Aliskiren targets multiple systems to alleviate cancer cachexia

  • Authors:
    • Chaoyi Wang
    • Dunwei Guo
    • Qiang Wang
    • Song You
    • Zhongpeng Qiao
    • Yong Liu
    • Hang Dai
    • Hua Tang
  • View Affiliations

  • Published online on: September 21, 2016     https://doi.org/10.3892/or.2016.5118
  • Pages: 3014-3022
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

To examine the effects of aliskiren, a small-molecule renin inhibitor, on cancer cachexia and to explore the underlying mechanisms. A cancer cachexia model was established by subcutaneously injecting C26 mouse colon carcinoma cells into isogenic BALB/c mice. Aliskiren was administered intragastrically [10 mg/kg body weight (BW)] on day 5 (as a preventive strategy, AP group) or on day 12 (as a therapeutic strategy, AT group) after C26 injection. Mice that received no C26 injection (healthy controls, HC group) or only C26 injection but not aliskiren (cancer, CA group) were used as controls. BW, tumor growth, whole body functions, and survival were monitored daily in half of the mice in each group, whereas serum, tumors, and gastrocnemius muscles were harvested from the other mice after sacrifice on day 20 for further analysis. Aliskiren significantly alleviated multiple cachexia‑associated symptoms, including BW loss, tumor burden, muscle wasting, muscular dysfunction, and shortened survival. On the molecular level, aliskiren antagonized cachexia‑induced activation of the renin‑angiotensin system (RAS), systematic and muscular inflammation, oxidative stress, and autophagy‑lysosome as well as ubiquitin‑proteasome stimulation. In addition, early administration of aliskiren before cachexia development (AP group) resulted in more robust effects in alleviating cachexia or targeting underlying mechanisms than administration after cachexia development (AT group). Aliskiren exhibited potent anti‑cachexia activities. These activities were achieved through the targeting of at least four mechanisms underlying cachexia development: RAS activation, increase in systematic inflammation, upregulation of oxidative stress, and stimulation of autophagy-lysosome pathway (ALP) and ubiquitin-proteasome pathway (UPP).

Introduction

Cancer cachexia is a complicated multifactorial syndrome featuring a progressive loss of skeletal muscle mass (with or without fat loss) that is associated with significant functional impairments (1). Cachexia occurs in ~60% of patients with advanced cancer and up to 80% of those in a terminal stage (2,3). The development of cachexia is divided into three stages: pre-cachexia, cachexia, and refractory cachexia. Cachexia significantly and adversely impacts the patient responses to treatment, quality of life, and survival. Cachexia is responsible for 20–30% of all cancer-related deaths (2). No treatments effectively alleviating cachexia have been developed to date, and thus, it is essential to explore its pathogenesis and underlying mechanisms, and develop therapeutic approaches targeting these mechanisms.

Muscle wasting is a prominent feature of cachexia and is closely associated with increased protein degradation. Studies have identified several mechanisms modulating protein degradation in muscles. One is the renin-angiotensin system (RAS): although most commonly associated with vasoconstriction and the regulation of blood pressure, angiotensin peptides angiotensin I (Ang I) and II are produced within skeletal muscle and directly or indirectly inhibit protein synthesis (4), promote protein degradation (5), stimulate cytokine production (6,7), and generate hydroxyl radicals (•OH) to exacerbate oxidative stress (8,9). Consistent with its functional consequences, the RAS is noted to be activated in conditions involving muscle wasting, including sarcopenia, chronic heart failure, chronic renal failure, and cancer (1013). Our previous study as well as that from Murphy et al (14), demonstrated the significance of the RAS in cancer cachexia (15), corroborating the potential of a RAS inhibitor/antagonist in treating cancer cachexia. A second mechanism is the autophagy-lysosome pathway (ALP) and ubiquitin-proteasome pathway (UPP): both are major pathways responsible for muscular protein degradation (16,17). Examples of genes critical for regulating these two machineries in muscles include Bnip3, Beclin1, and LC3, which are involved in different steps of autophagosome formation, and two muscle-specific ubiquitin E3 ligases, Atrogin1 and MuRF1 (17).

In this study, we examined the efficacy of a novel direct renin inhibitor, aliskiren, on cancer cachexia. Previous studies showed that aliskiren can potently inhibit inflammation, reduce oxidative stress, and protect against tissue damage (1820). However, minimal information is available on its actions in cancer cachexia. To address this issue, we established a cancer cachexia mouse model, assessed the prophylactic as well as therapeutic effects of aliskiren on cancer cachexia, and explored the molecular mechanisms underlying these effects.

Materials and methods

Ethics statement

The experimental protocols involving human samples were established according to the ethical guidelines of the Helsinki Declaration and approved by the Ethics Committee of Chongqing Medical University (Chongqing, China). The experimental protocols involving mice were approved by the Institutional Animal Care and Use Committee (IACUC) of Chongqing Medical University.

Cells and reagents

C26 mouse colon carcinoma cells were provided by the Research Center (the First Affiliated Hospital of Chongqing Medical University, Chongqing, China) and cultured in Dulbecco's Modified Eagle Medium (DMEM)-F12 supplemented with 10% fetal bovine serum (FBS; both from Hyclone, Logan, UT, USA).

Aliskiren was purchased from Novartis (San Diego, CA, USA). The following antibodies were used in this study: anti-Bnip1 (ab38621), anti-LC3 (ab168831; Abcam, Cambridge, MA, USA), anti-MuRF1 (sc-32920), anti-Atrogin1 (sc-33782; Santa Cruz Biotechnology, Santa Cruz, CA, USA), anti-Beclin1 (11306-1-AP), anti-GAPDH (10494-1-AP), and peroxidase-conjugated anti-rabbit IgG (SA00001-2; Proteintech, Wuhan, China). A real-time polymerase chain reaction (PCR) kit was purchased from Takara (Dalian, China), and a western immunoblot kit was purchased from Beyotime (Jiangsu, China).

Experimental animals and cachexia model

Healthy male BALB/c mice between 7 and 9 weeks of age and with a body weight (BW) of 21–26 g were purchased from the Experimental Animal Center (Chongqing Medical University). All mice were housed in a specific-pathogen-free facility at room temperature of 22±1°C on a 12/12-h light/dark cycle with access to food and water ad libitum.

To induce cancer cachexia, C26 cells growing in log phase were detached and resuspended as single cells in PBS at 1×107/ml, and 100 µl C26 cell suspension was injected subcutaneously into the left axilla of each mouse (N=48). In healthy controls (HC group), 100 µl PBS was injected into the mice (n=16). Mice receiving C26 injection were further divided into three groups (n=16/group): i) cancer-only group (CA): mice received PBS intragastrically on days 5 and 12 after C26 injection; ii) aliskiren prevention group (AP): mice received aliskiren (10 mg/kg BW) in PBS intragastrically on day 5 after C26 injection, when tumor nodules were palpable; and iii) aliskiren treatment group (AT): mice received aliskiren (10 mg/kg BW) in PBS intragastrically on day 12 after C26 injection, when cachexia was well-developed. Following C26 cell injection, all mice were monitored daily for the appearance of skin and hair, mental state, BW, and tumor growth until day 20, when eight mice from each group were sacrificed by CO2 euthanasia followed by cervical dislocation for sample collection. The tumor volume was calculated as V = ab2/2, where V is the tumor volume and a and b are the length and width of the tumor, respectively. The other eight from each group were continued to be monitored daily and used to assess whole-body functions (as described below) and survival time. According to the IACUC guidelines of Chongqing Medical University, mice were euthanized by CO2 asphyxiation followed by cervical dislocation when they presented signs of moribund conditions, including decreased response to stimuli, inability to remain upright, abnormal feeding behavior, evidence of severe muscle atrophy, and rough hair coat.

Assessment of grip strength, coordination, and locomotor activity

Whole body strength and coordination was assessed on day 20 after C26 cell injection using a TSE Grip Strength Meter and the rotarod performance test RotaRod Advanced (both from TSE Systems, Germany) as previously described (2123). Briefly, to measure grip strength, the mouse was allowed to hold on firmly to a grip that was mounted to a high-precision force sensor, and its tail was pulled backwards with a continuous movement until the grip was broken. The force value at this point was recorded. Ten measurements were taken for each animal within 2 min, and the maximum values were used for statistical analysis. For rotarod performance test, the mouse was placed on a horizontal rod that rotated from a starting speed of 4 rpm and accelerated at 1 rpm/8 sec until the mouse fell off the rod. The latency-to-fall was recorded. The test was repeated three times within 15 min for each animal, and the longest latency-to-fall was used for statistical analysis.

Locomotor activity was monitored using an infrared monitoring system (WV-CF314LCH; Panasonic, Japan) following the manufacturer's instructions.

Sample collection

On day 20 after C26 inoculation, eight mice from each group were sacrificed, with blood immediately collected from the retro-orbital venous sinus. The blood samples were allowed to sit at 4°C for 1 h, which was followed by centrifugation at 1,006 × g for 10 min at 4°C. The serum supernatant was then collected and stored at −80°C until future use. In addition to the serum sample, the tumor tissue as well as the bilateral gastrocnemius muscles were isolated and weighed. The left gastrocnemius muscle was fixed in 4% paraformaldehyde, and the right one was flash frozen and stored in liquid nitrogen until further use.

Measurement of cross-sectional area of gastrocnemius muscle

After 48 h of fixation in paraformaldehyde, the gastrocnemius muscle was embedded in paraffin, sectioned onto glass slides, and stained with hematoxylin and eosin (H&E) (Beyotime). Three slides for each sample were imaged under an optical microscope (×200 magnification, BX51TF; Olympus, Tokyo, Japan), with five random fields imaged for each slide, and the cross-cut area of the gastrocnemius muscle was measured using Image-Pro Plus 6.0 software (Media Cybernetics, Rockville, MD, USA).

Determination of serum Ang I and II levels and both serum and muscular tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) levels

To prepare muscle lysates, flash frozen muscles were suspended in buffer containing 10 mM HEPES (pH 7.9), 10 mM KCl, 2 mM MgCl2, 0.1 mM EDTA (pH 8.0), 1.0 mM DTT, 10% NP-40, and 0.5 mM phenylmethylsulfonyl fluoride (PMSF), and immediately homogenized on ice. The homogenized muscle tissue was centrifuged at 16,099 × g for 10 min at 4°C, and the supernatant was collected for future use. The levels of Ang I and II in serum and those of TNF-α and IL-6 in serum and in gastrocnemius muscle were determined using enzyme-linked immunosorbent assay (ELISA) kits (R&D Systems, Minneapolis, MN, USA) following the manufacturer's instructions.

Evaluation of oxidative stress in gastrocnemius muscle

To assess the oxidative stress level in gastrocnemius muscles, the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) and the levels of •OH and malondialdehyde (MDA) were determined using the corresponding test kits (Nanjing Jiancheng Biological Engineering Institute, Nanjing, China) following the manufacturer's instructions.

Real-time PCR analysis

Total RNA was extracted from flash frozen gastrocnemius muscles using TRIzol reagent (Takara) according to the manufacturer's instructions and reversed transcribed into cDNA. The real-time PCR reaction was set up using SYBR Premix Ex Taq II kit (Takara) and performed on an ABI Prism 7500 Sequence Detection System (Applied Biosystems, Foster City, CA, USA). The primer sequences used for real-time PCR analysis are listed in Table I. GAPDH was used as the internal control. The quantification of real-time PCR data was performed using the 2−ΔΔCt method, as previously described (24).

Table I

Primer sequences used in real-time polymerase chain reaction (PCR) analysis.

Table I

Primer sequences used in real-time polymerase chain reaction (PCR) analysis.

GeneSense primer (5′→3′)Antisense primer (5′→3′)
Bnip3 5′-GGGTTTTCCCCAAAGGAATA-3′ 5′-TGACCACCCAAGGTAATGGT-3′
LC3 5′-CGGCTTCCTGTACATGGTTT-3′5′-ATGT GGGTGCCTACGTTCTC-3′
Beclin1 5′-ATGGAGGGGTCTAAGGCGTC-3′ 5′-TGGGCTGTGGTAAGTAATGGA-3′
MuRF1 5′-GGAACACGAAGACGAGAAAATC-3′ 5′-TGGCTATTCTCCTTGGTCACTC-3′
Atrogin1 5′-GAAGAGAGCAGTATGGGGTCAC-3′ 5′-CTTGAGGGGAAAGRGAGACG-3′
Caspase-3 5′-TGGGACTGATGAGGAGATGGC-3′ 5′-TGCTGCAAAGGGACTGGATG-3′
GAPDH 5′-GGTGAAGGTCGGTGTGAACG-3′ 5′-CTCGCTCCTGGAAGATGGTG-3′
Western blotting

Total proteins from the flash frozen gastrocnemius muscle samples were extracted using radioimmunoprecipitation assay (RIPA)/PMSF lysis buffer (Beyotime). Equal amounts of total protein from different samples were separated by 10% sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis (PAGE), transferred to polyvinylidene fluoride (PVDF) membrane (Beyotime), probed with primary antibodies at 4°C overnight, followed by incubation with corresponding secondary antibodies. Signal detection and quantification were achieved using the UVP Bioimaging System (UVP, Inc., Upland, CA, USA). The relative protein level was expressed as the ratio of signal density of a target protein to that of the internal control protein (GAPDH).

Statistical analysis

Statistical analysis was performed using SPSS 17.0 software (SPSS, Inc., Chicago, IL, USA). Quantitative data were expressed as means ± standard deviation (SD). Differences between two groups were analyzed using Student-Newman-Keuls q-test, and those among multiple groups were analyzed using one-way analysis of variance (ANOVA). Survival analysis was performed using the log-rank test. P<0.05 was considered statistically significant.

Results

Aliskiren delays cachexia development, reduces tumor burden, and prolongs mouse survival

To examine the effects of aliskiren on cancer cachexia and the underlying mechanisms, we subcutaneously injected C26 colon cancer cells into isogenic BALB/c mice, a well-established experimental model of cancer cachexia (25). On day 5 after C26 cell inoculation, although tumor nodules were palpable through the skin, no significant differences in BW were noticed among the four groups of mice. On day 12, mice in the CA and AT groups showed obvious signs of cachexia, including rough skin, disheveled fur, reduced motility, and dramatic weight loss (P<0.05, compared with mice in the HC group). By day 20, mice in the CA group presented the most dramatic weight loss (P<0.01, compared with mice in the HC group), followed by those in the AT group (P<0.01, compared with mice in the HC group; P<0.05, compared with mice in the CA group), and the least weight loss was observed in mice of the AP group (P<0.01, compared with mice in the CA group; P<0.05, compared with mice in the AT group; Fig. 1A). Consistently, the percentage decrease in tumor-free body mass from day 0 to 20 was most significant in mice in the CA group (32.67%), and this percentage was significantly higher than that in the AT group (21.21%; P<0.05, compared with the CA group) or the AP group (12.42%; P<0.01, compared with the CA group; P<0.05, compared with the AT group; Fig. 1B).

With respect to tumor growth, aliskiren as a treatment agent (as in the AT group) or an early prevention strategy (as in the AP group) led to reductions in both tumor mass and tumor volume (P<0.05, compared with those in the CA group), with the most robust antitumor effects observed in the AP group (P<0.01, compared with mice in the CA group; P<0.05, compared with mice in the AT group; Fig. 1C–E).

Functionally, aliskiren resulted in significantly improved survival in the AT and AP groups (P<0.05, compared with the CA group), and the best survival was observed in the AP group (P<0.05, compared to the CA group; P<0.05, compared to the AT group; Fig. 1F).

Aliskiren improves whole-body strength, mobility and coordination, enhances locomotor activity, and inhibits muscle wasting

Whole-body strength was assessed on day 20 using a grip strength meter and mobility and coordination were assessed using the rotarod test. As shown in Fig. 3A and B, both grip strength and rotarod performance were significantly reduced in the CA group (P<0.01, compared with the HC group). Treatment with aliskiren on day 12 (AT group) improved grip strength and the latency-to-fall during the rotarod test by 21.17 and 16.98%, respectively (P<0.05, compared with the CA group), and prevention using aliskiren on day 5 (AP group) improved these two parameters by 39.06 and 34.56%, respectively (P<0.01, compared with the CA group; P<0.05, compared with the AT group). Similar alterations in locomotor activity also were noticed in different groups; that is, it was dramatically reduced in the CA group (P<0.01, compared with the HC group), but was enhanced following aliskiren interference (P<0.05, comparing the AT with the CA group; P<0.01, comparing the AP and CA groups), and the best locomotor activity among tumor-bearing mice was observed in the AP group (P<0.05, compared with the AT group; Fig. 2C).

On the histological level, C26 inoculation resulted in marked wasting in skeletal muscles, as represented by reductions in gastrocnemius mass by 40.11% and in the cross-cut area of gastrocnemius muscle by 36.23% (P<0.05, compared with the HC group). In contrast, aliskiren increased gastrocnemius mass by 15.47% in the AT group (P<0.05, compared with the CA group) and 41.14% in the AP group (P<0.05, compared with the CA group; P<0.01, compared with the AT group), respectively. It also increased the cross-cut area of gastrocnemius muscle by 24.12% in the AT group (P<0.05, compared with the CA group) and 41.14% in the AP group (P<0.05, compared with the CA group; P<0.01, compared with the AT group; Fig. 2D–G).

Aliskiren inhibits the RAS and controls the upregulation of pro-inflammatory cytokines TNF-α and IL-6 in both serum and skeletal muscles

To evaluate the effects of aliskiren on the RAS, we measured the serum levels of both Ang I and II, as shown in Table II. The development of cachexia in CA mice was associated with marked RAS activation, as demonstrated by the dramatic elevation of both Ang I and II in serum (P<0.01, compared with the HC group). In contrast, treatment with aliskiren, either before (AP group) or after (AT group) the development of cachexia, led to serum upregulation of Ang I and II to a much lower degree, with less upregulation observed in the AP group than in the AT group (P<0.05).

Table II

Changes in Ang I and II levels in serum and TNF-α and IL-6 levels in serum and gastrocnemius of mice in the indicated groups (n=8/group, means ± SD).

Table II

Changes in Ang I and II levels in serum and TNF-α and IL-6 levels in serum and gastrocnemius of mice in the indicated groups (n=8/group, means ± SD).

GroupAng I (ng/l)Ang II (ng/l)Serum (ng/l)
Gastrocnemius (pg/mg)
TNF-αIL-6TNF-αIL-6
HC9.62±1.0833.62±2.4214.18±2.012.65±0.547.24±1.741.27±0.61
CA25.73±1.11b78.95±4.15b 148.42±10.67b91.58±4.32b52.51±4.66b56.13±5.57b
AT14.70±1.07d44.93±3.31d 116.00±16.08d64.27±6.16d22.32±2.63d30.18±3.01d
AP13.35±1.15d37.65±1.83d,e85.85±10.53d,f45.52±8.77d,e15.82±2.46d,e22.95±2.38d,e

a P<0.05,

b P<0.01, compared with the HC group;

c P<0.05,

d P<0.01, compared with the CA group;

e P<0.05,

f P<0.01, compared with the AT group. Ang I, angiotensin I; IL-6, interleukin-6; TNF-α, tumor necrosis factor-α.

In addition, we also measured the serum and muscular levels of pro-inflammatory cytokines TNF-α as well as IL-6 in all four groups. Similar to the changes in serum Ang I and II levels, TNF-α and IL-6 levels in both the serum and gastrocnemius muscle were significantly higher in the CA group than in the HC group (P<0.01). Treatment with aliskiren after cachexia development (AT group) led to reductions in TNF-α and IL-6 in the serum by 21.84 and 29.82%, respectively, and in the muscle by 30.19 and 46.23%, respectively (P<0.05, compared with the CA group). More reductions were detected in the AP group: serum TNF-α and IL-6 by 42.16 and 50.29%, respectively, and muscular TNF-α and IL-6 by 69.87 and 59.11%, respectively (P<0.05, compared with both the CA and AT groups; Table II).

Aliskiren reduces oxidative stress associated with cancer cachexia

Following the development of cachexia, the level of oxidative stress significantly increased, as indicated by the reduced activity of antioxidative enzymes, SOD and GSH-Px, and the upregulated levels of •OH and MDA in gastrocnemius muscle (P<0.05, comparing the CA and HC groups). Following aliskiren treatment, the muscular activities of SOD and GSH-Px increased by 22.91 and 23.92%, respectively, whereas the levels of •OH and MDA decreased by 31.25 and 32.71%, respectively (P<0.05, compared with the CA group). Further reductions in oxidative stress were noted in the AP group, with 56.42 and 37.38% increases in the muscular activities of SOD and GSH-Px, respectively, and 41.86 and 42.93% decreases in •OH and MDA levels, respectively (P<0.01, compared with the CA group; P<0.05, compared with the AT group).

Aliskiren inhibits cachexia-induced ALP and UPP activation

To assess the status of ALP and UPP signaling in cancer cachexia, we examined the expression levels of genes related to these two signaling pathways on both the steady-state mRNA level and protein level. As shown in Fig. 4A, the mRNA levels of autophagy-related genes Bnip3, LC3, and Beclin1, as well as ubiquitin-proteasome-related genes MuRF1 and Atrogin1 in gastrocnemius muscle were significantly upregulated in mice of the CA group compared with these levels in HC mice (P<0.01). Upregulation of these genes was dramatically inhibited in the AT and AP groups, with more robust inhibition achieved in the AP group (P<0.05, compared with the AT group). Similar alterations were also observed in the protein levels of these proteins (Fig. 4B–F). In contrast to these changes, the steady-state mRNA level of the apoptosis-related gene caspase-3 was not markedly altered in tumor-bearing groups (Fig. 4A).

Discussion

Cachexia is a common complication in patients with malignant tumors and is characterized by increased catabolism, reduced anabolism, and excessive energy expenditure, which leads to progressive multi-organ failure and death. Despite extensive research on cancer cachexia, effective and satisfactory preventive and therapeutic approaches have yet to be developed. Given the significance of the RAS in cancer cachexia, we assessed the effects of a direct renin inhibitor, aliskiren, in this study. Using a mouse model of cancer cachexia, we showed that aliskiren effectively alleviated multiple symptoms associated with cachexia, including weight loss, tumor growth, and muscle wasting. Consequently, aliskiren treatments significantly improved whole-body functions and prolonged mouse survival. Mechanistically, aliskiren antagonizes multiple mechanisms underlying cachexia development, including RAS activation, upregulated systematic inflammation, increased oxidative stress, and enhanced ALP and UPP signaling.

Progressive skeletal muscle wasting is central for cachexia development and is mostly correlated with functional defects in these muscles, which increases the risk of postoperative complications, lowers quality of life, and reduces patient survival (26). Consistently, we showed that following cachexia development in C26 cell-inoculated mice, wasting of skeletal muscles, as demonstrated by loss of gastrocnemius mass and its cross-cut area, was associated with reduced grip strength, whole-body coordination, and locomotor activity. The administration of aliskiren as a therapeutic agent and more robustly as a preventive agent, significantly ameliorated muscle wasting and skeletal muscle functions, implying its prophylactic and therapeutic potential for cancer cachexia.

Chronic inflammation is an important feature of cancer cachexia. The inflammatory cytokines TNF-α and IL-6 play critical pathogenic roles in the development of cancer cachexia via multiple mechanisms, inducing anorexia, promoting muscular protein degradation, inhibiting protein synthesis, and enhancing energy expenditure (2729). Recent studies demonstrated that aliskiren achieves its anti-inflammatory property by inhibiting the RAS and reducing the release of inflammatory mediators (18,20). Consistent with these results, we showed that administration of aliskiren as a preventive or therapeutic agent significantly reduced the levels of TNF-α and IL-6 in both serum and skeletal muscle, suggesting anti-inflammation as a second mechanism for the anti-cachexia activity of aliskiren.

Oxidative stress is another important mechanism contributing to the development of anorexia and loss of skeletal muscles in cancer patients. Several causes have been linked to the pathogenesis of oxidative stress in cancer cachexia, including: i) anorexia reduces the intake of antioxidants; ii) alterations in metabolism reduces the synthesis of reducing agents; iii) upregulation of pro-inflammatory cytokines, such as TNF-α and IL-6, increases the production of reactive oxygen species (ROS); and ii) some anticancer medicines such as alkylating agents boost ROS generation (30). Of the endogenous antioxidants, SOD and GSH-Px are two important enzymes. Their activities reflect the body's capability to clear free radicals and protect against oxidative damage. MDA results from lipid degradation by ROS, and together with •OH, indicates the level of oxidative stress in an organism. In this study, we showed that cachexia led to significant downregulation of SOD and GSH-Px but upregulation of MDA and •OH in skeletal muscles, indicating an increase in oxidative stress. Aliskiren administration significantly reduced the level of oxidative stress in the mice by enhancing the activities of SOD and GSH-Px while reducing the levels of MDA and •OH. Consistently, Patel et al (19) and Thomas et al (31) demonstrated that aliskiren can inhibit oxidative stress by reducing the generation of free radicals. Therefore, targeting oxidative stress provides a third mechanism underlying the anti-cachexia activity of aliskiren.

Protein degradation in skeletal muscles under cancer cachexia is achieved mainly through two signaling pathways, ALP and UPP (32,33). Chronic inflammation and oxidative stress associated with cancer cachexia could upregulate autophagy-related genes and activate ALP (34,35). In this study, we found that cachexia development in mice led to significant upregulation of the ALP-related genes Bnip3, LC3 (increased LC3-II/LC3-I ratio), and Beclin1, indicating ALP activation. Following aliskiren administration, expression of these genes was markedly downregulated, implying reduced ALP activity. This finding is consistent with those of Weng et al and Zhang et al who reported that aliskiren inhibits ALP activity (36,37). Atrogin-1 and MuRF1 are two muscle-specific E3 ligases that critically control UPP activity, and the inhibition of these two enzymes has been demonstrated to alleviate the loss of skeletal muscles and ameliorate cancer cachexia (38,39). Here we showed that aliskiren reduced the mRNA expression levels of these two enzymes and thus improved cancer cachexia.

In summary, aliskiren is highly effective at ameliorating multiple symptoms associated with cancer cachexia. It inhibits weight loss, wasting of skeletal muscles, and whole-body dysfunctions, and results in improved locomotor activities as well as prolonged survival. On the molecular level, aliksiren targets multiple mechanisms underlying the pathogenesis of cancer cachexia, including RAS activation, chronic inflammation, increased oxidative stress, and stimulation of ALP and UPP signaling. Early administration of aliskiren as a preventive agent (before the development of cachexia) offers more robust anti-cachexia activities than later administration as a therapeutic agent (after the development of cachexia). This study proves the efficacy of aliskiren as an anti-cachexia agent in an experimental animal model, and the results support further testing of this agent in cancer patients in clinical settings.

Abbreviations:

RAS

renin-angiotensin system

ALP

autophagy-lysosome pathway

UPP

ubiquitin-proteasome pathway

H&E

hematoxylin and eosin

SOD

superoxide dismutase

GSH-Px

glutathione peroxidase

MDA

malondialdehyde

RIPA

radioimmunoprecipitation assay

PMSF

phenylmethylsulfonyl fluoride

SDS

sodium dodecyl sulfate

PAGE

polyacrylamide gel electrophoresis

PVDF

polyvinylidene fluoride

References

1 

Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL, Jatoi A, Loprinzi C, MacDonald N, Mantovani G, et al: Definition and classification of cancer cachexia: An international consensus. Lancet Oncol. 12:489–495. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Tan BH and Fearon KC: Cachexia: Prevalence and impact in medicine. Curr Opin Clin Nutr Metab Care. 11:400–407. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Tisdale MJ: Mechanisms of cancer cachexia. Physiol Rev. 89:381–410. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Russell ST, Sanders PM and Tisdale MJ: Angiotensin II directly inhibits protein synthesis in murine myotubes. Cancer Lett. 231:290–294. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Sanders PM, Russell ST and Tisdale MJ: Angiotensin II directly induces muscle protein catabolism through the ubiquitin-proteasome proteolytic pathway and may play a role in cancer cachexia. Br J Cancer. 93:425–434. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Ruiz-Ortega M, Lorenzo O, Suzuki Y, Rupérez M and Egido J: Proinflammatory actions of angiotensins. Curr Opin Nephrol Hypertens. 10:321–329. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Zhang L, Du J, Hu Z, Han G, Delafontaine P, Garcia G and Mitch WE: IL-6 and serum amyloid A synergy mediates angiotensin II-induced muscle wasting. J Am Soc Nephrol. 20:604–612. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Russell ST, Eley H and Tisdale MJ: Role of reactive oxygen species in protein degradation in murine myotubes induced by proteolysis-inducing factor and angiotensin II. Cell Signal. 19:1797–1806. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Sukhanov S, Semprun-Prieto L, Yoshida T, Michael Tabony A, Higashi Y, Galvez S and Delafontaine P: Angiotensin II, oxidative stress and skeletal muscle wasting. Am J Med Sci. 342:143–147. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Dalla Libera L, Ravara B, Angelini A, Rossini K, Sandri M, Thiene G, Battista Ambrosio G and Vescovo G: Beneficial effects on skeletal muscle of the angiotensin II type 1 receptor blocker irbesartan in experimental heart failure. Circulation. 103:2195–2200. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Neo JH, Ager EI, Angus PW, Zhu J, Herath CB and Christophi C: Changes in the renin angiotensin system during the development of colorectal cancer liver metastases. BMC Cancer. 10:1342010. View Article : Google Scholar : PubMed/NCBI

12 

Sun G, Haginoya K, Dai H, Chiba Y, Uematsu M, Hino-Fukuyo N, Onuma A, Iinuma K and Tsuchiya S: Intramuscular renin-angio-tensin system is activated in human muscular dystrophy. J Neurol Sci. 280:40–48. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Wang XH and Mitch WE: Mechanisms of muscle wasting in chronic kidney disease. Nat Rev Nephrol. 10:504–516. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Murphy KT, Chee A, Trieu J, Naim T and Lynch GS: Inhibition of the renin-angiotensin system improves physiological outcomes in mice with mild or severe cancer cachexia. Int J Cancer. 133:1234–1246. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Wang Q, Li C, Peng X, Kang Q, Deng D, Zhang L, Zheng Y, Wang C, Qiao Z, Guo D, et al: Combined treatment of carfilzomib and z-VAD-fmk inhibits skeletal proteolysis and apoptosis and ameliorates cancer cachexia. Med Oncol. 32:1002015. View Article : Google Scholar : PubMed/NCBI

16 

Acharyya S and Guttridge DC: Cancer cachexia signaling pathways continue to emerge yet much still points to the proteasome. Clin Cancer Res. 13:1356–1361. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Penna F, Baccino FM and Costelli P: Coming back: Autophagy in cachexia. Curr Opin Clin Nutr Metab Care. 17:241–246. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Del Fiorentino A, Cianchetti S, Celi A and Pedrinelli R: Aliskiren, a renin inhibitor, downregulates TNF-α-induced tissue factor expression in HUVECS. J Renin Angiotensin Aldosterone Syst. 11:243–247. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Patel RB, Prajapati KD, Sonara BM, Sharma MM, Patel HM, Pawar VD and Jain MR: Ameliorative potential of aliskiren in experimental colitis in mice. Eur J Pharmacol. 737:70–76. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Schmerbach K, Pfab T, Zhao Y, Culman J, Mueller S, Villringer A, Muller DN, Hocher B, Unger T and Thoene-Reineke C: Effects of aliskiren on stroke in rats expressing human renin and angiotensinogen genes. PLoS One. 5:e150522010. View Article : Google Scholar : PubMed/NCBI

21 

De Luca A, Pierno S, Liantonio A, Cetrone M, Camerino C, Fraysse B, Mirabella M, Servidei S, Rüegg UT and Conte Camerino D: Enhanced dystrophic progression in mdx mice by exercise and beneficial effects of taurine and insulin-like growth factor-1. J Pharmacol Exp Ther. 304:453–463. 2003. View Article : Google Scholar

22 

Ham DJ, Murphy KT, Chee A, Lynch GS and Koopman R: Glycine administration attenuates skeletal muscle wasting in a mouse model of cancer cachexia. Clin Nutr. 33:448–458. 2014. View Article : Google Scholar

23 

Murphy KT, Chee A, Trieu J, Naim T and Lynch GS: Importance of functional and metabolic impairments in the characterization of the C-26 murine model of cancer cachexia. Dis Model Mech. 5:533–545. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Schmittgen TD and Livak KJ: Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 3:1101–1108. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Tanaka Y, Eda H, Tanaka T, Udagawa T, Ishikawa T, Horii I, Ishitsuka H, Kataoka T and Taguchi T: Experimental cancer cachexia induced by transplantable colon 26 adenocarcinoma in mice. Cancer Res. 50:2290–2295. 1990.PubMed/NCBI

26 

Guo CB, Zhang W, Ma DQ, Zhang KH and Huang JQ: Hand grip strength: An indicator of nutritional state and the mix of postoperative complications in patients with oral and maxillofacial cancers. Br J Oral Maxillofac Surg. 34:325–327. 1996. View Article : Google Scholar : PubMed/NCBI

27 

Carson JA and Baltgalvis KA: Interleukin 6 as a key regulator of muscle mass during cachexia. Exerc Sport Sci Rev. 38:168–176. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Deans C and Wigmore SJ: Systemic inflammation, cachexia and prognosis in patients with cancer. Curr Opin Clin Nutr Metab Care. 8:265–269. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Tsoli M and Robertson G: Cancer cachexia: Malignant inflammation, tumorkines, and metabolic mayhem. Trends Endocrinol Metab. 24:174–183. 2013. View Article : Google Scholar

30 

Vaughan VC, Martin P and Lewandowski PA: Cancer cachexia: Impact, mechanisms and emerging treatments. J Cachexia Sarcopenia Muscle. 4:95–109. 2013. View Article : Google Scholar :

31 

Thomas CM, Yong QC, Seqqat R, Chandel N, Feldman DL, Baker KM and Kumar R: Direct renin inhibition prevents cardiac dysfunction in a diabetic mouse model: Comparison with an angiotensin receptor antagonist and angiotensin-converting enzyme inhibitor. Clin Sci (Lond). 124:529–541. 2013. View Article : Google Scholar

32 

Penna F, Costamagna D, Pin F, Camperi A, Fanzani A, Chiarpotto EM, Cavallini G, Bonelli G, Baccino FM and Costelli P: Autophagic degradation contributes to muscle wasting in cancer cachexia. Am J Pathol. 182:1367–1378. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Tardif N, Klaude M, Lundell L, Thorell A and Rooyackers O: Autophagic-lysosomal pathway is the main proteolytic system modified in the skeletal muscle of esophageal cancer patients. Am J Clin Nutr. 98:1485–1492. 2013. View Article : Google Scholar : PubMed/NCBI

34 

McClung JM, Judge AR, Powers SK and Yan Z: p38 MAPK links oxidative stress to autophagy-related gene expression in cachectic muscle wasting. Am J Physiol Cell Physiol. 298:C542–C549. 2010. View Article : Google Scholar :

35 

Rahman M, Mofarrahi M, Kristof AS, Nkengfac B, Harel S and Hussain SN: Reactive oxygen species regulation of autophagy in skeletal muscles. Antioxid Redox Signal. 20:443–459. 2014. View Article : Google Scholar

36 

Weng LQ, Zhang WB, Ye Y, Yin PP, Yuan J, Wang XX, Kang L, Jiang SS, You JY, Wu J, et al: Aliskiren ameliorates pressure overload-induced heart hypertrophy and fibrosis in mice. Acta Pharmacol Sin. 35:1005–1014. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Zhang W, Zhao G, Hu X, Wang M, Li H, Ye Y, Du Q, Yao J, Bao Z, Hong W, et al: Aliskiren-attenuated myocardium apoptosis via regulation of autophagy and connexin-43 in aged spontaneously hypertensive rats. J Cell Mol Med. 18:1247–1256. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Burckart K, Beca S, Urban RJ and Sheffield-Moore M: Pathogenesis of muscle wasting in cancer cachexia: Targeted anabolic and anticatabolic therapies. Curr Opin Clin Nutr Metab Care. 13:410–416. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Yuan L, Han J, Meng Q, Xi Q, Zhuang Q, Jiang Y, Han Y, Zhang B, Fang J and Wu G: Muscle-specific E3 ubiquitin ligases are involved in muscle atrophy of cancer cachexia: An in vitro and in vivo study. Oncol Rep. 33:2261–2268. 2015.PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 36 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang C, Guo D, Wang Q, You S, Qiao Z, Liu Y, Dai H and Tang H: Aliskiren targets multiple systems to alleviate cancer cachexia. Oncol Rep 36: 3014-3022, 2016
APA
Wang, C., Guo, D., Wang, Q., You, S., Qiao, Z., Liu, Y. ... Tang, H. (2016). Aliskiren targets multiple systems to alleviate cancer cachexia. Oncology Reports, 36, 3014-3022. https://doi.org/10.3892/or.2016.5118
MLA
Wang, C., Guo, D., Wang, Q., You, S., Qiao, Z., Liu, Y., Dai, H., Tang, H."Aliskiren targets multiple systems to alleviate cancer cachexia". Oncology Reports 36.5 (2016): 3014-3022.
Chicago
Wang, C., Guo, D., Wang, Q., You, S., Qiao, Z., Liu, Y., Dai, H., Tang, H."Aliskiren targets multiple systems to alleviate cancer cachexia". Oncology Reports 36, no. 5 (2016): 3014-3022. https://doi.org/10.3892/or.2016.5118