UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A...

182
UvA-DARE is a service provided by the library of the University of Amsterdam (http://dare.uva.nl) UvA-DARE (Digital Academic Repository) Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication Citation for published version (APA): Schooneman, M. G. (2015). Here, there and everywhere. A multi organ approach to acylcarnitine metabolism. General rights It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons). Disclaimer/Complaints regulations If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible. Download date: 02 Jul 2020

Transcript of UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A...

Page 1: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

UvA-DARE is a service provided by the library of the University of Amsterdam (http://dare.uva.nl)

UvA-DARE (Digital Academic Repository)

Here, there and everywhere. A multi organ approach to acylcarnitine metabolism

Schooneman, M.G.

Link to publication

Citation for published version (APA):Schooneman, M. G. (2015). Here, there and everywhere. A multi organ approach to acylcarnitine metabolism.

General rightsIt is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s),other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulationsIf you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, statingyour reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Askthe Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam,The Netherlands. You will be contacted as soon as possible.

Download date: 02 Jul 2020

Page 2: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

Marieke G. Schooneman

A multi organ approach to acylcarnitine metabolism.

HERETHERE EVERY

WHERE&

HERE, TH

ERE & EV

ERYW

HERE

Ma

rieke

G. Sc

hoo

nem

an

Page 3: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

A multi organ approach to acylcarnitine metabolism.

HERETHERE EVERY

WHERE&

Marieke G. Schooneman

K007_000000_HereThere and EveryWhere boekje.indd 1 09-02-15 08:30

Page 4: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

COLOFON

Here, there and everywhere. A multi organ approach to acylcarnitine metabolism.

Dissertation, University of Amsterdam, Amsterdam, the Netherlands

ISBN 978-90-9028866-6

Author Marieke G. Schooneman

Art direction Krista Rozema

Vormgeving Karin Jansen

Photography Chantal Spieard

Printing Drukkerij Aeroprint, Ouderkerk aan de Amstel

Copyright © 2015 Marieke G. Schooneman, Amsterdam, the Netherlands.

All rights reserved.

No part of this publication may be reproduced or transmitted in any form by any

means, without written permission of the author.

This dissertation was funded by the Gaucher Stichting, Fresenius-Kabi, Ipsen,

Astra Zeneca, Sanofi, Boehringer-Ingelheim, Goodlife Pharma and the University

of Amsterdam.

K007_000000_HereThere and EveryWhere boekje.indd 2 09-02-15 08:30

Page 5: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

Here, there and everywhere. A multi organ approach to acylcarnitine metabolism.

ACADEMISCH PROEFSCHRIFT

ter verkrijging van de graad van doctor

aan de Universiteit van Amsterdam

op gezag van de Rector Magnificus

Prof. Dr. D.C. van den Boom

ten overstaan van een door het College voor Promoties ingestelde

commissie, in het openbaar te verdedigen in de Agnietenkapel

op donderdag 2 april 2015, te 12:00 uur

door Marieke Guurtje Schooneman

geboren te Purmerend

K007_000000_HereThere and EveryWhere boekje.indd 1 09-02-15 08:30

Page 6: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

K007_000000_HereThere and EveryWhere boekje.indd 2 09-02-15 08:30

Page 7: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

Promotiecommissie

Promotores Prof. Dr. C.E.M. Hollak

Prof. Dr. R.J.A. Wanders

Copromotores Dr. M.R. Soeters

Dr. S.M. Houten

Overige leden Prof. Dr. J.A. Romijn

Prof. Dr. A.J. Verhoeven

Prof. Dr. C.J.M. de Vries

Prof. Dr. P. Schrauwen

Prof. Dr. A.K. Groen

Dr. M.J.M. Serlie

Faculteit der Geneeskunde

K007_000000_HereThere and EveryWhere boekje.indd 3 09-02-15 08:30

Page 8: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

K007_000000_HereThere and EveryWhere boekje.indd 4 09-02-15 08:30

Page 9: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

5

Table of contents

1. A short introduction 7

2. Acylcarnitines: reflecting or inflicting insulin resistance? 13

3. Plasma acylcarnitines inadequately reflect tissue acylcarnitine metabolism 31

4. Transorgan fluxes in a porcine model reveal a central role for liver 57 in acylcarnitine metabolism

5. Acylcarnitine kinetics in a fasting- and obesity-induced insulin 77 resistant mouse model

6. Assessment of plasma acylcarnitines before and after 93 weight loss in obese subjects

7. The impact of altered carnitine availability on fatty acid and glucose 113 metabolism in a diet-induced obesity mouse model

8. A multi organ perspective on acylcarnitines in various metabolic conditions 135

9. Appendix 149 Summary 151Nederlandse samenvatting 155PhD Portfolio 158Dankwoord 161Biografie 165

K007_000000_HereThere and EveryWhere boekje.indd 5 09-02-15 08:30

Page 10: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

A Short Introduction

C 1

K007_000000_HereThere and EveryWhere boekje.indd 6 09-02-15 08:30

Page 11: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

A Short Introduction

C 1

K007_000000_HereThere and EveryWhere boekje.indd 7 09-02-15 08:30

Page 12: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

K007_000000_HereThere and EveryWhere boekje.indd 8 09-02-15 08:30

Page 13: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

9A SHORT INTRODUCTION

We eat, therefore we are. The human body obtains energy via the breakdown of macro– nutrients, carbohydrates, lipids and proteins. The resulting sugars, fatty acids and amino acids can be oxidized via several metabolic pathways under different metabolic circumstances. In a postprandial state, we mainly oxidize carbohydrates because these macronutrients are predominant in most diets. But when carbohydrate oxidation (CHO) rates fall, either because most glucose is oxidized, or because glucose reserves are not replenished, we switch to lipids as main oxidative substrate (1). And in these days, a sub-stantial percentage of all human beings carry along large reserves of lipids. Lipids are stored mainly as triglycerides, and when needed for oxidation they are hydro-lyzed into glycerol and free fatty acids (FFA). FFAs are converted into acyl-CoAs, which are oxidized inside the mitochondrion via beta-oxidation. Because the mitochondrial membrane is impermeable to acyl-CoAs, carnitine is essential for the transport of acyl-CoAs into mitochondria (2, 3). Here, acyl-CoAs are transesterified to carnitine by the enzyme carnitine palmitoyltransferase-1 (CPT1) on the outer leaflet of the mitochondrial membrane. The resulting acylcarnitine is then shuttled over the membrane by carni-tine aylcarnitine translocase (CACT) towards the inner leaflet of the mitochondrial mem-brane, where CPT2 can release the acyl-CoA for further beta-oxidation (4). Due to its mitochondrial function, carnitine and acylcarnitines mainly reside within tissues (5). But as they can cross the cell membrane, they can be found in the plasma compartment as well. Here they form a characteristic acylcarnitine profile, which is considered to reflect the intracellular acyl-CoA pool. This profile has been studied extensively in the last few decades in relation to inherited metabolic diseases (6) and more recently for their pos-sible involvement in diet-induced metabolic derangements and insulin resistance (7-10). As stated earlier, FAO rates increase when CHO rates decline, for example during fasting. Therefore, acylcarnitine levels, being FAO-intermediates, increase in a fasted state (5, 11, 12). This shift from CHO towards FAO is accompanied by a physiological resistance to the insulin signal, mainly in skeletal muscle tissue, as the remaining glucose in plasma needs to be spared to fuel the central nervous system. Apart from fasting-induced in-sulin resistance, also obesity-associated insulin resistance is accompanied by a rise in acylcarnitine levels. Under circumstances of overfeeding and obesity, insulin resistance occurs as well and FAO rates remain high, even under fed conditions where glucose is readily available. This leads to high lipid levels in insulin sensitive tissues, which could potentially interfere directly with insulin signalling inside the cytosol, a theory referred to as lipotoxicity. Several lipid intermediates accumulate and impair insulin sensitivity, such as ceramides, gangliosides and diacylglycerol (7, 13-15). In addition, acylcarnitines accumulate due to high or incomplete FAO (8-10).Several studies have shown associations between acylcarnitines and insulin resistance. Here, insulin resistant states such as (prolonged) fasting and diet-induced obesity and type 2 diabetes mellitus were accompanied by elevated plasma acylcarnitines (9, 11, 12). Additionally several individual species showed correlations with markers of insulin re-sistance and glucose tolerance in both rodents and humans (99, 16-19). An important limitation of the proposed association between acylcarnitines and insulin resistance is that acylcarnitine profiles are often measured in plasma. Since acylcarnitine metabolism

K007_000000_HereThere and EveryWhere boekje.indd 9 09-02-15 08:30

Page 14: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

10 A SHORT INTRODUCTION

and FAO are cellular processes in insulin sensitive tissues, it remains to be determined what plasma acylcarnitines actually reflect. Moreover, much knowledge on acylcarnitine kinetics is lacking. Therefore a deeper understanding of acylcarnitine metabolism is cru-cial in the interpretation of the proposed associations with FAO derangements and in-sulin resistance.This thesis aims to elucidate the kinetic properties of acylcarnitines of different chain lengths in the plasma compartment. Furthermore we studied the interaction of acyl-carnitine metabolism between plasma and different insulin sensitive tissues. In all our studies we compared acylcarnitine metabolism in fasted, fed and a HFD-induced, insulin resistant state.

Thesis outlineChapter 2 is an introductory review on the existing literature on acylcarnitine metabolism in relation to insulin resistance. We included in vitro, animal and human studies. After this inventory of associations between acylcarnitines and insulin resistance, we tried to sub-sequently clarify what the alterations in acylcarnitine metabolism actually reflect. In order to study the relation between the plasma acylcarnitine profile and acylcarnitine metabolism on tissue level, chapter 3 focussed on correlations between the profiles in the different compartments in fed and fasted mice. Here we expected to find a relation between plasma acylcarnitines and muscle or liver acylcarnitines, as they are suggested to play an important role in acylcarnitine metabolism.Following chapter 3, the aim of chapter 4 was to determine the role of different organs in acylcarnitine metabolism. Therefore we measured fasted and postprandial trans organ fluxes of acylcarnitines in a catheterized and conscious pig model. The basic kinetics of acylcarnitines were studied in chapter 5, using stable C2- and C16-carnitine isotopes in mice with various degrees of insulin resistance. Here we tried to elucidate if acylcarnitine kinetics such as rates of appearance or elimination rates are different in various insulin sensitive states.In chapter 6, effects of weight loss on acylcarnitine levels were studied in relation to insulin sensitivity and energy expenditure in 60 obese human subjects. Based on the assumption that elevated acylcarnitine levels accompany insulin resistance, we expected that acylcarnitine levels would decrease along with improvements in glucose tolerance.Chapter 7 describes the effects of increasing carnitine levels in an obese model on FAO rates, energy expenditure and insulin sensitivity. Here we report the effects of increased carnitine availability by administration of the carnitine precursor gamma-butyrobetaine in lean and obese mice.Finally, chapter 8 discusses the results of the separate projects in relation to the current knowledge of acylcarnitine metabolism and if our results support the hypothesis on the role of acylcarnitines in the etiology of insulin resistance. The importance of acylcarni-tine tissue specificity, kinetics and substrates in different metabolic circumstances is put in perspective.

K007_000000_HereThere and EveryWhere boekje.indd 10 09-02-15 08:30

Page 15: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

11A SHORT INTRODUCTION

References1. Randle PJ, Garland PB, Hales CN, Newsholme EA. The glucose fatty-acid cycle. Its role in insulin sensi-

tivity and the metabolic disturbances of diabetes mellitus. Lancet. 1963;1(7285):785-9.2. Vaz FM, Wanders RJA. Carnitine biosynthesis in mammals. Biochemical Journal. 2002;361(3):417-29.3. Bremer J. Carnitine--metabolism and functions. Physiol Rev. 1983;63(4):1420-80.4. Violante S, Ijlst L, Te Brinke H, Tavares de Almeida I, Wanders RJ, Ventura FV, et al. Carnitine palmitoyl-

transferase 2 and carnitine/acylcarnitine translocase are involved in the mitochondrial synthesis and export of acylcarnitines. FASEB J. 2013;27(5):2039-44.

5. Schooneman MG, Achterkamp N, Argmann CA, Soeters MR, Houten SM. Plasma acylcarnitines inade-quately reflect tissue acylcarnitine metabolism. Biochim Biophys Acta. 2014.

6. Wanders R, Vreken P, den Boer M, Wijburg F, Van Gennip A, Ijlst L. Disorders of mitochondrial fatty acyl-CoA beta-oxidation. Journal of Inherited Metabolic Disease. 1999;22(4):442-87.

7. Muoio DM, Koves TR. Lipid-induced metabolic dysfunction in skeletal muscle. Novartis Found Symp. 2007;286:24-38.

8. Koves TR, Ussher JR, Noland RC, Slentz D, Mosedale M, Ilkayeva O, et al. Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance. Cell Metab. 2008;7(1):45-56.

9. Mihalik SJ, Goodpaster BH, Kelley DE, Chace DH, Vockley J, Toledo FG, et al. Increased levels of plasma acylcarnitines in obesity and type 2 diabetes and identification of a marker of glucolipotoxicity. Obe-sity (Silver Spring). 2010;18(9):1695-700.

10. Noland RC, Koves TR, Seiler SE, Lum H, Lust RM, Ilkayeva O, et al. Carnitine insufficiency caused by aging and overnutrition compromises mitochondrial performance and metabolic control. J Biol Chem. 2009;284(34):22840-52.

11. Hoppel CL, Genuth SM. Carnitine metabolism in normal-weight and obese human subjects during fasting. Am J Physiol. 1980;238(5):E409-E15.

12. Soeters MR, Sauerwein HP, Duran M, Wanders RJ, Ackermans MT, Fliers E, et al. Muscle acylcarnitines during short-term fasting in lean healthy men. Clin Sci (Lond). 2009;116(7):585-92.

13. Holland WL, Knotts TA, Chavez JA, Wang LP, Hoehn KL, Summers SA. Lipid mediators of insulin resis-tance. Nutr Rev. 2007;65(6 Pt 2):S39-S46.

14. Holland WL, Summers SA. Sphingolipids, insulin resistance, and metabolic disease: new insights from in vivo manipulation of sphingolipid metabolism. Endocr Rev. 2008;29(4):381-402.

15. Shulman GI. Cellular mechanisms of insulin resistance. The Journal of Clinical Investigation. 2000;106(2):171-6.

16. Adams SH, Hoppel CL, Lok KH, Zhao L, Wong SW, Minkler PE, et al. Plasma Acylcarnitine Profiles Suggest Incomplete Long-Chain Fatty Acid beta-Oxidation and Altered Tricarboxylic Acid Cycle Activity in Type 2 Diabetic African-American Women. The Journal of Nutrition. 2009;139(6):1073-81.

17. An J, Muoio DM, Shiota M, Fujimoto Y, Cline GW, Shulman GI, et al. Hepatic expression of malonyl-CoA decarboxylase reverses muscle, liver and whole-animal insulin resistance. Nat Med. 2004;10(3):268-74.

18. Newgard CB, An J, Bain JR, Muehlbauer MJ, Stevens RD, Lien LF, et al. A Branched-Chain Amino Ac-id-Related Metabolic Signature that Differentiates Obese and Lean Humans and Contributes to Insulin Resistance. Cell Metabolism. 2009;9(4):311-26.

19. Redman LM, Huffman KM, Landerman LR, Pieper CF, Bain JR, Muehlbauer MJ, et al. Effect of Caloric Restriction with and without Exercise on Metabolic Intermediates in Nonobese Men and Women. Journal of Clinical Endocrinology & Metabolism. 2011;96(2):E312-E21.

K007_000000_HereThere and EveryWhere boekje.indd 11 09-02-15 08:30

Page 16: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

Acylcarnitines: reflecting or inflicting insulin resistance?

Marieke G. Schooneman, Frédéric M. Vaz, Sander M. Houten, Maarten R. Soeters

Diabetes (2013) 62, 1-8

C 2

K007_000000_HereThere and EveryWhere boekje.indd 12 09-02-15 08:30

Page 17: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

Acylcarnitines: reflecting or inflicting insulin resistance?

Marieke G. Schooneman, Frédéric M. Vaz, Sander M. Houten, Maarten R. Soeters

Diabetes (2013) 62, 1-8

C 2

K007_000000_HereThere and EveryWhere boekje.indd 13 09-02-15 08:30

Page 18: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

K007_000000_HereThere and EveryWhere boekje.indd 14 09-02-15 08:30

Page 19: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

15ACYLCARNITINES: REFLECTING OR INFLICTING INSULIN RESISTANCE?

Introduction

The incidence of obesity and insulin resistance is growing and the increase in type 2 diabetes mellitus constitutes one of the biggest challenges for our healthcare systems. Many theories are proposed for the induction of insulin resistance in glucose and lipid metabolism and its metabolic sequelae. One of these mechanisms is lipotoxicity (1-4): here excess lipid supply and subsequent lipid accumulation in insulin sensitive tissues such as skeletal muscle interferes with insulin responsive metabolic pathways. Various lipid intermediates, like ceramides, gangliosides, diacylglycerol and other metabolites, have been held responsible for insulin resistance (2;3;5-10). These intermediates can exert such effects because they are signaling molecules and building blocks of cellular membranes, which harbor the insulin receptor. In addition, lipids play an important role in energy homeostasis. Fatty acids (FA) can be metabolized via mitochondrial FA oxida-tion (FAO), which yields energy (11). As such FAO competes with glucose oxidation in a process known as the glucose-fatty acid or Randle cycle (12).Muoio and colleagues proposed an alternative mechanism in which FAO rate outpaces the TCA cycle, thereby leading to the accumulation of intermediary metabolites such as acylcarnitines that may interfere with insulin sensitivity (1;13;14). This accumulation of acylcarnitines corroborates with some human studies showing that acylcarnitines are associated with insulin resistance (15-17). In addition, acylcarnitines have a long history in the diagnosis and neonatal screening of FAO defects and other inborn errors of metabo-lism (18). This knowledge may aid to understand the interaction between FAO and insulin resistance and fuel future research. In this review, we discuss the role of acylcarnitines in FAO and insulin resistance as emerging from animal and human studies.

Physiological role of Acylcarnitines

Carnitine biosynthesis and regulation of tissue carnitine contentTo guarantee continuous energy supply, the human body oxidizes considerable amounts of fat besides glucose. L-Carnitine transports activated long-chain FAs from the cytosol into the mitochondrion, and is therefore essential for FAO. Carnitine is mainly absorbed from the diet, but can be formed through biosynthesis (19). In several proteins, lysine residues are methylated to trimethyllysine (19). Four enzymes convert trimethyllysine into carnitine (19) of which the last step is the hydroxylation of butyrobetaine into carnitine by gamma-butyrobetaine dioxygenase (BBD). BBD is only present in human liver, kidney, and brain, which are the sites where actual carnitine biosynthesis takes place (19). Other tissues such as skeletal muscle must acquire carnitine from the blood. Treatment with a synthetic PPAR–alpha agonist increased BBD activity and carnitine levels in liver (20). This suggests that the nuclear receptor PPAR–alpha, which plays a crucial role in the adaptive response to fasting, is a regulator of (acyl)carnitine metabolism (20).The plasmalemmal carrier OCTN2 is responsible for cellular carnitine uptake in various organs including reabsorption from urine in the kidney. As is the case for BBD, OCTN2

K007_000000_HereThere and EveryWhere boekje.indd 15 09-02-15 08:30

Page 20: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

16

expression in liver is regulated by PPAR–alpha. A synthetic PPAR–alpha agonist increased OCTN2 expression in wild type mice and caused a rise in carnitine levels in plasma, liver, kidney, and heart (20). In PPAR–alpha -/- mice, low OCTN2 expression contributed to de-creased tissue and plasma carnitine levels (20).

The carnitine shuttleOnce inside the cell, FAs are activated by esterification to CoA. Then, the carnitine shut-tle transports long-chain acyl-CoAs into mitochondria via their corresponding carnitine ester (Figure 1) (21). Long-chain acyl-CoAs are converted to acylcarnitines by carnitine palmitoyltransferase 1 (CPT1), which exchanges the CoA moiety for carnitine. CPT1 is lo-cated at the outer mitochondrial membrane and three isoforms are known: CPT1a, 1b and 1c are encoded by separate genes (21). CPT1a is expressed in liver and most other abdominal organs, as well as human fibroblasts. CPT1b is selectively expressed in heart, skeletal muscle, adipose tissue and testes (11). CPT1c is only expressed in the endoplas-matic reticulum (and not the mitochondria) of neurons in the brain (22).

FIGURE 1 The carnitine shuttle After transportation into the cell by fatty acid transporters (FAT), fatty acids (FA) are activated

by esterification to CoA. Subsequently, carnitine palmitoyltransferase 1 (CPT1) exchanges the CoA moiety for carnitine (C).

The resulting acylcarnitine (AC) is transported across the inner mitochondrial membrane into the mitochondrion by carnitine

acylcarnitine translocase (CACT). Once inside, carnitine palmitoyltransferase 2 (CPT2) reconverts the acylcarnitine back into

free carnitine and a long-chain acyl-CoA that can undergo fatty acid oxidation for ATP production via the tricarboxylic acid

cycle (TCA) and respiratory chain (ETC) .

FAT

MITOCHONDRION

CARNITINESHUTTLE

FA

ACC

FATTY ACID SUPPLYLIPID FLUX

OCTN2

ß-OXIDATION

TCA

ETC

ATP

CYTOSOL

MITOCHONDRION

CYTOSOL

CPT1

CPT2

CACT

ACFA

C

CoA

CoA

C

AC

CoA

ß-OXIDATION

FACoA

ACYLCARNITINES: REFLECTING OR INFLICTING INSULIN RESISTANCE?

K007_000000_HereThere and EveryWhere boekje.indd 16 09-02-15 08:30

Page 21: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

17

CPT1 is an important regulator of FAO flux. Glucose oxidation after a meal leads to in-hibition of CPT1 activity via the FA-biosynthetic intermediate malonyl-CoA (23), which is produced by acetyl-CoA carboxylase (ACC) (24). There are two ACC isoforms. ACC1 plays a role in FA biosynthesis. ACC2 has been implicated in the regulation of FAO main-ly because of its localization to the outer mitochondrial membrane (25). Conversely, in the fasting state, activated AMP-activated protein kinase inhibits ACC resulting in falling malonyl-CoA levels, thereby permitting CPT1 activity and thus FAO. CPT1a is limiting for hepatic FAO and ketogenesis (26). Although the inhibition of malonyl-CoA on CPT1b is more potent than on CPT1a, no unequivocal evidence exists showing its control over muscle FAO (27).FAO is also regulated at the transcriptional level. PPAR-alpha, but also PPAR-beta/-delta, regulates the transcription of many enzymes involved in FAO. There is ample evidence that both PPARs participate in the transcriptional regulation of CPT1b (28-30). Regulation of CPT1a by PPAR is less prominent (21). After production of acylcarnitines by CPT1, the mitochondrial inner membrane transporter carnitine acylcarnitine translocase (CACT or SLC25A20) transports the acylcarnitines into the mitochondrial matrix. The FA transporter CD36 possibly facilitates transfer of acylcar-nitines from CPT1 to CACT (31). Finally, the enzyme CPT2 reconverts acylcarnitines back into free carnitine and long-chain acyl-CoAs, which can then be oxidized (21) (Figure 1).

Analysis of acylcarnitinesWith the introduction of tandem mass spectrometry (MS) in clinical chemistry in the 1990’s, it became relatively easy to measure acylcarnitine profiles. In these profiles, the mass-to-charge ratio reflects the length and composition of the acyl chain (32). This technique rapidly became the preferred screening test to diagnose inherited disorders in FAO, which lead to prominent changes in the acylcarnitine profile, with a pattern spe-cific for the deficient enzyme. More recently, acylcarnitine analysis is employed to inves-tigate more common metabolic derangements such as insulin resistance.Although most acylcarnitines are derived from FAO, they can be formed from almost any CoA ester (18). Other intermediates that yield acylcarnitines are ketone bodies (C4-3OH-car-nitine (33)), degradation products of lysine, tryptophane, valine, leucine and isoleucine (C3- and C5-carnitine and others), and carbon atoms from glucose (acetylcarnitine) (18).The standard acylcarnitine analysis using tandem MS cannot discriminate between ste-reoisomers and other isobaric compounds, which have the same nominal mass but a different molecular structure. These compounds can be separated using liquid chroma-tography-tandem MS (34). This is illustrated by C4-OH-carnitine, which can be derived from the CoA ester of the ketone body D-3-hydroxybutyrate, (D-C4-OH-carnitine), the FAO intermediate L-3-hydroxybutyryl-CoA (L-C4-OH-carnitine), and L-3-hydroxyisobu-tyryl-CoA, an intermediate in the degradation of valine (L-isoC4-OH-carnitine) (33).

The origin of plasma acylcarnitinesThe fact that acylcarnitines can be measured in plasma, illustrates that they are trans-ported across cell membranes. Two transporters have been implicated in the export of

ACYLCARNITINES: REFLECTING OR INFLICTING INSULIN RESISTANCE?

K007_000000_HereThere and EveryWhere boekje.indd 17 09-02-15 08:30

Page 22: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

18 ACYLCARNITINES: REFLECTING OR INFLICTING INSULIN RESISTANCE?

acylcarnitines. In addition to import, OCTN2 can export (acyl)carnitines (35). Also, the monocarboxylate transporter 9 (SLC16A9) may play a role in carnitine efflux (36). Although these putative transporters have been identified, the exact nature of this transport is unknown, but seems largely dependent on the intracellular acylcarnitine concentration (35). Early studies in rodent heart, liver, and brain mitochondria proved mitochondrial efflux of acylcarnitines and suggested this to be dependent on the substrate and tissue as well as the availability of alternative acyl-CoA utilizing reactions (37). In humans, acyl-carnitine efflux is exceptionally well evidenced by the acylcarnitine profiles of patients with a FAO defect (18). From a more physiological view, diets and fasting modulate the plasma acylcarnitine profile, which reflects changes in flux through the FAO pathway (13;16;38;39). However, exact rates of acylcarnitine production in relation to the FAO flux under different conditions remain to be determined.It is expected that muscle or liver contribute largely to acylcarnitine turnover. Early stud-ies showed that liver acylcarnitines correlated with plasma acylcarnitines in fasted ma-caques, but the individual chain lengths were not studied (40). A liver-plasma relation is plausible, considering that the liver accounts for most of the FAO activity during fasting. Human data are lacking, but muscle acylcarnitines did not correlate with plasma acylcar-nitines during short-term fasting (16).The physiological role of acylcarnitine efflux to the plasma compartment is unknown, but several scenarios are likely. Acylcarnitine formation prevents CoA trapping, allowing con-tinuation of CoA-dependent metabolic processes (21;41). In addition to plasma, acylcar-nitines are found also in bile and urine (42;43), suggesting that acylcarnitine efflux may serve as a detoxification process. Combined, the total daily bile and urine production of acylcarnitine is less than 200 μmol. This can be calculated to be less than 0.01 % of daily energy requirements which is a negligible amount in terms of potential energy loss. Moreover, intestinal re-uptake of bile acylcarnitines is possible. Alternatively, plasma acylcarnitines may serve as a means of transportation between cells or organs or sink for cellular/tissue acylcarnitine sequestration. Questions that remain are the contribution of specific tissues and organs to plasma acylcarnitine levels and the turnover rates of the individual acylcarnitine species in plasma.

Acylcarnitine metabolism in relation to insulin resistance

Current views on lipid metabolism in insulin resistanceFAO may be quantitatively and qualitatively different in insulin resistant subjects com-pared to healthy subjects, but a more pertinent conundrum is if increased FAO is either capable to limit insulin resistance via decreasing lipid accumulation or increasing insulin resistance via accumulation of incomplete FAO products such as acylcarnitines (1-3;13;14). Several theories describe mechanisms within the cytosol that can cause insulin resistance (figure 2). It has generally been accepted that chronic overnutrition leads to increased cytosolic lipid content of insulin responsive tissues (such as liver and skeletal muscle). This negatively affects the insulin sensitivity of these tissues by inhibiting insulin

K007_000000_HereThere and EveryWhere boekje.indd 18 09-02-15 08:30

Page 23: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

19ACYLCARNITINES: REFLECTING OR INFLICTING INSULIN RESISTANCE?

FIGURE 2. Mechanisms of lipid-induced insulin resistance After transportation into the cell, fatty acids (FA) can be stored,

oxidized or used as building blocks and signaling molecules (not all shown). Excess lipid supply and subsequent accumulation

in insulin sensitive tissues such as skeletal muscle, is proposed to interfere with different insulin responsive metabolic

pathways via various mechanisms. Firstly [1], increased intracellular lipid content inhibits insulin signaling via lipid

intermediates such as ceramides, diacylglycerol (DAG) or gangliosides (GM3), via effects on protein phosphatase A2 (PPA2)

and protein kinase B (AKT), protein kinase C (PKC) or effects on the insulin receptor in the cell membrane (1;3;5-8;44). Effects

of lipid intermediates on inhibitors of nuclear factor-kappa beta kinase subunit beta (IKKbeta) and c-Jun N-terminal kinase

1 (JnK1) are not depicted. The second mechanism [2] is a decreased number of functional mitochondria resulting in lower

FAO rates and increased accumulation of cytosolic lipid, again interfering with insulin sensitivity (2;9). Finally [3], metabolic

overload of mitochondria leads to incomplete beta-oxidation. Here, oxidation of fatty acids (FA) outpaces the tricarboxylic

acid cycle (TCA) and respiratory chain (RC), resulting in intramitochondrial accumulation of FAO intermediates like

acylcarnitines. These subsequently impinge on insulin signaling (1;48;50-56). Here, only the direct effects of acylcarnitines

on nuclear factor-kappa beta (NF-k beta) have been proposed (70).

TCAETC

ß-OXIDATION > TCA FLUX

ß-OXIDATION

$$

FA

IR

LIPID INDUCEDSTRESS

ACYLCARNITINESINTRAMUSCULAR KETONES

$$

REDUCED MITOCHONDRIAL DENSITY/FUNCTION ß-OXIDATION$

DAG

$

FA

FAT

2

3

CERAMIDE

$

PP2AAkt2

PKC

$

IMPAIRED INSULINSIGNALLING

1

LIPID FLUX

FFA SUPPLY

GM3

LCFA

FAT

NFκB?

$

K007_000000_HereThere and EveryWhere boekje.indd 19 09-02-15 08:30

Page 24: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

20

signaling via intermediates as ceramide, diacylglycerol, gangliosides and possible other long-chain FA-derived metabolites (1;3;5-8;44). Although contested now, cytosolic lipid accumulation was also suggested to arise from mitochondrial dysfunction and as a con-sequence decreased FAO rate (2;9;14;45;46). Likewise, increased levels of malonyl-CoA were suggested to limit the mitochondrial entrance of long-chain FAs by blocking CPT1; thus resulting in accumulating cytosolic long-chain FAs and decreasing FAO rate (10). Alternatively, more recent mechanistic (13;47;48) and metabolomic (49-54) studies asso-ciated obesity-induced insulin resistance with intramitochondrial disturbances. In this model, lipid overload leads to increased rather than decreased FAO in skeletal muscle. This coincides with accumulating acylcarnitines, an inability to switch to carbohydrate substrate and a depletion of TCA cycle intermediates suggesting that FAO flux does not match TCA cycle flux leading to incomplete FAO (13;47;48). In vitro interfering with FA uptake in L6 myocytes or a coordinate induction of FAO and TCA cycle enzymes by ex-ercise or PGC1alpha overexpression prevented insulin resistance (13;48). Moreover, us-ing carnitine to stimulate FAO without affecting the TCA cycle in these myocytes was dose dependently associated with insulin resistance (13). Zucker Diabetic Fatty (ZDF) rats, a model for more severe insulin resistance, had higher acylcarnitines but lower TCA cycle intermediates (such as citrate, malate and succinate) in skeletal muscle, again suggesting that increased FAO induces insulin resistance when not followed by propor-tionally increased TCA cycle activity (13). Additionally, the malonyl-CoA decarboxylase (MCD)-/- mouse that had decreased FAO due to higher malonyl-CoA concentrations, resisted diet-induced insulin resistance, which further implicated FAO in the pathogenesis of insulin resistance (13). The available studies on acylcarnitine metabolism and the relationship with insulin resistance will be discussed in the next sections with a focus on human studies.

The effect of increased lipid flux on mitochondrial FA uptake and oxidation:implications for insulin sensitivity.Insulin dependent type 2 diabetes mellitus patients had lower (~25%) carnitine concen-trations especially with longer standing or complicated disease (55;56). Interestingly, carnitine infusions increased FAO in lean healthy subjects, but only when high dose insu-lin was co-administered (57;58), which may be explained by an increased muscle OCTN2 expression under these conditions (59). The importance of insulin for cellular carnitine uptake is underscored by the finding that insulin and carnitine administration lowered muscle malonyl-CoA and lactate concentrations while muscle glycogen increased (58). These findings are supported by animal studies, which demonstrated that carnitine lev-els were diminished in skeletal muscle of multiple insulin resistant rat models. A high fat diet (HFD) exacerbated the age-related decrease of tissue carnitine content in these rats (primarily skeletal muscle, liver and kidney) (60). Moreover carnitine supplementation of HFD animals decreased plasma glucose levels and HOMA indices (60;61). Likewise, carni-tine supplementation improved insulin-stimulated glucose disposal in mouse models of diet-induced obesity and genetic diabetes (62). Recently it was shown that six months of carnitine supplementation improved glucose homeostasis in insulin-resistant humans (14).Although supplementation of carnitine possibly augments FAO and insulin sensitivity; the

ACYLCARNITINES: REFLECTING OR INFLICTING INSULIN RESISTANCE?

K007_000000_HereThere and EveryWhere boekje.indd 20 09-02-15 08:30

Page 25: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

21

lower carnitine levels in diabetes patients are unexplained. On the one hand carnitine uptake is insulin-dependent and therefore the a) absence of, or b) resistance to insulin may be the cause of lower carnitine levels. On the other hand, higher lipid load may lead to higher acylcarnitine concentrations and thus lower free carnitine.In addition, several studies reported on the carnitine shuttle and its effects on the rate of FAO in the development of insulin resistance. Obese subjects had lower CPT1 and citrate synthase content in muscle and lower FAO, suggesting that lesions at CPT1 and post-CPT1 events (i.e. mitochondrial content) may lower FAO in obesity (63). Although short-term inhibition of CPT1 with etomoxir in humans did not impede insulin sensitivity despite increased intramyocellular lipid accumulation (64), prolonged inhibition in rats resulted in the accumulation of intramyocellular lipid and increased insulin resistance while doubling adiposity despite feeding a low fat diet (65). These results all led to the assumption that low FAO rates due to decreased function of CPT1 were associated with insulin resistance, possibly caused by an accumulation of intramyocellular lipid inter-mediates and their interference with insulin signaling. Indeed, CPT1 activity increased after an endurance training program in obese subjects, coinciding with increased FAO, improved glucose tolerance and insulin sensitivity (66). However, this may also be ex-plained by the stimulatory effect of endurance training on mitochondrial function (i.e. TCA cycle and respiratory chain activity), thereby relieving the heavy lipid burden on mitochondria (48;67). In contrast to the model where excess FAO induces insulin re-sistance, these data suggest that decreasing mitochondrial FA uptake results in elevated intramuscular lipid levels and subsequent insulin resistance. However, increasing FAO by carnitine treatment in animals and humans permits mitochondrial FA uptake and oxidation that benefits insulin sensitivity. These observations will have to be reconciled with other studies that implicated incomplete FAO and acylcarnitine accumulation in the pathogenesis of insulin resistance.

Short chain acylcarnitines in insulin resistance Older work reported elevated acylcarnitine levels in obese insulin resistant subjects (15), but acylcarnitines were not suggested to be implicated in insulin resistance at that time. The shortest acylcarnitine, acetylcarnitine, is of particular interest since it may illustrate the controlling role of acetyl-CoA on substrate switching and thus metabolic flexibili-ty. The mitochondrial enzyme carnitine acetyltransferase (CrAT) converts acetyl-CoA to the membrane permeable acetylcarnitine and permits mitochondrial efflux of excess acetyl-CoA that otherwise could inhibit pyruvate dehydrogenase (68). Infusing intralipid decreased insulin sensitivity while increasing muscle acetylcarnitine (69). The same was true for plasma and muscle acetylcarnitine levels under high FAO conditions (starving) suggesting upregulation of CrAT to traffic acetyl-moieties (16). In contrast to lower CrAT expression in diabetic subjects (68), plasma acetylcarnitine levels showed significant positive correlation with HbA1c levels over a wide range of insulin sensitivity suggesting upregulation of CrAT in insulin resistant states (70).There is some complexity as both lipid and glucose oxidation funnel into acetylcarni-tine as supported by different findings (68;71). First, the insulin mediated suppression of

ACYLCARNITINES: REFLECTING OR INFLICTING INSULIN RESISTANCE?

K007_000000_HereThere and EveryWhere boekje.indd 21 09-02-15 08:30

Page 26: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

22

muscle acetylcarnitine occurred under high FAO conditions, but not postabsorptively (i.e. higher glucose availability) (16). Also, muscle acetylcarnitine correlated negatively with FAO in the postabsorptive state (71), whereas plasma acetylcarnitine correlated with plasma glucose levels in the postprandial state (72). In light of these data, the question is interesting if CrAT really favours fatty acid-derived acetyl-CoA over glucose-derived acetyl-CoA since this might imply intracellular compartmentalisation of acetyl-CoA (68). Moreover, glucose-derived acetyl-CoA can be carboxylated by ACC, producing the CPT1 inhibitor malonyl-CoA. Direct effects of FAO-derived acetyl-CoA on insulin action are unknown.C4-OH-carnitine (i.e. the carnitine ester of 3-hydroxybutyrate) has been proposed to cause insulin resistance: hepatic overexpression of malonyl-CoA decarboxylase (MCD) in rats on a high fat diet reversed whole-body, liver, and muscle insulin resistance whilst only decreasing C4-OH-carnitine within the acylcarnitine profile (47). In fasted humans, plas-ma and muscle C4-OH-carnitine increased (33). The increase in C4-OH-carnitine in these animal and human studies is quantitatively much more pronounced then the increase in acetylcarnitine; thus C4-OH-carnitine production may exert greater demands on cellular carnitine stores. Moreover ketone bodies yield acetyl-CoA, which stimulates PDK4 and thus inhibits glucose oxidation (73). In summary, under conditions characterized by higher FAO, elevated short chain acylcarnitines may reflect higher lipid fluxes but a direct relation to insulin resistance remains to be established.

Amino acid-derived acylcarnitines in insulin resistanceMetabolomics showed that branched-chain and aromatic amino acids (isoleucine, leu-cine, valine, tyrosine and phenylalanine) (74), significantly correlated with present or future diabetes (54;74;75). In line with this, the branched-chain amino acid-derived C3- and C5-carnitine, together with FA-derived C6- and C8-carnitine, were higher in obese and DM2 subjects compared to lean controls (17;54). In the same study, C4-dicarboxyl-carnitine (C4DC-carnitine), also derived from branched-chain amino acid metabolism, showed a positive correlation with basal glucose levels and HbA1c (17). In comparison to obese non insulin resistant subjects, DM2 subjects also had higher C3- and C5-carnitine levels compared to controls during insulin administration. Here, C3- but not C5-carni-tine correlated negatively with glucose disposal (17).At first glance, correlations of acylcarnitines to surrogate markers of insulin resistance fit with mitochondrial overload and incomplete FAO. Acylcarnitines, however, also directly reflect the oxidation rate of FA and amino acids, which is supported by human nutrition-al intervention studies (16;33;38;39). The uncertainty regarding the direct interference of short chain acylcarnitines and their metabolism with insulin signaling processes and insulin sensitivity warrants care when attributing a primary role for amino acid-derived acylcarnitines in the induction of insulin resistance.

Medium- and long-chain acylcarnitines: more evidence for insulin resistant effects?Long chain fatty acids such as palmitic acid were associated with insulin resistance, making a role for long chain acylcarnitines such as C16 in insulin resistance conceivable

ACYLCARNITINES: REFLECTING OR INFLICTING INSULIN RESISTANCE?

K007_000000_HereThere and EveryWhere boekje.indd 22 09-02-15 08:30

Page 27: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

23

(3;44). In 1980, Hoppel et al showed that the fasting-induced increase in plasma acylcar-nitines was restored upon refeeding in lean subjects within 24 hours opposed to four days in obese subjects suggesting an impaired metabolic flexibility in the latter (15). The hypothesis that obesity-induced alteration in the acylcarnitine profile are caused by incomplete FAO was based largely on two animal studies by the same group showing that long-chain acylcarnitine species (C16, C18:2, C18:1 and C18) were persistently increased in diet-induced obese rats, in both fed and fasted state (13;48). As reported for humans, most acylcarnitine species decreased upon refeeding in the chow fed control group, but not in the obese animals, suggesting they were incapable of adjusting their metabolism in response to refeeding. Although excessive and incomplete FAO can be responsible for insulin resistance, it can be argued that FAO probably must be in relative excess to oxidation in TCA and respiratory chain in order to guarantee continuous energy supply.Obese and insulin resistant humans had higher plasma long-chain acylcarnitine levels compared to lean controls (17). Upon insulin infusion, long-chain acylcarnitines de-creased overall, but to a lesser degree in the diabetic subjects. This was in agreement with lower resting energy expenditure (RER), indicating ongoing FAO or lipid flux (met-abolic inflexibility) (17). Moderate correlations between acylcarnitine profiles and vari-ous clinical characteristics (i.e. higher BMI, basal FFA levels, insulin sensitivity) point at a causal relationship. The DM2 subjects were unable to suppress acylcarnitines during insulin infusion in contrast to matched obese controls: therefore, elevated long-chain acylcarnitines in the diabetic group likely reflect increased lipid flux and illustrates the tight connection of acylcarnitines with FAO flux (17). Postprandially, plasma long-chain acylcarnitines did decrease in obese insulin resistant subjects, but the magnitude of this decrease correlated with both pre-meal insulin-me-diated glucose disposal rates and FAO and has been largely explained by nadir levels of C12:1, C14, and C14:1-carnitine (72). This showed that the more insulin sensitive subjects are, the more capable they are at metabolizing FAs. Metabolomics in healthy overweight calorie restricted subjects yielded comparable results: here acylcarnitines correlated significantly with plasma insulin and FFA levels albeit with low correlation coefficient (49).All in all, acylcarnitines with longer chain lengths are associated with insulin resistance, which seems logic in the light of known effects of long-chain FAs on insulin signaling. Indeed, acylcarnitines can reside in cell membranes since they are amphipathic mole-cules. Increasing chain length favors partitioning into the membrane phase (e.g. C16- and 18-carnitine) (76). It is interesting to speculate that long-chain acylcarnitines can inter-fere with insulin signaling directly within the cell membrane (3). On the other hand, acyl-carnitines seem to track with higher lipid flux and as such may only indicate higher FAO.

Acylcarnitines: reflecting or inflicting insulin resistance?The concept of lipotoxicity is generally accepted in the field of obesity-induced impair-ment of insulin sensitivity and more and more attention has attributed to intramitochon-drial alterations and impairments in FAO thereby focusing on acylcarnitines (1). Collected evidence shows that acylcarnitines have distinct functions in mitochondrial lipid metab-olism. The transmembrane export of acylcarnitines suggests that they not only prevent

ACYLCARNITINES: REFLECTING OR INFLICTING INSULIN RESISTANCE?

K007_000000_HereThere and EveryWhere boekje.indd 23 09-02-15 08:30

Page 28: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

24

the accumulation of noxious acyl-CoAs, but also reduce CoA trapping which is crucial for many metabolic pathways (21;41). Additionally, the metabolism of short chain acyl-carnitines and the interaction of acetyl-CoA and acetylcarnitine via CrAT may regulate the pyruvate dehydrogenase complex thereby affecting glucose oxidation (68). Besides mitochondrial need to liberate CoA and export acetyl-CoA, acylcarnitines may simply reflect the FAO flux.The concept of increased, though incomplete FAO by disproportional regulation of FAO, TCA cycle and respiratory chain is attractive to explain insulin resistance. However, there remains doubt about this mechanism and there is no proof that acylcarnitines play a role in the induction of insulin resistance itself. Acylcarnitines are present under physi-ological conditions and their levels vary according to dietary circumstances (13;16;38;39) The acylcarnitine fluxes are unknown but probably much lower than FAO flux. Moreover, it can be argued that flux of FAO probably will be in relative excess to downstream oxi-dation in TCA and respiratory chain to guarantee continuous substrate supply and allow fine tuning and anticipation for metabolic changes (e.g. activity). Otherwise, the organ-ism’s response to increased energy demands will be attenuated leading to more severe impairment of mitochondrial function as evidenced by the inherited FAO disorders.Observational studies associating different acylcarnitines to a variety of endpoints may yield new hypotheses but are unlikely to move the field forward from a mechanistic per-spective. Many questions are unanswered and some issues deserve particular attention. Tracer studies can quantify FAO flux and acylcarnitine production in different insulin resistant models on the cellular, tissue and whole organism level. Multiple animal and human models can help to investigate the effect of carnitine availability on insulin sen-sitivity. Mouse models for and humans with primary carnitine deficiency can be used to investigate the effect of carnitine availability on substrate switching and insulin sensi-tivity. In vitro work in muscle or liver cell lines is still important to dissect the influence of acylcarnitines on conventional insulin signaling or mechanisms of nutrient-induced mitochondrial stress. In this respect, different animal and human FAO disorders that accumulate acylcarnitines may undergo insulin sensitivity testing. The contribution of different organs to plasma acylcarnitines can be investigated using transorgan arte-rio-venous balance isotope dilution techniques under different conditions. Finally, we may set foot in new areas where acylcarnitines may have unexpected roles like inter-action with the insulin receptor in the plasma membrane or signaling in the gut when co-secreted with bile. Recently, magnetic resonance spectroscopy was shown to image tissue acetylcarnitine in humans enabling noninvasive techniques to assay tissue acetyl-carnitine (77). All these studies and more are necessary to decide to what extent acylcar-nitines are reflecting or inflicting insulin resistance.

ACYLCARNITINES: REFLECTING OR INFLICTING INSULIN RESISTANCE?

K007_000000_HereThere and EveryWhere boekje.indd 24 09-02-15 08:30

Page 29: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

25

References1. Muoio,DM, Koves,TR: Lipid-induced metabolic dysfunction in skeletal muscle. Novartis Found Symp

286:24-38, 20072. Morino,K, Petersen,KF, Shulman,GI: Molecular Mechanisms of Insulin Resistance in Humans and Their

Potential Links With Mitochondrial Dysfunction. Diabetes 55:S9-S15, 20063. Holland,WL, Knotts,TA, Chavez,JA, Wang,LP, Hoehn,KL, Summers,SA: Lipid mediators of insulin resis-

tance. Nutr Rev 65:S39-S46, 20074. Shulman,GI: Cellular mechanisms of insulin resistance. The Journal of Clinical Investigation 106:171-

176, 20005. Krssak,M, Falk Petersen,K, Dresner,A, DiPietro,L, Vogel,SM, Rothman,DL, Shulman,GI, Roden,M: In-

tramyocellular lipid concentrations are correlated with insulin sensitivity in humans: a 1H NMR spec-troscopy study. Diabetologia 42:113-116, 1999

6. Gray,RE, Tanner,CJ, Pories,WJ, MacDonald,KG, Houmard,JA: Effect of weight loss on muscle lipid con-tent in morbidly obese subjects. American Journal of Physiology - Endocrinology And Metabolism 284:E726-E732, 2003

7. Pan DA, Lillioja S, Kriketos AD, Milner MR, Baur LA, Bogardus C: Skeletal muscle triglyceride levels are inversely related to insulin action. Diabetes 46:983-988, 1997

8. Goodpaster,BH, Theriault,R, Watkins,SC, Kelley,DE: Intramuscular lipid content is increased in obesity and decreased by weight loss. Metabolism 49:467-472, 2000

9. Mootha,VK, Lindgren,CM, Eriksson,KF, Subramanian,A, Sihag,S, Lehar,J, Puigserver,P, Carlsson,E, Rid-derstrale,M, Laurila,E, Houstis,N, Daly,MJ, Patterson,N, Mesirov,JP, Golub,TR, Tamayo,P, Spiegelman,B, Lander,ES, Hirschhorn,JN, Altshuler,D, Groop,LC: PGC-1 alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet 34:267-273, 2003

10. Ruderman,NB, Saha,AK, Vavvas,D, Witters,LA: Malonyl-CoA, fuel sensing, and insulin resistance. American Journal of Physiology - Endocrinology And Metabolism 276:E1-E18, 1999

11. Eaton,S: Control of mitochondrial beta-oxidation flux. Prog Lipid Res 41:197-239, 200212. Hue,L, Taegtmeyer,H: The Randle cycle revisited: a new head for an old hat. American Journal of Phys-

iology - Endocrinology And Metabolism 297:E578-E591, 200913. Koves,TR, Ussher,JR, Noland,RC, Slentz,D, Mosedale,M, Ilkayeva,O, Bain,J, Stevens,R, Dyck,JR, New-

gard,CB, Lopaschuk,GD, Muoio,DM: Mitochondrial overload and incomplete fatty acid oxidation con-tribute to skeletal muscle insulin resistance. Cell Metab 7:45-56, 2008

14. Muoio,DM, Neufer,PD: Lipid-Induced Mitochondrial Stress and Insulin Action in Muscle. Cell Metabo-lism 15:595-605, 2012

15. Hoppel,CL, Genuth,SM: Carnitine metabolism in normal-weight and obese human subjects during fasting. Am J Physiol 238:E409-E415, 1980

16. Soeters,MR, Sauerwein,HP, Duran,M, Wanders,RJ, Ackermans,MT, Fliers,E, Houten,SM, Serlie,MJ: Mus-cle acylcarnitines during short-term fasting in lean healthy men. Clin Sci (Lond) 116:585-592, 2009

17. Mihalik,SJ, Goodpaster,BH, Kelley,DE, Chace,DH, Vockley,J, Toledo,FG, DeLany,JP: Increased levels of plasma acylcarnitines in obesity and type 2 diabetes and identification of a marker of glucolipotoxici-ty. Obesity (Silver Spring) 18:1695-1700, 2010

18. Rinaldo,P, Cowan,TM, Matern,D: Acylcarnitine profile analysis. Genetics in Medicine 10: 200819. Vaz,FM, Wanders,RJA: Carnitine biosynthesis in mammals. Biochem J 361:417-429, 200220. van Vlies,N, Ferdinandusse S, Wanders R J A, Vaz F M: PPARalfa-activation results in enhanced carni-

tine biosynthesis and OCTN2 expression. Biochim Biophys Acta 1767:11, 200721. Ramsay,RR, Gandour,RD, van der Leij,FR: Molecular enzymology of carnitine transfer and transport.

Biochim Biophys Acta 1546:21-43, 200122. Sierra,AY, Gratacos,E, Carrasco,P, Clotet,J, Urena,J, Serra,D, Asins,G, Hegardt,FG, Casals,N: CPT1c Is

Localized in Endoplasmic Reticulum of Neurons and Has Carnitine Palmitoyltransferase Activity. Jour-nal of Biological Chemistry 283:6878-6885, 2008

23. McGarry,JD, Mannaerts,GP, Foster,DW: A possible role for malonyl-CoA in the regulation of hepatic fatty acid oxidation and ketogenesis. The Journal of Clinical Investigation 60:265-270, 1977

24. Zammit,VA: The malonyl-CoA-long-chain acyl-CoA axis in the maintenance of mammalian cell func-tion. Biochem J 343:505-515, 1999

25. Castle,JC, Hara,Y, Raymond,CK, Garrett-Engele,P, Ohwaki,K, Kan,Z, Kusunoki,J, Johnson,JM: ACC2 Is Ex-pressed at High Levels Human White Adipose and Has an Isoform with a Novel N-Terminus. PLoS One 4:e4369, 2009

ACYLCARNITINES: REFLECTING OR INFLICTING INSULIN RESISTANCE?

K007_000000_HereThere and EveryWhere boekje.indd 25 09-02-15 08:30

Page 30: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

26

26. Drynan,L, Quant,PA, Zammit,VA: Flux control exerted by mitochondrial outer membrane carnitine pal-mitoyltransferase over beta-oxidation, ketogenesis and tricarboxylic acid cycle activity in hepatocytes isolated from rats in different metabolic states. Biochem J 317:791-795, 1996

27. Eaton S, Fukumoto K, Paladio Duran N, Pierro A, Spitz L, Quant PA, Bartlett K: Carnitine palmitoyl trans-ferase-I and the control of myocardial beta-oxidation flux. Biochemical Society Transactions245-250, 2001

28. Yu,GS, Lu,YC, Gulick,T: Co-regulation of Tissue-specific Alternative Human Carnitine Palmitoyltransferase Ib Gene Promoters by Fatty Acid Enzyme Substrate. Journal of Biological Chemistry 273:32901-32909, 1998

29. Brandt,JM, Djouadi,F, Kelly,DP: Fatty Acids Activate Transcription of the Muscle Carnitine Palmitoyl-transferase I Gene in Cardiac Myocytes via the Peroxisome Proliferator-activated Receptor alfa. Jour-nal of Biological Chemistry 273:23786-23792, 1998

30. Mascaro,C, Acosta,E, Ortiz,JA, Marrero,PF, Hegardt,FG, Haro,D: Control of Human Muscle-type Carni-tine Palmitoyltransferase I Gene Transcription by Peroxisome Proliferator-activated Receptor. Journal of Biological Chemistry 273:8560-8563, 1998

31. Bezaire,V, Bruce,CR, Heigenhauser,GJF, Tandon,NN, Glatz,JFC, Luiken,JJJF, Bonen,A, Spriet,LL: Iden-tification of fatty acid translocase on human skeletal muscle mitochondrial membranes: essential role in fatty acid oxidation. American Journal of Physiology - Endocrinology And Metabolism 290:E509-E515, 2006

32. Chace,DH, Kalas,TA, Naylor,EW: Use of Tandem Mass Spectrometry for Multianalyte Screening of Dried Blood Specimens from Newborns. Clin Chem 49:1797-1817, 2003

33. Soeters,MR, Serlie,MJ, Sauerwein,HP, Duran,M, Ruiter,JP, Kulik,W, Ackermans,MtT, Minkler,PE, Hop-pel,CL, Wanders,RJA, Houten,SM: Characterization of D-3-hydroxybutyrylcarnitine (ketocarnitine): an identified ketosis-induced metabolite. Metabolism 61:966-973, 2012

34. Minkler,PE, Stoll,MSK, Ingalls,ST, Yang,S, Kerner,J, Hoppel,CL: Quantification of Carnitine and Acylcar-nitines in Biological Matrices by HPLC Electrospray Ionization-Mass Spectrometry. Clin Chem 54:1451-1462, 2008

35. Pochini,L, Oppedisano,F, Indiveri,C: Reconstitution into liposomes and functional characterization of the carnitine transporter from renal cell plasma membrane. Biochimica et Biophysica Acta (BBA) - Biomembranes 1661:78-86, 2004

36. Suhre,K, Shin,SY, Petersen,AK, Mohney,RP, Meredith,D, Wagele,B, Altmaier,E, Deloukas,P, rdmann,J, rundberg,E, ammond,CJ, e Angelis,MH, astenmuller,G, ottgen,A, ronenberg,F, angino,M, eisinger,C, eitinger,T, ewes,HW, ilburn,MV, rehn,C, affler,J, ied,JS, omisch-Margl,W, amani,NJ, mall,KS, rich Wich-mann,H, hai,G, llig,T, pector,TD, damski,J, oranzo,N, ieger,C: Human metabolic individuality in biomed-ical and pharmaceutical research. Nature 477:54-60, 2011

37. Lysiak,W, Toth,PP, Suelter,CH, Bieber,LL: Quantitation of the efflux of acylcarnitines from rat heart, brain, and liver mitochondria. Journal of Biological Chemistry 261:13698-13703, 1986

38. Kien,CL, Everingham,KI, Stevens,D, Fukagawa,NK, Muoio,DM: Short-Term Effects of Dietary Fatty Acids on Muscle Lipid Composition and Serum Acylcarnitine Profile in Human Subjects. Obesity 19:305-311, 2011

39. Costa CG, De Almeida IT, Jacobs C: Dynamic Changes of Plasma Acylcarnitine Levels Induced by Fast-ing and Sunflower Oil Challenge Test in Children. Pediatric Research 46:440, 1999

40. Bell,FP, DeLucia,A, Bryant,LR, Patt,CS, Greenberg,HS: Carnitine metabolism in Macaca arctoides: the effects of dietary change and fasting on serum triglycerides, unesterified carnitine, esterified (acyl) carnitine, and beta-hydroxybutyrate. The American Journal of Clinical Nutrition 36:115-121, 1982

41. Lopaschuk GD, Belke DD, Gamble J, Itoi T, Schönekess BO: Regulation of fatty acid oxidation in the mammalian heart in health and disease. Biochim Biophys Acta 1213:263-276, 1994

42. Mueller,P, Schulze,A, Schindler,I, Ethofer,T, Buehrdel,P, Ceglarek,U: Validation of an ESI-MS/MS screen-ing method for acylcarnitine profiling in urine specimens of neonates, children, adolescents and adults. Clinica Chimica Acta 327:47-57, 2003

43. Chalmers RA, Roe CR, Stacey TE, Hoppel CL: Urinary excretion of l-carnitine and acylcarnitines by patients with disorders of organic acid metabolism: evidence for secondary insufficiency of l-carni-tine. Pediatric Research 18:1325-1328, 1984

44. Samuel,V, Shulman,G: Mechanisms for Insulin Resistance: Common Threads and Missing Links. Cell 148:852-871, 2012

45. Patti,ME, Butte,AJ, Crunkhorn,S, Cusi,K, Berria,R, Kashyap,S, Miyazaki,Y, Kohane,I, Costello,M, Sacco-ne,R, Landaker,EJ, Goldfine,AB, Mun,E, DeFronzo,R, Finlayson,J, Kahn,CR, Mandarino,LJ: Coordinated

ACYLCARNITINES: REFLECTING OR INFLICTING INSULIN RESISTANCE?

K007_000000_HereThere and EveryWhere boekje.indd 26 09-02-15 08:30

Page 31: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

27

reduction of genes of oxidative metabolism in humans with insulin resistance and diabetes: Potential role of PGC1 and NRF1. Proceedings of the National Academy of Sciences 100:8466-8471, 2003

46. Petersen,KF, Dufour,S, Befroy,D, Garcia,R, Shulman,GI: Impaired Mitochondrial Activity in the Insu-lin-Resistant Offspring of Patients with Type 2 Diabetes. N Engl J Med 350:664-671, 2004

47. An J., Muoio D.M., Shiota M., Fujimoto Y., Cline G.W., Shulman G.I., Koves T.R., Stevens R., Millington D., Newgard C.B.: Hepatic expression of malonyl-CoA decarboxylase reverses muscle, liver and whole-an-imal insulin resistance. Nat Med 10:268-274, 2004

48. Koves,TR, Li,P, An,J, Akimoto,T, Slentz,D, Ilkayeva,O, Dohm,GL, Yan,Z, Newgard,CB, Muoio,DM: Peroxi-some Proliferator-activated Receptor-gamma Co-activator 1alfa-mediated Metabolic Remodeling of Skeletal Myocytes Mimics Exercise Training and Reverses Lipid-induced Mitochondrial Inefficiency. Journal of Biological Chemistry 280:33588-33598, 2005

49. Redman,LM, Huffman,KM, Landerman,LR, Pieper,CF, Bain,JR, Muehlbauer,MJ, Stevens,RD, Wen-ner,BR, Kraus,VB, Newgard,CB, Kraus,WE, Ravussin,E: Effect of Caloric Restriction with and without Exercise on Metabolic Intermediates in Nonobese Men and Women. Journal of Clinical Endocrinology & Metabolism 96:E312-E321, 2011

50. Shah,SH, Hauser,ER, Bain,JR, Muehlbauer,MJ, Haynes,C, Stevens,RD, Wenner,BR, Dowdy,ZE, Grang-er,CB, Ginsburg,GS, Newgard,CB, Kraus,WE: High heritability of metabolomic profiles in families bur-dened with premature cardiovascular disease. Mol Syst Biol 5: 2009

51. Huffman,KM, Shah,SH, Stevens,RD, Bain,JR, Muehlbauer,M, Slentz,CA, Tanner,CJ, Kuchibhatla,M, Hou-mard,JA, Newgard,CB, Kraus,WE: Relationships between circulating metabolic intermediates and in-sulin action in overweight to obese, inactive men and women. Diabetes Care 32:1678-1683, 2009

52. Huffman,KM, Slentz,CA, Bateman,LA, Thompson,D, Muehlbauer,MJ, Bain,JR, Stevens,RD, Wenner,BR, Kraus,VB, Newgard,CB, Kraus,WE: Exercise-Induced Changes in Metabolic Intermediates, Hormones, and Inflammatory Markers Associated With Improvements in Insulin Sensitivity. Diabetes Care 34:174-176, 2011

53. Bain,JR, Stevens,RD, Wenner,BR, Ilkayeva,O, Muoio,DM, Newgard,CB: Metabolomics Applied to Diabe-tes Research. Diabetes 58:2429-2443, 2009

54. Newgard,CB, An,J, Bain,JR, Muehlbauer,MJ, Stevens,RD, Lien,LF, Haqq,AM, Shah,SH, Arlotto,M, Slen-tz,CA, Rochon,J, Gallup,D, Ilkayeva,O, Wenner,BR, Yancy,J, Eisenson,H, Musante,G, Surwit,RS, Milling-ton,DS, Butler,MD, Svetkey,LP: A Branched-Chain Amino Acid-Related Metabolic Signature that Differentiates Obese and Lean Humans and Contributes to Insulin Resistance. Cell Metabolism 9:311-326, 2009

55. Tamamogullari,N, Silig,Y, Icagasioglu,S, Atalay,A: Carnitine Deficiency in Diabetes Mellitus Complica-tions. Journal of Diabetes and its Complications 13:251-253, 2009

56. Poorabbas,A, Fallah,F, Bagdadchi,J, Mahdavi,R, Aliasgarzadeh,A, Asadi,Y, Koushavar,H, Vahed Jab-bari,M: Determination of free L-carnitine levels in type II diabetic women with and without complica-tions. Eur J Clin Nutr 61:892-895, 2007

57. Stephens,FB, Constantin-Teodosiu,D, Laithwaite,D, Simpson,EJ, Greenhaff,PL: A threshold exists for the stimulatory effect of insulin on plasma L-carnitine clearance in humans. Am J Physiol Endocrinol Metab 292:E637-E641, 2007

58. Stephens,FB, Constantin-Teodosiu,D, Laithwaite,D, Simpson,EJ, Greenhaff,PL: An acute increase in skeletal muscle carnitine content alters fuel metabolism in resting human skeletal muscle. J Clin En-docrinol Metab 91:5013-5018, 2006

59. Stephens,FB, Constantin-Teodosiu,D, Laithwaite,D, Simpson,EJ, Greenhaff,PL: Insulin stimulates L-carnitine accumulation in human skeletal muscle. FASEB J 20:377-379, 2006

60. Noland,RC, Koves,TR, Seiler,SE, Lum,H, Lust,RM, Ilkayeva,O, Stevens,RD, Hegardt,FG, Muoio,DM: Carni-tine insufficiency caused by aging and overnutrition compromises mitochondrial performance and metabolic control. J Biol Chem 284:22840-22852, 2009

61. van den Broek,NMA, Ciapaite,J, De Feyter,HMML, Houten,SM, Wanders,RJA, Jeneson,JAL, Nicolay,K, Prompers,JJ: Increased mitochondrial content rescues in vivo muscle oxidative capacity in long-term high-fat-diet-fed rats. The FASEB Journal 24:1354-1364, 2010

62. Power,R, Hulver,M, Zhang,J, Dubois,J, Marchand,R, Ilkayeva,O, Muoio,D, Mynatt,R: Carnitine revisited: potential use as adjunctive treatment in diabetes. Diabetologia 50:824-832, 2007

63. Kim,JY, Hickner,RC, Cortright,RL, Dohm,GL, Houmard,JA: Lipid oxidation is reduced in obese human skeletal muscle. Am J Physiol Endocrinol Metab 279:E1039-E1044, 2000

64. Timmers,S, Nabben,M, Bosma,M, van Bree,B, Lenaers,E, Van Beurden,D, Schaart,G, Westerterp-Plan-

ACYLCARNITINES: REFLECTING OR INFLICTING INSULIN RESISTANCE?

K007_000000_HereThere and EveryWhere boekje.indd 27 09-02-15 08:30

Page 32: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

28

tenga,MS, Langhans,W, Hesselink,MKC, Schrauwen-Hinderling,VB, Schrauwen,P: Augmenting muscle diacylglycerol and triacylglycerol content by blocking fatty acid oxidation does not impede insulin sensitivity. Proceedings of the National Academy of Sciences 2012

65. Dobbins,RL, Szczepaniak,LS, Bentley,B, Esser,V, Myhill,J, McGarry,JD: Prolonged inhibition of muscle carnitine palmitoyltransferase-1 promotes intramyocellular lipid accumulation and insulin resistance in rats. Diabetes 50:123-130, 2001

66. Bruce,CR, Thrush,AB, Mertz,VA, Bezaire,V, Chabowski,A, Heigenhauser,GJ, Dyck,DJ: Endurance train-ing in obese humans improves glucose tolerance and mitochondrial fatty acid oxidation and alters muscle lipid content. Am J Physiol Endocrinol Metab 291:E99-E107, 2006

67. Meex,RCR, Schrauwen-Hinderling,VB, Moonen-Kornips,E, Schaart,G, Mensink,M, Phielix,E, van de Weijer,T, Sels,JP, Schrauwen,P, Hesselink,MKC: Restoration of Muscle Mitochondrial Function and Met-abolic Flexibility in Type 2 Diabetes by Exercise Training Is Paralleled by Increased Myocellular Fat Storage and Improved Insulin Sensitivity. Diabetes 59:572-579, 2010

68. Muoio,DM, Noland,RC, Kovalik,JP, Seiler,SE, Davies,MN, DeBalsi,KL, Ilkayeva,OR, Stevens,RD, Kheter-pal,I, Zhang,J, Covington,JD, Bajpeyi,S, Ravussin,E, Kraus,W, Koves,TR, Mynatt,RL: Muscle-Specific De-letion of Carnitine Acetyltransferase Compromises Glucose Tolerance and Metabolic Flexibility. Cell Metabolism 15:764-777, 2012

69. Tsintzas,K, Chokkalingam,K, Jewell,K, Norton,L, Macdonald,IA, Constantin-Teodosiu,D: Elevated Free Fatty Acids Attenuate the Insulin-Induced Suppression of PDK4 Gene Expression in Human Skeletal Muscle: Potential Role of Intramuscular Long-Chain Acyl-Coenzyme A. Journal of Clinical Endocrinol-ogy & Metabolism 92:3967-3972, 2007

70. Adams,SH, Hoppel,CL, Lok,KH, Zhao,L, Wong,SW, Minkler,PE, Hwang,DH, Newman,JW, Garvey,WT: Plasma Acylcarnitine Profiles Suggest Incomplete Long-Chain Fatty Acid beta-Oxidation and Altered Tricarboxylic Acid Cycle Activity in Type 2 Diabetic African-American Women. The Journal of Nutrition 139:1073-1081, 2009

71. Ebeling,P, Tuominen,JA, Arenas,J, Garcia Benayas,C, Koivisto,VA: The association of acetyl-L-carnitine with glucose and lipid metabolism in human muscle in vivo: the effect of hyperinsulinemia. Metabolism 46:1454-1457, 1997

72. Ramos-Roman,MA, Sweetman,L, Valdez,MJ, Parks,EJ: Postprandial changes in plasma acylcarnitine concentrations as markers of fatty acid flux in overweight and obesity. Metabolism 61:202-212, 2012

73. Kerbey AL, Randle PJ, Cooper RH, Whitehouse S, Pask HT, Denton RM: Regulation of pyruvate dehydro-genase in rat heart. Mechanism of regulation of proportions of dephosphorylated and phosphorylated enzyme by oxidation of fatty acids and ketone bodies and of effects of diabetes: role of coenzyme A, acetyl-coenzyme A and reduced and oxidized nicotinamide-adenine dinucleotide. Biochem J 154:327-348, 1976

74. Fiehn,O, Garvey,WT, Newman,JW, Lok,KH, Hoppel,CL, Adams,SH: Plasma Metabolomic Profiles Reflec-tive of Glucose Homeostasis in Non-Diabetic and Type 2 Diabetic Obese African-American Women. PLoS One 5:e15234, 2010

75. Wang,TJ, Larson,MG, Vasan,RS, Cheng,S, Rhee,EP, McCabe,E, Lewis,GD, Fox,CS, Jacques,PF, Fernan-dez,C, O’Donnell,CJ, Carr,SA, Mootha,VK, Florez,JC, Souza,A, Melander,O, Clish,CB, Gerszten,RE: Me-tabolite profiles and the risk of developing diabetes. Nat Med 17:448-453, 2011

76. Ho,JK, Duclos,RI, Hamilton,JA: Interactions of acyl carnitines with model membranes. Journal of Lipid Research 43:1429-1439, 2002

77. Ren,J, Lakoski,S, Haller,RG, Sherry,AD, Malloy,CR: Dynamic monitoring of carnitine and acetylcarnitine in the trimethylamine signal after exercise in human skeletal muscle by 7T 1H-MRS. Magn Reson Med-n/a, 2012

ACYLCARNITINES: REFLECTING OR INFLICTING INSULIN RESISTANCE?

K007_000000_HereThere and EveryWhere boekje.indd 28 09-02-15 08:30

Page 33: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

K007_000000_HereThere and EveryWhere boekje.indd 29 09-02-15 08:30

Page 34: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

C 3Plasma acylcarnitines inadequately

reflect tissue acylcarnitine metabolism

Marieke G. Schooneman, Niki Achterkamp, Carmen A. Argmann, Maarten R. Soeters, Sander M. Houten

BBA Molecular and Cell Biology of Lipids (2014) 1841, 987-994

K007_000000_HereThere and EveryWhere boekje.indd 30 09-02-15 08:30

Page 35: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

C 3Plasma acylcarnitines inadequately

reflect tissue acylcarnitine metabolism

Marieke G. Schooneman, Niki Achterkamp, Carmen A. Argmann, Maarten R. Soeters, Sander M. Houten

BBA Molecular and Cell Biology of Lipids (2014) 1841, 987-994

K007_000000_HereThere and EveryWhere boekje.indd 31 09-02-15 08:30

Page 36: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

K007_000000_HereThere and EveryWhere boekje.indd 32 09-02-15 08:30

Page 37: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

33

Abstract

Acylcarnitines have been linked to obesity-induced insulin resistance. However the ma-jority of these studies have focused on acylcarnitines in plasma. It is currently unclear to what extent plasma levels of acylcarnitines reflect tissue acylcarnitine metabolism. We investigated the correlation of plasma acylcarnitine levels with selected tissue acylcar-nitines as measured with tandem mass spectrometry, in both fed and fasted BALB/cJ (BALB) and C57BL/6N (Bl6) mice. Fasting affected acylcarnitine levels in all tissues. These changes varied substantially between the different tissue compartments. No significant correlations were found between plasma acylcarnitine species and their tissue counter-parts in both mouse strains, with the exception of plasma C4OH-carnitine in BALB mice. We suggest that this lack of correlation is due to differences in acylcarnitine turnover rates between plasma and tissue compartments and the fact that the plasma acylcar-nitine profile is a composition of acylcarnitines derived from different compartments. Therefore, plasma acylcarnitine levels do not reflect tissue levels and should be interpret-ed with caution. A focus on tissue acylcarnitine levels is warranted in metabolic studies.

Introduction

The western lifestyle and concomitant obesity epidemic are the main causes of the in-creasing prevalence of insulin resistance, a condition characterized by decreased insulin sensitivity of tissues such as in liver and muscle. In obesity and overfeeding, high levels of circulating lipids can lead to lipid storage in non-adipose tissue [22]. This ectopic fat deposition can cause disturbances in lipid metabolism which may lead to insulin resis-tance, a concept referred to as lipotoxicity [10,11,22]. Several lipid intermediates like fatty acyl-CoAs, diacylglycerol (DAG), ceramides, gangliosides and free fatty acids (FFA) have been implicated in the development of insulin resistance [20,28]. Acylcarnitines have been suggested to result from incomplete fatty acid oxidation (FAO) and were also pro-posed to induce insulin resistance [1,12]. Acylcarnitines comprise of an acyl group esterified to L-carnitine, which enables them to cross the mitochondrial membrane. Given the abundance of different acyl groups, the resulting acylcarnitine profile is extensive. Most acyl groups are derived from FAO, but they can also originate from amino acid and glucose metabolism. L-carnitine is mainly absorbed from the diet, but can also be formed through biosynthesis in human liver, kid-ney and brain. Other tissues must therefore acquire carnitine from the circulation [37]. The plasma acylcarnitine profile is directly influenced by diet and metabolic status such as fasting, which is associated with altered FAO flux [7,15,17,30]. Acylcarnitine profiling has been and remains the golden standard in diagnosing many different inborn errors of metabolism including FAO disorders [5].Several recent studies show increased levels of multiple acylcarnitines in obese, insulin resistant subjects [11,17,20]. This has led to the hypothesis that accumulating acylcarni-tines can interfere with insulin signalling. Long-chain acylcarnitines [20,26,27], ketone body derived C4OH-carnitine [1,20,31] and branched-chain amino acid (BCAA) derived

PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 33 09-02-15 08:30

Page 38: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

34

species C3- and C5-carnitine [24,39] have been suggested to induce insulin resistance. In some of these studies, the investigators measured both in plasma and muscle tis-sue, but the majority focused only on plasma measurements of acylcarnitines and cor-relations between plasma and tissue were not investigated. Insulin resistance, however, occurs on a cellular level of the different organ and tissue compartments, and these compartments may play distinct roles in the development of whole body insulin resis-tance. Therefore it is important to understand if and how the carnitine and acylcarnitine pool in tissues is represented in the plasma compartment, and whether plasma acylcar-nitine profiles accurately reflect acylcarnitine profiles in any of the tissues implicated in insulin resistance.In a previous study, we showed that changes in plasma acylcarnitine levels upon fasting in lean healthy men did not reflect changes in acylcarnitine levels in muscle tissue [30]. The aim of this study was to investigate if and in what way acylcarnitines in plasma re-flect acylcarnitine profiles in mouse liver, muscle, heart, white adipose tissue (WAT) and brown adipose tissue (BAT). Therefore we studied the correlation of plasma acylcarni-tines with the acylcarnitines in these tissues, in both fed and fasted Balb/cJ (BALB) mice and a more insulin resistant model, C57BL/6N (Bl6) mice. We show that plasma acylcar-nitine levels poorly reflect tissue acylcarnitine levels.

Methods

Animal studiesMice were approximately 10 weeks of age at the time of the experiment. BALB mice were acquired from Harlan Laboratories and Bl6 mice were acquired from Charles River Lab-oratories. Mice were housed under standard conditions and fed a chow diet. After one week of acclimatization both the BALB and Bl6 groups (22 mice per strain) were split in two, of which one group was fed ad libitum whereas the other half was fasted overnight for approximately 17 to 19 hours to increase FAO and induce insulin resistance. Between 9 AM and 11 AM on the study day, all animals were anesthetized with pentobarbital (100 mg/kg i.p.). In the subsequent dissection, venous blood sampling was performed by cannu-lation of the caval vein. Liver, soleus muscle, gastrocnemius muscle, quadriceps femoris muscle, heart, gonadal white adipose tissue (WAT) and interscapular brown adipose tis-sue (BAT) were dissected and frozen in liquid nitrogen, later to be stored at -80°C. All experiments were approved by the institutional review board for animal experiments at the Academic Medical Center (Amsterdam, The Netherlands).

Laboratory analysesFor plasma acylcarnitine analysis, 25 μl of plasma was mixed with 50 μl of internal standard mixture (25 μl of 5 μM [3,3,3-2H3]C3-carnitine and 2 μM [6,6,6-2H3]C6-, [8,8,8 -2H3]C8-, [10,10,10-2H3]C10- and [16,16,16-2H3]C16-carnitine in acetonitril (ACN), and 25 μl of 26 μM [methyl-2H3]-L-carnitine in 10% ACN) [38]. The plasma samples were depro-teinized by addition of 250 μl ACN and subsequent vortex mixing. Next, samples were

PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 34 09-02-15 08:30

Page 39: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

35

centrifuged for 10 minutes at 4°C at a speed of 20.000 g. The supernatant was trans-ferred into 4 ml glass vials and evaporated under a stream of nitrogen at 40°C. After evaporation, 100 μl butylation reagent (4:1 mixture of 1-butanol and acetylchloride) was added and incubated for 15 minutes at 60°C. Again evaporation was performed at 40°C. The residue was dissolved in 100 μl ACN, vortex mixed and transferred to Gilson vials for tandem mass spectrometric analysis (Waters/Micromass Quattro Premier XE). Tissues were freeze-dried overnight and weighed afterwards. ACN (800 μl) and 50 μl of internal standard mixture (25 μl of 5 μM [3,3,3-2H3]C3-carnitine and 2 μM [6,6,6-2H3]C6-, [8,8,8-2H3]C8-, [10,10,10-2H3]C10- and [16,16,16-2H3]C16-carnitine in ACN, and 25 μl of 325 μM [methyl-2H3]-L-carnitine in 20% ACN) were added to the tissue [35]. The sample was homogenized by shaking twice with a 4mm metal ball using a TissueLyser II (Qiagen) for 30 seconds at frequency of 30/second. Samples were centrifuged for 10 minutes at 4°C at a speed of 20.000 g. The supernatant was transferred into 4 ml glass vials and evaporated under a stream of nitrogen at 40°C. After evaporation, 100 μl pro-pylation reagent (4:1 mixture of 1-propanol and acetylchloride) was added to the residue, vortex mixed and incubated for 15 minutes at 60°C. The derivatization reagent was evap-orated under nitrogen and the residue was dissolved in 100 μl ACN, vortex mixed and transferred to Gilson vials for tandem mass spectrometric analysis. To detect the acylcarnitines the scan range for butylated samples was 215-515 (m/z), and for propylated samples 200-750 (m/z). The common daughter ion of 85 was detected, which results in a spectrum of parent ions corresponding to (M+H)+. The area under each acylcarnitine peak (AAC) and that under the IS (AIS) was quantified using MassLynx 4.1. The ratio AAC /AIS was determined and multiplied by the amount of added internal standard to carry out a semi-quantitative analysis of acylcarnitines and hydroxyacylcarnitines. The results of tissue were normalized for dry tissue mass to compare individual samples.

Statistical analysisWe specifically analysed acylcarnitine species which we considered quantitatively, dietary or metabolically relevant. We therefore chose to analyze several short-chain species such as C0-, C2- and C4-, which reflect the free carnitine pool and breakdown products from FAO, glucose and amino acid metabolism. We also analyzed C4OH-car-nitine which in human muscle is mainly a ketone body derived acylcarnitine (D-stereo isomer of hydroxybutyrylcarnitine) [31] and several long-chain species of which C16- and C18:1-carnitine are derivatives of palmitate and oleate. Also, we analyzed C3- and C5-carnitine. C3-carnitine, or propionylcarnitine, can be derived from the BCAAs valine or isoleucine. C5-carnitine (isovalerylcarnitine or 2-methylbutyrylcarnitine) can be de-rived from the BCAAs leucine or isoleucine [39]. The data for each individual mouse are available as supplemental table 1 (Table S1).Acylcarnitine measurements were processed with Masslynx software version 4.1, and further analysed using Microsoft Office Excel 2003. MetaboAnalyst 2.0 was used for hierarchical clustering and heatmap generation [40,41]. We used the complete clustering algorithm with the Spearman correlations coefficient as the distance measure. Data were processed with range scaling methods (mean-centred and divided by the range of each

PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 35 09-02-15 08:30

Page 40: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

36

variable). Final statistical analysis was performed using SPSS statistical software program version 20.0. Data were evaluated by statistical correlations using bivariate Spearman’s correlation coefficients (two-tailed) and with independent T-tests. Data were consid-ered significant with a P value <0.05 (marked as *) or <0.01 (marked as **).For each of the four datasets (Bl6 or BALB, fasted or fed), we constructed an unweighted signed metabolite coexpression network using the approximately 15 unique metabolites measured across 8 different tissues. We followed the R tutorial as outlined previously [13,43]. In brief, in an unweighted signed coexpression network, nodes represent me-tabolites and the nodes are connected if the corresponding metabolites are positively co-expressed across the tissue samples according to a hard cutoff (positive spearman’s rank correlation coefficient > 0.75). This conservative threshold was determined based on the fact that the majority of the metabolite pair-wise correlations were highly signifi-cant at this cutoff. While coexpression networks can often use the absolute value of the correlation coefficient as an unsigned co-expression similarity measure, we employed a signed co-expression measure (sij) between metabolite expression profiles xi and xj, for which we used a simple transformation of the correlation: sij = (1+cor(xixj)/2). For the unweighted networks, we then generated an adjacency matrix, which contains binary information with entries of 1 or 0 depending on whether or not 2 nodes are adja-cent (connected) to one another. To group metabolites with coherent expression profiles into modules, we used average linkage hierarchical clustering, which uses the topological overlap measure (TOM) as dissimilarity. The topological overlap of two nodes reflects their similarity in terms of the commonality of the nodes they connect to [42]. A height cutoff value of 0.96 was chosen in the dendrogram so that the resulting branches corre-sponded to discrete diagonal blocks (modules) in the TOM plot with a minimum module size of 3 metabolites.To further reduce the dimensionality of the dataset, we summarized the expression within a single module to a single module representative called the module ‘eigen-metabolite’, which is essentially the mathematical equivalent to the first principal component. We correlated the module eigen-metabolite measures within a coexpression network to one another (spearman rank correlation) and displayed them in a heat map plot. All graphical plots were generated using the WGCNA [18,42] and the heatmap package in R [16].

Results

General characteristics of the fasting response in BALB and Bl6 miceUpon overnight fasting, BALB mice lost 12% of their body weight (25.8 grams ± 1.5 pre- and 22.5 ± 1.6 post-fasting, P < 0.001), whereas Bl6 mice lost 16% body weight (25.7 grams ± 1.0 pre- and 21.7 ± 0.9 post-fasting, P < 0.001). Fasting significantly induced ketosis in BALB (fasted beta-OH-butyrate 1.77 mmol/L ± 0.90 vs fed 0.12 mmol/L ± 0.05, p < 0.001) and Bl6 mice (fasted 1.06 mmol/L ± 0.36 vs fed 0.07 mmol/L ± 0.03, p < 0.001). Fasting ke-tosis was significantly higher in BALB compared to Bl6 mice (p < 0.05). Fasting increased plasma FFAs 1.3-fold in BALB mice (0.70 mmol/L ± 0.18 pre- and 0.88 mmol/L ± 0.18

PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 36 09-02-15 08:30

Page 41: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

37

post-fasting, P < 0.05) and 1.5-fold in Bl6 mice (0.46 mmol/L ± 0.07 pre- and 0.71 mmol/L ± 0.24 post-fasting, P < 0.01).

Acylcarnitine profiles in fed and fasted mice We used the acylcarnitine dataset of the Bl6 and BALB mice for two-dimensional hier-archical clustering (Figures 1 and S1). In one dimension, this analysis clustered the mice according to the fed or fasted condition. In the other dimension, clustering revealed clusters of acylcarnitines responding similarly to fasting. Overall, the levels of most acyl-carnitine species increased upon fasting regardless of tissue. This result illustrates that fasting is an excellent intervention to modulate acylcarnitine metabolism.

Acylcarnitine profiles in plasmaWhen compared to the fed condition, fasted Bl6 and BALB mice showed lower plas-ma levels of free carnitine, whereas the levels of most other acylcarnitine species (ace-tylcarnitine, C4-, C4OH-, C12-, C14:1-, C16- and C18:1-carnitine) increased significantly upon fasting (Figure 2, Table S1). The levels of C3- and C5-carnitine, two BCAA derived acylcarnitine species, responded differently to fasting in Bl6 and BALB mice. In BALBmice, C3-carnitine increased and C5-carnitine decreased in fasted compared to fed mice. In Bl6 mice, C3-carnitine did not change upon fasting, whereas C5-carnitine levels increased.

PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 37 09-02-15 08:30

Page 42: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

38 PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

Figure 2 A and B Fasting-induced changes in Bl6 mice in plasma and tissue acylcarnitine levels of a. C0-carnitine, b. C2-

carnitine, c. C4OH-carnitine and d. C16-carnitine. (* p < 0.05, ** p < 0.01, *** p < 0.001, ns not significant)

K007_000000_HereThere and EveryWhere boekje.indd 38 09-02-15 08:30

Page 43: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

39PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

Figure 1 Unsupervised hierarchical clustering of acylcarnitine levels of fed and fasted Bl6 mice. The colors in the heatmap

reflect the acylcarnitine abundance (mean-centred and divided by the range of each variable).

K007_000000_HereThere and EveryWhere boekje.indd 39 09-02-15 09:13

Page 44: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

40 PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

Figure 4 A network of tissue and plasma acyl carnitines Topological overlap measure plots for fed Bl6 (a) and fasted Bl6 mice

(b). The network analysis reveals distinct modules of connected acylcarnitines, which are indicated by color coding. Quad

denotes quadriceps muscle; Gastroc denotes gastrocnemius muscle; SCAC denotes shortchain acylcarnitines; LCAC denotes

longchain acylcarnitines. The Spearman correlation coefficient between the module eigenvectors are displayed in a heatmap.

Bl6 Fed

Green Blue Turquoise Grey Brown YellowGreenBlueTurquoiseGreyBrownYellow

*

*

Soleus/Gastroc/Quad LCAC

Plasma LCACSoleus SCAC

SCAC

Liver SCAC, LCACGastroc/WAT/BAT C0

Liver/Heart/WAT/BAT LCACBAT SCAC

Spearman Rank Correlation

-1 -0.5 0 0.5 1

* P value <0.05

Bl6 Fasted

Red Yellow Blue Green Black Grey BrownTurquoiseRedYellowBlueGreenBlackGreyBrownTurquoise

**

Liver LCAC

Plasma LCAC

WAT/BAT LCAC, SCACLiver/Quad/WAT/BAT C0

Plasma/Liver/Muscle SCAC

Soleus LCAC

Gastroc/Quad LCAC

Heart LCAC

K007_000000_HereThere and EveryWhere boekje.indd 40 09-02-15 09:13

Page 45: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

41

Supplemental data - part 1

Figure S1 Unsupervised hierarchical clustering of acylcarnitine levels. Heatmap of fed and fasted BALB mice. The colors in

the heatmap reflect the acylcarnitine abundance (mean-centred and divided by the range of each variable).

PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 41 09-02-15 09:13

Page 46: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

42

Figure S2 A network of tissue and plasma acylcarnitines Topological overlap measure plots for fed BALB (a) and fasted BALB mice (b). The network analysis reveals distinct modules of

connected acylcarnitines, which are indicated by color coding. Quad denotes quadriceps muscle; Gastroc denotes gastroc-

nemius muscle; SCAC denotes shortchain acylcarnitines; LCAC denotes long-chain acylcarnitines. The Spearman correlation

coefficient between the module eigenvectors are displayed in a heatmap.

Figure S2b: fasted BALB mice

Figure S2a: fed BALB mice

BALB Fed

Blue Grey Brown Green TurquoiseYellowBlueGreyBrownGreenTurquoiseYellow

*

Plasma/Liver LCAC

Soleus/Gastroc/Quad/Heart/BAT LCACSeveral organs SCAC

Soleus/Gastroc/Quad C0

WAT LCACSeveral organs SCAC

BAT SCAC

Spearman Rank Correlation

-1 -0.5 0 0.5 1

* P value <0.05

BALB Fasted

BlueTurquoiseGreenYellowBrownGrey

Blue Turquoise Green Yellow Brown Grey**

Plasma/Quad LCACPlasma/Several organs C4OH

Plasma/BAT LCACSeveral organs SCAC

Liver/Soleus/BAT C0

Soleus LCAC

Liver/WAT LCACGastroc/Quad/WAT C0Several organs SCAC

Gastroc/Quad/Heart LCACPlasma SCAC

PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 42 09-02-15 09:13

Page 47: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

43

Acylcarnitine profiles in liverIn liver, fasting levels of free carnitine were higher compared to fed levels in both BALB and Bl6 mice, whereas acetylcarnitine levels were lower in fasted liver of BALB and Bl6 mice (Figure 2, Table S1). C3-carnitine and C4OH-carnitine did not differ between fed and fasted mice. C5-carnitine levels decreased only in fasted BALB mice. Lastly, C16- and C18:1-carnitine were higher in fasted animals of both strains.

PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

Figure 2 C and D Fasting-induced changes in Bl6 mice in plasma and tissue acylcarnitine levels of a. C0-carnitine, b. C2-

carnitine, c. C4OH-carnitine and d. C16-carnitine. (* p < 0.05, ** p < 0.01, *** p < 0.001, ns not significant)

K007_000000_HereThere and EveryWhere boekje.indd 43 09-02-15 08:30

Page 48: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

44

Acylcarnitine profiles in skeletal and cardiac muscleIn muscle tissue (m. soleus, m. gastrocnemius and m. quadriceps femoris) free carnitine was lower when fasted (Figure 2, Table S1). However, levels of long-chain acylcarnitines did not change upon fasting. Acetylcarnitine levels differed between the three muscle types, reflecting the oxidative capacity of the muscles. Acetylcarnitine levels in soleus muscle, which can be considered as the most oxidative muscle tissue [8], were twice as high as in gastrocnemius and quadriceps muscle in the fed state. Whereas acetylcarni-tine levels increased in soleus muscle upon fasting, they decreased in quadriceps muscle and did not change in gastrocnemius muscle. C4OH-carnitine was higher in fasted ani-mals of both mouse strains, which is probably caused by the fasting induced ketosis, as was demonstrated for human muscle [31]. Acylcarnitines in heart did not show any differ-ences between fasted and fed BALB animals, with one exception being C4OH-carnitine, which was higher in the fasted group. In heart tissue of Bl6 mice, free carnitine was lower and all long chain species were higher compared to fed mice. Levels of BCAA derived species C3- and C5-carnitine did not significantly differ in muscle and heart tissue of fed and fasted BALB mice, whereas in fasted Bl6 mice both these acylcarnitine species were significantly higher in muscle and heart tissue when compared to fed Bl6 mice.

Acylcarnitine profiles in white and brown adipose tissueFree carnitine and all other examined acylcarnitine species in WAT and BAT were high-er in fasted BALB and Bl6 mice (Figure 2, Table S1). BAT showed the most pronounced response with a 5.5-fold increase in the level of acetylcarnitine and a 3.5- to 5-fold in-crease in the level of C16- and C18:1-carnitine.

Correlations between compartments

Correlations between plasma and tissue acylcarnitine levelsWe calculated individual correlation coefficients between plasma and tissue acylcar-nitines. In general, we found no significant correlations between plasma acylcarnitine species and their tissue counterparts in either mouse strain, which is illustrated by the correlation of plasma C0-, C2-, C4OH- and C16- with their counterparts in liver (Figure 3a) and soleus muscle (Figure 3b). Plasma free carnitine showed no significant correla-tions with free carnitine in any of the tissues. None of the short chain species showed significant correlations, with the exception of plasma C4OH-carnitine, which in fed Bl6 mice correlated with quadriceps femoris muscle (r2 0.622, p 0.031)(Table S2a). This cor-relation was even more pronounced in fasted BALB mice (Table S2b), which exhibited higher ketosis compared to Bl6 mice, with plasma C4OH-carnitine correlating with all tested muscle compartments (soleus muscle (r2 0.624, p 0.040), gastrocnemius mus-cle (r2 0.791, p 0.004) and quadriceps femoris muscle (r2 0.627, p 0.039)) and heart (r2 0.673, p 0.023). Interestingly plasma C4OH-carnitine correlated negatively with BAT (r2 -0.664, p 0.026). Lastly, none of the plasma long-chain acylcarnitine species correlated with their tissue counterparts in any metabolic state in both BALB and Bl6 mice.

PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 44 09-02-15 08:30

Page 49: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

45

Figure 3a

Figure 3b

Figure 3 Correlations of C0-, C2-, C4OH- and C16-carnitine in fasted Bl6 mice between a. plasma vs liver, and b. plasma vs

soleus muscle.

PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 45 09-02-15 08:30

Page 50: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

46

A network of tissue and plasma acylcarnitinesNetwork approaches attempt to syndicate large-scale datasets such as transcriptomics into informative relationships that otherwise are not readily apparent given the size of the dataset. In order to reveal these relationships in our acylcarnitine dataset, we con-structed unweighted signed metabolite coexpression networks for Bl6 and BALB in the fed and fasted condition. The networks are displayed as a plot of the topological overlap measure (TOM) that considers the pairwise correlation of metabolites in relation to other metabolites in the network. In all four conditions, our network analyses revealed distinct modules of connected acylcarnitines, which are indicated by color coding (Figure 4a, b and S2a, b). These modules differ between the fed and fasted condition, but also be-tween the Bl6 and BALB strain. In general the long-chain acylcarnitines grouped together according to the tissue they were analyzed in. For example, the long-chain acylcarnitines in most muscle tissues were connected in fed as well as fasted conditions, with the so-leus often forming a separate module. Indeed there were significant correlations for C16- and C18-carnitine in the examined muscles (table S3a, b), suggesting a coordinated long-chain acylcarnitine metabolism in striated muscle tissues. Short-chain acylcarni-tines grouped not only according to tissue, but also according to metabolite class.The variation within one metabolite module can be represented by a principal compo-nent, or module eigen-metabolite. This enables the analysis of the relations between the metabolite modules. The correlation coefficients of the metabolite modules in each TOM plot are displayed as a heatmap (Figure 4a, b and S2a, b). In fasted Bl6 mice, this analysis revealed a significant negative correlation between the blue module comprising the long-chain acylcarnitine species in quadriceps and gastrocnemius muscle, and the brown module, which represents the long-chain-acylcarnitines in liver.

PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 46 09-02-15 08:30

Page 51: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

47PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

Discussion

We analysed plasma acylcarnitine profiles in relation to their counterpart acylcarnitine profiles in tissue of both fed and fasted Bl6 and BALB mice. Despite the fact that acylcar-nitine levels differed between the fed and fasted state, no correlations could be estab-lished between plasma acylcarnitines and their tissue counterparts.We have studied both BALB and Bl6 mouse strains for their differences in insulin sen-sitivity. Bl6 mice are a generally preferred strain for metabolic studies, as they develop more severe insulin resistance upon high fat diet interventions [6], opposed to BALB mice [21]. In our study, both strains had comparable changes in acylcarnitine profiles under fed and fasted conditions. During fasting, glucose oxidation decreases and the organism enters a predominant lipid mobilizing (i.e. lipolysis) and oxidizing (i.e. whole body lipid oxidation and hepatic ketogenesis) state [32]. Our data showed organ specific changes in acylcarnitines upon a fasting intervention. In plasma, lower fasting free car-nitine levels were accompanied by a rise of mainly short-chain acylcarnitine species. A general shift from free to acylated carnitine is likely to explain this observation although the overall increase in acylcarnitine levels did not equal the decrease of free carnitine. Uptake of free carnitine by tissues is most likely another contributor to this decrease.In contrast to plasma, fasting increased hepatic free carnitine levels, which may reflect de novo synthesis by the liver. Here, fasting-induced peroxisome proliferator-activated receptor-alpha (PPAR-alpha) activation stimulates hepatic carnitine synthesis and up-take from plasma via induction of the Bbox1 and Slc22a5 gene, respectively [36]. Fast-ing lowered hepatic acetylcarnitine levels, which, combined with higher fasting plasma acetylcarnitine levels, suggests increased liver acetylcarnitine efflux. Acetylcarnitine was the most abundant of acylated carnitine species under both fed and fasted conditions. It can be speculated that acetylcarnitine serves to disseminate energy (via acetyl-CoA) or alternatively transports newly synthesized carnitine to other organs. Long-chain acylcarnitines all increased in liver upon fasting, and formed a highly con-nected metabolite module. As FFA increased upon fasting, this rise in hepatic long-chain acylcarnitines probably reflects the activation of these FFA and subsequently the con-version to an acylcarnitine for FAO and concomitant ketone body production in the liver.In muscle tissue, free carnitine was unchanged or tended to be slightly lower in fasted mice despite the changes in metabolic state, suggesting that supply of carnitine to the muscle compartment adequately responds to the demand in the fasted condition. We found that acetylcarnitine levels varied between different muscles, most likely reflecting muscle fiber type. Acetylcarnitine was highest in the fasted soleus muscle, which has predominantly type 1 oxidative muscle fibres and thus may reflect the high capacity for oxidizing lipids during fasting [25]. In contrast, acetylcarnitine levels in gastrocnemius and quadriceps muscle were unaffected or lower upon fasting. This might suggest lower FAO rates during fasting in these white and mixed muscle fibre types. Moreover high levels of acetylcarnitine and thus acetyl-CoA might inhibit the pyruvate dehydrogenase (PDH) complex and subsequent carbohydrate oxidation [23], underlining the potential importance of acetylcarnitine as metabolic regulator.

K007_000000_HereThere and EveryWhere boekje.indd 47 09-02-15 08:30

Page 52: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

48

In all muscle types C4OH-carnitine was strongly increased by fasting, being consistent with earlier studies showing a positive correlation between increased fasting levels of C4OH-carnitine in muscle tissue and plasma FFA levels in an insulin resistant state [1]. In human muscle, this acylcarnitine species has been shown to be mainly derived from the ketone body beta-hydroxybutyrate (D-3-hydroxybutyryl-carnitine/ketocarnitine) [31]. Although it has been suggested that skeletal muscle is capable of ketogenesis [1], the liver is most likely the predominant source of FAO-derived ketone body production and muscle C4OH-carnitine levels correlate significantly with the ketone body production rate in humans during fasting [31]. Long-chain acylcarnitines have been suggested to induce insulin resistance [20,22]. In this study, long-chain acylcarnitine levels in muscle were unchanged by fasting, with the exception of C14:1- and C18:1-carnitine which did increase upon fasting in several mus-cle types. Therefore the muscle compartment as a whole seems to maintain and war-rant stable acylcarnitine and free carnitine levels, reflecting an adaptive mechanism to prevent abrupt changes in energy production or CoA trapping as known from humans [30]. The long-chain acylcarnitines in muscle did form distinct modules, suggesting a coordinated long-chain acylcarnitine metabolism in striated muscle tissues. In fasted Bl6 mice, the module representing long-chain acylcarnitines in quadriceps and gastroc-nemius showed a strong negative correlation with liver long-chain acylcarnitines (Figure 4a). High long-chain acylcarnitines in liver could reflect higher hepatic FAO rates and thus higher production of ketone bodies, which on their turn suppress FAO in muscle, lowering muscle acylcarnitine levels.Heart acylcarnitine metabolism showed some resemblance with skeletal muscle. Chang-es upon fasting differed between BALB and Bl6 mice with the former showing no change except for C4OH-carnitine whereas the latter having lower free carnitine and higher lev-els of all other acylcarnitine species after fasting. As heart already relies on FAO for 70 to 80% of its energy requirements in the fed state [34], acylcarnitine metabolism is crucial for energy production in cardiac muscle. In absence of fatty acids, carnitine alone lowers acetyl-CoA/CoA ratios and thereby increases PDH activity and carbohydrate oxidation, whereas in presence of fatty acids it increases the acetyl-CoA/CoA ratio and thereby inhibits PDH activity in favor of FAO [4].The most consistent difference in acylcarnitine levels between the fed and fasted state was observed in WAT and BAT where all acylcarnitine species were significantly higher when fasted. Indeed, long- and short-chain acylcarnitine formed a distinct module in fasted Bl6 mice. We speculate that this increase in acylcarnitines reflects increased lip-olysis and as a consequence increased FAO rates. Indeed, stimulation of lipolysis in vis-ceral fat through the use of a PPAR-alpha agonist, increases FAO and energy expenditure in this type of adipose tissue [19]. Also treating cultured adipocytes with a specific isomer of conjugated linoleic acid [9], increases both lipolysis and FAO in these cells. Autophagy and accompanying lipolysis led to higher FAO rates in WAT [29]. All together these data may suggest that FAO is stimulated in WAT under lipolytic circumstances which may ex-plain the rise in acylcarnitines.

PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 48 09-02-15 08:30

Page 53: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

49PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

In contrast to WAT which functions mainly as a dynamic fat storage compartment and -supplier, BAT does not secrete FFA and has limited triglyceride storage capacity. FAO in BAT is used mainly for heat generation to maintain core body temperature [33]. This thermogenic function of BAT is essential in rodents and demands high energy supply to increase FAO rates in the abundant mitochondria [2]. Under fed conditions, BAT is capa-ble of oxidizing both glucose and fatty acids, but a high FAO capacity in BAT may explain the increased acylcarnitine levels in the fasted state.A potential limitation of our study is the effect of anesthesia or post-mortem tissue col-lection on acylcarnitine levels that could have masked significant correlations. Indeed, anesthesia using pentobarbital and post-mortem sampling collection have been report-ed to influence tissue and plasma acylcarnitine levels [3,14]. In our study all mice were comparable with each other, because they were anesthetized using a similar dose of pentobarbital. Blood withdrawal and organ collection were performed systematically, rapidly and in a specific order, equalizing the time lapse between blood and tissue har-vesting for all different tissues analyzed. Our data demonstrate fasting-induced changes in tissue and plasma acylcarnitine levels, illustrating that our method reliably detected physiological differences. Therefore potential correlation between plasma and tissue acylcarnitines should have been detected if present.In summary, we observed a lack of correlation between plasma acylcarnitines and the respective tissue acylcarnitine levels. This might be explained by the substantial differ-ences in the response of tissue acylcarnitines to fasting as discussed above. In addi-tion, the absence of correlation may be explained by a difference in the turnover rates of plasma acylcarnitines and tissue acylcarnitines. We speculate that the FAO induced change in individual acylcarnitines is considerably smaller in plasma when compared to tissue. Additionally the plasma acylcarnitine profile is also affected by the uptake of acyl-carnitines by tissues. This implies that the plasma acylcarnitine profile rather reflects a complex sum of past metabolic changes rather than the current metabolic status. Within this concept we hypothesize that plasma profiles are mainly composed of 1- the surplus of acylcarnitines which are cleared very effectively by muscle to prevent accumulation when FAO rates increase, 2- mobilized lipids from lipolysis in adipose, tissue in the form of acylcarnitines, 3- surplus and newly synthesized carnitine and acylcarnitines from the liver and 4- carnitine and acylcarnitines from other, probably smaller fat oxidizing compartments. In this study, we have shown that plasma acylcarnitine profiles did not correlate with tissue acylcarnitine profiles. However, acylcarnitine levels in different tissues did change under dietary (fasting) conditions and these changes were chain length and tissue de-pendent. Although fasting induced insulin resistance may differ from chronic diet-in-duced insulin resistance, these results have important consequences: since some studies showed associations of plasma acylcarnitines with various parameters of insulin sensitivity [11,17,20], the exact pathophysiological meaning of such correlations remains elusive. A focus on tissue acylcarnitines may be preferable in future studies.

K007_000000_HereThere and EveryWhere boekje.indd 49 09-02-15 08:30

Page 54: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

50

References1. An J., Muoio D.M., Shiota M., Fujimoto Y., Cline G.W., Shulman G.I., Koves T.R., Stevens R., Millington D., and

Newgard C.B., Hepatic expression of malonyl-CoA decarboxylase reverses muscle, liver and whole-ani-mal insulin resistance, Nat Med 10 (2004) pp. 268-274.

2. E. Arbuthnott, Brown adipose tissue: structure and function, Proceedings of the Nutrition Society 48 (1989) pp. 177-182.

3. E.P. Brass and Hoppel CL, In vivo studies of carnitine and fatty acid metabolism: Problems with use of anesthetics, Anal Biochem 110 (1981) pp. 77-81.

4. T.L. Broderick, H.A. Quinney, and G.D. Lopaschuk, Carnitine stimulation of glucose oxidation in the fatty acid perfused isolated working rat heart, Journal of Biological Chemistry 267 (1992) pp. 3758-3763.

5. D.H. Chace, T.A. Kalas, and E.W. Naylor, Use of Tandem Mass Spectrometry for Multianalyte Screening of Dried Blood Specimens from Newborns, Clin Chem 49 (2003) pp. 1797-1817.

6. S.M. Clee and A.D. Attie, The Genetic Landscape of Type 2 Diabetes in Mice, Endocrine Reviews 28 (2007) pp. 48-83.

7. Costa CG, De Almeida IT, and Jacobs C, Dynamic Changes of Plasma Acylcarnitine Levels Induced by Fasting and Sunflower Oil Challenge Test in Children, Pediatric Research 46 (1999) pp. 440-444.

8. M.T. Crow and M.J. Kushmerick, Chemical energetics of slow- and fast-twitch muscles of the mouse, The Journal of General Physiology 79 (1982) pp. 147-166.

9. L.J. den Hartigh, C.Y. Han, S. Wang, M. Omer, and A. Chait, 10E,12Z-conjugated linoleic acid impairs adi-pocyte triglyceride storage by enhancing fatty acid oxidation, lipolysis, and mitochondrial reactive oxy-gen species, Journal of Lipid Research (2013).

10. Z. Guo, Intramyocellular lipids: maker vs. marker of insulin resistance, Med Hypotheses 70 (2008) pp. 625-629.

11. W.L. Holland, T.A. Knotts, J.A. Chavez, L.P. Wang, K.L. Hoehn, and S.A. Summers, Lipid mediators of insulin resistance, Nutr Rev 65 (2007) p. S39-S46.

12. C.L. Hoppel and S.M. Genuth, Carnitine metabolism in normal-weight and obese human subjects during fasting, Am J Physiol 238 (1980) p. E409-E415.

13. S. Horvath, B. Zhang, M. Carlson, K.V. Lu, S. Zhu, R.M. Felciano, M.F. Laurance, W. Zhao, S. Qi, Z. Chen, Y. Lee, A.C. Scheck, L.M. Liau, H. Wu, D.H. Geschwind, P.G. Febbo, H.I. Kornblum, T.F. Cloughesy, S.F. Nelson, and P.S. Mischel, Analysis of Oncogenic Signaling Networks in Glioblastoma Identifies ASPM as a Molecular Target, Proceedings of the National Academy of Sciences of the United States of America 103 (2006) pp. 17402-17407.

14. Kerner J and L.L. Bieber, The effect of electrical stimulation, fasting and anesthesia on the carnitine(s) and acyl-carnitines of rat white and red skeletal muscle fibres, Comp Biochem Physiol B. 75 (1983) pp. 311-316.

15. C.L. Kien, K.I. Everingham, D. Stevens, N.K. Fukagawa, and D.M. Muoio, Short-Term Effects of Dietary Fatty Acids on Muscle Lipid Composition and Serum Acylcarnitine Profile in Human Subjects, Obesity 19 (2011) pp. 305-311.

16. Kolde R, A package for drawing pretty heatmaps in R, Pheatmap: Pretty Heatmaps (2013).17. T.R. Koves, J.R. Ussher, R.C. Noland, D. Slentz, M. Mosedale, O. Ilkayeva, J. Bain, R. Stevens, J.R. Dyck, C.B.

Newgard, G.D. Lopaschuk, and D.M. Muoio, Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance, Cell Metab 7 (2008) pp. 45-56.

18. Langfelder R and Horvath S, WGCNA: an R package for weighted correlation network analysis, BMC Bio-informatics 9 (2008).

19. M. Laplante, W.T. Festuccia, G. Soucy, Y. Gélinas, J. Lalonde, J.P. Berger, and Y. Deshaies, Mechanisms of the Depot Specificity of Peroxisome Proliferator Activated Receptor gamma Action on Adipose Tissue Metabolism, Diabetes 55 (2006) pp. 2771-2778.

20. S.J. Mihalik, B.H. Goodpaster, D.E. Kelley, D.H. Chace, J. Vockley, F.G. Toledo, and J.P. DeLany, Increased levels of plasma acylcarnitines in obesity and type 2 diabetes and identification of a marker of glucolipo-toxicity, Obesity (Silver Spring) 18 (2010) pp. 1695-1700.

21. M.K. Montgomery, N.L. Hallahan, S.H. Brown, M. Liu, T.W. Mitchell, G.J. Cooney, and N. Turner, Mouse strain-dependent variation in obesity and glucose homeostasis in response to high-fat feeding, Diabeto-logia 56 (2013) pp. 1129-1139.

22. D.M. Muoio and T.R. Koves, Lipid-induced metabolic dysfunction in skeletal muscle, Novartis Found Symp 286 (2007) pp. 24-38.

23. D.M. Muoio, R.C. Noland, J.P. Kovalik, S.E. Seiler, M.N. Davies, K.L. DeBalsi, O.R. Ilkayeva, R.D. Stevens, I.

PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 50 09-02-15 08:30

Page 55: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

51PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

Kheterpal, J. Zhang, J.D. Covington, S. Bajpeyi, E. Ravussin, W. Kraus, T.R. Koves, and R.L. Mynatt, Mus-cle-Specific Deletion of Carnitine Acetyltransferase Compromises Glucose Tolerance and Metabolic Flexibility, Cell Metabolism 15 (2012) pp. 764-777.

24. C.B. Newgard, J. An, J.R. Bain, M.J. Muehlbauer, R.D. Stevens, L.F. Lien, A.M. Haqq, S.H. Shah, M. Arlotto, C.A. Slentz, J. Rochon, D. Gallup, O. Ilkayeva, B.R. Wenner, J. Yancy, H. Eisenson, G. Musante, R.S. Surwit, D.S. Millington, M.D. Butler, and L.P. Svetkey, A Branched-Chain Amino Acid-Related Metabolic Signature that Differentiates Obese and Lean Humans and Contributes to Insulin Resistance, Cell Metabolism 9 (2009) pp. 311-326.

25. Picard M, Hepple RT, and Burelle Y, Mitochondrial functional specialization in glycolytic and oxidative muscle fibers: tailoring the organelle for optimal function, Am J Physiol Cell Physiol 302 (2012) pp. 629-641.

26. M.A. Ramos-Roman, L. Sweetman, M.J. Valdez, and E.J. Parks, Postprandial changes in plasma acylcarni-tine concentrations as markers of fatty acid flux in overweight and obesity, Metabolism 61 (2012) pp. 202-212.

27. L.M. Redman, K.M. Huffman, L.R. Landerman, C.F. Pieper, J.R. Bain, M.J. Muehlbauer, R.D. Stevens, B.R. Wenner, V.B. Kraus, C.B. Newgard, W.E. Kraus, and E. Ravussin, Effect of Caloric Restriction with and without Exercise on Metabolic Intermediates in Nonobese Men and Women, Journal of Clinical Endocri-nology & Metabolism 96 (2011) p. E312-E321.

28. M.G. Schooneman, F.M. Vaz, S.M. Houten, and M.R. Soeters, Acylcarnitines: Reflecting or Inflicting Insulin Resistance?, Diabetes 62 (2013) pp. 1-8.

29. R. Singh, Y. Xiang, Y. Wang, K. Baikati, A.M. Cuervo, Y.K. Luu, Y. Tang, J.E. Pessin, G.J. Schwartz, and M.J. Czaja, Autophagy regulates adipose mass and differentiation in mice, The Journal of Clinical Investigation 119 (2009) pp. 3329-3339.

30. M.R. Soeters, H.P. Sauerwein, M. Duran, R.J. Wanders, M.T. Ackermans, E. Fliers, S.M. Houten, and M.J. Serlie, Muscle acylcarnitines during short-term fasting in lean healthy men, Clin Sci (Lond) 116 (2009) pp. 585-592.

31. M.R. Soeters, M.J. Serlie, H.P. Sauerwein, M. Duran, J.P. Ruiter, W. Kulik, M.T. Ackermans, P.E. Minkler, C.L. Hoppel, R.J.A. Wanders, and S.M. Houten, Characterization of D-3-hydroxybutyrylcarnitine (ketocarni-tine): an identified ketosis-induced metabolite, Metabolism 61 (2012) pp. 966-973.

32. M.R. Soeters, P.B. Soeters, M.G. Schooneman, S.M. Houten, and J.A. Romijn, Adaptive reciprocity of lipid and glucose metabolism in human short-term starvation, American Journal of Physiology - Endocrinolo-gy And Metabolism 303 (2012) p. E1397-E1407.

33. B.M. Spiegelman and J.S. Flier, Obesity and the Regulation of Energy Balance, Cell 104 (2001) pp. 531-543.

34. H. Taegtmeyer, Energy metabolism of the heart: From basic concepts to clinical applications applica-tions, Current Problems in Cardiology 19 (1994) pp. 61-113.

35. T. Tyni, M. Pourfarzam, and D.M. Turnbull, Analysis of Mitochondrial Fatty Acid Oxidation Intermediates by Tandem Mass Spectrometry from Intact Mitochondria Prepared from Homogenates of Cultured Fibro-blasts, Skeletal Muscle Cells, and Fresh Muscle, Pediatr Res 52 (2002) pp. 64-70.

36. N. van Vlies, Ferdinandusse S, Wanders R J A, and Vaz F M, PPARalfa-activation results in enhanced car-nitine biosynthesis and OCTN2 expression, Biochim Biophys Acta 1767 (2007) p. 11.

37. F.M. Vaz and R.J.A. Wanders, Carnitine biosynthesis in mammals, Biochem. J. 361 (2002) pp. 417-429.38. Vreken P, van Lint AE, Bootsma AH, Overmars H, Wanders RJ, and van Gennip AH, Quantitative plasma

acylcarnitine analysis using electrospray tandem mass spectrometry for the diagnosis of organic acidae-mias and fatty acid oxidation defects., J Inherit Metab Dis 3 (1999) pp. 302-306.

39. T.J. Wang, M.G. Larson, R.S. Vasan, S. Cheng, E.P. Rhee, E. McCabe, G.D. Lewis, C.S. Fox, P.F. Jacques, C. Fernandez, C.J. O’Donnell, S.A. Carr, V.K. Mootha, J.C. Florez, A. Souza, O. Melander, C.B. Clish, and R.E. Gerszten, Metabolite profiles and the risk of developing diabetes, Nat Med 17 (2011) pp. 448-453.

40. J. Xia, R. Mandal, I.V. Sinelnikov, D. Broadhurst, and D.S. Wishart, MetaboAnalyst 2.0 -- a comprehensive server for metabolomic data analysis, Nucleic Acids Research (2012).

41. J. Xia, N. Psychogios, N. Young, and D.S. Wishart, MetaboAnalyst: a web server for metabolomic data analysis and interpretation, Nucleic Acids Research 37 (2009) p. W652-W660.

42. Yip A and Horvath S, Gene network interconnectedness and the generalized topological overlap measure, BMC Bioinformatics 8 (2007) p. 22.

43. Zhang B and Horvath S, A General Framework for Weighted Gene Co-Expression Network Analysis, Statis-tical Applications in Genetics and Molecular Biology 4 (2005).

K007_000000_HereThere and EveryWhere boekje.indd 51 09-02-15 08:30

Page 56: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

52 PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

Supplemental data - part 2

Table S1 Acylcarnitine levels in fed and fasted mice

Table S1a. Acylcarnitine levels in fed and fasted Bl6 mice

Bl6 mice C0 C2 C3 C4 C4OH C5 C12 C14:1 C16 C18:1

Plasma

fed mean 21.11 4.16 0.14 0.21 0.06 0.05 0.01 0.02 0.15 0.07

SD 3.57 0.74 0.06 0.18 0.02 0.02 0.00 0.01 0.03 0.02

fasted mean 10.94 5.18 0.13 0.23 0.14 0.07 0.03 0.09 0.50 0.43

SD 2.46 0.81 0.06 0.10 0.03 0.03 0.01 0.02 0.08 0.06

p 0.00 0.01 0.57 0.74 0.00 0.05 0.00 0.00 0.00 0.00

Liver

fed mean 785.76 161.72 9.90 31.88 5.59 3.96 0.58 0.42 3.49 3.14

SD 236.18 44.67 4.84 16.38 2.36 2.38 0.17 0.23 1.20 1.49

fasted mean 1020.73 121.80 10.75 8.23 5.80 2.99 1.96 2.74 14.91 13.24

SD 122.64 22.94 1.85 2.35 1.59 1.29 0.94 1.29 2.20 3.43

p 0.01 0.01 0.58 0.00 0.81 0.24 0.00 0.00 0.00 0.00

Soleus

fed mean 1963.75 368.84 9.47 12.19 5.57 1.09 5.04 6.51 47.73 28.61

SD 304.75 89.24 2.66 4.66 2.73 0.11 1.94 2.88 26.71 16.04

fasted mean 1644.47 555.83 11.32 27.02 49.00 1.57 5.54 9.43 59.78 55.48

SD 198.27 82.34 4.05 13.84 16.35 0.68 2.61 5.40 39.48 39.43

p 0.01 0.00 0.22 0.01 0.00 0.04 0.61 0.13 0.41 0.06

Gastroc- nemius

fed mean 10559.84 112.66 2.31 8.75 1.03 1.76 4.75 4.42 126.17 39.73

SD 799.39 15.10 0.43 1.84 0.18 0.25 2.05 2.00 54.64 15.75

fasted mean 12039.26 105.37 4.16 14.29 4.80 5.27 4.82 9.25 115.97 88.98

SD 1571.88 24.31 0.81 3.31 1.87 1.36 1.71 3.39 33.38 35.58

p 0.01 0.41 0.00 0.00 0.00 0.00 0.93 0.00 0.59 0.00

Quadri-ceps

fed mean 11504.66 137.45 2.63 9.42 1.22 1.47 4.67 4.69 189.61 72.61

SD 1929.83 19.49 0.49 3.27 0.35 0.31 1.91 2.23 75.77 35.33

fasted mean 10967.56 107.50 3.70 15.26 4.51 5.72 4.31 7.91 157.24 122.60

SD 1571.87 25.15 1.07 3.64 1.58 1.58 1.18 2.88 40.12 48.98

p 0.47 0.01 0.01 0.00 0.00 0.00 0.58 0.01 0.21 0.01

WAT

fed mean 14.38 0.94 0.09 0.14 0.05 0.06 0.02 0.02 0.21 0.11

SD 4.00 0.32 0.03 0.05 0.02 0.03 0.01 0.01 0.10 0.05

fasted mean 19.23 2.16 0.19 0.25 0.18 0.16 0.04 0.03 0.58 0.31

SD 6.64 0.94 0.08 0.14 0.07 0.10 0.03 0.01 0.17 0.09

p 0.05 0.00 0.00 0.02 0.00 0.01 0.01 0.01 0.00 0.00

BAT

fed mean 7204.51 40.90 1.11 4.65 1.67 0.51 0.46 0.56 4.54 3.99

SD 3594.46 25.57 0.93 3.91 0.79 0.35 0.31 0.41 3.55 2.21

fasted mean 15497.14 222.41 3.72 15.40 16.80 3.92 2.59 3.69 14.40 15.82

SD 9221.32 130.05 2.14 9.72 10.81 2.41 1.02 1.24 5.10 6.27

p 0.02 0.00 0.00 0.01 0.00 0.00 0.00 0.00 0.00 0.00

Heart

fed mean 2874.08 349.28 9.24 18.67 10.10 1.41 4.66 8.40 73.35 65.09

SD 385.66 214.12 4.55 23.35 18.84 0.94 2.43 4.41 47.36 50.43

fasted mean 2384.26 493.75 12.72 26.53 28.96 2.75 22.68 41.09 171.53 193.70

SD 462.17 159.64 8.69 9.56 16.44 1.44 18.94 37.57 138.62 169.88

p 0.01 0.08 0.25 0.30 0.02 0.02 0.01 0.02 0.05 0.03

K007_000000_HereThere and EveryWhere boekje.indd 52 09-02-15 08:30

Page 57: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

53PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

Table S1b. Acylcarnitine levels in fed and fasted BALB mice

BALB mice C0 C2 C3 C4 C4OH C5 C12 C14:1 C16 C18:1

Plasma

fed mean 32.37 7.04 0.21 0.80 0.09 0.12 0.02 0.03 0.16 0.09

SD 4.13 1.27 0.05 0.84 0.02 0.05 0.00 0.01 0.04 0.03

fasted mean 23.18 13.51 0.29 0.53 0.28 0.09 0.04 0.15 0.49 0.44

SD 2.98 3.22 0.09 0.19 0.06 0.03 0.01 0.03 0.08 0.06

p 0.00 0.00 0.02 0.31 0.00 0.05 0.00 0.00 0.00 0.00

Liver

fed mean 1071.92 262.84 23.84 81.58 6.25 10.15 1.05 0.77 6.57 5.41

SD 265.95 60.46 13.66 70.83 1.54 5.02 0.31 0.39 3.66 3.53

fasted mean 1318.07 179.65 24.39 11.45 6.17 5.97 3.45 4.82 16.97 13.73

SD 270.62 69.91 11.54 5.49 1.97 2.82 1.17 1.82 2.75 2.62

p 0.04 0.01 0.92 0.01 0.92 0.03 0.00 0.00 0.00 0.00

Soleus

fed mean 2169.38 518.82 8.94 30.70 7.96 2.40 9.48 11.79 123.39 75.02

SD 281.78 132.94 3.64 13.18 4.35 0.50 4.12 5.12 74.97 45.85

fasted mean 1673.51 725.09 8.81 57.51 52.07 2.96 7.42 10.61 145.96 150.84

SD 254.00 88.19 2.54 9.56 17.65 1.32 1.34 1.93 57.77 68.69

p 0.00 0.00 0.93 0.00 0.00 0.21 0.14 0.49 0.44 0.01

Gastroc- nemius

fed mean 1039.19 207.66 3.09 17.14 0.70 1.97 8.55 8.16 134.34 54.21

SD 148.62 39.22 1.34 5.85 0.23 1.18 3.76 3.60 56.31 22.66

fasted mean 884.00 193.43 2.80 25.05 3.10 2.33 10.02 12.99 196.64 171.78

SD 111.54 30.08 0.55 3.78 1.26 0.78 4.18 5.21 88.41 63.19

p 0.01 0.35 0.52 0.00 0.00 0.42 0.40 0.02 0.07 0.00

Quadri-ceps

fed mean 865.46 236.79 2.70 17.80 3.36 1.50 4.75 4.47 161.39 56.48

SD 120.74 40.46 0.44 4.87 1.23 0.74 1.15 1.26 78.26 28.03

fasted mean 650.85 197.76 2.80 22.77 11.08 1.56 6.10 8.66 210.61 143.88

SD 161.37 33.86 0.43 3.79 5.22 0.48 1.14 1.98 56.44 36.38

p 0.00 0.02 0.60 0.02 0.00 0.83 0.01 0.00 0.11 0.00

WAT

fed mean 70.80 4.96 0.38 0.69 0.37 0.46 0.13 0.09 0.81 0.40

SD 22.64 3.05 0.24 0.30 0.23 0.22 0.09 0.05 0.47 0.32

fasted mean 113.61 14.29 1.11 1.43 1.07 0.89 0.34 0.22 1.52 1.02

SD 50.86 7.09 0.57 0.79 0.52 0.68 0.25 0.10 0.61 0.57

p 0.02 0.00 0.00 0.01 0.00 0.07 0.03 0.00 0.01 0.01

BAT

fed mean 493.40 36.62 0.92 2.20 1.04 0.50 0.49 0.50 3.22 2.71

SD 287.12 21.06 1.20 1.26 0.61 0.38 0.18 0.23 1.27 0.60

fasted mean 1968.44 167.53 3.05 11.19 4.33 4.96 2.18 2.66 11.38 13.07

SD 1051.82 133.65 2.33 8.23 2.58 4.04 0.88 1.19 4.93 6.06

p 0.00 0.01 0.02 0.00 0.00 0.00 0.00 0.00 0.00 0.00

Heart

fed mean 4781.61 311.70 14.03 14.37 4.47 4.89 7.85 11.31 80.02 62.41

SD 2169.71 209.73 9.15 10.90 6.26 1.96 12.35 18.27 157.87 134.52

fasted mean 3258.49 655.09 16.43 18.63 35.11 5.98 16.89 26.48 102.90 109.02

SD 1176.71 367.32 8.09 8.20 25.82 3.21 17.32 25.99 96.61 102.63

p 0.06 0.02 0.52 0.31 0.00 0.35 0.18 0.13 0.69 0.37

K007_000000_HereThere and EveryWhere boekje.indd 53 09-02-15 08:30

Page 58: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

54 PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

Table S2 Individual Spearman’s correlations C4OH-carnitine in fasted mice

Table S2a. C4OH-carnitine correlations in fasted Bl6 mice

FASTED Bl6 BOHB C4OH PLASMA

C4OH LIVER

C4OH SOLEUS

C4OH GASTROC

C4OH QUAD

C4OH HEART

C4OH WAT

C4OH BAT

BOHB Corr. Coeff. 1.000 .243 .709* .882** .782** .591 .464 .127 .406

P . .471 .015 .000 .004 .056 .151 .709 .244

C4OH PLASMA Corr. Coeff. .243 1.000 -.028 .523 .216 .330 -.592 .110 -.324

P .471 . .936 .099 .524 .321 .055 .747 .361

C4OH LIVER Corr. Coeff. .709* -.028 1.000 .427 .527 .291 .436 .545 .503

P .015 .936 . .190 .096 .385 .180 .083 .138

C4OH SOLEUS Corr. Coeff. .882** .523 .427 1.000 .800** .627* .255 .036 .297

P .000 .099 .190 . .003 .039 .450 .915 .405

C4OH GASTROC

Corr. Coeff. .782** .216 .527 .800** 1.000 .709* .355 .300 .661*

P .004 .524 .096 .003 . .015 .285 .370 .038

C4OH QUAD Corr. Coeff. .591 .330 .291 .627* .709* 1.000 .064 -.045 .515

P .056 .321 .385 .039 .015 . .853 .894 .128

C4OH HEART Corr. Coeff. .464 -.592 .436 .255 .355 .064 1.000 -.018 .552

P .151 .055 .180 .450 .285 .853 . .958 .098

C4OH WAT Corr. Coeff. .127 .110 .545 .036 .300 -.045 -.018 1.000 .418

P .709 .747 .083 .915 .370 .894 .958 . .229

C4OH BAT Corr. Coeff. .406 -.324 .503 .297 .661* .515 .552 .418 1.000

P .244 .361 .138 .405 .038 .128 .098 .229 .

Correlation is significant at the 0.05 level (2-tailed).*

Correlation is significant at the 0.01 level (2-tailed).**

Table S2b. C4OH-carnitine correlations in fasted BALB mice

FASTED BALB BOHB C4OH PLASMA

C4OH LIVER

C4OH SOLEUS

C4OH GASTROC

C4OH QUAD

C4OH HEART

C4OH WAT

C4OH BAT

BOHB Corr. Coeff. 1.000 -.527 -.609* -.018 -.273 -.273 -.245 -.191 .382

P . .096 .047 .958 .417 .417 .467 .574 .247

C4OH PLASMA Corr. Coeff. -.527 1.000 .609* .624* .791** .627* .673* .391 -.664*

P .096 . .047 .040 .004 .039 .023 .235 .026

C4OH LIVER Corr. Coeff. -.609* .609* 1.000 .323 .591 .355 .545 .573 -.291

P .047 .047 . .332 .056 .285 .083 .066 .385

C4OH SOLEUS Corr. Coeff. -.018 .624* .323 1.000 .670* .743** .337 .191 -.533

P .958 .040 .332 . .024 .009 .311 .573 .091

C4OH GASTROC

Corr. Coeff. -.273 .791** .591 .670* 1.000 .709* .882** .364 -.436

P .417 .004 .056 .024 . .015 .000 .272 .180

C4OH QUAD Corr. Coeff. -.273 .627* .355 .743** .709* 1.000 .400 -.100 -.782**

P .417 .039 .285 .009 .015 . .223 .770 .004

C4OH HEART Corr. Coeff. -.245 .673* .545 .337 .882** .400 1.000 .509 -.264

P .467 .023 .083 .311 .000 .223 . .110 .433

C4OH WAT Corr. Coeff. -.191 .391 .573 .191 .364 -.100 .509 1.000 .164

P .574 .235 .066 .573 .272 .770 .110 . .631

C4OH BAT Corr. Coeff. .382 -.664* -.291 -.533 -.436 -.782** -.264 .164 1.000

P .247 .026 .385 .091 .180 .004 .433 .631 .

Correlation is significant at the 0.05 level (2-tailed).*

Correlation is significant at the 0.01 level (2-tailed).**

K007_000000_HereThere and EveryWhere boekje.indd 54 09-02-15 08:30

Page 59: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

55PLASMA ACYLCARNITINES INADEQUATELY REFLECT TISSUE ACYLCARNITINES

Table S3 Individual Spearman’s correlations long-chain acylcarnitines in fasted muscle

Table S3a Correlations long chain acylcarnitines in muscle of fasted Bl6

FASTED Bl6 C16 SOLEUS C16 GASTROC C16 QUAD C18 SOLEUS C18 GASTROC C18 QUAD

C16 SOLEUS Corr. Coeff. 1.000 .282 .273 .945** .600 .600

P . .401 .417 .000 .051 .051

C16 GASTROC Corr. Coeff. .282 1.000 .764** .136 .773** .827**

P .401 . .006 .689 .005 .002

C16 QUAD Corr. Coeff. .273 .764** 1.000 .136 .573 .809**

P .417 .006 . .689 .066 .003

C18 SOLEUS Corr. Coeff. .945** .136 .136 1.000 .573 .527

P .000 .689 .689 . .066 .096

C18 GASTROC Corr. Coeff. .600 .773** .573 .573 1.000 .864**

P .051 .005 .066 .066 . .001

C18 QUAD Corr. Coeff. .600 .827** .809** .527 .864** 1.000

P .051 .002 .003 .096 .001

Correlation is significant at the 0.05 level (2-tailed).*

Correlation is significant at the 0.01 level (2-tailed).**

Table S3b Correlations long chain acylcarnitines in muscle of fasted BALB

C16 SOLEUS C16 GASTROC C16 QUAD C18 SOLEUS C18 GASTROC C18 QUAD

C16 SOLEUS Corr. Coeff. 1.000 .282 .091 .782** .355 -.127

P . .401 .790 .004 .285 .709

C16 GASTROC Corr. Coeff. .282 1.000 .773** .527 .945** .482

P .401 . .005 .096 .000 .133

C16 QUAD Corr. Coeff. .091 .773** 1.000 .245 .718* .809**

P .790 .005 . .467 .013 .003

C18 SOLEUS Corr. Coeff. .782** .527 .245 1.000 .709* .055

P .004 .096 .467 . .015 .873

C18 GASTROC Corr. Coeff. .355 .945** .718* .709* 1.000 .482

P .285 .000 .013 .015 . .133

C18 QUAD Corr. Coeff. -.127 .482 .809** .055 .482 1.000

P .709 .133 .003 .873 .133

.

Correlation is significant at the 0.05 level (2-tailed).*

Correlation is significant at the 0.01 level (2-tailed).**

K007_000000_HereThere and EveryWhere boekje.indd 55 09-02-15 08:30

Page 60: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

C 4 Transorgan fluxes in a porcine model reveal a

central role for liver in acylcarnitine metabolism

Marieke G. Schooneman, Gabrie A.M. ten Have, Sander M. Houten, Nicolaas E.P. Deutz, Maarten R. Soeters

Submitted

K007_000000_HereThere and EveryWhere boekje.indd 56 09-02-15 08:30

Page 61: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

C 4 Transorgan fluxes in a porcine model reveal a

central role for liver in acylcarnitine metabolism

Marieke G. Schooneman, Gabrie A.M. ten Have, Sander M. Houten, Nicolaas E.P. Deutz, Maarten R. Soeters

Submitted

K007_000000_HereThere and EveryWhere boekje.indd 57 09-02-15 08:30

Page 62: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

K007_000000_HereThere and EveryWhere boekje.indd 58 09-02-15 08:30

Page 63: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

59TRANSORGAN FLUXES IN A PORCINE MODEL

Abstract

Acylcarnitines are derived from mitochondrial acyl-CoA metabolism, and have been as-sociated with diet-induced insulin resistance. However, plasma acylcarnitine profiles have been shown to poorly reflect whole body acylcarnitine metabolism.We aimed to clarify the individual role of different organ compartments in whole body acylcarnitine metabolism in a fasted and postprandial state in a porcine transorgan ar-teriovenous model. Twelve cross-bred pigs underwent surgery where intravascular catheters were posi-tioned before and after the liver, gut, hindquarter muscle compartment and the kidney. Before and after a mixed meal we measured acylcarnitine profiles at several time points and calculated transorgan acylcarnitine fluxes.Fasting plasma acylcarnitine concentrations correlated with hepatic transorgan fluxes of free, C2- and C16-carnitine. Transorgan acylcarnitine fluxes were small, except for a pronounced hepatic C2-carnitine production. The peak of the postprandial acylcarni-tine fluxes was between 60 to 90 minutes. Acylcarnitine production or release was seen in the gut and liver, and consisted mostly of C2-carnitine. Acylcarnitines were extracted by the kidney. No significant muscle acylcarnitine flux was observed.Liver has a key role in acylcarnitine metabolism with high fluxes of C2-carnitine both in the fasted and fed state, whereas the contribution of skeletal muscle is minor. These results further clarify the role of different organ compartments in the metabolism of different acylcarnitine species.

Introduction

Acylcarnitines are intermediates of mitochondrial acyl-CoA metabolism, which have gained much attention as markers of inherited metabolic diseases (28), and more recent-ly for their possible involvement in diet-induced insulin resistance and glucose intoler-ance (2, 9, 11, 13, 15, 20). Acylcarnitines are carboxylic acids of different chain lengths, derived from different substrates (eg. fatty acids, amino acids or acetyl-CoA) which are transesterified from CoA to L-carnitine, enabling them to enter or exit the mitochondri-on (27). Exchange of acylcarnitines also occurs over the cell membrane. This leads to a specific acylcarnitine profile in plasma, composed of acylcarnitines originating from and consumed by the different organs and compartments where the respective acyl-CoAs are metabolized (27). Historically, acylcarnitine profiles have been measured to detect mitochondrial fatty acid oxidation (FAO) disorders (28). More recently, studies have dis-cussed alterations in the acylcarnitine profile, mostly measured in plasma, in relation to deranged FAO and glucose intolerance or insulin resistance (2, 9, 11). As increased acyl-carnitine levels correlated with markers of glucose intolerance in obesity, acylcarnitines were proposed to potentially induce insulin resistance (7, 8, 22). However, we previously have shown that the plasma acylcarnitine profile should be in-terpreted with caution, as it does not reflect the acylcarnitine profile measured in a

K007_000000_HereThere and EveryWhere boekje.indd 59 09-02-15 08:30

Page 64: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

60 TRANSORGAN FLUXES IN A PORCINE MODEL

specific tissue (19, 23). To identify derangements of FAO involving acylcarnitines, these metabolite profiles should be measured in the specific tissue of interest. Apparently, the individual role of the plasma compartment and the different tissue compartments in whole body acylcarnitine metabolism is still poorly understood. We used a porcine model in which several intravascular catheters were positioned to measure transorgan fluxes in gut (portally drained viscera), liver, kidney and the hind-quarter skeletal muscle compartment. We measured fluxes of acylcarnitines in con-scious pigs on different time points in the fasted state and after a mixed meal. For this translational experimental setting, pigs are a very useful model, as their metabolism and gastrointestinal anatomy and physiology resembles that of humans (12). More specifically the transcriptional regulation of lipid metabolism, and subsequent FAO and acylcarnitine metabolism are comparable between pigs and humans (10, 17, 18). In this study we showed the importance of liver in whole body acylcarnitine metabolism and the relatively minor role for skeletal muscle of fasted and fed pigs.

Methods

Animals Twelve female crossbred piglets (20-25 kg, aged 8-12 weeks) were obtained from a commercial breeder in Rosenbaum Farms, Brenham, TX, USA. One week before sur-gery, animals were allowed to adapt to individual housing in galvanized bar runs (2 x 3 m), enriched with straw and toys during surgery recovery. Environmental temperature was 21-25°C, humidity level was not controlled and light was on for 12h per day (from 7AM to 7PM). A radio set was switched on during the light period. The piglets were fed with Harlan teklad Vegetarian Pig/Sow Grower (Harlan Laboratoria, Indianapolis, IN, USA). Food intake was 1 kg per day. Water was available ad libitum. The animal use protocol was approved by the Institutional Animal Care and Use Committee of Texas A&M University.

Surgery One week prior to the experiment the animals underwent surgery, where sev-eral indwelling catheters were placed, according to the technique described earlier by Deutz and Ten Have (figure 1)(5, 25). In short, the animals were fasted for 16 hours prior to surgery. On the day of surgery, anaesthesia was induced with an intramuscular dose of ti-letamine (3.3 mg/kg) and zolazepam (3.3mg/kg) (Telazol, Zoetis Inc, Kalamazoo, MI, USA). Animals were intubated and anaesthesia was maintained with isoflurane (2%). Before the start of the surgery a combination of lincomycin (6.25 mg/kg) and spectinomycin (12.5mg/kg, Linco-Spectin, Zoetis Inc, Kalamazoo, MI, USA) was administered intrave-nously as antimicrobial prophylaxis. Flunixin meglumine (2 mg/kg, Flunixamine, Zoetis Inc, Kalamazoo, MI, USA) was administered intravenously as anticoagulant and analgeti-cum. The abdominal cavity was approached by a median incision over the whole length of the abdomen. For muscle flux measurements, a sampling catheter was implanted into the inferior caval vein (V) with its tip 5 cm above the bifurcation, and a para-aminohip-puric acid (PAH) infusion catheter was implanted into the abdominal aorta with its tip 5 cm above the bifurcation. For kidney flux measurements, a catheter was implanted into

K007_000000_HereThere and EveryWhere boekje.indd 60 09-02-15 08:30

Page 65: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

61TRANSORGAN FLUXES IN A PORCINE MODEL

the left renal vein (R). A catheter for the infusion of PAH was implanted into the splenic vein. The catheter for splanchnic flux measurements was implanted via the portal vein with its tip in the liver hilus (P) and into a hepatic vein (H) by direct puncture. For infusion of the liquid meal, a feeding tube was implanted into the stomach. A jejunal Bishop Koop stoma was created approximately 25cm from the Treitz ligament. Finally, all catheters were secured in place by sutures and alpha-cyanoacrylate glue (Elmer’s Products Inc, Columbus, OH, USA) and were tunneled through the left abdominal wall. Before closure the abdominal cavity was flushed with iodine. The abdominal incision was closed in three layers with a continuous stitch and the pigs were fitted in a canvas harness to protect the catheters and to allow easy handling of the animals. All catheters were filled with 0.5 ml of a solution of gentamycine (20mg/ml) and alpha-chymotrypsin (225U/ml) to keep the catheters patent. For postoperative care, the animals were checked twice per day for 4 days after surgery, for overall behaviour, body temperature, catheter patency, and intra-venous administration of antibiotics (lincomycin 6.25 mg/kg and spectinomycin 12.5mg/kg) and an analgeticum (flunixin meglumine, 2 mg/kg). During the recovery period (7-10 days) animals were accustomed to a small movable cage (0.9x0.5x0.3m). The experiments were performed in this cage in conscious animals.

Figure 1 Placement of catheters. A= arterial line, placed in iliac circumflex profunda artery with its tip above the bifurcation

(PAH infusion for muscle flow) or above the right renal artery (blood collection); V= venous line, placed in iliac circumflex

profunda vein with its tip above the bifurcation (muscle blood collection or above the right renal vein for infusion); P= portal

line, placed in portal vein (splenic approach and tip in liver hilus); H= hepatic line, placed in hepatic vein by direct puncture

of the liver; R= renal line, placed in left renal vein; MV= mesenteric vein.

Experiment Food was withdrawn at 16:00 the day prior to the mixed meal experiment, so that the animals were fasted overnight until administration of the mixed test meal at 9:00 the next morning. In the morning the animals were weighed, temperature was measured. At 8:00, on time point t=-60’, a continuous infusion of 25 mM PAH at an infusion rate of 60 ml/hour was started. These flow measurements are needed for the final calculations of the transorgan fluxes on the individual time points. After 30 minutes, at t=-30’, a first jejunal mucosa biopsy was taken via the stoma, which was immediately frozen in liquid

A V

A

A

H

RMV

P

K007_000000_HereThere and EveryWhere boekje.indd 61 09-02-15 08:30

Page 66: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

62

nitrogen. Three basal blood withdrawals were performed at t=-10’, -5’ and 0 from all sampling catheters (A, P, H, V, R). Sampling was performed in the same order throughout the whole experiment. After the last basal blood withdrawal on t=0, the meal was ad-ministered directly into the stomach via the implanted gastric catheter. For a pig of 25 kg, the test meal consisted of 78 grams of Crude Whey protein isolate and 110 grams of carbohydrates mixed in water with a total volume of 600 ml, and additionally 22 grams of olive oil (30% of daily energy intake). We aimed for a maximum duration of administration of 5 minutes for the complete test meal. We performed another 9 blood withdrawals up to 240’ after the test meal. At t=60’ and t=180’ two additional mucosa biopsies were performed. After the experiment the pigs were placed in their original cages and were provided with their normal food and drinking water.

Sample preparation and laboratory analyses The blood samples were directly placed on ice. For the measurement of PAH concentrations 250 μL of blood was transferred to a tube containing 25 μL trichloroacetic acid (TCA), thoroughly vortexed and subsequently, together with the remaining blood samples, centrifuged at 8000 g for 5 minutes at 4°C. The supernatant of the TCA samples was pipetted into a clean tube. The other plasma samples were divided over different tubes. All samples were frozen in liquid nitrogen, later to be stored at -80°C. For the measurement of the plasma flow, PAH concentra-tions in TCA deproteinized plasma samples were compared with PAH standards and read out with a Microplate spectrophotometer (Spectramax, Molecular Devices Corporation, Sunnyvale, California, USA) running SoftmaxPro software (Molecular Devices Corpora-tion, Sunnyvale, California, USA) (1). Plasma acylcarnitine profiles were measured and an-alysed as previously described (19).

Calculations Blood flow was calculated using the dilution of PAH across the organs (3, 26). Here the amount of PAH infused per min in the splenic vein and the abdominal aor-ta was divided by the PAH concentration differences in venous blood downstream and arterial blood upstream of the organ measured. Concentrations of acylcarnitines were calculated from Masslynx measurements (Masslynx software version 4.1) in Microsoft Ex-cel 2011 version 14.4.4. C0 indicated free carnitine and Cx indicates the acylcarnitines of the respective fatty acids with a chain length of x. To calculate acylcarnitine fluxes over the organ compartments, the difference between plasma venous – arterial acylcarnitine concentration was multiplied by the mean plasma flow (4-6, 26). A positive value of the flux is therefore interpreted as (acyl)carnitine appearance or production and a negative flux value is interpreted as (acyl)carnitine disappearance or uptake. All additional analyses and statistics were performed using Microsoft Excel 2011 version 14.4.4 and Graphpad Prism software version 6.0c for Mac. A one-way ANOVA was per-formed to detect differences between AUCs and mean fluxes at specific time points between the four compartments. To test whether the flux at a certain time point differed from baseline in that same compartment, we performed a Wilcoxon paired t test. All data are expressed as means ± SEM. P-values < 0.05 are considered as statistically significant and depicted as *, and p-values < 0.01 are depicted as **.

TRANSORGAN FLUXES IN A PORCINE MODEL

K007_000000_HereThere and EveryWhere boekje.indd 62 09-02-15 08:30

Page 67: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

63

Results

Acylcarnitine fluxes in liver, muscle, kidney and the gutAcylcarnitine fluxes were calculated using the (acyl)carnitine concentrations in blood samples on the indicated time points (table 1) and the plasma flow which increased upon the meal in all compartments but muscle (data not shown). Concentrations of all acylcar-nitines were lower compared with human clinical laboratory reference values or plasma concentrations obtained in rodents (19).

C0 C2 C3 C5 C4OH C16 C18:1

t (min) sample

0 A 6.87 (± 0.41) 1.24 (± 0.04) 0.09 (± 0.02) 0.01 (± 0.00) 0.00 (± 0.00) 0.01 (± 0.00) 0.02 (± 0.00)

P 6.84 ( ± 0.43) 1.20 (± 0.06) 0.10 (± 0.02) 0.01 (± 0.00) 0.00 (± 0.00) 0.01 (± 0.00) 0.02 (± 0.00)

H 7.17 (± 0.33) 1.56 (± 0.11) 0.09 (± 0.01) 0.01 (± 0.00) 0.01 (± 0.00) 0.02 (± 0.00) 0.02 (± 0.00)

V 6.97 (± 0.34) 1.07 (± 0.05) 0.09 (± 0.02) 0.01 (± 0.00) 0.00 (± 0.00) 0.01 (± 0.00) 0.02 (± 0.00)

R 6.98 (± 0.43) 1.10 (± 0.06) 0.09 (± 0.02) 0.01 (± 0.00) 0.00 (± 0.00) 0.01 (± 0.00) 0.02 (± 0.00)

10 A 6.91 (± 0.36) 1.37 (± 0.07) 0.10 (± 0.01) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.02 (± 0.00)

P 6.86 (± 0.39) 1.41 (± 0.07) 0.11 (± 0.01) 0.01 (± 0.00) 0.01 (± 0.00) 0.02 (± 0.00) 0.02 (± 0.00)

H 7.20 (± 0.30) 1.85 (± 0.12) 0.10 (± 0.01) 0.01 (± 0.00) 0.01 (± 0.00) 0.02 (± 0.00) 0.02 (± 0.00)

V 7.14 (± 0.32) 1.26 (± 0.08) 0.09 (± 0.01) 0.01 (± 0.00) 0.01 (± 0.00) 0.02 (± 0.00) 0.02 (± 0.00)

R 6.62 (± 0.48) 1.17 (± 0.14) 0.10 (± 0.02) 0.01 (± 0.00) 0.00 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

20 A 6.71 (± 0.50) 1.40 (± 0.14) 0.11 (± 0.02) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.02 (± 0.00)

P 6.64 (± 0.50) 1.43 (± 0.18) 0.12 (± 0.02) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.02 (± 0.00)

H 7.60 (± 0.38) 2.15 (± 0.18) 0.12 (± 0.01) 0.02 (± 0.00) 0.01 (± 0.00) 0.02 (± 0.00) 0.02 (± 0.00)

V 6.93 (± 0.37) 1.23 (± 0.08) 0.11 (± 0.01) 0.01 (± 0.00) 0.00 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

R 6.94 (± 0.42) 1.26 (± 0.05) 0.11 (± 0.01) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

30 A 6.92 (± 0.35) 1.65 (± 0.13) 0.13 (± 0.01) 0.02 (± 0.00) 0.01 (± 0.00) 0.02 (± 0.00) 0.01 (± 0.00)

P 7.50 (± 0.36) 1.79 (± 0.13) 0.15 (± 0.01) 0.02 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.02 (± 0.00)

H 7.47 (± 0.41) 2.23 (± 0.11) 0.14 (± 0.02) 0.02 (± 0.00) 0.01 (± 0.00) 0.02 (± 0.00) 0.01 (± 0.00)

V 7.12 (± 0.39) 1.37 (± 0.08) 0.12 (± 0.01) 0.02 (± 0.00) 0.00 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

R 7.37 (± 0.26) 1.40 (± 0.11) 0.13 (± 0.01) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

45 A 7.27 (± 0.39) 1.75 (± 0.10) 0.14 (± 0.01) 0.02 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

P 7.13 (± 0.40) 1.77 (± 0.15) 0.16 (± 0.02) 0.03 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

H 6.90 (± 0.37) 2.17 (± 0.11) 0.15 (± 0.01) 0.03 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

V 7.02 (± 0.35) 1.40 (± 0.09) 0.13 (± 0.01) 0.02 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

R 7.04 (± 0.35) 1.44 (± 0.10) 0.13 (± 0.01) 0.02 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

60 A 7.27 (± 0.35) 1.75 (± 0.10) 0.15 (± 0.01) 0.03 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

P 7.08 (± 0.40) 1.90 (± 0.12) 0.16 (± 0.02) 0.03 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

H 7.26 (± 0.42) 2.30 (± 0.17) 0.15 (± 0.01) 0.03 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

V 7.10 (± 0.41) 1.44 (± 0.07) 0.14 (± 0.01) 0.02 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

R 7.05 (± 0.50) 1.45 (± 0.09) 0.14 (± 0.02) 0.02 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

90 A 6.95 (± 0.47) 1.72 (± 0.11) 0.14 (± 0.01) 0.02 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

P 6.94 (± 0.48) 1.72 (± 0.12) 0.16 (± 0.02) 0.03 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

H 7.01 (±0.39) 2.14 (± 0.15) 0.16 (± 0.02) 0.03 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

V 7.20 (± 0.40) 1.39 (± 0.08) 0.14 (± 0.01) 0.02 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

R 7.15 (± 0.47) 1.41 (± 0.09) 0.15 (± 0.01) 0.02 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00) 0.01 (± 0.00)

Table 1 Acylcarnitine concentrations Absolute plasma concentrations per time point and catheter. Acylcarnitines are

depicted by chain length (eg. C0 = free carnitine, C2 = C2-carnitine). A= arterial sample, P= portal sample, H= hepatic sample,

V= venous sample, R= renal sample. Values are means ± SD.

TRANSORGAN FLUXES IN A PORCINE MODEL

K007_000000_HereThere and EveryWhere boekje.indd 63 09-02-15 08:30

Page 68: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

64

Baseline We determined the mean acylcarnitine flux of 3 plasma samples prior to the meal (Figure 2a). In the fasted pigs, carnitine flux rates were small and did not differ be-tween the compartments. Liver showed production of other acylcarnitine species, which may be due to higher FAO in the fasted state. Here, acetylcarnitine was produced at high rates by the liver, whereas all other compartments showed uptake of acetylcarnitine at low rates. C3- and C5-carnitine were produced by the gut. In kidney, uptake was seen for the short chain species, which probably reflects excretion via the urine (Figure 2a).

Postprandial We measured postprandial fluxes by calculating the AUCs for the different acylcarnitine curves (figure 2b and c). Analysis of the net AUCs (consisting of the total positive AUC minus the total negative AUC) showed no differences in carnitine fluxes, whereas C2-carnitine fluxes from liver and kidney were significantly higher and lower respectively compared to the other compartments (figure 2b). Net AUCs of C3- and C5-carnitine showed uptake by the gut and liver compartment, which was only marginally found for C16- and C18:1-carnitine.To enable further interpretation of the net AUC with regard to different physiological functions of both acylcarnitine production and uptake, we calculated positive and negative AUCs separately (figure 2c). We found no differences in carnitine production or uptake. C2-carnitine was clearly produced by the liver and to a much lesser extent by the gut, but absorbed by muscle and kidney. C3- and C5-carnitine fluxes were similar with production or release by the gut and to a lesser extent liver.  However, C3 and C5 fluxes were much lower compared to C2 fluxes. The positive AUCs for C16-carnitine showedthat liver production differed significantly from kidney, and the uptake by liver and kidney differed significantly from muscle and gut. No differences in production were detected for C18:1-carnitine, but uptake by the liver and kidney both differed from uptake by the gut. Overall, the differences in production were greater, and flux rates were higher than those of negative fluxes. Moreover, on whole body level, the net production of C2-carni-tine was 2.5-fold higher than carnitine (cumulative net AUC for carnitine of 19516 versus 48233 for C2-carnitine), possibly suggesting a quantitative more important role for C2-carnitine in carnitine distribution compared to carnitine.

TRANSORGAN FLUXES IN A PORCINE MODEL

K007_000000_HereThere and EveryWhere boekje.indd 64 09-02-15 08:30

Page 69: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

65

-10

-5

0

5

10

15ba

selin

e C

0 flu

x(u

mol

/kg

BW

/min

)

PRODUCTIO

NUPTAKE

-10

0

10

20

30

40

base

line

C2

flux

(um

ol/k

g B

W/m

in)

** ** PRODUCTIO

NUPT.

-1.0

-0.5

0.0

0.5

1.0

base

line

C3

flux

(um

ol/k

g B

W/m

in)

****

PRODUCTIO

NUPTAKE

-0.3

-0.2

-0.1

0.0

0.1

0.2

0.3

base

line

C4

flux

(um

ol/k

g B

W/m

in)

*

*

*

PRODUCTIO

NUPTAKE

-0.2

-0.1

0.0

0.1

0.2

0.3

base

line

C5

flux

(um

ol/k

g B

W/m

in)

****

PRODUCTIO

NUPTAKE

-0.10

-0.05

0.00

0.05

0.10

0.15

base

line

C4O

H fl

ux(u

mol

/kg

BW

/min

)

** PRODUCTIO

NUPTAKE

0.00

0.04

0.08

0.12

0.16

base

line

C16

flux

(um

ol/k

g B

W/m

in)

PRODUCTIO

NU.

GUTMUSCLEKIDNEYLIVER

-0.1

0.0

0.1

0.2

base

line

C18

:1 fl

ux(u

mol

/kg

BW

/min

)

*

PRODUCTIO

NUPTAKE

TRANSORGAN FLUXES IN A PORCINE MODEL

Figure 2 Baseline flux and cummulative flux AUCs of acylcarnitines (A) baseline flux, (B) Net AUCs and (C) positive versus

negative AUCs of all organ compartments. Data represent mean ± SEM. P values < 0.05 are depicted as * and P values < 0.01

are depicted as **.

Figure 2a

K007_000000_HereThere and EveryWhere boekje.indd 65 09-02-15 08:30

Page 70: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

66 A SHORT INTRODUCTION

-3000

-2000

-1000

0

1000

2000

3000

net A

UC

C0-

carn

itine

PRODUCTIO

NUPTAKE

-4000

0

4000

8000

12000

net A

UC

C2-

carn

itine

**

**** PRO

DUCTION

UPTAKE

-100

0

100

200

300

net A

UC

C3-

carn

itine **

**

*

PRODUCTIO

NUPTAKE

-60

-40

-20

0

20

40

60

80

net A

UC

C5-

carn

itine **

**

*

PRODUCTIO

NUPTAKE

-20

-10

0

10

net A

UC

C16

-car

nitin

e *

PROD.

UPTAKE

GUTMUSCLEKIDNEYLIVER

-20

-10

0

10

20

net A

UC

C18

:1-c

arni

tine

*** PRO

DUCTION

UPTAKE

Figure 2b

K007_000000_HereThere and EveryWhere boekje.indd 66 09-02-15 08:30

Page 71: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

67A SHORT INTRODUCTION

-4000

-2000

0

2000

4000A

UC

Car

nitin

e

PRODUCTIO

NUPTAKE

-6000-4000-2000

02000400060008000

10000

AU

C C

2-ca

rniti

ne

**

** **

** ****

PRODUCTIO

NUPTAKE

-200-150-100-50

050

100150200250

AU

C C

3-ca

rniti

ne

** *** *

***

*PRO

DUCTION

UPTAKE

-80-60-40-20

020406080

AU

C C

5-ca

rniti

ne** *

*

** *** **

PRODUCTIO

NUPTAKE

-40

-30

-20

-10

0

10

20

AU

C C

16-c

arni

tine

*

**

***

PRODUCTIO

NUPTAKE

GUTMUSCLEKIDNEYLIVER

25

-30-25-20-15-10-505

101520

AU

C C

18:1

-car

nitin

e

****

PRODUCTIO

NUPTAKE

Figure 2c

K007_000000_HereThere and EveryWhere boekje.indd 67 09-02-15 08:30

Page 72: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

68

Individual acylcarnitine flux curves in liver, muscle, kidney and the gutAfter interpreting and testing all baseline and total postprandial fluxes in general, we subsequently interpreted the individual acylcarnitine curves with respect to time to detect and understand acylcarnitine flux patterns (figure 3a-f).

Carnitine The changes in flux of carnitine were most pronounced in liver, gut and kidney (figure 3a) in the first 60 minutes after the meal. The kidney and gut showed carnitine production after 30 minutes, of which only gut production is significantly higher com-

30 60 90 120 150 180 210 240

-50

-40

-30

-20

-10

10

20

30

40

Time (min)

C0-

carn

itine

flux

(um

ol/k

g B

W/m

in) C0 MUSCLE

C0 KIDNEYC0 LIVER

C0 GUT

**

**

30 60 90 120 150 180 210 240

-30

-20

-10

10

20

30

40

50

60

Time (min)

C2-

carn

itine

flux

(um

ol/K

G B

W/m

in)

30 60 90 120 150 180 210 240

-2

-1

0

1

2

Time (min)

C3-

carn

itine

flux

(um

ol/k

g B

W/m

in)

TRANSORGAN FLUXES IN A PORCINE MODEL

K007_000000_HereThere and EveryWhere boekje.indd 68 09-02-15 08:30

Page 73: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

69

pared to baseline (p 0.0068). At 45 minutes the liver shows uptake of carnitine (p 0.002). Both muscle and kidney show some minor non-significant fluctuations.

C2-carnitine The fluxes of C2-carnitine were very stable in contrast to the other acylcar-nitines (figure 3b). The liver and gut were mainly responsible for production of C2-car-nitine, of which the production by liver was the greatest by far. Upon the meal the gut showed continued production of C2-carnitine throughout the whole experiment with two modest production peaks within the first 60 minutes. Both muscle and kidney showed only uptake of C2-carnitine. This uptake tended to further increase upon the meal and remained stable throughout the rest of the experiment.

30 60 90 120 150 180 210 240

-0.5

0.5

Time (min)

C5-

carn

itine

flux

(um

ol/k

g B

W/m

in) *

*

30 60 90 120 150 180 210 240

-0.5

-0.4

-0.3

-0.2

-0.1

0.0

0.1

0.2

0.3

Time (min)

C16

-car

nitin

e flu

x (u

mol

/kg

BW

/min

)

*

30 60 90 120 150 180 210 240

-0.3

-0.2

-0.1

0.0

0.1

0.2

0.3

0.4

Time (min)

C18

:1-c

arni

tine

flux

(um

ol/k

g B

W/m

in)

**

Figure 3 Curves of carnitine and acylcarnitine fluxes through all organ compartments (A) for

Carnitine, (B) C2-carnitine, (C) C3-carnitine, (D) C5-

carnitine, (E) C16-carnitine and (F) C18:1-carnitine.

Data represent means ± SEM. P values < 0.05 are

depicted as * and P values < 0.01 are depicted as **.

TRANSORGAN FLUXES IN A PORCINE MODEL

K007_000000_HereThere and EveryWhere boekje.indd 69 09-02-15 08:30

Page 74: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

70

C3- and C5-carnitine The fluxes of C3- and C5-carnitine showed some similarities, possibly because they are both amino acid derived species (figure 3c and d). The meal affected the fluxes in the first 90 minutes. Both liver and gut showed production of C3- and C5-carnitine, of which only C5-carnitine production in liver reached significance at t=30 compared to baseline (p 0.01), and compared to kidney. Muscle and kidney showed uptake of C3- and C5-carnitine throughout the whole experiment. Kidney C5-carnitine uptake was greater at t=60 compared to baseline (p 0.02) and differed significantly from gut and liver fluxes. Overall we found a clear division of C3- and C5-carnitine production by liver and gut, and uptake by muscle and kidney, which is comparable to C2-carnitine.

C16- and C18:1-carnitine C16-carnitine and C18:1-carnitine are derived from the satu-rated palmitate and unsaturated oleate respectively. The latter was a substantial com-ponent of the mixed meal in the form of olive oil. However, these species showed similar flux patterns, in which the most pronounced changes occurred in the first 90 minutes (figure 3e and f). The liver showed an initial production of C16- and C18:1-carnitine in the first 30 minutes, followed by a significantly decreased flux (p 0.014 for C16-carnitine, and p 0.009 for C18:1-carnitine) reflecting uptake. The gut showed minor but overall produc-tion of C16- and C18:1-carnitine. In contrast to gut, kidney showed uptake throughout the whole experiment. The flux through the muscle compartment was minor for both species, and did not show differences from baseline either.In conclusion, liver and gut showed production of most acylcarnitines, which was signif-icant compared to baseline at some time points in the first hour after the meal. Kidney showed predominantly uptake that may fit with urinary excretion of acylcarnitines.

The relation between venous acylcarnitine concentrations and organ fluxesAs acylcarnitine profiles are mostly measured in plasma for clinical purposes, and most of the existing research on acylcarnitines involves plasma profiles, we were interested to what extent plasma acylcarnitine levels correlated with the fluxes through the different organ compartments. We correlated fluxes through liver, muscle, kidney and gut on t=0, 30 and 90 minutes with the venous concentrations of C0-, C2- and C16-carnitine. Plasma concentrations of acylcarnitines correlated significantly with the flux of these species through liver at time point t=0, in the fasted state (Figure 4). At time points t=30 and t=90, these correlations disappeared for all examined acylcarnitines. No correlation existed between plasma concentrations and the flux through muscle, kidney and gut on all time points (data not shown).

TRANSORGAN FLUXES IN A PORCINE MODEL

K007_000000_HereThere and EveryWhere boekje.indd 70 09-02-15 08:30

Page 75: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

71

6 7 8 9 10

-100

-50

0

50

100

Plasma C0-carnitine (umol/L) at t=0

Live

r C0-

carn

itine

flux

at t

=0(u

mol

/kg

BW

/min

)

p = 0.0138r = -0.80

1.0 1.5

-20

0

20

40

60

Plasma C2-carnitine (umol/L) at t=0

Live

r C2-

carn

itine

flux

at t

=0

(um

ol/k

g B

W/ m

in)

p = 0.0045r = 0.87

0.01 0.02 0.03 0.04

-0.2

0.0

0.2

0.4

0.6

0.8

Plasma C16-carnitine (umol/L) at t=0

Live

r C16

-car

nitin

e flu

x at

t=0

(um

ol/k

g B

W/m

in)

p = 0.0490r = 0.6485

Figure 4 Correlation between plasma concentrations and liver flux Correlations between absolute venous plasma

concentrations of C0-, C2- and C16-carnitine with liver flux on t=0.

TRANSORGAN FLUXES IN A PORCINE MODEL

K007_000000_HereThere and EveryWhere boekje.indd 71 09-02-15 08:30

Page 76: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

72

Discussion

In a catheterized porcine model we have investigated transorgan fluxes of acylcarnitines, and showed that the liver has a central role in whole body acylcarnitine metabolism, corroborating with our previous findings where plasma acylcarnitine profiles poorly re-flected tissue acylcarnitine metabolism, apart from an association between liver and plasma in the fasted state (19). Moreover, C2-carnitine prevails over free carnitine be-cause our results indicated that carnitine traffic in whole body acylcarnitine metabolism comprises predominantly of the C2-carnitine flux. Both carnitine and acetylcarnitine are transported over cell membranes by the organic cation transporter 2 (OCTN2), but it has been suggested that the presence of acetylcarnitine can inhibit the transport of carni-tine (24), suggesting a higher affinity for acetylcarnitine by OCTN2. This higher affinity for acetylcarnitine could explain why acetylcarnitine fluxes were higher than carnitine fluxes. Apart from a carnitine peak immediately after a meal, the most constant product was acetylcarnitine, of which the rates were only affected shortly after the mixed meal intervention.This finding is intriguing because acetylcarnitine can either be lipid, amino acid or car-bohydrate derived and this may change during a meal. In the fasted state acetylcarnitine is predominantly lipid-derived as FAO rates will be high. The test meal contained carbo-hydrates in excess of lipids, inducing both glucose and insulin response, changing the predominant oxidation substrate from lipids to carbohydrates. This yields acetyl-CoA which is converted to acetylcarnitine by carnitine acetyltransferase (CrAT), as ace-tyl-CoA can inhibit pyruvate dehydrogenase (PDH) activity, and thereby suppress glucose oxidation (14). Future research will be needed to clarify which substrate is responsible for acetyl-CoA and acetylcarnitine under different metabolic conditions and whether acetylcarnitine fluxes depict energy substrate or carnitine supply to tissues.The predominant positive fluxes of longer chain acylcarnitines in liver again underline the central role of liver in acylcarnitine metabolism. But if these acylcarnitines are either distributed by liver as substrate or only spill over from FAO activity in the liver is unclear. However, due to the low fluxes we consider a spill over effect to be highly likely. Last-ly, the significant relation between baseline plasma acylcarnitine concentrations and baseline liver uptake and production illustrate the relation between the plasma and liver compartment in the fasted state. Here, venous C2- and C16-carnitine plasma concentra-tions correlated positively with the flux of C2- and C16-carnitine respectively through liv-er, meaning that liver production of these acylcarnitine denominates the concentration in plasma. This corresponds with our former study where topological overlap measure plots showed an association between plasma and liver of fasted mice (19). Interestingly, the correlation between the plasma concentration and liver flux of carnitine was nega-tive and crossed the liver flux baseline axis, meaning that low plasma concentrations cor-respond with production of carnitine by the liver, whereas higher plasma concentrations correspond with uptake of carnitine by the liver. We hypothesize that this might reflect a feedback mechanism where, under a certain threshold, liver starts synthesizing carni-tine, and above this threshold the excess carnitine is taken up by liver. Again this under-

TRANSORGAN FLUXES IN A PORCINE MODEL

K007_000000_HereThere and EveryWhere boekje.indd 72 09-02-15 08:30

Page 77: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

73

pins the importance of the liver compartment in whole body acylcarnitine metabolism. The only other compartment which showed production of carnitine and acylcarnitines is the gut. It is well established that most carnitine is absorbed from the diet, which enters the circulation via the gut. Studies in pigs have shown that PPAR-alpha and sub-sequent CPT-1 activity are upregulated upon fasting (17). This PPAR-alpha activation can be necessary to orchestrate lipid oxidation pathways in the intestine itself in the fasted state, but might facilitate carnitine uptake as well. More recently a study in mice showed expression of yBBD in intestinal mucosa cells, suggesting that the intestinal tract is capa-ble of de novo synthesis of carnitine (21). However in the fasted baseline fluxes we found a negative flux in the gut, suggesting uptake of carnitine rather than production. We did find significant production of carnitine by the gut upon the meal. This might either reflect carnitine uptake from the meal or intestinal carnitine production. Although we were unable to reliably detect carnitine in the test food, we cannot exclude that the whey protein in the meal might contain carnitine. The other acylcarnitine fluxes after the meal were mostly positive and this is most evident for C3 and C5 carnitine that are amino acid derived. This may indicate direct acylcarnitine formation from the ingested macronutri-ents that are bound to carnitine in the intestinal mucosa cells. Kidney, one of the sites where carnitine is synthesized and where re-absorption of car-nitine takes place, is therefore known as a regulator of the whole body carnitine pool (16, 17, 27). Interestingly, the carnitine flux in kidney did not reflect clear production of carnitine from reabsorption. C2-carnitine was absorbed by kidney over the whole length of the experiment, probably for excretion (16). All other acylcarnitine were absorbed as well, suggesting excretion via the urine, although the kidney might also oxidize them as energy substrates. The capacity of the kidney for reabsorption is very efficient, and un-der normal dietary circumstances less than 5% of the filtered carnitine is excreted (16). Unfortunately this experimental model did not allow us to collect urine samples to mon-itor the excretion of carnitine and acylcarnitines via the urine. All together, despite the kidney’s capability of carnitine synthesis, we found the kidney to predominantly clear acylcarnitines from the circulation. We assumed that the muscle compartment has an important role in acylcarnitine me-tabolism. First, because the greatest carnitine pool in the body resides in muscle tis-sue as muscle predominantly oxidizes fatty acids when glucose reserves are depleted. Second, because acylcarnitines are being accused as potentially harmful metabolites in insulin resistance, which occurs in skeletal muscle as well. But in our previous study (19), we found that muscle acylcarnitine profiles do not correlate with acylcarnitines in other organs and compartments, and that muscle acylcarnitine profiles are hardly affected by metabolic interventions such as fasting or high fat diets (unpublished data). Indeed, the findings in this study confirm that the muscle compartment hardly interacts with other compartments or plasma. The fluxes are, apart from C2-carnitine, all very modest and not consistent, especially when considering that the concentrations of acylcarnitines in muscle are much higher compared with plasma and all other tissues. Here, C2-carnitine showed a negative flux, indicating some uptake. All together, for a compartment which contains approximately 75% of the total carnitine pool (27), the net fluxes in and out of

TRANSORGAN FLUXES IN A PORCINE MODEL

K007_000000_HereThere and EveryWhere boekje.indd 73 09-02-15 08:30

Page 78: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

74

muscle were low. We can only speculate why the muscle compartment is unaffected by all other participating organ compartments. Apparently muscle has a massive carnitine depot for FAO to take place, necessitating little, but constant influx. However, we must keep in mind that our experimental model does not consider the recycling of metab-olites, and that a balanced in- and efflux of acylcarnitines from muscle could cause a distorted reflection of the flux.

We demonstrate here that it is possible to study organ lipid fluxes in a larger animal with a metabolic phenotype comparable to humans. Our study demonstrates the importance of liver in whole body acylcarnitine metabolism, and the limited exchange with mus-cle. Moreover, we showed that acetylcarnitine is the most important metabolite when it comes to exchange of (acyl)carnitine.

References1. Agarwal R. Rapid microplate method for PAH estimation. American journal of physiology Renal physi-

ology 283: F236-241, 2002.2. An J, Muoio DM, Shiota M, Fujimoto Y, Cline GW, Shulman GI, Koves TR, Stevens R, Millington D, and

Newgard CB. Hepatic expression of malonyl-CoA decarboxylase reverses muscle, liver and whole-an-imal insulin resistance. Nat Med 10: 268-274, 2004.

3. Deutz NE, Bruins MJ, and Soeters PB. Infusion of soy and casein protein meals affects interorgan amino acid metabolism and urea kinetics differently in pigs. J Nutr 128: 2435-2445, 1998.

4. Deutz NE, Reijven PL, Athanasas G, and Soeters PB. Post-operative changes in hepatic, intestinal, splenic and muscle fluxes of amino acids and ammonia in pigs. Clin Sci (Lond) 83: 607-614, 1992.

5. Deutz NE, Ten Have GA, Soeters PB, and Moughan PJ. Increased intestinal amino-acid retention from the addition of carbohydrates to a meal. Clinical nutrition 14: 354-364, 1995.

6. Deutz NE, Welters CF, and Soeters PB. Intragastric bolus feeding of meals containing elementary, par-tially hydrolyzed or intact protein causes comparable changes in interorgan substrate flux in the pig. Clinical nutrition 15: 119-128, 1996.

7. Holland WL, Knotts TA, Chavez JA, Wang LP, Hoehn KL, and Summers SA. Lipid mediators of insulin resistance. Nutr Rev 65: S39-S46, 2007.

8. Kewalramani G, Bilan PJ, and Klip A. Muscle insulin resistance: assault by lipids, cytokines and local macrophages. Current Opinion in Clinical Nutrition and Metabolic Care 13: 382-390, 2010.

9. Koves TR, Ussher JR, Noland RC, Slentz D, Mosedale M, Ilkayeva O, Bain J, Stevens R, Dyck JR, Newgard CB, Lopaschuk GD, and Muoio DM. Mitochondrial overload and incomplete fatty acid oxidation con-tribute to skeletal muscle insulin resistance. Cell Metab 7: 45-56, 2008.

10. Luci S, Giemsa B, Kluge H, and Eder K. Clofibrate causes an upregulation of PPAR-{alpha} target genes but does not alter expression of SREBP target genes in liver and adipose tissue of pigs. American jour-nal of physiology Regulatory, integrative and comparative physiology 293: R70-77, 2007.

11. Mihalik SJ, Goodpaster BH, Kelley DE, Chace DH, Vockley J, Toledo FG, and DeLany JP. Increased levels of plasma acylcarnitines in obesity and type 2 diabetes and identification of a marker of glucolipotox-icity. Obesity (Silver Spring) 18: 1695-1700, 2010.

12. Miller ER and Ullrey DE. The pig as a model for human nutrition. Annual review of nutrition 7: 361-382, 1987.

13. Muoio DM and Koves TR. Lipid-induced metabolic dysfunction in skeletal muscle. Novartis Found Symp 286: 24-38, 2007.

14. Muoio DM, Noland RC, Kovalik JP, Seiler SE, Davies MN, DeBalsi KL, Ilkayeva OR, Stevens RD, Kheterpal I, Zhang J, Covington JD, Bajpeyi S, Ravussin E, Kraus W, Koves TR, and Mynatt RL. Muscle-Specific Deletion of Carnitine Acetyltransferase Compromises Glucose Tolerance and Metabolic Flexibility. Cell Metabolism 15: 764-777, 2012.

TRANSORGAN FLUXES IN A PORCINE MODEL

K007_000000_HereThere and EveryWhere boekje.indd 74 09-02-15 08:30

Page 79: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

75

15. Noland RC, Koves TR, Seiler SE, Lum H, Lust RM, Ilkayeva O, Stevens RD, Hegardt FG, and Muoio DM. Carnitine insufficiency caused by aging and overnutrition compromises mitochondrial performance and metabolic control. J Biol Chem 284: 22840-22852, 2009.

16. Rebouche C. Kinetics, pharmacokinetics, and regulation of L-carnitine and acetyl-L-carnitine metab-olism. Annals of the New York Academy of Sciences 1033: 30-41, 2004.

17. Ringseis R, Wege N, Wen G, Rauer C, Hirche F, Kluge H, and Eder K. Carnitine synthesis and uptake into cells are stimulated by fasting in pigs as a model of nonproliferating species. The Journal of Nutritional Biochemistry 20: 840-847, 2009.

18. Ringseis R, Wen G, and Eder K. Regulation of Genes Involved in Carnitine Homeostasis by PPARalpha across Different Species (Rat, Mouse, Pig, Cattle, Chicken, and Human). PPAR research 2012: 868317, 2012.

19. Schooneman MG, Achterkamp N, Argmann CA, Soeters MR, and Houten SM. Plasma acylcarnitines in-adequately reflect tissue acylcarnitine metabolism. Biochim Biophys Acta, 2014.

20. Schooneman MG, Vaz FM, Houten SM, and Soeters MR. Acylcarnitines: Reflecting or Inflicting Insulin Resistance? Diabetes 62: 1-8, 2013.

21. Shekhawat PS, Sonne S, Carter AL, Matern D, and Ganapathy V. Enzymes involved in L-carnitine biosyn-thesis are expressed by small intestinal enterocytes in mice: implications for gut health. Journal of Crohn’s & colitis 7: e197-205, 2013.

22. Shulman GI. Cellular mechanisms of insulin resistance. The Journal of Clinical Investigation 106: 171-176, 2000.

23. Soeters MR, Sauerwein HP, Duran M, Wanders RJ, Ackermans MT, Fliers E, Houten SM, and Serlie MJ. Muscle acylcarnitines during short-term fasting in lean healthy men. Clin Sci (Lond) 116: 585-592, 2009.

24. Tamai I, Ohashi R, Nezu J, Yabuuchi H, Oku A, Shimane M, Sai Y, and Tsuji A. Molecular and functional identification of sodium ion-dependent, high affinity human carnitine transporter OCTN2. J Biol Chem 273: 20378-20382, 1998.

25. Ten Have GA, Bost MC, Suyk-Wierts JC, van den Bogaard AE, and Deutz NE. Simultaneous measurement of metabolic flux in portally-drained viscera, liver, spleen, kidney and hindquarter in the conscious pig. Laboratory animals 30: 347-358, 1996.

26. Ten have GAM, Bost MCF, Suyk-Wierts JCAW, van den Bogaard AEJM, and Deutz NEP. Simultaneous measurement of metabolic flux in portally-drained viscera, liver, spleen, kidney and hindquarter in the conscious pig. Laboratory animals 30: 347-358, 1996.

27. Vaz FM and Wanders RJA. Carnitine biosynthesis in mammals. Biochemical Journal 361: 417-429, 2002.28. Wanders R, Vreken P, den Boer M, Wijburg F, Van Gennip A, and Ijlst L. Disorders of mitochondrial fatty

acyl-CoA beta-oxidation. Journal of Inherited Metabolic Disease 22: 442-487, 1999.

TRANSORGAN FLUXES IN A PORCINE MODEL

K007_000000_HereThere and EveryWhere boekje.indd 75 09-02-15 08:30

Page 80: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

C 5 Acylcarnitine kinetics in a fasting- and

obesity-induced insulin resistant mouse model

Marieke G. Schooneman, Sander M. Houten, Nicolaas E.P. Deutz, Maarten R. Soeters

Submitted

K007_000000_HereThere and EveryWhere boekje.indd 76 09-02-15 08:30

Page 81: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

C 5 Acylcarnitine kinetics in a fasting- and

obesity-induced insulin resistant mouse model

Marieke G. Schooneman, Sander M. Houten, Nicolaas E.P. Deutz, Maarten R. Soeters

Submitted

K007_000000_HereThere and EveryWhere boekje.indd 77 09-02-15 08:30

Page 82: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

K007_000000_HereThere and EveryWhere boekje.indd 78 09-02-15 08:30

Page 83: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

79ACYLCARNITINE KINETICS IN INSULIN RESISTANT MICE

Abstract

Acylcarnitines are derived from mitochondrial acyl-CoA metabolism, and have been studied in relation to diet-induced insulin resistance. Acylcarnitine kinetics, such as pro-duction rates and pool sizes, may aid in the understanding of the role of acylcarnitines under different metabolic circumstances. We aimed to determine acylcarnitine kinetics in various mouse models of insulin resistance.We studied 12 lean BALB/cJ mice and 10 lean versus 10 obese C57BL/6N mice. We infused a bolus of U-13C-C16-carnitine, and one week later D3-C2-carnitine in fasted and fed mice, either fed chow or high fat diet (HFD). Blood was sampled for up to 180 minutes, and after calculation of isotope enrichment, non-compartmental analysis was done.Both C2- and C16-carnitine kinetics allow single pool model analysis. A larger C16-car-nitine pool size was found in BALB/cJ mice (p=0.017), but other kinetics were compa-rable. In C57BL/6N mice, both fasting and HFD did not affect C16-carnitine kinetics. In contrast, C2-carnitine pool size and rate of appearance (Ra) were greater in fasted chow mice (p=0.007), along with a lower elimination constant k (p=0.016). A lower C2-carnitine Ra was found during HFD (p=0.02).C2 and C16-carnitine kinetics can be analysed with a single pool model. Our results sug-gest that HFD alters mainly C2-carnitine metabolism. We speculate that these findings reflect impaired substrate switching in HFD-induced insulin resistance.

Introduction

Acylcarnitines are fatty acid oxidation (FAO) intermediates that are studied extensively in relation to fatty acid oxidation disorders (1-4), and more recently in relation to diet-in-duced metabolic derangements (5-10). For these purposes, acylcarnitines are studied in tissues like heart, liver and skeletal muscle, but many studies mainly focus on plasma acylcarnitine profiles (9, 11). However, we have shown recently that plasma profiles do not reflect tissue acylcarnitine metabolism (12). Only a small fraction of the total acylcarni-tine pool is present in plasma, whereas the rest resides in tissues (12, 13). It is unknown how plasma concentrations relate to acylcarnitine transport between plasma and tis-sues. As only a few organs are capable of carnitine synthesis, and carnitine mainly is ab-sorbed from the diet, an efficient system for transportation and distribution of carnitine and acylcarnitines between tissues and plasma is required.Liver, kidney and brain are capable of de novo carnitine synthesis, as these organs con-tain the gamma-butyrobetaine dioxygenase (BBD) enzyme which catalyses the last step of carnitine synthesis from its precursor gamma-butyrobetaine (13). Carnitine is trans-ported into cells via the sodium dependent transporter OCTN2, found in fat oxidizing tis-sues (14, 15). OCTN2 also transports C2-carnitine and gamma-butyrobetaine with a higher affinity compared to carnitine, and their transport is competitively inhibited by carnitine (15). Once inside the cell, carnitine binds to an activated fatty acid in exchange for CoA by carnitine-palmitoyltransferase 1 (CPT-1) on the outer mitochondrial membrane, and the

K007_000000_HereThere and EveryWhere boekje.indd 79 09-02-15 08:30

Page 84: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

80 ACYLCARNITINE KINETICS IN INSULIN RESISTANT MICE

resulting acylcarnitine subsequently crosses the mitochondrial inner membrane via the carnitine-acylcarnitine-transporter (CACT). Here, carnitine-palmitoyltransferase 2 (CPT-2) exchanges the FFA again for a CoA, forming acyl-CoA which enters the beta-oxidation for energy production (3, 13).In the last decade there have been several studies proposing a theory of lipotoxicity, where intracellular lipids cause stress within the cytosol with impairment of the insulin signalling cascade and the induction of insulin resistance (5, 16-18). Within this concept, acylcarnitines were also proposed to interfere with insulin signalling (5, 6, 9). The high lipid flux in obesity could increase rates of incomplete FAO, causing an accumulation of acylcarnitines, which could distort insulin signalling in the cytosol and flow back into the plasma, as reflected by increased plasma acylcarnitine levels in obese, insulin re-sistant subjects (6, 9). To improve the interpretation of plasma acylcarnitines and how their appearance and concentration relates to the total acylcarnitine pool in tissues, we used stable acylcarnitine isotopes to study these metabolites in vivo as has been done from other metabolic processes in both human and animal studies (19-23). Here C16-car-nitine has been related to obesity-induced insulin resistance, whereas C2-carnitine is proposed to play a crucial role in switching between lipid and glucose metabolism (24, 25). We studied acylcarnitine metabolism with the stable isotopes U-13C-C16-carnitine and D3-C2-carnitine in fed and fasted, lean and obese mice. We infused these tracers in both BALB/cJ and C57BL/6N mice that differ in insulin sensitivity (26) to gain more insight in FAO in both insulin sensitivity and insulin resistance.

Methods

Tracer studies in lean miceTracer experiments were first performed in 12 lean and insulin sensitive BALB/cJ mice (26), half of which were fasted overnight whereas the other half were fed at libitum. At 8am after the overnight fast, conscious mice were weighed and one baseline blood sample of 10μL was drawn via tail bleeding. After blood sampling, mice were placed in a mouse restrainer and a bolus infusion of 125 μL of U-13C-C16-carnitine solution (1μg/mL in 0.9% NaCl) was injected via the tail vein. Blood was sampled via tail bleeding after 5, 10, 15, 20 and 30 minutes upon infusion. Blood samples of 10 μL were diluted in 90 μL of NaCl with EDTA (5.5 mmol/L in NaCl 0.9%) and directly placed on ice.

Tracer studies in lean versus obese miceWe repeated the experiment in 20 C57BL/6N mice that are more prone to develop in-sulin resistance (27). Mice were fed either chow or HFD for 8 weeks prior to the isotope infusions. After the diet we first performed an intra-peritoneal glucose tolerance test (IPGTT) to assess the insulin sensitivity upon the high fat diet in comparison with the con-trol group. After an overnight fast (~17 hours), mice were weighed and blood was taken via tail bleeding for a baseline (t=0) blood glucose level measurement (Bayer Contour). Subsequently 2g/kg bodyweight D-glucose (20% D-glucose in 0.9% NaCl) was injected

K007_000000_HereThere and EveryWhere boekje.indd 80 09-02-15 08:30

Page 85: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

81ACYLCARNITINE KINETICS IN INSULIN RESISTANT MICE

intraperitoneally. Blood glucose levels were determined after 15, 30, 45, 60, 90 and 120 minutes. Subsequently, one week after the glucose tolerance test, they were infused with U-13C-C16-carnitine and additionally one week later D3-C2-carnitine in fasted versus fed mice. Upon these infusions we prolonged the period of blood sampling which was performed after 15, 30, 45, 60, 90, 120 and 180 minutes, to secure the detection of pos-sible delayed effects after infusion.

Laboratory analysesFor measurement of the tracer/tracee ratio (TTR), we centrifuged the diluted blood samples and transferred 50 μL of diluted plasma into clean tubes. The plasma samples were deproteinized by addition of 500 μl acetonitrile (ACN) and subsequent vortex mix-ing. Next, samples were centrifuged for 10 minutes at 4°C at a speed of 20,000 g. The supernatant was transferred into 4 ml glass vials and evaporated under a stream of ni-trogen at 40°C. After evaporation, the residue was dissolved in 100 μl ACN, vortex mixed and transferred to Gilson vials for HPLC mass spectrometric analysis (Waters/Micromass Quattro Premier XE). TTRs were further calculated by dividing the peak area of the tracer (U-13C-C16- or D3-C2-carnitine) by the peak area of the endogenous tracee (C16- or C2-carnitine).

Calculations and Statistical AnalysisAdditional analyses and statistics were performed using Microsoft Excel 2011 version 14.4.4 and Graphpad Prism software version 6.0c. First, Excel was used for the inventory of mouse data, including body weight, bolus infusion volume (absolute and corrected for lean body mass) and the calculation of TTRs. Graphpad was used to generate and fit non-linear regression curves of the individual TTRs.We determined the proposed model for decay of the curve in Graphpad Prism. From the fitted curves we were able to determine the initial enrichment (Y0) and calculate the estimated pool size (Q), the elimination constant from the pool (k) and the rate of appearance (Ra). We observed that the data were best fitted in a single pool model. Ra was calculated as Ra = k • Q (28).Q is expressed as μmol/mouse, and the dose was the absolute amount of tracer in μmol, which was infused at time point 0. Additionally we calculated the anticipated Y0 as Trac-er dose/Total absolute [C16-carnitine](μmol/mouse). Total absolute [C16-carnitine] was calculated using the anticipated blood volume and lean body mass. Both blood volume and lean body mass are estimates based on the total body mass measured prior to iso-tope infusion. Mann-Whitney tests were performed to detect effects in the different mouse groups from the fasting or diet intervention. All data are expressed as means ± SEM. P-values < 0.05 are considered as statistically significant and depicted as *, and p-values < 0.01 are depicted as **.

K007_000000_HereThere and EveryWhere boekje.indd 81 09-02-15 08:30

Page 86: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

82

Results

Prior to the tracer infusion studies, we performed an intraperitoneal glucose tolerance test (IPGTT) in the C57BL/6N mice to monitor whether the HFD induced insulin resis-tance. Indeed, the mice on HFD showed impaired glucose tolerance when compared to the chow fed controls (p < 0.05 on all GTT time points, data not shown).

Y0: estimated ratio of enrichment by the tracer on time point zeroWe first determined the Y0, the estimated ratio of enrichment by the tracer on time point 0, from the slope of the TTR curves. Y0 values were high in all experiments. In the BALB/cJ mice, the Y0 was overall greater than 1, meaning that the infused amount of tracer ex-ceeded the endogenous plasma pool (figure 1a). The Y0 for C16-carnitine differed signifi-cantly between fed and fasted BALB/cJ mice (fed Y0: 4.13 ± 0.78 and fasted Y0: 1.26 ± 0.38, p=0.01). Thus, the plasma pool of endogenous C16-carnitine in the fasted state was ~3.3 times greater compared with the fed state.In the C57BL/6N mice, the Y0 for C16-carnitine was overall greater than 1 as well, and did not differ between all four groups (fed chow: Y0 5.81 ± 1.56; fasted chow: Y0 3.10 ± 0.92; fed HFD: Y0 5.44 ± 0.20; and fasted HFD: Y0 3.11 ± 0.81). This shows that fasting and HFD do not alter the endogenous C16-carnitine pool size of C57BL/6N mice opposed to BALB/cJ mice (figure 1b).The C2-carnitine Y0 of the chow fed C57BL/6N mice was much lower and did differ sig-nificantly between fed and fasted C57BL/6N mice (fed chow: Y0 1.59 ± 0.27 versus fasted chow: Y0 0.40 ± 0.06, p=0.008) but not in the HFD group (fed HFD: Y0 1.28 ± 0.36 ver-sus fasted Y0 mean TTR 1.16 ± 0.74) (figure 1c). Apparently, fasting induces a rise in the C2-carnitine concentration in lean mice as reflected by the lower Y0, which does not occur in HFD-fed obese mice. This corresponds with the results from our earlier study in which fasting induced a rise in the plasma C2-carnitine concentration in lean C57BL/6N mice and BALB/cJ mice (12).

Anticipated and actual pool size (Q)First we calculated the anticipated pool size for all mice by dividing the tracer dose by the anticipated Y0 value. When the actual pool size is compared with the anticipated pool size, we found that the actual pool is much larger (data not shown). The anticipated pool size is an estimate of the pool present in the plasma compartment. This difference between this estimated plasma pool size and the total pool residing in tissue, is con-sistent with the difference in measured concentrations of C2-carnitine and C16-carni-tine in both plasma and tissues of fasted and fed BALB/cJ and C57BL/6N mice (12). We have calculated the ratios between the actual pool size and the anticipated pool size of C16-carnitine and C2-carnitine (Supplemental figure S1a and S1b). For C16-carnitine we found ratios ranging from 5 in fed BALB/cJ mice to 48 in fasted BALB/cJ mice. In agreement with the pool sizes, ratios were overall much greater in fasted mice (Sup-plemental figure S1a), indicating that a greater pool size in plasma is accompanied by a greater fast-exchanging pool outside of the plasma as well. The ratio differed significantly

ACYLCARNITINE KINETICS IN INSULIN RESISTANT MICE

K007_000000_HereThere and EveryWhere boekje.indd 82 09-02-15 08:30

Page 87: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

83

between fasted and fed BALB/cJ mice (p=0.004) and fasted and fed HFD C57BL/6N mice (p=0.008). For C2-carnitine we found ratios ranging from 15 in chow fed C57BL/6N mice to 149 in HFD fasted C57BL/6N mice (Supplemental figure S1b). Here the ratio was signifi-cantly greater in fasted chow C57BL/6N mice compared to fed (p=0.008), and in fed HFD C57BL/6N mice compared to fed chow mice (p=0.03).The pool size of C16-carnitine in BALB/cJ mice was significantly greater in the fasted state compared to the fed (fed: 0.0040 μmol/mouse ± 0.0017 versus fasted: 0.0118 μmol/mouse ± 0.0021, p=0.0173)(figure 2a). In C57BL/6N mice, pool sizes of C16-carnitine did not differ between all groups (figure 2b). However for C2-carnitine, the pool size was significantly greater in fasted chow mice compared to fed chow mice (chow fed C2-car-nitine pool size Q 0.14 μmol/mouse ± 0.03 and chow fasted C2-carnitine pool size Q 0.54

FASTED BALB/cJ FED BALB/cJ0

1

2

3

4

5

6Y0

C16

-car

nitin

e*

FASTED CHOW FED CHOW FASTED HFD FED HFD012345678

Y0 C

16-c

arni

tine

FASTED CHOW FED CHOW FASTED HFD FED HFD0

1

2

Y0 C

2-ca

rniti

ne **

Figure 1 Y0 in fed versus fasted BALB/cJ and C57Bl6N mice (A) C16-carnitine Y0 in fed vs fasted BALB/cJ mice, (B)

C16-carnitine Y0 and (C) C2-carnitine Y0 in fed vs fasted Bl6 mice. Data expressed as means ± SEM. P values < 0.05 are

depicted as * and < 0.01 as **.

ACYLCARNITINE KINETICS IN INSULIN RESISTANT MICE

K007_000000_HereThere and EveryWhere boekje.indd 83 09-02-15 08:30

Page 88: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

84

μmol/mouse ± 0.09, p=0.007)(figure 2c). There was no statistical difference between C2-carnitine pool sizes of HFD mice.

Elimination constant k (1/min)K values were calculated by non-compartmental analysis of the TTR curves. In BALB/cJ mice, k values for C16-carnitine were overall much lower when compared to the k values for C16-carnitine in C57BL/6N mice (figure 3a and 3b) while the pool size of C16-carni-tine was higher in BALB/cJ mice compared to C57BL/6N mice (figure 2a and 2b). Both in the BALB/cJ mice and the C57BL/6N mice the k values for C16-carnitine did not differ, meaning that HFD or fasting does not affect the capacity for eliminating or process-ing C16-carnitine in both BALB/cJ and C57BL/6N mice (figure 3a and 3b). However for C2-carnitine the fasting intervention did have a significant effect on the k value in the

FASTED BALB/cJ FED BALB/cJ0.000

0.005

0.010

0.015

Pool

size

Q C

16-c

arni

tine

(um

ol/m

ouse

)

*

FASTED CHOW FED CHOW FASTED HFD FED HFD0.000

0.002

0.004

0.006

0.008

Pool

size

Q C

16-c

arni

tine

(um

ol/m

ouse

)

FASTED CHOW FED CHOW FASTED HFD FED HFD0.0

0.2

0.4

0.6

0.8

Pool

size

Q C

2-ca

rniti

ne

(um

ol/m

ouse

)

**

Figure 2 Poolsize Q in fed versus fasted BALB/cJ and C57Bl6N mice. (A) C16-carnitine poolsize (umol/mouse) in fed vs fasted

BALB/cJ mice, (B) C16-carnitine poolsize and (C) C2-carnitine poolsize in fed vs fasted Bl6 mice.

ACYLCARNITINE KINETICS IN INSULIN RESISTANT MICE

K007_000000_HereThere and EveryWhere boekje.indd 84 09-02-15 08:30

Page 89: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

85

chow fed animals, where k values decreased significantly (fed chow: k 0.18 ± 0.01 vs. fasted-chow: k 0.11 ± 0.01, p=0.016) (figure 3c). In HFD mice no difference in k values upon fasting was detected (fed HFD: k 0.15 ± 0.02 and fasted HFD: k 0.10 ± 0.02).

Rate of C16 and C2-carnitine appearanceThe Ra for C16-carnitine was overall lower in BALB/cJ mice and C57BL/6N mice when compared to the Ra for C2-carnitine in C57BL/6N mice (figure 4a, b and c). As for the k values (elimination constant), both in BALB/cJ mice and C57BL/6N mice the Ra for C16-carnitine was unchanged by the fasting or diet intervention (figure 4a and 4b). How-ever, in C57BL/6N mice the Ra for C16-carnitine trended towards higher levels in fasted mice. The Ra of C2-carnitine was significantly greater in chow fasted compared to chow fed C57BL/6N mice (fasted chow Ra: 0.05 μmol/min ± 0.01 versus fed chow Ra: 0.02

FASTED BALB/cJ FED BALB/cJ0.000

0.002

0.004

0.006

0.008

0.010

K (1

/min

)

FASTED CHOW FED CHOW FASTED HFD FED HFD0.00

0.01

0.02

0.03

0.04

0.05

K (1

/min

)

FASTED CHOW FED CHOW FASTED HFD FED HFD0.00

0.05

0.10

0.15

0.20

0.25

K (1

/min

)

*

Figure 3 Elimination of acylcarnitine (A) Elimination constant k of C16-carnitine in lean BALB/cJ mice, and in (B) lean and

obese C57Bl6N mice. (C) Elimination constant k of C2-carnitine in lean and obese C57Bl6N mice.

ACYLCARNITINE KINETICS IN INSULIN RESISTANT MICE

K007_000000_HereThere and EveryWhere boekje.indd 85 09-02-15 08:30

Page 90: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

86

μmol/min ± 0.00, p=0.008), and also compared to fasted HFD C57BL/6N mice (fasted HFD Ra: 0.02 μmol/min ± 0.01, p=0.02)(figure 4c). This suggests that fasting promotes the appearance of C2-carnitine in chow mice, but this effect is diminished in HFD mice.

Curve fitting and modellingIn the BALB/cJ mice the TTR curves were mono-exponential with a very flat slope, making the curves to appear linearly shaped. This implies that the concentration of the tracee only marginally influences the uptake or disposal rate (Curves not shown). For C57BL/6N mice, we found exponentially shaped curves for both C16- and C2-carnitine (Curves not shown). This points towards fractional uptake of the tracee, where high plasma concentrations increase uptake (or disposal) as well. The curves of all mice showed single compartment decay. As acylcarnitines are proven to be exchanged between plasma and tissues (14, 29, 30), this single compartment decay means that the plasma compartment might act as a single compartment together with the interstitium and the cell cytosol of tissues.

FASTED BALB/cJ FED BALB/cJ0.0000

0.0001

0.0002

0.0003

0.0004

Ra

(um

ol/m

in)

FASTED CHOW FED CHOW FASTED HFD FED HFD0.00000

0.00005

0.00010

0.00015

0.00020

0.00025

Ra

(um

ol/m

in)

FASTED CHOW FED CHOW FASTED HFD FED HFD0.00

0.02

0.04

0.06

0.08

Ra

(um

ol/m

in) **

*

Figure 4 Rate of appearance (Ra) of acylcarnitines (A) Ra of C16-carnitine in lean BALB/cJ mice, and in (B) lean and obese

C57Bl6N mice. (C) Ra of C2-carnitine in lean and obese C57Bl6N mice.

ACYLCARNITINE KINETICS IN INSULIN RESISTANT MICE

K007_000000_HereThere and EveryWhere boekje.indd 86 09-02-15 08:30

Page 91: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

87

Discussion

Here, we describe for the first time C2- and C16-carnitine kinetics using a stable isotope tracer approach. In our tracer experiments we compared two mouse strains, BALB/cJ, known for its resistance against diet-induced glucose intolerance and derangements in lip-id metabolism and C57BL/6N mice (26, 31). We analysed C2- and C16-carnitine with a single pool model, and showed that fasting and HFD did not affect C16-carnitine kinetics in BALB/cJ and C57BL/6N mice, but did affect C2-carnitine-kinetics in chow fed C57BL/6N mice. These effects were absent in HFD mice, indicating that HFD alters C2-carnitine metabolism.

Measurements of the initial enrichment showed that in fasted BALB/cJ mice the enrich-ment was lower, meaning that the pre-existing endogenous C16-carnitine pool must have been greater in fasted compared to fed mice. This was confirmed by a significantly great-er pool size Q in fasted BALB/cJ mice, which also corresponds with our assumptions that the C16-carnitine pool is larger under fasted conditions, as FAO rates will be higher when glucose oxidation rates are lower due to fasting (12). This greater C16-carnitine pool size fits with higher C16-carnitine concentrations in plasma and some tissues of fasted BALB/cJ mice (i.e. liver, WAT but not muscle) (12). Additionally, the ratio between the anticipat-ed pool and the actual pool was greatest in fasted BALB/cJ mice. This ratio reflects the proportion of the whole body pool size in relation to the calculated plasma pool size; thus a higher ratio reflects a higher proportion of ACs outside the plasma compartment. Therefore, the whole body C16-carnitine pool is larger in fasted BALB/cJ mice than in fed BALB/cJ mice. However, in C57BL/6N mice, fasting and HFD pool sizes trended towards higher levels but did not reach significance. This may illustrate differences in FAO rate or its efficiency since BALB/cJ and C57BL/6N mice differ in insulin sensitivity. When we compare these results with our study on correlations between plasma and tissue acylcarnitine profiles, we did find an increase of C16-carnitine concentrations in plasma and certain tissues (such as liver and WAT, but not muscle) of fasted C57BL/6N mice (12). This could imply that the pool size did not change upon fasting, but that there was a shift towards the plasma compartment and some tissue compartments as well. Alternatively, our group size in the current study may have been too small to reach significance.

Although fasting or diet did not affect the C16-carnitine pool in C57BL/6N mice, the C2-carnitine pool was higher in fasted chow but not HFD C57BL/6N mice. This differ-ence in C2-carnitine pool size could simply be a reflection of FAO rates, which may differ between the chow and HFD mice, given their differences in glucose tolerance (data not shown). Alternatively, the different C2-carnitine pool between fasted chow versus HFD mice can be explained by differences in oxidative substrates. The greater C2-carnitine pool size in fasted chow C57BL/6N mice is in agreement with C2-carnitine concentrations in plasma and almost all tissues (12). The acetyl-group of C2-carnitine can either be lip-id- or carbohydrate derived since both FAO and CHO yield acetyl-CoA as substrate (32). As the contribution of either FAO or CHO to the acetyl-CoA concentration can vary be-tween insulin sensitive versus insulin resistant conditions, we assume that the difference

ACYLCARNITINE KINETICS IN INSULIN RESISTANT MICE

K007_000000_HereThere and EveryWhere boekje.indd 87 09-02-15 08:30

Page 92: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

88

in C2-carnitine pool size is caused by a difference in oxidized substrate in the chow and HFD C57Bl/6N mice.

Interestingly in BALB/cJ mice, the elimination of C16-carnitine from the pool was overall much lower when compared to C57Bl6N mice. This demonstrates that elimination of acylcarnitines is not influenced by the size of the total pool, which was greater in BALB/cJ mice. This suggests that these insulin sensitive mice eliminate less C16-carnitine than the C57BL/6N mice, which are more prone to develop insulin resistance upon fasting. Additionally, both interventions of fasting and HFD did not affect the elimination rate of C16-carnitine, meaning that conditions of insulin resistance per sé do not determine elimination rates by tissue. This demonstrates that C16-carnitine is not eliminated from the pool with the purpose of being an energy substrate. Additionally the C16-carnitine TTR curves of the BALB/cJ mice were all almost linearly shaped, supporting that the C16-carnitine disposal is almost independent of the concentration. However we should consider that the elimination of C16-carnitine might also depend on the presence of other long-chain acylcarnitines using the same transporters as C16-carnitine. Thus, con-centrations of both C16-carnitine and that of other long-chain acylcarnitines might in-fluence elimination rates.

The exponential shape of the C16- and C2-carnitine TTR curves of C57BL/6N mice sug-gests concentration-driven elimination of the tracee. According to our current knowl-edge, no transporter is known for C16-carnitine on the cell membrane. For C2-carnitine the elimination rate was lower in fasted chow C57BL/6N mice compared to their fed counterparts. Assuming that in these fasted chow mice FAO rates were higher than in fed mice, this might indicate that the greater elimination of C2-carnitine in fed mice was mainly CHO driven. This would also explain why we did not find this difference in HFD fed mice. Here impaired metabolic flexibility could cause persistently higher FAO rates in the fed mice, and less elimination of C2-carnitine. Interestingly an inverse relation between lipid oxidation and C2-carnitine was suggested by Ebeling et al., supporting our assump-tions on C2-carnitine elimination (32).In concert with these findings on the elimination rates, the Ra of C16-carnitine was much lower than the Ra of C2-carnitine. This again underlines the quantitative importance of C2-carnitine, which might be distributed as either an energy substrate for the oxidation of acetyl-CoA, or a preferential way of distributing carnitine towards tissues. As for the elimination rates, we found no effect in general of both fasting and the diet for the Ra for C16-carnitine. Hence, production or release of C16-carnitine is not stimulated by insu-lin resistant or CHO-depleted conditions. The Ra for C2-carnitine was greater in fasted chow C57BL/6N mice than in the fed. Together with the elimination rates, this reflects more production rather than uptake of C2-carnitine in the fasted state, and more uptake rather than production in the fed state. Again, in HFD mice this difference was not de-tected, pointing at impaired substrate switching in these insulin resistant mice. In addition to the elimination rates, TTR curves in all mice showed single compartment decay for both C16- and C2-carnitine. Because the total pool size was much greater

ACYLCARNITINE KINETICS IN INSULIN RESISTANT MICE

K007_000000_HereThere and EveryWhere boekje.indd 88 09-02-15 08:30

Page 93: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

89

than the anticipated plasma pool, this implies that the plasma compartment forms one fast-exchanging pool with both interstitium and tissues. Probably, the exchange of acyl-carnitines between plasma and tissue is continuous and not driven by gradients, sug-gesting that the borders of this single pool lie at the mitochondrial membrane where CPT-1 and CPT-2 attach and detach CoA-groups in exchange of carnitine and vice versa (13). On the other hand, large differences exist between plasma and tissue concentra-tions, e.g. muscle tissue contains more than 100 times the amounts of acylcarnitines compared to plasma (12). It is likely that a large part of whole body acylcarnitine content resides intra-mitochondrial.

A limitation of the current study was the administered tracer amount. In all experiments the TTR values were high, indicating that we have exceeded the tracer dosage, which should be aimed at in tracer studies. However, the whole body acylcarnitine pool is much larger than the fraction in plasma (Supplemental figure 1a and 1b), so it is unlikely that the infused tracer dosage has influenced whole body acylcarnitine metabolism.

In summary, we have shown here that the kinetics of C16- and C2-carnitine differ, and that this might illustrate their different roles. In addition we showed that the two mouse strains show different kinetics as well, probably due to their differences in insulin sen-sitivity and lipid metabolism (26, 31). We demonstrated that C16-carnitine kinetics may reflect C16-carnitine as a by-product of FAO. In contrast, C2-carnitine kinetics showed that C2-carnitine is a- quantitatively more present and b- more dependent on the met-abolic state. Here C2-carnitine might function as a carnitine disposer or alternatively an oxidative substrate, which deserves more attention regarding its role in substrate switching and regulating glucose tolerance.

References1. Wanders R, Vreken P, den Boer M, Wijburg F, Van Gennip A, Ijlst L. Disorders of mitochondrial fatty

acyl-CoA beta-oxidation. Journal of Inherited Metabolic Disease. 1999;22(4):442-87.2. Chegary M, Brinke H, Ruiter JP, Wijburg FA, Stoll MS, Minkler PE, et al. Mitochondrial long chain fatty

acid beta-oxidation in man and mouse. Biochim Biophys Acta. 2009;1791(8):806-15.3. Houten SM, Wanders RJ. A general introduction to the biochemistry of mitochondrial fatty acid be-

ta-oxidation. J Inherit Metab Dis. 2010;33(5):469-77.4. van Vlies N, Tian L, Overmars H, Bootsma A, Kulik W, Wanders R, et al. Characterization of carnitine and

fatty acid metabolism in the long-chain acyl-CoA dehydrogenase-deficient mouse. Biochemical Jour-nal. 2005;387(1):185-93.

5. Muoio DM, Koves TR. Lipid-induced metabolic dysfunction in skeletal muscle. Novartis Found Symp. 2007;286:24-38.

6. Koves TR, Ussher JR, Noland RC, Slentz D, Mosedale M, Ilkayeva O, et al. Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance. Cell Metab. 2008;7(1):45-56.

7. An J, Muoio DM, Shiota M, Fujimoto Y, Cline GW, Shulman GI, et al. Hepatic expression of malonyl-CoA decarboxylase reverses muscle, liver and whole-animal insulin resistance. Nat Med. 2004;10(3):268-74.

8. Noland RC, Koves TR, Seiler SE, Lum H, Lust RM, Ilkayeva O, et al. Carnitine insufficiency caused by aging and overnutrition compromises mitochondrial performance and metabolic control. J Biol Chem. 2009;284(34):22840-52.

9. Mihalik SJ, Goodpaster BH, Kelley DE, Chace DH, Vockley J, Toledo FG, et al. Increased levels of plasma

ACYLCARNITINE KINETICS IN INSULIN RESISTANT MICE

K007_000000_HereThere and EveryWhere boekje.indd 89 09-02-15 08:30

Page 94: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

90

acylcarnitines in obesity and type 2 diabetes and identification of a marker of glucolipotoxicity. Obe-sity (Silver Spring). 2010;18(9):1695-700.

10. Schooneman MG, Vaz FM, Houten SM, Soeters MR. Acylcarnitines: Reflecting or Inflicting Insulin Resis-tance? Diabetes. 2013;62(1):1-8.

11. Soeters MR, Sauerwein HP, Duran M, Wanders RJ, Ackermans MT, Fliers E, et al. Muscle acylcarnitines during short-term fasting in lean healthy men. Clin Sci (Lond). 2009;116(7):585-92.

12. Schooneman MG, Achterkamp N, Argmann CA, Soeters MR, Houten SM. Plasma acylcarnitines inade-quately reflect tissue acylcarnitine metabolism. Biochim Biophys Acta. 2014.

13. Vaz FM, Wanders RJA. Carnitine biosynthesis in mammals. Biochemical Journal. 2002;361(3):417-29.14. Ohashi R, Tamai I, Yabuuchi H, Nezu J-I, Oku A, Sai Y, et al. Na+-Dependent Carnitine Transport by

Organic Cation Transporter (OCTN2): Its Pharmacological and Toxicological Relevance. Journal of Pharmacology and Experimental Therapeutics. 1999;291(2):778-84.

15. Tamai I, Ohashi R, Nezu J, Yabuuchi H, Oku A, Shimane M, et al. Molecular and functional identification of sodium ion-dependent, high affinity human carnitine transporter OCTN2. J Biol Chem. 1998;273(32):20378-82.

16. Holland WL, Knotts TA, Chavez JA, Wang LP, Hoehn KL, Summers SA. Lipid mediators of insulin resis-tance. Nutr Rev. 2007;65(6 Pt 2):S39-S46.

17. Shulman GI. Cellular mechanisms of insulin resistance. The Journal of Clinical Investigation. 2000;106(2):171-6.

18. Morino K, Petersen KF, Shulman GI. Molecular mechanisms of insulin resistance in humans and their potential links with mitochondrial dysfunction. Diabetes. 2006;55 Suppl 2:S9-S15.

19. Pouteau E, Nguyen P, Ballevre O, Krempf M. Production rates and metabolism of short-chain fatty acids in the colon and whole body using stable isotopes. The Proceedings of the Nutrition Society. 2003;62(1):87-93.

20. Meissner M, Herrema H, van Dijk TH, Gerding A, Havinga R, Boer T, et al. Bile acid sequestration reduces plasma glucose levels in db/db mice by increasing its metabolic clearance rate. PLoS One. 2011;6(11):e24564.

21. Mittendorfer B, Liem O, Patterson BW, Miles JM, Klein S. What does the measurement of whole-body fatty acid rate of appearance in plasma by using a fatty acid tracer really mean? Diabetes. 2003;52(7):1641-8.

22. Deutz NE, Bruins MJ, Soeters PB. Infusion of soy and casein protein meals affects interorgan amino acid metabolism and urea kinetics differently in pigs. J Nutr. 1998;128(12):2435-45.

23. Ramos-Roman MA, Sweetman L, Valdez MJ, Parks EJ. Postprandial changes in plasma acylcarnitine con-centrations as markers of fatty acid flux in overweight and obesity. Metabolism. 2012;61(2):202-12.

24. Muoio DM, Noland RC, Kovalik JP, Seiler SE, Davies MN, DeBalsi KL, et al. Muscle-Specific Deletion of Car-nitine Acetyltransferase Compromises Glucose Tolerance and Metabolic Flexibility. Cell Metabolism. 2012;15(5):764-77.

25. Seiler SE, Martin OJ, Noland RC, Slentz DH, Debalsi KL, Ilkayeva OR, et al. Obesity and lipid stress inhibit carnitine acetyltransferase activity. J Lipid Res. 2014;55(4):635-44.

26. Farrell GC, Mridha AR, Yeh MM, Arsov T, Van Rooyen DM, Brooling J, et al. Strain dependence of diet-in-duced NASH and liver fibrosis in obese mice is linked to diabetes and inflammatory phenotype. Liver in-ternational : official journal of the International Association for the Study of the Liver. 2014;34(7):1084-93.

27. Montgomery MK, Hallahan NL, Brown SH, Liu M, Mitchell TW, Cooney GJ, et al. Mouse strain-dependent variation in obesity and glucose homeostasis in response to high-fat feeding. Diabetologia. 2013;56(5):1129-39.

28. Wolfe RRCDL. Isotope Tracers in Metabolic Research: Principles and Practice of Kinetic Analysis. 2nd ed. Hoboken, New Jersey: John Wiley & Sons; 2005.

29. Ramsay RR, Gandour RD, van der Leij FR. Molecular enzymology of carnitine transfer and transport. Bio-chim Biophys Acta. 2001;1546(1):21-43.

30. Tamai I, Ohashi R, Nezu Ji, Yabuuchi H, Oku A, Shimane M, et al. Molecular and Functional Identification of Sodium Ion-dependent, High Affinity Human Carnitine Transporter OCTN2. Journal of Biological Chemis-try. 1998;273(32):20378-82.

31. Fearnside JF, Dumas ME, Rothwell AR, Wilder SP, Cloarec O, Toye A, et al. Phylometabonomic patterns of adaptation to high fat diet feeding in inbred mice. PLoS One. 2008;3(2):e1668.

32. Ebeling P, Tuominen JA, Arenas J, Garcia Benayas C, Koivisto VA. The association of acetyl-L-carnitine with glucose and lipid metabolism in human muscle in vivo: the effect of hyperinsulinemia. Metabolism. 1997;46(12):1454-7.

ACYLCARNITINE KINETICS IN INSULIN RESISTANT MICE

K007_000000_HereThere and EveryWhere boekje.indd 90 09-02-15 08:30

Page 95: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

91

Supplemental data

Figure S1 Ratios pool size and anticipated poolsize of (A) C16-carnitine and (B) C2-carnitine.

Figure S1a

Figure S1b

CHOW FASTE

D

CHOW FED

HFD FA

STED

HFD FE

D

BALB FA

STED

BALB FE

D0

20

40

60

80

Rat

io Q

/ant

icip

ated

QC

16-c

arni

tine

**

**

CHOW FASTE

D

CHOW FED

HFD FA

STED

HFD FE

D0

50

100

150

200

250

Rat

io Q

/ant

icip

ated

QC

2-ca

rniti

ne

**

*

ACYLCARNITINE KINETICS IN INSULIN RESISTANT MICE

K007_000000_HereThere and EveryWhere boekje.indd 91 09-02-15 08:30

Page 96: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

C 6 Assessment of plasma acylcarnitines before and

after weight loss in obese subjects

Marieke G. Schooneman, Antonella Napolitano, Sander M. Houten, Graeme K. Ambler, Peter R. Murgatroyd, Sam R. Miller, Carla E.M. Hollak, Chong Y. Tan,

Samuel Virtue, Antonio Vidal-Puig, Derek J. Nunez and Maarten R. Soeters

Submitted

K007_000000_HereThere and EveryWhere boekje.indd 92 09-02-15 08:30

Page 97: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

C 6 Assessment of plasma acylcarnitines before and

after weight loss in obese subjects

Marieke G. Schooneman, Antonella Napolitano, Sander M. Houten, Graeme K. Ambler, Peter R. Murgatroyd, Sam R. Miller, Carla E.M. Hollak, Chong Y. Tan,

Samuel Virtue, Antonio Vidal-Puig, Derek J. Nunez and Maarten R. Soeters

Submitted

K007_000000_HereThere and EveryWhere boekje.indd 93 09-02-15 08:30

Page 98: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

K007_000000_HereThere and EveryWhere boekje.indd 94 09-02-15 08:30

Page 99: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

95PLASMA ACYLCARNITINES BEFORE AND AFTER WEIGHT LOSS

Abstract

Acylcarnitines, fatty acid oxidation (FAO) intermediates, are implicated in diet-induced insulin resistance and type 2 diabetes mellitus, as increased levels are found in obese insulin resistant humans. Moreover plasma acylcarnitines have been associated with clinical parameters related to glucose metabolism, such as fasting glucose levels and HbA1c. We hypothesized that plasma acylcarnitines would correlate with energy expenditure, insulin sensitivity and other clinical parameters before and during a weight loss interven-tion. We expected acylcarnitine levels to decrease upon weight loss, along with improved insulin sensitivity.We therefore measured plasma acylcarnitines in 60 obese subjects before and after a 12 week weight loss intervention, and analysed the acylcarnitine profiles in relation to clinical parameters of glucose metabolism, insulin sensitivity and energy expenditure.Surprisingly, despite amelioration of HOMA-IR, plasma acylcarnitines levels increased during weight loss. HOMA-IR, energy expenditure and respiratory exchange ratio were not related to plasma acylcarnitines. However NEFA correlated strongly with several acyl-carnitines at baseline and during the weight loss intervention (p < 0.001). In our study acylcarnitines did not correlate with clinical parameters of glucose metab-olism during weight loss, questioning the role of acylcarnitines in the etiology of insulin resistance and subsequent type 2 diabetes mellitus.

Introduction

With the increased incidence of obesity and type 2 diabetes mellitus, many studies fo-cus on the interaction between lipid and glucose metabolism, and the relationship to insulin resistance. Within the concept of lipotoxicity increased lipid levels are proposed to interfere with insulin signalling, eventually leading to hyperglycemia. However, the ex-act mechanisms and the individual lipids that induce insulin resistance have not been characterised definitively. From a cellular point of view, lipotoxicity is thought to occur on a cytosolic level via lipid overload (e.g. ceramides, gangliosides or diacylglycerol) (1-4). Reduced mitochondrial content or capacity may result in elevated intracellular lipids (3, 5). Alternatively, increased fatty acid oxidation (FAO) rates that are not followed by increased tricarboxylic acid cycle (TCA) activity have been proposed to induce insulin re-sistance via accumulation of different mitochondrial metabolites such as acylcarnitines (1, 6, 7). This incomplete FAO is suggested to be manifested by altered plasma acylcarni-tine profiles, mainly increased levels of long chain acylcarnitines (1, 7, 8). Acylcarnitines are fatty acids esterified to carnitine by the outer membrane enzyme car-nitine palmitoyl tranferase 1 (CPT1) to enable transmitochondrial membrane transport of long-chain acyl-CoAs. Inside the mitochondrion, carnitine is exchanged for CoA via the inner mitochondrial membrane enzyme CPT2. CPT2 can convert acyl-CoAs into acylcar-nitines again, which can be shuttled back into the cytosol and exported to the plasma

K007_000000_HereThere and EveryWhere boekje.indd 95 09-02-15 08:30

Page 100: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

96 PLASMA ACYLCARNITINES BEFORE AND AFTER WEIGHT LOSS

compartment, ultimately contributing to the typical circulating profile of acylcarnitines (9). Consequently, acylcarnitines are excellent indicators of altered FAO as demonstrated by conditions in which lipid oxidation rates are elevated or when lipid oxidation is im-paired (e.g. short-term fasting and FAO disorders). Metabolomic studies have shown that acylcarnitines may be implicated in insulin resistance (7, 8, 10), as elevated acylcarnitine levels are found in both rodent models of dietary insulin resistance (7) and in obese, in-sulin resistant humans (8). Also several acylcarnitine species correlate moderately with clinical markers such as BMI, plasma glucose levels and insulin sensitivity in obese hu-mans with type 2 diabetes mellitus (8, 10).Here we report on plasma acylcarnitine concentrations before and during weight loss in obese humans. Based on the association of long- and short-chain acylcarnitines with type 2 diabetes mellitus, we hypothesized that these acylcarnitine levels would decrease with concomitant improvements in insulin sensitivity. Surprisingly, we observed that decreased body weight and improvements in insulin sensitivity were accompanied by increased plasma acylcarnitine levels. Moreover, we found that plasma acylcarnitines correlate strongly with plasma non-esterified fatty acids (NEFA).

Research design and methods

Design of the studySixty obese subjects were recruited for an outpatient study on weight loss prediction that has been reported elsewhere (11). In brief, subjects aged 20-55 years and BMI 30-40 kg/m2, without type 2 diabetes mellitus, history of childhood obesity or previous bar-iatric surgery, were included. After giving written informed consent, subjects were ran-domized to one of three 12-week weight loss interventions: (1) diet (-600 kcal/day) alone, (2) the same diet with moderate exercise (~10% of daily expenditure), and (3) the same diet with the centrally acting serotonin-norepinephrine reuptake inhibitor, sibutramine, which was approved for weight loss at the time this study was conducted. During the study, subjects visited the clinical unit at 0, 4 and 12 weeks at 07:00 hours a.m. after an overnight fast for the measurement of body weight, anthropometry, indirect calorime-try and blood sampling (e.g. plasma acylcarnitine, glucose, NEFA and insulin levels). The study was approved by the protocol review panel of GlaxoSmithKline, the Cambridge Lo-cal Research Ethics Committee (08/H0308/10) and the Wellcome Trust Clinical Research Facility Scientific Advisory Board.

Body composition and energy expenditureBody weight was measured in light clothing. Body composition was analysed by DXA (GE Lunar Prodigy, software version 12.2 (GE Healthcare, Madison, WI) and quantitative mag-netic resonance (Echo MRI-AH; Echo Medical Systems, Houston TX). Indirect calorimetry was performed using a ventilated canopy calorimetry instrument (GEM Nutrition, Dares-bury, UK) with the subject lying supine for 20 minutes before the measurement. Expired gas samples were analyzed every 30 seconds for 20 minutes. Gas exchanges of O2 and

K007_000000_HereThere and EveryWhere boekje.indd 96 09-02-15 08:30

Page 101: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

97PLASMA ACYLCARNITINES BEFORE AND AFTER WEIGHT LOSS

CO2 were computed to calculate respiratory exchange ratio (RER) and energy expendi-ture (EE; kJ/min using the following formula: 15.9131 x O2 consumption + 5.2069 x CO2 production x 0.9950) (12).

Laboratory analysesPlasma acylcarnitines samples were analysed as described in our previous study (13), and were processed with Masslynx software version 4.1.Glucose was measured using a hexokinase assay. Insulin was measured using a fluoro-metric autoDELFIA immunoassay. Plasma NEFA were analysed with a NEFA-HR(2) in vitro enzymatic colorimetric method (Wako Diagnostics, Richmond VA).

Statistical analysisAt baseline, one-way ANOVA with Bonferroni correction was performed. Pearson cor-relation analyses with Bonferroni correction were used to determine if plasma acylcar-nitine levels predicted clinical parameters. When significant, multiple regression analysis was done to establish which acylcarnitine had the greatest effect on a single clinical parameter. Differences between days 0, 28 and 84 for whole group and within subgroup data were analysed using repetitive ANOVA analysis with Bonferroni correction. Statisti-cal analysis was done with SPSS statistical software program version 20.0. Data are de-picted as mean and standard deviation.To analyse if changes in plasma acylcarnitine levels over time coincided with changes in clinical parameters, we used a Bayesian hierarchical model with fixed and random effects. Individual variables were modelled by linear regression over time. For instance, changes in weight were modelled as: W_i (t)= α_i^ω+ β_i^ω t+ ε_(it )^ω Here W_i (t) is the weight of subject i at time t, α_i^ω is the mean weight of subject i, β_i^ω t is the rate of change of the subject’s weight over time t, and ε_(it )^ω is a zero mean Gaussian error term: ε_it^ω ~ N(0,σ^2). Similarly, each of the acylcarnitines of interest was modelled for each individual patient by linear regression (C_i (t)= α_i^c+ β_i^c t+ ε_(it )^c, where C is any acylcarnitine of interest). To analyse the correlation over time between individual clinical parameters (for example, weight) and acylcarnitines, we modeled β_i^ω as a lin-ear regression over β_i^c. β_i^ω= α+ ββ_i^c+ ε_i where ε_i is again a zero mean Gaussian error term. We then applied Gibbs sampling to simulate from the posterior distribu-tion using standard software (Just Another Gibbs Sampler (JAGS, version 3.4.0, http://mcmc-jags.sourceforge.net/)). We used a single Markov chain with a burn-in of 100000 sweeps and then output the values alpha and beta for a further 1000000 sweeps. The proportion of sweeps when beta is greater than zero is then the posterior probability that variables are positively correlated, and the proportion of sweeps when beta < 0 is the posterior probability that variables are negatively correlated. This procedure was repeated for all clinical parameters and acylcarnitines of interest followed by the Bon-ferroni-Holm method to allow for multiple testing (14).

K007_000000_HereThere and EveryWhere boekje.indd 97 09-02-15 08:30

Page 102: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

98

Results

Whole group clinical data and plasma acylcarnitines at baseline (day 0)Table 1 shows the clinical characteristics of the subjects.and the levels of plasma acyl-carnitines, of which plasma C16- and C18:1-carnitine levels of our obese subjects were below clinical laboratory reference values. The levels of other acylcarnitines detected within the spectrum are given in the online supplemental data (Supplemental table 1).

Total Diet Exercise Sibutramine

Subjects (N) 60 20 21 19

Age (years) 40 (± 8.6) 41 (± 7) 41 (± 8) 40 (± 11)

Sex (m/f) 23/37 10/10 7/14 6/13

Bodyweight (kg) 100.9 (± 12.6) 105.0 (± 12.7) 98.7 (± 11.7) 99.2 (± 13.0)

BMI (kg/m2) 34.8 (± 2.7) 35.2 (± 2.2) 34.5 (± 3.0) 34.8 (± 2.8)

Fat mass (kg) 43.8 (± 7.6) 43.0 (± 9.1) 41.4 (± 11.5) 43.2 (± 8.4)

Lean mass (kg) 52.2 (±10.9) 56.4 (± 11.7) 52.2 (± 10.9) 51.0 (± 9.7)

EE (kJ/min) 5.2 (± 0.8) 5.4 (± 0.7) 5.2 (± 0.9) 5.1 (± 0.8)

RER 0.81 (± 0.05) 0.81 (± 0.04) 0.79 (± 0.04) 0.84 (± 0.06)a

FPG (mmol/L) 5.6 (± 0.5) 5.8 (± 0.5) 5.4 (± 0.3) 5.7 (± 0.6)a

Insulin (pmol/L) 14.7 (± 10.1) 13.2 (± 11.3) 13.2 (± 6.2) 17.9 (± 11.9)

HOMA-IR 3.7 (± 2.9) 3.5 (± 3.4) 3.2 (± 1.6) 4.6 (± 3.3)

NEFA (mmol/L) 0.5163 (±0.18) 0.49 (± 0.16)b 0.50 (± 0.18) 0.56 (± 0.19)a

Acylcarnitines Ref values

C0 (µmol/L) 32.9 (± 7.8) 33.5 (± 7.6) 34.3 (± 6.8) 31 (± 9.1) 22.30 – 54.80

C2 (µmol/L) 4.86 (± 1.68) 4.55 (± 0.96) 5.06 (± 1.13) 4.96 (± 2.56) 3.40 – 13.00

C4OH (µmol/L) 0.026 (± 0.021) 0.02 (± 0.012) 0.027 (± 0.012) 0.03 (± 0.031) 0.00 – 0.15

C10 (µmol/L) 0.18 (±0.11) 0.16 (± 0.10) 0.020 (± 0.13) 0.18 (± 0.08) 0.04 – 0.30

C14:1 (µmol/L) 0.079 (±0.035) 0.07 (± 0.03) 0.085 (± 0.036) 0.08 (± 0.036) 0.02 – 0.18

C16 (µmol/L) 0.024 (± 0.012) 0.021 (± 0.008) 0.026 (± 0.014) 0.024 (± 0.011) 0.06 – 0.24

C18:1 (µmol/L) 0.028 (±0.009) 0.029 (± 0.008) 0.028 (± 0.009) 0.028 (± 0.012) 0.06 – 0.28

Table 1 Baseline characteristics, clinical parameters and plasma acylcarnitine levelsBMI, body mass index; EE, energy expenditure; RER, respiratory exchange ratio; FPG, fasting plasma glucose; HOMA-IR,

homeostatic model assessment of insulin resistance; NEFA, non-esterified fatty acids. Acylcarnitine reference values were

acquired and validated by Laboratory Genetic Metabolic Diseases, Academic Medical Center, Amsterdam. Data are

represented as mean ± standard deviation; a = statistical significant between exercise and sibutramine, b = significant

between exercise and placebo.

To detect relationships between plasma acylcarnitines and clinical parameters at base-line, we performed Spearman correlation analyses. Our analysis showed that plasma NEFA correlated strongly with C2, C4OH, C14:1, C16 and C18:1-carnitine (Table 2 and figure 1). No other correlations were found (Supplemental table 2). Multiple regression identified C14:1 and C16-carnitine as contributors to the variation in plasma NEFA (Ta-ble 3). C4OH-carnitine is detectable after an overnight fast, but emerges predominantly during starvation (15). Therefore we repeated the regression analysis without C4OH-car-nitine after which only C16-carnitine remained significant.

PLASMA ACYLCARNITINES BEFORE AND AFTER WEIGHT LOSS

K007_000000_HereThere and EveryWhere boekje.indd 98 09-02-15 08:30

Page 103: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

99

FIGURE 1 Spearmans correlations between specific plasma acylcarnitine levels and plasma non esterified fatty acids (NEFA) for the whole group on day 0. The significance level shown is after Bonferroni correction.

0.0 0.5 1.0 1.50

20

40

60

80

C0

NEFA (mmol/L)

Acy

lcar

nitin

es(μ

mol

/L)

r 0.24, p 0.07

C4OH

0.0 0.5 1.0 1.50.00

0.05

0.10

0.15

0.20

NEFA (mmol/L)

Acy

lcar

nitin

es(μ

mol

/L)

r 0.40, p 0.00

C16

0.0 0.5 1.0 1.50.00

0.05

0.10

0.15

0.20

NEFA (mmol/L)

Acy

lcar

nitin

es(μ

mol

/L)

r 0.60, p 0.00

C2

0.0 0.5 1.0 1.50

5

10

15

NEFA (mmol/L)A

cylc

arni

tines

(μm

ol/L

)

r 0.50, p 0.00

C14:1

0.0 0.5 1.0 1.50.00

0.05

0.10

0.15

0.20

NEFA (mmol/L)

Acy

lcar

nitin

es(μ

mol

/L)

r 0.53, p 0.00

C18:1

0.0 0.5 1.0 1.50.00

0.05

0.10

0.15

0.20

NEFA (mmol/L)

Acy

lcar

nitin

es(μ

mol

/L)

r 0.46, p 0.00

PLASMA ACYLCARNITINES BEFORE AND AFTER WEIGHT LOSS

K007_000000_HereThere and EveryWhere boekje.indd 99 09-02-15 08:30

Page 104: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

100

Plasma NEFA

Spearmans rho p

C0 0.24 0.07

C2 0.50 0.00 *

C4OH 0.40 0.00 *

C10 0.22 0.11

C14:1 0.53 0.00 *

C16 0.60 0.00 *

C18:1 0.46 0.00 *

Table 2 Correlations of plasma NEFA with plasma acylcarnitines Spearman correlation analysis shows a strong correlation

of C2-, C4OH-, C14:1-, C16- and C18:1-carnitine with plasma NEFA at baseline. P-values are shown before Bonferroni

correction whereas * refers to p-values that remained significant after Bonferroni correction.

Analysis including C4OH-carnitine Analysis excluding C4OH-carnitine

Unstandardized Coefficient B

p Unstandardized Coefficient B

p

C0 -0.01 0.19 C0 -0.00 0.54

C2 0.05 0.12 C2 0.01 0.72

C4OH -3.40 0.08

C10 -0.39 0.16 C10 -0.25 0.36

C14:1 2.55 0.04 * C14:1 1.84 0.11

C16 2.51 0.04 * C16 3.07 0.01 *

C18:1 -0.13 0.91 C18:1 -0.16 0.89

Table 3 Multiple regression analysis of plasma NEFA levels and selected plasma acylcarnitines Multiple regression analysis

illustrates that variation in plasma NEFA is statistically driven by C14:1 and C16-carnitine. When C4OH-carnitine was omitted

(as it predominantly emerges during starvation), only C16-carnitine remained significant.

Whole group clinical data and plasma acylcarnitines during the weight loss interventionFigure 2 shows the effects of the weight loss interventions for the whole group, irre-spective of the treatment allocation. Overall, weight decreased between days 0 and 84 (~4.5 kg). As a consequence, BMI decreased between 0 and 28 days, with no additional reduction between days 28 and 84. HOMA-IR, FPG and plasma insulin levels improved between days 0 and 28, with no further improvement hereafter (figure 2). Plasma NEFA levels were unaffected by the intervention. Fat mass continued to decrease during the entire weight loss intervention period although this effect was most pronounced during the first 28 days. Lean body mass decreased (~0.6 kg) until 28 days only (figure 2). Sub-strate oxidation measurements showed that both EE and RER were lower after 28 days of treatment, with no further reduction afterwards (figure 2). FAO rates did not change upon weight loss. However protein oxidation rates and CHO rates both decreased after 28 days and remained at this level at 84 days (Supplemental figure 1). The decrease in pro-tein oxidation was minor, therefore we assume that the decrease in energy expenditure

PLASMA ACYLCARNITINES BEFORE AND AFTER WEIGHT LOSS

K007_000000_HereThere and EveryWhere boekje.indd 100 09-02-15 08:30

Page 105: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

101

FIGURE 2 Clinical parameters for the whole group on day 0, 28 and 84. HOMA-IR, homeostatic model

assessment of insulin resistance; BMI, body mass

index. Bars and whiskers represent mean and SEM. *

= P < 0.05 after Bonferroni correction.

WEIGHT

Day : 0 28 8490

95

100

105

110

** *

Kilo

gram

HOMA IR

Day : 0 28 842

3

4

5

6

**

HO

MA

IR

ENERGY EXPENDITURE

Day : 0 28 844.0

4.5

5.0

5.5

6.0*

*K

J/m

in

RESPIRATORY EXCHANGERATIO

Day : 0 28 84

0.7

0.8

0.8

0.9

**

KJ-

min

FASTING PLASMA GLUCOSE

Day : 0 28 845.0

5.2

5.4

5.6

5.8

6.0

**

mm

ol/L

INSULIN

Day : 0 28 845

10

15

20

25

*

pmol

/L

BMI

Day : 0 28 8430

32

34

36

38

40

**

kg/m

2

FAT MASS

Day : 0 28 8435

40

45

50

**

*

kg

LEAN MASS

Day : 0 28 8445

50

55

60

**

kg

WEIGHT

Day : 0 28 8490

95

100

105

110

** *

Kilo

gram

HOMA IR

Day : 0 28 842

3

4

5

6

**

HO

MA

IR

ENERGY EXPENDITURE

Day : 0 28 844.0

4.5

5.0

5.5

6.0*

*

KJ/

min

RESPIRATORY EXCHANGERATIO

Day : 0 28 84

0.7

0.8

0.8

0.9

**

KJ-

min

FASTING PLASMA GLUCOSE

Day : 0 28 845.0

5.2

5.4

5.6

5.8

6.0

**

mm

ol/L

INSULIN

Day : 0 28 845

10

15

20

25

*pm

ol/L

BMI

Day : 0 28 8430

32

34

36

38

40

**

kg/m

2

FAT MASS

Day : 0 28 8435

40

45

50

**

*

kg

LEAN MASS

Day : 0 28 8445

50

55

60

**

kg

WEIGHT

Day : 0 28 8490

95

100

105

110

** *

Kilo

gram

HOMA IR

Day : 0 28 842

3

4

5

6

**

HO

MA

IR

ENERGY EXPENDITURE

Day : 0 28 844.0

4.5

5.0

5.5

6.0*

*

KJ/

min

RESPIRATORY EXCHANGERATIO

Day : 0 28 84

0.7

0.8

0.8

0.9

**

KJ-

min

FASTING PLASMA GLUCOSE

Day : 0 28 845.0

5.2

5.4

5.6

5.8

6.0

**

mm

ol/L

INSULIN

Day : 0 28 845

10

15

20

25

*

pmol

/L

BMI

Day : 0 28 8430

32

34

36

38

40

**

kg/m

2

FAT MASS

Day : 0 28 8435

40

45

50

**

*

kg

LEAN MASS

Day : 0 28 8445

50

55

60

**

kg

WEIGHT

Day : 0 28 8490

95

100

105

110

** *

Kilo

gram

HOMA IR

Day : 0 28 842

3

4

5

6

**

HO

MA

IR

ENERGY EXPENDITURE

Day : 0 28 844.0

4.5

5.0

5.5

6.0*

*

KJ/

min

RESPIRATORY EXCHANGERATIO

Day : 0 28 84

0.7

0.8

0.8

0.9

**

KJ-

min

FASTING PLASMA GLUCOSE

Day : 0 28 845.0

5.2

5.4

5.6

5.8

6.0

**

mm

ol/L

INSULIN

Day : 0 28 845

10

15

20

25

*

pmol

/LBMI

Day : 0 28 8430

32

34

36

38

40

**

kg/m

2

FAT MASS

Day : 0 28 8435

40

45

50

**

*

kg

LEAN MASS

Day : 0 28 8445

50

55

60

**

kg

WEIGHT

Day : 0 28 8490

95

100

105

110

** *

Kilo

gram

HOMA IR

Day : 0 28 842

3

4

5

6

**

HO

MA

IR

ENERGY EXPENDITURE

Day : 0 28 844.0

4.5

5.0

5.5

6.0*

*

KJ/

min

RESPIRATORY EXCHANGERATIO

Day : 0 28 84

0.7

0.8

0.8

0.9

**

KJ-

min

FASTING PLASMA GLUCOSE

Day : 0 28 845.0

5.2

5.4

5.6

5.8

6.0

**

mm

ol/L

INSULIN

Day : 0 28 845

10

15

20

25

*

pmol

/L

BMI

Day : 0 28 8430

32

34

36

38

40

**

kg/m

2

FAT MASS

Day : 0 28 8435

40

45

50

**

*

kg

LEAN MASS

Day : 0 28 8445

50

55

60

**

kgWEIGHT

Day : 0 28 8490

95

100

105

110

** *

Kilo

gram

HOMA IR

Day : 0 28 842

3

4

5

6

**

HO

MA

IR

ENERGY EXPENDITURE

Day : 0 28 844.0

4.5

5.0

5.5

6.0*

*

KJ/

min

RESPIRATORY EXCHANGERATIO

Day : 0 28 84

0.7

0.8

0.8

0.9

**

KJ-

min

FASTING PLASMA GLUCOSE

Day : 0 28 845.0

5.2

5.4

5.6

5.8

6.0

**

mm

ol/L

INSULIN

Day : 0 28 845

10

15

20

25

*

pmol

/L

BMI

Day : 0 28 8430

32

34

36

38

40

**

kg/m

2

FAT MASS

Day : 0 28 8435

40

45

50

**

*

kg

LEAN MASS

Day : 0 28 8445

50

55

60

**

kg

WEIGHT

Day : 0 28 8490

95

100

105

110

** *

Kilo

gram

HOMA IR

Day : 0 28 842

3

4

5

6

**

HO

MA

IR

ENERGY EXPENDITURE

Day : 0 28 844.0

4.5

5.0

5.5

6.0*

*

KJ/

min

RESPIRATORY EXCHANGERATIO

Day : 0 28 84

0.7

0.8

0.8

0.9

**

KJ-

min

FASTING PLASMA GLUCOSE

Day : 0 28 845.0

5.2

5.4

5.6

5.8

6.0

**

mm

ol/L

INSULIN

Day : 0 28 845

10

15

20

25

*

pmol

/L

BMI

Day : 0 28 8430

32

34

36

38

40

**

kg/m

2

FAT MASS

Day : 0 28 8435

40

45

50

**

*

kg

LEAN MASS

Day : 0 28 8445

50

55

60

**

kgWEIGHT

Day : 0 28 8490

95

100

105

110

** *

Kilo

gram

HOMA IR

Day : 0 28 842

3

4

5

6

**

HO

MA

IR

ENERGY EXPENDITURE

Day : 0 28 844.0

4.5

5.0

5.5

6.0*

*

KJ/

min

RESPIRATORY EXCHANGERATIO

Day : 0 28 84

0.7

0.8

0.8

0.9

**

KJ-

min

FASTING PLASMA GLUCOSE

Day : 0 28 845.0

5.2

5.4

5.6

5.8

6.0

**

mm

ol/L

INSULIN

Day : 0 28 845

10

15

20

25

*

pmol

/L

BMI

Day : 0 28 8430

32

34

36

38

40

**

kg/m

2

FAT MASS

Day : 0 28 8435

40

45

50

**

*

kg

LEAN MASS

Day : 0 28 8445

50

55

60

**

kg

WEIGHT

Day : 0 28 8490

95

100

105

110

** *

Kilo

gram

HOMA IR

Day : 0 28 842

3

4

5

6

**

HO

MA

IR

ENERGY EXPENDITURE

Day : 0 28 844.0

4.5

5.0

5.5

6.0*

*

KJ/

min

RESPIRATORY EXCHANGERATIO

Day : 0 28 84

0.7

0.8

0.8

0.9

**

KJ-

min

FASTING PLASMA GLUCOSE

Day : 0 28 845.0

5.2

5.4

5.6

5.8

6.0

**

mm

ol/L

INSULIN

Day : 0 28 845

10

15

20

25

*

pmol

/L

BMI

Day : 0 28 8430

32

34

36

38

40

**

kg/m

2

FAT MASS

Day : 0 28 8435

40

45

50

**

*

kg

LEAN MASS

Day : 0 28 8445

50

55

60

**

kg

PLASMA ACYLCARNITINES BEFORE AND AFTER WEIGHT LOSS

K007_000000_HereThere and EveryWhere boekje.indd 101 09-02-15 08:30

Page 106: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

102

mainly resulted from lower CHO. Overall clinical improvements due to the weight loss intervention were greatest in the first 28 days, with no further improvement at 84 days. Whole group plasma acylcarnitine levels for days 0, 28 and 84 are shown in figure 3. The weight loss intervention increased carnitine levels after 84 days. C2-carnitine and C4OH-carnitine were higher after 28 days with no further increase at 84 days. C14:1- and

FIGURE 3 Plasma acylcarnitine levels for the whole group on day 0, 28 and 84. Bars and whiskers represent mean

and SEM. * = P < 0.05 after Bonferroni correction.

FREE CARNITINE

Day : 0 28 8410

20

30

40 **

umol

/L

C2-CARNITINE

Day : 0 28 842

4

6

8

**

umol

/L

C4OH-CARNITINE

Day : 0 28 84

0.02

0.04

0.06

0.08

**

umol

/L

C10-CARNITINE

Day : 0 28 840.1

0.2

0.3

0.4

**

*

umol

/L

C14:1-CARNITINE

Day : 0 28 84

0.10

0.15

0.20

umol

/L **

*

C16-CARNITINE

Day : 0 28 84

0.06

0.08

0.10

0.12

0.14

umol

/L * *

C18:1-CARNITINE

Day : 0 28 84

0.06

0.08

0.10

0.12

0.14

umol

/L

**

*

FREE CARNITINE

Day : 0 28 8410

20

30

40 **

umol

/L

C2-CARNITINE

Day : 0 28 842

4

6

8

**

umol

/L

C4OH-CARNITINE

Day : 0 28 84

0.02

0.04

0.06

0.08

**

umol

/L

C10-CARNITINE

Day : 0 28 840.1

0.2

0.3

0.4

**

*

umol

/L

C14:1-CARNITINE

Day : 0 28 84

0.10

0.15

0.20

umol

/L **

*

C16-CARNITINE

Day : 0 28 84

0.06

0.08

0.10

0.12

0.14

umol

/L * *

C18:1-CARNITINE

Day : 0 28 84

0.06

0.08

0.10

0.12

0.14

umol

/L

**

*

PLASMA ACYLCARNITINES BEFORE AND AFTER WEIGHT LOSS

K007_000000_HereThere and EveryWhere boekje.indd 102 09-02-15 08:30

Page 107: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

103

C18:1-carnitine showed a similar pattern over time with an initial increase after 28 days, followed by a decrease at 84 days, although at this point these levels were still higher compared with baseline. C16-carnitine was higher after 28 days followed again by a de-crease to baseline levels (figure 3).We used a Bayesian hierarchical model to investigate whether changes in plasma acylcarni-tine levels over time correlated with changes in weight, fat mass, lean mass, HOMA-IR, FPG, NEFA, RER and EE. We found that the increase over time in C4OH-, C16- and C18:1-carnitine correlated significantly with a reduction in both total and lean body mass over time (Sup-plemental table 2 and supplemental figure 2). Additionally over time NEFA levels correlated positively with the level of C16-carnitine (Supplemental table 2 and supplemental figure 2). We found no significant correlations for the other clinical parameters (data not shown).

Subgroup clinical data and plasma acylcarnitines at baseline (day 0)Table 1 shows the demographic data at baseline for the subgroups. Although in general the groups did not differ, there were a few exceptions prior to the intervention that were primarily driven by the exercise and sibutramine group. Plasma glucose and resting RER were both higher in the sibutramine group when compared to the exercise group. A dif-ference in RER was also found for FAO and CHO rates (Table 1). Plasma NEFA levels were lower in the exercise group when compared to the other two groups (Table 1).

Subgroup clinical data and plasma acylcarnitines during the weight loss interventionThe sibutramine group showed continued weight loss up to day 84 in contrast to the other two groups that only showed weight loss at day 28, but then remained stable up to day 84. The sibutramine group had a reduction in HOMA-IR at day 28, after which the levels plateaued in contrast to the exercise and placebo groups where HOMA-IR did not change during the study. Plasma NEFA did not change in any of the three groups. In the exercise and placebo groups, RER was not affected by the weight loss intervention, but the sibutramine group had a lower RER after 28 days that remained stable thereafter.Plasma acylcarnitine levels showed differential changes during the weight loss interven-tion. Overall the greatest effect was seen in the sibutramine group and a modest effect was seen for the exercise group. In the placebo group, carnitine did not change, whereas the exercise group showed a modest increase at day 84 compared to day 28. In the sibutra-mine group, carnitine increased steadily during the intervention becoming significant at day 84. In contrast to the placebo and exercise group, C2- and C4OH-carnitine levels in the sibutramine group showed an increase at 28 days and remained high at 84 days. In the placebo and sibutramine group, C10- and C14:1-carnitine initially increased at day 28 but not in the exercise group. C16- and C18:1-carnitine were higher at day 28 compared to day 0 in all groups, with the exception of C16-carnitine in the exercise group. At day 84, C10- and C18:1-carnitine were decreased again in the placebo group compared to day 28.We repeated the Bayesian hierarchical modelling to determine if changes in plasma acyl-carnitine levels over time correlated with changes in clinical parameters and found that the change in plasma C4OH- and C18:1-carnitine remained significantly correlated with weight change in the sibutramine group, but not in the other groups (data not shown).

PLASMA ACYLCARNITINES BEFORE AND AFTER WEIGHT LOSS

K007_000000_HereThere and EveryWhere boekje.indd 103 09-02-15 08:30

Page 108: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

104

Discussion

It has been suggested that acylcarnitines play a role in insulin resistance (1, 16). In this study we have shown in obese humans that upon weight loss the improvement in insulin sensitivity was accompanied by a significant increase in plasma acylcarnitine levels. Ad-ditionally, acylcarnitines correlated positively with NEFA at baseline and over time. Several studies have reported that increased plasma acylcarnitine levels associate with obesity and insulin resistance (7, 8, 10, 17). Adams et al reported that C2-carnitine, de-rived from both lipid and carbohydrate oxidation, correlated positively with HbA1c in diabetic subjects (10). Mihalik et al showed multiple correlations of short and longer chain acylcarnitines with glucose metabolism (8). Although they did not report C2-carni-tine values, C4-dicarboxylcarnitine (C4-DC-CN) strongly correlated with fasting plasma glucose levels and HbA1c. In our study, plasma NEFA correlated positively with plasma C16-carnitine and other species, namely C2-, C14:1- and C18:1-carnitine. In the context of this strong correlation of acylcarnitines with plasma NEFA, which are known to induce in-sulin resistance, the overall absence of correlations between acylcarnitines and markers of insulin sensitivity is remarkable (18, 19). Since plasma NEFA are indicative of lipolysis, acylcarnitines in plasma may reflect white adipose tissue (WAT) breakdown (20). As a re-sult, NEFA released from WAT could drive FAO rates generating acylcarnitines. The origin of the different acylcarnitines that correlate with plasma NEFA is intriguing as well. As discussed above, C2-carnitine is derived from both lipid and carbohydrate oxidation and our observed correlation between C2-carnitine and plasma NEFA suggests lipid as main source in our subjects (21, 22). C16- and C18:1-carnitine are derived from palmitate and oleate, which are the main human dietary fatty acids. Their correlation with NEFA may support lipolysis as a responsible mechanism, since these dietary fatty acids are stored in WAT. Finally, C14:1-carnitine is an interesting acylcarnitine since it is only produced after two cycles of beta-oxidation of C18:1-CoA (23). Therefore C14:1-car-nitine is a good marker of FAO. It remains unclear what the tissue origin of this acylcarni-tine is, but as FAO also takes place in WAT, C14:1-carnitine could still be derived from WAT (13, 24). Alternatively, plasma NEFA may reflect the load of fatty acids in general, thereby correlating with the most metabolically relevant acylcarnitines.Although we focussed on the observations at baseline, we studied the effects of a weight loss intervention on acylcarnitines and insulin resistance. Effects of weight loss on acyl-carnitine profiles have been described in only two studies, both of which studied lean subjects (25, 26). Redman et al compared caloric restriction with and without exercise in non-obese men and women, and showed no changes in acylcarnitines in the exercise group, but increased acylcarnitines in the group without exercise, accompanied by a greater improvement in insulin sensitivity analogous to our study (26). Here, caloric re-striction combined with exercise possibly improves the coupling of FAO and TCA flux, preventing acylcarnitines from accumulating. In contrast to our results, Falk-Petersen et al demonstrated in lean sedentary insulin resistant offspring of parents with type 2 dia-betes (25) that plasma acylcarnitine levels do not change after a 9 week hypocaloric diet, despite weight reduction and improved insulin sensitivity. However, these subjects dif-

PLASMA ACYLCARNITINES BEFORE AND AFTER WEIGHT LOSS

K007_000000_HereThere and EveryWhere boekje.indd 104 09-02-15 08:30

Page 109: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

105

fered from ours with respect to both body composition and insulin sensitivity. Whether differences in mitochondrial lipid flux handling or lipolysis explain the differential effects of the diet on plasma acylcarnitines remains elusive. Also, in these particular subjects, the initial insulin resistance may have been a result of increased intramyocellular lipids (IMCL) rather than acylcarnitines. Finally, the genetic susceptibility of insulin resistant offspring potentially blurs the etiological mechanism of insulin resistance (27). For both studies, it should be emphasized that plasma acylcarnitines do not reliably reflect indi-vidual tissue metabolite levels as we have shown recently (13, 28). Although NEFA tended to increase after 28 days, levels at 84 days were not affected by weight loss, which can be due to improved insulin sensitivity of WAT and suppression of lipolysis. Alternatively, as weight loss diminished after 84 days this may have caused greater spread of NEFA levels.Although HOMA-IR did not correlate with acylcarnitine levels, weight loss resulted in both increased acylcarnatine levels and improved HOMA-IR. Despite this improvement in insulin sensitivity, CHO rates decreased, probably reflecting the hypocaloric state and decreased carbohydrate intake. This causes an increase in FAO, directly inhibiting CHO via pyruvate dehydrogenase (PDH)(29).The changes in individual chain lengths of acylcarnitines are important because they re-veal some insight on physiological mechanisms in acylcarnitine metabolism during weight loss. Carnitine availability depends on dietary intake and endogenous synthesis. Both carnitine uptake via OCTN2 and carnitine synthesis are regulated by PPAR-alpha and are stimulated under hypocaloric conditions (16). The absence of increased carnitine after 28 days of intervention may reflect increased esterification to fatty acids and efflux of toxic lipid metabolites. This effect was probably lessened after 84 days when weight loss decreased and carnitine levels increased. Indeed, the increase in lipid derived acylcar-nitines, including C2-carnitine, was most pronounced after the first 4 weeks. Here the hypocaloric condition and subsequent weight loss could cause improved CrAT activity and increased C2-carnitine production, and long chain acylcarnitines that are released from the mitochondria and into the plasma compartment (30). When this diet induced CrAT activity declines again over time, more carnitine is released, explaining the delay in this increase of carnitine levels. These changes over time were not seen for the amino acid derived acylcarnitines, reflecting that under hypocaloric conditions lean body mass is more protected than fat mass (31).Overall, acylcarnitines increased and insulin sensitivity improved in our study and so our results dissociate insulin sensitivity from plasma acylcarnitines. We speculate that the elevated acylcarnitine levels on day 28 and day 84 may reflect lipolysis, due to the caloric deficit of the weight loss interventions as detailed above. Additionally it could reflect higher rates of FAO and CrAT activity, although we could not confirm this with measure-ments of whole body lipid oxidation or acylcarnitines in tissue. The effects on acylcarnitine levels and the clinical parameters such as weight loss, were greatest at 28 days and weakened at 84 days. It is often seen in weight loss studies that the initial effects outweigh the later effects, which may not only be due to changes in treatment compliance over time (32-34). Under hypocaloric conditions, whole body en-ergy expenditure decreases hampering further weight loss once a lower body weight is

PLASMA ACYLCARNITINES BEFORE AND AFTER WEIGHT LOSS

K007_000000_HereThere and EveryWhere boekje.indd 105 09-02-15 08:30

Page 110: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

106

attained (35-37). The increase in acylcarnitines over time also showed a relation with weight loss.Although we have analysed and reported on whole group changes over time because of the limited number of subjects, the weight loss intervention consisted of three arms (diet, sibutramine and exercise) (11). This is important because the weight loss interven-tions have divergent effects on mitochondrial physiology as described above (at least diet vs. exercise). Given that altered mitochondrial physiology may change in acylcar-nitines, we performed whole group analyses because of the primary interest in chang-es in acylcarnitines over time. However, the different interventions are intriguing and reveal subtle but clear effects on clinical parameters, and acylcarnitine metabolism in particular. Over time the sibutramine group lost more weight and had the highest acyl-carnitine levels, in contrast to more modest effects in the other two groups. This may confirm the idea that lipolysis, which is apparently greater when weight loss is greater, is reflected as higher levels of plasma acylcarnitines. Weight loss in the other two groups was comparable, but the rise in acylcarnitines was nearly absent upon weight loss in the exercise group. This may be due to more efficient mitochondrial fatty acid oxidation due to exercise (26, 38, 39), which could reduce accumulating acylcarnitines in the plasma compartment. Although the different interventions were extremely instructive for our understanding of acylcarnitine metabolism, the subgroups do have limitations, as the groups were relatively small, the exercise intervention was only modest and the mar-keting application for sibutramine has been withdrawn due to side effects of the drug (40). Additionally, HOMA-IR may be a crude measure for insulin sensitivity although it is clinical useful.In conclusion, we have found an increase in several acylcarnitine species in association with weight loss in obese human subjects, despite improvements in insulin sensitivity. It is likely that the level of plasma acylcarnitines is driven by the rate of lipolysis as well as improved efflux from cells, potentially resulting from improved CrAT activity and not by deranged mitochondrial FAO. The diet derived acylcarnitines, C16- and C18:1-carni-tine, seem the most relevant acylcarnitines at baseline and during weight loss. However, the importance of plasma acylcarnitines as clinical markers for insulin resistance seems negligible and the interpretation of changes in these lipid intermediates in relation to clinical status remains challenging.

References1. Muoio DM, Koves TR 2007 Lipid-induced metabolic dysfunction in skeletal muscle. Novartis Found

Symp 286:24-382. Holland WL, Knotts TA, Chavez JA, Wang LP, Hoehn KL, Summers SA 2007 Lipid mediators of insulin

resistance. Nutr Rev 65:S39-S463. Morino K, Petersen KF, Shulman GI 2006 Molecular Mechanisms of Insulin Resistance in Humans and

Their Potential Links With Mitochondrial Dysfunction. Diabetes 55:S9-S154. Shulman GI 2000 Cellular mechanisms of insulin resistance. The Journal of Clinical Investigation

106:171-1765. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, Puigserver P, Carlsson E, Rid-

derstrale M, Laurila E, Houstis N, Daly MJ, Patterson N, Mesirov JP, Golub TR, Tamayo P, Spiegelman B, Lander ES, Hirschhorn JN, Altshuler D, Groop LC 2003 PGC-1 alpha-responsive genes involved in oxi-

PLASMA ACYLCARNITINES BEFORE AND AFTER WEIGHT LOSS

K007_000000_HereThere and EveryWhere boekje.indd 106 09-02-15 08:30

Page 111: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

107

dative phosphorylation are coordinately downregulated in human diabetes. Nat Genet 34:267-2736. Muoio DM, Neufer PD 2012 Lipid-Induced Mitochondrial Stress and Insulin Action in Muscle. Cell Me-

tabolism 15:595-6057. Koves TR, Ussher JR, Noland RC, Slentz D, Mosedale M, Ilkayeva O, Bain J, Stevens R, Dyck JR, Newgard

CB, Lopaschuk GD, Muoio DM 2008 Mitochondrial overload and incomplete fatty acid oxidation con-tribute to skeletal muscle insulin resistance. Cell Metab 7:45-56

8. Mihalik SJ, Goodpaster BH, Kelley DE, Chace DH, Vockley J, Toledo FG, DeLany JP 2010 Increased levels of plasma acylcarnitines in obesity and type 2 diabetes and identification of a marker of glucolipotox-icity. Obesity (Silver Spring) 18:1695-1700

9. Ramsay RR, Gandour RD, van der Leij FR 2001 Molecular enzymology of carnitine transfer and trans-port. Biochim Biophys Acta 1546:21-43

10. Adams SH, Hoppel CL, Lok KH, Zhao L, Wong SW, Minkler PE, Hwang DH, Newman JW, Garvey WT 2009 Plasma Acylcarnitine Profiles Suggest Incomplete Long-Chain Fatty Acid beta-Oxidation and Altered Tricarboxylic Acid Cycle Activity in Type 2 Diabetic African-American Women. The Journal of Nutrition 139:1073-1081

11. Napolitano A, Miller SR, Murgatroyd PR, Delafont B, Brooke A, Elkhawad M, Tan CY, Virtue S, Vidal-Puig A, Nunez DJ 2012 Prediction of weight loss and regain following dietary, lifestyle, and pharmacologic intervention. Clinical pharmacology and therapeutics 91:1027-1034

12. Elia M, Livesey G 1992 Energy expenditure and fuel selection in biological systems: the theory and practice of calculations based on indirect calorimetry and tracer methods. World review of nutrition and dietetics 70:68-131

13. Schooneman MG, Achterkamp N, Argmann CA, Soeters MR, Houten SM 2014 Plasma acylcarnitines in-adequately reflect tissue acylcarnitine metabolism. Biochim Biophys Acta

14. Holm S 1979 A Simple Sequentially Rejective Multiple Test Procedure. Scand J Statist 6:65-7015. Soeters MR, Serlie MJ, Sauerwein HP, Duran M, Ruiter JP, Kulik W, Ackermans MT, Minkler PE, Hoppel

CL, Wanders RJA, Houten SM 2012 Characterization of D-3-hydroxybutyrylcarnitine (ketocarnitine): an identified ketosis-induced metabolite. Metabolism 61:966-973

16. Schooneman MG, Vaz FM, Houten SM, Soeters MR 2013 Acylcarnitines: Reflecting or Inflicting Insulin Resistance? Diabetes 62:1-8

17. An J, Muoio DM, Shiota M, Fujimoto Y, Cline GW, Shulman GI, Koves TR, Stevens R, Millington D, Newgard CB 2004 Hepatic expression of malonyl-CoA decarboxylase reverses muscle, liver and whole-animal insulin resistance. Nat Med 10:268-274

18. Boden G, Shulman GI 2002 Free fatty acids in obesity and type 2 diabetes: defining their role in the development of insulin resistance and beta-cell dysfunction. Eur J Clin Invest 32 Suppl 3:14-23

19. Corcoran MP, Lamon-Fava S, Fielding RA 2007 Skeletal muscle lipid deposition and insulin resistance: effect of dietary fatty acids and exercise. Am J Clin Nutr 85:662-677

20. Klein S, Sakurai Y, Romijn JA, Carroll RM 1993 Progressive alterations in lipid and glucose metabolism during short-term fasting in young adult men. Am J Physiol 265:E801-806

21. Ebeling P, Tuominen JA, Arenas J, Garcia Benayas C, Koivisto VA 1997 The association of acetyl-L-car-nitine with glucose and lipid metabolism in human muscle in vivo: the effect of hyperinsulinemia. Metabolism 46:1454-1457

22. Muoio DM, Noland RC, Kovalik JP, Seiler SE, Davies MN, DeBalsi KL, Ilkayeva OR, Stevens RD, Kheterpal I, Zhang J, Covington JD, Bajpeyi S, Ravussin E, Kraus W, Koves TR, Mynatt RL 2012 Muscle-Specific Deletion of Carnitine Acetyltransferase Compromises Glucose Tolerance and Metabolic Flexibility. Cell Metabolism 15:764-777

23. Chegary M, Brinke H, Ruiter JP, Wijburg FA, Stoll MS, Minkler PE, van Weeghel M, Schulz H, Hoppel CL, Wanders RJ, Houten SM 2009 Mitochondrial long chain fatty acid beta-oxidation in man and mouse. Biochim Biophys Acta 1791:806-815

24. Laplante M, Festuccia WT, Soucy G, Gélinas Y, Lalonde J, Berger JP, Deshaies Y 2006 Mechanisms of the Depot Specificity of Peroxisome Proliferator Activated Receptor gamma Action on Adipose Tissue Me-tabolism. Diabetes 55:2771-2778

25. Petersen KF, Dufour S, Morino K, Yoo PS, Cline GW, Shulman GI 2012 Reversal of muscle insulin resis-tance by weight reduction in young, lean, insulin-resistant offspring of parents with type 2 diabetes. Proceedings of the National Academy of Sciences 109:8236-8240

26. Redman LM, Huffman KM, Landerman LR, Pieper CF, Bain JR, Muehlbauer MJ, Stevens RD, Wenner BR, Kraus VB, Newgard CB, Kraus WE, Ravussin E 2011 Effect of Caloric Restriction with and without Exer-

PLASMA ACYLCARNITINES BEFORE AND AFTER WEIGHT LOSS

K007_000000_HereThere and EveryWhere boekje.indd 107 09-02-15 08:30

Page 112: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

108

cise on Metabolic Intermediates in Nonobese Men and Women. Journal of Clinical Endocrinology & Metabolism 96:E312-E321

27. Morino K, Petersen KF, Dufour S, Befroy D, Frattini J, Shatzkes N, Neschen S, White MF, Bilz S, Sono S, Pypaert M, Shulman GI 2005 Reduced mitochondrial density and increased IRS-1 serine phosphoryla-tion in muscle of insulin-resistant offspring of type 2 diabetic parents. The Journal of clinical investi-gation 115:3587-3593

28. Soeters MR, Sauerwein HP, Duran M, Wanders RJ, Ackermans MT, Fliers E, Houten SM, Serlie MJ 2009 Muscle acylcarnitines during short-term fasting in lean healthy men. Clin Sci (Lond) 116:585-592

29. Randle PJ, Priestman DA, Mistry SC, Halsall A 1994 Glucose fatty acid interactions and the regulation of glucose disposal. Journal of cellular biochemistry 55 Suppl:1-11

30. Seiler SE, Martin OJ, Noland RC, Slentz DH, Debalsi KL, Ilkayeva OR, An J, Newgard CB, Koves TR, Muoio DM 2014 Obesity and lipid stress inhibit carnitine acetyltransferase activity. J Lipid Res 55:635-644

31. Soeters MR, Soeters PB, Schooneman MG, Houten SM, Romijn JA 2012 Adaptive reciprocity of lipid and glucose metabolism in human short-term starvation. American Journal of Physiology - Endocrinology And Metabolism 303:E1397-E1407

32. Avenell A, Brown TJ, McGee MA, Campbell MK, Grant AM, Broom J, Jung RT, Smith WC 2004 What are the long-term benefits of weight reducing diets in adults? A systematic review of randomized con-trolled trials. Journal of human nutrition and dietetics : the official journal of the British Dietetic As-sociation 17:317-335

33. Anderson JW, Konz EC, Frederich RC, Wood CL 2001 Long-term weight-loss maintenance: a me-ta-analysis of US studies. Am J Clin Nutr 74:579-584

34. Douketis JD, Macie C, Thabane L, Williamson DF 2005 Systematic review of long-term weight loss stud-ies in obese adults: clinical significance and applicability to clinical practice. International journal of obesity 29:1153-1167

35. Camps SG, Verhoef SP, Westerterp KR 2013 Weight loss, weight maintenance, and adaptive thermo-genesis. Am J Clin Nutr 97:990-994

36. Astrup A, Gotzsche PC, van de Werken K, Ranneries C, Toubro S, Raben A, Buemann B 1999 Meta-anal-ysis of resting metabolic rate in formerly obese subjects. Am J Clin Nutr 69:1117-1122

37. Rosenbaum M, Hirsch J, Gallagher DA, Leibel RL 2008 Long-term persistence of adaptive thermogen-esis in subjects who have maintained a reduced body weight. Am J Clin Nutr 88:906-912

38. Dubé JJ, Amati F, Stefanovic-Racic M, Toledo FGS, Sauers SE, Goodpaster BH 2008 Exercise-induced alterations in intramyocellular lipids and insulin resistance: the athlete’s paradox revisited. American Journal of Physiology - Endocrinology And Metabolism 294:E882-E888

39. Meex RCR, Schrauwen-Hinderling VB, Moonen-Kornips E, Schaart G, Mensink M, Phielix E, van de Wei-jer T, Sels JP, Schrauwen P, Hesselink MKC 2010 Restoration of Muscle Mitochondrial Function and Metabolic Flexibility in Type 2 Diabetes by Exercise Training Is Paralleled by Increased Myocellular Fat Storage and Improved Insulin Sensitivity. Diabetes 59:572-579

40. 2010 Sibutramine (Meridia) withdrawn. The Medical letter on drugs and therapeutics 52:88

PLASMA ACYLCARNITINES BEFORE AND AFTER WEIGHT LOSS

K007_000000_HereThere and EveryWhere boekje.indd 108 09-02-15 08:30

Page 113: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

109

Supplemental data

Body weight

RER HOMA-IR

Fat mass

FPG FAO CHO

Spearman r p r p r p r p r p r p r p

C0 0,09 0,50 -0,01 0,94 -0,04 0,77 -0,05 0,72 0,02 0,87 -0,09 0,53 -0,09 0,53

C2 0,04 0,75 0,06 0,64 0,02 0,90 -0,05 0,71 0,10 0,46 -0,02 0,90 0,03 0,80

C4OH -0,01 0,91 0,01 0,94 0,11 0,40 0,06 0,66 0,19 0,16 0,07 0,60 0,03 0,80

C10 0,03 0,83 -0,02 0,90 -0,23 0,09 0,03 0,83 0,05 0,72 0,05 0,69 0,03 0,85

C14:1 0,07 0,61 0,06 0,68 -0,13 0,32 0,03 0,82 0,23 0,08 -0,01 0,93 0,09 0,53

C16 0,13 0,31 0,24 0,08 -0,23 0,08 0,04 0,75 0,06 0,67 -0,20 0,15 0,19 0,17

C18:1 -0,03 0,82 0,08 0,54 -0,19 0,14 -0,02 0,89 0,20 0,14 -0,04 0,78 0,10 0,48

Supplemental table 1 Spearman correlation analyses of acylcarnitines with clinical parameters at baseline. None of the

clinical parameters correlated with the level of plasma acylcarnitines at baseline, except for NEFA as shown in figure 1. RER,

respiratory exchange ratio; HOMA-IR, homeostatic model assessment of insulin resistance; FPG, fasting plasma glucose;

FAO, fatty acid oxidation; CHO, carbohydrate oxidation.

Supplemental figure 1 Clinical parameters on whole group level on day 0, 28 and 84. HOMA-B, homeostatic model

assessment of beta-cell function. Bars and whiskers represent

mean and SEM.

* = P < 0.05 after Bonferroni correction.

HOMA B

Day : 0 28 8450

100

150

200

*

mgr

/L

FATTY ACID OXIDATION

Day : 0 28 842.0

2.5

3.0

3.5

4.0

KJ/

min

ns

CARBOHYDRATE OXIDATION

Day : 0 28 840.5

1.0

1.5

2.0

2.5

3.0

**

KJ/

min

PROTEIN OXIDATION

Day : 0 28 840.0

0.2

0.4

0.6

0.8

1.0

**

KJ/

min

NON ESTERIFIED FATTY ACIDS

Day : 0 28 840.0

0.2

0.4

0.6

0.8

1.0

mg/

ml

ns

HOMA B

Day : 0 28 8450

100

150

200

*

mgr

/L

FATTY ACID OXIDATION

Day : 0 28 842.0

2.5

3.0

3.5

4.0

KJ/

min

ns

CARBOHYDRATE OXIDATION

Day : 0 28 840.5

1.0

1.5

2.0

2.5

3.0

**

KJ/

min

PROTEIN OXIDATION

Day : 0 28 840.0

0.2

0.4

0.6

0.8

1.0

**

KJ/

min

NON ESTERIFIED FATTY ACIDS

Day : 0 28 840.0

0.2

0.4

0.6

0.8

1.0

mg/

ml

ns

PLASMA ACYLCARNITINES BEFORE AND AFTER WEIGHT LOSS

K007_000000_HereThere and EveryWhere boekje.indd 109 09-02-15 08:30

Page 114: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

110

Acylcarnitine Body weight NEFA

C0 0.59 0.52

C2 0.01 0.84

C4OH 0.00 * 0.95

C10 0.15 0.88

C14:1 0.01 0.99

C16 0.01 1.00 *

C18:1 0.00 * 0.99

Supplemental table 2 Correlations between changes over time in acylcarnitines with weight loss and plasma NEFA levels.

Here the posterior probability is shown that change in body weight and the change in plasma NEFA levels are positively

correlated with change in plasma acylcarnitine levels. Only C18:1- and C4OH-carnitine remained significantly (negatively)

correlated with the change in body weight at the 1% level after Bonferroni-Holm correction. For changes over time in plasma

NEFA levels, only C16-carnitine was significantly positively correlated after Bonferroni-Holm correction at the 1% level.

P-values that remained significant after Bonferroni-Holm correction are marked by *.

Supplemental figure 2Figure S2 shows 3d density plots that depict the correlation between a. weight vs C4OH, b. weight vs C18:1 and c. NEFA vs C16.

The plots show the probability distributions for the variables alpha and beta whereas the volume under the density in a region

is the probability that an event (the true value of the parameters) lies in that region (equation; Y=alpha + beta*X + error). Alpha

is a ‘nuisance’ parameter: the intercept of the line on the y-axis and beta is the slope of the line. Thus, beta = 0 means no

correlation, beta > 0 means positive correlation and beta < 0 means negative correlation. Therefore the volume under the

density curve for beta > 0 is the probability of positive correlation between the parameters.

2a. Change in body weight versus change in

C4OH-carnitine

Alpha

−0.5

−0.4

−0.3

−0.2

Beta

−100

−50

0

Density

0.0

0.1

0.2

0.3

PLASMA ACYLCARNITINES BEFORE AND AFTER WEIGHT LOSS

K007_000000_HereThere and EveryWhere boekje.indd 110 09-02-15 08:30

Page 115: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

111PLASMA ACYLCARNITINES BEFORE AND AFTER WEIGHT LOSS

2b. Change in body weight versus change in

C18:1-carnitine

2c. Change in plasma NEFA levels versus

change in C16-carnitine

Alpha

−0.5

−0.4

−0.3

−0.2

Beta

−100

−50

0

Density

0.0

0.1

0.2

0.3

0.4

0.5

Alpha

−0.010−0.005

0.0000.005

0.010

0.015

Beta

0

5

Density

0

10

20

30

40

K007_000000_HereThere and EveryWhere boekje.indd 111 09-02-15 08:30

Page 116: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

C 7 The impact of altered carnitine

availability on fatty acid and glucose metabolism in diet-induced obese mice

Marieke G. Schooneman, Riekelt H. Houtkooper, Carla E.M. Hollak, Ronald J.A. Wanders, Frederic M. Vaz, Maarten R. Soeters and Sander M. Houten

Submitted

K007_000000_HereThere and EveryWhere boekje.indd 112 09-02-15 08:30

Page 117: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

C 7 The impact of altered carnitine

availability on fatty acid and glucose metabolism in diet-induced obese mice

Marieke G. Schooneman, Riekelt H. Houtkooper, Carla E.M. Hollak, Ronald J.A. Wanders, Frederic M. Vaz, Maarten R. Soeters and Sander M. Houten

Submitted

K007_000000_HereThere and EveryWhere boekje.indd 113 09-02-15 08:30

Page 118: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

K007_000000_HereThere and EveryWhere boekje.indd 114 09-02-15 08:30

Page 119: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

115THE IMPACT OF ALTERED CARNITINE AVAILABILITY

Abstract

Acylcarnitines are fatty acid oxidation (FAO) intermediates, which have been implicated in diet-induced insulin resistance. Elevated acylcarnitine levels are found in obese, in-sulin resistant humans and rodents, and coincide with lower free carnitine. We hypoth-esized that increasing free carnitine levels by administration of the carnitine precursor gamma-butyrobetaine (gBB) could facilitate FAO, thereby improving insulin sensitivity. C57BL/6N mice were fed a high fat or chow diet with or without gBB supplementation (n=10 per group). After 8 weeks of diet, indirect calorimetry, glucose tolerance and in-sulin sensitivity tests were performed. Then animals were dissected and AC profiles and carnitine biosynthesis intermediates were analyzed in plasma and tissues by tandem mass spectrometry (MS) and liquid chromatography MS MS. gBB supplementation was unable to prevent obesity and impaired glucose homeostasis in the HFD fed mice in spite of marked alterations in the acylcarnitine profiles in plasma and liver. Remarkably, gBB did not affect other tissue acylcarnitine profiles, most notably the muscle. Administration of a bolus acetylcarnitine also caused significant changes in plasma and liver, but not in muscle acylcarnitine profiles, again without effect on glucose tolerance. Altogether, increasing carnitine availability affects acylcarnitine profiles in plasma and liver but without affecting glucose tolerance or insulin sensitivity. This may be due to the lacking effect on muscle acylcarnitine profiles, as muscle tissue is an important contrib-utor to whole body insulin sensitivity. These results warrant caution on making associa-tions between plasma acylcarnitine levels and insulin resistance.

Introduction

The obesity epidemic increases the incidence of insulin resistance and type 2 diabetes mellitus. Several lipid intermediates such as diacylglycerol, ceramides, gangliosides and non esterified fatty acids (NEFA) have been implicated in the induction of insulin resis-tance, a concept known as lipotoxicity (1, 2). In addition, this lipid burden may increase (incomplete) fatty acid oxidation (FAO) leading to an accumulation of intermediates such as acylcarnitines that could impair insulin signaling. FAO in mitochondria depends on the carnitine shuttle for transport of fatty acids into these organelles. Carnitine is absorbed from the diet, but can also be synthesized de novo. Trimethyllysine is converted to L-car-nitine in four enzymatic reactions, where in the last step gamma-butyrobetaine (gBB) is hydroxylated to carnitine (3). In humans, this only occurs in liver, kidney and brain (4). In addition to the transport of activated fatty acids into mitochondria, carnitine is also needed for the efflux of acyl-derivatives out of the mitochondria (5). Carnitine deficiency was suggested to compromise mitochondrial function and FAO thereby inducing insulin resistance (5). Both oral and intravenous administration of L-carnitine in insulin resistant humans and rodents increase free carnitine levels (5-9). In insulin resistant rats, L-carnitine supplementation restored muscle and hepatic levels

K007_000000_HereThere and EveryWhere boekje.indd 115 09-02-15 08:30

Page 120: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

116

of free carnitine, and as a result from the carnitine treatment plasma glucose levels, HOMA-IR and mitochondrial function improved (5). In humans intravenous L-carnitine treatment increased carnitine levels in muscle tissue when co-administered with insulin (6, 8), and prolonged L-carnitine treatment increased plasma levels of free carnitine and acetylcarnitine leading to an improvement of glucose handling in insulin resistant sub-jects (9). These findings, however, need to be seen in the light of a majority of studies that only showed a positive effect of L-carnitine treatment on glucose tolerance when additional caloric restriction was implemented (10). The inability to switch from FAO to carbohydrate oxidation (CHO), a concept known as metabolic inflexibility (11, 12), is characteristic for the insulin resistant state (11, 13). Car-nitine acetyltransferase (CrAT) may play a pivotal role in substrate switching and glucose tolerance via the conversion of acetyl-CoA into acetylcarnitine and thereby regulating the activity of the pyruvate dehydrogenase complex (PDH) (9). The availability of carnitine also likely influences CrAT activity. Intriguingly, infusion of acetylcarnitine in both rats and humans improved glucose uptake rates and glucose storage (14, 15).Increasing carnitine availability may facilitate FAO and efflux of acyl-derivatives from the mitochondrion, thereby improving insulin sensitivity. Since gBB effectively increases carnitine levels (16, 17), we analyzed the effects of gBB administration on lipid and glu-cose metabolism in C57BL/6N mice on a high fat diet (HFD) versus a chow control diet. Subsequently we compared the effect of gBB administration with an acute infusion of acetylcarnitine on plasma and tissue acylcarnitine profiles and glucose tolerance.

Methods

Animal studies – general40 C57BL/6N mice (Charles River Laboratories), 10 weeks of age at the time of the exper-iment, were acclimatized for one week. Then, 20 mice were switched to an eight week high fat diet (HFD, Rodent Diet with 60% kcal from fat, 20% from carbohydrates and 20% from proteins, D12492, Research Diets Inc., NJ, USA), while the remainder of the cohort kept chow diet. Half of each group was supplemented with 50mg/kg/day gBB (420 mg gBB ((3-Carboxypropyl)trimethylammonium chloride, Sigma-Aldrich) in 1L water, based on 3ml water intake daily in a 25 gram mouse (17)). This resulted in four groups of 10 mice; chow + gBB, chow - gBB, HFD + gBB and HFD - gBB. Mice were weighed weekly and every two weeks whole blood samples were taken from the saphenous vein for carnitine con-centrations to monitor the effect of the gBB treatment. All experiments were approved by the institutional review board for animal experiments at the Academic Medical Center (Amsterdam, The Netherlands).

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

K007_000000_HereThere and EveryWhere boekje.indd 116 09-02-15 08:30

Page 121: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

117

Indirect calorimetryAfter 8 weeks of diet, we performed an indirect calorimetry (Phenomaster/Labmaster, TSE systems, Bad Homberg, Germany) for 48 hours in 8 mice per group. Two days prior to the calorimetry, the mice were first acclimatized to the experimental setup including feeding basket and drinking bottle by housing them individually in similar cages. During the indirect calorimetry several parameters were measured including VO2 ((ml/h)/kg), VCO2 ((ml/h)/kg), respiratory quotient (RQ, (VO2)/(VCO2)) and heat production ((kcal/h)/kg). Afterwards, mice were housed individually in standard cages for the remainder of the experiment. Measurements from the second day of calorimetry were used for data analysis.

Glucose tolerance and insulin sensitivity testsOne week after the indirect calorimetry an intraperitoneal glucose tolerance test (IP-GTT) was performed. After an overnight fast (~17 hours). mice were weighed and blood was taken via tail bleeding for a baseline (t=0) blood glucose level measurement (Bayer Contour). Subsequently 2g/kg bodyweight D-glucose (20% D-glucose in 0.9% NaCl) was injected intraperitoneally. Blood glucose levels were determined after 15, 30, 45, 60, 90 and 120 minutes.One week later an intraperitoneal insulin sensitivity test (IPIST) was performed. Mice were fasted for 4 hours and weighed where after blood was taken via tail bleeding for a baseline blood glucose level measurement (t=0). Then a bolus of Actrapid (0.75 IU/kg) was injected intraperitoneally. Blood glucose levels were determined at 15, 30, 45, 60, 90 and 120 minutes.

DissectionPrior to dissection in week 12, mice were fasted overnight for approximately 17 to 19 hours. Between 9AM and 11 AM on the study day, all animals were anesthetized with pen-tobarbital (100 mg/kg i.p.). In the subsequent dissection, venous blood sampling was performed by canulation of the caval vein. Liver, muscle (soleus, gastrocnemius, quad-riceps femoris, heart) and white adipose tissue (WAT)) were harvested, frozen in liquid nitrogen and stored at -80°C.

gBB versus acetylcarnitine experiment In a separate experiment, we compared the effect of gBB and a bolus of acetylcarnitine on glucose tolerance after IPGTT. We therefore divided 24 chow fed C57BL/6N mice in 3 groups. The first group was treated with gBB for eight weeks, similar to the original experiment. The second group received an i.p. injection of acetylcarnitine (250 mg/kg) 30 minutes prior to the IPGTT. The third group was a control group. We performed the IPGTT twice, after four and eight weeks of gBB treatment. Ten days after the second IP-GTT mice were dissected. Thirty minutes prior to dissection, mice in the acetylcarnitine group were again injected with a bolus of acetylcarnitine (250mg/kg).

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

K007_000000_HereThere and EveryWhere boekje.indd 117 09-02-15 08:30

Page 122: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

118

Laboratory analysesPlasma and tissue acylcarnitines samples were analysed as described in our previous study (18). Carnitine biosynthesis intermediates in plasma were measured using UPLC-tandem MS as described earlier (19, 20). Plasma NEFA were analysed using an enzymatic colori-metric method (NEFA-HR, Wako Diagnostics, Richmond, VA). Insulin levels in plasma were measured using the Ultra Sensitive Mouse Insulin ELISA Kit (Crystal Chem Inc, USA). qPCR was performed to measure OCTN2 expression in muscle tissue. Total mRNA was iso-lated from tissues using Trizol extraction. cDNA was produced using a first-strand cDNA synthesis kit. For the quantitative real-time PCR the LightCycler FastStart DNA Master SYBR Green I kit was used and the following primers: 5’-AGATTCGGGATATGCTGTTGG-3’ and 5’-AAAGCCTGGAAGAAGGAGGTC-3’ for 36B4 and 5’-TTCTGGTGCTTGTCTCACTG-3’ and 5’-TATGTTCGGCTTCCCATTCT-3’ for beta2M and 5’-CCACTCACCACCTCCTTGTT-3’ and 5’-AAGACCTGCAGGAAGCTGAA-3’ for OCTN2. All samples were analyzed in tripli-cate. To confirm that a single product was formed during PCR, melting curve analysis was performed. Values for OCTN2 were normalized against the values for the housekeeping genes 36B4 and beta2M to adjust for variations in the amount of input RNA.During dissection both liver tissue and a combined sample of gastrocnemius muscle and soleus muscle were snap frozen, stored immediately in liquid nitrogen, and freeze-dried overnight to measure acetyl- and free CoA as described before (21). Statistical analysisAcylcarnitine profile measurements were processed with Masslynx software version 4.1. Statistical analysis was performed using SPSS statistical software program version 20.0 and Graphpad Prism software version 6. A Student’s T test was performed to detect gBB treatment effects. A Holm-Sidak multiple T test was used to analyze the effect of gBB treatment on plasma free carnitine levels during the treatment period and plasma glu-cose levels during the IPGTTs and IPISTs. All graphs show means ± SD. Data was assessed as significant with a P value <0.05, marked as *, and <0.01 is showed as **.

Results

General effects of HFD and gBB treatmentFeeding the HFD caused a strong weight gain, but gBB supplementation did not affect body weight gain in both chow and HFD mice (Fig. 1a). In order to assess whether gBB supplementation increased carnitine over the entire study period, biweekly whole blood samples were collected during the first eight weeks of the treatment (Fig. 1b). Mice on a HFD had lower blood carnitine levels when compared to chow fed animals. gBB supple-mentation increased blood carnitine levels in HFD mice but in chow mice carnitine levels did not further increase. Thus despite increasing free carnitine levels effectively, gBB did not curb HFD-induced body weight.

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

K007_000000_HereThere and EveryWhere boekje.indd 118 09-02-15 08:30

Page 123: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

119

Effects of HFD and gBB treatment on acylcarnitine profiles in plasma and tissueAt the end of the study acylcarnitine profiles were measured in plasma and several organ compartments of all mice. The dietary conditions affected acylcarnitine profiles in both plasma and tissues (Fig. 2a-2f). In plasma of chow fed mice, only C0-, C2- and C4OH- (Fig. 2a) were significantly in-creased upon gBB treatment. In plasma of HFD mice, treatment with gBB caused a gen-eral increase of short- (including acetylcarnitine), medium- and long-chain ACs. The increase in C0-carnitine was significantly greater in the HFD mice compared to the chow mice. The ratio between C0- and C2-carnitine in plasma was significantly affected by gBB treatment in the HFD group but not in the chow group (Supplemental table S1a). Of the plasma carnitine biosynthesis intermediates, as expected, gBB levels increased significantly upon supplementation (Fig. 2b). The levels of TML, one of the precursors of gBB in the carnitine biosynthetic pathway, did not change upon gBB treatment (Fig. 2b).At the tissue level, gBB supplementation only had an effect on acylcarnitine levels in liver. Here, C0-, C2- and C4OH-carnitine significantly increased upon gBB treatment in the

0 2 4 6 820

30

40

50

Week of treatment

Bod

y w

eigh

t (gr

ams) - BB

+ BB- BB+ BB

Chow

HFD

week 2 week 4 week 6 week 80

10

20

30

40

50

C0-

carn

itine

(μm

ol/L

) - BB+ BB- BB+ BB

****

**

*

** **

Chow

HFD

Figure 1a

Figure 1b

Figure 1 gBB supplementation in a diet-induced obesity model (A) Body weight gain in HFD vs chow mice. (B) Carnitine levels

in whole blood.

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

K007_000000_HereThere and EveryWhere boekje.indd 119 09-02-15 08:30

Page 124: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

120

HFD group (Fig. 2c). In chow fed mice, only C4OH-carnitine was significantly increased in liver of gBB-treated animals. Although several acylcarnitine levels were lowered in HFD mice compared to chow controls, soleus muscle, gastrocnemius muscle and WAT did not show any significant effect of gBB treatment in all mice (Fig. 2d-f). In contrast to liver and

**- γBB+ γBB- γBB+ γBB

CHOW

HFD

**

****

*** **

** **

**

yBB TML0

1

2

3

mol

/L

- BB+ BB- BB+ BB

**ns**

Chow

HFD

- γBB+ γBB- γBB+ γBB

CHOW

HFD**

*** **

**

***

**

Figure 2a Plasma acylcarnitine profile

Figure 2b Carnitine intermediates

Figure 2c

Liver acylcarnitine profile

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

K007_000000_HereThere and EveryWhere boekje.indd 120 09-02-15 08:30

Page 125: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

121

muscle where C0/C2 ratios remained unchanged in both diets, the C0/C2 ratio in WAT was significantly lower after gBB treatment in the chow mice (Supplemental table S1a). We further investigated why muscle acylcarnitines were not affected by gBB treatment by measurement of mRNA expression of OCTN2 in muscle (Supplemental fig. S1). The

- γBB+ γBB- γBB+ γBB

CHOW

HFD

*

- γBB+ γBB- γBB+ γBB

CHOW

HFD

*

- γBB+ γBB- γBB+ γBB

CHOW

HFD

Figure 2d

Soleus acylcarnitine profile

Figure 2e

Gastrocnemius acylcarnitine

profile

Figure 2f WAT acylcarnitine profile

Figure 2 Acylcarnitine profiles upon gBB treatment. (A) Plasma acylcarnitine profiles, (B) plasma carnitine intermediates and

tissue acylcarnitine profiles in (C) liver, (D) soleus muscle, (E) gastrocnemius muscle and (F) WAT of fasted chow vs HFD mice.

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

K007_000000_HereThere and EveryWhere boekje.indd 121 09-02-15 08:30

Page 126: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

122

expression level of OCTN2 was not changed by gBB, suggesting that OCTN2 expression is not driven by altered carnitine concentrations in plasma.To summarize, HFD changed acylcarnitine profiles in plasma and tissues. gBB supple-mentation affected the acylcarnitine profile in plasma and, to a lesser extent, in liver. The changes in the plasma acylcarnitine profile were independent of plasma NEFA levels, which did not change upon the interventions (data not shown). Additionally the effects of gBB in plasma did not correspond with the effects on tissue level, which is in accordance with our previous study where plasma acylcarnitine profiles did not correlate with tissue acylcarnitine profiles (18).

Indirect calorimetry measurementsTo evaluate the effects of gBB supplementation on energy expenditure, we performed an indirect calorimetry measurement. Mice fed a HFD showed a lower respiratory exchange ratio compared to chow fed mice, indicating higher FAO rates upon HFD feeding. Treat-ment with gBB did not affect the RER in both groups (Supplemental fig. S2a), indicating that the metabolic substrate preference was not influenced by gBB supplementation. Energy expenditure (EE) was significantly higher in HFD mice when compared to chow controls, but when corrected for bodyweight EE did not differ upon HFD feeding (Sup-plemental fig. S2b) (22). Additionally treatment with gBB did not affect EE either in both the light and dark phase of the day. Thus gBB supplementation had no effect on energy expenditure and RER. These observation are consistent with the observation that gBB was unable to curb body weight on a HFD.

Effects of gBB on glucose tolerance and insulin sensitivityTo investigate whether carnitine availability affects glucose metabolism, we performed glucose tolerance and insulin sensitivity tests. Glucose tolerance was clearly impaired in HFD mice when compared to chow fed mice, but in both groups gBB treatment had no effect on glucose tolerance (Fig. 3a). Fasted plasma insulin levels showed a clear effect of the HFD on insulin resistance (Fig. 3b). However, gBB treatment did not have an addition-al effect on the insulin levels in both groups. Even though HFD fed mice were insulin re-sistant compared to chow fed mice, there was no effect of gBB treatment (Fig. 3c). These experiments indicate that gBB supplementation does not improve glucose homeostasis in a diet-induced obesity model.

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

K007_000000_HereThere and EveryWhere boekje.indd 122 09-02-15 08:30

Page 127: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

123

0 15 30 45 60 75 90 105 1200

5

10

15

20

25

time (minutes)

gluc

ose

(mm

ol/L

) - BB+ BB- BB+ BB

Chow

HFD

Chow - Chow + HFD - HFD +0

1

2

3

4

5

ns

ns

Plas

ma

insu

lin (μ

g/L)

+ BB

- BB+ BB- BB

Chow

HFD

0 15 30 45 60 75 90

5

10

15

time (minutes)

gluc

ose

(mm

ol/L

) - BB+ BB- BB+ BB

Chow

HFD

Figure 3a

Figure 3b

Figure 3c

Figure 3 Glucose tolerance and insulin sensitivity tests. Effect of gBB treatment on (A) glucose levels upon IPGTT, (B) fasting

plasma insulin levels or (C) glucose levels upon IPIST.

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

K007_000000_HereThere and EveryWhere boekje.indd 123 09-02-15 08:30

Page 128: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

124

gBB versus acetylcarnitine administrationNext we modulated acylcarnitine metabolism by administration of a bolus of acetylcarni-tine, and compared this to chronic gBB administration. The acylcarnitine profile in plas-ma showed pronounced alterations upon acetylcarnitine administration (Fig. 4a). Many acylcarnitines such as C0-, C2-, C3-, C5-, C4OH-, C16- and C18:1-carnitine, showed a marked increase of which C0- and C2-carnitine were the most pronounced with a 10- to 20-fold increase compared to controls. Comparable to the effects in plasma, the levels of hepatic C0-, C2-, C3-, C5-, C4OH-, C16- and C18:1-carnitine were increased by acetylcarnitine administration (Fig. 4b). In soleus and gastrocnemius muscle only free carnitine and acetylcarnitine levels were in-creased, and no other acylcarnitine species were affected by the intervention (Fig.4c and d). In addition to free and acetylcarnitine, C3- and C4OH-carnitine were increased in WAT as well (Fig. 4e). The C0/C2 ratio was significantly lowered by acetylcarnitine treatment in both plasma and tissues (Supplemental table S1b).After 4 weeks and 8 weeks of gBB treatment we tested glucose tolerance in all three groups of mice (Supplemental fig. S3). Both tests showed no significant differences be-tween the gBB-, acetylcarnitine- and control group. Therefore, despite pronounced effects on acylcarnitine profiles, acetylcarnitine infusion had no effect on glucose tolerance.

ControlγBBC2-carnitine

******** ****

****

****

********

ControlγBBC2-carnitine

******** ****

**** **** **** ****

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

Figure 4a Plasma acylcarnitine profile

Figure 4bLiver acylcarnitine profile

K007_000000_HereThere and EveryWhere boekje.indd 124 09-02-15 08:30

Page 129: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

125

ControlγBBC2-carnitine

********

Figure 4 Acylcarnitine profiles upon gBB vs acute acetylcarnitine treatment. (A) Plasma acylcarnitine profiles, and tissue

acylcarnitine profiles in (B) liver, (C) soleus muscle, (D) gastrocnemius muscle and (E) WAT of fasted chow fed mice.

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

Figure 4cSoleus acylcarnitine profile

****

****** Control

γBBC2-carnitine

Figure 4d

Gastrocnemius acylcarnitine

profile

ControlγBBC2-carnitine

****

**

*********

Figure 4e WAT acylcarnitine profile

K007_000000_HereThere and EveryWhere boekje.indd 125 09-02-15 08:30

Page 130: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

126

We measured the levels of acetyl-CoA and free CoA in both muscle and liver in order to establish the consequences of acetylcarnitine and gBB treatment on these potential regulators of PDH activity. We found an increase in acetyl-CoA levels in liver tissue of both gBB- and acetylcarnitine-treated mice, but not in their soleus and gastrocnemius muscle tissue (Fig. 5a and b). In liver, the increase in acetyl-CoA was accompanied by a decrease in free CoA, reflecting that the available acetyl-groups were bound to CoA, thereby lowering the amount of free CoA. This is also reflected by a significant decrease in the liver CoA/acetyl-CoA ratio (Fig. 5b). However, in muscle no effect was observed on free CoA levels (Fig.5c and d).

Acetyl-CoA CoA0

200

400

600

800

Con

cent

ratio

n in

live

r(p

mol

/mg

dry

wei

ght) Controls

yBBC2-carnitine***

**

*****

Controls yBB C2-carnitine0

1

2

3

4

Rat

io C

oA/A

cety

l-CoA

live

r

***

Figure 5a

Figure 5b

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

Figure 5 Free and acetyl-CoA in liver and muscle upon gBB versus acetylcarnitine treatment. (A) Acetyl-CoA and free CoA

concentrations in liver. (B) Ratios of free CoA versus acetyl-CoA and free carnitine versus acetylcarnitine in liver. (C) Acetyl-

CoA and free CoA concentrations in muscle. (D) Ratio of free CoA versus acetyl-CoA in muscle.

K007_000000_HereThere and EveryWhere boekje.indd 126 09-02-15 08:30

Page 131: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

127

Discussion

In this study, we aimed to increase carnitine availability via enteral gBB administration to investigate if this would improve FAO rates, body weight gain, energy expenditure and glucose homeostasis. By treating lean chow-fed mice and diet-induced obese, insulin resistant mice with the carnitine precursor gBB, we were able to modulate the (acyl)car-nitine levels in plasma and tissue. We showed that C0- and C2-levels were decreased in plasma of HFD mice and that these levels were both restored to chow control levels upon gBB treatment. This is in line with work in rats on a high fat diet, which have decreased plasma levels of free carnitine (5). It is intriguing that the proportional increase in plasma free carnitine in the HFD group is much greater than in the chow fed animals (Fig. 2a). Ultimately similar levels of free carnitine were achieved in chow and HFD mice after gBB treatment, suggesting a plateauing effect in which the kidney reaches a carnitine excre-tion threshold, above which excess carnitine is excreted (3). gBB treatment furthermore increased free carnitine levels and (mainly short-chain) acylcarnitine levels in plasma and liver, but not in muscle and adipose tissue.In the liver, gBB treatment resulted in an increase of most acylcarnitine species although this increase was less pronounced than plasma and absent for C16- and C18:1-carnitine.

Acetyl-CoA CoA0

50

100

150

200C

once

ntra

tion

in m

uscl

e(p

mol

/mg

dry

wei

ght) Controls

yBBC2-carnitine

ns

ns

Controls yBB C2-carnitine0.0

0.2

0.4

0.6

0.8

Rat

io C

oA/A

cety

l-CoA

mus

cle

ControlsyBBC2-carnitine

ns

Figure 5c

Figure 5d

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

K007_000000_HereThere and EveryWhere boekje.indd 127 09-02-15 08:30

Page 132: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

128

Oral administered gBB will efficiently be hydroxylated to L-carnitine in the liver, which explains an increase in hepatic free carnitine and subsequently acylated carnitines as well (23). Recently we showed that the hepatic acylcarnitine profile corresponds mod-estly with plasma long chain acylcarnitines, but not C0- and C2-carnitine (18). Therefore the corresponding effects in plasma and liver from gBB treatment might reflect hepatic carnitine synthesis and the subsequent plasma distribution of C0- and C2-carnitine. Remarkably, gBB treatment did not have any effect on acylcarnitine profiles in muscle tissue or WAT. As these tissues both lack the enzymes for carnitine synthesis (3) and must therefore actively import carnitine from the blood via the sodium dependent OCTN2 transporter, it is important to know which factors influence carnitine uptake. We showed that the expression of OCTN2 in muscle tissue was similar in all mice, from which we con-clude that the uptake of carnitine by muscle tissue to compensate for a potential carni-tine shortage is not hampered by decreased OCTN2 activity. Therefore despite similar OCTN2 expression levels, the transport of carnitine and acetylcarnitine into muscle and WAT does not seem to increase when the plasma concentrations of carnitine and acetyl-carnitine are higher. Additionally, the pool size of carnitine in plasma is only a fraction of that in muscle tissue, and the elevated plasma level might very well be too small to have a detectable effect on the muscle carnitine pool.Our findings partially correspond with the effects found in a study by Noland et al. where L-carnitine administration in obese rats caused similar alterations in muscle and plasma acylcarnitine profiles (5). But although they showed an improvement of insulin sensitivity after carnitine supplementation, in our study gBB treatment did not affect diet-induced insulin resistance despite the evident alterations of acylcarnitine profiles. This discrep-ancy might be explained by the fact that these authors studied rats on a 12-month HFD, whereas we studied mice that were only fed a HFD for 13 weeks. But even though our study did not involve longer high fat feeding, we did achieve severe obese and insulin resistant HFD animals. Acylcarnitine profiles in HFD animals differ from chow animals, and thus acylcarnitine profiles may reflect obesity. However, the changed acylcarnitine profile after gBB in our HFD animals was not accompanied by functional improvements in FAO and insulin sensitivity. Acylcarnitine profiles in plasma only have a modest relation to liver acylcarnitine profiles and do not correlate with profiles in muscle and adipose tissue; thus plasma acylcarnitine levels do not necessarily reflect the metabolic state in insulin sensitive tissues (18). Therefore these findings are in agreement with our results on the lack of effect of gBB on insulin resistance and energy expenditure.The absent effect of gBB could be explained by the fact that changed carnitine and acyl-carnitine availability does not influence insulin sensitivity and energy expenditure as long as the energy demand or metabolic state has not changed. Muoio et al. suggested that li-potoxicity and insulin resistance are not explained by reduced mitochondrial content or function but are a consequence of a supply of fatty acids exceeding the energy demand (24). In our study we did not change the energy demand in our animals, so the altered acylcarnitine profiles actually reflect excess carnitine and acylcarnitines, that do not influence insulin sensitivity.

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

K007_000000_HereThere and EveryWhere boekje.indd 128 09-02-15 08:30

Page 133: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

129

The effect of the HFD on acylcarnitine levels was comparable in plasma, liver, muscle and adipose tissue. Interestingly we could not reproduce the HFD-induced increase in long-chain acylcarnitines as shown by Koves et al. (25) as our mice showed an unchanged or decreased C16- and C18:1-carnitine level in plasma, liver and muscle. However, our mice were fasted overnight in contrast to the ad libitum fed rats in the study by Koves (25). Continuous HFD might result directly in higher long-chain acylcarnitines. Otherwise, the FAO rate may be adapted to a chronic elevated lipid flux in the HFD mice, which explains less accumulation of acylcarnitines. Accumulation of acylcarnitines upon fasting might also depend on the studied tissue compartment or species (11, 18). Recent studies have described the stimulating role of carnitine supplementation on the activity of CrAT, which produces short-chain acylcarnitine from their correspond-ing short-chain acyl-CoAs. By regulating acetyl-CoA/CoA and acetylcarnitine/carnitine ratios, CrAT influences glucose- and fat metabolism: this has already been implicated in the metabolic inflexibility observed in insulin resistance (5, 9, 26). Mechanistically, for-mation of acetylcarnitine is thought to alleviate the mitochondrium from high levels of acetyl-CoA thereby stimulating PDH activity (5, 9, 27). This influence of carnitine levels on CrAT activity is in agreement with the alterations we found in the plasma and liver acylcarnitine profiles, where free carnitine and specifically several short-chain acylcar-nitines but not long-chain acylcarnitines were increased upon gBB treatment. This could be due to the stimulating effect of free carnitine availability on CrAT activity (26).We compared gBB treatment with an acute high dose of acetylcarnitine in lean mice, which caused a strong increase of all acylcarnitine species in plasma and had some ef-fects on free carnitine and acetylcarnitine in tissue. We hypothesized that a high dose acetylcarnitine increased the concentration of acetyl-CoA, resulting in an increase in free carnitine. Increased levels of acetyl-CoA can inhibit both activity of the PDH com-plex and subsequently glucose uptake, leading to lower glucose tolerance. Both gBB and acetylcarnitine, however, did not affect glucose tolerance. As anticipated, acetyl-CoA was higher in liver upon both treatments, alongside with lower levels of free CoA. In mus-cle, however, both acetyl-CoA and free CoA were unaffected by acetylcarnitine or gBB treatment, which implies that PDH activity was not modulated. We speculate that mus-cle acetylcarnitine uptake may be insulin dependent similar to free carnitine uptake (8). Since our mice were fasted with low insulin levels, acetylcarnitine may not have reached muscle effectively.In conclusion, despite alterations in plasma acylcarnitine profiles, both gBB treatment and acetylcarnitine treatment had no effect on insulin sensitivity in lean and obese, in-sulin resistant Bl6 mice. As the effects on tissue acylcarnitine profiles were much more subtle or even absent, this might explain why an effect on insulin sensitivity is lacking. These results warrant caution on making associations between plasma acylcarnitine lev-els and insulin resistance.

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

K007_000000_HereThere and EveryWhere boekje.indd 129 09-02-15 08:30

Page 134: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

130

References1. Holland WL, Knotts TA, Chavez JA, Wang LP, Hoehn KL, Summers SA. Lipid mediators of insulin resis-

tance. Nutr Rev. 2007;65(6 Pt 2):S39-S46.2. Shulman GI. Cellular mechanisms of insulin resistance. The Journal of Clinical Investigation.

2000;106(2):171-6.3. Vaz FM, Wanders RJA. Carnitine biosynthesis in mammals. Biochemical Journal. 2002;361(3):417-29.4. Bremer J. Carnitine--metabolism and functions. Physiol Rev. 1983;63(4):1420-80.5. Noland RC, Koves TR, Seiler SE, Lum H, Lust RM, Ilkayeva O, et al. Carnitine insufficiency caused by

aging and overnutrition compromises mitochondrial performance and metabolic control. J Biol Chem. 2009;284(34):22840-52.

6. Stephens FB, Constantin-Teodosiu D, Laithwaite D, Simpson EJ, Greenhaff PL. A threshold exists for the stimulatory effect of insulin on plasma L-carnitine clearance in humans. Am J Physiol Endocrinol Metab. 2007;292(2):E637-E41.

7. Stephens FB, Constantin-Teodosiu D, Laithwaite D, Simpson EJ, Greenhaff PL. An acute increase in skeletal muscle carnitine content alters fuel metabolism in resting human skeletal muscle. J Clin En-docrinol Metab. 2006;91(12):5013-8.

8. Stephens FB, Constantin-Teodosiu D, Laithwaite D, Simpson EJ, Greenhaff PL. Insulin stimulates L-car-nitine accumulation in human skeletal muscle. FASEB J. 2006;20(2):377-9.

9. Muoio DM, Noland RC, Kovalik JP, Seiler SE, Davies MN, DeBalsi KL, et al. Muscle-Specific Deletion of Carnitine Acetyltransferase Compromises Glucose Tolerance and Metabolic Flexibility. Cell Metabo-lism. 2012;15(5):764-77.

10. Ringseis R, Keller J, Eder K. Role of carnitine in the regulation of glucose homeostasis and insulin sen-sitivity: evidence from in vivo and in vitro studies with carnitine supplementation and carnitine defi-ciency. European journal of nutrition. 2012;51(1):1-18.

11. Soeters MR, Lammers NM, Dubbelhuis PF, Ackermans M, Jonkers-Schuitema CF, Fliers E, et al. Inter-mittent fasting does not affect whole-body glucose, lipid, or protein metabolism. Am J Clin Nutr. 2009;90(5):1244-51.

12. Gao AW, Canto C, Houtkooper RH. Mitochondrial response to nutrient availability and its role in met-abolic disease. EMBO molecular medicine. 2014;6(5):580-9.

13. Hoppel CL, Genuth SM. Carnitine metabolism in normal-weight and obese human subjects during fasting. Am J Physiol. 1980;238(5):E409-E15.

14. Giancaterini A, De Gaetano A, Mingrone G, Gniuli D, Liverani E, Capristo E, et al. Acetyl-L-carnitine infusion increases glucose disposal in type 2 diabetic patients. Metabolism. 2000;49(6):704-8.

15. Ferreira MR, Camberos Mdel C, Selenscig D, Martucci LC, Chicco A, Lombardo YB, et al. Changes in hepatic lipogenic and oxidative enzymes and glucose homeostasis induced by an acetyl-L-carnitine and nicotinamide treatment in dyslipidaemic insulin-resistant rats. Clinical and experimental pharma-cology & physiology. 2013;40(3):205-11.

16. Rebouche CJ, Bosch EP, Chenard CA, Schabold KJ, Nelson SE. Utilization of dietary precursors for carnitine synthesis in human adults. The Journal of Nutrition. 1989;119(12):1907-13.

17. Rebouche CJ. Effect of dietary carnitine isomers and gamma-butyrobetaine on L-carnitine biosynthe-sis and metabolism in the rat. The Journal of Nutrition. 1983;113(10):1906-13.

18. Schooneman MG, Achterkamp N, Argmann CA, Soeters MR, Houten SM. Plasma acylcarnitines inade-quately reflect tissue acylcarnitine metabolism. Biochim Biophys Acta. 2014.

19. Celestino-Soper PB, Violante S, Crawford EL, Luo R, Lionel AC, Delaby E, et al. A common X-linked in-born error of carnitine biosynthesis may be a risk factor for nondysmorphic autism. Proc Natl Acad Sci U S A. 2012;109(21):7974-81.

20. Vaz FM, Melegh B, Bene J, Cuebas D, Gage DA, Bootsma A, et al. Analysis of Carnitine Biosynthesis Metabolites in Urine by HPLC-Electrospray Tandem Mass Spectrometry. Clinical Chemistry. 2002;48(6):826-34.

21. Demoz A, Garras A, Asiedu DK, Netteland B, Berge RK. Rapid method for the separation and detection of tissue short-chain coenzyme A esters by reversed-phase high-performance liquid chromatography. Journal of Chromatography B: Biomedical Sciences and Applications. 1995;667(1):148-52.

22. Tschop MH, Speakman JR, Arch JR, Auwerx J, Bruning JC, Chan L, et al. A guide to analysis of mouse energy metabolism. Nature methods. 2012;9(1):57-63.

23. Strijbis K, Vaz FM, Distel B. Enzymology of the carnitine biosynthesis pathway. IUBMB Life. 2010;62(5):357-62.

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

K007_000000_HereThere and EveryWhere boekje.indd 130 09-02-15 08:30

Page 135: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

131

24. Muoio DM, Neufer PD. Lipid-Induced Mitochondrial Stress and Insulin Action in Muscle. Cell Metabo-lism. 2012;15(5):595-605.

25. Koves TR, Ussher JR, Noland RC, Slentz D, Mosedale M, Ilkayeva O, et al. Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance. Cell Metab. 2008;7(1):45-56.

26. Seiler SE, Martin OJ, Noland RC, Slentz DH, Debalsi KL, Ilkayeva OR, et al. Obesity and lipid stress inhibit carnitine acetyltransferase activity. J Lipid Res. 2014;55(4):635-44.

27. Power R, Hulver M, Zhang J, Dubois J, Marchand R, Ilkayeva O, et al. Carnitine revisited: potential use as adjunctive treatment in diabetes. Diabetologia. 2007;50(4):824-32.

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

K007_000000_HereThere and EveryWhere boekje.indd 131 09-02-15 08:30

Page 136: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

132

Supplemental data

Table S1a

Mean C0/C2 ratios

CHOW HFD

- gBB + gBB - gBB + gBB

Plasma 2,18 (±0,59) 1,72 (±0,40) 3,44 (±0,81) 2,61 (±0,77) *

Liver 17,40 (±7,69) 12,45 (±5,47) 7,85 (±2,29) 7,31 (±1,71)

Soleus 4,64 (±1,43) 4,07 (±2,11) 3,98 (±1,51) 4,36 (±1,97)

Gastrocnemius 6,65 (±1,30) 7,07 (±1,22) 4,22 (±0,77) 4,69 (±1,15)

WAT 9,67 (±2,33) 7,08 (2,27) * 8,49 (±2,49) 6,75 (±1,05)

Table S1b

Mean C0/C2 ratios

Control gBB C2-carnitine

Plasma 2,31 (±0,49) 1,64 (±0,22)* 1,04 (±0,10)**

Liver 5,94 (±1,16) 4,97 (±0,69) 3,54 (±0,62)**

Soleus 3,31 (±1,50) 2,51 (±0,55) 1,62 (0,25)*

Gastrocnemius 9,08 (±3,25) 7,89 (±2,14) 3,44 (±0,51)**

WAT 5,89 (±1,16) 4,99 (±0,82) 0,92 (±0,12)**

Table S1 C0/C2-carnitine ratios in plasma and tissues (A) upon gBB treatment (B) upon gBB vs acetylcarnitine treatment.

Figure S1 OCTN2 expression in muscle. Effect of gBB treatment on the relative expression of OCTN2 in combined soleus and

gastrocnemius muscle.

- BBCHOW

+ BBCHOW

- BBHFD

+ BBHFD

0.00

0.02

0.04

0.06

Mus

cle

OC

TN2

rela

tive

expr

essi

on

ns

ns

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

K007_000000_HereThere and EveryWhere boekje.indd 132 09-02-15 08:30

Page 137: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

133

25 30 35 40-400

-200

0

200

400

weight (grams)

EE d

ivid

ed b

y bo

dy w

eigh

t (k

cal/h

/g)

p 0,88

25 30 35 40-200

-100

0

100

200

300

weight (grams)

EE d

ivid

ed b

y bo

dy w

eigh

t (k

cal/h

/g)

+ BB- BB

p 0,51

35 40 45 50 55-200

-100

0

100

200

weight (grams)

EE d

ivid

ed b

y bo

dy w

eigh

t (k

cal/h

/g)

p 0,45

35 40 45 50 55-300

-200

-100

0

100

200

weight (grams)

EE d

ivid

ed b

y bo

dy w

eigh

t (k

cal/h

/g)

p 0,37

CHOW LIGHT PHASE CHOW DARK PHASE

HFD LIGHT PHASE HFD DARK PHASE

Light phase Dark phase0.7

0.8

0.9

1.0

1.1R

ER- BB+ BB- BB+ BB

Chow

HFD

0 50 1000

5

10

15

20ControlyBBC2-carnitine

Time (min)

gluc

ose

(mm

ol/L

)

Figure S2 Indirect calorimetry (A) Respiratory exchange ratio, and (B) Resting energy expenditure measurements in light and

dark phase, corrected for body weight by ANCOVA.

Figure S3 Glucose tolerance after gBB vs acetylcarnitine treatment Plasma glucose levels upon an IPGTT after 8 weeks of

gBB treatment versus acute acetylcarnitine administration.

THE IMPACT OF ALTERED CARNITINE AVAILABILITY

Figure S2a

Figure S2b

K007_000000_HereThere and EveryWhere boekje.indd 133 09-02-15 08:30

Page 138: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

C 8 Discussion: A multi organ perspective

on acylcarnitine metabolism

K007_000000_HereThere and EveryWhere boekje.indd 134 09-02-15 08:30

Page 139: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

C 8 Discussion: A multi organ perspective

on acylcarnitine metabolism

K007_000000_HereThere and EveryWhere boekje.indd 135 09-02-15 08:30

Page 140: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

K007_000000_HereThere and EveryWhere boekje.indd 136 09-02-15 08:30

Page 141: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

137

Introduction

In the past few decades much of the metabolic research has focused on the relation between obesity and insulin resistance. Researchers strive after more understanding of, and control over the deranging body weight of the western population and all con-sequential diseases this problem brings along. Insulin resistance is one of the main de-rangements of overfeeding, and disturbed lipid metabolism is identified as one of the etiological factors of this derangement. Within the theory of lipotoxicity, where lipid intermediates such as ceramides, gangliosides and diacylglycerol accumulate in insulin sensitive tissues, acylcarnitines have also been suggested to interfere with insulin sig-nalling. This idea was based on several studies, showing elevated acylcarnitine levels in obese and insulin resistant humans and rodents (1-4). Impaired FAO, either low FAO or high but incomplete FAO, could lead to an accumulation of acylcarnitines inside the cell which could eventually end up in the plasma compartment (2, 4). Therefore, alterations in the plasma profile were thought to reflect acylcarnitine metabolism on the tissue level. But interestingly, acylcarnitine profiles in plasma did not correlate with levels in muscle tissue of fasted men (5). This discrepancy is relevant, as insulin resistance occurs at the tissue level, whereas acylcarnitine profiles are often measured in plasma. These find-ings, together with the proposed hypotheses, illustrate difficulties in the interpretation of altered acylcarnitine profiles. The interpretation of the plasma acylcarnitine profile requires knowledge of the origin and the kinetic properties of acylcarnitines in general, and additionally understanding of the reciprocal relation between different organs par-ticipating in acylcarnitine metabolism. This perspective further explores acylcarnitine metabolism in physiological and pathological conditions to clarify its involvement in in-sulin resistance and glucose intolerance.

Acylcarnitine metabolism in different speciesAcylcarnitine metabolism is extensively studied in humans and model organisms. Howev-er, important differences exist between species, including carnitine biosynthesis sites, but also acylcarnitine concentrations in general (6-10). Most important are the differenc-es between humans and rodents, which are the main model organisms in acylcarnitine research (7). An example is carnitine synthesis, which mainly occurs in human kidney in contrast to rodents where it mainly takes place in liver (10). Additionally, the expression of the enzyme carnitine acetyltransferase (CrAT) is highest in human liver while in ro-dents it is highly expressed in skeletal muscle (11). Comparing measurements of insulin sensitivity and energy expenditure of different species can be challenging as well (12). Even within one species these differences can be found, such as the weight gain upon a HFD, the regulation of glucose tolerance and possibly acylcarnitine metabolism between mouse strains (9, 13-15). These differences hamper a direct translation of rodent data to the human setting and vice versa.

A MULTI ORGAN PERSPECTIVE ON ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 137 09-02-15 08:30

Page 142: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

138

The origin and function of acylcarnitinesAcylcarnitines are intermediates of mitochondrial acyl-CoA oxidation, and comprise of an acyl group coupled to L-carnitine. L-carnitine enables transport of (mainly long chain) acyl-CoA over the mitochondrial membrane, which is otherwise impermeable to these acyl esters. Therefore L-carnitine fulfils a crucial role in the supply of substrates to the mitochondrion for energy production (10, 16). The acyl groups esterified to L-carnitine can originate from different substrates. Fatty acids are the most abundant species cou-pling to L-carnitine in order to be oxidized, as carnitine is essential for FAO. Therefore, acylcarnitines are generally specified as fatty acid oxidation (FAO) intermediates. How-ever, carbohydrates, amino acids and ketone bodies contribute to the formation of some acylcarnitines as well (3, 17-19). Acylcarnitines are often measured in a profile, and as such they are regarded as a family of metabolites. Historically, acylcarnitines were di-vided in acid-soluble and –insoluble species, later on specified as free carnitine, short chain- and long chain acylcarnitines (20). The introduction of tandem mass spectrome-try allowed the identification of individual acylcarnitine species, based on the molecular mass determined by the length of the acyl group attached to the carnitine molecule (21). Acylcarnitines can be divided based on their origin, plasma concentration or clinical rel-evance. As the origin of the acyl groups varies, they do not necessarily behave similarly or fulfil similar metabolic roles. Free carnitine is often decreased in obesity, possibly due to increased acylation of carnitine by the abundant amount of FFA present in plasma and tissues (3, 22). And as carnitine is mainly absorbed from the diet, specifically meat and dairy products, levels can be low in vegetarians and vegans (23). C2-carnitine main-ly derives its acetyl group from either lipids or carbohydrates, and therefore its level can vary upon different metabolic states as well (17): in chow fed mice that were fasted, C2-carnitine concentrations increased (9)(chapter 5), which was not observed when HFD fed mice were fasted (chapter 5). Of the remaining acylcarnitines, the presence of the individual species can vary in different metabolic states. As carnitine is crucial for FAO, an increase in the full range of acylcarnitines can be seen in FAO-stimulating conditions such as fasting or HFD-induced insulin resistance, both known for increased FAO rates along with decreased rates of carbohydrate oxidation (CHO) (3, 9). But other species follow different patterns, which can be based on their origin. Amino acid derived spe-cies such as C3- and C5-carnitine tend to increase upon meals containing protein (18)(chapter 4). And C4OH-carnitine, which is primarily ketone body derived, is mainly seen in ketotic and insulin resistant states (9, 19).As described above, we found substantial differences between C2- and C16-carnitine kinetics, which again can be explained by the origin of the substrate that is bound to the carnitine molecule. Here, parameters such as pool size, rate of appearance and elimination of C2-carnitine were affected by changes in metabolic state such as fast-ing or high fat diet feeding. This implies that C2-carnitine either is a transporter for carnitine towards tissues to meet increased carnitine requirements due to metabolic changes (chapter 4), or possibly as a carrier of the acetyl group. The pool size, rate of appearance or elimination of C16-carnitine did not change under different metabolic conditions, demonstrating that C16-carnitine may be a by-product of FAO rather than a

A MULTI ORGAN PERSPECTIVE ON ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 138 09-02-15 08:30

Page 143: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

139

carnitine carrier or energy substrate. These differences in kinetic properties of C2- and C16-carnitine illustrate that changes in acylcarnitine levels do not simply reflect overall metabolic changes such as alterations in FAO or insulin sensitivity. It also illustrates that acylcarnitine profiles reflect many different metabolic processes, and should be inter-preted per individual species.

Acylcarnitine profiles in plasma and tissuesPlasma is often the main compartment used for the analysis of the acylcarnitine profile. This profile has been considered as a reflection of whole body acylcarnitine metabolism for a long time, as many studies report plasma acylcarnitine alterations in relation to the metabolic condition in tissue (1, 3, 24). However, we have shown that the plasma profile does not represent a specific organ or tissue (9)(chapter 3). Although we did not find cor-relations between plasma and tissue acylcarnitines, we did find groups of acylcarnitines forming clusters within the compartments, which do relate to each other in a topological overlap measure plot, a technique often used to see how large sets of genes relate to each other (chapter 3). Here we showed that despite lacking correlations between plas-ma and muscle acylcarnitines, long chain acylcarnitines in four different compartments of striated muscle tissue form a cluster under fed and fasted circumstances, suggesting muscle specific coordinated acylcarnitine metabolism. The fact that we found no rela-tion between plasma and skeletal muscle corroborated with other studies, where plas-ma and muscle acylcarnitine levels did not correspond, acylcarnitine fluxes to and from muscle were low and administration of a carnitine precursor did not lead to changes in muscle acylcarnitine levels (5)(chapter 4 and 7). Long chain acylcarnitines in plasma did show some relation with long chain species in liver (9), which we recently confirmed in two studies where plasma acylcarnitines mostly resembled liver acylcarnitines as well (chapter 4 and 7). This relation between plasma and liver acylcarnitines was shown ear-lier in a study in fasted macaques, but here no individual chain lengths were measured (25). In conclusion, the finding that correlations between plasma and organs were ab-sent, corresponds with the differences in acylcarnitine fluxes through different organs (chapter 4), pointing out that these compartments play different roles in whole body acylcarnitine metabolism.

The interplay of different organs in acylcarnitine metabolismAs concluded in the previous paragraph, various concentrations, fluxes and kinetics are found in the organ compartments involved in acylcarnitine metabolism (5, 9)(chapter 4, 5 and 7). To understand these characteristics per organ, we need to understand the organ-specific role in whole body acylcarnitine metabolism under different metabolic circumstances. First, not every fatty acid oxidizing organ is capable of de novo carnitine synthesis (10, 26). This requires an efficient system of distribution and transportation, where organs can either supply or receive carnitine and where plasma facilitates the actual transport to and from organs (26). Within this system, the liver plays a central role in both the production and distribution of acylcarnitines. The capacity to synthesize carnitine in

A MULTI ORGAN PERSPECTIVE ON ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 139 09-02-15 08:30

Page 144: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

140 A MULTI ORGAN PERSPECTIVE ON ACYLCARNITINES

CARNITINE UPTAKE

DIETARY UPTAKE OFLIPID & CARNITINE

CARNITINE SYNTHESIS ,, REABSORPTION ,, DISTRIBUTION ,, EXCRETION

CARNITINE EXCRETION

LIPOLYSISACYLCARNITINES

LIPOGENESIS

UPTAKE C2-CARNITINEAND ACYLCARNITINES

FAO

Carnitinesynthesis

PLASMA

DIET FAO

Lipolysis

CHO

CARNITINE SYNTHESISDISTRIBUTION CARNITINE & ACYLCARNITINESHIGH C2-CARNITINE DISTRIBUTION

FAOCrAT

HIGH C2-CARNITINE DISTRIBUTION

CarnitinesynthesisCarnitinesynthesis

FASTEDFED

CACFA

CFA

FAAC

C

C2

C2FA

C

ACC2

C2

FA

FAFA

AC

ACCC

C2AC

C

h

Figure 1 provides an overview of different organs and their involvement in acylcarnitine metabolism under fed and fasted

conditions. Here, the liver has a main role in the synthesis and distribution of carnitine (via acetylcarnitine) and other

acylcarnitines. Plasma carnitine levels regulate hepatic carnitine production. Skeletal muscle is known for its high total

carnitine content that depends on carnitine uptake via plasma acetylcarnitine. Muscle takes up little other acylcarnitines and

hardly relates to other compartments. WAT showed a strong increase in all acylcarnitines upon fasting, possibly reflecting

lipolysis, as acylcarnitines correlated with FFA upon weight loss. The kidney has a distinct function in the synthesis and

reabsorption of carnitine. However, it takes up both carnitine and acylcarnitines for urinary excretion, thereby regulating the

carnitine pool. A major part of the total carnitine pool is absorbed from the diet by the gut. The gut releases acylcarnitines

as well, mainly upon feeding. It is unknown whether gut is capable of de novo carnitine synthesis. Finally, the plasma

compartment contains a profile which is a mixture of acylcarnitines from all participating tissues. But no specific tissue is

represented by the plasma acylcarnitine profile. However, it does have an important function in the exchange and distribution

between all different organs involved in acylcarnitine metabolism.

K007_000000_HereThere and EveryWhere boekje.indd 140 09-02-15 08:30

Page 145: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

141

the liver is well established in rodents, where liver contains high levels of the crucial enzyme for carnitine production, gamma-butyrobetaine dioxygenase (BBH) (26). This de novo hepatic carnitine production provides the liver with sufficient carnitine for higher FAO rates when needed, but also enables inter organ distribution of carnitine. As such, liver contains high concentrations of C2-carnitine, both in fed and fasted mice (9). More-over, the fluxes of C2-carnitine to and from the liver are highest in fed and fasted pigs, when compared to fluxes over skeletal muscle, gut and kidney (chapter 4). We found a negative correlation between venous plasma concentrations of carnitine and its flux over the liver, suggesting a feedback mechanism where low plasma carnitine concentrations correlate with high production of carnitine by the liver and vice versa. This feedback mechanism may be mediated by the intestinal carnitine uptake, where the liver might sense low portal vein carnitine levels and synthesize carnitine when needed. Carnitine synthesis itself is regulated by PPAR-alpha, which is an important regulator of fatty acid oxidation and is therefore abundantly expressed in fatty acid oxidizing tissues (6, 26, 27). Accordingly, it has been demonstrated that PPAR-alpha activation results in enhanced carnitine production (26). However, carnitine concentrations are not known to influence PPAR-alpha activity. An interesting aspect is that the expression of the carnitine trans-porter OCNT2 is rather low in liver when compared to muscle and kidney in humans (28). OCTN2 supposedly transports carnitine mainly into the cells, against a gradient of high carnitine concentrations inside and low concentrations outside of the cell. This could explain the relatively low hepatic OCTN2 expression, as liver is able to synthesize car-nitine and is therefore not depending on carnitine influx. It also suggests that efflux of carnitine does not occur via OCTN2 transport, but possibly by diffusion. In conclusion, the liver seems to play a key role in regulating the liver and plasma carnitine pool.Another organ capable of de novo carnitine synthesis is the kidney, which also contains the enzyme BBH (10, 29). In humans, the kidney is actually the main organ for carnitine production as it highly expressed BBOX1, the gene encoding for BBH (10, 29). We have shown carnitine production by the kidney in fed and fasted pigs (chapter 4). However, uptake of acylcarnitines by the kidney might be of similar importance, as shown by the negative fluxes in kidney, meaning acylcarnitine uptake in both fed and fasted state. The uptake of carnitine and C2-carnitine by the kidney may be mainly for excretion purpos-es and subsequent regulation of the acylcarnitine pool, as carnitine and acylcarnitines appear in urine as well (30, 31). Alternatively, it might facilitate FAO in the kidney itself.It is unknown whether the gut is capable of de novo carnitine synthesis. One mouse study has suggested that carnitine was produced in mouse intestines, but to our knowledge no other study has reproduced this finding so far (32). In our study on acylcarnitine fluxes in pigs, we did find a modest postprandial peak of the portal vein carnitine flux. However we were unable to determine whether this was de novo carnitine synthesis or uptake from the diet. The gut released other acylcarnitines as well, suggesting postprandial dis-tribution of dietary carnitine, lipids and protein.The skeletal muscle compartment is not capable of carnitine synthesis, and is therefore dependent on carnitine distribution from other organs via plasma (10). The need for car-nitine is high in muscle, as it relies heavily on FAO (5, 33). The acylcarnitine concentration

A MULTI ORGAN PERSPECTIVE ON ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 141 09-02-15 08:30

Page 146: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

142

in skeletal muscle is the highest of all studied organs, and approximately 95% of the to-tal carnitine pool resides here (9). Additionally the volume of the muscle compartment largely exceeds that of other organs, explaining the high percentage of the carnitine pool residing in muscle tissue. Therefore it was surprising to find the fluxes to and from the muscle compartment to be very low, except for the net C2-carnitine flux. The latter may be considered as either supply of carnitine or alternatively supply of acetyl-CoA (chapter 4). In general, muscle showed no correlations of acylcarnitine profiles with other tissues or plasma, nor did it show pronounced production or uptake of acylcarnitines (except for C2-carnitine). This demonstrates that muscle must interact only moderately with other compartments when it comes to acylcarnitine metabolism per se.

Intervening in the acylcarnitine poolAs carnitine is essential for the oxidation of fatty acids, it has been used and studied extensively in conditions where endogenous carnitine levels are low due to inborn errors of metabolism, leading to pathological carnitine deficiencies (34, 35). Additionally, carni-tine is often used as a supplement for potential beneficial effects on lipid oxidation and muscle performance (22, 36-40).We showed that the administration of the precursor of carnitine, gamma-butyrobetaine, in lean and obese mice, increased levels of carnitine and almost all plasma acylcarnitine species, as well as the shorter chain species in liver (chapter 7). But we were unable to interfere in the acylcarnitine levels of muscle or white adipose tissue (WAT). Even in obese mice, where FAO rates were potentially higher, no effect on carnitine levels was observed. When administering a high amount of C2-carnitine as a bolus in lean mice, acylcarnitine levels in plasma and liver showed a strong increase. But apart from an in-crease in carnitine and C2-carnitine, again the profiles in muscle or WAT were hardly affected. This illustrates how the compartments of muscle and adipose tissue appar-ently do not (fully) rely on constant supply of carnitine from the plasma and the other compartments. As we did not substantially interfere with the energy demand in these mice, it might be that the lacking effect in muscle and WAT resulted from an unchanged need for carnitine. Possibly, more pronounced effects after C2-carnitine administration could be observed in settings where carnitine is depleted or where metabolism is other-wise stressed. It has been suggested earlier by Muoio et al that mitochondrial capacity is sufficient to handle sudden increases in substrate flux (41). We assume that carnitine is present in muscle tissue in great excess, to secure the capacity for adjustment to mod-erately increased FAO rates. Additionally carnitine might be excessively present as a po-tential buffer for acetyl-CoA, as CrAT can regulate PDH activity by converting acetyl-CoA to C2-carnitine and vice versa. Another interesting aspect of the lacking effect of increased carnitine availability on muscle and WAT acylcarnitine profiles, is that our tracer studies showed that the acyl-carnitine pool behaves as a single pool, which exceeds the plasma compartment up to 50-fold. This means that the pool must include the interstitium and possibly the cyto-sol of certain tissues as well, and that exchange of acylcarnitines between plasma and interstitium or cytosol must occur unrestrained. We hypothesize that the boundary of

A MULTI ORGAN PERSPECTIVE ON ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 142 09-02-15 08:30

Page 147: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

143

the pool is the mitochondrial membrane where acylcarnitines are uncoupled, and the acyl-CoA enters beta-oxidation.

Acylcarnitines in various metabolic conditionsAs most acylcarnitines are mainly FAO derived intermediates, they are often studied un-der fasted circumstances as fasting induces higher FAO rates. But in HFD-induced obe-sity and insulin resistance in mice, FAO rates are often increased at the expense of CHO. Acylcarnitine levels are also found to be elevated in obese humans (3). A possible causal role for acylcarnitines in the induction of insulin resistance has been proposed. Here acylcarnitines accumulate as a consequence of increased or incomplete FAO in obesity. This hypothesis is supported by the observation of several statistically significant asso-ciations between specific acylcarnitine species and markers of glucose intolerance and insulin resistance, further suggesting a causal role (1, 3, 42-44). However, when studying acylcarnitines in insulin sensitive and insulin resistant mouse models, we were unable to reproduce some of the results from available studies. Moreover, the results from these studies are not always consistent. Therefore we question the direct role of acylcarnitines as modifiers of insulin sensitivity.In fasted HFD mice, plasma acylcarnitine levels were lower when compared to fasted chow mice (chapter 7). In liver, the shorter chain species were lower in HFD fed mice as well. However, in muscle and adipose tissue no difference was found between HFD and chow fed mice, in accordance with the absent effect of gamma-butyrobetaine adminis-tration. These effects could have been detectable when mice were not fasted. However, we also showed that in HFD mice, no differences in kinetics of both C2-carnitine and C16-carnitine were detected. In the chow-fed mice, there was a difference in C2-carni-tine kinetics, meaning that this effect is concealed by the HFD intervention. Moreover, changes in acylcarnitine profiles in the different studies were not associated with insulin resistance. We were unable to influence insulin resistance, glucose tolerance or energy expenditure by administering gamma-butyrobetaine (chapter 7). And in addi-tion, improvements in insulin sensitivity in obese humans on a weight loss intervention did not correlate with alterations in acylcarnitine levels (chapter 6). Here, changes in acylcarnitine levels did correlate with plasma FFA, suggesting that acylcarnitines may reflect WAT lipolysis during weight loss. We did not find any effect on production or elimination rates of C16-carnitine in diet- or fasting induced insulin resistant mice, even though this is one of the main species of interest in the discussion on acylcarnitines and insulin resistance (chapter 5). Again lowering the probability of acylcarnitines as causal factors in diet-induced insulin resistance.An exception to the modest contribution of most acylcarnitines is the role of C2-carni-tine in insulin resistance. Although we do not propose that C2-carnitine induces insulin resistance, it may have a crucial role in switching between lipid and carbohydrate oxi-dation. C2-carnitine can be produced by carnitine acetyltransferase (CrAT), an enzyme which converts acetyl-CoA into C2-carnitine and vice versa (22, 42, 45, 46). However the affinity of CrAT is higher for acetyl-CoA than for C2-carnitine, shifting the equilibrium towards C2-carnitine formation. As the presence of acetyl-CoA inhibits PDH activity,

A MULTI ORGAN PERSPECTIVE ON ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 143 09-02-15 08:30

Page 148: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

144

CrAT can buffer the amount of acetyl-CoA into C2-carnitine, in order to allow CHO via PDH. It has been suggested that CrAT function is lower in obese and insulin resistant subjects, leading to impaired substrate switching and metabolic inflexibility (42). It was demonstrated in a CrAT knockout mouse model that when CrAT activity is diminished, the ability to switch to CHO is impaired and FAO is the main source for energy produc-tion. Several studies including that of Muoio et al. (22, 42, 45) suggest that CrAT activity is mainly found in skeletal muscle and that skeletal muscle is the main source for C2-car-nitine. It was proposed that the high activity of CrAT causes a high efflux of acylcarni-tines from muscle tissue. However the expression and activity of CrAT varies between different species. In rodents, CrAT expression is indeed highest in skeletal muscle, but humans mainly express CrAT in liver tissue (11). The pigs in our study on trans organ fluxes, which exhibit CrAT activity in both liver and muscle (11), had low fluxes of all acylcarni-tines from muscle tissue. Therefore we question if muscle is indeed responsible for the largest fraction of the plasma acylcarnitine pool, as proposed by Noland et al. (22). In our study, much higher fluxes of C2-carnitine came from the liver. This could imply that the purpose of C2-carnitine formation in muscle and liver differs.

Conclusion

To summarize, our results do not directly implicate a causal role for acylcarnitines in insulin resistance. We even question if there is a relevant association between these metabolites and glucose tolerance, as levels of acylcarnitines varied under different metabolic circumstances and independent of the degree of insulin resistance. How- ever, we do think that there is a relevant association between C2-carnitine and glucose tolerance via the interplay between lipid and glucose oxidation.

References1. An J, Muoio DM, Shiota M, Fujimoto Y, Cline GW, Shulman GI, et al. Hepatic expression of malonyl-CoA

decarboxylase reverses muscle, liver and whole-animal insulin resistance. Nat Med. 2004;10(3):268-74.

2. Muoio DM, Koves TR. Lipid-induced metabolic dysfunction in skeletal muscle. Novartis Found Symp. 2007;286:24-38.

3. Mihalik SJ, Goodpaster BH, Kelley DE, Chace DH, Vockley J, Toledo FG, et al. Increased levels of plasma acylcarnitines in obesity and type 2 diabetes and identification of a marker of glucolipotoxicity. Obe-sity (Silver Spring). 2010;18(9):1695-700.

4. Koves TR, Ussher JR, Noland RC, Slentz D, Mosedale M, Ilkayeva O, et al. Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance. Cell Metab. 2008;7(1):45-56.

5. Soeters MR, Sauerwein HP, Duran M, Wanders RJ, Ackermans MT, Fliers E, et al. Muscle acylcarnitines during short-term fasting in lean healthy men. Clin Sci (Lond). 2009;116(7):585-92.

6. Ringseis R, Wen G, Eder K. Regulation of Genes Involved in Carnitine Homeostasis by PPAR-alpha across Different Species (Rat, Mouse, Pig, Cattle, Chicken, and Human). PPAR research. 2012;2012:868317.

7. Chegary M, Brinke H, Ruiter JP, Wijburg FA, Stoll MS, Minkler PE, et al. Mitochondrial long chain fatty acid beta-oxidation in man and mouse. Biochim Biophys Acta. 2009;1791(8):806-15.

A MULTI ORGAN PERSPECTIVE ON ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 144 09-02-15 08:30

Page 149: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

145

8. Su AI, Wiltshire T, Batalov S, Lapp H, Ching KA, Block D, et al. A gene atlas of the mouse and human protein-encoding transcriptomes. Proc Natl Acad Sci U S A. 2004;101(16):6062-7.

9. Schooneman MG, Achterkamp N, Argmann CA, Soeters MR, Houten SM. Plasma acylcarnitines inade-quately reflect tissue acylcarnitine metabolism. Biochim Biophys Acta. 2014.

10. Vaz FM, Wanders RJA. Carnitine biosynthesis in mammals. Biochemical Journal. 2002;361(3):417-29.11. Wu C, Orozco C, Boyer J, Leglise M, Goodale J, Batalov S, et al. BioGPS: an extensible and customizable

portal for querying and organizing gene annotation resources. Genome biology. 2009;10(11):R130.12. Butler AA, Kozak LP. A recurring problem with the analysis of energy expenditure in genetic models

expressing lean and obese phenotypes. Diabetes. 2010;59(2):323-9.13. Farrell GC, Mridha AR, Yeh MM, Arsov T, Van Rooyen DM, Brooling J, et al. Strain dependence of diet-in-

duced NASH and liver fibrosis in obese mice is linked to diabetes and inflammatory phenotype. Liver international : official journal of the International Association for the Study of the Liver. 2014;34(7):1084-93.

14. Clee SM, Attie AD. The Genetic Landscape of Type 2 Diabetes in Mice. Endocrine Reviews. 2007;28(1):48-83.

15. Montgomery MK, Hallahan NL, Brown SH, Liu M, Mitchell TW, Cooney GJ, et al. Mouse strain-dependent variation in obesity and glucose homeostasis in response to high-fat feeding. Diabetologia. 2013;56(5):1129-39.

16. Ramsay RR, Gandour RD, van der Leij FR. Molecular enzymology of carnitine transfer and transport. Biochim Biophys Acta. 2001;1546(1):21-43.

17. Ebeling P, Tuominen JA, Arenas J, Garcia Benayas C, Koivisto VA. The association of acetyl-L-carnitine with glucose and lipid metabolism in human muscle in vivo: the effect of hyperinsulinemia. Metabo-lism. 1997;46(12):1454-7.

18. Newgard CB, An J, Bain JR, Muehlbauer MJ, Stevens RD, Lien LF, et al. A Branched-Chain Amino Ac-id-Related Metabolic Signature that Differentiates Obese and Lean Humans and Contributes to Insulin Resistance. Cell Metabolism. 2009;9(4):311-26.

19. Soeters MR, Serlie MJ, Sauerwein HP, Duran M, Ruiter JP, Kulik W, et al. Characterization of D-3-hy-droxybutyrylcarnitine (ketocarnitine): an identified ketosis-induced metabolite. Metabolism. 2012;61(7):966-73.

20. Brass EP, Hoppel CL. Carnitine metabolism in the fasting rat. J Biol Chem. 1978;253(8):2688-93.21. Vreken P, van Lint A, Bootsma A, Overmars H, Wanders R, van Gennip A. Quantitative plasma acylcarni-

tine analysis using electrospray tandem mass spectrometry for the diagnosis of organic acidaemias and fatty acid oxidation defects. J Inherit Metab Dis. 1999;3(22):302-6.

22. Noland RC, Koves TR, Seiler SE, Lum H, Lust RM, Ilkayeva O, et al. Carnitine insufficiency caused by aging and overnutrition compromises mitochondrial performance and metabolic control. J Biol Chem. 2009;284(34):22840-52.

23. Delanghe J, De Slypere JP, De Buyzere M, Robbrecht J, Wieme R, Vermeulen A. Normal reference values for creatine, creatinine, and carnitine are lower in vegetarians. Clinical Chemistry. 1989;35(8):1802-3.

24. Hoppel CL, Genuth SM. Carnitine metabolism in normal-weight and obese human subjects during fasting. Am J Physiol. 1980;238(5):E409-E15.

25. Bell FP, DeLucia A, Bryant LR, Patt CS, Greenberg HS. Carnitine metabolism in Macaca arctoides: the effects of dietary change and fasting on serum triglycerides, unesterified carnitine, esterified (acyl) carnitine, and beta-hydroxybutyrate. The American Journal of Clinical Nutrition. 1982;36(1):115-21.

26. van Vlies N, Ferdinandusse S, Wanders R, Vaz F. PPARalfa-activation results in enhanced carnitine biosynthesis and OCTN2 expression. Biochim Biophys Acta. 2007;1767(9):11.

27. Leone. A Critical Role for the Peroxisome Proliferator-Activated Receptor alfa (PPAR alfa) in the Cellu-lar Fasting Response: The PPAR-alfa Null Mouse as a Model of Fatty Acid Oxidation Disorders. Proc Natl Acad Sci USA. 1999;96(13):5.

28. Tamai I, Ohashi R, Nezu Ji, Yabuuchi H, Oku A, Shimane M, et al. Molecular and Functional Identification of Sodium Ion-dependent, High Affinity Human Carnitine Transporter OCTN2. Journal of Biological Chemistry. 1998;273(32):20378-82.

29. Vaz FM, van Gool S, Ofman R, Ijlst L, Wanders RJ. Carnitine biosynthesis: identification of the cDNA encoding human gamma-butyrobetaine hydroxylase. Biochem Biophys Res Commun. 1998;250(2):506 -10.

A MULTI ORGAN PERSPECTIVE ON ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 145 09-02-15 08:30

Page 150: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

146

30. Vaz FM, Melegh B, Bene J, Cuebas D, Gage DA, Bootsma A, et al. Analysis of Carnitine Biosynthesis Metabolites in Urine by HPLC-Electrospray Tandem Mass Spectrometry. Clinical Chemistry. 2002;48(6):826-34.

31. Chalmers R, Roe C, Stacey T, Hoppel C. Urinary excretion of l-carnitine and acylcarnitines by patients with disorders of organic acid metabolism: evidence for secondary insufficiency of l-carnitine. Pedi-atric Research. 1984;18(12):1325-8.

32. Shekhawat PS, Sonne S, Carter AL, Matern D, Ganapathy V. Enzymes involved in L-carnitine biosynthe-sis are expressed by small intestinal enterocytes in mice: implications for gut health. Journal of Crohn’s & colitis. 2013;7(6):e197-205.

33. Soeters MR, Sauerwein HP, Dubbelhuis PF, Groener JE, Ackermans MT, Fliers E, et al. Muscle Adapta-tion to Short-Term Fasting in Healthy Lean Humans. Journal of Clinical Endocrinology & Metabolism. 2008;93(7):2900-3.

34. Wanders R, Vreken P, den Boer M, Wijburg F, Van Gennip A, Ijlst L. Disorders of mitochondrial fatty acyl-CoA beta-oxidation. Journal of Inherited Metabolic Disease. 1999;22(4):442-87.

35. Nezu J, Tamai I, Oku A, Ohashi R, Yabuuchi H, Hashimoto N, et al. Primary systemic carnitine deficiency is caused by mutations in a gene encoding sodium ion-dependent carnitine transporter. Nat Genet. 1999;21(1):91-4.

36. Keller J, Ringseis R, Priebe S, Guthke R, Kluge H, Eder K. Effect of L-carnitine on the hepatic transcript profile in piglets as animal model. Nutr Metab (Lond). 2011;8:76.

37. Rebouche CJ. Effect of dietary carnitine isomers and gamma-butyrobetaine on L-carnitine biosynthe-sis and metabolism in the rat. The Journal of Nutrition. 1983;113(10):1906-13.

38. Power RA, Hulver MW, Zhang JY, Dubois J, Marchand RM, Ilkayeva O, et al. Carnitine revisited: potential use as adjunctive treatment in diabetes. Diabetologia. 2007;50(4):824-32.

39. Giancaterini A, De Gaetano A, Mingrone G, Gniuli D, Liverani E, Capristo E, et al. Acetyl-L-carnitine infusion increases glucose disposal in type 2 diabetic patients. Metabolism. 2000;49(6):704-8.

40. Stephens FB, Constantin-Teodosiu D, Laithwaite D, Simpson EJ, Greenhaff PL. An acute increase in skeletal muscle carnitine content alters fuel metabolism in resting human skeletal muscle. J Clin En-docrinol Metab. 2006;91(12):5013-8.

41. Muoio DM, Neufer PD. Lipid-Induced Mitochondrial Stress and Insulin Action in Muscle. Cell Metabo-lism. 2012;15(5):595-605.

42. Muoio DM, Noland RC, Kovalik JP, Seiler SE, Davies MN, DeBalsi KL, et al. Muscle-Specific Deletion of Carnitine Acetyltransferase Compromises Glucose Tolerance and Metabolic Flexibility. Cell Metabo-lism. 2012;15(5):764-77.

43. Redman LM, Huffman KM, Landerman LR, Pieper CF, Bain JR, Muehlbauer MJ, et al. Effect of Caloric Restriction with and without Exercise on Metabolic Intermediates in Nonobese Men and Women. Journal of Clinical Endocrinology & Metabolism. 2011;96(2):E312-E21.

44. Ramos-Roman MA, Sweetman L, Valdez MJ, Parks EJ. Postprandial changes in plasma acylcarnitine concentrations as markers of fatty acid flux in overweight and obesity. Metabolism. 2012;61(2):202-12.

45. Marquis NR, Fritz IB. The Distribution of Carnitine, Acetylcarnitine, and Carnitine Acetyltransferase in Rat Tissues. J Biol Chem. 1965;240:2193-6.

46. Seiler SE, Martin OJ, Noland RC, Slentz DH, Debalsi KL, Ilkayeva OR, et al. Obesity and lipid stress inhibit carnitine acetyltransferase activity. J Lipid Res. 2014;55(4):635-44.

A MULTI ORGAN PERSPECTIVE ON ACYLCARNITINES

K007_000000_HereThere and EveryWhere boekje.indd 146 09-02-15 08:30

Page 151: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

K007_000000_HereThere and EveryWhere boekje.indd 147 09-02-15 08:30

Page 152: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

C 9 Appendix

K007_000000_HereThere and EveryWhere boekje.indd 148 09-02-15 08:30

Page 153: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

C 9 Appendix

K007_000000_HereThere and EveryWhere boekje.indd 149 09-02-15 08:30

Page 154: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

150

K007_000000_HereThere and EveryWhere boekje.indd 150 09-02-15 08:30

Page 155: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

151 SUMMARY

Summary

In this thesis, we investigated acylcarnitines in plasma and insulin sensitive tissues in fasted, fed and a high fat diet (HFD)-induced, insulin resistant state. Acylcarnitines are fatty acid oxidation intermediates. They comprise of a fatty acid bound to carnitine, so that the fatty acid can enter the otherwise impermeable mitochondrion for oxidation purposes. By studying acylcarnitine concentrations, kinetics, and fluxes, we aimed to understand whole body acylcarnitine metabolism under different metabolic circum-stances, including pathological conditions such as insulin resistance characterized by derangements in acylcarnitine profiles. A short background of acylcarnitines is present-ed in chapter 1, where general information on the function of carnitine and acylcarni-tines is provided. Finally, the thesis outline is presented.

In chapter 2, we reviewed acylcarnitines more extensively, in particular in relation to insulin resistance. As acylcarnitines are proposed to play a role in diet-induced insulin resistance, we discussed human and animal studies showing alterations in acylcarnitine metabolism in obese, insulin resistant subjects and correlations between acylcarnitines and markers of glucose intolerance. The theory of lipotoxicity is discussed, where the accumulation of lipid derived metabolites such as acylcarnitines, could interfere with in-sulin signaling. We concluded that most of the studies reviewed in chapter 2 only showed associations between acylcarnitines and insulin resistance, but an exact causal mech-anism has not yet been identified. This was the starting point for the following studies.

Many studies on altered acylcarnitine profiles, which we reviewed in chapter 2, showed predominantly alterations in plasma. But it was unknown whether the plasma acylcar-nitine profile represents the acylcarnitine profile in one specific, or multiple tissues, hampering the interpretation of these plasma profile alterations. In chapter 3 we inves-tigated plasma and tissue acylcarnitine profiles in fed and fasted C57BL/6N and BALB/cJ mice. Here, no significant correlations were found between plasma and all examined tissues, in both fed and fasted state. We concluded that plasma acylcarnitines do not ad-equately reflect tissue acylcarnitines, and therefore studies on acylcarnitine metabolism should focus on tissue measurements.

To further clarify the role of different organs in whole body acylcarnitine metabolism, in chapter 4 we investigated a pig model with intravascular catheters positioned before and after the liver, gut, hindquarter muscle compartment and the kidney. With a mixed meal test, we measured acylcarnitine profiles at several time points and calculated tran-sorgan acylcarnitine fluxes. Liver played a main role in acylcarnitine metabolism, as it synthesizes carnitine, and distributes and regulates the acylcarnitine pool. The high-est flux was found for C2-carnitine, especially from liver towards other organs. Muscle showed only minor fluxes besides C2-carnitine uptake. The kidney predominantly ex-tracted acylcarnitines for excretion. The gut provided the pool with carnitine from the diet. These results illustrated the importance of liver as main organ for the distribution of acylcarnitines, and in particular C2-carnitine.

K007_000000_HereThere and EveryWhere boekje.indd 151 09-02-15 08:30

Page 156: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

152 SUMMARY

To aid in the understanding of the role of acylcarnitines under different metabol-ic circumstances, we studied acylcarnitine kinetics using stable isotopes. Chapter 5 demonstrates the kinetics of C16- and C2-carnitine in various mouse models of insulin resistance. Both C2- and C16-carnitine kinetics allowed single pool model analysis. Al-though BALB/cJ mice had a greater pool size than C57BL/6N mice, other kinetics for C16-carnitine kinetics were not affected by mouse strain, fasting or HFD. In contrast, C2-carnitine kinetics in C57BL/6N mice were affected, with a greater pool size, rate of appearance and lower elimination in fasted chow mice. HFD lowered the rate of appear-ance of C2-carnitine. These results suggest that HFD alters mainly C2-carnitine metabo-lism, and that these findings reflect impaired substrate switching in HFD-induced insulin resistance.

As plasma acylcarnitines have been associated with clinical parameters related to glu-cose metabolism, such as fasting glucose levels and HbA1c, we hypothesized that plasma acylcarnitines would correlate with clinical parameters of insulin sensitivity and glucose tolerance. In chapter 6, we measured plasma acylcarnitines in 60 obese human subjects before and during weight loss, and analysed acylcarnitine profiles in relation to clinical parameters of glucose metabolism, insulin sensitivity and energy expenditure.We demonstrated that along with improved insulin sensitivity, plasma acylcarnitines did not correlate with clinical parameters of glucose metabolism during weight loss. Acyl-carnitines did correlate significantly with plasma non-esterified fatty acids, suggesting that acylcarnitines reflect lipolysis during weight loss. We demonstrated that high plas-ma acylcarnitine levels are not necessarily associated with insulin resistance, in contrast to some earlier studies discussed in chapter 2.

In chapter 7, we intervened in the carnitine pool by administration of the carnitine pre-cursor gamma-butyrobetaine, hypothesizing that increasing free carnitine levels could facilitate FAO and improve insulin sensitivity. We compared lean and obese C57BL/6N mice of which half of both groups were treated with gamma-butyrobetaine, and per-formed indirect calorimetry, glucose tolerance and insulin sensitivity tests. Increasing carnitine availability affected acylcarnitine profiles in plasma and liver. Glucose toler-ance or insulin sensitivity were not affected, possibly due to the lack of effect on muscle acylcarnitines, as this is an important contributor to whole body insulin sensitivity. A bolus of C2-carnitine had an even more pronounced effect on the profiles, again with-out effect on glucose tolerance. These results suggest that there is not an unequivocal association between plasma acylcarnitine levels and insulin resistance.

We discuss our results in chapter 8, combining the findings from the different studies to generate new hypotheses. Here we focus on the roles the different organs fulfill in whole body acylcarnitine metabolism, in particular the liver. Additionally we discuss the individual species, where we question whether acylcarnitines are indeed involved in the etiology of insulin resistance. The results of our studies could not confirm a causal role for acylcarnitines in the induction of insulin resistance. One exception is C2-carnitine,

K007_000000_HereThere and EveryWhere boekje.indd 152 09-02-15 08:30

Page 157: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

153 SUMMARY

as this species is abundantly present and plays an important role in substrate switching. Therefore we consider it as a species, which could be indirectly related to insulin re-sistance, as substrate switching is impaired in insulin resistant subjects. For the other species, we question whether they are causally involved in insulin resistance.

K007_000000_HereThere and EveryWhere boekje.indd 153 09-02-15 08:30

Page 158: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

K007_000000_HereThere and EveryWhere boekje.indd 154 09-02-15 08:30

Page 159: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

155NEDERLANDSE SAMENVATTING

Nederlandse samenvatting

In dit proefschrift hebben we gekeken naar acylcarnitine metabolisme in plasma en in-suline gevoelige weefsels, in een gevaste, gevoede en een hoog vet dieet-geïnduceerde, insuline resistente staat. Acylcarnitines zijn intermediairen van de vetzuuroxidatie die bestaan uit een vetzuur gekoppeld aan carnitine, om zo het mitochondrion te kunnen bereiken voor oxidatie aldaar. Door acylcarnitine concentraties, -kinetiek en -fluxen te bestuderen, hebben we getracht meer inzicht te krijgen in het totale acylcarnitine me-tabolisme in het lichaam onder verschillende metabole omstandigheden. Zo proberen we verstoorde acylcarnitine profielen beter te begrijpen in relatie tot pathologische condities zoals insuline resistentie. Een kort overzicht van de achtergrond van acylcar-nitines is geschetst in hoofdstuk 1, waar carnitine en acylcarnitines in algemenere zin worden besproken. We sluiten af met een globaal overzicht van de hoofdstukken in het proefschrift.

Hoofdstuk 2 geeft een uitgebreider overzicht van de bestaande acylcarnitine literatuur, met name in relatie tot insuline resistentie. Omdat acylcarnitines in verband worden gebracht met het ontstaan van dieet-geïnduceerde insulin resistentie, hebben we naar studies gekeken die verstoringen in het acylcarnitine metabolisme lieten zien in obese, insulin resistente mensen en diermodellen, en studies die verbanden lieten zien tussen acylcarnitines en kenmerken van glucose intolerantie. Ook bespreken we lipotoxicite-it, een bekende theorie waarin vetzuur metabolieten zoals acylcarnitines ophopen en de insuline signalering kunnen verstoren. We concluderen dat de bestaande literatu-ur hoofdzakelijk associaties aan het licht brengt, maar geen exact causaal mechanisme identificeert. Dit vormde de basis voor alle hierop volgende studies.

Veel studies die acylcarnitines in verband brengen met insuline resistentie, zoals bespro-ken in hoofdstuk 2, hebben voornamelijk gekeken naar plasma acylcarnitine profielen. Het is echter niet bekend welk weefsel het plasma profiel daadwerkelijk reflecteert. Dit bemoeilijkt de interpretatie van eventuele veranderingen in het plasma. In hoofdstuk 3 hebben we onderzocht of er correlaties bestaan tussen het plasma acylcarnitine prof-iel en de profielen in verschillende insuline gevoelige weefsels van gevaste en gevoede C57BL/6N en BALB/cJ muizen. Het plasma acylcarnitine profiel bleek met geen enkel profiel in weefsels te correleren, zowel in gevaste als in gevoede muizen. Dit leidde tot de conclusie dat plasma acylcarnitines geen reflectie zijn van wat er gebeurt op weefsel-niveau, en dat studies naar acylcarnitine metabolisme zich moet richten op metingen in een specifiek weefsel.

Om de rol van de verschillende organen betrokken bij acylcarnitine metabolisme verder te verduidelijken, hebben we in hoofdstuk 4 een varkensmodel gebruikt om trans-or-gaan acylcarnitine fluxen te meten. Hiervoor werden middels een operatie intraveneuze catheters geplaatst in varkens, voor en na de lever, darm, skeletspieren van de achter-poten en de nier. Voor en tijdens een maaltijd werden acylcarnitines gemeten en bijbe-

K007_000000_HereThere and EveryWhere boekje.indd 155 09-02-15 08:30

Page 160: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

156 NEDERLANDSE SAMENVATTING

horende fluxen berekend. Hieruit kwam naar voren dat de lever een centrale rol heeft in acylcarnitine metabolisme, als producent, distributeur en regulator van de acylcarnitine pool. C2-carnitine bleek de belangrijkste metaboliet uit het gehele acylcarnitine prof-iel, met zeer hoge fluxen vanuit de lever richting andere organen. Skeletspier liet kleine fluxen zien, en met name C2-opname. De nier toonde met name opname van acylcar-nitines voor de uitscheiding ervan. In de darm veroorzaakte de maaltijd positieve fluxen van bijna alle acylcarnitines, wat opname van carnitine vanuit het dieet suggereert. Deze bevindingen illustreren het belang van de lever in het totale acylcarnitine metabolisme, en het belang van C2-carnitine daarin als metaboliet.

Om acylcarnitine metabolisme onder verschillende metabole omstandigheden in nog meer detail te begrijpen, hebben we de kinetiek van acylcarnitines bestudeerd door middel van stabiele isotopen. Hoofdstuk 5 bevat kinetische studies van C16- en C2-car-nitine in muizen met een variabele mate van insuline gevoeligheid. Zowel de kinetiek van C16- en C2-carnitine kunnen worden geanalyseerd als 1-compartiments-model. Hoewel BALB/cJ muizen een grotere C16-carnitine pool size hadden, verschilde de C16-carnitine kinetiek niet tussen de muisstammen, voedingstoestanden of dieetsoort. Daarentegen was de kinetiek van C2-carnitine wel anders, met een grotere pool size, rate of appear-ance, en een lagere eliminatie constante in gevaste dieren op normaal dieet. Een hoog vet dieet verlaagde de rate of appearance van C2-carnitine. Deze resultaten impliceren dat een hoog vet dieet met name C2-carnitine metabolisme beïnvloedt, en dat dit mo-gelijk leidt tot een verminderde capaciteit om te switchen tussen suiker en vet als ener-gie substraat onder hoog vet dieet-geïnduceerde insuline resistentie.

Op basis van de vermeende associatie tussen acylcarnitines en klinische parameters van glucose metabolisme, verwachtten wij dat plasma acylcarnitines negatief correleren met insuline gevoeligheid en glucose tolerantie. In hoofdstuk 6 hebben wij acylcarnitine pro-fielen gemeten in het plasma van 60 obese proefpersonen, voor en tijdens gewichtsver-lies. Deze profielen analyseerden wij in relatie tot klinische parameters van glucose metabolisme, insuline gevoeligheid en de basale verbranding. Hier demonstreerden wij dat ondanks een verbetering in insuline gevoeligheid door gewichtsverlies, plasma acylcarnitine niveaus verhoogd waren en niet correleerden met glucose metabolisme parameters. Plasma acylcarnitines correleerden wel met vrije vetzuren in plasma, wat suggereert dat deze acylcarnitines mogelijk een reflectie zijn van verhoogde lipolyse bij gewichtsverlies. Op basis van deze resultaten vinden wij het minder aannemelijk dat acyl-carnitines een rol spelen in de etiologie van insuline resistentie.

In hoofdstuk 7 hebben we de carnitine pool beïnvloedt door middel van de toediening van de carnitine precursor gamma-butyrobetaine. Wij verwachtten dat het verhogen van carnitine niveaus de vetzuur oxidatie zou kunnen faciliteren en een beter verlopende vetzuur oxidatie vervolgens de insuline gevoeligheid zou kunnen verbeteren. Hiertoe vergeleken we slanke en obese C57BL/6N muizen die wel of geen gamma-butyrobeta-ine toegediend kregen, waarbij we vervolgens een indirecte caloriemetrie meting, een

K007_000000_HereThere and EveryWhere boekje.indd 156 09-02-15 08:30

Page 161: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

157NEDERLANDSE SAMENVATTING

glucose tolerantie- en een insuline gevoeligheidtest uitvoerden. Een verhoging van het carnitine niveau had een effect op het gehele acylcarnitine profiel in plasma en in lever. Glucose tolerantie en insuline gevoeligheid veranderden beide niet, mogelijk doordat er geen effect van gamma-butyrobetaine bereikt werd in skeletspier. Een eenmalige toe-diening van een hoge dosis C2-carnitine had een nog sterker effect op de acylcarnitine profielen, maar wederom zonder de glucose tolerantie te beïnvloeden.

We bediscussiëren onze resultaten in hoofdstuk 8, waar we de verschillende bevinding-en naast elkaar leggen om nieuwe hypotheses te genereren. Hier richten wij ons op de rol van de diverse organen in het totale acylcarnitine metabolisme, en daarin de lever in het bijzonder. Verder bespreken we de individuele acylcarnitine soorten, en bediscussiëren we of acylcarnitines inderdaad betrokken lijken te zijn bij het ontstaan van insuline re-sistentie. De resultaten uit onze studies bevestigden niet dat acylcarnitines betrokken zijn bij het induceren van insuline resistentie. Alleen C2-carnitine lijkt mogelijk wel een indirecte rol te spelen, daar deze acylcarnitine een belangrijke rol speelt in het switchen tussen vet en suiker als energie substraat. Voor de overige acylcarnitines vinden wij een causale rol in het ontstaan van insuline resistentie niet aannemelijk.

K007_000000_HereThere and EveryWhere boekje.indd 157 09-02-15 08:30

Page 162: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

158 PHD PORTFOLIO

Name PhD student: Marieke Guurtje Schooneman PhD period: June 2011 – Dec 2014 Name PhD supervisors: Prof. Dr. C.E.M. Hollak; Prof. Dr. R.J.A. Wanders Name PhD co-supervisors: Dr. S.M. Houten; Dr. M.R. Soeters

1. PhD training

Year Workload(Hours/ECTS)

General courses World of ScienceBROK (Basiscursus Regelgeving Klinisch Onderzoek)Practical BiostatisticsCrash course biomedical research

2011201220122012

0,70,91,10,4

Specific courses - Cursus Proefdierkunde- Stralingscursus

20112012

3,91,7

Seminars, workshops and master classesResearch meetings Endocrinology & MetabolismResearch meetings GMZ lab

2011-20142011-2014

41

PresentationsKeystone scientific symposium, Dublin, Ireland. Oral presentation & poster presentationADDRM NVDO, Oosterbeek. Oral presentationEASD Annual meeting, Barcelona, Spain. 3 poster presentationsESPEN congres, Leipzich, Germany. Poster presentationAMGRO, Lemmer. 3x oral presentations

Research meeting Endo&Meta. Oral presentationsResearch meeting GMZ lab. Oral presentations

2014

20132013

20132011, 2013, 20142011-20142011-2014

0,4

0,20,6

0,30,9

0,30,9

(Inter)national conferencesKeystone scientific symposium, Dublin, Ierland.ADDRM NVDO, Oosterbeek.EASD Annual meetingESPEN congres, Leipzich, Germany.AMGRO, Lemmer

20142012, 20132012, 201320132011-2014

1,70,63,410,9

PhD portfolio

K007_000000_HereThere and EveryWhere boekje.indd 158 09-02-15 08:30

Page 163: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

159PHD PORTFOLIO

3. Parameters of Esteem

Year

GrantsAMC PhD ScholarshipTjallingh Roorda FoundationAMC Young Talent Fund ScholarshipKeystone Scholarship travelfund

2010201120142014

4. Publications

- Schooneman MG, Vaz FM, Houten SM, Soeters MR (2013) Acylcarnitines: Reflecting or Inflicting Insulin Resistance? Diabetes 62(1): 1-8.

- Schooneman MG, Achterkamp N, Argmann CA, Soeters MR, Houten SM (2014) Plasma acylcarnitines inadequately reflect tissue acylcarnitine metabolism Biochim Biophys Acta. 1841, 987–994

- Schooneman MG, ten Have GAM, Houten SM, Deutz NEP, Soeters MR Transorgan fluxes in a porcine model reveal a central role for liver in acylcarnitine metabolism Submitted

- Schooneman MG, Houten SM, Deutz NEP, Soeters MR Acylcarnitine kinetics in a fasting- and obesity-induced insulin resistant mouse model Submitted

- Schooneman MG, Napolitano A, Houten SM, Ambler GK, Murgatroyd PR, Miller SR, Hollak CEM, Tan CY, Virtue S, Vidal-Puig A, Nunez DJ, Soeters MR Assessment of plasma acylcarnitines before and after weight loss in obese subjects Submitted

- Schooneman MG, Houtkooper RH, Hollak CEM, Wanders RJA, Vaz FM, Soeters MR, Houten SM The impact of altered carnitine availability on fatty acid and glucose metabolism in diet-induced obese mice Submitted

- Soeters MR, Soeters PB, Schooneman MG, Houten SM, Romijn JA (2012) Adaptive reciprocity of lipid and glucose metabolism in human short-term starvation American Journal of Physiology - Endocrinology And Metabolism 303(12): E1397-E1407

- Soeters MR, Soeters PB, Schooneman MG, Sobotka L The benefit of moderate hyperglycemia and insulin resistance in critical illness Submitted

2. Teaching

Year Workload (Hours/ECTS)

Supervising Niki Achterkamp, Correlation studyLeonie van der Geest, TMT studyGuido Bakker, PIGS study

20122012-20132013

111

K007_000000_HereThere and EveryWhere boekje.indd 159 09-02-15 08:30

Page 164: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

K007_000000_HereThere and EveryWhere boekje.indd 160 09-02-15 08:30

Page 165: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

161DANKWOORD

Dankwoord

Promoveren is en was een mooi avontuur, dat ik onderweg heb gedeeld met zoveel bij-zondere mensen.

Allereerst zoveel dank aan mijn beide promotores, Prof. Dr. C.E.M. Hollak en Prof. Dr. R.J.A. Wanders. Beste Carla, als AIO op een wat curieus, níet erfelijk stofwisselings-onderwerp, was ik voor jou initieel een vreemde eend in de bijt. Jij bleek als vrouwelijke academica een prachtig rolmodel, en een waardevolle, welkome aanvulling op mijn verder hoofdzakelijk masculiene begeleiding. Je scherpte, kritische blik, en je vermogen om al het handel-en te blijven toetsen op wetenschappelijke relevantie is bewonderenswaardig. Om nog maar te zwijgen over je goede smaak voor brilmonturen en schoenen. Dank voor al je aandacht en toewijding, en voor alle wel- en niet-wetenschappelijke gesprekken in jouw altijd warme kamer. Beste Ron, jouw begeleiding was weliswaar wat meer op afstand door die paar verdiepin-gen ertussen, maar zonder die begeleiding was mijn onderzoek nergens geweest. Heel veel dank dat ik gebruik heb mogen maken van het fantastische GMZ lab, met geweld-ige collega’s en de vele faciliteiten die elk artikel in mijn proefschrift van data hebben voorzien. Het voelt als een grote eer om onder zo’n ervaren wetenschapper te mogen promoveren.

Dank aan mijn beide co-promotores, Dr. M.R. Soeters en Dr. S.M. Houten. Beste Maarten. Ain’t no mountain high enough. Of je er nou tegenop moet fietsen in Zuid-Frankrijk, of dat je er één moet verplaatsen om iets te bereiken wat je graag wil; jij deinst niet terug voor een hoge berg. En het liefst neem je iedereen mee naar boven om te laten zien hoe mooi het daar is. Jij hebt mij met deze onuitputtelijke drive weten te enthousiasmeren en motiveren, met dit proefschrift tot gevolg. En ik ben zo trots op het resultaat! Dankjewel voor je zorg en aandacht, voor je kritiek (oh, al die ‘spreektaal’ in mijn manuscripten!) en je vertrouwen. Mijn promotie jaren waren een feestje!Beste Sander, toen ik jou ontmoette, had ik nog nooit een pipet vastgehouden. Vanaf het begin heb je mij met zoveel geduld en toewijding wegwijs gemaakt in de wereld der basale wetenschap. Jouw intensieve hulp bij de experimenten en het labwerk was van onschatbare waarde, en zeker ook de ruimte die je mij gaf om (keer op keer) fouten te maken. Een geweldige wetenschapper als jij met toch zoveel nuchterheid en bescheid-enheid, kom je niet vaak tegen. Ik ben dankbaar voor jouw fantastische begeleiding, die je zelfs voortzette toen je op avontuur ging naar New York. Hier ligt voor jou, Carmen en kleine Bas vast nog heel veel moois in het verschiet. Maar we missen je wel in het AMC!

Veel dank aan alle overige leden van de promotiecommissie, Prof. Dr. J.A. Romijn, Prof. Dr. A.J. Verhoeven, Prof. Dr. C.J.M. de Vries, Prof. Dr. A.K. Groen, Prof. Dr. P. Schrauwen en Dr. M.J. Serlie, voor het zitting nemen in de promotiecommissie, en het kritisch bestu-deren van mijn manuscript. Beste Prof. Dr. Romijn, wat een eer en groot genoegen om

K007_000000_HereThere and EveryWhere boekje.indd 161 09-02-15 08:30

Page 166: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

162 DANKWOORD

onder uw hoede internist te mogen worden in het AMC! Beste Prof. Dr. Groen, ik hoop dat we na mijn verdediging een minstens zo gezellige borrel kunnen drinken als die op het Keystone congres in Dublin.

Dank aan al mijn collega’s op F5, waar ik 3 en een half jaar met veel plezier heb gewerkt. Birgit, als we jou toch niet hadden! Gabor, je liefde voor Haribo is wanstaltig, maar je hebt heel wat saaie AMC-middagen opgevrolijkt, waarvoor dank (vooral onze grapjes over Dr. Soeters deden het altijd goed). Lieve mede-AIO’s Anke, Annegreet, Barbara, Bouwien, Charlotte, Dirk-Jan, Eelkje, Eveline, Karin, Kasper, Laura, Linda, Maarten, Martine, Murat, Pim, Ruth, Sam en Yvonne, het was gezellig. Tijdens verjaardagstaartjes, op congressen, borreltjes, kraambezoeken, barbeque’s, sinterklaasavondjes en pro-motiefeestjes: never a dull moment! Annegreet, wat leuk om nu als collega-assistenten interne verder te gaan! Eelkje, ook jij voegt je daar binnenkort bij, hoera! Kom je weer naast me zitten dan? Karin, zie ik je weer tijdens de interne-MDL-bespreking? Ruth, het zal wat drukker worden, maar ik maak graag tijd voor een mooi stuk rennen vanaf het AMC en een goeie borrel na afloop.

Sam, collega-AIO, kamergenoot, proefpersoon, co-promotorgenoot, en pindasoep-lief-hebber. Je bent als een goede rode wijn. Eerst wat stroef, maar met een mooie afdronk die veel interessanter is dan die van een doorsnee supermarktwijntje. Jouw aanwezigheid de afgelopen jaren was een verrijking, zowel op F5, in de zuurkast op F0 of bij een reusach-tige schnitzel met bier in Leipzich. En dan nog het andere lid van Groep Soeters; Hannah. Je hebt me verrast met jouw wereldbeeld, wat zo anders kan zijn dan dat van mij. Maar zeker niet minder leuk! Ik heb genoten van onze gezamenlijke congressen, stukjes ren-nen, Vietnamese etentjes en kopjes koffie op maandag. To be continued, zou ik zeggen.

Beste collega’s van het GMZ lab, dank voor jullie warme ontvangst op F0! Arno, Albert, Henk, Martin, Gerard, jullie geduld voor mijn soms ietwat gebrekkige biochemische kennis was onuitputtelijk. Zonder jullie hulp is het nog maar de vraag of ik ooit enige significantie had bereikt met mijn metingen. Beste Riekelt, ik heb genoten van de uren-lange muizendissecties, met slechte muziek en goede gesprekken. Fred, jouw carnitine expertise, je input op mijn manuscripten en het meedenken over mijn proeven en analyses was geweldig. Heleen en Simone, Sander’s twee rechter handen. Alles wat ik niet van Sander leerde, leerde ik van jullie. Zoveel dank! En ook voor alle leuke gesprekken op het lab als er weer eens 40 vials stonden droog te dampen. Desi, die roti op donderdag, man.. zo goed!

Prof. Dr. N.E.P. Deutz, beste Mick. Mijn tijd in Texas was misschien wel een van de grootste avonturen in mijn promotie jaren. Ik denk met een warm gevoel terug aan de ontvangst door jou en Gabrie in College Station. Je betrokkenheid bij mijn studies was geweldig. Texas heeft mijn hart wel een beetje veroverd, en ik kom dan ook graag nog eens terug voor een mooi experiment of een Texaanse bbq met Shiner Bock. Beste Gabrie, zonder jouw hulp was er geen varkensstudie geweest. Dank voor al je hulp bij deze prachtige

K007_000000_HereThere and EveryWhere boekje.indd 162 09-02-15 08:30

Page 167: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

163DANKWOORD

studie. En dank dat ik je fiets mocht lenen: ik blijf toch gewoon een Nederlander in Texas! Dear Danielle Nicole White, I’d probably still be running after a crazy pig if you hadn’t been there. I’ll never drink a margarita again without thinking of you.

En zoveel dank voor alle hulp bij mijn experimenten, geneeskundestudenten Niki, Leonie en Guido, en ook de proefdierverzorgers in het ARIA-S gebouw, Suzan, Lex en Nienke. Jullie hulp was onmisbaar!

En dan kom ik aan bij mijn paranimfen; ook wel laven voor intimi. Lieve Antony, wanneer jij binnenkomt en jouw polygoonjournaalstem door de kamer schalt, moet ik als vanzelf glimlachen. Ik zou je monologen boven de zuurkoolschotel bij een goede fles wijn voor geen goud willen missen. Je bent een fantastische vriend, en ik vind het een eer dat je me bijstaat in de Agnietenkapel.Lieve Chantal, ik vraag me af wat jij in een vorig leven zou zijn geweest; Boeddha, Jackie O. of Mrs. President herself? Je bent een warme, lichte, lieve, sterke en prachtige vrouw en vriendin. Je bent een geweldige fotografe (zie dit proefschrift!). Je bent als familie. En ik kan in mijn broek piesen van je grapjes. Dankjewel, lieve buv!

Lieve Krista! Naast dat je dit proefschrift prachtig ontworpen hebt als fantastische art-director, ben je een lieve vriendin, een mooie, nuchtere Drenthse vrouw, en bovenal een mede-‘ambitieuze moeder’. Ooit richten we een partij op! Met jou als lijsttrekker. Lieve Eva, het is soms best lastig om aan Anna en Wies uit te leggen dat tante Eva geen échte tante is. Maar wat ben je dan wel? Nou ja, you’re my person, you know.Lieve Polke, word nou gewoon ook internist, gezellig! Lieve Bas, jij hoort ook in dit dank-woord; jij maakte mijn studie zoveel leuker! Lieve Merlijn, ik geniet zo van jouw verwon-derende en bewonderende kijk op deze wereld. Lieve Fleur, we ontdekten elkaar wat later, maar ik ben blij dat we dat deden. Laten we daar weer eens een cocktail op drinken! Samen met Merlijn misschien?Lieve Johanna en Jurriaan, wat fijn om dat drukke leven met werk, jong gezin en eigen-wijze pre-puber-peuters te kunnen delen! En ja, hardlopen is een goede uitlaatklep: volgend jaar een gezamenlijke marathon?

En als ik het dan toch over hardlopen heb: lieve John, Michael, Salo, Frans, Eugenie, Mildred, Francesco, Kees, Urtha, Fred, Ineke, Jack, Peter, Joris, Joris en alle andere (ex-)Phanos-genoten. Het drukke leven van een promoverende moeder zou niet vol te houden zijn zonder die kilometers rennen. Op die zaterdagochtenden op de Blaricumse hei, kan het leven wat mij betreft niet veel mooier. Dankjulliewel! Lieve Monique, jij vervult hierin een bijzondere rol. We zijn al sinds 2007 elkaars personal coaches. We hebben al duizenden kilometers samen afgelegd, gepraat, gemopperd en genoten. Onze levens lijken soms gelijk op te gaan, met carrière maken, kinderen krijgen, marathons lopen. Laten we rennen tot we niet meer kunnen! Lieve Kees, je hebt mij geleerd te houden van alles wat oh zo mooi en soms zo lelijk is. Ik kan niet in woorden uitdrukken hoeveel mij dat gebracht heeft.

K007_000000_HereThere and EveryWhere boekje.indd 163 09-02-15 08:30

Page 168: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

164 DANKWOORD

Lieve Samma, jij leerde mij om niet alsmaar te rennen, maar soms gewoon te zijn. Je bent een bijzondere vrouw en vriendin.

Lieve Corine, wanneer ik dit proefschrift zal verdedigen, is het een kleine 9 maanden geleden dat ik je allerlaatste hartslag heb gevoeld. Hoewel ik je mis, ben je nog zoveel in mijn gedachten. Ik denk dat je er vol trots bij had gezeten in die Agnietenkapel. Op een bepaalde manier ben je er gewoon nog een beetje bij.

Lieve Menno & Tina, Roos & Jeroen, Daan & Ferry. Hoe ouder ik word, hoe meer ik mij realiseer wat een groot goed het is dat ons gezin zo ontzettend hecht is. We delen 2 fantastische ouders, 9 geweldige kinderen, en een heleboel lief en leed. En daar ben ik enorm dankbaar voor. Lieve Menno, je bent een amateur-meteoroloog, de droogste grappenmaker ter wereld, maar bovenal een fantastische broer. Lieve Roos, we zijn in vele opzichten best verschillend, maar in al jouw betrokkenheid, belangstelling en steun ben je zo vertrouwd en dichtbij. Wat een rijkdom om zo’n zus te hebben. Lieve Daan, je hebt ons gezin een kleine 30 jaar geleden aangevuld, en je bent nog steeds een volwaar-dig onderdeel van de familie. Wat fijn dat je er bent!Lieve Pappa en Mamma. Ons gezin is uniek, en inmiddels 19 man groot! En nu ik zelf kinderen heb, besef ik pas echt hoe fantastisch jullie ons grootgebracht hebben. Hoe- veel ik van jullie geleerd heb. Hoezeer ik door jullie gesteund ben. Met heel veel trots geef ik nu jullie waardevolle levenslessen door aan mijn eigen kleine Anna en Wies.

Lieve Tijs. Vraag je je wel eens af waar je destijds je steun aan hebt betuigd, toen ik op-perde om geen artdirector meer te zijn maar geneeskunde te gaan studeren? I’ve put you through a lot. En hoewel ik dingen doe op een dag waar jij nooit aan zou moeten denken, je interesse en enthousiasme zijn oneindig en fantastisch. Om over alle zorgen voor de meisjes waar ik door dat idioot drukke leven geen tijd voor heb nog maar te zwijgen.

Lieve Anna & Wies, mijn kleine meisjes. Anna, alweer zo groot, zo wijs. Als ik af mag gaan op jouw interessante vragen waar ik lang niet altijd een antwoord op weet, kan het niet anders of jij wordt ooit nog eens een onderzoekster. Lief meisje, je verrast me zo met al jouw talenten en interesses. Ik ben zo trots als een pauw! Kleine Wies, geboren tijdens deze promotie jaren, en ook jij bent alweer zo groot. En hoewel je het nog niet zo goed vertellen kan, kleurt jouw wereld die van mij elke dag een beetje mooier. Lieve meisjes, ik hou van jullie. Zó veel.

K007_000000_HereThere and EveryWhere boekje.indd 164 09-02-15 08:30

Page 169: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

165BIOGRAFIE

Biografie

Marieke Guurtje Schooneman werd geboren op 22 september 1979 en groeide op in de Beemster met haar ouders, broer, zus en pleegzus. Initieel wilde zij arts worden, maar het leven liep anders. Zij doorliep zonder middelbare schooldiploma de academie voor art direction en design. Hierna studeerde zij aan de Hallo©academie voor toegepaste creativiteit, om vervolgens art director te worden bij reclamebureau KesselsKramer in Amsterdam. Maar na enkele jaren in deze creatieve functie, deed een boeddhistische monnik in Chang Mai, Thailand haar inzien dat de reclame het niet was voor de rest van haar leven.

De wens om arts te worden bestond nog steeds: zo gebeurde het dat ze op haar 24e alsnog in de avonduren haar VWO diploma haalde. In september 2005 werd ze ingeloot voor geneeskunde aan het AMC. De studie doorliep ze, iets ouder dan haar medestu-denten, met véél overtuiging, voorspoed en plezier.

Na het behalen van haar artsenbul in juni 2011, is zij promotieonderzoek gaan doen onder begeleiding van promotores Prof. Carla Hollak en Prof. Ronald Wanders en co- promotores Dr. Maarten Soeters en Dr. Sander Houten, aan de afdeling Endocrinologie & Metabolisme in het AMC. Sinds 1 januari 2015 is zij in opleiding tot internist in het AMC. Samen met Mattijs heeft zij twee prachtige dochters, Anna en Wies, van 6 en 3 jaar oud. Vanuit de Amsterdamse Pijp, waar zij woont, rent ze vele rondjes langs de Amstel richting Ouderkerk en verder. In september 2014 liep zij in Berlijn haar 7e marathon in een PR van 3:19:57.

K007_000000_HereThere and EveryWhere boekje.indd 165 09-02-15 08:30

Page 170: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

K007_000000_HereThere and EveryWhere boekje.indd 166 09-02-15 08:30

Page 171: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

167A SHORT INTRODUCTION

K007_000000_HereThere and EveryWhere boekje.indd 167 09-02-15 09:13

Page 172: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

K007_000000_HereThere and EveryWhere boekje.indd 168 09-02-15 09:13

Page 173: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

169A SHORT INTRODUCTION

K007_000000_HereThere and EveryWhere boekje.indd 169 09-02-15 09:13

Page 174: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

170 A SHORT INTRODUCTION

K007_000000_HereThere and EveryWhere boekje.indd 170 09-02-15 09:13

Page 175: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

171A SHORT INTRODUCTION

K007_000000_HereThere and EveryWhere boekje.indd 171 09-02-15 09:13

Page 176: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

172 A SHORT INTRODUCTION

K007_000000_HereThere and EveryWhere boekje.indd 172 09-02-15 09:13

Page 177: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

173A SHORT INTRODUCTION

K007_000000_HereThere and EveryWhere boekje.indd 173 09-02-15 09:13

Page 178: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

174 A SHORT INTRODUCTION

K007_000000_HereThere and EveryWhere boekje.indd 174 09-02-15 09:13

Page 179: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

175A SHORT INTRODUCTION

K007_000000_HereThere and EveryWhere boekje.indd 175 09-02-15 09:13

Page 180: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

176 A SHORT INTRODUCTION

K007_000000_HereThere and EveryWhere boekje.indd 176 09-02-15 09:13

Page 181: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

Carnitine (kar´nətēn´)

From carne (Spanish, Italian) or caro (Latin)

1. f. meat; asada roast beef;

de carnero lamb;

de puerco pork;

de ternera veal;

2. flesh, muscular tissue of the body

K007_000000_HereThere and EveryWhere boekje.indd 177 09-02-15 09:13

Page 182: UvA-DARE (Digital Academic Repository) Here, there and ...€¦ · Here, there and everywhere. A multi organ approach to acylcarnitine metabolism Schooneman, M.G. Link to publication

K007_000000_HereThere and EveryWhere boekje.indd 178 09-02-15 09:13