You are on page 1of 7

Mutation Research 690 (2010) 57–63

Contents lists available at ScienceDirect


Mutation Research/Fundamental and Molecular
Mechanisms of Mutagenesis
journal homepage: www.elsevier.com/locate/molmut
Community address: www.elsevier.com/locate/mutres

Role of PPAR-gamma in inflammation. Prospects for therapeutic intervention by


food components夽
Harry Martin ∗
The New Zealand Institute for Plant & Food Research Limited, Palmerston North 4474, New Zealand

a r t i c l e i n f o a b s t r a c t

Article history: Peroxisome proliferator activated receptor gamma (PPAR␥) is a ligand-dependent transcription factor
and a member of the nuclear receptor superfamily. Acting as sensors of hormones, vitamins, endoge-
nous metabolites and xenobiotic compounds, the nuclear receptors control the expression of a very
Keywords: large number of genes. PPAR␥ has been known for some time to regulate adipocyte differentiation, fatty
Peroxisome proliferator activated receptor acid storage and glucose metabolism, and is a target of anti-diabetic drugs. More recently, PPAR␥ has
gamma (PPAR␥)
been recognized as playing a fundamentally important role in the immune response through its abil-
Inflammatory disease
ity to inhibit the expression of inflammatory cytokines and to direct the differentiation of immune cells
Diet
towards anti-inflammatory phenotypes. A feature of PPAR␥ is the structural diversity of its ligands, which
encompass endogenous metabolites, dietary compounds and synthetic drugs. The high and increasing
incidence of inflammatory and allergic disease, coupled with encouraging results from recent clinical
trials, suggest that natural PPAR␥ agonists found in foods may be beneficial to human health by acting
as anti-inflammatory molecules. PPAR␥ is therefore not only a target of the pharmaceutical industry,
but also of great potential interest to the food industry, since it is activated by several natural dietary
constituents. The prospects for dietary intervention in inflammatory disease have improved somewhat
over the last few years, and are reviewed here.
© 2009 Elsevier B.V. All rights reserved.

1. Introduction—PPAR␥ and its role in inflammation and prevalence of these illnesses in Asia are increasing [2–4]. The
risk of developing colorectal carcinoma is increased for individu-
Inflammatory disease is a broad term encompassing a wide als with inflammatory bowel disease [5]. In geographical regions
range of pathological conditions affecting many organs and tis- where diets are high in n-3 polyunsaturated fatty acids (PUFAs)
sues. Inflammatory diseases include inflammatory bowel disease, cancer incidence is low [6]. It has been suggested that some of the
atherosclerosis, rheumatoid arthritis, multiple sclerosis, asthma protective effect of dietary n-3 PUFAs may be due to their anti-
and psoriasis. The incidence of atopic illness including asthma has inflammatory, PPAR␥ activating properties [7].
increased considerably over recent decades [1]. Ulcerative Colitis The peroxisome proliferator activated receptors (PPARs) are
and the less prevalent, but more severe, Crohn’s disease, were both a subset of the nuclear receptor superfamily. Unlike the classi-
considered to be the diseases of Western societies but the incidence cal hormone-activated receptors such as the estrogen receptor,
which is located in the cytoplasm and translocates to the nucleus
after binding of the activating ligand, the PPAR receptors reside
DOI of original article:10.1016/j.mrfmmm.2009.06.009. in the nucleus bound to DNA response elements [8]. The nuclear
Abbreviations: 15d-PGJ2 , 15-deoxy-12,14 prostaglandin J2; 5-ASA, 5- location of the PPAR receptor is typical of metabolite-activated
aminosalicylic acid; c9, t11, cis-9 trans-11; CLA, conjugated linoleic acid; DHA, nuclear receptors. There are three forms of PPAR receptor: alpha,
docosahexaenoic acid; EPA, eicosapentaenoic acid; IgA, immunoglobulin A; IL, beta/delta and gamma [9], all of which form obligate heterodimers
interleukin; PPAR␥, peroxisome proliferator activated receptor gamma; PUFA,
with the retinoid-X-receptor (RXR-receptor). PPAR␣ is the tar-
polyunsaturated fatty acid; t10, c12, trans-10, cis-12; TNF-␣, tumour necrosis factor
alpha. get for hypolipidaemic drugs known as fibrates. PPAR␤/␦ is
夽 An error resulted in this article appearing in a previous volume of this journal. now emerging as an important regulator of bowel cell prolifera-
The article is reprinted here for the reader’s convenience and for the continuity of tion/differentiation. The tissue expression of the three PPAR forms
this special issue. The details of the original publication are as follows [Martin, 2009. is different but overlapping. The functions of PPAR␥ and PPAR␣ also
Role of PPAR-gamma in inflammation. Prospects for therapeutic intervention by
overlap somewhat. PPAR␥ is expressed in a range of tissues includ-
food components. Mutation Research 669, 1–7. 10.1016/j.mrfmmm.2009.06.009].
∗ Tel.: +64 6 953 7747; fax: +64 6 953 7701. ing adipocytes, skeletal muscle cells, osteoclasts, osteoblasts and
E-mail address: hmartin@hortresearch.co.nz. several immune-type cells. Unsurprisingly, germ-line knockout of

0027-5107/$ – see front matter © 2009 Elsevier B.V. All rights reserved.
doi:10.1016/j.mrfmmm.2009.09.009
58 H. Martin / Mutation Research 690 (2010) 57–63

PPAR␥ is lethal. Four transcripts of the PPAR␥ gene are found in underlying these bizarre symptoms was traced to the default syn-
human tissue as a result of tissue specific variations in promoters thesis of toxic, highly inflammatory oxidised lipids in the milk of
and splicing [10–12]. However in normal cells only two proteins, mice lacking the endothelial PPAR␥ gene. These oxidised lipids
PPARG-1 and PPARG-2 are expressed. PPARG-1 protein can be pro- were generated due to a huge excess of lipoxygenase activity, nor-
duced from transcripts PPARG-1, PPARG-3 and PPARG-4. PPARG-1 mally repressed by the endothelial PPAR␥ gene. Accumulation of
protein is widely expressed whereas PPARG-2 is mainly restricted the inflammatory lipid mediators in the skin of the suckling mice
to adipocytes [13]. caused inflammation and subsequent alopecia. Skin inflammation
PPAR␥ is activated, not only by ligand (agonist) binding, but was confirmed as the cause of alopecia when topical aspirin cured
also by phosphorylation, which increases the ligand-independent the inflammation and prevented the hair loss [21]. These exam-
transcriptional activity [14]. Apart from its role as a transcrip- ples suggest that systemic inflammation is the default physiological
tion factor, PPAR␥ also acts as a trans-repressor of macrophage condition, held in check by PPAR␥ under normal circumstances.
inflammatory genes [15]. In this mechanism the ligand-dependent Animal studies and human trials of PPAR␥ activating drugs, nor-
sumoylation of PPAR␥ represses inflammatory gene expression. mally used to treat diabetes, have shown these compounds to have
Binding of sumoylated PPAR␥ to a DNA-bound repressor complex great potential as anti-inflammatory drugs. In a mouse model of
blocks the expression of inflammatory gene products by prevent- asthma, pioglitazone, a PPAR␥ activator, was found to be as effec-
ing the 19S proteasome-mediated degradation of the repressor tive as dexamethasone, a corticosteroid commonly used in asthma
complex. Ligand-independent activation of PPAR␤/␦ can suppress treatment [22]. In a rat model of rheumatoid arthritis known as
bowel disease by down regulation of inflammatory signalling [16]. adjuvant induced arthritis, the PPAR␥ activating drugs pioglita-
Evidence of the role of PPAR␥ in inflammatory disease is appar- zone and rosiglitazone, were found to reduce bone erosion and
ent from genetic screening of patients suffering from multiple inflammatory bone loss [23]. Using Type II diabetic rats as a model
sclerosis (MS), an autoimmune disease of the central nervous of inflammatory renal disease, pioglitazone reduced nephropathy
system. Various genetic linkages to immune genes have been rec- by an anti-inflammatory mechanism [24]. The successes of PPAR␥
ognized for MS, notably the receptors for interleukin (IL) 2 and activators in animal models of the autoimmune disease multi-
IL7. Very recently a PPAR␥ polymorphism has been added to ple sclerosis (MS) have given strong support to the use of PPAR␥
the list of MS-linked genes. In a study of 116 patients and 211 agonists in human trials [25]. The use of rosiglitazone in human
healthy age-matched controls, the Ala/Ala genotype of the PPAR␥ inflammatory bowel disease gave beneficial results in an Ulcera-
Pro12Ala polymorphism was associated with a 10-year, statisti- tive Colitis clinical trial [26]. The US National Institutes of Health
cally significantly delayed onset of disease in patients with MS are currently recruiting participants for a trial of pioglitazone in
[17]. The anti-inflammatory effects of the Pro12Ala polymorphism the inflammatory diseases rheumatoid arthritis and atherosclero-
are corroborated by a recent study of atherosclerosis, a chronic, sis (clinical trial identifier NCT00554853). A trial of pioglitazone in
macrophage-mediated, inflammatory disease of the arterial blood asthma (clinical trial identifier NCT00787644) is currently in the
vessels. This 10-year follow-up study of men with coronary artery planning phase.
disease showed that carriers of the Pro12Ala allele of PPAR␥ have PPAR␥ is of particular interest to the food industry because
less widespread atherosclerosis and are considerably protected PPAR␥ activators and their precursors e.g. linolenic and linoleic
against 10-year vascular morbidity and mortality [18]. acid are abundant in several foods. In addition, certain isoforms
The use of PPAR␥ knockout mice has provided strong evidence of linoleic acid, notably the trans-10, cis-12 isoform are normally
for the role of natural PPAR␥ ligands in controlling inflammation. rare in the diet but present in higher concentration in certain syn-
In a seminal study, colitis was induced in mice deficient in colonic thetic food supplements used for weight loss [27]. The t10, c12
PPAR␥ by two unrelated mechanisms, either by treatment with oral isoform of CLA is effective at inducing weight loss in mice, but it
dextran sodium sulfate or by transfer of CD4+ CD45RBhi T-cells. In does this by antagonising PPAR␥ and inducing the stress response
the former approach colitis is induced by chemical irritation of the in adipocytes [28,29]. On the other hand, the cis-9, trans-11 iso-
mucosa and is dependent on macrophage activation and epithelial form of linoleic acid, which is commonly found in natural food
cell apoptosis whereas in the latter method, colitis is mediated by products, inhibits allergic airway sensitization and inflammation
antigen driven TH1-cell activation. In both of these models, wild in mice [30] via a PPAR␥ dependent mechanism. The mechanisms
type mice responded to dietary conjugated linoleic acid (CLA) by by which dietary PPAR␥ activators may ameliorate inflammatory
clinical amelioration of colitis while the colonic PPAR␥ knockout disease are discussed.
mice were unresponsive [19].
The anti-inflammatory effects of PPAR␥ are closely linked with
its anti-diabetic effects. Mice with a macrophage-specific dele- 2. Anti-inflammatory effects of PPAR␥ ligands—synthetic
tion of PPAR␥ were tested for their ability to resist a challenge and natural
with Leishmania major, a parasitic infection which requires an
M1 (cell-mediated) type response from the host for immunity to PPAR␥ ligands can be divided into various categories: nat-
occur. Mice lacking macrophage PPAR␥ were not only less sus- ural/synthetic, endogenous/exogenous or covalent/reversible. To
ceptible to Leishmania spp. infection, as expected, but the same demonstrate the variety of PPAR␥ ligands, the structures of the
animals were also prone to diabetic illness [20]. Selective knockout ligands discussed in this article are shown in Fig. 1.
of macrophage PPAR␥ resulted in a reduced capacity for skele- PPAR␥ agonists known as thiazolidinediones are commonly pre-
tal muscle to metabolise fatty acids and sugars. The expression of scribed for the treatment of diabetes but have been investigated
transcription factors and co-activator proteins necessary for mito- for their anti-inflammatory effects. PPAR␥ protein was identified
chondrial biogenesis were also greatly reduced. Thus macrophage in the antigen presenting cells, monocytes and macrophages and
PPAR␥ activation not only induces differentiation of macrophages synthetic PPAR␥ agonists including pioglitazone, troglitazone and
into the non-inflammatory, M2 type but also has profound effects rosiglitazone were shown to suppress production of inflammatory
on the metabolic status of the whole mouse. cytokines by these cells [31,32]. Subsequently, PPAR␥ was identi-
A similar demonstration of the profound physiological effect of fied in dendritic cells which are potent and highly differentiated,
selective knockout (KO) of the PPAR␥ gene, this time selectively professional antigen presenting cells. The same thiazolidinedione
removing the gene from endothelial cells, resulted in the whole- compounds were demonstrated to decrease dendritic cell secre-
trunk hair loss in the offspring of the KO mice. The mechanism tion of IL12, a potent TH1-type inflammatory cytokine [33,34].
H. Martin / Mutation Research 690 (2010) 57–63 59

Fig. 1. The diversity of PPAR␥ ligands.

In addition, the synthesis of TH1-cell but not TH2-cell recruiting reduced renal scarring and inflammation in a mouse model of renal
chemokines was also suppressed by thiazolidinediones [33]. An fibrosis [39]. In a rat model of post-operative brain inflammation,
mRNA profiling study using mouse macrophages demonstrated rosiglitazone attenuated myeloperoxidase activity and qualita-
that rosiglitazone acted via PPAR␥ as a general repressor of a tively decreased expression of the cytokine markers of inflamma-
broad range of lipopolysaccharide and interferon-␥ target genes. tion, IL1␤ and tumour necrosis factor ␣ (TNF-␣) [40]. The beneficial
Taken together, these studies provide strong evidence of the anti- effects of rosiglitazone in brain inflammation are tempered by the
inflammatory function of PPAR␥ through its ability to suppress fact that it has poor penetrance through the blood–brain barrier.
TH1-type cytokine production in macrophages and dendritic cells Conceivably its effects are mediated outside the brain, perhaps in
[35]. the brain vasculature. In a mouse model of gastro-intestinal can-
Beneficial effects of the PPAR␥ activator, rosiglitazone, were didiasis, rosiglitazone prevented gut colonization by activation of
reported in two recent clinical trials for the treatment of Ulcerative PPAR␥ [41]. This effect was mediated by increased expression of
Colitis [36,37]. the mannose-receptor on mucosal macrophages. Using a rat model
Animal models of inflammation are now being used to explore of pancreatitis, it was shown that the PPAR␥ agonists 15-deoxy-
the utility of PPAR␥ agonists in other inflammatory diseases. In 12,14 prostaglandin J2 (15d-PGJ2 ) and troglitazone both suppress
a mouse model of systemic lupus erthythematosus, rosiglitazone pancreatic inflammation which was measured by reduced pancre-
reduced autoantibody production, renal disease, and atheroscle- atic weight and lowered levels of the inflammatory cytokines IL6
rosis [38]. The PPAR␥ agonist troglitazone, a thiazolidinedione, and Transforming-Growth-Factor-1␤ [42].
60 H. Martin / Mutation Research 690 (2010) 57–63

5-Aminosalicylic acid (5-ASA) is widely used in the treatment to induce weight loss. The safety of this treatment is controversial
of inflammatory bowel disease. The anti-inflammatory mechanism due to the high-content of t10, c12 linoleic acid in some synthetic
of 5-ASA has been shown to be dependent on PPAR␥ activation preparations. Recently, the probiotic bacterium Lactobacillus crispa-
in a PPAR␥ +/− mouse model of colitis [43]. These data were also tus M247, was shown to induce increased PPAR␥ expression and
confirmed by culture of human colonic biopsies. activity in the intestinal epithelial cells of mice [50]. The medi-
These human clinical trials and animal studies show that PPAR␥ ator of the PPAR␥ inducing activity was identified as bacterially
agonists can have systemic anti-inflammatory effects. Similar stud- derived hydrogen peroxide. Bacterial strains unable to produce
ies using natural dietary PPAR␥ ligands corroborate the drug-based H2 O2 were also unable to induce PPAR␥ expression while antioxi-
evidence. The dietary PUFAs and PPAR␥ ligands, docosahexaenoic dants and H2 O2 scavengers blocked the effect. These data illustrate
acid (DHA), eicosapentaenoic acid (EPA) and ␥-linolenic acid were the important point that probiotic bacteria can increase PPAR␥
used to treat the human enterocyte cell line, Caco-2 and human activity either by the metabolism of precursors into PPAR␥ ago-
dendritic cells, during exposure of the cells to IL1␤ and lipopolysac- nists or by the synthesis of compounds which enhance PPAR␥ gene
charide. Not only was secretion of the pro-inflammatory cytokines, expression.
IL6 and IL8, reduced by these PUFAs, but the expression of PPAR␥ The dependence of PPAR␥ function on the colonizing microbiota
was itself enhanced [44]. The central role of PPAR␥ in these cellu- of the gut was demonstrated when Enterococcus faecalis isolated
lar responses to PUFAs was confirmed by the use of GW 9662, a from newborn babies was shown to induce phosphorylation of
synthetic PPAR␥ antagonist, which blocked the anti-inflammatory PPAR␥ in primary colonic cells and colonic cell lines. Phospho-
actions of the PUFAs. In a recent animal feeding trial in pigs, rylation of PPAR␥ activated transcription of anti-inflammatory
linseed oil significantly reduced the expression of TNF-␣, a pro- target genes, notably including the anti-inflammatory IL10 [51].
inflammatory cytokine and marker of inflammation, and increased The PPAR␥ antagonist GW9662 specifically blocked these effects.
muscle mass [45]. Linseed oil’s main components are the PPAR␥ The authors did not identify which product of Enterococcus faecalis
agonists linoleic and linolenic acid. In this study, PPAR␥ concentra- was responsible for the PPAR␥ phosphorylation but suggested CLA
tion in the spleen and muscle negatively correlated with TNF-␣ in as a likely mediator.
the serum. A meta-analysis of several studies concluded that CLA is mod-
X-ray crystallographic studies of PPAR␥ with bound ligands has erately beneficial for human health [52]. As discussed earlier,
provided great insight into the mechanism of PPAR␥ activation. macrophage PPAR␥ has both anti-inflammatory and anti-diabetic
The crystal structures showed that the ligand binding pocket can actions. It is therefore feasible that the dietary benefits of CLA, seen
accommodate two fatty acids simultaneously. More remarkable in the meta-analysis, derive from the transformation of unconju-
was the finding that oxo-fatty acid metabolites [46] and the anti- gated linoleic acid to CLA by gut bacteria. In an in vitro study of 30
inflammatory 15-deoxy-12,14 prostaglandin J2 (15d-PGJ2 ) [47] strains of gram-positive intestinal bacteria, certain species, notably
bind covalently to the sulfhydryl group of at Cys285 in the ligand Roseburia, were capable of converting the unconjugated form of
binding domain by a Michael addition mechanism. The covalently linoleic acid (cis-9, cis-12, 18:2) to the immediate precursors of
bound ligands were shown to be particularly effective PPAR␥ acti- the health promoting cis-9, trans-11 form. An in vitro, biochemical
vators. study showed that the variability among bacterial strains was sub-
The variety and apparent lack of selectivity of the PPAR␥ ligand stantial, some strains having almost none of the required linoleate
binding pocket has led to concern about which ligands should be isomerase activity necessary to perform this metabolic step [53].
considered physiologically relevant. The discovery of the covalent- The implications from this work are that variations in the gut flora
binding nature of certain ligands strongly supports the credibility among individuals may well influence their inflammatory and dia-
of these ligands as being physiologically important since reversibly betic status.
binding ligands are much more dependant on concentration for CLA isoforms differ in terms of health benefit and PPAR␥ activa-
activity than covalent binders. This is because a reversible ligand tion. In primary cultures of human adipocytes the t10–c12 isoform
relies on a having either a high affinity or a high concentration caused adipocyte delipidation, insulin resistance and inflammation
to achieve appreciable binding to its receptor i.e. to establish a by acting as a PPAR␥ antagonist. These activities were not associ-
dynamic equilibrium in which the receptor function is affected. By ated with the more common and natural cis-9, trans-11 (c9, t11)
contrast, a covalent binder with only moderate affinity is more able isoform of CLA [54]. The pro-inflammatory effects of the trans-10,
to influence receptor function since, once covalently bound, it does cis-12 (t10, c12) CLA isoform in human adipocytes were subse-
not dissociate from the receptor. Penicillin and aspirin, are classic quently shown to be reversible by resveratrol [55] in an in vitro,
examples of covalently binding drugs. cell-based study. A 16-week dietary trial comparing c9, t11 CLA
with the synthetic form t10, c12 CLA in a group of 75 healthy post-
menopausal women, found that the t10, c12 isoform increased
3. Dietary PPAR␥ activators, their sources and levels of inflammatory markers including C-reactive protein and
bioavailability fibrinogen [56].
A study of the effects of long term feeding of c9, t11 CLA to
Linoleic acid is a well known component of many foods and rats gave two compelling examples of beneficial immune modu-
is present in vegetables, fruits, nuts, grains and seeds among lation from an analysis of their immune response to ovalbumin
other dietary sources. Linoleic and linolenic acids are highly orally [57]. Firstly, the ovalbumin-specific response of the splenocytes
bioavailable to the plasma and the brain. Indeed the exceptional was enhanced 50%, while at the same time, the polyclonal spleno-
bioavailability characteristics of these fatty acids have lead to their cyte response to mitogen stimulation was reduced by up to
design as drug-delivery vehicles in emulsions with anti-HIV pro- 20%. Secondly, the intestinal anti-ovalbumin immunoglobulin A
tease treatments to enhance drug delivery to plasma and brain (IgA) response was increased by 75%. Although these effects may
[48], demonstrated using a mouse model. The conjugated form of be mediated, in part, by PPAR␥, no evidence was presented in
linoleic acid cis-9, trans-11, a well researched PPAR␥ ligand, was this particular study to test PPAR␥ involvement in the immuno-
shown to be produced naturally from linoleic acid by the action logically beneficial effects of dietary c9, t11 CLA. However the
of gut flora, especially probiotic bacteria [49]. In the same study anti-lymphoproliferative effects of the dietary PUFAs and PPAR␥
using human HT-29 colonic carcinoma cells, CLA was also shown ligands, linoleic acid and DHA have been demonstrated on human
to enhance expression of PPAR␥. CLA is sold as a dietary supplement dendritic cells [58]. In this cell-based, in vitro study, specific block-
H. Martin / Mutation Research 690 (2010) 57–63 61

ing of PPAR␥ activity with the antagonist GW9662, also blocked the in mice. The importance of PPAR␥ in inflammatory disease is
anti-inflammatory effects of the PUFAs. evident from the success of PPAR␥ activating drugs in animal dis-
Although the evidence for PPAR␥ mediated anti-inflammatory ease models and human Ulcerative Colitis trials. Furthermore, the
effects is convincing, interpretation of the data from CLA feed- genetic association between PPAR␥ polymorphisms and diseases
ing trial is complicated by the fact that CLA is also an activator such as multiple sclerosis and atherosclerosis confirms the role
of PPAR␣ and PPAR␤/␦ [59,60]. Likewise, in the mRNA profiling of PPAR␥ in human inflammatory disease. These data support the
study using rosiglitazone to stimulate PPAR␥ receptors in mouse hypothesis that PPAR␥ activation in healthy individuals suppresses
macrophages, it was apparent, using PPAR␥ deficient macrophages, systemic inflammation. The prospects that dietary factors may have
that PPAR␦ was activated at higher concentrations of rosiglitazone beneficial effects in inflammatory disease are good, since various
[35]. Similarly, in a study using normal and PPAR␣ deficient mice, dietary ligands such as conjugated (c9, t11) linoleic acid or DHA,
the PPAR␥ ligand, pioglitazone was shown to suppress inflamma- are present in certain foods. Pre-biotic foods and probiotic bacte-
tion through a PPAR␣ dependent mechanism [61]. Thus the human ria play an important role in influencing the immune phenotype
health benefits of PPAR␥ agonists, in the diet and from pharmaceu- through their ability to convert food components into PPAR␥ ago-
tical sources, are probably not restricted to their PPAR␥ activating nists and to synthesise compounds de novo which stimulate PPAR␥
properties. Furthermore the metabolic fate of CLA in humans is not gene expression. The discovery of new dietary PPAR␥ activators
fully understood but it is apparent that the conversion of CLA to coupled with understanding of the subtle mechanisms by which
conjugated arachidonic acid is substantially more efficient in rats these compounds influence PPAR␥ activity promises to be an excit-
than in humans [62]. Differences in CLA metabolism may therefore ing field in the immediate future—with enormous implications for
explain the apparent disagreement between animal trials which human health.
suggest CLA induces weight loss and human trials which are equiv-
ocal on this issue [62]. Role of funding source
Dietary phytochemicals with quite different structures and
properties from the unsaturated fatty acids discussed so far, Internal funding from Plant and Food Research.
have been characterised from oregano by activity-based frac-
tionation. Various flavonoids and other phytochemicals from
oregano were characterised for their agonist and antagonist PPAR␥ Conflict of interest
activities. Compounds were first identified as ligands using a
PPAR␥ fluorescence-polarization assay and subsequently classi- None.
fied as agonist or antagonist, based on their ability to inhibit
rosiglitazone-induced cellular responses. The dietary flavonoids References
quercetin, luteolin, rosmarinic acid and diosmetin were found
to be PPAR␥ antagonists, naringenin and apigenin were modu- [1] S.F. Bloomfield, R. Stanwell-Smith, R.W.R. Crevel, J. Pickup, Too clean, or not too
clean: the hygiene hypothesis and home hygiene, Clinical and Experimental
lators and biochanin A was found to be a PPAR␥ agonist [63]. Allergy 36 (2006) 402–425.
Although the bioavailability of these phytochemicals needs to [2] K.L. Goh, S.D. Xiao, Inflammatory bowel disease: a survey of the epidemiology
be addressed, the approach demonstrates that many common in Asia, Journal of Digestive Diseases 10 (2009) 1–6.
[3] E.V. Loftus, Clinical epidemiology of inflammatory bowel disease: inci-
phytochemicals have the potential to influence PPAR␥ activity, dence, prevalence, and environmental influences, Gastroenterology 126 (2004)
due to the promiscuous nature of the binding site. The notion 1504–1517.
of potentially harmful over-activation of PPAR␥ by dietary con- [4] B.A. Jacobsen, J. Fallingborg, H.H. Rasmussen, K.R. Nielsen, A.M. Drewes, E.
Puho, G.L. Nielsen, H.T. Sorensen, Increase in incidence and prevalence of
stituents was raised by Adapala, who suggested that prolonged inflammatory bowel disease in northern Denmark: a population-based study,
consumption of curcumin, a polyphenol and principal compo- 1978–2002, European Journal of Gastroenterology & Hepatology 18 (2006)
nent of the spice turmeric, suppressed the immune response and 601–606.
[5] A.B. Carter, S.A. Misyak, R. Hontecillas, J. Bassaganya-Riera, Dietary modula-
exacerbated Leishmania donovani infection in mice [64]. However,
tion of inflammation-induced colorectal cancer through PPAR␥, PPAR Research
curcumin is not itself a PPAR␥ ligand and probably exerts its 2009 (2009), Article ID 498352.
anti-inflammatory effects by other mechanisms such as upregu- [6] C.P.J. Caygill, M.J. Hill, Fish, N-3 fatty-acids and human colorectal and breast-
cancer mortality, European Journal of Cancer Prevention 4 (1995) 329–332.
lated expression of PPAR␥ [65,66]. Zingerone, the active ingredient
[7] C.D. Allred, D.R. Talbert, R.C. Southard, X. Wang, M.W. Kilgore, PPAR gamma 1
in ginger, which is structurally similar to curcumin, is also anti- as a molecular target of eicosapentaenoic acid in human colon cancer (HT-29)
inflammatory through its ability to increase PPAR␥ expression cells, Journal of Nutrition 138 (2008) 250–256.
[67]. [8] C.K. Glass, M.G. Rosenfeld, The coregulator exchange in transcriptional func-
tions of nuclear receptors, Genes & Development 14 (2000) 121–141.
Apoptosis delivers anti-inflammatory signals to macrophages [9] C.K. Glass, S. Ogawa, Combinatorial roles of nuclear receptors in inflammation
while necrosis is pro-inflammatory [68]. The sumoylation of PPAR␥ and immunity, Nature Reviews Immunology 6 (2006) 44–55.
in mouse macrophages exposed, in vitro, to apoptotic cells results [10] T.T. Wang, J. Xu, X.F. Yu, R.C. Yang, Z.C. Han, Peroxisome proliferator-activated
receptor gamma in malignant diseases, Critical Reviews in Oncology Hematol-
in a phenotype shift in the macrophages from pro-inflammatory to ogy 58 (2006) 1–14.
anti-inflammatory [69]. Diets which contain pro-apoptotic com- [11] L. Michalik, J. Auwerx, J.P. Berger, V.K. Chatterjee, C.K. Glass, F.J. Gonzalez,
pounds may not only protect against cancer, but also reduce P.A. Grimaldi, T. Kadowaki, M.A. Lazar, S. O’Rahilly, C.N.A. Palmer, J. Plutzky,
J.K. Reddy, B.M. Spiegelman, B. Staels, W. Wahli, International Union of Phar-
inflammation [70–72]. The dietary PUFA and PPAR␥ ligand EPA has macology. LXI. Peroxisome proliferator-activated receptors, Pharmacological
a PPAR␥ mediated apoptotic effect in HT-29 colon cancer cells [7] in Reviews 58 (2006) 726–741.
vitro and CLA has been reported to induce apoptosis in the human [12] C. Bruedigam, M. Koedam, H. Chiba, M. Eijken, J. van Leeuwen, Evidence for mul-
tiple peroxisome proliferator-activated receptor gamma transcripts in bone:
breast cancer cell line SKBr3 [73]. A synergistic effect may therefore
fine-tuning by hormonal regulation and mRNA stability, FEBS Letters 582
occur when cells are exposed to compounds such as these, which (2008) 1618–1624.
are both pro-apoptotic and also anti-inflammatory. [13] P. Escher, O. Braissant, S. Basu-Modak, L. Michalik, W. Wahli, B. Desvergne, Rat
PPARs: quantitative analysis in adult rat tissues and regulation in fasting and
refeeding, Endocrinology 142 (2001) 4195–4202.
[14] C. Diradourian, J. Girard, J.P. Pegorier, Phosphorylation of PPARs: from molec-
4. Conclusion ular characterization to physiological relevance, Biochimie 87 (2005) 33–38.
[15] G. Pascual, A.L. Fong, S. Ogawa, A. Gamliel, A.C. Li, V. Perissi, D.W. Rose, T.M.
Willson, M.G. Rosenfeld, C.K. Glass, A SUMOylation-dependent pathway medi-
The tissue-specific knockout of the PPAR␥ gene has revealed the ates transrepression of inflammatory response genes by PPAR-gamma, Nature
pivotal role of PPAR␥ in regulating inflammation and metabolism 437 (2005) 759–763.
62 H. Martin / Mutation Research 690 (2010) 57–63

[16] J.M. Peters, H.E. Hollingshead, F.J. Gonzalez, Role of peroxisome-proliferator- [41] A. Coste, C. Lagane, C. Filipe, H. Authier, A. Gales, J. Bernad, V. Douin-Echinard,
activated receptor beta/delta (PPAR beta/delta) in gastrointestinal tract J.C. Lepert, P. Balard, M.D. Linas, J.F. Arnal, J. Auwerx, B. Pipy, IL-13 attenuates
function and disease, Clinical Science 115 (2008) 107–127. gastrointestinal candidiasis in normal and immunodeficient RAG-2(−/−) mice
[17] L. Klotz, S. Schmidt, R. Heun, T. Klockgether, H. Kolsch, Association of the PPAR via peroxisome proliferator-activated receptor-gamma activation, Journal of
gamma gene polymorphism Pro12Ala with delayed onset of multiple sclerosis, Immunology 180 (2008) 4939–4947.
Neuroscience Letters 449 (2009) 81–83. [42] J.H. Yu, K.H. Kim, H. Kim, SOCS 3 and PPAR-gamma ligands inhibit the expres-
[18] J.J. Regieli, J.W. Jukema, P.A. Doevendans, A.H. Zwinderman, Y. van der Graaf, J.J. sion of IL-6 and TGF-beta 1 by regulating JAK2/STAT3 signaling in pancreas,
Kastelein, D.E. Grobbee, PPAR gamma variant influences angiographic outcome International Journal of Biochemistry & Cell Biology 40 (2008) 677–688.
and 10-year cardiovascular risk in male symptomatic coronary artery disease [43] C. Rousseaux, B. Lefebvre, L. Dubuquoy, P. Lefebvre, O. Romano, J. Auwerx, D.
patients, Diabetes Care 32 (2009) 839–844. Metzger, W. Wahli, B. Desvergne, G.C. Naccari, P. Chavatte, A. Farce, P. Bulois,
[19] J. Bassaganya-Riera, K. Reynolds, S. Martino-Catt, Y.Z. Cui, L. Hennighausen, F. A. Cortot, J.F. Colombel, P. Desreumaux, Intestinal antiinflammatory effect
Gonzalez, J. Rohrer, A.U. Benninghoff, R. Hontecillas, Activation of PPAR gamma of 5-aminosalicylic acid is dependent on peroxisome proliferator-activated
and delta by conjugated linoleic acid mediates protection from experimental receptor-gamma, Journal of Experimental Medicine 201 (2005) 1205–
inflammatory bowel disease, Gastroenterology 127 (2004) 777–791. 1215.
[20] J.I. Odegaard, R.R. Ricardo-Gonzalez, M.H. Goforth, C.R. Morel, V. Subrama- [44] R. Marion-Letellier, M. Butler, P. Dechelotte, R.J. Playford, S. Ghosh, Comparison
nian, L. Mukundan, A.R. Eagle, D. Vats, F. Brombacher, A.W. Ferrante, A. of cytokine modulation by natural peroxisome proliferator-activated receptor
Chawla, Macrophage-specific PPAR gamma controls alternative activation and gamma ligands with synthetic ligands in intestinal-like Caco-2 cells and human
improves insulin resistance, Nature 447 (2007), pp. 1116-U1112. dendritic cells-potential for dietary modulation of peroxisome proliferator-
[21] Y.H. Wan, A. Saghatelian, L.W. Chong, C.L. Zhang, B.F. Cravatt, R.M. Evans, Mater- activated receptor gamma in intestinal inflammation, American Journal of
nal PPAR gamma protects nursing neonates by suppressing the production of Clinical Nutrition 87 (2008) 939–948.
inflammatory milk, Genes & Development 21 (2007) 1895–1908. [45] F.R. Huang, Z.P. Zhan, J. Luo, S.W. Jiang, J. Peng, Duration of feeding linseed
[22] V.R. Narala, R. Ranga, M.R. Smith, A.A. Berlin, T.J. Standiford, N.W. Lukacs, R.C. diet influences peroxisome proliferator-activated receptor gamma and tumor
Reddy, Pioglitazone is as effective as dexamethasone in a cockroach allergen- necrosis factor gene expression, and muscle mass of growing-finishing bar-
induced murine model of asthma, Respiratory Research 8 (2007). rows, Livestock Science 119 (2008) 194–201.
[23] M. Koufany, D. Moulin, A. Bianchi, M. Muresan, S. Sebillaud, P. Netter, G. [46] T. Itoh, L. Fairall, K. Amin, Y. Inaba, A. Szanto, B.L. Balint, L. Nagy, K. Yamamoto,
Weryha, J.Y. Jouzeau, Anti-inflammatory effect of antidiabetic thiazolidine- J.W.R. Schwabe, Structural basis for the activation of PPAR gamma by oxidized
diones prevents bone resorption rather than cartilage changes in experimental fatty acids, Nature Structural & Molecular Biology 15 (2008) 924–931.
polyarthritis, Arthritis Research & Therapy 10 (2008). [47] T. Waku, T. Shiraki, T. Oyama, Y. Fujimoto, K. Maebara, N. Kamiya, H. Jingami, K.
[24] G.J. Ko, Y.S. Kang, S.Y. Han, M.H. Lee, H.K. Song, K.H. Han, H.K. Kim, J.Y. Morikawa, Structural insight into PPAR[gamma] activation through covalent
Han, D.R. Cha, Pioglitazone attenuates diabetic nephropathy through an modification with endogenous fatty acids, Journal of Molecular Biology 385
anti-inflammatory mechanism in type 2 diabetic rats, Nephrology Dialysis (2009) 188–199.
Transplantation 23 (2008) 2750–2760. [48] T.K. Vyas, A. Shahiwala, M.M. Amiji, Improved oral bioavailability and brain
[25] J.J. Brightt, C.C. Walline, S. Kanakasabai, S. Chakraborty, Targeting PPAR as a transport of Saquinavir upon administration in novel nanoemulsion formula-
therapy to treat multiple sclerosis, Expert Opinion on Therapeutic Targets 12 tions, International Journal of Pharmaceutics 347 (2008) 93–101.
(2008) 1565–1575. [49] J.B. Ewaschuk, J.W. Walker, H. Diaz, K.L. Madsen, Bioproduction of conjugated
[26] J.D. Lewis, G.R. Lichtenstein, J.J. Deren, B.E. Sands, S.B. Hanauer, J.A. Katz, B. Lash- linoleic acid by probiotic bacteria occurs in vitro and in vivo in mice, Journal of
ner, D.H. Present, S. Chuai, J.H. Ellenberg, L. Nessel, G.D. Wu, Rosiglitazone for Nutrition 136 (2006) 1483–1487.
active ulcerative colitis: a randomized placebo-controlled trial, Gastroenterol- [50] S. Voltan, D. Martines, M. Elli, P. Brun, S. Longo, A. Porzionato, V. Macchi, R.
ogy 134 (2008) 688–695. D’Inca, M. Scarpa, G. Palu, G.C. Sturniolo, L. Morelli, I. Castagliuolo, Lactobacillus
[27] X.R. Zhou, C.H. Sun, J.R. Liu, D. Zhao, Dietary conjugated linoleic acid increases crispatus M247-derived H2 O2 acts as a signal transducing molecule activating
PPAR gamma gene expression in adipose tissue of obese rat, and improves peroxisome proliferator activated receptor-gamma in the intestinal mucosa,
insulin resistance, Growth Hormone & Igf Research 18 (2008) 361–368. Gastroenterology 135 (2008) 1216–1227.
[28] P.C. LaRosa, J.J.M. Riethoven, H. Chen, Y. Xia, Y. Zhou, M. Chen, J. Miner, M.E. [51] A. Are, L. Aronsson, S.G. Wang, G. Greicius, Y.K. Lee, J.A. Gustafsson, S. Pet-
Fromm, Trans-10, cis-12 conjugated linoleic acid activates the integrated stress tersson, V. Arulampalam, Enterococcus faecalis from newborn babies regulate
response pathway in adipocytes, Physiological Genomics 31 (2007) 544–553. endogenous PPAR gamma activity and IL-10 levels in colonic epithelial cells,
[29] H. Poirier, J.S. Shapiro, R.J. Kim, M.A. Lazar, Nutiritional supplementation with in: Proceedings of the National Academy of Sciences of the United States of
trans-10, cis-12-conjugated linoleic acid induces inflammation of white adi- America 105, 2008, pp. 1943–1948.
pose tissue, Diabetes 55 (2006) 1634–1641. [52] L.D. Whigham, A.C. Watras, D.A. Schoeller, Efficacy of conjugated linoleic acid
[30] A. Jaudszus, M. Krokowski, P. Mockel, Y. Darcan, A. Avagyan, P. Matricardi, G. for reducing fat mass: a meta-analysis in humans, American Journal of Clinical
Jahreis, E. Hamelmann, Cis-9, trans-11-conjugated linoleic acid inhibits allergic Nutrition 85 (2007) 1203–1211.
sensitization and airway inflammation via a PPAR gamma-related mechanism [53] E. Devillard, F.M. McIntosh, S.H. Duncan, R.J. Wallace, Metabolism of linoleic
in mice, Journal of Nutrition 138 (2008) 1336–1342. acid by human gut bacteria: different routes for biosynthesis of conjugated
[31] M. Ricote, A.C. Li, T.M. Willson, C.J. Kelly, C.K. Glass, The peroxisome proliferator- linoleic acid, Journal of Bacteriology 189 (2007) 2566–2570.
activated receptor-gamma is a negative regulator of macrophage activation, [54] A. Kennedy, S. Chung, K. LaPoint, O. Fabiyi, M.K. McIntosh, Trans-10, cis-12
Nature 391 (1998) 79–82. conjugated linoleic acid antagonizes ligand-dependent PPAR gamma activ-
[32] C.Y. Jiang, A.T. Ting, B. Seed, PPAR-gamma agonists inhibit production of mono- ity in primary cultures of human adipocytes, Journal of Nutrition 138 (2008)
cyte inflammatory cytokines, Nature 391 (1998) 82–86. 455–461.
[33] P. Gosset, A.S. Charbonnier, P. Delerive, J. Fontaine, B. Staels, J. Pestel, A.B. Tonnel, [55] A. Kennedy, A. Overman, K. LaPoint, R. Hopkins, T. West, C.C. Chuang, K. Mar-
F. Trottein, Peroxisome proliferator-activated receptor gamma activators affect tinez, D. Bell, M. McIntosh, Conjugated linoleic acid-mediated inflammation
the maturation of human monocyte-derived dendritic cells, European Journal and insulin resistance in human adipocytes are attenuated by resveratrol, Jour-
of Immunology 31 (2001) 2857–2865. nal of Lipid Research 50 (2009) 225–232.
[34] C. Faveeuw, S. Fougeray, V. Angeli, J. Fontaine, G. Chinetti, P. Gosset, P. [56] T. Tholstrup, M. Raff, E.M. Straarup, P. Lund, S. Basu, J.M. Bruun, An oil mixture
Delerive, C. Maliszewski, M. Capron, B. Staels, M. Moser, F. Trottein, Peroxi- with trans-10, cis-12 conjugated linoleic acid increases markers of inflamma-
some proliferator-activated receptor gamma activators inhibit interleukin-12 tion and in vivo lipid peroxidation compared with cis-9, trans-11 conjugated
production in murine dendritic cells, FEBS Letters 486 (2000) 261–266. linoleic acid in postmenopausal women, Journal of Nutrition 138 (2008)
[35] J.S. Welch, M. Ricote, T.E. Akiyama, F.J. Gonzalez, C.K. Glass, PPAR gamma and 1445–1451.
PPAR delta negatively regulate specific subsets of lipopolysaccharide and IFN- [57] C. Ramirez-Santana, C. Castellote, M. Castell, M. Rivero, M. Rodriguez-Palmero,
gamma target genes in macrophages, in: Proceedings of the National Academy A. Franch, F.J. Perez-Cano, Long-term feeding of the cis-9,trans-11 isomer of
of Sciences of the United States of America 100, 2003, pp. 6712–6717. conjugated linoleic acid reinforces the specific immune response in rats, Journal
[36] J.D. Lewis, G.R. Lichtenstein, J.J. Deren, B.E. Sands, S.B. Hanauer, J.A. Katz, B. Lash- of Nutrition 139 (2009) 76–81.
ner, D.H. Present, S. Chuai, J.H. Ellenbergr, L. Nessel, G.D. Wu, Rosiglitazone for [58] F. Zapata-Gonzalez, F. Rueda, J. Petriz, P. Domingo, F. Villarroya, J. Diaz-Delfin,
active ulcerative colitis: a randomized placebo-controlled trial, Gastroenterol- M.A. de Madariaga, J.C. Domingo, Human dendritic cell activities are modulated
ogy 134 (2008) 688–695. by the omega-3 fatty acid, docosahexaenoic acid, mainly through PPAR gamma:
[37] H.L. Liang, Q. Ouyang, A clinical trial of combined use of rosiglitazone and 5- RXR heterodimers: comparison with other polyunsaturated fatty acids, Journal
aminosalicylate for ulcerative colitis, World Journal of Gastroenterology 14 of Leukocyte Biology 84 (2008) 1172–1182.
(2008) 114–119. [59] S.Y. Moya-Camarena, J.P. Vanden Heuvel, S.G. Blanchard, L.A. Leesnitzer, M.A.
[38] T. Aprahamian, R.G. Bonegio, C. Richez, K. Yasuda, L.K. Chiang, K. Sato, K. Walsh, Belury, Conjugated linoleic acid is a potent naturally occurring ligand and acti-
I.R. Rifkin, The peroxisome proliferator-activated receptor gamma agonist vator of PPAR alpha, Journal of Lipid Research 40 (1999) 1426–1433.
rosiglitazone ameliorates murine lupus by induction of adiponectin, Journal [60] S.Y. Moya-Camarena, J.P. Vanden Heuvel, M.A. Belury, Conjugated linoleic acid
of Immunology 182 (2009) 340–346. activates peroxisome proliferator-activated receptor a and B subtypes but
[39] T. Kawai, T. Masaki, S. Doi, T. Arakawa, Y. Yokoyama, T. Doi, N. Kohno, N. Yorioka, does not induce hepatic peroxisome proliferation in Sprague–Dawley rats,
PPAR-gamma agonist attenuates renal interstitial fibrosis and inflammation Biochimica Et Biophysica Acta: Molecular and Cell Biology of Lipids 1436 (1999)
through reduction of TGF-beta, Laboratory Investigation 89 (2009) 47–58. 331–342.
[40] A. Hyong, V. Jadhav, S. Lee, W. Tong, J. Rowe, J.H. Zhang, J.P. Tang, Rosiglitazone, [61] G. Orasanu, O. Ziouzenkova, P.R. Devchand, V. Nehra, O. Hamdy, E.S. Hor-
a PPAR gamma agonist, attenuates inflammation after surgical brain injury in ton, J. Plutzky, The peroxisome proliferator-activated receptor-gamma agonist
rodents, Brain Research 1215 (2008) 218–224. pioglitazone represses inflammation in a peroxisome proliferator-activated
H. Martin / Mutation Research 690 (2010) 57–63 63

receptor-alpha-dependent manner in vitro and in vivo in mice, Journal of the receptor activation by a short-term feeding of zingerone in aged rats, Journal
American College of Cardiology 52 (2008) 869–881. of Medicinal Food 12 (2009) 345–350.
[62] M. Plourde, S. Jew, S.C. Cunnane, P.J.H. Jones, Conjugated linoleic acids: why the [68] R.E. Cocco, D.S. Ucker, Distinct modes of macrophage recognition for apoptotic
discrepancy between animal and human studies? Nutrition Reviews 66 (2008) and necrotic cells are not specified exclusively by phosphatidylserine exposure,
415–421. Molecular Biology of the Cell 12 (2001) 919–930.
[63] M. Mueller, B. Lukas, J. Novak, T. Simoncini, A.R. Genazzani, A. Jung- [69] C. Jennewein, A.M. Kuhn, M.V. Schmidt, V. Meilladec-Jullig, A. von Knethen, F.J.
bauert, Oregano: a source for peroxisome proliferator-activated receptor Gonzalez, B. Brune, Sumoylation of peroxisome proliferator-activated receptor
gamma antagonists, Journal of Agricultural and Food Chemistry 56 (2008) gamma by apoptotic cells prevents lipopolysaccharide-induced NCoR removal
11621–11630. from kappa B binding sites proinflammatory cytokines, Journal of Immunology
[64] N. Adapala, M.M. Chan, Long-term use of an antiinflammatory, curcumin, sup- 181 (2008) 5646–5652.
pressed type 1 immunity and exacerbated visceral leishmaniasis in a chronic [70] J. Ju, X.P. Hao, M.J. Lee, J.D. Lambert, G. Lu, H. Xiao, H.L. Newmark, C.S. Yang,
experimental model, Laboratory Investigation 88 (2008) 1329–1339. A gamma-tocopherol-rich mixture of tocopherols inhibits colon inflammation
[65] A.M. Siddiqui, X.X. Cui, R.Q. Wu, W.F. Dong, M. Zhou, M.W. Hu, H.H. and carcinogenesis in azoxymethane and dextran sulfate sodium-treated mice,
Simms, P. Wang, The anti-inflammatory effect of curcumin in an exper- Cancer Prevention Research 2 (2009) 143–152.
imental model of sepsis is mediated by up-regulation of peroxisome [71] E.H. Jeffery, M. Araya, Physiological effects of broccoli consumption, Phyto-
proliferator-activated receptor-gamma, Critical Care Medicine 34 (2006) 1874– chemistry Reviews 8 (2009) 283–298.
1882. [72] T. Tanaka, Y. Yasui, R. Ishigamori-Suzuki, T. Oyama, Citrus compounds inhibit
[66] V.R. Narala, M.R. Smith, R.K. Adapala, R. Ranga, K. Panati, B.B. Moore, T. Leff, inflammation- and obesity-related colon carcinogenesis in mice, Nutrition and
V.D. Reddy, A.K. Kondapi, R.C. Reddy, Curcumin is not a ligand for peroxisome Cancer: An International Journal 60 (2008) 70–80.
proliferator-activated receptor-␥, Gene Therapy & Molecular Biology 13 (2009) [73] X.L. Yuan, F. He, Q. Chen, X.L. Yang, D.P. Yang, D.M. Wang, L. Zhong, Studies
20–25. on PPAR gamma signal pathway of conjugated linoleic acid isomers induce
[67] S.W. Chung, M.K. Kim, J.H. Chung, D.H. Kim, J.S. Choi, S. Anton, A.Y. Seo, K.Y. Park, apoptosis of human breast cancer cell line SKBr3, Progress in Biochemistry and
T. Yokozawa, S.H. Rhee, B.P. Yu, H.Y. Chung, Peroxisome proliferator-activated Biophysics 36 (2009) 491–499.

You might also like