Open Access

Expression of factors and key components associated with the PI3K signaling pathway in colon cancer

  • Authors:
    • Hua Chen
    • Junyi Gao
    • Zhenhua Du
    • Xuequn Zhang
    • Fei Yang
    • Wei Gao
  • View Affiliations

  • Published online on: February 13, 2018     https://doi.org/10.3892/ol.2018.8044
  • Pages: 5465-5472
  • Copyright: © Chen 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

The pathophysiology of colorectal cancer (CRC) has not been fully elucidated. The dysregulation of the phosphatidylinositol 3‑kinase (PI3K) signaling pathway frequently contributes to the tumorigenesis and progression of human cancer. The aim of the present study was to explore the expression and clinical significance of a number of associated factors and key components of the PI3K signaling pathway, including phosphatidylinositol‑4,5‑bisphosphate 3‑kinase catalytic subunit α (p110α), phosphorylated protein kinase B (p‑Akt) Ser473, p‑mammalian target of rapamycin (mTOR) Ser2448, cyclin D1, cyclin dependent kinase (CDK)4, RELA proto‑oncogene, nuclear factor‑κβ subunit (p65), Ras and extracellular signal‑regulated kinase (ERK)1/2 in human CRC. The expression of target proteins was detected using immunohistochemistry (IHC) in 65 CRC cases and 15 colonic adenoma cases. The association between the expression of target proteins and clinical pathological parameters was analyzed using a χ² test. IHC results revealed that the expression of all target proteins was significantly increased in CRC tissues compared with in colonic adenoma tissues (P<0.05). No significant associations were observed between the expression of p110α, p‑Akt Ser473, p‑mTOR Ser2448 and sex, age, differentiation, lymph node metastasis or Tumor‑Node‑Metastasis staging (P>0.05). Cyclin D1, CDK4 and Ras were revealed to be expressed significantly higher in poorly differentiated CRC compared with moderately differentiated CRC (P<0.05). Expression of p65 and ERK1/2 were significantly increased in cancer tissues with lymph node metastasis compared with cancer tissues without lymph node metastasis (P<0.05). These results suggest that the target proteins may all participate in the tumorigenesis of CRC. Furthermore, cyclin D1, CDK4, Ras, p65 and ERK1/2 may be important in the progression of CRC. The results of the present study may provide novel predictive factors and therapeutic targets for CRC.

Introduction

Colorectal cancer (CRC) is becoming one of the most common malignancies globally (1). Each year, almost 1.4 million new CRC cases occur and 700,000 individuals succumb to mortality from CRC globally (13). Despite advancements in surgery, radiation therapy, chemotherapy and targeted therapy, the survival rate of patients remains low (4,5). Although extensive studies have demonstrated that CRC is a multi-factorial disease and is associated with physical, chemical, biological and genetic factors (6), the pathophysiology of CRC remains to be fully elucidated.

The phosphatidylinositol 3-kinase (PI3K) signaling pathway serves crucial functions in normal cellular processes, including cell proliferation, growth, apoptosis and motility (7). PI3K, protein kinase B (Akt) and mammalian target of rapamycin (mTOR) represent key nodes in the PI3K pathway. Class IA PI3K is a heterodimeric lipid kinase composed of a catalytic subunit, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (p110α) and a regulatory subunit (p85) (8). Akt is the downstream effector of PI3K and regulates the effects of PI3K on tumor growth and progression (9,10). The phosphorylation at serine 473 in its C-terminus fully activates Akt, and the activity of Akt may be evaluated using antibodies against phosphorylated (p)-Akt-S473 (11,12). An evolutionarily conserved serine/threonine kinase known as mTOR is the downstream target of Akt signaling. Akt may directly phosphorylate serine 2448 in mTOR (9,13). Akt serves important functions in cell proliferation. Cyclin D1 is a key cell cycle regulatory protein and its expression levels are important in the G1/S phase transition (7,14). The activation of Akt may phosphorylate and inhibit glycogen synthesis kinase-3 β, which results in the stabilization of cyclin D1 (14). Activated Akt may control the assembly of cyclin D1-cyclin dependent kinase (CDK)4 complexes by modulating cyclin D1 levels and contributing to the regulation of cell cycle progression (15). Akt may also affect cell survival by the indirect regulation of the nuclear factor-κβ (NF-κβ) signaling pathway (7). However, the elevated expression of RELA proto-oncogene, NF-κβ subunit (p65), a subunit of activated NF-κβ, may increase the phosphorylation and expression of Akt in the NIH3T3 cell line and primary endothelial cells (16). The Ras oncoprotein is one of the main regulators of the NF-κβ signaling pathway, and in its active conformation, Ras may phosphorylate the Raf/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) and the PI3K/Akt/mTOR signaling pathways to regulate normal cell growth and malignant transformation (17). Studies have demonstrated that the PI3K/Akt/mTOR and the Raf/MEK/ERK cascades are associated, with multiple points of convergence, cross-talk and feedback loops (10,17). ERK1/2 may mediate the phosphorylation of the essential scaffolding protein of mTOR complex 1 (C1) to promote mTOR signaling (18). The associated factors and key components of the P13K pathway include p110α, Akt, mTOR, cyclinD1, p65, Ras and ERK1/2; the aforementioned studies suggest that these factors may constitute a biochemical network to regulate cellular functions.

Numerous studies have demonstrated that the altered expression and/or activation of the aforementioned factors contribute to the tumorigenesis and progression of various types of human cancer, including CRC (7,10,13,14,17,1931). However, systemic research on the expression and function of all these factors in CRC is limited.

In the present study, the expression of the associated factors and key components of the PI3K pathway in patients with CRC, including p110α, p-Akt Ser473, p-mTOR Ser2448, cyclin D1, CDK4, p65, Ras and ERK1/2, were explored. Their clinical significance in CRC was additionally analyzed.

Materials and methods

Patients and controls

Between January 2013 and December 2014, a total of 65 CRC and 15 colonic adenoma tissue samples were collected from the Department of Pathology of the Jinan Central Hospital Affiliated to Shandong University (Shandong, China) for a retrospective study. All tissues were confirmed by postoperative pathology. The group of colonic adenoma cases comprised 7 females and 8 males with an age range of 34–82 years (mean age, 64.2). The group of patients with CRC consisted of 34 males and 31 females, and the mean age was 66 years (range, 32–88 years). A total of 36 of the tumors were moderately differentiated and 29 cases were poorly differentiated according to pathology reports. According to the tumor-node-metastasis (TNM) staging system (32), there were 3 cases of stage I, 25 cases of stage II, 24 cases of stage III and 13 cases of stage IV. Lymph node metastasis occurred in 33 CRC cases. The present study was approved by the Institutional Review Board of Jinan Central Hospital Affiliated to Shandong University, and written informed consent was obtained from all patients or the patient's family.

Immunohistochemistry (IHC)

IHC was performed to detect the expression of p110α, p-Akt Ser473, p-mTOR Ser2448, cyclin D1, CDK4, p65, Ras and ERK1/2 in CRC and colonic adenoma tissues. All tissue samples were 10% formalin-fixed at room temperature for 1 h, paraffin-embedded, cut into 4-µm sections and then placed on slides pretreated with 3-aminopropyltriethoxysilane. Following deparaffinization and rehydration (xylene for 10 min; 100, 95, 75 and 50% EtOH for 2 min each; PBS for 5 min), the sections were microwaved for heat-induced epitope retrieval at 100°C for 3 min in a citrate buffer solution (10 mM sodium citrate and 0.05% Tween 20; pH 6.0). The sections were then washed using PBS and the endogenous peroxidase activity was inhibited with 3% hydrogen peroxide for 10 min. The specimens were then blocked with PBS containing 5% normal goat serum (cat no., ab7481; Abcam, Shanghai, China) at 37°C for 30 min. Subsequently, the sections were incubated with antibodies overnight at 4°C. The primary antibodies used included a rabbit anti-p110α antibody (cat no. 4249; 1:200), a rabbit anti-p-Akt Ser473 antibody (cat no. 4060; 1:200), a rabbit anti-p-mTOR Ser2448 antibody (cat no. 2976; 1:200), a rabbit anti-cyclin D1 antibody (cat no. 2978; 1:200), a rabbit anti-CDK4 antibody (cat no. 12790; 1:200), a rabbit anti-NF-κB p65 antibody (cat no. 8242; 1:200), a rabbit anti-Erk1/2 antibody (cat no. 4695; 1:200; all Cell Signaling Technology, Inc., Danvers, MA, USA) and a rabbit anti-Ras (H,K,N) antibody (cat no. LS-C176193; 1:200; LifeSpan BioSciences, Inc., Seattle, WA, USA). Following rinsing with PBS, the slides were incubated at room temperature for 30 min with a secondary anti-rabbit antibody conjugated with horseradish peroxidase (1:200; cat no., sc-2357; Santa Cruz Biotechnology, Inc., Dallas, TX, USA). The slides were then exposed to 3,3′-diaminobenzidine for visualization, and hematoxylin at room temperature for 5 min, for nuclear counterstaining.

Evaluation of immunostaining parameters

Tissue sections were evaluated at a ×200 magnification using light microscopy by two pathologists in a blinded manner. Slides with debatable evaluations were re-evaluated until a consensus was reached. First, proportion scores were assigned to represent the estimated proportion of positive tumor cells (0, <5%; 1, 6–25%; 2, 26–50%; 3, 51–75%; 4, ≥76%). Secondly, intensity scores were assigned to represent the average intensity of the positive tumor cells (0, no color; 1, pale yellow; 2, tan; 3, brown). The proportion and intensity scores were then multiplied to obtain total scores. A total of 5 typical regions were assessed for each sample and the average scores were obtained. For further statistical analysis, all specimens were divided into four groups: Negative expression (−), 0–1; weak positive expression (+), 2–4; positive expression (++), 5–8; or strong positive expression (+++) >9. Scores with a range from 1–2 were also added to the positive expression group.

Statistical analysis

SPSS 17.0 (SPSS, Inc., Chicago, IL, USA) software package was used for statistical analyses. The χ2 test was performed to analyze the differences in various target proteins between CRC tissues and colonic adenoma tissues, in addition to associations between the expression levels of the various proteins and the clinical parameters. P<0.05 was considered to indicate a statistically significant difference.

Results

Expression of p110α, p-Akt Ser473, cyclin D1, CDK4, p-mTOR Ser2448, p65, Ras and ERK1/2 in CRC tissues and colonic adenoma tissues

IHC staining results revealed that, of the 65 CRC cases, 52 cases (80.00%) were p110α-positive, 48 cases (73.85%) were p-Akt Ser473-positive, 45 cases (69.23%) were cyclin D1-positive, 41 cases (63.08%) were CDK4-positive, 51 cases (78.46%) were p-mTOR Ser2448-positive, 43 cases (66.15%) were p65-positive, 40 cases (61.54%) were Ras-positive, and 46 cases (70.77%) were ERK1/2-positive. However, positive staining of p110α, p-Akt Ser473, cyclin D1, CDK4, p-mTOR Ser2448, p65, Ras and ERK1/2 was only observed in 3 cases (20.00%), 5 cases (33.33%), 2 cases (13.33%), 3 cases (20.00%), 4 cases (26.67%), 5 cases (33.33%), 4 cases (26.67%) and 4 cases (26.67%) of 15 colon adenoma cases, respectively. The expression of target proteins in CRC tissues were all significantly increased compared with in colon adenoma tissues (Table I; P<0.05). The localization of p110α and p-Akt Ser473 were primarily in the cytoplasm of CRC cells (Fig. 1A and B). The location of p-mTOR Ser2448 was mainly in the perinuclear and cytoplasmic regions of CRC cells, and in the nuclei of colonic adenoma cells (Fig. 1C). Cyclin D1 and CDK4 presented in the nuclei of CRC cells (Fig. 2A and B). The localization of p65 was mainly in the nuclei and cytoplasm of CRC cells (Fig. 2C). Ras and ERK1/2 were also observed in the cytoplasm of CRC cells (Fig. 3A and B).

Table I.

Expression of different targets in colon cancer and colonic adenoma tissues.

Table I.

Expression of different targets in colon cancer and colonic adenoma tissues.

Groups++++++Positive numberPositive rate (%)P-value
p110α <0.001
  Colon cancer13222285280.00
  Colonic adenoma12210320.00
p-Akt Ser473 0.001
  Colon cancer17301444873.85
  Colonic adenoma10500533.33
Cyclin D1 <0.001
  Colon cancer20261364569.23
  Colonic adenoma13200213.33
CDK4 0.005
  Colon cancer24261144163.08
  Colonic adenoma12210320.00
p-mTOR Ser2448 0.002
  Colon cancer142216135178.46
  Colonic adenoma11121426.67
NF-κβ 0.021
  Colon cancer22171794366.15
  Colonic adenoma10311533.33
RAS
  Colon cancer25161594061.540.022
  Colonic adenoma11211426.67
ERK1/2 0.019
  Colon cancer19251924670.77
  Colonic adenoma11121426.67

[i] CDK, cyclin dependent kinase; ERK, extracellular signal-regulated kinase; p65, RELA proto-oncogene, nuclear factor-Κβ subunit; p-, phosphorylated; Akt, protein kinase B; mTOR, mammalian target of rapamycin; p110α, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α.

Association between target proteins and clinicopathological characteristics in CRC

Statistical analysis results indicated that p110α, p-Akt Ser473 and p-mTOR Ser2448 expression were not significantly associated with sex, age, degree of differentiation, lymphatic metastasis or TNM staging of patients with CRC (P>0.05; Table II). No significant association was identified between cyclin D1 or CDK4 expression and sex, age and lymphatic metastasis (P>0.05; Table III). The positive expression of cyclin D1 and CDK4 in stage I and stage II of CRC were significantly increased compared with in stage III and stage IV (P<0.05; Table III). Tumors in poorly differentiated CRC presented a significantly increased positive expression of cyclin D1 or CDK4 compared with tumors in moderately differentiated CRC (P<0.05; Table III).

Table II.

Association between the expression of p110α, p-Akt Ser473 and p-mTOR Ser2448 and clinicopathological characteristics.

Table II.

Association between the expression of p110α, p-Akt Ser473 and p-mTOR Ser2448 and clinicopathological characteristics.

p110α p-Akt Ser473 p-mTOR Ser2448



Parametersn++++++P-value++++++P-value++++++P-value
Sex 0.081 0.628 0.445
  Male3481574 91762 811114
  Female3157154 81382 61159
Age, years 0.831 0.988 0.494
  ≥6041716144 111974 101498
  <60246684 61170 4875
Degree of differentiation 0.494 0.385 0.191
  Moderate36710154 91593 710109
  Poor2961274 81551 71264
Lymphatic metastasis 0.07 0.498 0.728
  Yes3358164 121584 51558
  No3281464 91560 97115
TNM staging 0.053 0.102 0.263
  I+II2871353 91450 96103
  III+IV3769175 81694 516610

[i] p-, phosphorylated; Akt, protein kinase B; mTOR, mammalian target of rapamycin; TNM, tumor-node-metastasis; p110α, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α.

Table III.

Association between the expression of cyclin D1, CDK4 and p65 and clinicopathological characteristics.

Table III.

Association between the expression of cyclin D1, CDK4 and p65 and clinicopathological characteristics.

Cyclin D1 CDK4 p65



Parametern++++++P-value++++++P-value++++++P-value
Sex 0.48 0.823 0.163
  Male34111481 121462 14974
  Female3191255 121252 88105
Age, years 0.487 0.701 0.329
  ≥60411415102 161663 141485
  <602461134 81051 8394
Degree of differentiation 0.01 0.008 0.69
  Moderate36161271 181341 14886
  Poor2941465 61373 8993
Lymphatic metastasis 0.562 0.067 0.008
  Yes3371295 91473 61188
  No32131441 151241 16691
TNM staging 0.002 0.034 0.006
  I+II28131221 141040 15571
  III+IV37714115 101674 712108

[i] CDK, cyclin dependent kinase; p65, RELA proto-oncogene, nuclear factor-κβ subunit; TNM, tumor-node-metastasis.

The expression of p65 was significantly increased in cancer tissues with lymph node metastasis compared with in cancer tissues without lymph node metastasis (P<0.05; Table III). The expression of p65 significantly decreased in stage I and stage II of CRC compared with stage III and stage IV of CRC (P<0.05; Table III). However, the expression of p65 was not associated with sex, age and degree of differentiation (P>0.05; Table III). The positive expression of Ras was significantly increased in poorly differentiated CRC compared with moderately differentiated CRC (P<0.05; Table IV). However, the expression of RAS was not associated with sex, age, TNM staging or lymphatic metastasis (P>0.05; Table IV). The expression of ERK1/2 was significantly increased in lymphatic metastasized tissues compared with non-lymphatic metastasized tissues (P<0.05; Table IV). The expression of ERK1/2 significantly decreased in stage I and stage II of CRC compared with stage III and stage IV of CRC (P<0.05; Table IV). There was no significant association indicated between the expression of ERK1/2 and sex, age or degree of differentiation (P>0.05; Table IV).

Table IV.

Association between the expression of Ras and ERK1/2 and clinicopathological characteristics.

Table IV.

Association between the expression of Ras and ERK1/2 and clinicopathological characteristics.

Ras ERK1/2


Parametern++++++P-value++++++P-value
Sex 0.962 0.527
  Male3413975 139111
  Female3112784 61681
Age, years 0.820 0.977
  ≥6041168125 1216112
  <60249834 7980
Degree of differentiation 0.002 0.308
  Moderate3617694 914112
  Poor2981065 101180
Lymphatic metastasis 0.426 0.033
  Yes33111281 613131
  No3214478 131261
TNM staging 0.260 0.036
  I+II2812368 13861
  III+IV37131391 617131

[i] ERK, extracellular signal-regulated kinase; TNM, tumor-node-metastasis.

Discussion

In the present study, the expression of the associated factors and key components of the PI3K signaling pathway in CRC tissues and colonic adenoma tissues were evaluated, and their clinical significances analyzed. These components include p110α, p-Akt Ser473, p-mTOR Ser2448, cyclin D1, CDK4, p65, Ras and ERK1/2. The expression of the target proteins was all significantly increased in CRC tissues compared with in colonic adenoma tissues (P<0.05). The expression of cyclin D1, CDK4 and Ras were significantly increased in poorly differentiated CRC compared with moderately differentiated CRC (P<0.05). The expression of p65 and ERK1/2 were significantly increased in cancer tissues with lymph node metastasis compared with in cancer tissues without lymph node metastasis (P<0.05).

p110α protein is the catalytic subunit of PI3K. The amplification of the genes encoding p110α have been demonstrated in ovarian, breast and pancreatic cancer cells (14). A previous study demonstrated that clustered regions of point mutations in the p110α catalytic subunit occurred in 20–30% of the breast, colon, brain and gastric tumors examined (10). Additionally, the overexpression of the regulatory subunit of PI3K (p85) protein and mutations in the gene encoding p85 have been identified in CRC (13,21). The aforementioned previous studies provide direct evidence for the alteration of PI3K in human cancer types. In the present study, it was revealed that the expression of p110α is significantly increased in CRC tissues compared with in colonic adenoma tissues (P<0.05; Table I). However, its expression was not associated with the sex, age, degree of differentiation, lymphatic metastasis or TNM staging of patients with CRC (P>0.05; Table II). The results of the present study suggest that p110α may be involved in the tumorigenesis of CRC.

Akt is the downstream effector of PI3K and regulates the effects of PI3K on tumor growth and progression (9,10). The complete activation of Akt requires phosphorylation at Serine 473 in its C-terminus (11,12). Akt phosphorylation has been observed in patients with non-small cell lung cancer (22,23), and Akt phosphorylation in human CRC was associated with the clinicopathological parameters of patients (24,25). In the present study, it was revealed that the expression of p-Akt Ser473 was significantly increased in CRC tissues compared with in colonic adenoma tissues (P<0.05; Table I). However, its expression was not associated with the sex, age, degree of differentiation, lymphatic metastasis or TNM staging of patients with CRC (P>0.05; Table II). The results of the present study suggest that p-Akt Ser473 may be involved in the tumorigenesis of CRC.

mTOR is the downstream target of Akt signaling and Akt may directly phosphorylate its Serine 2448 (10). The abnormal expression of p-mTOR Ser2448 has been linked to tumor progression in a number of types of human cancer, including CRC (13,26). In the present study, it was revealed that the expression of p-mTOR Ser2448 was significantly increased in CRC tissues compared with in colonic adenoma tissues (P<0.05; Table I). However, its expression was not associated with the sex, age, degree of differentiation, lymphatic metastasis or TNM staging of patients with CRC (P>0.05; Table II). The results of the present study suggest that p-mTOR Ser2448 may be involved in the tumorigenesis of CRC.

Cyclin D1 is a key cell cycle regulatory protein, and may form a complex with CDK4 in order to govern the cell cycle and its progression (7,14). Akt may indirectly regulate the levels of cyclin D1 and the assembly of cyclin D1-CDK4 complexes (14,15). The altered expression of cyclin D1 and CDK4 has been demonstrated in a variety of tumor types, including colon tumors (2729). They may be involved in early events in gastric tumorigenesis (29). In the present study, it was revealed that the expression levels of cyclin D1 and CDK4 were significantly increased in CRC tissues compared with those in colonic adenoma tissues (P<0.05; Table I). The expression levels of cyclin D1 and CDK4 were significantly increased in poorly differentiated CRC compared with moderately differentiated CRC (P<0.05). The results of the present study suggest that cyclin D1 and CDK4 may participate in the tumorigenesis and progression of CRC.

The NF-κβ signaling pathway serves important functions in oncogenesis by regulating the expression of genes associated with the development and progression of cancer (20). Akt may affect cell survival by indirectly regulating the NF-κβ signaling pathway (7). p65, the subunit of activated NF-κβ, has been demonstrated to regulate the phosphorylation and expression of Akt (16). An abnormal expression of p65 was observed in pancreatic cancer, in which a high expression of p65 indicated poor patient survival (30). In the present study, it was revealed that the expression of p65 was significantly increased in CRC tissues compared with that in colonic adenoma tissues (P<0.05; Table I). The expression of p65 was significantly increased in cancer tissues with lymph node metastasis compared with in cancer tissues without lymph node metastasis (P<0.05; Table III). These results suggest that p65 may be associated with the tumorigenesis and progression of CRC.

Ras proteins may regulate the phosphorylation of the Raf/MEK/ERK and the PI3K/Akt/mTOR signaling pathways (17). They exhibit essential functions in controlling a number of the malignant characteristics of transformed cells, for example in breast cancer (17,18). The activated mutations in the Ras genes themselves or alterations in upstream or downstream signaling components occurred in various types of human cancer, including CRC (10,33). KRAS proto-oncogene, GTPase (KRAS) is a member of the Ras family, and its mutations are predictors for resistance to cetuximab therapy in CRC (34). KRAS expression was significantly increased in carcinomatous lesions of the pancreas compared with in normal ducts or ductal hyperplasia (35). The antibody used in the present study may detect the expression of the three members of the Ras family (namely, HRAS proto-oncogene, GTPase, KRAS and NRAS proto-oncogene, GTPase). It was revealed that the expression of Ras was significantly increased in CRC tissues compared with in colonic adenoma tissues (P<0.05; Table I). The positive expression rate of Ras was significantly increased in poorly differentiated CRC compared with that in moderately differentiated CRC (P<0.05; Table IV). The results of the present study suggest that Ras may participate in the tumorigenesis and progression of CRC.

ERK1 and ERK2 are MAP kinases and serve important functions in normal physiological development and carcinogenesis (17,36). ERK1/2 may mediate the phosphorylation of the essential scaffolding protein of mTORC1 in order to promote mTOR signaling (18). ERK1 expression may be an early marker of cervical carcinogenesis (37). In the present study, it was revealed that the expression of ERK1/2 was significantly increased in CRC tissues compared with in colonic adenoma tissues (P<0.05; Table I). The expression of ERK1/2 was significantly increased in lymphatic metastasized tissues compared with non-lymphatic metastasized tissues (P<0.05; Table IV). The results of the present study suggest that ERK1/2 may participate in the tumorigenesis and progression of CRC.

In conclusion, the results of the present study demonstrated that p110α, p-Akt Ser473, p-mTOR Ser2448, cyclin D1, CDK4, p65, Ras and ERK1/2 may all participate in the tumorigenesis of CRC. Additionally, cyclin D1, CDK4, Ras, p65 and ERK1/2 may be important in the progress of CRC. These results may provide novel predictive factors and therapeutic targets for CRC.

Acknowledgements

The present study was supported by the Science and Technology Plan of Shandong (grant no. 2016GSF118008).

References

1 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics. 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Stein U, Walther W, Arlt F, Schwabe H, Smith J, Fichtner I, Birchmeier W and Schlag PM: MACC1, a newly identified key regulator of HGF-MET signaling, predicts colon cancer metastasis. Nat Med. 15:59–67. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Ferlay J, Shin HR, Bray F, Forman D, Mathers C and Parkin DM: Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 127:2893–2917. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Board RE and Valle JW: Metastatic colorectal cancer: Current systemic treatment options. Drugs. 67:1851–1867. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Ferlay J, Autier P, Boniol M, Heanue M, Colombet M and Boyle P: Estimates of the cancer incidence and mortality in Europe in 2006. Ann Oncol. 18:581–592. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Hansen JD, Kumar S, Lo WK, Poulsen DM, Halai UA and Tater KC: Ursodiol and colorectal cancer or dysplasia risk in primary sclerosing cholangitis and inflammatory bowel disease: A meta-analysis. Dig Dis Sci. 58:3079–3087. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Vivanco I and Sawyers CL: The phosphatidylinositol 3-Kinase Akt pathway in human cancer. Nat Rev Cancer. 2:489–501. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Yang SX, Polley E and Lipkowitz S: New insights on PI3K/Akt pathway alterations and clinical outcomes in breast cancer. Cancer Treat Rev. 45:87–96. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Ciruelos Gil EM: Targeting the PI3K/Akt/mTOR pathway in estrogen receptor-positive breast cancer. Cancer Treat Rev. 40:862–871. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Shaw RJ and Cantley LC: Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature. 441:424–430. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Sarbassov DD, Guertin DA, Ali SM and Sabatini DM: Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science. 307:1098–1101. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Badve S and Nakshatri H: Role of Akt isotypes in breast cancer. J Pathol. 229:e12013. View Article : Google Scholar : PubMed/NCBI

13 

Johnson SM, Gulhati P, Rampy BA, Han Y, Rychahou PG, Doan HQ, Weiss HL and Evers BM: Novel expression patterns of PI3K/Akt/mTOR signaling pathway components in colorectal cancer. J Am Coll Surg. 210:767–778. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Luo J, Manning BD and Cantley LC: Targeting the PI3K-Akt pathway in human cancer: Rationale and promise. Cancer Cell. 4:257–262. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Li Y, Dowbenko D and Lasky LA: AKT/PKB phosphorylation of p21Cip/WAF1 enhances protein stability of p21Cip/WAF1 and promotes cell survival. J Biol Chem. 277:11352–11361. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Meng F and D'Mello SR: NF-kappaB stimulates Akt phosphorylation and gene expression by distinct signaling mechanisms. Biochim Biophys Acta. 1630:35–40. 2003. View Article : Google Scholar : PubMed/NCBI

17 

Saini KS, Loi S, de Azambuja E, Metzger-Filho O, Saini ML, Ignatiadis M, Dancey JE and Piccart-Gebhart MJ: Targeting the PI3K/Akt/mTOR and Raf/MEK/ERK pathways in the treatment of breast cancer. Cancer Treat Rev. 39:935–946. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Carriere A, Romeo Y, Acosta-Jaquez HA, Moreau J, Bonneil E, Thibault P, Fingar DC and Roux PP: ERK1/2 phosphorylate Raptor to promote Ras-dependent activation of mTOR complex 1 (mTORC1). J Biol Chem. 286:567–577. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Okkenhaug K, Graupera M and Vanhaesebroeck B: Targeting PI3K in Cancer: Impact on tumor cells, their protective Stroma, Angiogenesis, and Immunotherapy. Cancer Discov. 6:1090–1105. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Dolcet X, Llobet D, Pallares J and Matias-Guiu X: NF-κB in development and progression of human cancer. Virchows Arch. 446:475–482. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Bader AG, Kang S, Zhao L and Vogt PK: Oncogenic PI3K deregulates transcription and translation. Nat Rev Cancer. 5:921–929. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Lee SH, Kim HS, Park WS, Kim SY, Lee KY, Kim SH, Lee JY and Yoo NJ: Non-small cell lung cancers frequently express phosphorylated Akt; an immunohistochemical study. APMIS. 110:587–592. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Cappuzzo F, Magrini E, Ceresoli GL, Bartolini S, Rossi E, Ludovini V, Gregorc V, Ligorio C, Cancellieri A, Damiani S, et al: Akt phosphorylation and gefitinib efficacy in patients with advanced non-small-cell lung cancer. J Natl Cancer Inst. 96:1133–1141. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Khaleghpour K, Li Y, Banville D, Yu Z and Shen SH: Involvement of the PI 3-kinase signaling pathway in progression of colon adenocarcinoma. Carcinogenesis. 25:241–248. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Itoh N, Semba S, Ito M, Takeda H, Kawata S and Yamakawa M: Phosphorylation of Akt/PKB is required for suppression of cancer cell apoptosis and tumor progression in human colorectal carcinoma. Cancer. 94:3127–3134. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Müller J, Ehlers A, Burkhardt L, Sirma H, Steuber T, Graefen M, Sauter G, Minner S, Simon R, Schlomm T, et al: Loss of pSer2448-mTOR expression is linked to adverse prognosis and tumor progression in ERG-fusion-positive cancers. Int J Cancer. 132:1333–1340. 2013. View Article : Google Scholar : PubMed/NCBI

27 

McKay JA, Douglas JJ, Ross VG, Curran S, Murray GI, Cassidy J and McLeod HL: Cyclin D1 protein expression and gene polymorphism in colorectal cancer. Aberdeen Colorectal Initiative. Int J Cancer. 88:77–81. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Motohashi M, Wakui S, Muto T, Suzuki Y, Shirai M, Takahashi H and Hano H: Cyclin D1/cdk4, estrogen receptors α and β, in N-methyl-N'-nitro-N-nitrosoguanidine-induced rat gastric carcinogenesis: Immunohistochemical study. J Toxicol Sci. 36:373–378. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Lee KH, Lee HE, Cho SJ, Cho YJ, Lee HS, Kim JH, Nam SY, Chang MS, Kim WH and Lee BL: Immunohistochemical analysis of cell cycle-related molecules in gastric carcinoma: Prognostic significance, correlation with clinicopathological parameters, proliferation and apoptosis. Pathobiology. 75:364–372. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Weichert W, Boehm M, Gekeler V, Bahra M, Langrehr J, Neuhaus P, Denkert C, Imre G, Weller C, Hofmann HP, et al: High expression of RelA/p65 is associated with activation of nuclear factor-kappaB-dependent signaling in pancreatic cancer and marks a patient population with poor prognosis. Br J Cancer. 97:523–530. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Uzgare AR, Kaplan PJ and Greenberg NM: Differential expression and/or activation of P38MAPK, erk1/2, and jnk during the initiation and progression of prostate cancer. Prostate. 55:128–139. 2003. View Article : Google Scholar : PubMed/NCBI

32 

Noone AM, Schussler N, Negoita S, Adamo M, Cronin K, Cyr J, Gress D, Grove C, Kosary C, Liu B, et al: Availability of TNM staging data elements in the medical record and training needs assessment: Results from the 2014 SEER Training Needs Assessment for TNM study. J Registry Manag. 42:40–47. 2015.PubMed/NCBI

33 

Downward J: Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 3:11–22. 2003. View Article : Google Scholar : PubMed/NCBI

34 

Lievre A, Bachet JB, Le Corre D, Boige V, Landi B, Emile JF, Coté JF, Tomasic G, Penna C, Ducreux M, et al: KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res. 66:3992–3995. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Apple SK, Hecht JR, Lewin DN, Jahromi SA, Grody WW and Nieberg RK: Immunohistochemical evaluation of K-ras, p53, and HER-2/neu expression in hyperplastic, dysplastic, and carcinomatous lesions of the pancreas: Evidence for multistep carcinogenesis. Hum Pathol. 30:123–129. 1999. View Article : Google Scholar : PubMed/NCBI

36 

Poulikakos PI and Solit DB: Resistance to MEK inhibitors: Should we co-target upstream? Sci Signal. 4:pe162011. View Article : Google Scholar : PubMed/NCBI

37 

Branca M, Ciotti M, Santini D, Bonito LD, Benedetto A, Giorgi C, Paba P, Favalli C, Costa S, Agarossi A, et al: Activation of the ERK/MAP kinase pathway in cervical intraepithelial neoplasia is related to grade of the lesion but not to high-risk human papillomavirus, virus clearance, or prognosis in cervical cancer. Am J Clin Pathol. 122:902–911. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2018
Volume 15 Issue 4

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
Chen H, Gao J, Du Z, Zhang X, Yang F and Gao W: Expression of factors and key components associated with the PI3K signaling pathway in colon cancer. Oncol Lett 15: 5465-5472, 2018
APA
Chen, H., Gao, J., Du, Z., Zhang, X., Yang, F., & Gao, W. (2018). Expression of factors and key components associated with the PI3K signaling pathway in colon cancer. Oncology Letters, 15, 5465-5472. https://doi.org/10.3892/ol.2018.8044
MLA
Chen, H., Gao, J., Du, Z., Zhang, X., Yang, F., Gao, W."Expression of factors and key components associated with the PI3K signaling pathway in colon cancer". Oncology Letters 15.4 (2018): 5465-5472.
Chicago
Chen, H., Gao, J., Du, Z., Zhang, X., Yang, F., Gao, W."Expression of factors and key components associated with the PI3K signaling pathway in colon cancer". Oncology Letters 15, no. 4 (2018): 5465-5472. https://doi.org/10.3892/ol.2018.8044