Open Access

SMAD‑6, ‑7 and ‑9 are potential molecular biomarkers for the prognosis in human lung cancer

  • Authors:
    • Shuxian Pan
    • Guangming Zhou
    • Wentao Hu
    • Hailong Pei
  • View Affiliations

  • Published online on: July 9, 2020     https://doi.org/10.3892/ol.2020.11851
  • Pages: 2633-2644
  • Copyright: © Pan 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

SMADs, a family of proteins that function as signal transducers and transcriptional regulators to regulate various signaling pathways, including the transforming growth factor‑β signaling pathway, are similar to the mothers against decapentaplegic family of genes and the sma gene family in Caenorhabditis elegans. SMADs generate context‑dependent modulation by interacting with various sequence‑specific transcription factors, such as E2F4/5, c‑Fos, GATA3, YY1 and SRF, which have been found to serve a key role in lung carcinoma oncogenesis and progression. However, the prognostic values of the eight SMADs in lung cancer have not been fully understood. In the present study, the expression levels and survival data of SMADs in patients with lung carcinoma from the Oncomine, Gene Expression Profiling Interactive Analysis, Kaplan‑Meier plotter and cBioPortal databases were downloaded and analyzed. It was found that the mRNA expression levels of SMAD‑6, ‑7 and ‑9 were decreased in lung adenocarcinoma and squamous cell carcinoma compared with that in adjacent normal tissues, while there was no significant difference in SMADs 1‑5. Survival analysis revealed that not only were low transcriptional levels of SMAD‑6, ‑7 and ‑9 associated with low overall survival but they also had prognostic role for progression‑free survival and post‑progression survival (P<0.05) in patients with lung carcinoma. In conclusion, the present study demonstrated that SMAD‑6, ‑7 and ‑9 are potential biomarkers for the prognosis of patients with lung carcinoma.

Introduction

Lung carcinoma, the most commonly diagnosed cancer (11.6% of total cases) and the leading cause of cancer-associated mortality (18.4% of total mortalities) worldwide, according to the GLOBOCAN 2018 estimates of cancer incidence and mortality (1), is characterized by a high degree of malignancy, metastasis and high mortality rate (1). Lung carcinoma is the most diagnosed cancer and the leading cause of cancer-associated mortality in men (1), and the third most common cancer and second cause of cancer-associated mortality in women, worldwide (1). Most lung cancer tumors have no obvious symptoms in the early stage and are therefore less likely to be detected; thus, the majority of patients with lung carcinoma are diagnosed at an advanced stage, which reduces the efficacy of treatment and the 5-year survival rate is low (2,3). Therefore, novel potential prognostic biomarkers and drug targets are required to improve the outcome and individualized treatments of lung cancer.

SMADs are a family of genes that encode signal transducers and transcriptional modulators, which mediate various signaling pathways, such as TGF-β/SMAD, BMP/SMAD, ERK/MAPK, Hippo, JAK/STAT and Wnt/β-catenin (4). In mammals, there are eight SMAD proteins, which are sub-divided into three types: Receptor-regulated SMADs (R-SMADs), common-mediator SMADs and inhibitory SMADs (46). SMADs can recognize and bind several sequence-specific and context-dependent transcriptional regulation factors, such as FoxH1, Sp1, YY1 and p53, which have been found to participate in various biological processes, including cell proliferation, apoptosis, differentiation, as well as tumor progression and immune regulation processes (79). The majority of signaling pathways regulated by SMADs, such as TGF-β/SMAD, BMP/SMAD, ERK/MAPK, JAK/STAT and Wnt/β-catenin, are deregulated in various human malignant carcinomas, including lung carcinoma, malignant melanoma, colorectal cancer, kidney cancer, breast cancer, ovarian cancer and prostate cancer (1017).

A total of seven mammalian SMAD proteins have been reported to participate in the regulation of lung carcinoma tumorigenesis or progression. The SMAD-1 gene participates in negative regulation of the Akt/GSK3β pathway to maintain the cancer stem cell-like characteristic of cancer stem cells in non-small cell lung carcinoma (NSCLC) (18). Yang et al (19) found that SMAD1 knockdown inhibited epithelial-mesenchymal transition (EMT) induced by fine particulate matter (PM2.5) in human lung carcinoma cells. In addition, Tang et al (20) reported that transcriptional activation of SMAD-2 and −3 facilitates the growth and metastasis of lung carcinoma through enhanced transforming growth factor (TGF)-β1-induced EMT and from the generation of the angiogenic factors, such as vascular endothelial growth factor and connective tissue growth factor. SMAD4 is phosphorylated(p) at Tyrosine 95 by an oncogenic tyrosine kinase ALK, preventing it from binding to DNA and eliciting TGF-β-induced tumor suppressing responses during lung cancer tumorigenesis (21). Phosphorylated (p)-SMAD-9 expression levels were found to be markedly enhanced and associated with the metastatic potential of non-small cell lung carcinoma A549 subclones from experimental brain metastases through four rounds of intracardiac injection of A549 cells or its derivatives into athymic nude mice (22). Inhibitory SMADs, including SMAD-6 and −7, have been found to be involved in lung carcinoma by regulating the stability or activity of TGF-β receptors, including the modulation of TβRI ubiquitination and degradation, by which TGF-β signaling is regulated and functions in lung cancer growth and metastasis (6,23). However, the underlying mechanisms by which SMADs are involved in the regulation of lung carcinoma are not fully understood.

At present, the dysregulated mRNA expression levels of SMAD proteins in human lung cancer and their associations with lung carcinoma prognosis have not been investigated. In the present study, bioinformatics analysis was used to investigate the roles of SMAD proteins in human lung carcinoma. The expression patterns and mutations of different SMAD proteins in patients with lung carcinoma were analyzed, from the vast number of gene expression data that has been previously published, to identify SMAD expression patterns and potential prognostic values in human lung carcinoma.

Materials and methods

Oncomine analysis

The Oncomine gene expression array dataset (www.oncomine.org), was used for analyzing the expression levels of SMADs in different carcinomas, including bladder cancer, brain and central nervous system cancer, breast cancer, cervical cancer, colorectal cancer, esophageal cancer, gastric cancer, head and neck cancer, kidney cancer, leukemia, liver cancer, lung cancer, lymphoma, melanoma, myeloma, ovarian cancer, pancreatic cancer, prostate cancer and sarcoma (24). A comparison of SMAD mRNA expression levels between clinical tumor specimens and adjacent normal tissues was performed using unpaired two-tailed Student's t-tests (number of samples: GSE19188: 10, GSE10072: 107, GSE32863: 116, GSE31210: 246 and GSE7670: 64). The cut-off values were set as follows, P<0.01 and fold change >2, respectively.

Gene expression omnibus (GEO) dataset analysis

GSE19188 (25), GSE10072 (26), GSE32863 (25), GSE31210 (25), GSE7670 (27) were downloaded from the GEO database (www.ncbi.nlm.nih.gov/geo/) to analyze the mRNA expression levels of all SMAD subtypes in lung neoplasm tissues and adjacent normal lung tissues (28). These datasets were obtained by searching using the following key words: ‘lung cancer AND SMAD’ or ‘lung cancer AND gene’. To avoid generating less reliable results, the 5 datasets were batch normalized in the R computing environment in RStudio (version 1.2.5001) using the sva package and merged to reduce the variability (2931).

Gene expression profiling interactive analysis (GEPIA) dataset

GEPIA is an online cancer database used for analyzing RNA sequences and expression levels for 9,736 lung cancer samples and 8,587 normal samples from The Cancer Genome Atlas (TCGA) and GTEx projects, based on a standard processing pipeline (32). GEPIA can be used to analyze differential expression in tumor and adjacent normal tissues, tumor type or clinical stage, patient survival analysis, correlation, dimensionality reduction (reducing the dimensionality of high dimensional expression datasets while maintaining most of the variances based on principal component analysis) and similar gene detection (32).

Kaplan-Meier plotter

The prognostic value of the SMADs transcriptional levels was assessed using the online database Kaplan-Meier plotter (www.kmplot.com) (33,34), which contains gene expression data and survival information for 2,437 patients with NSCLC. Overall survival (OS), progression-free survival (PFS) and post-progression survival (PPS) were analyzed after the patients had been divided into 2 groups (high and low) based on the median expression value, using Kaplan-Meier curves, with the log rank test to determine any significant difference. The hazard ratio (HR) and 95% confidence intervals (CI) were also calculated. Only SMAD datasets selected using the JetSet best probe package in R were used in Kaplan-Meier analysis.

TCGA data and cBioPortal

TGCA includes sequencing and pathology data for 30 different cancer types, including glioblastoma multiforme, head and neck, kidney clear cell, lung adenocarcinoma, lung squamous cell carcinoma and medulloblastoma (35). Using cBioPortal (36), SMAD analyses were conducted using the provisional lung adenocarcinoma TCGA dataset. Using the Genomic Identification of Significant Targets in Cancer package (version 1.12.0) which can identify mutations, putative copy number alterations, this was included with the z-scores (also known as standard score, it is obtained by dividing the difference between a number and an average by the standard deviation. In statistics, the standard score is the sign number of the standard deviation of the value of an observation or data point higher than the average of the observed value or measured value. Its value is positively correlated with mRNA expression level) (37) of mRNA expression levels [RNA sequencing V2 RSEM (RNA-Seq by Expectation Maximization)] (38) and the protein expression level data (using reverse phase protein array data) to create the genomic profile for 522 patients with lung adenocarcinoma. The co-expression network was plotted using cBioPortal. The functional roles of the target host gene of SMADs were predicted using Gene Ontology (GO) enrichment analysis of three elements: Biological process, cellular composition and molecular function. Kyoto Encyclopedia of Genes and Genomes (KEGG) function analysis of genes associated with SMAD alterations was performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID; http://david.ncifcrf.gov/summary.jsp).

Statistical analysis

Student's t-tests was used to compare mRNA expression levels in the Oncomine and GEO databases. Kaplan-Meier plotter and Cramér-von Mises tests were used to analyze survival. By defining disease state (tumor or normal) as a variable, GEPIA analysis of variance was performed using one-way ANOVA. Two-tailed Student's t-test was used to compare the expression levels of SMAD mRNA between clinical tumor specimens and adjacent normal tissues. Spearman's correlation analysis was used to evaluate the correlation between gene expression levels in TCGA database. P<0.05 was considered to indicate a statistically significant difference.

Results

Variation of SMADs expression levels among different types of lung carcinoma and adjacent normal tissues

The expression levels of the eight SMADs of various cancer types and in adjacent normal samples were compared using the Oncomine database (Fig. 1). The transcriptional levels of SMAD-6, −7 and −9 were significantly decreased in patients with lung carcinoma (P<0.05). No significant expression difference was observed for SMAD-1/-2/-3/-4/-5 between lung cancer tissues and normal tissues. With respect to lung adenocarcinoma, the expression levels of SMAD-6, −7 and −9 were significantly decreased in cancer tissues compared with that in adjacent normal tissues, with a fold change of −4.622, −3.508 and −2.285, respectively (P<0.05; Table I) (3941). In particular, only the expression levels of SMAD-7 were decreased compared with that in the adjacent normal samples in all types of lung cancer; SMAD-7 was found to be decreased in squamous cell lung carcinoma by −3.813-fold, in small cell lung carcinoma with a fold change of −4.568 and in lung carcinoid tumor with a fold change of −6.114 (Table I). There were no significant differences in the transcription levels for the remaining SMADs (P>0.05) (2527).

Table I.

Significant changes of SMAD transcriptional levels between different types of lung cancer and normal lung tissues using the Oncomine database.

Table I.

Significant changes of SMAD transcriptional levels between different types of lung cancer and normal lung tissues using the Oncomine database.

GenesTypes of lung cancer vs. normal tissueFold changeP-valuet-test valueAuthor, year(Refs.)
SMAD-1Lung adenocarcinoma vs. normal1.185>0.051.555Beer et al, 2002(25)
SMAD-2Squamous cell lung cancer vs. normal1.205>0.051.525Garber et al, 2001(26)
Large cell lung carcinoma vs. normal1.070>0.050.668Garber et al, 2001(26)
Large cell lung carcinoma vs. normal1.412>0.051.272Garber et al, 2001(26)
Small cell lung carcinoma vs. normal−1.069>0.05−0.427Garber et al, 2001(26)
SMAD-3Lung adenocarcinoma vs. normal1.213>0.051.104Beer et al, 2002(25)
SMAD-4Lung adenocarcinoma vs. normal−1.110>0.05−1.405Beer et al, 2002(25)
SMAD-5Lung adenocarcinoma vs. normal1.415>0.051.239Yamagata et al, 2003(27)
Squamous cell carcinoma vs. normal1.302>0.050.962Yamagata et al, 2003(27)
Large cell carcinoma vs. normal1.398>0.051.073Yamagata et al, 2003(27)
SMAD-6Lung adenocarcinoma vs. normal−4.622<0.0001−12.757Selamat et al, 2012(39)
SMAD-7Lung adenocarcinoma vs. normal−3.508<0.0001−5.632Bhattacharjee et al, 2001(40)
Squamous cell carcinoma vs. normal−3.813<0.0001−4.561Bhattacharjee et al, 2001(40)
Small cell lung carcinoma vs. normal−4.568<0.0001−6.228Bhattacharjee et al, 2001(40)
Lung carcinoid tumor vs. normal−6.114<0.0001−7.804Bhattacharjee et al, 2001(40)
SMAD-9Lung adenocarcinoma vs. normal−2.285<0.0001−5.387Okayama et al, 2012(41)
SMAD mRNA expression levels are associated with the clinicopathological parameters of patients with lung carcinoma

GEPIA and GEO were used to compare the mRNA transcriptional levels of SMAD proteins between lung carcinoma tissues and adjacent normal tissues. The results from GEPIA analysis showed that there were significantly lower expression levels of SMAD-6, −7 and −9 in lung adenocarcinoma and squamous cell lung carcinoma tissues compared with normal tissues, while there were no significant differences in the expression levels for SMAD1-5 (Fig. 2A and C). The results from GEO analysis showed similar results, and SMAD-7 showed significantly increased expression in ADC, LCC and SCC samples as compared with normal tissues (Fig. 2B). The transcriptional levels of SMADs in different pathological stages of lung adenocarcinoma and squamous cell carcinoma tissues were also analyzed. The mRNA expression levels of SMAD-6 and −9 were significantly different at stages I–IV, according to the TNM staging system of American Joint Committee on Cancer (AJCC) in 2010 (42), while there were no significant differences in the expression levels of SMAD-1, −2, −3, −4, −5 and −7 (Fig. 3).

Association between decreased mRNA expression levels of SMAD-6, −7 and −9 with the prognosis of patients with lung carcinoma

The association between the mRNA expression levels of SMAD proteins and the survival of patients with NSCLC was analyzed using Kaplan-Meier plotter, and the results revealed that the patients with low mRNA expression levels of SMAD-5, −6, −7 and −9 had poor OS, PFS and PPS rates (from left to right: OS, PFS and PPS) (P<0.05, Fig. 4), suggesting their potential roles as a tumor suppressor, except that SMAD6 is not significantly associated with PPS. SMAD-2, −4 was identified as a tumor suppressor by OS analysis; however, the results for SMAD-3 were ambiguous, which was demonstrated to be a tumor suppressor for PPS but an oncogene for PFS.

Changes in SMAD protein expression levels and their networks in patients with lung carcinoma

Alterations, associations and networks of SMAD proteins were analyzed using the cBioPortal online tool for lung adenocarcinoma. Of the 522 patients with lung adenocarcinoma, 208 (40%) had alterations in SMADs; while >2 changes were detected in 83 samples (Fig. 5A). Among the eight SMADs, SMAD-4 has the most alterations (with ‘mRNA low’ being the most abundant type) while SMAD-6 has the least changes. Deep deletion was identified in all SMADs except SMAD-1. Missense mutations were identified in SMAD-2/3/4/5/6/9 but not SMAD-1/7. Calculation of the correlations between each of the SMADs in lung adenocarcinoma was performed using cBioPortal online tool based on their mRNA expression levels. The results indicated that SMAD-2 was significantly correlated with SMAD4 (R=0.55; P<0.001) and SMAD7 (R=0.4; P<0.001), while SMAD-1, −3, −5, −6 and −9 were not significantly associated with any other SMADs (Fig. 5B). The interaction network between SMADs and the 49 most-confirmed altered neighbors was then constructed, demonstrating that cell cycle-associated genes, such as CDK4, BCL-9, MYC, CREBBP and E2F5, were associated with changes to SMAD expression levels (Fig. 5C).

Using the Database for Annotation, Visualization and Integrated Discovery (https://david.ncifcrf.gov/summary.jsp), the functions of the SMADs and genes associated with SMAD expression level changes were predicted. The functional roles of the target host genes of SMADs were predicted using Gene Ontology (GO) enrichment analysis of three elements: Biological process, cellular composition and molecular function. It was found that all SMAD mRNA expression changes in lung adenocarcinoma were found to be enriched in cellular metabolic processes, biochemical processes, multicellular progression and biochemical activity-related proteins, such as stress (Fig. 6A). Among the proteins associated with disease and drug sensitivity, it was found that the SMAD proteins were involved in biological activities such as ‘lung cancer (smoking interaction)’, ‘severe aortic features in Marfan syndrome’ and ‘immune response to anthrax vaccine’ (Fig. 6B).

Discussion

The majority of studies investigating SMAD proteins are related to the TGF-β signaling pathway, which functions in tumorigenesis and tumor progression as well as tumor immunosuppression (11,4347). Dysregulation of several SMAD proteins has been proved in numerous types of cancer. It has been reported that SMAD1 gene polymorphisms influence colorectal cancer susceptibility (48), while SMAD2 signaling is enhanced in chemoresistant colorectal cancer cells (49). Tone et al (50) identified R361G mutation of SMAD4 in primary low-grade serous ovarian carcinoma samples, which is situated within MH2 domain and is speculated to affect the functional specificity and selectivity of SMAD4 protein. SMAD5 was found to be overexpressed in human breast cancer cells and was involved in the induction of cancer stem cell-like phenotype (51). Further, SMAD7 expression is decreased by the overexpression of SETDB1 in human breast cancer, while upregulation of SMAD7 by SETDB1 knockdown inhibits breast cancer metastasis (52). However, bioinformatics analysis of SMAD proteins associated with human lung cancer has not been performed, to the best of our knowledge. The present study reported the mRNA expression levels and survival prediction potential of SMAD proteins in human lung cancer for the first time.

SMAD1 has been found to be involved in both the positive modulation of EMT in human malignant lung neoplasm cells and in the maintenance of stem-like cell traits (18,19). Immunohistochemical analyses was performed to determine the protein expression levels of SMAD1 in 60 cases of lung cancer tissues (lung cancer group), 25 cases of normal alveolus tissues (alveolus control group) and 29 cases of normal bronchial tissues (bronchial control group), and the results revealed markedly lower expression levels of SMAD1 in the lung cancer group compared with that in normal tissue (53). Furthermore, the expression levels of SMAD-1 protein were significantly associated with lung carcinoma differentiation and lymphatic metastasis (53). However, according to the present analysis, there were no significant differences in the expression levels of SMAD-1 between either lung adenocarcinoma or lung squamous cell carcinoma and adjacent normal tissues. The results from Liu et al (18) and Yang et al (19) were collected from lung cancer cell lines, which might account for these differences. In addition, the difference between the findings from the present study (based on 650 cases of lung cancer tissues) and with that from Gao et al (53) (based on 60 cases of lung cancer tissues) may be caused by the different sample size.

The regulatory functions of SMAD-2 and −3 have been explored extensively in lung carcinoma. It was reported that SMAD-2 and −3 function as key R-SMAD proteins mediating the TGF-β/bone morphogenetic protein signaling pathway to facilitate EMT of lung carcinoma cells, as well as tumorigenesis, invasion and metastasis (20,5458). Chen et al (59) also reported that a higher transcriptional level of p-SMAD-2 in stromal fibroblasts predicted less favorable survival in patients with pathological stage I to IIIA NSCLC, according to TNM staging system of American Joint Committee on Cancer (AJCC) in 2010 (42). SMAD3 was also reported to facilitate the progression of NSCLC by upregulating PAX6 expression (60). However, in the present bioinformatics study there were no significant differences in the expression levels of SMAD-2 and −3 between lung carcinoma and adjacent normal tissues. SMAD-2 was identified as a tumor suppressor using OS analysis; however, the results for SMAD-3 were ambiguous, as SMAD3 was demonstrated to be a tumor suppressor for PPS but an oncogene for PFS.

SMAD-4 binds to other active R-SMAD proteins (SMAD-1/2/3/5/8) to form homo- and heterotypic complexes that accumulate in the nucleus and regulate transcription of target genes, including Nanog, CDKN1C, CDKN2B, Nodal, PAI-1, Lefty1 and SMAD7 (6). SMAD-4 appears to function as a tumor suppressor in lung carcinoma and is associated with the differentiation status of lung carcinoma tissues (61,62). It was found that the p.R361C mutation in SMAD-4 serves a role in downregulating the TGF-β signaling pathway, causing a loss of growth inhibition and transcriptional activation mediated by SMADs, and it is hypothesized that the mutation p.R361C in SMAD-4 plays a crucial part in lung oncogenesis (63). It has been reported that the SMAD4 protein expression levels in NSCLC tissues are significantly lower compared with that in a normal tracheal-bronchial epithelium (P<0.05) using immunohistochemistry and that SMAD4 knockdown initiates and promotes lung carcinoma progression (64). In addition, suppression of SMAD4 protein expression has been found to be associated with the promotion of growth and invasion and metastasis of lung cancer (6567).

In the present study, SMAD-4 was shown to be downregulated in both lung adenocarcinoma and lung squamous cell carcinoma; however, this difference was not significant. OS analysis also demonstrated that SMAD-4 functioned as a tumor suppressor, which is consistent with the genetic alteration analysis in lung adenocarcinoma (cBioPortal) shown in Fig. 5A (with both missense and truncating mutations as putative drivers). A possible explanation for the tumor suppressor function of SMAD-4 may be due to the missense mutation sites, given that no difference in expression levels was observed between cancer tissues and adjacent normal tissues, similar with that for SMAD-2. This hypothesis is also in accordance with a previous report indicating that some mutations in SMAD-2 and −4 (D450H, del1434-6 in SMAD-2 and R420H, R441P in SMAD-4) are associated with progression in lung cancer (68), which further supports the present finding that SMAD-2 is significantly correlated with SMAD-4 (Fig. 5B).

It was observed using a two-stage case-control study, with 4,680 cases and controls, that there is a significant association between SMAD-5 rs12719482 and increased risk of lung carcinoma, suggesting that SMAD-5 participates in the modulation of lung cancer tumorigenesis. It is speculated that SMAD-5 rs12719482 is a candidate marker for lung cancer susceptibility, which decreases the expression of SMAD-5 through binding has-miR-1270, hsa-miR-571 or hsa-miR-920 (69). This may also explain why the OS analysis of the present study suggested that SMAD-5 functions as a tumor suppressor despite the fact that there was no significant difference in SMAD-5 expression levels.

With regards to SMAD-9, which is another R-SMAD protein, there have been no previously published studies investigating its function in the regulation of lung cancer to the best of our knowledge. The present findings indicated that no significant mRNA expression level changes were observed for SMAD-5; however, the expression levels of SMAD-9 was found to be significantly decreased in lung adenocarcinoma and lung squamous cell carcinoma tissues compared with that in adjacent normal tissues, which suggests that SMAD-9 may also serve as a tumor suppressor in lung cancer progression. In addition, the survival analysis also suggested that low expression levels of SMAD-5 and −9 were significantly associated with poor OS, PFS and PPS in all patients with lung carcinoma, which agrees with previous reports demonstrating the tumor suppressor roles of SMAD-5 and −9 (6971). However, as there was no significant association between SMAD-5 expression levels and the cancer stage of lung carcinoma (Fig. 3), the value of SMAD5 as a potential biomarker is limited.

The two inhibitory SMAD proteins, SMAD-6 and −7, were found to be downregulated in both human lung adenocarcinoma and lung squamous cell carcinoma tissues in the present study. Low expression levels of SMAD6 were associated with poor OS and PFS, but not PPS, in all patients with lung carcinoma, whilst low expression levels of SMAD-7 were associated with poor OS, PFS and PPS. These findings indicate the roles of SMAD-6 and −7 as tumor suppressors in human lung cancer, which agrees with previous published findings regarding these functions in lung cancer (46,72). Moreover, it was found that SMAD7 is markedly correlated with SMAD-2 as shown in Fig. 5B, which is also consistent with the finding that the expression of SMAD-2 is regulated by inhibitory SMAD-7 activity in a negative feedback in cancer cells (73).

As shown in Fig. 5, different deep deletions were found for SMAD-6, −7 and −9 in clinical lung adenocarcinoma samples, which is in accordance with the present survival analysis. Missense mutations were also identified for SMAD-6 and −9 but not SMAD-7, suggesting that both deep deletions and missense mutations may serve an important role in lung cancer progression.

In conclusion, the expression levels and prognostic values of SMADs in lung cancer were analyzed in the present study, which improved the understanding of the regulation of lung cancer. The results demonstrated that decreased expression levels of SMAD-6, −7 and −9 might function in the tumorigenesis and development of lung carcinoma, and that these SMADs may also function as biomarkers to identify patients with a high-risk of lung carcinoma. Nevertheless, there are still some limitations for the present study, for example, all results were obtained by bioinformatics analyses and no experimental validation was performed. It is suggested that both in vitro and in vivo experimental validation is required for the confirmation of the three molecules as bona fide biomarkers for lung carcinoma, which should be conducted in future studies. Furthermore, experiments need to be performed to validate the tumor suppressive functions of the three SMADs in both lung cancer cells and transplanted lung tumor models. The experimental validation and the functional deciphering of the three SMADs are of great importance to better understand the functions of TGF-β signaling in human lung cancer.

Acknowledgements

Not applicable.

Funding

This study was supported by The National Key R and D Program of China (grant nos. 2018YFC0115704 and 2018YFC0115705) and The National Natural Science Foundation of China (grant nos. 11405235 and 81602794).

Availability of data and materials

The datasets generated and/or analyzed during the present study are available in the [online] repository, [http://ualcan.path.uab.edu/analysis.html, https://www.oncomine. org/resource/main.html, https://www.cancer.gov/about-nci/organization/ccg/research/structural-genomics/tcga, https://www.cbioportal.org/].

Authors' contributions

GZ, WH and HP conceived and designed the study. SP conducted the bioinformatics analyses. WH and SP drafted the initial manuscript. All authors read and approved the final manuscript.

Ethic approval and consent to participate

The present study was conducted according to The Declaration of Helsinki. All the datasets were retrieved from open databases and published literature.

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 

Weller DP, Peake MD and Field JK: Presentation of lung cancer in primary care. NPJ Prim Care Respir Med. 29:212019. View Article : Google Scholar : PubMed/NCBI

3 

Zhao Y, Wang W, Liang H, Yang CJ, D'Amico T, Ng CSH, Liu CC, Petersen RH, Rocco G, Brunelli A, et al: The optimal treatment for stage IIIA-N2 non-small cell lung cancer: A network meta-analysis. Ann Thorac Surg. 107:1866–1875. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Budi EH, Duan D and Derynck R: Transforming growth factor-β receptors and smads: Regulatory complexity and functional versatility. Trends Cell Biol. 27:658–672. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Luo K: signaling cross talk between TGF-β/Smad and other signaling pathways. Cold Spring Harb Perspect Biol. 9:a0221372017. View Article : Google Scholar : PubMed/NCBI

6 

Yu Y and Feng XH: TGF-β signaling in cell fate control and cancer. Curr Opin Cell Biol. 61:56–63. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Kamato D, Do BH, Osman N, Ross BP, Mohamed R, Xu S and Little PJ: Smad linker region phosphorylation is a signalling pathway in its own right and not only a modulator of canonical TGF-β signalling. Cell Mol Life Sci. 77:243–251. 2020. View Article : Google Scholar : PubMed/NCBI

8 

Silvestri L, Nai A, Dulja A and Pagani A: Hepcidin and the BMP-SMAD pathway: An unexpected liaison. Vitam Horm. 110:71–99. 2019. View Article : Google Scholar : PubMed/NCBI

9 

Dai J, Xu M, Zhang X, Niu Q, Hu Y, Li Y and Li S: Bi-directional regulation of TGF-β/Smad pathway by arsenic: A systemic review and meta-analysis of in vivo and in vitro studies. Life Sci. 220:92–105. 2019. View Article : Google Scholar : PubMed/NCBI

10 

Eckhardt BL, Cao Y, Redfern AD, Chi LH, Burrows AD, Roslan S, Sloan EK, Parker BS, Loi S, Ueno NT, et al: Activation of canonical BMP4-SMAD7 signaling suppresses breast cancer metastasis. Cancer Res. 80:1304–1315. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Lang C, Dai Y, Wu Z, Yang Q, He S, Zhang X, Guo W, Lai Y, Du H, Wang H, et al: SMAD3/SP1 complex-mediated constitutive active loop between lncRNA PCAT7 and TGF-β signaling promotes prostate cancer bone metastasis. Mol Oncol. 14:808–828. 2020. View Article : Google Scholar : PubMed/NCBI

12 

Zhang Y, Zeng Y, Liu T, Du W, Zhu J, Liu Z and Huang JA: The canonical TGF-β/Smad signalling pathway is involved in PD-L1-induced primary resistance to EGFR-TKIs in EGFR-mutant non-small-cell lung cancer. Respir Res. 20:1642019. View Article : Google Scholar : PubMed/NCBI

13 

Tuncer E, Calcada RR, Zingg D, Varum S, Cheng P, Freiberger SN, Deng CX, Kleiter I, Levesque MP, Dummer R and Sommer L: SMAD signaling promotes melanoma metastasis independently of phenotype switching. J Clin Invest. 129:2702–2716. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Jung M, Lee JH, Lee C, Park JH, Park YR and Moon KC: Prognostic implication of pAMPK immunohistochemical staining by subcellular location and its association with SMAD protein expression in clear cell renal cell carcinoma. Cancers (Basel). 11:16022019. View Article : Google Scholar

15 

Leng Z, Li Y, Zhou G, Lv X, Ai W, Li J and Hou L: Krüppel-like factor 4 regulates stemness and mesenchymal properties of colorectal cancer stem cells through the TGF-β1/Smad/snail pathway. J Cell Mol Med. 24:1866–1877. 2020. View Article : Google Scholar : PubMed/NCBI

16 

Bai Y, Li LD, Li J, Chen RF, Yu HL, Sun HF, Wang JY and Lu X: A FXYD5/TGF-β/SMAD positive feedback loop drives epithelial-to-mesenchymal transition and promotes tumor growth and metastasis in ovarian cancer. Int J Oncol. 56:301–314. 2020.PubMed/NCBI

17 

Chen J, Deng Y, Ao L, Song Y, Xu Y, Wang CC, Choy KW, Tony Chung KH, Du Q, Sui Y, et al: The high-risk HPV oncogene E7 upregulates miR-182 expression through the TGF-β/Smad pathway in cervical cancer. Cancer Lett. 460:75–85. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Liu CW, Li CH, Peng YJ, Cheng YW, Chen HW, Liao PL, Kang JJ and Yeng MH: Snail regulates Nanog status during the epithelial-mesenchymal transition via the Smad1/Akt/GSK3β signaling pathway in non-small-cell lung cancer. Oncotarget. 5:3880–3894. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Yang D, Ma M, Zhou W, Yang B and Xiao C: Inhibition of miR-32 activity promoted EMT induced by PM2.5 exposure through the modulation of the Smad1-mediated signaling pathways in lung cancer cells. Chemosphere. 184:289–298. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Tang YN, Ding WQ, Guo XJ, Yuan XW, Wang DM and Song JG: Epigenetic regulation of Smad2 and Smad3 by profilin-2 promotes lung cancer growth and metastasis. Nat Commun. 6:82302015. View Article : Google Scholar : PubMed/NCBI

21 

Zhang Q, Xiao M, Gu S, Xu Y, Liu T, Li H, Yu Y, Qin L, Zhu Y, Chen F, et al: ALK phosphorylates SMAD4 on tyrosine to disable TGF-β tumour suppressor functions. Nat Cell Biol. 21:179–189. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Rao G, Pierobon M, Kim IK, Hsu WH, Deng J, Moon YW, Petricoin EF, Zhang YW, Wang Y and Giaccone G: Inhibition of AKT1 signaling promotes invasion and metastasis of non-small cell lung cancer cells with K-RAS or EGFR mutations. Sci Rep. 7:70662017. View Article : Google Scholar : PubMed/NCBI

23 

Yu J, Lei R, Zhuang X, Li X, Li G, Lev S, Segura MF, Zhang X and Hu G: MicroRNA-182 targets SMAD7 to potentiate TGFβ-induced epithelial-mesenchymal transition and metastasis of cancer cells. Nat Commun. 7:138842016. View Article : Google Scholar : PubMed/NCBI

24 

Liu W, Ouyang S, Zhou Z, Wang M, Wang T, Qi Y, Zhao C, Chen K and Dai L: Identification of genes associated with cancer progression and prognosis in lung adenocarcinoma: Analyses based on microarray from oncomine and the cancer genome atlas databases. Mol Genet Genomic Med. 7:e005282019.PubMed/NCBI

25 

Beer DG, Kardia SL, Huang CC, Giordano TJ, Levin AM, Misek DE, Lin L, Chen G, Gharib TG, Thomas DG, et al: Gene-expression profiles predict survival of patients with lung adenocarcinoma. Nat Med. 8:816–824. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Garber ME, Troyanskaya OG, Schluens K, Petersen S, Thaesler Z, Pacyna-Gengelbach M, van de Rijn M, Rosen GD, Perou CM, Whyte RI, et al: Diversity of gene expression in adenocarcinoma of the lung. Proc Natl Acad Sci USA. 98:13784–13789. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Yamagata N, Shyr Y, Yanagisawa K, Edgerton M, Dang TP, Gonzalez A, Nadaf S, Larsen P, Roberts JR, Nesbitt JC, et al: A training-testing approach to the molecular classification of resected non-small cell lung cancer. Clin Cancer Res. 9:4695–4704. 2003.PubMed/NCBI

28 

Pei YF, Xu XN, Wang ZF, Wang FW, Wu WD, Geng JF and Liu XQ: Methyl-CpG binding domain protein 2 inhibits the malignant characteristic of lung adenocarcinoma through the epigenetic modulation of 10 to 11 translocation 1 and miR-200s. Am J Pathol. 189:1065–1076. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Loraine AE, Blakley IC, Jagadeesan S, Harper J, Miller G and Firon N: Analysis and visualization of RNA-Seq expression data using RStudio, bioconductor, and integrated genome browser. Methods Mol Biol. 1284:481–501. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Benso A, Di Carlo S, Politano G, Savino A and Hafeezurrehman H: Building gene expression profile classifiers with a simple and efficient rejection option in R. BMC Bioinformatics. 12 (Suppl 13):S32011. View Article : Google Scholar : PubMed/NCBI

31 

Leek JT, Johnson WE, Parker HS, Jaffe AE and Storey JD: The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics. 28:882–883. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res 45 (W1). W98–W102. 2017. View Article : Google Scholar

33 

Israel Y, Rachmiel A, Gourevich K and Nagler R: Kaplan-Meier analysis of salivary gland tumors: Prognosis and long-term survival. J Cancer Res Clin Oncol. 145:2123–2130. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Kim J: Drawing guideline for JKMS manuscript (01) Kaplan-Meier curve and survival analysis. J Korean Med Sci. 34:e352019. View Article : Google Scholar : PubMed/NCBI

35 

Sun CC, Zhou Q, Hu W, Li SJ, Zhang F, Chen ZL, Li G, Bi ZY, Bi YY, Gong FY, et al: Transcriptional E2F1/2/5/8 as potential targets and transcriptional E2F3/6/7 as new biomarkers for the prognosis of human lung carcinoma. Aging (Albany NY). 10:973–987. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Unberath P, Knell C, Prokosch HU and Christoph J: Developing new analysis functions for a translational research platform: Extending the cBioPortal for cancer genomics. Stud Health Technol Inform. 258:46–50. 2019.PubMed/NCBI

37 

Tarca AL, Romero R, Erez O, Gudicha DW, Than NG, Benshalom-Tirosh N, Pacora P, Hsu CD, Chaiworapongsa T, Hassan SS and Gomez-Lopez N: Maternal whole blood mRNA signatures identify women at risk of early preeclampsia: A longitudinal study. J Matern Fetal Neonatal Med. 1–12. 2020.(Online ahead of print). View Article : Google Scholar

38 

Li B and Dewey CN: RSEM: Accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 12:3232011. View Article : Google Scholar : PubMed/NCBI

39 

Selamat SA, Chung BS, Girard L, Zhang W, Zhang Y, Campan M, Siegmund KD, Koss MN, Hagen JA, Lam WL, et al: Genome-scale analysis of DNA methylation in lung adenocarcinoma and integration with mRNA expression. Genome Res. 22:1197–1211. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Bhattacharjee A, Richards WG, Staunton J, Li C, Monti S, Vasa P, Ladd C, Beheshti J, Bueno R, Gillette M, et al: Classification of human lung carcinomas by mRNA expression profiling reveals distinct adenocarcinoma subclasses. Proc Natl Acad Sci USA. 98:13790–13795. 2001. View Article : Google Scholar : PubMed/NCBI

41 

Okayama H, Kohno T, Ishii Y, Shimada Y, Shiraishi K, Iwakawa R, Furuta K, Tsuta K, Shibata T, Yamamoto S, et al: Identification of genes upregulated in ALK-positive and EGFR/KRAS/ALK-negative lung adenocarcinomas. Cancer Res. 72:100–111. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Rami-Porta R, Asamura H, Travis WD and Rusch VW: Lung cancer-major changes in the American joint committee on cancer eighth edition cancer staging manual. CA Cancer J Clin. 67:138–155. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Gurrapu S, Franzolin G, Fard D, Accardo M, Medico E, Sarotto I, Sapino A, Isella C and Tamagnone L: Reverse signaling by semaphorin 4C elicits SMAD1/5- and ID1/3-dependent invasive reprogramming in cancer cells. Sci Signal. 12:eaav20412019. View Article : Google Scholar : PubMed/NCBI

44 

Girolami I, Veronese N, Smith L, Caruso MG, Reddavide R, Leandro G, Demurtas J and Nottegar A: The activation status of the TGF-β transducer Smad2 is associated with a reduced survival in gastrointestinal cancers: A systematic review and meta-analysis. Int J Mol Sci. 20:38312019. View Article : Google Scholar

45 

Liang C, Xu J, Meng Q, Zhang B, Liu J, Hua J, Zhang Y, Shi S and Yu X: TGFB1-induced autophagy affects the pattern of pancreatic cancer progression in distinct ways depending on SMAD4 status. Autophagy. 16:486–500. 2020. View Article : Google Scholar : PubMed/NCBI

46 

Jeon HS, Dracheva T, Yang SH, Meerzaman D, Fukuoka J, Shakoori A, Shilo K, Travis WD and Jen J: SMAD6 contributes to patient survival in non-small cell lung cancer and its knockdown reestablishes TGF-beta homeostasis in lung cancer cells. Cancer Res. 68:9686–9692. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Ma L, Jiang H, Xu X, Zhang C, Niu Y, Wang Z, Tao Y, Li Y, Cai F, Zhang X, et al: Tanshinone IIA mediates SMAD7-YAP interaction to inhibit liver cancer growth by inactivating the transforming growth factor beta signaling pathway. Aging (Albany NY). 11:9719–9737. 2019. View Article : Google Scholar : PubMed/NCBI

48 

Karmokar PF, Asaduzzaman M, Islam MS, Shahriar M and Shabnaz S: The influence of SMAD1 gene polymorphisms on colorectal cancer susceptibility in Bangladeshi population: A case-control study. FEBS Open Bio. 8:3402018.

49 

Cheng M, Jiang Y, Yang H, Zhao D, Li L and Liu X: FLNA promotes chemoresistance of colorectal cancer through inducing epithelial-mesenchymal transition and smad2 signaling pathway. Am J Cancer Res. 10:403–423. 2020.PubMed/NCBI

50 

Tone AA, McConechy MK, Yang W, Ding J, Yip S, Kong E, Wong KK, Gershenson DM, Mackay H, Shah S, et al: Intratumoral heterogeneity in a minority of ovarian low-grade serous carcinomas. BMC Cancer. 14:9822014. View Article : Google Scholar : PubMed/NCBI

51 

Opyrchal M, Gil M, Salisbury JL, Goetz MP, Suman V, Degnim A, McCubrey J, Haddad T, Iankov I, Kurokawa CB, et al: Molecular targeting of the Aurora-A/SMAD5 oncogenic axis restores chemosensitivity in human breast cancer cells. Oncotarget. 8:91803–91816. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Ryu TY, Kim K, Kim SK, Oh JH, Min JK, Jung CR, Son MY, Kim DS and Cho HS: SETDB1 regulates SMAD7 expression for breast cancer metastasis. BMB Rep. 52:139–144. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Gao YQ, Liu M and Zhang H: Expression profiles of Smad1 protein in lung cancer tissues and normal tissues and its effect on lung cancer incidence. J Biol Regul Homeost Agents. 30:165–171. 2016.PubMed/NCBI

54 

Chae DK, Ban E, Yoo YS, Kim EE, Baik JH and Song EJ: MIR-27a regulates the TGF-β signaling pathway by targeting SMAD2 and SMAD4 in lung cancer. Mol Carcinog. 56:1992–1998. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Sun W, Ma Y, Chen P and Wang D: MicroRNA-10a silencing reverses cisplatin resistance in the A549/cisplatin human lung cancer cell line via the transforming growth factor-β/Smad2/STAT3/STAT5 pathway. Mol Med Rep. 11:3854–3859. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Wang Z, Lu Y, Sheng B, Ding Y and Cheng X: Catalpol inhibits TGF-β1-induced epithelial-mesenchymal transition in human non-small-cell lung cancer cells through the inactivation of Smad2/3 and NF-κB signaling pathways. J Cell Biochem. Sep 11–2018.(Online ahead of print).

57 

Wei Y, Li D, Wang D, Qiu T and Liu K: WITHDRAWN: Evaluation of microRNA-203 in bone metastasis of patients with non-small cell lung cancer through TGF-β/SMAD2 expression. Oncol Rep. Sep 21–2017.(Online ahead of print). View Article : Google Scholar

58 

Zhang JX, Zhai JF, Yang XT and Wang J: MicroRNA-132 inhibits migration, invasion and epithelial-mesenchymal transition by regulating TGFβ1/Smad2 in human non-small cell lung cancer. Eur Rev Med Pharmacol Sci. 20:3793–3801. 2016.PubMed/NCBI

59 

Chen Y, Xing P, Chen Y, Zou L, Zhang Y, Li F and Lu X: High p-Smad2 expression in stromal fibroblasts predicts poor survival in patients with clinical stage I to IIIA non-small cell lung cancer. World J Surg Oncol. 12:3282014. View Article : Google Scholar : PubMed/NCBI

60 

Qian Z, Zhang QK, Hu Y, Zhang T, Li J, Liu Z, Zheng H, Gao Y, Jia W, Hu A, et al: Investigating the mechanism by which SMAD3 induces PAX6 transcription to promote the development of non-small cell lung cancer. Respir Res. 19:2622018. View Article : Google Scholar : PubMed/NCBI

61 

Ziemke M, Patil T, Nolan K, Tippimanchai D and Malkoski SP: Reduced Smad4 expression and DNA topoisomerase inhibitor chemosensitivity in non-small cell lung cancer. Lung Cancer. 109:28–35. 2017. View Article : Google Scholar : PubMed/NCBI

62 

Chen H, Wang JW, Liu LX, Yan JD, Ren SH, Li Y and Lu Z: Expression and significance of transforming growth factor-β receptor type II and DPC4/Smad4 in non-small cell lung cancer. Exp Ther Med. 9:227–231. 2015. View Article : Google Scholar : PubMed/NCBI

63 

D'Haene N, Le Mercier M, Salmon I, Mekinda Z, Remmelink M and Berghmans T: SMAD4 mutation in small cell transformation of epidermal growth factor receptor mutated lung adenocarcinoma. Oncologist. 24:9–13. 2019. View Article : Google Scholar : PubMed/NCBI

64 

Haeger SM, Thompson JJ, Kalra S, Cleaver TG, Merrick D, Wang XJ and Malkoski SP: Smad4 loss promotes lung cancer formation but increases sensitivity to DNA topoisomerase inhibitors. Oncogene. 35:577–586. 2016. View Article : Google Scholar : PubMed/NCBI

65 

Zeng YY, Zhu JJ, Shen D, Qin H, Lei Z, Li W, Liu Z and Huang JA: MicroRNA-205 targets SMAD4 in non-small cell lung cancer and promotes lung cancer cell growth in vitro and in vivo. Oncotarget. 8:30817–30829. 2017. View Article : Google Scholar : PubMed/NCBI

66 

Takahashi K, Nishikawa S, Miyata R, Noguchi M, Ishikawa H, Yutaka Y, Nakajima D, Hamaji M, Ohsumi A, Menju T, et al: Tranilast inhibits TGF-beta-induced EMT and invasion/metastasis via the suppression of smad4 in lung cancer cell lines. Ann Oncol 29:. (Suppl 8):viii1–viii13. 2018.

67 

Lee CC, Yang WH, Li CH, Cheng YW, Tsai CH and Kang JJ: Ligand independent aryl hydrocarbon receptor inhibits lung cancer cell invasion by degradation of Smad4. Cancer Lett. 376:211–217. 2016. View Article : Google Scholar : PubMed/NCBI

68 

Yanagisawa K, Uchida K, Nagatake M, Masuda A, Sugiyama M, Saito T, Yamaki K, Takahashi T and Osada H: Heterogeneities in the biological and biochemical functions of Smad2 and Smad4 mutants naturally occurring in human lung cancers. Oncogene. 19:2305–2311. 2000. View Article : Google Scholar : PubMed/NCBI

69 

Zhang Z, Wang J, Zeng X, Li D, Ding M, Guan R, Yuan L, Zhou Q, Guo M, Xiong M, et al: Two-stage study of lung cancer risk modification by a functional variant in the 3′-untranslated region of SMAD5 based on the bone morphogenetic protein pathway. Mol Clin Oncol. 8:38–46. 2018.PubMed/NCBI

70 

Ngeow J, Yu W, Yehia L, Niazi F, Chen J, Tang X, Heald B, Lei J, Romigh T, Tucker-Kellogg L, et al: Exome sequencing reveals germline SMAD9 mutation that reduces phosphatase and tensin homolog expression and is associated with hamartomatous polyposis and gastrointestinal ganglioneuromas. Gastroenterology. 149:886–889 e5. 2015. View Article : Google Scholar : PubMed/NCBI

71 

Zhang Q, Gan H, Song W, Chai D and Wu S: MicroRNA-145 promotes esophageal cancer cells proliferation and metastasis by targeting SMAD5. Scand J Gastroenterol. 53:769–776. 2018. View Article : Google Scholar : PubMed/NCBI

72 

Shi JQ, Wang B, Cao XQ, Wang YX, Cheng X, Jia CL, Wen T, Luo BJ and Liu ZD: Circular RNA_LARP4 inhibits the progression of non-small-cell lung cancer by regulating the expression of SMAD7. Eur Rev Med Pharmacol Sci. 24:1863–1869. 2020.PubMed/NCBI

73 

Jin L, Zhu C, Wang X, Li C, Cao C, Yuan J and Li S: Urocortin attenuates TGFβ1-induced Snail1 and slug expressions: Inhibitory role of Smad7 in Smad2/3 signaling in breast cancer cells. J Cell Biochem. 116:2494–2503. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2020
Volume 20 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Pan S, Zhou G, Hu W and Pei H: SMAD‑6, ‑7 and ‑9 are potential molecular biomarkers for the prognosis in human lung cancer. Oncol Lett 20: 2633-2644, 2020
APA
Pan, S., Zhou, G., Hu, W., & Pei, H. (2020). SMAD‑6, ‑7 and ‑9 are potential molecular biomarkers for the prognosis in human lung cancer. Oncology Letters, 20, 2633-2644. https://doi.org/10.3892/ol.2020.11851
MLA
Pan, S., Zhou, G., Hu, W., Pei, H."SMAD‑6, ‑7 and ‑9 are potential molecular biomarkers for the prognosis in human lung cancer". Oncology Letters 20.3 (2020): 2633-2644.
Chicago
Pan, S., Zhou, G., Hu, W., Pei, H."SMAD‑6, ‑7 and ‑9 are potential molecular biomarkers for the prognosis in human lung cancer". Oncology Letters 20, no. 3 (2020): 2633-2644. https://doi.org/10.3892/ol.2020.11851