Download Fulltext - Jultika

Document related concepts

NADH:ubiquinone oxidoreductase (H+-translocating) wikipedia , lookup

Protein wikipedia , lookup

Oxidative phosphorylation wikipedia , lookup

Gene regulatory network wikipedia , lookup

Interactome wikipedia , lookup

Evolution of metal ions in biological systems wikipedia , lookup

Paracrine signalling wikipedia , lookup

Silencer (genetics) wikipedia , lookup

Signal transduction wikipedia , lookup

Citric acid cycle wikipedia , lookup

Gene expression wikipedia , lookup

Magnesium transporter wikipedia , lookup

Amino acid synthesis wikipedia , lookup

Butyric acid wikipedia , lookup

Lipid signaling wikipedia , lookup

Point mutation wikipedia , lookup

Endogenous retrovirus wikipedia , lookup

Enzyme wikipedia , lookup

Western blot wikipedia , lookup

Metalloprotein wikipedia , lookup

Specialized pro-resolving mediators wikipedia , lookup

Protein–protein interaction wikipedia , lookup

Artificial gene synthesis wikipedia , lookup

Biochemistry wikipedia , lookup

Biosynthesis wikipedia , lookup

Expression vector wikipedia , lookup

Metabolism wikipedia , lookup

Hepoxilin wikipedia , lookup

Proteolysis wikipedia , lookup

Two-hybrid screening wikipedia , lookup

Mitochondrion wikipedia , lookup

Fatty acid synthesis wikipedia , lookup

Biosynthesis of doxorubicin wikipedia , lookup

Mitochondrial replacement therapy wikipedia , lookup

Fatty acid metabolism wikipedia , lookup

Transcript
A 519
OULU 2008
U N I V E R S I T Y O F O U L U P. O. B . 7 5 0 0 F I - 9 0 0 1 4 U N I V E R S I T Y O F O U L U F I N L A N D
U N I V E R S I TAT I S
S E R I E S
SCIENTIAE RERUM NATURALIUM
Professor Mikko Siponen
HUMANIORA
University Lecturer Elise Kärkkäinen
TECHNICA
Professor Hannu Heusala
MEDICA
Professor Olli Vuolteenaho
SCIENTIAE RERUM SOCIALIUM
ACTA
UN
NIIVVEERRSSIITTAT
ATIISS O
OU
ULLU
UEEN
NSSIISS
U
Zhijun Chen
E D I T O R S
Zhijun Chen
A
B
C
D
E
F
G
O U L U E N S I S
ACTA
A C TA
A 519
CHARACTERIZATION OF
THE 2-ENOYL THIOESTER
REDUCTASE OF
MITOCHONDRIAL FATTY
ACID SYNTHESIS TYPE II
IN MAMMALS
Senior Researcher Eila Estola
SCRIPTA ACADEMICA
Information officer Tiina Pistokoski
OECONOMICA
University Lecturer Seppo Eriksson
EDITOR IN CHIEF
Professor Olli Vuolteenaho
PUBLICATIONS EDITOR
Publications Editor Kirsti Nurkkala
ISBN 978-951-42-8979-8 (Paperback)
ISBN 978-951-42-8980-4 (PDF)
ISSN 0355-3191 (Print)
ISSN 1796-220X (Online)
FACULTY OF SCIENCE,
DEPARTMENT OF BIOCHEMISTRY, UNIVERSITY OF OULU;
BIOCENTER OULU,
UNIVERSITY OF OULU
A
SCIENTIAE RERUM
RERUM
SCIENTIAE
NATURALIUM
NATURALIUM
ACTA UNIVERSITATIS OULUENSIS
A Scientiae Rerum Naturalium 519
ZHIJUN CHEN
CHARACTERIZATION OF THE
2-ENOYL THIOESTER REDUCTASE
OF MITOCHONDRIAL FATTY ACID
SYNTHESIS TYPE II IN MAMMALS
Academic dissertation to be presented, with the assent of
the Faculty of Science of the University of Oulu, for public
defence in Auditorium GO101, Linnanmaa, on December
4th, 2008, at 12 noon
O U L U N Y L I O P I S TO, O U L U 2 0 0 8
Copyright © 2008
Acta Univ. Oul. A 519, 2008
Supervised by
Professor Kalervo Hiltunen
Reviewed by
Docent Peter Mattjus
Docent Sami Väisänen
ISBN 978-951-42-8979-8 (Paperback)
ISBN 978-951-42-8980-4 (PDF)
http://herkules.oulu.fi/isbn9789514289804/
ISSN 0355-3191 (Printed)
ISSN 1796-220X (Online)
http://herkules.oulu.fi/issn03553191/
Cover design
Raimo Ahonen
OULU UNIVERSITY PRESS
OULU 2008
Chen, Zhijun, Characterization of the 2-enoyl thioester reductase of mitochondrial
fatty acid synthesis type II in mammals
Faculty of Science, Department of Biochemistry, University of Oulu, P.O.Box 3000, FI-90014
University of Oulu, Finland; Biocenter Oulu, University of Oulu, P.O. Box 5000, FI-90014
University of Oulu, Finland
Acta Univ. Oul. A 519, 2008
Oulu, Finland
Abstract
A data base search using the amino acid sequence of Saccharomyces cerevisiae Etr1p, the last
enzyme of mitochondrial fatty acid synthesis type II (FAS II), revealed a highly similar human
protein, NRBF-1. Expression of NRBF-1 in a yeast etr1? strain rescued its respiratory deficiency.
NRBF-1 resides in mitochondria in cultured HeLa cells. The recombinant NRBF-1 is
enzymatically active, reducing 2E-enoyl-CoAs to acyl-CoAs in an NADPH-dependent manner.
Altogether, our data showed that NRBF-1 is a mitochondrial 2-enoyl-CoA reductase/2-enoyl
thioester reductase (MECR/ETR1), the human functional counterpart of yeast Etr1p. In addition,
MECR was also isolated from bovine heart. It turns out that mammals contain a mitochondrial
FAS II pathway, in addition to cytoplasmic FAS I.
To investigate the functional mechanism of MECR/ETR1 at the molecular level, the protein
was crystallized and the crystal structure determined. The apo-structure of MECR/ETR1 contains
two sulfates in the nucleotide binding site and the domain arrangement resembles the NADPHcontaining holo-structure of yeast Etr1p. The predicted mode of NADPH-binding and kinetic data
suggest that Tyr94 and Trp311 play critical roles in catalysis. A pocket was found in the structure
extending away from the catalytic site that can accommodate fatty acyl chains up to 16 carbons.
An acyl carrier protein (ACP) binding site was also suggested.
To study the physiological function of mouse Mecr, two lines of transgenic mice
overexpressing Mecr were generated. The Mecr transgenic mice developed cardiac and
mitochondrial abnormalities. The phenotyping was carried out using echocardiography, heart
perfusion, histology, and endurance testing. Our results suggest Mecr plays a role in mitochondrial
and heart function. Therefore, inappropriate expression of the genes of FAS II may result in the
development of cardiomyopathy.
Keywords: biochemistry, lipid, MECR/ETR1, mitochondria, structural enzymology,
transgenic mice
Acknowledgements
The work has been carried out at Biocenter Oulu and Department of Biochemistry,
University of Oulu, during the years 2001–2008.
I would like to express my deepest gratitude to my supervisor Professor
Kalervo Hiltunen for his excellent guidance and constant support during this
study. He shared with me his hand-on experiences from protein purification from
animal tissues to communication skills. He has also provided numerous
opportunities for me to learn different methods and to use multiple techniques to
tackle biological questions. I have also learned from him to be optimistic and
persistent. I am grateful to Professor Rik Wierenga for helping me to understand
structural enzymology and for guidance in the structure related projects. I would
like to thank Professor Yari Ylänne for his invaluable comments during and after
evaluations by the thesis committee, influencing not only my projects but also my
career. I wish to thank Professor Seppo Vainio for his kind advice and numerous
great ideas concerning my mouse projects as well as potential future topics. I also
wish to express my special thanks to Professor Raili Myllylä for her
encouragement and her very constructive suggestions. I wish to thank Professor
Lloyd Ruddock for sharing with me skills in biochemistry and tips in molecular
biology and cell biology. My special thanks also to Dr. Ilkka Miinalainen and Dr.
Antti Haapalainen for their excellent work during collaboration through this study.
I am very grateful to Dr. Sami Väisänen and Dr. Peter Mattjus for acting as
reviewers of this thesis and providing constructive comments and Dr. Deborah
Kaska for the revision of the language.
I wish to warmly acknowledge Dr. Raija Soininen in the Biocenter Oulu
mouse facility, Professor Ilmo Hassinen in the department of medical
biochemistry, Professor Heikki Roskoho in department of pharmacology and
toxicology, Professor Petri Kovanen in the Wihuri Research Institute, and
Professor Walter Koch in Thomas Jefferson University for their important
guidance. I would like to acknowledge Dr. Hanna Leskinen, Dr. Raija Sormunen,
and Dr. Pasi Tavi for teaching me how to study the cardiac phenotype of
transgenic mice. I wish to thank Dr. Alexander Kastaniotis for sharing with me
the essence of yeast genetics and kind suggestions in writing. I would like to
thank Dr. Hanna-Marja Voipio in the Laboratory Animal Centre University of
Oulu, Professor Timo Nevalainen in the National Animal Center Kuopio
University for teaching me how to handle animals. I wish to thank Professor Aner
Gurvitz at the Medical University of Vienna for important comments on scientific
5
writing. I wish to thank Dr. Erkki Liimatta, Dr. Regina Pudas, and Satyan Sharma,
Professor Vasily Antonenkov, Dr. Tuomo Glumoff, Dr. Pekka Kilpeläinen, Dr.
Ulrich Bergmann, Dr. Päivi Pirila, Dr. Steffen Ohlmeier, Dr. Sakari Kellokumpu,
Dr. Aki Manninen, Dr. André Juffer and Dr. Apaja Pirjo for their help and
suggestions.
I wish to thank all the current and former members of the KH group, as well
as colleagues in Oulu University: Professor Yongmei Qin, Petri Lackman, Dr.
Mari Ylianttila, Eija Selkälä, Aare Rokka, François Pujol, Dr. Tiina Kotti, Dr.
Juha Torkko, Samuli Kursu, Dr. Kaija Autio, Dr. Kristian Koski, Tatu Haataja,
Miia Vapola, Dr. Silke Grunau, Maija Malin, Ville Ratas, Leena Ollitervo, Marika
Kamps, Virpi Hannas, Johanna Moilanen, Katri Näppä, Fumi Suomi, Antonina
Shvetsova, Antti Isomursu, Dr. Kalle Savolainen, Dr. Prasenjit Bhaumik, Dr.
Jukka Taskinen, Dr. Tiila-Riikka Kiema, Dr. Ari-pekka Kvist, Kyösti Keränen,
Jaakko Keskitalo, Pia Askonen, Anneli Kaattari, Tuula Koret, Anne Vainionpää,
Jonna Ranta, Sanna Kauppinen, Eija Nissinen, Tanja Sankala, Sanna Haapalainen,
Tarja Huhta, Vivii Majava, Antti Salo, Parasad Kasaragod, Mirva Peltoniemi, Van
Dat Nguyen, Heli Alanen, Marco Casteleijn, Ilkka Pietilä, and many others. I also
wish to thank Chinese friends in Biocenter Oulu: Chunguang, Weidong, Hongmin,
Jindong, Yanfeng, Shaobing, and Lijun for their help.
I wish to acknowledge my M.Sc. mentors Professor Yi Li, Professor
Zhangliang Chen, Dr. Xiaotian Ming and Professor Changfa Xu and Professor
Lanxian Wang at Peking University for their important guidance.
I am deeply grateful to my father Weimiao Chen, my mother Judan Jin for
their great love and great support. I wish also to express the gratitude to my
father-in-law Professor Desheng Wang, and mother-in-law Xiuzheng Hu for their
enormous support. My warmest thanks belong to my dear wife Hui Wang, and my
dear children, Zhuotong, Zhuokun. The thesis could not have been possible
without their great love, great understanding and great support.
This work was financially supported by grants from the Sigrid Jusélius
Foundation, the Academy of Finland, the NordForsk under the Nordic Centres of
Excellence programme in Food, Nutrition and Health, Project (070010)
MitoHealth”, CIMO and scholarships from the Finnish Cultural Foundation, and
the Oulu University Scholarship Foundation.
Oulu, 2008
6
Zhijun Chen
Abbreviations
ACC
ACP
ACPS
AMPK
ATP
CoA
CPT
Cr
CrP
DAPI
DTP
EF
ETR
FAO
FAS
GFP
HSD
HTD
IEM
IVS
KAS
KAR
kDa
LVEDD
LVESD
LVFS
MCAT
MCD
mDa
MDR
MECR
MFE-2
αMHC
MRF
MT-I
acetyl-CoA carboxylase
acyl carrier protein
holo-ACP synthase
5'-adenosine monophosphate-activated protein kinase
adenosine triphosphate
coenzyme A
carnitine palmitoyltransferse
creatine
creatine phosphate
4', 6-diamidino-2'-phenylindole dihydrochloride
dicarboxylate transport protein
ejection fraction
enoyl thioester reductase
fatty acid oxidation
fatty acid synthesis
green fluorescent protein
hydroxysteroid dehydrogenase
3R-hydroxyacyl-ACP dehydratase
immunoelectron microscopy
interventricular septum
β-ketoacyl synthase
β-ketoacyl reductase
kilo Dalton
left ventricular end-diastolic
left ventricular end-systolic
LV fractional shorting
malonyl-CoA:ACP transacylase (malonyltransferase)
malonyl-CoA decarboxylase
mega Dalton
medium-chain dehydrogenase/reductase
mitochondrial 2-enoyl-CoA reductase
multifunctional enzyme type 2
alpha myosin heavy chain
mitochondrial respiratory function
metallothionein-I
7
MTF-I
NAD(P)
NR
NRBF-1
PBS
PCR
PCC
PDC
PPT
PW
RT-PCR
SDR
SDS-PAGE
TCA
TEM
TG
tg
wt
8
metal-regulatory factor-1
nicotinamide adenine dinucleotide (phosphate)
nuclear receptor
nuclear receptor binding factor 1
phosphate buffered saline
polymerase chain reaction
propionyl-CoA carboxylase
pyruvate dehydrogenase complex
phosphopantetheine:protein transferase
posterior wall
reverse template-PCR
short-chain dehydrogenase/reductase
sodium dodecyl sulfate polyacrylamide gel electrophoresis
tricarboxylic acid
transmission electron microscopy
triacylglycerol
transgenic
wild type
List of original articles
This thesis is based on the following articles, which are referred to in the text by
their Roman numerals:
I
Miinalainen IJ*, Chen ZJ, Torkko JM, Pirilä PL, Sormunen RT, Bergmann U, Qin
YM & Hiltunen JK (2003) Characterization of 2-enoyl thioester reductase from
mammals – an ortholog of Ybr026p/Mrf1’p of the yeast mitochondrial FAS type II.
J Biol Chem 278: 20154–20161.
II Chen ZJ, Pudas R, Sharma S, Smart O, Juffer A, Hiltunen JK, Wierenga RK &
Haapalainen AM (2008) Structure and enzymology of 2-enoyl thioester reductase of
human FAS II: New insights into substrate binding and catalysis. J Mol Biol 379:
830–844.
III Chen ZJ, Leskinen H, Liimatta E, Sormunen RT, Miinalainen IJ, Hassinen IE &
Hiltunen JK (2008) Overexpression of Mecr, a gene of mitochondrial FAS II in heart
leads to cardiac dysfunction in mouse. Manuscript.
*Publication I has been used as part of the dissertation of Ilkka Miinalainen
(ISBN 9514282442).
9
10
Contents
Abstract
Acknowledgements
5
Abbreviations
7
List of original articles
9
Contents
11
1 Introduction
15
2 Review of the literature
17
2.1 Polyunsaturated fatty acids (PUFA), branched-chain fatty acids,
and trans fatty acids ................................................................................ 17
2.2 The transcriptional regulation of lipid metabolism in mammals............. 18
2.2.1 Retinoid X receptors (RXRs) ....................................................... 19
2.2.2 SREBP.......................................................................................... 19
2.2.3 Carbohydrate responsive element-binding protein
(ChREBP)..................................................................................... 19
2.2.4 X-box binding protein-1 (XBP1).................................................. 20
2.2.5 Peroxisome proliferator-activated receptors (PPARs) .................. 20
2.2.6 Peroxisome proliferator-activated receptor γ coactivator
(PGC) ........................................................................................... 21
2.2.7 RIP140.......................................................................................... 21
2.2.8 Other regulators in lipid metabolism ............................................ 22
2.3 The organelles in mammalian cells......................................................... 22
2.3.1 The single membrane organelles .................................................. 22
2.3.2 The double membrane organelles................................................. 23
2.3.3 Mitochondria ................................................................................ 23
2.4 The components of mitochondrial FAS II............................................... 25
2.4.1 Acyl carrier protein....................................................................... 25
2.4.2 Phosphopantetheine:protein transferase (PPTase) ........................ 25
2.4.3 Malonyl-CoA:ACP transacylase (malonyltransferase;
MCAT) ......................................................................................... 26
2.4.4 β-Ketoacyl synthase (KAS) .......................................................... 27
2.4.5 β-Ketoacyl reductase (KAR) ........................................................ 27
2.4.6 3R-Hydroxyacyl-ACP dehydratases (HTD)................................. 28
2.4.7 2-Enoyl thioester reductase (ETR1) ............................................. 28
2.4.8 The FAS II components of Caenorhabditis elegans...................... 29
2.5 Where is malonyl-CoA generated in mammalian mitochondria? ........... 30
11
2.5.1 Acetyl-CoA carboxylase 2 (ACC2) .............................................. 31
2.5.2 Propionyl-CoA carboxylase (PCC)............................................... 32
2.5.3 Putative malonyl-CoA synthetase................................................. 32
2.5.4 Putative malonyl-CoA:ACP transacylase ..................................... 34
2.6 Structural aspects of FAS II components ................................................ 34
2.6.1 Short chain dehydrogenase/reductase (SDR) ............................... 34
2.6.2 Medium-chain dehydrogenase/reductase (MDR) ......................... 36
2.6.3 Interactions of FAS II enzymes and ACP ..................................... 37
2.6.4 Cofactor specificity....................................................................... 37
2.6.5 Substrate binding pocket .............................................................. 38
2.7 Myocardial metabolism and cardiomyopathies....................................... 38
2.7.1 Mitochondrial energy metabolism and heart failure..................... 38
2.7.2 Energy metabolism in ischemic cardiomyopathy ......................... 39
2.7.3 Energy metabolism in dilated cardiomyopathy and
hypertrophic cardiomyopathy....................................................... 39
2.7.4 Energy metabolism in diabetic cardiomyopathy and other
cardiomyopathies.......................................................................... 40
3 Outlines of the present study
41
4 Materials and methods
43
4.1 Northern blot analysis (I) ........................................................................ 43
4.2 Cloning and mutagenesis (I, II, III)......................................................... 43
4.3 Protein expression and purification (I, II) ............................................... 43
4.4 Enzymatic assay and determination of kinetic parameters for
MECR/ETR1 and its mutated forms (I, II).............................................. 44
4.5 Reaction product analysis (I)................................................................... 44
4.6 Fluorescence microscopy (I) ................................................................... 44
4.7 TEM analysis of ybr026c∆ cells expressing MECR (I) .......................... 45
4.8 Crystallization of MECR/ETR1 (II)........................................................ 45
4.9 Data collection, structure determination and refinement (II) .................. 45
4.10 Modeling of NADPH and fatty acyl thioester in the active site of
MECR/ETR1 (II) .................................................................................... 46
4.11 Animal care (III) ..................................................................................... 46
4.12 Generation of metallothionein-I (MT-I)-Mecr overexpressing
mice (III) ................................................................................................. 46
4.13 Southern blot genotyping (III)................................................................. 46
4.14 Reverse template PCR and real time PCR (III)....................................... 47
4.15 Histological staining (III) ........................................................................ 47
12
4.16 Echocardiographic analysis (III) ............................................................. 47
4.17 Heart perfusion (III) ................................................................................ 47
4.18 Metabolite assays (III) ............................................................................ 48
4.19 Exercise performance test (III)................................................................ 48
4.20 Isolation of heart mitochondria (I, III) .................................................... 48
4.21 Statistical analysis (III) ........................................................................... 48
5 Results
51
5.1 Biochemical properties of MECR/ETR1 ................................................ 51
5.1.1 Yeast etr1 mutant complementation ............................................. 51
5.1.2 Protein purification....................................................................... 51
5.1.3 End product analysis..................................................................... 51
5.1.4 Subcellular localization ................................................................ 52
5.1.5 Tissue distribution ........................................................................ 52
5.2 Structural enzymology of MECR/ETR1 ................................................. 52
5.2.1 The overall structure..................................................................... 52
5.2.2 Substrate binding pocket .............................................................. 53
5.2.3 Binding of ACP ............................................................................ 53
5.2.4 Kinetics......................................................................................... 53
5.3 Mecr transgenic mice .............................................................................. 54
5.3.1 Transgenic mice............................................................................ 54
5.3.2 Generation of Mecr oxerexpressing mice..................................... 55
5.3.3 Echocardiography......................................................................... 56
5.3.4 Heart perfusion ............................................................................. 56
5.3.5 Exercise endurance test ................................................................ 56
6 Discussion
57
6.1 2-Enoyl thioester reductases (ETRs)....................................................... 57
6.2 Lipoic acid and other products................................................................ 58
6.3 CoA and ACP .......................................................................................... 59
6.4 FAS II enzymes exist at low levels in mammalian mitochondria ........... 60
6.5 FAS II and its physiological function...................................................... 61
7 Conclusions
63
References
65
Original articles
83
13
14
1
Introduction
In living cells, two distinct fatty acid synthesis systems have been revealed so far,
namely, fatty acid synthesis types I (FAS I) and II (FAS II) (Rock & Cronan
1996).
In bacteria and in the plastids (chloroplasts) of plants, FAS II serves as the
major route for generating cellular fatty acids with reactions catalyzed by a series
of discrete enzymes (White et al. 2005). The bacterial FAS II system produces the
β-hydroxy fatty acyl groups for lipid A of lipopolysaccharides, intermediates used
in vitamin (biotin, lipoic acid, etc) synthesis, fatty acyl moieties used for protein
modifications and acyl groups for the synthesis of cellular triacylglycerol (TG)
and phospholipids (Rock & Cronan 1996). Progress has been made recently in the
development of antibacterials using bacterial FAS II proteins as targets (Heath &
Rock 2004).
In contrast, in the cells of fungi, mammals, certain mycobacteria, and
Corynebacterium ammoniagenes, the vast majority of endogenously derived fatty
acids are synthesized in the cytosol through a large polyfunctional enzyme (FAS
I). This enzyme is heterododecameric (α6β6) in fungi and homodimeric (α2) in
mammals, and in which each of the reactions is catalyzed by distinct domains in
the enzyme (Tsukamoto et al. 1983, Smith 1994, Rock & Cronan 1996,
Schweizer & Hofmann 2004).
An additional biosynthetic process, also conserved in mammals, is the
recently identified mitochondrial fatty acid synthesis (Torkko et al. 2001, Joshi et
al. 2003, Zhang et al. 2003a, Kastaniotis et al. 2004, Cronan et al. 2005, Zhang et
al. 2005, Autio et al. 2007). In this system, the required reactions are catalyzed by
separate enzymes and, therefore, the system resembles the well-characterized
bacterial FAS II (White et al. 2005). The endoplasmic reticulum (ER) of
eukaryotes also contains a fatty acid chain elongation system, producing longchain and very long-chain fatty acids (Cinti et al. 1992, Das et al. 2000, Moon &
Horton 2003).
The last enzyme of mammalian FAS II is represented by mitochondrial enoylCoA reductase/2-enoyl thioester reductase (MECR/ETR1; EC 1.3.1.38). This
enzyme catalyzes the reaction (Torkko et al. 2001):
2E-enoyl-(ACP/CoA) + NADPH + H+ → acyl-(ACP/CoA) + NADP+
The present thesis is concerned with the characterization of this enzyme in
humans and mice with reference to fatty acid biosynthesis. To gain more insight
15
into the function of mammalian mitochondrial FAS II, biochemical, structural,
and genetic characterizations of the mammalian MECR/ETR1 were undertaken.
16
2
Review of the literature
2.1
Polyunsaturated fatty acids (PUFA), branched-chain fatty acids,
and trans fatty acids
Fatty acids (FAs) are among the key components of lipids and over 1000 different
FAs have been found in nature to date. Mammalian tissues contain some 250
different FAs, 20–30 of which are major FAs. Notably, the concentration of free
FAs in human blood serum is on a micromolar level (Jungling & Kammermeier
1988).
Most FAs are straight-chain compounds, the simplest of which is saturated
FA. However, many straight-chain FAs contain double bonds, ranging from 1 to 6
or more, with a free methyl between two double bonds. The double bonds have
significant influence on the physical properties (e.g., the melting point) of FAs.
Moreover, the positions of double bonds in FAs also affect the biochemical
properties of FAs (Michaud et al. 2002). When counting from the ω-end of FAs,
the first double bond which mammals can introduce into the existing covalent
chain is in the ∆9 position. Therefore, mammals are completely dependent on
dietary intake of ∆3 and ∆6 (poly)-unsaturated FAs, which, however, can be remodified in the body to longer chain FAs with a higher degree of unsaturation
through a retroconversion pathway. The retroconversion requires participation of
enzymes in both the ER and peroxisomes using unsaturated FAs such as 18:3n-3
as substrates. For instance, docosahexaenoic acid (22:6n-3), which is critical for
maintaining the function of the nervous system (Salem et al. 2001), arises either
exogenously or from eicosapentaenoic acid (EPA, C20:5n-3) through chain
elongation and desaturation in the ER followed by chain contraction in
peroxisomal β-oxidation. Moreover, PUFAs can suppress glycolytic and lipogenic
genes by inhibiting the translocation of the carbohydrate responsive elementbinding protein (ChREBP) from the cytosol to the nucleus (Dentin et al. 2005).
Notably, diet affects the n-3 PUFA/n-6 PUFA ratio of cell membranes.
Additionally, exogenous PUFA supply regulates endogenous monounsaturated
fatty acid (MUFA) production through modulation of ∆9-desaturase activity by
interfering with the function of sterol regulatory element binding proteins
(SREBP) (Tabor et al. 1999).
Another group of FAs is branched-chain FAs which can be found from
microbial and animal lipids. In mammals, branched-chain FAs may act as bound
17
components of hair fibers for maintaining hair hydrophobicity (Jones & Rivett
1997) or esterified to cholesterol, binding to certain enzymes of protein
biosynthesis (Tuhackova & Hradec 1985, Hradec & Dufek 1994). The branchedchain FAs can be associated also with brain function and act as an inhibitor of the
growth of cancer (Yang et al. 2000, Kniazeva et al. 2004).
Trans fatty acids (TFAs) are special unsaturated straight-chain compounds.
Most of the trans fatty acids (TFAs) of tissues originate from processed vegetable
fats or from milk. TFAs sometimes will raise LDL and lower HDL in serum and
also raise the risk of cardiovascular disease by an unclear mechanism (Gebauer et
al. 2007).
2.2
The transcriptional regulation of lipid metabolism in mammals
Cellular lipid homeostasis is tightly regulated at both the transcriptional and posttranscriptional levels and is connected closely to glucose metabolism. Many
metabolic genes are transcribed at a basic level but can be activated or repressed
in response to changes in the nutritional or physiological state (White et al. 2008).
Many transcription factors in lipid metabolism belong to the superfamily of
nuclear receptors (NRs), which consist of an N-terminal domain, a DNA binding
domain, a hinge region, a ligand binding domain, and a C-terminal domain
(Chawla et al. 2001) (Fig. 1). Most of the known NRs are mainly regulated
through ligand interaction. Upon ligand binding, these NRs are dissociated from
heat shock proteins, translocated into the nucleus and bind to hormone response
elements. The ligands can be either endogenous or exogenous, and can act as
agonists or antagonists. NRs also recruit co-activators to the promoter or enhancer
regions of target genes. In contrast to this, the apo form of NRs recruits corepressors and thus inhibits gene expression. Notably, in the nucleus, regardless of
ligand binding, NRs form heterodimers or homodimers. Furthermore, NRs
together with co-activators/repressors can activate histone (de)acetylases, and
other chromatin remodeling complexes, hence change chromatin structure to
promote or inhibit RNA polymerase II access to the target promoter.
DNA binding
NH2
Fig. 1. Schematic structure of a typical NR.
18
ligand binding
COOH
2.2.1 Retinoid X receptors (RXRs)
Specifically, retinoid X receptors (RXRs) can regulate liver X receptors (LXRs),
peroxisome proliferators-activated receptors (PPARs), and farnesoid X receptors
(FXR). LXR forms heterodimers with RXR where both subunits can be
stimulated by ligands (Willy et al. 1995, Svensson et al. 2003). LXR regulates
sterol adsorption in the intestine, cholesterol efflux from peripheral cells,
cholesterol transport in the bloodstream, and cholesterol secretion into bile. LXR
also enhances lipogenesis and increases serum triacylglycerol (TG) levels. Bile
acids can activate FXR, and thus regulate many metabolic pathways (Lee et al.
2006).
2.2.2 SREBP
Another group of transcription factors is the basic helix-loop-helix leucine zipper
proteins (bHLH/LZ), such as, SREBP and upstream stimulating factors (USF)
(Goldstein et al. 2006). SREBPs are master regulators of lipid homeostasis
through binding to the sterol regulatory element (SRE) in the promoter of target
genes. Fourteen enzymes in the cholesterol synthesis pathway are more or less
regulated by SREBP (mainly -1a and -2). Via interaction with the SREBP
complex, both cholesterol and its oxidized forms can inhibit the transport of the
SREBP cleavage-activating protein/SREBP (SCAP/SREBP) to the Golgi
(Nohturfft et al. 1999). Specifically, SREBP-1a regulates both cholesterol and
fatty acid synthesis (FAS); SREBP-1c regulates hepatic and adipose tissue FAS
and glucose metabolism; moreover, SREBP-2 responds to cellular cholesterol
levels (Shimano et al. 1997, Joseph et al. 2002).
2.2.3 Carbohydrate responsive element-binding protein (ChREBP)
The ChREBP is another bHLH/LZ transcription factor (Yamashita et al. 2001),
which can directly regulate glycolytic and lipogenic gene expression through
binding to their promoters sequences (Ishii et al. 2004). Interestingly, like many
other transcription factors, ChREBP forms heterodimers with the Max-like
protein X (Mlx) to generate a functional complex (Stoeckman et al. 2004, Ma et
al. 2005, Ma et al. 2006). The roles of ChREBP/Mlx in different tissues remain to
be identified (Postic et al. 2007).
19
2.2.4 X-box binding protein-1 (XBP1)
XBP1 is a key regulator of the unfolded protein response in the ER in mammals
(Ron & Walter 2007). Upon ER stress in mammals, an unconventional spliced
version of XBP1 mRNA induced by the proximal sensor and endoribonuclease
IRE1α generates an active transcription factor, binding to the promoter of ER
chaperone genes (Lee et al. 2003, Shaffer et al. 2004, Acosta-Alvear et al. 2007).
Most recently, XBP1 was identified as a novel transcription factor governing
hepatic lipogenesis by activating the transcription of key lipogenic genes in the
liver, and can be induced by a high-carbohydrate diet. XBP1 liver conditional
knock-out mice have a significant decrease in de novo hepatic lipid synthesis,
which further leads to decreases in serum TG, cholesterol, and free FAs (Lee et al.
2008).
2.2.5 Peroxisome proliferator-activated receptors (PPARs)
Like many other NRs, PPARs can respond to dietary lipids and form heterodimers
with the RXRs and act as transcription factors. PPARs are modulated through
ligand/coactivator/corepressor binding, which can stimulate or inhibit their
function (Yu & Reddy 2007). There are three PPAR isoforms. PPARα is highly
expressed in liver, heart and kidney and acts as a mediator of FAs metabolism by
upregulating enzymes of fatty acid oxidation (FAO), or downregulating enzymes
of FAS such as acetyl-CoA carboxylase 1 (ACC1) (Lee et al. 2002). For instance,
when ACC1 is down-regulated, the concentration of malonyl-CoA decreases,
resulting in the activation of carnitine palmitoyltransferase I (CPT-1), and
consequently more FAs are transported into mitochondria and used for FAO.
PPARγ is highly expressed in adipose tissue and the immune system.
Expression of PPARγ in fibroblasts and muscle cells leads to adipocyte
differentiation (Tontonoz et al. 1994, Hu et al. 1995), which may be achieved
through interaction with PPARγ/RXR, C/EBPs and ADD-1/SREBP-1 and result
in the expression of adipocyte-specific genes such as aP2, leptin, and lipoprotein
lipase (Auwerx 1999, Rosen & Spiegelman 2000). PPARγ knockout mice show a
phenotype of early embryonic lethality, and the embryos are devoid of adipose
tissue (Barak et al. 1999), suggesting that PPARγ plays a role in the development
of multiple tissues. The ligands of PPARγ have not yet been identified
conclusively, but the reported ligands include the thiazolidinediones (TZDs)
(Kletzien et al. 1992), PUFAs (Xu et al. 1999), eicosanoids (Yu et al. 1995), and
20
many others. PPARγ is associated with both lipid and glucose metabolism and is
regarded as a potential drug target for the treatment of multiple diseases including
diabetes, cancer and inflammation (Houseknecht et al. 2002).
PPARδ is ubiquitously expressed, but not as well-characterized as the other
two PPARs. Wang and coauthors generated PPARδ transgenic mice using an aP-2
promoter which is adipocyte-specific (Wang et al. 2004). Notably, this form of
PPARδ contains a VP-tag that converts the PPARδ into an active transcription
factor even when a ligand is not present. The PPARδ transgenic mice show a lean
phenotype and a depletion of serum lipids due to activation of a set of target
genes required for FAO and energy dissipation in brown and white adipose tissues.
In contrast to this, PPARδ knockout mice show reduced energy uncoupling and
are prone to obesity (Wang et al. 2003). These transgenic studies suggest that
PPARδ serves as a key regulator of fat burning.
2.2.6 Peroxisome proliferator-activated receptor γ coactivator (PGC)
PGC-1s can enhance the transcriptional activity of transcription factors like
PPARs by directing protein-protein interaction (Finck & Kelly 2007). Specifically,
PGC-1α is a coactivator of PPARγ and other NRs (Wu et al. 1999) and a regulator
of mitochondrial biogenesis, thermogenesis, gluconeogenesis, and other catabolic
processes (Wu et al. 1999). In particular, PGC-1α is capable of promoting cardiac
mitochondrial biogenesis (Lehman et al. 2000). PGC-1α functions as an adaptor
or scaffold to recruit other coactivators/proteins that remodel chromatin or modify
DNA polymerase II (Finck & Kelly 2007). Recently, the new PGC-1 family
members, PGC-1β and a PGC-related coactivator have also been identified
(Andersson & Scarpulla 2001, Lin et al. 2002) and they are actively expressed in
high-capacity mitochondria tissues such as heart, muscle and brown adipose
tissue (Puigserver et al. 1998, Lin et al. 2002).
2.2.7 RIP140
RIP140 is actively expressed mainly in metabolic tissues such as adipose tissue,
liver, and muscle (Leonardsson et al. 2004). RIP140 is a ligand-dependent corepressor of energy expenditure such as FAO, oxidative phosphorylation,
mitochondrial biogenesis, and glucose uptake (Feige & Auwerx 2007), and
functions as a scaffold protein via interaction with NRs such as ERRα (Powelka
et al. 2006). Furthermore, through interaction with a dehydrogenase CtBp,
21
RIP140 can respond to changes in levels of NADH/NAD+ (Zhang et al. 2002), by
which it senses the cellular redox status. Interestingly, RIP140 itself is heavily
modulated by post-translational modifications such as phosphorylation (Gupta et
al. 2005), methylation (Mostaqul Huq et al. 2006), and acetylation (Vo et al.
2001). RIP140 shares many target genes with PGC-1, but may have opposing
effects (White et al. 2008). RIP140 is a negative regulator of cellular respiration
and mitochondrial biogenesis (Powelka et al. 2006).
2.2.8 Other regulators in lipid metabolism
Many other factors are also regulators of lipid metabolism. For instance,
steroid/thyroid hormones and vitamins A/D can act as ligands of NRs. Other
intracellular regulators are also actively involved in lipid metabolism, such as
SRC-2/3, CBP, Med1, etc. However, these factors will not be addressed here.
2.3
The organelles in mammalian cells
Mammalian cells contain cytoplasm and organelles, separated by either a single
or double membrane.
2.3.1 The single membrane organelles
The majority of organelles within mammalian cells contain a single membrane.
These include the ER, peroxisomes, endosomes, lysosomes, and Golgi complex.
The ER is a network of interconnected membranes. The major lipid fraction
of the ER membrane is formed by glycerophospholipids (van Meer 2005). The
main function of the ER is the synthesis of lipids including FAs, TG, cholesterol,
and phospholipids, membrane proteins, and secreted proteins. For example, long
chain and very long chain FAs are produced by the ER elongation system. In
mammals, the ER is also the place where ceramide is synthesized (Hoetzl et al.
2007). Further, one of the protein quality-control systems also resides in the ER,
thus dysfunction of this system leads to the accumulation of misfolded proteins in
the lumen of the ER, and ultimately contributes to vascular, cardiac and other
diseases (Glembotski 2007).
Peroxisomes are roughly spherical organelles, 0.2–1.0 μm in diameter. One of
their main functions is the β-oxidation of FAs (Schrader & Yoon 2007), yielding
acetyl groups which then are transported into the mitochondria, while the energy
22
released during this process is partially converted into heat. Peroxisomes can also
oxidize (poly)unsaturated enoyl-CoA esters which requires the participation of
several auxiliary enzymes such as ∆3, ∆2-enoyl-CoA isomerase, 2,4-dienoyl-CoA
reductase, and ∆3,5 ∆2,4-dienoyl-CoA isomerase (Hiltunen et al. 1996).
Endosomes contain over 10% sphingolipids and 30–40% cholesterol (van
Meer 2005). Notably, late endosomes contain locally produced unique lipids, such
as lysobisphosphatidic acid (Matsuo et al. 2004).
Lysosomes are the “digestive” machinery within mammalian cells. They
digest different kinds of molecules taken up by cells via endocytosis,
phagocytosis, and autophagy. Dysfunction of enzymes in lysosomes can result in
some 30 different inherited lysosomal storage disorders in humans (von Figura &
Hasilik 1986).
The Golgi complex picks up the protein-loaded vesicles that bud from the ER.
The Golgi complex is responsible for the processing and sorting of membrane
components and secreted proteins. Moreover, the ER and Golgi complex are
organelles which are responsible for glycosylation, sulfation, and phosphorylation
of secretory and membrane-bound proteins, proteoglycans, and lipids (Hirschberg
et al. 1998). For instance, ceramide is converted to sphingomyelin and complex
glycosphingolipids on the inner, non-cytosolic surface of Golgi cisterns (Hoetzl et
al. 2007). The trans Golgi network (TGN) has a lipid composition similar to that
of the endosomes (van Meer 2005).
2.3.2 The double membrane organelles
The nucleus is the largest organelle in mammalian cells, and the composition of
proteins within the outer and inner membrane is different. The two membranes
are fused at nuclear pores. The nucleus functions as a container of genomic DNA,
and is the site of replication of the genetic material, and the synthesis of RNAs
(Lodish H. 2003).
Further organelles within mammalian cells containing double membranes are
mitochondria (see below).
2.3.3 Mitochondria
In many mammalian cells, mitochondria occupy up to 25% of the volume of the
cytosol. Cardiac mitochondria range from 0.1 to 20 μm in size (Fig. 2). Again,
like in the nucleus, the composition of the two membranes differs significantly.
23
The outer mitochondrial membrane consists of approximately 50% proteins
including porins which make it highly permeable, whereas the inner
mitochondrial membrane contains around 80% proteins which confer several
types of activities: they carry out the oxidation reactions of the respiratory chain
as well as ATP synthesis, and function as metabolite transport and proteinimporting. Additionally, the inner membrane is rich in unusual phospholipids, the
cardiolipins. Furthermore, the surface area of the inner membrane is much larger
than that of the outer membrane due to a number of extensively folded cristae
(Lodish H. 2003). In contrast to the outer membrane, there is no porin in the inner
mitochondrial membrane. This inner membrane is highly impermeable to ions
and/or molecules which cross the barrier through transporters. As a consequence,
a membrane potential across the inner membrane is generated. Furthermore,
mitochondria may play a role in the cellular homeostasis of Ca2+, similar to the
role of the ER (Romagnoli et al. 2007). Mitochondria are also the major location
of iron-sulfur clusters biosynthesis (Scheffler 2001).
Fig. 2. Ultrastructure of mouse heart mitochondria visualized by transmission electron
microscopy (TEM), bar represents 5 μm, arrow points to cristae (picture from Dr. Raija
Sormunen).
The mitochondrial matrix contains hundreds of enzymes, ribosomes, tRNA, and
mitochondrial DNA. Many of the enzymes are involved in intermediary and
energy metabolism such as oxidation/synthesis of FAs, the tricarboxylic acid
(TCA) cycle, pyruvate oxidation, urea cycle, ketone body formation, and steroid
metabolism. Human mitochondrial DNA contains 37 genes, including 24 tRNA or
rRNA genes as well as 13 polypeptide-encoding genes (Anderson et al. 1981).
24
Proteomic studies revealed that about half of the mammalian mitochondrial
proteins are ubiquitously expressed, whereas the rest are tissue specific (Mootha
et al. 2003).
2.4
The components of mitochondrial FAS II
Mitochondrial FAS II in mammals consists of multiple components including
several enzymes and cofactors (Fig. 3).
2.4.1 Acyl carrier protein
The acyl carrier protein (ACP) in mammals is a small acidic protein. ACP
transfers acyl groups via a thioester linkage to the sulfhydryl group of
4’-phosphopantetheine (Jordan & Cronan 1997). ACP from mammals was
isolated first as a component of mitochondrial complex I (Runswick et al. 1991),
but later a soluble ACP pool was also described (Cronan et al. 2005). ACP is
needed for assembly of the peripheral arm and the membrane arm of complex I
(Schulte 2001). Two-dimensional NMR revealed that Escherichia coli ACP has a
defined but flexible tertiary structure dominated by three major parallel α-helices
(Flaman et al. 2001). Even though the binding of FAs has little influence on ACP
conformation, it stabilizes ACP against denaturation at alkaline pH (Rock &
Cronan 1979, Rock et al. 1981, Cronan 1982, Jones et al. 1987, Flaman et al.
2001). ACP is highly expressed in heart and skeletal muscle (Triepels et al. 1999).
Unlike the mitochondrial ACP, which functions as a structurally independent
component, an ACP-like domain is a part of cytoplasmic FAS I (Stuible et al.
1998).
2.4.2 Phosphopantetheine:protein transferase (PPTase)
Activation of ACP is mediated by the holo-ACP synthase (ACPS) or
phosphopantetheine:protein transferase (PPTase) (Xu et al. 2001).
4’-Phosphopantetheine is transferred from CoA to the hydroxyl group of a
specific serine on apo-ACP in a Mg2+-dependent reaction (Flugel et al. 2000). The
addition of 4’-phosphopantetheine to ACP does not result in any observable
difference in the folding of the apo and holo forms of the protein (Xu et al. 2001).
The 4’-phosphopantetheine contains dual functions: (i) it activates acyl groups by
25
thioester linkage, and (ii) acts as a flexible arm that allows translocation of
intermediates between catalytic sites of FAS pathways (Stuible et al. 1998).
In bacteria, ACP and the ACP-like domain of multiple-domain polypeptides
can be modified by five different PPTases, namely, the enterobactin synthetase of
E. coli by EntD (Lambalot et al. 1996, Gehring et al. 1997), the surfactin
synthetase of Bacillus subtilis by Sfp (Lambalot et al. 1996), and o195 (Lambalot
et al. 1996), type II FAS of E. coli by ACPS (Lambalot & Walsh 1995), and type I
FAS enzymes of Brevibacterium ammoniagenes by Ppt1p (Stuible et al. 1997).
Notably, the substrate for o195 has not yet been determined.
Three classes of PPTases in yeast, namely, Fas2, Ppt2, and Lys5, are
responsible for FAS I (Schuller et al. 1992), mitochondrial FAS II (Stuible et al.
1998), or lysine biosynthesis (Borell & Bhattacharjee 1988), respectively. The
Ppt2p (YPL148C) has a low molecular mass and is responsible specifically for
phosphopantetheinylation of the mitochondrial apo-ACP. It is still unclear
whether the event takes place in the mitochondria or in the cytosol prior to entry
of the ACP into the mitochondria. Ppt2 contains a relatively weak mitochondrial
target signal and unprocessed Ppt2 is enzymatically active (Stuible et al. 1998). It
would be worthwhile to design and implement experiments that precisely localize
Ppt2p in yeast cells.
In mammals, a single PPTase may be responsible for posttranslational
modification of carrier proteins in three different metabolic pathways: (i) the
cytosolic FAS I, (ii) the mitochondrial FAS II, and (iii) the lysine degradative
pathway (Joshi et al. 2003). Moreover, the human PPTase localizes predominantly
in the cytoplasm, suggesting that posttranslational modification of carrier proteins
takes place in the cytosol, regardless of their ultimate destination within the cell.
Therefore, the mitochondrial-apo-ACP may be phosphopantetheinylated prior to
importation into the mitochondria (Joshi et al. 2003).
2.4.3 Malonyl-CoA:ACP transacylase (malonyltransferase; MCAT)
The yeast gene encoding MCAT has been identified by its homology to the
bacterial enzyme (Schneider et al. 1997). Disruption of MCAT led to a respiratory
deficient phenotype. Again, there has not been report regarding the biochemical
properties of the yeast Mcat1p. However, the kinetic characterization of its human
ortholog was done using a recombinant protein expressed in an insect cell system
(Zhang et al. 2003a).
26
2.4.4 β-Ketoacyl synthase (KAS/CEM1)
A gene CEM1 encoding β-ketoacyl synthase (KAS) (condensing enzyme) was
identified from Saccharomyces cerevisiae in 1993. Inactivation of CEM1 leads to
a respiratory deficient phenotype (Harington et al. 1993). However, the lipid
composition of the CEM1 mutant strain showed no major changes, indicating that
FAS II in yeast may produce only specialized lipids instead of synthesizing the
bulk of the FAs (Harington et al. 1993). Moreover, the biochemical properties of
the yeast Cem1p have not been characterized. Importantly, Zhang et al cloned and
purified the human ortholog of Cem1p and studied its kinetics. Their results
showed that the human CEM/KAS uses acetyl-ACP as a primer, and accepts C2C14 substrates with a maximal catalytic efficiency with C6 and C10 substrates
(Zhang et al. 2005). Surprisingly, mitochondrial KAS from Arabidopsis thaliana
utilizes neither acetyl-CoA nor acetyl-ACP as a primer, but prefers malonyl-ACP.
Interestingly, this A. thaliana mitochondrial CEM1 showed a bimodal substrate
preference with maxima at C-8 and C14-16 (Yasuno et al. 2004).
2.4.5 β-Ketoacyl reductase (KAR)
E. coli β-ketoacyl reductase (FabG) is a tetramer with a monomeric molecular
mass of 25.5 kDa. The N-terminally His-tagged recombinant FabG was purified
and showed KAR activity in an NADPH-dependent manner (Heath & Rock 1995).
The apo and liganded crystal structures of FabG have been solved. The NADPH
binding causes a conformational change, which leads to reorganization of the
active site and promotes binding of the ACP substrate. This property is not
characteristic of most of the other known short-chain dehydrogenase/reductase
(SDR) enzymes (Price et al. 2001), although, like many other SDRs, FabG
contains a conserved triad of Ser138-Tyr151-Lys155 residues in its catalytic site
(Price et al. 2001). Another KAR ortholog, NodG in Rhizobium meliloti is a
component of a FAS II-like pathway that is critical for acylation of a “host-specific”
signaling molecule – oligosaccharide (Hopwood & Sherman 1990), which restricts
the infectivity of the bacterium (Lerouge et al. 1990, Spaink et al. 1991, Slabas et
al. 1992).
S. cerevisiae Oar1 (Oar1p) is homologous to FabG and has been
characterized based on the analysis of the respiratory deficient phenotype of its
mutant strain, whereas the biochemical properties of Oar1p have not yet been
determined (Schneider et al. 1997). However, neither a homology-based cloning
27
nor a large scale yeast genetic complemention screening using human cDNA
libraries was successfully in cloning of the human counterpart of Oar1p, HsKAR.
The leaves of antisense Kar treated plants were curled and slightly smaller at
maturity than controls (O'Hara et al. 2000). The plant KAR can also catalyze
reduction of acetoacetyl-CoA in a NADPH-dependent manner. The crystal
structure of Brassica napus KAR has been determined at 2.3 Å resolution in a
binary complex with an NADP+ cofactor. Each subunit of the homotetramer has a
dinucleotide-binding fold. Like other SDRs, the BnKAR has a classical conserved
triad of Ser154-Tyr167-Lys171 residues in the catalytic site and conformation
changes are induced by substrate binding. The structure is similar to another SDR
enzyme, the bacterial enoyl-ACP reductase (FabI) (Fisher et al. 2000).
2.4.6 3R-Hydroxyacyl-ACP dehydratases (HTD)
The first eukaryotic 3R-hydroxyacyl-ACP dehydratase (HTD) identified using
genetic tools was the yeast mitochondrial HTD. This protein is a component of
the mitochondrial FAS II in yeast (Kastaniotis et al. 2004). In human
mitochondria, a bicistronic gene RPP14 can be transcribed into a single mRNA
but it encodes two proteins, namely, ribonuclease P (RNase P) and 3-hydroxyacyl
thioester hydratase (HTD2) (Autio et al. 2007). The data suggested a link between
mitochondrial FAS and RNA processing in vertebrates. Interestingly,
Mycobacterium tuberculosis contains a gene cluster encompassing HadA, HadB,
and HadC. These proteins can form two types of heterodimers, HadA–HadB and
HadB–HadC, in which HadB apparently is the catalytic subunit (Sacco et al.
2007). The heterodimers (HadA–HadB and HadB–HadC) act as 3R-hydroxyacylACP dehydratases and show distinct chain length specificity in FAS II. The
interaction between proteins was essential for stabilizing HadB and potentially
confers the specificity of the catalysis.
2.4.7 2-Enoyl thioester reductase (MECR/ETR1)
In E. coli, the last step of FAS II is catalyzed by a SDR enzyme, FabI (Heath &
Rock 1995, Bergler et al. 1996, Heath et al. 1999). In yeast, the corresponding
reaction of the mitochondrial FAS II is catalyzed by Etr1p which is a MDR
enzyme (Torkko et al. 2001, Airenne et al. 2003). Interestingly, Candida
tropicalis contains two isoforms of Etr1p, which are encoded by two different
genes but differ by only three amino acids (Torkko et al. 2003). As the last
28
enzyme of FAS II in mammals, MECR/ETR1 was identified on the basis of
similarities to the fungal protein, Etr1p. It is also known as nuclear receptor
binding factor 1, Nrbf1’. MECR/ETR1 was localized on 1p36.1-p35.1. The
murine protein is encoded by Mecr, at CCDS18715 on chromosome 4.
2.4.8 The FAS II components of Caenorhabditis elegans
Y56A3A.19 encodes a mitochondrial ACP in C. elegans. In the larval stage, it is
mainly expressed in the intestine, whereas in the adult worm, it is predominantly
expressed in the pharynx (McKay et al. 2003, Hunt-Newbury et al. 2007). The
RNAi phenotype of Y56A3A.19 includes early larval arrest/lethality (Simmer et
al. 2003, Sonnichsen et al. 2005) and slow growth (Kamath et al. 2003).
F37C12.3 potentially encodes another mitochondrial ACP in C. elegans. The
RNAi phenotype of F37C12.3 includes slow growth (Kamath et al. 2003, Simmer
et al. 2003), embryonic lethality (Maeda et al. 2001, Kamath et al. 2003), larval
arrest/lethality (Maeda et al. 2001, Rual et al. 2004, Sonnichsen et al. 2005), as
well as small (Maeda et al. 2001) and sterile progeny (Maeda et al. 2001).
C50D2.9, F10G8.9, dhs-11/25, and Y48A6B.9 potentially encode malonylCoA:ACP transacylase, β-ketoacyl ACP synthase, β-ketoacyl reductase, and
MECR/ETR1 of mitochondrial FAS, respectively. However, no phenotype has
been observed from high throughput knock-down studies (Fraser et al. 2000,
Gonczy et al. 2000, Kamath et al. 2003, Rual et al. 2004, Sonnichsen et al. 2005).
Interestingly, W09H1.5 potentially encodes an additional mitochondrial
2-enoyl thioester reductase. In the larval stage, it is expressed in the pharynx,
intestine, body wall muscle, nerve ring, head neurons, tail neurons, as well as in
unidentified cells in the tail; in the adult stage, it is detectable in pharynx,
intestine, vulval muscle, body wall muscle, nerve ring; head neurons, tail neurons,
and similarly in unidentified cells in the tail (McKay et al. 2003, Hunt-Newbury
et al. 2007). RNAi of W09H1.5 showed an anti-aging phenotype, namely, an
extended life span (Hamilton et al. 2005).
29
Fig. 3. Mitochondrial FAS spiral in mammals. A question mark means either the
enzymes responsible for the steps have not been identified or the sources/products
are unknown. Notably, C8 is one product of FAS II used for the synthesis of lipoic acid,
however, another possible product(s) of this pathway is unknown. β-Ketoacyl
thioester reductase (KAR) is also missing. The source of mitochondrial malonylACP/CoA is unknown.
2.5
Where is malonyl-CoA generated in mammalian mitochondria?
Analysis of the enzymatic properties of FAS II enzymes suggests that
mitochondrial FAS can synthesize fatty acyl groups in a malonyl-CoA/ACPdependent manner (Zhang et al. 2003a, Yasuno et al. 2004, Zhang et al. 2005).
However, it is not yet known where the malonyl-CoA/ACP in mitochondria is
30
generated, and whether malonyl-CoA/ACP is necessary for de novo
mitochondrial FAS (Fig. 3). In mammals, malonyl-CoA is used for the synthesis
of FAs within the mitochondria, peroxisomes, and cytosol. Malonyl-CoA is not
only an intermediate in the de novo FA biosynthesis or elongation, but also an
important cellular signal through inhibition of carnitine palmitoyltransferase 1
(CPT1), which regulates mitochondrial β-oxidation (Saggerson 2008). Several
enzymes are described below that are putative candidates for synthesizing
malonyl-CoA/ACP within mitochondria in mammals.
2.5.1 Acetyl-CoA carboxylase 2 (ACC2)
Biotin-dependent carboxylases include pyruvate carboxylase, propionyl-CoA
carboxylase (PCC), β-methylcrotonyl-CoA carboxylase and acetyl-CoA
carboxylases (ACCs) (Cole et al. 1994, Schrick & Lingrel 2001). Among them,
ACC1 is the catalytic enzyme responsible for the first reaction of FAS, producing
malonyl-CoA and is tightly regulated by phosphorylation-de-phosphorylation and
cellular metabolites (Kim et al. 1989). Overproduction of ACC activity results in
a significant increase in the rate of FAS in E. coli (Davis et al. 2000), indicating
its important regulatory role in FAS. The carnitine palmitoyltransferases
(CPTs)/malonyl-CoA/ACCs system in mammals is also involved in the sensing of
mitochondrial FAs via CPTs and ACCs, which can be further regulated by insulinstimulated protein kinase (Alam & Saggerson 1998, Heesom et al. 1998).
Mammalian ACC1 is a 265 kDa protein highly expressed in white adipose tissue
and lactating mammary gland (Bianchi et al. 1990, Widmer et al. 1996), whereas
ACC2 in mammals is a 280 kDa protein enriched in heart, skeletal muscle and
brown adipose tissue (Abe et al. 1998). ACC2 knockout mice show a higher FAO
rate and reduced fat content (Abu-Elheiga et al. 2003). ACC2, a mitochondria
associated protein, is 75% identical in sequence to the cytosolic ACC1 except for
the N-terminal region, where ACC2 has an extra ~200 amino acids (Ha et al.
1996). Interestingly, the N-terminus of ACC2 acts as a mitochondrial targeting
signal and is immediately followed by a 100-aa hydrophilic stretch. It was
suggested that the fuction of the leader sequence is solely to anchor ACC2 in the
membrane of mitochondria, with the bulk of the polypeptide facing the cytosol
(Abu-Elheiga et al. 2000). Moreover, it is also possible that ACC2 is associated
with the contact sites of the outer and inner mitochondrial membranes. This
association can be readily reversed, which may be the reason why previous
attempts to establish a subcellular localization of ACC2 have been difficult, as the
31
isolation procedure should not include a harsh treatment. Notably, native ACC2
can be purified using avidin-sepharose chromatography from a heart nonmitochondria-homogenate (after mitochondria were removed from the
homogenate) (Thampy 1989). This evidence also indicates that ACC2 resides
primarily in the in cytosol rather than in the mitochondrial matrix or membrane.
This localization argues against the role of ACC2 as a generator of
intramitochondrial malonyl-CoA.
2.5.2 Propionyl-CoA carboxylase (PCC)
PCCs from Myxococcus xanthus, plant and Yarrowia lipolytica can also accept the
acetyl-CoA substrate with a Km of 0.25 mM for M. xanthus, 0.11–0.26 mM for
plant and 0.26 mM for Y. lipolytica, respectively (Mishina et al. 1976, Nikolau &
Hawke 1984, Kimura et al. 1998). Notably, MxPCC has a much higher affinity
toward propionyl-CoA (Km = 0.032 mM) (Kimura et al. 1998). The relative
carboxylation rates of MxPCC were 100% for propionyl-CoA and 14.3% for
acetyl-CoA (Kimura et al. 1998). In mammals, PCC is abundant in the heart and
also accepts acetyl-CoA as a substrate in addition to propionyl-CoA (Scholte et al.
1986).
In certain species, a single enzyme with dual-substrate specificity catalyzes
the carboxylation of both acetyl- and propionyl-CoA. The dcm-1 mutant cells of
M. xanthus with a deficiency in propionyl-CoA carboxylase activity had
decreased levels of long-chain FAs (C16 to C18) compared to wild-type cells
(Kimura et al. 1997). PCCs from Mycobacterium smegmatis, Streptomyces
erythreus, and Turbatrix aceti also catalyze the carboxylation of acetyl-CoA to
yield malonyl-CoA (Meyer & Meyer 1978, Henrikson & Allen 1979, Hunaiti &
Kolattukudy 1982).
2.5.3 Putative malonyl-CoA synthetase
Even though the possible source of malonyl-CoA in mammalian mitochondria
might be carboxylation of acetyl-CoA by ACCs or PCC, there exists another
mechanism that might produce malonyl-CoA or malonyl-ACP in mammalian
mitochondria.
In Rhizobium leguminosarum, a mat operon consisting of three consecutive
genes (matABC) has been described. The mat encodes enzymes involved in the
uptake and conversion of malonate to acetyl-CoA through malonyl-CoA (An et al.
32
2002). The matB encodes malonyl-CoA synthetase. A human protein
(LOC197322) related to MatB in amino acid sequence (identity 31%), containing
a putative mitochondrial signal, can be found from databases. The human
homolog might function as a malonyl-CoA synthetase like MatB in R.
leguminosarum.
The matA encodes a malonyl-CoA decarboxylase that is homologous to the
human malonyl-CoA decarboxylase (MCD) (identity 30%). Although some
studies suggested that human MCD is mainly localized in peroxisomes and
cytoplasm (Sacksteder KA, 1999), the rat MCD apparently contains a
mitochondrial targeting sequence in the N-terminus and a peroxisomal (Ser-LysLeu) targeting signal in the C-terminus (Voilley et al. 1999). This is consistent
with the notion that the rat MCD is mainly localized in the mitochondrial matrix
but a small fraction of the activity is extra-mitochondrial (Hamilton & Saggerson
2000, Kerner & Hoppel 2002). Moreover, further studies suggested that human
MCD is localized in both mitochondria and peroxisomes (FitzPatrick et al. 1999).
Recent studies showed that the rat MCD is present in three subcellular
compartments: cytosol, mitochondria and peroxisomes (Joly et al. 2005). The
activities of MCD as well as ACCs are important determinants of the cytosolic
concentration of malonyl-CoA. The cytosolic localization of the MCD suggests
that MCD by its effect on the concentration of malonyl-CoA can function as a
regulator of fat oxidation and lipid partitioning.
MatC encodes a putative dicarboxylate carrier protein in R. leguminosarum,
however, no homologous protein can be found in the human genome of either
human or that of any other organism so far, except for Xanthomonas axonopodis,
Xanthomonas campestris, Streptomyces coelicolor, and Streptomyces
hygroscopicus.
The next question concerns the origin of malonate in mitochondria. One
study on the origin of free malonate in the brain suggests that it could be the result
of the following sequential reactions: acetyl-CoA → malonyl-CoA → malonate.
The first reaction could be catalyzed by the cytosolic ACC, and the final step
might occur by transfer of the CoA-group from malonyl-CoA to succinate and/or
acetoacetate (Riley et al. 1991), although the enzyme catalyzing this reaction is
still unknown (Succinyl-CoA:acetoacetate-CoA transferase is supposed to be a
mitochondrial enzyme). Malonate can enter the mitochondrial matrix via the
dicarboxylate transport protein (DTP). DTP is located in the inner mitochondrial
membrane and is capable of taking up malonate, malate and succinate. The
33
mitochondrial DTPs from yeast, plants and animals show high similarity (Kaplan
& Pedersen 1985, Vivekananda et al. 1988, Kakhniashvili et al. 1997).
In addition, Wada and Baldet suggested that pea mitochondria do not possess
ACC (Wada et al. 1997). Instead, they take up and utilize malonate but not acetate
or malonyl-CoA from outside the mitochondria (Wada et al. 1997). This indicates
that mitochondria may contain a different system to produce malonyl-CoA
compared to FAS in the cytosol and chloroplasts that uses ACC to generate
malonyl-CoA (Sasaki et al. 1997).
Recently, Stephens and coauthors described a FAS in vitro reconstitution
system by incubating isolated mitochondria from Trypanosoma brucei with
radiolabeled FAs and [14C]malonyl-CoA or [14C]malonic acid (Stephens et al.
2007). The data suggested cytosolic malonyl-CoA instead of malonate is the
source of mitochondrial FAS in T. brucei, in contrast to the plant mitochondrial
FAS system (Gueguen et al. 2000).
2.5.4 Putative malonyl-CoA:ACP transacylase
The initiating steps of FAS II in Plasmodium falciparum are catalyzed by pfACP,
pfMCAT, and pfKASIII (Prigge et al. 2003). MCAT, also called FabD, produces
malonyl-ACP from malonyl-CoA and ACP. β-Ketoacyl-ACP synthase III (KASIII,
also called FabH) catalyzes the condensation of malonyl-ACP and acyl-CoA
(typically acetyl-CoA) forming a 3-ketoacyl-ACP product (Prigge et al. 2003). In
2002, we picked up a human MCAT candidate (CAB94798) with an identity of
31% via database searching. This human protein is targeted to mitochondria and
contains a GXSXG and some other consensus sequences common to various acyl
hydrolases. In 2003, Zhang and coworkers showed that the human recombinant
protein indeed contains malonyltransferase activity in vitro (Zhang et al. 2003a)
2.6
Structural aspects of FAS II components
2.6.1 Short chain dehydrogenase/reductase (SDR)
SDR enzymes were originally found in lower life forms, and subsequent studies
indicated that a number of human proteins also belong to the SDR family. So far
around 3000 members of this group have been defined including 63 human
34
proteins (Jornvall et al. 2003). The “short” refers to the length of the polypeptide
chain of SDRs rather than the substrates.
SDRs catalyze NAD(P)(H)-dependent oxidation/reductions. Although they
are divergent in terms of substrate specificities and sequence identities, SDRs
possess highly similar α/β folding patterns with a central β-sheet, typical of the
Rossmann-fold (Oppermann et al. 2003). SDRs can be further divided into two
families, namely, “classical” (~250 residues) and “extended” (~350 residues)
(Jornvall et al. 1995). SDRs contain a common GXXXGXG pattern in the
N-terminal region, which is characteristic of NAD(P)(H) dependent enzymes,
while the C-terminus constitutes the substrate binding site (Kallberg et al. 2002).
Recent studies proposed a catalytic tetrad of Asn, Ser, Tyr, Lys residues, probably
forming a proton relay system (Oppermann et al. 2003). Two examples of SDRs
are described below.
3S-Hydroxyacyl-CoA dehydrogenases (HAD) or 17β-hydroxysteroid
dehydrogenase type 10 (17βHSD10)
3S-Hydroxyacyl-CoA dehydrogenase type I (HAD) of the FAO system in
mammals consists of two identical monomers with a molecular mass of ~35 kDa.
Another mitochondrial enzyme that contains similar activity is the
3S-hydroxyacyl-CoA dehydrogenase type II (HADII), which is a homotetramer
with a molecular mass of ~27 kDa for each subunit (Kobayashi et al. 1996). Both
proteins show broad tissue distribution. The HADII gene is localized in
chromosome Xp11.2, where a multigene domain escapes X inactivation (He et al.
1998, Miller & Willard 1998). Unlike type I and most other FAO matrix enzymes,
HADII does not contain a cleavable mitochondrial signal (Furuta et al. 1997).
This is similar to another mitochondrial enzyme, 3-ketoacyl-CoA thiolase
(Arakawa et al. 1990).
As the first mitochondrial enzyme found to catalyze the oxidoreduction of
steroid hormones, HADII was also called 17β-hydroxysteroid dehydrogenase type
10 (17βHSD10). Nevertheless, the HADII/17βHSD10 is different from other
17βHSDs, in that it is unable to catalyze the reduction of either estrone or
androstenedione by NAD(P)H (He et al. 1999). HADII shows broad substrate
specificity, whereas its enzymatic activities are solely based on NAD(H), which
can not be replaced by NADP(H). The ratio of the kcat of alcohol dehydrogenase,
17βHSD and 3S-HAD was 1:0.3:1028 (He et al. 2000b), suggesting it has a major
role in FAO. It is noteworthy that 3α-adiol is the best steroid substrate for HADII,
35
which means HADII preferably acts in oxidization rather than reduction,
regulating 5α-DHT levels in tissues (He et al. 2000a). However, there is no clear
sequence similarity between HADII and other cytosolic NADP(H) dependent
3α-HADs.
Interestingly, inactivation of scully, the Drosophila counterpart of HADII,
leads to the presence of cytoplasmic lipid inclusions and scarce mitochondria in
spermatocytes. Furthermore, photoreceptors of the scully mutant fly contain
aberrant mitochondria and accumulations of membranous material (Torroja et al.
1998). Also significant is the fact that the amino acid sequence of HADII is
identical to the endoplasmic reticulum amyloid β-peptide-binding protein
(ERAB), which is associated with Alzheimer’s disease (He et al. 1998). A recent
study shows that the reduced expression of HADII in human caused by a C→A
mutation, leads to mental retardation, choreoathetosis, and abnormal behavior
(Lenski et al. 2007).
17β-hydroxysteroid dehydrogenase type 4 (17βHSD4) or peroxisomal
multifunctional enzyme type 2 (MFE-2)
The peroxisomal multifunctional enzyme type 2 (MFE-2; EC1.1.1.62) catalyzes
the second and third reactions of the peroxisomal spiral of FAO (Hiltunen et al.
1992). MFE-2 contains 3R-hydroxyacyl-CoA dehydrogenase activity as well as
2E-enoyl-CoA hydratase-2 activities.
Mammalian MFE-2 shows broad substrate specificity. It is also called
17β-hydroxysteroid dehydrogenase type 4 (17βHSD4), oxidizing 17β-estradiol,
17β-diol, and 5-androstene-3β (Leenders et al. 1994, Adamski et al. 1995).
Moreover, MFE-2 also oxidizes 24 hydroxylated intermediates in bile acid
synthesis, and the 3-hydroxyl on 2-methyl branched chain fatty acyl-CoAs
(Dieuaide-Noubhani et al. 1996, Novikov et al. 1997).
2.6.2 Medium-chain dehydrogenase/reductase (MDR)
The MDR family has approximately 1000 members including 23 human proteins
(Jornvall et al. 2003). The biological roles of the MDRs include participating in
metabolic pathways, the defense system against toxic compounds, and regulatory
functions (Jornvall et al. 2003).
Like SDRs, MDRs also utilize NAD(P)(H), and some members have one or
two zinc ions either at the catalytic or at another site-stabilizing the structure
36
(Nordling et al. 2002). There are two types of MDRs, with/without Zn2+ at the
active site (Jornvall et al. 2003). MDRs can be further divided into eight families.
The zinc-containing MDRs are as follows: alcohol dehydrogenase (ADH),
cinnamyl alcohol dehydrogenase (CAD), polyol dehydrogenases (PDH), and
yeast alcohol dehydrogenase (YADH). The non-zinc-containing MDRs are the
following: acyl-CoA reductase (ACR), leukotriene B4 dehydrogenase (LTD),
mitochondrial response proteins (MRF), and quinone oxidoreductases (QOR)
(Nordling et al. 2002). Yeast Etr1p and human MECR/ETR1 belong to the
mitochondrial respiratory function (MRF) family. MDR enzymes are more
closely addressed in the attached papers and manuscript.
2.6.3 Interactions of FAS II enzymes and ACP
FAS II enzymes in bacteria do not contain any sequence similarity that defines a
common ACP binding site, but rather an electropositive/hydrophobic surface to
interact with helix α2 of ACP. Notably, this special patch is a ubiquitous feature of
bacterial FAS II enzymes (Zhang et al. 2003b). In the structure of FabG (the
3-ketoacyl-ACP reductase of FAS II), two conserved residues Arg129 and Arg172,
which are located in the hydrophobic patch adjacent to the catalytic site play a
role in the interaction with α2 of ACP (Zhang et al. 2003b).
2.6.4 Cofactor specificity
Most NAD(P) dependent dehydrogenases contain a Rossmann fold for binding of
NAD(P)H (Smiley et al. 1971).
17βHSD1 is a SDR member preferring NADP(H) over NAD(H).
Mutagenesis replacements showed that Ser12 and Leu36 are important for
selection of the cofactor (Huang et al. 2001). Replacement of Leu36 with aspartic
acid altered the preference to NAD(H).
Some SDRs show dual cofactor acceptance and therefore may use different
coenzymes in different tissues/organelles (Kallberg et al. 2002). Moreover,
bovine retinol dehydrogenase (RDH) prefers NAD(H), whereas RDH from rat is
NADP(H) dependent (Simon et al. 1995, Peralba et al. 1999). Modeling
experiments suggested that a polar residue Thr61 in the bovine protein and a
corresponding basic residue Lys64 in the rat enzyme may be responsible for the
different preferences (Tsigelny & Baker 1996).
37
The numbers of NAD(H) dependent and NADP(H) dependent SDR members
are similar in plant, mouse, and human; however, in yeast, worm and fly, the
majority of SDRs is NADP(H) dependent (Kallberg et al. 2002).
2.6.5 Substrate binding pocket
Many enzymes contain a substrate binding pocket, which can be classified
according to its shape, such as crevice-like, funnel-like and tunnel-like (Yang et al.
2002). The shape, size, and hydrophobicity of the pocket may determine the
substrate preference (e.g., chain length) of enzymes. Mutagenesis can be applied
to alter the chain length specificities of the enzymes and may result in engineered
enzymes with new features (Cahoon et al. 1997, Klein et al. 1997, Whittle &
Shanklin 2001, Schmitt et al. 2002, Stinnett et al. 2007).
2.7
Myocardial metabolism and cardiomyopathies
Cardiomyopathy is the deterioration of the function of the myocardium, leading to a
high risk of arrhythmia and/or sudden cardiac death. Cardiomyopathies can be
categorized into two groups: extrinsic and intrinsic cardiomyopathies (Richardson et
al. 1996). The most common cause of extrinsic cardiomyopathy is ischemia,
which is a weakness in the heart muscle due to inadequate oxygen delivery to the
myocardium. Moreover, many systemic diseases (e.g., diabetes) can lead to
cardiomyopathy. Intrinsic cardiomyopathies have mainly two forms. In dilated
cardiomyopathy, the heart (especially the left ventricle) is enlarged and the
systolic function is reduced. In hypertrophic cardiomyopathies, the left ventricle
wall thickens, which results in reduced blood flow and in heart dysfunction.
Cardiomyopathies arise from different genetic and environmental conditions,
and are an important contributor to heart failure. Mutations in a number of
metabolic enzymes and contractile proteins can generate cardiomyopathies. In the
case of dilated cardiomyopathy, the genetic causes are still poorly understood.
2.7.1 Mitochondrial energy metabolism and heart failure
Mitochondria oxidize FAs and carbohydrates and generate the reducing
equivalents, NADH and reduced flavin adenine dinucleotide (FADH2). These
reducing equivalents transfer electrons into the respiratory chain, linked to the
generation of ATP. The cardiac energy metabolism is elaborately coupled to ATP
38
production/hydrolysis, and any imbalance of this coupling could results in cardiac
diseases.
The ATP requirement of mouse heart is the highest among mouse organs, and
ATP insufficiency is frequently associated with impairment of contractile function
(Nascimben et al. 1996). For instance, studies on human patients and genemodified mice suggested causal links between derangements in mitochondrial
energy metabolism and cardiac dysfunction (Finck & Kelly 2007). Thus,
improving the efficiency of ATP production could have therapeutic benefit (Taha
2007).
2.7.2 Energy metabolism in ischemic cardiomyopathy
Ischemic cardiomyopathy is associated with insufficiency of oxygen delivery to
meet the energy demands of the heart during physical or psychological stress
(Taha 2007). The outcome of this shortage of oxygen supply is an inhibition of
respiratory function, forcing the heart to rely more heavily on anaerobic
glycolysis (Stanley et al. 1997). Consequently, glucose uptake is stimulated and
the ratios of mitochondrial acetyl-CoA:CoA and NADH:NAD+ are raised,
inhibiting the pyruvate dehydrogenase complex (PDC). Therefore, pyruvate stays
in the cytoplasm and is converted to lactate, leading to intracellular acidosis (Liu
et al. 1996). A consequence of the intracellular acidosis is that the Na+/H+
exchanger, and the Na+-HCO3− transporter are activated, resulting in a rise in
intracellular Na+ level. This leads to activation of the Na+/Ca2+ exchanger, leading
to a rise in the intracellular [Ca2+], which may be detrimental to the cell. This
finally is translated into myocardial ATP being used for the maintenance of
intracellular Ca2+ homeostasis rather than for contraction (Taha 2007).
Modulation of the balance between FAs and glucose metabolism has been a target
of intensive research concerning the treatment of ischemic cardiomyopathy.
2.7.3 Energy metabolism in dilated cardiomyopathy and
hypertrophic cardiomyopathy
Dilated cardiomyopathy means that the heart becomes enlarged and weakened
and the heart work output is decreased, consequently triggering a cascade of
compensatory physiological responses. Patients show reduced respiratory
function, higher levels of cardiac glucose uptake and a decrease in FA uptake.
39
In hypertrophic cardiomyopathy patients, mitochondrial respiratory function
is reduced. This mimics the metabolic profile of the fetal heart, resulting in
intracellular acidosis similar to ischemic cardiomyopathy. 5’-Adenosine
monophosphate-activated protein kinase (AMPK) detects changes in the cellular
energy status through monitoring the rise and fall in ratios of ATP:AMP, and
CrP:Cr. AMPK can be activated by AMP or via phosphorylation by other kinases.
The activated AMPK then activates ATP-generating processes and inhibits energy
consuming processes (Hardie & Carling 1997). Mutation of AMPK may lead to
the Wolff-Parkinson-White (WPW) syndrome (Blair et al. 2001).
2.7.4 Energy metabolism in diabetic cardiomyopathy and other
cardiomyopathies
A possible cause of diabetic cardiomyopathy is that insulin resistance leads to
impaired glucose uptake and enhanced FAs uptake, making it the substrate of
choice (Taha 2007). The increased level of FAs in myocytes results in activation
of PPARα, facilitating FAO and inhibiting glucose oxidation. This shift in the
“substrate preference” causes the oxidative capacity of the heart to be exceeded,
potentially increasing the production of reactive oxygen species, and/or causing
lipotoxicity.
Restrictive cardiomyopathy is characterized by “stiffening” of the
myocardium, which leads to impaired diastolic function (Richardson et al. 1996).
The genetic causes of this disease have yet to be clarified. Arrhythmogenic right
ventricular dysplasia is other type of cardiomyopathy caused by genetic mutations
(dominant/recessive), leading to apoptotic loss of cardiac myocytes which are
replaced by fatty or fibro-fatty tissue in the right ventricle (Corrado et al. 2000).
40
3
Outlines of the present study
The purpose of this study was to identify and characterize a mammalian enoyl
thioester reductase, MECR/ETR1.
More specifically the aims were:
1.
2.
3.
Characterization of the biochemical properties of MECR/ETR1, with focus
on its existence and localization in mammalian tissues and organelles, and on
its enzymatic activity.
Investigation of the crystal structure of MECR/ETR1 and to provide insight
into its substrate binding and catalytic mechanism.
To shed a light on the physiological function of MECR/ETR1, through
generation of Mecr overexpressing mice, and characterization of their
phenotype.
41
42
4
Materials and methods
The materials and methods have been described in more detail in the original
articles.
4.1
Northern blot analysis (I)
A human Multiple Tissue Northern BlotTM from various human tissues was
hybridized with 32P-labeled MECR cDNA. Human β-actin cDNA was used as a
control sample. Hybridized fragments were visualized by autoradiography at
−70 °C for 24 h using X-OmatTM AR imaging film.
4.2
Cloning and mutagenesis (I, II, III)
cDNAs were amplified from isolated RNA with the RobustRT-PCR kit, and the
PCR products ligated into the Acceptor Vector. The DNA fragments were further
subcloned into pBluescript SK(+) (pSK), pET3a, pET23, or pYE352 vectors. For
making fusion constructs, the DNA fragment was digested and subcloned inframe with the 5' end of GFP/His tag cDNAs. The pET23::MECR construct was
used as a template for generating mutant variants using a site-directed
mutagenesis kit.
4.3
Protein expression and purification (I, II)
The expression plasmids were transformed into BL21 (DE3) pLysS E. coli cells.
After induction, the cells were washed with 1 × phosphate buffered saline (PBS)
and stored at −20 ºC until used. Cells were resuspended, sonicated in breaking
buffer and the disrupted cell suspension centrifuged. For non-tagged protein, the
supernatant was applied to a Q-Sepharose column, followed by 2',5'-ADPSepharose 4B, Resource S and Superdex 200. For His-tagged proteins, the
supernatant was loaded onto a Ni-NTA column. The column was washed first
with 1x binding buffer and then with washing buffer. Bound proteins were eluted
with a linear imidazole gradient. Fractions containing MECR/ETR1 or its mutant
forms were pooled, concentrated and applied to Superdex 200. The peak fractions
were combined, concentrated and stored at −70 ºC until used. For isolation of wt
proteins from bovine tissues, the soluble extract of purified mitochondria was
prepared using sonication, followed by ultracentrifugation. The supernatant was
43
dialyzed and applied to an ADP-Sepharose column. The fractions containing
positive signals were pooled, dialyzed, and subjected to immunoisolation. A
suspension of protein A-coated Dynabeads was washed and incubated with antiHsMECRAb. The antibody-loaded beads were washed and transferred to the
dialyzed sample containing BtMECR. The beads were then separated, washed,
and transferred to SDS sample buffer. The beads were removed after incubation,
and the sample was acetylamidated. After electrophoresis, bands were excised,
destained, and dried. The gel pieces were rehydrated with trypsin solution and
incubated overnight. The digested peptides were recovered by extraction. Extracts
were pooled, dried, and dissolved, and subjected to matrix-assisted laser
desorption ionization-time of flight-mass spectroscopy (MALDI-TOF MS).
4.4
Enzymatic assay and determination of kinetic parameters for
MECR/ETR1 and its mutated forms (I, II)
The kinetic constants were determined by monitoring the decrease of absorbance
of NADPH at 340 nm minus 385 nm. The substrates from 2E-C4-CoA to 2E-C16CoA were used. In particular, 2E-octenoyl-CoA was chosen to determine the
kinetic values of mutated variants. The kinetic data were transferred to
Lineweaver-Burk plots using GraFit computer software.
4.5
Reaction product analysis (I)
MECR/ETR1 was incubated in the presence of 2E-hexenoyl-CoA and NADPH.
Aliquots were acidified, and samples from the aliquots were subjected to reversed
phase chromatography. The column was eluted with an isocratic flow of 0.2
ml/min with buffers A (25 mM CH3COONH4, pH 5.5) and B (80% CH3CN and
25 mM CH3COONH4, pH 5.5) at an 83:17 ratio. Metabolites in the eluted peaks
were identified by MALDI-TOF MS.
4.6
Fluorescence microscopy (I)
Attached HeLa cells were grown on glass coverslips in Dulbecco's modified
Eagle's media. Transfection of the HeLa cells was done using the FuGENE 6
reagent and the transfected cells were treated with a mitochondria-selective
fluorescent dye and fixed with paraformaldehyde and permeabilized. Nuclear
44
DNA was counterstained with 1 µg/ml 4',6-diamidino-2'-phenylindole
dihydrochloride (DAPI), followed by analysis with a fluorescence microscope.
4.7
TEM analysis of ybr026c∆ cells expressing MECR (I)
S. cerevisiae ybr026c∆ cells overexpressing MECR or a truncated form of MECR
were transferred from overnight cultures in synthetic medium and grown for 17 h.
BJ1991 wild-type cells and ybr026c∆ cells were grown overnight and then
transferred to oleic acid medium. The cells were fixed with 2.5% glutaraldehyde
and immersed in 2% agar. Yeast cells in agar blocks were further post-fixed,
dehydrated in acetone, and embedded in Embed 812. Thin sections of 80 nm were
cut and examined using TEM.
4.8
Crystallization of MECR/ETR1 (II)
The solution for crystallization contained NADP+ and CdCl2. Crystals were
obtained at 295 K in three to four weeks by the hanging drop vapor diffusion
method in 2 M ammonium sulfate and 100 mM Tris, pH 8.5.
4.9
Data collection, structure determination and refinement (II)
X-ray diffraction data was collected in EMBL/DESY, Hamburg, Germany. The
measurements were made on a single crystal in a nitrogen stream. The data were
processed using the iMOSFLM, SCALA, CHAINSAW, DM of the CCP4 program
package and PHASER. Initial phases were determined from molecular
replacement solution obtained with the program PHASER using CtEtr1p as a
model. The resulting model was subjected to simulated annealing with CNS
followed by the refinement cycles with REFMAC. The quality of the maps
improved considerably when the BUSTER-TNT software system was
implemented into refinement steps with NCS restraints; this allowed further
construction of some of the problematic loops with fragmented electron density.
The final structure was analyzed with PROCHECK and WHATIF.
45
4.10 Modeling of NADPH and fatty acyl thioester in the active site of
MECR/ETR1 (II)
NADPH was modeled in the active site of MECR/ETR1. The complex was
energy minimized using the steepest descent followed by conjugate gradient
minimization procedures. The ligand was docked. The complexes were then
subjected to 2 ns of molecular dynamics simulations to equilibrate the complexes.
The MD simulations were carried out with the Gromacs 3.2.1 package using a
ffG43a1 force field. The topology of the ligand was obtained from the PRODRG
server. The LINCS algorithm was employed to constrain bonds. After energy
minimizations, a short (10 ps) position restraining run for further equilibation and
subsequently a full MD run were carried out.
4.11 Animal care (III)
The animal experiments were carried out in the barrier and in the animal facility
of the Department of Biochemistry, University of Oulu. All the experiments were
performed in accordance with the Animal Care and Use Committee of Oulu
University.
4.12 Generation of metallothionein-I (MT-I)-Mecr overexpressing
mice (III)
A cDNA of mouse Mecr with the poly A signal of Mecr was fused to a vector
containing the mouse MT-1 promoter followed by a fragment of the untranslated
region of the human ornithine decarboxylase (ODC) gene. The construct was
linearized and microinjected into the pronuclei of fertilized eggs that were then
transferred to C57Bl/6J recipient mothers. Transgenic founders and their progeny
were identified by genomic PCR and/or southern blot.
4.13 Southern blot genotyping (III)
Mouse genomic DNA was digested by BamHI. The probe (1 kb) was made by
digestion of the construct with PvuII. The intensity of the 6kb band is
proportional to the copy number of the transgene.
46
4.14 Reverse template PCR and real time PCR (III)
The cDNAs were produced with a First Strand cDNA Synthesis Kit using total
RNA isolated from tissues. Semiquantitative real time PCR was performed,
GAPDH was used as an internal control for normalizing the gene expression.
4.15 Histological staining (III)
The heart tissue samples were fixed and embedded in paraffin, and sections were
stained with hematoxylin-eosin/TUNEL. The sections were examined using light
microscopy. For TEM, tissue samples from mouse heart were fixed, in 1%
glutaraldehyde and 4% formaldehyde in 0.1 M phosphate buffer, pH 7.2,
dehydrated in acetone and embedded Embed 812. Thin sections of 80 nm were
cut and examined by TEM.
4.16 Echocardiographic analysis (III)
The dimensions of the cardiac chambers and left ventricular function were
assessed using transthoracic echocardiography. A short-axis view of the left
ventricle at the level of the papillary muscles was obtained by using 2D imaging
for the M-mode recording. Left ventricular end-systolic (LVESD) and enddiastolic (LVEDD) dimensions as well as the thickness of the interventricular
septum (IVS) and posterior wall (PW) were measured from the M-mode tracings.
LV fractional shortening (LVFS) and ejection fraction (EF) were estimated from
the M-mode LV dimensions by applying the following formulas:
LVFS (%) = [(LVEDD − LVESD)/LVEDD] × 100
and
EF (%) = [(LVEDD3 − LVESD3)/LVEDD3] × 100.
4.17 Heart perfusion (III)
Perfusion of isolated hearts was performed retrogradely with a Krebs-Henseleit
perfusion medium followed by the Langendorff procedure (Chain et al. 1969).
Left ventricular pressure was monitored through the ventricular wall. The
pressure wave signal was fed to a Lab-PC+ data acquisition card and heart
function parameters (heart rate, peak systolic pressure, diastolic pressure and peak
47
pressure development) were calculated on-line with custom designed software
and stored on a computer at 4-s intervals. Coronary flow was measured during the
experiments by using a drop counter with an analog frequency output. Oxygen
consumption was calculated from the arteriovenous concentration difference
multiplied by coronary flow.
4.18 Metabolite assays (III)
The heart was quick-frozen with aluminum clamps precooled in liquid nitrogen,
and stored in liquid N2 until processed. The frozen heart tissue was immersed in
8% (wt/vol) HClO4 in 40% (vol/vol) ethanol precooled to −20 °C, and
homogenized immediately with an Ultra-Turrax homogenizer. After
centrifugation, the supernatant was neutralized with 3.75 mol/L K2CO3 containing
5 mol/L triethanolamine hydrochloride. Creatine phosphate (CrP), creatine (Cr),
ATP, ADP, AMP and inorganic phosphate levels were measured in the supernatant
by enzymatic assays.
4.19 Exercise performance test (III)
Mice were familiarized with running on a motorized running belt two days prior
to the test. The treadmill was started at a low speed with low slope. The speed and
inclination were raised gradually. Exercise was stopped when the mice were
exhausted. Performance was assessed based on three parameters: the duration of
the run, the distance run, and vertical work performance.
4.20 Isolation of heart mitochondria (I, III)
Crude mitochondria were isolated from heart and were purified further by
isopycnic density gradient centrifugation on a Percoll gradient. The gradient was
unloaded from the bottom of the tube, and marker enzyme activities for
mitochondria, microsomes, and cytosol were measured.
4.21 Statistical analysis (III)
Values are expressed as mean ± standard deviation (SD) and analyzed by the
Student’s t test. The homodynamic variables were analyzed with 2-way repeatedmeasures ANOVA. Echocardiographic measurements were analyzed using
48
normalized values with 1-way ANOVA followed by the Student-Newman-Keul
post hoc test. A value of P < 0.05 was considered statistically significant.
49
50
5
Results
5.1
Biochemical properties of MECR/ETR1
5.1.1 Yeast etr1 mutant complementation
Yeast etr1∆ strains display a petite phenotype and can not grow on nonfermentable carbon sources such as glycerol. Expression of full length MECR can
rescue the phenotype, whereas expression of the truncated form, without the
mitochondrial targeting signal, can not restore the respiratory function of the
etr1∆ strains (Fig. 7 in I). Further, TEM shows that etr1∆ cells contain only
rudimentary mitochondria, whereas MECR overexpressing cells contain enlarged
mitochondria compared to the mitochondria from wild-type cells (Fig. 8 in I).
5.1.2 Protein purification
The recombinant truncated forms of MECR/ETR1 and its bovine counterpart
were purified to apparent homogeneity using a bacterial overexpression system
and multiple types of columns (Fig. 3 in I). Further, truncated forms of
MECR/ETR1 and their mutant variants with C-terminal histidine tags were also
purified to apparent homogeneity using Ni-NTA and gel filtration columns. In
addition, the wild-type protein was also isolated from bovine heart mitochondria
using a 2’,5’-ADP-Sepharose affinity column and immunoprecipitation (Fig. 6 in
I), expanding the list of mitochondrial FAS components found in mammals, even
though they are apparently not abundant.
5.1.3 End product analysis
MECR/ETR1 was incubated with 2E-hexenoyl-CoA and NADPH. At various
time points, samples were taken from the reaction for HPLC analysis. The
disappearance of 2E-hexenoyl-CoA was accompanied by the oxidation of
NADPH with the appearance of a metabolite. MALDI-TOF/MS analysis showed
that the accumulating metabolite (865.7 m/z) was 2 mass units larger than
2E−hexenoyl-CoA (863.7 m/z), consistent with the prediction that the product is
hexanoyl-CoA (Fig. 4 in I). The result indicates that MECR/ETR1 is able to
reduce 2E-enoyl-CoA to its saturated counterpart.
51
5.1.4 Subcellular localization
Previously it was suggested that mammalian MECR/ETR1 is a nuclear localized
protein (Masuda et al. 1998). To reinvestigate the subcellular localization of the
protein, the full length cDNA of MECR/ETR1 carrying a cDNA encoding EGFP
at the 3’end was expressed in HeLa cells. The mitochondria were stained with
MitoTracker Red, and nuclear DNA was stained with DAPI. A punctuated pattern
of green fluorescence was superimposed on the MitoTracker staining (Fig. 5A and
D in I), indicating that the chimeric MECR/ETR1-EGFP was targeted into
mitochondria. Notably, the cells expressing EGFP alone showed a diffuse uniform
fluorescence pattern, which was not superimposed on the MitoTracker staining
(Fig. 5E and H in I).
5.1.5 Tissue distribution
The expression levels of MECR/ETR1 in different human tissues were analyzed
by Northern blot. The signal of MECR/ETR1 (1.4 kb) was observed in multiple
samples, namely, heart, brain, placenta, liver, skeletal muscle, kidney and
pancreas. The mRNA of β-actin was used as a control. Importantly, the highest
expression levels of MECR/ETR1 were observed in the high energy demanding
tissues such as heart and skeletal muscle, suggesting a role for MECR/ETR1 in
energy metabolism in these tissues (Fig. 2A in I).
5.2
Structural enzymology of MECR/ETR1
5.2.1 The overall structure
The structure of MECR/ETR1 was refined at 2.41 Å resolution. Like most other
MDR enzymes, the apo structure of MECR/ETR1 forms a dimer. Each monomer
is composed of two domains, namely, the N-terminal (catalytic, Arg42-Val166)
and the C-terminal (cofactor binding, Asn167-Trp311) domains, with the
exception of the last 61 residues (after W311) which fold back to the catalytic
domain (Fig. 3 in II). The two domains are separated by a deep cleft. The cofactor
binding domain contains βαβαβ motifs and is arranged into a Rossmann fold that
is a classical nucleotide binding motif. Two sulfates from the crystallization buffer
bound to the groove of the NADPH binding site, making the structure adopt a
ligand-bound form (a closed conformation) (Fig. 4 in II). The
52
191
AlaSerAsnSerGlyValGly197 motif interacts with the pyrophosphate moiety of
NADPH. Simulation of the MECR/ETR1–NADPH complex suggested that
Arg216 and Arg218 are important for the interaction and for cofactor specificity.
5.2.2 Substrate binding pocket
A substrate binding pocket was found in the structure of MECR/ETR1 (Fig. 5 in
II). Three residues lining the fatty acyl binding pocket were studied by
mutagenesis and kinetic characterization. Ile129 is a conserved residue localized
at the end of the substrate binding pocket. Gly165 and Phe324 are residues in the
middle of the pocket. Three mutant variants, namely, Ile129Met, Gly165Ser, and
Phe324Tyr display a similar level of activity toward the C8 substrate compared to
the wild type protein. Interestingly, they show a much lower activity toward long
chain substrates such as C14 and C16 compared to the wild type MECR (Table 4
in II), confirming that the observed pocket is real and can be manipulated.
5.2.3 Binding of ACP
In the fatty acyl–ACP substrate, the fatty acyl moiety is covalently bound to the
pantetheine moiety of ACP. The fatty acyl moiety can enter the catalytic site of
MECR/ETR1 via a narrow cleft (Fig. 7 in II). The length of the cleft is
approximately equal to the length of an extended pantetheine moiety (~14 Å).
Further, a groove is formed near the entrance of the cleft. The ~15 Å wide groove
contains a few basic residues (Lys248, Lys252, Arg274, Arg278 and Lys303),
which may interact with the acidic residues of the α2 helix of ACP.
5.2.4 Kinetics
Bimodal distribution of catalytic efficiency of wild-type MECR/ETR1
MECR/ETR1 showed a high Km for short chain substrates (C4 and C6) and a low
Km for medium and long chain substrates (C8–C16) (Table 2 in II). Specifically,
the Km values decrease from 74.2 to 3.6 μM when the chain length of the
substrate increases from C4 to C12, although the Km values for C14 and C16 are
slightly higher than that for C12. Furthermore, MECR/ETR1 shows higher
turnover for the C8 and C12 substrates than for C6, C10, and C14 and other
53
substrates as well. Altogether, MECR/ETR1 shows a bimodal distribution of
catalytic efficiencies and a higher affinity and turnover of MECR/ETR1 for
medium- and long-chain substrates than for short-chain substrates.
Catalytic residues
To probe the role of several residues in the active site (Fig. 6 in II), the mutated
forms (Ser85Ala, Tyr94Phe, Thr170Ala, Trp311Ala, and Trp311Leu) were
purified and their kinetic properties were determined (Table 3 in II). Tyr94Phe
and Trp311Ala showed a substantial increase in Km (10.2 μM for wild-type,
78.5 μM for Tyr94Phe, and 56.5 μM for Trp311Ala) and decrease in kcat (3.0 s−1
for wild-type, 0.011 s−1 for Tyr94Phe, and 0.31 s−1 for Trp311Ala), suggesting that
they are important for substrate binding and catalysis. Further, Trp311Leu showed
a higher kcat/Km than that of Trp311Ala, suggesting that the size of the Trp311 side
chain is relevant for the catalysis. The mutagenesis of Ser85 and Thr170 to
alanines reduced the catalytic efficiency to approximately 30% of the wild-type,
suggesting they are not very critical for the catalysis.
5.3
Mecr transgenic mice
5.3.1 Transgenic mice
Gordon and coauthors succeeded in genetic transformation of mouse embryos by
microinjection of DNA (Gordon et al. 1980). Soon after that, a number of
transgenic mice were generated using pronuclear microinjection. Transgenic and
knockout mouse models can be used to study gene expression and human
diseases making them essential tools for biomedical research. Furthermore, gene
trapped mice can be used to find new genes (Ward et al. 2000).
A transgenic construct usually forms a transgene array with multiple copies
before integration into a chromosome, where it is attacked by an exonuclease
resulting in single strand exposure. The complementary regions of the exposed
strands hybridize to and form duplexes with genomic DNA, and then a repair
process mediates integration via DNA synthesis and ligation. Integration often
occurs through illegitimate recombination between poorly matched sequences.
The strands in the region are covalently closed by well characterized repair
reactions involving further nuclease action, ligation and probably DNA synthesis.
54
A replication “eye”, which is formed by the replication forks diverging from a
chromosomal origin of replication, might be invaded by the transgenic construct.
The frequency of mosaicism among transgenic mice produced by embryo
microinjection is around 65%. Many of the embryos develop normally after
microinjection and are born alive, normally between 5 and 25% of injected
embryos incorporate the foreign DNA into their chromosomes (Bishop 1999).
5.3.2 Generation of Mecr oxerexpressing mice
Transcription of the mammalian MT-I genes is activated by heavy metal load via
the metal-regulatory transcription factor-I (MTF-I), which is expressed at similar
levels in all tissues, except in the testes where the mRNA level is apparently
higher than in other tissues (Auf der Maur et al. 2000). The current model
suggests that the zinc-finger domain of MTF-1 directly (and reversibly) binds to
zinc (Andrews et al. 2001). The protein then adopts a DNA-binding conformation
and translocates to the nucleus, where it binds to metal-response elements in a
few gene promoters (e.g., MT-I), leading to increased transcription.
Ralph L. Brinster is the pioneer of using mouse MT-I promoter to generate
transgenic animals. In one of his publications, 70% of transgenic mice for
overexpression of a growth hormone gene showed high levels of expression, and
the expression level is influenced by the site of integration (Palmiter et al. 1983).
Brinster also tried to overexpress müllerian inhibiting substance (MIS) using the
mouse MT-I promoter, and therefore generated nine transgenic founders (7 male
and 2 female) (Behringer et al. 1990). Two male transgenic mice lines showed no
overexpression, while all the other transgenic mouse lines overexpressed the
target gene, and thus have disorders in their reproduction systems. Miller and coauthors used the mouse MT-I promoter for expression of the growth hormone
gene in swine (Miller et al. 1989). Eight of nine transgenic animals showed
overexpression of the target genes with significant variation in the expression
level.
To study the function of Mecr in a physiological context in mouse, a
construct was prepared under the control of the MT-I promoter, which is
ubiquitously active at low levels in tissues. Two transgenic founders (tg60 and
tg62) were generated (Fig. 1a and 1b in III). Transgenic line tg60 showed lower
level of expression (1.72) in terms of mRNA than transgenic line tg62 (13.3) (Fig.
1c in III).
55
5.3.3 Echocardiography
Both Mecr transgenic lines had significantly reduced cardiac systolic function
(Fig. 3 in III). The ejection fraction was 95.4% in wild-type, 88.1% in tg60 and
85.3% in tg62; (P < 0.01, wt vs. both tg groups). The fractional shortening was
66.8% in wild-type, 52.2% in tg60 and 48.5% in tg62 (P < 0.01, wt vs. both tg
groups). The thickness of the left ventricular posterior wall was 1.2 mm in wildtype vs. 0.8 mm in tg62; P < 0.05. In line tg62, left ventricular dilatation was seen,
as the diameter of the left ventricle was 2.9 mm in wild-type, 3.1 mm in tg60,
p = 0.47, and 3.4 mm in tg62, P < 0.05.
5.3.4 Heart perfusion
The heart perfusion experiments were monitored mainly in three levels (Fig. 4 in
III). At the endogenous frequency (3.2–3.8 Hz), no significant difference in the
work output was found between Mecr transgenic hearts (36.2 ± 11.6) and wildtype (38.3 ± 5.9), although the mean value of the heart rate for transgenic mice is
somewhat slower than that of wild-type mice. At 5 Hz, the work output was lower
in the Mecr transgenic hearts (55.3 ± 24.6) than in wild-type (65.6 ± 11.8). At 7.5
Hz, the difference in the work output between two groups was increased
(58.0 ± 25.1 for transgenic vs 81.9 ± 10.2 for wild-type). Moreover, in the wildtype hearts a linear correlation (P < 0.005) between heart rate and work output
was observed, which is not the case for the transgenic hearts. When the pacing
was increased to 10 Hz, the wild-type hearts responded, but the Mecr transgenic
hearts did not. After pacing at 7.5 Hz the transgenic hearts show a lower mean
value of the CrP/Pi ratio (1.26 ± 0.17 for transgenic vs 1.40 ± 0.43 for wild-type).
5.3.5 Exercise endurance test
When the mice were challenged to exercise on a treadmill, the Mecr tg62 mice
and wild-type mice differed significantly in three parameters (Fig. 5 in III), the
running distance (396 ± 85 m for tg62 vs. 740 ± 150 m for wt, P < 0.005), vertical
distance (118 ± 43 m for tg62 vs. 301 ± 81 m for wt, P < 0.005) and vertical work
performance (3.62 ± 1.62 kg·m for tg62 vs. 8.45 ± 1.85 kg·m for wt, P < 0.005).
56
6
Discussion
6.1
2-Enoyl thioester reductases (ETRs)
The last step of mitochondrial FAS is catalyzed by 2-enoyl thioester reductase
(ETR), producing saturated FAs which can enter the next round of FAS reactions
or can be used for final function and further processing. There are several types of
ETRs found in nature, which differ between organisms as well as between cellular
compartments.
In bacteria, there are at least four ETRs, namely, FabI (E. coli and Bacillus
subtilis), FabK (Streptococcus pneumoniae), FabL (B. subtilis) and FabV (Vibrio
cholerae). FabI belongs to the SDR superfamily and uses NADH as a cofactor
(Rock & Cronan 1996). A chlorinated bisphenol called triclosan can inhibit FabI
by forming a FabI–NAD+–triclosan ternary complex (Heath et al. 1999), whereas
FabK, FabL and FabV are all triclosan-resistant (Massengo-Tiasse & Cronan
2008). FabK is distinct from FabI in amino acid sequence and uses flavin
mononucleotide (FMN) and NADH as cofactors (Marrakchi et al. 2002).
Moreover, the overall structure of FabK is similar to ETR of the ER fatty acid
elongation system in fungi (Saito et al. 2008). Each monomer of dimeric FabK
has a triose phosphate isomerase (TIM) barrel fold. AG205, a phenylimidazole
derivative inhibitor binds to a hydrophobic pocket in the catalytic site of FabK
probably competing with NADH for binding to FabK (Saito et al. 2008).
Interestingly, B. subtilis contains both FabI and FabL, which share a low level of
sequence similarity (Heath & Rock 2000). FabV is 60% larger than the typical
SDRs in length, and shows a strong preference for NADH over NADPH as a
cofactor (Massengo-Tiasse & Cronan 2008).
Fungal cytosolic FAS is a 2.6-megadalton barrel-shaped α6β6 complex,
containing two separated reaction chambers, each chamber having three sets of
active sites (Jenni et al. 2006). This organization may be more suitable for the
coordinated biosynthesis of FAs compared to that of bacterial type II FAS. The
ETR domain of fungal FAS contains binding sites for both FMN and NADPH as
cofactors (Fox & Lynen 1980). Interestingly, the FMN-binding part consists of a
TIM-barrel fold; and the NADPH-binding part is predominantly formed by a
α-helical domain, clearly different from a Rossmann fold (Jenni et al. 2006). The
yeast mitochondrial Etr1p is a member of MDR family. Interestingly, Candida
tropicalis contains two mitochondrial ETRs, forming both homodimers and
57
heterodimers (Torkko et al. 2003). It is noteworthy that the structure of the yeast
Etr1p is very similar to that of its mammalian functional counterpart
MECR/ETR1, an MDR enzyme. The ETR of the ER-elongation system in yeast is
Tsc13p (Kohlwein et al. 2001), which apparently bears no similarity to ETRs of
other FAS systems in yeast. Tsc13p contains six membrane-spanning domains,
and the putative catalytic residues of Tsc13p may lie on the cytosolic face of the
ER, partially embedded within the ER membrane (Paul et al. 2007). In addition,
Tsc13p and other enzymes in the elongation pathway are probably organized in a
complex (Tehlivets et al. 2007).
In mammals, the cytosolic FAS I is formed by a 0.54 mDa X-shaped
intertwined α2 homodimer of a single 270 kDa polypeptide. The ETR domain of
mammalian FAS I belongs to the MDR superfamily rather than to the bacterial
type SDR; and it is structurally very similar to a zinc-free bacterial quinone
reductase (QORTt) from Thermus thermophilus HB8 (Maier et al. 2006). The
ETR domain of mammalian FAS I contains a NADPH-binding Rossmann fold as
well as a substrate binding domain (Maier et al. 2008). The mitochondrial ETR
(MECR/ETR1) in mammals is the most extensively studied. It forms a dimer with
a molecular mass of 37 kDa for each monomer. MECR/ETR1 shows a bimodal
distribution of catalytic specificity with maxima at C8 and C12. The ETR in
mammalian ER shows the highest activity with 2-hexenoyl-CoA (Podack et al.
1974), although its molecular and biochemical nature has not yet been determined.
The peroxisomal ETR in mammals is a monomeric SDR with a molecular mass
of 32.5 kDa, and displays a maximum activity with the medium chain length
substrate (C10) (Das et al. 2000).
Interestingly, the respiratory deficient phenotype of yeast deficient in Etr1p, a
dimeric member of MDR family, can be rescued by mitochondrially targeted FabI,
a tetrameric member of SDR family (Torkko et al. 2001). This observation
suggested that the enzyme activity per se is more important than the structural
properties of the protein complexes in vivo.
6.2
Lipoic acid and other products
Structural and kinetic studies show that HsmtKAS and MECR/ETR1 have broad
substrate acceptance (from C4 to C16) (Zhang et al. 2005, Christensen et al. 2007,
Chen et al. 2008). This suggests that the physiological role of the mitochondrial
FAS may not be limited to the previously proposed role – making octanoic acid,
the lipoic acid precursor (Harington et al. 1993, Brody et al. 1997, Wada et al.
58
1997, Witkowski et al. 2007). In fact, some reports also claimed that
mitochondrial FAS may produce longer chain products, which might be linked to
the remodeling of cardiolipins (Zensen et al. 1992, Schneider et al. 1995) or
mitochondrial phospholipids (Schneider et al. 1995, Guler et al. 2008). Lipoic
acid is a critical component of the E2 subunit of the mitochondrial α-ketoacid
dehydrogenase complexes or linked to the H-protein of the glycine cleavage
system (Schonauer et al. 2008). In mammals, in addition to the dietary source,
there is a lipoic acid synthase which is indispensable for embryonic development
(Yi & Maeda 2005). Deletion of S. cerevisiae Etr1 resulted in respiration
deficiency (Torkko et al. 2001), which may be at least partially due to the
insufficiency of lipoic acid production. Altogether, one of the main products of
mitochondrial FAS is apparently C8 which is used for making lipoic acid.
Nevertheless, another longer chain product is possible even though it has not yet
been determined conclusively, especially in mammals. It is noteworthy that the
catalytic efficiency of mammalian enzymes of mitochondrial FAS is much higher
toward longer chain substrates (> C8) compared to that of the yeast counterparts
(Torkko et al. 2003, Chen et al. 2008), suggesting a possibility that the function of
the mammalian pathway may be different from the yeast one. An RNAi
knockdown experiment would provide some information about the role of
mammalian mitochondria on lipoic acid synthesis.
6.3
CoA and ACP
During FA metabolism, fatty acyls frequently form thiol esters with
4'-phosphopantetheine moiety of coenzymes (CoA or ACP). Fatty acyl
CoA/ACPs attach to enzymes transiently and transfer the covalent fatty acyl tails
between distinct catalytic sites via a phosphopantethiene chain, the flexibility and
length of which allows the intermediate transfer to process efficiently.
CoA is synthesized in five steps from pantothenic acid (vitamin B5), which is
also the source of 4’-phosphopantethiene, the prosthetic group of ACP (Leonardi
et al. 2005). Prokaryotic and eukaryotic CoA synthetic enzymes (CoaA, CoaB,
CoaC, CoaD, CoaE) are different in their primary sequences although they
display the same catalytic activities. For instance, CoaD and CoaE are two
separated enzymes in prokaryotes whereas they are fused to form a bifunctional
protein in mammals. The concentration of CoAs is tightly regulated. In the
mammalian heart, CoA can reach ~100 nmol/g tissue, which is lower than in liver
(~130–430 nmol/g) but higher than in most of other tissues (Tahiliani & Beinlich
59
1991). Interestingly, tumors have low CoA levels (McAllister et al. 1988). Within
a mammalian cell, the concentration of CoA is around 0.08 mM in the cytosol,
3.5 mM in mitochondria, 0.7 mM in peroxisomes (Leonardi et al. 2005).
In FAS systems, the fatty acyl-ACPs are the substrates, which are different
from FAO systems where only fatty acyl-CoAs are accepted as substrates. In the
organelles such as mitochondria where both FAO and FAS present, the nature of
substrates becomes very important to separate the two pathways. Notably, even
though FAS enzymes apparently contain recognition sites for the interaction with
ACP, many of them can also accept fatty acyl-CoA substrates at least in vitro. It is
to be determined if these FAS enzymes can use fatty acyl-CoAs substrates in vivo.
If this is the case, the mitochondrial FAS may also play a role in the regulation of
FAO in addition to its primary role in the synthesis of specialized FAs.
6.4
FAS II enzymes exist at low levels in mammalian mitochondria
The isolation of ACP from bovine heart mitochondria was the first piece of
evidence suggesting the existence of the FAS II enzyme in mammals (Runswick
et al. 1991). In agreement with this suggestion, we were able to isolate
MECR/ETR1 from bovine heart mitochondria.
It is important to note that FAS II components in mammals are probably of
low abundance compared to β-oxidation enzymes in mitochondria (unpublished
observation). The fact that many of the proteomic studies do not reveal any
components of mitochondrial FAS II supports this hypothesis (Ohlmeier et al.
2004, Rezaul et al. 2005). Carroll et al revealed an ACP which is the substrate
shuttling component of mitochondrial FAS, through analysis of the subunit
composition of complex I from bovine heart mitochondria (Carroll et al. 2003).
Most recently, Pagliarini et al identified several mammalian FAS II components
(e.g., MECR/ETR1, KAS/CEM1, malonyl CoA:ACP acyltransferase), during the
investigation of 1098 putative mitochondrial proteins from mouse tissues using
multiple advanced techniques (Pagliarini et al. 2008). Nevertheless, in bovine
heart mitochondria, the amount of BtMECR/ETR1 protein (and most likely other
components as well) is much less than that of 3-hydroxyacyl-CoA dehydrogenase
or other β-oxidation enzymes (data not shown). This is line with the notion that
the end products of mitochondrial FAS are most likely certain specialized lipids,
instead of the bulk of the FAs within the cells.
60
6.5
FAS II and its physiological function
Inactivation of each of the yeast FAS II genes results in loss of cytochromes and
defects in mitochondrial respiration (Harington et al. 1993, Schneider et al. 1995,
Schneider et al. 1997, Torkko et al. 2001, Kastaniotis et al. 2004), indicating that
eukaryotic mitochondrial FAS is important for maintaining the function of these
organelles. Moreover, mitochondrial FAS members are mainly expressed in
mitochondria-enriched tissues, such as heart and skeletal muscle (Joshi et al. 2003,
Zhang et al. 2003a, Zhang et al. 2005, Autio et al. 2007). Mitochondrial FAS is
linked genetically to RNA processing in vertebrates (Autio et al. 2007) and
functionally to RNA processing in yeast (Schonauer et al. 2008). In addition,
yeast mutants with compromised mitochondrial FAS function are synthetically
petite (synthetically lethal on non-fermentable carbon source) with mutations in
the mitochondrial RNA processing and translation apparatus (Kursu et al,
personal communication).
Overexpression of Mecr leads to a change in mitochondrial morphology
including the increased density of cristae. The average size of the mitochondria
from Mecr transgenic mice was also enlarged, partially resembles the ETR1
overexpressing phenotype in yeast, although the difference between transgenic
and wild-type groups maybe not be as large as that in yeast groups.
The consequence of overexpression of Mecr in mice ultimately leads to the
impairment of heart function. In particular, systolic function was significantly
reduced. It will be interesting to know if aberrant expression of other components
of mitochondrial FAS can result in heart dysfunction in mice. Furthermore, recent
studies showed that Mecr is linked to kidney development in Xenopus (Kyuno et
al. 2008). Therefore, it is also important to investigate the physiological roles of
FAS II in other mitochondria-containing tissues/organs.
61
62
7
Conclusions
This study aimed to characterize 2E-enoyl thioester reductase of mitochondrial
FAS II in mammals. To achieve this goal, three major approaches were used.
The mammalian protein MECR/ETR1, previously called NRFB-1, was
purified, and localized in mammalian mitochondria using biochemical approaches.
The protein displays 2E-enoyl thioester reductase activity.
Using structure enzymological approaches, MECR/ETR1 was crystallized
and the structure of MECR/ETR1 was solved. A substrate binding pocket of this
enzyme was revealed based on the results of structural, mutagenesis and kinetic
analysis. An ACP binding site was suggested in this study. In addition, a bimodal
distribution of activity was shown based on the kinetic properties of the wild type
enzyme, supporting the possibility of two major products of human mitochondrial
FAS.
To study the physiological significance of mitochondrial FAS in mammals,
Mecr transgenic mice were generated. The Mecr-overexpressing mice showed an
impairment of heart systolic function and damaged mitochondrial ultrastructure,
as demonstrated using histology, echocardiography, heart perfusion, and an
endurance performance test. It was proposed that FAS II in mammals may play a
role in the function of mitochondria and the heart. Thus a new set of genes that
may be involved in cardiomyopathy was suggested. Finally, we provided the first
mammalian model for studying mitochondrial FAS II.
In conclusion, our work on MECR/ETR1, the last enzyme of mitochondrial
FAS, provides further evidence that FAS II is highly conserved and very likely to
also be functional in mammals.
63
64
References
Abe K, Shinohara Y & Terada H (1998) Isolation and characterization of cDNA encoding
rat heart type acetyl-CoA carboxylase. Biochim Biophys Acta 1398: 347–352.
Abu-Elheiga L, Brinkley WR, Zhong L, Chirala SS, Woldegiorgis G & Wakil SJ (2000)
The subcellular localization of acetyl-CoA carboxylase 2. Proc Natl Acad Sci USA 97:
1444–1449.
Abu-Elheiga L, Oh W, Kordari P & Wakil SJ (2003) Acetyl-CoA carboxylase 2 mutant
mice are protected against obesity and diabetes induced by high-fat/high-carbohydrate
diets. Proc Natl Acad Sci USA 100: 10207–10212.
Acosta-Alvear D, Zhou Y, Blais A, Tsikitis M, Lents NH, Arias C, Lennon CJ, Kluger Y
& Dynlacht BD (2007) XBP1 controls diverse cell type- and condition-specific
transcriptional regulatory networks. Mol Cell 27: 53–66.
Adamski J, Normand T, Leenders F, Monte D, Begue A, Stehelin D, Jungblut PW & de
Launoit Y (1995) Molecular cloning of a novel widely expressed human 80 kDa 17
beta-hydroxysteroid dehydrogenase IV. Biochem J 311: 437–443.
Airenne TT, Torkko JM, Van den plas S, Sormunen RT, Kastaniotis AJ, Wierenga RK &
Hiltunen JK (2003) Structure-function analysis of enoyl thioester reductase involved
in mitochondrial maintenance. J Mol Biol 327: 47–59.
Alam N & Saggerson ED (1998) Malonyl-CoA and the regulation of fatty acid oxidation
in soleus muscle. Biochem J 334: 233–241.
An JH, Lee HY, Ko KN, Kim ES & Kim YS (2002) Symbiotic effects of deltamatB
Rhizobium leguminosarum bv. trifolii mutant on clovers. Mol Cells 14: 261–266.
Anderson S, Bankier AT, Barrell BG, de Bruijn MH, Coulson AR, Drouin J, Eperon IC,
Nierlich DP, Roe BA, Sanger F, Schreier PH, Smith AJ, Staden R & Young IG (1981)
Sequence and organization of the human mitochondrial genome. Nature 290: 457–465.
Andersson U & Scarpulla RC (2001) Pgc-1-related coactivator, a novel, serum-inducible
coactivator of nuclear respiratory factor 1-dependent transcription in mammalian cells.
Mol Cell Biol 21: 3738–3749.
Andrews GK, Lee DK, Ravindra R, Lichtlen P, Sirito M, Sawadogo M & Schaffner W
(2001) The transcription factors MTF-1 and USF1 cooperate to regulate mouse
metallothionein-I expression in response to the essential metal zinc in visceral
endoderm cells during early development. EMBO J 20: 1114–1122.
Arakawa H, Amaya Y & Mori M (1990) The NH2-terminal 14-16 amino acids of
mitochondrial and bacterial thiolases can direct mature ornithine carbamoyltransferase
into mitochondria. J Biochem (Tokyo) 107: 160–164.
Auf der Maur A, Belser T, Wang Y, Gunes C, Lichtlen P, Georgiev O & Schaffner W
(2000) Characterization of the mouse gene for the heavy metal-responsive
transcription factor MTF-1. Cell Stress Chaperones 5: 196–206.
Autio KJ, Kastaniotis AJ, Pospiech H, Miinalainen IJ, Schonauer MS, Dieckmann CL &
Hiltunen JK (2008) An ancient genetic link between vertebrate mitochondrial fatty
acid synthesis and RNA processing. FASEB J 22: 569–578.
Auwerx J (1999) PPARgamma, the ultimate thrifty gene. Diabetologia 42: 1033–1049.
65
Barak Y, Nelson MC, Ong ES, Jones YZ, Ruiz-Lozano P, Chien KR, Koder A & Evans
RM (1999) PPAR gamma is required for placental, cardiac, and adipose tissue
development. Mol Cell 4: 585–595.
Behringer RR, Cate RL, Froelick GJ, Palmiter RD & Brinster RL (1990) Abnormal sexual
development in transgenic mice chronically expressing mullerian inhibiting substance.
Nature 345: 167–170.
Bergler H, Fuchsbichler S, Hogenauer G & Turnowsky F (1996) The enoyl-[acyl-carrierprotein] reductase (FabI) of Escherichia coli, which catalyzes a key regulatory step in
fatty acid biosynthesis, accepts NADH and NADPH as cofactors and is inhibited by
palmitoyl-CoA. Eur J Biochem 242: 689–694.
Bianchi A, Evans JL, Iverson AJ, Nordlund AC, Watts TD & Witters LA (1990)
Identification of an isozymic form of acetyl-CoA carboxylase. J Biol Chem 265:
1502–1509.
Bishop J (1999) Transgenic mammals. Pearson education limited, Edinburgh Gate, Harlow,
England.
Blair E, Redwood C, Ashrafian H, Oliveira M, Broxholme J, Kerr B, Salmon A, OstmanSmith I & Watkins H (2001) Mutations in the gamma(2) subunit of AMP-activated
protein kinase cause familial hypertrophic cardiomyopathy: evidence for the central
role of energy compromise in disease pathogenesis. Hum Mol Genet 10: 1215–1220.
Borell CW & Bhattacharjee JK (1988) Cloning and biochemical characterization of LYS5
gene of Saccharomyces cerevisiae. Curr Genet 13: 299–304.
Brody S, Oh C, Hoja U & Schweizer E (1997) Mitochondrial acyl carrier protein is
involved in lipoic acid synthesis in Saccharomyces cerevisiae. FEBS Lett 408: 217–
220.
Cahoon EB, Lindqvist Y, Schneider G & Shanklin J (1997) Redesign of soluble fatty acid
desaturases from plants for altered substrate specificity and double bond position.
Proc Natl Acad Sci USA 94: 4872–4877.
Carroll J, Fearnley IM, Shannon RJ, Hirst J & Walker JE (2003) Analysis of the subunit
composition of complex I from bovine heart mitochondria. Mol Cell Proteomics 2:
117–126.
Chain EB, Mansford KR & Opie LH (1969) Effects of insulin on the pattern of glucose
metabolism in the perfused working and Langendorff heart of normal and insulindeficient rats. Biochem J 115: 537–546.
Chawla A, Repa JJ, Evans RM & Mangelsdorf DJ (2001) Nuclear receptors and lipid
physiology: opening the X-files. Science 294: 1866–1870.
Christensen CE, Kragelund BB, von Wettstein-Knowles P & Henriksen A (2007) Structure
of the human beta-ketoacyl [ACP] synthase from the mitochondrial type II fatty acid
synthase. Protein Sci 16: 261–272.
Cinti DL, Cook L, Nagi MN & Suneja SK (1992) The fatty acid chain elongation system
of mammalian endoplasmic reticulum. Prog Lipid Res 31: 1–51.
Cole H, Reynolds TR, Lockyer JM, Buck GA, Denson T, Spence JE, Hymes J & Wolf B
(1994) Human serum biotinidase. cDNA cloning, sequence, and characterization. J
Biol Chem 269: 6566–6570.
66
Corrado D, Basso C & Thiene G (2000) Arrhythmogenic right ventricular cardiomyopathy:
diagnosis, prognosis, and treatment. Heart 83: 588–595.
Cronan JE, Fearnley IM & Walker JE (2005) Mammalian mitochondria contain a soluble
acyl carrier protein. FEBS Lett 579: 4892–4896.
Cronan JE, Jr. (1982) Molecular properties of short chain acyl thioesters of acyl carrier
protein. J Biol Chem 257: 5013–5017.
Das AK, Uhler MD & Hajra AK (2000) Molecular cloning and expression of mammalian
peroxisomal trans-2-enoyl-coenzyme A reductase cDNAs. J Biol Chem 275: 24333–
24340.
Davis MS, Solbiati J & Cronan JE, Jr. (2000) Overproduction of acetyl-CoA carboxylase
activity increases the rate of fatty acid biosynthesis in Escherichia coli. J Biol Chem
275: 28593–28598.
Dentin R, Benhamed F, Pegorier JP, Foufelle F, Viollet B, Vaulont S, Girard J & Postic C
(2005) Polyunsaturated fatty acids suppress glycolytic and lipogenic genes through
the inhibition of ChREBP nuclear protein translocation. J Clin Invest 115: 2843–2854.
Dieuaide-Noubhani M, Novikov D, Baumgart E, Vanhooren JC, Fransen M, Goethals M,
Vandekerckhove J, Van Veldhoven PP & Mannaerts GP (1996) Further
characterization of the peroxisomal 3-hydroxyacyl-CoA dehydrogenases from rat liver.
Relationship between the different dehydrogenases and evidence that fatty acids and
the C27 bile acids di- and tri-hydroxycoprostanic acids are metabolized by separate
multifunctional proteins. Eur J Biochem 240: 660–666.
Feige JN & Auwerx J (2007) Transcriptional coregulators in the control of energy
homeostasis. Trends Cell Biol 17: 292–301.
Finck BN & Kelly DP (2007) Peroxisome proliferator-activated receptor gamma
coactivator-1 (PGC-1) regulatory cascade in cardiac physiology and disease.
Circulation 115: 2540–2548.
Fisher M, Kroon JT, Martindale W, Stuitje AR, Slabas AR & Rafferty JB (2000) The
X-ray structure of Brassica napus beta-keto acyl carrier protein reductase and its
implications for substrate binding and catalysis. Structure 8: 339–347.
FitzPatrick DR, Hill A, Tolmie JL, Thorburn DR & Christodoulou J (1999) The molecular
basis of malonyl-CoA decarboxylase deficiency. Am J Hum Genet 65: 318–326.
Flaman AS, Chen JM, Van Iderstine SC & Byers DM (2001) Site-directed mutagenesis of
acyl carrier protein (ACP) reveals amino acid residues involved in ACP structure and
acyl-ACP synthetase activity. J Biol Chem 276: 35934–35939.
Flugel RS, Hwangbo Y, Lambalot RH, Cronan JE, Jr. & Walsh CT (2000) Holo-(acyl
carrier protein) synthase and phosphopantetheinyl transfer in Escherichia coli. J Biol
Chem 275: 959–968.
Fox JL & Lynen F (1980) Characterization of the flavoenzyme enoyl reductase of fatty
acid synthetase from yeast. Eur J Biochem 109: 417–424.
Fraser AG, Kamath RS, Zipperlen P, Martinez-Campos M, Sohrmann M & Ahringer J
(2000) Functional genomic analysis of C. elegans chromosome I by systematic RNA
interference. Nature 408: 325–330.
67
Furuta S, Kobayashi A, Miyazawa S & Hashimoto T (1997) Cloning and expression of
cDNA for a newly identified isozyme of bovine liver 3-hydroxyacyl-CoA
dehydrogenase and its import into mitochondria. Biochim Biophys Acta 1350: 317–
324.
Gebauer SK, Psota TL & Kris-Etherton PM (2007) The diversity of health effects of
individual trans fatty acid isomers. Lipids 42: 787–799.
Gehring AM, Bradley KA & Walsh CT (1997) Enterobactin biosynthesis in Escherichia
coli: isochorismate lyase (EntB) is a bifunctional enzyme that is
phosphopantetheinylated by EntD and then acylated by EntE using ATP and
2,3-dihydroxybenzoate. Biochemistry 36: 8495–8503.
Glembotski CC (2007) Endoplasmic reticulum stress in the heart. Circ Res 101: 975–984.
Goldstein JL, DeBose-Boyd RA & Brown MS (2006) Protein sensors for membrane
sterols. Cell 124: 35–46.
Gonczy P, Echeverri C, Oegema K, Coulson A, Jones SJ, Copley RR, Duperon J, Oegema
J, Brehm M, Cassin E, Hannak E, Kirkham M, Pichler S, Flohrs K, Goessen A, Leidel
S, Alleaume AM, Martin C, Ozlu N, Bork P & Hyman AA (2000) Functional
genomic analysis of cell division in C. elegans using RNAi of genes on chromosome
III. Nature 408: 331–336.
Gordon JW, Scangos GA, Plotkin DJ, Barbosa JA & Ruddle FH (1980) Genetic
transformation of mouse embryos by microinjection of purified DNA. Proc Natl Acad
Sci USA 77: 7380–7384.
Gueguen V, Macherel D, Jaquinod M, Douce R & Bourguignon J (2000) Fatty acid and
lipoic acid biosynthesis in higher plant mitochondria. J Biol Chem 275: 5016–5025.
Guler JL, Kriegova E, Smith TK, Lukes J & Englund PT (2008) Mitochondrial fatty acid
synthesis is required for normal mitochondrial morphology and function in
Trypanosoma brucei. Mol Microbiol 67: 1125–42.
Gupta P, Huq MD, Khan SA, Tsai NP & Wei LN (2005) Regulation of co-repressive
activity of and HDAC recruitment to RIP140 by site-specific phosphorylation. Mol
Cell Proteomics 4: 1776–1784.
Ha J, Lee JK, Kim KS, Witters LA & Kim KH (1996) Cloning of human acetyl-CoA
carboxylase-beta and its unique features. Proc Natl Acad Sci USA 93: 11466–11470.
Hamilton B, Dong Y, Shindo M, Liu W, Odell I, Ruvkun G & Lee SS (2005) A systematic
RNAi screen for longevity genes in C. elegans. Genes Dev 19: 1544–1555.
Hamilton C & Saggerson ED (2000) Malonyl-CoA metabolism in cardiac myocytes.
Biochem J 350: 61–67.
Hardie DG & Carling D (1997) The AMP-activated protein kinase–fuel gauge of the
mammalian cell? Eur J Biochem 246: 259–273.
Harington A, Herbert CJ, Tung B, Getz GS & Slonimski PP (1993) Identification of a new
nuclear gene (CEM1) encoding a protein homologous to a beta-keto-acyl synthase
which is essential for mitochondrial respiration in Saccharomyces cerevisiae. Mol
Microbiol 9: 545–555.
68
He XY, Merz G, Mehta P, Schulz H & Yang SY (1999) Human brain short chain
L-3-hydroxyacyl coenzyme A dehydrogenase is a single-domain multifunctional
enzyme. Characterization of a novel 17beta-hydroxysteroid dehydrogenase. J Biol
Chem 274: 15014–15019.
He XY, Merz G, Yang YZ, Pullakart R, Mehta P, Schulz H & Yang SY (2000a) Function
of human brain short chain L-3-hydroxyacyl coenzyme A dehydrogenase in androgen
metabolism. Biochim Biophys Acta 1484: 267–277.
He XY, Schulz H & Yang SY (1998) A human brain L-3-hydroxyacyl-coenzyme A
dehydrogenase is identical to an amyloid beta-peptide-binding protein involved in
Alzheimer's disease. J Biol Chem 273: 10741–10746.
He XY, Yang YZ, Schulz H & Yang SY (2000b) Intrinsic alcohol dehydrogenase and
hydroxysteroid dehydrogenase activities of human mitochondrial short-chain
L-3-hydroxyacyl-CoA dehydrogenase. Biochem J 345: 139–143.
Heath RJ & Rock CO (1995) Enoyl-acyl carrier protein reductase (fabI) plays a
determinant role in completing cycles of fatty acid elongation in Escherichia coli. J
Biol Chem 270: 26538–26542.
Heath RJ & Rock CO (2000) A triclosan-resistant bacterial enzyme. Nature 406: 145–146.
Heath RJ & Rock CO (2004) Fatty acid biosynthesis as a target for novel antibacterials.
Curr Opin Investig Drugs 5: 146–153.
Heath RJ, Rubin JR, Holland DR, Zhang E, Snow ME & Rock CO (1999) Mechanism of
triclosan inhibition of bacterial fatty acid synthesis. J Biol Chem 274: 11110–11114.
Heesom KJ, Moule SK & Denton RM (1998) Purification and characterisation of an
insulin-stimulated protein-serine kinase which phosphorylates acetyl-CoA
carboxylase. FEBS Lett 422: 43–46.
Henrikson KP & Allen SH (1979) Purification and subunit structure of propionyl
coenzyme A carboxylase of Mycobacterium smegmatis. J Biol Chem 254: 5888–5891.
Hiltunen JK, Filppula SA, Koivuranta KT, Siivari K, Qin YM & Hayrinen HM (1996)
Peroxisomal beta-oxidation and polyunsaturated fatty acids. Ann N Y Acad Sci 804:
116–128.
Hiltunen JK, Wenzel B, Beyer A, Erdmann R, Fossa A & Kunau WH (1992) Peroxisomal
multifunctional beta-oxidation protein of Saccharomyces cerevisiae. Molecular
analysis of the fox2 gene and gene product. J Biol Chem 267: 6646–6653.
Hirschberg CB, Robbins PW & Abeijon C (1998) Transporters of nucleotide sugars, ATP,
and nucleotide sulfate in the endoplasmic reticulum and Golgi apparatus. Annu Rev
Biochem 67: 49–69.
Hoetzl S, Sprong H & van Meer G (2007) The way we view cellular (glyco)sphingolipids.
J Neurochem 103 Suppl 1: 3–13.
Hopwood DA & Sherman DH (1990) Molecular genetics of polyketides and its
comparison to fatty acid biosynthesis. Annu Rev Genet 24: 37–66.
Houseknecht KL, Cole BM & Steele PJ (2002) Peroxisome proliferator-activated receptor
gamma (PPARgamma) and its ligands: a review. Domest Anim Endocrinol 22: 1–23.
69
Hradec J & Dufek P (1994) Determination of cholesteryl 14-methylhexadecanoate in blood
serum by reversed-phase high-performance liquid chromatography. J Chromatogr B
Biomed Appl 660: 386–389.
Hu E, Tontonoz P & Spiegelman BM (1995) Transdifferentiation of myoblasts by the
adipogenic transcription factors PPAR gamma and C/EBP alpha. Proc Natl Acad Sci
USA 92: 9856–9860.
Huang YW, Pineau I, Chang HJ, Azzi A, Bellemare V, Laberge S & Lin SX (2001)
Critical residues for the specificity of cofactors and substrates in human estrogenic
17beta-hydroxysteroid dehydrogenase 1: variants designed from the three-dimensional
structure of the enzyme. Mol Endocrinol 15: 2010–2020.
Hunaiti AR & Kolattukudy PE (1982) Isolation and characterization of an acyl-coenzyme
A carboxylase from an erythromycin-producing Streptomyces erythreus. Arch
Biochem Biophys 216: 362–371.
Hunt-Newbury R, Viveiros R, Johnsen R, Mah A, Anastas D, Fang L, Halfnight E, Lee D,
Lin J, Lorch A, McKay S, Okada HM, Pan J, Schulz AK, Tu D, Wong K, Zhao Z,
Alexeyenko A, Burglin T, Sonnhammer E, Schnabel R, Jones SJ, Marra MA, Baillie
DL & Moerman DG (2007) High-throughput in vivo analysis of gene expression in
Caenorhabditis elegans. PLoS Biol 5: e237.
Ishii S, Iizuka K, Miller BC & Uyeda K (2004) Carbohydrate response element binding
protein directly promotes lipogenic enzyme gene transcription. Proc Natl Acad Sci
USA 101: 15597–15602.
Jenni S, Leibundgut M, Maier T & Ban N (2006) Architecture of a fungal fatty acid
synthase at 5 A resolution. Science 311: 1263–1267.
Joly E, Bendayan M, Roduit R, Saha AK, Ruderman NB & Prentki M (2005) MalonylCoA decarboxylase is present in the cytosolic, mitochondrial and peroxisomal
compartments of rat hepatocytes. FEBS Lett 579: 6581–6586.
Jones LN & Rivett DE (1997) The role of 18-methyleicosanoic acid in the structure and
formation of mammalian hair fibres. Micron 28: 469–485.
Jones PJ, Holak TA & Prestegard JH (1987) Structural comparison of acyl carrier protein
in acylated and sulfhydryl forms by two-dimensional 1H NMR spectroscopy.
Biochemistry 26: 3493–3500.
Jordan SW & Cronan JE, Jr. (1997) A new metabolic link. The acyl carrier protein of lipid
synthesis donates lipoic acid to the pyruvate dehydrogenase complex in Escherichia
coli and mitochondria. J Biol Chem 272: 17903–17906.
Jornvall H, Nordling E & Persson B (2003) Multiplicity of eukaryotic ADH and other
MDR forms. Chem Biol Interact 143–144: 255–261.
Jornvall H, Persson B, Krook M, Atrian S, Gonzalez-Duarte R, Jeffery J & Ghosh D (1995)
Short-chain dehydrogenases/reductases (SDR). Biochemistry 34: 6003–6013.
Joseph SB, Laffitte BA, Patel PH, Watson MA, Matsukuma KE, Walczak R, Collins JL,
Osborne TF & Tontonoz P (2002) Direct and indirect mechanisms for regulation of
fatty acid synthase gene expression by liver X receptors. J Biol Chem 277: 11019–
11025.
70
Joshi AK, Zhang L, Rangan VS & Smith S (2003) Cloning, expression, and
characterization of a human 4'-phosphopantetheinyl transferase with broad substrate
specificity. J Biol Chem 278: 33142–33149.
Jungling E & Kammermeier H (1988) A one-vial method for routine extraction and
quantification of free fatty acids in blood and tissue by HPLC. Anal Biochem 171:
150–157.
Kakhniashvili D, Mayor JA, Gremse DA, Xu Y & Kaplan RS (1997) Identification of a
novel gene encoding the yeast mitochondrial dicarboxylate transport protein via
overexpression, purification, and characterization of its protein product. J Biol Chem
272: 4516–4521.
Kallberg Y, Oppermann U, Jornvall H & Persson B (2002) Short-chain
dehydrogenases/reductases (SDRs). Eur J Biochem 269: 4409–4417.
Kamath RS, Fraser AG, Dong Y, Poulin G, Durbin R, Gotta M, Kanapin A, Le Bot N,
Moreno S, Sohrmann M, Welchman DP, Zipperlen P & Ahringer J (2003) Systematic
functional analysis of the Caenorhabditis elegans genome using RNAi. Nature 421:
231–237.
Kaplan RS & Pedersen PL (1985) Isolation and reconstitution of the n-butylmalonatesensitive dicarboxylate transporter from rat liver mitochondria. J Biol Chem 260:
10293–10298.
Kastaniotis AJ, Autio KJ, Sormunen RT & Hiltunen JK (2004) Htd2p/Yhr067p is a yeast
3-hydroxyacyl-ACP dehydratase essential for mitochondrial function and morphology.
Mol Microbiol 53: 1407–1421.
Kerner J & Hoppel CL (2002) Radiochemical malonyl-CoA decarboxylase assay: activity
and subcellular distribution in heart and skeletal muscle. Anal Biochem 306: 283–289.
Kim KH, Lopez-Casillas F, Bai DH, Luo X & Pape ME (1989) Role of reversible
phosphorylation of acetyl-CoA carboxylase in long-chain fatty acid synthesis. FASEB
J 3: 2250–2256.
Kimura Y, Kojyo T, Kimura I & Sato M (1998) Propionyl-CoA carboxylase of
Myxococcus xanthus: catalytic properties and function in developing cells. Arch
Microbiol 170: 179–184.
Kimura Y, Sato R, Mimura K & Sato M (1997) Propionyl coenzyme A carboxylase is
required for development of Myxococcus xanthus. J Bacteriol 179: 7098–7102.
Klein RR, King G, Moreau RA & Haas MJ (1997) Altered acyl chain length specificity of
Rhizopus delemar lipase through mutagenesis and molecular modeling. Lipids 32:
123–130.
Kletzien RF, Clarke SD & Ulrich RG (1992) Enhancement of adipocyte differentiation by
an insulin-sensitizing agent. Mol Pharmacol 41: 393–398.
Kniazeva M, Crawford QT, Seiber M, Wang CY & Han M (2004) Monomethyl branchedchain fatty acids play an essential role in Caenorhabditis elegans development. PLoS
Biol 2: E257.
Kobayashi A, Jiang LL & Hashimoto T (1996) Two mitochondrial 3-hydroxyacyl-CoA
dehydrogenases in bovine liver. J Biochem (Tokyo) 119: 775–782.
71
Kohlwein SD, Eder S, Oh CS, Martin CE, Gable K, Bacikova D & Dunn T (2001) Tsc13p
is required for fatty acid elongation and localizes to a novel structure at the nuclearvacuolar interface in Saccharomyces cerevisiae. Mol Cell Biol 21: 109–125.
Kyuno J, Masse K & Jones EA (2008) A functional screen for genes involved in Xenopus
pronephros development. Mech Dev 125: 571–586.
Lambalot RH, Gehring AM, Flugel RS, Zuber P, LaCelle M, Marahiel MA, Reid R,
Khosla C & Walsh CT (1996) A new enzyme superfamily – the phosphopantetheinyl
transferases. Chem Biol 3: 923–936.
Lambalot RH & Walsh CT (1995) Cloning, overproduction, and characterization of the
Escherichia coli holo-acyl carrier protein synthase. J Biol Chem 270: 24658–24661.
Lee AH, Iwakoshi NN & Glimcher LH (2003) XBP-1 regulates a subset of endoplasmic
reticulum resident chaperone genes in the unfolded protein response. Mol Cell Biol 23:
7448–7459.
Lee AH, Scapa EF, Cohen DE & Glimcher LH (2008) Regulation of hepatic lipogenesis
by the transcription factor XBP1. Science 320: 1492–1496.
Lee FY, Lee H, Hubbert ML, Edwards PA & Zhang Y (2006) FXR, a multipurpose
nuclear receptor. Trends Biochem Sci 31: 572–580.
Lee Y, Yu X, Gonzales F, Mangelsdorf DJ, Wang MY, Richardson C, Witters LA &
Unger RH (2002) PPAR alpha is necessary for the lipopenic action of hyperleptinemia
on white adipose and liver tissue. Proc Natl Acad Sci USA 99: 11848–11853.
Leenders F, Adamski J, Husen B, Thole HH & Jungblut PW (1994) Molecular cloning and
amino acid sequence of the porcine 17 beta-estradiol dehydrogenase. Eur J Biochem
222: 221–227.
Lehman JJ, Barger PM, Kovacs A, Saffitz JE, Medeiros DM & Kelly DP (2000)
Peroxisome proliferator-activated receptor gamma coactivator-1 promotes cardiac
mitochondrial biogenesis. J Clin Invest 106: 847–856.
Lenski C, Kooy RF, Reyniers E, Loessner D, Wanders RJ, Winnepenninckx B, Hellebrand
H, Engert S, Schwartz CE, Meindl A & Ramser J (2007) The reduced expression of
the HADH2 protein causes X-linked mental retardation, choreoathetosis, and
abnormal behavior. Am J Hum Genet 80: 372–377.
Leonardi R, Zhang YM, Rock CO & Jackowski S (2005) Coenzyme A: back in action.
Prog Lipid Res 44: 125–153.
Leonardsson G, Steel JH, Christian M, Pocock V, Milligan S, Bell J, So PW, MedinaGomez G, Vidal-Puig A, White R & Parker MG (2004) Nuclear receptor corepressor
RIP140 regulates fat accumulation. Proc Natl Acad Sci USA 101: 8437–8442.
Lerouge P, Roche P, Faucher C, Maillet F, Truchet G, Prome JC & Denarie J (1990)
Symbiotic host-specificity of Rhizobium meliloti is determined by a sulphated and
acylated glucosamine oligosaccharide signal. Nature 344: 781–784.
Lin J, Puigserver P, Donovan J, Tarr P & Spiegelman BM (2002) Peroxisome proliferatoractivated receptor gamma coactivator 1beta (PGC-1beta ), a novel PGC-1-related
transcription coactivator associated with host cell factor. J Biol Chem 277: 1645–1648.
Liu B, Clanachan AS, Schulz R & Lopaschuk GD (1996) Cardiac efficiency is improved
after ischemia by altering both the source and fate of protons. Circ Res 79: 940–948.
72
Lodish H. BA, Matsudaira P, Kaiser CA, Krieger M, Scott MP, Zipursky, Darnell J. (2003)
Molecular Cell Biology. W.H. Freeman and Company, New York.
Ma L, Robinson LN & Towle HC (2006) ChREBP*Mlx is the principal mediator of
glucose-induced gene expression in the liver. J Biol Chem 281: 28721–28730.
Ma L, Tsatsos NG & Towle HC (2005) Direct role of ChREBP.Mlx in regulating hepatic
glucose-responsive genes. J Biol Chem 280: 12019–12027.
Maeda I, Kohara Y, Yamamoto M & Sugimoto A (2001) Large-scale analysis of gene
function in Caenorhabditis elegans by high-throughput RNAi. Curr Biol 11: 171–176.
Maier T, Jenni S & Ban N (2006) Architecture of mammalian fatty acid synthase at 4.5 A
resolution. Science 311: 1258–1262.
Maier T, Leibundgut M & Ban N (2008) The crystal structure of a mammalian fatty acid
synthase. Science 321: 1315–1322.
Marrakchi H, Zhang YM & Rock CO (2002) Mechanistic diversity and regulation of Type
II fatty acid synthesis. Biochem Soc Trans 30: 1050–1055.
Massengo-Tiasse RP & Cronan JE (2008) Vibrio cholerae FabV defines a new class of
enoyl-acyl carrier protein reductase. J Biol Chem 283: 1308–1316.
Masuda N, Yasumo H, Furusawa T, Tsukamoto T, Sadano H & Osumi T (1998) Nuclear
receptor binding factor-1 (NRBF-1), a protein interacting with a wide spectrum of
nuclear hormone receptors. Gene 221: 225–233.
Matsuo H, Chevallier J, Mayran N, Le Blanc I, Ferguson C, Faure J, Blanc NS, Matile S,
Dubochet J, Sadoul R, Parton RG, Vilbois F & Gruenberg J (2004) Role of LBPA and
Alix in multivesicular liposome formation and endosome organization. Science 303:
531–534.
McAllister RA, Fixter LM & Campbell EH (1988) The effect of tumour growth on liver
pantothenate, CoA, and fatty acid synthetase activity in the mouse. Br J Cancer 57:
83–86.
McKay SJ, Johnsen R, Khattra J, Asano J, Baillie DL, Chan S, Dube N, Fang L,
Goszczynski B, Ha E, Halfnight E, Hollebakken R, Huang P, Hung K, Jensen V,
Jones SJ, Kai H, Li D, Mah A, Marra M, McGhee J, Newbury R, Pouzyrev A, Riddle
DL, Sonnhammer E, Tian H, Tu D, Tyson JR, Vatcher G, Warner A, Wong K, Zhao Z
& Moerman DG (2003) Gene expression profiling of cells, tissues, and developmental
stages of the nematode C. elegans. Cold Spring Harb Symp Quant Biol 68: 159–169.
Meyer H & Meyer F (1978) Acyl-coenzyme A carboxylase of the free-living nematode
Turbatrix aceti. 2. Its catalytic properties and activation by monovalent cations.
Biochemistry 17: 1828–1833.
Michaud AL, Diau GY, Abril R & Brenna JT (2002) Double bond localization in minor
homoallylic fatty acid methyl esters using acetonitrile chemical ionization tandem
mass spectrometry. Anal Biochem 307: 348–360.
Miller AP & Willard HF (1998) Chromosomal basis of X chromosome inactivation:
identification of a multigene domain in Xp11.21-p11.22 that escapes X inactivation.
Proc Natl Acad Sci USA 95: 8709–8714.
73
Miller KF, Bolt DJ, Pursel VG, Hammer RE, Pinkert CA, Palmiter RD & Brinster RL
(1989) Expression of human or bovine growth hormone gene with a mouse
metallothionein-1 promoter in transgenic swine alters the secretion of porcine growth
hormone and insulin-like growth factor-I. J Endocrinol 120: 481–488.
Mishina M, Kamiryo T, Tanaka A, Fukui S & Numa S (1976) Acetyl-coenzyme-A
carboxylase of Candida Lipolytica. 1. Purification and properties of the enzyme. Eur J
Biochem 71: 295–300.
Moon YA & Horton JD (2003) Identification of two mammalian reductases involved in the
two-carbon fatty acyl elongation cascade. J Biol Chem 278: 7335–7343.
Mootha VK, Bunkenborg J, Olsen JV, Hjerrild M, Wisniewski JR, Stahl E, Bolouri MS,
Ray HN, Sihag S, Kamal M, Patterson N, Lander ES & Mann M (2003) Integrated
analysis of protein composition, tissue diversity, and gene regulation in mouse
mitochondria. Cell 115: 629–640.
Mostaqul Huq MD, Gupta P, Tsai NP, White R, Parker MG & Wei LN (2006) Suppression
of receptor interacting protein 140 repressive activity by protein arginine methylation.
EMBO J 25: 5094–5104.
Nascimben L, Ingwall JS, Pauletto P, Friedrich J, Gwathmey JK, Saks V, Pessina AC &
Allen PD (1996) Creatine kinase system in failing and nonfailing human myocardium.
Circulation 94: 1894–1901.
Nikolau BJ & Hawke JC (1984) Purification and characterization of maize leaf acetylcoenzyme A carboxylase. Arch Biochem Biophys 228: 86–96.
Nohturfft A, DeBose-Boyd RA, Scheek S, Goldstein JL & Brown MS (1999) Sterols
regulate cycling of SREBP cleavage-activating protein (SCAP) between endoplasmic
reticulum and Golgi. Proc Natl Acad Sci USA 96: 11235–11240.
Nordling E, Jornvall H & Persson B (2002) Medium-chain dehydrogenases/reductases
(MDR). Family characterizations including genome comparisons and active site
modeling. Eur J Biochem 269: 4267–4276.
Novikov D, Dieuaide-Noubhani M, Vermeesch JR, Fournier B, Mannaerts GP & Van
Veldhoven PP (1997) The human peroxisomal multifunctional protein involved in bile
acid synthesis: activity measurement, deficiency in Zellweger syndrome and
chromosome mapping. Biochim Biophys Acta 1360: 229–240.
O'Hara P, Slabas AR & Fawcett T (2000) Modulation of fatty acid biosynthesis by
antisense beta-keto reductase expression. Biochem Soc Trans 28: 613–615.
Ohlmeier S, Kastaniotis AJ, Hiltunen JK & Bergmann U (2004) The yeast mitochondrial
proteome, a study of fermentative and respiratory growth. J Biol Chem 279: 3956–
3979.
Oppermann U, Filling C, Hult M, Shafqat N, Wu X, Lindh M, Shafqat J, Nordling E,
Kallberg Y, Persson B & Jornvall H (2003) Short-chain dehydrogenases/reductases
(SDR): the 2002 update. Chem Biol Interact 143–144: 247–253.
Pagliarini DJ, Calvo SE, Chang B, Sheth SA, Vafai SB, Ong SE, Walford GA, Sugiana C,
Boneh A, Chen WK, Hill DE, Vidal M, Evans JG, Thorburn DR, Carr SA & Mootha
VK (2008) A mitochondrial protein compendium elucidates complex I disease
biology. Cell 134: 112–123.
74
Palmiter RD, Norstedt G, Gelinas RE, Hammer RE & Brinster RL (1983) Metallothioneinhuman GH fusion genes stimulate growth of mice. Science 222: 809–814.
Paul S, Gable K & Dunn TM (2007) A six-membrane-spanning topology for yeast and
Arabidopsis Tsc13p, the enoyl reductases of the microsomal fatty acid elongating
system. J Biol Chem 282: 19237–19246.
Peralba JM, Cederlund E, Crosas B, Moreno A, Julia P, Martinez SE, Persson B, Farr s J,
Pares X & Jornvall H (1999) Structural and enzymatic properties of a gastric
NADP(H)- dependent and retinal-active alcohol dehydrogenase. J Biol Chem 274:
26021–26026.
Podack ER, Lakomek M, Saathoff G & Seubert W (1974) On the mechanism and control
of the malonyl-CoA-dependent chain elongation of fatty acids. Characterization of
hexenoyl-CoA reductase from liver and adrenal cortex as a constituent of the
microsomal chain elongation. Eur J Biochem 45: 13–23.
Postic C, Dentin R, Denechaud PD & Girard J (2007) ChREBP, a transcriptional regulator
of glucose and lipid metabolism. Annu Rev Nutr 27: 179–192.
Powelka AM, Seth A, Virbasius JV, Kiskinis E, Nicoloro SM, Guilherme A, Tang X,
Straubhaar J, Cherniack AD, Parker MG & Czech MP (2006) Suppression of
oxidative metabolism and mitochondrial biogenesis by the transcriptional corepressor
RIP140 in mouse adipocytes. J Clin Invest 116: 125–136.
Price AC, Zhang YM, Rock CO & White SW (2001) Structure of beta-ketoacyl-[acyl
carrier protein] reductase from Escherichia coli: negative cooperativity and its
structural basis. Biochemistry 40: 12772–12781.
Prigge ST, He X, Gerena L, Waters NC & Reynolds KA (2003) The initiating steps of a
type II fatty acid synthase in Plasmodium falciparum are catalyzed by pfACP,
pfMCAT, and pfKASIII. Biochemistry 42: 1160–1169.
Puigserver P, Wu Z, Park CW, Graves R, Wright M & Spiegelman BM (1998) A coldinducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell 92:
829–839.
Rezaul K, Wu L, Mayya V, Hwang SI & Han D (2005) A systematic characterization of
mitochondrial proteome from human T leukemia cells. Mol Cell Proteomics 4: 169–
181.
Richardson P, McKenna W, Bristow M, Maisch B, Mautner B, O'Connell J, Olsen E,
Thiene G, Goodwin J, Gyarfas I, Martin I & Nordet P (1996) Report of the 1995
World Health Organization/International Society and Federation of Cardiology Task
Force on the Definition and Classification of cardiomyopathies. Circulation 93: 841–
842.
Riley KM, Dickson AC & Koeppen AH (1991) The origin of free brain malonate.
Neurochem Res 16: 117–122.
Rock CO & Cronan JE (1996) Escherichia coli as a model for the regulation of dissociable
(type II) fatty acid biosynthesis. Biochim Biophys Acta 1302: 1–16.
Rock CO & Cronan JE, Jr. (1979) Re-evaluation of the solution structure of acyl carrier
protein. J Biol Chem 254: 9778–9785.
75
Rock CO, Cronan JE, Jr. & Armitage IM (1981) Molecular properties of acyl carrier
protein derivatives. J Biol Chem 256: 2669–2674.
Romagnoli A, Aguiari P, De Stefani D, Leo S, Marchi S, Rimessi A, Zecchini E, Pinton P
& Rizzuto R (2007) Endoplasmic reticulum/mitochondria calcium cross-talk. Novartis
Found Symp 287: 122–131; discussion 129–131.
Ron D & Walter P (2007) Signal integration in the endoplasmic reticulum unfolded protein
response. Nat Rev Mol Cell Biol 8: 519–529.
Rosen ED & Spiegelman BM (2000) Molecular regulation of adipogenesis. Annu Rev Cell
Dev Biol 16: 145–171.
Rual JF, Ceron J, Koreth J, Hao T, Nicot AS, Hirozane-Kishikawa T, Vandenhaute J,
Orkin SH, Hill DE, van den Heuvel S & Vidal M (2004) Toward improving
Caenorhabditis elegans phenome mapping with an ORFeome-based RNAi library.
Genome Res 14: 2162–2168.
Runswick MJ, Fearnley IM, Skehel JM & Walker JE (1991) Presence of an acyl carrier
protein in NADH:ubiquinone oxidoreductase from bovine heart mitochondria. FEBS
Lett 286: 121–124.
Sacco E, Covarrubias AS, O'Hare H M, Carroll P, Eynard N, Jones TA, Parish T, Daffe M,
Backbro K & Quemard A (2007) The missing piece of the type II fatty acid synthase
system from Mycobacterium tuberculosis. Proc Natl Acad Sci USA 104: 14628–
14633.
Saggerson D (2008) Malonyl-CoA, a Key Signaling Molecule in Mammalian Cells. Annu
Rev Nutr 28: 253–272.
Saito J, Yamada M, Watanabe T, Iida M, Kitagawa H, Takahata S, Ozawa T, Takeuchi Y
& Ohsawa F (2008) Crystal structure of enoyl-acyl carrier protein reductase (FabK)
from Streptococcus pneumoniae reveals the binding mode of an inhibitor. Protein Sci
17: 691–699.
Salem N, Jr., Litman B, Kim HY & Gawrisch K (2001) Mechanisms of action of
docosahexaenoic acid in the nervous system. Lipids 36: 945–959.
Sasaki Y, Kozaki A & Hatano M (1997) Link between light and fatty acid synthesis:
thioredoxin-linked reductive activation of plastidic acetyl-CoA carboxylase. Proc Natl
Acad Sci USA 94: 11096–11101.
Scheffler IE (2001) A century of mitochondrial research: achievements and perspectives.
Mitochondrion 1: 3–31.
Schmitt J, Brocca S, Schmid RD & Pleiss J (2002) Blocking the tunnel: engineering of
Candida rugosa lipase mutants with short chain length specificity. Protein Eng 15:
595–601.
Schneider R, Brors B, Burger F, Camrath S & Weiss H (1997) Two genes of the putative
mitochondrial fatty acid synthase in the genome of Saccharomyces cerevisiae. Curr
Genet 32: 384–388.
Schneider R, Massow M, Lisowsky T & Weiss H (1995) Different respiratory-defective
phenotypes of Neurospora crassa and Saccharomyces cerevisiae after inactivation of
the gene encoding the mitochondrial acyl carrier protein. Curr Genet 29: 10–17.
76
Scholte HR, Luyt-Houwen IE, Dubelaar ML & Hulsmann WC (1986) The source of
malonyl-CoA in rat heart. The calcium paradox releases acetyl-CoA carboxylase and
not propionyl-CoA carboxylase. FEBS Lett 198: 47–50.
Schonauer MS, Kastaniotis AJ, Hiltunen JK & Dieckmann CL (2008) Intersection of RNA
Processing and the Type II Fatty Acid Synthesis Pathway in Yeast Mitochondria. Mol
Cell Biol 28: 6646–6657.
Schrader M & Yoon Y (2007) Mitochondria and peroxisomes: Are the 'Big Brother' and
the 'Little Sister' closer than assumed? Bioessays 29: 1105–1114.
Schrick JJ & Lingrel JB (2001) cDNA cloning, mapping and expression of the mouse
propionyl CoA carboxylase beta (pccb), the gene for human type II propionic
acidaemia. Gene 264: 147–152.
Schuller HJ, Fortsch B, Rautenstrauss B, Wolf DH & Schweizer E (1992) Differential
proteolytic sensitivity of yeast fatty acid synthetase subunits alpha and beta
contributing to a balanced ratio of both fatty acid synthetase components. Eur J
Biochem 203: 607–614.
Schulte U (2001) Biogenesis of respiratory complex I. J Bioenerg Biomembr 33: 205–212.
Schweizer E & Hofmann J (2004) Microbial type I fatty acid synthases (FAS): major
players in a network of cellular FAS systems. Microbiol Mol Biol Rev 68: 501–517.
Shaffer AL, Shapiro-Shelef M, Iwakoshi NN, Lee AH, Qian SB, Zhao H, Yu X, Yang L,
Tan BK, Rosenwald A, Hurt EM, Petroulakis E, Sonenberg N, Yewdell JW, Calame
K, Glimcher LH & Staudt LM (2004) XBP1, downstream of Blimp-1, expands the
secretory apparatus and other organelles, and increases protein synthesis in plasma
cell differentiation. Immunity 21: 81–93.
Shimano H, Shimomura I, Hammer RE, Herz J, Goldstein JL, Brown MS & Horton JD
(1997) Elevated levels of SREBP-2 and cholesterol synthesis in livers of mice
homozygous for a targeted disruption of the SREBP-1 gene. J Clin Invest 100: 2115–
2124.
Simmer F, Moorman C, van der Linden AM, Kuijk E, van den Berghe PV, Kamath RS,
Fraser AG, Ahringer J & Plasterk RH (2003) Genome-wide RNAi of C. elegans using
the hypersensitive rrf-3 strain reveals novel gene functions. PLoS Biol 1: E12.
Simon A, Hellman U, Wernstedt C & Eriksson U (1995) The retinal pigment epithelialspecific 11-cis retinol dehydrogenase belongs to the family of short chain alcohol
dehydrogenases. J Biol Chem 270: 1107–1112.
Slabas AR, Chase D, Nishida I, Murata N, Sidebottom C, Safford R, Sheldon PS, Kekwick
RG, Hardie DG & Mackintosh RW (1992) Molecular cloning of higher-plant
3-oxoacyl-(acyl carrier protein) reductase. Sequence identities with the nodG-gene
product of the nitrogen-fixing soil bacterium Rhizobium meliloti. Biochem J 283:
321–326.
Smiley IE, Koekoek R, Adams MJ & Rossmann MG (1971) The 5 A resolution structure
of an abortive ternary complex of lactate dehydrogenase and its comparison with the
apo-enzyme. J Mol Biol 55: 467–475.
Smith S (1994) The animal fatty acid synthase: one gene, one polypeptide, seven enzymes.
FASEB J 8: 1248–1259.
77
Sonnichsen B, Koski LB, Walsh A, Marschall P, Neumann B, Brehm M, Alleaume AM,
Artelt J, Bettencourt P, Cassin E, Hewitson M, Holz C, Khan M, Lazik S, Martin C,
Nitzsche B, Ruer M, Stamford J, Winzi M, Heinkel R, Roder M, Finell J, Hantsch H,
Jones SJ, Jones M, Piano F, Gunsalus KC, Oegema K, Gonczy P, Coulson A, Hyman
AA & Echeverri CJ (2005) Full-genome RNAi profiling of early embryogenesis in
Caenorhabditis elegans. Nature 434: 462–469.
Spaink HP, Sheeley DM, van Brussel AA, Glushka J, York WS, Tak T, Geiger O,
Kennedy EP, Reinhold VN & Lugtenberg BJ (1991) A novel highly unsaturated fatty
acid moiety of lipo-oligosaccharide signals determines host specificity of Rhizobium.
Nature 354: 125–130.
Stanley WC, Lopaschuk GD, Hall JL & McCormack JG (1997) Regulation of myocardial
carbohydrate metabolism under normal and ischaemic conditions. Potential for
pharmacological interventions. Cardiovasc Res 33: 243–257.
Stephens JL, Lee SH, Paul KS & Englund PT (2007) Mitochondrial fatty acid synthesis in
Trypanosoma brucei. J Biol Chem 282: 4427–4436.
Stinnett L, Lewin TM & Coleman RA (2007) Mutagenesis of rat acyl-CoA synthetase 4
indicates amino acids that contribute to fatty acid binding. Biochim Biophys Acta
1771: 119–125.
Stoeckman AK, Ma L & Towle HC (2004) Mlx is the functional heteromeric partner of the
carbohydrate response element-binding protein in glucose regulation of lipogenic
enzyme genes. J Biol Chem 279: 15662–15669.
Stuible HP, Meier S & Schweizer E (1997) Identification, isolation and biochemical
characterization of a phosphopantetheine:protein transferase that activates the two
type-I fatty acid synthases of Brevibacterium ammoniagenes. Eur J Biochem 248:
481–487.
Stuible HP, Meier S, Wagner C, Hannappel E & Schweizer E (1998) A novel
phosphopantetheine:protein transferase activating yeast mitochondrial acyl carrier
protein. J Biol Chem 273: 22334–22339.
Svensson S, Ostberg T, Jacobsson M, Norstrom C, Stefansson K, Hallen D, Johansson IC,
Zachrisson K, Ogg D & Jendeberg L (2003) Crystal structure of the heterodimeric
complex of LXRalpha and RXRbeta ligand-binding domains in a fully agonistic
conformation. EMBO J 22: 4625–4633.
Tabor DE, Kim JB, Spiegelman BM & Edwards PA (1999) Identification of conserved ciselements and transcription factors required for sterol-regulated transcription of
stearoyl-CoA desaturase 1 and 2. J Biol Chem 274: 20603–20610.
Taha M, Lopaschuk GD (2007) Alterations in energy metablism in cardiomyopathies.
Annals of medicine 39: 594–607.
Tahiliani AG & Beinlich CJ (1991) Pantothenic acid in health and disease. Vitam Horm 46:
165–228.
Tehlivets O, Scheuringer K & Kohlwein SD (2007) Fatty acid synthesis and elongation in
yeast. Biochim Biophys Acta 1771: 255–270.
78
Thampy KG (1989) Formation of malonyl coenzyme A in rat heart. Identification and
purification of an isozyme of A carboxylase from rat heart. J Biol Chem 264: 17631–
17634.
Tontonoz P, Hu E & Spiegelman BM (1994) Stimulation of adipogenesis in fibroblasts by
PPAR gamma 2, a lipid-activated transcription factor. Cell 79: 1147–1156.
Torkko JM, Koivuranta KT, Kastaniotis AJ, Airenne TT, Glumoff T, Ilves M, Hartig A,
Gurvitz A & Hiltunen JK (2003) Candida tropicalis expresses two mitochondrial
2-enoyl thioester reductases that are able to form both homodimers and heterodimers.
J Biol Chem 278: 41213–41220.
Torkko JM, Koivuranta KT, Miinalainen IJ, Yagi AI, Schmitz W, Kastaniotis AJ, Airenne
TT, Gurvitz A & Hiltunen KJ (2001) Candida tropicalis Etr1p and Saccharomyces
cerevisiae Ybr026p (Mrf1'p), 2-enoyl thioester reductases essential for mitochondrial
respiratory competence. Mol Cell Biol 21: 6243–6253.
Torroja L, Ortuno-Sahagun D, Ferrus A, Hammerle B & Barbas JA (1998) scully, an
essential gene of Drosophila, is homologous to mammalian mitochondrial type II
L-3-hydroxyacyl-CoA dehydrogenase/amyloid-beta peptide-binding protein. J Cell
Biol 141: 1009–1017.
Triepels R, Smeitink J, Loeffen J, Smeets R, Buskens C, Trijbels F & van den Heuvel L
(1999) The human nuclear-encoded acyl carrier subunit (NDUFAB1) of the
mitochondrial complex I in human pathology. J Inherit Metab Dis 22: 163–173.
Tsigelny I & Baker ME (1996) Structures important in NAD(P)(H) specificity for
mammalian retinol and 11-Cis-retinol dehydrogenases. Biochem Biophys Res
Commun 226: 118–127.
Tsukamoto Y, Wong H, Mattick JS & Wakil SJ (1983) The architecture of the animal fatty
acid synthetase complex. IV. Mapping of active centers and model for the mechanism
of action. J Biol Chem 258: 15312–15322.
Tuhackova Z & Hradec J (1985) The role of cholesteryl 14-methylhexadecanoate in the
function of eukaryotic peptide elongation factor 1. Eur J Biochem 146: 365–370.
Wada H, Shintani D & Ohlrogge J (1997) Why do mitochondria synthesize fatty acids?
Evidence for involvement in lipoic acid production. Proc Natl Acad Sci USA 94:
1591–1596.
van Meer G (2005) Cellular lipidomics. EMBO J 24: 3159–3165.
Wang YX, Lee CH, Tiep S, Yu RT, Ham J, Kang H & Evans RM (2003) Peroxisomeproliferator-activated receptor delta activates fat metabolism to prevent obesity. Cell
113: 159–170.
Wang YX, Zhang CL, Yu RT, Cho HK, Nelson MC, Bayuga-Ocampo CR, Ham J, Kang H
& Evans RM (2004) Regulation of muscle fiber type and running endurance by
PPARdelta. PLoS Biol 2: e294.
Ward CM, Mahler J, Maronpot RR, Sundberg JP, Frederichson RM (2000) Pathology of
genetically engineered mice. Iowa Sate University Press.
White R, Morganstein D, Christian M, Seth A, Herzog B & Parker MG (2008) Role of
RIP140 in metabolic tissues: connections to disease. FEBS Lett 582: 39–45.
79
White SW, Zheng J, Zhang YM & Rock (2005) The structural biology of type II fatty acid
biosynthesis. Annu Rev Biochem 74: 791–831.
Whittle E & Shanklin J (2001) Engineering delta 9-16:0-acyl carrier protein (ACP)
desaturase specificity based on combinatorial saturation mutagenesis and logical
redesign of the castor delta 9-18:0-ACP desaturase. J Biol Chem 276: 21500–21505.
Widmer J, Fassihi KS, Schlichter SC, Wheeler KS, Crute BE, King N, Nutile-McMenemy
N, Noll WW, Daniel S, Ha J, Kim KH & Witters LA (1996) Identification of a second
human acetyl-CoA carboxylase gene. Biochem J 316: 915–922.
Willy PJ, Umesono K, Ong ES, Evans RM, Heyman RA & Mangelsdorf DJ (1995) LXR, a
nuclear receptor that defines a distinct retinoid response pathway. Genes Dev 9: 1033–
1045.
Witkowski A, Joshi AK & Smith S (2007) Coupling of the de novo fatty acid biosynthesis
and lipoylation pathways in mammalian mitochondria. J Biol Chem 282: 14178–
14185.
Vivekananda J, Beck CF & Oliver DJ (1988) Monoclonal antibodies as tools in membrane
biochemistry. Identification and partial characterization of the dicarboxylate
transporter from pea leaf mitochondria. J Biol Chem 263: 4782–4788.
Vo N, Fjeld C & Goodman RH (2001) Acetylation of nuclear hormone receptor-interacting
protein RIP140 regulates binding of the transcriptional corepressor CtBP. Mol Cell
Biol 21: 6181–6188.
Voilley N, Roduit R, Vicaretti R, Bonny C, Waeber G, Dyck JR, Lopaschuk GD & Prentki
M (1999) Cloning and expression of rat pancreatic beta-cell malonyl-CoA
decarboxylase. Biochem J 340: 213–217.
von Figura K & Hasilik A (1986) Lysosomal enzymes and their receptors. Annu Rev
Biochem 55: 167–193.
Wu Z, Puigserver P, Andersson U, Zhang C, Adelmant G, Mootha V, Troy A, Cinti S,
Lowell B, Scarpulla RC & Spiegelman BM (1999) Mechanisms controlling
mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1.
Cell 98: 115–124.
Xu GY, Tam A, Lin L, Hixon J, Fritz CC & Powers R (2001) Solution structure of B.
subtilis acyl carrier protein. Structure 9: 277–287.
Xu HE, Lambert MH, Montana VG, Parks DJ, Blanchard SG, Brown PJ, Sternbach DD,
Lehmann JM, Wisely GB, Willson TM, Kliewer SA & Milburn MV (1999) Molecular
recognition of fatty acids by peroxisome proliferator-activated receptors. Mol Cell 3:
397–403.
Yamashita H, Takenoshita M, Sakurai M, Bruick RK, Henzel WJ, Shillinglaw W, Arnot D
& Uyeda K (2001) A glucose-responsive transcription factor that regulates
carbohydrate metabolism in the liver. Proc Natl Acad Sci USA 98: 9116–9121.
Yang J, Koga Y, Nakano H & Yamane T (2002) Modifying the chain-length selectivity of
the lipase from Burkholderia cepacia KWI-56 through in vitro combinatorial
mutagenesis in the substrate-binding site. Protein Eng 15: 147–152.
80
Yang Z, Liu S, Chen X, Chen H, Huang M & Zheng J (2000) Induction of apoptotic cell
death and in vivo growth inhibition of human cancer cells by a saturated branchedchain fatty acid, 13-methyltetradecanoic acid. Cancer Res 60: 505–509.
Yasuno R, von Wettstein-Knowles P & Wada H (2004) Identification and molecular
characterization of the beta-ketoacyl-[acyl carrier protein] synthase component of the
Arabidopsis mitochondrial fatty acid synthase. J Biol Chem 279: 8242–8251.
Yi X & Maeda N (2005) Endogenous production of lipoic acid is essential for mouse
development. Mol Cell Biol 25: 8387–8392.
Yu K, Bayona W, Kallen CB, Harding HP, Ravera CP, McMahon G, Brown M & Lazar
MA (1995) Differential activation of peroxisome proliferator-activated receptors by
eicosanoids. J Biol Chem 270: 23975–23983.
Yu S & Reddy JK (2007) Transcription coactivators for peroxisome proliferator-activated
receptors. Biochim Biophys Acta 1771: 936–951.
Zensen R, Husmann H, Schneider R, Peine T & Weiss H (1992) De novo synthesis and
desaturation of fatty acids at the mitochondrial acyl-carrier protein, a subunit of
NADH:ubiquinone oxidoreductase in Neurospora crassa. FEBS Lett 310: 179–181.
Zhang L, Joshi AK, Hofmann J, Schweizer E & Smith S (2005) Cloning, expression, and
characterization
of
the
human
mitochondrial
beta-ketoacyl
synthase.
Complementation of the yeast CEM1 knock-out strain. J Biol Chem 280: 12422–
12429.
Zhang L, Joshi AK & Smith S (2003a) Cloning, expression, characterization, and
interaction of two components of a human mitochondrial fatty acid synthase.
Malonyltransferase and acyl carrier protein. J Biol Chem 278: 40067–40074.
Zhang Q, Piston DW & Goodman RH (2002) Regulation of corepressor function by
nuclear NADH. Science 295: 1895–1897.
Zhang YM, Wu B, Zheng J & Rock CO (2003b) Key residues responsible for acyl carrier
protein and beta-ketoacyl-acyl carrier protein reductase (FabG) interaction. J Biol
Chem 278: 52935–52943.
81
82
Original articles
I
Miinalainen IJ, Chen ZJ, Torkko JM, Pirilä PL, Sormunen RT, Bergmann U, Qin YM
& Hiltunen JK (2003) Characterization of 2-enoyl thioester reductase from mammals
– an ortholog of Ybr026p/Mrf1’p of the yeast mitochondrial FAS type II. J Biol Chem
278: 20154–20161.
II Chen ZJ, Pudas R, Sharma S, Smart O, Juffer A, Hiltunen JK, Wierenga RK &
Haapalainen AM (2008) Structure and enzymology of 2-enoyl thioester reductase of
human FAS II: New insights into substrate binding and catalysis. J Mol Biol 379:
830–844.
III Chen ZJ, Leskinen H, Liimatta E, Sormunen RT, Miinalainen IJ, Hassinen IE &
Hiltunen JK (2008) Overexpression of Mecr, a gene of mitochondrial FAS II in heart
leads to cardiac dysfunction in mouse. Manuscript.
Reprinted with permission from the American Society for Biochemistry and
Molecular Biology (I) and Elsevier (II).
Original publications are not included in the electronic version of the dissertation.
83
84
ACTA UNIVERSITATIS OULUENSIS
SERIES A SCIENTIAE RERUM NATURALIUM
502.
Asikkala, Janne (2008) Application of ionic liquids and microwave activation in
selected organic reactions
503.
Rinta-aho, Marko (2008) On monomial exponential sums in certain index 2 cases
and their connections to coding theory
504.
Sallinen, Pirkko (2008) Myocardial infarction. Aspects relating to endogenous and
exogenous melatonin and cardiac contractility
505.
Xin, Weidong (2008) Continuum electrostatics of biomolecular systems
506.
Pyhäjärvi, Tanja (2008) Roles of demography and natural selection in molecular
evolution of trees, focus on Pinus sylvestris
507.
Kinnunen, Aulis (2008) A Palaeoproterozoic high-sulphidation epithermal gold
deposit at Orivesi, southern Finland
508.
Alahuhta, Markus (2008) Protein crystallography of triosephosphate isomerases:
functional and protein engineering studies
509.
Reif, Vitali (2008) Birds of prey and grouse in Finland. Do avian predators limit or
regulate their prey numbers?
510.
Saravesi, Karita (2008) Mycorrhizal responses to defoliation of woody hosts
511.
Risteli, Maija (2008) Substrate specificity of lysyl hydroxylase isoforms and
multifunctionality of lysyl hydroxylase 3
512.
Korsu, Kai (2008) Ecology and impacts of nonnative salmonids with special
reference to brook trout (Salvelinus fontinalis Mitchill) in North Europe
513.
Laitinen, Jarmo (2008) Vegetational and landscape level responses to water level
fluctuations in Finnish, mid-boreal aapa mire – aro wetland environments
514.
Viljakainen, Lumi (2008) Evolutionary genetics of immunity and infection in social
insects
515.
Hurme, Eija (2008) Ecological knowledge towards sustainable forest management.
Habitat requirements of the Siberian flying squirrel in Finland
516.
Kaukonen, Risto (2008) Sulfide-poor platinum-group element deposits. A
mineralogical approach with case studies and examples from the literature
517.
Rokka, Aare (2008) Solute traffic across the mammalian peroxisomal
membrane—the role of Pxmp2
518.
Sharma, Satyan (2008) Computational Studies on Prostatic Acid Phosphatase
Book orders:
OULU UNIVERSITY PRESS
P.O. Box 8200, FI-90014
University of Oulu, Finland
Distributed by
OULU UNIVERSITY LIBRARY
P.O. Box 7500, FI-90014
University of Oulu, Finland
A 519
OULU 2008
U N I V E R S I T Y O F O U L U P. O. B . 7 5 0 0 F I - 9 0 0 1 4 U N I V E R S I T Y O F O U L U F I N L A N D
U N I V E R S I TAT I S
S E R I E S
SCIENTIAE RERUM NATURALIUM
Professor Mikko Siponen
HUMANIORA
University Lecturer Elise Kärkkäinen
TECHNICA
Professor Hannu Heusala
MEDICA
Professor Olli Vuolteenaho
SCIENTIAE RERUM SOCIALIUM
ACTA
UN
NIIVVEERRSSIITTAT
ATIISS O
OU
ULLU
UEEN
NSSIISS
U
Zhijun Chen
E D I T O R S
Zhijun Chen
A
B
C
D
E
F
G
O U L U E N S I S
ACTA
A C TA
A 519
CHARACTERIZATION OF
THE 2-ENOYL THIOESTER
REDUCTASE OF
MITOCHONDRIAL FATTY
ACID SYNTHESIS TYPE II
IN MAMMALS
Senior Researcher Eila Estola
SCRIPTA ACADEMICA
Information officer Tiina Pistokoski
OECONOMICA
University Lecturer Seppo Eriksson
EDITOR IN CHIEF
Professor Olli Vuolteenaho
PUBLICATIONS EDITOR
Publications Editor Kirsti Nurkkala
ISBN 978-951-42-8979-8 (Paperback)
ISBN 978-951-42-8980-4 (PDF)
ISSN 0355-3191 (Print)
ISSN 1796-220X (Online)
FACULTY OF SCIENCE,
DEPARTMENT OF BIOCHEMISTRY, UNIVERSITY OF OULU;
BIOCENTER OULU,
UNIVERSITY OF OULU
A
SCIENTIAE RERUM
RERUM
SCIENTIAE
NATURALIUM
NATURALIUM