Clinica Chimica Acta 431 (2014) 131–142

Contents lists available at ScienceDirect

Clinica Chimica Acta journal homepage: www.elsevier.com/locate/clinchim

Invited critical review

Postprandial hypertriglyceridemia as a coronary risk factor Jan Borén a,⁎, Niina Matikainen b,c, Martin Adiels a, Marja-Riitta Taskinen b a b c

Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden HUCH Heart and Lung Centre, Research Programs Unit Diabetes and Obesity, Cardiovascular Research Group, Finland Department of Endocrinology, Helsinki University Central Hospital, Diabetes & Obesity, University of Helsinki, Helsinki, Finland

a r t i c l e

i n f o

Article history: Received 13 November 2013 Received in revised form 10 January 2014 Accepted 11 January 2014 Available online 6 February 2014 Keywords: Postprandial hypertriglyceridemia Cardiovascular risk factor Triglyceride-rich lipoproteins

a b s t r a c t Postprandial hypertriglyceridemia is now established as an important risk factor for cardiovascular disease (CVD). This metabolic abnormality is principally initiated by overproduction and/or decreased catabolism of triglyceride-rich lipoproteins (TRLs) and is a consequence of predisposing genetic variations and medical conditions such as obesity and insulin resistance. Accumulation of TRLs in the postprandial state promotes the retention of remnant particles in the artery wall. Because of their size, most remnant particles cannot cross the endothelium as efficiently as smaller low-density lipoprotein (LDL) particles. However, since each remnant particle contains approximately 40 times more cholesterol compared with LDL, elevated levels of remnants may lead to accelerated atherosclerosis and CVD. The recognition of postprandial hypertriglyceridemia in the clinical setting has been severely hampered by technical difficulties and the lack of established clinical protocols for investigating postprandial lipemia. In addition, there are currently no internationally agreed management guidelines for this type of dyslipidemia. Here we review the mechanism for and consequences of excessive postprandial hypertriglyceridemia, epidemiological evidence in support of high triglycerides and remnant particles as risk factors for CVD, the definition of hypertriglyceridemia, methods to measure postprandial hypertriglyceridemia and apolipoproteins and, finally, current and future treatment opportunities. © 2014 Elsevier B.V. All rights reserved.

Contents 1. 2. 3. 4. 5.

6. 7. 8.

9. 10. 11.

12.

Introduction . . . . . . . . . . . . . . . . . . . . . . . . Chylomicrons and VLDL particles . . . . . . . . . . . . . . Uptake of dietary fat and formation of CMs . . . . . . . . . . Regulation of the secretion of apoB-containing lipoproteins . . Metabolism of TRLs in the plasma and hepatic uptake . . . . . 5.1. Intravascular lipolysis of TRLs . . . . . . . . . . . . 5.2. ApoCIII: a key regulator of TRL metabolism . . . . . . 5.3. Hepatic removal of remnant lipoproteins . . . . . . . Definition of hypertriglyceridemia . . . . . . . . . . . . . . Genetic variants causing postprandial lipemia . . . . . . . . Epidemiological and clinical evidence for high TG levels in CVD 8.1. Evidence for a link between fasting TG and CVD . . . . 8.2. Evidence for a link between non-fasting TG and CVD . . Epidemiological evidence for remnant particles in CVD . . . . Postprandial lipids in populations with CVD: case–control studies Assessment of postprandial TRL metabolism . . . . . . . . . 11.1. Measurement of non-fasting TG levels . . . . . . . . . 11.2. Calculated remnant cholesterol . . . . . . . . . . . . 11.3. Oral fat tolerance test . . . . . . . . . . . . . . . . 11.4. Alternative markers of postprandial lipemia . . . . . . 11.5. Measurement of apoB48 . . . . . . . . . . . . . . . Current and future therapeutic approaches . . . . . . . . . . 12.1. Management of hypertriglyceridemia . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

⁎ Corresponding author at: Wallenberg Laboratory, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden. Tel.: +46 313422949. E-mail address: [email protected] (J. Borén). 0009-8981/$ – see front matter © 2014 Elsevier B.V. All rights reserved. http://dx.doi.org/10.1016/j.cca.2014.01.015

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . .

132 132 132 132 133 133 133 133 133 133 134 134 136 136 136 137 137 137 137 137 138 138 138

132

12.2. Management of postprandial lipemia 12.3. Potential future therapies . . . . . . 13. Concluding summary . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . .

J. Borén et al. / Clinica Chimica Acta 431 (2014) 131–142

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

1. Introduction Despite a dramatic reduction in mortality from cardiovascular disease (CVD) in recent decades, CVD is still the major killer in the western world. In particular, individuals with obesity-associated disturbances in metabolism, such as insulin resistance and type 2 diabetes (T2D), remain at high risk of cardiovascular events [1]. The mechanisms behind this increased risk are not fully understood. Epidemiological studies have identified non-fasting (postprandial) triglyceride (TG) concentrations as a clinically significant risk factor for CVD [2–4]. TGs are carried in chylomicrons (CMs) and very low-density lipoproteins (VLDLs), which are synthesized in the intestine and liver, respectively. Lipolysis of these triglyceride-rich lipoproteins (TRLs) by lipoprotein lipase (LPL) results in the formation of smaller remnant particles that are TG depleted and enriched in cholesteryl esters [5]. For many years, CMs and CM remnants were thought to be the major culprits in postprandial hyperlipidemia [6,7]. However, the major increase in the postprandial lipoproteins after food intake occurs in the liver-derived VLDL remnant particles [8,9].

2. Chylomicrons and VLDL particles TRLs in the plasma consist of CMs carrying TG from the diet, liverderived VLDLs and their respective remnant particles. TRLs consist of a core of neutral lipids (mainly TG but also some cholesteryl esters) surrounded by a monolayer of phospholipids, free cholesterol and proteins. Each TRL particle contains one molecule of apolipoprotein B (apoB), one of the largest proteins in the mammalian cell and the ligand for the low-density lipoprotein (LDL) receptor [10,11]. ApoB exists in two forms, apoB100 and apoB48, which are coded by the same gene. ApoB48 corresponds to the amino-terminal 48% of apoB100 and is formed in the intestine through editing of apoB100 mRNA by an enzyme called apobec-1. Thus, apoB48 is present on CMs and CM remnants, and apoB100 on VLDL, intermediate-density lipoprotein (IDL) and LDL. Although about 80% of the increase in TGs after a fat-load meal comes from apoB48-containing lipoproteins [8], approximately 80% of the increase in particle number is accounted for by VLDL particles [12,13]. ApoB48- and apoB100-containing particles are cleared from the circulation by common pathways and therefore compete for clearance [14]. Increased secretion of VLDL from the liver is therefore an important predictor of postprandial accumulation of CMs and CM remnants [15].

3. Uptake of dietary fat and formation of CMs The human intestine is equipped to efficiently absorb dietary fat predominantly in the form of TGs. After eating a meal, TG is hydrolyzed by lipase to yield fatty acids (FAs) and monoacylglycerol, which are then absorbed into the enterocytes. Within the enterocyte, FAs can be: (1) used for synthesis of cholesteryl esters or phospholipids; (2) oxidized; (3) re-esterified to form TG for incorporation into CMs; or (4) stored as TGs in cytoplasmic lipid droplets. The multistep assembly of CMs within the enterocyte is not as well characterized as the assembly process of VLDL in hepatocytes, but it is presumed that these processes are similar and dependent on the microsomal triglyceride transfer protein (MTTP) for lipidation of apoB [16–21]. Over the past 10 years, two striking characteristics of enterocyte-TG processing have emerged:[19] (1) lipids secreted at the very onset of a meal are those that were consumed in an earlier meal, suggesting the

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

. . . .

138 138 139 139

presence of an enterocyte storage pool for TG; and (2) a cephalic phase release of CMs is linked to oral stimulation by food intake. The first phenomenon noted is a rise in blood TG concentrations within 10 to 30 min of the consumption of a fat-containing meal [22]. This early rise in TG is termed ‘the early peak’ to delineate it from the primary postprandial peak of blood TG that occurs 3 to 4 h after the start of a meal. The early peak occurs before the meal fat has been absorbed into the enterocytes [23], and is more likely to occur when the previous evening meal was high in fat [23,24]. Parks and coworkers have elegantly shown via utilization of stable isotopes that 10–12% of TGs consumed in the previous evening meal appear in new CMs, first appearing 15 to 20 min after the onset of morning food consumption [19]. This observation indicates that the time between a meal and formation of CMs from TG stored in the intra-enterocyte pool can extend to at least 16 h [24]. The second phenomenon is connected to the overall cephalic phase response [25–28]. The early meal-induced rise in CM-TG concentration can occur when fat is merely tasted and not consumed [29], and also when only glucose is consumed [30]. The existence of an oral taste sensor for lipids is intriguing and has led to the generation of hypotheses on a taste–gut–brain axis, which is currently an active area of investigation [31].

4. Regulation of the secretion of apoB-containing lipoproteins The secretion of TRLs has been extensively studied, but mainly in liver cells. It is today well known that the secretion of VLDL is reduced by insulin, and hepatic insulin resistance is linked to an oversecretion of VLDL [32–38]. Increased liver fat in humans is linked to overproduction of large TG-rich VLDL (VLDL1 particles) [34], which is not surprising given that VLDL formation is dependent on lipid availability. The most common form of liver steatosis is non-alcoholic fatty liver disease [39,40]. This is related to insulin resistance and T2D, and probably explains (at least partly) the dyslipidemia that is observed in subjects with insulin resistance and T2D [41–44]. The results from several groups have clarified at a molecular level that T2D drives VLDL secretion through multiple pathways [38,45]. T2D is characterized by selective insulin resistance: insulin fails to suppress lipolysis in the adipose tissue and FoxO1 in the liver [46], but is still able to activate mammalian target of rapamycin complex 1 (mTORC1) [47,48]. The disinhibition of FoxO1 leads to increased expression of microsomal triglyceride transfer protein (MTTP) and apoCIII, thereby promoting apoB secretion [38,49]. At the same time, the stimulation of mTORC1 in the liver leads to activation of sterol regulatory element binding protein 1c (SREBP-1c) and increased lipogenesis, as well as suppression of sortilin [50]. Because sortilin increases apoB degradation, the suppression of sortilin further promotes apoB secretion [51,52]. Finally, the increase in lipogenesis, coupled with the increased flux of FAs to the liver, expands the cytoplasmic pool of TGs available to lipidate apoB [53], driving the formation of VLDL1 particles. The overall result is increased secretion of apoB in the form of VLDL1 particles in T2D. Thus, the increased level of plasma TGs in T2D is achieved mainly by an increased VLDL1-TG pool that is the effect of an increased number of particles rather than increased particle size [32,36]. The intestinal hormone glucagon-like peptide 1 (GLP-1) is important in the regulation of postprandial glucose levels [20,54–56], and emerging data indicate that GLP-1 also modulates lipid metabolism. GLP-1 has been shown to play an important role in the assembly and secretion of CMs

J. Borén et al. / Clinica Chimica Acta 431 (2014) 131–142

[57]. Pharmacological augmentation of GLP-1 receptor signaling by GLP-1 agonists or dipeptidyl peptidase 4 (DPP-4) inhibition reduces intestinal lipoprotein secretion in experimental studies [58], suggesting that the GLP-1 receptor is essential for the action of GLP-1 on CM metabolism. Furthermore, a recent study in healthy humans showed that the GLP-1 receptor agonist exenatide can suppress intestinal lipoprotein particle production [59]. GLP-2 is produced by posttranslational proteolytic cleavage of proglucagon from intestinal L-cells [60]. In addition to its intestinotropic effects, GLP-2 has been shown to modulate lipid metabolism. In hamsters, administration of human GLP-2 augmented postprandial lipids by increasing CM secretion in vivo and TRL-apoB48 secretion ex vivo in cultured jejunal fragments [61]. This increased apoB48 secretion was not accompanied by enhanced apoB48 synthesis or increased MTTP mRNA expression. Rather, it was attributed to increased FA uptake by posttranslational modification of the FA transport protein CD36. The direct role of GLP-2 on postprandial lipoprotein metabolism in the intestine and liver, independent of its effects on glucagon and gastric acid secretion, remains to be determined. 5. Metabolism of TRLs in the plasma and hepatic uptake 5.1. Intravascular lipolysis of TRLs After secretion of TRLs from the intestine and liver, TGs are removed from the lipoproteins by LPL allowing the delivery of free FAs to muscle and adipose tissue. As the TGs are removed and density increases [62], CMs become CM remnants, and large TG-rich VLDL1 particles become smaller VLDL2 and subsequently IDL. IDL can be further hydrolyzed by hepatic lipase (HL) to LDL, which is catabolized mainly by hepatic uptake of LDL through LDL receptors [63]. Since the TRLs contain a substantial amount of cholesteryl esters, the smaller remnant particles formed by TG hydrolysis are enriched in cholesteryl esters [5]. 5.2. ApoCIII: a key regulator of TRL metabolism ApoCIII inhibits LPL activity and is therefore closely correlated with increased concentrations of serum and total VLDL-TG [64]. Epidemiological studies have demonstrated that apoCIII and LDL-apoCIII independently predict coronary heart disease (CHD) [65,66], and that lifelong deficiency of apoCIII has a cardioprotective effect [67]. The gene expression of apoCIII is decreased by insulin [68,69], peroxisome proliferator-activated receptor-α (PPARα) [70], and farnesoid X receptor (FXR) [71]. In contrast, Caron et al. recently showed that glucose stimulates expression of apoCIII via hepatic nuclear factor-4 (HNF4) and carbohydrate-responsive element binding protein (ChREBP) [72]. Thus, in states of insulin resistance, the inhibitory role of insulin on apoCIII expression may be lost and higher glucose levels would further stimulate apoCIII expression. Increased plasma free FA delivery to the liver would exacerbate this problem, resulting in defective LPL-mediated lipolysis of TRLs and reduced remnant lipoprotein clearance. Thus, dysregulated apoCIII synthesis and secretion could play a major role in the genesis of the diabetic, insulin-resistant dyslipidemia. In addition, accumulation of apoCIII-enriched apoB-containing lipoproteins might further aggravate the CVD risk [73,74]. Paradoxically, despite an increased plasma concentration of apoCIII in subjects with T2D, the concentration of VLDL-apoCIII does not increase in concert with core lipids [75]. Hiukka et al. described a relative deficiency of apoCIII in all TRL species in subjects with T2D [76]. Thus, T2D does not appear to be associated with high concentrations of apoCIII-containing TRLs, despite increased LDL-apoCIII in T2D subjects [74]. 5.3. Hepatic removal of remnant lipoproteins The plasma level of TG depends not only on the synthesis and lipolysis of the TRLs, but also on the removal of the remnant particles.

133

The CM remnants are normally cleared by the liver through binding to syndecan-1 heparan sulfate proteoglycans (HSPGs) and the LDL receptor [77–82]. The importance of the HSPG pathway has been questioned, but compelling evidence now exists proving that dysfunction of the HSPG syndecan-1 in animal models disrupts defective hepatic remnant clearance [77]. Furthermore, Williams and coworkers have shown that accelerated degradation of HSPG following hepatic induction of a heparan sulfate 6-O-endosulfatase (SULF2) in T2D mice suppresses uptake of remnant lipoproteins [83,84]. The relevance of SULF2 in humans has recently been demonstrated in studies showing that the genetic variation rs2281279 in SULF2 associates with postprandial clearance of TG-rich remnant particles and TG levels in healthy subjects [85,86]. In subjects with T2D, hepatic uptake of VLDL, IDL and LDL is also decreased, resulting in increased plasma residence time of these lipoproteins [87–89]. Thus, individuals with insulin resistance exhibit an impaired lipid tolerance with a severely delayed postprandial lipemia due to suppressed removal of TRL remnants. Although CMs and CM remnants were for many years thought to be the major culprits in postprandial hyperlipidemia [6], the VLDL remnant particles account for the major increase in number of postprandial lipoproteins after food intake [8]. Kinetic studies have demonstrated that CM is the preferred substrate over VLDL1 for LPL-mediated clearance [15]. This finding is in line with earlier studies showing that infusion of CM-like TG emulsions leads to accumulation of apoB100 particles [90]. Furthermore, modest induction of LPL activity induces lipolysis of CM but not of VLDL, whereas more pronounced induction of LPL induces hydrolysis of both CM and VLDL [91]. 6. Definition of hypertriglyceridemia The tradition has been to measure TG after an overnight fast and guidelines have defined a desirable ‘normal’ level of fasting serum TG to be b 1.7 mmol/l (b150 mg/dL) [1,92]. The classification of elevated serum TG is generally based on fasting values, which vary in different guidelines [93]. Fasting TG cut-off values for diagnosis of hypertriglyceridemia have varied from 1.7 to 2.3 mmol/l (from 150 mg/dL to 199 mg/dL). Recently, a re-definition of hypertriglyceridemic states has been proposed to simplify the diagnosis of hypertriglyceridemia (Box 1). 7. Genetic variants causing postprandial lipemia As the metabolic machinery of postprandial TRL species comprises several steps at the intestine level, in the circulation and in the liver the genetic control of these processes has been associated with several variants in multiple genes modifying different steps. The current approach has been to look at identified polymorphisms of specific candidate genes regulating the metabolic steps. The small number of subjects and variability of study designs have further hampered the conclusions reflected in the conflicting results observed between different genetic association studies. The majority of reports have focused on genes regulating lipid absorption, apolipoproteins, and clearance pathways. These genes include NPC1L1, FABP2, FATP, CD36, MTTP, APOAI, APOA4, APOA5, APOE, APO5, APOC1 and APOCIII, CETP, ANGPLT4, PLIN, LPL, HL, GPIHBP1, LDLR, SRB1, ABCA1 and TCF7L2. The Box 1 Definition of hypertriglyceridemia. Adapted from reference [183]. Definition

Value of triglycerides

Normal Mild-to-moderate hypertriglyceridemia Severe hypertriglyceridemia

b1.7 mmol/L (150/dL) 1.7–10.0 mmol/L (150–885 mg/dL) N10.0 mmol/L (885 mg/dL)

134

J. Borén et al. / Clinica Chimica Acta 431 (2014) 131–142

results has been recently reviewed [94–96] highlighting the complex genetic basis of postprandial lipemia. Recently Mendelian randomization approach was used to examine the genetic background of non-fasting remnant cholesterol [97]. Genetic variants modulating non-fasting remnant cholesterol and the risk for ischemic heart disease included TRIB1, GCKR and APOA5. 8. Epidemiological and clinical evidence for high TG levels in CVD The significance of fasting and postprandial TGs in CVD has been debated since Zilversmit's proposal in 1979 that CMs and their remnants are atherogenic [98]. Today it is well accepted that, because of their size, most remnant particles cannot cross the endothelium as efficiently as smaller LDL particles [99]. However, since remnant particles not only contain TGs but also approximately 40 times higher levels of cholesteryl esters per particle compared with LDL [99], elevated levels of remnants may lead to accelerated atherosclerosis and CVD (Fig. 1). In the 1970s, the significance of an elevated TG concentration as a risk marker was largely neglected. The skepticism was based on inconsistent epidemiologic associations between fasting TG levels and CVD and also because subjects with familial chylomicromia (type V hyperlipidemia) and extremely high TG levels (up to 25–300 mmol/L) do not demonstrate enhanced vascular disease [100] (although opposite findings in populations with very high TGs also exist [101,102]). In recent years, the recognition of biological pathways, Mendelian randomization studies and large epidemiologic findings support strong links between remnant cholesterol and atherosclerosis [1](and Box 2). Largely because of technical challenges to accurately measure remnant particle concentrations, the controversy still exists as to whether fasting or postprandial TG measurements reflect the atherogenic potential of diverse TRL metabolic pathways and if measurement of serum TG represents an independent risk factor for CVD. Data accumulated from epidemiological studies to date demonstrate that: (1) TG levels are increased among populations with established CVD or with multiple risk factors predisposing to CVD; and (2) follow-up of subjects in prospective population-based studies link postprandial (and, less robustly, fasting) TG and remnant cholesterol to incidence of CVD events or death [1].

Many patients with CVD remain at high risk of events even when the LDL-cholesterol goal has been achieved with lipid-lowering therapy [1,103]. Studies attempting to explain the residual risk have mostly focused on fasting TG measures. The EUROASPIRE III demonstrated that more than a third of CVD patients have elevated TG levels despite many of them receiving antihyperlipidemic treatment [104]. A study from the Swedish diabetes register reported a similar proportion of hypertriglyceridemic subjects among subjects with T2D [105]. Furthermore, the Prospective Cardiovascular Münster (PROCAM) study found that hypertriglyceridemia is twice as prevalent in myocardial infarction survivors compared with control subjects [106]. Together, these data indicate that high TG levels cluster in populations with established CVD despite lipid-lowering therapy. 8.1. Evidence for a link between fasting TG and CVD Several meta-analyses have evaluated the association between fasting TG and CVD, mostly in western populations. Fasting TG was modestly but independently associated with CVD in a meta-analysis of 17 prospective studies with 2900 CHD endpoints [107]. This study demonstrated that 1 mmol/L increase in fasting TG is associated with a 14% increase in the risk of CVD [107]. Sarwar et al. [108] reported data from two nested case–control comparisons from population-based cohorts [the Reykjavik and the European Prospective Investigation of Cancer (EPIC)-Norfolk studies] and included a total of 3582 incident cases of fatal and non-fatal CHD and 6175 controls. The study gave important insights into the long-term stability of log TG values (within-person correlation coefficients of 0.64 [95% confidence interval (CI), 0.60–0.68] over 4 years and 0.63 [95% CI, 0.57–0.70] over 12 years), with variations that were similar to those reported for blood pressure and total serum cholesterol. After adjustment for baseline values of several established risk factors, the odds ratio (OR) for CHD was 1.76 (95% CI, 1.39–2.21) in the Reykjavik study and 1.57 (95% CI, 1.10–2.24) in the EPIC-Norfolk study between individuals in the top and bottom TG tertiles. These results were in line with those reported in an accompanying meta-analysis involving a total of 10,158 incident CHD cases from 262,525 participants from 29 studies (adjusted OR, 1.72; 95% CI,

Fig. 1. Mechanisms for how TRL induce CVD. Atherogenesis is initiated by subendothelial retention of lipoproteins [184,185]. Large VLDL and CM are too large to enter the arterial wall, but smaller remnants and LDL penetrate the arterial intima and bind to artery wall proteoglycans. This explains why patients who are homozygous for rare deleterious mutations in the LPL gene, causing chylomicronemia with extremely high levels of large chylomicrons and VLDLs, have an increased risk of pancreatitis, but not an increased risk of CVD [100]. Remnants may also be involved in the development of CVD by other pathways than by accumulation of cholesterol in the arterial wall. For example, by inducing endothelial dysfunction, such as impaired vasodilation and enhanced inflammatory response (see review [186]). The accumulation of TRLs generates cholesterol-ester enriched small dense HDL and LDL. The HDL is removed from the circulation leading to low HDL cholesterol. The cholesterol-enriched LDL display increased binding affinity to artery wall proteoglycans [187].

J. Borén et al. / Clinica Chimica Acta 431 (2014) 131–142

135

Box 2 Epidemiologic studies reporting non-fasting triglycerides (TG) or remnant lipoproteins and the risk for cardiovascular morbidity and mortality. Study [ref.]

Population

Follow-up

Norwegian counties study [111]

43,641 men and 42, 600 women free of CVD

Prospective, 27 years

Copenhagen Random population sample City Heart of 6391 men and 7581 study women [2,112]

Copenhagen Random population sample City Heart of 6372 men and 7579 study women [113]

The Women's Health study [3]

26,509 initially healthy US women of which 6391 had non-fasting samples

The Framingham study [117] Kugiyama et al. [118]

1567 women offspring of the original Framingham cohort: 83 with and 1484 without CVD 147 consecutive patients with CAD

The Honolulu Heart study [119]

1156 Japanese-American men

Main outcomes

HRs (95%CI) per 1 mmol/L increase in non-fasting TGs for all causes, CVD, IHD, and stroke mortality: Women: 1.16 (1.13–1.20), 1.20 (1.14–1.27), 1.26 (1.19–1.34) and 1.09 (0.96–1.23) Men: 1.03 (1.01–1.04), 1.03 (1.00–1.05), 1.03 (1.00–1.06) and 0.99 (0.92–1.07). Prospective, HRs (95%CI) for total mortality 31 years by non-fasting TGs: (TG b 1 mmol/L: HR 1) TG 1.0–1.99 mmol/L: 1.1 (95%CI: 1.0–1.2) in women and 1.1 (95%CI: 1.1–1.2) in men TG 2.0–2.99 mmol/L: 1.3 (95%CI: 1.2–1.4) in women and 1.2 (95%CI: 1.1–1.4) in men TG 3.0–3.99 mmol/L: 1.4 (95%CI: 1.2–1.7) in women and 1.3 (95%CI: 1.1–1.4) in men TG 4.0–4.99 mmol/L: 1.4 (95%CI: 1.1–1.9) in women and 1.4 (95%CI: 1.2–1.6) in men TG N 5 mmol/L: 2.0 (95%CI: 1.5–2.7) in women and 1.5 (95%CI: 1.2–1.7) in men Prospective, HRs (95%CI) for ischemic stroke 33 years by non-fasting TGs: (TG b 1 mmol/L: HR 1) TG 1.0–1.99 mmol/L: 1.2 (95%CI: 0.9–1.7) in women and 1.2 (95%CI: 0.8–1.7) in men TG N 5 mmol/L: 3.9 (95%CI: 1.3–11.1) in women and 2.3 (95%CI: 1.2–4.3) in men Prospective, HR for CVD event by non-fasting 11 years TG: 2nd tertile: 1.44 (95% CI 0.90–2.29) 3rd tertile: 1.98 (95% CI 1.21–3.25) Cross-sectional RLP-chol + 15.6%; P b 0.0001 and RLP-TG +27.0%; P b 0.0002 in women with prevalent CVD Prospective OR for developing coronary follow-up until event: coronary event or 2nd tertile of remnant levels 2.43 36 months (95%CI: 1.1–5.8) 3rd tertile of remnant levels 6.38 (95%CI:2.3–17.6) Prospective, RLP-chol (P = 0.0022) and RLP17 years TG (P = 0.0045) predicted CHD risk independent of non-lipid risk factors

Remarks Adjustment for major cardiovascular risk factors attenuated the effect

The best predictor for MI in women was non-fasting TG and in men non-fasting cholesterol

The remnant cholesterol increased stepwise as a function of non-fasting TG and cholesterol in cross-sectional analysis of 53,629 subjects (see also Fig. 1.)

TG measured 2 to 4 h postprandially had the strongest association with CVD events (fully adjusted HR [95% CI] for highest vs. lowest tertiles of levels, 4.48 [1.98–10.15] [P b .001 for trend]) Adjusted RLP-chol was significantly associated with prevalent CVD in women in logistic regression analysis Remnant levels were independent predictors of future coronary event in multivariate model

RLP-chol and RLP-TG were only significant lipid predictors of CHD in models with total, HDL and LDL cholesterols as additional covariates

CI, confidence interval; CHD, coronary heart disease; CVD, cardiovascular disease; HR, hazard ratio; MI, myocardial infarction; OR, odds ratio; RLP, remnant-like particles.

136

J. Borén et al. / Clinica Chimica Acta 431 (2014) 131–142

1.56–1.90) [108]. The Emerging Risk Factors Collaboration meta-analysis [109] of 68 prospective long-term studies with more than 302,000 people and 2.79 million years of follow-up demonstrated comparable hazard ratios (HRs) but could not confirm an independent role of TG as a CVD risk factor. This study demonstrated that the HR for CHD with triglyceride was 1.37 (95% CI, 1.31–1.42) after adjustment for non-lipid risk factors, but that it was reduced to 0.99 (95% CI, 0.94–1.05) after further adjustment for high-density lipoprotein (HDL)-cholesterol and nonHDL-cholesterol. Adjusted HRs in subgroups of fasted [1.02 (95% CI 0.95–1.09)] and non-fasted [0.92 (95% CI 0.82–1.03)] subjects did not differ [109]. Observations in Asian Pacific populations are in line with those in western populations [110]. Interestingly, decreased TG values during follow-up were associated with lowered CVD risk in young men. These studies together indicate moderate but highly significant associations between fasting TG values and CHD risk. 8.2. Evidence for a link between non-fasting TG and CVD Prospective epidemiologic studies on non-fasting TG levels in response to normal food intake include three large-scale studies, the Copenhagen City Heart study [2], Women's Health study [3] and the Norwegian counties study [111], all of which found highly significant associations between HRs for cardiovascular events with increases in non-fasting TG. Although the variable timing of lipid sampling and nutrient exposure within and between these studies may attenuate the effect size, these studies were able to provide quantitative information about exposure to postprandial remnant particles and the risk for CVD. The Copenhagen City Heart study [2], a prospective study with 26–31 years' follow-up of 13,000 untreated individuals from the Danish general population, found that non-fasting TG levels above 5 mmol/L (compared with values under 1 mmol/L in age-adjusted analyses) were associated with a 17-fold (women) and five-fold (men) increased risk of myocardial infarction and a four-fold (women) and two-fold (men) increased risk of early death. Furthermore, the findings in the 31 year follow-up of this study were in line with previous data with non-fasting cholesterol and TG being similarly associated with increased risk of myocardial infarction and ischemic heart disease [112], and non-fasting TG also with ischemic stroke [113]. Unexpectedly, elevated non-fasting TG, but not non-fasting cholesterol, was associated with total mortality [112]. In the Women's Health study [3], comprising 26,509 initially healthy US women and 6391 non-fasting samples, the follow-up demonstrated

Lipoprotein cholesterol,mmol/L

7.0

9. Epidemiological evidence for remnant particles in CVD The estimate of circulating atherogenic particles is improved by measuring non-fasting remnant lipoprotein cholesterol in addition to TG. The strongest evidence and validation of non-fasting lipid measurements are from the Copenhagen City Heart study [2,114]. After 31 years of follow-up, non-fasting cholesterol remained the best predictor of myocardial infarction in men [112]. Remnant-like particles (RLP) are isolated from serum samples by the immune adsorption method with the monoclonal antibodies to apoAI and apoB [115], and measurement of fasting RLP-cholesterol may be used as a surrogate to estimate circulating remnant particles [116]. In a subset of 1567 women from the Framingham Heart study, RLPcholesterol was independently associated with prevalent coronary artery disease (CAD) [117]. Furthermore, high levels of RLPcholesterol were predictive for future events independent of other risk factors in a Japanese population with CAD [118]. The Honolulu Heart study [119], with 1156 Japanese-American men and 17 years of follow-up, showed that TG, RLP-TG and RLP-cholesterol predicted CHD independently of other lipid risk factors. In this study, log fasting TG correlated strongly with both RLP-TG and RLPcholesterol (r = 0.93 and r = 0.88, respectively) suggesting that RLP measurements do not add to the prognostic value of fasting TG [119]. An association between remnant particles and ischemic stroke has been analyzed only indirectly in the Copenhagen City Heart study. A stepwise increase in LDL-cholesterol only above levels of 9 mmol/L predicted ischemic stroke in men but not women. Instead, the adjusted non-fasting TG levels correlated with ischemic stroke and remnant cholesterol concentrations, suggesting that TG as a marker for remnant cholesterol should be the treatment target in these patients [113]. 10. Postprandial lipids in populations with CVD: case–control studies

6.0 5.0 4.0 3.0 2.0 1.0 0.0

TG, mmol/L < 1 Number 14906

a strong independent relationship between non-fasting TG levels and cardiovascular events. In fully adjusted models, the HRs for increasing tertiles of non-fasting TG levels were 1.44 (95% CI 0.90–2.29) and 1.98 (95% CI 1.21–3.25). In contrast, fasting TG levels showed little independent relationship. Finally, in the Norwegian counties study [111], an association between non-fasting TG and risk of CVD death was observed in women but not men after controlling for risk factors other than HDL-cholesterol. The Copenhagen City Heart and the Norwegian counties studies both found non-fasting TG to predict future CVD better in women than men, which is in line with the robust data from the Women's Health study. Together, these studies underline the epidemiological link between non-fasting TG and both events and death due to vascular causes.

1–1.99

2–2.99

3–3.99

4–4.99

5–5.99

28,041

10,722

3,826

1,418

982

Fig. 2. Lipoprotein cholesterol as a function of increasing levels of non-fasting triglycerides. Red = remnant cholesterol, blue = LDL-cholesterol and green = HDL-cholesterol. Adapted from reference [97].

Case–control studies comparing the magnitude and time-frame of postprandial lipemia in CHD patients are few and limited in number of subjects. These studies used highly variable fat loads, sampling times and methodology for analysis, and are therefore not directly comparable. Data accumulated from rather small studies on CVD clearly demonstrate abnormal postprandial TG and remnant particle metabolism in both subjects with vascular disease and their relatives (reviewed in [94]). The main findings of these studies are that CAD is associated with delayed TG and retinyl-ester clearance (a sign of impaired CM remnant metabolism) [120]. More specifically, late TG responses 6 and 8 h after a meal are 68% accurate in predicting the presence or absence of CAD by logistic regression [121]. However, in subjects with T2D, the findings are more conflicting (reviewed in [122]). More recently, fasting apoB48 measurements have been reported to correlate with prevalence of CAD [123] and to predict both new-onset and chronic CAD [124].

J. Borén et al. / Clinica Chimica Acta 431 (2014) 131–142

B 15

10

5

0 0

5

10

15

Plasma ApoB48 (Elisa) (mg/dl)

Total lipoprotein ApoB48 (SDS PAGE) (mg/dl*h)

Total lipoprotein ApoB48 (SDS PAGE) (mg/dl)

A

137

150

100

50

0 0

50

100

150

Plasma ApoB48 (Elisa) (mg/dl*h)

Fig. 3. Comparison of total apoB48 in plasma measured by ELISA, and quantification of apoB48 in the TRL fractions measured by SDS-PAGE (sum of IDL, VLDL2, VLDL1 and chylomicron fractions). 10 subjects were studied at 6 time points (0, 2, 3, 4, 6 and 8 h) after a standardized mixed meal. Panel A shows the correlation between the data point at 4 h for each individual (r = 0.59, P b 0.01). Panel B shows the correlation between the calculated area under the curve (AUC) (r = 0.88, P b 0.001) (unpublished data).

11. Assessment of postprandial TRL metabolism Emerging evidence in support of the power of non-fasting TG levels to predict CVD risk and postprandial dysmetabolism identifies an urgent need to define accurate and standardized methodology to assess different components of TRLs in the postprandial state. The heterogeneity of TRLs with respect to size and apolipoprotein composition represents a challenge. In addition, there is a need to set clinical reference standards for each component, and the methodology to measure these markers should be applicable to large-scale clinical intervention trials. 11.1. Measurement of non-fasting TG levels Recent indications suggest that non-fasting TG concentration is a potentially good surrogate marker that reflects the burden of postprandial dysmetabolism [114,125]. However, the use of non-fasting TG values in clinical practice has been hampered by a lack of standardization of non-fasting TG measures (with respect to the time since last meal) and population-based reference values. It should be recognized that intra-individual variation of plasma TG values is between 15 and 30% [126] and seems to increase during the day [127]. Recent data have demonstrated that changes of non-fasting TG values taken at random time-points after normal food intake seem to be modest with a mean maximum increase of 0.3 mmol/L [125]. This contrasts with the observed increase of 1–2 mmol/L after a fat tolerance test [128,129]. An expert panel statement has proposed that a non-fasting TG concentration of b2.0 mmol/(180 mg/dL) is considered to be desirable [129]. If non-fasting TG levels exceed 2.2 mmol/L (200 mg/dL), further testing by repeat measurements of fasting TG or a fat tolerance test is recommended [92,129]. 11.2. Calculated remnant cholesterol Recently, the calculated remnant cholesterol values in non-fasting state have been introduced as a surrogate market of postprandial TRL load. This parameter comprises all TRL particles, including not only VLDL and CMs but also their remnants [130]. The equation for the calculation is: remnant cholesterol = total cholesterol − [LDL-cholesterol and HDL-cholesterol]. LDL-cholesterol is recommended to be measured using a direct assay but can also be estimated using the Friedewald equation. In a study of 73,513 subjects from Copenhagen, remnant cholesterol values correlated strongly with plasma TG (Fig. 2). Remnant cholesterol values associated negatively with nonfasting HDL-cholesterol but the strength of the correlation with LDLcholesterol was less robust as remnant cholesterol showed more variability than respective LDL-cholesterol values [97]. A 1 mmol/l (39 mg/dL) increase in non-fasting remnant cholesterol is associated with a 2.8-fold increased risk of ischemic heart disease [97]. Thus,

calculated remnant cholesterol emerges as a useful parameter for CVD risk prediction as its assessment is easy in clinical practice and allows analyses of many samples in large clinical trials. 11.3. Oral fat tolerance test The oral fat tolerance test (OFTT) has been used to follow the dynamics of postprandial lipoprotein metabolism in parallel with the use of the oral glucose tolerance (OGTT) test to evaluate glucose tolerance. The major determinants of the postprandial TG response to a test meal are the ambient fasting concentration of serum TG and the fat content and quality of the meal. However, the composition of the fat-rich test meal has been highly variable, from a mixed meal mimicking a rich breakfast to liquid cream shakes, making it difficult to compare the different studies. Based on a systematic meta-analysis of 119 studies [131], an OFTT meal consisting of 75 g fat is recommended to be served after an 8 hour fast. In normal subjects, postprandial TG values increase for up to 3 or 4 h after a meal and remain elevated for up to 6 or 8 h. The response is exaggerated and delayed in subjects with various metabolic disorders or genetic variants causing hypertriglyceridemia. The TG response to an OFTT is commonly analyzed from sequential blood samples (taken every 1 or 2 h for up to 6 or 8 h) as the area under the curve (AUC) or incremental AUC (iAUC) (i.e. ignoring the area beneath the fasting concentration). The fasting TG measurement is represented by the lowest TG value in a setting of regular daily meals and snacks. In a normal population, a strong correlation exists between the fasting and postprandial serum TG response. The lack of a standardized meal and sampling times has hampered the assessment of the postprandial TG response and no standard reference ranges exist for either TG-AUC or TG-iAUC values. Thus, no consensus exists on the definition and assessment of postprandial lipemia in contrast to the widely used OGTT to diagnose glucose tolerance. However, data from the meta-analysis indicates that a single TG measurement 4 h after an OFTT can be used for assessment of postprandial lipemia, and that a TG concentration b2.5 mmol/L (220 mg/dL) can be considered a desirable postprandial TG value [129]. 11.4. Alternative markers of postprandial lipemia Two methods to quantitate RLP-cholesterol and RLP-TG are available [132]. The first assay is based on immune-specific separation of particles containing apoB100 and apoA-I using antibodies coupled to Sepharose4B [115,133]. All apoB48-containing particles and partly lipolyzed apoB100-containing VLDL remnants are recovered in the RLP fraction, and both cholesterol and TG content are measured [134]. Both RLPcholesterol and RLP-TG have been shown to increase after an OFFT. Interestingly, VLDL remnants rather than CM remnants are the major

138

J. Borén et al. / Clinica Chimica Acta 431 (2014) 131–142

contributors to RLP-TG [135]. Although RLP-cholesterol has been reported to predict cardiovascular events in patients with CAD, this assay has not been used to any great extent in clinical practice [136]. The second assay system uses a specific detergent to modify CM and VLDL remnants to allow direct measurements of cholesterol content in these particles [137,138]. However, the available data based on this specific assay are currently limited [139]. 11.5. Measurement of apoB48 The measurement of apoB48 reflects the number of CM particles as each CM carries a single molecule of apoB48 [140]. Thus, apoB48 is an exclusive marker to reflect the dynamics of CMs and their remnants. The major problem in the quantification of apoB48 is that its concentration in fasting plasma is very low compared with apoB100. As expected, plasma apoB48 concentrations increase in parallel with those of plasma TG following an OFTT [141]. The most common method to quantitate apoB involves sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) combined with Coomassie Brilliant Blue to stain the apoB48 and apoB100 bands [142]. This method enables measurement of apoB48 in different TRL fractions [Svedberg flotation rate (Sf) N400, Sf 60–400 and Sf 20–60] [143]. The sensitivity can be improved by immunoblotting with monoclonal and polyclonal antibodies to apoB48 [144]. However, the method is impractical to measure apoB48 in large population samples because of the time-consuming procedure of gel casting combined with different techniques for gel scanning that result in major variability between laboratories. Several enzyme-linked immunosorbent assays (ELISAs) have been introduced that allow the measurement of apoB48 in both fasting and postprandial samples [145,146]. However, the low concentration of apoB48 (about 20% of the total apoB100) hampers the measurements of apoB48 in different TRL subspecies [147]. The lack of internationally recognized standardized assays and population-based reference ranges has so far prevented the utilization of apoB48 measurements in clinical practice and randomized trials. However, the development of ELISAs with increased sensitivity and accuracy indicates their potential for use in large clinical cohorts. We recently used an ELISA to measure plasma total apoB48 in response to a standardized mixed meal and showed that the plasma values correlated well with apoB48 in the TRL fractions measured by SDS-PAGE (Fig. 3) (Matikainen et al. Unpublished results). 12. Current and future therapeutic approaches 12.1. Management of hypertriglyceridemia Current guidelines acknowledge elevated fasting and postprandial TG levels as important risk factors for CVD [1,148]. It is also well established that the risk of pancreatitis is clinically significant if plasma TG values exceed 10 mmol/L, and that subjects with such high TG levels need immediate pharmacotherapy in addition to intense dietary changes and restriction of calories and fat content in the diet. Data from randomized clinical trials on the clinical benefits of lowering TG are much less robust than for the benefits of lowering LDL-cholesterol with statins [148]. Importantly, the lack of high-grade evidence for benefits of TG-lowering drugs is reflected in the lack of specific target goals for both fasting and postprandial lipid values. Consequently, the control of secondary factors and lifestyle modification are considered to be the first-line approach of the clinical management of both fasting hypertriglyceridemia and postprandial lipemia. Appropriate dietary changes limiting fat content, caloric restriction resulting in weight loss, restriction of alcohol intake and increased exercise are fundamental for management of hypertriglyceridemia [1,148–151]. There are multiple examples of beneficial effects of lifestyle modifications on postprandial dysmetabolism, but the data are based on relatively small case–control studies [149–151]. As statin

treatment is the well-established therapy to reduce CVD risk and outcomes, elevated LDL-cholesterol is also the primary target for lipidlowering therapy in subjects with hypertriglyceridemia. Consequently, intensification of statin therapy is recommended as the first pharmacological step to reduce elevated TG levels in individuals with high or very high CVD risk based on global risk assessment, particularly in subjects with the metabolic syndrome and diabetes [148,152]. Overall, statin therapy decreases TG levels modestly but reductions of up to 30% may be seen depending on baseline TG level and the dose of statin [153]. Fibrates, niacin and 3-omega fatty acids have been commonly used to reduce plasma TG, particularly in subjects with high residual risk on statin therapy [1,149,150]. The clinical utility of niacin has been questioned following results from both the AIM-HIGH [154] and the HPS2-THRIVE [155] studies, and the European Medicines Agency (EMA) recently suspended niacin/laropiprant products in Europe [152,156]. Likewise, data from randomized clinical trials of 3-omega fatty acids have not confirmed clinical benefits on CVD outcomes [157,158]. Thus, fibrates remain the only available option as add-on therapy to statins to reduce the residual risk, and evidence in support of these drugs is from subgroup analyses of the ACCORD [159] and FIELD [160] trials as well as meta-analyses [161]. 12.2. Management of postprandial lipemia Current guidelines do not stipulate the atherogenic lipid profile in the postprandial state as a target for therapy nor do they give any target values for the parameters of postprandial lipemia. This is because randomized clinical trials of lipid-lowering therapy have not specified any of the biomarkers for postprandial lipemia as an endpoint. The available data from lipid-lowering studies are from small and shortterm case–control studies and do not provide high-grade evidence suitable for guidelines, although the results may be useful for treating individual patients. Fibrate studies have given consistent results reporting significant lowering of postprandial TG levels in response to a standardized fat challenge [129,151]. Furthermore, fenofibrate also reduces apoB48 and biomarkers for remnants [162]. Recently, the lipid response was assessed in a pre-specified subgroup (n = 139) of the ACCORD study [163]. The repeat measurement of biomarkers was performed after at least four months on fenofibrate or placebo added to the statin therapy. Fenofibrate therapy significantly reduced both the responses of plasma TG and apoB48 measured as the iAUC. Available data indicate that statins also reduce postprandial TG values [129], but the effect seems to be variable depending on the dose and the efficacy of the statin. Recent data suggest that ezetimibe, which exerts its effect on cholesterol lowering by inhibition of cholesterol absorption, also reduces postprandial lipemia [164–166]. Interestingly, ezetimibe has been reported to reduce apoB48 secretion in humans, suggesting a direct effect on TRL metabolism in enterocytes [167]. Although fenofibrate seems to be the best option to add on to statin therapy for reduction of postprandial lipemia, there is an urgent need for large-scale intervention studies specifying biomarkers of postprandial lipemia as a therapeutic target and assessing the benefits on cardiovascular endpoints. 12.3. Potential future therapies Emerging evidence suggests that incretin-based therapies may not only improve glucose metabolism but may also influence postprandial lipid metabolism. GLP-1 and -2, which are secreted after meals and modulate insulin secretion [168,169], have both been reported to participate in the regulation of intestinal CM secretion [170]. Pharmacological augmentation of GLP-1 receptor signaling by GLP-1 agonists or DPP-4 inhibition reduces intestinal lipoprotein secretion in experimental studies, suggesting that incretin-based therapies may ameliorate

J. Borén et al. / Clinica Chimica Acta 431 (2014) 131–142

dyslipidemia and thus improve cardiovascular risk profile in patients with T2D. Recently, both GLP-1 agonists and DPP-4 inhibitors have been reported to suppress the TG and apoB48 responses to a standardized fat-rich meal in patients with T2D [56,59,171–174]. Thus, the available data suggest that the incretin-based therapies have a class effect on intestinal lipid metabolism and offer a potential strategy to reduce postprandial lipemia [175]. Improved understanding of the physiology and genetic regulation of postprandial lipid metabolism may identify new targets for therapy [97,176]. This prospect is highlighted by the genetic approaches to target LPL expression as well as apoCIII, apoB and proprotein convertase subtilisin/kexin type 9 (PCSK9) [150]. Recently, intramuscular injection of the human LPL S447X has been approved by the EMA [177]. Furthermore, administration of the alipogene tiparvovec reduced markedly postprandial lipemia in patients with LPL deficiency [178–181]. Antisense oligonucleotide inhibition of apoCIII has also been reported to reduce plasma TG in rodents and primates (both non-human and human) [182]. 13. Concluding summary In the postprandial state, TRLs comprise CMs synthesized in the intestine, VLDL particles produced in the liver and their cholesterol-rich remnants. Postprandial hypertriglyceridemia can be initiated by both overproduction and/or defective catabolism of TRLs and is caused by both genetic variations and non-genetic factors such as obesity or T2D. Remnants can accumulate in the arterial wall and will deliver more cholesterol than LDL particles. Recent data strongly indicate that both non-fasting triglycerides and remnant cholesterol are not only CHD risk factors but are causally linked to atherogenesis. To date, no randomized clinical trials have addressed the effects of TRL-lowering measures on CVD outcomes, thus hampering recommendations for management. This is partly due to the lack of consensus on clinical measures of biomarkers to screen and assess the postprandial burden of TRLs. Likewise, the failure of clinical intervention trials that aimed to reduce CVD endpoints by lowering plasma TG has raised the urgent need for more extensive clinical endpoint data specifically testing the benefits of lowering postprandial hypertriglyceridemia and remnant cholesterol. However, novel therapy concepts utilizing anti-sense oligonucleotides or siRNA to target regulatory genes of postprandial lipid metabolism provide hope for future management. References [1] Chapman MJ, Ginsberg HN, Amarenco P, et al. Triglyceride-rich lipoproteins and high-density lipoprotein cholesterol in patients at high risk of cardiovascular disease: evidence and guidance for management. Eur Heart J 2011;32:1345–61. [2] Nordestgaard BG, Benn M, Schnohr P, Tybjaerg-Hansen A. Nonfasting triglycerides and risk of myocardial infarction, ischemic heart disease, and death in men and women. JAMA 2007;298:299–308. [3] Bansal S, Buring JE, Rifai N, Mora S, Sacks FM, Ridker PM. Fasting compared with nonfasting triglycerides and risk of cardiovascular events in women. JAMA 2007;298:309–16. [4] Freiberg JJ, Tybjaerg-Hansen A, Jensen JS, Nordestgaard BG. Nonfasting triglycerides and risk of ischemic stroke in the general population. JAMA 2008;300:2142–52. [5] Dallinga-Thie GM, Franssen R, Mooij HL, et al. The metabolism of triglyceride-rich lipoproteins revisited: new players, new insight. Atherosclerosis 2010;211:1–8. [6] Zilversmit DB. A proposal linking atherogenesis to the interaction of endothelial lipoprotein lipase with triglyceride-rich lipoproteins. Circ Res 1973;33:633–8. [7] Moreton JR. Atherosclerosis and alimentary hyperlipidemia. Science 1947;106:190–1. [8] Cohn JS, Johnson EJ, Millar JS, et al. Contribution of apoB-48 and apoB-100 triglyceride-rich lipoproteins (TRL) to postprandial increases in the plasma concentration of TRL triglycerides and retinyl esters. J Lipid Res 1993;34:2033–40. [9] Nakajima K, Nakano T, Tokita Y, et al. Postprandial lipoprotein metabolism: VLDL vs chylomicrons. Clin Chim Acta Int J Clin Chem 2011;412:1306–18. [10] Olofsson SO, Wiklund O, Boren J. Apolipoproteins A-I and B: biosynthesis, role in the development of atherosclerosis and targets for intervention against cardiovascular disease. Vasc Health Risk Manag 2007;3:491–502. [11] Olofsson SO, Boren J. Apolipoprotein B: a clinically important apolipoprotein which assembles atherogenic lipoproteins and promotes the development of atherosclerosis. J Intern Med 2005;258:395–410.

139

[12] Karpe F, Bell M, Bjorkegren J, Hamsten A. Quantification of postprandial triglyceriderich lipoproteins in healthy men by retinyl ester labeling and simultaneous measurement of apolipoproteins B-48 and B-100. Arterioscler Thromb Vasc Biol 1995;15:199–207. [13] Schneeman BO, Kotite L, Todd KM, Havel RJ. Relationships between the responses of triglyceride-rich lipoproteins in blood plasma containing apolipoproteins B-48 and B-100 to a fat-containing meal in normolipidemic humans. Proc Natl Acad Sci U S A 1993;90:2069–73. [14] Brunzell JD, Hazzard WR, Porte Jr D, Bierman EL. Evidence for a common, saturable, triglyceride removal mechanism for chylomicrons and very low density lipoproteins in man. J Clin Invest 1973;52:1578–85. [15] Adiels M, Matikainen N, Westerbacka J, et al. Postprandial accumulation of chylomicrons and chylomicron remnants is determined by the clearance capacity. Atherosclerosis 2012;222:222–8. [16] Boren J, Taskinen MR, Olofsson SO, Levin M. Ectopic lipid storage and insulin resistance: a harmful relationship. J Intern Med 2013;274:25–40. [17] Olofsson SO, Boren J. Apolipoprotein B secretory regulation by degradation. Arterioscler Thromb Vasc Biol 2012;32:1334–8. [18] Abumrad NA, Davidson NO. Role of the gut in lipid homeostasis. Physiol Rev 2012;92:1061–85. [19] Lambert JE, Parks EJ. Postprandial metabolism of meal triglyceride in humans. Biochim Biophys Acta 1821;2012:721–6. [20] Xiao C, Lewis GF. Regulation of chylomicron production in humans. Biochim Biophys Acta 1821;2012:736–46. [21] Ramasamy I. Recent advances in physiological lipoprotein metabolism. Clin Chem Lab Med 2013:1–33. [22] Mattes RD. Oral fat exposure increases the first phase triacylglycerol concentration due to release of stored lipid in humans. J Nutr 2002;132:3656–62. [23] Robertson MD, Henderson RA, Vist GE, Rumsey RD. Extended effects of evening meal carbohydrate-to-fat ratio on fasting and postprandial substrate metabolism. Am J Clin Nutr 2002;75:505–10. [24] Chavez-Jauregui RN, Mattes RD, Parks EJ. Dynamics of fat absorption and effect of sham feeding on postprandial lipema. Gastroenterology 2010;139:1538–48. [25] Hiraoka T, Fukuwatari T, Imaizumi M, Fushiki T. Effects of oral stimulation with fats on the cephalic phase of pancreatic enzyme secretion in esophagostomized rats. Physiol Behav 2003;79:713–7. [26] Mattes RD. Is there a fatty acid taste? Annu Rev Nutr 2009;29:305–27. [27] Ahren B, Holst JJ. The cephalic insulin response to meal ingestion in humans is dependent on both cholinergic and noncholinergic mechanisms and is important for postprandial glycemia. Diabetes 2001;50:1030–8. [28] Teff KL, Engelman K. Oral sensory stimulation improves glucose tolerance in humans: effects on insulin, C-peptide, and glucagon. Am J Physiol 1996;270:R1371–9. [29] Mattes RD. Brief oral stimulation, but especially oral fat exposure, elevates serum triglycerides in humans. Am J Physiol Gastrointest Liver Physiol 2009;296:G365–71. [30] Robertson MD, Parkes M, Warren BF, et al. Mobilisation of enterocyte fat stores by oral glucose in humans. Gut 2003;52:834–9. [31] Khan NA, Besnard P. Oro-sensory perception of dietary lipids: new insights into the fat taste transduction. Biochim Biophys Acta 2009;1791:149–55. [32] Adiels M, Olofsson SO, Taskinen MR, Boren J. Overproduction of very low-density lipoproteins is the hallmark of the dyslipidemia in the metabolic syndrome. Arterioscler Thromb Vasc Biol 2008;28:1225–36. [33] Adiels M, Westerbacka J, Soro-Paavonen A, et al. Acute suppression of VLDL1 secretion rate by insulin is associated with hepatic fat content and insulin resistance. Diabetologia 2007;50:2356–65. [34] Adiels M, Taskinen MR, Packard C, et al. Overproduction of large VLDL particles is driven by increased liver fat content in man. Diabetologia 2006;49:755–65. [35] Adiels M, Olofsson SO, Taskinen MR, Boren J. Diabetic dyslipidaemia. Curr Opin Lipidol 2006;17:238–46. [36] Adiels M, Packard C, Caslake MJ, et al. A new combined multicompartmental model for apolipoprotein B-100 and triglyceride metabolism in VLDL subfractions. J Lipid Res 2005;46:58–67. [37] Adiels M, Boren J, Caslake MJ, et al. Overproduction of VLDL1 driven by hyperglycemia is a dominant feature of diabetic dyslipidemia. Arterioscler Thromb Vasc Biol 2005;25:1697–703. [38] Haas ME, Attie AD, Biddinger SB. The regulation of ApoB metabolism by insulin. Trends Endocrinol Metab 2013;24:391–7. [39] Musso G, Gambino R, Cassader M. Recent insights into hepatic lipid metabolism in non-alcoholic fatty liver disease (NAFLD). Prog Lipid Res 2009;48:1–26. [40] Ratziu V, Bellentani S, Cortez-Pinto H, Day C, Marchesini G. A position statement on NAFLD/NASH based on the EASL 2009 special conference. J Hepatol 2010;53: 372–84. [41] Kissebah AH, Alfarsi S, Evans DJ, Adams PW. Integrated regulation of very low density lipoprotein triglyceride and apolipoprotein-B kinetics in non-insulindependent diabetes mellitus. Diabetes 1982;31:217–25. [42] Duvillard L, Pont F, Florentin E, Galland-Jos C, Gambert P, Verges B. Metabolic abnormalities of apolipoprotein B-containing lipoproteins in non-insulin-dependent diabetes: a stable isotope kinetic study. Eur J Clin Invest 2000;30:685–94. [43] Ouguerram K, Magot T, Zair Y, et al. Effect of atorvastatin on apolipoprotein B100 containing lipoprotein metabolism in type-2 diabetes. J Pharmacol Exp Ther 2003;306:332–7. [44] Cummings MH, Watts GF, Umpleby AM, et al. Increased hepatic secretion of very-lowdensity lipoprotein apolipoprotein B-100 in NIDDM. Diabetologia 1995;38:959–67. [45] Sparks JD, Sparks CE, Adeli K. Selective hepatic insulin resistance, VLDL overproduction, and hypertriglyceridemia. Arterioscler Thromb Vasc Biol 2012;32:2104–12. [46] Kamagate A, Dong HH. FoxO1 integrates insulin signaling to VLDL production. Cell Cycle 2008;7:3162–70.

140

J. Borén et al. / Clinica Chimica Acta 431 (2014) 131–142

[47] Wan M, Leavens KF, Saleh D, et al. Postprandial hepatic lipid metabolism requires signaling through Akt2 independent of the transcription factors FoxA2, FoxO1, and SREBP1c. Cell Metab 2011;14:516–27. [48] Leavens KF, Birnbaum MJ. Insulin signaling to hepatic lipid metabolism in health and disease. Crit Rev Biochem Mol Biol 2011;46:200–15. [49] Kamagate A, Qu S, Perdomo G, et al. FoxO1 mediates insulin-dependent regulation of hepatic VLDL production in mice. J Clin Invest 2008;118:2347–64. [50] Ai D, Baez JM, Jiang H, et al. Activation of ER stress and mTORC1 suppresses hepatic sortilin-1 levels in obese mice. J Clin Invest 2012;122:1677–87. [51] Strong A, Ding Q, Edmondson AC, et al. Hepatic sortilin regulates both apolipoprotein B secretion and LDL catabolism. J Clin Invest 2012;122:2807–16. [52] Strong A, Rader DJ. Sortilin as a regulator of lipoprotein metabolism. Curr Atheroscler Rep 2012;14:211–8. [53] Andersson L, Bostrom P, Ericson J, et al. PLD1 and ERK2 regulate cytosolic lipid droplet formation. J Cell Sci 2006;119:2246–57. [54] Holst JJ, Vilsboll T, Deacon CF. The incretin system and its role in type 2 diabetes mellitus. Mol Cell Endocrinol 2009;297:127–36. [55] Van Gaal LF, Gutkin SW, Nauck MA. Exploiting the antidiabetic properties of incretins to treat type 2 diabetes mellitus: glucagon-like peptide 1 receptor agonists or insulin for patients with inadequate glycemic control? Eur J Endocrinol 2008;158:773–84. [56] Ussher JR, Drucker DJ. Cardiovascular biology of the incretin system. Endocr Rev 2012;33:187–215. [57] Xiao C, Hsieh J, Adeli K, Lewis GF. Gut–liver interaction in triglyceride-rich lipoprotein metabolism. Am J Physiol Endocrinol Metab 2011;301:E429–46. [58] Hsieh J, Longuet C, Baker CL, et al. The glucagon-like peptide 1 receptor is essential for postprandial lipoprotein synthesis and secretion in hamsters and mice. Diabetologia 2010;53:552–61. [59] Xiao C, Bandsma RH, Dash S, Szeto L, Lewis GF. Exenatide, a glucagon-like peptide-1 receptor agonist, acutely inhibits intestinal lipoprotein production in healthy humans. Arterioscler Thromb Vasc Biol 2012;32:1513–9. [60] Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology 2007;132:2131–57. [61] Hsieh J, Longuet C, Maida A, et al. Glucagon-like peptide-2 increases intestinal lipid absorption and chylomicron production via CD36. Gastroenterology 2009;137: 997–1005 1005 e1001-1004. [62] Goldberg IJ. Lipoprotein lipase and lipolysis: central roles in lipoprotein metabolism and atherogenesis. J Lipid Res 1996;37:693–707. [63] Brown MS, Goldstein JL. A receptor-mediated pathway for cholesterol homeostasis. Science 1986;232:34–47. [64] Marcoux C, Tremblay M, Fredenrich A, Davignon J, Cohn JS. Lipoprotein distribution of apolipoprotein C-III and its relationship to the presence in plasma of triglyceriderich remnant lipoproteins. Metabolism 2001;50:112–9. [65] Sacks FM, Alaupovic P, Moye LA, et al. VLDL, apolipoproteins B, CIII, and E, and risk of recurrent coronary events in the cholesterol and recurrent events (CARE) trial. Circulation 2000;102:1886–92. [66] Lee SJ, Campos H, Moye LA, Sacks FM. LDL containing apolipoprotein CIII is an independent risk factor for coronary events in diabetic patients. Arterioscler Thromb Vasc Biol 2003;23:853–8. [67] Pollin TI, Damcott CM, Shen H, et al. A null mutation in human APOC3 confers a favorable plasma lipid profile and apparent cardioprotection. Science 2008; 322:1702–5. [68] Chen M, Breslow JL, Li W, Leff T. Transcriptional regulation of the apoC-III gene by insulin in diabetic mice: correlation with changes in plasma triglyceride levels. J Lipid Res 1994;35:1918–24. [69] Altomonte J, Cong L, Harbaran S, et al. Foxo1 mediates insulin action on apoC-III and triglyceride metabolism. J Clin Invest 2004;114:1493–503. [70] Hertz R, Bishara-Shieban J, Bar-Tana J. Mode of action of peroxisome proliferators as hypolipidemic drugs. Suppression of apolipoprotein C-III. J Biol Chem 1995;270:13470–5. [71] Claudel T, Inoue Y, Barbier O, et al. Farnesoid X receptor agonists suppress hepatic apolipoprotein CIII expression. Gastroenterology 2003;125:544–55. [72] Caron S, Verrijken A, Mertens I, et al. Transcriptional activation of apolipoprotein CIII expression by glucose may contribute to diabetic dyslipidemia. Arterioscler Thromb Vasc Biol 2011;31(3):513–9. [73] Ginsberg HN, Brown WV. Apolipoprotein CIII: 42 years old and even more interesting. Arterioscler Thromb Vasc Biol 2011;31:471–3. [74] Hiukka A, Stahlman M, Pettersson C, et al. ApoCIII-enriched LDL in type 2 diabetes displays altered lipid composition, increased susceptibility for sphingomyelinase, and increased binding to biglycan. Diabetes 2009;58:2018–26. [75] Lee SJ, Moye LA, Campos H, Williams GH, Sacks FM. Hypertriglyceridemia but not diabetes status is associated with VLDL containing apolipoprotein CIII in patients with coronary heart disease. Atherosclerosis 2003;167:293–302. [76] Hiukka A, Fruchart-Najib J, Leinonen E, Hilden H, Fruchart JC, Taskinen MR. Alterations of lipids and apolipoprotein CIII in very low density lipoprotein subspecies in type 2 diabetes. Diabetologia 2005;48:1207–15. [77] Williams KJ, Chen K. Recent insights into factors affecting remnant lipoprotein uptake. Curr Opin Lipidol 2010;21:218–28. [78] Williams KJ. Molecular processes that handle — and mishandle — dietary lipids. J Clin Invest 2008;118:3247–59. [79] Mahley RW, Ji ZS. Remnant lipoprotein metabolism: key pathways involving cellsurface heparan sulfate proteoglycans and apolipoprotein E. J Lipid Res 1999;40:1–16. [80] MacArthur JM, Bishop JR, Stanford KI, et al. Liver heparan sulfate proteoglycans mediate clearance of triglyceride-rich lipoproteins independently of LDL receptor family members. J Clin Invest 2007;117:153–64.

[81] Williams KJ, Fless GM, Petrie KA, Snyder ML, Brocia RW, Swenson TL. Mechanisms by which lipoprotein lipase alters cellular metabolism of lipoprotein(a), low density lipoprotein, and nascent lipoproteins. Roles for low density lipoprotein receptors and heparan sulfate proteoglycans. J Biol Chem 1992;267:13284–92. [82] Fuki IV, Kuhn KM, Lomazov IR, et al. The syndecan family of proteoglycans. Novel receptors mediating internalization of atherogenic lipoproteins in vitro. J Clin Invest 1997;100:1611–22. [83] Chen K, Liu ML, Schaffer L, et al. Type 2 diabetes in mice induces hepatic overexpression of sulfatase 2, a novel factor that suppresses uptake of remnant lipoproteins. Hepatology 2010;52:1957–67. [84] Hassing HC, Mooij H, Guo S, et al. Inhibition of hepatic sulfatase-2 in vivo: a novel strategy to correct diabetic dyslipidemia. Hepatology 2012;55:1746–53. [85] Hassing C, Mooij H, Dallinga-Thie G, et al. Genetic variation in SULF2 is associated with plasma triglyceride levels in subjects with type 2 diabetes. XVI International Symposium on Atherosclerosis — ISA2012. Sydney; 2012. [86] Matikainen N, Burza MA, Romeo S, et al. Genetic variation in SULF2 is associated with postprandial clearance of triglyceride-rich remnant particles and triglyceride levels in healthy subjects. PLoS One 2013;8:e79473. [87] Riches FM, Watts GF, Naoumova RP, Kelly JM, Croft KD, Thompson GR. Hepatic secretion of very-low-density lipoprotein apolipoprotein B-100 studied with a stable isotope technique in men with visceral obesity. Int J Obes Relat Metab Disord J Int Assoc Study Obes 1998;22:414–23. [88] Chan DC, Watts GF, Redgrave TG, Mori TA, Barrett PH. Apolipoprotein B-100 kinetics in visceral obesity: associations with plasma apolipoprotein C-III concentration. Metabolism 2002;51:1041–6. [89] Watts GF, Barrett PH, Ji J, et al. Differential regulation of lipoprotein kinetics by atorvastatin and fenofibrate in subjects with the metabolic syndrome. Diabetes 2003;52:803–11. [90] Bjorkegren J, Packard CJ, Hamsten A, et al. Accumulation of large very low density lipoprotein in plasma during intravenous infusion of a chylomicron-like triglyceride emulsion reflects competition for a common lipolytic pathway. J Lipid Res 1996;37:76–86. [91] van Beek AP, van Barlingen HH, de Ruijter-Heijstek FC, et al. Preferential clearance of apoB-48-containing lipoproteins after heparin-induced lipolysis is modulated by lipoprotein lipase activity. J Lipid Res 1998;39:322–32. [92] Miller M, Stone NJ, Ballantyne C, et al. Triglycerides and cardiovascular disease: a scientific statement from the American Heart Association. Circulation 2011;123: 2292–333. [93] Brahm A, Hegele RA. Hypertriglyceridemia. Nutrient 2013;5:981–1001. [94] Lopez-Miranda J, Williams C, Lairon D. Dietary, physiological, genetic and pathological influences on postprandial lipid metabolism. Br J Nutr 2007;98: 458–73. [95] Jackson KG, Poppitt SD, Minihane AM. Postprandial lipemia and cardiovascular disease risk: interrelationships between dietary, physiological and genetic determinants. Atherosclerosis 2012;220:22–33. [96] Perez-Martinez P, Delgado-Lista J, Perez-Jimenez F, Lopez-Miranda J. Update on genetics of postprandial lipemia. Atheroscler Suppl 2010;11:39–43. [97] Varbo A, Benn M, Tybjaerg-Hansen A, Jorgensen AB, Frikke-Schmidt R, Nordestgaard BG. Remnant cholesterol as a causal risk factor for ischemic heart disease. J Am Coll Cardiol 2013;61:427–36. [98] Zilversmit DB. Atherogenesis: a postprandial phenomenon. Circulation 1979;60: 473–85. [99] Proctor SD, Vine DF, Mamo JC. Arterial retention of apolipoprotein B(48)- and B(100)containing lipoproteins in atherogenesis. Curr Opin Lipidol 2002;13:461–70. [100] Brunzell JD. Familial lipoprotein lipase deficiency and other causes of the chylomicronemia syndrome. In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. The metabolic and molecular bases of inherited disease. New York: McGraw-Hill; 1995. p. 1913–32. [101] Benlian P, De Gennes JL, Foubert L, Zhang H, Gagne SE, Hayden M. Premature atherosclerosis in patients with familial chylomicronemia caused by mutations in the lipoprotein lipase gene. N Engl J Med 1996;335:848–54. [102] Neil HA, Cooper J, Betteridge DJ, et al. All-cause and cardiovascular mortality in treated patients with severe hypertriglyceridaemia: a long-term prospective registry study. Atherosclerosis 2010;211:618–23. [103] Miller M, Cannon CP, Murphy SA, Qin J, Ray KK, Braunwald E. Impact of triglyceride levels beyond low-density lipoprotein cholesterol after acute coronary syndrome in the PROVE IT-TIMI 22 trial. J Am Coll Cardiol 2008;51:724–30. [104] Kotseva K, Wood D, De Backer G, De Bacquer D, Pyorala K, Keil U. EUROASPIRE III: a survey on the lifestyle, risk factors and use of cardioprotective drug therapies in coronary patients from 22 European countries. Eur J Cardiovasc Prev Rehabil 2009;16:121–37. [105] Eriksson M, Zethelius B, Eeg-Olofsson K, et al. Blood lipids in 75,048 type 2 diabetic patients: a population-based survey from the Swedish National diabetes register. Eur J Cardiovasc Prev Rehabil 2011;18:97–105. [106] Assmann G, Cullen P, Schulte H. Non-LDL-related dyslipidaemia and coronary risk: a case–control study. Diab Vasc Dis Res 2010;7:204–12. [107] Hokanson JE, Austin MA. Plasma triglyceride level is a risk factor for cardiovascular disease independent of high-density lipoprotein cholesterol level: a meta-analysis of population-based prospective studies. J Cardiovasc Risk 1996;3:213–9. [108] Sarwar N, Danesh J, Eiriksdottir G, et al. Triglycerides and the risk of coronary heart disease: 10,158 incident cases among 262,525 participants in 29 Western prospective studies. Circulation 2007;115:450–8. [109] Di Angelantonio E, Sarwar N, Perry P, et al. Major lipids, apolipoproteins, and risk of vascular disease. JAMA 2009;302:1993–2000. [110] Patel A, Barzi F, Jamrozik K, et al. Serum triglycerides as a risk factor for cardiovascular diseases in the Asia-Pacific region. Circulation 2004;110:2678–86.

J. Borén et al. / Clinica Chimica Acta 431 (2014) 131–142 [111] Lindman AS, Veierod MB, Tverdal A, Pedersen JI, Selmer R. Nonfasting triglycerides and risk of cardiovascular death in men and women from the Norwegian counties study. Eur J Epidemiol 2010;25:789–98. [112] Langsted A, Freiberg JJ, Tybjaerg-Hansen A, Schnohr P, Jensen GB, Nordestgaard BG. Nonfasting cholesterol and triglycerides and association with risk of myocardial infarction and total mortality: the Copenhagen City Heart study with 31 years of follow-up. J Intern Med 2011;270:65–75. [113] Varbo A, Nordestgaard BG, Tybjaerg-Hansen A, Schnohr P, Jensen GB, Benn M. Nonfasting triglycerides, cholesterol, and ischemic stroke in the general population. Ann Neurol 2011;69:628–34. [114] Langsted A, Freiberg JJ, Nordestgaard BG. Fasting and nonfasting lipid levels: influence of normal food intake on lipids, lipoproteins, apolipoproteins, and cardiovascular risk prediction. Circulation 2008;118:2047–56. [115] Nakajima K, Saito T, Tamura A, et al. Cholesterol in remnant-like lipoproteins in human serum using monoclonal anti apo B-100 and anti apo A-I immunoaffinity mixed gels. Clin Chim Acta Int J Clin Chem 1993;223:53–71. [116] Fujioka Y, Ishikawa Y. Remnant lipoproteins as strong key particles to atherogenesis. J Atheroscler Thromb 2009;16:145–54. [117] McNamara JR, Shah PK, Nakajima K, et al. Remnant-like particle (RLP) cholesterol is an independent cardiovascular disease risk factor in women: results from the Framingham Heart study. Atherosclerosis 2001;154:229–36. [118] Kugiyama K, Doi H, Takazoe K, et al. Remnant lipoprotein levels in fasting serum predict coronary events in patients with coronary artery disease. Circulation 1999;99:2858–60. [119] Imke C, Rodriguez BL, Grove JS, et al. Are remnant-like particles independent predictors of coronary heart disease incidence? The Honolulu Heart study. Arterioscler Thromb Vasc Biol 2005;25:1718–22. [120] Karpe F, Boquist S, Tang R, Bond GM, de Faire U, Hamsten A. Remnant lipoproteins are related to intima-media thickness of the carotid artery independently of LDL cholesterol and plasma triglycerides. J Lipid Res 2001;42:17–21. [121] Patsch JR, Miesenbock G, Hopferwieser T, et al. Relation of triglyceride metabolism and coronary artery disease. Studies in the postprandial state. Arterioscler Thromb 1992;12:1336–45. [122] Enkhmaa B, Ozturk Z, Anuurad E, Berglund L. Postprandial lipoproteins and cardiovascular disease risk in diabetes mellitus. Curr Diabetes Rep 2010;10:61–9. [123] Masuda D, Sugimoto T, Tsujii K, et al. Correlation of fasting serum apolipoprotein B48 with coronary artery disease prevalence. Eur J Clin Invest 2012;42:992–9. [124] Mori K, Ishida T, Yasuda T, et al. Fasting serum concentration of apolipoprotein B48 represents residual risks in patients with new-onset and chronic coronary artery disease. Clin Chim Acta Int J Clin Chem 2013;421:51–6. [125] Langsted A, Nordestgaard BG. Nonfasting lipids, lipoproteins, and apolipoproteins in individuals with and without diabetes: 58 434 individuals from the Copenhagen General Population study. Clin Chem 2011;57:482–9. [126] Smith SJ, Cooper GR, Myers GL, Sampson EJ. Biological variability in concentrations of serum lipids: sources of variation among results from published studies and composite predicted values. Clin Chem 1993;39:1012–22. [127] Klop B, Cohn JS, van Oostrom AJ, van Wijk JP, Birnie E, Castro Cabezas M. Daytime triglyceride variability in men and women with different levels of triglyceridemia. Clin Chim Acta Int J Clin Chem 2011;412:2183–9. [128] Cohn JS, McNamara JR, Cohn SD, Ordovas JM, Schaefer EJ. Plasma apolipoprotein changes in the triglyceride-rich lipoprotein fraction of human subjects fed a fatrich meal. J Lipid Res 1988;29:925–36. [129] Kolovou GD, Mikhailidis DP, Kovar J, et al. Assessment and clinical relevance of non-fasting and postprandial triglycerides: an expert panel statement. Curr Vasc Pharmacol 2011;9:258–70. [130] Ooi TC, Nordestgaard BG. Methods to study postprandial lipemia. Curr Vasc Pharmacol 2011;9:302–8. [131] Mihas C, Kolovou GD, Mikhailidis DP, et al. Diagnostic value of postprandial triglyceride testing in healthy subjects: a meta-analysis. Curr Vasc Pharmacol 2011;9:271–80. [132] Dominiczak MH, Caslake MJ. Apolipoproteins: metabolic role and clinical biochemistry applications. Ann Clin Biochem 2011;48:498–515. [133] Nakano T, Tokita Y, Nagamine T, et al. Measurement of serum remnant-like lipoprotein particle-triglyceride (RLP-TG) and RLP-TG/total TG ratio using highly sensitive triglyceride assay reagent. Clin Chim Acta Int J Clin Chem 2011;412:71–8. [134] Nakajima K, Nakano T, Moon HD, et al. The correlation between TG vs remnant lipoproteins in the fasting and postprandial plasma of 23 volunteers. Clin Chim Acta Int J Clin Chem 2009;404:124–7. [135] Nakajima K, Nakano T, Tokita Y, et al. The characteristics of remnant lipoproteins in the fasting and postprandial plasma. Clin Chim Acta Int J Clin Chem 2012;413:1077–86. [136] Nakamura T, Obata JE, Hirano M, et al. Predictive value of remnant lipoprotein for cardiovascular events in patients with coronary artery disease after achievement of LDL-cholesterol goals. Atherosclerosis 2011;218:163–7. [137] Miyauchi K, Kayahara N, Ishigami M, et al. Development of a homogeneous assay to measure remnant lipoprotein cholesterol. Clin Chem 2007;53:2128–35. [138] Yoshida H, Kurosawa H, Hirowatari Y, et al. Characteristic comparison of triglyceride-rich remnant lipoprotein measurement between a new homogenous assay (RemL-C) and a conventional immunoseparation method (RLP-C). Lipids Health Dis 2008;7:18. [139] Schaefer EJ. Limitations of automated remnant lipoprotein cholesterol assay for diagnostic use. Clin Chem 2009;55:2061–2 author reply 2062–2063. [140] Phillips ML, Pullinger C, Kroes I, et al. A single copy of apolipoprotein B-48 is present on the human chylomicron remnant. J Lipid Res 1997;38:1170–7. [141] Smith D, Watts GF, Dane-Stewart C, Mamo JC. Post-prandial chylomicron response may be predicted by a single measurement of plasma apolipoprotein B48 in the fasting state. Eur J Clin Invest 1999;29:204–9.

141

[142] Kotite L, Bergeron N, Havel RJ. Quantification of apolipoproteins B-100, B-48, and E in human triglyceride-rich lipoproteins. J Lipid Res 1995;36:890–900. [143] Karpe F, Hamsten A. Determination of apolipoproteins B-48 and B-100 in triglyceride-rich lipoproteins by analytical SDS-PAGE. J Lipid Res 1994;35:1311–7. [144] Smith D, Proctor SD, Mamo JC. A highly sensitive assay for quantitation of apolipoprotein B48 using an antibody to human apolipoprotein B and enhanced chemiluminescence. Ann Clin Biochem 1997;34(Pt 2):185–9. [145] Sakai N, Uchida Y, Ohashi K, et al. Measurement of fasting serum apoB-48 levels in normolipidemic and hyperlipidemic subjects by ELISA. J Lipid Res 2003;44:1256–62. [146] Kinoshita M, Kojima M, Matsushima T, Teramoto T. Determination of apolipoprotein B-48 in serum by a sandwich ELISA. Clin Chim Acta Int J Clin Chem 2005;351:115–20. [147] Jackson KG, Williams CM. Apolipoprotein B-48: comparison of fasting concentrations measured in normolipidaemic individuals using SDS-PAGE, immunoblotting and ELISA. Atherosclerosis 2004;176:207–17. [148] Catapano AL, Reiner Z, De Backer G, et al. ESC/EAS guidelines for the management of dyslipidaemias: the Task Force for the management of dyslipidaemias of the European Society of Cardiology (ESC) and the European Atherosclerosis Society (EAS). Atherosclerosis 2011;217:3–46. [149] Berglund L, Brunzell JD, Goldberg AC, et al. Evaluation and treatment of hypertriglyceridemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2012;97:2969–89. [150] Watts GF, Ooi EM, Chan DC. Demystifying the management of hypertriglyceridaemia. Nat Rev Cardiol 2013;10:648–61. [151] Chan DC, Pang J, Romic G, Watts GF. Postprandial hypertriglyceridemia and cardiovascular disease: current and future therapies. Curr Atheroscler Rep 2013;15:309. [152] Ryden L, Grant PJ, Anker SD, et al. ESC Guidelines on diabetes, pre-diabetes, and cardiovascular diseases developed in collaboration with the EASD: the Task Force on diabetes, pre-diabetes, and cardiovascular diseases of the European Society of Cardiology (ESC) and developed in collaboration with the European Association for the Study of Diabetes (EASD). Eur Heart J 2013;34:3035–87. [153] Schaefer EJ, McNamara JR, Tayler T, et al. Comparisons of effects of statins (atorvastatin, fluvastatin, lovastatin, pravastatin, and simvastatin) on fasting and postprandial lipoproteins in patients with coronary heart disease versus control subjects. Am J Cardiol 2004;93:31–9. [154] Boden WE, Probstfield JL, Anderson T, et al. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med 2011;365:2255–67. [155] HPS2-THRIVE randomized placebo-controlled trial in 25673 high-risk patients of ER niacin/laropiprant: trial design, pre-specified muscle and liver outcomes, and reasons for stopping study treatment. Eur Heart J 2013;34:1279–91. [156] Song WL, FitzGerald GA. Niacin, an old drug with a new twist. J Lipid Res 2013;54:2586–94. [157] Rizos EC, Ntzani EE, Bika E, Kostapanos MS, Elisaf MS. Association between omega3 fatty acid supplementation and risk of major cardiovascular disease events: a systematic review and meta-analysis. JAMA 2012;308:1024–33. [158] Jump DB, Depner CM, Tripathy S. Omega-3 fatty acid supplementation and cardiovascular disease. J Lipid Res 2012;53:2525–45. [159] Ginsberg HN, Elam MB, Lovato LC, et al. Effects of combination lipid therapy in type 2 diabetes mellitus. N Engl J Med 2010;362:1563–74. [160] Scott R, O'Brien R, Fulcher G, et al. Effects of fenofibrate treatment on cardiovascular disease risk in 9,795 individuals with type 2 diabetes and various components of the metabolic syndrome: the Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) study. Diabetes Care 2009;32:493–8. [161] Jun M, Foote C, Lv J, et al. Effects of fibrates on cardiovascular outcomes: a systematic review and meta-analysis. Lancet 2010;375:1875–84. [162] Chan DC, Wong AT, Yamashita S, Watts GF. Apolipoprotein B-48 as a determinant of endothelial function in obese subjects with type 2 diabetes mellitus: effect of fenofibrate treatment. Atherosclerosis 2012;221:484–9. [163] Reyes-Soffer G, Ngai CI, Lovato L, et al. Effect of combination therapy with fenofibrate and simvastatin on postprandial lipemia in the ACCORD lipid trial. Diabetes Care 2013;36:422–8. [164] Bozzetto L, Annuzzi G, Corte GD, et al. Ezetimibe beneficially influences fasting and postprandial triglyceride-rich lipoproteins in type 2 diabetes. Atherosclerosis 2011;217:142–8. [165] Hajer GR, Dallinga-Thie GM, van Vark-van der Zee LC, Visseren FL. The effect of statin alone or in combination with ezetimibe on postprandial lipoprotein composition in obese metabolic syndrome patients. Atherosclerosis 2009;202:216–24. [166] Kikuchi K, Nezu U, Inazumi K, et al. Double-blind randomized clinical trial of the effects of ezetimibe on postprandial hyperlipidaemia and hyperglycaemia. J Atheroscler Thromb 2012;19:1093–101. [167] Tremblay AJ, Lamarche B, Hogue JC, Couture P. Effects of ezetimibe and simvastatin on apolipoprotein B metabolism in males with mixed hyperlipidemia. J Lipid Res 2009;50:1463–71. [168] Phillips LK, Prins JB. Update on incretin hormones. Ann N Y Acad Sci 2011;1243: E55–74. [169] Holst JJ, Orskov C. Incretin hormones — an update. Scand J Clin Lab Invest Suppl 2001;234:75–85. [170] Hein GJ, Baker C, Hsieh J, Farr S, Adeli K. GLP-1 and GLP-2 as yin and yang of intestinal lipoprotein production: evidence for predominance of GLP-2-stimulated postprandial lipemia in normal and insulin-resistant states. Diabetes 2013;62:373–81. [171] Matikainen N, Manttari S, Schweizer A, et al. Vildagliptin therapy reduces postprandial intestinal triglyceride-rich lipoprotein particles in patients with type 2 diabetes. Diabetologia 2006;49:2049–57. [172] Bunck MC, Corner A, Eliasson B, et al. One-year treatment with exenatide vs. insulin glargine: effects on postprandial glycemia, lipid profiles, and oxidative stress. Atherosclerosis 2010;212:223–9.

142

J. Borén et al. / Clinica Chimica Acta 431 (2014) 131–142

[173] Tremblay AJ, Lamarche B, Deacon CF, Weisnagel SJ, Couture P. Effect of sitagliptin therapy on postprandial lipoprotein levels in patients with type 2 diabetes. Diabetes Obes Metab 2011;13:366–73. [174] Eliasson B, Moller-Goede D, Eeg-Olofsson K, et al. Lowering of postprandial lipids in individuals with type 2 diabetes treated with alogliptin and/or pioglitazone: a randomised double-blind placebo-controlled study. Diabetologia 2012;55:915–25. [175] Farr S, Adeli K. Incretin-based therapies for treatment of postprandial dyslipidemia in insulin-resistant states. Curr Opin Lipidol 2012;23:56–61. [176] Huff MW, Hegele RA. Apolipoprotein C-III: going back to the future for a lipid drug target. Circ Res 2013;112:1405–8. [177] Kaeppel C, Beattie SG, Fronza R, et al. A largely random AAV integration profile after LPLD gene therapy. Nat Med 2013;19:889–91. [178] Haddley K. Alipogene tiparvovec for the treatment of lipoprotein lipase deficiency. Drugs Today (Barc) 2013;49:161–70. [179] Burnett JR, Hooper AJ. Alipogene tiparvovec, an adeno-associated virus encoding the Ser(447)X variant of the human lipoprotein lipase gene for the treatment of patients with lipoprotein lipase deficiency. Curr Opin Mol Ther 2009;11:681–91. [180] Carpentier AC, Frisch F, Labbe SM, et al. Effect of alipogene tiparvovec (AAV1LPL(S447X)) on postprandial chylomicron metabolism in lipoprotein lipasedeficient patients. J Clin Endocrinol Metab 2012;97:1635–44. [181] Gaudet D, Methot J, Dery S, et al. Efficacy and long-term safety of alipogene tiparvovec (AAV1-LPLS447X) gene therapy for lipoprotein lipase deficiency: an open-label trial. Gene Ther 2013;20:361–9.

[182] Graham MJ, Lee RG, Bell 3rd TA, et al. Antisense oligonucleotide inhibition of apolipoprotein C-III reduces plasma triglycerides in rodents, nonhuman primates, and humans. Circ Res 2013;112:1479–90. [183] Panel EASC. The polygenic nature of hypertriglyceridaemia: implications for definition, diagnosis and management. Lancet Diabetes Endocrinol 2013. http:// dx.doi.org/10.1016/S2213-8587(13)70191-8 [in press]. [184] Skalen K, Gustafsson M, Rydberg EK, et al. Subendothelial retention of atherogenic lipoproteins in early atherosclerosis. Nature 2002;417:750–4. [185] Tabas I, Williams KJ, Boren J. Subendothelial lipoprotein retention as the initiating process in atherosclerosis: update and therapeutic implications. Circulation 2007;116:1832–44. [186] Varbo A, Benn M, Nordestgaard BG. Remnant cholesterol as a cause of ischemic heart disease: evidence, definition, measurement, atherogenicity, high risk patients, and present and future treatment. Pharmacol Ther Mar. 2014;141(3): 358–67. [187] Flood C, Gustafsson M, Pitas RE, Arnaboldi L, Walzem RL, Boren J. Molecular mechanism for changes in proteoglycan binding on compositional changes of the core and the surface of low-density lipoprotein-containing human apolipoprotein B100. Arterioscler Thromb Vasc Biol 2004;24:564–70.

Postprandial hypertriglyceridemia as a coronary risk factor.

Postprandial hypertriglyceridemia is now established as an important risk factor for cardiovascular disease (CVD). This metabolic abnormality is princ...
838KB Sizes 2 Downloads 0 Views