ArticlePDF Available

Uncoupling protein 3 transcription is regulated by peroxisome proliferator-activated receptor α in the adult rodent heart

Authors:

Abstract and Figures

Relatively little is known concerning the regulation of uncoupling proteins (UCPs) in the heart. We investigated in the adult rodent heart 1) whether changes in workload, substrate supply, or cytokine (TNF-alpha) administration affect UCP-2 and UCP-3 expression, and 2) whether peroxisome proliferator-activated receptor alpha (PPARalpha) regulates the expression of either UCP-2 or UCP-3. Direct comparisons were made between cardiac and skeletal muscle. UCP-2, UCP-3, and PPARalpha expression were reduced when cardiac workload was either increased (pressure overload by aortic constriction) or decreased (mechanical unloading by heterotopic transplantation). Similar results were observed during cytokine administration. Reduced dietary fatty acid availability resulted in decreased expression of both cardiac UCP-2 and UCP-3. However, when fatty acid (the natural ligand for PPARalpha) supply was increased (high-fat feeding, fasting, and STZ-induced diabetes), cardiac UCP-3 but not UCP-2 expression increased. Comparable results were observed in rats treated with the specific PPARalpha agonist WY-14,643. The level of cardiac UCP-3 but not UCP-2 expression was severely reduced (20-fold) in PPARalpha-/- mice compared to wild-type mice. These results suggest that in the adult rodent heart, UCP-3 expression is regulated by PPARalpha. In contrast, cardiac UCP-2 expression is regulated in part by a fatty acid-dependent, PPARalpha-independent mechanism.
Content may be subject to copyright.
Uncoupling protein 3 transcription is regulated by
peroxisome proliferator-activated receptor in the
adult rodent heart
MARTIN E. YOUNG, SARITA PATIL, JUN YING, CHRISTOPHE DEPRE,
HARLEEN SINGH AHUJA,* GREGORY L. SHIPLEY,* STANISLAW M. STEPKOWSKI,
PETER J. A. DAVIES,* AND HEINRICH TAEGTMEYER
1
Division of Cardiology, *Department of Integrative Biology, and the
Division of Organ
Transplantation, University of Texas Houston Medical Center, Houston, Texas 77030, USA
ABSTRACT Relatively little is known concerning the
regulation of uncoupling proteins (UCPs) in the heart.
We investigated in the adult rodent heart 1) whether
changes in workload, substrate supply, or cytokine
(TNF-) administration affect UCP-2 and UCP-3 ex-
pression, and 2) whether peroxisome proliferator-acti-
vated receptor (PPAR) regulates the expression of
either UCP-2 or UCP-3. Direct comparisons were made
between cardiac and skeletal muscle. UCP-2, UCP-3,
and PPAR expression were reduced when cardiac
workload was either increased (pressure overload by
aortic constriction) or decreased (mechanical unload-
ing by heterotopic transplantation). Similar results were
observed during cytokine administration. Reduced di-
etary fatty acid availability resulted in decreased expres-
sion of both cardiac UCP-2 and UCP-3. However, when
fatty acid (the natural ligand for PPAR) supply was
increased (high-fat feeding, fasting, and STZ-induced
diabetes), cardiac UCP-3 but not UCP-2 expression
increased. Comparable results were observed in rats
treated with the specific PPAR agonist WY-14,643.
The level of cardiac UCP-3 but not UCP-2 expression
was severely reduced (20-fold) in PPAR
/
mice
compared to wild-type mice. These results suggest that
in the adult rodent heart, UCP-3 expression is regu-
lated by PPAR. In contrast, cardiac UCP-2 expression
is regulated in part by a fatty acid-dependent, PPAR-
independent mechanism.—Young, M. E., Patil, S.,
Ying, J., Depre, C., Ahuja, H. S., Shipley, G. L.,
Stepkowski, S. M., Davies, P. J. A., Taegtmeyer H.
Uncoupling protein 3 transcription is regulated by
peroxisome proliferator-activated receptor in the
adult rodent heart. FASEB J. 15, 833–845 (2001)
Key Words: diabetes fasting fatty acids hypertrophy un-
loading
Very little is known about either the function or the
regulation of uncoupling proteins (UCPs) in the adult
heart. There are three known members of the family of
UCPs. Thermogenin, also known as UCP-1, was first
discovered in brown adipose tissue as the principal
mediator of nonshivering thermogenesis (1). UCP-1 is
a transmembrane protein found in the inner mitochon-
drial membrane, where it uncouples ATP synthesis and
mitochondrial electron transport, dissipating the en-
ergy as heat (2). More recently, two genes have been
described with high sequence homology to UCP-1—
UCP-2 and UCP-3 (3–5)—both of which have been
shown to possess uncoupling activity in cells (6). UCP-2
is ubiquitously expressed whereas UCP-3 is highly ex-
pressed in skeletal muscle, adipose tissue, and, to a
lesser extent, the heart. Although much research has
focussed on UCPs in skeletal muscle, the mechanism(s)
by which cardiac UCP expression alter(s) require elu-
cidation.
There are four primary hypotheses concerning the
physiological roles of UCP-2 and UCP-3 (7): thermo-
genesis, regulation of fatty acid oxidation, regulation of
ATP synthesis, and reduction of mitochondrial reactive
oxygen species (ROS) generation. Whether UCPs in
the heart play a role in one or several of the above
mentioned functions is unknown. The roles of UCPs in
tissues, such as skeletal muscle, as well as white and
brown adipocytes have been investigated by determin-
ing the changes in gene expression of the UCPs in
response to various stimuli. For example, increases in
plasma free fatty acid levels result in increased skeletal
muscle UCP expression, suggesting a role in fatty acid
utilization and/or prevention of lipotoxicity and insu-
lin resistance (7–9). Thus, determining UCP expres-
sion in the heart in response to altered workload (in
which ATP demand, substrate utilization, and ROS
generation are altered) and/or substrate availability
might aid an understanding of the role of cardiac
UCPs. In addition, tumor necrosis factor (TNF-) has
been shown to increase skeletal muscle UCP expres-
sion, suggesting a role in cytokine-induced thermogen-
esis (10). TNF- levels are known to increase in the
failing heart(11), which has also been described as
‘energy starved’ (12). Whether increased TNF- levels
result in increased UCP expression and whether the
1
Correspondence: Division of Cardiology,, University of
Texas Houston Medical School, 6431 Fannin, MSB 1.246,
Houston, TX 77030, USA. E-mail: ht@heart.med.uth.tmc.edu
8330892-6638/01/0015-0833 © FASEB
latter affects the efficiency of the failing heart are
unknown.
The purpose of the present study was twofold. First,
we wanted to investigate both physiological and patho-
physiologic situations under which cardiac UCP-2 and
UCP-3 expression might be altered. These included
increased and decreased workload and altered sub-
strate availability (e.g., fasting/refeeding, altered di-
etary fatty acid composition, streptozotocin-induced
diabetes), as well as cytokine (TNF-) challenge. Sec-
ond, we were curious about the potential role of
peroxisome proliferator-activated receptor (PPAR,a
nuclear transcription factor that has been suggested to
play a role in skeletal muscle and adipocyte UCP
expression) in the changes observed in UCP expres-
sion. We 1) measured PPAR expression under condi-
tions in which UCP expression altered; 2) determined
the effects of specific activation of PPAR in vivo, with
WY-14,643, on UCP expression; and 3) measured the
level of UCP expression in PPAR
/
mice. Where
possible, we made direct comparisons between cardiac
and skeletal muscle to determine the similarities and
differences of UCP control in these two tissues. The
results show that during increased availability of fatty
acids, the heart increases the expression of UCP-3 only,
with no effect on cardiac UCP-2 expression. In contrast,
in skeletal muscle, the expression of both UCP-2 and
UCP-3 is increased with PPAR activation. UCP-3, but
not UCP-2, expression was severely reduced in the
heart of PPAR
/
mice compared to wild-type mice.
Together, these findings strongly implicate PPAR as
the major regulator of UCP-3, but not UCP-2, expres-
sion in the adult rodent heart. During mechanical
unloading and pressure overloading of the heart,
UCP-2, UCP-3, and PPAR expression all decrease. The
results provide new hypotheses for the roles of UCPs in
the adult heart.
MATERIALS AND METHODS
Animals
Male Sprague-Dawley rats (200–225 g initial weight) were
kept in the Animal Care Center of the University of Texas
Houston Medical School under controlled conditions
(231°C; 12 h light/12 h dark cycle), and received standard
laboratory chow and water ad libitum unless otherwise stated.
Changes in workload
In the first series of experiments, cardiac unloading was
induced by heterotopic transplantation of a rat heart into the
abdomen of a recipient rat, as described earlier (13, 14). After
2 wk, the animals were anesthetized and both donor (unload-
ed) and recipient (control) hearts were removed, freeze
clamped, and stored at 80°C prior to RNA extraction.
In the second series of experiments, cardiac pressure
overload was induced by banding the ascending aorta, with a
20-gauge needle, as described previously (14, 15). In control
animals, sham operations were performed without banding of
the aorta. Either 7 or 9 days after aortic constriction (to be
specified), the animals were anesthetized, hearts were re-
moved, freeze-clamped, and stored at 80°C.
Changes in substrate availability
In the first set of experiments, rats were fed either a high-
carbohydrate/low-fat (HC/LF) diet or a low-carbohydrate/
high-fat (LC/HF) diet (Purina Mills). These isocaloric diets
varied only in the proportion of energy obtained from
carbohydrate and fat. The contribution of carbohydrate, fat
and protein to total energy available were 71%, 6% and 23%
for the HC/LF diet, and 24%, 53% and 23% for the LC/HF
diet, respectively. The source of carbohydrate was a combina-
tion of sucrose and dextrin, while the source of fat was a
combination of corn oil and lard. Nonnutritive fiber was also
increased in the LC/HF diet. The length of time in which the
rats were fed the special diets is specified for individual
experiments. In selected experiments, soleus (skeletal) mus-
cle was removed in addition to the heart.
The effects of fasting and refeeding on cardiac gene
expression were investigated. Rats were fasted for either 1 or
2 days, after which the heart and soleus muscles were isolated.
A subset of fasted rats were refed with the HC/LF diet for an
additional 4 days.
Diabetes was induced through a single injection of strep-
tozotocin [STZ; 55 mg/kg intravenous (i.v.)]. Control ani-
mals were administered with buffer (Hank’s buffer; Life
Technologies, Inc., Grand Island, N.Y.) only. Five, 7, 14, or
182 days (6 months) after the initial injection, the animals
were anesthetized, hearts were removed, freeze-clamped, and
stored at
80°C. Animals were considered diabetic if their
blood glucose level was greater than 300 mg per deciliter.
Pharmacological interventions
To test the effects of specific PPAR activation, WY-14,643 was
added to standard powdered Purina rodent chow at a con-
centration of 0.01% (w/w). Rats were fed a WY-14,643-
containing diet for 4 days. Control animals received powered
rodent chow only.
In a separate set of experiments, rats received a single i.v.
(tail vein) injection with TNF- (human recombinant, 30
g/kg body weight in 1 ml 0.9% NaCl); controls were
injected with NaCl only. Twelve hours later, heart and soleus
muscles were removed. As cytokine administration decreases
rodent food intake (which can potentially affect gene expres-
sion), rats were injected at 07.00 h. During the following 12 h
period in the light, rodent ingestion is minimal. To ensure
standardized experimental conditions, food was withdrawn
from all animals at the time of injection.
PPAR
/
mice
Isolated RNA from hearts of age matched wild-type and
PPAR
/
mice was a kind gift from Dr L. Nagy (The Salk
Institute for Biological Studies, Gene Expression Laboratory,
La Jolla, Calif.).
RNA extraction and quantitative reverse transcription-
polymerase chain reaction (RT-PCR)
RNA extraction and quantitative RT-PCR of samples was
performed using previously described methods well estab-
lished in our laboratory (14, 16–18). Specific quantitative
assays were designed from the rat sequences available in
GenBank (Table 1). Primers and probes were designed from
unconserved sequences of the genes (allowing for isoform
834 Vol. 15 March 2001 YOUNG ET AL.The FASEB Journal
specificity). The correlation between C
t
(the number of PCR
cycles required for the fluorescent signal to reach a detection
threshold) and the amount of standard was linear overa5log
range of RNA for all assays (Fig. 1 illustrates the values for
rat/mouse UCP-2 and rat UCP-3). The level of transcripts for
the constitutive housekeeping gene product cyclophilin was
quantitatively measured in each sample to control for sample-
to-sample differences in RNA concentration. PCR data are
reported as the number of transcripts per number of cyclo-
philin molecules.
Statistical analysis
Data are presented as the mean se. Statistically significant
differences between groups were calculated by the Student’s
t test. A value of P0.05 was considered significant.
RESULTS
Mechanical unloading and pressure overload both
decrease cardiac UCP expression
Decreased workload results in significant atrophy over
the course of 2 wk, at which time there is a 55%
decrease in the heart weight-to-body weight ratio (14).
The expression of UCP-2 and UCP-3 were decreased in
unloaded hearts (Fig. 2A, B). Pressure overload re-
sulted in cardiac hypertrophy (after 1 wk the heart
weight to body weight ratios were 3.420.06 vs.
2.980.05 in the experimental and control groups;
P0.05). Hypertrophy also decreased the expression of
UCP-2 and UCP-3 (Fig. 2C, D). In all hearts, levels of
UCP-2 expression were 100-fold higher compared to
UCP-3 (Fig. 2).
Mechanical unloading decreases cardiac PPAR
expression
A possible mechanism by which reduced cardiac work-
load results in decreased UCP expression is a decrease
in the level of PPAR expression. Unloading reduced
cardiac PPAR expression by half (0.1370.027 vs.
0.2820.018 in experimental and control groups;
P0.05).
TABLE 1. Primer and probe sequences used in the quantitative PCR for UCP-2, UCP-3, PPAR, PDK-2, PDK-4, muscle CPT-I, iNOS,
and cyclophilin
Gene Primer/probe Sequence
r/m UCP-2
a
Forward 5-TCATCAAAGATACTCTCCTGAAAGC-3
Reverse 5-TGACGGTGGTGCAGAAGC-3
Probe 5-FAM-TGACAGACGACCTCCCTTGCCACT-TAMRA-3
r UCP-3 Forward 5-GTGACCTATGACATCATCAAGGA-3
Reverse 5-GCTCCAAAGGCAGAGACAAAG-3
Probe 5-FAM-CTGGACTCTCACCTGTTCACTGACAACTTCC-TAMRA-3
m UCP-3 Forward 5-TGCTGAGATGGTGACCTACGA-3
Reverse 5-CCAAAGGCAGAGACAAAGTGA-3
Probe 5-FAM-AAGTTGTCAGTAAACAGGTGAGACTCCAGCAA-TAMRA-3
r PPAR Forward 5-ACTACGGAGTTCACGCATGTG-3
Reverse 5-TTGTCGTACACCAGCTTCAGC-3
Probe 5-FAM-AGGCTGTAAGGGCTTCTTTCGGCG-TAMRA-3
r PDK-2 Forward 5-TCAGCAAGTTCTCCCCGTC-3
Reverse 5-ATGAAGTTTTCTCGCAGGCA-3
Probe 5-FAM-TGCTGGATCCGAAGTCTAGAAACTGCTTCAT-TAMRA-3
r PDK-4 Forward 5-TTCACACCTTCACCACATGC-3
Reverse 5-AAAGGGCGGTTTTCTTGATG-3
Probe 5-FAM-CGTGGCCCTCATGGCATTCTTG-TAMRA-3
r muscle CPT-I Forward 5-ATCATGTATCGCCGCAAACT-3
Reverse 5-ACCCATGTGCTCCTACCAGAT-3
Probe 5-FAM-TCAAGCCGGTAATGGCACTGGG-TAMRA-3
r iNOS Forward 5-GAGAAGCTGAGGCCCAGG-3
Reverse 5-ACCTTCCGCATTAGCACAGA-3
Probe 5-FAM-CAGTCTTGGTGAAAGCGGTGTTCTTTG-TAMRA-3
r/m Cyclophilin Forward 5-CTGATGGCGAGCCCTTG-3
Reverse 5-TCTGCTGTCTTTGGAACTTTGTC-3
Probe 5-FAM-CGCGTCTGCTTCGAGCTGTTTGCA-TAMRA-3
a
r denotes a rat-specific assay; m denotes a mouse-specific assay; r/m denotes a rat- and mouse-compatible assay.
Figure 1. Sensitivity of the quantitative assays for UCP-2 and
UCP-3. Each graph shows the C
t
obtained with various
amounts of standard RNA molecules. The relation between
the number of standard molecules and the C
t
was linear over
the range investigated (from 10
3
to 10
7
).
835CARDIAC UCP GENE EXPRESSION
Pressure overload induced changes in UCP
expression are dependent on dietary fatty acids
The involvement of PPAR in decreased UCP expres-
sion during pressure overload was investigated in two
ways. First, we measured the expression of PPAR
during overloading. Second, we determined whether
the availability of the natural ligand for PPAR (fatty
acids) affected pressure overload induced alterations in
UCP expression. Therefore, rats were fed one of two
diets: either a high-carbohydrate/low-fat (HC/LF) diet
or a low-carbohydrate/high-fat (LC/HF) diet. On day 7
after the initiation of the special diets, half of the rats
underwent aortic banding, whereas the other half were
operated without banding. Rats were maintained on
their specific diets for 9 days after surgery, then the
hearts were isolated. Aortic banding resulted in similar
degrees of cardiac hypertrophy in both groups (Table
2).
Hearts isolated from sham-operated rats fed the
LC/HF diet compared to sham-operated rats fed the
HC/LF diet possessed significantly higher levels of both
UCP-2 and UCP-3 expression (Table 2). However, the
fold induction of UCP-3 was far greater than that of
UCP-2. As observed during standard laboratory chow
feeding, banding significantly reduced the expression
of both UCP-2 and UCP-3 when rats were fed the
LC/HF diet (Table 2). In contrast, pressure overload
did not decrease the cardiac expression of either UCP-2
or UCP-3 for rats fed the HC/LF diet (Table 2).
Banding resulted in a significant decrease in PPAR
expression for rats fed either diet (Table 2). Further-
more, rats fed the HC/LF diet showed significantly
higher cardiac PPARexpression compared to rats fed
the LC/HF diet (Table 2).
Increased dietary fat increases cardiac UCP-3, but not
UCP-2, expression; heart vs. skeletal muscle
To investigate the time course over which cardiac UCP
expression changes, rats were fed the LC/HF diet for
various lengths of time (1, 2, 4, or 8 days), after which
the expression of cardiac UCP-2 and –3 as well as
PPAR were measured (Fig. 3). At all time points,
feeding with the LC/HF diet had no effect on cardiac
UCP-2 expression (Fig. 3A). In contrast, UCP-3 expres-
sion was rapidly induced by the LC/HF diet (within
24 h), and this induction was maintained (Fig. 3B). In
the case of PPAR, the LC/HF diet repressed expres-
sion, an effect that was partially normalized after 8 days
of continuous feeding (Fig. 3C).
We also measured UCP and PPAR expression in
soleus muscle (Fig. 3D–F). Feeding of rats with the
LC/HF diet resulted in a rapid increase in soleus
muscle UCP-2 and UCP-3 expression (Fig. 3D, E). This
increase was maintained for the full duration of the
feeding study. PPAR expression was not affected by
the LC/HF diet (Fig. 3F).
Figure 2. UCP-2 and UCP-3 expression during cardiac un-
loading and overloading. Altered expression of UCP-2 and
UCP-3 in unloaded (A, B) and overloaded (C, D) hearts.
Values are shown as the mean se for 9 or 10 separate
observations. All values are normalized against the expression
of the housekeeping gene cyclophilin. *P 0.05 vs. control.
TABLE 2. Effects of banding and altered dietary fatty acid content on body weight (BW), heart weight (HW), hypertrophy (HW/BW
ratio), and expression of UCP-2, UCP-3, and PPAR
HC/LF sham HC/LF banded LC/HF sham LC/HF banded
Body weight (g) 255 7 257 6 267 6 262 6
Heart weight (g) 0.81 0.03 0.99 0.09* 0.85 0.02 1.02 0.07
$
HW/BW
a
3.16 0.08 3.91 0.24* 3.17 0.07 3.90 0.13
$
UCP-2/cyclo 0.278 0.024 0.306 0.021 0.360 0.018* 0.285 0.019
$
UCP-3/cyclo
b
0.214 0.041 0.345 0.061 1.147 0.215*** 0.719 0.115
$
PPAR/cyclo 0.146 0.007 0.106 0.014* 0.092 0.006** 0.074 0.004***
$
a
HW/BW ratio is multiplied by a factor of 1000.
b
UCP-3/cyclo ratio is multiplied by a factor of 100. The table shows the body weight (BW), heart weight (BW), and heart weight to body
weight ratio (HW/BW; a marker of cardiac hypertrophy) for sham operated and banded animals fed either a high-carbohydrate/low-fat
(HC/LF) diet or a low-carbohydrate/high-fat (LC/HF) diet, as well as the expression of UCP-2, UCP-3, and PPAR. Duration of diet feeding
was a total of 16 days (7 days before and 9 days after surgery). Values are shown as the mean se for 5–10 observations. All gene expression
values are normalized against the expression of the housekeeping gene cyclophilin (cyclo).
* P 0.05, ** P 0.01 and *** P 0.001 vs. HC/LF control.
$
P 0.05 vs. LC/HF control.
836 Vol. 15 March 2001 YOUNG ET AL.The FASEB Journal
Fasting and refeeding modulates cardiac UCP-3, but
not UCP-2, expression; heart vs. skeletal muscle
Rats were fasted for either 1 or 2 days, after which a
subset was refed for an additional 4 days on the HC/LF
diet. Fasting had no effect on cardiac UCP-2 expres-
sion, although refeeding with the HC/LF diet resulted
in a significant decrease in expression (Fig. 4A.In
contrast, fasting significantly increased UCP-3 expres-
sion by threefold within 24 h (Fig. 4B). This increased
UCP-3 expression was severely blunted when fasting
continued for 2 days (Fig. 4B). Refeeding returned
cardiac UCP-3 expression to normal levels (Fig. 4B).
Fasting for either 1 or 2 days significantly lowered the
expression of cardiac PPAR, an effect that was re-
versed on refeeding (Fig. 4C).
To investigate further differences in UCP regulation
between cardiac and skeletal muscle, we also deter-
mined the effects of fasting and refeeding on soleus
muscle UCP expression. Fasting resulted in significant
increases in UCP-2 and -3 expression (Fig. 4D, E,
respectively). In the case of UCP-3, this increase ob-
served after 1 day was severely reduced at the second
day of fasting, a result similar to that observed in the
heart (Fig. 4E, B). Refeeding normalized soleus muscle
UCP (both -2 and -3) expression (Fig. 4D, E, respective-
ly). Soleus muscle PPAR expression was not affected
by either fasting or refeeding (Fig. 4F).
STZ-induced diabetes increases cardiac UCP-3
expression
STZ-induction of diabetes through pancreatic -cell
destruction resulted in significant elevations in plasma
glucose levels at 5 (3.12-fold; P0.001), 7 (4.45-fold;
P0.001), 14 (3.18-fold; P0.001), and 182 (5.00-fold;
P0.001) days after STZ injection (compared to age-
matched controls). UCP-2 expression in the heart was
not affected by STZ-induced diabetes (Fig. 5A). How-
ever, cardiac UCP-3 expression increased rapidly in
STZ-induced diabetes and remained elevated at all time
points investigated (Fig. 5B). The highest fold induc-
tion in cardiac UCP-3 expression occurred 14 days after
the initial STZ injection (6.4-fold). Despite a mainte-
nance in the induction of UCP-3 at day 182 (4.6-fold),
there was a substantial decrease in the absolute level of
cardiac UCP-3 expression in both control and STZ
diabetic animals (Fig. 5B). Although there was no
significant difference in cardiac UCP-2 expression with
respect to time, there was a trend for older animals to
show reduced expression (Fig. 5A). Thus, cardiac UCP
expression declined with age.
We also investigated the effects of STZ-induced dia-
betes on cardiac PPARgene expression. After either 5
or 7 days of diabetes, there is no effect on PPAR
expression (Fig. 5C). After 14 days of diabetes there is
a decrease in PPARexpression, although this effect is
Figure 3. Time course for altered cardiac and soleus muscle UCP-2, UCP-3, and PPARexpression during increased dietary fat
content. The time course for altered UCP-2, UCP-3, and PPAR expression in cardiac (A—C) and soleus (D—F) muscle in
response to feeding on the low-carbohydrate/high-fat (LC/HF) diet. Values are shown as the mean se for five separate
observations. All values are normalized against the expression of the housekeeping gene cyclophilin. *P0.05, **P0.01, and
***P0.001 vs. control (day zero).
837CARDIAC UCP GENE EXPRESSION
not significant (Fig. 5C). However, there is a significant
decrease in PPAR expression at 182 days after the
induction of diabetes (Fig. 5C). Age alone had no effect
on cardiac PPAR expression (Fig. 5C).
The PPAR agonist WY-14,643 alters heart and soleus
muscle gene expression
Rats were fed either control diet or diet containing the
PPAR agonist WY-14,643 (0.01% w/w) for 4 days, after
which cardiac and soleus muscle UCP-2, UCP-3,
PPAR, PDK-2, PDK-4, and muscle CPT-I expression
were measured. WY-14,643 caused an increase in soleus
muscle UCP-2 expression, with no effect on cardiac
UCP-2 expression (Fig. 6A). Both cardiac and soleus
muscle UCP-3 expression was increased by WY-14,643
(3.5- and 13.4-fold, respectively; Fig. 6B). PPAR ex-
pression was not affected by WY-14,643 in either heart
or soleus muscle (Fig. 6C). To determine the effective-
ness of WY-14,643 feeding, we measured pyruvate de-
hydrogenase kinase-4 (PDK-4) and muscle-specific car-
nitine palmitoyltransferase-I (CPT-I) expression, genes
known to be induced by PPAR activation (19, 20).
PDK-4 and muscle CPT-I expression increased in re-
sponse to WY-14,643 feeding in both heart and soleus
muscle (Fig. 6C, F). PDK-2, whose expression is not
believed to be regulated by PPAR (19), was not
affected by WY-14,643 (Fig. 6D).
TNF- decreases cardiac UCP-2, UCP-3, and PPAR
expression
We investigated whether cytokine exposure alters car-
diac gene expression by injecting rats with TNF-.
TNF- administration (30 g/kg; i.v.) resulted in a
significant decrease in cardiac PPAR expression after
12h(Fig. 7C). Reduced PPARexpression in response
to TNF- was accompanied by decreased cardiac UCP-2
and UCP-3 expression (Figs. 7A, B). Similarly, TNF-
administration resulted in decreased soleus muscle
UCP-3 and PPARexpression (Fig. 7B, C). In contrast,
soleus muscle UCP-2 expression increased (Fig. 7A). To
determine the effectiveness of TNF- administration,
cardiac and soleus muscle inducible nitric oxide syn-
Figure 4. Effects of fasting and refeeding on cardiac and soleus muscle UCP-2, UCP-3, and PPAR expression. The effects of
fasting (either 1 or 2 days) and refeeding on UCP-2, UCP-3, and PPAR expression in cardiac (A—C) and soleus (D—F) muscle.
Animals were refed the high-carbohydrate/low-fat (HC/LF) diet. Values are shown as the mean se for four or five separate
observations. All values are normalized against the expression of the housekeeping gene cyclophilin. *P0.05, **P0.01, and
***P0.001 vs. control (day zero).
838 Vol. 15 March 2001 YOUNG ET AL.The FASEB Journal
thase (iNOS) expression was also measured. TNF-
increased both cardiac and soleus muscle iNOS expres-
sion to similar extents (Fig. 7D).
Genetic mutation of the PPAR gene decreases
expression of cardiac UCP-3, but not UCP-2
We investigated whether PPAR was essential for the
expression of cardiac UCP-2 and UCP-3 by comparing
wild-type and PPAR
/
mouse hearts. The level of
cardiac UCP-2 and UCP-3 in wild-type mice were rela-
tively similar (Table 3). There was a 20-fold lower level
of cardiac UCP-3 expression in PPAR
/
mice com
-
pared to wild-type hearts (Table 3). No differences were
observed in the level of UCP-2 expression in hearts
isolated from PPAR
/
mice compared to those iso
-
lated from wild-type mice (Table 3).
DISCUSSION
We show that cardiac UCP-2 and UCP-3 expression
changes in response to altered physiological and patho-
physiologic states; in the case of UCP-3, these changes
are regulated by PPAR. In contrast to soleus muscle,
cardiac UCP-2 expression does not significantly change
in response to high-fat feeding, fasting, diabetes, or
WY-14,643 treatment, all of which lead to PPAR acti-
vation. However, UCP-3 expression increases in cardiac
and soleus muscle under the same conditions. Expres-
sion of both UCP-3 and PPAR decrease during cardiac
mechanical unloading and pressure overloading. Mu-
tation of the PPAR gene results in a near complete
inhibition of cardiac UCP-3 expression, with no effect
on UCP-2 expression. These results suggest that in the
adult rodent heart, UCP-3 expression is regulated by
PPAR whereas UCP-2 expression is not.
Cardiac UCP-2 expression decreased during mechan-
ical unloading and pressure overloading. Furthermore,
decreased UCP-2 expression during pressure overload
was dependent on the presence of dietary fatty acids.
Decreased dietary fatty acid intake alone reduced car-
diac UCP-2 and UCP-3 expression. These results sug-
gest that UCP-2 expression in the heart is regulated, at
least in part, by a fatty acid-dependent, PPAR-indepen-
Figure 5. Time course for altered cardiac UCP-2, UCP-3, and
PPAR expression by streptozotocin-induced diabetes. The
effects of streptozotocin (STZ; 55 mg/kg i.v.) -induced dia-
betes on cardiac UCP-2 (A), UCP-3 (B), and PPAR (C)
expression either 5, 7, 14, or 182 days after the initial
injection. Values are shown as the mean se for five separate
observations. All values are normalized against the expression
of the housekeeping gene cyclophilin. *P 0.05, **P 0.01,
and ***P 0.001 vs. age-matched controls.
839CARDIAC UCP GENE EXPRESSION
dent mechanism. Last, we show that TNF- reduces the
expression of both UCP-2 and UCP-3 in the heart,
suggesting that TNF- induced UCP expression in the
failing heart is not involved in energetic dysfunction.
The results of the present study must be interpreted
in a wider context. The uncoupling of mitochondrial
electron transport from ADP phosphorylation causes a
collapse of the proton gradient across the inner mito-
chondrial membrane and thereby limits ATP genera-
tion through oxidative phosphorylation. Instead, the
potential energy ‘stored’ by the gradient is liberated as
heat, an essential process in nonshivering thermogen-
esis (1, 2). Such a proton leak is catalyzed by UCPs.
Potential roles for UCP-2 and UCP-3 have been sug-
gested for skeletal muscle and adipose tissue, primarily
from studies investigating regulation of their expres-
sion in response to different stimuli. For example, cold
exposure, thyroid hormone, TNF-, elevated dietary fat
composition, insulin-dependent diabetes mellitus, and
specific PPAR agonists have all been shown to increase
skeletal muscle UCP expression (8–10, 21–23). In
contrast, exercise training lowers skeletal muscle UCP
expression (23). These results, plus consideration of
the sheer mass of skeletal muscle in the body, have
implicated skeletal muscle UCPs in the processes of
heat generation, obesity, and perhaps maintenance of
insulin sensitivity (9, 24, 25). However, the role(s) of
UCPs in the adult heart have not been addressed and
are more difficult to rationalize, which gives rise to the
following considerations.
Altered cardiac workload affects both UCP-2 and
UCP-3 expression
In response to increased or decreased workload, the heart
increases glucose utilization and decreases fatty acid utili-
zation (26–28). This change is mirrored by changes in the
expression of several genes encoding metabolic enzymes,
including decreases in the expression of PDK-4 (M. E.
Young et al., unpublished observation), muscle-specific
CPT-I, and medium chain acyl CoA dehydrogenase
(MCAD) in the heart (14, 29). All three genes are
regulated by PPAR in the heart (19, 20, 30, 31). The
present study has found that PPAR expression decreases
during both unloading and overloading (see Results
section and Table 2), which is responsible for the ob-
served changes in PDK-4, CPT-I, and MCAD. Likewise,
cardiac expression of both UCP-2 and UCP-3 decreased
during unloading and overloading (Fig. 2). This decrease
in UCP expression depended on the presence of fatty
acids, as pressure overloaded hearts isolated from animals
Figure 6. Effects of the specific PPARagonist WY-14,643, on cardiac and soleus muscle UCP-2, UCP-3, PPAR, PDK-2, PDK-4,
and muscle CPT-I expression. The effects of feeding animals with a diet containing WY-14,643 (0.01% w/w) on cardiac and
soleus muscle UCP-2 (A), UCP-3 (B), PPAR (C), PDK-2 (D), PDK-4 (E), and muscle CPT-I (F) expression. Values are shown
as the mean se for six separate observations. All values are normalized against the expression of the housekeeping gene
cyclophilin. *P 0.05, **P 0.01, and ***P 0.001 vs. tissue-matched control.
840 Vol. 15 March 2001 YOUNG ET AL.The FASEB Journal
fed a low-fat diet (HC/LF) did not possess decreased UCP
expression compared to diet-matched controls, despite a
decrease in PPAR expression (Table 2). These results
initially suggested to us that PPARacts in the decreased
UCP expression during hypertrophy only in the presence
of its ligand (fatty acids). Similarly, the decreased PPAR
expression during unloading could potentially result in
decreased UCP expression. Consistent with the decrease
observed in UCP expression during pressure overload,
exercise training, which is associated with cardiac hyper-
trophy, reduces UCP expression in the heart (as well as in
skeletal muscle) (23).
Fatty acid availability influences both cardiac and
skeletal muscle UCP expression
High-fat feeding, fasting, and insulin-dependent diabe-
tes mellitus would be expected to activate PPAR (30,
32, 33). In certain experiments, a direct comparison
was made between expression of UCPs in heart and a
slow-twitch skeletal muscle (soleus) that is relatively
similar to the heart (34, 35). Still, there were differ-
ences between the muscles. High-fat feeding, fasting,
and diabetes all increased the expression of cardiac
UCP-3 expression but had no effect on UCP-2 expres-
sion (Figs. 3–5). These results agree with a recent study
that investigated cardiac UCP expression during diabe-
tes and fasting (36). In contrast to the heart, high-fat
feeding and fasting caused induction of both UCP-2
and UCP-3 in soleus muscle (Figs. 3 and 4); previously
reported studies have shown both UCP-2 and UCP-3
expression to increase in skeletal muscle during STZ-
induced diabetes (21). The fold induction of UCP-3 in
soleus muscle was greater than that of the heart during
high-fat feeding and fasting. Specific activation of
PPAR through feeding with WY-14,643 resulted in a
similar pattern of UCP expression observed with eleva-
tion of plasma fatty acid levels: increased UCP-3 expres-
sion in both heart and soleus muscles, with the largest
induction in the latter, whereas UCP-2 was induced
only in soleus, not heart, muscle (Fig. 6).
Cardiac UCP-3, but not UCP-2, expression is
dependent on PPAR signaling in the adult rodent
heart
Table 4 summarizes the relationship between PPAR
and the expression of cardiac UCP-2 and UCP-3, as
observed in the present study. Decreased PPAR ex-
pression during cardiac unloading and overloading
occurs in concert with decreased expression of UCP-2
and UCP-3. Furthermore, the decreased UCP expres-
sion during hypertrophy is dependent on the presence
of fatty acids in the diet, providing evidence for the
hypothesis that this decrease is related to the reduced
PPARexpression. Similarly, on low-fat feeding (either
postfasting or comparing the HC/LF and LC/HF feed-
ing effects), when PPAR activation should be reduced,
UCP-2 and UCP-3 expression both decrease (Fig. 4 and
Table 2). However, situations in which PPAR is stim-
ulated (high-fat feeding, fasting, diabetes, and WY-
14,643 feeding) increase the expression of cardiac
UCP-3, with no effect on cardiac UCP-2 expression.
These observations are consistent with the hypothesis
that UCP-3 expression is regulated by PPAR. One
possible explanation for these observations was that in
the adult rat heart, UCP-2, but not UCP-3, expression is
maximal through PPAR signaling. Thus, a further
stimulation of PPAR would have no effect on UCP-2
expression, yet reduced PPARsignaling would reduce
UCP-2 expression. To investigate this hypothesis fur-
ther, we determined the level of expression of cardiac
UCP-2 and UCP-3 in PPAR
/
mice. In this model,
expression of various PPAR-regulated genes is de-
creased (37). Because we find reduced expression of
TABLE 3. Expression of cardiac UCP-2 and UCP-3 in
PPAR
/
and wild-type mice
Wild-type PPAR
/
UCP-2/cyclo 0.248 0.034 0.259 0.076
UCP-3/cyclo 0.193 0.074 0.010 0.002*
a
Expression of UCP-2 and UCP-3 in wild-type and PPAR
/
hearts. Values are shown as the mean se for four (wild-type) or five
(PPAR
/
) separate observations. All values are normalized against
the expression of the housekeeping gene cyclophilin (cyclo).
* P 0.05 vs wild-type.
Figure 7. Effects of TNF- administration on cardiac and
soleus muscle UCP-2, UCP-3, PPAR and iNOS expression.
The effects of TNF- injection (30 g/kg; i.v.) on cardiac and
soleus muscle UCP-2 (A), UCP-3 (B), PPAR (C), and iNOS
(D) expression. Values are shown as the mean se for six
separate observations. All values are normalized against the
expression of the housekeeping gene cyclophilin. *P 0.05
and **P 0.01 vs. tissue-matched control.
841CARDIAC UCP GENE EXPRESSION
UCP-3, but not UCP-2, in PPAR
/
mice, these results
provide evidence that UCP-2 expression in the adult
rodent heart is not regulated by PPAR. Instead, there
must be a fatty acid-dependent, PPAR-independent
mechanism modulating cardiac UCP-2 expression,
which is maximally activated under normal physiologi-
cal circumstances. PPAR, another member of the
family of PPAR transcription factors, does not appear to
regulate UCP-3 expression in the same way as PPAR in
the heart. Treatment of adult rats with the PPAR
agonist troglitazone (0.1% w/w in the diet for 4 days),
has no effect on UCP-3 (nor UCP-2) expression in the
heart (M. E. Young et al., unpublished observation).
This lack of an effect by troglitazone treatment (which
did induce known PPAR-regulated genes in the skel-
etal muscle) on cardiac UCP gene expression is most
likely due to the very low abundance of PPAR in the
heart, as compared with both adipose tissue and skele-
tal muscle.
TNF- reduces cardiac UCP expression
TNF- is known to cause contractile dysfunction (38),
and the expression of TNF- increases in the failing
heart (11). Furthermore, TNF- antagonism appears to
have beneficial effects in subjects with heart failure
(39). TNF-has been shown to induce UCP expression
in skeletal muscle (10). As overexpression of UCPs
might limit mitochondrial ATP production, we hypoth-
esized that TNF--induced cardiac UCP expression
could result in the contractile dysfunction observed
previously. However, administration of TNF- to adult
rats caused a significant reduction in cardiac UCP
expression (Fig. 7). Whether reduced cardiac UCP
expression in response to TNF- administration was
due to a direct effect of TNF- on the cardiomyocyte or
to a systemic effect cannot be determined in the
present study. It is also still possible that local TNF-
generation by the cardiomyocyte, as observed in failing
myocardium, induces UCP expression.
Complexity of cardiac and skeletal muscle UCP gene
expression regulation
There are obvious differences in the regulation of
UCP-2 and UCP-3 expression, some of which are sum-
marized in Table 4. Under physiological conditions,
cardiac UCP-2 expression is 100-fold greater than that
of UCP-3. Expression of UCP-3 is regulated by PPAR
in the adult rodent heart, whereas UCP-2 is not. On the
second day of exposure to elevated fatty acid levels (for
both the high-fat feeding and fasting experiments),
both cardiac and soleus muscle UCP-3 expression fall
transiently (Figs. 3 and 4). As fatty acid exposure
continues, UCP-3 expression increases again (Fig. 3).
Increased age was associated with decreased cardiac
UCP expression. In addition, TNF- increases soleus
muscle UCP-2 expression while decreasing cardiac
UCP-2 expression (Fig. 7). Thus, multiple mechanisms
for the regulation of UCP expression must operate in
heart and skeletal muscle. These might include altered
expression and/or activity in PPAR (as observed in
the heart for the present study), PPAR dimerization
partners (e.g., RXR) or coactivators (e.g., PGC-1),
which confer specificity between UCP-2 and UCP-3
promoter regions, or even as yet unidentified PPAR-
independent mechanisms (4042). A study published
during the preparation of this manuscript suggests that
cardiac UCP-2 expression is regulated by PPAR (43).
In this study, neonatal cardiomyocytes were cultured in
the presence of triiodothyronine, fatty acids or WY-
14,643, resulting in increased UCP-2 expression. From
these observations, it was concluded that increased
cardiac UCP-2 expression on birth was due to com-
bined stimulation of the thyroid hormone receptor and
PPAR. However, at birth there is a substantial increase
in mitochondrial biogenesis, which might explain the
increased expression of a mitochondrial protein such
as UCP-2 (44). It should be noted that the results
observed in the present study are not consistent with
altered mitochondrial biosynthesis (e.g., acute pressure
overload results in increased mitochondrial biogenesis
whereas UCP expression decreases; 45). It is possible
that neonatal cardiomyocytes in culture possess a spe-
cific factor (e.g., a PPAR dimerization partner or
coactivator) that is not present in the adult heart.
Increased fatty acid availability reduces cardiac
PPAR expression
The present results suggest that conditions associated
with increased cardiac fatty acid utilization (high-fat
feeding, fasting, and diabetes) result in decreased
PPAR expression, a mechanism not acutely observed
in skeletal muscle (Figs. 3–5). Two hypotheses can be
drawn regarding the potential mechanism by which this
phenomenon occurs. First, increased signaling through
PPARmight cause the altered expression of a protein
that affects the expression of PPAR. This autoregula-
tion mechanism would therefore be PPAR-depen-
dent. A second, PPAR-independent mechanism can
TABLE 4. Relationship between PPAR and the expression of
UCP-2 and UCP-3 in the heart
a
Model
PPAR
(expression/ligand
availability)
UCP-2
expression
UCP-3
expression
Unloading 2Expression 22
Pressure overload 2 Expression 22
TNF administration 2 Expression 22
PPAR
/
mouse
2 Expression 2
High-fat feeding 1 Ligand 1
Fasting 1 Ligand 1
STZ-induced
diabetes 1 Ligand 1
WY-14,643 treatment 1 Ligand 1
a
1 represents an increase, 2 represents a decrease, and
represents no change.
842 Vol. 15 March 2001 YOUNG ET AL.The FASEB Journal
be postulated wherein fatty acids activate a specific
pathway that regulates the expression of PPAR. Thus,
the first mechanism is fatty acid independent and
PPAR dependent whereas the latter mechanism is
fatty acid dependent and PPAR independent. To
investigate which mechanism was responsible for the
observed results, we used a specific PPAR agonist
(WY-14,643) that would lead to PPAR activation in the
absence of elevated fatty acid levels. WY-14,643 in-
creased cardiac and skeletal muscle UCP-3, PDK-4, and
muscle CPT-I expression, suggesting PPAR was acti-
vated. In contrast to fatty acids, WY-14,643 feeding had
no effect on cardiac PPAR expression (or soleus
muscle PPAR expression). The results are consistent
with the hypothesis that a fatty acid-dependent mecha-
nism is responsible for the observed down-regulation of
cardiac PPAR. Further evidence toward this hypothe-
sis is the observation that hearts isolated from the obese
Zucker rat, a model of insulin resistance in which
plasma fatty acid levels are elevated, have severely
reduced PPAR expression (46). It could be hypothe-
sized that repression of PPAR expression by fatty acids
serves as a mechanism to prevent excessive fluctuations
in PPAR signaling in this tissue during altered fatty
acid availability.
Potential roles of UCPs in the adult rodent heart
The present study has focused on uncovering the
mechanisms by which gene expression of the uncou-
pling proteins is regulated in the adult heart during
both physiological and pathophysiological conditions.
Determination of UCP protein levels (requiring anti-
body generation) and altered mitochondrial function is
beyond the scope of the present study. It is believed
that elucidating the mechanisms by which the heart
alters the expression of the UCPs might aid in the
understanding of their function. Possible major func-
tions for UCPs in the heart include thermogenesis,
regulation of fatty acid oxidation, regulation of ATP
synthesis, and reduction of ROS formation. Due to its
size (compared with the body), the heart is unlikely to
play a role in global thermogenesis. UCP-2 expression
is relatively insensitive to substrate availability, suggest-
ing it plays little function in cardiac fatty acid utilization
(which increases, for example, in diabetes). However,
UCP-3 is highly responsive to fatty acids. Whether
UCP-3 induction helps prevent lipotoxicity when fatty
acid levels are high or acts in an antioxidant capacity
when oxidative metabolism increases (fatty acids can-
not be metabolized anaerobically) are distinct possibil-
ities. When the heart requires increased efficiency, for
example, during increased or decreased workload or
substrate limitation (as observed during low dietary
fatty acid availability, as fatty acids are the primary fuel
for the heart under physiological conditions), cardiac
UCP expression decreases. If cardiac UCPs affect mito-
chondrial ATP synthesis, decreased UCP expression
would be expected to increase mitochondrial effi-
ciency. Last, evidence suggests that uncoupling pro-
teins can act as antioxidants (47, 48). The heart is a
continuously contracting organ, with high oxidative
metabolism. The observation that cardiac UCP-2 ex-
pression is relatively high (even higher than skeletal
muscle) and relatively constant during various condi-
tions suggests that it has an essential, constitutive role
in the heart, such as prevention of ROS formation.
Indirect evidence for this hypothesis comes from the
observations that TNF-administration, which causes a
rapid decrease in cardiac UCP-2 and UCP-3 expression,
is known to increase oxidative stress (49).
Limitations of the study
Whether changes in UCP gene expression result in
changes in either UCP protein or activity or ultimately
lead to altered cardiac function has not been deter-
mined. Future studies are required to address these
issues. For example, the expression of UCP-2 in the
heart is 100-fold greater than that of UCP-3. There-
fore, if both UCP-2 and UCP-3 possess uncoupling
activity, what is the physiological significance of UCP-3
induction during increased fatty acid availability? It is
possible that UCP-2 and UCP-3 possess different intrin-
sic activities, are differentially regulated post-transcrip-
tionally, or are located within different regions of the
inner mitochondrial membrane, akin to the light har-
vesting complexes and photosystems of chloroplasts.
Whether allosteric factors other than guanosine nucle-
otides and fatty acids differentially affect the activity of
UCP-2 and UCP-3 is unknown. Another concern is that
each intervention investigated in the present study
could potentially affect multiple factors in these com-
plex in vivo models. For example, nervous activity,
workload, and multiple growth factor and cytokine
signaling cascades are all altered in both the unloaded
and pressure overloaded heart. In addition, various
hormonal alterations occur during nutritional manip-
ulation and diabetes. Even in knockout mice, compen-
satory mechanisms become activated, allowing for ad-
aptation in the absence of a specific gene. All these
factors could potentially affect cardiac UCP gene ex-
pression. However, one common factor that links car-
diac UCP-3 (but not UCP-2) expression with these
diverse animal models is the transcription factor
PPAR (see Table 4). Although the present study has
not directly measured PPAR protein or activity, previ-
ous studies have shown that increased fatty acid avail-
ability results in PPARactivation (30, 32, 33) and that
reduced PPAR transcription—for example, during
cardiac hypertrophy—is associated with reduced activ-
ity and expression of PPAR-regulated genes (50).
CONCLUSIONS
In the adult rodent heart, expression of UCP-3 but not
UCP-2 is regulated by PPAR. In contrast to skeletal
muscle, in which UCP expression alters dramatically in
response to substrate availability, heart UCP expression
843CARDIAC UCP GENE EXPRESSION
does not fluctuate very much. The expression of UCP-2,
the major UCP isoform in the rat heart (100-fold
higher expression compared to UCP-3), changes rela-
tively little in response to dramatic alterations in sub-
strate availability. Uncoupling of skeletal muscle mito-
chondria may be important for the utilization of fatty
acids, whereas the uncoupling of cardiac mitochondria
might play more of a role in an antioxidant capacity,
preventing over-reduction of the electron transport
chain. In situations in which increased cardiac effi-
ciency is required, such as unloading, overloading, and
decreased fatty acid availability, cardiac UCP expression
is decreased. Whether other factors are able to signifi-
cantly increase the expression of uncoupling proteins
in the adult heart is unknown.
We wish to thank Patrick H. Guthrie and Wenhao Chen for
technical assistance. This work was supported in part by
grants from the NIH (HL-43133 and HL-61483) and from the
American Heart Association National Center. C.D. was a
Daland Fellow of the American Philosophical Society.
REFERENCES
1. Ricquier, D., Thibault, J., Bouillaud, F., and Kuster, Y. (1983)
Molecular approach to thermogenesis in brown adipose tissue.
Cell-free translation of mRNA and characterization of the
mitochondrial uncoupling protein. J. Biol. Chem. 258, 6675–
6677
2. Palou, A., Pico, C., Bonet, M., and Oliver, P. (1998) The
uncoupling protein, thermogenin. Int. J. Biochem. Cell Biol. 30,
7–11
3. Fleury, C., Neverova, M., Collins, S., Raimbault, S., Champigny,
O., Levi-Meyrueis, C., Bouillaud, F., Seldin, M., Surwit, R.,
Ricquier, D., and Warden, C. (1997) Uncoupling protein-2: a
novel gene linked to obesity and hyperinsulinemia. Nat. Genet.
15, 269–272
4. Boss, O., Samec, S., Paoloni-Giacobino, A., Rossier, C., Dulloo,
A., Seydoux, J., Muzzin, P., and Giacobino, J. (1997) Uncou-
pling protein-3: a new member of the mitochondrial carrier
family with tissue-specific expression. FEBS Lett. 408, 39–42
5. Vidal-Puig, A., Solanes, G., Grujic, D., Flier, J., and Lowell, B.
(1997) UCP3: an uncoupling protein homologue expressed
preferentially and abundantly in skeletal muscle and brown
adipose tissue. Biochem. Biophys. Res. Commun. 235, 79–82
6. Jaburek, M., Varecha, M., Gimeno, R., Dembski, M., Jezek, P.,
Zhang, M., Burn, P., Tartaglia, L., and Garlid, K. (1999)
Transport function and regulation of mitochondrial uncou-
pling proteins 2 and 3. J. Biol. Chem. 274, 26003–26007
7. Boss, O., Hagen, T., and Lowell, B. (2000) Uncoupling proteins
2 and 3. Potential regulators of mitochondrial energy metabo-
lism. Diabetes 49, 143–156
8. Brun, S., Carmona, M., Mampel, T., Vinas, O., Giralt, M.,
Iglesias, R., and Villarroya, F. (1999) Activators of peroxisome
proliferator-activated receptor-alpha induce the expression of
the uncoupling protein-3 gene in skeletal muscle: a potential
mechanism for the lipid intake-dependent activation of uncou-
pling protein-3 gene expression at birth. Diabetes 48, 1217–1222
9. Samec, S., Seydoux, J., and Dulloo, A. (1999) Post-starvation
gene expression of skeletal muscle uncoupling protein 2 and
uncoupling protein 3 in response to dietary fat levels and fatty
acid composition: a link with insulin resistance. Diabetes 48,
436441
10. Busquets, S., Sanchis, D., Alvarez, B., Ricquier, D., Lopez-
Soriano, F., and Argiles, J. M. (1998) In the rat, tumor necrosis
factor alpha administration results in an increase in both UCP2
and UCP3 mRNAs in skeletal muscle: a possible mechanism for
cytokine-induced thermogenesis? FEBS Lett. 440, 348–350
11. Torre-Amione, G., Kapadia, S., Lee, J., Durand, J. B., Bies, R. D.,
Young, J. B., and Mann, D. L. (1996) Tumor necrosis factor-
alpha and tumor necrosis factor receptors in the failing human
heart. Circulation 93, 704–711
12. Katz, A. (1998) Is the failing heart energy depleted? Cardiol.
Clin. 16, 633–644
13. Ono, K., and Lindsey, E. S. (1969) Improved technique of heart
transplantation in rats. J. Thorac. Cardiovasc. Surg. 57, 225–229
14. Depre, C., Shipley, G., Chen, W., Han, Q., Doenst, T., Moore,
M., Stepkowski, S., Davies, P., and Taegtmeyer, H. (1998)
Unloaded heart in vivo replicates fetal gene expression of
cardiac hypertrophy. Nature Med. 4, 1269–1275
15. Kleinman, L., Wechsler, A., Rembert, J., Fedor, J., and Green-
field, J. (1978) A reproducible model of moderate to severe
concentric left ventricular hypertrophy. Am. J. Physiol. 234,
H515–H519
16. Heid, C. A., Stevens, J., Livak, K. J., and Williams, P. M. (1996)
Real time quantitative PCR. Genome Res. 6, 986–994
17. Gibson, U. E. M., Heid, C. A., and Williams, P. M. (1996) A novel
method for real time quantitative RT-PCR. Genome Res. 6,
995–1001
18. Chomczynski, P., and Sacchi, N. (1987) Single-step method of
RNA isolation by acid guanidium thiocyanate-phenol-chloro-
form extraction. Anal. Biochem. 162, 159–169
19. Wu, P., Inskeep, K., Bowker-Kinley, M., Popov, K., and Harris, R.
(1999) Mechanism responsible for inactivation of skeletal mus-
cle pyruvate dehydrogenase complex in starvation and diabetes.
Diabetes 48, 1593–1599
20. Brandt, J., Djouadi, F., and Kelly, D. (1998) Fatty acids activate
transcription of the muscle carnitine palmitoyltransferase I gene
in cardiac myocytes via the peroxisome proliferator-activated
receptor alpha. J. Biol. Chem. 273, 23786–23792
21. Kageyama, H., Suga, A., Kashiba, M., Oka, J., Osaka, T., Kashiwa,
T., Hirano, T., Nemoto, K., Namba, Y., Ricquier, D., Giacobino,
J., and Inoue, S. (1998) Increased uncoupling protein-2 and -3
gene expressions in skeletal muscle of STZ-induced diabetic
rats. FEBS Lett. 440, 450 453
22. Larkin, S., Mull, E., Miao, W., Pittner, R., Albrandt, K., Moore,
C., Young, A., Denaro, M., and Beaumount, K. (1997) Regula-
tion of the third member of the uncoupling protein family,
UCP3, by cold and thyroid hormone. Biochem. Biophys. Res.
Commun. 240, 222–227
23. Boss, O., Samec, S., Desplanches, D., Mayet, M., Seydoux, J.,
Muzzin, P., and Giacobino, J. (1998) Effect of endurance
training on mRNA expression of uncoupling proteins 1, 2, and
3 in the rat. FASEB J. 12, 335–339
24. Schrauwen, P., Walder, K., and Ravussin, E. (1999) Human
uncoupling proteins and obesity. Obes. Res. 7, 97–105
25. Krook, A., Digby, J., O’Rahilly, S., Zierath, J., and H, W.-H.
(1998) Uncoupling protein 3 is reduced in skeletal muscle of
NIDDM patients. Diabetes 47, 1528–1531
26. Kagaya, Y., Kanno, Y., Takeyama, D., Ishide, N., Maruyama, Y.,
Takahashi, T., Ido, T., and Takishima, T. (1990) Effects of
long-term pressure overload on regional myocardial glucose
and free fatty acid uptake in rats. A quantitative autoradio-
graphic study. Circulation 81, 1353–1361
27. Allard, M., Schonekess, B., Henning, S., English, D., and Lop-
aschuk, G. (1994) Contribution of oxidative metabolism and
glycolysis to ATP production in hypertrophied hearts. Am. J.
Physiol. 267, H742–H750
28. Doenst, T., Depre, C., Goodwin, G. W., Wang, M., Stepkowski,
S. M., and Taegtmeyer, H. (1999) Left ventricular unloading in
vivo shifts myocardial substrate preference in vitro. Circulation
100 (Suppl. I), I-344
29. Sack, M. N., Rader, T. A., Park, S., Bastin, J., McCune, S. A.,
and Kelly, D. P. (1996) Fatty acid oxidation enzyme gene
expression is downregulated in the failing heart. Circulation
94, 2837–2842
30. Leone, T., Weinheimer, C., and Kelly, D. (1999) A critical role
for the peroxisome proliferator-activated receptor alpha
(PPARalpha) in the cellular fasting response: the PPARalpha-
null mouse as a model of fatty acid oxidation disorders. Proc.
Natl. Acad. Sci. USA 96, 7473–7478
31. Wu, P., Sato, J., Zhao, Y., Jaskiewicz, J., Popov, K., and Harris, R.
(1998) Starvation and diabetes increase the amount of pyruvate
dehydrogenase kinase isoenzyme 4 in rat heart. Biochem. J. 329,
197–201
32. Kersten, S., Seydoux, J., Peters, J., Gonzalez, F., Desvergne, B.,
and Wahli, W. (1999) Peroxisome proliferator-activated recep-
844 Vol. 15 March 2001 YOUNG ET AL.The FASEB Journal
tor alpha mediates the adaptive response to fasting. J. Clin.
Invest. 103, 1489–1498
33. Forman, B., Chen, J., and Evans, R. (1997) Hypolipidemic
drugs, polyunsaturated fatty acids, and eicosanoids are ligands
for peroxisome proliferator-activated receptors alpha and delta.
Proc. Natl. Acad. Sci. USA 94, 4312–4317
34. Armstrong, R., and Laughlin, M. (1983) Blood flows within and
among rat muscles as a function of time during high speed
treadmill exercise. J. Physiol. (London) 344, 189–208
35. Hom, F., and Goodner, C. (1984) Insulin dose-response char-
acteristics among individual muscle and adipose tissues mea-
sured in the rat in vivo with 3[H]2-deoxyglucose. Diabetes 33,
153–159
36. Hidaka, S., Kakuma, T., Yoshimatsu, H., Sakino, H., Fukuchi, S.,
and Sakata, T. (1999) Streptozotocin treatment upregulates
uncoupling protein 3 expression in the rat heart. Diabetes 48,
430 435
37. Aoyama, T., Peters, J., Iritani, N., Nakajima, T., Furihata, K.,
Hashimoto, T., and Gonzalez, F. (1998) Altered constitutive
expression of fatty acid-metabolizing enzymes in mice lacking
the peroxisome proliferator-activated receptor alpha (PPARal-
pha). J. Biol. Chem. 273, 5678–5684
38. Hegewisch, S., Weh, H., and Hossfeld, D. (1990) TNF-induced
cardiomyopathy. Lancet 335, 294–295
39. Deswal, A., Bozkurt, B., Seta, Y., Parilti-Eiswirth, S., Hayes, F.,
Blosch, C., and Mann, D. (1999) Safety and efficacy of a soluble
P75 tumor necrosis factor receptor (Enbrel, etanercept) in
patients with advanced heart failure. Circulation 99, 3224–3226
40. Schoonjans, K., Staels, B., and Auwerx, J. (1996) The peroxi-
some proliferator activated receptors (PPARS) and their effects
on lipid metabolism and adipocyte differentiation. Biochim.
Biophys. Acta 1302, 93–109
41. Boss, O., Bachman, E., Vidal-Puig, A., Zhang, C., Peroni, O., and
Lowell, B. (1999) Role of the beta(3)-adrenergic receptor
and/or a putative beta(4)-adrenergic receptor on the expres-
sion of uncoupling proteins and peroxisome proliferator-acti-
vated receptor-gamma coactivator-1. Biochem. Biophys. Res. Com-
mun. 261, 870 876
42. Wu, Z., Puigserver, P., Andersson, U., Zhang, C., Adelmant, G.,
Mootha, V., Troy, A., Cinti, S., Lowell, B., Scarpulla, R., and
Spiegelman, B. (1999) Mechanisms controlling mitochondrial
biogenesis and respiration through the thermogenic coactivator
PGC-1. Cell 98, 115–124
43. Van Der Lee, K., Willemsen, P., Van Der Vusse, G., and Van
Bilsen, M. (2000) Effects of fatty acids on uncoupling protein-2
expression in the rat heart. FASEB J. 14, 495–502
44. Legato, M. (1979) Cellular mechanisms of normal growth in the
mammalian heart. II. A quantitative and qualitative comparison
between the right and left ventricular myocytes in the dog from
birth to five months of age. Circ. Res. 44, 263–279
45. Meerson, F., and Pomoinitsky, V. (1972) The role of high-
energy phosphate compounds in the development of cardiac
hypertrophy. J. Mol. Cell Cardiol. 4, 571–597
46. Zhou, Y., Grayburn, P., Karim, A., Shimabukuro, M., Higa, M.,
Baetens, D., Orci, L., and Unger, R. (2000) Lipotoxic heart
disease in obese rats: implications for human obesity. Proc. Natl.
Acad. Sci. USA 97, 1784–1789
47. Korshunov, S., Korkina, O., Ruuge, E., Skulachev, V., and
Starkov, A. (1998) Fatty acids as natural uncouplers preventing
generation of O
2
.-
and H
2
O
2
by mitochondria in the resting
state. FEBS Lett. 435, 215–218
48. Negre-Salvayre, A., Hirtz, C., Carrera, G., Cazenave, R., Troly,
M., Salvayre, R., Penicaud, L., and Casteilla, L. (1997) A role for
uncoupling protein-2 as a regulator of mitochondrial hydrogen
peroxide generation. FASEB J. 11, 809 815
49. Goossens, V., Grooten, J., De Vos, K., and Fiers, W. (1995)
Direct evidence for tumor necrosis factor-induced mitochon-
drial reactive oxygen intermediates and their involvement in
cytotoxicity. Proc. Natl. Acad. Sci. USA 92, 8115–8119
50. Barger, P., Brandt, J., Leone, T., Weinheimer, C., and Kelly, D.
(2000) Deactivation of peroxisome proliferator-activated recep-
tor-alpha during cardiac hypertrophic growth. J. Clin. Invest.
105, 1723–1730
Received for publication May 24, 2000.
Revised for publication July 27, 2000.
845CARDIAC UCP GENE EXPRESSION
... We then tested, whether FOXO1 or KLF5 ablation in T1D affects the expression of a PPARα target, UCP3 (uncoupling protein 3). 38 Cardiac UCP3 mRNA and protein levels were increased in C57BL/6 mice with T1D ( Figure XXIA, XXIB, and XXIC in the Data Supplement) that had higher cardiac PPARα expression, whereas Ucp2 expression was not altered ( Figure XXIA in the Data Supplement). Both precursor and cleaved (activated) SREBP-1 (sterol regulatory-element binding protein 1), which is a negative regulator of UCP3 in hyperinsulinemia, 39 . ...
... 37 The lack of downregulation of cardiac PPARα in diabetic mice that underwent KLF5 inhibition may be due to compensation by FOXO1-mediated stimulation of PPARα expression that we also show or other pathways. Similarly, expression of cardiac UCP3, which is a PPARα target, 38 was increased in mice with T1D, and KLF5 inhibition did not reverse it. Upregulation of cardiac UCP3 expression in mice with T1D was also consistent with inhibition of SREBP-1 activation that we also observed. ...
Article
Full-text available
Rationale: Diabetic cardiomyopathy (DbCM) is a major complication in type-1 diabetes (T1D), accompanied by altered cardiac energetics, impaired mitochondrial function and oxidative stress. Previous studies indicate that T1D is associated with increased cardiac expression of Krüppel-like factor-5 (KLF5) and Peroxisome Proliferator Activated Receptor (PPAR)α that regulate cardiac lipid metabolism. Objective: In this study, we investigated the involvement of KLF5 in DbCM and its transcriptional regulation. Methods and Results: KLF5 mRNA levels were assessed in isolated cardiomyocytes from cardiovascular patients with diabetes and was higher compared with non-diabetic individuals. Analyses in human cells and diabetic mice with cardiomyocyte-specific FOXO1 deletion showed that FOXO1 bound directly on the KLF5 promoter and increased KLF5 expression. Diabetic mice with cardiomyocyte-specific FOXO1 deletion had lower cardiac KLF5 expression and were protected from DbCM. Genetic, pharmacologic gain and loss of KLF5 function approaches and AAV-mediated Klf5 delivery in mice showed that KLF5 induces DbCM. Accordingly, the protective effect of cardiomyocyte FOXO1 ablation in DbCM was abolished when KLF5 expression was rescued. Similarly, constitutive cardiomyocyte-specific KLF5 overexpression caused cardiac dysfunction. KLF5 caused oxidative stress via direct binding on NADPH oxidase (NOX)4 promoter and induction of NOX4 expression. This was accompanied by accumulation of cardiac ceramides. Pharmacologic or genetic KLF5 inhibition alleviated superoxide formation, prevented ceramide accumulation and improved cardiac function in diabetic mice. Conclusions: Diabetes-mediated activation of cardiomyocyte FOXO1 increases KLF5 expression, which stimulates NOX4 expression, ceramide accumulation and causes DbCM.
... Patients with metabolic syndrome have an increased sterol-regulatory elementbinding protein (SREBP)-1c/peroxisome proliferator-activated receptor-γ (PPARγ) pathway, which is correlated with decreased ejection fraction (EF) and elevated lipid accumulation [63]. High levels of fatty acids and elevated PPAR-α are associated with uncoupling protein 3 (UCP-3) overexpression [64]. Uncoupling is a process reducing mitochondrial ATP production which correlates with the impaired contractility of the heart muscle [65]. ...
Article
Full-text available
Cardiovascular complications are the most deadly and cost-driving effects of diabetes mellitus (DM). One of them, which is steadily attracting attention among scientists, is diabetes-induced heart failure, also known as diabetic cardiomyopathy (DCM). Despite significant progress in the research concerning the disease, a universally accepted definition is still lacking. The pathophysiology of the processes accelerating heart insufficiency in diabetic patients on molecular and cellular levels also remains elusive. However, the recent interest concerning extracellular vesicles (EVs) has brought promise to further clarifying the pathological events that lead to DCM. In this review, we sum up recent investigations on the involvement of EVs in DCM and show their therapeutic and indicatory potential.
... Although PPARα is the dominant isoform in the heart, PPARβ and γ can also modulate FA metabolism [36]. PPARα stimulates the transcription of lipoprotein lipase, involved in FA uptake and β-oxidation in the mitochondria [37][38][39][40]. In addition, overexpression of PPARα in the heart favors FA metabolism and induces insulin resistance [41]. ...
Article
Full-text available
Cardiovascular disease is the leading cause of mortality globally with at least 26 million people worldwide living with heart failure (HF). Metabolism has been an active area of investigation in the setting of HF since the heart demands a high rate of ATP turnover to maintain homeostasis. With the advent of -omic technologies, specifically metabolomics and lipidomics, HF pathologies have been better characterized with unbiased and holistic approaches. These techniques have identified novel pathways in our understanding of progression of HF and potential points of intervention. Furthermore, sodium-glucose transport protein 2 inhibitors, a drug that has changed the dogma of HF treatment, has one of the strongest types of evidence for a potential metabolic mechanism of action. This review will highlight cardiac metabolism in both the healthy and failing heart and then discuss the metabolic effects of heart failure drugs.
... Excessive FA oxidation increases ATP expenditure for futile cycling of metabolic intermediates, inhibits ATP shuttling from mitochondria to the cytosol, and increases the expression of mitochondrial uncoupling protein (UCP) 3 through PPARα, thereby dissipating the mitochondrial proton gradient and deteriorating the ATP production efficiency (56,57). Finally, these changes produce oxidative stress and mitochondrial dysfunction (58). ...
Article
Full-text available
Individuals with diabetes mellitus (DM) disclose a higher incidence and a poorer prognosis of heart failure (HF) than non-diabetic people, even in the absence of other HF risk factors. The adverse impact of diabetes on HF likely reflects an underlying “diabetic cardiomyopathy” (DM–CMP), which may by exacerbated by left ventricular hypertrophy and coronary artery disease (CAD). The pathogenesis of DM-CMP has been a hot topic of research since its first description and is still under active investigation, as a complex interplay among multiple mechanisms may play a role at systemic, myocardial, and cellular/molecular levels. Among these, metabolic abnormalities such as lipotoxicity and glucotoxicity, mitochondrial damage and dysfunction, oxidative stress, abnormal calcium signaling, inflammation, epigenetic factors, and others. These disturbances predispose the diabetic heart to extracellular remodeling and hypertrophy, thus leading to left ventricular diastolic and systolic dysfunction. This Review aims to outline the major pathophysiological changes and the underlying mechanisms leading to myocardial remodeling and cardiac functional derangement in DM-CMP.
... The altered fatty acid-metabolizing proteins expression in PPARα −/− is associated with myocardial damage and fibrosis [98]. In rodent cardiomyocytes, chronic exposure to elevated FFAs reduces the expression of PPAR-α which inhibits FFA oxidation and increases intracellular fat accumulation reducing heart function [99,100]. Like PPAR-α, PPAR-β/δ is abundantly expressed in the heart playing a different role in myocardial metabolism since its overexpression enhances the capacity for myocardial glucose utilization and increases glycolytic gene expression. ...
Article
Full-text available
Diabetic patients are predisposed to diabetic cardiomyopathy, a specific form of cardiomyopathy which is characterized by the development of myocardial fibrosis, cardiomyocyte hypertrophy, and apoptosis that develops independently of concomitant macrovascular and microvascular diabetic complications. Its pathophysiology is multifactorial and poorly understood and no specific therapeutic guideline has yet been established. Diabetic cardiomyopathy is a challenging diagnosis, made after excluding other potential entities, treated with different pharmacotherapeutic agents targeting various pathophysiological pathways that need yet to be unraveled. It has great clinical importance as diabetes is a disease with pandemic proportions. This review focuses on the potential mechanisms contributing to this entity, diagnostic options, as well as on potential therapeutic interventions taking in consideration their clinical feasibility and limitations in everyday practice. Besides conventional therapies, we discuss novel therapeutic possibilities that have not yet been translated into clinical practice.
... 17 Accordingly, the myocardial expression of Pparα (peroxisome proliferator-activated receptor α) which promotes FA uptake and utilization, was significantly elevated in diabetic WT, but not in mGCH1-Tg, compared with sham-injected controls ( Figure 7A). Similarly, the LV content of the mitochondrial UCP3 (uncoupling protein-3), a downstream target of PPARα, 18 was 50% higher in diabetic WT at 4 weeks post-DM induction ( Figure IIIA in the Data Supplement) in keeping with an early switch to FA metabolism. However, the increase in myocardial UCP3 was much greater in WT by 12 weeks post-DM induction ( Figure 7B) as was that of PDK4 (pyruvate dehydrogenase kinase 4; Figure IIIB in the Data Supplement), implying that myocardial metabolism was increasingly compromised as the duration of DM increased. ...
Article
Full-text available
Rationale: In diabetic patients, heart failure with predominant left ventricular (LV) diastolic dysfunction is a common complication for which there is no effective treatment. Oxidation of the nitric oxide synthase (NOS) co-factor tetrahydrobiopterin (BH4) and dysfunctional NOS activity have been implicated in the pathogenesis of the diabetic vascular and cardiomyopathic phenotype. Objective: Using mice models and human myocardial samples, we evaluated whether and by which mechanism increasing myocardial BH4 availability prevented or reversed LV dysfunction induced by diabetes. Methods and Results: In contrast to the vascular endothelium, BH4 levels, superoxide production and NOS activity (by liquid chromatography) did not differ in the LV myocardium of diabetic mice or in atrial tissue from diabetic patients. Nevertheless, the impairment in both cardiomyocyte relaxation and [Ca2+]i decay and in vivo LV function (echocardiography and tissue Doppler) that developed in wild type mice (WT) 12 weeks post-DM induction (streptozotocin, 42-45mg/kg) was prevented in mice with elevated myocardial BH4 content secondary to overexpression of GTP-cyclohydrolase 1 (mGCH1-Tg) and reversed in WT mice receiving oral BH4 supplementation from the 12th to the 18th week after DM induction. The protective effect of BH4 was abolished by CRISPR/Cas9-mediated knockout of neuronal NOS (nNOS) in mGCH1-Tg. In HEK cells, S-nitrosoglutathione led to a PKG-dependent increase in plasmalemmal density of the insulin-independent glucose transporter, GLUT-1. In cardiomyocytes, mGCH1 overexpression induced a NO/sGC/PKG-dependent increase in glucose uptake via GLUT-1, which was instrumental in preserving mitochondrial creatine kinase activity, oxygen consumption rate, LV energetics (by 31P MRS) and myocardial function. Conclusions: We uncovered a novel mechanism whereby myocardial BH4 prevents and reverses LV diastolic and systolic dysfunction associated with diabetes via a nNOS-mediated increase in non-insulin dependent myocardial glucose uptake and utilization. These findings highlight the potential of GCH1/BH4-based therapeutics in human diabetic cardiomyopathy.
Article
BACKGROUND Strategies to increase cellular NAD ⁺ (oxidized nicotinamide adenine dinucleotide) level have prevented cardiac dysfunction in multiple models of heart failure, but molecular mechanisms remain unclear. Little is known about the benefits of NAD ⁺ -based therapies in failing hearts after the symptoms of heart failure have appeared. Most pretreatment regimens suggested mechanisms involving activation of sirtuin, especially Sirt3 (sirtuin 3), and mitochondrial protein acetylation. METHODS We induced cardiac dysfunction by pressure overload in SIRT3-deficient (knockout) mice and compared their response with nicotinamide riboside chloride treatment with wild-type mice. To model a therapeutic approach, we initiated the treatment in mice with established cardiac dysfunction and found nicotinamide riboside chloride improved mitochondrial function and blunted heart failure progression. Similar benefits were observed in wild-type and knockout mice. Boosting NAD ⁺ level improved the function of NAD(H) redox-sensitive SDR (short-chain dehydrogenase/reductase) family proteins. Upregulation of Mrpp2 (mitochondrial ribonuclease P protein 2), a multifunctional SDR protein and a subunit of mitochondrial ribonuclease P, improves mitochondrial DNA transcripts processing and electron transport chain function. Activation of SDRs in the retinol metabolism pathway stimulates RXRα (retinoid X receptor α)/PPARα (proliferator-activated receptor α) signaling and restores mitochondrial oxidative metabolism. Downregulation of Mrpp2 and impaired mitochondrial ribonuclease P were found in human failing hearts, suggesting a shared mechanism of defective mitochondrial biogenesis in mouse and human heart failure. CONCLUSIONS These findings identify SDR proteins as important regulators of mitochondrial function and molecular targets of NAD ⁺ -based therapy. Furthermore, the benefit is observed regardless of Sirt3-mediated mitochondrial protein deacetylation, a widely held mechanism for NAD ⁺ -based therapy for heart failure. The data also show that NAD ⁺ -based therapy can be useful in pre-existing heart failure.
Chapter
Mechanical stretch activates several intracellular signalling networks and modulates gene expressions which initiate structural and functional remodelling in cardiomyocytes. Various durations, loads and frequencies of mechanical forces induce differing response mechanisms. Cardiomyocytes remodel in response to mechanical stress both during cardiac development as well as in disease conditions. In both of these situations, the cytoskeleton has a key role in sensing mechanical stress and in mediating adaptive or maladaptive changes within the cardiomyocyte. Cytoskeleton which is highly sensitive and adaptive to mechanical forces acts as a signal integrator for mechanical and structural inputs. The information is transmitted throughout the cardiomyocyte to ensure that the cell respond and adapt appropriately. Mechanical stretch induces secretion or synthesis of molecules with autocrine and paracrine effects. Energy metabolism is also altered in adult cardiomyocytes, which hypertrophy in response to mechanical stress.
Article
Full-text available
A new method of total RNA isolation by a single extraction with an acid guanidinium thiocyanate-phenol-chloroform mixture is described. The method provides a pure preparation of undegraded RNA in high yield and can be completed within 4 h. It is particularly useful for processing large numbers of samples and for isolation of RNA from minute quantities of cells or tissue samples.
Article
Full-text available
Prolonged deprivation of food induces dramatic changes in mammalian metabolism, including the release of large amounts of fatty acids from the adipose tissue, followed by their oxidation in the liver. The nuclear receptor known as peroxisome proliferator-activated receptor α (PPARα) was found to play a role in regulating mitochondrial and peroxisomal fatty acid oxidation, suggesting that PPARα may be involved in the transcriptional response to fasting. To investigate this possibility, PPARα-null mice were subjected to a high fat diet or to fasting, and their responses were compared with those of wild- type mice. PPARα-null mice chronically fed a high fat diet showed a massive accumulation of lipid in their livers. A similar phenotype was noted in PPARα-null mice fasted for 24 hours, who also displayed severe hypoglycemia, hypoketonemia, hypothermia, and elevated plasma free fatty acid levels, indicat- ing a dramatic inhibition of fatty acid uptake and oxidation. It is shown that to accommodate the increased requirement for hepatic fatty acid oxidation, PPARα mRNA is induced during fasting in wild- type mice. The data indicate that PPARα plays a pivotal role in the management of energy stores during fasting. By modulating gene expression, PPARα stimulates hepatic fatty acid oxidation to supply sub- strates that can be metabolized by other tissues.
Article
Full-text available
A mitochondrial protein called uncoupling protein (UCP1) plays an important role in generating heat and burning calories by creating a pathway that allows dissipation of the proton electrochemical gradient across the inner mitochondrial membrane in brown adipose tissue, without coupling to any other energy-consuming process1. This pathway has been implicated in the regulation of body temperature, body composition and glucose metabolism2. However, UCP1-containing brown adipose tissue is unlikely to be involved in weight regulation in adult large-size animals and humans living in a thermoneutral environment (one where an animal does not have to increase oxygen consumption or energy expenditure to lose or gain heat to maintain body temperature), as there is little brown adipose tissue present3. We now report the discovery of a gene that codes for a novel uncoupling protein, designated UCP2, which has 59% amino-acid identity to UCP1, and describe properties consistent with a role in diabetes and obesity. In comparison with UCP1, UCP2 has a greater effect on mitochondrial membrane potential when expressed in yeast. Compared to UCP1, the gene is widely expressed in adult human tissues, including tissues rich in macrophages, and it is upregulated in white fat in response to fat feeding. Finally, UCP2 maps to regions of human chromosome 11 and mouse chromosome 7 that have been linked to hyperinsulinaemia and obesity. Our findings suggest that UCP2 has a unique role in energy balance, body weight regulation and thermoregulation and their responses to inflammatory stimuli.
Article
Full-text available
Endurance exercise training has been shown to decrease diet-induced thermogenesis (DIT) in rats and humans. In rodents, most thermogenesis is thought to occur in brown adipose tissue via activation of the uncoupling protein-1 (UCP1) and in skeletal muscle. Since the level of UCP1 mRNA in rat BAT was reported to be unmodified by exercise training, the newly described uncoupling proteins UCP2 and UCP3 could be responsible for the decreased DIT in trained rats. UCP3 mRNA levels in endurance-trained rats were found to be reduced by 76% and 59% in tibialis anterior and soleus muscles, respectively. UCP2 mRNA levels were also decreased in tibialis anterior and in heart by 54% and 41%, respectively. Neither white adipose tissue UCP2 nor brown adipose tissue UCP1, UCP2, and UCP3 mRNA levels were modified. The results of this study show that a need for a higher metabolic efficiency is associated with decreased mRNA expression of the uncoupling proteins in skeletal and heart muscles, which would decrease energy dissipation in these tissues. The down-regulation of UCP3 and UCP2 expressions might also contribute to the rapid weight gain known to occur when exercise training ceased.
Article
Full-text available
To determine the mechanism of the cardiac dilatation and reduced contractility of obese Zucker Diabetic Fatty rats, myocardial triacylglycerol (TG) was assayed chemically and morphologically. TG was high because of underexpression of fatty acid oxidative enzymes and their transcription factor, peroxisome proliferator-activated receptor-α. Levels of ceramide, a mediator of apoptosis, were 2–3 times those of controls and inducible nitric oxide synthase levels were 4 times greater than normal. Myocardial DNA laddering, an index of apoptosis, reached 20 times the normal level. Troglitazone therapy lowered myocardial TG and ceramide and completely prevented DNA laddering and loss of cardiac function. In this paper, we conclude that cardiac dysfunction in obesity is caused by lipoapoptosis and is prevented by reducing cardiac lipids.
Article
This study invesigatec whether metaboiic condition known to after the activity and phosphorylation sate ofthe pyranvale dehydrog. Chferentiation, and apoptosis. Investigation of this bypotheis haidentified a sphingomyeling cycle, wlerey the action of a number of agoaist load is sphingortyelin aydroysis and ceranale genecation. Once geterated cearateide are throughia ceramide activated protem pho-phatade (CAPP)to dlich its dowtist team effects. Although, CAPP is a prime candidate for me diating cetamice signaling in mammalian cells. Its identily and its rennection to the down-tream targets of revamide activation remain eladive. Moleculat characterization of CAPP is therfore. a prelnde to a better inderstanding of the mechanist y which ceramide causes its downstream effects. our study demonstraten that at ImAl concentration some divaten cation, such as Mgappear to have a stimuarorv effect on both basal a CAPP phosphatase intivities. while others, such as Mn2+ and Zn2+, were inhibitony towards both phosphatase activities. Moreover, ather divalent cations werw lested and it was found that all were ingibitory towards CAPP activiy. On the contary monavalent cations had no effect on CAPP, inducd, they actnally stimulated basal phosphatase artivities. When EDTA and FGTA wree tested. It was observed that EDLA, dose dependently, abolished CAPP activity, while it had aecffect npon basal phosphatase activity. However. EGTA was much less protin in inhilating CAPP. From theae data, it is possible to condude that CAPP is a metal dependent protein and that the metal colactor is must Edely not Ca 2+. These findings indiacate that regulation of expression of PDK4 is an important control mecmnism for rong term control of the activity of the pyruvate dehyarogeuasw cemplex it tatheart.
Article
Both natural (laurate) and artificial (m-chlorocarbonylcyanide phenylhydrazone; CCCP) uncouplers strongly inhibit O⋅−2 and H2O2 formation by rat heart mitochondria oxidizing succinate. Carboxyatractylate, an ATP/ADP antiporter inhibitor, abolishes the laurate inhibition, the CCCP inhibition being unaffected. Atractylate partially releases the inhibition by laurate and decelerates the releasing effect of carboxyatractylate. GDP is much less effective than carboxyatractylate in releasing the laurate inhibition of reactive oxygen species (ROS) formation. Micromolar laurate concentrations arresting the ROS formation cause strong inhibition of reverse electron transfer from succinate to NAD+, whereas State 4 respiration and the transmembrane electric potential difference (ΔΨ) level are affected only slightly. It is suggested that (i) free fatty acids operate as natural `mild uncouplers' preventing the transmembrane electrochemical H+ potential difference (ΔμH+) from being above a threshold critical for ROS formation by complex I and, to a lesser degree, by complex III of the respiratory chain, and (ii) it is the ATP/ADP-antiporter, rather than uncoupling protein 2, that is mainly involved in this antioxidant mechanism of heart muscle mitochondria.