Sei sulla pagina 1di 9

Clin Chem Lab Med 2000; 38(1):3–11 © 2000 by Walter de Gruyter · Berlin · New York

Review

Regulation of Lipid and Lipoprotein Metabolism by PPAR Activators

Philippe Gervois, Inés Pineda Torra, Jean-Charles UCPs uncoupling proteins; VLDL very low density
Fruchart and Bart Staels lipoprotein.
Département d’Athérosclérose, INSERM U.325, Institut
Pasteur de Lille et Faculté de Pharmacie, Université de
Lille II, Lille, France Introduction

Numerous epidemiological and intervention studies


The peroxisome proliferator-activated receptors have firmly established the causative relationship be-
(PPARs) are ligand-activated transcription factors be- tween dyslipidemia and coronary artery disease (CAD).
longing to the nuclear hormone receptor superfamily. Hypercholesterolemia (high LDL-cholesterol levels) is
PPARa, the first identified PPAR family member, is considered a major risk factor for CAD (1, 2). Hypoal-
principally expressed in tissues exhibiting high rates of phalipoproteinemia (low plasma HDL), alone or com-
b-oxidation such as liver, kidney, heart and muscle. bined with hypertriglyceridemia, constitutes another
PPARg, on the other hand, is expressed at high levels important risk factor as well (3). Furthermore, elevated
in adipose tissue. PPARs are activated by dietary fatty plasma triglyceride levels have been recently identified
acids and eicosanoids, as well as by pharmacological as an independent factor associated with increased
drugs, such as fibrates for PPARa and glitazones for mortality in patients with established CAD (4). Fibrates
PPARg. PPARa mediates the hypolipidemic action of fi- are hypolipidemic drugs currently used in the treat-
brates in the treatment of hypertriglyceridemia and ment of dyslipidemia (5). Coronary angiography trials
hypoalphalipoproteinemia. PPARa is considered a ma- have demonstrated that administration of fibrates re-
jor regulator of intra- and extracellular lipid metabo- duces the progression of CAD (6, 7). Results from the
lism. Upon fibrate activation, PPARa down-regulates recently published Veterans Affairs High-Density
hepatic apolipoprotein C-III and increases lipoprotein Lipoprotein Cholesterol Intervention Trial, a secondary
lipase gene expression, key players in triglyceride me- prevention trial with the fibrate derivative gemfibrozil,
tabolism. In addition, PPARa activation increases demonstrated a highly significant reduction in cardio-
plasma HDL cholesterol via the induction of hepatic vascular events in patients with low baseline HDL-C
apolipoprotein A-I and apolipoprotein A-II expression concentrations treated with gemfibrozil (8).
in humans. Glitazones exert a hypotriglyceridemic ac- Although fibrates have been widely used in clinical
tion via PPARg-mediated induction of lipoprotein li- practice as hypolipidemic drugs, their mechanism of
pase expression in adipose tissue. PPARs play also a action was only recently elucidated. Fibrates also be-
role in intracellular lipid metabolism by up-regulating long to a class of compounds termed peroxisome pro-
the expression of enzymes involved in conversion of liferators which are able to induce peroxisome prolifer-
fatty acids in acyl-coenzyme A esters, fatty acid entry ation and hepatomegaly resulting, after long-term
into mitochondria and peroxisomal and mitochondrial treatment, in hepatocarcinogenesis in rodents (9). This
fatty acid catabolism. These observations have pro- effect is accompanied by the transcriptional induction
vided the molecular basis leading to a better under- of genes implicated in peroxisomal β-oxidation and oc-
standing of the mechanism of action of fibrates and curs through activation of specific transcription factors
glitazones on lipid and lipoprotein metabolism and named peroxisome proliferator-activated receptors
identify PPARs as attractive targets for the rational de- (PPARs) (10, 11). Although the induction of peroxisome
sign of more potent lipid-lowering drugs. proliferation by fibrates in rodents requires PPARα (12),
activation of PPARα by fibrates does not lead to perox-
Key words: Nuclear receptors; Fibrates; Lipoprotein
isome proliferation in humans. By contrast, PPARα ac-
metabolism.
tivators exert potent lipid lowering activities. Results
Abbreviations: ACO acyl-CoA oxidase; ACS acyl-CoA from studies conducted over the past five years have
synthetase; apo apolipoprotein; CAD coronary artery identified PPARs as key regulators of lipid and lipopro-
disease; FA fatty acid; FAS fatty acid synthase; FATP tein metabolism and as mediators of lipid effects of fi-
fatty acid transport protein; HDL high density lipopro- brate and glitazone action (13). In this report, we will re-
tein; LCAT lecithin:cholesterol acyltransferase; LDL view recent advances on the role of PPARs in lipid and
low density lipoprotein; LPL lipoprotein lipase; oxLDL lipoprotein metabolism.
oxidized low density lipoprotein; PPARs peroxisome
proliferator-activated receptors; PPRE peroxisome
proliferator response element; RXR retinoic X recep-
tor; SR-BI scavenger receptor BI; TG triglycerides;

Brought to you by | University of Arizona


Authenticated
Download Date | 5/28/15 10:38 AM
4 Gervois et al.: Role of PPARs in lipid and lipoprotein metabolism

PPARs: Transcription Factors and Nuclear Receptors

PPARs are members of the nuclear hormone receptor


superfamily and are ligand-activated transcription fac-
tors (13) involved in translating the effects of lipid solu-
ble factors such as hormones, vitamins, fatty acids (FA)
and various drugs into the gene expression level.
Three distinct PPARs, α, β(δ) and γ, each encoded by a
separate gene and displaying different tissue (14–17)
and developmental (18) expression patterns, have
been identified. PPARα is preferentially expressed in
liver, heart and kidney while PPARγ is expressed at high
levels in adipose tissue. All PPARs are furthermore also Fig. 1 Mechanism of transcriptional gene regulation by
expressed, to variable extents, in a wide variety of PPARs.
other cells and tissues. Whereas PPARα modulates the Activated PPARs form a heterodimeric complex with the
transcription of genes implicated in lipid and lipopro- retinoic X receptor (RXR) which subsequently binds to a PPRE
tein metabolism in liver (5), PPARγ mediates adipogen- located within the regulatory sequence of target genes and
therefore inducing gene transcription. PPARs negatively regu-
esis (19). Recent findings suggest that PPARγ activation
late gene transcription through protein-protein interaction
induces the terminal differentiation of human breast
leading to interference with the STAT (not represented for the
cancer cells (20) and may modulate the development of clarity of the figure), NF-ΚB and Fos/Jun pathways.
colorectal cancer (21–23). PPARs are activated by nat-
ural ligands derived from FAs such as 8(S) hydroxye-
icosatetraenoic acid, 8(S) hydroxyeicosapentaenoic netic and environmental factors. The transcriptional
acid and leukotriene B4 for PPARα (24) and regulation of PPARγ occurs at least in part, through the
prostaglandin-J2 and components of oxidized low- use of alternative promoters, which results, both in ro-
density lipoprotein (oxLDL) for PPARγ (24, 25). More- dents and humans (43, 44), in the production of two
over, hypolipidemic drugs of the fibrate class and an- distinct proteins, PPARγ1 and PPARγ2, with distinct ac-
tidiabetic glitazones constitute synthetic ligands for tivation capacities (45). In addition, glucocorticoids are
PPARα and PPARγ respectively (24). Natural and syn- implicated in the regulation of PPARγ expression dur-
thetic ligands have also been identified for PPARδ (26, ing differentiation of 3T3-L1 preadipocytes (46).
27). Following ligand binding, PPAR interacts with the Finally, PPAR activity may be modulated at the pro-
retinoic X receptor (RXR) to form a PPAR/RXR het- tein level. Protein phosphorylation constitutes a com-
erodimeric complex which binds to a specific response mon post-translational mechanism of regulation. Both
element, termed peroxisome proliferator response ele- PPARα and PPARγ are regulated by physiological phos-
ment (PPRE), located in the regulatory region of target phorylation, thus affecting their transactivation poten-
genes (Figure 1). Although the majority of PPREs iden- tial, and modulating their biological functions (47–52).
tified so far consist of a direct repeat of the canonical The production of isoforms with repressive activity on
AGGTCA sequence spaced by 1 nucleotide (DR-1) (13), the wild type receptor represents another mechanism
DR-2 elements may also function as PPRE (28). PPARs of regulation of PPAR signalling occurring at the pro-
positively regulate target genes through this DNA bind- tein level. Such an isoform, resulting from alternative
ing dependent mechanism. In addition, PPARs have splicing and giving rise to a truncated protein, has been
been shown to repress gene transcription in a DNA identified for PPARα (41, 42). This variant acts in a lig-
binding independent manner. PPARs interfere with the and independent manner and alters PPARα wild type
NF-κB, STAT and AP-1 transcription pathways, proba- transcriptional capacity.
bly due to protein-protein interaction between the tran-
scription factors leading to the formation of inactive
complexes (29–34) (Figure 1). The interactions between Regulation of Intracellular Lipid Metabolism by PPARs
PPARα with Jun and p65, partners of the AP-1 and NF-
κB transcription factor complexes respectively, result PPARα plays a key role in intracellular FA metabolism
in the inhibition of AP-1 and NF-κB protein binding to (Figure 2). High level of PPARα expression is observed
their respective response elements (33, 34). in tissues with elevated FA catabolism. PPARα regu-
PPAR activity may be regulated at the transcrip- lates the expression of genes coding for enzymes im-
tional, post-transcriptional and at the protein level. In plicated in the peroxisomal β-oxidation pathway such
rats, PPARα expression is regulated by hormones, such as acyl-CoA oxidase (ACO), multi-functional enzyme
as glucocorticoids, insulin and leptin, by physiological and 3 ketoacyl-CoA thiolase (13). Moreover, these en-
stimuli such as stress and fasting and follows a diurnal zymes have been shown to influence PPARα ligand me-
rhythm (35–40). Although little is known about factors tabolism (53, 54). PPARα also modulates genes in-
regulating PPARα expression in humans, the observa- volved in FA uptake, activation to acyl-CoA esters,
tion that PPARα expression in liver varies significantly mitochondrial β-oxidation and ketone body synthesis
among individuals (41, 42) suggests that PPARα is also (13, 55). Intracellular FA concentrations are controlled,
strongly regulated at the gene level in humans by ge- in part, by the activity of the FA transport protein

Brought to you by | University of Arizona


Authenticated
Download Date | 5/28/15 10:38 AM
Gervois et al.: Role of PPARs in lipid and lipoprotein metabolism 5

FA flux from peripheral tissues, such as adipose tissue,


to the liver (Figure 2). Interestingly, it is likely that
PPARα plays also a role in adipose tissue as a mediator
of leptin-induced lipolysis (37). Indeed, hyperleptine-
mia in rodents depletes adipocyte fat, while it up-regu-
lates FA oxidation enzymes, UCPs and PPARα, which is
normally low in adipocytes.
Whereas PPARα is involved in hepatic lipid metabo-
lism, PPARγ modulates lipid homeostasis through its
function in adipose tissue. Several key genes for
adipocyte differentiation and lipid storage contain a
PPRE. Gene expression of aP2 (66), phosphoenol pyru-
vate carboxykinase (67), ACS (68, 69), FATP (56) and
LPL (70) is regulated by PPARγ. The induction of LPL
Fig. 2 PPARα enhances FA catabolism by inducing genes promotes FA delivery to adipocytes while induction of
(bold) involved in FA uptake (FATP), esterification (ACS) and β- FATP and ACS results in enhanced FA uptake by the
oxidation (β-ox). PPARα activation enhances FA β-oxidation adipocyte. These actions contribute to enhanced TG
and therefore diminishes the FA pool to be incorporated into synthesis and accumulation in adipose tissue suggest-
TG-rich lipoproteins. Consequently, PPARα stimulates lipid ing that PPARγ activation maintains the mature
flux by controlling the FA flux from peripheral tissues such as adipocyte phenotype and participates in lipid home-
adipose tissue to the liver. ostasis.
Bold arrows indicate pathways enhanced by PPARα activa-
tors. FA, fatty acid; TG, triglycerides; LPL, lipoprotein lipase;
FAS, FA synthase.
Regulation of Lipoprotein Metabolism by PPARs

Metabolic effects of PPAR activators


(FATP), which controls the entry of FA through the cell
Studies carried out both in rodents and humans led to
membrane, and by acyl-CoA synthetase (ACS) which
the establishment of four major metabolic pathways
traps FA inside the cells by their conversion to ester de-
affected by fibrates and explaining their effects on
rivatives. PPARα activation mediates the induction of
lipoprotein metabolism (Figure 3). These are: (1) Induc-
FATP expression in liver and intestine and the up-regu-
tion of lipoprotein lipolysis as a result of either an in-
lation of ACS expression in liver and kidney (56). The
crease in intrinsic lipoprotein lipase activity or an in-
implication of PPARα in FA transport was further
creased accessibility of TG-rich lipoprotein particles for
demonstrated by the lack of induction of FATP and FA
lipolysis due to reduced TG-rich lipoprotein apo C-III
translocase mRNA in liver by PPARα activators in
content (71, 72). (2) Limitation of hepatic TG synthesis
PPARα-null mice (57).
and VLDL production due to increased FA uptake, en-
FA metabolism is regulated by the rate of mitochon-
hanced FA catabolism and reduced FA synthesis (56,
drial FA uptake. PPAR α has been demonstrated to af-
68, 73). (3) Increase in LDL particle removal as a result
fect FA import into mitochondria by up-regulating the
of changes in plasma LDL composition and subsequent
expression of muscle- (58–60) and liver-type α-carni-
increase in LDL affinity for its receptor leading to en-
tine palmitoyltransferase I genes (39). Interestingly,
hanced LDL catabolism (74). (4) Increase in HDL pro-
specific inhibition of mitochondrial FA import in
duction and stimulation of reverse cholesterol trans-
PPARα-null mice causes hepatic and cardiac lipid accu-
port.
mulation, hypoglycemia and death in 100% of males
Fibrates increase the production of apolipoprotein
and 25% of females (61). Furthermore, PPARα-deficient
A-I and A-II in human liver (13, 75–77), which leads to
mice fed a high fat diet showed a massive accumula-
tion of lipids in liver (39, 62) highlighting the crucial role
PPARα plays in lipid metabolism. A concordant pheno-
type was observed in PPARα-deficient mice fasted for
24 hours who displayed hypoglycemia, hypoketone-
mia, and elevated plasma FA levels (39). Recent studies
revealed that PPARα might increase energy expendi-
ture by up-regulating the expression of uncoupling
proteins (UCPs) (63–65). These data strongly argue in
favour of a critical role for PPARα in the regulation of
lipid metabolism. Through their effect on the expres-
sion of FA transporter and FA oxidation genes, PPARα
activators direct the FA flux to the β-oxidation pathway
and therefore diminish the FA pool to be incorporated
to triglyceride (TG)-rich lipoproteins. Consequently, Fig. 3 Metabolic effects of fibrates on lipid and lipoprotein
PPARα maintains lipid homeostasis by controlling the metabolism.

Brought to you by | University of Arizona


Authenticated
Download Date | 5/28/15 10:38 AM
6 Gervois et al.: Role of PPARs in lipid and lipoprotein metabolism

increased plasma HDL concentrations and enhanced is also able to increase apo C-III expression (89). Sec-
reverse cholesterol transport. ond, apo C-III repression by fibrates might occur via
Glitazones, antidiabetic agents, are primarily studied PPARα-dependent enhanced expression of a negative
for their effect on glucose homeostasis. Nevertheless, regulator of transcription. Fibrates induce the hepatic
these drugs exert hypotriglyceridemic action both in expression of human and rat Rev-erbα, a nuclear or-
rodents and in humans by increasing lipolysis and phan receptor which is a strong repressor of transcrip-
clearance of TG-rich lipoproteins (78–83). This effect tion (28, 90). The induction of Rev-erbα is mediated by
may be attributed, at least in part, to the induction of PPARα which interacts with a PPRE located within the
adipose tissue LPL expression (70, 84). human Rev-erbα promoter (28). Such a mechanism is
consistent with the observation that Rev-erbα-deficient
PPARs and TG metabolism mice exhibit increased plasma TG and apo C-III con-
centrations and elevated hepatic apo C-III mRNA levels
The role of PPARs in TG metabolism is convincingly es-
(91). Finally, as already shown for other target genes
tablished (13). The hypotriglyceridemic effect of PPARα
(29, 32, 92, 93), PPARα might repress transcription of
activators is the result of increased lipoprotein lipolysis
apo C-III by interacting with other transcription factors
and enhanced FA oxidation. The process of TG clear-
leading to the formation of inactive complexes thereby
ance is directly under the control of LPL and apo C-III.
limiting the induction of apo C-III expression. Further
PPARα activators repress apo C-III expression (85, 86)
studies are required to elucidate the exact role of
and induce LPL in liver (70) (Figure 4). Moreover,
PPARα in the regulation of apo C-III gene expression.
PPARα enhances FA catabolic rate, which subsequently
The induction by glitazones of LPL gene expression
affects TG synthesis and VLDL production (13).
in adipocytes (70) is mediated by PPARγ, which inter-
PPARα plays a central role in hepatic apo C-III gene
acts with a PPRE present both in the rodent and human
regulation as demonstrated using PPARα-deficient
LPL promoter (70). The increase in LPL production will
mice (87). Hepatic apo C-III gene transcription is con-
enhance clearance of plasma TGs and FA delivery to
trolled by numerous nuclear receptors, such as HNF-4,
the adipose tissue. This mechanism of action likely
ARP-I, Ear/COUP-TF (88) and RXRα (88, 89). HNF-4,
constitutes the basis for the hypotriglyceridemic action
RXRα and PPARα can activate apo C-III transcription
of PPARγ activators.
through a common DNA binding site of the upstream
regulatory region of the apo C-III gene, whereas ARP-1
and Ear/COUP-TF function as repressors of transcrip- PPARs and HDL metabolism
tion. Several mechanisms may be involved to explain
Both apo A-I and HDL are inversely correlated with the
the effects of fibrates on apo C-III gene regulation. First,
incidence of CAD (94). There is evidence supporting the
the repression of apo C-III gene expression by fibrates
protective role of HDL through the removal and the re-
might be the result of a competition between transcrip-
cycling of cholesterol excess from peripheral tissues to
tion factors. Substitution of the strong activator HNF-4
liver. PPARα appears to be the major isoform impli-
by a less active PPAR/RXR complex may explain a re-
cated in HDL metabolism. PPARα activators affect HDL
duction of apo C-III transcriptional activity. The fine-
metabolism in an opposite manner in rodents and hu-
tuning of apo C-III regulation will then depend on the
mans. Whereas fibrate treatment of rats lowers plasma
relative abundance of PPARα and HNF-4. Against this
HDL, an increase is generally observed in humans
hypothesis is the observation that the RXR homodimer
(95–97). Such an increase in HDL plasma levels is re-
lated, at least in part, to changes in apo A-I and apo A-II
gene expression in liver (Figure 4). Apo A-I and apo A-
II gene transcription is up-regulated in humans by
PPARα activators (75–77). By contrast, these genes are
repressed in rodents (77, 98). Recent studies in trans-
genic mice lacking PPARα univocally confirmed the im-
plication of PPARα in the regulation of apo A-I and HDL
metabolism in rodent species (87, 99). Basal plasma
HDL cholesterol, hepatic apo A-I mRNA and apo A-I
plasma levels are higher in PPARα-deficient mice, com-
pared to wild type control mice. Treatment with fibrates
lowered both hepatic apo A-I mRNA levels and apo A-I
plasma levels in wild type mice, whereas no effect was
observed in PPARα-deficient mice. Studies on the reg-
ulation of hepatic apo A-I gene expression, carried out
in human apo A-I transgenic mice, demonstrated an in-
Fig. 4 Implication of PPARα in the transcriptional regulation
of human genes involved in lipoprotein metabolism. A-I, A-II
duction of plasma HDL and human apo A-I concentra-
and C-III, apolipoproteins; LPL, lipoprotein lipase; HDL, high- tions by fibrates (77). It was shown that the effects on
density lipoprotein; TG, triglycerides; PPRE, peroxisome pro- human apo A-I mRNA levels are due to an enhanced
liferator response element. (+), activation of transcription; (–) transcription rate. Moreover, treatment of human pri-
repression of transcription. mary hepatocytes with fibrates increases mRNA levels

Brought to you by | University of Arizona


Authenticated
Download Date | 5/28/15 10:38 AM
Gervois et al.: Role of PPARs in lipid and lipoprotein metabolism 7

and secretion of apo A-I, supporting the hypothesis lation of gene expression by fibrates, namely the in-
that a direct action of fibrates on liver human apo A-I duction of expression of a negative regulator of gene
expression results in an increase in plasma apo A-I and transcription.
HDL-cholesterol. To determine whether fibrates in- Fibrates also influence the expression of human apo
crease HDL and apo A-I concentrations without induc- A-II, the other major protein constituent of HDL (75).
ing hepatomegaly and peroxisome proliferation, their Administration of fenofibrate to patients with CAD re-
effects were tested in rabbits, an animal model more sults in a marked increase in plasma apo A-II concen-
resistant to peroxisome proliferation than rodents trations. This increase in plasma apo A-II is due to a di-
(100). In normal rabbits, plasma A-I levels remain un- rect effect on hepatic apo A-II production, since fibrates
changed upon fibrate treatment. By contrast, adminis- induce apo A-II mRNA levels both in primary cultures
tration of fibrates to transgenic human apo A-I rabbits of human hepatocytes and in human hepatoblastoma
results in increased plasma HDL and human apo A-I HepG2 cells (75). The induction of apo A-II mRNA levels
concentrations due to the induction of human apo A-I is followed by an increase in apo A-II secretion in both
gene expression in liver (100). These actions occured cell culture systems. PPARα binds with high affinity to
independently of alterations in TG concentrations and a DR-1-type PPRE located in the human apo A-II pro-
without affecting liver weight or peroxisomal acyl-CoA moter, thereby activating apo A-II gene transcription.
oxidase activity. These data demonstrate a direct ac- These data demonstrate that fibrates increase human
tion of PPARα activators on apo A-I expression result- apo A-II plasma levels by stimulating transcription of
ing in increased human apo A-I concentrations and its gene through the interaction of activated PPARα
HDL plasma levels and decreased plasma apo A-I and with the apo A-II-PPRE.
HDL-cholesterol in rodents. The beneficial effects of fi- In addition to influencing the transcription of HDL
brates on plasma lipoprotein homeostasis can thus oc- apolipoproteins, fibrates also affect the expression of
cur dissociated from potentially deleterious peroxi- enzymes and receptors regulating HDL metabolism. In
some proliferation. rats, fibrates decrease the production of both the major
The molecular mechanism underlying the gene reg- HDL apolipoproteins as well as of HDL-remodelling en-
ulation of apo A-I by fibrates was elucidated by VuDac zymes, such as hepatic lipase and lecithin:cholesterol
et al. (76). The authors demonstrated a direct involve- acyltransferase (LCAT) (101–103). Furthermore, PPAR
ment of PPARα in the control of apo A-I gene transcrip- agonists regulate the expression of HDL receptors.
tion since PPARα binds to a PPRE located in the A site Murine scavenger receptor BI (SR-BI) and its human
of the human apo A-I gene promoter (Figure 5). In ro- homologue CLA-1 have been identified as HDL recep-
dents, the mechanism of negative regulation of apo A- tors, which bind HDL with high affinity and mediate the
I gene expression by fibrates appears to be more com- selective uptake of cholesteryl esters in liver and
plex. The lack of rat apo A-I transcriptional induction is steroidogenic tissues (104, 105). In addition, studies
due to 3 nucleotide differences between the rat and the demonstrating that the rate of cholesterol efflux medi-
human apo A-I promoter A sites which prevent PPARα ated by HDL or serum is correlated with cellular SR-BI
from binding to the rodent A site (90). By contrast, Rev- expression levels (106) suggest that SR-BI may pro-
erbα binds to a negative response element adjacent to mote cholesterol removal from peripheral cells includ-
the TATA box present only in the rat apo A-I promoter, ing macrophages. Therefore, SR-BI/CLA-1 may play a
but which is lacking in the human apo A-I promoter role in the transport of cholesterol by HDL from periph-
(90). Since fibrates induce rev-erbα expression, rodent eral tissues to the liver, which is known as the reverse
apo A-I expression may be repressed via this mecha- cholesterol transport pathway (107). Interestingly,
nism. The results from these studies allowed the eluci- since PPARs are expressed in human macrophages
dation of the mechanisms explaining the opposite reg- (108), the regulation of CLA-1/SR-BI by PPAR activators
ulation of human vs. rodent apo A-I in response to was recently investigated (109). Treatment of differen-
fibrate through the implication of Rev-erbα and PPARα. tiated human macrophages with PPAR activators re-
These data provide new insight into the negative regu- sulted in the induction of CLA-1 expression. Further-
more, SR-BI is induced in aortas of apo E-deficient mice
upon treatment with PPAR activators. Together this
data point to a major role of PPARs in the regulation of
HDL metabolism.

Conclusion

Investigations over the last five years have consider-


ably improved our knowledge on the metabolic conse-
quences of the activation of the PPAR signalling path-
ways. The important role of PPARs in regulating lipid
Fig. 5 Scheme summarizing our current understanding of and lipoprotein metabolism and in mediating the ac-
the species-specific regulation of apo A-I gene transcription tions of pharmacological compounds is becoming
by fibrates. more precisely understood, even though the mecha-

Brought to you by | University of Arizona


Authenticated
Download Date | 5/28/15 10:38 AM
8 Gervois et al.: Role of PPARs in lipid and lipoprotein metabolism

nism of negative regulation of certain genes such as on lipid metabolism and adipocyte differentiation.
apo C-III by PPARs requires further mechanistic stud- Biochim Biophys Acta 1996; 1302:93–109.
ies. Taking into account the effects that currently clini- 14. Cullingford TE, Bhakoo K, Peuchen S, Dolphin CT, Patel R,
and Clark JB. Distribution of mRNAs encoding the perox-
cally used PPAR activators have on energy and lipid
isome proliferator-activated receptor alpha, beta, and
homeostasis, PPARs can be considered as promising
gamma and the retinoid X receptor alpha, beta, and
pharmacological targets for the rational development gamma in rat central nervous system. J Neurochem
of novel compounds useful in the treatment of dyslipi- 1998; 70:1366–75.
demia and related disorders. 15. Braissant O, Foufelle F, Scotto C, Dauca M, and Wahli W.
Differential expression of peroxisome proliferator-acti-
vated receptors (PPARs): tissue distribution of PPAR-al-
References pha, -beta, and -gamma in the adult rat. Endocrinology
1996; 137:354–66.
1. Randomised trial of cholesterol lowering in 4444 patients 16. Auboeuf D, Rieusset J, Fajas L, Vallier P, Frering V, Riou
with coronary heart disease: the Scandinavian Simvas- JP, et al. Tissue distribution and quantification of the ex-
tatin Survival Study (4S). Lancet 1994; 344:1383–9. pression of mRNAs of peroxisome proliferator-activated
2. Shepherd J, Cobbe SM, Ford I, Isles C, Lorimer A, Mac- receptors and liver X receptor-alpha in humans: no alter-
Farlane P, et al.Prevention of coronary heart disease with ation in adipose tissue of obese and NIDDM patients. Di-
pravastatin in men with hypercholesterolemia. West abetes 1997; 46:1319–27.
Scotland Coronary Prevention Study Group. N Engl J 17. Dreyer C, Krey G, Keller H, Givel F, Helftenbein G, and
Med 1995; 333:1301–7. Wahli W. Control of the peroxisomal beta-oxidation path-
3. Saku K, Zhang B, Ohta T, and Arakawa K. Quantity and way by a novel family of nuclear hormone receptors. Cell
function of high density lipoprotein as an indicator of 1992; 68:879–87.
coronary atherosclerosis. J Am Coll Cardiol 1999; 18. Braissant O, and Wahli W. Differential expression of per-
33:436–43. oxisome proliferator-activated receptor- alpha, -beta, and
4. Haim M, Benderly M, Brunner D, Behar S, Graff E, Re- -gamma during rat embryonic development. Endocrinol-
icher-Reiss H, et al. Elevated serum triglyceride levels and ogy 1998; 139:2748–54.
long-term mortality in patients with coronary heart dis- 19. Spiegelman BM. PPAR-gamma: adipogenic regulator and
ease: the Bezafibrate Infarction Prevention (BIP) Registry. thiazolidinedione receptor. Diabetes 1998; 47:507–14.
Circulation 1999; 100:475–82. 20. Mueller E, Sarraf P, Tontonoz P, Evans RM, Martin KJ,
5. Staels B, Dallongeville J, Auwerx J, Schoonjans K, Leit- Zhang M, et al. Terminal differentiation of human breast
ersdorf E, and Fruchart JC. Mechanism of action of fi- cancer through PPAR gamma. Mol Cell 1998; 1:465–70.
brates on lipid and lipoprotein metabolism. Circulation 21. Lefebvre AM, Chen I, Desreumaux P, Najib J, Fruchart JC,
1998; 98:2088–93. Geboes K, et al. Activation of the peroxisome proliferator-
6. Ericsson CG, Hamsten A, Nilsson J, Grip L, Svane B, and activated receptor gamma promotes the development of
de Faire U. Angiographic assessment of effects of bezafi- colon tumors in C57BL/6J-APCMin/+ mice. Nature Med
brate on progression of coronary artery disease in young 1998; 4:1053–7.
male postinfarction patients. Lancet 1996; 347:849–53. 22. Saez E, Tontonoz P, Nelson MC, Alvarez JG, Ming UT,
7. Frick MH, Syvanne M, Nieminen MS, Kauma H, Majahalme Baird SM, et al. Activators of the nuclear receptor
S, Virtanen V, et al. Prevention of the angiographic pro- PPARgamma enhance colon polyp formation. Nature
gression of coronary and vein-graft atherosclerosis by Med 1998; 4:1058–61.
gemfibrozil after coronary bypass surgery in men with low 23. Sarraf P, Mueller E, Jones D, King FJ, DeAngelo DJ, Par-
levels of HDL cholesterol. Lopid Coronary Angiography tridge JB, et al. Differentiation and reversal of malignant
Trial (LOCAT) Study Group. Circulation 1997; 96:2137–43. changes in colon cancer through PPARgamma. Nature
8. Rubins HB, Robins SJ, Collins D, Fye CL, Anderson JW, Med 1998; 4:1046–52.
Elam MB, et al. Gemfibrozil for the secondary prevention 24. Willson TM, and Wahli W. Peroxisome proliferator-acti-
of coronary heart disease in men with low levels of high- vated receptor agonists. Curr Opin Chem Biol 1997;
density lipoprotein cholesterol. Veterans Affairs High- 1:235–41.
Density Lipoprotein Cholesterol Intervention Trial Study 25. Nagy L, Tontonoz P, Alvarez JG, Chen H, and Evans RM.
Group. N Engl J Med 1999; 341:410–8. Oxidized LDL regulates macrophage gene expression
9. Reddy J, and Qureshi S. Tumorigenicity of the hypolipi- through ligand activation of PPARγ. Cell 1998; 93:229–40.
daemic peroxisome proliferator ethyl-alpha-p-chlorophe- 26. Xu HE, Lambert MH, Montana VG, Parks DJ, Blanchard
noxyisobutyrate (clofibrate) in rats. Br J Cancer 1979; SG, Brown PJ, et al. Molecular recognition of fatty acids
40:476–82. by peroxisome proliferator-activated receptors. Mol Cell
10. Issemann I, and Green S. Activation of a member of the 1999; 3:397–403.
steroid hormone receptor superfamily by peroxisome 27. Berger J, Leibowitz MD, Doebber TW, Elbrecht A, Zhang
proliferators. Nature 1990; 347:645–50. B, Zhou G, et al. Novel peroxisome proliferator-activated
11. Vanden Heuvel JP. Peroxisome proliferator-activated re- receptor (PPAR) gamma and PPARdelta ligands pro-
ceptors: a critical link among fatty acids, gene expression duce distinct biological effects. J Biol Chem 1999;
and carcinogenesis. J Nutr 1999; 129:575S-80S. 274:6718–25.
12. Lee SS, Pineau T, Drago J, Lee EJ, Owens JW, Kroetz DL, 28. Gervois P, Chopin-Delannoy S, Fadel A, Dubois G, Kosykh
et al. Targeted disruption of the alpha isoform of the per- V, Fruchart JC, et al. Fibrates increase human REV-ERBal-
oxisome proliferator-activated receptor gene in mice re- pha expression in liver via a novel peroxisome prolifera-
sults in abolishment of the pleiotropic effects of peroxi- tor-activated receptor response element. Mol Endocrinol
some proliferators. Mol Cell Biol 1995; 15:3012–22. 1999; 13:400–9.
13. Schoonjans K, Staels B, and Auwerx J. The peroxisome 29. Staels B, Koenig W, Habib A, Merval R, Lebret M, Pineda
proliferator activated receptors (PPARs) and their effects Torra I, et al. Activation of human aortic smooth-muscle

Brought to you by | University of Arizona


Authenticated
Download Date | 5/28/15 10:38 AM
Gervois et al.: Role of PPARs in lipid and lipoprotein metabolism 9

cells is inhibited by PPARalpha but not by PPARgamma 45. Werman A, Hollenberg A, Solanes G, Bjorbaek C, Vidal-
activators. Nature 1998; 393:790–3. Puig AJ, and Flier JS. Ligand-independent activation do-
30. Ricote M, Li AC, Willson TM, Kelly CJ, and Glass CK. The main in the N terminus of peroxisome proliferator-acti-
peroxisome proliferator-activated receptor-gamma is a vated receptor gamma (PPARgamma). Differential
negative regulator of macrophage activation. Nature activity of PPARgamma1 and -2 isoforms and influence of
1998; 391:79–82. insulin. J Biol Chem 1997; 272:20230–5.
31. Jiang C, Ting AT, and Seed B. PPAR-gamma agonists in- 46. Wu Z, Bucher NL, and Farmer SR. Induction of peroxi-
hibit production of monocyte inflammatory cytokines. some proliferator-activated receptor gamma during the
Nature 1998; 391:82–6. conversion of 3T3 fibroblasts into adipocytes is mediated
32. Sakai M, Matsushima-Hibiya Y, Nishizawa M, and Nishi S. by C/EBPbeta, C/EBPdelta, and glucocorticoids. Mol Cell
Suppression of rat gluthatione transferase P expression Biol 1996; 16:4128–36.
by peroxisome proliferators:interaction between Jun and 47. Juge-Aubry CE, Hammar E, Siegrist-Kaiser C, Pernin A,
peroxisome proliferator-activated receptor α. Cancer Res Takeshita A, Chin WW, et al. Regulation of the transcrip-
1995; 53:5370–6. tional activity of the peroxisome proliferator-activated
33. Delerive P, Martin F, Chinetti G, Trottein F, Fruchart JC, Na- receptor alpha by phosphorylation of a ligand-indepen-
jib J, et al. PPAR activators inhibit thrombin-induced en- dent trans-activating domain. J Biol Chem 1999;
dothelin-1 production in human vascular endothelial cells 274:10505–10.
by inhibiting the AP-1 signalling pathways. Circ Res 1999; 48. Shao D, Rangwala SM, Bailey ST, Krakow SL, Reginato
85:394–402. MJ, and Lazar MA. Interdomain communication regulat-
34. Delerive P, De Bosscher K, Besnard S, Vanden Berghe W, ing ligand binding by PPAR-gamma. Nature 1998;
Peters JM, Gonzalez FJ, et al. PPARα negatively regulates 396:377–80.
the vascular wall inflammatory gene response by nega- 49. Shalev A, Siegrist-Kaiser CA, Yen PM, Wahli W, Burger
tive cross-talk with transcription factors NF−κB and AP-1. AG, Chin WW, et al. The peroxisome proliferator-acti-
J Biol Chem 1999; 274:32048–54. vated receptor alpha is a phosphoprotein: regulation by
35. Lemberger T, Staels B, Saladin R, Desvergne B, Auwerx J, insulin. Endocrinology 1996; 137:4499–502.
and Wahli W. Regulation of the peroxisome proliferator- 50. Adams M, Reginato MJ, Shao D, Lazar MA, and Chatter-
activated receptor alpha gene by glucocorticoids. J Biol jee VK. Transcriptional activation by peroxisome prolifer-
Chem 1994; 269:24527–30. ator-activated receptor gamma is inhibited by phospho-
36. Steineger HH, Sorensen HN, Tugwood JD, Skrede S, Spy- rylation at a consensus mitogen- activated protein kinase
devold O, and Gautvik KM. Dexamethasone and insulin site. J Biol Chem 1997; 272:5128–32.
demonstrate marked and opposite regulation of the 51. Hu E, Kim JB, Sarraf P, and Spiegelman BM. Inhibition of
steady-state mRNA level of the peroxisomal proliferator- adipogenesis through MAP kinase-mediated phosphory-
activated receptor (PPAR) in hepatic cells. Hormonal lation of PPARgamma. Science 1996; 274:2100–3.
modulation of fatty acid-induced transcription. Eur J 52. Camp HS, Tafuri SR, and Leff T. c-Jun N-terminal kinase
Biochem 1994; 225:967–74. phosphorylates peroxisome proliferator-activated recep-
37. Wang MY, and Unger RH. Novel form of lipolysis induced tor-γ1 and negatively regulates its transcriptional activity.
by leptin. J Biol Chem 1999; 274:17541–4. Endocrinology 1999; 140:392–7.
38. Zhou YT, Wang ZW, Higa M, Newgard CB, and Unger RH. 53. Hashimoto T, Fujita T, Usuda N, Cook W, Qi C, Peters JM,
Reversing adipocyte differentiation: implications for et al. Peroxisomal and mitochondrial fatty acid beta-oxi-
treatment of obesity. Proc Natl Acad Sci USA 1999; dation in mice nullizygous for both peroxisome prolifera-
96:2391–5. tor-activated receptor alpha and peroxisomal fatty Acyl-
39. Kersten S, Seydoux J, Peters JM, Gonzalez FJ, Desvergne CoA oxidase. Genotype correlation with fatty liver
B, and Wahli W. Peroxisome proliferator-activated recep- phenotype. J Biol Chem 1999; 274:19228–36.
tor alpha mediates the adaptive response to fasting. J 54. Qi C, Zhu Y, Pan J, Usuda N, Maeda N, Yeldandi AV, et al.
Clin Invest 1999; 103:1489–98. Absence of spontaneous peroxisome proliferation in
40. Lemberger T, Saladin R, Vazquez M, Assimacopoulos F, enoyl-CoA Hydratase/L-3-hydroxyacyl-CoA dehydroge-
Staels B, Desvergne B, et al. Expression of the peroxi- nase-deficient mouse liver. Further support for the role of
some proliferator-activated receptor α gene is stimulated fatty acyl-CoA oxidase in PPAR alpha ligand metabolism.
by stress and follows a diurnal rhythm. J Biol Chem 1996; J Biol Chem 1999; 274:15775–80.
271:1764–9. 55. Aoyama T, Peters JM, Iritani N, Nakajima T, Furihata K,
41. Gervois P, Pineda Torra I, Chinetti G, Dubois G, Grötzinger Hashimoto T, et al. Altered constitutive expression of
T, Fruchart JC, et al. A truncated human PPARα splice fatty acid-metabolizing enzymes in mice lacking the per-
variant with dominant negative activity. Mol Endocrinol oxisome proliferator-activated receptor alpha (PPARal-
1999; 13:1535–49. pha). J Biol Chem 1997; 273:5678–84.
42. Palmer CN, Hsu MH, Griffin KJ, Raucy JL, and Johnson 56. Martin G, Schoonjans K, Lefebvre AM, Staels B, and Auw-
EF. Peroxisome proliferator activated receptor-alpha ex- erx J. Coordinate regulation of the expression of the fatty
pression in human liver. Mol Pharmacol 1998; 53:14–22. acid transport protein and acyl-CoA synthetase genes by
43. Zhu Y, Qi C, Korenberg JR, Chen XN, Noya D, Rao MS, et PPARalpha and PPARgamma activators. J Biol Chem
al. Structural organization of mouse peroxisome prolifer- 1997; 272:28210–7.
ator-activated receptor gamma (mPPAR gamma) gene: 57. Motojima K, Passilly P, Peters JM, Gonzalez FJ, and
alternative promoter use and different splicing yield two Latruffe N. Expression of putative fatty acid transporter
mPPAR gamma isoforms. Proc Natl Acad Sci USA 1995; genes are regulated by peroxisome proliferator-acti-
92:7921–5. vated receptor alpha and gamma activators in a tissue-
44. Fajas L, Auboeuf D, Raspe E, Schoonjans K, Lefebvre AM, and inducer-specific manner. J Biol Chem 1998;
Saladin R, et al. The organization, promoter analysis, and 273:16710–4.
expression of the human PPARgamma gene. J Biol Chem 58. Mascaro C, Acosta E, Ortiz JA, Marrero PF, Hegardt FG,
1997; 272:18779–89. and Haro D. Control of human muscle-type carnitine

Brought to you by | University of Arizona


Authenticated
Download Date | 5/28/15 10:38 AM
10 Gervois et al.: Role of PPARs in lipid and lipoprotein metabolism

palmitoyltransferase I gene transcription by peroxisome fenofibrate administration. Atherosclerosis 1989;


proliferator-activated receptor. J Biol Chem 1998; 77:139–49.
273:8560–73. 73. D’Costa MA, and Angel A. Inhibition of hormone-stimu-
59. Brandt JM, Djouadi F, and Kelly DP. Fatty acids activate lated lipolysis by clofibrate. A possible mechanism for its
transcription of the muscle carnitine palmitoyltrans- hypolipidemic action. J Clin Invest 1975; 55:138–48.
ferase I gene in cardiac myocytes via the peroxisome 74. Caslake MJ, Packard CJ, Gaw A, Murray E, Griffin BA, Val-
proliferator-activated receptor alpha. J Biol Chem 1998; lance BD, et al. Fenofibrate and LDL metabolic hetero-
273:23786–92. geneity in hypercholesterolemia. Arterioscler Thromb
60. Yu GS, Lu YC, and Gulick T. Co-regulation of tissue-spe- 1993; 13:702–11.
cific alternative human carnitine palmitoyltransferase 75. Vu-Dac N, Schoonjans K, Kosykh V, Dallongeville J,
Ibeta gene promoters by fatty acid enzyme substrate. J Fruchart JC, Staels B, et al. Fibrates increase human
Biol Chem 1998; 273:32901–9. apolipoprotein A-II expression through activation of the
61. Djouadi F, Weinheimer CJ, Saffitz JE, Pitchford C, Bastin peroxisome proliferator-activated receptor. J Clin Invest
J, Gonzalez FJ, et al. A gender-related defect in lipid me- 1995; 96:741–50.
tabolism and glucose homeostasis in peroxisome prolif- 76. Vu-Dac N, Schoonjans K, Laine B, Fruchart JC, Auwerx J,
erator- activated receptor alpha- deficient mice. J Clin In- and Staels B. Negative regulation of the human
vest 1998; 102:1083–91. apolipoprotein A-I promoter by fibrates can be attenu-
62. Leone TC, Weinheimer CJ, and Kelly DP. A critical role for ated by the interaction of the peroxisome proliferator-ac-
the peroxisome proliferator-activated receptor alpha tivated receptor with its response element. J Biol Chem
(PPARalpha) in the cellular fasting response: The PPARal- 1994; 269:31012–8.
pha-null mouse as a model of fatty acid oxidation disor- 77. Berthou L, Duverger N, Emmanuel F, Langouet S, Auwerx
ders. Proc Natl Acad Sci USA 1999; 96:7473–8. J, Guillouzo A, et al. Opposite regulation of human versus
63. Kelly LJ, Vicario PP, Thompson GM, Candelore MR, Doeb- mouse apolipoprotein A-I by fibrates in human
ber TW, Ventre J, et al. Peroxisome proliferator-activated apolipoprotein A-I transgenic mice. J Clin Invest 1996;
receptors gamma and alpha mediate in vivo regulation of 97:2408–16.
uncoupling protein (UCP-1, UCP-2, UCP-3) gene expres- 78. Stevenson RW, Hutson NJ, Krupp MN, Volkmann RA, Hol-
sion. Endocrinology 1998; 139:4920–7. land GF, Eggler JF, et al. Actions of novel antidiabetic
64. Brun S, Carmona MC, Mampel T, Vinas O, Giralt M, Igle- agent englitazone in hyperglycemic hyperinsulinemic
sias R, et al. Activators of peroxisome proliferator-acti- ob/ob mice. Diabetes 1990; 39:1218–27.
vated receptor-alpha induce the expression of the uncou- 79. Sohda T, Mizuno K, Momose Y, Ikeda H, Fujita T, and Me-
pling protein-3 gene in skeletal muscle: a potential guro K. Studies on antidiabetic agents. 11. Novel thiazo-
mechanism for the lipid intake-dependent activation of lidinedione derivatives as potent hypoglycemic and hy-
uncoupling protein-3 gene expression at birth. Diabetes polipidemic agents. J Med Chem 1992; 35:2617–26.
1999; 48:1217–22. 80. Kemnitz JW, Elson DF, Roecker EB, Baum ST, Bergman
65. Tsuboyama-Kasaoka N, Takahashi M, Kim H, and Ezaki O. RN, and Meglasson MD. Pioglitazone increases insulin
Up-regulation of liver uncoupling protein-2 mRNA by ei- sensitivity, reduces blood glucose, insulin, and lipid lev-
ther fish oil feeding or fibrate administration in mice. els, and lowers blood pressure, in obese, insulin- resis-
Biochem Biophys Res Commun 1999; 257:879–85. tant rhesus monkeys. Diabetes 1994; 43:204–11.
66. Tontonoz P, Hu E, Graves RA, Budavari AI, and Spiegel- 81. Young PW, Cawthorne MA, Coyle PJ, Holder JC, Holman
man BM. mPPAR gamma 2: tissue-specific regulator of an GD, Kozka IJ, et al. Repeat treatment of obese mice with
adipocyte enhancer. Genes Dev 1994; 8:1224–34. BRL 49653, a new potent insulin sensitizer, enhances in-
67. Tontonoz P, Hu E, Devine J, Beale EG, and Spiegelman sulin action in white adipocytes. Association with in-
BM. PPAR gamma 2 regulates adipose expression of the creased insulin binding and cell-surface GLUT4 as mea-
phosphoenolpyruvate carboxykinase gene. Mol Cell Biol sured by photoaffinity labeling. Diabetes 1995;
1995; 15:351–7. 44:1087–92.
68. Schoonjans K, Watanabe M, Suzuki H, Mahfoudi A, Krey 82. Lohray BB, Bhushan V, Rao BP, Madhavan GR, Murali N,
G, Wahli W, et al. Induction of the acyl-coenzyme A syn- Rao KN, et al. Novel euglycemic and hypolipidemic
thetase gene by fibrates and fatty acids is mediated by a agents. 1. J Med Chem 1998; 41:1619–30.
peroxisome proliferator response element in the C pro- 83. Reddy KA, Lohray BB, Bhushan V, Reddy AS, Mamidi NV,
moter. J Biol Chem 1995; 270:19269–76. Reddy PP, et al. Novel antidiabetic and hypolipidemic
69. Schoonjans K, Staels B, Grimaldi P, and Auwerx J. Acyl- agents. 5. Hydroxyl versus benzyloxy containing chro-
CoA synthetase mRNA expression is controlled by fibric- man derivatives. J Med Chem 1999; 42:3265–78.
acid derivatives, feeding and liver proliferation. Eur J 84. Lefebvre AM, Peinado-Onsurbe J, Leitersdorf I, Briggs
Biochem 1993; 216:615–22. MR, Paterniti JR, Fruchart JC, et al. Regulation of lipopro-
70. Schoonjans K, Peinado-Onsurbe J, Lefebvre AM, Hey- tein metabolism by thiazolidinediones occurs through a
man RA, Briggs M, Deeb S, et al. PPARalpha and distinct but complementary mechanism relative to fi-
PPARgamma activators direct a distinct tissue-specific brates. Arterioscler Thromb Vasc Biol 1997; 17:1756–64.
transcriptional response via a PPRE in the lipoprotein li- 85. Hertz R, Bishara-Shieban J, and Bar-Tana J. Mode of ac-
pase gene. EMBO J 1996; 15:5336–48. tion of peroxisome proliferators as hypolipidemic drugs.
71. Heller F, and Harvengt C. Effects of clofibrate, bezafibrate, Suppression of apolipoprotein C-III. J Biol Chem 1995;
fenofibrate and probucol on plasma lipolytic enzymes in 270:13470–5.
normolipaemic subjects. Eur J Clin Pharmacol 1983; 86. Staels B, Vu-Dac N, Kosykh V, Saladin R, Fruchart JC, Dal-
25:57–63. longeville J, et al. Fibrates down-regulate apolipoprotein
72. Malmendier CL, Lontie JF, Delcroix C, Dubois DY, Magot C-III expression independent of induction of peroxisomal
T, and De Roy L. Apolipoproteins C-II and C-III metabolism Acyl co-enzyme A oxidase. J Clin Invest 1995; 95:705–12.
in hypertriglyceridemic patients. Effect of a drastic 87. Peters JM, Hennuyer N, Staels B, Fruchart JC, Fievet C,
triglyceride reduction by combined diet restriction and Gonzalez FJ, et al. Alterations in lipoprotein metabolism

Brought to you by | University of Arizona


Authenticated
Download Date | 5/28/15 10:38 AM
Gervois et al.: Role of PPARs in lipid and lipoprotein metabolism 11

in peroxisome proliferator-activated receptor alpha-defi- pha-isoform deficiency leads to progressive dyslipidemia


cient mice. J Biol Chem 1997; 272:27307–12. with sexually dimorphic obesity and steatosis. J Biol
88. Ladias JA, Hadzopoulou-Cladaras M, Kardassis D, Cardot Chem 1998; 273:29577–85.
P, Cheng J, Zannis V, et al. Transcriptional regulation of 100. Hennuyer N, Poulain P, Madsen L, Berge RK, Houdebine
human apolipoprotein genes ApoB, ApoCIII, and ApoAII LM, Branellec D, et al. Beneficial effects of fibrates on
by members of the steroid hormone receptor superfamily apolipoprotein A-I metabolism occur independently of
HNF- 4, ARP-1, EAR-2, and EAR-3. J Biol Chem 1992; any peroxisome proliferative response. Circulation 1999;
267:15849–60. 99:2445–51.
89. Vu-Dac N, Gervois P, Pineda Torra I, Fruchart JC, Kosykh 101. Staels B, van Tol A, Andreu T, and Auwerx J. Fibrates in-
V, Kooistra T, et al. Retinoids increase human apo C-III ex- fluence the expression of genes involved in lipoprotein
pression at the transcriptional level via the retinoid X re - metabolism in a tissue-selective manner in the rat. Arte-
ceptor. Contribution to the hypertriglyceridemic action of rioscler Thromb 1992; 12:286–94.
retinoids. J Clin Invest 1998; 102:625–32. 102. Staels B, Peinado-Onsurbe J, and Auwerx J. Down-regu-
90. Vu-Dac N, Chopin-Delannoy S, Gervois P, Bonnelye E, lation of hepatic lipase gene expression and activity by
Martin G, Fruchart JC, et al. The nuclear receptors perox- fenofibrate. Biochim Biophys Acta 1992; 1123:227–30.
isome proliferator-activated receptorα and Rev-erbα me- 103. Staels B, van Tol A, Skretting G, and Auwerx J.
diate the species-specific regulation of apolipoprotein A-I Lecithin:cholesterol acyltransferase gene expression is
expression by fibrates. J Biol Chem 1998; 273:25713–20. regulated in a tissue-selective manner by fibrates. J Lipid
91. Duez H, Mansen A, Fruchart JC, Vennstrom B, Fievet C, Res 1992; 33:727–35.
and Staels B. Rev-erbα : a nuclear receptor with a role in 104. Acton S, Rigotti A, Landschulz KT, Xu S, Hobbs HH, and
lipoprotein metabolism. Circulation 1998; 98:17,I- Krieger M. Identification of scavenger receptor SR-BI as a
449:2363. high density lipoprotein receptor. Science 1996;
92. Poynter ME, and Daynes RA. Peroxisome proliferator-ac- 271:518–20.
tivated receptor alpha activation modulates cellular re- 105. Calvo D, Gomez-Coronado D, Lasuncion MA, and Vega
dox status, represses nuclear factor-kappaB signaling, MA. CLA-1 is an 85-kD plasma membrane glycoprotein
and reduces inflammatory cytokine production in aging. that acts as a high- affinity receptor for both native (HDL,
J Biol Chem 1998; 273:32833–41. LDL, and VLDL) and modified (OxLDL and AcLDL)
93. Zhou YC, and Waxman DJ. Cross-talk between janus ki- lipoproteins. Arterioscler Thromb Vasc Biol 1997;
nase-signal transducer and activator of transcription 17:2341–9.
(JAK-STAT) and peroxisome proliferator-activated recep- 106. Ji Y, Jian B, Wang N, Sun Y, Moya ML, Phillips MC, et al.
tor-alpha (PPARα) signalling pathways. Growth hormone Scavenger receptor BI promotes high density lipopro-
inhibition of PPARα transcriptional activity mediated by tein-mediated cellular cholesterol efflux. J Biol Chem
STAT5b. J Biol Chem 1999; 274:2672–81. 1997; 272:20982–5.
94. Miller GJ, and Miller NE. Plasma-high-density-lipoprotein 107. Trigatti B, Rayburn H, Vinals M, Braun A, Miettinen H,
concentration and development of ischaemic heart-dis- Penman M, et al. Influence of the high density lipoprotein
ease. Lancet 1975; 1:16–9. receptor SR-BI on reproductive and cardiovascular
95. Balfour JA, McTavish D, and Heel RC. Fenofibrate. A re- pathophysiology. Proc Natl Acad Sci USA 1999;
view of its pharmacodynamic and pharmacokinetic prop- 96:9322–7.
erties and therapeutic use in dyslipidaemia. Drugs 1990; 108. Chinetti G, Griglio S, Antonucci M, Pineda Torra I,
40:260–90. Delerive P, Majd Z, et al. Activation of proliferator-acti-
96. Malmendier CL, and Delcroix C. Effects of fenofibrate on vated receptors alpha and gamma induces apoptosis of
high and low density lipoprotein metabolism in heterozy- human monocyte-derived macrophages. J Biol Chem
gous familial hypercholesterolemia. Arteriosclerosis 1998; 273:25573–80.
1985; 55:161–9. 109. Gbaguidi F, Chinetti G, Griglio S, Antonucci M, Fruchart
97. Bard JM, Parra HJ, Camare R, Luc G, Ziegler O, Dachet C, JC, Chapman J, et al. Regulation of CLA-1 (CD36 and
et al. A multicenter comparison of the effects of simvas- LIMP II analogous I) by activators of peroxisome prolifer-
tatin and fenofibrate therapy in severe primary hypercho- ator activated receptors (PPARs). Atherosclerosis 1999;
lesterolemia, with particular emphasis on lipoproteins 144: Suppl 1:112.
defined by their apolipoprotein composition. Metabolism
1992; 41:498–503. Received 18 October 1999; accepted in present form
98. Berthou L, Saladin R, Yaqoob P, Branellec D, Calder P, 4 January 2000
Fruchart JC, et al. Regulation of rat liver apolipoprotein A-
I, apolipoprotein A-II and acyl-coenzyme A oxidase gene Corresponding author: Bart Staels, INSERM U.325,
expression by fibrates and dietary fatty acids. Eur J Département d’Athérosclérose, Institut Pasteur de Lille 1,
Biochem 1995; 232:179–87. rue du Professeur Calmette, BP245, 59019 Lille Cedex, France
99. Costet P, Legendre C, More J, Edgar A, Galtier P, and Tel:+33-3-20-87-73-88, Fax: +33-3-20-87-73-60
Pineau T. Peroxisome proliferator-activated receptor al- Email: Bart.Staels@pasteur-lille.fr

Brought to you by | University of Arizona


Authenticated
Download Date | 5/28/15 10:38 AM

Potrebbero piacerti anche