jordan
- Charles Bronfman Institute for Personalized Medicine and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Ron Do
- Charles Bronfman Institute for Personalized Medicine and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| |
Collapse
|
107
|
Balder JW, Rimbert A, Zhang X, Viel M, Kanninga R, van Dijk F, Lansberg P, Sinke R, Kuivenhoven JA. Genetics, Lifestyle, and Low-Density Lipoprotein Cholesterol in Young and Apparently Healthy Women. Circulation 2018; 137:820-831. [DOI: 10.1161/circulationaha.117.032479] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/08/2018] [Indexed: 01/02/2023]
Abstract
Background:
Atherosclerosis starts in childhood but low-density lipoprotein cholesterol (LDL-C), a causal risk factor, is mostly studied and dealt with when clinical events have occurred. Women are usually affected later in life than men and are underdiagnosed, undertreated, and understudied in cardiovascular trials and research. This study aims at a better understanding of lifestyle and genetic factors that affect LDL-C in young women.
Methods:
We randomly selected for every year of age 8 women with LDL-C ≤1st percentile (≤50 mg/dL) and 8 women with LDL-C ≥99th percentile (≥186 mg/dL) from 28 000 female participants aged between 25 to 40 years of a population-based cohort study. The resulting groups include 119 and 121 women, respectively, of an average 33 years of age. A gene-sequencing panel was used to assess established monogenic and polygenic origins of these phenotypes. Information on lifestyle was extracted from questionnaires. A healthy lifestyle score was allocated based on a recently developed algorithm.
Results:
Of the women with LDL-C ≤1st percentile, 19 (15.7%) carried mutations that are causing monogenic hypocholesterolemia and 60 (49.6%) were genetically predisposed to low LDL-C on the basis of an extremely low weighted genetic risk score. In comparison with control groups, a healthier lifestyle was not associated with low LDL-C in women without genetic predispositions. Among women with LDL-C ≥99th percentile, 20 women (16.8%) carried mutations that cause familial hypercholesterolemia, whereas 25 (21%) were predisposed to high LDL-C on the basis of a high-weighted genetic risk score. The women in whom no genetic origin for hypercholesterolemia could be identified were found to exhibit a significantly unfavorable lifestyle in comparison with controls.
Conclusions:
This study highlights the need for early assessment of the cardiovascular risk profile in apparently healthy young women to identify those with LDL-C ≥99th percentile for their age: first, because, in this study, 17% of the cases were molecularly diagnosed with familial hypercholesterolemia, which needs further attention; second, because our data indicate that an unfavorable lifestyle is significantly associated with severe hypercholesterolemia in genetically unaffected women, which may also need further attention.
Collapse
Affiliation(s)
- Jan-Willem Balder
- Department of Pediatrics, Section Molecular Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (J.-W.B., A.R., P.L., J.A.K.)
- Department of Vascular Medicine, University Medical Center Groningen, University of Groningen, the Netherlands (J.-W.B.)
| | - Antoine Rimbert
- Department of Pediatrics, Section Molecular Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (J.-W.B., A.R., P.L., J.A.K.)
| | - Xiang Zhang
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands (X.Z.)
| | - Martijn Viel
- Department of Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (M.V., R.K., F.v.D., R.S.)
| | - Roan Kanninga
- Department of Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (M.V., R.K., F.v.D., R.S.)
| | - Freerk van Dijk
- Department of Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (M.V., R.K., F.v.D., R.S.)
| | - Peter Lansberg
- Department of Pediatrics, Section Molecular Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (J.-W.B., A.R., P.L., J.A.K.)
| | - Richard Sinke
- Department of Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (M.V., R.K., F.v.D., R.S.)
| | - Jan Albert Kuivenhoven
- Department of Pediatrics, Section Molecular Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (J.-W.B., A.R., P.L., J.A.K.)
| |
Collapse
|
108
|
Sánchez-Hernández RM, Prieto-Matos P, Civeira F, Lafuente EE, Vargas MF, Real JT, Goicoechea FG, Fuentes FJ, Pocovi M, Boronat M, Wägner AM, Masana L. Autosomal recessive hypercholesterolemia in Spain. Atherosclerosis 2018; 269:1-5. [PMID: 29245109 DOI: 10.1016/j.atherosclerosis.2017.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/21/2017] [Accepted: 12/05/2017] [Indexed: 10/18/2022]
Abstract
BACKGROUND AND AIMS Autosomal recessive hypercholesterolemia (ARH) is a very rare disease, caused by mutations in LDL protein receptor adaptor 1 (LDLRAP1). It is characterized by high levels of low-density lipoprotein cholesterol (LDL-C) and increased risk of premature cardiovascular disease. We aimed to characterize ARH in Spain. METHODS Data were collected from the Dyslipidemia Registry of the Spanish Atherosclerosis Society. A literature search was performed up to June 2017, and all diagnostic genetic studies for familial hypercholesterolemia of Spain were reviewed. RESULTS Seven patients with ARH were identified, 6 true homozygous and one compound heterozygous with a novel mutation: c.[863C>T];p.[Ser288Leu]. High genetic heterogeneity was found in this cohort. True homozygous subjects for LDLRAP1 have more severe phenotypes than the compound heterozygous patient, but similar to patients with homozygous familial hypercholesterolemia (HoFH). Cardiovascular disease was present in 14% of the ARH patients. LDL-C under treatment was above 185 mg/dl and the response to PCSK9 inhibitors was heterogeneous. Finally, the estimated prevalence in Spain is very low, with just 1 case per 6.5 million people. CONCLUSIONS ARH is a very rare disease in Spain, showing high genetic heterogeneity, similarly high LDL-C concentrations, but lower incidence of ASCVD than HoFH.
Collapse
Affiliation(s)
- Rosa María Sánchez-Hernández
- Sección de Endocrinología y Nutrición, Complejo Hospitalario Universitario Insular Materno Infantil de Gran Canaria, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.
| | - Pablo Prieto-Matos
- Unidad de Endocrinología Pediátrica, Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca, Spain
| | - Fernando Civeira
- Unidad Clínica y de Investigación en Lípidos y Arterioesclerosis, Hospital Universitario Miguel Servet, IIS Aragón, Centro de Investigación Biomedica en Red de Enfermedades Cardiovasculares (CIBERCV), Universidad de Zaragoza, Zaragoza, Spain
| | - Eduardo Esteve Lafuente
- Servicio Endocrinología y Nutrición, Hospital Universitari de Girona Dr. Josep Trueta, Spain
| | | | - José T Real
- Servicio de Endocrinología y Nutrición, Hospital Clínico Valencia, Departamento de Medicina, Universidad de Valencia, INCLIVA, Centro de Investigación Biomedica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERDEM), Spain
| | | | - Francisco J Fuentes
- Hospital Universitario Reina Sofía, Universidad de Córdoba, Centro de Investigación Biomédica en Red de Fisiopatolgía de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Instituto Maimónedes de Investigación Biomédica de Córdoba (IMIBIC), Spain
| | - Miguel Pocovi
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, IIS Aragón, CIBERCV, Zaragoza, Spain
| | - Mauro Boronat
- Sección de Endocrinología y Nutrición, Complejo Hospitalario Universitario Insular Materno Infantil de Gran Canaria, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Ana María Wägner
- Sección de Endocrinología y Nutrición, Complejo Hospitalario Universitario Insular Materno Infantil de Gran Canaria, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Luis Masana
- Unidad de Medicina Vascular y Metabolica, Unidad de Investigación en Lipidos y Arterioesclerosis, Hospital Universitario "Sant Joan", Universitat Rovira i Virgili, IISPV, CIBERDEM, Reus, Madrid, Spain
| |
Collapse
|
109
|
New Sequencing technologies help revealing unexpected mutations in Autosomal Dominant Hypercholesterolemia. Sci Rep 2018; 8:1943. [PMID: 29386597 PMCID: PMC5792649 DOI: 10.1038/s41598-018-20281-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/15/2018] [Indexed: 01/25/2023] Open
Abstract
Autosomal dominant hypercholesterolemia (ADH) is characterized by elevated LDL-C levels leading to coronary heart disease. Four genes are implicated in ADH: LDLR, APOB, PCSK9 and APOE. Our aim was to identify new mutations in known genes, or in new genes implicated in ADH. Thirteen French families with ADH were recruited and studied by exome sequencing after exclusion, in their probands, of mutations in the LDLR, PCSK9 and APOE genes and fragments of exons 26 and 29 of APOB gene. We identified in one family a p.Arg50Gln mutation in the APOB gene, which occurs in a region not usually associated with ADH. Segregation and in-silico analysis suggested that this mutation is disease causing in the family. We identified in another family with the p.Ala3396Thr mutation of APOB, one patient with a severe phenotype carrying also a mutation in PCSK9: p.Arg96Cys. This is the first compound heterozygote reported with a mutation in APOB and PCSK9. Functional studies proved that the p.Arg96Cys mutation leads to increased LDL receptor degradation. This work shows that Next-Generation Sequencing (exome, genome or targeted sequencing) are powerful tools to find new mutations and identify compound heterozygotes, which will lead to better diagnosis and treatment of ADH.
Collapse
|
110
|
D'Erasmo L, Minicocci I, Nicolucci A, Pintus P, Roeters Van Lennep JE, Masana L, Mata P, Sánchez-Hernández RM, Prieto-Matos P, Real JT, Ascaso JF, Lafuente EE, Pocovi M, Fuentes FJ, Muntoni S, Bertolini S, Sirtori C, Calabresi L, Pavanello C, Averna M, Cefalu AB, Noto D, Pacifico AA, Pes GM, Harada-Shiba M, Manzato E, Zambon S, Zambon A, Vogt A, Scardapane M, Sjouke B, Fellin R, Arca M. Autosomal Recessive Hypercholesterolemia: Long-Term Cardiovascular Outcomes. J Am Coll Cardiol 2018; 71:279-288. [PMID: 29348020 DOI: 10.1016/j.jacc.2017.11.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 10/16/2017] [Accepted: 11/06/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Autosomal recessive hypercholesterolemia (ARH) is a rare lipid disorder characterized by premature atherosclerotic cardiovascular disease (ASCVD). There are sparse data for clinical management and cardiovascular outcomes in ARH. OBJECTIVES Evaluation of changes in lipid management, achievement of low-density lipoprotein cholesterol (LDL-C) goals and cardiovascular outcomes in ARH. METHODS Published ARH cases were identified by electronic search. All corresponding authors and physicians known to treat these patients were asked to provide follow-up information, using a standardized protocol. RESULTS We collected data for 52 patients (28 females, 24 males; 31.1 ± 17.1 years of age; baseline LDL-C: 571.9 ± 171.7 mg/dl). During a mean follow-up of 14.1 ± 7.3 years, there was a significant increase in the use of high-intensity statin and ezetimibe in combination with lipoprotein apheresis; in 6 patients, lomitapide was also added. Mean LDL-C achieved at nadir was 164.0 ± 85.1 mg/dl (-69.6% from baseline), with a better response in patients taking lomitapide (-88.3%). Overall, 23.1% of ARH patients reached LDL-C of <100 mg/dl. During follow-up, 26.9% of patients had incident ASCVD, and 11.5% had a new diagnosis of aortic valve stenosis (absolute risk per year of 1.9% and 0.8%, respectively). No incident stroke was observed. Age (≥30 years) and the presence of coronary artery disease at diagnosis were the major predictors of incident ASCVD. CONCLUSIONS Despite intensive treatment, LDL-C in ARH patients remains far from targets, and this translates into a poor long-term cardiovascular prognosis. Our data highlight the importance of an early diagnosis and treatment and confirm the fact that an effective treatment protocol for ARH is still lacking.
Collapse
Affiliation(s)
- Laura D'Erasmo
- Department of Internal Medicine and Clinical Specialties, Sapienza University of Rome, Rome, Italy.
| | - Ilenia Minicocci
- Department of Internal Medicine and Clinical Specialties, Sapienza University of Rome, Rome, Italy
| | - Antonio Nicolucci
- Center for Outcomes Research and Clinical Epidemiology, Coreresearch, Inc., Pescara, Italy
| | - Paolo Pintus
- Dipartimento Internistico, Centro per le Malattie Dismetaboliche e l'Arteriosclerosi, Cagliari, Italy
| | | | - Luis Masana
- Research Unit on Lipids and Atherosclerosis, Vascular Medicine and Metabolism Unit, Sant Joan University Hospital, Universitat Rovira i Virgili, IISPV, Reus, Spain, and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Pedro Mata
- Fundación Hipercoesterolaemia Familiar, Madrid, Spain
| | - Rosa Maria Sánchez-Hernández
- Sección de Endocrinología y Nutrición, Complejo Hospitalario Universitario Insular Materno Infantil de Gran Canaria, Instituto Universitario de Investigación Biomédica y Sanitaria (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Pablo Prieto-Matos
- Unidad de Endocrinología Pediátrica Hospital Universitario de Salamanca Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Josè T Real
- Servicio de Endocrinología y Nutrición, Hospital Clínico Valencia, Valencia, Spain, and Department of Medicine, University of Valencia, INCLIVA, CIBERDEM, Madrid, Spain
| | - Juan F Ascaso
- Servicio de Endocrinología y Nutrición, Hospital Clínico Valencia, Valencia, Spain, and Department of Medicine, University of Valencia, INCLIVA, CIBERDEM, Madrid, Spain
| | | | - Miguel Pocovi
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza & IIS Aragón, CIBERCV, Zaragoza, Spain
| | - Francisco J Fuentes
- Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, Universidad de Córdoba, Córdoba, Spain, and Centro de Investigación Biomédica en Red de Fisiopatolgía de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Sandro Muntoni
- Department of Biomedical Sciences, University of Cagliari and Centre for Metabolic Diseases and Atherosclerosis, The ME.DI.CO Association, Cagliari, Italy
| | - Stefano Bertolini
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Cesare Sirtori
- Center E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Universita' degli Studi di Milano, and Dyslipidemia Center, Niguarda Hospital, Milan, Italy
| | - Laura Calabresi
- Center E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Universita' degli Studi di Milano, and Dyslipidemia Center, Niguarda Hospital, Milan, Italy
| | - Chiara Pavanello
- Center E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Universita' degli Studi di Milano, and Dyslipidemia Center, Niguarda Hospital, Milan, Italy
| | - Maurizio Averna
- Dipartimento Biomedico di Medicina Interna e Specialistica, Università di Palermo, Palermo, Italy
| | - Angelo Baldassare Cefalu
- Dipartimento Biomedico di Medicina Interna e Specialistica, Università di Palermo, Palermo, Italy
| | - Davide Noto
- Dipartimento Biomedico di Medicina Interna e Specialistica, Università di Palermo, Palermo, Italy
| | - Adolfo Arturo Pacifico
- Unità Operativa Diabetologia e Malattie Metaboliche, Azienda Ospedaliero Universitaria, Sassari, Italy
| | - Giovanni Mario Pes
- Department of Clinical and Experimental Medicine, University of Sassari, Sassari, Italy
| | - Mariko Harada-Shiba
- Department of Molecular Innovation in Lipidology, National Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | | | | | | | - Anja Vogt
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität (LMU) Klinikum der Universität München, Munich, Germany
| | - Marco Scardapane
- Center for Outcomes Research and Clinical Epidemiology, Coreresearch, Inc., Pescara, Italy
| | - Barbara Sjouke
- Department of Internal and Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Renato Fellin
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Marcello Arca
- Department of Internal Medicine and Clinical Specialties, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
111
|
Shirahama R, Ono T, Nagamatsu S, Sueta D, Takashio S, Chitose T, Fujisue K, Sakamoto K, Yamamoto E, Izumiya Y, Kaikita K, Hokimoto S, Hori M, Harada-Shiba M, Kajiwara I, Ogawa H, Tsujita K. Coronary Artery Plaque Regression by a PCSK9 Antibody and Rosuvastatin in Double-heterozygous Familial Hypercholesterolemia with an LDL Receptor Mutation and a PCSK9 V4I Mutation. Intern Med 2018; 57:3551-3557. [PMID: 30555118 PMCID: PMC6355420 DOI: 10.2169/internalmedicine.1060-18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The low-density lipoprotein-cholesterol (LDL-C) level of a 38-year-old man diagnosed with acute coronary syndrome was 257 mg/dL. The administration of a proprotein convertase subtilisin-kexin type 9 (PCSK9) antibody in addition to rosuvastatin plus ezetimibe was initiated, reducing his LDL-C level to 37 mg/dL. A genetic analysis revealed both an LDL receptor (LDLR) mutation and a PCSK9 V4I mutation. Nine months after revascularization, intravascular ultrasound revealed plaque regression in the coronary arteries. LDLR/PCSK9 mutation carriers are prone to coronary artery disease. Intensive LDL-C lowering by including PCSK9 antibody was associated with coronary plaque regression, suggesting the expectation of prognosis improvement.
Collapse
Affiliation(s)
- Ryo Shirahama
- Department of Cardiovascular Medicine, Arao City Hospital, Japan
| | - Takamichi Ono
- Department of Cardiovascular Medicine, Arao City Hospital, Japan
| | - Suguru Nagamatsu
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Daisuke Sueta
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Seiji Takashio
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Tadasuke Chitose
- Department of Cardiovascular Medicine, Arao City Hospital, Japan
| | - Koichiro Fujisue
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Kenji Sakamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Eiichiro Yamamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Yasuhiro Izumiya
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Koichi Kaikita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Seiji Hokimoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Mika Hori
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, Japan
| | - Mariko Harada-Shiba
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, Japan
| | - Ichiro Kajiwara
- Department of Cardiovascular Medicine, Arao City Hospital, Japan
| | - Hisao Ogawa
- National Cerebral and Cardiovascular Center, Japan
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Japan
| |
Collapse
|
112
|
Genome-Wide Association Studies and Risk Scores for Coronary Artery Disease: Sex Biases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1065:627-642. [PMID: 30051411 DOI: 10.1007/978-3-319-77932-4_38] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Phenotypic sex differences in coronary artery disease (CAD) and its risk factors have been apparent for many decades in basic and clinical research; however, whether these are also present at the gene level and thus influence genome-wide association and genetic risk prediction studies has often been ignored. From fundamental and medical standpoints, this is critically important to assess in order to fully understand the underlying genetic architecture that predisposes to CAD and better predict disease outcomes based on the interaction between genes, sex effects, and environment. In this chapter we aimed to (1) integrate the history and latest research from genome-wide association studies for CAD and clinical and genetic risk scores for prediction of CAD, (2) highlight sex-specific differences in these areas of research, and (3) discuss reasons why sex differences have often not been considered and, where present, why sex differences exist at genetic and phenotypic levels and how important they are for consideration in future research. While we find interesting examples of sex differences in effects of genetic variants on CAD, genome-wide association and genetic risk studies have typically not tested for sex-specific effects despite mounting evidence from diverse fields that these are likely very important to consider at both the genetic and phenotypic levels. In-depth testing for sex effects in large-scale genome-wide association studies that include autosomal and often excluded sex chromosomes alongside parallel improvements in resolution of sex-specific differences for risk factors and disease outcomes for CAD has the potential to substantially improve clinical and genetic risk prediction studies. Developing sex-tailored genetic risk scores as has been done recently for other disorders might be also warranted for CAD. In the era of precision medicine, this level of accuracy is essential for such a common and costly disease.
Collapse
|
113
|
Hegele RA. Learning From Patients With Ultrarare Conditions. J Am Coll Cardiol 2018; 71:289-291. [DOI: 10.1016/j.jacc.2017.09.1158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 09/19/2017] [Indexed: 01/19/2023]
|
114
|
Spina R, Noto D, Barbagallo CM, Monastero R, Ingrassia V, Valenti V, Baschi R, Pipitone A, Giammanco A, La Spada MP, Misiano G, Scrimali C, Cefalù AB, Averna MR. Genetic epidemiology of autosomal recessive hypercholesterolemia in Sicily: Identification by next-generation sequencing of a new kindred. J Clin Lipidol 2018; 12:145-151. [PMID: 29153781 DOI: 10.1016/j.jacl.2017.10.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/18/2017] [Accepted: 10/18/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Autosomal recessive hypercholesterolemia (ARH) is a rare inherited lipid disorder. In Sardinia, differently from other world regions, the mutated allele frequency is high. It is caused by mutations in the low-density lipoprotein receptor adaptor protein 1 gene. Fourteen different mutations have been reported so far; in Sardinia, 2 alleles (ARH1 and ARH2) explain most of the cases. Four ARH patients, all carriers of the ARH1 mutation, have been identified in mainland Italy and 2 in Sicily. OBJECTIVE The objectives of the study were to improve the molecular diagnosis of familial hypercholesterolemia (FH) and to estimate the frequency of the ARH1 allele in 2 free-living Sicilian populations. METHODS We sequenced by targeted next-generation sequencing 20 genes related to low-density lipoprotein metabolism in 50 hypercholesterolemic subjects. Subjects from 2 free-living populations from Northern (Ventimiglia Heart Study, 848 individuals) and Southern Sicily (Zabut Zabùt Aging Project, 1717 individuals) were genotyped for ARH1 allele. RESULTS We identified 1 homozygous carrier of the ARH1 mutation among the 50 hypercholesterolemic outpatients. Population-based genotyping of ARH1 in 2565 subjects allowed the identification of 1 heterozygous carrier. The overall estimated allele frequency of ARH1 in Sicily was 0.0002 (0.02%). CONCLUSIONS The identification of a new case of ARH in Sicily among 50 clinically diagnosed FH highlights the importance of next-generation sequencing analysis as tool to improve the FH diagnosis. Our results also indicate that ARH1 carrier status is present in ∼1:2500 of Sicilian inhabitants, confirming that ARH is extremely rare outside Sardinia.
Collapse
Affiliation(s)
- Rossella Spina
- Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy; Molecular Biology Diagnostic Laboratory, Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), Department of Diagnostic Laboratory, AOUP "Paolo Giaccone", Palermo, Italy
| | - Davide Noto
- Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
| | - Carlo M Barbagallo
- Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
| | - Roberto Monastero
- Department of Experimental Biomedicine and Clinical Neuroscience (BioNeC), University of Palermo, Palermo, Italy
| | - Valeria Ingrassia
- Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy; Molecular Biology Diagnostic Laboratory, Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), Department of Diagnostic Laboratory, AOUP "Paolo Giaccone", Palermo, Italy
| | - Vincenza Valenti
- Molecular Biology Diagnostic Laboratory, Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), Department of Diagnostic Laboratory, AOUP "Paolo Giaccone", Palermo, Italy
| | - Roberta Baschi
- Department of Experimental Biomedicine and Clinical Neuroscience (BioNeC), University of Palermo, Palermo, Italy
| | - Antonina Pipitone
- Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
| | - Antonina Giammanco
- Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
| | - Maria P La Spada
- Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
| | - Gabriella Misiano
- Molecular Biology Diagnostic Laboratory, Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), Department of Diagnostic Laboratory, AOUP "Paolo Giaccone", Palermo, Italy
| | - Chiara Scrimali
- Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
| | - Angelo B Cefalù
- Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy; Molecular Biology Diagnostic Laboratory, Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), Department of Diagnostic Laboratory, AOUP "Paolo Giaccone", Palermo, Italy.
| | - Maurizio R Averna
- Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy; Molecular Biology Diagnostic Laboratory, Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), Department of Diagnostic Laboratory, AOUP "Paolo Giaccone", Palermo, Italy.
| |
Collapse
|
115
|
Scicali R, Di Pino A, Platania R, Purrazzo G, Ferrara V, Giannone A, Urbano F, Filippello A, Rapisarda V, Farruggia E, Piro S, Rabuazzo AM, Purrello F. Detecting familial hypercholesterolemia by serum lipid profile screening in a hospital setting: Clinical, genetic and atherosclerotic burden profile. Nutr Metab Cardiovasc Dis 2018; 28:35-43. [PMID: 28958694 DOI: 10.1016/j.numecd.2017.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 07/04/2017] [Accepted: 07/10/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND AIMS Familial hypercholesterolemia (FH) is underdiagnosed and public cholesterol screening may be useful to find new subjects. In this study, we aim to investigate the prevalence of FH patients in a hospital screening program and evaluate their atherosclerotic burden using intima-media thickness (IMT). METHODS AND RESULTS We screened 1575 lipid profiles and included for genetic analysis adults with a low-density lipoprotein (LDL) cholesterol >190 mg/dL and triglycerides <200 mg/dL and first-degree child relatives with LDL cholesterol >160 mg/dL and triglycerides <200 mg/dL. The diagnosis of FH was presumed by Dutch Lipid Clinic Network (DLCN) criteria and confirmed by the presence of the genetic variant. Mean common carotid intima-media thickness (IMT) was assessed using consensus criteria. After confirming LDL cholesterol value and excluding secondary hypercholesterolemia, 56 subjects with a DLCN ≥4 performed genetic analysis. Of these, 26 had an FH genetic variant. The proportion of patients with a mutation having a DLCN score of 6-8 was 75%; in individuals with a DLCN score >8 it was 100%. Mean IMT was higher in FH patients compared to non FH (0.73 [0.61-0.83] vs 0.71 [0.60-0.75] mm, p < 0.01). Moreover, we detected two mutations not previously described. Finally, simple regression analysis showed a correlation of IMT with LDL cholesterol >190 mg/dL and corneal arcus (p < 0.01 and p < 0.001, respectively). CONCLUSIONS A hospital screening was useful to detect FH subjects with increased atherosclerosis. Also, next-generation sequencing was able to detect new FH mutations.
Collapse
Affiliation(s)
- R Scicali
- Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - A Di Pino
- Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - R Platania
- Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - G Purrazzo
- Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - V Ferrara
- Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - A Giannone
- Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - F Urbano
- Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - A Filippello
- Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - V Rapisarda
- Department of Clinical and Experimental Medicine, Section of Occupational Medicine, University of Catania, Italy
| | - E Farruggia
- Occupational Medicine Division, Garibaldi Hospital of Catania, Catania, Italy
| | - S Piro
- Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - A M Rabuazzo
- Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - F Purrello
- Department of Clinical and Experimental Medicine, University of Catania, Italy.
| |
Collapse
|
116
|
Pek SLT, Dissanayake S, Fong JCW, Lin MX, Chan EZL, Tang JIS, Lee CW, Ong HY, Sum CF, Lim SC, Tavintharan S. Spectrum of mutations in index patients with familial hypercholesterolemia in Singapore: Single center study. Atherosclerosis 2017; 269:106-116. [PMID: 29353225 DOI: 10.1016/j.atherosclerosis.2017.12.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 12/13/2017] [Accepted: 12/21/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Familial hypercholesterolemia (FH) is an autosomal dominant genetic disease characterized by the presence of high plasma low density lipoproteins cholesterol (LDL-c). Patients with FH, with mutation detected, are at increased risk of premature cardiovascular disease compared to those without mutations. The aim of the study was to assess the type of mutations in patients, clinically diagnosed with FH in Singapore. METHODS Patients (probands) with untreated/highest on-treatment LDL-c>4.9 mmol/l were recruited (June 2015 to April 2017). Anthropometric, biochemical indices, blood and family history were collected. DNA was extracted and Next Generation Sequencing (NGS) was performed in 26 lipid-related genes, including LDLR, APOB and PCSK9, and validated using Sanger. Multiplex-ligation probe analyses for LDLR were performed to identify large mutation derangements. Based on HGVS nomenclature, LDLR mutations were classified as "Null"(nonsense, frameshift, large rearrangements) and "Defective"(point mutations which are pathogenic). RESULTS Ninety-six probands were recruited: mean age: (33.5 ± 13.6) years. 52.1% (n = 50) of patients had LDLR mutations, with 15 novel mutations, and 4.2% (n = 4) had APOB mutations. Total cholesterol (TC) and LDL-c were significantly higher in those with LDLR mutations compared to APOB and no mutations [(8.53 ± 1.52) vs. (6.93 ± 0.47) vs. (7.80 ± 1.32)] mmol/l, p = 0.012 and [(6.74 ± 0.35) vs. (5.29 ± 0.76) vs. (5.98 ± 1.23)] mmol/l, p=0.005, respectively. Patients with "null LDLR" mutations (n = 13) had higher TC and LDL-c than "defective LDLR" mutations (n = 35): [(9.21 ± 1.60) vs. (8.33 ± 1.41)]mmol/l, p = 0.034 and [(7.43 ± 1.47) vs. (6.53 ± 1.21)]mmol/l, p=0.017, respectively. CONCLUSIONS To our knowledge, this is the first report of mutation detection in patients with clinically suspected FH by NGS in Singapore. While percentage of mutations is similar to other countries, the spectrum locally differs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Chee Wan Lee
- Cardiology, Khoo Teck Puat Hospital, 768828, Singapore
| | - Hean Yee Ong
- Cardiology, Khoo Teck Puat Hospital, 768828, Singapore
| | - Chee Fang Sum
- Diabetes Centre, Admiralty Medical Centre, 730676, Singapore; Division of Endocrinology, Khoo Teck Puat Hospital, 768828, Singapore
| | - Su Chi Lim
- Clinical Research Unit, Khoo Teck Puat Hospital, 768828, Singapore; Diabetes Centre, Admiralty Medical Centre, 730676, Singapore; Division of Endocrinology, Khoo Teck Puat Hospital, 768828, Singapore; Saw Swee Hock School of Public Health, National University Hospital, 117549, Singapore
| | - Subramaniam Tavintharan
- Clinical Research Unit, Khoo Teck Puat Hospital, 768828, Singapore; Diabetes Centre, Admiralty Medical Centre, 730676, Singapore; Division of Endocrinology, Khoo Teck Puat Hospital, 768828, Singapore.
| |
Collapse
|
117
|
A novel indel variant in LDLR responsible for familial hypercholesterolemia in a Chinese family. PLoS One 2017; 12:e0189316. [PMID: 29228028 PMCID: PMC5724832 DOI: 10.1371/journal.pone.0189316] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/22/2017] [Indexed: 01/10/2023] Open
Abstract
Familial hypercholesterolemia (FH) is an inherited disorder characterized by elevation of serum cholesterol bound to low-density lipoprotein. Mutations in LDLR are the major factors responsible for FH. In this study, we recruited a four-generation Chinese family with FH and identified the clinical features of hypercholesterolemia. All affected individuals shared a novel indel mutation (c.1885_1889delinsGATCATCAACC) in exon 13 of LDLR. The mutation segregated with the hypercholesterolemia phenotype in the family. To analyze the function of the indel, we established stable clones of mutant and wild-type LDLR in Hep G2 cells. The mutant LDLR was retained in the endoplasmic reticulum (ER) and failed to glycosylate via the Golgi. Moreover, the membrane LDLR was reduced and lost the ability to take up LDL. Our data also expand the spectrum of known LDLR mutations.
Collapse
|
118
|
van Schie MC, Jainandunsing S, van Lennep JER. Monogenetic disorders of the cholesterol metabolism and premature cardiovascular disease. Eur J Pharmacol 2017; 816:146-153. [DOI: 10.1016/j.ejphar.2017.09.046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 09/05/2017] [Accepted: 09/28/2017] [Indexed: 12/13/2022]
|
119
|
Morad FA, Rashidi OM, Sadath SS, Al-Allaf FA, Athar M, Alama MN, Edris SE, Bondagji NS, Shaik NA, Banaganapalli B, Awan Z. In Silico Approach to Investigate the Structural and Functional Attributes of Familial Hypercholesterolemia Variants Reported in the Saudi Population. J Comput Biol 2017; 25:170-181. [PMID: 29172679 DOI: 10.1089/cmb.2017.0018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Familial hypercholesterolemia (FH) is a metabolic disorder that leads primarily to premature cardiovascular diseases, the main cause of mortality in Saudi Arabia (SA). FH is underreported and underdiagnosed in SA with statistical evidence of high expected prevalence in such a consanguineous community. Lacking knowledge of which and how these alterations are actually impacting lipid metabolism is one of the main reasons why FH is insufficiently diagnosed in the region. The aim of this study was to develop a fast prediction approach using an integrated bioinformatics method for future screening of the potential causative variants from national registries. A total of 21 variants were detected with majority rate in LDLR (81%). Variants were classified based on the type of mutation. Missense variants resulting in amino acid changes, c.1429G>A (p.D477N), c.1474G>A (p.D492N), c.1731G>T (p.W577C), and c.1783C>T (p.R595W) in LDLR gene, in addition to c.9835A>G (p.S3279G) in APOB, were shown to be deleterious by concordant analysis. Furthermore, functional interaction deformities showed a significant loss and gain of energies in the mutated proteins. These findings will help in distinguishing the most harmful mutations needed to be screened for clinically diagnosed FH patients in SA. Such computational research is necessary to avoid time consumption and the usage of expensive biological experiments. This can be a fast track to facilitate the future filtering and screening of causative mutations from national registries.
Collapse
Affiliation(s)
- Fatima A Morad
- 1 Department of Biology, Genomic and Biotechnology Section, Faculty of Science, King Abdulaziz University , Jeddah, Kingdom of Saudi Arabia
| | - Omran M Rashidi
- 2 Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University , Jeddah, Kingdom of Saudi Arabia
| | - Saida S Sadath
- 3 Department of Biochemistry, Faculty of Science, King Abdulaziz University , Jeddah, Kingdom of Saudi Arabia
| | - Faisal A Al-Allaf
- 4 Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University , Makkah, Kingdom of Saudi Arabia .,5 Department of Science and Technology, Umm Al-Qura University , Makkah, Kingdom of Saudi Arabia
| | - Mohammad Athar
- 4 Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University , Makkah, Kingdom of Saudi Arabia .,5 Department of Science and Technology, Umm Al-Qura University , Makkah, Kingdom of Saudi Arabia
| | - Mohamed N Alama
- 6 Adult Interventional Cardiology, Cardiology Unit, King Abdulaziz University Hospital (KAUH) , Jeddah, Kingdom of Saudi Arabia
| | - Sherif E Edris
- 1 Department of Biology, Genomic and Biotechnology Section, Faculty of Science, King Abdulaziz University , Jeddah, Kingdom of Saudi Arabia
| | - Nabeel S Bondagji
- 7 Obstetrics and Gynecology Unit, King Abdul Aziz University Hospital , Jeddah, Kingdom of Saudi Arabia
| | - Noor A Shaik
- 8 Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD) , Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Babajan Banaganapalli
- 8 Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD) , Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Zuhier Awan
- 2 Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University , Jeddah, Kingdom of Saudi Arabia
| |
Collapse
|
120
|
Bertolini S, Pisciotta L, Fasano T, Rabacchi C, Calandra S. The study of familial hypercholesterolemia in Italy: A narrative review. ATHEROSCLEROSIS SUPP 2017; 29:1-10. [DOI: 10.1016/j.atherosclerosissup.2017.07.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
121
|
Rashidi OM, H Nazar FA, Alama MN, Awan ZA. Interpreting the Mechanism of APOE (p.Leu167del) Mutation in the Incidence of Familial Hypercholesterolemia; An In-silico Approach. Open Cardiovasc Med J 2017; 11:84-93. [PMID: 29204218 PMCID: PMC5688386 DOI: 10.2174/1874192401711010084] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 06/30/2017] [Accepted: 07/08/2017] [Indexed: 11/22/2022] Open
Abstract
Background: Apolipoprotein E (APOE) gene is a ligand protein in humans which mediates the metabolism of cholesterol by binding to the low-density lipoprotein receptor (LDLR). P.Leu167del mutation in APOE gene was recently connected with Familial Hypercholesterolemia, a condition associated with premature cardiovascular disease. The consequences of this mutation on the protein structure and its receptor binding capacity remain largely unknown. Objective: The current study aims to further decipher the underlying mechanism of this mutation using advanced software-based algorithms. The consequences of disrupting the leucine zipper by this mutation was studied at the structural and functional level of the APOE protein. Methods: 3D protein modeling for both APOE and LDLR (wild types), along with APOE (p.Leu167del) mutant type were generated using homology modeling template-based alignment. Structural deviation analysis was performed to evaluate the spatial orientation and the stability of the mutant APOE structure. Molecular docking analysis simulating APOE-LDLR protein interaction was carried out, in order to evaluate the impact of the mutation on the binding affinity. Result: Structural deviation analysis for APOE mutated model showed low degree of deviance scoring root-mean-square deviation, (RMSD) = 0.322 Å. Whereas Docking simulation revealed an enhanced molecular interaction towards the LDLR with an estimation of +171.03 kJ/mol difference in binding free energy. Conclusion: This in-silico study suggests that p.Leu167del is causing the protein APOE to associate strongly with its receptor, LDLR. This gain-of-function is likely hindering the ability of LDLR to be effectively recycled back to the surface of the hepatocytes to clear cholesterol from the circulation therefore leading to FH.
Collapse
Affiliation(s)
- Omran Mohammed Rashidi
- Department of Clinical Biochemistry. Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fatima Amanullah H Nazar
- Department of Biology, Genomic and Biotechnology Section. Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohamed Nabil Alama
- Adult interventional cardiology, Cardiology unit, King Abdulaziz University Hospital (KAUH), Jeddah, Saudi Arabia
| | - Zuhier Ahmed Awan
- Department of Clinical Biochemistry. Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
122
|
Alallaf F, H.Nazar FA, Alnefaie M, Almaymuni A, Rashidi OM, Alhabib K, Alnouri F, Alama MN, Athar M, Awan Z. The Spectrum of Familial Hypercholesterolemia (FH) in Saudi Arabia: Prime Time for Patient FH Registry. Open Cardiovasc Med J 2017; 11:66-75. [PMID: 28868092 PMCID: PMC5564019 DOI: 10.2174/1874192401711010066] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 04/27/2017] [Accepted: 05/17/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Familial hypercholesterolemia (FH) is a life-threatening inherited condition. Untreated patients have the risk to develop raised plasma levels of cholesterol, atherosclerosis and cardiovascular disease (CVD). If diagnosed and treated early in life, the pathological consequences due to atherosclerosis could be avoided and patients with FH can have an anticipated normal life. Mounting evidence suggests that FH is underdiagnosed and undertreated in all populations. The underlying molecular basis of FH is the presence of mutations in one or more genes in the low-density lipoprotein receptor (LDLR), apolipoprotein B (APOB) or proprotein convertase subtilisin/kexin 9 (PCSK9). However, their prevalence is largely unknown in Saudi Arabia but given the high rates of consanguinity, the prevalence appears to be higher. Furthermore, the high prevalence of obesity and diabetes mellitus in Saudi Arabia increases the vascular disease burden in FH cases by adding additional CVD risk factors. OBJECTIVE This article explores the spectrum of FH-causing mutations in the highly consanguineous Saudi community, the need for establishing the Saudi FH registry, the challenges in creating gene databases, and cascade screening. CONCLUSION The establishment of FH registry and genetic testing should raise awareness not only among healthcare professionals, but the general population as well. It also helps to provide the best treatment regimen in a cost effective manner to this under-recognised population of FH patients.
Collapse
Affiliation(s)
- Faisal Alallaf
- Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University, Mekkah. Saudi Arabia
| | - Fatima Amanullah H.Nazar
- Department of Biology, Genomic and Biotechnology Section, Faculty of Science, King Abdulaziz University, Jeddah. Saudi Arabia
| | - Majed Alnefaie
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah. Saudi Arabia
| | - Adel Almaymuni
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah. Saudi Arabia
| | - Omran Mohammed Rashidi
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah. Saudi Arabia
| | - Khalid Alhabib
- Interventional Cardiology, King Fahad Cardiac Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Fahad Alnouri
- Cardiovascular Prevention and Rehabilitation Unit, Prince Sultan Cardiac Centre, Riyadh, Saudi Arabia
| | - Mohamed-Nabil Alama
- Adult interventional cardiology, Cardiology unit, King Abdulaziz University Hospital (KAUH), Jeddah, Saudi Arabia
| | - Mohammad Athar
- Department of Science and Technology, Umm Al-Qura University, Mekkah, Saudi Arabia
| | - Zuhier Awan
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah. Saudi Arabia
| |
Collapse
|
123
|
Lasam G, Shambhu S, Fishberg R. Heterozygous Familial Hypercholesterolemia With APOE Haplotype: A Prospective Harbinger of a Catastrophic Cardiovascular Event. Cardiol Res 2017; 8:117-122. [PMID: 28725328 PMCID: PMC5505295 DOI: 10.14740/cr548w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/22/2017] [Indexed: 12/12/2022] Open
Abstract
We report a very young man with heterozygous familial hypercholesterolemia (FH) with APOE haplotype and a significant cardiac family history who underwent cardiac catheterization for intermittent episodes of exertional dyspnea and was noted to have a severe triple vessel coronary artery disease (CAD). He underwent coronary artery bypass graft (CABG) surgery which was uneventful. He was discharged on antiplatelet, beta blocker, nitrate, and statin. On routine health maintenance evaluation, he had no cardiac complaints and had been tolerating well his activities of daily living.
Collapse
Affiliation(s)
- Glenmore Lasam
- Department of Medicine, Atlantic Health System-Overlook Medical Center, Summit, NJ 07901 USA
| | - Siddesh Shambhu
- Department of Medicine, Atlantic Health System-Overlook Medical Center, Summit, NJ 07901 USA
| | - Robert Fishberg
- Section of Cardiology, Atlantic Health System-Overlook Medical Center, Summit, NJ 07901 USA
| |
Collapse
|
124
|
Buonuomo PS, Iughetti L, Pisciotta L, Rabacchi C, Papadia F, Bruzzi P, Tummolo A, Bartuli A, Cortese C, Bertolini S, Calandra S. Timely diagnosis of sitosterolemia by next generation sequencing in two children with severe hypercholesterolemia. Atherosclerosis 2017; 262:71-77. [DOI: 10.1016/j.atherosclerosis.2017.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 04/14/2017] [Accepted: 05/03/2017] [Indexed: 01/27/2023]
|
125
|
Khera AV, Kathiresan S. Genetics of coronary artery disease: discovery, biology and clinical translation. Nat Rev Genet 2017; 18:331-344. [PMID: 28286336 PMCID: PMC5935119 DOI: 10.1038/nrg.2016.160] [Citation(s) in RCA: 432] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Coronary artery disease is the leading global cause of mortality. Long recognized to be heritable, recent advances have started to unravel the genetic architecture of the disease. Common variant association studies have linked approximately 60 genetic loci to coronary risk. Large-scale gene sequencing efforts and functional studies have facilitated a better understanding of causal risk factors, elucidated underlying biology and informed the development of new therapeutics. Moving forwards, genetic testing could enable precision medicine approaches by identifying subgroups of patients at increased risk of coronary artery disease or those with a specific driving pathophysiology in whom a therapeutic or preventive approach would be most useful.
Collapse
Affiliation(s)
- Amit V Khera
- Division of Cardiology, Department of Medicine and Center for Genomic Medicine, Massachusetts General Hospital; Cardiovascular Disease Initiative, Broad Institute of Harvard and Massachusetts Institute of Technology, 185 Cambridge Street, CPZN 5.252, Boston, Massachusetts 02114, USA
| | - Sekar Kathiresan
- Division of Cardiology, Department of Medicine and Center for Genomic Medicine, Massachusetts General Hospital; Cardiovascular Disease Initiative, Broad Institute of Harvard and Massachusetts Institute of Technology, 185 Cambridge Street, CPZN 5.252, Boston, Massachusetts 02114, USA
| |
Collapse
|
126
|
März W, Grammer TB, Delgado G, Kleber ME. Angeborene Störungen im Lipoproteinstoffwechsel. Herz 2017; 42:449-458. [DOI: 10.1007/s00059-017-4578-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
127
|
|
128
|
Futema M, Cooper JA, Charakida M, Boustred C, Sattar N, Deanfield J, Lawlor DA, Timpson NJ, Humphries SE, Hingorani AD. Screening for familial hypercholesterolaemia in childhood: Avon Longitudinal Study of Parents and Children (ALSPAC). Atherosclerosis 2017; 260:47-55. [PMID: 28349888 PMCID: PMC5414415 DOI: 10.1016/j.atherosclerosis.2017.03.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 02/18/2017] [Accepted: 03/05/2017] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND AIMS Familial hypercholesterolaemia (FH) is an autosomal-dominant disease with frequency of 1/500 to 1/250 that leads to premature coronary heart disease. New approaches to identify FH mutation-carriers early are needed to prevent premature cardiac deaths. In a cross-sectional study of the Avon Longitudinal Study of Parents and Children (ALSPAC), we evaluated the biochemical thresholds for FH screening in childhood, and modelled a two-stage biochemical and sequencing screening strategy for FH detection. METHODS From 5083 ALSPAC children with cholesterol measurement at age nine years, FH genetic diagnosis was performed in 1512 individuals, using whole-genome or targeted sequencing of known FH-causing genes. Detection rate (DR) and false-positive rate (FPR) for proposed screening thresholds (total-cholesterol > 1.53, or LDL-C > 1.84 multiples of the median (MoM)) were assessed. RESULTS Six of 1512 sequenced individuals had an FH-causing mutation of whom five had LDL-C > 1.84 MoM, giving a verification-bias corrected DR of 62.5% (95% CI: 25-92), with a FPR of 0.2% (95% CI: 0.1-0.4). The DR for the TC cut-point of 1.53 MoM was 25% (95% CI: 3.2-65.1) with a FPR of 0.4% (95% CI: 0.2-0.6). We estimated 13 of an expected 20 FH mutation carriers (and 13 of the 20 parental carriers) could be detected for every 10,000 children screened, with false-positives reliably excluded by addition of a next generation sequencing step in biochemical screen-positive samples. CONCLUSIONS Proposed cholesterol thresholds for childhood FH screening were less accurate than previously estimated. A sequential strategy of biochemical screening followed by targeted sequencing of FH genes in screen-positive children may help mitigate the higher than previously estimated FPR and reduce wasted screening of unaffected parents.
Collapse
Affiliation(s)
- Marta Futema
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, London, UK
| | - Jackie A Cooper
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, London, UK
| | - Marietta Charakida
- National Centre for Cardiovascular Prevention and Outcomes, Institute of Cardiovascular Science, University College London (UCL), London, UK
| | - Christopher Boustred
- North East Thames Regional Genetics Service, Great Ormond Street Hospital for Children, London, UK
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow, Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - John Deanfield
- National Centre for Cardiovascular Prevention and Outcomes, Institute of Cardiovascular Science, University College London (UCL), London, UK
| | - Debbie A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK; School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK; School of Social and Community Medicine, University of Bristol, Bristol, UK
| | | | - Steve E Humphries
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, London, UK; UCL Genetics Institute, Department of Genetics, Environment and Evolution, University College London, London, UK
| | - Aroon D Hingorani
- Genetic Epidemiology Group, Institute of Cardiovascular Science, Farr Institute for Health Informatics, University College London, London, UK.
| |
Collapse
|
129
|
Schmidt AF, Pearce LS, Wilkins JT, Overington JP, Hingorani AD, Casas JP. PCSK9 monoclonal antibodies for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2017; 4:CD011748. [PMID: 28453187 PMCID: PMC6478267 DOI: 10.1002/14651858.cd011748.pub2] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Despite the availability of effective drug therapies that reduce low-density lipoprotein (LDL)-cholesterol (LDL-C), cardiovascular disease (CVD) remains an important cause of mortality and morbidity. Therefore, additional LDL-C reduction may be warranted, especially for patients who are unresponsive to, or unable to take, existing LDL-C-reducing therapies. By inhibiting the proprotein convertase subtilisin/kexin type 9 (PCSK9) enzyme, monoclonal antibodies (PCSK9 inhibitors) may further reduce LDL-C, potentially reducing CVD risk as well. OBJECTIVES Primary To quantify short-term (24 weeks), medium-term (one year), and long-term (five years) effects of PCSK9 inhibitors on lipid parameters and on the incidence of CVD. Secondary To quantify the safety of PCSK9 inhibitors, with specific focus on the incidence of type 2 diabetes, cognitive function, and cancer. Additionally, to determine if specific patient subgroups were more or less likely to benefit from the use of PCSK9 inhibitors. SEARCH METHODS We identified studies by systematically searching the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, and Web of Science. We also searched Clinicaltrials.gov and the International Clinical Trials Registry Platform and screened the reference lists of included studies. We identified the studies included in this review through electronic literature searches conducted up to May 2016, and added three large trials published in March 2017. SELECTION CRITERIA All parallel-group and factorial randomised controlled trials (RCTs) with a follow-up time of at least 24 weeks were eligible. DATA COLLECTION AND ANALYSIS Two review authors independently reviewed and extracted data. When data were available, we calculated pooled effect estimates. MAIN RESULTS We included 20 studies with data on 67,237 participants (median age 61 years; range 52 to 64 years). Twelve trials randomised participants to alirocumab, three trials to bococizumab, one to RG7652, and four to evolocumab. Owing to the small number of trials using agents other than alirocumab, we did not differentiate between types of PCSK9 inhibitors used. We compared PCSK9 inhibitors with placebo (thirteen RCTs), ezetimibe (two RCTs) or ezetimibe and statins (five RCTs).Compared with placebo, PCSK9 inhibitors decreased LDL-C by 53.86% (95% confidence interval (CI) 58.64 to 49.08; eight studies; 4782 participants; GRADE: moderate) at 24 weeks; compared with ezetimibe, PCSK9 inhibitors decreased LDL-C by 30.20% (95% CI 34.18 to 26.23; two studies; 823 participants; GRADE: moderate), and compared with ezetimibe and statins, PCSK9 inhibitors decreased LDL-C by 39.20% (95% CI 56.15 to 22.26; five studies; 5376 participants; GRADE: moderate).Compared with placebo, PCSK9 inhibitors decreased the risk of CVD events, with a risk difference (RD) of 0.91% (odds ratio (OR) of 0.86, 95% CI 0.80 to 0.92; eight studies; 59,294 participants; GRADE: moderate). Compared with ezetimibe and statins, PCSK9 inhibitors appeared to have a stronger protective effect on CVD risk, although with considerable uncertainty (RD 1.06%, OR 0.45, 95% CI 0.27 to 0.75; three studies; 4770 participants; GRADE: very low). No data were available for the ezetimibe only comparison. Compared with placebo, PCSK9 probably had little or no effect on mortality (RD 0.03%, OR 1.02, 95% CI 0.91 to 1.14; 12 studies; 60,684 participants; GRADE: moderate). Compared with placebo, PCSK9 inhibitors increased the risk of any adverse events (RD 1.54%, OR 1.08, 95% CI 1.04 to 1.12; 13 studies; 54,204 participants; GRADE: low). Similar effects were observed for the comparison of ezetimibe and statins: RD 3.70%, OR 1.18, 95% CI 1.05 to 1.34; four studies; 5376 participants; GRADE: low. Clinical event data were unavailable for the ezetimibe only comparison. AUTHORS' CONCLUSIONS Over short-term to medium-term follow-up, PCSK9 inhibitors reduced LDL-C. Studies with medium-term follow-up time (longest median follow-up recorded was 26 months) reported that PCSK9 inhibitors (compared with placebo) decreased CVD risk but may have increased the risk of any adverse events (driven by SPIRE-1 and -2 trials). Available evidence suggests that PCSK9 inhibitor use probably leads to little or no difference in mortality. Evidence on relative efficacy and safety when PCSK9 inhibitors were compared with active treatments was of low to very low quality (GRADE); follow-up times were short and events were few. Large trials with longer follow-up are needed to evaluate PCSK9 inhibitors versus active treatments as well as placebo. Owing to the predominant inclusion of high-risk patients in these studies, applicability of results to primary prevention is limited. Finally, estimated risk differences indicate that PCSK9 inhibitors only modestly change absolute risks (often to less than 1%).
Collapse
Affiliation(s)
- Amand F Schmidt
- University College LondonInstitute of Cardiovascular Science222 Euston Road, Room 206LondonUKNW1 2DA
| | - Lucy S Pearce
- London School of Hygiene & Tropical MedicineDepartment of Non‐communicable Disease EpidemiologyKeppel StreetLondonUKWC1 E7HT
| | - John T Wilkins
- Northwestern University Feinberg School of MedicineThe Department of Medicine (Cardiology) and the Department of Preventive MedicineSuite 1400 680 N. Lakeshore DriveChicagoUSA60611
| | | | - Aroon D Hingorani
- University College LondonInstitute of Cardiovascular Science222 Euston Road, Room 206LondonUKNW1 2DA
| | - Juan P Casas
- University College LondonFarr Institute of Health Informatics Research222 Euston RoadLondonUKNW1 2DA
| |
Collapse
|
130
|
Abstract
Despite thousands of genetic loci identified to date, a large proportion of genetic variation predisposing to complex disease and traits remains unaccounted for. Advances in sequencing technology enable focused explorations on the contribution of low-frequency and rare variants to human traits. Here we review experimental approaches and current knowledge on the contribution of these genetic variants in complex disease and discuss challenges and opportunities for personalised medicine.
Collapse
Affiliation(s)
- Lorenzo Bomba
- Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, CB10 1HH, UK
| | - Klaudia Walter
- Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, CB10 1HH, UK
| | - Nicole Soranzo
- Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, CB10 1HH, UK. .,Department of Haematology, University of Cambridge, Hills Rd, Cambridge, CB2 0AH, UK. .,The National Institute for Health Research Blood and Transplant Unit (NIHR BTRU) in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Strangeways Research Laboratory, Wort's Causeway, Cambridge, CB1 8RN, UK.
| |
Collapse
|
131
|
Elsafadi M, Manikandan M, Alajez NM, Hamam R, Dawud RA, Aldahmash A, Iqbal Z, Alfayez M, Kassem M, Mahmood A. MicroRNA-4739 regulates osteogenic and adipocytic differentiation of immortalized human bone marrow stromal cells via targeting LRP3. Stem Cell Res 2017; 20:94-104. [PMID: 28340487 DOI: 10.1016/j.scr.2017.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 02/25/2017] [Accepted: 03/01/2017] [Indexed: 12/16/2022] Open
Abstract
Understanding the regulatory networks underlying lineage differentiation and fate determination of human bone marrow stromal cells (hBMSC) is a prerequisite for their therapeutic use. The goal of the current study was to unravel the novel role of the low-density lipoprotein receptor-related protein 3 (LRP3) in regulating the osteogenic and adipogenic differentiation of immortalized hBMSCs. Gene expression profiling revealed significantly higher LRP3 levels in the highly osteogenic hBMSC clone imCL1 than in the less osteogenic clone imCL2, as well as a significant upregulation of LRP3 during the osteogenic induction of the imCL1 clone. Data from functional and gene expression assays demonstrated the role of LRP3 as a molecular switch promoting hBMSC lineage differentiation into osteoblasts and inhibiting differentiation into adipocytes. Interestingly, microRNA (miRNA) expression profiling identified miR-4739 as the most under-represented miRNA (-36.11 fold) in imCL1 compared to imCL2. The TargetScan prediction algorithm, combined with functional and biochemical assays, identified LRP3 mRNA as a novel target of miR-4739, with a single potential binding site for miR-4739 located in the LRP3 3' UTR. Regulation of LRP3 expression by miR-4739 was subsequently confirmed by qRT-PCR, western blotting, and luciferase assays. Over-expression of miR-4739 mimicked the effects of LRP3 knockdown on promoting adipogenic and suppressing osteogenic differentiation of hBMSCs. Hence, we report for the first time a novel biological role for the LRP3/hsa-miR-4739 axis in balancing osteogenic and adipocytic differentiation of hBMSCs. Our data support the potential utilization of miRNA-based therapies in regenerative medicine.
Collapse
Affiliation(s)
- Mona Elsafadi
- Stem Cell Unit, Department of Anatomy, College of Medicine,King Saud University, Riyadh 11461, Saudi Arabia; KMEB, Department of Endocrinology, University Hospital of Odense, University of Southern Denmark, Winslowsparken 25.1, DK-5000 Odense C, Denmark.
| | - Muthurangan Manikandan
- Stem Cell Unit, Department of Anatomy, College of Medicine,King Saud University, Riyadh 11461, Saudi Arabia
| | - Nehad M Alajez
- Stem Cell Unit, Department of Anatomy, College of Medicine,King Saud University, Riyadh 11461, Saudi Arabia.
| | - Rimi Hamam
- Stem Cell Unit, Department of Anatomy, College of Medicine,King Saud University, Riyadh 11461, Saudi Arabia
| | - Raed Abu Dawud
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 12713, Saudi Arabia
| | - Abdullah Aldahmash
- Stem Cell Unit, Department of Anatomy, College of Medicine,King Saud University, Riyadh 11461, Saudi Arabia; Prince Naif Health Research Center, King Saud University, Riyadh 11461, Saudi Arabia.
| | - Zafar Iqbal
- Department of Basic Sciences, College of applied medical sciences, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), National Guard Health Affairs, Al Ahsa, Saudi Arabia
| | - Musaad Alfayez
- Stem Cell Unit, Department of Anatomy, College of Medicine,King Saud University, Riyadh 11461, Saudi Arabia.
| | - Moustapha Kassem
- Stem Cell Unit, Department of Anatomy, College of Medicine,King Saud University, Riyadh 11461, Saudi Arabia; KMEB, Department of Endocrinology, University Hospital of Odense, University of Southern Denmark, Winslowsparken 25.1, DK-5000 Odense C, Denmark.
| | - Amer Mahmood
- Stem Cell Unit, Department of Anatomy, College of Medicine,King Saud University, Riyadh 11461, Saudi Arabia.
| |
Collapse
|
132
|
Gabunia K, Herman AB, Ray M, Kelemen SE, England RN, DeLa Cadena R, Foster WJ, Elliott KJ, Eguchi S, Autieri MV. Induction of MiR133a expression by IL-19 targets LDLRAP1 and reduces oxLDL uptake in VSMC. J Mol Cell Cardiol 2017; 105:38-48. [PMID: 28257760 DOI: 10.1016/j.yjmcc.2017.02.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/21/2017] [Accepted: 02/27/2017] [Indexed: 10/20/2022]
Abstract
The transformation of vascular smooth muscle cells [VSMC] into foam cells leading to increased plaque size and decreased stability is a key, yet understudied step in atherogenesis. We reported that Interleukin-19 (IL-19), a novel, anti-inflammatory cytokine, attenuates atherosclerosis by anti-inflammatory effects on VSMC. In this work we report that IL-19 induces expression of miR133a, a muscle-specific miRNA, in VSMC. Although previously unreported, we report that miR133a can target and reduce mRNA abundance, mRNA stability, and protein expression of Low Density Lipoprotein Receptor Adaptor Protein 1, (LDLRAP1), an adaptor protein which functions to internalize the LDL receptor. Mutations in this gene lead to LDL receptor malfunction and cause the Autosomal Recessive Hypercholesterolemia (ARH) disorder in humans. Herein we show that IL-19 reduces lipid accumulation in VSMC, and LDLRAP1 expression and oxLDL uptake in a miR133a-dependent mechanism. We show that LDLRAP1 is expressed in plaque and neointimal VSMC of mouse and human injured arteries. Transfection of miR133a and LDLRAP1 siRNA into VSMC reduces their proliferation and uptake of oxLDL. miR133a is significantly increased in plasma from hyperlipidemic compared with normolipidemic patients. Expression of miR133a in IL-19 stimulated VSMC represents a previously unrecognized link between vascular lipid metabolism and inflammation, and may represent a therapeutic opportunity to combat vascular inflammatory diseases.
Collapse
Affiliation(s)
- Khatuna Gabunia
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Allison B Herman
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Mitali Ray
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Sheri E Kelemen
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Ross N England
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Raul DeLa Cadena
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - William J Foster
- Departments of Ophthalmology & Bioengineering, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Katherine J Elliott
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Satoru Eguchi
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Michael V Autieri
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States.
| |
Collapse
|
133
|
Durst R, Ibe UK, Shpitzen S, Schurr D, Eliav O, Futema M, Whittall R, Szalat A, Meiner V, Knobler H, Gavish D, Henkin Y, Ellis A, Rubinstein A, Harats D, Bitzur R, Hershkovitz B, Humphries SE, Leitersdorf E. Molecular genetics of familial hypercholesterolemia in Israel–revisited. Atherosclerosis 2017; 257:55-63. [DOI: 10.1016/j.atherosclerosis.2016.12.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 12/10/2016] [Accepted: 12/16/2016] [Indexed: 10/20/2022]
|
134
|
Fahy EF, McCarthy E, Steinhagen-Thiessen E, Vaughan CJ. A case of autosomal recessive hypercholesterolemia responsive to proprotein convertase subtilisin/kexin 9 inhibition. J Clin Lipidol 2017; 11:287-288. [DOI: 10.1016/j.jacl.2016.10.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 10/03/2016] [Accepted: 10/03/2016] [Indexed: 11/15/2022]
|
135
|
Seidah NG, Abifadel M, Prost S, Boileau C, Prat A. The Proprotein Convertases in Hypercholesterolemia and Cardiovascular Diseases: Emphasis on Proprotein Convertase Subtilisin/Kexin 9. Pharmacol Rev 2017; 69:33-52. [PMID: 27920219 DOI: 10.1124/pr.116.012989] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
The secretory proprotein convertase (PC) family comprises nine members, as follows: PC1/3, PC2, furin, PC4, PC5/6, paired basic amino acid cleaving enzyme 4, PC7, subtilisin kexin isozyme 1/site 1 protease (SKI-1/S1P), and PC subtilisin/kexin type 9 (PCSK9). The first seven PCs cleave their substrates at single/paired basic residues and exhibit specific and often essential functions during development and/or in adulthood. The essential SKI-1/S1P cleaves membrane-bound transcription factors at nonbasic residues. In contrast, PCSK9 cleaves itself once, and the secreted inactive protease drags the low-density lipoprotein receptors (LDLR) and very LDLR (VLDLR) to endosomal/lysosomal degradation. Inhibitory PCSK9 monoclonal antibodies are now prescribed to treat hypercholesterolemia. This review focuses on the implication of PCs in cardiovascular functions and diseases, with a major emphasis on PCSK9. We present a phylogeny of the PCs and the analysis of PCSK9 haplotypes in modern and archaic human species. The absence of PCSK9 in mice led to the discovery of a sex- and tissue-specific subcellular distribution of the LDLR and VLDLR. PCSK9 inhibition may have other applications because it reduces inflammation and sepsis in a LDLR-dependent manner. Our present understanding of the cellular mechanism(s) that enables PCSK9 to induce the degradation of receptors is reviewed, as well as the consequences of its key natural mutations. The PCSK9 ongoing clinical trials are reviewed. Finally, how the other PCs may impact cardiovascular disease and the metabolic syndrome, and become relevant targets, is discussed.
Collapse
Affiliation(s)
- Nabil G Seidah
- Laboratory of Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal, affiliated to Université de Montréal, QC, Canada (N.G.S., A.P.); LVTS, INSERM U1148, Hôpital Xavier-Bichat, Paris, France (M.A., C.B.); Laboratory of Biochemistry and Molecular Therapeutics, Faculty of Pharmacy, Pôle Technologie-Santé, Saint-Joseph University, Beirut, Lebanon (M.A.); Department of Integrative Biology, Center for Theoretical Evolutionary Genomics, University of California Berkeley, Berkeley, California (S.P.); Department of Biology, Stanford University, Stanford, California (S.P.); and Département de Génétique, AP-HP, CHU Xavier Bichat, and Université Paris Diderot, Paris, France (C.B.)
| | - Marianne Abifadel
- Laboratory of Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal, affiliated to Université de Montréal, QC, Canada (N.G.S., A.P.); LVTS, INSERM U1148, Hôpital Xavier-Bichat, Paris, France (M.A., C.B.); Laboratory of Biochemistry and Molecular Therapeutics, Faculty of Pharmacy, Pôle Technologie-Santé, Saint-Joseph University, Beirut, Lebanon (M.A.); Department of Integrative Biology, Center for Theoretical Evolutionary Genomics, University of California Berkeley, Berkeley, California (S.P.); Department of Biology, Stanford University, Stanford, California (S.P.); and Département de Génétique, AP-HP, CHU Xavier Bichat, and Université Paris Diderot, Paris, France (C.B.)
| | - Stefan Prost
- Laboratory of Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal, affiliated to Université de Montréal, QC, Canada (N.G.S., A.P.); LVTS, INSERM U1148, Hôpital Xavier-Bichat, Paris, France (M.A., C.B.); Laboratory of Biochemistry and Molecular Therapeutics, Faculty of Pharmacy, Pôle Technologie-Santé, Saint-Joseph University, Beirut, Lebanon (M.A.); Department of Integrative Biology, Center for Theoretical Evolutionary Genomics, University of California Berkeley, Berkeley, California (S.P.); Department of Biology, Stanford University, Stanford, California (S.P.); and Département de Génétique, AP-HP, CHU Xavier Bichat, and Université Paris Diderot, Paris, France (C.B.)
| | - Catherine Boileau
- Laboratory of Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal, affiliated to Université de Montréal, QC, Canada (N.G.S., A.P.); LVTS, INSERM U1148, Hôpital Xavier-Bichat, Paris, France (M.A., C.B.); Laboratory of Biochemistry and Molecular Therapeutics, Faculty of Pharmacy, Pôle Technologie-Santé, Saint-Joseph University, Beirut, Lebanon (M.A.); Department of Integrative Biology, Center for Theoretical Evolutionary Genomics, University of California Berkeley, Berkeley, California (S.P.); Department of Biology, Stanford University, Stanford, California (S.P.); and Département de Génétique, AP-HP, CHU Xavier Bichat, and Université Paris Diderot, Paris, France (C.B.)
| | - Annik Prat
- Laboratory of Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal, affiliated to Université de Montréal, QC, Canada (N.G.S., A.P.); LVTS, INSERM U1148, Hôpital Xavier-Bichat, Paris, France (M.A., C.B.); Laboratory of Biochemistry and Molecular Therapeutics, Faculty of Pharmacy, Pôle Technologie-Santé, Saint-Joseph University, Beirut, Lebanon (M.A.); Department of Integrative Biology, Center for Theoretical Evolutionary Genomics, University of California Berkeley, Berkeley, California (S.P.); Department of Biology, Stanford University, Stanford, California (S.P.); and Département de Génétique, AP-HP, CHU Xavier Bichat, and Université Paris Diderot, Paris, France (C.B.)
| |
Collapse
|
136
|
Arca M. Old challenges and new opportunities in the clinical management of heterozygous familial hypercholesterolemia (HeFH): The promises of PCSK9 inhibitors. Atherosclerosis 2017; 256:134-145. [DOI: 10.1016/j.atherosclerosis.2016.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 08/29/2016] [Accepted: 09/01/2016] [Indexed: 12/17/2022]
|
137
|
Subcellular Trafficking of Mammalian Lysosomal Proteins: An Extended View. Int J Mol Sci 2016; 18:ijms18010047. [PMID: 28036022 PMCID: PMC5297682 DOI: 10.3390/ijms18010047] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 12/15/2016] [Accepted: 12/18/2016] [Indexed: 01/02/2023] Open
Abstract
Lysosomes clear macromolecules, maintain nutrient and cholesterol homeostasis, participate in tissue repair, and in many other cellular functions. To assume these tasks, lysosomes rely on their large arsenal of acid hydrolases, transmembrane proteins and membrane-associated proteins. It is therefore imperative that, post-synthesis, these proteins are specifically recognized as lysosomal components and are correctly sorted to this organelle through the endosomes. Lysosomal transmembrane proteins contain consensus motifs in their cytosolic regions (tyrosine- or dileucine-based) that serve as sorting signals to the endosomes, whereas most lysosomal acid hydrolases acquire mannose 6-phosphate (Man-6-P) moieties that mediate binding to two membrane receptors with endosomal sorting motifs in their cytosolic tails. These tyrosine- and dileucine-based motifs are tickets for boarding in clathrin-coated carriers that transport their cargo from the trans-Golgi network and plasma membrane to the endosomes. However, increasing evidence points to additional mechanisms participating in the biogenesis of lysosomes. In some cell types, for example, there are alternatives to the Man-6-P receptors for the transport of some acid hydrolases. In addition, several “non-consensus” sorting motifs have been identified, and atypical transport routes to endolysosomes have been brought to light. These “unconventional” or “less known” transport mechanisms are the focus of this review.
Collapse
|
138
|
Tao W, Moore R, Smith ER, Xu XX. Endocytosis and Physiology: Insights from Disabled-2 Deficient Mice. Front Cell Dev Biol 2016; 4:129. [PMID: 27933291 PMCID: PMC5122593 DOI: 10.3389/fcell.2016.00129] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/27/2016] [Indexed: 01/29/2023] Open
Abstract
Disabled-2 (Dab2) is a clathrin and cargo binding endocytic adaptor protein, and cell biology studies revealed that Dab2 plays a role in cellular trafficking of a number of transmembrane receptors and signaling proteins. A PTB/PID domain located in the N-terminus of Dab2 binds the NPXY motif(s) present at the cytoplasmic tails of certain transmembrane proteins/receptors. The membrane receptors reported to bind directly to Dab2 include LDL receptor and its family members LRP1 and LRP2 (megalin), growth factor receptors EGFR and FGFR, and the cell adhesion receptor beta1 integrin. Dab2 also serves as an adaptor in signaling pathways. Particularly, Dab2 facilitates the endocytosis of the Ras activating Grb2/Sos1 signaling complex, controls its disassembly, and thereby regulates the Ras/MAPK signaling pathway. Cellular analyses have suggested several diverse functions for the widely expressed proteins, and Dab2 is also considered a tumor suppressor, as loss or reduced expression is found in several cancer types. Dab2 null mutant mice were generated and investigated to determine if the findings from cellular studies might be important and relevant in intact animals. Dab2 conditional knockout mice mediated through a Sox2-Cre transgene have no obvious developmental defects and have a normal life span despite that the Dab2 protein is essentially absent in the mutant mice. The conditional knockout mice were grossly normal, though more recent investigation of the Dab2-deficient mice revealed several phenotypes, which can be accounted for by several previously suggested mechanisms. The studies of mutant mice established that Dab2 plays multiple physiological roles through its endocytic functions and modulation of signal pathways.
Collapse
Affiliation(s)
- Wensi Tao
- Sylvester Comprehensive Cancer Center and Department of Cell Biology, Graduate Program in Cell and Developmental Biology, University of Miami School of Medicine Miami, FL, USA
| | - Robert Moore
- Sylvester Comprehensive Cancer Center and Department of Cell Biology, Graduate Program in Cell and Developmental Biology, University of Miami School of Medicine Miami, FL, USA
| | - Elizabeth R Smith
- Sylvester Comprehensive Cancer Center and Department of Cell Biology, Graduate Program in Cell and Developmental Biology, University of Miami School of Medicine Miami, FL, USA
| | - Xiang-Xi Xu
- Sylvester Comprehensive Cancer Center and Department of Cell Biology, Graduate Program in Cell and Developmental Biology, University of Miami School of Medicine Miami, FL, USA
| |
Collapse
|
139
|
Abstract
Genetic variation arises through multiple different alleles that vary in frequency and severity of effect. Mutations that give rise to Mendelian disorders, such as the LDL receptor (LDLR) mutations that result in familial hypercholesterolaemia, are efficiently winnowed from the population by purifying selection and are almost inevitably rare. Conversely, alleles that are common in the population (such that homozygotes for the minor allele are present even in modest sample sizes) typically have very modest phenotypic effects. Mutations in the gene for proprotein convertase subtilisin/kexin type 9 (PCSK9) represent an unusual but informative exception in that they are relatively common but have large effects on phenotype. Loss-of-function mutations in PCSK9 occur in ~2.5% of African Americans and are associated with large reductions in coronary heart disease (CHD) risk. The development of agents to inhibit PCSK9 demonstrates the utility of translating genetics into clinical therapeutics. Attempts to identify genes responsible for hypercholesterolaemia have used traditional linkage analysis, which requires samples collected from multiple families with defects in the same gene, or genome-wide association, which requires thousands of samples from the population. More recently, whole-exome sequencing studies have revealed loss-of-function mutations in ANGPTL3 associated with pan-hypolipidemia, and in APOC3 that confer protection against CHD. The application of whole-exome sequencing to large populations or to carefully selected patients can streamline the discovery of causal genetic mutations.
Collapse
Affiliation(s)
- J C Cohen
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
140
|
Abstract
Familial hypercholesterolemia (FH) is a common genetic disorder that can manifest clinically as both the severe homozygous (HoFH) form that often presents in childhood and the commoner heterozygous (HeFH) form that is typically identified in adults. The majority of genetic causes are due to defects in low-density lipoprotein (LDL) receptor synthesis and action. Until recently, it was exceedingly difficult to achieve the goal of a 50% reduction in LDL-cholesterol or LDL-C < 70-100 in these patients. Established therapies include statins, niacin, bile-acid sequestrants, and ezetimibe in various combinations. The recent advent of monoclonal antibodies to PCSK9 (evolocumab and alirocumab) has revolutionized the management of FH and results in a substantial reduction in LDL-C and also reductions in Lp(a). In addition, the previous ushering in of antisense therapy against apoB (mipomersen) and inhibition of microsomal transfer protein (lomitapide) for use in HoFH greatly enhanced our ability to manage refractory hypercholesterolemia in these patients. Hence, the therapeutic landscape for this common disorder has changed dramatically for these patients, with a strong promise for a reduction in cardiovascular events.
Collapse
Affiliation(s)
- P Barton Duell
- 1 Knight Cardiovascular Institute, Oregon Health Sciences University , Portland, Oregon
| | - Ishwarlal Jialal
- 2 California Northstate University College of Medicine , Elk Grove, California
| |
Collapse
|
141
|
Disabled-2 Determines Commitment of a Pre-adipocyte Population in Juvenile Mice. Sci Rep 2016; 6:35947. [PMID: 27779214 PMCID: PMC5078790 DOI: 10.1038/srep35947] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 10/06/2016] [Indexed: 01/01/2023] Open
Abstract
Disabled-2 (Dab2) is a widely expressed clathrin binding endocytic adaptor protein and known for the endocytosis of the low-density lipoprotein (LDL) family receptors. Dab2 also modulates endosomal Ras/MAPK (Erk1/2) activity by regulating the disassembly of Grb2/Sos1 complexes associated with clathrin-coated vesicles. We found that the most prominent phenotype of Dab2 knockout mice was their striking lean body composition under a high fat and high caloric diet, although the weight of the mutant mice was indistinguishable from wild-type littermates on a regular chow. The remarkable difference in resistance to high caloric diet-induced weight gain of the dab2-deleted mice was presented only in juvenile but not in mature mice. Investigation using Dab2-deficient embryonic fibroblasts and mesenchymal stromal cells indicated that Dab2 promoted adipogenic differentiation by modulation of MAPK (Erk1/2) activity, which otherwise suppresses adipogenesis through the phosphorylation of PPARγ. The results suggest that Dab2 is required for the excessive calorie-induced differentiation of an adipocyte progenitor cell population that is present in juvenile but depleted in mature animals. The finding provides evidence for a limited pre-adipocyte population in juvenile mammals and the requirement of Dab2 in the regulation of Ras/MAPK signal in the commitment of the precursor cells to adipose tissues.
Collapse
|
142
|
Pal R, Ke Q, Pihan GA, Yesilaltay A, Penman ML, Wang L, Chitraju C, Kang PM, Krieger M, Kocher O. Carboxy-terminal deletion of the HDL receptor reduces receptor levels in liver and steroidogenic tissues, induces hypercholesterolemia, and causes fatal heart disease. Am J Physiol Heart Circ Physiol 2016; 311:H1392-H1408. [PMID: 27694217 DOI: 10.1152/ajpheart.00463.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/13/2016] [Indexed: 01/15/2023]
Abstract
The HDL receptor SR-BI mediates the transfer of cholesteryl esters from HDL to cells and controls HDL abundance and structure. Depending on the genetic background, loss of SR-BI causes hypercholesterolemia, anemia, reticulocytosis, splenomegaly, thrombocytopenia, female infertility, and fatal coronary heart disease (CHD). The carboxy terminus of SR-BI (505QEAKL509) must bind to the cytoplasmic adaptor PDZK1 for normal hepatic-but not steroidogenic cell-expression of SR-BI protein. To determine whether SR-BI's carboxy terminus is also required for normal protein levels in steroidogenic cells, we introduced into SR-BI's gene a 507Ala/STOP mutation that produces a truncated receptor (SR-BIΔCT). As expected, the dramatic reduction of hepatic receptor protein in SR-BIΔCT mice was similar to that in PDZK1 knockout (KO) mice. Unlike SR-BI KO females, SR-BIΔCT females were fertile. The severity of SR-BIΔCT mice's hypercholesterolemia was intermediate between those of SR-BI KO and PDZK1 KO mice. Substantially reduced levels of the receptor in adrenal cortical cells, ovarian cells, and testicular Leydig cells in SR-BIΔCT mice suggested that steroidogenic cells have an adaptor(s) functionally analogous to hepatic PDZK1. When SR-BIΔCT mice were crossed with apolipoprotein E KO mice (SR-BIΔCT/apoE KO), pathologies including hypercholesterolemia, macrocytic anemia, hepatic and splenic extramedullary hematopoiesis, massive splenomegaly, reticulocytosis, thrombocytopenia, and rapid-onset and fatal occlusive coronary arterial atherosclerosis and CHD (median age of death: 9 wk) were observed. These results provide new insights into the control of SR-BI in steroidogenic cells and establish SR-BIΔCT/apoE KO mice as a new animal model for the study of CHD.
Collapse
Affiliation(s)
- Rinku Pal
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Qingen Ke
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - German A Pihan
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Ayce Yesilaltay
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts; and
| | - Marsha L Penman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts; and
| | - Li Wang
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts; and
| | - Chandramohan Chitraju
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts
| | - Peter M Kang
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Monty Krieger
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts; and
| | - Olivier Kocher
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts;
| |
Collapse
|
143
|
Priest JR, Knowles JW. Standards of Evidence and Mechanistic Inference in Autosomal Recessive Hypercholesterolemia. Arterioscler Thromb Vasc Biol 2016; 36:1465-6. [DOI: 10.1161/atvbaha.116.307714] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- James R. Priest
- From the Divisions of Pediatric Cardiology (J.R.P.) and Cardiovascular Medicine (J.W.K.), Stanford Cardiovascular Institute, Stanford University School of Medicine, CA; and The FH Foundation, Pasadena, CA (J.W.K.)
| | - Joshua W. Knowles
- From the Divisions of Pediatric Cardiology (J.R.P.) and Cardiovascular Medicine (J.W.K.), Stanford Cardiovascular Institute, Stanford University School of Medicine, CA; and The FH Foundation, Pasadena, CA (J.W.K.)
| |
Collapse
|
144
|
Thedrez A, Sjouke B, Passard M, Prampart-Fauvet S, Guédon A, Croyal M, Dallinga-Thie G, Peter J, Blom D, Ciccarese M, Cefalù AB, Pisciotta L, Santos RD, Averna M, Raal F, Pintus P, Cossu M, Hovingh K, Lambert G. Proprotein Convertase Subtilisin Kexin Type 9 Inhibition for Autosomal Recessive Hypercholesterolemia-Brief Report. Arterioscler Thromb Vasc Biol 2016; 36:1647-50. [PMID: 27079874 DOI: 10.1161/atvbaha.116.307493] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/31/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Proprotein convertase subtilisin kexin type 9 (PCSK9) inhibitors lower low-density lipoprotein (LDL) cholesterol in the vast majority of patients with autosomal dominant familial hypercholesterolemia. Will PCSK9 inhibition with monoclonal antibodies, in particular alirocumab, be of therapeutic value for patients with autosomal recessive hypercholesterolemia (ARH)? APPROACH AND RESULTS Primary lymphocytes were obtained from 28 genetically characterized ARH patients and 11 controls. ARH lymphocytes treated with mevastatin were incubated with increasing doses of recombinant PCSK9 with or without saturating concentrations of alirocumab. Cell surface LDL receptor expression measured by flow cytometry and confocal microscopy was higher in ARH than in control lymphocytes. PCSK9 significantly reduced LDL receptor expression in ARH lymphocytes albeit to a lower extent than in control lymphocytes (25% versus 76%, respectively), an effect reversed by alirocumab. Fluorescent LDL cellular uptake, also measured by flow cytometry, was reduced in ARH lymphocytes compared with control lymphocytes. PCSK9 significantly lowered LDL cellular uptake in ARH lymphocytes, on average by 18%, compared with a 46% reduction observed in control lymphocytes, an effect also reversed by alirocumab. Overall, the effects of recombinant PCSK9, and hence of alirocumab, on LDL receptor expression and function were significantly less pronounced in ARH than in control cells. CONCLUSIONS PCSK9 inhibition with alirocumab on top of statin treatment has the potential to lower LDL cholesterol in some autosomal recessive hypercholesterolemia patients.
Collapse
Affiliation(s)
- Aurélie Thedrez
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Barbara Sjouke
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Maxime Passard
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Simon Prampart-Fauvet
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Alexis Guédon
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Mikael Croyal
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Geesje Dallinga-Thie
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Jorge Peter
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Dirk Blom
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Milco Ciccarese
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Angelo B Cefalù
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Livia Pisciotta
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Raul D Santos
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Maurizio Averna
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Frederick Raal
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Paolo Pintus
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Maria Cossu
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Kees Hovingh
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Gilles Lambert
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| |
Collapse
|
145
|
Al Rasadi K, Almahmeed W, AlHabib KF, Abifadel M, Farhan HA, AlSifri S, Jambart S, Zubaid M, Awan Z, Al-Waili K, Barter P. Dyslipidaemia in the Middle East: Current status and a call for action. Atherosclerosis 2016; 252:182-187. [PMID: 27522462 DOI: 10.1016/j.atherosclerosis.2016.07.925] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 07/10/2016] [Accepted: 07/27/2016] [Indexed: 12/18/2022]
Abstract
The increase in the cardiovascular disease (CVD)-associated mortality rate in the Middle East (ME) is among the highest in the world. The aim of this article is to review the current prevalence of dyslipidaemia and known gaps in its management in the ME region, and to propose initiatives to address the burden of dyslipidaemia. Published literature on the epidemiology of dyslipidaemia in the ME region was presented and discussed at an expert meeting that provided the basis of this review article. The high prevalence of metabolic syndrome, diabetes, familial hypercholesterolaemia (FH) and consanguineous marriages, in the ME region, results in a pattern of dyslipidaemia (low high-density lipoprotein cholesterol and high triglycerides) that is different from many other regions of the world. Early prevention and control of dyslipidaemia is of paramount importance to reduce the risk of developing CVD. Education of the public and healthcare professionals and developing preventive programs, FH registries and regional guidelines on dyslipidaemia are the keys to dyslipidaemia management in the ME region.
Collapse
Affiliation(s)
- Khalid Al Rasadi
- Department of Clinical Biochemistry, Sultan Qaboos University Hospital, Muscat, Oman.
| | - Wael Almahmeed
- Heart and Vascular Institute -Cleveland Clinic, Abu Dhabi, United Arab Emirates
| | - Khalid F AlHabib
- Department of Cardiac Sciences, King Fahad Cardiac Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Marianne Abifadel
- Laboratory of Biochemistry and Molecular Therapeutics, Faculty of Pharmacy, Saint-Joseph University of Beirut, Beirut, Lebanon
| | - Hasan Ali Farhan
- Baghdad Teaching Hospital, Medical City, Iraqi Board for Medical Specialization, Baghdad, Iraq
| | - Saud AlSifri
- Department of Internal Medicine, Alhada Armed Forces Hospital, Taif, Saudi Arabia
| | - Selim Jambart
- St Joseph University Faculty of Medicine and Hotel Dieu Hospital, Beirut, Lebanon
| | - Mohammad Zubaid
- Department of Medicine, Faculty of Medicine, Kuwait University, Kuwait
| | - Zuhier Awan
- Department of Clinical Biochemistry, King Abdulaziz University, Abdullah Sulayman, Jeddah, Saudi Arabia
| | - Khalid Al-Waili
- Department of Clinical Biochemistry, Sultan Qaboos University Hospital, Muscat, Oman
| | - Philip Barter
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| |
Collapse
|
146
|
Foody JM, Vishwanath R. Familial hypercholesterolemia/autosomal dominant hypercholesterolemia: Molecular defects, the LDL-C continuum, and gradients of phenotypic severity. J Clin Lipidol 2016; 10:970-986. [DOI: 10.1016/j.jacl.2016.04.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 04/19/2016] [Accepted: 04/26/2016] [Indexed: 01/17/2023]
|
147
|
Rabacchi C, Bigazzi F, Puntoni M, Sbrana F, Sampietro T, Tarugi P, Bertolini S, Calandra S. Phenotypic variability in 4 homozygous familial hypercholesterolemia siblings compound heterozygous for LDLR mutations. J Clin Lipidol 2016; 10:944-952.e1. [DOI: 10.1016/j.jacl.2016.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/11/2016] [Accepted: 04/12/2016] [Indexed: 12/31/2022]
|
148
|
Pariani MJ, Knowles JW. Integration of Clinical Genetic Testing in Cardiovascular Care. CURRENT GENETIC MEDICINE REPORTS 2016. [DOI: 10.1007/s40142-016-0094-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
149
|
Dianat N, Weber A, Dubart-Kupperschmitt A. [Human pluripotent stem cells and liver disorders]. Biol Aujourdhui 2016; 210:19-26. [PMID: 27286577 DOI: 10.1051/jbio/2016006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Indexed: 11/14/2022]
Abstract
The liver is associated with many diseases including metabolic and cholestatic diseases, cirrhosis as well as chronic and acute hepatitis. However, knowledge about the mechanisms involved in the pathophysiology of these diseases remains limited due to the restricted access to liver biopsies and the lack of cellular models derived from patients. The liver is the main organ responsible for the elimination of xenobiotics and thus hepatocytes have a key role in toxicology and pharmacokinetics. The induced pluripotent stem cells generated from patients with monogenic metabolic disorders, for which the corresponding gene is identified, are relevant in vitro models for the study of the mechanisms involved in generation of pathologies and also for drug screening. Towards this aim, robust protocols for generating liver cells, such as hepatocytes and cholangiocytes, are essential. Our study focused on familial hypercholesterolemia disease modeling, as well as on establishing a protocol for generation of functional cholangiocytes from pluripotent stem cells.
Collapse
Affiliation(s)
- Noushin Dianat
- INSERM U1193, Hôpital Paul Brousse, 94807 Villejuif, France - UMR S1193, Université Paris-Sud, Hôpital Paul Brousse, 94800 Villejuif, France - Département hospitalo-universitaire Hepatinov, Hôpital Paul Brousse, 94807 Villejuif, France
| | - Anne Weber
- INSERM U1193, Hôpital Paul Brousse, 94807 Villejuif, France - UMR S1193, Université Paris-Sud, Hôpital Paul Brousse, 94800 Villejuif, France - Département hospitalo-universitaire Hepatinov, Hôpital Paul Brousse, 94807 Villejuif, France
| | - Anne Dubart-Kupperschmitt
- INSERM U1193, Hôpital Paul Brousse, 94807 Villejuif, France - UMR S1193, Université Paris-Sud, Hôpital Paul Brousse, 94800 Villejuif, France - Département hospitalo-universitaire Hepatinov, Hôpital Paul Brousse, 94807 Villejuif, France
| |
Collapse
|
150
|
Merchán A, Ruiz ÁJ, Campo R, Prada CE, Toro JM, Sánchez R, Gómez JE, Jaramillo NI, Molina DI, Vargas-Uricoechea H, Sixto S, Castro JM, Quintero AE, Coll M, Slotkus S, Ramírez A, Pachajoa H, Ávila FA, Alonso K R. Hipercolesterolemia familiar: artículo de revisión. REVISTA COLOMBIANA DE CARDIOLOGÍA 2016. [DOI: 10.1016/j.rccar.2016.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|