You are on page 1of 17

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/293638020

Chylomicrons: Advances in biology, pathology, laboratory testing, and


therapeutics

Article  in  Clinica Chimica Acta; International Journal of Clinical Chemistry · February 2016


DOI: 10.1016/j.cca.2016.02.004

CITATIONS READS

37 2,420

4 authors:

Josep Julve Jesús Maria Martin-Campos


IIB Sant Pau IR-Sant Pau - Sant Pau Institute of Biomedical Research
131 PUBLICATIONS   2,044 CITATIONS    54 PUBLICATIONS   1,089 CITATIONS   

SEE PROFILE SEE PROFILE

Joan Carles Escolà-Gil Francisco Blanco-Vaca


IIB Sant Pau Hospital de la Santa Creu i Sant Pau; Universitat Autònoma de Barcelona
183 PUBLICATIONS   3,025 CITATIONS    256 PUBLICATIONS   5,676 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

LDL and HDL as inductors of m-RCT View project

Lipotoxicity and microvascular disease: contribution to myocardial damage in clinical and experimental models of diabetes View project

All content following this page was uploaded by Joan Carles Escolà-Gil on 18 March 2016.

The user has requested enhancement of the downloaded file.


This article appeared in a journal published by Elsevier. The attached
copy is furnished to the author for internal non-commercial research
and education use, including for instruction at the authors institution
and sharing with colleagues.
Other uses, including reproduction and distribution, or selling or
licensing copies, or posting to personal, institutional or third party
websites are prohibited.
In most cases authors are permitted to post their version of the
article (e.g. in Word or Tex form) to their personal website or
institutional repository. Authors requiring further information
regarding Elsevier’s archiving and manuscript policies are
encouraged to visit:
http://www.elsevier.com/authorsrights
Author's personal copy

Clinica Chimica Acta 455 (2016) 134–148

Contents lists available at ScienceDirect

Clinica Chimica Acta

journal homepage: www.elsevier.com/locate/clinchim

Invited critical review

Chylomicrons: Advances in biology, pathology, laboratory testing,


and therapeutics
Josep Julve a,b,c,⁎,1, Jesús M. Martín-Campos a,b,c,⁎,1, Joan Carles Escolà-Gil a,b,c, Francisco Blanco-Vaca a,b,c,d
a
Institut de Recerca de l'HSCSP – Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain
b
Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
c
CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain
d
Hospital de la Santa Creu i Sant Pau, Servei de Bioquímica, Barcelona, Spain

a r t i c l e i n f o a b s t r a c t

Article history: The adequate absorption of lipids is essential for all mammalian species due to their inability to synthesize some
Received 29 December 2015 essential fatty acids and fat-soluble vitamins. Chylomicrons (CMs) are large, triglyceride-rich lipoproteins that
Received in revised form 1 February 2016 are produced in intestinal enterocytes in response to fat ingestion, which function to transport the ingested lipids
Accepted 6 February 2016
to different tissues. In addition to the contribution of CMs to postprandial lipemia, their remnants, the degrada-
Available online 8 February 2016
tion products following lipolysis by lipoprotein lipase, are linked to cardiovascular disease. In this review, we will
Keywords:
focus on the structure–function and metabolism of CMs. Second, we will analyze the impact of gene defects re-
Chylomicron ported to affect CM metabolism and, also, the role of CMs in other pathologies, such as atherothrombotic cardio-
Triglyceride vascular disease and diabetes mellitus. Third, we will provide an overview of the laboratory tests currently used
Enterocyte to study CM disorders, and, finally, we will highlight current treatments in diseases affecting CMs.
Type I hyperlipidemia © 2016 Elsevier B.V. All rights reserved.
Type V hyperlipidemia
Abetalipoproteinemia
Hypobetalipoproteinemia
Chylomicron retention disease
Atherosclerosis
Laboratory testing
Therapy

Contents

1. Structure and function of chylomicrons . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135


1.1. Physico-chemical and functional characteristics of CMs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135
1.2. Lipid composition of CMs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
1.3. CM apolipoproteins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
1.3.1. Apolipoprotein B-48 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
1.3.2. Apolipoprotein A-I . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
1.3.3. Apolipoprotein A-II . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
1.3.4. Apolipoprotein A-IV . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
1.3.5. Apolipoprotein A-V . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 137
1.3.6. Apolipoproteins C . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138
1.3.7. Apolipoprotein E . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138

Abbreviations: ACAT, acyl CoA acyl transferase; Angptl3, angiopoietin-like protein 3; Angptl4, angiopoietin-like protein 4; Angptl8, angiopoietin-like protein 8; apo, apolipoprotein;
apobec 1, apoB mRNA editing enzyme 1; CM, chylomicron; CMs, chylomicrons; COPII, coat protein complex II; DGAT, diglyceride acyltransferase; FABP, fatty acid binding protein; GLP-
1, glucagon-like peptide-1; GPIHBP1, glycosylphosphatidylinositol-anchored high-density lipoprotein binding protein 1; HDL, high-density lipoproteins; HSPG, heparan sulfate proteogly-
can; LDL, low-density lipoproteins; LDLR, LDL receptor; LR11/SorLA1, LDL receptor relative with 11 ligand binding repeats/sorting protein-related receptors containing LDLR class A
repeats; LRP, LDL receptor-related protein; LMF1, lipase maturation factor 1; MGAT, monoglyceride acyltransferase; MTTP, microsomal triglyceride transfer protein; NPC1L1,
Niemann–Pick C1-like 1; PCV, preCM transport vesicles; PPAR-alpha, peroxisome proliferator-activated receptor alpha; PUFAs, polyunsaturated fatty acids; Sar1b, secretion-associated,
Ras-related GTPase 1B; SORT1, sortilin-1; SR-BI, scavenger receptor class B type 1; TRL, triglyceride-rich lipoprotein; TRLs, triglyceride-rich lipoproteins; VLDL, very low-density lipopro-
teins; VLDLR, VLDL receptor.
⁎ Corresponding authors at: Grup bases metabòliques del risc cardiovascular, pavelló 17. Institut de Recerca de l'HSCSP-IIB Sant Pau, C/ Antoni M. Claret 167, 08025 Barcelona, Spain.
E-mail addresses: jjulve@santpau.cat (J. Julve), jmartinca@santpau.cat (J.M. Martín-Campos).
1
Both authors contributed equally.

http://dx.doi.org/10.1016/j.cca.2016.02.004
0009-8981/© 2016 Elsevier B.V. All rights reserved.
Author's personal copy

J. Julve et al. / Clinica Chimica Acta 455 (2016) 134–148 135

2. Metabolism of CM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139
2.1. Intracellular metabolic determinants of synthesis and secretion of CM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139
2.1.1. Fatty acid transporters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139
2.1.2. Fatty acid binding proteins (FABPs) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139
2.1.3. Enzymes involved in triglyceride synthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139
2.1.4. MTTP . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139
2.1.5. Coat protein complex II (COPII) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139
2.1.6. Sortilin-1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139
2.2. Extracellular metabolic determinants of catabolism and clearance of CMs and their remnants . . . . . . . . . . . . . . . . . . . . . . 140
2.2.1. Lipoprotein lipase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140
2.2.2. Hepatic lipase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140
2.2.3. Glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1 (GPIHBP1) . . . . . . . . . . . . . . . . . 140
2.2.4. Lipase maturation factor (LMF) 1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140
2.3. Receptors for CM remnant clearance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140
2.3.1. LDLR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140
2.3.2. LDL receptor-related proteins (LRPs) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140
3. Pathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 141
3.1. Hyperchylomicronemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 141
3.1.1. Monogenic hyperchylomicronemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 141
3.1.2. Polygenic hyperchylomicronemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 141
3.1.3. Autoimmune hyperchylomicronemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142
3.2. Hypochylomicronemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142
3.2.1. Chylomicron retention disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142
3.2.2. Abetalipoproteinemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142
3.2.3. Familial hypobetalipoproteinemia (FHBL) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142
3.2.4. Familial combined hypolipidemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142
3.3. Chylomicron remnants and cardiovascular disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 143
3.4. Regulation of intestinal chylomicron production in type 2 diabetes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 143
4. Laboratory tests for CM analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 143
4.1. Biochemical testing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 144
4.2. Genetic testing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 144
5. Treatment of chylomicron disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 144
5.1. Hyperchylomicronemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 144
5.1.1. Dietary treatment of hyperchylomicronemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 144
5.1.2. Drug treatment of hyperchylomicronemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 145
5.1.3. Gene therapy of hyperchylomicronemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 145
5.2. Hypochylomicronemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 145
5.2.1. Dietary treatment of hypochilomicronemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 145
Disclosures . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 145
Conflicts of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 145
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 145
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 145

1. Structure and function of chylomicrons Although postprandial concentrations of triglycerides may not be
found to be significantly influenced by the type of meal, the extent of
In healthy humans, dietary fat, including lipids and fat-soluble vita- postprandial CM response is highly dependent on the meal-fat mixtures
mins, is efficiently absorbed by the small intestine. Thus, postprandial and, therefore, the nature and amount of different dietary components,
lipemia refers to the systemic elevation of triglyceride-rich lipoprotein especially fatty acids [7]. For instance, meal mixtures enriched in n-3
(TRL) levels after a meal. The induction of postprandial lipemia is due polyunsaturated fatty acids (PUFAs) generally decrease the postprandi-
in part to an elevation in the serum concentration of chylomicrons al triglyceride response compared with those rich in saturated and
(CMs) [1]. monounsaturated fatty acids [8–10]. Independent studies assessing
CMs transport most of the dietary fatty acids (mainly long-chain the impact on the serum levels of CMs after chronic consumption of iso-
fatty acids) and their remnants, resulting from the triglyceride hydroly- caloric diets rich in saturated fatty acids, n-3 PUFAs, and n-6 PUFAs re-
sis of these TRLs by the lipolytic enzyme lipoprotein lipase, and are also vealed that the serum levels of CMs increased with saturated fatty
present in circulation during the postabsorptive state and cleared main- acids compared with n-6 PUFAs and n-3 PUFAs, respectively. Although
ly by liver receptors [2]. CM production rates were not assessed, subjects on the n-6 PUFA or
n-3 PUFA diets displayed enhanced lipolysis compared with those on
1.1. Physico-chemical and functional characteristics of CMs a saturated fatty acid diet, suggesting an increased in CM clearance.
Recent evidence suggests that enterocyte-triglyceride processing also
CMs are the largest lipoproteins found in circulation [3]. Their size influences postprandial lipemia [11]. Firstly, some evidence supports the
has been found to be significantly dependent on the fed/fasted state notion that the initial rise of lipids (within 10 to 30 min) secreted right
[4], the rate of fat absorption, and the type and amount of fat absorbed after a fatty meal—and preceding the primary postprandial peak of
[5,6]. Conversely, the number of TRL particles produced during the post- serum triglycerides that occurs 3 to 4 h after the start of a meal—actually
prandial state is only modestly increased compared with those released correspond to those consumed in the previous meal. Of note, this strong-
under fasting conditions [4]. ly suggests the existence of triglyceride storage within enterocytes. Sec-
The increase in postprandial lipids is influenced by gender, genetics, ondly, the release of CMs in response to the cephalic phase is induced by
age, body size, exercise, weight loss, and metabolic syndrome [7]. an oral stimulation, which occurs when fat is yet not consumed. This last
Author's personal copy

136 J. Julve et al. / Clinica Chimica Acta 455 (2016) 134–148

finding has led to the hypothesis of a taste-gut-brain axis, which may


also be involved in regulating the serum levels of triglycerides in re-
sponse to oral taste.

1.2. Lipid composition of CMs

Quantitative analysis shows that CMs have a central lipid core main-
ly composed of triglycerides and monoglycerides (85–92%), free and es-
terified cholesterol (1–3%), phospholipids (6–12%), and traces of fatty
acids [12]. In general, the fatty acid composition of postprandial CM tri-
glycerides fairly closely reflects that of ingested fat [5,13]. However, sev-
eral studies report that the fatty acid composition of a fatty meal is not
always reflected in the fatty acid composition of CM triglycerides. For
instance, low- and medium-chain fatty acid species (mainly C10:0,
C12:0, and C14:0) in the diet are frequently poorer substrates for re-
esterification into triglycerides within the enterocyte; therefore, they
are not transported to a significant extent into CMs but are rather
absorbed and rapidly delivered via the portal system to the liver, [5,14].

1.3. CM apolipoproteins

Apolipoproteins are the key protein constituents of CMs. Although


these proteins generally represent only less than 2% of the CMs' total
mass, they determine the intra- and extra-cellular metabolic fate of
these lipoproteins [15].

1.3.1. Apolipoprotein B-48


The apolipoprotein (apo) B-48 is the main, nonexchangeable CM pro-
tein and plays a critical role in CMs' synthesis, assembly, and secretion
[8]. Under fasting conditions, the serum levels of apoB-48 are very low
or undetectable in most individuals. In human beings, apoB-48 is exclu-
sively produced in the small intestine in response to dietary fat through a
unique mRNA-editing event that converts codon 2153 (CAA, encoding a
glutamine) into a premature stop codon (UAA). This stop codon induces
Fig. 1. Biology and pathology of the formation and packaging of CMs in the enterocyte.
the translation of a truncated apoB, apoB-48, which contains 48% of the
Graphic symbology: open, double-surrounded ellipsoids indicate gene targets. Red
mature apoB primary sequence. The C-to-U site-specific editing of apoB circles indicate gene targets with known causal loss-of-function mutations: MTTP in
mRNA is accomplished by a large multiprotein complex, which includes abetalipoproteinemia (1) and Sar1 GTPase in chylomicron retention disease (2). A curly
a deaminase, the apoB mRNA editing enzyme (apobec)-1, and the black line was used to indicate the apoB-48 protein in nascent CM. Abbreviations used
include ACAT-2, acyl CoA acyltransferase; apo, apolipoprotein; CE, cholesteryl ester; CM,
apobec-1 complementation factor, among other factors. Importantly,
chylomicron; COPII, coat protein complex II; DGAT, diglyceride acyltransferase; FA, fatty
APOBEC1 is highly expressed in the small intestine but is absent in the acids; FC, free cholesterol; FABP, fatty acid binding protein; MG, monoglyceride; MGAT,
liver in humans. Consequently, all the apoB produced in the small intes- monoglyceride acyltransferase; MTTP, microsomal triglyceride transfer protein; NPC1L1,
tine essentially corresponds to apoB-48. The absence of the C-terminal Niemann–Pick C1-like 1; PCV, pre-CM transport vesicle; PLIN, perilipin; TG, triglycerides.
portion of apoB in apoB-48 significantly determines its metabolic role. (For interpretation of the references to color in this figure legend, the reader is referred
to the web version of this article.)
For instance, the C-terminal domain of apoB-48 does not contain the
LDL receptor (LDLR) binding domain [16], and it is therefore unable to
mediate the clearance of this type of lipoproteins via LDLR. apoA-II have been found to be directly correlated with those of triglyc-
erides in healthy individuals. The deficiency of apoA-II in mice is associ-
1.3.2. Apolipoprotein A-I ated with the reduced serum levels of TRLs and the increased catabolism
ApoA-I is the main protein constituent of circulating HDL [17]. Intes- of TRL remnants [22]. The overexpression of human apoA-II in mice pro-
tinal CMs are also a source of significant amounts of circulating apoA-I. motes postprandial hypertriglyceridemia in independently transgenic
ApoA-I is generated in the endoplasmic reticulum of enterocytes but is mice [23–25], this being mainly attributed to a defective catabolism of
transported to the Golgi separately from CM vesicles and added to the circulating postprandial CMs. Because excess apoA-II associates with
CMs before the mature particle is secreted into the mesenteric lymph CMs in circulation [24], it was first suggested that its accumulation in
[18] (Fig. 1). Its suppression does not reduce triglyceride release from these particles might directly convert them in poorer lipolytic sub-
cells [19], thereby indicating that the assembly of apoA-I may not be re- strates [23,26]. More recently, it has been proposed that an altered pro-
quired for an appropriate lipidation of CMs. teome of the HDL from mice overexpressing apoA-II [23,25] might also
contribute to an impaired trafficking of apoC-II, C-III, and E from HDL to
1.3.3. Apolipoprotein A-II CMs.
ApoA-II is mainly synthesized in the liver [20,21] and, to a lesser ex-
tent, in the intestine. The role of apoA-II as a determinant of triglyceride 1.3.4. Apolipoprotein A-IV
metabolism is supported by several observations in studies on humans ApoA-IV is the largest member of the exchangeable apolipoprotein
and experimental models [20,21], even though whether this is a result family [27]. It is most highly produced by enterocytes in response to
of a function over CMs, VLDL, or both is unclear. For instance, there is lipid absorption [28], with minor amounts made in the liver [29]. It is se-
a significant association of the −265C allele in the APOA2 gene with de- creted in association with nascent CM particles into the mesenteric
creased serum apoA-II concentration and the enhanced postprandial lymph [30]. During the intestinal assembly of CMs, several lines of evi-
metabolism of large TRL (i.e., VLDL). The postprandial serum levels of dence support that apoA-IV is incorporated into CMs at an early stage,
Author's personal copy

J. Julve et al. / Clinica Chimica Acta 455 (2016) 134–148 137

and within the endoplasmic reticulum, and participate in the intestinal 1.3.5. Apolipoprotein A-V
assembly and secretion of these lipoproteins [31,32] (Fig. 1). First, apoA- ApoA-V is mainly expressed in the liver and is found mainly bound
IV intestinal synthesis is increased during the absorption of long-chain to TRLs and HDL at a very low serum concentration (20–500 ng/mL)
fatty acids, which require CM assembly, but not during the absorption [34]. This protein is recognized as a potent regulator of triglyceride me-
of short-chain fatty acids, which are directly transported into portal circu- tabolism and leads to the enhanced lipolysis of circulating TRL and the
lation rather than packaged into CMs. Second, apoA-IV secretion into clearance of their remnants [34]. This role has been, in part, attributed
lymph declines when intestinal CM formation is inhibited, and it increases to its ability to promote a greater interaction between both the TRL
together with CM secretion when the inhibitor is removed. Third, the in- and lipoprotein lipase, which might enhance the catabolism of TRLs by
hibition of CM formation has also been found to result in the accumula- stimulating the lipoprotein lipase-mediated lipolysis of these lipopro-
tion of intracellular lipid droplets containing both apoB-48 and apoA-IV, teins. Several factors might be involved in the apoA-V-mediated modu-
thus showing that these apoliproteins associate with nascent primordial lation of lipoprotein lipase action [34]. First, it has been suggested that
CMs. Fourth, apoA-IV overexpression induced triglyceride packaging in the interaction between apoA-V and glycosylphosphatidylinositol-
CMs in vitro. This indicates that apoA-IV has a role in the stabilization of anchored high-density lipoprotein binding protein 1 (GPIHBP1) might
the expanding lipid interfaces during the formation of primordial CMs in- favor the triglyceride hydrolysis of TRLs in mediating the interaction of
side the enterocyte, enabling the enlargement of these particles. Fifth and apoA-V with lipoprotein lipase (Fig. 2).
last, dietary fat absorption results in an increase in the serum concentra- ApoA-V has also been shown to facilitate the binding of TRL rem-
tion of apoA-IV, whereas it remains unchanged in patients with defective nants to LDLR family members and thereby the clearance of these parti-
intestinal lipid absorption in response to a fatty meal. cles from circulation (Fig. 3). In particular, sortilin-1 (SORT1), which is a
Despite this solid body of evidence, neither the overexpression nor multiligand receptor in this family, has been reported to bind lipopro-
deficiency of apoA-IV in experimental animals has been shown to influ- tein lipase and apoA-V. Lastly, apoA-V has also been described to medi-
ence fat absorption or production [31,32], thus suggesting that this pro- ate the hepatic heparan-sulfate-mediated clearance of TRL remnants.
tein exerts an auxiliary role in these processes or, at least, one function Few studies have analyzed the effect of apoA-V on the serum levels
that can be substituted by other proteins. On the other hand, it has also of triglycerides after a fatty meal. The serum levels of apoA-V appear
been proposed that this protein influences serum CM clearance. For in- to be closely related to the magnitude of postprandial lipemia and in-
stance, apoA-IV modulates the activation of lipoprotein lipase activity crease after a meal challenge, this finding reflecting an association
by apoC-II [33], whereas its absence causes a delay in the CM clearance with CMs [35], possibly as part of a regulatory mechanism to cover the
in mice [31]. During the lipolysis of the CM triglycerides, most apoA-IV increasing need of postprandial lipolysis [36]. The increase in the
detaches from the CMs. It has been reported that approximately 25% serum levels of apoA-V after a meal challenge might be, in part,
transfers to serum HDL and the remaining percentage is found in the accounted for by the action of PPAR-alpha. For instance, PPAR-alpha is
lipoprotein-depleted serum [31]. Recent reports show that apoA-IV activated after a fat load [37], and this transcription factor is involved
also exerts an effect on the regulation of satiety and appetite [32], there- in the upregulation of APOA5 [38,39].
by suggesting a unique role for this protein in integrating feeding be- The potential role of apoA-V on the control of the absorption of
havior and intestinal lipid absorption. dietary fats by the gut still remains enigmatic. Interestingly, apoA-V

Fig. 2. Biology and pathology of chylomicron triglyceride catabolism. Graphic symbology: open, double-surrounded ellipsoids indicate gene targets. Red circles indicate gene targets with
known causal loss-of-function mutations in familial hyperchylomicronemia: LPL (1), apoC-II (2), GPIHBP1 (3), LMF1 (4), apoA-V (5). FA, fatty acids; GPIHBP1, glycophosphatidylinositol
HDL-binding protein-1; HSPG, heparan sulfate proteoglycans; LMF-1, lipase maturation factor-1; LPL, lipoprotein lipase; TG, triglycerides. (For interpretation of the references to color in
this figure legend, the reader is referred to the web version of this article.)
Author's personal copy

138 J. Julve et al. / Clinica Chimica Acta 455 (2016) 134–148

TRLs are known to stabilize and protect lipoprotein lipase from


inactivating factors, including the angiopoietin-like protein 4 (Angptl4).
The addition of apoC-I to TRLs (either CM or synthetic lipid emulsion)
significantly interferes with their protective effect on lipoprotein lipase,
which becomes more prone to inactivation by Angptl4 in vitro [45].

1.3.6.2. Apolipoprotein C-II. This protein is mainly synthesized in the liver


and to a lesser extent in the small intestine [46]. Apart from HDL, apoC-II
is also bound to CMs and VLDL, becoming one of the key components in
the metabolism of TRLs [42]. The importance of apoC-II as an activator of
lipoprotein lipase has been unequivocally demonstrated in patients
with genetic defects in their structure or production and in transgenic
animals [42,46]. ApoC-II is therefore required for maximal rates of TRL
lipolysis [47] (Fig. 2). However, the mechanism whereby this protein ac-
tivates lipoprotein lipase still remains elusive. For instance, it is under
discussion whether apoC-II binds or not directly to lipoprotein lipase
[46].
The hepatic expression of APOC2 is upregulated by a high-fat diet
and by agonists for PPAR-alpha. Similar to the concept described for
apoA-V (see Section 3.2.4), oleoylethanolamide, a potent PPAR-alpha li-
gand, which is overproduced by enterocytes in response to food intake
[39], might activate PPAR-alpha and induce the transcription rate of
APOC2. Consistent with this, an increase in the serum levels of apoC-II
has been reported under postprandial conditions [33].

1.3.6.3. Apolipoprotein C-III. ApoC-III is a component and one of the most


important markers of TRL [48]. It is the most abundant apoC in human
serum, and its concentration is positively associated with that of triglyc-
erides. Apart from TRLs, it is also found to be associated with HDL and,
though to a lesser degree, with LDL. This protein is synthesized by the
liver and to a lesser extent by the intestine and, along with the APOA1,
APOA4, and APOA5 genes, belongs to a gene cluster that has been impli-
cated as a potential genetic determinant for variations in triglycerides.
Several lines of evidence involve apoC-III as a major player contributing
to the development of hypertriglyceridemia [42,45,48]. First, apoC-III
inhibits lipoprotein lipase activity (Fig. 2). Although the mechanism of
Fig. 3. Biology and pathology of the hepatic clearance of remnant chylomicrons. Graphic this action is not yet understood, it may act by interfering with the bind-
symbology: open, double-surrounded ellipsoids indicate gene targets. Red circles indicate ing of lipoprotein lipase to lipids, which would result in a decreased ac-
gene targets with known causal loss-of-function mutations: apoE (1) and apoA-V (2) in tivity and enhanced inactivation of the enzyme. Second, apoC-III also
dysbetalipoproteinemia, and HL and/or LPL (3), and LRP1 (4). Abbreviations used include
interferes with the binding of apoB-100 and apoE to hepatic receptors,
CE, cholesteryl ester; CM, chylomicron; HL, hepatic lipase; HSPG, heparan sulfate
proteoglycans; LDLR, LDL receptor; LRP1, LDLR-related protein-1; LPL, lipoprotein lipase;
leading to a decreased lipoprotein remnant clearance by receptor-
TG, triglycerides; TRL, triglyceride-rich lipoprotein. (For interpretation of the references to mediated endocytosis. ApoC-III also impairs the binding of lipoproteins
color in this figure legend, the reader is referred to the web version of this article.) to glycosaminoglycans.
APOC3 is also a PPAR gene target [48]. In contrast to its action on the
deficiency in mice has been experimentally demonstrated to increase expression of APOC2 and APOA5 (see Sections 1.3.6.2 and 1.3.5, respec-
the production rate of CMs by the small intestine [40]. This could be tively), activated PPAR-alpha inhibits the transcription of the gene,
due to the presence of apoA-V in the bile [41]. which results in a concomitant reduction of both the serum levels of
this protein and those of triglycerides.
1.3.6. Apolipoproteins C
1.3.7. Apolipoprotein E
The human apoCs (i.e., C-I, C-II, and C-III) are distributed among all
In the circulation, TRLs (i.e., CMs and VLDL) are immediately hydro-
lipoprotein classes, especially TRL and HDL [42]. Nascent apoCs are
lyzed by lipoprotein lipase, leading to the generation of TRL remnants.
largely released in their lipid-poor form and rapidly associate with cir-
During lipolysis, these remnants recruit HDL-derived apoE, which will
culating lipoproteins. Under fasting conditions, apoCs are mainly associ-
eventually facilitate their rapid clearance through specific endocytic re-
ated with HDL, whereas in the fed state they preferentially redistribute
ceptors [49] (Fig. 3). Human apoE serves as a ligand for the LDL receptor
to serum CMs and VLDL.
and the LDL receptor-related protein [49], promoting the efficient clear-
ance of TRL from circulation. The LDLR preferentially mediates the clear-
1.3.6.1. Apolipoprotein C-I. ApoC-I is the smallest exchangeable protein of ance of apoB-containing lipoproteins, which include the LDL and
the family and is synthesized in the liver [15,43,44]. Several experimen- different types of TRLs containing apoB-100, or combinations of apoB-
tal studies support a role of apoC-I in TRL metabolism [42]. For instance, 48 and apoE [49], while LRP preferentially interacts with apoE [50].
apoC-I inhibits the lipoprotein lipase activity in vitro, and transgenic ApoE also binds to specific subsets of the sulfate groups of the heparan
mice overexpressing the human protein display hypertriglyceridemia. sulfate chains and mediates the clearance of TRLs by hepatic HSPGs
Like apoC-III (see Section 1.3.6.3), apoC-I also inhibits the clearance of [50,51] even though this property is not exclusive for apoE. Other pro-
TRLs and their remnants by impairing interactions with their receptors, teins associated with TRLs (i.e., apoB-48 [52], apoB-100 [53], and
this action being in part related to the apoC-I-induced displacement of apoA-V [34] as well as the lipolytic enzymes lipoprotein lipase and he-
apoE. patic lipase [54]) also interact with HSPGs. This interaction may
Author's personal copy

J. Julve et al. / Clinica Chimica Acta 455 (2016) 134–148 139

contribute to sequestrating the TRL remnants in the space of Disse be- respect to non-deficient subjects [55]. Of note, however, the size of post-
fore they are rapidly internalized by hepatocytes either directly or indi- prandial CMs from CD36-deficient patients is smaller than those from
rectly via binding to endocytic receptors (i.e., LDLR and LRP). non-deficient subjects, thereby indicating a potential role of CD36 in
The endocytic uptake of TRLs mediated by the LRP is more complex postprandial CM formation rather than in fatty acid uptake [8].
than that of the classical LDLR-mediated endocytosis. It has been sug-
gested that LRP-mediated internalization might prevail in the liver during 2.1.2. Fatty acid binding proteins (FABPs)
the postprandial state [49]. During the LRP-mediated internalization, Within the enterocyte, cytosolic FABPs (L-FABP and I-FABP) promote
TRLs are thought to be hydrolyzed in peripheral endosomes, where the targeted delivery of fatty acids to specific metabolic sites during post-
core lipids and apoB-48 are targeted to lysosomes. Conversely, most of prandial lipemic response [8]. Different findings suggest a role of L-FABP
the TRL-derived apoE content is maintained in peripheral recycling in intracellular lipid trafficking [8] (Fig. 1). This protein displays an aux-
endosomes, thus maintaining the production and release to serum of iliary role, along with CD36, in the exportation of nascent CMs from the
apoE-containing HDL, which in turn accelerate CM remnant enrichment endoplasmic reticulum to the Golgi via PCV. Consistent with this, L-
with apoE. FABP-deficient mice accumulate triglycerides within the enterocytes
concomitantly with a reduction in triglyceride release into circulation.
2. Metabolism of CM However, there are no reports on human mutations or variants within
the FABP1 gene leading to a dysfunctional L-FABP. Regarding other
Dietary lipid trafficking compressed from enterocyte absorption FABP, patients bearing a genetic sequence variant in the human gene
until their delivery into systemic cells is a complex, multistep process (FABP2) present elevated serum triglyceride levels [8]. Although the im-
involving the participation of several key molecular players. We will pact on postprandial lipemia was not studied in these patients, there
mainly focus on proteins involved in clinically relevant alterations were no alterations in intestinal fatty acid absorption, thus showing
influencing CMs and will distinguish between the intracellular (see that FABP2 may not be essential in this process [8].
Section 2.1) and extracellular (see Section 2.2) determinants.
2.1.3. Enzymes involved in triglyceride synthesis
2.1. Intracellular metabolic determinants of synthesis and secretion of CM Dietary fatty acids and monoglycerides are intracellularly esterified
within the enterocytes by monoglyceride acyltransferases (MGATs, cod-
During CM assembly and secretion, dietary free fatty acids (mainly ified by MGAT2 and MGAT3) to form diglycerides. Subsequently, newly
long-chain fatty acids) are internalized across the luminal membrane of synthesized diglycerides are acylated by diglyceride acyltransferases
enterocytes via passive diffusion and, to a lesser extent, by different (DGATs) to form triglycerides [8] (Fig. 1).
fatty acid transporters [8]. Fatty acids within the enterocyte are trans- Functional studies conducted in MGAT2-deficient mice, the main
ferred to the endoplasmic reticulum by intracellular fatty acid binding isoform in mice, reveal that the absence of this enzyme results in a di-
proteins (FABP), L-FABP (FABP-1), and I-FABP (FABP-2), where fatty minished postprandial triglyceridemia [56]. In relation with the other
acids are re-esterified into triglycerides (Fig. 1). In parallel, the transport enzyme involved in triglyceride synthesis during CM packaging, an im-
of dietary cholesterol occurs through the brush border by specific choles- paired DGAT function attributed to mutations within the DGAT1 [8] has
terol transporters (i.e., Niemann–Pick C1-like 1 – NPC1L1 – and Scavenger been found in a family with congenital diarrheal disorder [57]. In con-
receptor class B type 1—SR-BI). Free cholesterol may be either transported trast, mice lacking DGAT1 show only delayed fat absorption. This appar-
back to the intestinal lumen by ABCG5/G8 or esterified by the enzyme ent disparity could possibly be explained by the fact that mice also
acyl CoA acyl transferase (ACAT)-2. Inside the endoplasmic reticulum, express DGAT2 in the intestine, which may compensate, at least in
newly synthesized apoB-48 acquires triglycerides, cholesteryl esters, part, for the deficiency in DGAT1.
and phospholipids in a process catalyzed by the enzyme microsomal tri-
glyceride transfer protein (MTTP) to produce the primordial CMs [8] 2.1.4. MTTP
(Fig. 1). Further lipidation of these newly produced primordial CMs by MTTP controls apoB lipidation in the endoplasmic reticulum in both
MTTP form preCMs. During this process, these lipoproteins further ac- the liver and small intestine and therefore participates in the formation
quire apoA-IV (see Fig. 1). PreCMs are then exported to the Golgi in of VLDL and CMs, respectively [58]. MTTP acts as a chaperone to assist in
preCM transport vesicles (PCV) from the endoplasmic reticulum in a pro- apoB folding and catalyzing the addition of triglycerides, esterified cho-
cess modulated by CD36 and L-FABP. When PCVs reach the Golgi, they lesterol, and phospholipids to the apoB-48 to produce primordial CMs
fuse with the cis-Golgi in a process controlled by both the secretion- [8,59] (Fig. 1).
associated, Ras-related GTPase 1B (Sar1b) and the soluble N-ethyl
maleimide sensitive-factor attachment protein receptor proteins 2.1.5. Coat protein complex II (COPII)
(SNARE). In the Golgi, preCMs further acquire apoA-I and, to a lesser ex- Newly synthesized and properly folded proteins are transported
tent, apoA-II to form mature CMs. Subsequently, mature CMs are from the endoplasmic reticulum to the Golgi via COPII vesicles [60].
exported as vesicles from the Golgi to the basolateral surface of the The intracellular traffic of apoB-48-containing CMs is dependent on
enterocyte and are released into the lymph, from where they will reach the COPII complex, which allows the vesicle budding from the endo-
the blood. plasmic reticulum membrane to transport newly synthesized proteins
to the Golgi. Secretion-associated, Ras-related GTPase 1B (Sar1b),
2.1.1. Fatty acid transporters which is encoded by the gene SARA2, is one of the main components
As previously stated (see Section 2.1), the fatty acid absorption by of this multicomplex protein and plays a pivotal role in the assembly, or-
enterocytes is mainly produced via passive diffusion (Fig. 1). Albeit ganization, and function of the COPII. In the context of CM packaging, it
minor, fatty acids are also transported via certain fatty acid transporters has been proposed that Sar1b might gather COPII, which in turn assem-
located at the luminal membrane of enterocytes [18]. One of these bles the lipid vesicles to transport primordial CM particles to the Golgi
transporters is CD36, which is abundantly expressed in the small intes- (pre-CM transport vesicle, PCV in fig. 1). Sar1b overexpression enhances
tine and where it is thought to exert a role in fat absorption. This is con- intestinal lipoprotein trafficking and sorting through the stimulation of
sistent with its expression pattern, which is abundant in the proximal CM assembly and release (extensively reviewed in [8]).
intestine, and its apical localization within the luminal surface of
enterocytes. CD36 deficiency in humans was not found to be associated 2.1.6. Sortilin-1
with altered fatty acid absorption but, rather, with higher postprandial Sortilin-1 (SORT1) is one of the members of the multi-ligand vacuo-
serum levels of triglycerides and apoB-48-containing lipoproteins in lar protein sorting 10 receptor family. It is primarily localized in the
Author's personal copy

140 J. Julve et al. / Clinica Chimica Acta 455 (2016) 134–148

trans Golgi and early endosomes. Although it mainly functions as a traf- 2.2.2. Hepatic lipase
ficking receptor to sort lysosomal hydrolases to the lysosome, a small Hepatic lipase is a member of the lipase gene family that is mainly
fraction of this receptor is translocated to the cell membrane, where it synthesized and secreted by hepatocytes [71]. This enzyme may hydro-
may act as an uptake receptor, mediate the signal transduction of lyze triglycerides and phospholipids in different lipoproteins, but its
neurotrophins, or be cleaved by a specific secretase to release a soluble function is critical in the conversion of intermediate-density lipopro-
extracellular domain of still unknown function [61] SORT1 has been re- teins (IDLs) to low-density lipoproteins (LDL) and in the conversion of
ported to bind LDL and facilitate its uptake and degradation. Also, SORT1 triglyceride-rich HDL into triglyceride-poor HDL. Apart from its catalytic
has been reported to bind lipoprotein lipase and receptor-associated action, it has also been described to act as a ligand of lipoproteins, there-
protein (RAP) [61], apoA-V [62], and apoE [63]. Beyond its role in the by facilitating the removal of remnant lipoproteins (Fig. 3). In this re-
clearance of LDL particles from circulation, it has been proposed that gard, it has been suggested that hepatic lipase may mediate the
SORT1 may also modulate hepatic VLDL secretion [61]. Since this recep- interaction between remnant lipoproteins and cell surface receptors
tor binds apoB-100, but not apoB-48 [64], it could be speculated that it and/or HSPGs.
would not display any role in CM synthesis.
2.2.3. Glycosylphosphatidylinositol-anchored high-density lipoprotein-
binding protein 1 (GPIHBP1)
2.2. Extracellular metabolic determinants of catabolism and clearance of Human GPIHBP1 is expressed in the same tissues that also express
CMs and their remnants lipoprotein lipase [65]. Although it was first identified as an HDL binding
protein, it directs the transendothelial transport of lipoprotein lipase to
In circulation, CMs acquire apoC-II, C-III, and E mainly from HDL and help anchor CMs to the endothelium. The acidic domain of GPIHBP1 ap-
are subjected to triglyceride hydrolysis by lipoprotein lipase (Fig. 2). The pears to be critical for lipoprotein lipase binding, since its replacement
function of this enzyme is modulated by apoC-II, which enhances the leads to an impaired ability of GPIHBP1 to bind both lipoprotein lipase
catalytic rate of the enzyme and is inhibited by apoC-III. During this hy- and CMs (Fig. 2). Interestingly, mutations in the heparin-binding do-
drolytic process, a substantial portion of phospholipid, apoAs, and Cs is mains of both LPL and APOA5 abrogate the binding of these proteins to
removed from the delipidated TRLs and is transferred to the HDL GPIHBP1. GPIHBP1 also prevents lipoprotein lipase inhibition by
fraction. Angptl3 and Angptl4 [72], thereby providing another mechanism to
The physiological importance of other determinants of CM clear- preserve the lipoprotein lipase function.
ance has been revealed from genetic defects in humans in recent
years [65] (Fig. 2), including the lipase maturation factor (LMF) 1, 2.2.4. Lipase maturation factor (LMF) 1
glycosylphosphatidylinositol-anchored high-density lipoprotein- LMF1 is a chaperone required for the proper posttranslationally fold-
binding protein 1 (GPIHBP1), and apoA-V. In addition, the hydrolysis ing and processing of lipoprotein lipase and hepatic lipase and, therefore,
of CM triglycerides originates remnant CMs, which are removed is critical for their function [65]. LMF1 is a membrane-bound protein that
from circulation by the liver. This is, in part, due to the enrichment is localized in the lumen of endoplasmic reticulum. Although mutations
of these CM remnants with apoE, which promotes their binding to in this gene are rare, they are associated with a reduced expression of
HSPG on the luminal membrane of hepatocytes, and it is the protein both lipases in patients with severe hypertriglyceridemia (Fig. 2). The
ligand recognized by LDLR and LRP [66], a process which is facilitated fact that the tissue distribution of LMF1 is not restricted only to the tis-
by hepatic lipase [67,68]. sues synthesizing these two lipases suggests that LMF1 may exert
broader biological functions.

2.2.1. Lipoprotein lipase 2.3. Receptors for CM remnant clearance


Lipoprotein lipase is a main determinant for postprandial lipemia
[69]. This lipase is primarily produced by the parenchymal cells of adi- The liver-dependent clearance from the circulation of CMs and VLDL
pose tissue, skeletal muscle, and myocardium. It is anchored to heparan remnants via LDLR and LRP receptors is apoE-dependent [73]. In contrast,
sulfate chains (i.e., HSPGs) on the luminal surface of vascular endotheli- apoB-48 does not contribute to receptor recognition. Triglyceride-rich
um, where it hydrolyzes the triglycerides of postprandial CMs and large VLDL produced by the liver is also cleared from circulation competing
VLDL (Fig. 2). with CMs. The delayed clearance of CMs is concomitantly accompanied
The expression of lipoprotein lipase is regulated in a tissue/cell- by a delay in the clearance of VLDL, since the LDLR preferentially takes
specific manner and in response to metabolic demands in different tis- up CM remnants. In this regard, another additional potential participant
sues. During triglyceride hydrolysis, the released fatty acids are then in the clearance of postprandial VLDL is the VLDL receptor (VLDLR).
taken up and reesterified for storage or oxidized to provide energy to HSPGs and, in particular, transmembrane syndecans have also been
adipose tissue and muscle, while some fatty acids remain in the circula- proposed to mediate the binding and uptake of CM remnants by the
tion bound to albumin. The active form of lipoprotein lipase is a homo- liver [74].
dimer, which may eventually dissociate to produce inactive monomers.
The function of lipoprotein lipase is modulated by several factors 2.3.1. LDLR
(depicted in Fig. 2). First, apoC-II enhances its action, while apoC-III is LDLR binds apoB-100 and/or apoE-containing lipoproteins. There-
the endogenous inhibitor of this enzyme [8] (see Sections 1.3.6.2 and fore, it accounts for the bulk clearance of serum lipoproteins into the
1.3.6.3). Second, genetic mutations (see Section 3.1.1) have also re- liver, including the apoE-containing CM remnants [73] (Fig. 3). LDLR de-
vealed the physiological importance of the lipase maturation factor ficiency in both patients and animal models does not result in a defec-
(LMF1), GPIHBP1, and apoA-V (encoded by APOA5) in regulating the ca- tive clearance of CM remnants, indicating the existence of alternative
tabolism of CMs [65]. apoE-specific remnant receptors for their clearance [66].
Lipoprotein lipase is inhibited by the angiopoietin-like proteins 3, 4,
and 8 (Angptl3, Angptl4, and Angptl8, respectively) [69]. The mecha- 2.3.2. LDL receptor-related proteins (LRPs)
nisms of action of both Angptl3 and Angptl4 in disabling lipoprotein li- One of the main candidates for apoE-specific CM remnant receptors
pase are distinct [70]. Angptl3 reduces the lipoprotein lipase activity is the LRP1 [73]. LRP1 belongs to the LRP family that represents a group
without inducing its inactivation, whereas Angptl4 irreversibly sup- of structurally related transmembrane proteins involved in a diverse
presses lipoprotein lipase activity. Angptl8 inhibits lipoprotein lipase ei- range of biological activities [73]. In this regard, the discovery that
ther directly or indirectly by promoting the activation of Angptl3 [69]. LRP1 binds apoE led to the notion that it acts as a remnant receptor
Author's personal copy

J. Julve et al. / Clinica Chimica Acta 455 (2016) 134–148 141

[73] (Fig. 3). Consistently, LRP1 deficiency or inhibition does result in a consisting of fasting serum triglyceride N1000 mg/dL, concomitant
decreased clearance of CM remnants. with the presence of CMs.
Aside from LRP1, the LRP family also includes other members, such
as LRP1b, LRP2 (also called megalin), LRP4 (also called multiple epider- 3.1.1. Monogenic hyperchylomicronemia
mal growth factor-like domains protein 7 or MEGF7), LRP5/6, LRP8 (also Type I hyperlipidemia, or familial hyperchylomicronemia, is a rare
called apo E receptor 2), the VLDL receptor (VLDLR), and LR11/SorLA1 disease with a prevalence of ~1 case in 1,000,000 inhabitants [89], char-
(LDL receptor relative with 11 ligand binding repeats/sorting protein- acterized by the early onset of severe hypertriglyceridemia associated
related receptors containing LDLR class A repeats). Of note, it has been with chylomicronemia but without an increase in VLDL. It is mainly
recently proposed that apoA-V might mediate in the binding of this due to rare homozygous or double heterozygous, loss-of-function LPL
last sortilin class to the HSPG of endothelial cell surfaces [75]. gene variants (HLP1A, OMIM 238600) (Fig. 2). Also, mutations in sever-
al additional loci, which encode for proteins involved in the activity, as-
sembly, or transport of lipoprotein lipase, have been identified in
3. Pathology
patients with familial hyperchylomicronemia without LPL mutations.
According to the OMIM database, less common causes of familial
There are some disorders in which CMs are involved. CMs are
chylomicronemia (also represented in Fig. 2), are rare, loss-of-function
considered to be present in blood serum when triglyceride concen-
variants in APOC2 (HLP1B, OMIM 207750), GPIHBP1 (HLP1D, OMIM
tration is above 1000 mg/dL (N 11.3 mmol/L) [76]. Severe hypertri-
615947), LMF1 (combined lipase deficiency, OMIM 246650), or APOA5
glyceridemia is also defined by the same criteria. The prevalence of
(late on-set hyperchylomicronemia, OMIM 144650), as well as the pres-
hyperchylomicronemia in North America is between 1, 5 and 4 per
ence in blood serum of circulating lipoprotein lipase inhibitors (HLP1C,
1000 individuals [77]. Hyperchylomicronemia is a well known cause
OMIM 118830). However, in the latter, only one affected family has
of acute pancreatitis. Although the exact pathophysiological mechanism
been described [90] as presenting an autosomal dominant mode of
remains unclear [78], pancreatic ischemia and acidosis—due to an im-
inheritance.
pairment in the blood flow due to increased viscosity—have been pro-
Recently, two studies of the molecular diagnosis of hyperchylo-
posed. Hypertriglyceridemia was considered causative of the attack in
micronemia in a clinical laboratory setting have been reported. In a sam-
up to 10% of all pancreatitis episodes and in more than a half of gesta-
ple of 29 patients with severe hypertriglyceridemia from Spain [91],
tional pancreatitis patients [79]. It is noteworthy that five of the six clas-
44.8% were homozygous or compound heterozygous for rare, loss-of-
sical hyperlipidemic phenotypes show HTG, but only those defined
function variants in the LPL gene and could be classified as HLP1A, and
with high CM concentrations (types I and V) may trigger acute pancre-
10.3% were heterozygous or compound heterozygous with a rare and
atitis [79].
common variant. Additionally, 6.9% of the patients were homozygous
Also, there is epidemiological and clinical evidence that fasting, and
or compound heterozygous for loss-of-function variants in APOA5. In
to a greater extent non-fasting, hypertriglyceridemia are important risk
the other study, in a sample of 149 severe HTG patients from Italy
factors for cardiovascular disease (CVD) [11,80,81]. Although CMs are
[92], the proportion of homozygous or compound heterozygous for
too large to cross the endothelium, their smaller remnant particles can
loss-of-function variants in the LPL gene was 20.1%, while 14.1% were
penetrate the arterial wall, contributing to plaque formation and induc-
heterozygous for rare LPL gene variants. The higher number of HLP1A
ing endothelial dysfunction [82]. The atherogenic potential role of TRL
in the first study could be explained by the higher proportion of new-
remnants is clearly demonstrated in the type III hyperlipidemia pheno-
borns in the Spanish sample, as HLP1 has an earlier onset than other
type, a remnant hyperlipidemia where unequivocal accelerated athero-
forms of hyperchylomicronemia.
sclerosis and CVD have been described. CMs and VLDL share the same
clearance pathway in a competitive manner [83], but CMs seem to be
3.1.2. Polygenic hyperchylomicronemia
the preferred substrate of lipoprotein lipase [84].
3.1.2.1. Type V hyperlipidemia. The presence of fasting CMs is also a char-
3.1. Hyperchylomicronemia acteristic of type V hyperlipidemia (HLP5, OMIM 144650), but this form
also presents with a concomitant increase in VLDL and is usually ob-
The hyperchylomicronemia syndrome is a disorder characterized by served in adults rather than children. Type V hyperlipidemia is thought
extreme hypertriglyceridemia, the presence of chylomicrons, and one to be the result of complex interactions between genetics and environ-
or more of the following clinical manifestations: eruptive xanthomas, mental factors, with the former including heterozygous, loss-of-func-
lipemia retinalis, hepatosplenomegaly, recurrent abdominal pain, and/ tion mutations in LPL (in approximately 10% of affected individuals)
or acute pancreatitis [85]. Although CMs are usually considered to be together with contributions from more common variants in APOA5,
present when serum triglyceride concentrations are above 1000 mg/dL, APOE, or ANGPTL3 [93,94]. In a study of 86 Dutch patients with severe
the symptoms associated with the chylomicronemia syndrome almost HTG, 26% of the cases were carriers of only common variants in LPL
always occur at higher triglyceride levels [86]. Early diagnosis of severe and APOA5 [95]. In a Spanish population, the common LPL variants
chylomicronemia syndrome is crucial to avoid pancreatitis-derived con- p.Asp36Asn and p.Asn318Ser, APOA5 p.Ser19Trp, and APOE4 were inde-
sequences, including abdominal pain and diabetes mellitus. pendently associated with an increase in serum triglyceride levels [96],
There are genetic causes of hyperchylomicronemia, but the ma- although an additive effect was observed among the LPL p.Asp36Asn,
jority of patients seem to have a combination of a common genetic APOA5 p.Ser19Trp, and APOE4 variants. Therefore, type V hyperlipid-
predisposition and an acquired cause of hypertriglyceridemia. Famil- emia is thought to be of a polygenic nature in most cases, where the
ial combined hyperlipidemia, familial hypertriglyceridemia and, oc- individual's environment is obviously relevant and patients with genet-
casionally dysbetalipoproteinemia, in combination with untreated ic susceptibility (e.g., those with heterozygous mutations in the LPL
diabetes mellitus, high-saturated fat/high carbohydrate diet, certain gene) in combination with pregnancy, excessive alcohol intake, obesity,
drug therapies (e.g., estrogen therapy, antiretroviral drugs), or ex- uncontrolled type 1 or 2 diabetes mellitus, or different medications
cessive alcohol consumption account for nearly 90% of the cases of (i.e., estrogens, glucocorticoids, tamoxifen, 13-cis-retinoic acid, antire-
hyperchylomicronemia in adults [85,86]. troviral therapies) cooperate in developing the disease [89,97].
As mentioned above, five of the six classical HLP phenotypes initially In this context, it is known that an accumulation of rare variants
classified by Fredrickson have HTG as a defining component, and contributes to the heritability of complex traits among individuals at
the chylomicronemia syndrome is a feature of two of them—type I the extreme of a lipid phenotype [98]. It has been estimated that ap-
and type V HLP [87,88]—which have a partially common phenotype proximately 53% of new missense variants in humans have mildly
Author's personal copy

142 J. Julve et al. / Clinica Chimica Acta 455 (2016) 134–148

deleterious effects and give rise to many low-frequency deleterious alle- malabsorption, as this suggests a primary defect in chylomicron
lic variants [99], the accumulation of which can produce a burden of var- production.
iants with small effect size. In a recent study performed in healthy male Inherited causes of hypochylomicronemia are caused by very rare
adults, 42 SNPs in 23 genes explained 88% of the variance in chylomi- mutations in the genes involved in the packaging and secretion of
cron response to dietary fat [100], some of them affecting fatty acid up- apoB-containing lipoprotein particles, resulting in either a strong de-
take and triglyceride synthesis (e.g., CD36, ELOVL5, SLC27A5, and crease or the absence of CMs, VLDL, and LDL in blood serum. Three
SLC27A6), CM clearance (LPL, APOB, APOA5, and LIPC), insulin and carbo- inherited diseases affecting chylomicrons can be distinguished: CM re-
hydrate metabolism (IRS1, INSIG2, and TCF7L2), HDL function (APOA1 tention disease (CMRD, OMIM 246700), abetalipoproteinemia (ABL,
and ABCA1), and appetite and body weight regulation (MC4R). Further OMIM 200100), and familial hypobetalipoproteinemia (FHBL1, OMIM
investigations are needed to test whether common variants in these 615558).
genes accumulate in chylomicronemia, but in the study by Martín-Cam-
pos et al. [91], 28% subjects with severe HTG were heterozygous for loss- 3.2.1. Chylomicron retention disease
of-function variants in LPL or a combination of common, small effect This disease, also called Anderson disease, is an autosomal recessive
variants in LPL, APOA5, APOE, and GPIHBP1. disorder characterized by an intestinal defect in lipid transport due to a
failure of CM formation in enterocytes, which results in severe malab-
3.1.2.2. Dysbetalipoproteinemia. Another polygenic disorder involving sorption with steatorrhea, fat-soluble vitamin deficiency, low blood
TRL remnants is dysbetalipoproteinemia (HLP3, OMIM 107741). cholesterol levels, and failure to thrive in infancy. Chylomicron reten-
Type III hyperlipidemia is caused by the conjunction of a defect in the tion disease is caused by homozigous and compound heterozygous mu-
clearance of remnant particles, mainly due to homozygosity for the tations in SAR1B, a gene encoding Sar1 homolog B GTPase [108] (Fig. 1).
binding-defective apoE2 isoform or other rare APOE variants, with Sar1b is one of the subunits of the coat protein (COPII) complex, which
other environmental and/or genetic factors that cause the overproduc- has been found to be critical for the vesicular transport of apoB-48-
tion of triglyceride-rich particles or a reduction in LDL receptor activity containing particles from endoplasmic reticulum to the Golgi.
[101] (Fig. 3). This would be the reason why only 10–20% of E2/E2 sub-
jects develop type III hyperlipidemia and 30–40% of them have a muta- 3.2.2. Abetalipoproteinemia
tion in APOA5 as a secondary factor [88,102]. In contrast, less than 5% of Familial abetalipoproteinemia is an autosomal recessive disorder
type III hyperlipidemia patients have a monogenic, dominant form due involving the improper packaging and secretion of apoB-containing
to a mutation in APOE, which would not require other factors to develop particles as a root cause. It is caused by homozygous and compound het-
the disease. Type III hyperlipidemia patients usually exhibit fasting HTG erozygous mutations in the gene encoding the large subunit of the
between 300 and 1000 mg/dL due to the impairment in hepatic rem- MTTP [109] (Fig. 1). MTTP is a heterodimer composed of the multifunc-
nant clearance and a putative impairment in lipoprotein lipase activa- tional protein disulfide isomerase (P4HB), which is related with Cole-
tion (i.e., from the presence of reduced-expression APOA5 mutants), Carpenter syndrome and is a unique, large subunit of MTTP. This sub-
which could induce hyperchylomicronemia frequently, at least during unit binds to amino acids 1–264 and 512–721 or 270–570 of apoB—an
the postprandial state [103] (Fig. 3). interaction important for the initiation of translocation of the nascent
apoB chain to the endoplasmic reticulum and for the addition of lipids
to this chain, causing the defective cellular secretion of the apoB. The in-
3.1.3. Autoimmune hyperchylomicronemia cidence of familial abetalipoproteinemia is less than 1 in 1 million [110].
Some autoimmune diseases, such as systemic lupus erythematosus,
systemic sclerosis, polymyositis, and rheumatoid arthritis, have shown 3.2.3. Familial hypobetalipoproteinemia (FHBL)
the presence of anti-lipoprotein lipase antibodies, which could explain FHBL1 is, in contrast to chylomicron retention disease and abeta-
some hyperlipidemias observed during rheumatic diseases [104]. Circu- lipoproteinemia, an autosomal co-dominant disorder. The majority of
lating anti-lipoprotein lipase antibodies could bind the enzyme familial hypobetalipoproteinemic patients are heterozygotes, carriers
and have an inhibitory effect on activity and, therefore, impair triglycer- of a truncating mutation in APOB that results low serum lipids, especial-
ide degradation and lead to hypertriglyceridemia. Anti-lipoprotein li- ly LDL cholesterol and intestinal and hepatic triglyceride accumulation
pase antibodies could be present both in normotriglyceridemic and as a result of the decreased export of triglycerides [111]. Homozygous
hyperchylomicronemic subjects without autoimmune disease [105]; subjects are rare and present, as in ABL, a near absence of apoB-
however, although the proportion of anti-lipoprotein lipase antibodies containing lipoproteins, with more severe clinical manifestations in
was significantly higher in hyperchylomicronemic subjects, it would ex- childhood, such as failure to thrive, intestinal fat malabsorption causing
plain only a minority of sporadic cases. In a study of 63 HLP5 patients, liposoluble vitamin deficiency, and nonalcoholic liver steatosis.
27% presented an anti-lipoprotein lipase serum level above the 95th
percentile of a control population, but less than 10% displayed a sub- 3.2.4. Familial combined hypolipidemia
stantial inhibition of serum lipolysis [105]. In another study of 44 pa- Familial combined hypolipidemia (FHBL2, OMIM 605019) is caused
tients with HTG, anti-lipoprotein lipase antibodies were present in by homozigous and compound heterozygous mutations in the ANGPTL3
only 2 of them [106]. It is interesting to point out that in the first descrip- gene and, in addition to low apoB-containing lipoproteins, includes low
tion of an autoimmune hyperchylomicronemia due to a defect of lipo- HDL cholesterol [112–115]. Angptl3 is secreted and expressed almost
protein lipase activity, in a 35-year-old woman with severe HLP1 exclusively in the liver and in mice plays a role in the inhibition of lipo-
[107], the circulating anti-lipoprotein lipase antibodies were bound to protein lipase and endothelial lipase (encoded by LIPG) [116]—two key
CMs, thus emphasizing their ability to transport lipoprotein lipase. enzymes in the metabolism of triglyceride-rich particles and HDL. In a
population study, common loss-of-function mutations in the ANGPTL3
3.2. Hypochylomicronemia gene either interfered with the synthesis/secretion of the protein or
failed to inhibit lipoprotein lipase activity in vitro but did not reduce
Hypochylomicronemia is defined as the low level or absence of post- HDL cholesterol (HDL-C) [117]. Serum Angptl3 has also been strongly
prandial CMs, and it can result from genetic or acquired causes. correlated with hepatic lipase (encoded by LIPC) activity and serum
As hypolipidemia is frequently in association with different diseases, HDL cholesterol in healthy Japanese subjects [118] but not with serum
especially those that are serious and consumptive, the diagnosis of triglycerides [118–120] or lipoprotein lipase activity [118]. However,
hypochylomicronemia is usually considered in the presence of very the complete absence of Angptl3 results in an increased lipoprotein li-
low serum lipids either in healthy subjects or in concomitance with pase activity, and decreased serum free fatty acids [121].Thus, the effect
Author's personal copy

J. Julve et al. / Clinica Chimica Acta 455 (2016) 134–148 143

of Angptl3 on lipid metabolism seems to involve different, as of yet not exacerbated in some metabolic diseases, such as type 1 and 2 diabetes.
completely understood, mechanisms. However, the potential therapeutic effects of remnant lipoproteins-
lowering measures on cardiovascular outcomes remain largely un-
3.3. Chylomicron remnants and cardiovascular disease known, and further studies are needed in this field.

Postprandial hyperlipidemia is considered an important risk factor


for cardiovascular disease. Indeed, three prospective epidemiologic 3.4. Regulation of intestinal chylomicron production in type 2 diabetes
studies found highly significant associations between non-fasting tri-
glycerides and the risk of adverse cardiovascular events [80,81,122], The most common causes of elevated cholesterol and triglycerides in
and, also, strong evidence supports the role of cholesterol in remnant li- remnant lipoproteins are obesity and poorly controlled diabetes
poproteins as a clinically significant cardiovascular risk factor for CVD mellitus [124]. The increased secretion of liver-derived VLDL in insulin
[123,124]. The non-fasting triglycerides and remnant cholesterol are resistance states has been extensively investigated, and this explains,
mainly associated to CMs synthesized in the intestine and the liver- at least in part, the dyslipidemic profile in type 2 diabetic patients
derived VLDLs and also their corresponding remnant lipoproteins. As [11]. However, other studies have also shown an abnormal metabolism
commented above, both VLDL and CMs may compete for LPL—being of CMs in type 2 diabetic patients [132]. More importantly, insulin-
the CMs' preferred substrate—thereby prolonging the blood residence resistance is strongly associated with intestinal-derived apoB-48 pro-
time of VLDL particles [125]. For this reason, although 80% of the triglyc- duction rate in insulin-resistant subjects and type 2 diabetic patients
eride increase after a fat-load meal is associated to CMs [126], the main [132,147]. It should be noted that NPC1L1 and MTTP expression were
increase in particle number is due to an increase in VLDL particles [125, found to be increased in duodenal biopsies of type 2 diabetic patients,
127]. However, CMs and large VLDLs are too large to penetrate into the whereas ABCG5 and ABCG8 were downregulated [148]—thereby indi-
arterial intima, and only their remnants penetrate and, eventually, accu- cating that diabetic patients could have increased intestinal cholesterol
mulate in the atherosclerotic lesions [128]. This is in line with the fact absorption and increased levels of enterocyte cholesterol for CM assem-
that hyperchylomicronemia syndromes display increased risk of pan- bly. These findings are consistent with the positive effects of ezetimibe,
creatitis but in most cases no increase in cardiovascular disease [124]. the main drug targeting intestinal NPC1L1, on postprandial apoB-48
It should also be noted that postprandial hyperlipidemia may promote levels in dyslipidemic type 2 diabetic patients [149]. Furthermore, rem-
atherosclerosis indirectly by increasing the small, dense LDL and reduc- nant CMs were highly retained in arteries from diabetic rats, and this ef-
ing HDL [11]. fect was reversed by ezetimibe [133]. As described in Section 2.1.5,
Fasting serum apoB-48 levels were significantly higher in coronary Sar1b plays a pivotal role in the routing of apoB-48-containing CMs
disease patients compared with those without [129,130]. Importantly, from the endoplasmic reticulum to the Golgi apparatus. Sar1b expres-
apoB-48 is markedly increased in patients with early atherosclerosis sion has been recently associated to obesity and insulin-resistance in
having a new onset of lesion progression [130]. Consistent with these mice fed a high-fat diet by promoting CM production [150] (Fig. 1).
findings, apoB-48 levels under fasting conditions have also been signifi- One of the main incretins in humans is the glucagon-like peptide-1
cantly correlated with carotid intima-media thickness in normolipidemic (GLP-1), which is produced by intestinal enteroendocrine K-cells and L-
subjects [131]. Furthermore, both type 1 and type 2 diabetic patients ex- cells located in the intestinal mucosa next to the enterocytes [151]. The
hibited a greater total serum apoB-48 compared with that of controls ability of GLP-1 to prevent postprandial triglyceride increase has been
[132–135], and the severity of angiographically coronary artery disease demonstrated in healthy humans given GLP-1 infusions [152]. Further-
correlated with the postprandial response of apoB-48 in type 2 diabetic more, fat intake is a strong stimulus of GLP-1 secretion, whereas type 2
patients [136]. Importantly, this alteration was prevented in type 2 dia- diabetic patients displayed a reduced and delayed postprandial response
betic patients by improving their metabolic control [137]. However, in- [153]. The importance GLP-1 receptor stimulation for normalizing post-
ternationally standardized assays and normal reference ranges are prandial CM levels has been supported by the positive effects of the
needed for extending the use of apoB-48 determinations to clinical GLP-1 receptor agonist exenatide on intestinal CM secretion in healthy
practice. subjects [154]. Exenatide also reduced postprandial apoB-48 levels in
The potential atherogenic effects of remnant CMs have also been type 2 diabetic patients [155]. The mechanisms whereby GLP-1 prevents
demonstrated in vivo. Therefore, early studies with rabbits demonstrat- CM production remain poorly understood. However, the positive thera-
ed that carotid-perfused remnant CMs and LDL were retained within peutic effects may involve a GLP-1-mediated increase in insulin secretion
the subendothelial space [128]. The retention of cholesterol was partic- and impaired gastric emptying or a direct effect of the GLP-1 on
ularly high within the intima of heritable hyperlipidemic Watanabe rab- enterocytes [151]. Importantly, insulin inhibits apoC-III expression,
bits, particularly that of apoB-48-containing lipoproteins [128]. This which may overcome the unrestrained apoC-III production in the
finding, together with the presence of triglyceride-containing remnant insulin-resistance state [156], thereby enhancing the lipoprotein lipase-
lipoproteins in human atherosclerotic plaques [138–140], strongly indi- mediated lipolysis of CMs and promoting their liver uptake and clearance.
cate that remnant CMs may promote the initiation and progression of Recent evidence also supports the importance of GLP-1 neural accessibil-
lesion development by itself. The mechanism whereby the CM particles ity for controlling intestinal CM production, since the intracerebroven-
may promote atherosclerosis has also been extensively investigated tricular injection of the GLP-1 receptor agonist exendin 4 prevented
in vitro. Indeed, remnant CMs contribute to macrophage foam cell for- apoB-48 accumulation in hamsters via central melanocortin-4 receptors
mation after being taken up by multiple receptor-mediated routes [157].
[141–143]. Remnant CMs may also promote an increase in the inflam-
matory state of monocytes, thereby enhancing their susceptibility to en-
dothelium adhesion and invasion of the artery wall (reviewed in [144]). 4. Laboratory tests for CM analysis
Finally, some evidence suggests that remnant CMs may promote rodent
vascular smooth muscle cell proliferation [145,146], but the importance The potentially fatal risk of acute pancreatitis associated with
of these changes in human cells is unknown. hyperchylomicronemia makes their diagnosis and treatment very im-
In summary, the current consensus view supports the notion that portant. Secondary causes of chylomicronemia include hypothyroidism,
CM remnant particles are critical cardiovascular risk factors. Strong evi- unhealthy diet, excessive ethanol use, renal disease, HIV infection,
dence indicates that remnant CMs are proatherogenic lipoproteins, Cushing's syndrome, sarcoidosis and mieloma as well as different treat-
most likely by promoting macrophage foam cell formation and mono- ments such as antiretroviral therapy, estrogens, tamoxifen, 13-cis-
cyte dysfunction. It is also recognized that these actions are likely retinoic acid, and some antihypertensive or antipsychotic drugs [76].
Author's personal copy

144 J. Julve et al. / Clinica Chimica Acta 455 (2016) 134–148

4.1. Biochemical testing The determination of the main protein marker of chylomicrons,
apoB-48, is possible by electrophoresis of isolated TRL [162], especially
A laboratory diagnosis of hyperchylomicronemia is assumed when if followed by the Western blot. However, this methodology is imprac-
triglyceride levels exceed 1000 mg/dL. Ten to twelve hours of fast is usu- tical for clinical laboratory use, and enzyme-linked assays have been de-
ally recommended for the optimal assessment of serum triglyceride veloped. The low concentration of apoB-48 with respect to that of apoB-
values. 100 and the lack of internationally recognized standardized assays have
The method to directly demonstrate and quantitate hyperchylo- prevented stronger clinical evaluation of the assays [11].
micronemia requires the isolation of CMs by ultracentrifugation, typi-
cally at 10,000 g, for 30 min in saline solution at a density of 4.2. Genetic testing
1.006 g/mL. After this time period, CMs' float can be isolated and its
composition measured. This allows for the determination of the amount Genetic testing may be used to confirm clinical diagnosis, establish
of triglycerides or cholesterol associated with the CMs. This method of the carriers within families, and carry out genetic counseling for mono-
isolating chylomicrons is also useful for measuring (in the serum with- genic diseases affecting chylomicrons. Genetic testing requires previous
out chylomicrons) other clinical chemistry components that can be in- in-depth clinical, familial, and biochemical data to allow a diagnostic hy-
terfered with by hyperchylomicronemia. pothesis. This will be the basis for selecting which gene or genes will be
When hyperchylomicronemia is suspected in a serum sample, a clas- studied and in which sequential order and to predict how many patho-
sical way of confirming its existence is to keep the sample overnight at genic mutations will be expected depending on whether a recessive or a
4 °C and then look for CMs as they will float due to lower density with codominant or a dominant disorder is foreseen (Table 1). In most cases,
respect to the whole serum. As most measurements in clinical chemis- the method used will be the PCR of exons and exon–intron boundaries
try testing are performed in fasted patients, the presence of CMs may and Sanger sequencing. It is possible, and even likely, that in the near fu-
be unexpected; in these cases, hyperchylomicronemic patients should ture some assays could result from simultaneous gene testing using
first be asked about their period of fasting prior to blood sampling. next-generation sequencing methods. Gross deletion/insertions are in-
Only serious impairments of chylomicron metabolism will be de- frequent in monogenic disorders affecting chylomicrons, but methods
tected by traditional fasting testing, and this has been the basis for pro- to detect them should be considered if point mutations are either unde-
posing studies during a postprandial period. However, and contrary to tected or insufficient to explain the phenotype.
what was thought for many years, the VLDL remnants rather than the
CM remnants are the major component of the TRLs during the postpran- 5. Treatment of chylomicron disorders
dial state [158]. This should be kept in mind when using parameters that
are informative of the postprandial state such as non-fasting triglycer- 5.1. Hyperchylomicronemia
ides or calculated, non-fasting remnant cholesterol [11]. At least two
methods to quantitate remnant-like cholesterol and triglycerides are Hyperchylomicronemia is present mainly in type I and type V hyper-
available. One is based on the antibody-specific separation of lipopro- lipidemia. While the former is usually present in children and is charac-
teins containing apoB and A-I coupled to Sepharose 4B. All apoB-48- terized by a decrease in VLDL, the latter is usually seen in adults and is
and apoB-100-containing lipoproteins are recovered in the remnant also characterized by a concomitant increase in VLDL. In both cases,
fraction, where cholesterol and triglycerides can be measured [159]. the main risk is that of acute pancreatitis, which is especially high
The other system uses a specific detergent to modify CMs and VLDL with serum triglycerides N 1000 mg/dL.
remnants to allow cholesterol determination in these particles [160]. Al- Agents known to increase endogenous triglyceride concentration
though remnant-like particles have been reported to predict cardiovas- are to be avoided, including alcohol, oral estrogens, diuretics, isotreti-
cular events in patients with coronary artery disease [161], available noin, glucocorticoids, sertraline, and beta-adrenergic blocking agents.
data based on these two methods is limited [11].
Oral fat tolerance tests have also been used to follow postprandial 5.1.1. Dietary treatment of hyperchylomicronemia
lipemia, measuring mainly triglycerides as a marker. However, the Treatment for type I hyperlipidemia includes chronic dietary fat re-
lack of a standardized meal, sampling times, and reference ranges has striction (fat b 15% of total energy intake or 20 g/day) together with
limited the extension of these tests in a clinical setting [11]. medium-chain fatty acids that are thought to arrive directly to the

Table 1
Genetic diagnosis of monogenic diseases affecting chylomicrons.

Disease OMIM Gene or genes Inheritance Pathogenic Other lipoproteins affected


affected⁎ by the disease
mutations expected

Hyperchylomicronemia
Familial hyperchylomicronemia HLP1A, #238600 LPL
HLP1B, #207750 APOC2
HLP1D, #615947 GPIHBP1 Recessive Homozygous or compound heterozygous No
#144650 APOA5
#246650 LMF1⁎⁎

Hypochylomicronemia
Abetalipoproteinemia ABL, #200100 MTTP Recessive Homozygous or compound heterozygous Mainly all apoB-containing lipoproteins
Anderson disease or chylomicron CMRD, #246700 SARA2 Recessive Homozygous or compound heterozygous Mainly all apoB-containing lipoproteins
retention disease
Familial hypobetalipoproteinemia FHBL1 #615558 APOB Codominant Homozygous and heterozygous forms All apoB-containing lipoproteins
Familial combined hypolipidemia FHBL2, #605019 ANGPTL3⁎⁎ Recessive Homozygous or compound heterozygous Mainly all apoB-containing lipoproteins,
but HDL cholesterol may be decreased
⁎ In monogenic disorders only one gene contains the mutations; if mutations in more than one gene are the potential cause of the disease, those are usually studied depending on the
frequency (where the first studied is the one whose mutations are the most frequent cause of the disease).
⁎⁎ Mutations in these genes also directly affect HDL metabolism.
Author's personal copy

J. Julve et al. / Clinica Chimica Acta 455 (2016) 134–148 145

liver independently of chylomicron transport. Fish oil supplements are 5.2. Hypochylomicronemia
contraindicated in familial deficiency of lipoprotein lipase because
they contribute to CM levels [163]. 5.2.1. Dietary treatment of hypochilomicronemia
Treatment for type V hyperlipidemia depends on the level of triglyc- Monogenic diseases altering CM synthesis or secretion have in
erides. When fasting triglycerides N500 mg/dL patient need to be sub- common lipid malabsorption, which can lead to liposoluble vitamin
jected to therapeutic lifestyle change and be considered for drug deficiency by insufficient arrival to the tissues and steatorrhea,
treatment [76]. In hyperchylomicronemic patients with fasting triglyc- reflecting intestinal lipid malabsorption [110]. It is thus important to
erides N500 mg/dL and abdominal pain, or fasting triglycerides consider decreases in dietary fat intake, to supplement with medium-
N1000 mg/dL, the patients need to be hospitalized without receiving al- change fatty acids, and to add complements of liposoluble vitamins to
iments by mouth and be hydrated with intravenously administered prevent complications—especially neurological ones, as these may be
fluids to decrease serum triglycerides. Low-doses of insulin should irreversible.
also be administered (with glucose in non diabetic patients to prevent
hypoglycemia) [76]. Disclosures

The authors declare that there is no duality of interest associated


5.1.2. Drug treatment of hyperchylomicronemia with this manuscript.
Drug options for hyperchylomicronemia are the same as those for
hypertriglyceridemia treatment, even though maximal doses or drug
Conflicts of interest
combination could be tried.
Omega-3 fatty acids, at pharmaceutical doses (3–4 g/day) de-
All authors have read the journal's policy on disclosure of potential
crease serum triglycerides by about 30% and, therefore, have been
conflicts of interest. The authors declare that there is no duality of inter-
used in hypertriglyceridemia [164]. The mechanism of action in-
est associated with this article.
cludes the suppression of adipose tissue inflammation, which is
thought to increase insulin sensitivity and decrease the lipolysis me-
diated by the lipase-sensitive hormone. Also, omega-3 fatty acids in- Acknowledgments
crease lipoprotein lipase activity and fatty acid oxidation in adipose
tissue, skeletal muscle, and the heart, thus reducing fatty acid arrival This work was supported by Ministerio de Sanidad y Consumo,
to the liver, which is a major input for triglyceride and VLDL synthe- Instituto de Salud Carlos III, and FEDER “Una manera de hacer Europa”
sis and secretion. If omega-3 fatty acids are insufficient for control- (grant numbers: CP13-00070 to JJ; PI1401648 to FBV and JMMC; PI12-
ling hypertriglyceridemia, hypotriglyceridemic drugs can be added 0291 to JC-E). JJ is the recipient of a Miguel Servet Type 1 contract
to the treatment. Fibrates and nicotinic acid are medications (CP13-00070). CIBER de Diabetes y Enfermedades Metabólicas Asociadas
approved to treat hypertriglyceridemia in the context of a type V (CIBERDEM) is a project of the Instituto de Salud Carlos III.
hyperlipidemia [76]. Fibrates may decrease serum triglyceride up
to 50% by a variety of mechanisms mediated by PPAR-alpha activa- References
tion. Their use is contraindicated in those with hepatic or renal
[1] K. Nakajima, T. Nagamine, M.Q. Fujita, M. Ai, A. Tanaka, E. Schaefer, Apolipoprotein
dysfunction. Major side effects include lithogenicity and potentia- B-48: a unique marker of chylomicron metabolism, Adv. Clin. Chem. 64 (2014)
tion of the effects of oral anticoagulants. Nicotinic acid can decrease 117–177.
triglycerides by up to 35%, but it is not used as a primary agent for [2] J. Iqbal, M.M. Hussain, Intestinal lipid absorption, Am. J. Physiol. Endocrinol. Metab.
296 (2009) E1183–E1194.
chylomicronemic patients. Chylomicronemia in the setting of diabe- [3] W.J. Lossow, F.T. Lindgren, J.C. Murchio, G.R. Stevens, L.C. Jensen, Particle size and
tes can be improved by adequate insulinization. protein content of six fractions of the Sf 20 plasma lipoproteins isolated by density
There is poor or absent response to drugs in patients with type I gradient centrifugation, J. Lipid Res. 10 (1969) 68–76.
[4] H. Hayashi, K. Fujimoto, J.A. Cardelli, D.F. Nutting, S. Bergstedt, P. Tso, Fat feeding
hyperlipidemia.
increases size, but not number, of chylomicrons produced by small intestine, Am.
J. Physiol. 259 (1990) G709–G719.
[5] P. Wood, K. Imaichi, J. Knowles, G. Michaels, L. Kinsell, The lipid composition of
human plasma chylomicrons, J. Lipid Res. 5 (1964) 225–231.
5.1.3. Gene therapy of hyperchylomicronemia
[6] S.W. Sakr, N. Attia, M. Haourigui, et al., Fatty acid composition of an oral load affects
The first gene therapy treatment was recently approved by the chylomicron size in human subjects, Br. J. Nutr. 77 (1997) 19–31.
European Union for the treatment of lipoprotein lipase deficiency in [7] D. Lairon, J. Lopez-Miranda, C. Williams, Methodology for studying postprandial
cases of severe or multiple acute pancreatitis despite diet restriction, lipid metabolism, Eur. J. Clin. Nutr. 61 (2007) 1145–1161.
[8] S. Dash, C. Xiao, C. Morgantini, G.F. Lewis, New insights into the regulation of chy-
under the name of alipogene tiparvovec (Glybera®). It is based on a lomicron production, Annu. Rev. Nutr. 35 (2015) 265–294.
human LPL gene-replacement approach that uses adenovirus as a carri- [9] E. Griffo, L. Di Marino, L. Patti, et al., Test meals rich in marine long-chain n-3 poly-
er of the human gene [165]. Thus, this treatment requires previous ge- unsaturated fatty acids increase postprandial chylomicron response, Nutr. Res. 34
(2014) 661–666.
netic diagnosis that proves the existence of mutations that inactive [10] T. Miyoshi, Y. Noda, Y. Ohno, et al., Omega-3 fatty acids improve postprandial
lipoprotein lipase. lipemia and associated endothelial dysfunction in healthy individuals—a random-
Glybera® is administered once by multiple intramuscular injec- ized cross-over trial, Biomed. Pharmacother. 68 (2014) 1071–1077.
[11] J. Boren, N. Matikainen, M. Adiels, M.R. Taskinen, Postprandial hypertriglyc-
tions, and, although triglyceride levels returned to pre-treatment eridemia as a coronary risk factor, Clin. Chim. Acta 431 (2014) 131–142.
levels within 16–26 weeks after administration, a 6-year follow-up [12] M.M. Hussain, M.H. Kedees, K. Singh, H. Athar, N.Z. Jamali, Signposts in the assem-
demonstrated a clinically relevant reduction in the incidence of doc- bly of chylomicrons, Front. Biosci. 6 (2001) D320–D331.
[13] T. Karupaiah, K. Sundram, Modulation of human postprandial lipemia by changing
umented pancreatitis and acute abdominal pain events consistent
ratios of polyunsaturated to saturated (P/S) fatty acid content of blended dietary
with pancreatitis [166]. fats: a cross-over design with repeated measures, Nutr. J. 12 (2013) 122.
A different approach was followed recently in three patients with li- [14] M.P. Bonham, K.M. Linderborg, A. Dordevic, et al., Lipidomic profiling of chylomi-
cron triacylglycerols in response to high fat meals, Lipids 48 (2013) 39–50.
poprotein lipase deficiency and recurrent pancreatitis who were treated
[15] R.W. Mahley, T.L. Innerarity, S.C. Rall Jr., K.H. Weisgraber, Plasma lipoproteins: apo-
with an inhibitor of APOC3 mRNA (ISIS 304801), which was injected sub- lipoprotein structure and function, J. Lipid Res. 25 (1984) 1277–1294.
cutaneously once weekly for 13 weeks. Serum triglycerides were reduced [16] Y.L. Marcel, T.L. Innerarity, C. Spilman, R.W. Mahley, A.A. Protter, R.W. Milne, Map-
up to 86%, and all patients had triglycerides b500 mg/dL [167]—an ap- ping of human apolipoprotein B antigenic determinants, Arteriosclerosis 7 (1987)
166–175.
proach that could also be of interest in other patients with hypertriglyc- [17] G. Schonfeld, E. Bell, D.H. Alpers, Intestinal apoproteins during fat absorption, J.
eridemia [168]. Clin. Invest. 61 (1978) 1539–1550.
Author's personal copy

146 J. Julve et al. / Clinica Chimica Acta 455 (2016) 134–148

[18] N.A. Abumrad, N.O. Davidson, Role of the gut in lipid homeostasis, Physiol. Rev. 92 [49] J. Heeren, U. Beisiegel, T. Grewal, Apolipoprotein E recycling: implications for dys-
(2012) 1061–1085. lipidemia and atherosclerosis, Arterioscler. Thromb. Vasc. Biol. 26 (2006) 442–448.
[19] A. Jaschke, B. Chung, D. Hesse, et al., The GTPase ARFRP1 controls the lipidation of [50] K.J. Williams, K. Chen, Recent insights into factors affecting remnant lipoprotein
chylomicrons in the Golgi of the intestinal epithelium, Hum. Mol. Genet. 21 (2012) uptake, Curr. Opin. Lipidol. 21 (2010) 218–228.
3128–3142. [51] J.C. Gonzales, P.L. Gordts, E.M. Foley, J.D. Esko, Apolipoproteins E and AV mediate
[20] F. Blanco-Vaca, J.C. Escola-Gil, J.M. Martin-Campos, J. Julve, Role of apoA-II in lipid lipoprotein clearance by hepatic proteoglycans, J. Clin. Invest. 123 (2013)
metabolism and atherosclerosis: advances in the study of an enigmatic protein, J. 2742–2751.
Lipid Res. 42 (2001) 1727–1739. [52] C. Flood, M. Gustafsson, P.E. Richardson, S.C. Harvey, J.P. Segrest, J. Boren, Identifi-
[21] J.M. Martin-Campos, J.C. Escola-Gil, V. Ribas, F. Blanco-Vaca, Apolipoprotein A-II, cation of the proteoglycan binding site in apolipoprotein B48, J. Biol. Chem. 277
genetic variation on chromosome 1q21-q24, and disease susceptibility, Curr. (2002) 32228–32233.
Opin. Lipidol. 15 (2004) 247–253. [53] K.H. Weisgraber, S.C. Rall Jr., Human apolipoprotein B-100 heparin-binding sites, J
[22] W. Weng, J.L. Breslow, Dramatically decreased high density lipoprotein cholesterol, Biol Chem 262 (1987) 11097–11103.
increased remnant clearance, and insulin hypersensitivity in apolipoprotein A-II [54] J.S. Hill, D. Yang, J. Nikazy, L.K. Curtiss, J.T. Sparrow, H. Wong, Subdomain chimeras
knockout mice suggest a complex role for apolipoprotein A-II in atherosclerosis of hepatic lipase and lipoprotein lipase. localization of heparin and cofactor bind-
susceptibility, Proc. Natl. Acad. Sci. U. S. A. 93 (1996) 14788–14794. ing, J. Biol. Chem. 273 (1998) 30979–30984.
[23] E. Boisfer, G. Lambert, V. Atger, et al., Overexpression of human apolipoprotein A-II [55] D. Masuda, K. Hirano, H. Oku, et al., Chylomicron remnants are increased in the
in mice induces hypertriglyceridemia due to defective very low density lipoprotein postprandial state in CD36 deficiency, J. Lipid Res. 50 (2009) 999–1011.
hydrolysis, J. Biol. Chem. 274 (1999) 11564–11572. [56] C.L. Yen, M.L. Cheong, C. Grueter, et al., Deficiency of the intestinal enzyme acyl
[24] S. Dugue-Pujol, X. Rousset, D. Pastier, et al., Human apolipoprotein A-II associates CoA: monoacylglycerol acyltransferase-2 protects mice from metabolic disorders
with triglyceride-rich lipoproteins in plasma and impairs their catabolism, J. Lipid induced by high-fat feeding, Nat. Med. 15 (2009) 442–446.
Res. 47 (2006) 2631–2639. [57] J.T. Haas, H.S. Winter, E. Lim, et al., DGAT1 mutation is linked to a congenital diar-
[25] J. Julve, J.C. Escola-Gil, N. Rotllan, et al., Human apolipoprotein A-II determines plas- rheal disorder, J. Clin. Invest. 122 (2012) 4680–4684.
ma triglycerides by regulating lipoprotein lipase activity and high-density lipopro- [58] J. Iqbal, L.L. Rudel, M.M. Hussain, Microsomal triglyceride transfer protein enhances
tein proteome, Arterioscler. Thromb. Vasc. Biol. 30 (2010) 232–238. cellular cholesteryl esterification by relieving product inhibition, J. Biol. Chem. 283
[26] L.W. Castellani, C.N. Nguyen, S. Charugundla, et al., Apolipoprotein AII is a regulator (2008) 19967–19980.
of very low density lipoprotein metabolism and insulin resistance, J. Biol. Chem. [59] D.M. Mitchell, M. Zhou, R. Pariyarath, et al., Apoprotein B100 has a prolonged inter-
283 (2008) 11633–11644. action with the translocon during which its lipidation and translocation change
[27] X. Deng, R.G. Walker, J. Morris, W.S. Davidson, T.B. Thompson, Role of conserved from dependence on the microsomal triglyceride transfer protein to indepen-
proline residues in human apolipoprotein A-IV structure and function, J. Biol. dence, Proc. Natl. Acad. Sci. U. S. A. 95 (1998) 14733–14738.
Chem. 290 (2015) 10689–10702. [60] B. Antonny, R. Schekman, ER export: public transportation by the COPII coach, Curr.
[28] X. Pan, M.M. Hussain, Gut triglyceride production, Biochim. Biophys. Acta 2012 Opin. Cell Biol. 13 (2001) 438–443.
(1821) 727–735. [61] A. Strong, K. Patel, D.J. Rader, Sortilin and lipoprotein metabolism: making sense
[29] S.K. Karathanasis, I. Yunis, V.I. Zannis, Structure, evolution, and tissue-specific syn- out of complexity, Curr. Opin. Lipidol. 25 (2014) 350–357.
thesis of human apolipoprotein AIV, Biochemistry 25 (1986) 3962–3970. [62] S.K. Nilsson, S. Christensen, M.K. Raarup, R.O. Ryan, M.S. Nielsen, G. Olivecrona, En-
[30] P.H. Green, R.M. Glickman, J.W. Riley, E. Quinet, Human apolipoprotein A-IV. Intes- docytosis of apolipoprotein A-V by members of the low density lipoprotein recep-
tinal origin and distribution in plasma, J. Clin. Invest. 65 (1980) 911–919. tor and the VPS10p domain receptor families, J. Biol. Chem. 283 (2008)
[31] A.B. Kohan, F. Wang, C.M. Lo, M. Liu, P. Tso, ApoA-IV: current and emerging roles in 25920–25927.
intestinal lipid metabolism, glucose homeostasis, and satiety, Am. J. Physiol. [63] A.S. Carlo, C. Gustafsen, G. Mastrobuoni, et al., The pro-neurotrophin receptor
Gastrointest. Liver Physiol. 308 (2015) G472–G481. sortilin is a major neuronal apolipoprotein E receptor for catabolism of amyloid-
[32] F. Wang, A.B. Kohan, C.M. Lo, M. Liu, P. Howles, P. Tso, Apolipoprotein A-IV: a pro- beta peptide in the brain, J. Neurosci. 33 (2013) 358–370.
tein intimately involved in metabolism, J. Lipid Res. 56 (2015) 1403–1418. [64] M. Kjolby, O.M. Andersen, T. Breiderhoff, et al., Sort1, encoded by the cardiovascu-
[33] I.J. Goldberg, C.A. Scheraldi, L.K. Yacoub, U. Saxena, C.L. Bisgaier, Lipoprotein ApoC- lar risk locus 1p13.3, is a regulator of hepatic lipoprotein export, Cell Metab. 12
II activation of lipoprotein lipase. Modulation by apolipoprotein A-IV, J. Biol. Chem. (2010) 213–223.
265 (1990) 4266–4272. [65] A.J. Brahm, R.A. Hegele, Chylomicronaemia—current diagnosis and future thera-
[34] S.K. Nilsson, J. Heeren, G. Olivecrona, M. Merkel, Apolipoprotein A-V; a potent tri- pies, Nat. Rev. Endocrinol. 11 (2015) 352–362.
glyceride reducer, Atherosclerosis 219 (2011) 15–21. [66] T.E. Willnow, Mechanisms of hepatic chylomicron remnant clearance, Diabet. Med.
[35] V. Pruneta-Deloche, G. Ponsin, L. Groisne, J. Fruchart-Najib, M. Lagarde, P. Moulin, 14 (Suppl. 3) (1997) S75–S80.
Postprandial increase of plasma apoAV concentrations in Type 2 diabetic patients, [67] P. Diard, M.I. Malewiak, D. Lagrange, S. Griglio, Hepatic lipase may act as a ligand in
Atherosclerosis 181 (2005) 403–405. the uptake of artificial chylomicron remnant-like particles by isolated rat hepato-
[36] P. Almeda-Valdes, D. Cuevas-Ramos, R. Mehta, et al., Factors associated with post- cytes, Biochem. J. 299 (Pt 3) (1994) 889–894.
prandial lipemia and apolipoprotein A-V levels in individuals with familial com- [68] G.M. Dallinga-Thie, R. Franssen, H.L. Mooij, et al., The metabolism of triglyceride-rich
bined hyperlipidemia, BMC Endocr. Disord. 14 (2014) 90. lipoproteins revisited: new players, new insight, Atherosclerosis 211 (2010) 1–8.
[37] J. Fu, S. Gaetani, F. Oveisi, et al., Oleylethanolamide regulates feeding and body [69] S. Kersten, Physiological regulation of lipoprotein lipase, Biochim. Biophys. Acta
weight through activation of the nuclear receptor PPAR-alpha, Nature 425 2014 (1841) 919–933.
(2003) 90–93. [70] L. Shan, X.C. Yu, Z. Liu, et al., The angiopoietin-like proteins ANGPTL3 and ANGPTL4
[38] N. Vu-Dac, P. Gervois, H. Jakel, et al., Apolipoprotein A5, a crucial determinant of inhibit lipoprotein lipase activity through distinct mechanisms, J. Biol. Chem. 284
plasma triglyceride levels, is highly responsive to peroxisome proliferator- (2009) 1419–1424.
activated receptor alpha activators, J. Biol. Chem. 278 (2003) 17982–17985. [71] J. Kobayashi, K. Miyashita, K. Nakajima, H. Mabuchi, Hepatic lipase: a comprehen-
[39] J. Fu, G. Astarita, S. Gaetani, et al., Food intake regulates oleoylethanolamide forma- sive view of its role on plasma lipid and lipoprotein metabolism, J. Atheroscler.
tion and degradation in the proximal small intestine, J. Biol. Chem. 282 (2007) Thromb. 22 (2015) 1001–1011.
1518–1528. [72] W.K. Sonnenburg, D. Yu, E.C. Lee, et al., GPIHBP1 stabilizes lipoprotein lipase and
[40] L.S. Zhang, M. Xu, Q. Yang, R.O. Ryan, P. Howles, P. Tso, Apolipoprotein A-V defi- prevents its inhibition by angiopoietin-like 3 and angiopoietin-like 4, J. Lipid Res.
ciency enhances chylomicron production in lymph fistula mice, Am. J. Physiol. 50 (2009) 2421–2429.
Gastrointest. Liver Physiol. 308 (2015) G634–G642. [73] A.P. Lillis, L.B. Van Duyn, J.E. Murphy-Ullrich, D.K. Strickland, LDL receptor-related
[41] L.S. Zhang, H. Sato, Q. Yang, et al., Apolipoprotein A-V is present in bile and its se- protein 1: unique tissue-specific functions revealed by selective gene knockout
cretion increases with lipid absortion in Sprague–Dawley rats, Am. J. Physiol. studies, Physiol. Rev. 88 (2008) 887–918.
Gastrointest. Liver Physiol. 309 (2015) G918–G925. [74] K.I. Stanford, J.R. Bishop, E.M. Foley, et al., Syndecan-1 is the primary heparan sul-
[42] M.C. Jong, M.H. Hofker, L.M. Havekes, Role of ApoCs in lipoprotein metabolism: fate proteoglycan mediating hepatic clearance of triglyceride-rich lipoproteins in
functional differences between ApoC1, ApoC2, and ApoC3, Arterioscler. Thromb. mice, J. Clin. Invest. 119 (2009) 3236–3245.
Vasc. Biol. 19 (1999) 472–484. [75] E. Mendoza-Barbera, J. Julve, S.K. Nilsson, et al., Structural and functional analysis of
[43] E.J. Schaefer, S. Eisenberg, R.I. Levy, Lipoprotein apoprotein metabolism, J. Lipid Res. APOA5 mutations identified in patients with severe hypertriglyceridemia, J. Lipid
19 (1978) 667–687. Res. 54 (2013) 649–661.
[44] E. Windler, R.J. Havel, Inhibitory effects of C apolipoproteins from rats and humans [76] D.A. Leaf, Chylomicronemia and the chylomicronemia syndrome. A practical guide
on the uptake of triglyceride-rich lipoproteins and their remnants by the perfused to treatment, Am. J. Med. 121 (2008) 10–12.
rat liver, J. Lipid Res. 26 (1985) 556–565. [77] D. Gaudet, J. Signorovitch, E. Swallow, et al., Medical resource use and cost associ-
[45] M. Larsson, E. Vorrsjo, P. Talmud, A. Lookene, G. Olivecrona, Apolipoproteins C-I ated with chylomicronemia, J. Med. Econ. 16 (2013) 657–666.
and C-III inhibit lipoprotein lipase activity by displacement of the enzyme from [78] R.J. Havel, Pathogenesis, differentiation and management of hypertriglyceridemia,
lipid droplets, J. Biol. Chem. 288 (2013) 33997–34008. Adv. Intern. Med. 15 (1969) 117–154.
[46] A.A. Kei, T.D. Filippatos, V. Tsimihodimos, M.S. Elisaf, A review of the role of apoli- [79] P. Valdivielso, A. Ramirez-Bueno, N. Ewald, Current knowledge of
poprotein C-II in lipoprotein metabolism and cardiovascular disease, Metabolism hypertriglyceridemic pancreatitis, Eur. J. Intern. Med. 25 (2014) 689–694.
61 (2012) 906–921. [80] B.G. Nordestgaard, M. Benn, P. Schnohr, A. Tybjaerg-Hansen, Nonfasting triglycer-
[47] J.C. LaRosa, R.I. Levy, P. Herbert, S.E. Lux, D.S. Fredrickson, A specific apoprotein ides and risk of myocardial infarction, ischemic heart disease, and death in men
activator for lipoprotein lipase, Biochem. Biophys. Res. Commun. 41 (1970) and women, JAMA 298 (2007) 299–308.
57–62. [81] S. Bansal, J.E. Buring, N. Rifai, S. Mora, F.M. Sacks, P.M. Ridker, Fasting compared
[48] G.D. Norata, S. Tsimikas, A. Pirillo, A.L. Catapano, Apolipoprotein C-III: from patho- with nonfasting triglycerides and risk of cardiovascular events in women, JAMA
physiology to pharmacology, Trends Pharmacol. Sci. 36 (2015) 675–687. 298 (2007) 309–316.
Author's personal copy

J. Julve et al. / Clinica Chimica Acta 455 (2016) 134–148 147

[82] X.Y. Zheng, L. Liu, Remnant-like lipoprotein particles impair endothelial function: [112] K. Musunuru, J.P. Pirruccello, R. Do, et al., Exome sequencing, ANGPTL3 mutations,
direct and indirect effects on nitric oxide synthase, J. Lipid Res. 48 (2007) and familial combined hypolipidemia, N. Engl. J. Med. 363 (2010) 2220–2227.
1673–1680. [113] J.M. Martin-Campos, R. Roig, C. Mayoral, et al., Identification of a novel mutation in
[83] J.D. Brunzell, W.R. Hazzard, D. Porte Jr., E.L. Bierman, Evidence for a common, sat- the ANGPTL3 gene in two families diagnosed of familial hypobetalipoproteinemia
urable, triglyceride removal mechanism for chylomicrons and very low density li- without APOB mutation, Clin. Chim. Acta 413 (2012) 552–555.
poproteins in man, J. Clin. Invest. 52 (1973) 1578–1585. [114] L. Pisciotta, E. Favari, L. Magnolo, et al., Characterization of three kindreds with fa-
[84] M. Adiels, N. Matikainen, J. Westerbacka, et al., Postprandial accumulation of chy- milial combined hypolipidemia caused by loss-of-function mutations of ANGPTL3,
lomicrons and chylomicron remnants is determined by the clearance capacity, Circ. Cardiovasc. Genet. 5 (2012) 42–50.
Atherosclerosis 222 (2012) 222–228. [115] I. Minicocci, A. Montali, M.R. Robciuc, et al., Mutations in the ANGPTL3 gene and fa-
[85] J.D. Brunzell, E.L. Bierman, Chylomicronemia syndrome. Interaction of genetic and milial combined hypolipidemia: a clinical and biochemical characterization, J. Clin.
acquired hypertriglyceridemia, Med. Clin. North Am. 66 (1982) 455–468. Endocrinol. Metab. 97 (2012) E1266–E1275.
[86] J.D. Brunzell, Familial lipoprotein lipase deficiency and other causes of the [116] T. Shimizugawa, M. Ono, M. Shimamura, et al., ANGPTL3 decreases very low den-
chylomicronemia syndrome, in: C.R. Scriver, A.L. Beaudet, W.S. Sly, D. Valle sity lipoprotein triglyceride clearance by inhibition of lipoprotein lipase, J. Biol.
(Eds.), The Metabolic and Molecular Basis of Inherited Disease, 7 ed.McGraw- Chem. 277 (2002) 33742–33748.
Hill, New York 1995, pp. 1913–1932. [117] S. Romeo, W. Yin, J. Kozlitina, et al., Rare loss-of-function mutations in ANGPTL
[87] D.S. Fredrickson, An international classification of hyperlipidemias and family members contribute to plasma triglyceride levels in humans, J. Clin. Invest.
hyperlipoproteinemias, Ann. Intern. Med. 75 (1971) 471–472. 119 (2009) 70–79.
[88] R.A. Hegele, Plasma lipoproteins: genetic influences and clinical implications, Nat. [118] K. Nakajima, J. Kobayashi, H. Mabuchi, et al., Association of angiopoietin-like pro-
Rev. Genet. 10 (2009) 109–121. tein 3 with hepatic triglyceride lipase and lipoprotein lipase activities in human
[89] M.R. Hayden, Y. Ma, Molecular genetics of human lipoprotein lipase deficiency, plasma, Ann. Clin. Biochem. 47 (2010) 423–431.
Mol. Cell. Biochem. 113 (1992) 171–176. [119] M. Shimamura, M. Matsuda, H. Yasumo, et al., Angiopoietin-like protein3 regulates
[90] J.D. Brunzell, N.E. Miller, P. Alaupovic, et al., Familial chylomicronemia due to a cir- plasma HDL cholesterol through suppression of endothelial lipase, Arterioscler.
culating inhibitor of lipoprotein lipase activity, J. Lipid Res. 24 (1983) 12–19. Thromb. Vasc. Biol. 27 (2007) 366–372.
[91] J.M. Martin-Campos, J. Julve, R. Roig, et al., Molecular analysis of chylomicronemia [120] H.D. Moon, K. Nakajima, K. Kamiyama, K. Takanashi, I. Sakurabayashi, T. Nagamine,
in a clinical laboratory setting: diagnosis of 13 cases of lipoprotein lipase deficien- Higher frequency of abnormal serum angiopoietin-like protein 3 than abnormal
cy, Clin. Chim. Acta 429 (2014) 61–68. cholesteryl ester transfer protein in Japanese hyperalphalipoproteinemic subjects,
[92] C. Rabacchi, L. Pisciotta, A.B. Cefalu, et al., Spectrum of mutations of the LPL gene Clin. Chim. Acta 398 (2008) 99–104.
identified in Italy in patients with severe hypertriglyceridemia, Atherosclerosis [121] M.R. Robciuc, M. Maranghi, A. Lahikainen, et al., Angptl3 deficiency is associated
241 (2015) 79–86. with increased insulin sensitivity, lipoprotein lipase activity, and decreased
[93] A.R. Rahalkar, F. Giffen, B. Har, et al., Novel LPL mutations associated with lipopro- serum free fatty acids, Arterioscler. Thromb. Vasc. Biol. 33 (2013) 1706–1713.
tein lipase deficiency: two case reports and a literature review, Can. J. Physiol. [122] A.S. Lindman, M.B. Veierod, A. Tverdal, J.I. Pedersen, R. Selmer, Nonfasting triglyc-
Pharmacol. 87 (2009) 151–160. erides and risk of cardiovascular death in men and women from the Norwegian
[94] J. Wang, H. Cao, M.R. Ban, et al., Resequencing genomic DNA of patients with severe Counties Study, Eur. J. Epidemiol. 25 (2010) 789–798.
hypertriglyceridemia (MIM 144650), Arterioscler. Thromb. Vasc. Biol. 27 (2007) [123] C. Imke, B.L. Rodriguez, J.S. Grove, et al., Are remnant-like particles independent
2450–2455. predictors of coronary heart disease incidence? The Honolulu Heart study,
[95] R.P. Surendran, M.E. Visser, S. Heemelaar, et al., Mutations in LPL, APOC2, APOA5, Arterioscler. Thromb. Vasc. Biol. 25 (2005) 1718–1722.
GPIHBP1 and LMF1 in patients with severe hypertriglyceridaemia, J. Intern. Med. [124] A. Varbo, M. Benn, A. Tybjaerg-Hansen, A.B. Jorgensen, R. Frikke-Schmidt, B.G.
272 (2012) 185–196. Nordestgaard, Remnant cholesterol as a causal risk factor for ischemic heart dis-
[96] M.J. Ariza, M.A. Sanchez-Chaparro, F.J. Baron, et al., Additive effects of LPL, APOA5 ease, J. Am. Coll. Cardiol. 61 (2013) 427–436.
and APOE variant combinations on triglyceride levels and hypertriglyceridemia: [125] B.O. Schneeman, L. Kotite, K.M. Todd, R.J. Havel, Relationships between the re-
results of the ICARIA genetic sub-study, BMC Med. Genet. 11 (2010) 66. sponses of triglyceride-rich lipoproteins in blood plasma containing apolipopro-
[97] C.F. Semenkovich, S.H. Chen, M. Wims, C.C. Luo, W.H. Li, L. Chan, Lipoprotein lipase teins B-48 and B-100 to a fat-containing meal in normolipidemic humans, Proc.
and hepatic lipase mRNA tissue specific expression, developmental regulation, and Natl. Acad. Sci. U. S. A. 90 (1993) 2069–2073.
evolution, J. Lipid Res. 30 (1989) 423–431. [126] J.S. Cohn, E.J. Johnson, J.S. Millar, et al., Contribution of apoB-48 and apoB-100
[98] C.T. Johansen, J. Wang, A.D. McIntyre, et al., Excess of rare variants in non-genome- triglyceride-rich lipoproteins (TRL) to postprandial increases in the plasma con-
wide association study candidate genes in patients with hypertriglyceridemia, Circ. centration of TRL triglycerides and retinyl esters, J. Lipid Res. 34 (1993)
Cardiovasc. Genet. 5 (2012) 66–72. 2033–2040.
[99] G.V. Kryukov, L.A. Pennacchio, S.R. Sunyaev, Most rare missense alleles are delete- [127] F. Karpe, M. Bell, J. Bjorkegren, A. Hamsten, Quantification of postprandial
rious in humans: implications for complex disease and association studies, Am. J. triglyceride-rich lipoproteins in healthy men by retinyl ester labeling and simulta-
Hum. Genet. 80 (2007) 727–739. neous measurement of apolipoproteins B-48 and B-100, Arterioscler. Thromb.
[100] C. Desmarchelier, J.C. Martin, R. Planells, et al., The postprandial chylomicron triac- Vasc. Biol. 15 (1995) 199–207.
ylglycerol response to dietary fat in healthy male adults is significantly explained [128] S.D. Proctor, J.C. Mamo, Intimal retention of cholesterol derived from apolipopro-
by a combination of single nucleotide polymorphisms in genes involved in triacyl- tein B100- and apolipoprotein B48-containing lipoproteins in carotid arteries of
glycerol metabolism, J. Clin. Endocrinol. Metab. 99 (2014) E484–E488. Watanabe heritable hyperlipidemic rabbits, Arterioscler. Thromb. Vasc. Biol. 23
[101] L. Berglund, J.D. Brunzell, A.C. Goldberg, et al., Evaluation and treatment of (2003) 1595–1600.
hypertriglyceridemia: an Endocrine Society clinical practice guideline, J. Clin. [129] D. Masuda, T. Sugimoto, K. Tsujii, et al., Correlation of fasting serum apolipoprotein
Endocrinol. Metab. 97 (2012) 2969–2989. B-48 with coronary artery disease prevalence, Eur. J. Clin. Investig. 42 (2012)
[102] J.M. Martin-Campos, N. Rico, R. Bonet, C. Mayoral, J. Ordonez-Llanos, F. Blanco- 992–999.
Vaca, Apolipoprotein A5 S19W may play a role in dysbetalipoproteinemia in pa- [130] K. Mori, T. Ishida, T. Yasuda, et al., Fasting serum concentration of apolipoprotein
tients with the Apo E2/E2 genotype, Clin. Chem. 52 (2006) 1974–1975. B48 represents residual risks in patients with new-onset and chronic coronary ar-
[103] M.S. Weintraub, S. Eisenberg, J.L. Breslow, Dietary fat clearance in normal subjects tery disease, Clin. Chim. Acta 421 (2013) 51–56.
is regulated by genetic variation in apolipoprotein E, J. Clin. Invest. 80 (1987) [131] K. Nakatani, T. Sugimoto, D. Masuda, et al., Serum apolipoprotein B-48 levels are
1571–1577. correlated with carotid intima-media thickness in subjects with normal serum tri-
[104] C.E. Rodrigues, E. Bonfa, J.F. Carvalho, Review on anti-lipoprotein lipase antibodies, glyceride levels, Atherosclerosis 218 (2011) 226–232.
Clin. Chim. Acta 411 (2010) 1603–1605. [132] J.C. Hogue, B. Lamarche, A.J. Tremblay, J. Bergeron, C. Gagne, P. Couture, Evidence of
[105] M. Moret, V. Pruneta-Deloche, A. Sassolas, C. Marcais, P. Moulin, Prevalence and func- increased secretion of apolipoprotein B-48-containing lipoproteins in subjects
tion of anti-lipoprotein lipase auto-antibodies in type V hyperchylomicronemia, Ath- with type 2 diabetes, J. Lipid Res. 48 (2007) 1336–1342.
erosclerosis 208 (2010) 324–327. [133] R. Mangat, J.W. Su, J.E. Lambert, et al., Increased risk of cardiovascular disease in
[106] J.F. de Carvalho, V.S. Viana, E.F. Neto, R.D. Santos, E. Bonfa, Anti-lipoprotein lipase Type 1 diabetes: arterial exposure to remnant lipoproteins leads to enhanced de-
antibodies in patients with hypertriglyceridemia without associated autoimmune position of cholesterol and binding to glycated extracellular matrix proteoglycans,
disease, Isr. Med. Assoc. J. 13 (2011) 350–353. Diabet. Med. 28 (2011) 61–72.
[107] V. Pruneta, P. Moulin, F. Labrousse, P.J. Bondon, G. Ponsin, F. Berthezene, Char- [134] P. Valdivielso, S. Puerta, J. Rioja, et al., Postprandial apolipoprotein B48 is associated
acterization of a new case of autoimmune type I hyperlipidemia: long-term re- with asymptomatic peripheral arterial disease: a study in patients with type 2 di-
mission under immunosuppressive therapy, J. Clin. Endocrinol. Metab. 82 abetes and controls, Clin. Chim. Acta 411 (2010) 433–437.
(1997) 791–796. [135] E. Lapice, P. Cipriano, L. Patti, G. Romano, O. Vaccaro, A.A. Rivellese, Fasting apolipo-
[108] B. Jones, E.L. Jones, S.A. Bonney, et al., Mutations in a Sar1 GTPase of COPII vesicles protein B48 is associated with asymptomatic peripheral arterial disease in type 2
are associated with lipid absorption disorders, Nat. Genet. 34 (2003) 29–31. diabetic subjects: a case–control study, Atherosclerosis 223 (2012) 504–506.
[109] J.R. Wetterau, L.P. Aggerbeck, M.E. Bouma, et al., Absence of microsomal triglycer- [136] N. Mero, R. Malmstrom, G. Steiner, M.R. Taskinen, M. Syvanne, Postprandial metab-
ide transfer protein in individuals with abetalipoproteinemia, Science 258 (1992) olism of apolipoprotein B-48- and B-100-containing particles in type 2 diabetes
999–1001. mellitus: relations to angiographically verified severity of coronary artery disease,
[110] J. Lee, R.A. Hegele, Abetalipoproteinemia and homozygous hypobetalipoproteinemia: Atherosclerosis 150 (2000) 167–177.
a framework for diagnosis and management, J. Inherit. Metab. Dis. 37 (2014) [137] C. Phillips, G. Murugasu, D. Owens, P. Collins, A. Johnson, G.H. Tomkin, Improved
333–339. metabolic control reduces the number of postprandial apolipoprotein B-48-
[111] A.J. Hooper, K. Robertson, P.H. Barrett, K.G. Parhofer, F.M. van Bockxmeer, J.R. containing particles in type 2 diabetes, Atherosclerosis 148 (2000) 283–291.
Burnett, Postprandial lipoprotein metabolism in familial hypobetalipoproteinemia, [138] B.H. Chung, G. Tallis, V. Yalamoori, G.M. Anantharamaiah, J.P. Segrest, Liposome-
J. Clin. Endocrinol. Metab. 92 (2007) 1474–1478. like particles isolated from human atherosclerotic plaques are structurally and
Author's personal copy

148 J. Julve et al. / Clinica Chimica Acta 455 (2016) 134–148

compositionally similar to surface remnants of triglyceride-rich lipoproteins, [153] T. Vilsbøll, T. Krarup, C.F. Deacon, S. Madsbad, J.J. Holst, Reduced postprandial con-
Arterioscler. Thromb. 14 (1994) 622–635. centrations of intact biologically active glucagon-like peptide 1 in type 2 diabetic
[139] S. Pal, K. Semorine, G.F. Watts, J. Mamo, Identification of lipoproteins of intestinal patients, Diabetes 50 (2001) 609–613.
origin in human atherosclerotic plaque, Clin. Chem. Lab. Med. 41 (2003) 792–795. [154] C. Xiao, R.H. Bandsma, S. Dash, L. Szeto, G.F. Lewis, Exenatide, a glucagon-like
[140] T. Nakano, K. Nakajima, M. Niimi, et al., Detection of apolipoproteins B-48 and B- peptide-1 receptor agonist, acutely inhibits intestinal lipoprotein production in
100 carrying particles in lipoprotein fractions extracted from human aortic athero- healthy humans, Arterioscler. Thromb. Vasc. Biol. 32 (2012) 1513–1519.
sclerotic plaques in sudden cardiac death cases, Clin. Chim. Acta 390 (2008) 38–43. [155] E.A. Schwartz, J. Koska, M.P. Mullin, I. Syoufi, D.C. Schwenke, P.D. Reaven, Exenatide
[141] C.H. Floren, A. Chait, Uptake of chylomicron remnants by the native LDL receptor in suppresses postprandial elevations in lipids and lipoproteins in individuals with
human monocyte-derived macrophages, Biochim. Biophys. Acta 665 (1981) impaired glucose tolerance and recent onset type 2 diabetes mellitus, Atheroscle-
608–611. rosis 212 (2010) 217–222.
[142] C.L. Elsegood, S. Pal, P.D. Roach, J.C. Mamo, Binding and uptake of chylomicron [156] J. Altomonte, L. Cong, S. Harbaran, et al., Foxo1 mediates insulin action on apoC-III
remnants by primary and THP-1 human monocyte-derived macrophages: deter- and triglyceride metabolism, J. Clin. Invest. 114 (2004) 1493–1503.
mination of binding proteins, Clin. Sci. (Lond.) 101 (2001) 111–119. [157] S. Farr, C. Baker, M. Naples, et al., Central Nervous system regulation of intestinal lipo-
[143] C.L. Elsegood, J.C. Mamo, An investigation by electron microscopy of chylomicron protein metabolism by glucagon-like peptide-1 via a brain-gut axis, Arterioscler.
remnant uptake by human monocyte-derived macrophages, Atherosclerosis 188 Thromb. Vasc. Biol. 35 (2015) 1092–1100.
(2006) 251–259. [158] K. Nakajima, T. Nakano, Y. Tokita, et al., Postprendial lipoprotein metabolism: VLDL
[144] K.M. Botham, C.P. Wheeler-Jones, Postprandial lipoproteins and the molecular reg- vs chylomicrons, Clin. Chim. Acta 412 (2011) 1306–1318.
ulation of vascular homeostasis, Prog. Lipid Res. 52 (2013) 446–464. [159] K. Nakajima, T. Saito, A. Tamura, et al., Cholesterol in remnant-like lipoproteins in
[145] K. Domoto, T. Taniguchi, H. Takaishi, et al., Chylomicron remnants induce mono- human serum using monoclonal anti apo B-100 and anti apo A-I immunoaffinity
cyte chemoattractant protein-1 expression via p38 MAPK activation in vascular mixed gels, Clin. Chim. Acta 223 (1993) 53–71.
smooth muscle cells, Atherosclerosis 171 (2003) 193–200. [160] K. Miyauchi, N. Kayahara, M. Ishigami, et al., Development of a homogeneous assay
[146] Y. Takahashi, Y. Fujioka, T. Takahashi, et al., Chylomicron remnants regulate early to measure remnant lipoprotein cholesterol, Clin. Chem. 53 (2007) 2128–2135.
growth response factor-1 in vascular smooth muscle cells, Life Sci. 77 (2005) [161] T. Nakamura, J.E. Obata, M. Hirano, et al., Predictive value of remnant lipoprotein
670–682. for cardiovascular events in patients with coronary artery disease after achieve-
[147] H. Duez, B. Lamarche, K.D. Uffelman, R. Valero, J.S. Cohn, G.F. Lewis, Hyperinsulinemia ment of LDL-cholesterol goals, Atherosclerosis 218 (2011) 163–167.
is associated with increased production rate of intestinal apolipoprotein B-48- [162] M.L. Phillips, C. Pullinger, I. Kroes, et al., A single copy of apolipoprotein B-48 is
containing lipoproteins in humans, Arterioscler. Thromb. Vasc. Biol. 26 (2006) present on the human chylomicron remnant, J. Lipid Res. 38 (1997) 1170–1177.
1357–1363. [163] J.D. Brunzell, Familial Lipoprotein Lipase, in: R.A. Pagon, M.P. Adam, H.H. Ardinger,
[148] S. Lally, C.Y. Tan, D. Owens, G.H. Tomkin, Messenger RNA levels of genes involved et al., (Eds.), University of Washinton, Seattle (WA), 1999.
in dysregulation of postprandial lipoproteins in type 2 diabetes: the role of [164] G.C. Shearer, O.V. Savinova, W.S. Harris, Fish-oil—how does it reduce plasma
Niemann–Pick C1-like 1, ATP-binding cassette, transporters G5 and G8, and of mi- triglicerides? Biochim. Biophys. Acta 182 (2012) 843–851.
crosomal triglyceride transfer protein, Diabetologia 49 (2006) 1008–1016. [165] V. Ferreira, J. Twisk, K. Kwikkers, et al., Immune responses to intramuscular admin-
[149] L. Bozzetto, G. Annuzzi, G.D. Corte, et al., Ezetimibe beneficially influences fasting istration of alipogene tiparvovec (AAV1-LPL(S447X)) in a phase II clinical trial of
and postprandial triglyceride-rich lipoproteins in type 2 diabetes, Atherosclerosis lipoprotein lipase deficiency gene therapy, Hum. Gene Ther. 25 (2014) 180–188.
2011 (2017) 142–148. [166] L.J. Scott, Alipogene tiparvovec: a review of its use in adults with familial lipopro-
[150] E. Levy, S. Spahis, C. Garofalo, et al., Sar1b transgenic male mice are more suscepti- tein lipase deficiency, Drugs 75 (2015) 175–182.
ble to high-fat diet-induced obesity, insulin insensitivity and intestinal chylomi- [167] D. Gaudet, D. Brisson, K. Tremblay, et al., Targeting APOC3 in the familial
cron overproduction, J. Nutr. Biochem. 25 (2014) 540–548. chylomicronemic syndrome, N. Engl. J. Med. 371 (2014) 2200–2206.
[151] S. Farr, K. Adeli, Incretin-based therapies for treatment of postprandial dyslipid- [168] D. Gaudet, V.J. Alexander, B.F. Baker, et al., Antisense inhibition of apolipoprotein C-
emia in insulin-resistant states, Curr. Opin. Lipidol. 23 (2012) 56–61. III in patients with hypertriglyceridemia, N. Engl. J. Med. 373 (2015) 438–447.
[152] J.J. Meier, A. Gethmann, O. Götze, et al., Glucagon-like peptide 1 abolishes the post-
prandial rise in triglyceride concentrations and lowers levels of non-esterified fatty
acids in humans, Diabetologia 49 (2006) 452–458.

View publication stats

You might also like