Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Research Article

Are Peroxisome Proliferator-Activated Receptors Involved in Skeletal


Muscle Wasting during Experimental Cancer Cachexia?
Role of B2-Adrenergic Agonists
Gemma Fuster, Sı́lvia Busquets, Elisabet Ametller, Mireia Olivan, Vanessa Almendro,
Cibely Cristine Fontes de Oliveira, Maite Figueras, Francisco J. López-Soriano,
and Josep M. Argilés
Cancer Research Group, Departament de Bioquı́mica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain

Abstract view, especially if there are different mediators involved. Taking


this into consideration, a lack of knowledge about signaling
Implantation of the Yoshida AH-130 ascites hepatoma to rats
pathways and transcription factors involved in muscle wasting
resulted in a decrease in muscle weight 7 days after the
exists. Some work has postulated a role for nuclear factor-nB
inoculation of the tumor. These changes were associated with
(NF-nB) in muscle wasting associated with cytokines (7) and
increases in the mRNA content for both peroxisome prolifer-
tumor-derived factors (8). Other transcription factors, such as
ator-activated receptor (PPAR) ; and PPARD in skeletal muscle.
activator protein-1 (AP-1) and CCAAT/enhancer binding protein
The increase in gene expression for these transcription factors
(C/EBP), have also been involved in sepsis-induced muscle
was related to increases in the expression of several genes
cachexia (9). Results from our laboratory indicate that the
involved in fatty acid transport, activation, and oxidation.
transcription factor AP-1 could also be involved during cancer
Tumor burden also resulted in increases in PPAR; coactivator-
cachexia (10, 11). Not much attention has been focused on the role
1A gene expression and pyruvate dehydrogenase kinase 4. All
of peroxisome proliferator-activated receptors (PPAR) in skeletal
these changes in lipid metabolism genes suggest that a
muscle. These transcription factors are associated with changes in
metabolic shift occurs in skeletal muscle of tumor-bearing rats
lipid metabolism as well as UCP expression (12) and apoptosis (13).
toward a more oxidative phenotype. Formoterol treatment to
PPARs are transcription factors belonging to the superfamily of
tumor-bearing rats resulted in an amelioration of all the
nuclear receptors. Three isoforms (a, y, and g) have been described
changes observed as a result of tumor burden. Administration
(14). They act on DNA response elements as heterodimers with the
of this B2-adrenergic agonist also resulted in a decrease in
nuclear retinoic acid receptor. Their natural activating ligands are
mRNA content of muscle PPARA, PPARD, and PPAR;, as well as
fatty acids and lipid-derived substrates. PPARa is present in liver,
in mRNA levels of many of the genes involved in both lipid and
heart, and, to a lesser extent, skeletal muscle; when activated, it
mitochondrial metabolism. All these results suggest an
promotes fatty acid oxidation, ketone body synthesis, and glucose
involvement of the different PPARs as transcription factors
sparing. PPARg is expressed in adipose tissue, lower intestine,
related with muscle wasting and also indicate that a possible
skeletal muscle, and immune cells; activation of PPARg induces the
mode of action of the anticachectic compound formoterol may
differentiation of preadipocytes into adipocytes and stimulates
involve a normalization of the levels of these transcription
triglyceride storage. The PPARs are thus major regulators of lipid
factors. [Cancer Res 2007;67(13):6512–9]
and glucose metabolism, allowing adaptation to the prevailing
nutritional environment (14). PPARy has a broad expression
Introduction pattern in adult and is expressed very early during embryogenesis
Muscle wasting is a common feature in many pathologic states, (15). These past few years, it has been shown that treatment with
including infection and cancer (1). Muscle wasting, the main trend PPARy agonists normalizes blood lipids and also reduces insulin
of cachexia, is responsible for the death of at least 30% of cancer resistance and adiposity in rodents and primates. Utilization of
patients (2). Although we know the main events related with both cellular and animal models revealed that this nuclear receptor
muscle wasting [activation of myofibrillar protein degradation, plays a central role in the control of fatty acid burning in adipose
induction of apoptosis, and activation of uncoupling proteins tissue and skeletal muscle. Furthermore, PPARy seemed to be
(UCP); refs. 3, 4], we have contradictory evidence about the possible important for adaptive response of skeletal muscle to environ-
mediators involved. Indeed, whereas involvement of different mental changes, such as physical exercise (15).
cytokines, mainly tumor necrosis factor-a (TNFa) and interleu- h2-adrenergic agonists are potent muscle growth promoters in
kin-6 (IL-6), has been postulated, other studies describe a more many animal species (16, 17), resulting in skeletal muscle
direct role for tumor-derived factors, such as proteolysis-inducing hypertrophy (18–20) and reducing body fat content (21, 22).
factor (PIF) and lipid-mobilizing factor (5, 6). The intracellular Interestingly, results from our laboratory clearly indicate that
signaling pathway may have a key role, from a therapeutic point of formoterol is a very efficient agent preventing muscle weight loss in
tumor-bearing rats (23). In vivo treatment can effectively reverse
muscle wasting loss decreasing protein degradation and increasing
Requests for reprints: Josep M. Argilés, Cancer Research Group, Departament de
the rate of protein synthesis in skeletal muscle, therefore favoring
Bioquı́mica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, protein accretion (23).
Diagonal 645, 08028 Barcelona, Spain. Phone: 34-934021002; Fax: 34-934021559; Bearing this in mind, the aim of the present investigation was to
E-mail: jargiles@ub.edu.
I2007 American Association for Cancer Research. ascertain if tumor burden induces any changes in PPARs gene
doi:10.1158/0008-5472.CAN-07-0231 transcription in skeletal muscle and if these changes are associated

Cancer Res 2007; 67: (13). July 1, 2007 6512 www.aacrjournals.org

Downloaded from cancerres.aacrjournals.org on July 9, 2018. © 2007 American Association for Cancer
Research.
PPARs and Cancer Cachexia

Table 1. Food intake, body weight, and muscle weight in tumor-bearing rats

Experimental groups

C, n (mean F SE) C + F, n (mean F SE) TB, n (mean F SE) TB + F, n (mean F SE) P, ANOVA

IBW-FBW 5 (49 F 3a) 5 (50 F 2a) 6 (11 F 5b) 7 (17 F 5b) <0.001
Food intake 5 (133 F 1a) 5 (131 F 2a) 6 (106 F 1b) 7 (108 F 1b) <0.001
Muscle weights
GSN 5 (718 F 10.5a) 4 (774 F 8b) 5 (505 F 17c) 7 (611 F 18d) <0.001
Soleus 5 (52 F 2.2a) 4 (52 F 1.6a) 5 (41 F 2.7b) 6 (44 F 1.4b) 0.001
EDL 5 (59 F 2a) 5 (60 F 1.6a) 5 (42 F 1.8b) 6 (51 F 2c) <0.001
Tibialis 5 (236 F 2.5a) 5 (266 F 4.5b) 5 (171 F 4.8c) 7 (200 F 9d) <0.001

NOTE: For further details, see Materials and Materials. n is the number of animals. Food intake (grams) refers to the ingestion for each rat during the
period of the experiment before sacrifice (7 d). Initial body weight-final body weight without tumor is expressed as grams. Tissue weights are expressed
as mg/100 g of initial body weight. Formoterol was given for 7 d s.c. (0.3 mg/kg body weight). Statistical significance of the results by one-way ANOVA
and statistically significant difference by post-hoc Duncan test. Different superscripts indicate differences between groups.
Abbreviations: IBW, initial body weight; FBW, final body weight without tumor; GSN, gastrocnemius; C, control; F, formoterol-treated animals; TB,
tumor-bearing animals.

with alterations in gene transcription of different types of proteins TTTCTGTG-3¶ and 5¶-GCCTTCAGTTTGCTTTCCAG-3¶ (gi: 16758425). Ampli-
involved in lipid metabolism. A second objective in the present fication conditions consisted in 5 s of denaturation at 94jC, 9 s of annealing
investigation was to see if these changes could be reversed by at 60jC, and 9 s of extension at 72jC for each step for 45 cycles for FAT,
administration of the h2-agonist formoterol, a molecule that shows PGC1a, PDK4, and ACS4 and for FATP consisted in 5 s of denaturation at 94jC,
9 s of annealing at 60jC, 9 s of extension at 72jC, and 3 s at 87jC to detect
a clear anticachectic action in skeletal muscle during cancer (23).
amplification product avoiding unspecific products after each cycle, for
45 cycles. Myosin heavy-chain I (MHCI) and MHCIIA primers were designed
Materials and Methods by Mortensen et al. (27). To avoid the detection of possible contamination
Animals. Male Wistar rats (Interfauna) of 5 weeks of age were used. The by genomic DNA, primers were designed in different exons. The real-time PCR
animals were maintained at 22 F 2jC with a regular light-dark cycle (light was done using a commercial kit (LightCycler FastStart DNA Master PLUS
on, 08:00–20:00) and had free access to food and water. The food intake was SYBR Green I, Roche). The relative amount of all mRNA was calculated using
measured daily. All animal manipulations were made in accordance with comparative C T method. 18S mRNA was used as the invariant control for all
the European Community guidelines for the use of laboratory animals. studies.
Tumor inoculation and treatment. Rats were divided into two groups, Protein extraction and Western blotting. For PDK4 analysis, EDL and
namely controls and tumor hosts. The latter received an i.p. inoculum of soleus muscles were homogenized in 10 mmol/L HEPES (pH 7.5),
108 AH-130 Yoshida ascites hepatoma cells obtained from tumors grow- containing 10 mmol/L MgCl2, 5 mmol/L KCl, 0.1 mmol/L EDTA, 0.1%
ing in the exponential phase (24). Both groups were further divided into Triton X-100, 1 mmol/L DTT, and 5 AL of a protease inhibitor cocktail
treated and untreated, the former being given daily i.p. dose of formoterol (Sigma)/mL of buffer. They were then centrifuged at 7,000 rpm for 5 min at
(0.3 mg/kg body weight), dissolved in physiologic solution, and the latter a 4jC, and the supernatants were collected. For MCPTI, PGC1a, and
corresponding volume of solvent. On day 7 after tumor transplantation, the mitochondrial complex II analysis, an enriched mitochondrial/nuclear
animals were weighed and anesthetized with an i.p. injection of ketamine/ fraction was obtained: the pellets being resuspended in 500 AL of buffer
xylazine mixture (3:1; Imalgene and Rompun, respectively). The tumor was [10 mmol/L HEPES (pH 7.9) containing 0.5 mol/L NaCl, 1.5 mmol/L MgCl2,
harvested from the peritoneal cavity and its volume and cellularity were 0.2 mmol/L EDTA, 30% glycerol, 1 mmol/L DTT, 5 AL of a protease inhibitor
evaluated. Tissues were rapidly excised, weighed, and frozen in liquid nitrogen. cocktail/mL of buffer] and left for 30 min on ice. They were later
Biochemicals. They were all reagent grade and obtained either from centrifuged at 7,000 rpm for 5 min at 4jC, and the supernatants were
Roche S.A. or from Sigma Chemical Co. collected. About FAT analysis, an enriched plasma membrane was obtained
RNA isolation. Total RNA from soleus and extensor digitorum longus from the total protein extracts; these were obtained by ultracentrifugation
(EDL) muscle was extracted by TriPure kit (Roche), a commercial modifica- at 47,000 rpm for 1 h at 4jC. Protein concentrations were determined
tion of the acid guanidinium isothiocyanate/phenol/chloroform method (25). according to the method of bicinchoninic acid (Pierce). Equal amounts of
Real-time PCR. First-strand cDNA was synthesized from total RNA protein (50 or 100 Ag) were heat denatured in sample loading buffer
with oligonucleotide dT15 primers and random primers p(dN)6 by using a [50 mmol/L Tris-HCl (pH 6.8), 100 mmol/L DTT, 2% SDS, 0.1% bromphenol
cDNA synthesis kit (Transcriptor Reverse Transcriptase, Roche). Analysis of blue, 10% glycerol], resolved by SDS-PAGE (10% polyacrylamide, 0.1% SDS),
mRNA levels for PPARa, PPARy, PPARg, muscle carnitine palmitoyltrans- and transferred to Immobilon membranes (Immobilon polyvinylidene
ferase-I (MCPTI), and CPTII were done with primers designed to detect gene difluoride, Millipore). The filters were blocked with 5% PBS-nonfat dry milk
products as described previously (26). Oligonucleotide sequences for fatty and then incubated with polyclonal antibodies: anti-PDK4, anti-PGC1a,
acid translocase (FAT) were 5¶-CTCTGACATTTGCAGGTC-3¶ and 5¶-CAC- anti-MCPTI, anti-FAT (Santa Cruz Biotechnology), and monoclonal anti-
AGGCTTTCCTTCTTTGC-3¶ (gi: 98674378), for fatty acid transport protein body (mAb) anti-OxPhos complex II (Molecular Probes). Polyclonal anti-
(FATP) were 5¶-GCATGGATGATCGGCTATTT-3¶ and 5¶-GATGTTCCCTGC- bodies anti-Na+/K+ ATPase a-subunit (Developmental Studies Hybridoma
TGAGTGGT-3¶ (gi: 18811712), for PPARg coactivator-1a (PGC1a) were Bank), anti–glyceraldehyde-3-phosphate dehydrogenase, and mAb anti-
5¶-AAGGTCCCCAGGCAGTAGAT-3¶ and 5¶-TTCAGACTCCCGCTTCTCAT-3¶ porin (Calbiochem) were used as invariant controls for the different studies.
(gi: 13786187), for pyruvate dehydrogenase kinase 4 (PDK4) were 5¶-CCTT- Donkey anti-mouse peroxidase-conjugated IgG (Jackson ImmunoResearch,
TGCTGGTTTTGGTTA-3¶ and 5¶-CACCAGTCATCAGCCTCAGA-3¶ (gi: Laboratories, Inc.), rabbit anti-goat peroxidase-conjugated IgG (Acris
16758425), and for acyl-CoA synthetase 4 (ACS4) were 5¶-CACCATTGCCAT- Antibodies GmbH), and goat anti-rabbit horseradish peroxidase conjugate

www.aacrjournals.org 6513 Cancer Res 2007; 67: (13). July 1, 2007

Downloaded from cancerres.aacrjournals.org on July 9, 2018. © 2007 American Association for Cancer
Research.
Cancer Research

(Bio-Rad) were used as secondary antibodies. The membrane-bound anorexia present in the cancer patients (29). Bearing this in
immune complexes were detected by an enhanced chemiluminescence mind, it is obvious that a good understanding of the molecular
system (EZ-ECL). mechanisms involved in the signaling of these mediators may be
Statistical analysis. Statistical analysis of the data was done by means of
very positive in the design of the therapeutic strategy. This is
one-way ANOVA.
especially relevant because different mediators may be sharing
the same signaling pathways (30, 31). However, very few
Results and Discussion investigations have focused on the signaling of these mediators
Muscle wasting during different pathologic conditions exerts a in skeletal muscle, especially about transcription factors. Penner
negative effect on survival and quality of life in the patient. et al. (32) described, in a model of sepsis, the involvement of
Therefore, understanding the molecular mechanisms and the NF-nB and AP-1 in muscle wasting. Other reports, using expe-
different mediators involved provides for the design of efficient rimental cancer models, have also suggested that NF-nB is
therapeutic approaches (28). From this point of view, the therapy involved in the signaling of muscle wasting (30, 31). In our own
against wasting during cachexia has concentrated on either laboratory, we have shown recently that there is an increased
increasing food intake or normalizing the persistent metabolic activation of AP-1 in skeletal muscle of tumor-bearing rats,
alterations that take place in the patient. It is difficult to apply a therefore suggesting that this factor is indeed involved in the
therapeutic approach based on the neutralization of the muscle events that take place during cancer cachexia (10).
potential mediators involved in muscle wasting (i.e., TNFa, Indeed, the i.m. administration of adenoviruses carrying TAM-67
IL-6, IFN-g, and PIF) because many of them are involved at the [a negative-dominant of c-jun (AP-1)] resulted in an improve-
same time in promoting the metabolic alterations and the ment of the muscle weight during tumor growth (11).

Table 2. Gene expression of different proteins related to lipid metabolism

Experimental groups

C, n (mean F SE) C + F, n (mean F SE) TB, n (mean F SE) TB + F, n (mean F SE) P, ANOVA

(A) Skeletal muscle mRNA content of the different PPARs


PPARa
EDL 4 (100 F 13a) 5 (49 F 10b) 5 (102 F 26a) 5 (23 F 18b) <0.05
Soleus 4 (100 F 24ab) 4 (111 F 12ab) 5 (137 F 12a) 4 (76 F 22b) 0.1
PPARg
EDL 4 (100 F 8ab) 5 (45 F 5ac) 4 (148 F 31b) 5 (30 F 12c) <0.05
Soleus 5 (100 F 28 )
a
6 (119 F 6a) 6 (308 F 31b) 6 (77 F 19a) <0.05
PPARy
EDL 5 (100 F 12a) 5 (93 F 9a) 4 (241 F 20b) 5 (85 F 22a) <0.001
Soleus 4 (100 F 13a) 5 (109 F 12a) 5 (476 F 46b) 6 (140 F 29ab) 0.09

(B) Skeletal muscle mRNA content of the different genes related to lipid metabolism
Fatty acid transport and activation
FAT
EDL 4 (100 F 18a) 4 (22 F 11a) 5 (315 F 34b) 5 (26 F 10a) <0.05
Soleus 4 (100 F 6ab) 5 (57 F 28a) 4 (186 F 12c) 4 (135 F 5b) <0.001
FATP
EDL 4 (100 F 9a) 4 (51 F 10b) 4 (151 F 17c) 4 (31 F 14b) <0.001
Soleus 4 (100 F 22 )
a
5 (68 F 15 )a
5 (181 F 14b) 4 (105 F 11a) <0.05
ACS4
EDL 5 (100 F 18a) 5 (68 F 6ab) 4 (100 F 14a 5 (58 F 9b) <0.05
Soleus 4 (100 F 5a) 5 (160 F 9b) 4 (138 F 7bc) 5 (123 F 9ac) <0.05
Oxidation
MCPTI
EDL 4 (100 F 11a) 4 (38 F 5b) 3 (253 F 38c) 5 (35 F 5b) <0.001
Soleus 5 (100 F 7a) 6 (110 F 5a) 4 (216 F 17b) 4 (145 F 10a) <0.001
CPTII
EDL 3 (100 F 16a) 4 (40 F 9b) 3 (188 F 19c) 5 (20 F 16b) <0.001
Soleus 4 (100 F 12 )
a
6 (91 F 6 )
a
4 (156 F 11b) 4 (112 F 20a) <0.05

NOTE: For further details, see Materials and Methods. n is the number of animals. The results are expressed as a percentage of controls. Statistical
significance of the results by one-way ANOVA and statistically significant difference by post-hoc Duncan test. Different superscripts indicate differences
between groups.
Abbreviations: C, control; F, formoterol-treated animals; TB, tumor-bearing animals; FAT, fatty acid translocase; PPAR, peroxisome proliferator-activated
receptor; FATP, fatty acid transport protein; ACS4, acyl-CoA synthetase 4; MCPTI, muscle carnitine palmitoyltransferase I; CPTII, carnitine
palmitoyltransferase II.

Cancer Res 2007; 67: (13). July 1, 2007 6514 www.aacrjournals.org

Downloaded from cancerres.aacrjournals.org on July 9, 2018. © 2007 American Association for Cancer
Research.
PPARs and Cancer Cachexia

Table 3. Skeletal muscle protein content of different proteins related to lipid metabolism

Experimental groups

C, n (mean F SE) C + F, n (mean F SE) TB, n (mean F SE) TB + F, n (mean F SE) P, ANOVA

FAT
EDL 4 (100 F 14ab) 6 (99 F 17ab) 3 (198 F 97a) 3 (66 F 12b) ns
Soleus 5 (100 F 5a) 7 (108 F 17ab) 5 (160 F 25b) 3 (99 F 18a) 0.09
MCPTI
EDL 5 (100 F 7ab) 6 (38 F 14a) 5 (256 F 93b) 3 (97 F 12ab) <0.05
Soleus 4 (100 F 16a) 7 (94 F 22a) 5 (319 F 50b) 4 (185 F 35a) <0.001
PDK4
EDL 4 (100 F 16a) 4 (77 F 6a) 3 (175 F 21b) 4 (99 F 29a) <0.001
Soleus 5 (100 F 20) 6 (109 F 13) 3 (170 F 31) 4 (135 F 55) ns
PGC1a
EDL 5 (100 F 11a) 6 (92 F 13a) 4 (341 F 83b) 4 (131 F 20a) <0.05
Soleus 4 (100 F 9a) 7 (64 F 9a) 4 (196 F 26b) 3 (95 F 21a) <0.001

NOTE: For further details, see Materials and Methods. n is the number of animals. The results are expressed as a percentage of controls. Statistical
significance of the results by one-way ANOVA and statistically significant difference by post-hoc Duncan test. Different superscripts indicate differences
between groups.
Abbreviations: C, control; F, formoterol-treated animals; TB, tumor-bearing animals; FAT, fatty acid translocase; MCPTI, muscle carnitine
palmitoyltransferase I; PDK4, pyruvate dehydrogenase kinase-4; PGC1a, PPAR coactivator-1a; ns, nonsignificant differences.

In spite of the involvement of PPARs in several metabolic path- Table 1 shows the effect of tumor growth on body weight, food
ways that are altered during cancer cachexia, no information is intake, and muscle weights in animals bearing the Yoshida AH-130
available on the mRNA levels of these transcription factors in skeletal ascites hepatoma. The growth of this tumor causes in the host a
muscle during this catabolic situation. This was the aim of the rapid and progressive loss of body weight and tissue waste,
present investigation: to elucidate any changes of PPARs in skeletal particularly in skeletal muscle (33). Acceleration of tissue protein
muscle during tumor growth and also to relate them to the changes breakdown accounts for most of the waste in the AH-130 bearers
of mRNA content of different genes involved in lipid metabolism. (24, 34). The effects of the tumor on weight loss were very

Figure 1. A, skeletal muscle protein


content of mitochondrial complex II. Porin
was used as invariant control. B, soleus
mRNA content of the different MHCs. 18S
was used as invariant control. For further
details, see Materials and Methods.
Columns, mean of four animals in each
group; bars, SE. The results are expressed
as a percentage of controls. C, control;
F, formoterol-treated animals; TB,
tumor-bearing animals. Statistical
significance of the results by one-way
ANOVA and statistically significant
difference by post-hoc Duncan test. a, b,
and c, differences between groups.

www.aacrjournals.org 6515 Cancer Res 2007; 67: (13). July 1, 2007

Downloaded from cancerres.aacrjournals.org on July 9, 2018. © 2007 American Association for Cancer
Research.
Cancer Research

Figure 2. A, mRNA content of PDK4 and


PGC1a in EDL muscle of tumor-bearing
animals. 18S was used as invariant control.
B, mRNA content of PDK4 and PGC1a in
soleus muscle of tumor-bearing animals.
18S was used as invariant control. For
further details, see Materials and Methods.
Columns, mean values of four animals in
each group; bars, SE. The results are
expressed as a percentage of controls.
Statistical significance of the results by
one-way ANOVA and statistically
significant difference by post-hoc Duncan
test. a and b, differences between groups.

significant, with a decrease of 28% in the tumor-bearing group. muscle of tumor-bearing rats (35). In addition, all these changes in
This effect on body weight was accompanied by a marked anorexia: lipid metabolism genes clearly suggest that a metabolic shift occurs
the tumor-bearing rats ate 20% less than the control ones (Table 1). in skeletal muscle of tumor-bearing rats toward a more oxidative
It can also be seen that the implantation of the tumor resulted in a phenotype. Previous studies have shown that PPARy is involved in
general decrease of muscle weights (30% gastrocnemius, 29% EDL, muscle fiber composition; an increase in this transcription factor
28% tibialis, and 21% soleus) 7 days after the inoculation of the determines a more oxidative fiber phenotype (36). Bearing this in
tumor. These changes were associated with increases in the mRNA mind, the results found in this study clearly agree with this
content for PPARg (48% in EDL and 208% in soleus) and PPARy previously reported data. In fact, as can be seen in Fig. 1A, tumor
(141% in EDL and 376% in soleus) but no changes in PPARa mRNA burden resulted in a clear increase (49%) in oxidative muscle fibers,
content (Table 2A). It is interesting to note here that PPARy is as measured by the expression of the MHCI gene (27). Conversely,
especially important for adaptable response in skeletal muscle and the presence of the tumor did not have any effects on the mRNA
precisely the largest changes occur at the level of this transcription content of MHCIIA, a clear glycolytic marker (Fig. 1A; ref. 27). The
factor (15). The increase in gene expression for these two same tendency toward a more oxidative phenotype in the muscles
transcription factors was related with increases in the expression of tumor-bearing rats was confirmed when analyzing the
of genes involved in fatty acid transport activation and oxidation. mitochondrial complex II protein content as observed in Fig. 1B
Thus, FAT and FATP (both proteins associated with fatty acid (37). PGC1a is a transcriptional coregulator that coordinates the
cellular transport) were increased 215% and 51% in EDL and 86% formation/maintenance of slow twitch fibers in skeletal myocytes,
and 81% in soleus, respectively (Table 2B). In addition, FAT protein and this seems to be directly controlled by PPARy (37). In addition,
content was also increased in soleus (60%) of tumor-bearing PPARs seem to independently regulate specific PDK isoforms
animals (Table 3). Similarly ACS4 (the protein that activates the transcript levels, which are likely to impart important metabolic
intracellular fatty acids for their subsequent use in oxidation mediation of fuel utilization by the muscle. Experiments to date
pathways) was increased 38% in soleus (Table 2B). Finally, MCPTI suggest that PDK4 is the major isoenzyme responsible for changes
and CPTII mRNA content (both involved in the transport of fatty in pyruvate dehydrogenase complex activity in response to
acid across the mitochondrial membrane) were also increased as a various different metabolic conditions (38). Tumor burden also
result of tumor burden by 153% and 116% for MCPTI in EDL and resulted in increases in the PGC1a gene expression (83% and
soleus, respectively, and 88% and 56% for CPTII in EDL and soleus, 102% in EDL and soleus, respectively) and PDK4 (373% and 172%
respectively. MCPTI protein content was also increased in EDL in EDL and soleus, respectively; Fig. 2). In addition PDK4 and
(156%) and in soleus (219%) muscles of tumor-bearing animals PGC1a protein content was also increased both in EDL (75% and
(Table 3). All these results agree with previous reports that have 241%, respectively) and in soleus (70% and 96%, respectively)
already suggested that lipid metabolism is increased in skeletal muscles of tumor-bearing animals (Table 3). These data agree

Cancer Res 2007; 67: (13). July 1, 2007 6516 www.aacrjournals.org

Downloaded from cancerres.aacrjournals.org on July 9, 2018. © 2007 American Association for Cancer
Research.
PPARs and Cancer Cachexia

with the work by Puigserver et al. (39) that stated that cytokine- It is interesting to note that formoterol also had an effect on
induced activation of PGC1a in culture muscle cells or muscle nontumor-bearing animals (Table 3A and B), therefore suggesting
in vivo causes increased respiration and expression of genes that the action [possibly linked with variations of the intracellular
linked to mitochondrial uncoupling and energy expenditure. It is concentration of cyclic AMP (cAMP) in cells] may be linked with
therefore possible to suggest that the cytokine changes related changes in PPARs (42, 43). In fact, a previous report using the h2-
with cancer cachexia are altering the mRNA content of PGC1a agonist phenylephrine already suggests this possibility (44). On the
and this may affect the transcriptional activity of both PPARg and other hand, it is well known that h2-agonists also affect lipid
PPARy. In connection with this, it is interesting to remark that metabolism on adipose tissue, decreasing lipid synthesis and
the activation of UCP2 and UCP3 is also observed in tumor- favoring lipid oxidation (45), but this is the first report that clearly
bearing rats (40). This could be very well linked with PPARs shows a possible action of h2-agonists on skeletal muscle lipid
activation, possibly through PGC1a coactivation. metabolism. The effects of the h2-agonists via h2-adrenergic
Formoterol, a h2-adrenergic agonist, is a very efficient agent receptors increase cAMP and interfere with PPARs gene expression
preventing muscle weight loss in tumor-bearing rats (23). Bearing (Fig. 3). However, another possibility is that formoterol actually
this in mind, we decided to see if the effects of formoterol on interferes with the signaling of TNFa on skeletal muscle cells.
muscle wasting during experimental tumor growth were connected Indeed, a report already suggests that the h2-agonist clenbuterol
with changes in PPARs and the genes involved in lipid metabolism decreases the circulating levels and activity of TNFa (46). This
studied above. The results obtained seem to indicate that makes particular sense for the tumor model involved in this study
formoterol treatment can normalize the changes induced by the because the Yoshida AH-130 ascites hepatoma seems to be highly
tumor (Fig. 2; Tables 1–3). Several studies have shown that h2- dependent on the increase of TNFa circulating levels (47). In
adrenergic agonists act on skeletal muscle by changing the fiber addition, in this study, formoterol treatment also reduced the
composition toward a more glycolytic pattern (41). Taking this into TNFa mRNA content in gastrocnemius muscle [tumor, 185 F 15a
account, the different effects of formoterol on different types of arbitrary units (4); tumor treated with formoterol, 115 F 9b
muscles were studied; therefore, these studies were done in a arbitrary units (3); P < 0.05]. (Statistical significance of the results
predominantly oxidative muscle (soleus) and in a predominantly by one-way ANOVA and statistically significant difference by post-
glycolytic muscle (EDL). The results found here are in line with hoc Duncan test; different superscripts indicate differences
previously described observations (41). As seen in Fig. 1A, between groups).
formoterol treatment resulted in a significant increase in MHCIIA The data presented here, in a way, complicate the understanding
(a glycolytic marker) in mRNA soleus content, in both control and of the different transcription factors in muscle wasting because
tumor-bearing animals. In addition, formoterol also significantly some reports have suggested that activation of different PPARs may
decreased mitochondrial complex II content in tumor-bearing rats be useful in preventing metabolic alterations linked with
(Fig. 1B), this observation supporting further the change in fiber inflammation by cytokines (48). However, other reports suggest
composition. the opposite, emphasizing that PPARs may have a role in mediating

Figure 3. Hypothetical involvement of


the different transcription factors during
muscle wasting in cancer cachexia. ARb2,
h2-adrenoceptor; IL-6R, IL-6 receptor;
PIFR, PIF receptor; TNFR1, TNF receptor
1; TNFR2, TNF receptor 2.

www.aacrjournals.org 6517 Cancer Res 2007; 67: (13). July 1, 2007

Downloaded from cancerres.aacrjournals.org on July 9, 2018. © 2007 American Association for Cancer
Research.
Cancer Research

cytokine action in skeletal muscle (39). In addition, several studies tissue during cancer cachexia. It becomes clear that future research
(49) indicate that PPARs can partially interfere in NF-nB signaling into this field is necessary and may provide important tools to
in skeletal muscle, therefore controlling to some extent the action design effective drugs for the treatment of wasting in skeletal
exerted by cytokines through these transcription factors (Fig. 3). muscle during pathologic conditions.
Bearing all this in mind, one has to look at the plethora of
transcription factors involved in muscle wasting during cancer
cachexia because possibly some of them play a more important Acknowledgments
role in some cases, depending on the species, the tumor model, and
Received 1/18/2007; revised 3/22/2007; accepted 4/12/2007.
the tumor stage. It is perfectly conceivable to understand that by Grant support: Ministerio de Educación y Ciencia Dirección General de
agonizing PPARg and PPARy, one can produce a reduction in Investigación Cientı́fica y Técnica BFI2002-02186 (G. Fuster); Ministerio de Sanidad
muscle weight partly through the interference in the NF-nB y Consumo Instituto de Salud Carlos III grant 03/0100 (E. Ametller); La Marató TV3
grant 04/1010 (M. Olivan); Programme Alhan, the European Union Programme of High
signaling system. It can also be suggested that the elevation of the Level Scholarships for Latin America, scholarship E05D059293BR (C.C. Fontes de
different PPARs during muscle wasting in cancer may be a counter Oliveira); Fondo de Investigaciones Sanitarias de la Seguridad Social of the Ministerio
de Sanidad y Consumo grant 06/0907; and Ministerio de Ciencia y Tecnologı́a grant
metabolism triggered by the host to control an excessive SAF 4744-2005.
degradation of muscle protein and favoring lipid utilization for The costs of publication of this article were defrayed in part by the payment of page
muscle. charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
In conclusion, the results presented here contribute to a better We thank Industriale Chimica s.r.l. for providing micronized formoterol fumarate;
understanding of the role of transcription factors in the muscle Dr. Carles Barceló-Vidal for his statistical support.

References 14. Ferre P. The biology of peroxisome proliferator- 28. Argilés JM, Almendro V, Busquets S, López-Soriano
activated receptors: relationship with lipid metabolism FJ. The pharmacological treatment of cachexia. Curr
1. Argilés JM, Alvarez B, López-Soriano FJ. The metabolic and insulin sensitivity. Diabetes 2004;53:S43–50. Drug Targets 2004;5:265–77.
basis of cancer cachexia. Med Res Rev 1997;17:477–98. 15. Grimaldi PA. Regulatory role of peroxisome prolifer- 29. Argilés JM, Busquets S, Garcı́a-Martı́nez C, López-
2. Warren S. The immediate causes of death in cancer. ator-activated receptor y (PPARy) in muscle metabo- Soriano FJ. Mediators involved in the cancer anorexia-
Am J Med Sci 1932;184:610–5. lism. A new target for metabolic syndrome treatment? cachexia syndrome: past, present, and future. Nutrition
3. Argilés JM, Moore-Carrasco R, Fuster G, Busquets S, Biochimie 2005;87:5–8. 2005;21:977–85.
López-Soriano FJ. Cancer cachexia: the molecular 16. Stock MJ, Rothwell NJ. Effects of h-adrenergic 30. Whitehouse AS, Tisdale MJ. Increased expression of
mechanisms. Int J Biochem Cell Biol 2003;35:405–9. agonists on metabolism and body composition. In: the ubiquitin-proteasome pathway in murine myotubes
4. Argilés JM, Busquets S, López-Soriano FJ. The role of Buttery PJ, Hayes NB, Lindsay DB, editors. Control and by proteolysis-inducing factor (PIF) is associated with
uncoupling proteins in pathophysiological states. Bio- manipulation of animal growth. London: Butterworths; activation of the transcription factor NF-nB. Br J Cancer
chem Biophys Res Commun 2002;293:1145–52. 1985. p. 249–57. 2003;89:1116–22.
5. Lorite MJ, Smith HJ, Arnold JA, Morris A, Thompson 17. Kim YS, Sainz RD. h-Adrenergic agonists and 31. Wyke SM, Tisdale MJ. NF-nB mediates proteolysis-
MG, Tisdale MJ. Activation of ATP-ubiquitin-dependent hypertrophy of skeletal muscles. Life Sci 1992;50: inducing factor induced protein degradation and
proteolysis in skeletal muscle in vivo and murine 397–407. expression of the ubiquitin-proteasome system in
myoblasts in vitro by a proteolysis-inducing factor 18. Agbenyega ET, Wareham AC. Effect of clenbuterol on skeletal muscle. Br J Cancer 2005;92:711–21.
(PIF). Br J Cancer 2001;85:297–302. skeletal muscle atrophy in mice induced by the 32. Penner CG, Gang G, Wray C, Fischer JE, Hasselgren
6. Todorov PT, McDevitt TM, Meyer DJ, Ueyama H, glucocorticoid dexamethasone. Comp Biochem Physiol PO. The transcription factors NF-nb and AP-1 are
Ohkubo I, Tisdale MJ. Purification and characterization Comp Physiol 1992;102:141–5. differentially regulated in skeletal muscle during sepsis.
of a tumor lipid-mobilizing factor. Cancer Res 1998;58: 19. Hinkle RT, Hodge KM, Cody DB, Sheldon RJ, Kobilka Biochem Biophys Res Commun 2001;281:1331–6.
2353–8. BK, Isfort RJ. Skeletal muscle hypertrophy and anti- 33. Llovera M, Garcı́a-Martı́nez C, Agell N, López-Soriano
7. Guttridge DC, Mayo MW, Madrid LV, Wang CY, atrophy effects of clenbuterol are mediated by the h2- FJ, Argilés JM. Muscle wasting associated with cancer
Baldwin AS, Jr. NF-nB-induced loss of MyoD messenger adrenergic receptor. Muscle Nerve 2002;25:729–34. cachexia is linked to an important activation of the
RNA: possible role in muscle decay and cachexia. 20. Wineski LE, von Deutsch DA, Abukhalaf IK, Pitts SA, ATP-dependent ubiquitin-mediated proteolysis. Int J
Science 2000;289:2363–6. Potter DE, Paulsen DF. Muscle-specific effects of Cancer 1995;61:138–41.
8. Wyke SM, Russell ST, Tisdale MJ. Induction of hindlimb suspension and clenbuterol in mature male 34. Tessitore L, Bonelli G, Baccino FM. Early develop-
proteasome expression in skeletal muscle is attenuated rats. Cells Tissues Organs 2002;171:188–98. ment of protein metabolic perturbations in the liver and
by inhibitors of NF-nB activation. Br J Cancer 2004;91: 21. Yang YT, McElligot MA. Multiple actions of h- skeletal muscle of tumour-bearing rats. Biochem J 1987;
1742–50. adrenergic agonists on skeletal muscle and adipose 241:153–9.
9. Penner G, Gang G, Sun X, Wray C, Hasselgren PO. tissue. Biochem J 1989;261:1–10. 35. Seelaender MC, Nascimento CM, Curi R, Williams JF.
C/EBP DNA-binding activity is upregulated by a 22. Mersmann HJ. Overview of the effects of h-adrener- Studies on the lipid metabolism of Walker 256 tumour-
glucocorticoid-dependent mechanism in septic mus- gic receptor agonists on animal growth including bearing rats during the development of cancer cachexia.
cle. Am J Physiol Regul Integr Comp Physiol 2002;282: mechanisms of action. J Anim Sci 1998;76:160–72. Biochem Mol Biol Int 1996;39:1037–47.
R439–44. 23. Busquets S, Figueras MT, Fuster G, et al. Anticachec- 36. Luquet S, Lopez-Soriano J, Holst D, et al. Roles of
10. Costelli P, Muscaritoli M, Bossola M, et al. Skeletal tic effects of formoterol: a drug for potential treatment peroxisome proliferator-activated receptor y (PPARy) in
muscle wasting in tumor-bearing rats is associated of muscle wasting. Cancer Res 2004;64:6725–31. the control of fatty acid catabolism. A new target for the
with MyoD down-regulation. Int J Oncol 2005;26: 24. Tessitore L, Costelli P, Bonetti G, Baccino FM. Cancer treatment of metabolic syndrome. Biochimie 2004;86:
1663–8. cachexia, malnutrition, and tissue protein turnover in 833–7.
11. Moore-Carrasco R, Garcı́a-Martı́nez C, Busquets S, experimental animals. Arch Biochem Biophys 1993;306: 37. Schuler M, Ali F, Chambon C, et al. PGC1a expression
et al. The AP-1/cjun signaling cascade is involved in 52–8. is controlled in skeletal muscles by PPARh, whose
muscle differentiation: implications in muscle wasting 25. Chomczynski P, Sacchi N. Single-step method of RNA ablation results in fiber-type switching, obesity, and type
during cancer cachexia. FEBS Lett 2006;580:691–6. isolation by acid guanidinium thiocyanate phenol 2 diabetes. Cell Metab 2006;4:407–14.
12. Dressel U, Allen TL, Pippal JB, Rohde PR, Lau P, chloroform extraction. Anal Biochem 1987;162:156–9. 38. Abbot EL, McCormack JG, Reynet C, Hassall DG,
Muscat GE. The peroxisome proliferator-activated 26. Almendro V, Busquets S, Ametller E, et al. Effects of Buchan KW, Yeaman SJ. Diverging regulation of pyruvate
receptor h/y agonist, GW501516, regulates the expres- interleukin-15 on lipid oxidation. Disposal of an oral dehydrogenase kinase isoform gene expression in
sion of genes involved in lipid catabolism and energy [14C]-triolein load. Biochim Biophys Acta 2006;1761: cultured human muscle cells. FEBS J 2005;272:3004–14.
uncoupling in skeletal muscle cells. Mol Endocrinol 37–42. 39. Puigserver P, Rhee J, Lin J, et al. Cytokine stimulation
2003;17:2477–93. 27. Mortensen OH, Frandsen L, Schjerling P, Nishimura of energy expenditure through p38 MAP kinase
13. Chinetti G, Griglio S, Antonucci M, et al. Activation of E, Grunnet N. PGC-1a and PGC-1h have both similar activation of PPARg coactivator-1. Mol Cell 2001;8:
proliferator-activated receptors a and g induces apo- and distinct effects on myofiber switching toward an 971–82.
ptosis of human monocyte-derived macrophages. J Biol oxidative phenotype. Am J Physiol Endocrinol Metab 40. Sanchı́s D, Busquets S, Alvarez B, Ricquier D, López-
Chem 1998;273:25573–80. 2006;291:E807–16. Soriano FJ, Argilés JM. Skeletal muscle UCP2 and UCP3

Cancer Res 2007; 67: (13). July 1, 2007 6518 www.aacrjournals.org

Downloaded from cancerres.aacrjournals.org on July 9, 2018. © 2007 American Association for Cancer
Research.
PPARs and Cancer Cachexia

gene expression in a rat cancer cachexia model. FEBS 44. Planavila A, Rodrı́guez-Calvo R, Jové M, et al. 47. Costelli P, Carbó N, Tessitore L, et al. Tumor
Lett 1998;436:415–8. Peroxisome proliferator-activated receptor h/y activa- necrosis factor-a mediates changes in tissue protein
41. Stevens L, Firinga C, Gohlsch B, Bastide B, Mounier Y, tion inhibits hypertrophy in neonatal rat cardiomyo- turnover in a rat cancer cachexia model. J Clin Invest
Pette D. Effects of unweighting and clenbuterol on cytes. Cardiovasc Res 2005;65:832–41. 1993;92:2783–9.
myosin light and heavy chains in fast and slow muscles 45. Orcutt AL, Cline TR, Mills SE. Influence of the h2- 48. Moller DE, Berger JP. Role of PPARs in the regulation
of rat. Am J Physiol Cell Physiol 2000;279:C1558–63. adrenergic agonist clenbuterol on insulin-stimulated of obesity-related insulin sensitivity and inflammation.
42. Herzig S, Hedrick S, Morantte I, Koo SH, Galimi F, lipogenesis in mouse adipocytes. Domest Anim Endo- Int J Obes Relat Metab Disord 2003;3:S17–21.
Montminy M. CREB controls hepatic lipid metabolism crinol 1989;6:59–69. 49. Jove M, Laguna JC, Vazquez-Carrera M. Agonist-induced
through nuclear hormone receptor PPAR-g. Nature 46. Izeboud CA, Monshouwer M, van Miert AS, Witkamp activation releases peroxisome proliferator-activated re-
2003;426:190–3. RF. The h-adrenoceptor agonist clenbuterol is a potent ceptor h/y from its inhibition by palmitate-induced nuclear
43. Farmer SR. Regulation of PPARg activity during inhibitor of the LPS-induced production of TNF-a and factor-nB in skeletal muscle cells. Biochim Biophys Acta
adipogenesis. Int J Obes 2005;1:S13–6. IL-6 in vitro and in vivo . Inflamm Res 1999;48:497–502. 2005;1734:52–61.

www.aacrjournals.org 6519 Cancer Res 2007; 67: (13). July 1, 2007

Downloaded from cancerres.aacrjournals.org on July 9, 2018. © 2007 American Association for Cancer
Research.
Are Peroxisome Proliferator-Activated Receptors Involved in
Skeletal Muscle Wasting during Experimental Cancer
Cachexia? Role of β2-Adrenergic Agonists
Gemma Fuster, Sílvia Busquets, Elisabet Ametller, et al.

Cancer Res 2007;67:6512-6519.

Updated version Access the most recent version of this article at:
http://cancerres.aacrjournals.org/content/67/13/6512

Cited articles This article cites 48 articles, 7 of which you can access for free at:
http://cancerres.aacrjournals.org/content/67/13/6512.full#ref-list-1

Citing articles This article has been cited by 1 HighWire-hosted articles. Access the articles at:
http://cancerres.aacrjournals.org/content/67/13/6512.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Subscriptions Department at pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, use this link
http://cancerres.aacrjournals.org/content/67/13/6512.
Click on "Request Permissions" which will take you to the Copyright Clearance Center's
(CCC)
Rightslink site.

Downloaded from cancerres.aacrjournals.org on July 9, 2018. © 2007 American Association for Cancer
Research.

You might also like