NOD‑like receptor X1, tumor necrosis factor receptor‑associated factor 6 and NF‑κB are associated with clinicopathological characteristics in gastric cancer

  • Authors:
    • Zhe Fan
    • Jiyong Pan
    • Hai Wang
    • Yewei Zhang
  • View Affiliations

  • Published online on: January 14, 2021     https://doi.org/10.3892/etm.2021.9640
  • Article Number: 208
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gastric cancer (GC) is a common cancer worldwide and its precise mechanism is largely unknown. The aim of the present study was to evaluate the expression levels of NOD‑like receptor X1 (NLRX1), tumor necrosis factor receptor‑associated factor 6 (TRAF6) and NF‑κB in GC and normal gastric tissue samples to determine the association with the clinicopathological features of GC. GC and adjacent normal gastric tissues from 60 patients with GC were analyzed using immunohistochemistry and western blotting analysis. In addition, the association between NLRX1, TRAF6 and NF‑κB expression levels were investigated by Spearman's correlation analysis. The results revealed that NLRX1 protein expression levels were downregulated in the GC tissues compared with the normal gastric tissues (P<0.05). Conversely, TRAF6 and NF‑κB protein expression levels were upregulated in the GC tissues compared with the normal gastric tissues (P<0.05). A significant difference was identified between GC patients with high and low expression levels of each protein in the tumor size, vascular invasion, neural invasion, lymph node metastasis, differentiation, gross stage and clinical stage. In addition, a negative correlation was observed between NLRX1 and TRAF6, and NLRX1 and NF‑κB expression levels, while a positive correlation was observed between TRAF6 and NF‑κB expression levels. In conclusion, NLRX1 expression levels were discovered to be downregulated in GC tissues. The expression levels of NLRX1, TRAF6 and NF‑κB were also significantly associated with the clinicopathological characteristics of GC, and the aforementioned results indicated that NLRX1 may be a biomarker in assessing GC.

Introduction

Gastric cancer (GC) is a challenging disease for general surgeons to manage and is one of the most common types of cancer worldwide, and GC has the third leading mortality rate worldwide, causing 723,000 deaths every year (1,2). In developing countries, 70% of deaths result from GC compared with 40% in China (3). Changes in lifestyle and eating habits, for example unhealthy diet, may increase the possibility of the incidence of GC (4). GC is more prevalent in East Asia than other geographic areas (5). Surgery, including open surgery and minimally invasive surgery, is the main treatment option for GC; however, as <10% patients with GC in developing countries are diagnosed early, there is a poor survival rate (6,7). Thus, the mechanisms underlying GC require further investigation.

Previous studies have demonstrated that NF-κB served a crucial role during the progression of GC (8,9). The NF-κB complex is activated in the cytoplasm following the degradation of inhibitor (I)κB, and is then involved in the nuclear physiologic response (10). Tumor necrosis factor receptor-associated factor 6 (TRAF6), which belongs to the TRAF family (11), is an adaptor protein that has important roles in innate immune responses and is a participator in the activatory process of the NF-κB signaling pathway (12). In addition, Han et al (11) reported that TRAF6 promoted the invasion and metastasis of GC and was an index for GC prognosis; Sun et al (13) concluded that the expression levels of TRAF6 in the skeletal muscle of patients with GC were significantly upregulated; and Maeda et al (14) suggested that Helicobacter pylori (H. pylori) may lead to NF-κB activation through a TRAF6 intracellular signaling pathway, which to the authors' best knowledge, is the only report of a relationship between NF-κB and TRAF6 in GC.

The NOD-like receptor (NLR) family serves a crucial role in immune defense and inflammation (15,16). A new member of the NLR family member, NLRX1, has been identified as a protein localized to the membrane of the mitochondria (16,17). Previous studies have revealed that NLRX1 suppressed tumorigenesis by inhibiting NF-κB signaling (18,19); however, the role of NLRX1, the correlation between NLRX1, TRAF6 and NF-κB expression levels, and the relationship between NLRX1 and the clinicopathological characteristics of GC have not been established.

In the present study, the expression levels of NLRX1, TRAF6 and NF-κB in GC tissues were determined and the association between these three proteins and the clinicopathological characteristics of gastric adenocarcinoma (GA) were examined. The present research may offer insight into novel molecular mechanisms underlying GC, specifically GA.

Materials and methods

Patients studies

The present study included 60 patients (age range, 34-83 years; 51 males and 9 females), who were diagnosed with GA based on post-operative pathologic evaluations at The Third People's Hospital of Dalian (Dalian, China) between October 2017 and April 2019. All patient demographic and clinicopathological data were recorded (Table I). The patients were diagnosed with GA by gastroscopy. Patients that received pre-operative chemotherapy or radiotherapy were excluded from the present study. The tissues were obtained at the time of surgery and immediately stored at -80˚C. GA and adjacent normal gastric tissues (>5 cm from the tumor) were collected. Written informed consent regarding information and tissue samples were acquired from the patients. The present study was approved by the Ethics Committee of The Third People's Hospital of Dalian (approval no. 2017-KY-004).

Table I

Demographic and clinicopathological characteristics of the patients.

Table I

Demographic and clinicopathological characteristics of the patients.

Clinicopathological features
Age, years (mean ± SD)66.5±11.4
Male, number (%)51(85)
Female, number (%)9(15)
Gross type, number 
     Early GC8
     Advanced GC52
Tumor size, number 
     <5 cm29
     ≥5 cm31
Differentiation, number 
     Moderately-to-well33
     Poorly differentiated27
Lymph node metastasis, number 
     Positive31
     Negative29
Vascular invasion, number 
     Yes35
     No25
Nerve invasion, number 
     Yes26
     No34
Depth of infiltration, number 
     T1 or T218
     T3 or T442
Clinical stage, number 
     I/II34
     III/IV26

[i] GC, gastric cancer; T, tumor.

The histologic grade was evaluated using the World Health Organization tumor classification: i) grade well if gland tissue is present, possibly including metaplasia; ii) grade poorly if highly irregular glands are indistinguishable; and iii) grade moderately if the condition is between the grade well and grade poorly classifications (20).

Immunohistochemistry (IHC)

IHC staining was performed to analyze the expression levels of NLRX1, TRAF6 and NF-κB in GC and normal gastric tissues. The tissues were fixed in 10% formalin for 24 h at room temperature, Subsequently, the tissues were embedded in paraffin and cut into 4-µm thick sections. Paraffin sections were placed in an incubator at 60˚C for 120 min. Xylene was used for deparaffinization and ethanol was used for rehydration at room temperature. Then the sections were subsequently incubated at 37˚C with 3% H2O2 for 10 min to block endogenous peroxidase activity. Antigen retrieval was performed by boiling the sections with 0.01 M citric acid buffer (pH 6.0) at 95˚C for 20 min and the sections were then blocked using goat serum (cat. no. SP-9000; OriGene Technologies, Inc.) at 37˚C for 15 min. Subsequent incubation was performed with primary antibodies against NLRX1 (1:100; cat. no. ABP57527; Abbkine Scientific Co., Ltd.), TRAF6 (1:100; cat. no. ABP52637; Abbkine Scientific Co., Ltd.) and NF-κB (1:100; cat. no. ABP51957; Abbkine Scientific Co., Ltd.) overnight in a humidified chamber at 4˚C. Negative controls were performed using PBS. Following the primary antibody incubation, the samples were then incubated with HRP-conjugated anti-rabbit secondary antibody (cat. no. SP-9000; OriGene Technologies, Inc.) at 37˚C for 15 min. Finally, DAB (cat. no. ZLI-9018; OriGene Technologies, Inc.) was added for 5 min and the tissue sections were counterstained with hematoxylin for 20 sec (both at room temperature).

IHC-stained slides were accessed by two pathologists who were blinded to the nature of the research using a light microscope (Nikon Eclipse Ni-E; Nikon Corporation; magnification, x400). The stained slides were evaluated semi-quantitatively and the intensity of staining was categorized into four levels as follows: No staining=0; weak staining=1; moderate staining=2; and strong staining=3. Positive stained cells were scored as follows: 1, 0-25; 2, 26-50; 3, 51-75; and 4, 76-100%. The final score was based on the above scores (score of positive stained cells multiplied by the intensity of staining) (21). There were four categories based on the final staining scores as follows: 0, -; 1-4, +; 5-8, ++; and 9-12, +++. The results were evaluated as negative and moderate for- and + (grouped as negative) and positive for ++ and +++ (21).

Western blotting

Total protein was extracted using RIPA lysis buffer, proteinase inhibitor, phosphatase inhibitors and PMSF (all from Beyotime Institute of Biotechnology). Proteins (30 µg; quantified using the BCA method) and the molecular weight marker were separated by 10% SDS-PAGE (cat. no. P0015A; Beyotime Institute of Biotechnology). The separated proteins were subsequently transferred onto PVDF membranes, which were blocked with 5% non-fat milk in 0.1% Tween-20 in TBS (TBST) for 1 h at room temperature, and incubated overnight at 4˚C with rabbit polyclonal anti-NLRX1 (1:1,000), anti-TRAF6 (1:1,000), anti-NF-κB (1:500) and anti-β-actin (1:1,000; cat. no. ABP57456; Abbkine Scientific Co., Ltd.) diluted in TBST. Following the primary antibody incubation, the membranes were washed three times with TBS-T for 10 min and incubated for 1 h at room temperature with an anti-rabbit IgG HRP-conjugated secondary antibody (1:5,000; cat. no. A21020; Abbkine Scientific Co., Ltd.). BeyoECL Plus (Beyotime Institute of Biotechnology) was used to visualize the protein bands. All results were analyzed using Gel-Pro Analyzer (version 4.0; Media Cybernetics, Inc.).

Statistical analysis

Data are presented as the mean ± SD of three experimental repeats. Statistical analyses were performed using SPSS 20.0 software (IBM Corp.) and GraphPad Prism 7.0 software (GraphPad Software, Inc.). A χ2 and Fisher's exact test were used to determine the associations between NLRX1, TRAF6 and NF-κB expression levels and the clinicopathological parameters. A paired Student's t-test was used for the western blotting analysis. Spearman's rank correlation was used for correlation analyses. P<0.05 was considered to indicate a statistically significant difference.

Results

Demographic and clinicopathological characteristics of the enrolled patients

Demographic information was collected; the average age of the patients was 66.5±11.4 years, and there were 51 males and 9 females in the present study. Clinicopathological characteristics were also collected, including age, sex, tumor size, histologic grade, lymph node metastasis, vascular invasion, nerve invasion, depth of infiltration (22) and clinical stage (Table I).

NLRX1, TRAF6 and NF-κB protein expression levels in GC and normal gastric tissues

To investigate the roles of NLRX1, TRAF6 and NF-κB in the progression of GC, IHC staining was performed to evaluate the changes in the protein expression levels. IHC staining of NLRX1 was markedly reduced in the GC tissues compared with the normal tissues (Fig. 1). In contrast, IHC staining of TRAF6 and NF-κB were markedly increased in the GC tissues compared with the normal tissues.

Association between NLRX1, TRAF6 and NF-κB expression levels and clinicopathological characteristics

NLRX1, TRAF6 and NF-κB IHC staining of GC samples and clinicopathological characteristics are presented in Table II. There were no significant differences identified between positive or negative expression levels of NLRX1, TRAF6 or NF-κB for either patient sex or age (≤65 or >65 years). However, significant differences were identified between positive or negative NLRX1, TRAF6 or NF-κB expression levels for tumor size (<5 cm or ≥5 cm), vascular invasion (yes or no), neural invasion (yes or no), lymph node metastasis (yes or no), differentiation (poorly or moderately-to-well differentiated), gross stage (early or advanced) and clinical stage (I/II or III/IV) (23).

Table II

Association between NLRX1, TRAF6 and NF-κB expression levels and the clinicopathologic features of patients with gastric cancer.

Table II

Association between NLRX1, TRAF6 and NF-κB expression levels and the clinicopathologic features of patients with gastric cancer.

A, NLRX1 expression levels
Clinicopathological variablesNumber of patientsPositive expressionNegative expressionP-value
Age, years   0.955
     ≤6527819 
     >65331023 
Sex   0.431
     Male511437 
     Female945 
Tumor size, cm   0.012
     <5281315 
     ≥532527 
Vascular invasion   0.010
     Yes35629 
     No251213 
Neural invasion   <0.001
     Yes26125 
     No341717 
Lymph node metastasis   0.011
     Yes29425 
     No311417 
Differentiation   0.001
     Poorly27225 
     Moderately-to-well331617 
Gross stage   0.005
     Early862 
     Advanced521141 
Clinical stage   <0.001
     I/II341717 
     III/IV26125 
B, TRAF6 expression levels
Clinicopathological variablesNumber of patientsPositive expressionNegative expressionP-value
Age, years   0.180
     ≤65271710 
     >65332013 
Sex   0.284
     Male513318 
     Female945 
Tumor size, cm   <0.001
     <5281018 
     ≥532275 
Vascular invasion   <0.001
     Yes35296 
     No25817 
Neural invasion   0.002
     Yes26224 
     No341519 
Lymph node metastasis   <0.001
     Yes29254 
     No311219 
Differentiation   0.001
     Poorly27234 
     Moderately-to-well331419 
Gross stage   <0.001
     Early808 
     Advanced523715 
Clinical stage   0.002
     I/II341519 
     III/IV26224 
C, NF-κB expression levels
Clinicopathological variablesNumber of patientsPositive expressionNegative expressionP-value
Age, years   0.957
     ≤65271710 
     >65332112 
Sex   0.111
     Male513813 
     Female945 
Tumor size, cm   0.020
     <5281216 
     ≥532266 
Vascular invasion   0.002
     Yes35287 
     No251015 
Neural invasion   0.017
     Yes26215 
     No341717 
Lymph node metastasis   0.007
     Yes29245 
     No311516 
Differentiation   0.015
     Poorly27225 
     Moderately-to-well331617 
Gross stage   <0.001
     Early808 
     Advanced523913 
Clinical stage   0.017
     I/II341717 
     III/IV26215 

[i] NLRX1, NOD-like receptor X1; TRAF6, tumor necrosis factor receptor-associated factor 6.

NLRX1, TRAF6 and NF-κB expression levels based on western blotting

The results from the western blotting experiments revealed that NLRX1 expression levels were significantly downregulated in the GC tissues compared with the normal gastric tissues (Fig. 2). Conversely, TRAF6 and NF-κB expression levels were discovered to be significantly upregulated in the GC tissues compared with the normal tissues.

Correlation analysis between NLRX1 and TRAF6, NLRX1 and NF-κB and TRAF6 and NF-κB expression levels

Spearman's rank correlation was used to determine the correlation between NLRX1 and TRAF6, NLRX1 and NF-κB and TRAF6 and NF-κB expression levels. A negative correlation was identified between NLRX1 and TRAF6 expression levels (Table III), NLRX1 and NF-κB protein expression levels (Table IV) and a positive correlation between TRAF6 and NF-κB protein expression levels in GC tissues (Table V).

Table III

Correlation between NLRX1 and TRAF6 expression levels.

Table III

Correlation between NLRX1 and TRAF6 expression levels.

 NLRX1 expression 
 Staining score, (n)- (23)+ (19)++ (14)+++ (4)Correlation coefficientP-value
TRAF6 expression- (3)--21 rs=-0.635P<0.001
 + (20)2783  
 ++ (18)864-  
 +++ (19)136--  

[i] NLRX1, NOD-like receptor X1; TRAF6, tumor necrosis factor receptor-associated factor 6.

Table IV

Correlation between NLRX1 and NF-κB expression levels.

Table IV

Correlation between NLRX1 and NF-κB expression levels.

 NLRX1 expression 
 Staining score, (n)- (23)+ (19)++ (14)+++ (4)Correlation coefficientP-value
NF-κB P65 expression- (4)-112 rs=-0.530P<0.001
 + (18)3681  
 ++ (17)6731  
 +++ (21)1452-  

[i] NLRX1, NOD-like receptor X1.

Table V

Correlation between TRAF6 and NF-κB expression levels.

Table V

Correlation between TRAF6 and NF-κB expression levels.

 TRAF6 expression 
 Staining score, (n)- (3)+ (20)++ (17)+++ (20)Correlation coefficientP-value
NF-κB P65 expression- (4)13-- rs=0.781P<0.001
 + (18)2142-  
 ++ (17)-377  
 +++ (21)--813  

[i] TRAF6, tumor necrosis factor receptor-associated factor 6.

Discussion

The worldwide incidence of GC has decreased markedly in recent decades (24). Data from the 2014 Chinese National Cancer Center (NCCRC) have also revealed a decreased incidence of GA (25). Nevertheless, the poor prognosis among patients with GC remains a serious threat to global health. The incidence of GC varies by country and is 2-3-fold higher in males compared with females (26). In the present study, the male-to-female GC ratio was 51:9 (5.7-fold higher in males). The 2014 NCCRC reported a high incidence of GC in the 60-79-year group (27), which coincides with the age data (66.5±11.4 years) obtained in the present research.

NLRX1 is a negative regulator of antiviral immunity during the early stage of virus infection (16). With additional research involving NLRX1, other functions of NLRX1 have been discovered, such as its ability to regulate inflammation (28), metabolism (29) and development of histiocytic sarcoma (18). Previous studies have also reported that NLRX1 served as a tumor suppressor in colorectal cancer (30,31). For example, the deletion of NLRX1 in intestinal epithelial cells did not alter the architecture of the intestines, but there was an increased susceptibility among NLRX1-/- mice for developing colitis-associated colorectal cancer (30,31). Wang et al (32) also concluded that NLRX1 expression levels were downregulated in hepatocellular carcinoma tissues and that NLRX1 expression levels may be used as a prognostic marker in HCC hepatectomy. Castano-Rodriguez et al (33) reported that NLRX1 expression levels were downregulated in H. pylori-infected gastric tissues. However, the role of NLRX1 in GC has not been elucidated.

By examining IHC staining and western blotting of NLRX1 in GC and normal gastric tissues, the present study revealed that NLRX1 expression levels were downregulated in GC tissues, indicating that NLRX1 may be a tumor suppressor. The changes in NLRX1 expression levels were significantly associated with tumor size, vascular invasion, neural invasion, lymph node metastasis, differentiation, gross stage and clinical stage, which indicated that NLRX1 may serve as an index in assessing GC. However, no statistical differences were observed between NLRX1 expression levels and the age or sex of the patient. A previous study concluded that NLRX1 was a tumor suppressor in primary solid tumors of the breast (34). Hu et al (19) also reported that downregulated expression levels of NLRX1 were associated with liver cancer prognosis.

NF-κB is a pivotal mammalian transcription factor that was discovered to exert protumorigenic effects in liver (35), lung (36), breast (37) and prostate (38) cancer, in addition to in GC (39). In normal cells, NF-κB is located in the cytosol in the form of an inactive complex bound to IκBα. Once stimulated, IκBα is phosphorylated and separated from NF-κB for degradation (40). NF-κB has been demonstrated to remain in an active status in pancreatic cancer cells (41). The results of the present study revealed that NF-κB expression levels were upregulated in GC tissues through IHC staining and western blotting, which is consistent with the aforementioned studies.

microRNA-146a was discovered to upregulate NF-κB by targeting TRAF6 in human cervical cancer (42). In multiple myeloma, TRAF6 was discovered to mediate NF-κB activation and was suggested as a potential therapeutic target (43). Both of these studies illustrated that TRAF6 could regulate NF-κB as an upstream gene. However, to the best of our knowledge, the relationship between TRAF6 and NF-κB in GC remains to be clarified. It has also been reported that in response to lipopolysaccharide, NLRX1 interacted with TRAF6 and negatively regulated NF-κB activation in 293T cells (16,44). Allen et al (16) demonstrated that in response to viral infection, NLRX1 decreased the inflammatory responses by interacting with TRAF6.

TRAF6, as a TRAF protein family member, activates IκB kinase, thus resulting in the degradation of IκB and the activation of NF-κB (45,46). Therefore, a positive correlation is suggested to exist between TRAF6 and NF-κB. In the present study, a positive correlation was also discovered and TRAF6 and NF-κB expression levels, as tumor promotors, were increased and correlated with tumor size, vascular and neural invasion, lymph node metastasis, differentiation, gross stage and clinical stage.

In the present study, a negative correlation between NLRX1 and TRAF6 was discovered using Spearman's rank correlation. Therefore, it was speculated that NLRX1 may also interact with TRAF6 to exert an antitumor effect in GC.

In conclusion, NLRX1 expression levels were discovered to be downregulated in GC tissues and the expression levels of NLRX1 were associated with the clinicopathological characteristics of GC. A negative correlation was identified between NLRX1 and TRAF6/NF-κB expression levels, while a positive correlation was observed between TRAF6 and NF-κB expression levels. Thus, NLRX1 may be a potential biomarker for the diagnosis of GC and warrants further investigation.

Acknowledgements

Not applicable.

Funding

The present study was funded by the Dalian Medical Science Research Project (grant no. 1711038), the National Natural Science Foundation of China (grant nos. 81701965 and 81872255), the Key Medical Talents Fund of Jiangsu Province (grant no. 2016KJQWZDRC-03) and the Natural Science Foundation of Liaoning Province (grant nos. 20180550116 and 2019-MS-069).

Availability of data and materials

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

Authors' contributions

ZF and JP designed the study. JP contributed to the collection and storage of the tissues. HW analyzed the pathology. ZF, JP, HW and YZ performed the remaining experiments. YZ revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of The Third People's Hospital of Dalian (approval no. 2017-KY-004). Written informed consent regarding information and tissue samples were acquired from the patients.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 513:202–209. 2014.PubMed/NCBI View Article : Google Scholar

2 

Gong Z, Mu Y, Chen J, Chu H, Lian P, Wang C, Wang J and Jiang L: Expression and significance of cyclophilin J in primary gastric adenocarcinoma. Anticancer Res. 37:4475–4481. 2017.PubMed/NCBI View Article : Google Scholar

3 

Wadhwa R, Song S, Lee JS, Yao Y, Wei Q and Ajani JA: Gastric cancer-molecular and clinical dimensions. Nat Rev Clin Oncol. 10:643–655. 2013.PubMed/NCBI View Article : Google Scholar

4 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016.PubMed/NCBI View Article : Google Scholar

5 

McGuire S: World Cancer Report 2014 Geneva, Switzerland: World Health Organization, International Agency for Research on Cancer, WHO Press, 2015. Adv Nutr. 7:418–419. 2016.PubMed/NCBI View Article : Google Scholar

6 

Graziosi L, Marino E and Donini A: Minimally invasive surgery for advanced gastric cancer: Are we sure? Gastric Cancer. 20:1013–1014. 2017.PubMed/NCBI View Article : Google Scholar

7 

Peddanna N, Holt S and Verma RS: Genetics of gastric cancer. Anticancer Res. 15:2055–2064. 1995.PubMed/NCBI

8 

Lv Y, Zhao Y, Wang X, Chen N, Mao F, Teng Y, Wang T, Peng L, Zhang J, Cheng P, et al: Increased intratumoral mast cells foster immune suppression and gastric cancer progression through TNF-α-PD-L1 pathway. J Immunother Cancer. 7(54)2019.PubMed/NCBI View Article : Google Scholar

9 

Xie Y, Li F, Li Z and Shi Z: miR-135a suppresses migration of gastric cancer cells by targeting TRAF5-mediated NF-κB activation. Onco Targets Ther. 12:975–984. 2019.PubMed/NCBI View Article : Google Scholar

10 

Deptala A, Bedner E, Gorczyca W and Darzynkiewicz Z: Activation of nuclear factor kappa B (NF-kappaB) assayed by laser scanning cytometry (LSC). Cytometry. 33:376–382. 1998.PubMed/NCBI View Article : Google Scholar

11 

Han F, Zhang L, Qiu W and Yi X: TRAF6 promotes the invasion and metastasis and predicts a poor prognosis in gastric cancer. Pathol Res Pract. 212:31–37. 2016.PubMed/NCBI View Article : Google Scholar

12 

Inoue J, Gohda J and Akiyama T: Characteristics and biological functions of TRAF6. Adv Exp Med Biol. 597:72–79. 2007.PubMed/NCBI View Article : Google Scholar

13 

Sun YS, Ye ZY, Qian ZY, Xu XD and Hu JF: Expression of TRAF6 and ubiquitin mRNA in skeletal muscle of gastric cancer patients. J Exp Clin Cancer Res. 31(81)2012.PubMed/NCBI View Article : Google Scholar

14 

Maeda S, Yoshida H, Ogura K, Mitsuno Y, Hirata Y, Yamaji Y, Akanuma M, Shiratori Y and Omata M: H. pylori activates NF-kappaB through a signaling pathway involving IkappaB kinases, NF-kappaB-inducing kinase, TRAF2, and TRAF6 in gastric cancer cells. Gastroenterology. 119:97–108. 2000.PubMed/NCBI View Article : Google Scholar

15 

Takeuchi O and Akira S: Innate immunity to virus infection. Immun Rev. 227:75–86. 2009.PubMed/NCBI View Article : Google Scholar

16 

Allen IC, Moore CB, Schneider M, Lei Y, Davis BK, Scull MA, Gris D, Roney KE, Zimmermann AG, Bowzard JB, et al: NLRX1 protein attenuates inflammatory responses to infection by interfering with the RIG-I-MAVS and TRAF6-NF-κB signaling pathways. Immunity. 34:854–865. 2011.PubMed/NCBI View Article : Google Scholar

17 

Moore CB, Bergstralh DT, Duncan JA, Lei Y, Morrison TE, Zimmermann AG, Accavitti-Loper MA, Madden VJ, Sun L, Ye Z, et al: NLRX1 is a regulator of mitochondrial antiviral immunity. Nature. 451:573–577. 2008.PubMed/NCBI View Article : Google Scholar

18 

Coutermarsh-Ott S, Simmons A, Capria V, LeRoith T, Wilson JE, Heid B, Philipson CW, Qin Q, Hontecillas-Magarzo R, Bassaganya-Riera J, et al: NLRX1 suppresses tumorigenesis and attenuates histiocytic sarcoma through the negative regulation of NF-κB signaling. Oncotarget. 7:33096–33110. 2016.PubMed/NCBI View Article : Google Scholar

19 

Hu B, Ding GY, Fu PY, Zhu XD, Ji Y, Shi GM, Shen YH, Cai JB, Yang Z, Zhou J, et al: NOD-like receptor X1 functions as a tumor suppressor by inhibiting epithelial-mesenchymal transition and inducing aging in hepatocellular carcinoma cells. J Hematol Oncol. 11(28)2018.PubMed/NCBI View Article : Google Scholar

20 

Fléjou JF: WHO Classification of digestive tumors: The fourth edition. Ann Pathol. 31 (5 Suppl):S27–S31. 2011.PubMed/NCBI View Article : Google Scholar : (In French).

21 

Zheng M, Zang S, Xie L, Fang X, Zhang YU, Ma X, Liu J, Lin D and Huang A: Rheb phosphorylation is involved in p38-regulated/activated protein kinase-mediated tumor suppression in liver cancer. Oncol Lett. 10:1655–1661. 2015.PubMed/NCBI View Article : Google Scholar

22 

Lyros O, Thomaidis T, Muller M, Sivanathan V, Grimminger P, Lang H, Gockel I, Hartmann JT and Moehler M: External Validation of the Proposed Kiel Staging System and Comparison with the Old (6th edition) and the Currently Used (7th edition) TNM Classification in Gastric Cancer. Oncol Res Treat. 41:122–128. 2018.PubMed/NCBI View Article : Google Scholar

23 

Harino Y, Imura S, Kanemura H, Morine Y, Fujii M, Ikegami T, Uehara H and Shimada M: Role of tumor angiogenesis in gallbladder carcinoma: With special reference to thymidine phosphorylase. Int J Clin Oncol. 13:452–457. 2008.PubMed/NCBI View Article : Google Scholar

24 

Bertuccio P, Chatenoud L, Levi F, Praud D, Ferlay J, Negri E, Malvezzi M and La Vecchia C: Recent patterns in gastric cancer: A global overview. Int J Cancer. 125:666–673. 2009.PubMed/NCBI View Article : Google Scholar

25 

Wang FH, Shen L, Li J, Zhou ZW, Liang H, Zhang XT, Tang L, Xin Y, Jin J, Zhang YJ, et al: The Chinese society of clinical oncology (CSCO): Clinical guidelines for the diagnosis and treatment of gastric cancer. Cancer Commun (Lond). 39(10)2019.PubMed/NCBI View Article : Google Scholar

26 

Makino Y, Nishimura Y, Oshita S, Mizosoe T and Akihiro T: Storage in high-barrier pouches increases the sulforaphane concentration in broccoli florets. PLoS One. 13(e0192342)2018.PubMed/NCBI View Article : Google Scholar

27 

Chen W, Sun K, Zheng R, Zeng H, Zhang S, Xia C, Yang Z, Li H, Zou X and He J: Cancer incidence and mortality in China, 2014. Chin J Cancer Res. 30:1–12. 2018.PubMed/NCBI View Article : Google Scholar

28 

Ma D, Zhao Y, She J, Zhu Y, Zhao Y, Liu L and Zhang Y: NLRX1 alleviates lipopolysaccharide-induced apoptosis and inflammation in chondrocytes by suppressing the activation of NF-κB signaling. Int Immunopharmacol. 71:7–13. 2019.PubMed/NCBI View Article : Google Scholar

29 

Scantlebery AML, Uil M, Butter LM, Poelman R, Claessen N, Girardin SE, Florquin S, Roelofs JJTH and Leemans JC: NLRX1 does not play a role in diabetes nor the development of diabetic nephropathy induced by multiple low doses of streptozotocin. PLoS One. 14(e0214437)2019.PubMed/NCBI View Article : Google Scholar

30 

Soares F, Tattoli I, Rahman MA, Robertson SJ, Belcheva A, Liu D, Streutker C, Winer S, Winer DA, Martin A, et al: The mitochondrial protein NLRX1 controls the balance between extrinsic and intrinsic apoptosis. J Biol Chem. 289:19317–19330. 2014.PubMed/NCBI View Article : Google Scholar

31 

Tattoli I, Killackey SA, Foerster EG, Molinaro R, Maisonneuve C, Rahman MA, Winer S, Winer DA, Streutker CJ, Philpott DJ and Girardin SE: NLRX1 acts as an epithelial-intrinsic tumor suppressor through the modulation of TNF-mediated proliferation. Cell Rep. 14:2576–2586. 2016.PubMed/NCBI View Article : Google Scholar

32 

Wang X, Yang C, Liao X, Han C, Yu T, Huang K, Yu L, Qin W, Zhu G, Su H, et al: NLRC and NLRX gene family mRNA expression and prognostic value in hepatocellular carcinoma. Cancer Med. 6:2660–2672. 2017.PubMed/NCBI View Article : Google Scholar

33 

Castaño-Rodríguez N, Kaakoush NO, Goh KL, Fock KM and Mitchell HM: The NOD-like receptor signalling pathway in Helicobacter pylori infection and related gastric cancer: A case-control study and gene expression analyses. PLoS One. 9(e98899)2014.PubMed/NCBI View Article : Google Scholar

34 

Singh K, Poteryakhina A, Zheltukhin A, Bhatelia K, Prajapati P, Sripada L, Tomar D and Singh R, Singh AK, Chumakov PM and Singh R: NLRX1 acts as tumor suppressor by regulating TNF-α induced apoptosis and metabolism in cancer cells. Biochim Biophys Acta. 1853:1073–1086. 2015.PubMed/NCBI View Article : Google Scholar

35 

Zhang M, Pan L, Xu D, Cao C, Shi R, Han S, Liu J, Li X and Li M: The NFκB signaling pathway serves an important regulatory role in Klebsiella pneumoniae liver abscesses. Exp Ther Med. 15:5443–5449. 2018.PubMed/NCBI View Article : Google Scholar

36 

Ahmmed B, Khan MN, Nisar MA, Kampo S, Zheng Q, Li Y and Yan Q: Tunicamycin enhances the suppressive effects of cisplatin on lung cancer growth through PTX3 glycosylation via AKT/NF-κB signaling pathway. Int J Oncol. 54:431–442. 2019.PubMed/NCBI View Article : Google Scholar

37 

Bishop RT, Marino S, de Ridder D, Allen RJ, Lefley DV, Sims AH, Wang N, Ottewell PD and Idris AI: Pharmacological inhibition of the IKKε/TBK-1 axis potentiates the anti-tumour and anti-metastatic effects of Docetaxel in mouse models of breast cancer. Cancer Lett. 450:76–87. 2019.PubMed/NCBI View Article : Google Scholar

38 

Marino S, Bishop RT, Carrasco G, Logan JG, Li B and Idris AI: Pharmacological inhibition of NFκB reduces prostate cancer related osteoclastogenesis in vitro and osteolysis ex vivo. Calcif Tissue Int. 105:193–204. 2019.PubMed/NCBI View Article : Google Scholar

39 

Fu J, Yu L, Luo J, Huo R and Zhu B: Paeonol induces the apoptosis of the SGC-7901 gastric cancer cell line by downregulating ERBB2 and inhibiting the NF-κB signaling pathway. Int J Mol Med. 42:1473–1483. 2018.PubMed/NCBI View Article : Google Scholar

40 

Novack DV: Role of NF-κB in the skeleton. Cell Res. 21:169–182. 2011.PubMed/NCBI View Article : Google Scholar

41 

Jeong Y, Lim JW and Kim H: Lycopene inhibits reactive oxygen species-mediated NF-κB signaling and induces apoptosis in pancreatic cancer cells. Nutrients. 11(762)2019.PubMed/NCBI View Article : Google Scholar

42 

Li T, Li M, Xu C, Xu X, Ding J, Cheng L and Ou R: miR-146a regulates the function of Th17 cell differentiation to modulate cervical cancer cell growth and apoptosis through NF-κB signaling by targeting TRAF6. Oncol Rep. 41:2897–2908. 2019.PubMed/NCBI View Article : Google Scholar

43 

Morgan JJ, McAvera RM and Crawford LJ: TRAF6 silencing attenuates multiple myeloma cell adhesion to bone marrow stromal cells. Int J Mol Sci. 20(702)2019.PubMed/NCBI View Article : Google Scholar

44 

Xia X, Cui J, Wang HY, Zhu L, Matsueda S, Wang Q, Yang X, Hong J, Songyang Z, Chen ZJ and Wang RF: NLRX1 negatively regulates TLR-induced NF-κB signaling by targeting TRAF6 and IKK. Immunity. 34:843–853. 2011.PubMed/NCBI View Article : Google Scholar

45 

Paik JH, Jang JY, Jeon YK, Kim WY, Kim TM, Heo DS and Kim CW: MicroRNA-146a downregulates NFκB activity via targeting TRAF6 and functions as a tumor suppressor having strong prognostic implications in NK/T cell lymphoma. Clin Cancer Res. 17:4761–4771. 2011.PubMed/NCBI View Article : Google Scholar

46 

Yang WL, Wang J, Chan CH, Lee SW, Campos AD, Lamothe B, Hur L, Grabiner BC, Lin X, Darnay BG and Lin HK: The E3 ligase TRAF6 regulates Akt ubiquitination and activation. Science. 325:1134–1138. 2009.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

March-2021
Volume 21 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fan Z, Pan J, Wang H and Zhang Y: NOD‑like receptor X1, tumor necrosis factor receptor‑associated factor 6 and NF‑κB are associated with clinicopathological characteristics in gastric cancer. Exp Ther Med 21: 208, 2021
APA
Fan, Z., Pan, J., Wang, H., & Zhang, Y. (2021). NOD‑like receptor X1, tumor necrosis factor receptor‑associated factor 6 and NF‑κB are associated with clinicopathological characteristics in gastric cancer. Experimental and Therapeutic Medicine, 21, 208. https://doi.org/10.3892/etm.2021.9640
MLA
Fan, Z., Pan, J., Wang, H., Zhang, Y."NOD‑like receptor X1, tumor necrosis factor receptor‑associated factor 6 and NF‑κB are associated with clinicopathological characteristics in gastric cancer". Experimental and Therapeutic Medicine 21.3 (2021): 208.
Chicago
Fan, Z., Pan, J., Wang, H., Zhang, Y."NOD‑like receptor X1, tumor necrosis factor receptor‑associated factor 6 and NF‑κB are associated with clinicopathological characteristics in gastric cancer". Experimental and Therapeutic Medicine 21, no. 3 (2021): 208. https://doi.org/10.3892/etm.2021.9640