Viewpoint. Genetics of Dyslipidemia. Keywords

Similar documents
Genetic and biochemical characteristics of patients with hyperlipidemia who require LDL apheresis

Nature Genetics: doi: /ng.3561

Behind LDL: The Metabolism of ApoB, the Essential Apolipoprotein in LDL and VLDL

Appendix F Simon Broome Diagnostic criteria for index individuals and relatives

Low-density lipoproteins cause atherosclerotic cardiovascular disease (ASCVD) 1. Evidence from genetic, epidemiologic and clinical studies

Zuhier Awan, MD, PhD, FRCPC

Central role of apociii

There are many ways to lower triglycerides in humans: Which are the most relevant for pancreatitis and for CV risk?

Lipid Metabolism in Familial Hypercholesterolemia

Pathophysiology of Lipid Disorders

LIPOPROTEINE ATEROGENE E ANTI-ATEROGENE ATEROGENE

Treatment of Atherosclerosis in 2007

Focus on FH (Familial Hypercholesterolemia) Joshua W. Knowles, MD PhD for PCNA May, 2013

Low-density lipoprotein as the key factor in atherogenesis too high, too long, or both

UnitedHealthcare Pharmacy Clinical Pharmacy Programs

UnitedHealthcare Pharmacy Clinical Pharmacy Programs

High density lipoprotein metabolism

Disclosures. Pediatric Dyslipidemia Casey Elkins, DNP, NP C, CLS, FNLA. Learning Objectives. Atherogenesis. Acceptable Values

Update on Familial Hypercholesterolemia: Diagnosis, Cardiovascular Risk, and Novel Therapeutics

Familial hypercholesterolaemia

The overall frequency of major

Genetic Dyslipidemia and Cardiovascular Diseases

Familial hypercholesterolaemia in children and adolescents

DIAGNOSIS AND TREATMENT OF FH CHILDREN. O. GUARDAMAGNA Dipartimento di Scienze della Sanità Pubblica e Pediatriche UNIVERSITA DI TORINO

. Non HDL-c : Downloaded from ijdld.tums.ac.ir at 18:05 IRDT on Friday March 22nd Non HDL LDL. . LDL Non HDL-c

1. Which one of the following patients does not need to be screened for hyperlipidemia:

ATP III (Adult Treatment Panel III) CLASSIFICATION C IN ADULTS

Comparison of Abnormal Cholesterol in Children, Adolescent & Adults in the United States, : Review

Metabolism and Atherogenic Properties of LDL

ScreenPro FH: From the Czech MedPed to International Collaboration. ScreenPro FH Is a Participating Project

Cost effectiveness of cascade testing for FH:

Very high cholesterol from birth: are target LDL cholesterol levels now achievable with new treatments?

Cascade Screening for FH: the U.S. experience

Kynamro (mipomersen)

Management of Lipid Disorders in Children and Adolescents

REPATHA (PCSK9 INHIBITORS)

Metabolism, Atherogenic Properties and Agents to reduce Triglyceride-Rich Lipoproteins Manfredi Rizzo, MD, PhD

Drug Class Prior Authorization Criteria PCSK9 Inhibitors

The inhibition of CETP: From simply raising HDL-c to promoting cholesterol efflux and lowering of atherogenic lipoproteins Prof Dr J Wouter Jukema

Regence. Medical Policy Manual. Date of Origin: May Topic: Genetic Testing for Lipoprotein(a) Variant(s) as a Decision Aid for Aspirin Treatment

THE CLINICAL BIOCHEMISTRY OF LIPID DISORDERS

Familial Hypercholesterolemia What a cardiologist should know

2.5% of all deaths globally each year. 7th leading cause of death by % of people with diabetes live in low and middle income countries

Density Lipoprotein (HDL) and Corneal Opacity Caused by Lecithin-Cholesterol

Disclosures. Background 1 What is Known MENOPAUSE, ESTROGENS, AND LIPOPROTEIN PARTICLES. Background 2 What is Not Known 10/2/2017

Comprehensive Treatment for Dyslipidemias. Eric L. Pacini, MD Oregon Cardiology 2012 Cardiovascular Symposium

Tina-quant Lipoprotein (a) Gen. 2 For accurate and reliable assessment of cardiovascular risk

PCSK9 Inhibition: From Genetics to Patients

sad EFFECTIVE DATE: POLICY LAST UPDATED:

Dyslipidemia. (Med-341)

Screening for dyslipidemias in children and adolescents

Chapter (5) Etiology of Low HDL- Cholesterol

THE IMPACT OF FAMILIAL HYPERCHOLESTEROLEMIA ON CARDIOVASCULAR DISEASE

Diagnosis and Management of Familial Hypercholesterolaemia Position Statement

Identification and management of familial hypercholesterolaemia (FH) - An overview

Apolipoprotein B in the Risk Assessment and Management of Cardiovascular Disease. Original Policy Date

Distinguishing FH from non-fh: Right therapy, right patient, right time. Joshua W. Knowles, MD, PhD Stanford and the FH Foundation

Juxtapid and Kynamro

Comparison of Lipoprotein (a) and Apolipoproteins in Children with and without Familial History of Premature Coronary Artery Disease

Hypertriglyceridemia. Ara Metjian, M.D. Resident s Report 20 December 2002

Measurement of Serum Intermediate Density Lipoproteins (Remnant-like Particles) Original Policy Date

*Address Correspondence to Päivi Pajukanta, MD, PhD, Associate Professor, Department of

Lipids and cardiovascular disease 3 Triglycerides and cardiovascular disease

An update on lipidology and cardiovascular risk management. Lipids, Metabolism & Vascular Risk Section - Royal Society of Medicine

The Cardiovascular Institute Mount Sinai School of Medicine, New York

ADMINISTRATIVE POLICY AND PROCEDURE

PCSK9 Agents Drug Class Prior Authorization Protocol

Appendix E Health Economic modelling

LIPIDS AND CHOLESTEROL - RISK FACTORS TO A POLICE UNIT FROM BRASOV

Lipoproteins Metabolism

Approach to Dyslipidemia among diabetic patients

raised triglycerides and residual cardiovascular risk

Current Cholesterol Guidelines and Treatment of Residual Risk COPYRIGHT. J. Peter Oettgen, MD

The Role of Apolipoprotein CIII in Coronary Artery Disease. Disclosures

CETP inhibition: pros and cons. Philip Barter The Heart Research Institute Sydney, Australia

Fasting or non fasting?

High-Density Lipoprotein Subclass Testing in the Diagnosis and Management of Cardiovascular Disease. Original Policy Date

C h a p t e r 1 9 Protective HDL Cholesterol : Modalities to Elevate it

Is it really that simple? Alyssa Hasty, PhD Associate Professor Molecular Physiology and Biophysics

Proprotein Convertase Subtilisin/Kexin type 9(PCSK9) Inhibitors Prior Authorization with Quantity Limit Program Summary

Katsuyuki Nakajima, PhD. Member of JCCLS International Committee

DYSLIPIDEMIA. Michael Brändle, Stefan Bilz

Lipoprotein (a): Is it important for Friedewald formula?

The Addition of Ezetimibe to Statin therapy in. Patients with Homozygous Familial. Hypercholesterolaemia

Role of diet in prevention and treatment of hypercholesterolaemia/hypertriglyceridaemia in children C. Hartman (IL)

The Target is LDL, HDL not so much

Lipoprotein (a) Disclosures 2/20/2013. Lipoprotein (a): Should We Measure? Should We Treat? Health Diagnostic Laboratory, Inc. No other disclosures

Considerations and Controversies in the Management of Dyslipidemia for ASCVD Risk Reduction

Progress in the care of familial hypercholesterolaemia: 2016

Homozygous Familial Hypercholesterolemia: phenotype rules! Commentary on the study of Raal et al.

Thematic Review Series: Genetics of Human Lipid Diseases. Genetic causes of high and low serum HDL-cholesterol

Repatha. Repatha (evolocumab) Description

LIPID CLUB Rome, 2014

Request for Prior Authorization for PCSK9 inhibitor therapy Website Form Submit request via: Fax

Lipid/Lipoprotein Structure and Metabolism (Overview)

A Naturally Randomized Trial Comparing the Effect of Genetic Variants that Mimic CETP Inhibitors and Statins on the Risk of Cardiovascular Disease.

Hypertriglyceridemia: Why, When, and How to Treat. Gregory Cohn, MD, FNLA, FASPC

Lipids Board Review. Ira Goldberg, MD New York University School of Medicine. Which of the following is the best initial therapy choice?

Managing Dyslipidemia and ASCVD Risk: Confusion, Controversy Consensus

High lipoprotein(a) as a possible cause of clinical familial hypercholesterolaemia: a prospective cohort study

Transcription:

Genetics of Dyslipidemia Diego García-Giustiniani 1,2 and Ricardo Stein 3,4,5 Health in Code 1, La Coruña, Spain; Grupo de Investigación Cardiovascular de La Universidad de La Coruña, La Coruña, Spain 2 ; Grupo de Pesquisa em Cardiologia do Exercício do Hospital de Clínicas de Porto Alegre 3 ; Serviço de Cardiologia do Hospital de Clínicas de Porto Alegre Universidade Federal do Rio Grande do Sul 4 ; Vitta Centro de Bem Estar Físico 5, Porto Alegre, RS Brazil Lipoproteins constitute a set of proteins and lipids, organized to facilitate the transport of lipids through blood plasma. Elevated or decreased levels of these lipoproteins may be related to genetic alterations in 40% to 60% of cases. 1 This fact explains why it is common to find lipid abnormalities in several members of the same family. The elevated levels of these composite organic biomolecules are responsible for approximately 50% of the attributable risk for the development of atherosclerotic cardiovascular diseases, 2,3 a process in which other phenotypes with a hereditary component also participate, such as diabetes, obesity and metabolic syndrome. Population studies of genetic association identified more than one hundred genes that could have a direct impact on lipid levels. 3 These genes affect plasma levels of total cholesterol, low-density lipoprotein-cholesterol (LDL-c) and high-density lipoprotein cholesterol (HDL-c), as well as of triglycerides, being capable of establishing complex phenotypes. However, among these genes identified in the association studies, there is a previously known and well characterized group of genes responsible for the development of monogenic dyslipidemias. In such cases, the variant in a single gene clearly explains the phenotype. It is noteworthy that even when isolated, these mutations determine diseases considered rare and the impact produced by variants of these genes tend to be high, resulting in extreme values of lipid levels. Considering that cardiovascular atherosclerotic disease is a process that starts in childhood and progresses throughout life, 4 the early identification of risk factors is very important. In this sense, the diagnosis of phenotypes determined by monogenic diseases of lipid metabolism deserves special attention, considering that, when they produce extreme phenotypes, they can be associated with the development of early atherosclerosis of rapid progression. In fact, genetic studies using Next-Generation Sequencing (NGS), carried out in patients with early acute myocardial infarction (AMI) (that occurring before 50 years of age in men and 60 in women) have determined that the presence of rare genetic variants in the LDLR gene resulted in a 4-fold higher risk of this event in their carriers. On the other hand, the rare variants in the APOA5 gene, associated with high triglyceride levels, resulted in a 3-fold higher risk. Moreover, approximately 2% of the cases were carriers of a clearly pathogenic mutation in LDLR, suggesting that these patients had a diagnosis of familial hypercholesterolemia. 2 In a broader sense, it is estimated that approximately 5% of AMIs in patients aged < 60 years may be due to familial hypercholesterolemia, a figure that can increase up to 20% when acute coronary events affect individuals younger than 45 years. 5 It is noteworthy that even in the case of monogenic diseases, mutations identified in many of the genes frequently responsible for these diseases show significant variability in phenotypic expression and the associated risk, either in carriers from the same family or among carriers of the same mutation that belong to different populations. 6-9 Occasionally, this fact may hinder or delay diagnosis in patients to whom only clinical criteria are applied, since in those circumstances, lipid levels may overlap those observed in the general population. The great variability in clinical expression in patients with a certain variant is due to the effect of other factors, which would act as modifiers for the expression of the accountable mutation, a fact that has been demonstrated in several occasions. Incidentally, this is the case of variants in genes such as ApoB, PCSK9, LRP1 and Lp(a), as well as in carriers with mutations in LDLR diagnosed with familial hypercholesterolemia, just to name a few examples. 6-11 The participation of genetic variants as phenotypic expression modifiers is explained by the fact that the genes involved can be independently associated, being capable of promoting an increase or decrease in the lipoprotein levels that they affect. In other words, the interactions of their effects attenuate or increase lipid levels, as well as the associated risk in carriers. Similarly, certain variants may influence not only the phenotypic expression, but also the response to drug treatment, determining that the intervention be or not effective in its goal of reducing the associated risk. 6,7 Keywords Dyslipidemias / genetics; Hyperlipoproteinemia Type II; Dyslipidemias / diagnosis. Mailing Address: Ricardo Stein HCPA UFRGS. João Caetano, 20/402, Petrópolis. Postal Code 90470260, Porto Alegre, RS Brazil E-mail: rstein@cardiol.br Manuscript received January 08, 2016; revised manuscript January 12, 2016; accepted January 20, 2016. DOI: 10.5935/abc.20160074 Genes Associated with Alterations in LDL-C Levels Familial Hypercholesterolemia This familial disease is one of the most prevalent monogenic disorders, being identified in approximately 1 in 500 individuals in the general population. Interestingly, in some European subpopulations, it has been estimated that the frequency is even higher, around 1 in 250 individuals. 8 434

In the past, the characterization of familial hypercholesterolemia was carried out using only clinical and laboratory criteria. However, knowledge of this strategy limitations, regarding its low sensitivity for the diagnosis, 9,10 motivated changes in two of the three main diagnostic criteria of this disease entity. According to the criteria established by the Dutch Lipid Clinic Criteria (Table 1) and the Simon Broome Diagnostic Criteria, the identification of pathogenic mutations, when added to additional clinical criteria, has been used to consider the diagnosis of familial hypercholesterolemia as a definitive one. Incidentally, the identification of pathogenic mutations in probands constitutes the first step to start the genetic screening in this individual s family. This approach is of utmost importance in pediatric and adolescent family members, in which the clinical criteria may not be well defined, although the disease may have already started to exert its effects at the vascular level. Regarding the diagnosis and management of this disease, such perspective has been supported by guidelines established by important medical societies, such as the Canadian, 11 the British (National Institute for Health and Clinical Excellence NICE), 12 the Australian / New Zealander 13 and the Spanish society, 14 among others. The genes for which there is demonstrated evidence of association with familial hypercholesterolemia are LDLR, ApoB, PCSK9 and LDLRAP1. The main lipoprotein alteration associated with the phenotype is the increase in the LDL lipoprotein levels. However, some variations in the ApoB and PCSK9 genes may determine the decrease of this lipoprotein. It is noteworthy that the phenotype determined by the three genes, LDLR, ApoB and PCSK9, shows a pattern of autosomal dominant transmission, while the phenotype determined by mutations in the gene LDLRAP1 has an exclusive autosomal recessive transmission. Phytosterolemia This is one of the main differential diagnoses in patients with suspected familial hypercholesterolemia. This disease is caused by the abnormal absorption of cholesterol and accumulation of sterols, especially those of plant origin (hence its name). The clinical features of this disease include the presence of xanthomas and premature coronary atherosclerosis, as well as hemolytic anemia and/or liver disease. The genes associated with this disease are ABCG5 and ABCG8. Regarding other genes that alter the metabolism of this lipoprotein, it includes a group associated with a decrease in its levels (familial hypobetalipoproteinemia), with a possible protective effect for cardiovascular disease in carriers. 15 The genes associated with these phenotypes are ANGPTL3, MTTP and MYLIP. Table 1 Dutch Lipid Clinic Criteria for the diagnosis of familial hypercholesterolemia (score) Criteria Score Family history First-degree relative with early coronary disease (men < 55 years and women < 60 years) 1 First-degree relative with LDL-c levels > 210 mg/dl First-degree relative with tendon xanthomas and/or corneal arc < 45 years 2 Family member < 18 years with LDL-c 150 mg/dl Personal history Patients with early coronary disease (men < 55 years and women < 60 years) 2 Patient with early cerebrovascular or peripheral artery disease (men < 55 years and women < 60 years) 1 Physical examination Tendon xanthomas 6 Corneal arc in individuals < 45 years 4 Laboratory analysis LDL-c 330 mg/dl 8 LDL-c 250-329 mg/dl 5 LDL-c 190-249 mg/dl 3 LDL-c 155-189 mg/dl 1 Genetic analysis Functional mutation in one of these genes: LDLR, APOB or PCSK9 8 Diagnosis of familial hypercholesterolemia Definite diagnosis: 8 points Probable diagnosis: 6-7 points Possible diagnosis: 3-5 points LDL-c: low-density lipoprotein-cholesterol. 435

Genes Associated with Triglyceride Metabolism Alterations The role of hypertriglyceridemia caused by genetic factors as a risk factor for atherosclerotic cardiovascular disease has been well established. In a recent study, which included a large cohort of patients with early AMI (before age 50), in which sequencing was carried out through NGS, carriers of APOA5 gene variants had an increased risk up to 3.3-fold higher when compared to non-carriers. 16 A similar association had been previously established in another large cohort (the Copenhagen Cohort), in which the LPL gene was also investigated. In this observational study, it was determined that for every increase of 1 mmol/l (39 mg/dl) in triglyceride rich cholesterol (remaining), which is mediated by genetic cause, an increase in risk for coronary artery disease (CAD) was 2.8 higher than in controls. 17 The main genes associated with increased levels of triglycerides (as main phenotype) are APOA5, APOC2, APOE, GPD1, GPIHBP1, HNF1A, LMF1, LPL and SLC25A40. It is noteworthy that a group of rare variants in the APOC3 gene was associated with a reduction in triglyceride levels, promoting reduction of up to 40% in the risk of CAD. 18 Genes related with HDL-c levels Epidemiological studies of the 1990s established an inverse association between HDL-c levels and CAD risk. 19 At present, what is known is that this association seems to be more related to the quality of the HDL-c particles, due to structural abnormalities secondary to mutations, than the absolute amount of this lipoprotein 20,21 a fact that may explain the recent questions about the meaning of HDL-c levels on the development of CAD. 22 The typical example of the structure/function association is that represented by a mutation affecting apolipoprotein A1 (ApoA1 ratio), a major constituent of HDL lipoprotein, which is associated with an effect opposite to that expected. For instance, the mutation (Arg173Cys) determines the production of ApoA1 dimers, determining that HDL-c levels be extremely low. However, these particles are very efficient in reverse cholesterol transport, which makes the carriers have a very low incidence of ischemic heart disease, although an extremely low quantitatively measured HDL-c. Nevertheless, low levels of HDL-c of monogenic etiology determine a group of diseases that have a high prevalence of early atherosclerotic cardiovascular disease. The genes associated with low HDL-c of monogenic etiology are APOA1, ABCA1, LCAT, SAR1B and ABCG1. APOA1 is the gene responsible for the phenotype of familial hypoalphalipoproteinemia, an entity with autosomal dominant etiology. The ABCA1 gene, in turn, is associated with Tangier disease, which is autosomal recessive. Similarly, the LCAT genes, which are associated with fish-eye disease, and the SAR1B gene, which is related to chylomicron-retention disease, also occur with decreased levels of HDL-c. On the other hand, there is a group of genes associated with increased levels of HDL-c. Nevertheless, there is no evidence of a protective effect against atherosclerosis. One explanation for this finding might be related to the fact that these genetic variants alter the distribution of subclasses of HDL-c particles, increasing the less protective ones. 23 This information may be relevant to determine whether the increase of this lipoprotein may be considered when calculating the cardiovascular risk associated with the lipoprotein ratios. The genes associated with increased HDL-C of genetic etiology are CETP, LIPC, PLTP and SCARB1. Secondary Causes of Dyslipidemia Lipodystrophies Lipodystrophies constitute a heterogeneous group of rare diseases of which common characteristic is the selective loss of adipose tissue. However, they predispose to the development of metabolic complications similar to those seen in obese individuals. Among these, are alterations in the lipid metabolism (increased triglycerides and reduced HDL-c) as well as insulin resistance and diabetes mellitus, entities associated with increased risk of premature atherosclerosis. 24,25 The main genes associated with these diseases are LPL, AGPAT2, BSCL2, CAV1, CIDEC, LMNA, PLIN1, PPARG, PTRF and ZMPSTE24. Other Genes of Interest Statin-induced myopathy Muscle pain secondary to treatment with statins is the adverse effect most often associated with the use of these drugs. It is estimated that it affects 1 to 10% of treated patients, 26 being an underdiagnosed complication. 27 The severity and risk for the development of major complications, such as rhabdomyolysis, are related with the combined use of statins, dose and factors such as age and gender, as well as the concomitant use of other drugs affecting the bioavailability of these compounds. The genetic determinants of associated risk consist of genes encoding enzymes related to the metabolism of these drugs, as well as carriers and genes associated with mitochondrial dysfunction. All these factors add up to those effects caused by statins themselves, as a cause of muscle damage. 28 The genes with the highest level of association evidence with this complication are AMPD1, COQ2, CPT2, CYP2D6, PPARA, PYGM, SLC22A8 and SLCO1B1. Conclusion Primary dyslipidemias or those without apparent cause can be classified genotypically or phenotypically through biochemical analysis. In genotypic classification, dyslipidemias are divided into monogenic (caused by mutations in a single gene) and polygenic (caused by associations of multiple mutations, which would not be of great impact when alone). 29 The knowledge of the molecular basis of dyslipidemias allows their correct diagnosis. In this scenario, the institution of an optimal drug therapy may be better founded. Moreover, genetic diagnosis in an index case can trigger a family investigation process, which allows early detection, therapeutic guidance and consequent reduction of cardiovascular risk in such individuals. 436

Author contributions Conception and design of the research: García-Giustiniani D and Stein R. Acquisition of data: García-Giustiniani D and Stein R. Analysis and interpretation of the data: García-Giustiniani D and Stein R. Statistical analysis: García- Giustiniani D and Stein R. Writing of the manuscript: García- Giustiniani D and Stein R. Critical revision of the manuscript for intellectual content: García-Giustiniani D and Stein R. Supervision / as the major investigador: García-Giustiniani D and Stein R. Authors Potential Conflicts of Interest Diego-García-Giustiane works at a company that can directly or indirectly benefit from the study results. Ricardo Stein received a grant from the manufacturer of a product in the area of cardiovascular genetics. Sources of Funding There were no external funding sources for this study. Study Association This article is part of the thesis of Pos Doctoral abroad (Spain) submitted by Ricardo Stein, from Programa de Pós-graduação em Cardiologia e Ciências Cardiovasculares da UFRGS. References 1. Weiss LA, Pan L, Abney M, Ober C. The sex-specific genetic architecture of quantitative traits in humans. Nat Genet. 2006;38(2):218-22. 2. Di Angelantonio E, Sarwar N, Perry P, Kaptoge S, Ray KK, Thompson A, et al. Emerging risk factors collaboration: major lipids, apolipoproteins, and risk of vascular disease. JAMA. 2009;302(18):1993-2000. 3. Asselbergs FW, Guo Y, van Iperen EP, Sivapalaratnam S, Tragante V, Chen YD, et al. Large-scale gene-centric meta-analysis across 32 studies identifies multiple lipid loci. Am J Hum Genet. 2012;2;91(5):823-38. 4. Berenson GS, Srinivasan SR, Bao W, Newman WP 3 rd, Tracy RE, Wattigney WA. Association between multiple cardiovascular risk factors and the early development of atherosclerosis. Bogalusa Heart Study. N Engl J Med. 1998;338(23):1650-6. 5. Hopkins PN, Toth PP, Ballantyne CM, Rader DJ; National Lipid Association Expert Panel on Familial Hypercholesterolemia. Familial hypercholesterolemias: prevalence, genetics, diagnosis and screening recommendations from the National Lipid Association Expert Panel on Familial Hypercholesterolemia. J Clin Lipidol. 2011;5(3 Suppl):S9-17. 6. Peters BJ, Pett H, Klungel OH, Stricker BH, Psaty BM, Glazer NL, et al. Genetic variability within the cholesterol lowering pathway and the effectiveness of statins in reducing the risk of MI. Atherosclerosis. 2011;217(2):458-64. 7. Choumerianou DM, Dedoussis GV. Familial hypercholesterolemia and response to statin therapy according to LDLR genetic background. Clin Chem Lab Med. 2005;43(8):793-801. 8. Nordestgaard BG, Chapman MJ, Humphries SE, Ginsberg HN, Masana L, Descamps OS, et al; European Atherosclerosis Society Consensus Panel. Familial hypercholesterolaemia is underdiagnosed and undertreated in the general population: guidance for clinicians to prevent coronary heart disease: consensus statement of the European Atherosclerosis Society. Eur Heart J. 2013;34(45):3478-3490. 9. Kwiterovich PO Jr, Fredrickson DS, Levy RI. Familial hypercholesterolemia (one form of familial type II hyperlipoproteinemia). A study of its biochemical, genetic and clinical presentation in childhood. J Clin Invest. 1974;53(5):1237-49. 10. Marks D, Thorogood M, Neil HA, Humphries SE. A review on the diagnosis, natural history, and treatment of familial hypercholesterolaemia. Atherosclerosis. 2003;168(1):1-14. 11. Genest J, Hegele RA, Bergeron J, Brophy J, Carpentier A, Couture P, et al. Canadian Cardiovascular Society position statement on familial hypercholesterolemia. Can J Cardiol. 2014;30(12):1471-81. 12. National Institute for Health and Care Excellence (NICE). Familial hypercholesterolaemia: identification and management. [on line]. [Cited in 2015 Jun 10]. Available from: https://www.nice.org.uk/guidance/cg71 13. Index of/up-content/uploads/2013/03. [on line]. [Cited in 2015 May 21]. Available from: http://www.csanz.edu.au/wp-content/uploads/2013/03/ download_document.png 14. Mata P, Alonso R, Ruiz A, Gonzalez-Juanatey JR, Badimón L, Díaz-Díaz JL, et al. [Diagnosis and treatment of familial hypercholesterolemia in Spain: consensus document]. Atención Primaria. 2015;47;(1):56-65. 15. Sankatsing RR, Fouchier SW, de Haan S, Hutten BA, de Groot E, Kastelein JJ, et al. Hepatic and cardiovascular consequences of familial hypobetalipoproteinemia. Arterioscler Thromb Vasc Biol. 2005;25(9):1979-84. 16. Do R, Stitziel NO, Won HH, Jørgensen AB, Duga S, Angelica Merlini P, et al. Exome sequencing identifies rare LDLR and APOA5 alleles conferring risk for myocardial infarction. Nature. 2015;518(7537):102-6. 17. Varbo A, Benn M, Tybjaerg-Hansen A, Jørgensen AB, Frikke-Schmidt R, Nordestgaard BG. Remnant cholesterol as a causal risk factor for ischemic heart disease. J Am Coll Cardiol. 2013;61(4):427-36. 18. Jørgensen AB, Frikke-Schmidt R, Nordestgaard BG, Tybjærg-Hansen A. Loss-of-function mutations in APOC3 and risk of ischemic vascular disease. N Engl J Med. 2014;371(1):32-41. 19. Assmann G, Schulte H, von Eckardstein A, Huang Y. High-density lipoprotein cholesterol as a predictor of coronary heart disease risk. The PROCAM experience and pathophysiological implications for reverse cholesterol transport. Atherosclerosis. 1996;124 Suppl:S11-20. 20. Sirtori CR, Calabresi L, Franceschini G, Baldassarre D, Amato M, Johansson J, et al. Cardiovascular status of carriers of the apolipoprotein A-I(Milano) mutant: the Limone sul Garda study. Circulation. 2001;103(15):1949-54. 21. Rader DJ. Lecithin: cholesterol acyltransferase and atherosclerosis: another high-density lipoprotein story that doesn t quite follow the script. Circulation. 2009;120(7):549-52. 22. Voight BF, Peloso GM, Orho-Melander M, Frikke-Schmidt R, Barbalic M, Jensen MK, et al. Plasma HDL cholesterol and risk of myocardial infarction: a mendelian randomisation study. Lancet. 2012;380(9841):572-80. Erratum in: Lancet. 2012;380(9841):564. 23. Calabresi L, Gomaraschi M, Simonelli S, Bernini F, Franceschini G. HDL and atherosclerosis: Insights from inherited HDL disorders. Biochim Biophys Acta. 2015;1851(1):13-8. 437

24. Simha V, Garg A. Lipodystrophy: lessons in lipid and energy metabolism. Curr Opin Lipidol. 2006;17(2):162-9. 25. Hegele RA. Premature atherosclerosis associated with monogenic insulin resistance. Circulation. 2001;103(18):2225-9. 26. Thompson PD, Clarkson P, Karas RH. Statin-associated myopathy. JAMA. 2003;289(13):1681-90. 27. Law M, Rudnicka AR. Statin safety: a systematic review. Am J Cardiol. 2006;97(8A):52C 60C. 28. Feng Q, Wilke RA, Baye TM. Individualized risk for statin-induced myopathy: current knowledge, emerging challenges and potential solutions. Pharmacogenomics. 2012;13(5):579-94. 29. Sposito AC, Caramelli B, Fonseca FA, Bertolami MC, Afiune Neto A, Souza AD, et al. [IV Brazilian Guideline for dyslipidemia and atherosclerosis prevention: Department of Atherosclerosis of Brazilian Society of Cardiology]. Arq Bras Cardiol. 2007;88 Suppl 1:2-19. 438