Open Access

Activin A promotes cell proliferation, invasion and migration and predicts poor prognosis in patients with colorectal cancer

  • Authors:
    • Nobuya Daitoku
    • Yuji Miyamoto
    • Yukiharu Hiyoshi
    • Ryuma Tokunaga
    • Yuki Sakamoto
    • Hiroshi Sawayama
    • Takatsugu Ishimoto
    • Yoshifumi Baba
    • Naoya Yoshida
    • Hideo Baba
  • View Affiliations

  • Published online on: April 20, 2022     https://doi.org/10.3892/or.2022.8318
  • Article Number: 107
  • Copyright: © Daitoku et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Activin A is a member of the transforming growth factor‑β superfamily of cytokines and displays various pathophysiological activities, including regulation of muscle catabolism and atrophy. Activin A expression is upregulated in several human cancer types and in certain pathologies, its expression is associated with poor prognosis. In the present study, activin A expression was assessed in colorectal cancer (CRC) tissue specimens from 157 patients with primary CRC and the relationship between activin A levels and clinicopathological characteristics, including skeletal muscle mass, and prognosis, was determined. Furthermore, the effects of knockdown of endogenous or exposure to exogenous activin A on the malignant behavior of human CRC cell lines were investigated in vitro. The results indicated that activin A mRNA was significantly upregulated in CRC tumor tissues compared with normal intestinal epithelium. High activin A expression was significantly associated with shorter cancer‑specific survival (P=0.047) and overall survival (P=0.014). According to a multivariate analysis, tumor activin A levels were an independent prognostic factor for overall survival (P=0.001). However, activin A mRNA levels were not associated with the skeletal muscle index. The in vitro experiments demonstrated that exposure to exogenous activin A increased the proliferation, invasion and migration of CRC cell lines, whereas knockdown of endogenous activin A had the opposite effects. In conclusion, activin A is an autocrine and paracrine regulator of CRC cell proliferation and high tumor expression of activin A is associated with poor prognosis in patients with CRC.

Introduction

Colorectal cancer (CRC) is currently the third most common cause of cancer-related mortality in the economically developed world and is on track to increase in ranking in the coming decades (1). Surgical resection in combination with systemic chemotherapy offers the only hope of cure or long-term survival for patients with CRC. However, the disease recurs in ~30% of patients and better treatment options are required to improve prognosis (2). Only a small number of specific diagnostic or therapeutic tools are currently available, to a large part due to the currently limited understanding of the molecular pathogenesis of the disease. Although certain molecular targeted therapies have proven efficacious in CRC (37), there is an urgent requirement to identify novel therapeutic targets. Furthermore, as CRC has a high relapse rate even early after radical resection, there is a requirement to identify additional biomarkers that may complement those currently available to predict early postoperative recurrence and poor prognosis for patients with CRC (3,5,6,8).

Previous studies by our group reported that sarcopenia is an independent unfavorable prognostic factor after curative resection in patients with CRC (9,10). Sarcopenia, defined as a decrease in muscle mass associated with aging and disease, is common in cancer patients (11), and other studies also indicated that it is a poor prognostic factor for various types of cancer (10,1214). Sarcopenia is associated with decreased survival in patients with CRC undergoing curative resection (9), suggesting that understanding the molecular events underlying skeletal muscle degradation may identify potential novel therapeutic targets for these patients.

One molecule associated with the regulation of skeletal muscle mass is activin A, a member of the transforming growth factor-β (TGF-β) family of proteins. In addition to promoting skeletal muscle degradation and atrophy, activin A displays an array of biological activities (15). Under physiological conditions, activin A exerts its effects on cells by binding to type II receptors, which induces its dimerization with type I receptors. Once engaged, the activated type I receptor complex, which has serine/threonine kinase activity, phosphorylates SMAD2/3 and recruits SMAD4, the main signal transducers of the TGF-β family receptors. The SMAD complex then translocates to the nucleus where it promotes transcription of a panel of genes involved in the regulation of cell development and proliferation, including muscle catabolism (16).

Activin A is expressed and secreted by a number of human cancer cell lines (17) and its overexpression has been associated with poor prognosis in various malignant tumor types, including esophageal adenocarcinoma, lung cancer and gastric cancer (1822). In the present study, activin A expression was determined in CRC tissues and its association with skeletal muscle mass and its prognostic significance were examined. In addition, the mechanisms underlying the involvement of activin A in CRC were explored in vitro.

Materials and methods

Patients and tissue samples

The study population consisted of 157 patients with CRC who underwent surgical resection at the Department of Gastroenterological Surgery, Kumamoto University Hospital (Kumamoto, Japan) between January 2008 and December 2012. The mean observation period for the cohort was 57 months (range, 1–91 months). The clinical characteristics of the 157 patients are summarized in Table I. This study included 83 males and 74 females ranging in age from 34 to 86 years. The patients underwent imaging examination, such as colonoscopy and enhanced computed tomography, for CRC diagnosis and staging prior to surgery. The diagnosis was pathologically confirmed using biopsy specimens. Patients who had received preoperative chemotherapy or emergency surgery were excluded. CRC tissue or paired normal epithelial tissue was obtained at the time of surgical resection, snap-frozen and stored at −80°C until use. The present retrospective, non-interventional, observational study was approved by the institutional ethics committee of Kumamoto University Hospital (14 June 2019/approval no. 1047) and performed in accordance with the Declaration of Helsinki from 1975.

Table I.

Characteristics of the patients (n=157).

Table I.

Characteristics of the patients (n=157).

FactorValue
Age, years64 (34–86)
Sex
  Male83
  Female74
Body mass index, kg/m222.6 (15.7-34.2)
SMI, cm2/m247.0 (28.4-85.0)
Location
  Colon113
  Rectum44
Tumor stage
  T124
  T228
  T373
  T432
Lymph node metastasis
  Present59
  Absent98
Lymphatic invasion
  Present51
  Absent106
Venous invasion
  Present67
  Absent90
pStage
  I33
  II49
  III39
  IV36
CEA, ng/ml24.7 (0.6-18021)
CA19-9, U/ml39.7 (0.6-34708)

[i] Values are expressed as median (range) or n. SMI, skeletal muscle index; CEA, carcinoembryonic antigen; CA19-9, carbohydrate antigen 19-9.

Validation analysis in The Cancer Genome Atlas (TCGA) database

To validate the association between activin A expression and prognosis for patients with CRC, the activin expression data and related clinical information of patients with CRC were obtained from the TCGA database (http://www.cbioportal.org). The patients with CRC in the TCGA dataset were divided into two groups according to the median activin A expression. The cumulative overall survival (OS) rate of the patients was determined using Kaplan-Meier survival analysis with a log-rank test.

Cell lines and cell culture

The human CRC cell lines LoVo and SW480 were purchased from RIKEN Bioresource Center Cell Bank and the Japanese Collection of Research Bioresource Cell Bank, respectively. LoVo and SW480 cells were cultured in Ham's and RPMI media (both from Wako Pure Chemical Industries, Ltd.), respectively, supplemented with 10% fetal bovine serum (Mediatech, Inc.). Cells were cultured at 37°C in a humidified atmosphere with 5% CO2 and were confirmed to be negative for mycoplasma infection prior to use.

Measurement of skeletal muscle area

The skeletal muscle area was retrospectively measured on preoperative computed tomography scans at the third lumbar vertebra (L3) level in the inferior direction with the patient in the supine position. In brief, a three-dimensional image analysis system (Volume Analyzer SYNAPSE VINCENT; Fujifilm Medical) was used to measure pixels using a window width of −30 to 150 HU to delineate the muscle compartments and compute the cross-sectional area of each in centimeters squared (cm2). The cross-sectional area of the muscle (cm2) at the L3 level computed from each image was normalized by the square of the height (m2) to obtain the skeletal muscle index (SMI) expressed in cm2/m2 (9,10).

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from frozen tissue samples or CRC cell lines using TRIzol (Invitrogen; Thermo Fisher Scientific, Inc.) and the concentration of purified RNA was measured by comparing absorbance at 260 nm (A260) and A280 using a Nanodrop® 2000 spectrophotometer (Thermo Fisher Scientific, Inc.). cDNA was generated from total RNA using a ReverTra Ace qPCR RT kit (Toyobo Life Science) according to the manufacturer's protocol and was subsequently used as a PCR template. Real-time q-PCR was performed as described previously (21). mRNA levels were measured in technical duplicates and the relative level of activin A mRNA was calculated as the fold change relative to β-actin (ACTB) mRNA. The samples were quantified using the 2−ΔΔCq method (23). The primers used were as follows: Activin A (INHBA) forward, 5′-CCTCGGAGATCATCACGTTT-3′ and reverse, 5′-CCCTTTAAGCCCACTTCCTC-3′; and ACTB forward, 5′-ATTGGCAATGAGCGGTTC-3′ and reverse, 5′-CGTGGATGCCACAGGACT-3′.

Immunohistochemical (IHC) staining

CRC and normal epithelial samples were formalin-fixed and paraffin-embedded. Blocks were cut into 3-µm sections, which were deparaffinized and rehydrated. Activin A antigen was retrieved by autoclaving in a pH 9 buffer solution for 15 min. Subsequently, the sections were incubated overnight at 4°C with goat anti-activin A antibody (1:100 dilution; cat. no. A1594; MilliporeSigma). The sections were washed in PBS and incubated with horseradish peroxidase-conjugated mouse anti-goat secondary antibody (1:50 dilution; cat. no. K8000; EnVision goat; Dako; Agilent Technologies, Inc.) at room temperature for 30 min. Color development was achieved by the addition of 3,3′-diaminobenzidine [Wako Tablet; cat. no. 040-27001 (5 mg); Dako; Agilent Technologies, Inc.] followed by counterstaining with hematoxylin.

Cell transfection

A total of two activin A-specific small interfering RNAs (siRNAs; siActivin A #1 and #2; Silencer Select s7434 and s7436; Thermo Fisher Scientific, Inc.) and a negative control siRNA (siCtrl, Stealth RNAi; Invitrogen; Thermo Fisher Scientific, Inc.) were employed. Pilot experiments were performed to determine the optimal siRNA concentration (10 µM) for inhibition of activin A expression to <30% of the levels in siCtrl-transfected cells. Cells were seeded in 6-well plates at a density of 105 cells/well in 2.5 ml medium and incubated for 24 h. The cells were then transfected with 10 µM siActivin A or siCtrl using Lipofectamine® RNAiMAX Transfection Reagent (Thermo Fisher Scientific, Inc.) in accordance with the manufacturer's protocol. After 48 h of transfection, the supernatant was removed, the cells were washed with PBS and the experiments were performed.

Cell proliferation assay

Cell proliferation was measured using a Cell Counting Kit-8 (CCK-8; Dojindo Molecular Technologies, Inc.) according to the manufacturer's protocol. LoVo and SW480 cells were seeded in 96-well plates at a density of 3.0×103 cells/well in 100 µl medium and incubated overnight at 37°C. Aliquots of 10 µl/well CCK-8 solution were then added to the cells after 0, 24, 48, 72 or 96 h of incubation and the plates were incubated for an additional 90 min. The absorbance at 450 nm was then read using a microplate reader (SPECTRAmax PLUS 384 microplate spectrophotometer; Scientific Equipment Source). Each experiment was performed in triplicate.

Invasion assay

Cell invasion was measured assay using BioCoat Matrigel invasion chambers (24-well plates, 8-µm pore size; BD Biosciences) according to the manufacturer's protocol. In brief, LoVo and SW480 cells were resuspended in medium at a concentration of 105 cells/ml and 500 µl of the cell suspension was placed in the upper chambers. The same medium supplemented with 10% fetal bovine serum (FBS; Mediatech) was placed in the lower chamber and the plates were incubated for 22 h. The cells on the upper surface of the membrane were then removed using a cotton swab and the cells on the lower surface were fixed with 100% ice-cold methanol for 2 min, followed by staining with toluidine blue for 2 min at room temperature. The membrane was rinsed with water and examined using a microscope (MRP-3001; R&D Systems, Inc.). The number of invaded cells was counted in five microscopic fields per membrane (magnification, ×40).

Migration assay

Migration was measured using 6-well plates coated with 200 µl/well Matrigel® (BD Biosciences). LoVo and SW480 cells were resuspended at a concentration of 4.0×104 cells/ml in each medium, plated at 200 µl/well and allowed to adhere for 12 h. The plates were then imaged with a KEYENCE BZ-X700 all-in-one fluorescence microscope equipped with a CO2 and temperature-controlled chamber and time-lapse tracking system (Keyence Corporation). Phase contrast images were acquired every 10 min for 24 h and converted to video files using a BZ-X Analyzer (Keyence Corporation). Cell migration was analyzed using video editing analysis software VW-H2MA (Keyence Corporation) and the tracking data were processed using Excel 2010 (Microsoft Corporation) to generate xy coordinate plots and allow measurement of the distance moved. Migration distance was calculated by randomly selecting three cells in each well, tracking their movement for 15 sec and plotting the average value (n=3) of the distances moved on a graph.

Statistical analysis

Continuous variables are expressed as the median and range. Continuous and categorical variables were compared using the Mann-Whitney U-test and χ2 test, respectively. Survival analyses were performed using the Kaplan-Meier method with the log-rank test. The correlation between activin A mRNA levels and SMI was assessed by calculating Spearman's rank correlation coefficient ρ. OS was calculated as the duration from the date of surgery until death or the last follow-up. Cancer-specific survival (CSS) was calculated from the time of diagnosis to the time of death from any cancer or last follow-up. Variables with significance at P<0.05 in the univariate analysis were included in multivariate analysis using stepwise backward elimination procedures. The Cox proportional hazards model for multivariate analysis was used. All statistical analyses were performed using JMP version 13.1 (SAS Institute, Inc.). All P-values were two-sided and P<0.05 was considered to indicate statistical significance. The term ‘prognostic marker’ is used according to the REMARK guidelines (24).

Results

Associations between activin A expression in CRC tissues and clinicopathological characteristics

To determine whether the expression level of activin A is elevated in CRC, RT-qPCR analysis of 157 matched pairs of CRC and normal epithelial tissue samples was performed. Activin A mRNA expression was significantly higher in CRC tissues than in normal epithelia (P<0.001; Fig. 1). To assess the associations between activin A mRNA levels and clinicopathological factors, patients were assigned to high (n=78) and low (n=79) activin A expression groups using the median value as the cut-off. However, none of the clinicopathological factors examined, including tumor location and metastasis/invasion status, was significantly associated with activin A mRNA levels in tumor tissues (Table II).

Table II.

Patients' characteristics and clinicopathological factors in patients with colorectal cancer according to activin A expression.

Table II.

Patients' characteristics and clinicopathological factors in patients with colorectal cancer according to activin A expression.

Activin A mRNA expression

FactorLow (n=78)High (n=79)P-value
Age ≥75 years25 (32)30 (38)0.585
Female sex39 (50)35 (44)0.162
Body mass index ≥25.0 kg/m219 (24)19 (24)0.933
Location in rectum13 (17)29 (37)0.055
Tumor stage T3-T455 (71)60 (76)0.221
Lymph node metastasis29 (37)30 (38)0.778
Lymphatic invasion26 (33)25 (32)0.502
Venous invasion30 (38)37 (47)0.425
CEA >3.4 ng/ml24 (31)28 (35)0.440
CA19-9 >37.0 U/ml12 (15)15 (19)0.590

[i] Values are expressed as n (%). CEA, carcinoembryonic antigen; CA19-9, carbohydrate antigen 19-9.

Correlation between tumor expression of activin A and SMI

Next, the association between activin A mRNA expression and the SMI was assessed using Spearman's rank correlation analysis. As presented in Fig. 2, there were no significant correlations between activin A mRNA expression in CRC tissues and the SMI for the full patient cohort (n=157, ρ=0.037, P=0.651), males (n=93, ρ=0.083, P=0.938) or females (n=64, ρ=0.189, P=0.141). Thus, the elevated expression of activin A in CRC tumors appeared to be unrelated to the SMI.

Association between activin A expression and patient survival

Kaplan-Meier curves were generated to assess the OS and CSS of patients with CRC according to activin A tumor expression levels (Fig. 3). It was indicated that patients with high tumor expression of activin A had significantly poorer OS (P=0.014) and CSS (P=0.047, log-rank test) than patients with low tumor expression. Next, factors associated with poor OS were evaluated by univariate and multivariate Cox regression analyses. Univariate analysis revealed that an age of ≥75 years, tumor stage, lymph node metastasis, CA19-9 level >37 U/l and high activin A expression were significantly associated with poor OS (Table III) and multivariate analysis demonstrated that an age of ≥75 years [hazard ratio (HR)=4.678, P=0.009], lymph node metastasis (HR=3.372, P=0.009), CA19-9 level >37 (HR=3.591, P=0.015) and high activin A expression (HR=4.287, P=0.001) were independent risk factors for poor OS (Table III). To validate the present results, the association of activin A mRNA levels with survival of 443 patients with CRC was determined using a dataset from the TCGA database. Kaplan-Meier analysis confirmed that high activin A expression (n=333) was significantly associated with poor OS (P=0.039; Fig. S1).

Table III.

Univariate and multivariate analyses of factors influencing overall survival in colorectal cancer.

Table III.

Univariate and multivariate analyses of factors influencing overall survival in colorectal cancer.

Univariate analysisMultivariate analysis


FactorHR95%CIP-valueHR95%CIP-value
Age ≥75 years1.8011.108-2.8980.0184.6782.018-11.150.009
Female sex1.1850.733-1.9020.485
Body mass index ≥25.0 kg/m20.5640.271-1.0520.073
Location in rectum0.9330.552-1.5290.788
Tumor stage T3-T42.9491.218-9.7040.0142.7110.839-12.120.100
Lymph node metastasis2.8351.754-4.673<0.0013.3721.344-9.0280.009
CEA >3.4 ng/ml1.0050.624-1.6140.982
CA19-9 >37.0 U/ml2.0341.166-3.4080.0143.5911.313-9.0480.015
Activin A high2.5431.157-5.9820.0204.2871.776-11.140.001

[i] HR, hazard ratio; CEA, carcinoembryonic antigen; CA19-9, carbohydrate antigen 19-9.

Proliferation, invasion and migration of human CRC cell lines exposed to activin A in vitro

Next, in vitro experiments were performed to clarify the biological activities of activin A in CRC cells. The subcellular pattern of activin A expression in CRC cells was determined by IHC staining of sections of resected specimens. Activin A expression was not present in normal epithelium but in certain CRC tissues. Activin A staining was observed throughout the cytoplasm of CRC cells (Fig. S2). The present results thus indicated that high activin A expression is associated with unfavorable patient outcomes, but not with sarcopenia, as indicated by the SMI analysis. Therefore, it was next queried whether exposure to activin A directly affects the malignant behaviors of CRC cells in vitro. First, RT-qPCR analysis of a panel of human CRC cell lines was performed, which indicated that LoVo and SW480 cell lines expressed significantly higher levels of endogenous activin A mRNA than the other cell lines tested (Fig. S3). LoVo and SW480 cells were exposed to exogenous activin A at 10 ng/ml for up to 96 h and the effects on cell proliferation, migration and invasion were analyzed. Cells exposed to activin A displayed significantly increased proliferation (P<0.05; Fig. 4A), invasion (P<0.05; Fig. 4B) and migration (P<0.0; Fig. 4C) compared with untreated control cells. Similar results were obtained in SW620 cells, which had lower activin A expression than LoVo cells (Fig. S4). Thus, activin A may act on CRC cells to promote behaviors associated with malignancy.

Proliferation, invasion and migration of human CRC cell lines subjected to activin A knockdown

As LoVo and SW480 cells express high endogenous levels of activin A (Fig. S3), it was next investigated whether siRNA-mediated knockdown of activin A affected malignant cell phenotypes in vitro. Two independent activin A-targeting siRNAs (siActivin A #1 and #2) were evaluated and confirmed by RT-qPCR analysis to effectively suppress activin A mRNA expression to levels <30% of those in LoVo or SW480 cells transfected with an siCtrl sequence (Fig. S5).

Compared with siCtrl-transfected cells, siActivin A #1- or #2-transfected LoVo and SW480 cells exhibited significantly decreased proliferation (P<0.05; Fig. 5A), invasion (P<0.05; Fig. 5B) and migration (P<0.05; Fig. 5C). These results suggested that endogenous activin A expression contributed to the malignant behavior of CRC cells.

Discussion

Activin A has a number of important physiological roles, including induction of differentiation during vertebrate embryogenesis, neuronal differentiation and skeletal muscle cell degradation. Activin A circulates in the blood and is secreted by the gonads, pituitary gland and placenta. Previous studies reported that high tumor expression of activin A is associated with poor prognosis in patients with certain types of gastrointestinal cancer (1821). Consistent with this, the present study demonstrated that activin A was highly expressed in CRC tissues compared with matched normal intestinal epithelium and that high expression was an independent predictor of poor prognosis for patients with CRC.

In the early stages of epithelial cancers, TGF-β family cytokines function as tumor suppressors and inhibit cell proliferation. However, as the cancer progresses, TGF-β cytokines become tumor promoters and induce cancer metastasis by enhancing cell migration and invasion. Numerous studies have examined the involvement of activin A in cancer malignancy (22,25,26). For instance, Hoda et al (25) reported that activin A is involved in the malignant transformation of pleural mesothelioma through regulation of cyclin D. The results of the present study provide support for these earlier findings.

Antagonism of Activin A receptor type IIB, the main cell surface receptor for activin A, has been reported to suppress cachexia and prolong survival in a mouse model (26). This is consistent with the role of activin A in regulating skeletal muscle degradation, suggesting that activin A released from cancer tissues may be associated with the sarcopenia observed in numerous patients with CRC. However, in the present study, no significant correlation between tumor activin A expression levels and SMI was obtained. One possible explanation is that SMI is strongly affected by age and sex, with higher SMIs observed in males and young adults compared with those observed in females and older individuals, respectively (27). In addition, Loumaye et al (28) reported that the blood level of activin A was associated with skeletal muscle density in patients with CRC or lung cancer and high blood levels were associated with poor prognosis. Zhong et al (29) reported that serum activin A secreted by pancreatic adenocarcinoma cells was associated with cachexia and poor prognosis. Thus, activin A secreted by tumor cells and other tissues may be involved in the regulation of skeletal muscle mass.

The present study suggested that high expression of activin A was associated with poor prognosis but not with SMI, which suggested that any contribution of activin A to CRC would occur via alternative mechanisms. Indeed, exposure of two CRC cell lines to exogenous activin A directly enhanced the proliferation, invasion and migration of the cells, while knockdown of endogenous activin A had the opposite effects. Furthermore, IHC staining suggested that activin A protein was detected predominantly in the cytoplasm, suggesting that it may be secreted by the CRC cells. These data indicated that CRC cells may both secrete activin A and respond to extracellular activin A, and it may be speculated that activin A therefore functions as both an autocrine and paracrine modulator of CRC malignancy.

Matsuzaki (30) reported that activin A is involved in the metastasis and invasion of cancer cells by promoting c-Myc transcription factor activity and matrix metalloproteinase-9 production via its effects on SMAD signaling. The mechanism by which activin A exposure and knockdown affects the behavior of the CRC cells assessed in the present study remains to be elucidated; however, it is reasonable to assume that it may have effects on SMAD pathway activity. The results of the present study support those of previous reports indicating that activin A contributes to the malignant behaviors of CRC cell lines, including their proliferation, migration and invasion.

The current study has certain limitations, including its retrospective design and the fact that it was performed at a single institution. However, the association between activin A expression in CRC tumors and OS was validated using a dataset from the TCGA. Furthermore, only SMI was used as an indicator of sarcopenia, although there are other indicators of sarcopenia. However, the method using abdominal CT scan was considered to be the best means of evaluating sarcopenia in patients with gastrointestinal cancer, as CT has the great advantage of being routinely performed for staging and follow-up of cancer. Furthermore, Albano et al (31) reported that CT is probably the easiest and most promising modality for the evaluation of sarcopenia. In addition, there are certain reports on the prognosis of CRC (9,10). Furthermore, the results of the in vitro experiments were also confirmed with clinical samples. The effect of activin A should be evaluated in a dose-dependent manner. In additional invasion and migration assays, treatment of LoVo and SW620 cells with 10 ng/ml activin A significantly increased their invasive ability compared with treatment with 100 ng/ml activin A; activin A did not increase invasion or migration in a dose-dependent manner (data not shown). These findings may be associated with activin A receptors and their nuclear translocation. Another limitation is that activin A secreted by the CRC cell lines into the culture medium was not removed prior to the experiments, which may have affected the results. However, given that exogenous activin A was added at a high concentration and that the opposite effects on cell phenotypes were observed upon activin A knockdown, it may be assumed that activin A secreted by the tumor cells would have only had a minor effect on the results.

In conclusion, the present study demonstrated that activin A promotes the proliferation, invasion and migration of CRC cell lines and that high expression of activin A in tumor tissues correlates with poor prognosis in patients with CRC.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding authors on reasonable request.

Authors' contributions

YM and HB designed and directed the study. ND performed the laboratory experiments and collected all the clinicopathological data. YH, RT and YS assisted with the collection of clinicopathological data. HS, TI, YB, NY supported the experiments. ND and YM were responsible for the statistical analysis and wrote the manuscript. TI, YB, NY and HB supervised the study. ND, YM and HB confirmed the authenticity of all the raw data. All authors have read and approved the final version of the manuscript.

Ethics approval and consent to participate

This retrospective and observational study was approved by the institutional ethics committee of Kumamoto University Hospital (14 June 2019/approval no. 1047) and performed in accordance with the Declaration of Helsinki from 1975. Informed consent for sample use was obtained from the patients and families according to institutional review board protocols.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

2 

van der Stok EP, Spaander MCW, Grunhagen DJ, Verhoef C and Kuipers EJ: Surveillance after curative treatment for colorectal cancer. Nat Rev Clin Oncol. 14:297–315. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Tokunaga R, Imamura Y, Nakamura K, Uchihara T, Ishimoto T, Nakagawa S, Iwatsuki M, Baba Y, Sakamoto Y, Miyamoto Y, et al: Carbohydrate antigen 19-9 is a useful prognostic marker in esophagogastric junction adenocarcinoma. Cancer Med. 4:1659–1666. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Daitoku N, Miyamoto Y, Tokunaga R, Sakamoto Y, Hiyoshi Y, Iwatsuki M, Baba Y, Iwagami S, Yoshida N and Baba H: Controlling nutritional status (CONUT) score is a prognostic marker in metastatic colorectal cancer patients receiving first-line chemotherapy. Anticancer Res. 38:4883–4888. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Daitoku N, Miyamoto Y, Sakamoto Y, Tokunaga R, Hiyoshi Y, Nagai Y, Iwatsuki M, Iwagami S, Yoshida N and Baba H: Prognostic significance of serum p53 antibody according to KRAS status in metastatic colorectal cancer patients. Int J Clin Oncol. 25:651–659. 2020. View Article : Google Scholar : PubMed/NCBI

6 

Taniguchi H, Yamazaki K, Yoshino T, Muro K, Yatabe Y, Watanabe T, Ebi H, Ochiai A, Baba E and Tsuchihara K; Japanese Society of Medical Oncology, : Japanese society of medical oncology clinical guidelines: RAS (KRAS/NRAS) mutation testing in colorectal cancer patients. Cancer Sci. 106:324–327. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Yoshino T, Arnold D, Taniguchi H, Pentheroudakis G, Yamazaki K, Xu RH, Kim TW, Ismail F, Tan IB, Yeh KH, et al: Pan-Asian adapted ESMO consensus guidelines for the management of patients with metastatic colorectal cancer: A JSMO-ESMO initiative endorsed by CSCO, KACO, MOS, SSO and TOS. Ann Oncol. 29:44–70. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Forones NM and Tanaka M: CEA and CA 19-9 as prognostic indexes in colorectal cancer. Hepatogastroenterology. 46:905–908. 1999.PubMed/NCBI

9 

Miyamoto Y, Baba Y, Sakamoto Y, Ohuchi M, Tokunaga R, Kurashige J, Hiyoshi Y, Iwagami S, Yoshida N, Yoshida M, et al: Sarcopenia is a negative prognostic factor after curative resection of colorectal cancer. Ann Surg Oncol. 22:2663–2668. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Miyamoto Y, Baba Y, Sakamoto Y, Ohuchi M, Tokunaga R, Kurashige J, Hiyoshi Y, Iwagami S, Yoshida N, Watanabe M and Baba H: Negative impact of skeletal muscle loss after systemic chemotherapy in patients with unresectable colorectal cancer. PLoS One. 10:e01297422015. View Article : Google Scholar : PubMed/NCBI

11 

Evans WJ: Skeletal muscle loss: Cachexia, sarcopenia, and inactivity. Am J Clin Nutr. 91:1123S–1127S. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Kitano Y, Yamashita YI, Saito Y, Nakagawa S, Okabe H, Imai K, Komohara Y, Miyamoto Y, Chikamoto A, Ishiko T and Baba H: Sarcopenia affects systemic and local immune system and impacts postoperative outcome in patients with extrahepatic cholangiocarcinoma. World J Surg. 43:2271–2280. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Harada K, Ida S, Baba Y, Ishimoto T, Kosumi K, Tokunaga R, Izumi D, Ohuchi M, Nakamura K, Kiyozumi Y, et al: Prognostic and clinical impact of sarcopenia in esophageal squamous cell carcinoma. Dis Esophagus. 29:627–633. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Levolger S, van Vugt JL, de Bruin RW and JN IJ: Systematic review of sarcopenia in patients operated on for gastrointestinal and hepatopancreatobiliary malignancies. Br J Surg. 102:1448–1458. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Gaddykurten D, Tsuchida K and Vale W: Activins and the receptor serine kinase superfamily. Recent Prog Horm Res. 50:109–129. 1995.PubMed/NCBI

16 

Miyamoto Y, Hanna DL, Zhang W, Baba H and Lenz HJ: Molecular pathways: Cachexia signaling-a targeted approach to cancer treatment. Clin Cancer Res. 22:3999–4004. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Lokireddy S, Wijesoma IW, Bonala S, Wei M, Sze SK, McFarlane C, Kambadur R and Sharma M: Myostatin is a novel tumoral factor that induces cancer cachexia. Biochem J. 446:23–36. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Lyu S, Jiang C, Xu R, Huang Y and Yan S: INHBA upregulation correlates with poorer prognosis in patients with esophageal squamous cell carcinoma. Cancer Manag Res. 10:1585–1596. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Oshima T, Yoshihara K, Aoyama T, Hasegawa S, Sato T, Yamamoto N, Akito N, Shiozawa M, Yoshikawa T, Numata K, et al: Relation of INHBA gene expression to outcomes in gastric cancer after curative surgery. Anticancer Res. 34:2303–2309. 2014.PubMed/NCBI

20 

Katayama Y, Oshima T, Sakamaki K, Aoyama T, Sato T, Masudo K, Shiozawa M, Yoshikawa T, Rino Y, Imada T and Masuda M: Clinical significance of INHBA gene expression in patients with gastric cancer who receive curative resection followed by adjuvant S-1 chemotherapy. In Vivo. 31:565–571. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Okano M, Yamamoto H, Ohkuma H, Kano Y, Kim H, Nishikawa S, Konno M, Kawamoto K, Haraguchi N, Takemasa I, et al: Significance of INHBA expression in human colorectal cancer. Oncol Rep. 30:2903–2908. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Hoda MA, Rozsas A, Lang E, Klikovits T, Lohinai Z, Torok S, Berta J, Bendek M, Berger W, Hegedus B, et al: High circulating activin a level is associated with tumor progression and predicts poor prognosis in lung adenocarcinoma. Oncotarget. 7:13388–13399. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Sauerbrei W, Taube SE, McShane LM, Cavenagh MM and Altman DG: Reporting recommendations for tumor marker prognostic studies (REMARK): An abridged explanation and elaboration. J Natl Cancer Inst. 110:803–811. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Hoda MA, Munzker J, Ghanim B, Schelch K, Klikovits T, Laszlo V, Sahin E, Bedeir A, Lackner A, Dome B, et al: Suppression of activin a signals inhibits growth of malignant pleural mesothelioma cells. Br J Cancer. 107:1978–1986. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Zhou X, Wang JL, Lu J, Song Y, Kwak KS, Jiao Q, Rosenfeld R, Chen Q, Boone T, Simonet WS, et al: Reversal of cancer cachexia and muscle wasting by ActRIIB antagonism leads to prolonged survival. Cell. 142:531–543. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Janssen I, Heymsfield SB and Ross R: Low relative skeletal muscle mass (sarcopenia) in older persons is associated with functional impairment and physical disability. J Am Geriatr Soc. 50:889–896. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Loumaye A, de Barsy M, Nachit M, Lause P, van Maanen A, Trefois P, Gruson D and Thissen JP: Circulating activin A predicts survival in cancer patients. J Cachexia Sarcopenia Muscle. 8:768–777. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Zhong XL, Pons M, Poirier C, Jiang Y, Liu J, Sandusky GE, Shahda S, Nakeeb A, Schmidt CM, House MG, et al: The systemic activin response to pancreatic cancer: Implications for effective cancer cachexia therapy. J Cachexia Sarcopenia Muscle. 10:1083–1101. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Matsuzaki K: Smad phosphoisoform signaling specificity: The right place at the right time. Carcinogenesis. 32:1578–1588. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Albano D, Messina C, Vitale J and Sconfienza LM: Imaging of sarcopenia: Old evidence and new insights. Eur Radiol. 30:2199–2208. 2020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2022
Volume 47 Issue 6

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
Daitoku N, Miyamoto Y, Hiyoshi Y, Tokunaga R, Sakamoto Y, Sawayama H, Ishimoto T, Baba Y, Yoshida N, Baba H, Baba H, et al: Activin A promotes cell proliferation, invasion and migration and predicts poor prognosis in patients with colorectal cancer. Oncol Rep 47: 107, 2022
APA
Daitoku, N., Miyamoto, Y., Hiyoshi, Y., Tokunaga, R., Sakamoto, Y., Sawayama, H. ... Baba, H. (2022). Activin A promotes cell proliferation, invasion and migration and predicts poor prognosis in patients with colorectal cancer. Oncology Reports, 47, 107. https://doi.org/10.3892/or.2022.8318
MLA
Daitoku, N., Miyamoto, Y., Hiyoshi, Y., Tokunaga, R., Sakamoto, Y., Sawayama, H., Ishimoto, T., Baba, Y., Yoshida, N., Baba, H."Activin A promotes cell proliferation, invasion and migration and predicts poor prognosis in patients with colorectal cancer". Oncology Reports 47.6 (2022): 107.
Chicago
Daitoku, N., Miyamoto, Y., Hiyoshi, Y., Tokunaga, R., Sakamoto, Y., Sawayama, H., Ishimoto, T., Baba, Y., Yoshida, N., Baba, H."Activin A promotes cell proliferation, invasion and migration and predicts poor prognosis in patients with colorectal cancer". Oncology Reports 47, no. 6 (2022): 107. https://doi.org/10.3892/or.2022.8318