Peroxisome proliferator-activated receptor delta (PPAR-δ) is a nuclear receptor transcription factor that regulates gene expression during development and disease states, such as cancer. However, the precise role of PPAR-δ during tumorigenesis is not well understood. Recent data suggest that PPAR-δ may have context-specific oncogenic and tumor-suppressive roles depending on the tissue, cell-type, or diet-induced physiology in question. For example, in the intestine, pro-obesity diets, such as a high-fat diet (HFD), are associated with increased colorectal cancer incidence. Interestingly, many of the effects of an HFD in the stem and progenitor cell compartment are driven by a robust PPAR-δ program and contribute to the early steps of intestinal tumorigenesis. Importantly, the PPAR-δ pathway or its downstream mediators may serve as therapeutic intervention points or biomarkers in colon cancer that arise in patients who are obese. Although potent PPAR-δ agonists and antagonists exist, their clinical utility may be enhanced by uncovering how PPAR-δ mediates tumorigenesis in diverse tissues and cell types as well as in response to diet. Clin Cancer Res; 22(23); 5636–41. ©2016 AACR.

Peroxisome proliferator-activated receptor delta (PPAR-δ) belongs to the peroxisome proliferator-activated receptor (PPAR) subfamily of nuclear receptor transcription factors that is composed of three members (alpha, beta/delta, and gamma; ref. 1). PPARs are critical regulators of metabolism and exhibit tissue and cell type–specific expression patterns and functions that can be altered by physiologic cues. Although PPAR isoforms have highly homologous structures and overlapping functions, distinct selective ligands have been identified to activate each family member. PPAR-α is the first member of the family that was cloned and characterized in mouse liver, where it regulates lipid homeostasis and reduces triacylglycerol levels. PPAR-γ has been extensively studied in adipose tissue, where it controls adipogenesis and lipid metabolism and enhances glucose metabolism (1). PPAR-δ is highly expressed in the intestinal crypts, keratinocytes, and oxidative skeletal muscle fibers (2) and is a master regulator of lipid metabolism (3). For example, PPAR-δ–mediated metabolic regulation protects against diet-induced obesity and metabolic syndrome, and it enhances skeletal muscle endurance and function (4, 5). Furthermore, dietary interventions, such as fasting (3), and inflammatory cytokines, such as TNFα and IFNγ (6), augment PPAR-δ signaling. Thus, PPAR-δ couples diverse dietary, nutritional, or physiologic inputs with organismal metabolism (7–9).

Although long-chain fatty acids and their derivatives were initially recognized as endogenous activators of the PPAR-δ pathway, the actual nature of the physiologic ligands of PPAR-δ remains elusive. In the past decade, in addition to fatty acids, derivatives of arachidonic acid (10), lipoperoxidation products (11), and all-trans retinoic acid (12) have been proposed to activate the PPAR-δ pathway. Although a general consensus is lacking, two major models have been postulated regarding how nuclear PPAR-δ senses its ligand(s). In the first model, it is proposed that the ligands are delivered from the cytoplasm to the nucleus by fatty acid–binding proteins (FABP; refs. 13, 14), and in the second model, it is suggested that the ligands are products of active nuclear lipid metabolism (15). Importantly, several very potent and selective small-molecule PPAR-δ agonists, including GW501516, GW0742 (16), and MBX-8025 (17), have been developed and widely used experimentally to decipher the biological significance of PPAR-δ–mediated transcriptional activity (Fig. 1).

Figure 1.

Mechanisms of PPAR-δ–mediated gene regulation. Dietary fatty acids, synthetic small-molecule agonists (GW501516), intracellular fatty acids, and metabolites activate the PPAR-δ pathway. Two major models have been proposed regarding delivery of PPAR-δ ligands. First, fatty acid–binding proteins (FABP) can transport fatty acids to the nucleus. Second, ligands are produced in the nucleus as a result of nuclear lipid metabolism. Ligand binding activates PPAR-δ and leads to the regulation of several important biological processes. First, PPAR-δ partners with retinoid X receptor (RXR) and binds to peroxisome proliferator response elements (PPRE) to activate the expression of genes involved in lipid metabolism (1). In addition, PPAR-δ interacts with β-catenin, which is a key transcription factor for cell fate determination and assists β-catenin to induce gene expression to modulate stemness in response to dietary cues, such as fatty acids (2). Finally, PPAR-δ inhibits inflammatory gene expression by interfering with the NF-κB pathway (3).

Figure 1.

Mechanisms of PPAR-δ–mediated gene regulation. Dietary fatty acids, synthetic small-molecule agonists (GW501516), intracellular fatty acids, and metabolites activate the PPAR-δ pathway. Two major models have been proposed regarding delivery of PPAR-δ ligands. First, fatty acid–binding proteins (FABP) can transport fatty acids to the nucleus. Second, ligands are produced in the nucleus as a result of nuclear lipid metabolism. Ligand binding activates PPAR-δ and leads to the regulation of several important biological processes. First, PPAR-δ partners with retinoid X receptor (RXR) and binds to peroxisome proliferator response elements (PPRE) to activate the expression of genes involved in lipid metabolism (1). In addition, PPAR-δ interacts with β-catenin, which is a key transcription factor for cell fate determination and assists β-catenin to induce gene expression to modulate stemness in response to dietary cues, such as fatty acids (2). Finally, PPAR-δ inhibits inflammatory gene expression by interfering with the NF-κB pathway (3).

Close modal

PPAR-δ controls transcription both directly and indirectly (1). Direct regulation involves partnering with retinoid X receptor and binding to peroxisome proliferator response elements in the enhancers and promoters of its target genes (Fig. 1). Furthermore, PPAR-δ–mediated transcriptional activation relies on ligand binding and concomitant epigenetic mechanisms that include displacement of corepressors and recruitment of coactivator-associated chromatin remodeling factors, which orchestrate the framework necessary for transcriptional initiation (18). Over the past decade, PPAR-δ or its direct transcriptional targets have been demonstrated to bear on diverse cellular functions, such as stem cell maintenance, cellular differentiation, metabolism, and inflammation.

PPAR-δ also regulates many genes indirectly by affecting the activity of other transcription factors through physical interaction or through signal transduction pathways that affect the function of other transcription factors (Fig. 1). For example, PPAR-δ is found to interfere with the NF-κB pathway (an important pathway in controlling inflammatory gene expression) and attenuate inflammation (19). Moreover, PPAR-δ cooperates with β-catenin (a key transcription factor for cell fate determination) in bone turnover and intestinal tumorigenesis (7, 20).

Gene expression analysis studies in combination with chromatin immunoprecipitation sequencing have identified direct and indirect PPAR-δ–regulated genes (21, 22). Well-documented direct targets of PPAR-δ include genes involved in fatty acid oxidation and lipid metabolism, such as Cpt1a, Fabp1, and Pdk4, whereas proinflammatory cytokines, such as TNFα and IL6, are regulated indirectly. Overall, PPAR-δ controls the expression of a large set of genes through different mechanisms that are influenced by PPAR-δ expression levels, interaction with additional transcription factors, as well as the abundance and nature of its ligands. These different levels of control confer versatility to PPAR-δ–mediated gene regulation in myriad tissues and cell types during development and disease states such as cancer.

PPAR-δ in development and cancer

Several developmental abnormalities have been reported in PPAR-δ–null mice, including reduced gestational and postnatal body weight as well as abnormalities in multiple tissues such as adipose tissue, skin, and intestine (23, 24). In adipose tissue, PPAR-δ deficiency leads to a reduction in adipocyte numbers and PPAR-γ expression levels (as noted, PPAR-γ is the PPAR family member that controls adipogenesis; ref. 23). Although PPAR-δ is not directly implicated in the regulation of adipogenesis, it contributes to the metabolic response of adipose tissue to dietary fatty acids (8, 23). Although PPAR-δ is expendable for homeostatic function in the skin, it plays a critical role in skin repair after injuries that disrupt skin integrity. Also, it is understood that agonist-activated PPAR-δ hampers cellular proliferation and induces keratinocyte differentiation (24, 25). Thus, these findings are consistent with the notion that PPAR-δ signaling may possess antitumorigenic roles in the skin by preventing the growth and promoting the differentiation of (pre-) malignant cells through antagonizing the expression of proteins known to contribute to keratinocyte hyperproliferation and skin tumorigenesis (24–27). However, these antitumorigenic effects of PPAR-δ signaling in the skin may be context specific, as they are observed in only some models of skin cancer (28, 29).

In the intestine, PPAR-δ performs critical regulatory functions and modulates intestinal physiology in multiple cell types, yet the underlying mechanisms are poorly understood. For instance, PPAR-δ was identified as a crucial factor in Paneth cell differentiation, thus implicating it as a significant player in intestinal homeostasis (30). Paneth cells not only possess antimicrobial functions but also constitute a niche component for intestinal stem cells (ISC) that reside at the bottom of the crypt, a location where PPAR-δ expression is the highest in the intestine (30, 31). By influencing Paneth cell differentiation, PPAR-δ regulates a significant support cell of the ISCs, which drives the rapid renewal of the intestinal epithelium. In addition, PPAR-δ contributes to intestinal physiology through the control of lipid absorption, cholesterol trafficking, and enteroendocrine cell function in more differentiated or specialized intestinal cell types (32–34).

Initial reports on the role of PPAR-δ in intestinal tumorigenesis suggested that PPAR-δ is a direct transcriptional target of the Wnt/β-catenin pathway, a pathway that governs intestinal maintenance and tumorigenesis. Adenomatous polyposis coli (APC) orchestrates the destruction of cytoplasmic β-catenin in the absence of an upstream Wnt activation signal to tightly control β-catenin nuclear localization and activity. APC inactivation leads to strong activation of the β-catenin pathway and is the most common initiating oncogenic event of intestinal dysplasia (35). Although early studies demonstrated induction of PPAR-δ in intestinal tumors that were formed upon loss of APC or by chemical mutagenesis, subsequent reports in both rodents and humans failed to establish an unequivocal induction of PPAR-δ expression or activity in intestinal cancers (36). Several studies have used either whole-body or intestinal-specific genetic ablation of PPAR-δ to delineate the precise function of PPAR-δ in tumor-prone Apcmin/+ mice (mice with nonsense mutation in Apc gene that results in a truncated protein) or chemically induced models of intestinal tumorigenesis. Whereas some studies found a protumorigenic role for PPAR-δ, others posited an antitumorigenic function. Conflicting results from these loss-of-function studies have raised important questions regarding the role of PPAR-δ in intestinal tumorigenesis (23, 37–39). In contrast, however, gain-of-function studies that used the PPAR-δ agonist GW501516 have more consistently demonstrated that PPAR-δ activation mediates a protumorigenic phenotype in the intestine (40).

These conflicting findings regarding PPAR-δ necessity in intestinal tumorigenesis may be due to different genetic strategies used to disrupt PPAR-δ function. Studies that supported an antitumorigenic role for PPAR-δ in Apcmin/+ mice use germline disruption of PPAR-δ exon 8 (37, 38). Whereas ablation of PPAR-δ by disrupting exon 4 failed to decrease tumor incidence in Apcmin/+ mice but led to a reduction in size of the largest intestinal adenomas (23), deletion of PPAR-δ exon 4 and part of exon 5 significantly diminished tumor numbers in Apcmin/+ mice, and exon 4 deletion reduced dysplastic lesions in Apcmin/+ mice exposed to chemical injury (39, 41). Some of the discrepancy likely arises from the fact that disruption of exon 8 (the last PPAR-δ exon) may produce a hypomorphic protein, while disruption of exon 4 (an essential portion of the PPAR-δ DNA-binding domain) ablates PPAR-δ activity (42). Finally, it is important to point out that these studies used germline PPAR-δ mutants. More recently, intestine-specific PPAR-δ loss (targeting exon 4) decreased the numbers of dysplastic lesions in response to chemical mutagenesis (42). Another possible confounder in these studies is the use of different genetic backgrounds, which is known to influence tumor susceptibility in Apcmin/+ mice and in chemical mutagenesis models (1).

Dietary regulation of stemness and tumor initiation

As discussed above, accumulating evidence underscores the importance of the PPAR-δ pathway in health and disease. However, little is known about how PPAR-δ contributes to the regulation of stemness and tumor initiation in response to diet-induced physiologies. Diet is a modifiable lifestyle factor that has a profound impact on mammalian physiology, health, and disease, including cancer. Long-lived mammalian tissue–specific stem cells play a key role in how tissues adapt to diverse diet-induced physiologic states. Such stem cells dynamically remodel tissue composition in response to physiologic cues by altering the balance between self-renewal and differentiation divisions (43). Factors that increase somatic stem cell self-renewal division rate are implicated to elevate cancer risk (44, 45). This suggests that dietary interventions that boost stem cell self-renewal increase the potential pool of cells that can undergo mutagenesis and give rise to cancer.

Consistent with this notion, we recently demonstrated that a pro-obesity, high-fat diet (HFD)–activated PPAR-δ program augments the numbers, proliferation, and function of ISCs in the mouse intestine, providing a possible explanation to the long-sought link between diet-induced obesity and cancer (7, 46). In addition, intestinal adaptations to a long-term HFD not only enabled ISCs to acquire niche independence (i.e., initiate mini intestines in culture without need of their Paneth niche) but also allowed non–stem cell progenitors to obtain stem cell attributes. These changes effectively augmented the number of cells within the intestine that possessed stemness and that could initiate tumors upon loss of the APC tumor suppressor gene (7). Moreover, enforced PPAR-δ activation mimicked many of these effects of an HFD. As mentioned, there are differing opinions on the role of PPAR-δ in intestinal tumor initiation. However, these findings demonstrate that PPAR-δ activation in ISCs and progenitors has protumorigenic effects in the intestine and raises the possibility that PPAR-δ inhibition in the setting of an HFD may dampen tumor initiation, progression, or both.

An important question is whether an HFD has similar stem cell–enhancing effects in other tissues. Interestingly, in contrast to the enhanced stemness observed in the intestine, an HFD decreases neural progenitor cells proliferation and hippocampal neurogenesis by increasing lipid peroxidation and reducing brain-derived neutrophic factor (BDNF) levels in the hippocampus (47). On the other hand, an HFD alters the differentiation potential of bone marrow–derived mesenchymal stem cells, subcutaneous adipose-derived stem cells, and infrapatellar fat pad–derived stem cells. Notably, the fatty acid constituents of the diet, in part, mediated this effect, implicating a possible role for PPAR-δ signaling (48). Also, the impact of an HFD in hematopoiesis is context dependent. A recent study demonstrated that a maternal HFD compromises the expansion of the fetal hematopoietic stem/progenitor cell (HSPC) pool and hematopoietic lineage specification. Conversely, adult hematopoiesis is augmented in response to HFD-induced obesity (49, 50). The fact that PPAR-δ–activated fatty acid oxidation drives hematopoietic stem cell self-renewal (51) suggests that HFD-mediated regulation of hematopoiesis may involve PPAR-δ function. Nonetheless, whether HFD or PPAR-δ is linked to tumor initiation in these tissues warrants further investigation.

Despite the fact that preclinical studies have provided evidence that PPAR-δ agonists possess therapeutic value in the treatment of several metabolic diseases, no FDA-approved agonists for PPAR-δ currently exist (5, 9). This is in contrast to PPAR-α and PPAR-γ, which have FDA-approved agonists that are standard of care for metabolic disorders, such as hyperlipidemia and diabetes. Although clinical trials have revealed that short-term administration of the PPAR-δ agonists GW501516 (NCT00841217; ref. 52) and MBX-8025 (NCT00701883; ref. 53) had beneficial effects in patients with metabolic syndrome, preclinical long-term studies in mice have indicated that PPAR-δ agonists might have protumorigenic effects, thus dampening enthusiasm for these compounds in human trials.

Several studies have suggested that PPAR-δ expression has prognostic value for cancer patients, including those with colorectal cancer (54, 55). However, there are inconsistencies in the reported PPAR-δ expression levels of different human tumors compared with normal tissue as well as how PPAR-δ expression correlates with prognosis. For instance, a study that examined 52 patients with colorectal cancer found that PPAR-δ and COX-2 expression in tumors negatively correlated with patient survival (54). However, another study with 141 patients with colorectal cancer revealed that higher expression of PPAR-δ in primary colorectal tumors was associated with lower expression of Ki67 (a cellular marker for proliferation), a higher frequency of stage I cases, a lower frequency of late-stage cases, and a lower rate of lymph node metastasis (55). More studies are required to precisely ascertain the molecular mechanisms that PPAR-δ employs during cancer initiation and progression and how they affect patient prognosis.

PPAR-δ polymorphisms have been associated with physical performance (56), diabetes (57), obesity (58), and cancer (59, 60). A recent study focusing on patients with colorectal cancer identified seven novel variants among 22 inherited or acquired PPARD variants. Interestingly, four recurrent variants were detected in or adjacent to exon 4, which encodes an essential portion of the PPAR-δ DNA-binding domain, suggesting that these variants might have functional significance (60). Although these variants may have clinical importance, the effect of PPAR-δ polymorphisms on gene expression is still poorly understood.

Our recent data indicate that HFD-activated PPAR-δ engages a specific β-catenin program that involves expression of genes implicated in intestinal tumorigenesis, such as Jag1 and Bmp4, in ISCs and progenitors (7). Future studies need to address whether increased expression of Jag1 or Bmp4 has any prognostic or correlative value in colorectal cancer that arises in patients who are obese. Furthermore, it will be important to determine the functions of PPAR-δ in the setting of diverse tissues, cell types, diets, and risk factors that are associated with cancer in humans. The fact that PPAR-δ links dietary regulation of stemness to tumor initiation in the intestine raises the possibility of a new therapeutic approach in the treatment of intestinal cancers in obesity (7), such as exploiting possible PPAR-δ dependencies in tumors that arise in an HFD or obesity. Uncovering the cellular and molecular mechanisms by which PPAR-δ drives its effects may lead to new therapeutic insights, especially in those cancers with high PPAR-δ activity.

No potential conflicts of interest were disclosed.

Conception and design: S. Beyaz

Writing, review, and/or revision of the manuscript: S. Beyaz, Ö.H. Yilmaz

This work was supported by the NIHR00 AG045144, V Foundation V Scholar Award, the Sidney Kimmel Scholar Award, the Pew-Stewart Trust Scholar Award, Kathy and Curt Marble Cancer Research Fund, and American Federation of Aging Research (to Ö.H. Yilmaz).

1.
Neels
JG
,
Grimaldi
PA
. 
Physiological functions of peroxisome proliferator-activated receptor beta
.
Physiol Rev
2014
;
94
:
795
858
.
2.
Girroir
EE
,
Hollingshead
HE
,
He
P
,
Zhu
B
,
Perdew
GH
,
Peters
JM
. 
Quantitative expression patterns of peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) protein in mice
.
Biochem Biophys Res Commun
2008
;
371
:
456
61
.
3.
Holst
D
,
Luquet
S
,
Nogueira
V
,
Kristiansen
K
,
Leverve
X
,
Grimaldi
PA
. 
Nutritional regulation and role of peroxisome proliferator-activated receptor delta in fatty acid catabolism in skeletal muscle
.
Biochim Biophys Acta
2003
;
1633
:
43
50
.
4.
Wang
YX
,
Zhang
CL
,
Yu
RT
,
Cho
HK
,
Nelson
MC
,
Bayuga-Ocampo
CR
, et al
Regulation of muscle fiber type and running endurance by PPARdelta
.
PLoS Biol
2004
;
2
:
e294
.
5.
Wang
YX
,
Lee
CH
,
Tiep
S
,
Yu
RT
,
Ham
J
,
Kang
H
, et al
Peroxisome-proliferator-activated receptor delta activates fat metabolism to prevent obesity
.
Cell
2003
;
113
:
159
70
.
6.
Tan
NS
,
Michalik
L
,
Noy
N
,
Yasmin
R
,
Pacot
C
,
Heim
M
, et al
Critical roles of PPAR beta/delta in keratinocyte response to inflammation
.
Genes Dev
2001
;
15
:
3263
77
.
7.
Beyaz
S
,
Mana
MD
,
Roper
J
,
Kedrin
D
,
Saadatpour
A
,
Hong
SJ
, et al
High-fat diet enhances stemness and tumorigenicity of intestinal progenitors
.
Nature
2016
;
531
:
53
8
.
8.
Bastie
C
,
Luquet
S
,
Holst
D
,
Jehl-Pietri
C
,
Grimaldi
PA
. 
Alterations of peroxisome proliferator-activated receptor delta activity affect fatty acid-controlled adipose differentiation
.
J Biol Chem
2000
;
275
:
38768
73
.
9.
Luquet
S
,
Gaudel
C
,
Holst
D
,
Lopez-Soriano
J
,
Jehl-Pietri
C
,
Fredenrich
A
, et al
Roles of PPAR delta in lipid absorption and metabolism: a new target for the treatment of type 2 diabetes
.
Biochim Biophys Acta
2005
;
1740
:
313
7
.
10.
Naruhn
S
,
Meissner
W
,
Adhikary
T
,
Kaddatz
K
,
Klein
T
,
Watzer
B
, et al
15-hydroxyeicosatetraenoic acid is a preferential peroxisome proliferator-activated receptor beta/delta agonist
.
Mol Pharmacol
2010
;
77
:
171
84
.
11.
Riahi
Y
,
Sin-Malia
Y
,
Cohen
G
,
Alpert
E
,
Gruzman
A
,
Eckel
J
, et al
The natural protective mechanism against hyperglycemia in vascular endothelial cells: roles of the lipid peroxidation product 4-hydroxydodecadienal and peroxisome proliferator-activated receptor delta
.
Diabetes
2010
;
59
:
808
18
.
12.
Schug
TT
,
Berry
DC
,
Shaw
NS
,
Travis
SN
,
Noy
N
. 
Opposing effects of retinoic acid on cell growth result from alternate activation of two different nuclear receptors
.
Cell
2007
;
129
:
723
33
.
13.
Huang
H
,
Starodub
O
,
McIntosh
A
,
Kier
AB
,
Schroeder
F
. 
Liver fatty acid-binding protein targets fatty acids to the nucleus. Real time confocal and multiphoton fluorescence imaging in living cells
.
J Biol Chem
2002
;
277
:
29139
51
.
14.
Tan
NS
,
Shaw
NS
,
Vinckenbosch
N
,
Liu
P
,
Yasmin
R
,
Desvergne
B
, et al
Selective cooperation between fatty acid binding proteins and peroxisome proliferator-activated receptors in regulating transcription
.
Mol Cell Biol
2002
;
22
:
5114
27
.
15.
Martelli
AM
,
Fala
F
,
Faenza
I
,
Billi
AM
,
Cappellini
A
,
Manzoli
L
, et al
Metabolism and signaling activities of nuclear lipids
.
Cell Mol Life Sci
2004
;
61
:
1143
56
.
16.
Sznaidman
ML
,
Haffner
CD
,
Maloney
PR
,
Fivush
A
,
Chao
E
,
Goreham
D
, et al
Novel selective small molecule agonists for peroxisome proliferator-activated receptor delta (PPARdelta)–synthesis and biological activity
.
Bioorg Med Chem Lett
2003
;
13
:
1517
21
.
17.
Bays
HE
,
Schwartz
S
,
Littlejohn
T
 III
,
Kerzner
B
,
Krauss
RM
,
Karpf
DB
, et al
MBX-8025, a novel peroxisome proliferator receptor-delta agonist: lipid and other metabolic effects in dyslipidemic overweight patients treated with and without atorvastatin
.
J Clin Endocrinol Metab
2011
;
96
:
2889
97
.
18.
Viswakarma
N
,
Jia
Y
,
Bai
L
,
Vluggens
A
,
Borensztajn
J
,
Xu
J
, et al
Coactivators in PPAR-regulated gene expression
.
PPAR Res
2010
;
2010
:
250126
.
19.
Barroso
E
,
Eyre
E
,
Palomer
X
,
Vazquez-Carrera
M
. 
The peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) agonist GW501516 prevents TNF-alpha-induced NF-kappaB activation in human HaCaT cells by reducing p65 acetylation through AMPK and SIRT1
.
Biochem Pharmacol
2011
;
81
:
534
43
.
20.
Scholtysek
C
,
Katzenbeisser
J
,
Fu
H
,
Uderhardt
S
,
Ipseiz
N
,
Stoll
C
, et al
PPARbeta/delta governs Wnt signaling and bone turnover
.
Nat Med
2013
;
19
:
608
13
.
21.
Khozoie
C
,
Borland
MG
,
Zhu
B
,
Baek
S
,
John
S
,
Hager
GL
, et al
Analysis of the peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) cistrome reveals novel co-regulatory role of ATF4
.
BMC Genomics
2012
;
13
:
665
.
22.
Adhikary
T
,
Kaddatz
K
,
Finkernagel
F
,
Schonbauer
A
,
Meissner
W
,
Scharfe
M
, et al
Genomewide analyses define different modes of transcriptional regulation by peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta)
.
PLoS One
2011
;
6
:
e16344
.
23.
Barak
Y
,
Liao
D
,
He
W
,
Ong
ES
,
Nelson
MC
,
Olefsky
JM
, et al
Effects of peroxisome proliferator-activated receptor delta on placentation, adiposity, and colorectal cancer
.
Proc Natl Acad Sci U S A
2002
;
99
:
303
8
.
24.
Peters
JM
,
Lee
SS
,
Li
W
,
Ward
JM
,
Gavrilova
O
,
Everett
C
, et al
Growth, adipose, brain, and skin alterations resulting from targeted disruption of the mouse peroxisome proliferator-activated receptor beta(delta)
.
Mol Cell Biol
2000
;
20
:
5119
28
.
25.
Michalik
L
,
Wahli
W
. 
Peroxisome proliferator-activated receptors (PPARs) in skin health, repair and disease
.
Biochim Biophys Acta
2007
;
1771
:
991
8
.
26.
Bility
MT
,
Devlin-Durante
MK
,
Blazanin
N
,
Glick
AB
,
Ward
JM
,
Kang
BH
, et al
Ligand activation of peroxisome proliferator-activated receptor beta/delta (PPAR beta/delta) inhibits chemically induced skin tumorigenesis
.
Carcinogenesis
2008
;
29
:
2406
14
.
27.
Kim
DJ
,
Akiyama
TE
,
Harman
FS
,
Burns
AM
,
Shan
W
,
Ward
JM
, et al
Peroxisome proliferator-activated receptor beta (delta)-dependent regulation of ubiquitin C expression contributes to attenuation of skin carcinogenesis
.
J Biol Chem
2004
;
279
:
23719
27
.
28.
Zhu
B
,
Ferry
CH
,
Blazanin
N
,
Bility
MT
,
Khozoie
C
,
Kang
BH
, et al
PPARbeta/delta promotes HRAS-induced senescence and tumor suppression by potentiating p-ERK and repressing p-AKT signaling
.
Oncogene
2014
;
33
:
5348
59
.
29.
Montagner
A
,
Delgado
MB
,
Tallichet-Blanc
C
,
Chan
JS
,
Sng
MK
,
Mottaz
H
, et al
Src is activated by the nuclear receptor peroxisome proliferator-activated receptor beta/delta in ultraviolet radiation-induced skin cancer
.
EMBO Mol Med
2014
;
6
:
80
98
.
30.
Varnat
F
,
Heggeler
BB
,
Grisel
P
,
Boucard
N
,
Corthesy-Theulaz
I
,
Wahli
W
, et al
PPARbeta/delta regulates paneth cell differentiation via controlling the hedgehog signaling pathway
.
Gastroenterology
2006
;
131
:
538
53
.
31.
Sato
T
,
van Es
JH
,
Snippert
HJ
,
Stange
DE
,
Vries
RG
,
van den Born
M
, et al
Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts
.
Nature
2011
;
469
:
415
8
.
32.
Oliver
WR
 Jr
,
Shenk
JL
,
Snaith
MR
,
Russell
CS
,
Plunket
KD
,
Bodkin
NL
, et al
A selective peroxisome proliferator-activated receptor delta agonist promotes reverse cholesterol transport
.
Proc Natl Acad Sci U S A
2001
;
98
:
5306
11
.
33.
Daoudi
M
,
Hennuyer
N
,
Borland
MG
,
Touche
V
,
Duhem
C
,
Gross
B
, et al
PPARbeta/delta activation induces enteroendocrine L cell GLP-1 production
.
Gastroenterology
2011
;
140
:
1564
74
.
34.
Poirier
H
,
Niot
I
,
Monnot
MC
,
Braissant
O
,
Meunier-Durmort
C
,
Costet
P
, et al
Differential involvement of peroxisome-proliferator-activated receptors alpha and delta in fibrate and fatty-acid-mediated inductions of the gene encoding liver fatty-acid-binding protein in the liver and the small intestine
.
Biochem J
2001
;
355
:
481
8
.
35.
He
TC
,
Chan
TA
,
Vogelstein
B
,
Kinzler
KW
. 
PPARdelta is an APC-regulated target of nonsteroidal anti-inflammatory drugs
.
Cell
1999
;
99
:
335
45
.
36.
Peters
JM
,
Yao
PL
,
Gonzalez
FJ
. 
Targeting peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) for cancer chemoprevention
.
Curr Pharmacol Rep
2015
;
1
:
121
8
.
37.
Harman
FS
,
Nicol
CJ
,
Marin
HE
,
Ward
JM
,
Gonzalez
FJ
,
Peters
JM
. 
Peroxisome proliferator-activated receptor-delta attenuates colon carcinogenesis
.
Nat Med
2004
;
10
:
481
3
.
38.
Reed
KR
,
Sansom
OJ
,
Hayes
AJ
,
Gescher
AJ
,
Winton
DJ
,
Peters
JM
, et al
PPARdelta status and Apc-mediated tumourigenesis in the mouse intestine
.
Oncogene
2004
;
23
:
8992
6
.
39.
Wang
D
,
Fu
L
,
Ning
W
,
Guo
L
,
Sun
X
,
Dey
SK
, et al
Peroxisome proliferator-activated receptor delta promotes colonic inflammation and tumor growth
.
Proc Natl Acad Sci U S A
2014
;
111
:
7084
9
.
40.
Gupta
RA
,
Wang
D
,
Katkuri
S
,
Wang
H
,
Dey
SK
,
DuBois
RN
. 
Activation of nuclear hormone receptor peroxisome proliferator-activated receptor-delta accelerates intestinal adenoma growth
.
Nat Med
2004
;
10
:
245
7
.
41.
Wang
D
,
Wang
H
,
Guo
Y
,
Ning
W
,
Katkuri
S
,
Wahli
W
, et al
Crosstalk between peroxisome proliferator-activated receptor delta and VEGF stimulates cancer progression
.
Proc Natl Acad Sci U S A
2006
;
103
:
19069
74
.
42.
Zuo
X
,
Peng
Z
,
Moussalli
MJ
,
Morris
JS
,
Broaddus
RR
,
Fischer
SM
, et al
Targeted genetic disruption of peroxisome proliferator-activated receptor-delta and colonic tumorigenesis
.
J Natl Cancer Inst
2009
;
101
:
762
7
.
43.
Mihaylova
MM
,
Sabatini
DM
,
Yilmaz
OH
. 
Dietary and metabolic control of stem cell function in physiology and cancer
.
Cell Stem Cell
2014
;
14
:
292
305
.
44.
Tomasetti
C
,
Vogelstein
B
. 
Cancer etiology. Variation in cancer risk among tissues can be explained by the number of stem cell divisions
.
Science
2015
;
347
:
78
81
.
45.
Wu
S
,
Powers
S
,
Zhu
W
,
Hannun
YA
. 
Substantial contribution of extrinsic risk factors to cancer development
.
Nature
2016
;
529
:
43
7
.
46.
Finucane
MM
,
Stevens
GA
,
Cowan
MJ
,
Danaei
G
,
Lin
JK
,
Paciorek
CJ
, et al
National, regional, and global trends in body-mass index since 1980: systematic analysis of health examination surveys and epidemiological studies with 960 country-years and 9.1 million participants
.
Lancet
2011
;
377
:
557
67
.
47.
Park
HR
,
Park
M
,
Choi
J
,
Park
KY
,
Chung
HY
,
Lee
J
. 
A high-fat diet impairs neurogenesis: involvement of lipid peroxidation and brain-derived neurotrophic factor
.
Neurosci Lett
2010
;
482
:
235
9
.
48.
Wu
CL
,
Diekman
BO
,
Jain
D
,
Guilak
F
. 
Diet-induced obesity alters the differentiation potential of stem cells isolated from bone marrow, adipose tissue and infrapatellar fat pad: the effects of free fatty acids
.
Int J Obes
2013
;
37
:
1079
87
.
49.
Trottier
MD
,
Naaz
A
,
Li
Y
,
Fraker
PJ
. 
Enhancement of hematopoiesis and lymphopoiesis in diet-induced obese mice
.
Proc Natl Acad Sci U S A
2012
;
109
:
7622
9
.
50.
Singer
K
,
DelProposto
J
,
Morris
DL
,
Zamarron
B
,
Mergian
T
,
Maley
N
, et al
Diet-induced obesity promotes myelopoiesis in hematopoietic stem cells
.
Mol Metab
2014
;
3
:
664
75
.
51.
Ito
K
,
Carracedo
A
,
Weiss
D
,
Arai
F
,
Ala
U
,
Avigan
DE
, et al
A PML-PPAR-delta pathway for fatty acid oxidation regulates hematopoietic stem cell maintenance
.
Nat Med
2012
;
18
:
1350
8
.
52.
Ooi
EM
,
Watts
GF
,
Sprecher
DL
,
Chan
DC
,
Barrett
PH
. 
Mechanism of action of a peroxisome proliferator-activated receptor (PPAR)-delta agonist on lipoprotein metabolism in dyslipidemic subjects with central obesity
.
J Clin Endocrinol Metab
2011
;
96
:
E1568
76
.
53.
Choi
YJ
,
Roberts
BK
,
Wang
X
,
Geaney
JC
,
Naim
S
,
Wojnoonski
K
, et al
Effects of the PPAR-delta agonist MBX-8025 on atherogenic dyslipidemia
.
Atherosclerosis
2012
;
220
:
470
6
.
54.
Yoshinaga
M
,
Taki
K
,
Somada
S
,
Sakiyama
Y
,
Kubo
N
,
Kaku
T
, et al
The expression of both peroxisome proliferator-activated receptor delta and cyclooxygenase-2 in tissues is associated with poor prognosis in colorectal cancer patients
.
Dig Dis Sci
2011
;
56
:
1194
200
.
55.
Yang
L
,
Zhang
H
,
Zhou
ZG
,
Yan
H
,
Adell
G
,
Sun
XF
. 
Biological function and prognostic significance of peroxisome proliferator-activated receptor delta in rectal cancer
.
Clin Cancer Res
2011
;
17
:
3760
70
.
56.
Hautala
AJ
,
Leon
AS
,
Skinner
JS
,
Rao
DC
,
Bouchard
C
,
Rankinen
T
. 
Peroxisome proliferator-activated receptor-delta polymorphisms are associated with physical performance and plasma lipids: the HERITAGE Family Study
.
Am J Physiol Heart Circ Physiol
2007
;
292
:
H2498
505
.
57.
Andrulionyte
L
,
Peltola
P
,
Chiasson
JL
,
Laakso
M
;
STOP-NIDDM Study Group
. 
Single nucleotide polymorphisms of PPARD in combination with the Gly482Ser substitution of PGC-1A and the Pro12Ala substitution of PPARG2 predict the conversion from impaired glucose tolerance to type 2 diabetes: the STOP-NIDDM trial
.
Diabetes
2006
;
55
:
2148
52
.
58.
Shin
HD
,
Park
BL
,
Kim
LH
,
Jung
HS
,
Cho
YM
,
Moon
MK
, et al
Genetic polymorphisms in peroxisome proliferator-activated receptor delta associated with obesity
.
Diabetes
2004
;
53
:
847
51
.
59.
Yang
Y
,
Burke
RV
,
Jeon
CY
,
Chang
SC
,
Chang
PY
,
Morgenstern
H
, et al
Polymorphisms of peroxisome proliferator-activated receptors and survival of lung cancer and upper aero-digestive tract cancers
.
Lung Cancer
2014
;
85
:
449
56
.
60.
Ticha
I
,
Gnosa
S
,
Lindblom
A
,
Liu
T
,
Sun
XF
. 
Variants of the PPARD gene and their clinicopathological significance in colorectal cancer
.
PLoS One
2013
;
8
:
e83952
.