Uploaded by Denis Souza

dnp metabolic

advertisement
obesity reviews
Mitochondrial uncoupling as a target for drug
development for the treatment of obesity
J. A. Harper1,2, K. Dickinson3 and M. D. Brand1
1
Summary
Cambridge CB2 2XY, UK; 2Department of
Mitochondrial proton cycling is responsible for a significant proportion of basal
or standard metabolic rate, so further uncoupling of mitochondria may be a good
way to increase energy expenditure and represents a good pharmacological target
for the treatment of obesity. Uncoupling by 2,4-dinitrophenol has been used in
this way in the past with notable success, and some of the effects of thyroid
hormone treatment to induce weight loss may also be due to uncoupling. Diet
can alter the pattern of phospholipid fatty acyl groups in the mitochondrial membrane, and this may be a route to uncoupling in vivo. Energy expenditure can be
increased by stimulating the activity of uncoupling protein 1 (UCP1) in brown
adipocytes either directly or through b3-adrenoceptor agonists. UCP2 in a number
of tissues, UCP3 in skeletal muscle and the adenine nucleotide translocase have
also been proposed as possible drug targets. Specific uncoupling of muscle or
brown adipocyte mitochondria remains an attractive target for the development
of antiobesity drugs.
MRC Dunn Human Nutrition Unit, Hills Road,
Biochemistry, University of Cambridge, 80
Tennis Court Road, Cambridge CB2 1GA, UK;
3
Knoll Ltd, Pennyfoot Street, Nottingham NG1
1GF, UK
Received 14 May 2001; revised 12 June
2001; accepted 15 June 2001
Address reprint requests to: Dr Martin Brand,
MRC Dunn Human Nutrition Unit, Hills Road,
Cambridge CB2 2XY, UK.
E-mail: martin.brand@mrc-dunn.cam.ac.uk
Keywords: Uncoupling, Mitochondria, Standard Metabolic Rate, Obesity.
obesity reviews (2001) 2, 255–265
Pharmaceutical interest in uncoupling
Obesity is a disease resulting from a prolonged positive
imbalance between energy intake and energy expenditure
resulting in the storage of fat. The rapidly increasing worldwide incidence of obesity and its association with serious
comorbid diseases means it is beginning to replace undernutrition and infectious diseases as the most significant
contributor to ill health in the developed world (1). Weight
loss, induced by dieting, has been shown to be successful
in reducing the health consequences of obesity but unfortunately >90% of individuals who lose weight through
dietary control eventually return to their original weight
(2). Pharmacological treatment may therefore be desirable
for those patients with associated comorbid conditions
who have been unable to control their obesity through diet
and exercise.
Any treatment for obesity has to reduce energy intake,
increase energy expenditure or combine both effects.
Current therapies for obesity predominantly lead to
decreased energy intake either by acting at satiety centres in the brain (e.g. sibutramine) (3–5) or by reducing
the efficiency of intestinal absorption (e.g. orlistat) (6,7).
In addition to reducing energy intake, sibutramine increases standard metabolic rate (SMR) profoundly in rodents,
but increased energy expenditure appears to be only a
minor component of its activity in humans (8,9). Exercise is the most practical and potentially easiest way
to increase energy output. Studies have shown a sevenfoldincreased risk of the incidence of overweight in those
with a physical activity ratio (total energy expenditure: resting metabolic rate) of <1.8 (10). The main
benefit of exercise is to increase resting metabolic rate,
and overall energy expenditure, by a greater amount
than that resulting directly from the exercise (11).
Pharmacological agents that increase metabolic rate by
increasing uncoupling of mitochondrial oxidative phosphorylation are likely to mimic this beneficial effect of
exercise on resting metabolic rate and could provide
a useful adjunct to agents acting to reduce satiety;
© 2001 The International Association for the Study of Obesity. obesity reviews 2, 255–265
255
256 Mitochondrial uncoupling as a treatment for obesity
J. A. Harper et al.
this review will consider how such uncoupling can be
achieved.
As discussed below, uncoupling of mitochondria represents a particularly attractive target, as there is already
excellent proof of concept for this approach in humans
using 2,4-dinitrophenol (DNP) (12) and in animals using
b3-adrenoceptor agonists (13), or overexpression of UCP3
(uncoupling protein 3) (14). However, there are problems
associated with the use of chemical uncouplers like DNP.
Many of these problems may result from inappropriate
activities in critical tissues, and more selective uncoupling
would be desirable.
Skeletal muscle represents a particularly attractive
target for directed uncoupling due to the large muscle
mass, which accounts for approximately 15–20% of SMR
(15,16). The maximal aerobic capacity of a human is
generally estimated to be up to 12 times SMR in untrained
subjects (17). Most of this increase can be directly attributed to skeletal muscle respiratory activity and it is clear
that muscle can greatly increase its metabolic activity.
Doubling metabolic rate by modestly uncoupling skeletal
muscle should produce few adverse side-effects as this
increase would only be equivalent to mild exercise (actually equivalent to approximately the difference between
lying down and standing up (17)). Indeed, support for this
view has been obtained using proteins that may naturally
uncouple mitochondria (uncoupling proteins 1 and 3).
High expression of human UCP3 in mouse skeletal muscle
led to decreased weight gain despite increased food intake
(14), and expression of UCP1 in mouse skeletal muscle led
to improvements in insulin sensitivity and resistance to
obesity on a high fat diet (18).
Standard metabolic rate
Basal metabolic rate (BMR) is the minimal calorific requirement for normal life in an organism in the absence of
external stimulation, work and growth. It is measured
under rigorous conditions at thermoneutrality, in a postabsorptive state. Many physiological processes continue
in this minimal state, including ventilation and blood circulation. At the cellular level, the reactions that make up
BMR include protein turnover, ion cycling across the
plasma membrane, turnover of nucleic acids and lipids, and
proton cycling across the mitochondrial inner membrane
(uncoupling) (19,20). Resting metabolic rate (RMR) is
measured as BMR but not in the post-absorptive state (17).
These measurements are not suitable in animals (particularly ectotherms), therefore standard metabolic rate (SMR)
is used. SMR is measured under defined conditions which
are not necessarily those of BMR; in particular, temperature may vary. SMR varies considerably between species.
It is primarily dependant on body mass to approximately
the 0.75th power (21,22), so mass-specific SMR decreases
obesity reviews
as body mass increases (22,23). For example, the SMR per
gram of a mouse (body mass 0.05 kg) is approximately 10fold higher than the SMR per gram of a horse (body mass
500 kg). The field metabolic rate (FMR) is the metabolic
rate of an animal in its natural environment. Factors such
as digestion, hunting, reproduction, growth and temperature regulation increase the metabolic rate around twofold
in humans and around fourfold in other mammals (20),
irrespective of body size. In other words, around one
quarter of field energy expenditure in mammals is due to
SMR (21,24,25). The differences in mass-specific SMR
between species indicate the existence of regulatory mechanisms that may be amenable to pharmacological manipulation. Some of these differences in SMR may be caused by
differences in mitochondrial proton cycling.
What is uncoupling?
Mitochondria are normally responsible for 90% of cellular
oxygen consumption and the majority of adenosine triphosphate (ATP) production. The flow of electrons from reduced
substrate to oxygen is coupled by a proton electrochemical
gradient across the mitochondrial inner membrane to the
synthesis of ATP from adenosine diphosphate (ADP) and
phosphate (Fig. 1). This process of oxidative phosphorylation can be subdivided into two distinct parts: the generation
of the proton electrochemical gradient by the respiratory
chain and the synthesis of ATP by the Fo-F1 ATP synthase
using the potential energy stored in the gradient. However,
not all of the available energy is coupled to ATP synthesis.
Instead, much is lost by uncoupled reactions when protons
move from the cytosol back into the mitochondrial matrix
via pathways which circumvent the ATP synthase and other
uses of the electrochemical gradient. There are two sorts of
uncoupling. Basal uncoupling is not acutely regulated and is
present in all mitochondria, whereas inducible proton conductance is catalysed by proteins, tightly regulated, and
found in discrete cell types (Fig. 1).
Physiological significance of mitochondrial
proton cycling
Proton cycling is a major contributor to SMR, responsible
for around 26% of the oxygen consumed in resting rat
hepatocytes (26) and about 52% in perfused, resting rat
skeletal muscle (15). Multiplication of these values by the
contribution of each tissue to SMR suggests that proton
cycling accounts for 20–25% of rat SMR (15). This makes
proton cycling the largest single contributor to SMR.
Under field conditions, ATP turnover increases significantly and the ATP synthase competes with proton leak for
the same proton electrochemical energy. It is important to
discover whether proton cycling still represents a significant proportion of energy expenditure when ATP synthe-
© 2001 The International Association for the Study of Obesity. obesity reviews 2, 255–265
obesity reviews
Mitochondrial uncoupling as a treatment for obesity J. A. Harper et al.
257
Figure 1 Chemiosmotic proton circuits
across the inner membrane of isolated
mitochondria. Substrate oxidation consists of
substrate transport, substrate metabolism
and the electron transport chain, and leads
to proton pumping from the matrix to the
intermembrane space, setting up a proton
electrochemical gradient. In the lower
circuit, return of protons is coupled to
adenosine triphosphate (ATP) production via
the ATP synthase. The upper circuits show
uncoupled proton leak through the basal
leak pathway or through an inducible leak
pathway (represented by a protein, such as
an uncoupling protein (UCP)). Uncoupling
by 2,4-dinitrophenol effectively increases the
basal leak pathway whereas uncoupling by
UCPs increases the inducible pathway.
sis is increased. Rolfe and co-workers (16) showed that
when respiration rate was doubled by stimulating ureagenesis and gluconeogenesis in hepatocytes and muscle contraction in perfused rat hindquarters, the rate of proton
cycling stayed fairly constant. Of course, proton cycling
dropped as a proportion of total respiration rate, but only
to 22% in hepatocytes and 34% in muscle (16). Estimates
from these experiments suggest that proton cycling in
animals under more plausibly physiological conditions
uses around 15–20% of total energy consumption and so
remains a substantial proportion of SMR.
SMR varies with body mass, and proton cycling is an
important component of SMR. Does proton cycling change
in parallel with SMR, or does it change less or more than
SMR? Components of SMR such as the urinary excretion
of endogenous nitrogen and sulphur (27) and the rates of
respiration, circulation and renal activity (28,29) remain a
constant proportion of SMR as body mass varies. Only
relatively recently has the contribution of proton cycling to
SMR been examined in animals of different body mass.
Porter and Brand (30) found that proton conductance in
liver mitochondria decreased with increasing body mass in
mammals. However, the proportion of energy expended via
proton cycling was approximately the same in hepatocytes
from all mammals (31,32). Around 70% of the proton
conductance difference was because of less mitochondrial
inner membrane area in cells from larger animals (33). The
remaining difference was some intrinsic property of the
membrane. Because this intrinsic property can be altered
according to body mass it might be modifiable by drugs. If
proton leak could be stimulated in some way, then more
energy would be dissipated during synthesis of ATP. This
© 2001 The International Association for the Study of Obesity. obesity reviews 2, 255–265
258 Mitochondrial uncoupling as a treatment for obesity
J. A. Harper et al.
partial uncoupling would cause an increase in SMR and an
alteration in the balance between energy input and output
if energy intake did not rise to compensate. At comparable
energy intakes, a 1% change in BMR in humans would
lead to loss or gain of about 1 kg of adipose tissue per year,
so over a long period it could cause or combat obesity.
Mechanism of proton leak: passive diffusion
To consider how to increase mitochondrial uncoupling as
a means to increase BMR and reduce obesity, it would be
helpful to understand how protons re-enter the mitochondrial matrix without passing through the ATP synthase.
Diffusion of protons through the phospholipid bilayer is
one possible mechanism of basal proton conductance. To
judge the physiological importance of this process, phospholipids from mitochondria with different proton conductance were extracted and reformed into liposomes
and the proton conductance of the bilayer was examined
(34). This experiment allowed two issues to be resolved;
first, the percentage of proton conductance that could
be explained by proton diffusion across the bilayer; and
second, the effect of the phospholipid fatty acyl composition of the bilayer on the conductance. The results showed
that passive diffusion in liposomes could account for only
2.5% to 25% of the proton conductance of mitochondria
with the same phospholipid fatty acyl composition, implying that some other property of the membrane, such as the
presence of proteins, was important in determining the
conductance. There was no significant difference in proton
conductance between liposomes with very different phospholipid fatty acyl compositions, despite a known relationship between the phospholipid fatty acyl composition
of intact mitochondria and proton conductance (34).
Perhaps acyl composition is important in mitochondria,
but loss of phospholipid asymmetry or of some specific
protein causes the effect to be lost in liposomes.
Mitochondrial membranes containing a higher ratio of
polyunsaturated fatty acyl groups (PUFA) to monounsaturated fatty acyl groups (MUFA) may allow a higher molecular activity of membrane proteins, so membrane acyl
composition could affect metabolic rate. In particular,
n-3 PUFAs have been strongly linked to SMR (35). As
described above, mammals with a smaller body mass have
higher mass-specific SMR and higher mitochondrial proton
conductance than larger mammals. Surface area and fatty
acyl composition of mitochondria both vary with body
mass (33,35), so either factor could affect proton conductance. Around two-thirds of the difference in proton conductance of mitochondria from mammals of different body
mass is due to the amount of mitochondrial membrane, and
one-third is due to some difference (protein or phospholipid) in membrane composition (33). Dietary fatty acids
can alter the fatty acyl composition of the mitochondrial
obesity reviews
inner membrane (36), and it is conceivable that diet, or
pharmaceutical agents that alter the PUFA : MUFA ratio
of fatty acyl groups in the membrane, could affect mitochondrial uncoupling and hence modify BMR and weight
gain. For example, fish oil high in n-3 PUFA has been suggested to limit obesity (37,38). Oils that contain high levels
of n-3 fatty acids and purport to be beneficial are already
on sale, although evidence that they effect weight loss is
lacking (39).
Artificial uncoupling by dinitrophenol
Uncoupling of mitochondria as a treatment for obesity is not
unprecedented; the artificial uncoupler 2,4-dinitrophenol
(DNP) has been used for this purpose for many years
(12,40). DNP is a lipid-soluble weak acid which acts as a
protonophore because it can cross membranes protonated,
lose its proton and return as the anion, then reprotonate
and repeat the cycle. In this way, it increases the basal
proton conductance of mitochondria and uncouples. The
effect of DNP derivatives was first noted in 1885 when
scientists saw thermogenic effects of Martius Yellow (a
dinitro-alpha-napthol), a coal tar dye used in the 19th
century to give the impression that food was rich in eggs
(41).
DNP was introduced as a drug in the 1930s and used
with considerable success, though reports of side-effects
(cataracts) and some deaths from overdose led to it being
chased off the market by the US Food and Drug Administration (FDA) in 1938. This use of DNP to treat obesity
was stimulated by observations of its toxicity in French
munitions workers during World War I (the French commonly used a mixture of 40% dinitrophenol and 60%
trinitrophenol for their munitions). In animals, it was
shown that the drug promoted a direct stimulation of cellular respiration and a consequent rise in body temperature. It led to the almost immediate onset of rigor mortis
when death was promoted by large doses (42,43).
A series of controlled trials in obese patients were
prompted by these promising mode of action studies. The
most extensive were carried out during the 1930s at Stanford University, CA, USA (44–47), though others also performed careful studies (48). Interpretation of this work can
be complicated, as the doses of DNP had to be optimized
for each patient (due to the steep dose response and patient
to patient variability) and were usually increased as the
studies progressed. This means the DNP-sensitive patients
on low doses of the drug would show enhanced efficacy
compared with the population (and vice versa for patients
on high does of DNP). However, there was a clear dependence of metabolic rate on DNP dose (Fig. 2). There was
an average 11% increase in metabolic rate for each dosage
increment of 0.1 g of DNP (44,48). Doses up to 0.5 g
(about 5 mg kg-1) were generally well-tolerated apart from
© 2001 The International Association for the Study of Obesity. obesity reviews 2, 255–265
obesity reviews
Mitochondrial uncoupling as a treatment for obesity J. A. Harper et al.
Figure 2 Effects of dinitrophenol on metabolic rate and weight loss in
humans. Replotted from data given in (44).
patients almost always reporting a feeling of warmth
together with increased perspiration (45,46,48). Between
about 5–10 mg kg-1, patients reported profuse sweating
but, surprisingly, there was no evidence of increased body
temperature or heart rate. Doses above 10 mg kg-1 led to
increased heart rate, respiration rate and excessive body
temperature rises (45). Single doses of 3–5 mg kg-1 produced an increase in resting metabolic rate of 20–30%
within the first hour. This was maintained for 24 h after
which a gradual fall became apparent. Daily dosing of
3–5 mg kg-1 produced a gradual increase of efficacy: an
average 40% increase in metabolic rate was observed after
a few weeks that was then maintained, with no sign of tolerance, for at least 10 weeks (45).
Weight loss in these studies, where no attempt was made
to control diet, was reported to be variable. Of 170 treated
patients, a mean of 7.8 kg weight loss was recorded, averaging 0.64 kg week-1 (44) (Fig. 2). There were no clearly
reported effects on food consumption. In contrast to the
use of thyroid extract (also in common use at the time
to treat obesity), DNP did not promote urinary nitrogen
excretion, so the assumption was made that weight loss
could be attributed to a specific loss of fat (47). One potential complication in the interpretation of weight loss data
is the ability of the drug to promote oedema. This effect
was reported to account for a common apparent failure to
continue to lose weight in the face of a maintained increase
in metabolic rate (48). For some patients, withdrawing
DNP led to a rapid weight loss that was attributed to loss
of excess body water after which DNP dosing could be
resumed with its former effectiveness. Therapeutic doses of
DNP had no effects on blood pressure or heart rate in
normal patients. Interestingly, a subset of 30 hypertensive
patients exhibited average falls of 9.4% in systolic and
12.6% in diastolic blood pressure (44). These improvements in hypertension were also noted at doses of DNP
insufficient to cause weight loss (48). Some studies were
also performed in diabetic patients with inconsistent results
259
but there did appear to be improvements in glucose tolerance after long-term dosing (48). The reported potency of
DNP to treat obesity (and associated comorbid conditions)
in these early trials compares well with current treatments
for obesity (3,4,6).
This ability of dinitrophenol to produce good reductions
in body weight, without the need for dietary restriction, led
to its widespread use to treat obesity. In the absence of
formal regulatory controls it is not surprising that it was
soon prescribed by inexperienced physicians with no access
to the metabolic rate measurements necessary to determine
optimal doses. DNP was even included in a variety of
‘antifat nostrums’ that could be used by the public without
medical consultation. By 1934 it was estimated that a total
of 100 000 patients had been treated (40).
Given the steep dose dependence of metabolic rate and
the widespread use of DNP it is perhaps not surprising that
a number of people were ‘literally cooked to death’ in the
1930s due to accidental or deliberate overdose (40). It was
argued at the time that the reported deleterious effects of
DNP were remarkably few (when given at the correct therapeutic doses), given the large number of patients who had
taken it. The authors did, however, note a 7% incidence of
severe skin rashes, necessitating discontinuation of treatment. There was no overt liver or kidney damage, but the
same authors voiced some concerns about the incidence of
agranulocytosis though they saw no cases in their own
long-term studies (44). Of more concern, a number of cases
of cataracts were reported in women in 1935 (49,50). In
1938 the FDA acquired more powers to prosecute manufacturers of misbranded therapies and announced that the
use of a variety of patent medicines (including DNP) could
lead to prosecution (12). These threats led to a withdrawal,
in 1938, of the DNP-containing nostrums from the market
as well as an end to the official clinical use of DNP. Interestingly, however, there are reports on the Internet that
describe the use of DNP by US clinics (who avoided illegal
interstate transportation of DNP for human use by synthesizing it within each state), and give detailed protocols
for its use amongst the designer drug community of bodybuilders and those willing to risk self-administration of
DNP to lower body mass.
Given the age of the publications describing the mode
of action in rodents, one of us (KD) recently re-evaluated
the effects of DNP in rats and confirmed the relative insensitivity of this species and the particularly steep dose
response to increase metabolic rate. In contrast to humans,
DNP did not significantly increase metabolic rate in rats
when dosed orally at 10 mg kg-1 (data not shown) but a
good, comparatively long lasting, increase was seen at
30 mg kg-1 that was comparable in magnitude to a rodent
b3-adrenoceptor agonist (Fig. 3) (51). Doses of 100 mg kg-1
produced an escalating hyperthermia that necessitated
killing the animals to avoid distress.
© 2001 The International Association for the Study of Obesity. obesity reviews 2, 255–265
260 Mitochondrial uncoupling as a treatment for obesity
obesity reviews
J. A. Harper et al.
Figure 3 Effect of dinitrophenol (30 mg kg-1)
and the b3-adrenoceptor agonist ZD7114
(1 mg kg-1) on oxygen consumption in female
Wistar rats (mean ± SEM, n = 8). Oxygen
consumption was measured using indirect
closed circuit calorimetry at the thermoneutral
temperature of the rat (29°C). Animals were
temporarily removed from the chambers for
oral dosing with drugs or vehicle at 100 min
(51).
Thyroid hormones
Thyroid hormones have a long history in the treatment
of obesity (52) and are positively correlated with BMR
(53,54). At the cellular level, hepatocytes isolated from
hyperthyroid rats have twice the respiration rate of euthyroid controls. About half of the increase is caused by
increased mitochondrial proton cycling, which is partly
due to greater proton conductance and partly to a greater
driving force (proton electrochemical gradient) (55). At the
subcellular level, mitochondria prepared from the liver of
hyperthyroid animals have increased proton permeability
compared with those from euthyroid animals (56,57).
In the 1950s the respiratory stimulation in isolated
mitochondria was attributed to direct uncoupling by thyroid hormones using the same mechanism as DNP (58).
However, this effect only occurred in vitro with supraphysiological concentrations of hormones (59). There are
now two main hypotheses for the stimulation of respiration and proton conductance in mitochondria and cells by
thyroid hormones (60).
In the first hypothesis, thyroid hormones act directly at
the membrane level, rigidifying the bilayer and leading to
a compensatory change in the fatty acyl composition of the
phospholipids. Alternatively, thyroid hormones could alter
membrane composition through transcriptional regulation
of desaturases and other enzymes. In either case, the change
in phospholipid fatty acyl composition alters the proton
conductance and degree of uncoupling of the mitochondrial inner membrane. In support of this hypothesis, the
polyunsaturation of mitochondrial phospholipid acyl
chains increases in hyperthyroidism (36,60,61), and liver
mitochondrial proton conductance correlates with polyunsaturation of mitochondrial phospholipid acyl chains
(33,62). Hyperthyroid mitochondria also have less cholesterol and increased cardiolipin (63). Changes in the area of
mitochondrial inner membrane are also implicated in the
change in measured proton conductance (57).
In the second hypothesis, thyroid hormones act through
transcriptional regulation and affect proton conductance
through expression of specific proteins (64). For example,
the levels of UCP2 and UCP3 mRNA increase in response
to thyroid treatment (65–69).
Supra-physiological doses of thyroid hormones are
no longer used in the treatment of obesity because of
unwanted side-effects such as tachycardia, increased heart
weight (70,71), thyroid atrophy (72) and a negative nitrogen balance, that is a loss of lean body mass (muscle)
(73,74). They may cause loss of water and muscle rather
than loss of fat and adipose tissue.
Inducible uncoupling catalysed by
specific proteins
Uncoupling protein 1
The ability of hibernators, cold-adapted rodents and
new-born mammals to produce heat and increase SMR by
non-shivering thermogenesis results mostly or only from
the activity of uncoupling protein 1 (UCP1) in mitochondria in brown adipose tissue (BAT) (75,76). The identification of UCP1 as the mediator of uncoupling followed from
© 2001 The International Association for the Study of Obesity. obesity reviews 2, 255–265
obesity reviews
Mitochondrial uncoupling as a treatment for obesity J. A. Harper et al.
the observation of greater uncoupling of BAT mitochondria
than of liver and heart mitochondria (77,78). Importantly,
this increased uncoupling was abolished by albumin and
by purine nucleotides, indicating that it could be regulated
in vivo by free fatty acids and nucleotides (79). The ability
of UCP1 to transport protons was demonstrated by mitochondrial swelling experiments (77,79). A 32-kDa protein
was observed to be specifically and highly expressed in BAT
mitochondria. Photo-affinity labelling identified a 32-kDa
protein as the binding site of purine nucleotides and a putative uncoupling protein was proposed (80). Subsequently,
the 32 kDa protein was purified from hamster and rat
(81,82). The cDNA was sequenced and cloned and the
protein’s proton translocating activity was demonstrated in
transgenic yeast mitochondria (83) and liposomes (84–87).
After these experiments it was generally accepted that
UCP1 was responsible for the regulated uncoupling of BAT.
Further experiments were performed with transgenic
mice (88). A mouse model in which all UCP1 had been
removed by homologous recombination was created by
Enerbäck and co-workers (89). These UCP1-deficient mice
were more sensitive to cold and showed a greatly reduced
effect of b3-adrenoreceptor agonists, indicating reduced
thermogenic capacity. However, when they were fed
normal or high-fat diets they were not obese. There was an
increase in the adiposity of the BAT, as expected if it did
not have to oxidize fatty acids as fast to maintain a normal
proton electrochemical gradient. Interestingly, other studies
in transgenic mice using a diphtheria toxin gene linked
to a UCP1 promoter to genetically ablate BAT did show
obesity linked to loss of BAT activity (90). As the obesity
in this UCP1-DTA model seemed to result predominantly
from hyperphagia it appears that the differences between
the two models resides in other (non-thermogenic) activities of BAT. Other studies using targeted expression of
UCP1 to mouse muscle (18) or white fat (91) demonstrated
a resistance to the development of obesity and an improvement in comorbid conditions consistent with increased
uncoupling activities in these tissues.
b3-Adrenergic receptor agonists
There has been considerable interest within the pharmaceutical industry in developing specific b3-adrenoceptor
agonists that would selectively lead to the activation of
uncoupling through UCP1 in brown fat. The ability to
selectively uncouple should avoid many of the side-effects
that might occur with DNP. The activity of the b3-adrenoceptor to activate lipolysis in both white and brown fat
leading to a consequent activation of UCP1 mediated lipolysis in brown fat is now well understood (92–94). Numerous studies in rodents have shown that b3-adrenoceptor
agonists produce profound improvements in insulin sensitivity in a number of diabetic animal models and lead to
261
substantial weight loss due to selective fat loss (13,95).
These rodent studies provide excellent evidence that activation of uncoupling by brown fat represents an important
potential human therapeutic target. Unfortunately, clinical
studies have yet to provide proof of concept in humans,
though clinical trials are still in progress. It has proved particularly difficult to develop b3-adrenoceptor agonists that
lack activity against b1 and b2-adrenoceptors (13,95). This
difficulty, coupled with the very low levels of brown fat in
adult humans has made the results of the various clinical
trials difficult to interpret. However, if b3-adrenoceptor
agonists prove capable of reactivating dormant brown fat,
or if conversion of white to brown adipose tissue becomes
possible, then a suitably selective agent may show relevant
activity in humans.
Uncoupling proteins 2 and 3
UCP1 is restricted to brown adipocytes, and was originally
thought to be the only transporter capable of uncoupling
mitochondria (96–98). However, UCP1 probes occasionally gave weak signals in other tissues (93), suggesting the
presence of homologues. UCP2 was discovered because of
its relatively high sequence identity to UCP1 (59%). UCP2
mRNA has been found in many tissues, including cardiac
muscle, BAT, white adipose tissue, skeletal muscle, kidney,
non-parenchymal liver cells, lung, placenta, pancreatic b
cells and the immune system. However, mRNA expression
does not necessarily imply protein expression, as UCP2
protein was not detected in heart, skeletal muscle, liver or
BAT (99). UCP3 was also discovered by database searching. Human UCP3 is 59% identical to human UCP1 and
72% identical to human UCP2. UCP3 is restricted to BAT
and skeletal muscle (100), both tissues that make a major
contribution to thermogenesis, making it a more enticing
target for drug development than UCP2.
Evidence that UCP2 and UCP3 are responsible for basal
proton conductance comes from experiments employing
high expression of mammalian UCP2 and UCP3 in yeast
(101–103) and transgenic overexpression of human UCP3
in mice (14). Such high expression lowers mitochondrial
membrane potential and increases state 4 respiration, indicating uncoupling. It inhibits the growth of yeast and
protects against obesity in hyperphagic mice. However,
the normal concentrations of UCP2 (99) and UCP3
(104,105)protein expressed in mammalian tissues are very
low. The amounts expressed in transgenic yeast (104) or
UCP3-overexpressing mice (14) are strongly supra-physiological and there is good evidence that the uncoupling is an
artefact not related to the physiological functions of UCP2
and UCP3 (105,106) (S. Cadenas et al. unpublished data; J.
A. Harper et al. unpublished data). Proteoliposomes have
also been used in studies of the function and regulation of
UCP1 and its homologues (107–110). These studies found
© 2001 The International Association for the Study of Obesity. obesity reviews 2, 255–265
262 Mitochondrial uncoupling as a treatment for obesity
J. A. Harper et al.
different nucleotide sensitivities and low turnover numbers,
but they did identify ubiquinone as an essential cofactor for
uncoupling in vitro by UCP1 and its homologues (111,112).
The UCPs have been knocked out in mice, avoiding interpretational problems resulting from the requirement for
overexpressed protein to be correctly inserted and folded
in membrane. Two studies (113,114) found that UCP2
mRNA was increased in BAT of UCP1 knockout mice with
no increased uncoupling. However, UCP2 protein was not
measured and may not have changed. The proton conductance of skeletal muscle mitochondria from UCP3 knockout mice was decreased (115,116), indicating that UCP3 is
responsible for at least some of the proton conductance in
skeletal muscle. However, we have not been able to confirm
these observations (S. Cadenas et al. unpublished data).
Despite the reported effects in genetically modified mice,
natural changes in UCP2 and UCP3 mRNA and UCP3
protein levels do not alter mitochondrial proton conductance. For example, UCP2 and UCP3 mRNA increase in
muscle from starved rats (117) despite a known depression
in thermogenesis (118). UCP3 mRNA increases fourfold
and UCP3 protein doubles but there is no change in the
mitochondrial proton conductance (119).
Taken together, the evidence suggests that UCP1, UCP2
and UCP3 are not responsible for the basal proton conductance of mitochondria. Clearly, UCP1 causes inducible
uncoupling in BAT mitochondria, and UCP2 and UCP3
may yet be found to catalyse a similar uncoupling that is
induced by unidentified agonists. The UCPs remain important potential targets for specific pharmacological uncoupling as a treatment for obesity. Other members of the
mitochondrial carrier family have also been implicated in
the uncoupling of mitochondria. In particular, the adenine
nucleotide transporter is responsible for uncoupling by free
fatty acids (120,121) and AMP (122). These carriers may
also be potential drug targets.
Is the uncoupling of mitochondria a viable
target for drug development?
Uncoupling mitochondria is an effective way to increase
thermogenesis and basal metabolic rate and it can lead
to a substantial reduction in body weight by loss of fat
deposits. This has been shown in humans taking DNP
orally and in transgenic mice overexpressing human UCP3
or UCP1 in muscle. Proof of concept has therefore been
unambiguously established, showing that uncoupling can
increase energy expenditure without compensatory mechanisms increasing food intake to the extent that they nullify
the uncoupling.
However, using pharmacological agents to uncouple
all mitochondria throughout the body may be a high-risk
treatment, because it might compromise energy homeostasis in tissues such as heart and brain. On the other hand,
obesity reviews
active tissues like these may be less susceptible to mild
uncoupling than less active ones like resting muscle or
resting BAT because proton conductance has much less
control over respiration rate in active mitochondria (123).
The small difference between the effective and the fatal
doses of DNP, as well as side-effects resulting from its nonselective actions, mean that it is not itself a suitable antiobesity drug. Tissue selectivity and safety need to be improved.
The UCP3-overexpressing mouse shows that selective
uncoupling of muscle mitochondria is sufficient for a strong
anti-obesity effect. Specific uncoupling of brown adipose
tissue mitochondria through UCP1 or of muscle mitochondria through UCP3 or in some other way remains a
viable and attractive target for the development of drugs
for the treatment of obesity.
Acknowledgements
JAH was supported by a BBSRC CASE award with Knoll
Ltd.
References
1. Kopelman PG. Obesity as a medical problem. Nature 2000;
404: 635–643.
2. Wadden TA. Treatment of obesity by moderate and severe
caloric restriction. Results of clinical research trials. Ann Intern
Med 1993; 119: 688–693.
3. Bray GA, Blackburn GL, Ferguson JM, Greenway FL, Jain AK,
Mendel CM, Mendels J, Ryan DH, Schwartz SL, Scheinbaum ML,
Seaton TB. Sibutramine produces dose-related weight loss. Obes
Res 1999; 7: 189–198.
4. James WP, Astrup A, Finer N, Hilsted J, Kopelman P, Rossner
S, Saris WH, Van Gaal LF. Effect of sibutramine on weight maintenance after weight loss: a randomised trial. STORM Study
Group. Sibutramine Trial of Obesity Reduction and Maintenance.
Lancet 2000; 356: 2119–2125.
5. Ryan DH. Use of sibutramine and other noradrenergic and
serotonergic drugs in the management of obesity. Endocrine 2000;
13: 193–199.
6. Sjostrom L, Rissanen A, Andersen T, Boldrin M, Golay A,
Koppeschaar HP, Krempf M. Randomised placebo-controlled trial
of orlistat for weight loss and prevention of weight regain in obese
patients. European Multicentre Orlistat Study Group. Lancet
1998; 352: 167–172.
7. Hvizdos KM, Markham A. Orlistat: a review of its use in the
management of obesity. Drugs 1999; 58: 743–760.
8. Connoley IP, Lui Y, Frost I, Reckless IP, Heal DJ, Stock MJ.
Thermogenic effects of sibutramine and its metabolites. Br J Pharmacol 1999; 126: 1487–1495.
9. Danforth E Jr. Sibutramine and thermogenesis in humans. Int
J Obes Relat Metab Disord 1999; 23: 1007–1008.
10. Ferro-Luzzi A, Martino L. The Origins and Consequences of
Obesity. Wiley: Chichester, 1996.
11. Poehlman ET. A review: exercise and its influence on resting
energy metabolism in man. Med Sci Sports Exerc 1989; 21:
515–525.
12. Parascandola J, Dinitrophenol and bioenergetics: a historical
perspective. Mol Cell Biochem 1974; 5: 69–77.
© 2001 The International Association for the Study of Obesity. obesity reviews 2, 255–265
obesity reviews
Mitochondrial uncoupling as a treatment for obesity J. A. Harper et al.
13. Weyer C, Gautier JF, Danforth E Jr. Development of
b3-adrenoceptor agonists for the treatment of obesity and
diabetes-an update. Diabetes Metab 1999; 25: 11–21.
14. Clapham JC, Arch JR, Chapman H, Haynes A, Lister C,
Moore GB, Piercy V, Carter SA, Lehner I, Smith SA, Beeley LJ,
Godden RJ, Herrity N, Skehel M, Changani KK, Hockings PD,
Reid DG, Squires SM, Hatcher J, Trail B, Latcham J, Rastan S,
Harper AJ, Cadenas S, Buckingham JA, Brand MD, Abuin A. Mice
overexpressing human uncoupling protein-3 in skeletal muscle are
hyperphagic and lean. Nature 2000; 406: 415–418.
15. Rolfe DFS, Brand MD. Contribution of mitochondrial proton
leak to skeletal muscle respiration and to standard metabolic rate.
Am J Physiol 1996; 271: C1380–C1389.
16. Rolfe DFS, Newman JM, Buckingham JA, Clark MG, Brand
MD. Contribution of mitochondrial proton leak to respiration rate
in working skeletal muscle and liver and to SMR. Am J Physiol
1999; 276: C692–C699.
17. Blaxter K. Energy Metabolism in Animals and Man. Cambridge University Press: Cambridge, 1989.
18. Li B, Nolte LA, Ju JS, Han DH, Coleman T, Holloszy JO,
Semenkovich CF. Skeletal muscle respiratory uncoupling prevents
diet-induced obesity and insulin resistance in mice. Nat Med 2000;
6: 1115–1120.
19. Brand MD. The contribution of the leak of protons across the
mitochondrial inner membrane to standard metabolic rate. J Theor
Biol 1990; 145: 267–286.
20. Hulbert AJ, Else PL. Mechanisms underlying the cost of living
in animals. Annu Rev Physiol 2000; 62: 207–235.
21. Hemmingsen AM. Energy metabolism as related to body size
and respiratory surfaces and its evolution. Rep Steno Mem Hosp
Nord Insulinab 1960; 9: 1–110.
22. Kleiber M. Body size and metabolism. Hilgardia 1932; 6:
315–353.
23. Brody S, Procter RC. Relation between basal metabolism and
mature body weight in different species of mammals and birds.
Univ Miss Agric Exp Sta Res Bull 1932; 166: 89–101.
24. Nagy KA. Field metabolic rate and food requirement scaling
in mammals and birds. Ecol Mono 1987; 57: 111–128.
25. Nagy KA. Field bioenergetics of mammals: what determines
field metabolic rates. Aust J Zool 1994; 42: 43–53.
26. Rolfe DFS, Hulbert AJ, Brand MD. Characteristics of mitochondrial proton leak and control of oxidative phosphorylation in
the major oxygen-consuming tissues of the rat. Biochim Biophys
Acta 1994; 1188: 405–416.
27. Brody S. Bioenergetics and Growth. Hafner: Canada,
1945.
28. Calder WA. Size, Function and Life History. Harvard University Press: London, 1984.
29. Peters RH. The Ecological Implications of Body Size. Cambridge University Press: Cambridge, 1983.
30. Porter RK, Brand MD. Body mass dependence of H+ leak in
mitochondria and its relevance to metabolic rate. Nature 1993;
362: 628–630.
31. Brand MD, Couture P, Else PL, Withers KW, Hulbert AJ. Evolution of energy metabolism. Proton permeability of the inner
membrane of liver mitochondria is greater in a mammal than in a
reptile. Biochem J 1991; 275: 81–86.
32. Porter RK, Brand MD. Causes of differences in respiration
rate of hepatocytes from mammals of different body mass. Am J
Physiol 1995; 269: R1213–R1224.
33. Porter RK, Hulbert AJ, Brand MD. Allometry of mitochondrial proton leak: influence of membrane surface area and
fatty acid composition. Am J Physiol 1996; 271: R1550–R1560.
34. Brookes PS, Hulbert AJ, Brand MD. The proton permeability
of liposomes made from mitochondrial inner membrane phos-
263
pholipids: no effect of fatty acid composition. Biochim Biophys
Acta 1997; 1330: 157–164.
35. Hulbert AJ, Else PL. Membranes as possible pacemakers of
metabolism. J Theor Biol 1999; 199: 257–274.
36. Pehowich DJ. Thyroid hormone status and membrane n-3
fatty acid content influence mitochondrial proton leak. Biochim
Biophys Acta 1999; 1411: 192–200.
37. Belzung F, Raclot T, Groscolas R. Fish oil n-3 fatty acids selectively limit the hypertrophy of abdominal fat depots in growing
rats fed high-fat diets. Am J Physiol 1993; 264: R1111–R1118.
38. Hun CS, Hasegawa K, Kawabata T, Kato M, Shimokawa T,
Kagawa Y. Increased uncoupling protein 2 mRNA in white
adipose tissue, and decrease in leptin, visceral fat, blood glucose,
and cholesterol in KK-Ay mice fed with eicosapentaenoic and
docosahexaenoic acids in addition to linolenic acid. Biochem
Biophys Res Commun 1999; 259: 85–90.
39. Erasmus U. (2001). Udo’s Choice 3,6,9 Perfected Oil Blend
[WWW document] URL http://www.udoerasmus.com/
40. Tainter ML, Cutting WC, Stockton AB. Use of dinitrophenol
in nutritional disorders: a critical survey of clinical results. Am J
Public Health 1934; 24: 1045–1053.
41. Cazeneuze P, Lepine R. Compt Rend Soc Biol 1885; 101:
1167–1169.
42. Magne H, Mayer A, Plantefol L. Studies on the action of dinitrophenol 1–2–4 (Thermol). Ann Physiol Physicochem Biol 1932;
8: 1–167.
43. Cutting WC, Tainter ML. Actions of dinitrophenol. Proc Soc
Exper Biol & Med 1932; 29: 1268.
44. Tainter ML, Stockton AB, Cutting WC. Dinitrophenol in the
treatment of obesity: Final report. J Am Med Assoc 1935; 101:
322–336.
45. Cutting WC, Mehrtens HG, Tainter ML. Actions and uses of
dinitrophenol: Promising metabolic applications. J Am Med Assoc
1933; 101: 193–195.
46. Tainter ML, Stockton AB, Cutting WC. Use of dinitrophenol
in obesity and related conditions: a progress report. J Am Med
Assoc 1933; 101: 1472–1475.
47. Cutting WC, Tainter ML. Metabolic actions of dinitrophenol
with the use of balanced and unbalanced diets. J Am Med Assoc
1933; 101: 2099–2102.
48. Simkins S. Dinitrophenol and desiccated thyroid in the treatment of obesity: a comprehensive clinical and laboratory study. J
Am Med Assoc 1937; 108: 2110–2118.
49. Boardman WW. Rapidly developing cataract after dinitrophenol. J Am Med Assoc 1935; 105: 108.
50. Horner W, Jones R, Boardman WW. Cataracts following the
use of dinitrophenol. J Am Med Assoc 1935; 105: 108–110.
51. Anthony DM, Dickinson K, Jones RB, Heal DJ. Metabolic
actions of 2,4-dinitrophenol: – a re-evaluation of activity in-vitro
and in-vivo. In: Obesity Regulation Energy Homeostasis Keystone
Symposia. Keystone Symposia: Silverthorne, CO, 2001, p 218.
52. Byrom FB. Nature of myxoedema. Clin Sc 1933; 1: 273–285.
53. Astrup A, Buemann B, Toubro S, Ranneries C, Raben A. Low
resting metabolic rate in subjects predisposed to obesity: a role for
thyroid status. Am J Clin Nutr 1996; 63: 879–883.
54. Stenlof K, Sjostrom L, Fagerberg B, Nystrom E, Lindstedt G.
Thyroid hormones, procollagen III peptide, body composition and
basal metabolic rate in euthyroid individuals. Scand J Clin Lab
Invest 1993; 53: 793–803.
55. Harper ME, Brand MD. The quantitative contributions of
mitochondrial proton leak and ATP turnover reactions to the
changed respiration rates of hepatocytes from rats of different
thyroid status. J Biol Chem 1993; 268: 14850–14860.
56. Hafner RP, Nobes CD, McGown AD, Brand MD. Altered
relationship between protonmotive force and respiration rate in
© 2001 The International Association for the Study of Obesity. obesity reviews 2, 255–265
264 Mitochondrial uncoupling as a treatment for obesity
J. A. Harper et al.
non-phosphorylating liver mitochondria isolated from rats of
different thyroid hormone status. Eur J Biochem 1988; 178:
511–518.
57. Brand MD, Steverding D, Kadenbach B, Stevenson PM,
Hafner RP. The mechanism of the increase in mitochondrial
proton permeability induced by thyroid hormones. Eur J Biochem
1992; 206: 775–781.
58. Drabkin DL. Cytochrome c metabolism and liver regeneration. Influence of thyroid gland and thyroxine. J Biol Chem 1949;
182: 335–349.
59. Tata JR, Ernster L, Lindberg O, Arrhenius E, Pederson S,
Hedman R. The action of thyroid hormones at the cell level.
Biochem J 1963; 86: 408–428.
60. Hulbert AJ. Thyroid hormones and their effects: a new perspective. Biol Rev Camb Philos Soc 2000; 75: 519–631.
61. Hoch FL. Lipids and thyroid hormones. Prog Lipid Res 1988;
27: 199–270.
62. Brookes PS, Buckingham JA, Tenreiro AM, Hulbert AJ, Brand
MD. The proton permeability of the inner membrane of liver mitochondria from ectothermic and endothermic vertebrates and from
obese rats: correlations with standard metabolic rate and phospholipid fatty acid composition. Comp Biochem Physiol B
Biochem Mol Biol 1998; 119: 325–334.
63. Paradies G, Ruggiero FM, Petrosillo G, Quagliariello E.
Enhanced cytochrome oxidase activity and modification of lipids
in heart mitochondria from hyperthyroid rats. Biochim Biophys
Acta 1994; 1225: 165–170.
64. Soboll S. Thyroid hormone action on mitochondrial energy
transfer. Biochim Biophys Acta 1993; 1144: 1–16.
65. Masaki T, Yoshimatsu H, Kakuma T, Hidaka S, Kurokawa
M, Sakata T. Enhanced expression of uncoupling protein 2 gene
in rat white adipose tissue and skeletal muscle following chronic
treatment with thyroid hormone. FEBS Lett 1997; 418: 323–326.
66. Lanni A, Beneduce L, Lombardi A, Moreno M, Boss O,
Muzzin P, Giacobino JP, Goglia F. Expression of uncoupling
protein-3 and mitochondrial activity in the transition from
hypothyroid to hyperthyroid state in rat skeletal muscle. FEBS Lett
1999; 444: 250–254.
67. Lanni A, De Felice M, Lombardi A, Moreno M, Fleury C,
Ricquier D, Goglia F. Induction of UCP2 mRNA by thyroid
hormones in rat heart. FEBS Lett 1997; 418: 171–174.
68. Gong DW, He Y, Karas M, Reitman M. Uncoupling protein3 is a mediator of thermogenesis regulated by thyroid hormone,
beta3-adrenergic agonists, and leptin. J Biol Chem 1997; 272:
24129–24132.
69. Larkin S, Mull E, Miao W, Pittner R, Albrandt K, Moore C,
Young A, Denaro M, Beaumont K. Regulation of the third member
of the uncoupling protein family, UCP3, by cold and thyroid
hormone. Biochem Biophys Res Commun 1997; 240: 222–227.
70. Klein I, Levey GS. New perspectives on thyroid hormone,
catecholamines, and the heart. Am J Med 1984; 76: 167–172.
71. Burgi U, Burgi-Saville ME, Burgherr J, Clement M, Lauber K.
T3 plus high doses of beta-blockers: effects on energy intake, body
composition, bat and heart in rats. Int J Obes 1990; 14:
1023–1038.
72. Mittleman RE, Goldberg RB, Nadji M. Severe thyroid atrophy
due to prolonged ingestion of thyroid hormone for treatment of
obesity. South Med J 1984; 77: 268–270.
73. Kyle LH, Ball MF, Doolan PD. Effect of thyroid hormone on
body composition in myxedema and obesity. N Engl J Med 1966;
275: 12–17.
74. Abraham RR, Densem JW, Davies P, Davie MW, Wynn V. The
effects of triiodothyronine on energy expenditure, nitrogen balance
and rates of weight and fat loss in obese patients during prolonged
caloric restriction. Int J Obes 1985; 9: 433–442.
obesity reviews
75. Nicholls DG, Locke RM. Thermogenic mechanisms in brown
fat. Physiol Rev 1984; 64: 1–64.
76. Nedergaard J, Golozoubova V, Matthias A, Asadi A,
Jacobsson A, Cannon B. UCP1: the only protein able to mediate
adaptive non-shivering thermogenesis and metabolic inefficiency.
Biochim Biophys Acta 2001; 1504: 82–106.
77. Nicholls DG. The effective proton conductance of the inner
membrane of mitochondria from brown adipose tissue. Dependency on proton electrochemical potential gradient. Eur J Biochem
1977; 77: 349–356.
78. Nicholls DG, Lindberg O. Brown-adipose-tissue mitochondria. The influence of albumin and nucleotides on passive ion permeabilities. Eur J Biochem 1973; 37: 523–530.
79. Nicholls DG. Hamster brown-adipose-tissue mitochondria.
Purine nucleotide control of the ion conductance of the inner membrane, the nature of the nucleotide binding site. Eur J Biochem
1976; 62: 223–228.
80. Heaton GM, Wagenvoord RJ, Kemp AJ, Nicholls DG. Brownadipose-tissue mitochondria: photoaffinity labelling of the regulatory site of energy dissipation. Eur J Biochem 1978; 82: 515–521.
81. Lin CS, Hackenberg H, Klingenberg EM. The uncoupling
protein from brown adipose tissue mitochondria is a dimer. A
hydrodynamic study. FEBS Lett 1980; 113: 304–306.
82. Lin CS, Klingenberg M. Characteristics of the isolated purine
nucleotide binding protein from brown fat mitochondria. Biochemistry 1982; 21: 2950–2956.
83. Bathgate B, Freebairn EM, Greenland AJ, Reid GA. Functional expression of the rat brown adipose tissue uncoupling
protein in Saccharomyces cerevisiae. Mol Microbiol 1992; 6:
363–370.
84. Jezek P, Orosz DE, Garlid KD. Reconstitution of the uncoupling protein of brown adipose tissue mitochondria. Demonstration of GDP-sensitive halide anion uniport. J Biol Chem 1990;
265: 19296–19302.
85. Katiyar SS, Shrago E. Reconstitution of purified brown
adipose tissue mitochondria uncoupling protein: demonstration of
separate identity of nucleotide binding and proton translocation
sites by chemical probes. Proc Natl Acad Sci USA 1989; 86:
2559–2562.
86. Klingenberg M, Winkler E. The reconstituted isolated uncoupling protein is a membrane potential driven H+ translocator.
EMBO J 1985; 4: 3087–3092.
87. Strieleman PJ, Schalinske KL, Shrago E. Fatty acid activation
of the reconstituted brown adipose tissue mitochondria uncoupling protein. J Biol Chem 1985; 260: 13402–13405.
88. Harper ME, Himms-Hagen J. Mitochondrial efficiency:
lessons learned from transgenic mice. Biochim Biophys Acta 2001;
1504: 159–172.
89. Enerbäck S, Jacobsson A, Simpson EM, Guerra C, Yamashita
H, Harper ME, Kozak LP. Mice lacking mitochondrial uncoupling
protein are cold-sensitive but not obese. Nature 1997; 387: 90–94.
90. Melnyk A, Harper ME, Himms-Hagen J. Raising at thermoneutrality prevents obesity and hyperphagia in BAT- ablated
transgenic mice. Am J Physiol 1997; 272: R1088–R1093.
91. Kopecky J, Clarke G, Enerback S, Spiegelman B, Kozak LP.
Expression of the mitochondrial uncoupling protein gene from the
aP2 gene promoter prevents genetic obesity. J Clin Invest 1995;
96: 2914–2923.
92. Strosberg AD, Pietri-Rouxel F. Function and regulation of the
b3-adrenoceptor. Trends Pharmacol Sci 1996; 17: 373–381.
93. Ricquier D, Bouillaud F. The uncoupling protein homologues:
UCP1, UCP2, UCP3, StUCP and AtUCP. Biochem J 2000; 345:
161–179.
94. Nicholls DG, Rial E. A history of the first uncoupling protein,
UCP1. J Bioenerg Biomembr 1999; 31: 399– 406.
© 2001 The International Association for the Study of Obesity. obesity reviews 2, 255–265
obesity reviews
Mitochondrial uncoupling as a treatment for obesity J. A. Harper et al.
95. Arch JR, Wilson S. Prospects for b3-adrenoceptor agonists in
the treatment of obesity and diabetes. Int J Obes Relat Metab
Disord 1996; 20: 191–199.
96. Henningfield MF, Swick RW. Immunochemical detection and
quantitation of brown adipose tissue uncoupling protein. Biochem
Cell Biol 1987; 65: 245–251.
97. Lean ME, Branch WJ, James WP, Jennings G, Ashwell M.
Measurement of rat brown-adipose-tissue mitochondrial uncoupling protein by radioimmunoassay: increased concentration after
cold acclimation. Biosci Rep 1983; 3: 61–71.
98. Ricquier D, Barlet JP, Garel JM, Combes-George M, Dubois
MP. An immunological study of the uncoupling protein of brown
adipose tissue mitochondria. Biochem J 1983; 210: 859–866.
99. Pecqueur C, Alves-Guerra MC, Gelly C, Levi-Meyrueis C,
Couplan E, Collins S, Ricquier D, Bouillaud F, Miroux B. Uncoupling Protein 2, in vivo distribution, induction upon oxidative
stress, and evidence for translational regulation. J Biol Chem 2001;
276: 8705–8712.
100. Sivitz WI, Fink BD, Donohoue PA. Fasting and leptin
modulate adipose and muscle uncoupling protein: divergent effects
between messenger ribonucleic acid and protein expression.
Endocrinology 1999; 140: 1511–1519.
101. Fleury C, Neverova M, Collins S, Raimbault S, Champigny
O, Levi-Meyrueis C, Bouillaud F, Seldin MF, Surwit RS, Ricquier
D, Warden CH. Uncoupling protein-2: a novel gene linked to
obesity and hyperinsulinemia. Nat Genet 1997; 15: 269–272.
102. Gonzalez-Barroso MM, Fleury C, Arechaga I, Zaragoza P,
Levi-Meyrueis C, Raimbault S, Ricquier D, Bouillaud F, Rial E.
Activation of the uncoupling protein by fatty acids is modulated
by mutations in the C-terminal region of the protein. Eur J
Biochem 1996; 239: 445–450.
103. Hinz W, Faller B, Gruninger S, Gazzotti P, Chiesi M. Recombinant human uncoupling protein-3 increases thermogenesis in
yeast cells. FEBS Lett 1999; 448: 57–61.
104. Stuart JA, Harper JA, Brindle KM, Jekabsons MB, Brand
MD. Physiological levels of mammalian uncoupling protein 2 do
not uncouple yeast mitochondria. J Biol Chem 2001; 22: 22.
105. Stuart JA, Brand MD, Brindle KM, Jekabsons MB, Harper
JA. Evidence that the uncoupling observed with UCP2 and UCP3
in yeast mitochondria is an artefact. Obesity Regulation Energy
Homeostasis Keystone Symposia. Keystone Symposia: Silverthorne, CO, 2001, pp 218.
106. Stuart JA, Harper JA, Jekabsons MB, Brindle KM, Brand
MD. A mitochondrial uncoupling artefact can be caused by
expression of uncoupling protein 1 in yeast. Biochem J 2001; 356:
779–789.
107. Echtay KS, Liu Q, Caskey T, Winkler E, Frischmuth K,
Bienengraber M, Klingenberg M. Regulation of UCP3 by
nucleotides is different from regulation of UCP1. FEBS Lett 1999;
450: 8–12.
108. Jaburek M, Varecha M, Gimeno RE, Dembski M, Jezek P,
Zhang M, Burn P, Tartaglia LA, Garlid KD. Transport function
and regulation of mitochondrial uncoupling proteins 2 and 3. J
Biol Chem 1999; 274: 26003–26007.
265
109. Echtay KS, Winkler E, Bienengraeber M, Klingenberg M.
Site-directed mutagenesis identifies residues in uncoupling protein
(UCP1) involved in three different functions. Biochemistry 2000;
39: 3311–3317.
110. Klingenberg M, Echtay KS. Uncoupling proteins: the issues
from a biochemist point of view. Biochim Biophys Acta 2001;
1504: 128–143.
111. Echtay KS, Winkler E, Klingenberg M. Coenzyme Q is an
obligatory cofactor for uncoupling protein function. Nature 2000;
408: 609–613.
112. Echtay KS, Winkler E, Frischmuth K, Klingenberg M.
Uncoupling proteins 2 and 3 are highly active H+ transporters and
highly nucleotide sensitive when activated by coenzyme Q
(ubiquinone). Proc Natl Acad Sci USA 2001; 98: 1416–1421.
113. Matthias A, Jacobsson A, Cannon B, Nedergaard J. The
bioenergetics of brown fat mitochondria from UCP1-ablated
mice. UCP1 is not involved in fatty acid-induced de-energization
(‘uncoupling’). J Biol Chem 1999; 274: 28150–28160.
114. Monemdjou S, Kozak LP, Harper ME. Mitochondrial proton
leak in brown adipose tissue mitochondria of Ucp1- deficient mice
is GDP insensitive. Am J Physiol 1999; 276: E1073–E1082.
115. Gong DW, Monemdjou S, Gavrilova O, Leon LR, MarcusSamuels B, Chou CJ, Everett C, Kozak LP, Li C, Deng C, Harper
ME, Reitman ML. Lack of obesity and normal response to fasting
and thyroid hormone in mice lacking uncoupling protein-3. J Biol
Chem 2000; 275: 16251–16257.
116. Vidal-Puig AJ, Grujic D, Zhang CY, Hagen T, Boss O, Ido
Y, Szczepanik A, Wade J, Mootha V, Cortright R, Muoio DM,
Lowell BB. Energy metabolism in uncoupling protein 3 gene
knockout mice. J Biol Chem 2000; 275: 16258–16266.
117. Samec S, Seydoux J, Dulloo AG. Role of UCP homologues
in skeletal muscles and brown adipose tissue: mediators of thermogenesis or regulators of lipids as fuel substrate? FASEB J 1998;
12: 715–724.
118. Ma SW, Foster DO. Starvation-induced changes in metabolic
rate, blood flow, and regional energy expenditure in rats. Can J
Physiol Pharmacol 1986; 64: 1252–1258.
119. Cadenas S, Buckingham JA, Samec S, Seydoux J, Din N,
Dulloo AG, Brand MD. UCP2 and UCP3 rise in starved rat skeletal muscle but mitochondrial proton conductance is unchanged.
FEBS Lett 1999; 462: 257–260.
120. Andreyev A, Bondareva TO, Dedukhova VI, Mokhova
EN, Skulachev VP, Volkov NI. Carboxyatractylate inhibits the
uncoupling effect of free fatty acids. FEBS Lett 1988; 226: 265–
269.
121. Skulachev VP. Uncoupling: new approaches to an old
problem of bioenergetics. Biochim Biophys Acta 1998; 1363:
100–124.
122. Cadenas S, Buckingham JA, St-Pierre J, Dickinson K, Jones
RB, Brand MD. AMP decreases the efficiency of skeletal-muscle
mitochondria. Biochem J 2000; 351: 307–311.
123. Hafner RP, Brown GC, Brand MD. Thyroid-hormone
control of state-3 respiration in isolated rat liver mitochondria.
Biochem J 1990; 265: 731–734.
© 2001 The International Association for the Study of Obesity. obesity reviews 2, 255–265
Download