Open Access

The prognostic value of ADRA1 subfamily genes in gastric carcinoma

  • Authors:
    • Tingan Wang
    • Yuzhou Qin
    • Hao Lai
    • Weiyuan Wei
    • Zhao Li
    • Yang Yang
    • Mingwei Huang
    • Jiansi Chen
  • View Affiliations

  • Published online on: July 24, 2019     https://doi.org/10.3892/ol.2019.10660
  • Pages: 3150-3158
  • Copyright: © Wang 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

Adrenergic receptor α1 (ADRA1) subfamily members, including ADRA1A, ADRA1B and ADRA1D, are understood to participate in cardiac disease and benign prostatic hyperplasia. In addition, adrenergic signals in cell pathways can promote the development of cancer. However, little is understood regarding the associations between ADRA1 subfamily members and gastric carcinoma (GC). The present study investigated the prognostic value of the ADRA1 subfamily genes in GC. Data from a total of 379 patients with GC were obtained from The Cancer Genome Atlas and Gene Expression Omnibus databases. Kaplan‑Meier analysis and Cox regression analysis were used to determine associations with overall survival (OS) and to evaluate the median survival time using hazard ratios (HRs) and 95% confidence intervals (CIs). Multivariate survival analysis revealed that low expression levels of ADRA1A (HR, 0.595; 95% CI, 0.426‑0.831; adjusted P=0.002) ADRA1B (HR, 0.576; 95% CI, 0.412‑0.805; adjusted P=0.001) and ADRA1D (HR, 0.559; 95% CI, 0.398‑0.787; adjusted P=0.001) were associated with a favourable OS. Joint‑effects analysis demonstrated that combinations of low expression levels of ARDA1A, ARDA1B and ARDA1D were significantly associated with a favourable OS. Overall, the current results suggested that the mRNA expression levels of ARDA1 subfamily members may serve as potential prognostic markers for GC.

Introduction

Gastric carcinoma (GC) is one of the most common types of malignancy and is the second leading cause of cancer-related mortality worldwide (1). Despite advances in surgical techniques, adjuvant chemotherapy, radiotherapy and immune therapy, the morbidity and mortality rates remain high (2). Therefore, new strategies to improve the diagnosis and prognosis of GC are urgently required.

Tumour metastasis is a complex process that serves a vital role in the progression and outcome of the cancer (3). Therefore, novel effective strategies that target metastasis are required. Previous studies have suggested that adrenergic receptor antagonists could be used in novel therapeutic approaches in the treatment of various types of cancer, including prostate and breast carcinoma (4,5). Tumour metastasis involves the migration of cancer cells from the primary tumour via lymphoid/hematopoietic pathways and is regulated by exogenous signalling molecules, including G protein coupled receptor (GPCR) ligands, chemokines and neurotransmitters (6,7). A previous study has reported that norepinephrine, a stress-associated neurotransmitter, is a potent inducer of migration in cancer cell lines (8).

Adrenergic receptor antagonists include two major groups; α receptor and β receptor specific antagonists (9,10). Adrenergic receptor α1 (ADRA1) receptor is classified into three different subtypes ADRA1A, ADRA1B and ADRA1D, that differ in their tissue distribution, cell signalling, pharmacology and physiological roles (11). ADRA1 receptors are ubiquitous in the majority of human tissues; ADRA1B is predominantly expressed on the cell surface, ADRA1A is mainly expressed on the cell surface and intracellularly, and ADRA1D is primarily localized perinuclear (12).

It is well-known that ADRA1 is a member of the GPCR family, which interacts with a heterotrimeric G protein containing the Gαq/11/14/16 subunits (13). The Gαq subunit is the main activator of phospholipase CE, which promotes the cleavage of phosphatidylinositol-4,5-diphosphate into diacylglycerol and inositol-1,4,5-triphosphate. This subsequently promotes the release of Ca2+ from intracellular stores to activate protein kinase C (PKC) (14).

Previous studies have suggested that adrenergic signals in cells can promote the development of cancer (15,16). However, little is understood regarding the associations between the expression levels of ARDA1A, ARDA1B and ARDA1D and the risk of GC. Therefore, the aim of the present study was to identify the associations between the expression levels of individual ADRA1 subtypes and GC prognosis. The current study may provide insights into the potential functional roles of ADRA1 subtypes in GC.

Materials and methods

Patient and disease characteristics

Metabolic gEne Rapid Visualizer (http://merav.wi.mit.edu/) was used to generate boxplots of the expression levels of ADRA1 subtypes in normal gastric tissues and primary GC tumours. Subsequently, clinical data were obtained for 379 patients with GC, including sex, grade (17), age, Tumour-Node-Metastasis (TNM) stage (18), targeted therapy, events (metastases or deaths), survival time, mortality status and mRNA expression levels of ADRA1A, ADRA1B and ADRA1D, from OncoLnc (http://www.oncolnc.org/) and Gene Expression Omnibus (GEO; http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE14520) databases (19,20). The data presented here are based on studies published on The Cancer Genome Atlas (TCGA) (21,22). To analyse the difference in the expression levels in cancer compared with the adjacent tissues, three genes (ADRA1A, ADRA1B and ADRA1D0 were searched in the GEPIA (http://gepia.cancer-pku.cn/) database separately.

Correlation and functional enrichment analysis of the ADRA1 subfamily

The online Database for Annotation, Visualization, and Integrated Discovery (DAVID; v.6.8; http://david.ncifcrf.gov/tools.jsp) (23) was used to perform functional enrichment. This included gene ontology (GO) functional analysis and Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway analysis. In the present study, GO functional analysis consisted of molecular function, cellular component, and biological process terms. KEGG pathway analysis was performed on the ADRA1 gene subfamily. The gene function prediction website GeneMANIA (http://genemania.org/) was used to construct gene-gene interaction networks (24).

Survival analysis

Using the TCGA data, 379 patients with GC were divided into high- and low-expression groups according to the 50% cut-off values (median values). Overall survival (OS) and median survival time (MST) were applied to estimate patient prognosis. Kaplan-Meier analysis with a log-rank test was used to identify associations between ADRA1A, ADRA1B and ADRA1D mRNA expression levels and patient survival. Cox regression analysis was then used to evaluate statistically significant factors, including age and TNM stage.

Joint-effects analysis

In the TCGA database, the expression levels of ADRA1A, ADRA1B and ADRA1D were significantly different between tumour and non-tumour tissues. Therefore, joint-effects analysis was performed with the following combinations: i) ADRA1A and ADRA1B; i) ADRA1A and ADRA1D; iii) ADRA1B and ADRA1D; and iv) ADRA1A, ADRA1B and ADRA1D (Table I). A Cox regression model was adjusted for TNM stage, age and sex in keeping with the aforementioned combinations.

Table I.

Grouping according to the expression levels of two or three selected genes.

Table I.

Grouping according to the expression levels of two or three selected genes.

GroupComposition
ILow ARDA1A + low ARDA1B
IILow ARDA1A + high ARDA1B
High ARDA1A + low ARDA1B
IIIHigh ARDA1A + high ARDA1B
IVLow ARDA1A + low ARDA1D
VLow ARDA1A + high ARDA1D
High ARDA1A + low ARDA1D
VIHigh ARDA1A + high ARDA1D
VIILow ARDA1B + low ARDA1D
VIIILow ARDA1B + high ARDA1D
High ARDA1B + low ARDA1D
IXHigh ARDA1B + high ARDA1D
XLow ARDA1A + low ARDA1B + low ARDA1D
XIHigh ARDA1A + low ARDA1B + high ARDA1D
Low ARDA1A+ high ARDA1B + high ARDA1D
Low ARDA1A + low ARDA1B + low ARDA1D
High ARDA1A + high ARDA1B + high ARDA1D
High ARDA1A + low ARDA1B + low ARDA1D
Low ARDA1A + high ARDA1B + low ARDA1D
XIIHigh ARDA1A + high ARDA1B + high ARDA1D

[i] ADRA1, adrenergic receptor α1.

Statistical analysis

OS was evaluated by Kaplan-Meier survival analysis followed by a log-rank test. Cox proportional hazards regression model was used for univariate and multivariate survival analysis. GraphPad Prism v.7.0 (GraphPad Prism, Inc.) was used to construct vertical scatter plots and survival curves. All statistical analyses were performed using SPSS v.22.0 software and compared using an independent-samples t-test (IBM, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

Clinicopathological features of the patient cohort

Detailed characteristics of the 379 included patients obtained from the TCGA database are presented in Table II. It was identified that TNM stage and age were significantly associated with MST (P<0.001 and P=0.007, respectively); however, no significant associations were revealed for grade, sex or targeted therapy (all P>0.05; Table II).

Table II.

Clinical data for the 379 patients with gastric carcinoma.

Table II.

Clinical data for the 379 patients with gastric carcinoma.

VariablePatients (n=379)No. of events (%)MST, daysHR (95% CI)Log-rank P-value
Grade
  1 + 214652 (35.6)1,294Ref.0.07
  323399 (42.5)8011.333 (0.977–1.818)
Sex
  Male247104 (42.1)2,030Ref.0.229
  Female13247 (35.6)8741.236 (0.875–1.745)
Age, years
  ≥6012438 (30.6)475Ref.0.007
  <60255113 (44.3)7920.604 (0.418–0.874)
TNM stage
  I5012 (24.0)2,1970.270 (0.133–0.550)<0.001
  II11933 (27.7)1,6860.372 (0.214–0.645)
  III16478 (47.6)7660.649 (0.400–1.053)
  IV3421 (61.8)476Ref.
  NA12
Targeted therapy
  No18879 (42.0)805Ref.0.063
  Yes16259 (38.4)1,2940.726 (0.518–1.018)
  NA29

[i] MST, median survival time; HR, hazard ratio; CI, confidence interval; Ref, reference group; NA, not available; TNM, Tumor-Node-Metastasis.

Analysis of ADRA1 subfamily gene expression in GC

Expression levels of ADRA1A, ADRA1B and ADRA1D in primary GC tissues and normal tissues were analysed using the online tool (GEPIA; http://gepia.cancer-pku.cn/) and illustrated by boxplots (Fig. 1A-C). The expression levels of ADRA1A were higher in normal gastric tissues compared with primary gastric tumours. However, no marked differences in the expression levels of ADRA1B and ADRA1D were revealed between primary GC tumours and normal tissues. The low expression groups and high expression groups for these genes obtained from the GEO and TCGA databases were analysed by Scatter diagrams and compared using an independent-samples t-test (Fig. 1D).

GO and KEGG pathway analysis of the ADRA1 subfamily

The results of GO analysis of ADRA1A, ADRA1B and ADRA1D including molecular function, cellular component and biological process terms, are presented in Fig. 2B. KEGG enrichment analysis identified certain signalling pathways associated with ADRA1 subfamily members, such as ‘Calcium signalling pathway’, ‘cGMP-PKG signalling pathway’ and mitogen-activated protein kinase (MAPK) signalling (Fig. 2B). Furthermore, the GeneMANIA website (http://genemania.org/) was used to analyse interaction networks of ADRA1A, ADRA1B and ADRA1D. The interaction network between the ADRA1 subfamily and other genes presented in Fig. 2A.

Association of ADRA1 subfamily members with survival

Cox proportional hazards regression analysis of the ADRA1 members revealed that the mRNA expression levels of all three genes were significantly associated with MST in patients with GC (Table III). As aforementioned in Table I, TNM stage and age were also identified as prognostic factors in TCGA GC cohort. Therefore, a multivariate Cox regression model was employed next, in order to explore whether the expression levels of the ADRA1 subfamily genes were independent prognostic factors following adjustment for age and TNM stage. The results revealed that the lower expression levels of ADRA1A [hazard ratio (HR), 0.595; 95% CI, 0.426–0.831; adjusted P=0.002], ADRA1B (HR, 0.576; 95% CI, 0.412–0.805; adjusted P=0.001) and ADRA1D (HR, 0.559; 95% CI, 0.398–0.787; adjusted P=0.001) were significantly associated with favourable MST in patients with GC, following adjustment for age and TNM stage (Table III). The survival curves of ADRA1A, ADRA1B and ADRA1D are presented in Fig. 2C-E; these revealed that low expression levels of these genes were significantly associated with a favourable OS in patients with GC (P=0.002, P=0.006 and P=0.002, respectively).

Table III.

Survival analysis according to the expression levels of ADRA1 subfamily genes.

Table III.

Survival analysis according to the expression levels of ADRA1 subfamily genes.

GenePatients (n=379)No. of events (%)MST, daysCrude HR (95% CI)Crude P-valueAdjusted HR (95% CI)aAdjusted P-valuea
ADRA1A
  Low18961 (32.1)1,686Ref.0.001Ref.0.002
  High18990 (47.6)6690.601 (0.434–0.834) 0.595 (0.426–0.831)
  NA1
ADRA1B
  Low18962 (32.9)1,407Ref.0.006Ref.0.001
  High18989 (47.1)7120.637 (0.460–0.881) 0.0576 (0.412–0.805)
  NA1
ADRA1D
  Low18958 (30.7)1,407Ref.0.002Ref.0.001
  High18993 (49.2)6750.601 (0.433–0.835) 0.559 (0.398–0.787)
  NA1

a Adjusted for age and Tumour-Node-Metastasis stage. ADRA1, adrenergic receptor α1; MST, median survival time; HR, hazard ratio; CI, confidence interval; Ref, reference group; NA, not available.

Joint-effects analysis of the ADRA1 subfamily

Based on the influence of ADRA1A, ADRA1B and ADRA1D on patient survival identified by multivariate analysis, a joint-effects model was employed to evaluate the combined effects of these genes on the OS of patients with GC. According to the different expression levels of ADRA1A, ADRA1B and ADRA1D, 12 groups were generated, as presented in Table I. Group XII, with high expression levels of ARDA1A, ARDA1B and ARDA1D, exhibited the shortest MST of 552 days (adjusted P=0.001). Group X, with low expression levels of ARDA1A, ARDA1B and ARDA1D, exhibited the highest MST of 2,100 days (adjusted P=0.002). The detailed joint-effects analysis results are presented in Table IV.

Table IV.

Joint-effects analysis of the prognostic value of combinations of ADRA1A, ADRA1B and ADRA1D expression levels in gastric carcinoma.

Table IV.

Joint-effects analysis of the prognostic value of combinations of ADRA1A, ADRA1B and ADRA1D expression levels in gastric carcinoma.

GroupPatients (n=379)MST, daysCrude P-valueCrude HR (95% CI)Adjusted P-valueaAdjusted HR (95% CI)a
I1481,407<0.0010.480 (0.327–0.705)<0.0010.444 (0.297–0.664)
II1142,1000.0050.552 (0.364–0.838)0.0040.531 (0.347–0.813)
III113560<0.001Ref.<0.001Ref.
IV1151,686<0.0010.420 (0.271–0.651)<0.0010.406 (0.260–0.633)
V1471,2940.0830.729 (0.510–1.043)0.0310.665 (0.458–0.963)
VI1146330.001Ref.<0.001Ref.
VII1211,407<0.0010.481 (0.319–0.725)<0.0010.433 (0.285–0.659)
VIII1349400.0660.708 (0.490–1.023)0.0030.563 (0.385–0.825)
IX1215620.002Ref.<0.001Ref.
X832,1000.0050.490 (0.300–0.802)0.0020.457 (0.279–0.749)
XI2151,2940.0100.625 (0.438–0.893)0.0010.541 (0.376–0.780)
XII785520.006Ref.0.001Ref.

a Adjusted for age and Tumour-Node-Metastasis stage. MST, median survival time; HR, hazard ratio; CI, confidence interval; ADRA1, adrenergic receptor α1; Ref, reference group.

Associated survival curves, presented in Fig. 3, were evaluated by Kaplan-Meier analysis with a log-rank test. Low expression levels of ADRA1A, ADRA1B and ADRA1D (group X) were identified to be significantly associated with favourable OS. By contrast, high expression levels of ADRA1A, ADRA1B and ADRA1D (group XII) were revealed to be significantly associated with poor OS (all P<0.05; Fig. 3).

Discussion

Cancer metastasis can be influenced by numerous factors, including intracellular signalling molecules and extracellular components, including cytokines, the extracellular matrix and neurotransmitters (25). It has previously been suggested that the nervous system serves an important role in the progression of cancer. It has been reported that the sympathetic and parasympathetic nervous systems participate in the development and distribution of cancer (26).

Adrenergic neurotransmitters, including norepinephrine (NE) and epinephrine (E), have been reported to promote cell migration and invasion in various types of cancer (6,27). Previous studies have demonstrated that NE/E may promote tumour progression by promoting angiogenesis (27). Furthermore, the tumour stromal cells in the microenvironment of cancer may be affected by the nervous system (28). A number of neurotransmitter receptor-associated genes have been identified to be closely associated with the proliferation and survival of tumour cells by a large-scale complementary DNA transfection screening (29).

It is understood that NE/E exert their physiological functions predominantly by the α and β adrenergic receptors (30). The ability of NE/E to induce invasion and anoikis resistance of cancer cells can be mediated by both ADRA1A and ADRB2 receptors (31). ADRA1 mediates the role of endogenous adrenaline and NE in multiple target cells. NE induces proton release and calcium flux, and activates phospholipase C, PKC and extracellular signal-regulated kinase 1/2 pathways to promote cell proliferation (32). In addition, ADRs transactivate epidermal growth factor receptor (EGFR) signalling via specific matrix metalloproteinases (MMPs) or a disintegrin and metalloproteinase hydrolysis of latent ligands, including heparin-binding EGF-like growth factor (HB-EGF) (33). For the first time, Li et al (30) demonstrated that the ADRA1A and ADRB2 can inhibit EGFR signalling in cancer. EGFR is overexpressed in the majority of adenocarcinoma and squamous cell carcinoma cases and can be selectively targeted by pharmacological inhibitors currently used in the clinic (34).

It has been reported that ADRA1 can promote the metastasis of cancer, and continuously activated ADRA induces cell apoptosis via p53 (35). The tumour suppressor protein p53 serves an important role in cellular regulation and acts as an important mediator of apoptotic cell death (36). Evidence suggests that p53 not only induces transcription of pro-apoptotic proteins but also activates the mitochondrial cell death pathway (37). Previous studies have demonstrated that ADRA1A is also associated with reactive oxygen species (ROS) production via the EGFR and the nicotinamide adenine dinucleotide phosphate oxidase signalling pathways (38).

However, little is understood regarding the association between ADRA1 mRNA expression and the prognosis of GC. Using data from TCGA and GEO databases that included mRNA expression profiles of the ADRA1 genes and the associated clinical information from patients with GC, the present study investigated the associations between ADRA1 family members expression and patient prognosis. In addition, the current study assessed whether any ADRA1 genes, alone or in combination, could be used as biomarkers for predicting the prognosis of GC. The results suggested that the expression levels of ADRA1A in normal tissue were higher compared with that in primary tumour tissue. In addition, survival analysis demonstrated that low expression levels of ADRA1A, ADRA1B or ADRA1D were associated with a favourable OS in patients with GC. Furthermore, joint-effects analysis demonstrated that the combination of low levels of all three ADRA1A, ADRA1B and ADRA1D was significantly associated with a favourable OS. By contrast, the combination of high expression levels of ADRA1A, ADRA1B and ADRA1D was associated with a poor OS.

Furthermore, functional analysis and KEGG enrichment identified specific signalling pathways associated with ADRA1 genes including ‘Calcium signalling pathway’, ‘cGMP-PKG signalling pathway’ and mitogen-activated protein kinase (MAPK) signalling. These pathways serve important roles in cancer. For example, studies have demonstrated that numerous biological functions are regulated by MAPK signalling, including cell proliferation, apoptosis and metastasis (39,40). In addition, cGMP/PKGIβ regulates breast cancer cell migration and invasion through the actin/myosin-associated protein, caldesmon (CaD) (41). Analysis of gene-gene interactions showed that certain genes were associated with members of the ADRA1 subfamily [G protein-coupled receptor kinases (GRK)1, GRK4, GRK5, GRK6, GRK7, ARRB1, DRD4, GNAQ, ADRBK2] and some of these genes serve important roles in the regulation of tumour biology. For example, GRKs can modulate GPCR signalling by interacting with the ligand-activated GPCR and phosphorylating its intracellular domain (42). It has been demonstrated that GPCRs affect multiple aspects of cancer biology, such as vascular remodelling, invasion and migration (42).

Peng et al (43) reported that ADRA1A was highly expressed in the peripheral serum of patients with hysterocarcinoma and associated with tumour stage and lymph node metastasis status. Powe et al (44) demonstrated that ADRA1B expression was associated with breast cancer progression and prognosis. Notably, the expression levels of ADRA1A were higher in normal gastric tissues compared with primary gastric tumour tissues in the current study. However, the present study also revealed that low levels of ADRA1 were favourable for patient outcome. There may be a number of reasons for these contradictory results. Firstly, it is understood that certain genes act as tumour suppressors during the early stage of tumorigenesis, while they serve as tumour promoters in later stages. For example, transforming growth factor-β (TGF-β) is a cytokine that exhibits dual activities in numerous types of cancer (45). TGF-β is an important mediator of cancer invasion, metastasis and angiogenesis; however, it also exhibits antitumor functions (46). Another possible explanation may be that ADRA1 serves different roles in gastric tumour tissue and in normal tissue. Despite the low expression of ADRA1 in gastric cancer tumour tissues, it also serves a role in promoting cancer cell proliferation and metastasis (47). Therefore, the present results require further experimental validation in order to fully elucidate the role of ADRA1 in GC.

In conclusion, the results of the present study demonstrated that the expression levels of ADRA1A, ADRA1B and ADRA1D were associated with the prognosis of gastric cancer. High expression levels of the ADRA1 family were associated with a poor prognosis and a significant reduction in survival rate. These results suggested that an increased expression of the ADRA1 family may somehow promote tumour metastasis. Accordingly, the levels of ADRA1 genes may serve as potential markers for prognosis evaluation.

The present study has certain limitations. Firstly, clinical information and gene expression data used in the study were obtained from a public database and the data were limited. Second, the low expression of ADRA1 in gastric tumour tissues and its role in predicting prognosis is difficult to understand. Due to insufficient funds, the present study did not perform verification experiments. Therefore, the specific mechanism of ADRA1 in gastric cancer tissues requires further experimental validation. Despite these limitations, to the best of our knowledge, the current study is the first to systematically investigate the diagnostic and prognostic values of ADRA1 genes in GC. In summary, ADRA1 genes may serve as potential novel indicators for the prediction of the prognosis of patients with GC.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81760521).

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the author on reasonable request.

Authors' contributions

JC, TW and MH designed the study, YY and ZL performed the analysis and prepared the manuscript. TW wrote the manuscript. WW performed the analysis. YQ and HL helped perform the analysis with constructive discussions. All authors approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Van Cutsem E, Sagaert X, Topal B, Haustermans K and Prenen H: Gastric cancer. Lancet. 388:2654–2664. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2015. CA Cancer J Clin. 65:5–29. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Ye J, Xu J, Li Y, Huang Q, Huang J, Wang J, Zhong W and Lin X, Chen W and Lin X: DDAH1 mediates gastric cancer cell invasion and metastasis via Wnt/β-catenin signaling pathway. Mol Oncol. 11:1208–1224. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Harris AM, Warner BW, Wilson JM, Becker A, Rowland RG, Conner W, Lane M, Kimbler K, Durbin EB, Baron AT and Kyprianou N: Effect of alpha1-adrenoceptor antagonist exposure on prostate cancer incidence: An observational cohort study. J Urol. 178:2176–2180. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Algazi M, Plu-Bureau G, Flahault A, Dondon MG and Le MG: Is beta-blocker treatment associated with a decrease in the risk of cancer. Lett Drug Des Discov. 3:653–661. 2006. View Article : Google Scholar

6 

Entschladen F, Drell TL IV, Lang K, Joseph J and Zaenker KS: Tumour-cell migration, invasion, and metastasis: Navigation by neurotransmitters. Lancet Oncol. 5:254–258. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Entschladen F, Drell TL IV, Lang K, Joseph J and Zaenker KS: Neurotransmitters and chemokines regulate tumor cell migration: Potential for a new pharmacological approach to inhibit invasion and metastasis development. Curr Pharm Des. 11:403–411. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Lang K, Drell TL IV, Lindecke A, Niggemann B, Kaltschmidt C, Zaenker KS and Entschladen F: Induction of a metastatogenic tumor cell type by neurotransmitters and its pharmacological inhibition by established drugs. Int J Cancer. 112:231–238. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Dorn GW II and Liggett SB: Pharmacogenomics of beta-adrenergic receptors and their accessory signaling proteins in heart failure. Clin Transl Sci. 1:255–262. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Rosini M, Bolognesi ML, Giardina D, Minarini A, Tumiatti V and Melchiorre C: Recent advances in alpha1-adrenoreceptor antagonists as pharmacological tools and therapeutic agents. Curr Top Med Chem. 7:147–162. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Hein P and Michel MC: Signal transduction and regulation: Are all alpha1-adrenergic receptor subtypes created equal? Biochem Pharmacol. 73:1097–1106. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Docherty JR: Subtypes of functional alpha1-adrenoceptor. Cell Mol Life Sci. 67:405–417. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Hubbard KB and Hepler JR: Cell signalling diversity of the Gqalpha family of heterotrimeric G proteins. Cell Signal. 18:135–150. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Theroux TL, Esbenshade TA, Peavy RD and Minneman KP: Coupling efficiencies of human alpha 1-adrenergic receptor subtypes: Titration of receptor density and responsiveness with inducible and repressible expression vectors. Mol Pharmacol. 50:1376–1387. 1996.PubMed/NCBI

15 

Fitzgerald PJ: Is norepinephrine an etiological factor in some types of cancer? Int J Cancer. 124:257–263. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Adissu HA and Schuller HM: Antagonistic growth regulation of cell lines derived from human lung adenocarcinomas of Clara cell and aveolar type II cell lineage: Implications for chemoprevention. Int J Oncol. 24:1467–1472. 2004.PubMed/NCBI

17 

Gobbi PG, Villano L, Pozzoli D, Bergonzi M, Vanoli A, Tava F, Dionigi P and Corazza GR: Improving the AJCC/TNM classification for use in early gastric cancer. J Gastrointest Surg. 15:935–941. 2011. View Article : Google Scholar : PubMed/NCBI

18 

In H, Solsky I, Palis B, Langdon-Embry M, Ajani J and Sano T: Validation of the 8th edition of the AJCC TNM staging system for gastric cancer using the national cancer database. Ann Surg Oncol. 24:3683–3691. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Roessler S, Jia HL, Budhu A, Forgues M, Ye QH, Lee JS, Thorgeirsson SS, Sun Z, Tang ZY, Qin LX and Wang XW: A unique metastasis gene signature enables prediction of tumor relapse in early-stage hepatocellular carcinoma patients. Cancer Res. 70:10202–10212. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Roessler S, Long EL, Budhu A, Chen Y, Zhao X, Ji J, Walker R, Jia HL, Ye QH, Qin LX, et al: Integrative genomic identification of genes on 8p associated with hepatocellular carcinoma progression and patient survival. Gastroenterology. 142:957–966.e12. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Shaul YD, Yuan B, Thiru P, Nutter-Upham A, McCallum S, Lanzkron C, Bell GW and Sabatini DM: MERAV: A tool for comparing gene expression across human tissues and cell types. Nucleic Acids Res. 44(D1): D560–D566. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Anaya J: OncoLnc: Linking TCGA survival data to mRNAs, miRNAs, and lncRNAs. PeerJ Comp Sci. 2:e672016. View Article : Google Scholar

23 

Huang da W, Sherman BT and Lempicki RA: Bioinformatics enrichment tools: Paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37:1–13. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Warde-Farley D, Donaldson SL, Comes O, Zuberi K, Badrawi R, Chao P, Franz M, Grouios C, Kazi F, Lopes CT, et al: The GeneMANIA prediction server: Biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res. 38:W214–W220. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Mínguez B, Hoshida Y, Villanueva A, Toffanin S, Cabellos L, Thung S, Mandeli J, Sia D, April C, Fan JB, et al: Gene-expression signature of vascular invasion in hepatocellular carcinoma. J Hepatol. 55:1325–1331. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Magnon C, Hall SJ, Lin J, Xue X, Gerber L, Freedland SJ and Frenette PS: Autonomic nerve development contributes to prostate cancer progression. Science. 341:12363612013. View Article : Google Scholar : PubMed/NCBI

27 

Schuller HM: Neurotransmitter receptor-mediated signaling pathways as modulators of carcinogenesis. Prog Exp Tumor Res. 39:45–63. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Thaker PH, Han LY, Kamat AA, Arevalo JM, Takahashi R, Lu C, Jennings NB, Armaiz-Pena G, Bankson JA, Ravoori M, et al: Chronic stress promotes tumor growth and angiogenesis in a mouse model of ovarian carcinoma. Nat Med. 12:939–944. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Wan D, Gong Y, Qin W, Zhang P, Li J, Wei L, Zhou X, Li H, Qiu X, Zhong F, et al: Large-scale cDNA transfection screening for genes related to cancer development and progression. Proc Natl Acad Sci USA. 101:15724–15729. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Li J, Yang XM, Wang YH, Feng MX, Liu XJ, Zhang YL, Huang S, Wu Z, Xue F, Qin WX, et al: Monoamine oxidase A suppresses hepatocellular carcinoma metastasis by inhibiting the adrenergic system and its transactivation of EGFR signaling. J Hepatol. 60:1225–1234. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Perez DM: The adrenergic receptors: In the 21st century. NewYork: Humana Press; 2006

32 

Liu W, Wang X, Mei Z, Gong J, Huang L, Gao X, Zhao Y, Ma J and Qian L: BNIP3L promotes cardiac fibrosis in cardiac fibroblasts through [Ca2+]i-TGF-β-Smad2/3 pathway. Sci Rep. 7:19062017. View Article : Google Scholar : PubMed/NCBI

33 

Oganesian A, Yarov-Yarovoy V, Parks WC and Schwinn DA: Constitutive coupling of a naturally occurring human alpha1a- adrenergic receptor genetic variant to EGFR transactivation pathway. Proc Natl Acad Sci USA. 108:19796–19801. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Salazar R and Ciardiello F: Optimizing anti-EGFR therapy in colorectal cancer. Clin Cancer Res. 21:5415–5416. 2015. View Article : Google Scholar : PubMed/NCBI

35 

García-Cazarín ML, Smith JL, Clair DK and Piascik MT: The alpha1D-adrenergic receptor induces vascular smooth muscle apoptosis via a p53-dependent mechanism. Mol Pharmacol. 74:1000–1007. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Okada H and Mak TW: Pathways of apoptotic and non-apoptotic death in tumour cells. Nat Rev Cancer. 4:592–603. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Liu B, Chen Y and St Clair DK: Ros and p53: A versatile partnership. Free Radic Biol Med. 44:1529–1535. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Xiao L, Pimentel DR, Wang J, Singh K, Colucci WS and Sawyer DB: Role of reactive oxygen species and NAD(P)H oxidase in alpha(1)-adrenoceptor signaling in adult rat cardiac myocytes. Am J Physiol Cell Physiol. 282:C926–C934. 2002. View Article : Google Scholar : PubMed/NCBI

39 

Yoda Y, Takeshima H, Niwa T, Kim JG, Ando T, Kushima R, Sugiyama T, Katai H, Noshiro H and Ushijima T: Integrated analysis of cancer-related pathways affected by genetic and epigenetic alterations in gastric cancer. Gastric Cancer. 18:65–76. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Wang Z, Wang W, Xu S, Wang S, Tu Y, Xiong Y, Mei J and Wang C: The role of MAPK signaling pathway in the Her-2-positive meningiomas. Oncol Rep. 36:685–695. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Schwappacher R, Rangaswami H, Su-You J, Hassad A, Spitler R and Casteel DE: cGMP-dependent protein kinase Iβ regulates breast cancer cell migration and invasion via interaction with the actin/myosin-associated protein caldesmon. J Cell Sci. 126:1626–1636. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Yu S, Sun L, Jiao Y and Lee LTO: The role of G protein-coupled receptor kinases in cancer. Int J Biol Sci. 14:189–203. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Peng L, Peng W, Hu P and Zhang HF: Clinical significance of expression levels of serum ADRA1A in hysterocarcinoma patients. Oncol Lett. 15:9162–9166. 2018.PubMed/NCBI

44 

Powe DG, Voss MJ, Habashy HO, Zänker KS, Green AR, Ellis IO and Entschladen F: Alpha- and beta-adrenergic receptor (AR) protein expression is associated with poor clinical outcome in breast cancer: An immunohistochemical study. Breast Cancer Res Treat. 130:457–463. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Huang JJ and Blobe GC: Dichotomous roles of TGF-β in human cancer. Biochem Soc Trans. 44:1441–1454. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Costanza B, Umelo IA, Bellier J, Castronovo V and Turtoi A: Stromal modulators of TGF-β in cancer. J Clin Med. 6(pii): E72017. View Article : Google Scholar : PubMed/NCBI

47 

Tatsuta M, Iishi H, Baba M, Yano H, Sakai N, Uehara H, Hirasawa R and Nakaizumi A: Alpha1-adrenoceptor stimulation enhances experimental gastric carcinogenesis induced by N-methyl-N′-nitro-N-nitrosoguanidine in Wistar rats. Int J Cancer. 77:467–469. 1998. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2019
Volume 18 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
Wang T, Qin Y, Lai H, Wei W, Li Z, Yang Y, Huang M and Chen J: The prognostic value of ADRA1 subfamily genes in gastric carcinoma. Oncol Lett 18: 3150-3158, 2019
APA
Wang, T., Qin, Y., Lai, H., Wei, W., Li, Z., Yang, Y. ... Chen, J. (2019). The prognostic value of ADRA1 subfamily genes in gastric carcinoma. Oncology Letters, 18, 3150-3158. https://doi.org/10.3892/ol.2019.10660
MLA
Wang, T., Qin, Y., Lai, H., Wei, W., Li, Z., Yang, Y., Huang, M., Chen, J."The prognostic value of ADRA1 subfamily genes in gastric carcinoma". Oncology Letters 18.3 (2019): 3150-3158.
Chicago
Wang, T., Qin, Y., Lai, H., Wei, W., Li, Z., Yang, Y., Huang, M., Chen, J."The prognostic value of ADRA1 subfamily genes in gastric carcinoma". Oncology Letters 18, no. 3 (2019): 3150-3158. https://doi.org/10.3892/ol.2019.10660