Zum Hauptinhalt springen

Functions and clinical significance of KLRG1 in the development of lung adenocarcinoma and immunotherapy

Han, Zhihai ; Yang, Xiaodong ; et al.
In: BMC Cancer, Jg. 21 (2021), Heft 1, S. 1-16
Online unknown

Functions and clinical significance of KLRG1 in the development of lung adenocarcinoma and immunotherapy 

Background: As a marker of differentiation, Killer cell lectin like receptor G1 (KLRG1) plays an inhibitory role in human NK cells and T cells. However, its clinical role remains inexplicit. This work intended to investigate the predictive ability of KLRG1 on the efficacy of immune-checkpoint inhibitor in the treatment of lung adenocarcinoma (LUAD), as well as contribute to the possible molecular mechanisms of KLRG1 on LUAD development. Methods: Using data from the Gene Expression Omnibus, the Cancer Genome Atlas and the Genotype-Tissue Expression, we compared the expression of KLRG1 and its related genes Bruton tyrosine kinase (BTK), C-C motif chemokine receptor 2 (CCR2), Scm polycomb group protein like 4 (SCML4) in LUAD and normal lung tissues. We also established stable LUAD cell lines with KLRG1 gene knockdown and investigated the effect of KLRG1 knockdown on tumor cell proliferation. We further studied the prognostic value of the four factors in terms of overall survival (OS) in LUAD. Using data from the Gene Expression Omnibus, we further investigated the expression of KLRG1 in the patients with different responses after immunotherapy. Results: The expression of KLRG1, BTK, CCR2 and SCML4 was significantly downregulated in LUAD tissues compared to normal controls. Knockdown of KLRG1 promoted the proliferation of A549 and H1299 tumor cells. And low expression of these four factors was associated with unfavorable overall survival in patients with LUAD. Furthermore, low expression of KLRG1 also correlated with poor responses to immunotherapy in LUAD patients. Conclusion: Based on these findings, we inferred that KLRG1 had significant correlation with immunotherapy response. Meanwhile, KLRG1, BTK, CCR2 and SCML4 might serve as valuable prognostic biomarkers in LUAD.

Keywords: KLRG1; Lung adenocarcinoma; Prognosis; Immunotherapy

Supplementary Information The online version contains supplementary material available at https://doi.org/10.1186/s12885-021-08510-3.

Key points

KLRG1 inhibited the progress of LUAD.

The expression of KLRG1 had correlation with immunotherapy response.

KLRG1, BTK, CCR2 and SCML4 might serve as valuable prognostic biomarkers in LUAD.

Background

Lung cancer is the principal cause of cancer deaths worldwide [[1]]. Most patients have advanced disease when they are diagnosed with lung cancer. Patients in early-stage can receive surgical, chemo- or radiation therapy, but over 90% of the patients will unavoidably have disease recurrence. The overall 5-year survival rate is lower than 60% [[2]].

Non-small cell lung cancers (NSCLCs), account for 85% of lung tumors, include a variety of cancer types, such as squamous cell cancers (LUSCs), adenocarcinomas (LUADs), and large cell lung cancers. Among them, LUSCs and LUADs are the largest NSCLC subgroups. Meanwhile, lung adenocarcinoma (LUAD) is the most heterogeneous and aggressive among all NSCLC subtypes. LUAD is the most common type of lung cancers among nonsmokers. The incidence of LUAD is higher among women than men, and it is more likely to happen in younger people than other types of lung cancer. In the past few decades, LUAD has replaced LUSC as the most frequent histological subtype [[4]].

LUADs originate from cells that secrete surfactant ingredients. The most important morphological features of LUADs include acinar, solid, papillary, micropapillary, and invasive mucinous types. At the same time, a small part of LUADs shows colloid, enteric or fetal features. The staining of thyroid transcription factor 1 (TTF-1/NKX2–1) or napsin-A (NAPSA) can be used to support the diagnosis when the morphological feature of adenocarcinoma is unclear. The sensitivity of the two markers is approximately 80% for the identification of LUAD [[5]].

Although chemotherapy, radiotherapy, targeted therapy and immunotherapy have made huge progress in the past decade, the prevention, early detection and treatment of LUAD are still facing great challenges. More research is needed to understand the molecular mechanisms facilitating the development of lung carcinogenesis.

The clinical development of immune-checkpoint inhibitors has created an exhilarant era of anticancer therapies. Durable responses have been seen in patients with lung cancer, melanoma and other malignancies [[6]]. Pembrolizumab, an anti-PD-1 antibody, in combination with pemetrexed, was approved by European Medicines Agency (EMA) as first-line treatment of metastatic LUAD. Atezolizumab, an anti-PD-L1 antibody, in combination with carboplatin/paclitaxel/bevacizumab was granted Food and Drug Administration (FDA) approval in untreated LUAD patients. Although monotherapy with anti-PD-1 or PD-L1 drugs is usually well tolerated, the combination treatment increases the risk of immune-related adverse events [[8]]. So, biomarkers with predictive role need to be developed to augment patient benefit, diminish the risk of toxicity, and guide the combination approaches [[9]]. Although the expression of PD-L1 on tumor cells positivity improves the clinical benefit population, PD-L1 detection alone is not satisfactory for patient selection and efficacy prediction in most malignancies [[11]]. The investigation of powerful biomarkers to help predict the response and clinical benefit of immune-checkpoint inhibitors is crucial to further advance the field of precision immunotherapy [[12]].

In this study, through the analysis of the database and clinical samples, we found four markers (KLRG1, BTK, CCR2 and SCML4) which may play important roles in the development of lung adenocarcinoma. Knockdown of KLRG1 promoted the proliferation of A549 and H1299 lung tumor cells. Additionally, the expression of KLRG1 is positively correlated with the efficacy of immune-checkpoint inhibitors. Collectively, KLRG1 may be a powerful biomarker to support the diagnosis and predict the clinical benefit of immunotherapy in lung adenocarcinoma.

Methods

Extraction and analysis of data from the TCGA database

The raw data of RNAseqv2 in 518 LUAD cases were downloaded from The Cancer Genome Atla (TCGA) database. Their clinicopathological information, including age at initial pathologic diagnosis, smoking history, gender, nodal status, pathologic stage, residual tumors, recurrence status, relapse-free survival (RFS) in days, overall survival (OS) status, and OS in days was downloaded. GEPIA2 (http://gepia2.cancer-pku.cn) [[14]] and Xena Browser (http://xena.ucsc.edu/) [[15]], two online interactive web server for analyzing the RNA sequencing data of tumors and normal samples from the TCGA and The Genotype-Tissue Expression (GTEx) projects, were used to analyze the expression profiles and prognostic value of selected genes. And the log2(TPM + 1) was used the show the expression value of the selected genes.

Prediction of related genes

The co-expression analysis module of cBioPortal [[16]] is able to extract genes that are co-expressed with KLRG1 in the database of Lung Adenocarcinoma (TCGA, PanCancer Atlas). The mRNA expression, RSEM (Batch normalized from Illumina HiSeq_RNASeqV2) was used in the co-expression analysis. The associated genes that were identified by cBioPortal were subjected to follow-up pathway analysis.

Cell line

The A549 human lung adenocarcinoma and H1299 human lung carcinoma cell lines were obtained from the American Type Culture Collection. Cells were maintained in Dulbecco's modified Eagle's medium (DMEM) basic medium supplemented with 10% fetal bovine serum and 1% antibiotics at 37 °C with 5% CO2.

Establishment of stable KLRG1 knockdown cells

HEK-293 T cells in T175 flasks were transfected with the packaging vector psPAX2 (9.4 μg), envelope plasmid pVSVG (9.4 μg), and transfer plasmid (18.8 μg) containing the shRNA targeting KLRG1. Following 72 h, the HEK-293 T medium containing the virus was collected and concentrated, and then transferred to A549 and H1299 plate (5 × 106 cells). The medium was replaced after 72 h and cells containing the integrated virus were selected with puromycin (50 ng/μl). The knockdown efficiency were validated by RT-PCR and Western blotting. Sequences of used shRNAs were as follow: KLRG1-shRNA-1: 5′- CCGGGATTGGTCTGAGGAACAATTCCTCGAGGAATTGTTCCTCAGACCAATCTTTTTTG-3′, KLRG1-shRNA-2: 5′- CCGGGATCTGTCATGTATCCCTAAACTCGAGTTTAGGGATACATGACAGATCTTTTTTG-3′, scrambled-shRNA: 5′- CCGGGCGCGATAGCGCTAATAATTTCTCGAGAAATTATTAGCGCTATCGCGCTTTTTTG-3′.

Western blotting

The knockdown and control A549 or H1299 cells (1 × 106 cells per well) were plated in 12-well plates and cultured for 24 h, then cellular KLRG1 protein levels were evaluated in total cell extracts by Western blot analysis. Blots were developed using enhanced chemiluminescence (ECL) and detected by exposure to chemiluminescence-sensitive film. Antibodies against KLRG1 (rabbit polyclonal), β-actin (mouse monoclonal, AC-15) were from Sigma-Aldrich (St. Louis, MO, USA). The protein bands without overexposure were quantified relative to β-actin expression using ImageJ software (NIH, Bethesda, MD, USA). We obtain the absolute intensity (AI) for each experimental band of KLRG1. Relative intensity (RI) for each experimental band was calculated by normalizing the experimental AI to the corresponding loading control (β-actin) AI.

RNA extraction and qRT-PCR

Total RNAs were extracted with TRIzol reagent (Invitrogen, Carlsbad, CA, USA), reverse transcribed with PrimeScrip RT-PCR Kit (Takara Biotechnology Co., Ltd., Dalian, China), followed by qRT-PCR with SYBR Premix Ex Taq (Takara Biotechnology Co., Ltd., Dalian, China). QuantStudio 5 real-time PCR system (Applied Biosystems, Grand Island, NY) were used to examine the gene of interest mRNA expression. The amplification cycling conditions were 95 °C for 2 min; 40 cycles of 95 °C for 10 s, 60 °C for 40 s. Control of the RT reactions was performed by omitting DNA template in the negative controls. The data were analysed by comparative CT method and three replicates were performed. The primers for KLRG1 were forward 5′-CCAGACCGCTGGATGAAATATG-3′ and reverse 5′-CTGATTGTCCGTTATCACAAGGA-3′. The primers for BTK were forward 5′- TCTGAAGCGATCCCAACAGAA-3′ and reverse 5′-TGCACGGTCAAGAGAAACAGG-3′. The primers for CCR2 were forward 5′-CCACATCTCGTTCTCGGTTTATC-3′ and reverse 5′-CAGGGAGCACCGTAATCATAATC-3′. The primers for SCML4 were forward 5′-TCACTCCACGCCTATGAAGAT-3′ and reverse 5′-GGGTTTCCGCCCTCTTTTC-3′. The primers for ACTB (Beta-Actin) were forward 5′-CTGTCCCTGTATGCCTCTG-3′ and reverse 5′-ATGTCACGCACGATTTCC-3′.

ACTB was used as an internal control.

Cell proliferation analysis

Control and the KLRG1-knockdown A549 cells (2 × 103 cells per well) were plated in 48-well plates treated with E-Cadherin Antibody or IgG1 (Sinobiological, Beijing, China), and then cultured for 72 h. Cell proliferation was determined using the colorimetric Cell Counting Kit-8 (CCK-8, Dojindo Laboratories, Kumamoto, Japan). The absorbance was measured using Varioskan LUX (Thermo Fisher, Waltham, MA, USA) at a wavelength of 450 nm.

Receiver operating characteristic (ROC) curve analysis

The diagnostic value of the expression levels of KLRG1, BTK, CCR2 and SCML4 in LUADs was studied by analyzing the expression data from 483 LUADs and 347 normal tissues. Specificity and sensitivity were plotted on the x- and y-axes, respectively. The area under curve (AUC) was calculated to assess the ability of the expression levels of KLRG1, BTK, CCR2 and SCML4 to predict the outcome of patients with LUAD.

PPI network analysis and functional enrichment

Protein–protein (PPI) interactions network can visualize the patterns of molecular interactions and help to explain the mechanisms underlying phenotypes. PPI network analysis was performed using the online database STRING (https://string-db.org/) [[18]]. And the Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were analysed by STRING.

Statistical analysis

Assays were repeated in 2 or more biological experiments with each data point being the average of a minimum of 3 technical replicates. Statistical analysis was conducted using GraphPad Prism 6.0 (GraphPad Inc., La Jolla, California) or SPSS 20.0 software package (SPSS Inc., Chicago, Illinois). Group comparison was performed using two-tailed unpaired Student's t-test. Prognostic factors were evaluated using univariate Cox regression analysis. The diagnostic and prognostic value of KLRG1, BTK, CCR2 and SCML4 expression in LUAD was judged using receiver operating characteristic (ROC) curves. Kaplan–Meier curves of OS and RFS were generated using GraphPad Prism. P values are derived from Log-rank (Mantel-Cox) test for all the survival analysis. P < 0.05 was considered statistically significant.

Results

The expression of KLRG1 is correlated with immunotherapy response and overall survival in LUA...

To find the potential biomarkers related to the development and immunotherapy response of lung adenocarcinoma. We analysed the data from 1 previous array (GSE93157) that compared gene expression profiles from 65 patients with melanoma, lung nonsquamous(N = 22), squamous cell lung or head and neck cancers who were treated with the approved PD1-targeting antibodies pembrolizumab or nivolumab [[19]]. The clinical and pathological characteristics of 22 lung nonsquamous patients were shown in Table 1. Among them, 59% (N = 13) of the patients had relatively good response including SD (stable disease), PR (partial response) and CR (complete response). And 41% (N = 9) of the patients showed PD (progressive disease). We analysed the differential expression genes between different responses. Then, we studied the differential survival genes in LUAD using data from GEPIA2. There were four genes had overlap in the two databases, which were KLRG1, CDK1, IL1R2, BTK and CCR2. Among them, we found that KLRG1 had the most significant correlation with the drug response after anti-PD1 treatment. KLRG1 expression was significantly higher in LUAD patients which showed better responses (SD, PR and CR) compared those patients with PD (Fig. 1a). The associations between the expression levels of KLRG1 and the clinicopathological characteristics of 22 lung nonsquamous patients were shown in Table 2. By conducting multivariate analysis, we found that, the expression level of KLRG1 was independent prognostic factor to predict the progression-free survival time (PFS) (P = 0.014) (Table 3). Furthermore, we investigated the association between KLRG1 expression and progression-free survival outcomes in lung nonsquamous patients after immunotherapy. The result showed that the high expression group had significantly better PFS compared to the low expression group (P = 0.0069) (Fig. 1b). Meanwhile, ROC curves and AUC analysis were performed to evaluate the prognostic performance. The results indicated that the performance of the KLRG1 had high sensitivity and specificity (Fig. 1c). These results suggested that KLRG1 maybe a potential biomarker to predict the immunotherapy response in LUAD patients.

Table 1 Clinical–pathologic characteristics of the 22 nonsquamous lung carcinoma patients evaluated in this study

N (%)

N

22

Age, median (range)

58 (42–79)

Sex

Male

7 (32%)

Female

15 (68%)

Previous lines

0

4 (18%)

1

8 (37%)

2

6 (27%)

≥ 3

4(18%)

Biopsy

Archival

10 (45%)

Baseline

12 (55%)

Drug response

CR

1 (5%)

PR

5 (22%)

SD

7 (32%)

PD

9 (41%)

Smoking

Current smoker

8 (36%)

Former smoker

11 (50%)

Never smoker

3 (14%)

ECOG

0

5 (23%)

1

17 (77%)

Drug

Nivolumab

14 (64%)

Pembrolizumab

8 (36%)

PFS, median

3.03

Lung cancer EGFR status

EGFR mutated

1 (5%)

EGFR wild-type

21 (95%)

Lung cancer ALK status

ALK rearranged

0 (0%)

ALK not rearranged

21 (95%)

NA

1 (5%)

Abbreviations: CR complete response, PR partial response, SD stable disease, PD progression disease, PFS progression-free survival, ALK anaplastic lymphoma kinase, NA not applicable

Graph: Fig. 1 Influence of KLRG1 expression on LUAD survival and immunotherapy responses. A Expression of KLRG1 in 22 nonsquamous lung carcinoma patients with different response after PD-1 blockade. P values are derived from two-tailed unpaired Student's t-test. B Kaplan–Meier curves of progression-free survival in the 22 nonsquamous lung carcinoma patients based on the mRNA expression of KLRG1. C Receiver operating characteristic curves for estimating the prognostic value of KLRG1 after PD-1 blockade. D Expression of KLRG1 in LUAD and normal lung tissues. The method for differential analysis is one-way ANOVA, using disease state (Tumor or Normal) as variable for calculating differential expression. E Kaplan–Meier curves of OS in LUAD based on the mRNA expression of KLRG1. F Kaplan–Meier curves of RFS in LUAD based on the mRNA expression of KLRG1. Data represent mean ± SD. **, P < 0.01, ***, P < 0.001

Table 2 Association of KLRG1 expression levels with clinicopathologic variables of 22 lung cancer patients

Outcome

KLRG1 expression

Low

High

P value

Age / year

0.838

<58

5

6

≥ 58

6

5

Gender

0.798

Male

5

9

Female

6

2

Drug

0.794

Nivolumab

7

7

Pembrolizumab

4

4

Smoking history / Category

0.123

Current smoker & Former smoker

1

2

Never smoker

10

9

EGFR

0.285

Mutated

1

0

Wild-type

10

11

ECOG

0.882

0

2

3

1

9

8

State

0.030

PD

8

1

CR, PR, SD

3

10

P values are derived from one-way analysis of variance. Abbreviations: CR complete response, PR partial response, SD stable disease, PD progression disease

Table 3 Statistically significant associations of KLRG1 expression and other clinicopathologic variables with progression-free survival

Outcome

PFS

HR

95%CI

P value

KLRG1 expression level (High vs. Low)

0.139

0.029–0.668

0.014

Drug (Nivolumab vs. Pembrolizumab)

8.781

1.332–57.878

0.024

Biopsy (Archival vs. Baseline)

0.248

0.094–1.842

0.248

Smoking history (Current smoker & Former smoker vs. Never smoker)

2.930

0.311–27.575

0.347

Gender (Male vs. Female)

0.263

0.048–1.447

0.125

Age (< 58 vs. ≥58)

0.417

0.110–1.579

0.198

ECOG (0 vs. 1)

0.480

0.093–2.480

0.381

P values are derived from Cox regression analysis

Meanwhile, using data from GEPIA2, we also found KLRG1 belonged to the most differential survival genes in LUAD. Using RNA-seq data in TCGA and GTEx projects, we compared the KLRG1 expression between cancerous and normal lung tissues. Results showed that LUAD tissues (N = 483) had significantly decreased KLRG1 expression compared to normal controls (N = 347, Fig. 1d). By generating Kaplan-Meier survival curves, we analyzed the association between KLRG1 expression and OS/RFS in patients with LUAD. The LUAD patients were divided into high/low KLRG1 expression group by using the best cutoff model. Results showed that the high KLRG1 expression group had significantly better overall survival (OS) (P < 0.01) and relapse-free survival (RFS) (P < 0.05) compared to the low KLRG1 expression group (Fig. 1e and f).

BTK, CCR2 and SCML4 are positively co-expressed with KLRG1 in LUAD

We next conducted the co-expression analysis in cBioPortal database, and found that BTK, CCR2 and SCML4 had a strong expression correlation with KLRG1 (Fig. 2a, b and c). Furthermore, BTK, CCR2 and SCML4 were among the top factors which expression can significantly influence the overall survival outcomes in LUAD. To further investigate the role of BTK, CCR2 and SCML4 in LUAD, we compared the expression of BTK, CCR2 and SCML4 between cancerous and normal lung tissues using the RNA-seq data in TCGA and GTEx projects, respectively. In the data cohort, RNA-seq was performed in 483 LUAD tissues and 347 normal tissues. The plots chart showed that BTK, CCR2 and SCML4 was significantly downregulated in LUAD tissues compared with the normal controls (Fig. 2d, e and f). Heatmap also showed that KLRG1, BTK, CCR2 and SCML4 expressions were significantly higher in normal tissues than in LUAD tissues (Fig. 2g).

Graph: Fig. 2 BTK, CCR2 and SCML4 were co-expressed with KLRG1 and downregulated in LUAD. A-C Regression analysis of the correlation between KLRG1 expression and BTK (A), CCR2 (B), SCML4 (C) expression, respectively. P values are derived from Spearman and Pearson correlation analysis. D-F Expression of BTK (D), CCR2 (E), SCML4 (F) in LUAD and normal lung tissues, respectively. P values are derived from one-way ANOVA. G Heatmap of KLRG1, BTK, CCR2 and SCML4 expression in LUAD patients and normal lung tissues. ***, P < 0.001

Knockdown of KLRG1 promotes the proliferation of A549 and H1299 tumor cells

To further understand the role of KLRG1 in the development of LUAD, we generated stable KLRG1 knockdown cells by transfection with KLRG1-specific short hairpin RNAs (shRNAs). The knockdown efficiency of KLRG1 in A549 and H1299 cells were confirmed by both qRT-PCR and Western blotting (Fig. 3a-d). Moreover, we investigated the effect of KLRG1 knockdown on the proliferation of A549 and H1299 cells. The results showed that knockdown of KLRG1 enhanced the proliferation of A549 (Fig. 3e) and H1299 (Fig. 3f) tumor cells. The shRNA2 did not influence the proliferation of H1299 tumor cells because of the low knockdown efficiency in H1299 tumor cells. These results indicated that KLRG1 may promote the development of LUAD.

Graph: Fig. 3 Knockdown of KLRG1 in A549 and H1299 lung tumor cells. A-B The KLRG1 knockdown efficiency in A549 was evaluated by qRT-PCR, n = 3 for each group (A) and Western blot (B). C-D The KLRG1 knockdown efficiency in H1299 was evaluated by qRT-PCR, n = 3 for each group (C) and Western blot (D). The full-length gels are presented in Supplementary Figure 1 and Figure 2. E-F The effect of KLRG1-knockdown on the proliferation of A549 (E) and H1299 (F) tumor cells, n = 4 for each group. P values are derived from two-tailed unpaired Student's t-test. Data represent mean ± SD. **, P < 0.01, ***, P < 0.001, ns, no significance

ROC analysis of the BTK, CCR2, SCML4 and KLRG1 expression in patients with LUAD

Since the four factors were downregulated in LUAD samples compared with controls, we next explored whether the four factors may serve as potential diagnostic biomarkers in LUAD. Diagnostic ROC curves and AUC analysis were performed to evaluate the diagnostic performance. The results indicated that the performance of the KLRG1 [AUC, 0.570; 95% CI (confidence interval),0.528–0.612] (Fig. 4a) and CCR2 (AUC, 0.515; 95% CI, 0.473–0.557) (Fig. 4b) were not satisfactory, but BTK (AUC, 0.871; 95% CI, 0.789–0.846) (Fig. 4c) and SCML4 (AUC, 0.810; 95% CI, 0.781–0.839) (Fig. 4d) had high sensitivity and specificity. It suggested that the BTK and SCML4 may have huge value in the auxiliary diagnosis of LUAD patients.

Graph: Fig. 4 The diagnostic value of KLRG1, CCR2, BTK and SCML4 expression in LUAD. A-D Receiver operating characteristic curves for estimating the diagnostic value of KLRG1 (A), CCR2 (B), BTK (C), and SCML4 (D). E-H the KLRG1 (E), CCR2 (F), BTK (G) and SCML4 (H) expressions in different pathological stages of LUAD

Association between KLRG1, BTK, CCR2, SCML4 expression and the demographic and clinicopatholo...

Then, we investigated the associations between the expression levels of KLRG1, BTK, CCR2, SCML4 and the clinicopathological characteristics of 499 patients with LUAD. The results showed that the expression level of CCR2 was associated with age (P = 0.040; Table 4). Meanwhile, the expression level of CCR2 and SCML4 was associated with gender (P = 0.029; P = 0.006, respectively; Table 1). Notably, the expression levels of KLRG1, BTK, CCR2, SCML4 were all associated with neoplasm disease stage (American Joint Committee on Cancer Code) (P = 0.001; P = 0.022; P = 0.004; P = 0.014, respectively; Table 4). And a lower expression levels of KLRG1, BTK, CCR2, SCML4 were all associated with advanced neoplasm disease stage (Fig. 4e-h). By conducting multivariate analysis, we found that, in addition to neoplasm disease stage and diagnosis age, the expression levels of KLRG1, BTK, CCR2 and SCML4 were all independent prognostic factor for OS (hazard ratio, HR = 1.658 and P = 0.002 for KLRG1; HR = 1.889 and P < 0.001 for BTK; HR = 1.922 and P < 0.001 for CCR2; HR = 1.638 and P = 0.001 for SCML4) in LUAD patients (Table 5).

Table 4 The association between KLRG1, BTK, CCR2, SCML4 expression and the demographic and clinicopathological parameters

Outcome a

KLRG1 expression

BTK expression

CCR2 expression

SCML4 expression

Low

High

P value

Low

High

P value

Low

High

P value

Low

High

P value

Age / year

0.250

0.082

0.040

0.172

<65

145

73

132

86

134

84

96

122

≥ 65

160

111

142

129

136

135

97

174

Gender

0.710

0.156

0.029

0.006

Male

146

84

137

93

140

90

105

125

Female

165

104

141

128

134

135

92

177

Mutations / number

0.615

0.067

0692

0.103

≤ 230

92

50

90

54

77

67

56

88

>230

52

25

40

35

39

36

37

38

Smoking history / Categoryb

0.628

0.112

0.280

0.937

1

40

31

32

39

32

39

26

45

2, 3, 4, 5

262

152

239

175

235

179

165

249

Neoplasm Disease Stage

0.001

0.022

0.004

0.014

I, II

224

162

202

184

196

190

141

245

III, IV, V

82

23

72

33

72

33

54

51

P values are derived from one-way analysis of variance a A total of 499 samples were analyzed. Age: N = 489, NA = 10; Stage: N = 491, NA = 8; Mutations: N = 219, NA = 280; Smoking History: N = 485, NA = 14. N: Number; NA: Not Applicable b Smoking history: 1: lifelong non-smoker; 2: current smoker; 3. Current reformed smoker (for> 15 years); 4. Current reformed smoker (for≤15 years); 5. Current reformed smoker (duration not specified)

Table 5 Statistically significant associations of KLRG1, BTK, CCR2 and SCML4 expressions with overall survival in LUAD patients

Outcome

OS

HR

95% CI

P value

KLRG1 expression level (high vs. low)

1.658

1.202–2.288

0.002

BTK expression level (high vs. low)

1.889

1.381–2.585

< 0.001

CCR2 expression level (high vs. low)

1.922

1.404–2.631

< 0.001

SCML4 expression level (high vs. low)

1.638

1.225–2.191

0.001

Diagnosis Age < 65 vs. ≥ 65

3.553

1.122–11.253

0.031

Neoplasm Disease Stage I, II vs. III, IV, V

1.788

1.391–2.298

< 0.001

Mutation Count

< 230 vs. ≥ 230

1.001

0.850–1.178

0.995

Gender

Female vs. Male

1.043

0.779–1.396

0.779

Smoking History Categorya

1 vs. 2, 3, 4, 5

0.966

0.681–1.371

0.847

P values are derived from Cox regression analysis aSmoking history: 1: lifelong non-smoker; 2: current smoker; 3. Current reformed smoker (for> 15 years); 4. Current reformed smoker (for≤15 years); 5. Current reformed smoker (duration not specified)

Prognostic value of the BTK, CCR2, SCML4 and KLRG1 in patients with LUAD

To examine the association between BTK, CCR2 and SCML4 expression and survival outcomes in LUAD, respectively, we extracted the survival data in TCGA database. The LUAD patients were divided into high/low expression group by using the best cutoff model. For all the three genes, results showed that the high expression groups had significantly better OS compared to the low expression groups, respectively (P < 0.0001 for all, Fig. 5a, b and c). Furthermore, we combined the expression of the four factors (KLRG1, BTK, CCR2 and SCML4), the patients were divided into three groups. Group 1 included 110 patients which had all low expression of the four factors. Group 2 included 137 patients with three factors low expression among the four. Group 3 included the rest of the patients (N = 253). Strikingly, the patients which had all low expression of the four factors showed the worst overall survival (P < 0.0001, median survival time = 29.5, 41.4, 66.7, respectively, Fig. 5d). These results indicated that the expression of the four factors could be powerful biomarkers to predict the LUAD patient survival.

Graph: Fig. 5 Kaplan-Meier survival curves for assessing the prognostic value of KLRG1, BTK, CCR2 and SCML4. Kaplan–Meier analysis for the OS of LUAD patients according to distinct BTK (A), CCR2 (B) and SCML4 (C) expression level. D Kaplan–Meier analysis for the OS of LUAD patients according to the expression of the four factors

Protein-protein interaction network analysis

In order to study how these factors were involved in the development of LUAD, STRING was performed to construct the protein-protein interactions among KLRG1, BTK, CCR2 and SCML4. The number of nodes was 24 and number of edges was 80. The PPI enrichment P-value was 7.13e-10. In the PPI network, KLRG1, BTK and CCR2 had close connections through CD19 (CD19 molecule), CDH1 (Cadherin 1), FYN (Src family tyrosine kinase), PLCG2 (Phospholipase C gamma 2) and LCP2 (Lymphocyte cytosolic protein 2) (Fig. 6a). Due to few studies based on SCML4, it showed an independent relationship in the PPI network. Furthermore, we did functional analysis based on the PPI network. The top processes and pathways were showed in Table 6. To further investigate the interaction of the four factors, we analyzed the expression levels of BTK, CCR2 and SCLM4 in KLRG1 knockdown A549 tumor cells. The results showed that knockdown of KLRG1 also reduced the expression of BTK and CCR2 in A549 tumor cells (Fig. 6b and c). On the other hand, the expression of SCLM4 did not show significant changes (Fig. 6d). E-cadherin was reported as the ligand of KLRG1. And the treatment of E-Cadherin antibody enhanced the proliferation of A549 tumor cells (Fig. 6e). The tumor promotion effect was eliminated in KLRG1 knockdown A549 cells (Fig. 6e). This indicated that KLRG1 may influence the proliferation of tumor cells through the biding of E-cadherin. As KLRG1 was abundantly expressed on the surface of T cells and NK cells, we speculated that the expression of KLRG1 on tumor cells would competitively bind E-cadherin in tumor microenvironment which would promote the activity of T cells and NK cells. We conducted the co-expression analysis in cBioPortal database, and found that IFNG and CD274 had a good expression correlation with KLRG1 (Fig. 6f and g). The results indicated that the high expression of KLRG1 on tumor cells may induce the activation of immune cells. Altogether, these results revealed that KLRG1, BTK and CCR2 may interact through cell surface receptor signaling pathway to influence the proliferation of tumor cells or affect the immune response indirectly.

Graph: Fig. 6 The interaction between KLRG1, BTK and CCR2 influences the proliferation of lung tumor cells. A Protein-protein interaction analysis of KLRG1, BTK, CCR2, SCML4 and their-related genes. Each node represents a different gene. Each line represents a connection between two different genes. B-D The expression of BTK (B), CCR2 (C) and SCML4 (D) in KLRG1-knockdown A549 tumor cells were evaluated by qRT-PCR, n = 3 for each group, P values are derived from two-tailed unpaired Student's t-test. E The effect of E-Cadherin antibody or IgG1 treatment on the proliferation of A549 tumor cells treated with scramble or KLRG1 shRNA, n = 4 for each group, P values are derived from two-tailed unpaired Student's t-test. F-G Regression analysis of the correlation between KLRG1 and IFNG (F) or CD274 (G), P values are derived from Spearman and Pearson correlation analysis. Data represent mean ± SD. **, P < 0.01, ***, P < 0.001, ns, no significance

Table 6 Functional and pathway enrichment analysis

Category

ID

Term Description

Observed gene count

Background gene count

P-value

Included Gene

Biological Process (GO)

GO:0007166

Cell Surface Receptor Signaling Pathway

31

2198

2.11E-23

BTK,CCR2,KLRG1

GO:0002376

Immune System Process

28

2370

2.94E-18

BTK,CCR2,KLRG1

GO:0007165

Signal Transduction

32

4738

1.10E-15

BTK,CCR2,KLRG1

GO:0006955

Immune Response

23

1560

1.11E-15

BTK,CCR2,KLRG1

GO:0051716

Cellular Response to Stimulus

33

6212

7.71E-14

BTK,CCR2,KLRG1

GO:0006952

Defense Response

18

1234

1.82E-11

BTK,CCR2,KLRG1

GO:0045087

Innate Immune Response

14

676

1.68E-10

BTK,KLRG1

GO:0006950

Response to Stress

22

3267

1.95E-08

BTK,CCR2,KLRG1

GO:0050794

Regulation of Cellular Process

34

10,484

2.12E-08

BTK,CCR2,KLRG1,SCML4

GO:0006954

Inflammatory Response

9

482

2.51E-06

BTK,CCR2,KLRG1

Molecular Function (GO)

GO:0005515

Protein Binding

29

6605

1.32E-07

BTK,CCR2

GO:0005102

Signaling Receptor Binding

15

1513

7.57E-07

BTK,CCR2

GO:0005488

Binding

31

11,878

0.001

BTK,CCR2,KLRG1

GO:0042802

Identical Protein Binding

9

1754

0.0128

BTK,CCR2

Cellular Component (GO)

GO:0071944

Cell Periphery

23

5254

2.02E-05

BTK,CCR2,KLRG1

GO:0005886

Plasma Membrane

22

5159

5.39E-05

BTK,CCR2,KLRG1

GO:0005623

Cell

34

16,271

0.011

BTK,CCR2,KLRG1,SCML4

GO:0016020

Membrane

23

8420

0.0169

BTK,CCR2,KLRG1

GO:0005622

Intracellular

31

14,286

0.037

BTK,CCR2,KLRG1,SCML4

KEGG Pathways

hsa04662

B Cell Receptor Signaling Pathway

8

71

3.96E-11

BTK

hsa04062

Chemokine Signaling Pathway

8

181

2.36E-08

CCR2

hsa04664

Fc Epsilon Ri Signaling Pathway

6

67

5.25E-08

BTK

hsa05340

Primary Immunodeficiency

5

37

1.53E-07

BTK

Reactome Pathways

HSA-168256

Immune System

27

1925

4.00E-19

BTK,CCR2,KLRG1

HSA-1280218

Adaptive Immune System

16

733

8.03E-13

BTK,KLRG1

HSA-168249

Innate Immune System

17

1012

5.02E-12

BTK,CCR2

HSA-162582

Signal Transduction

22

2605

1.53E-10

BTK,CCR2

UniProt Keywords

KW-0391

Immunity

8

507

5.56E-05

BTK,KLRG1

KW-1003

Cell Membrane

16

3208

0.00064

BTK,CCR2,KLRG1

KW-0597

Phosphoprotein

25

8066

0.0018

BTK,CCR2,SCML4

KW-0399

Innate Immunity

4

309

0.0141

BTK,KLRG1

P values are derived from false discovery rate corrections

Discussion

In the past few decades, experts have explored the mechanisms of lung adenocarcinoma formation and development through extensive basic and clinical research. The treatment of LUAD also has made huge progress. However, it is an urgent need to develop biomarkers to predict and monitor the response and clinical benefit of immunotherapy in LUAD patients. In this study, through the analyze of the data from the Gene Expression Omnibus, the Cancer Genome Atlas and the Genotype-Tissue Expression, we found that the expression of KLRG1, BTK, CCR2 and SCML4 was significantly downregulated in LUAD tissues compared to normal controls. And the expression of these four factors significantly predicted the overall survival time in patients with LUAD. Furthermore, low expression of KLRG1 also correlated with poor responses in LUAD patients after immune-checkpoint inhibitor treatment.

Killer-cell lectin like receptor G1 (KLRG1) is expressed on NK cells and antigen-experienced T cells and has been assumed to be a marker of senescence [[20]]. Despite the extensive use of KLRG1 as a marker of differentiation, KLRG1 possesses an immune receptor tyrosine-based inhibitory motif (ITIM) in its cytoplasmic domain, suggesting that it may play an inhibitory role in human NK cells and T cells [[21]]. KLRG1 has been little studied in human tumor samples. There is only one paper published in Oncotarget which investigated the expression of KLRG1 on tumor cells. In that study, it showed that KLRG1 expression is upregulated in human tumor samples after a variety of therapies, including radiation, endocrine therapy, chemotherapy, and immunotherapy [[23]]. And we are the first study to focus on the direct role of KLRG1 on tumor cells. In our study, we found that low expression of KLRG1 also correlated with poor responses in LUAD patients after immune-checkpoint inhibitor treatment. Direct assessment of PD-L1 expression on tumor cells is a logical biomarker for the prediction of treatment response to anti-PD-1 or anti-PD-L1 therapies. However, patients show PD-L1-negative by immunohistochemistry can still achieve clinical benefit with anti-PD-1 or anti-PD-L1 therapies. KLRG1-mediated inhibition of NK cell function reveals that KLRG1/ligand interactions inhibit the cytolytic activity of polyclonal human NK cells by interfering with both degranulation and interferon γ (IFNγ) release [[24]]. And blocking KLRG1 signaling during TCR activation using antibodies against its ligand, E-cadherin, enhances proliferative activity of T cells [[25]]. Meanwhile, some study already described that immune gene signatures, especially those induced by IFNγ, might be robust biomarkers for predicting clinical benefit to anti-PD-1 or anti-PD-L1 therapies [[6]]. Consistent with these studies, it led us to propose that the expression of KLRG1 on LUAD tumor cells will competitively bind to its ligand E-cadherin with T cells or NK cells. The decreased level of E-cadherin in the tumor microenvironment will reduce the inhibitory role of KLRG1 on T cells and NK cells, which leads to high release of degranulation and interferon γ. Consequently, the anti-tumor effect of the immune system is strengthened. On the other hand, the expression of KLRG1 was directly related to the overall survival and progress free survival of LUAD patients. So, KLRG1 may also play a directly role to the tumor differentiation and progression. Our results also indicated that knockdown of KLRG1 promoted the proliferation of A549 lung cancer cells. And E-cadherin maybe the ligand of KLRG1 on tumor cells. How KLRG1 influences tumor cells proliferation is an interesting question which deserves further study. On the other hand, the expression of KLRG1 is not very high in lung tumor tissues which is indeed a limitation as a biomarker. So, we need sensitive methods, such as PCR, to detect the expression of KLRG1 in tumor tissues.

Bruton's tyrosine kinase (BTK), a Tec family non-receptor protein kinase, plays a crucial role in B-cell activation, proliferation, maturation, differentiation, and survival [[26]]. BTK has emerged as a novel molecular target in some B-cell leukemias and lymphomas where it is commonly overexpressed [[27]]. A Phase 1b/2 Study investigated the efficacy of the Bruton tyrosine kinase inhibitor Ibrutinib and the PD-L1 inhibitor Durvalumab in patients with pretreated solid tumors which includes 28 non-small cell lung cancer patients. The results indicated that the combination of ibrutinib and durvalumab did not show meaningful activity in any of the tumor types studied, and, therefore, recruitment was stopped due to lack of efficacy [[28]]. These results indicated that BTK may play different roles in LUAD compared with B-cell malignancies. The expression of BTK in different tumor types also implies this point. BTK is overexpressed in Diffuse Large B-cell Lymphoma (DLBCL) than normal tissues, but has a lower expression in LUAD and LUSC than normal tissues. So, it is really an interesting question that how BTK influences the development of lung cancer.

The CCL2/CCR2 signaling axis is first characterized as a chemotactic molecule with physiological regulating roles in inflammation. And the CCL2/CCR2 axis has generated increasing interest in recent years due to its association with the progression of cancer. On the other hand, CCL2/CCR2 has been shown to exert both pro- and anti-tumor effects [[29]]. CCL2 may also act to attract antitumor immune cells and is required for efficient immunosurveillance, implying that the inhibition of CCL2 may promote neocarcinogenesis as well as the development of metastases [[30]–[32]]. In our study, the expression of CCR2 was positively correlated with patients' survival which indicated that CCL2/CCR2 axis may play anti-tumor effects in LUAD. Very little research is focused on SCML4 (Scm Polycomb Group Protein Like 4). But a study shown that SCML4 makes functional contributions to processes critical for atherosclerosis (endothelial cell activation and survival, inflammation, and adhesion) and decreased expression of SCML4 exacerbates endothelial dysfunction and vascular remodeling in a rat model [[33]].

In our study, we found that KLRG1, BTK, CCR2 and SCML4 were co-expressed in LUAD patients. Knockdown of KLRG1 enhanced the proliferation of A549 lung tumor cells. And low expression of the four factors associated with unfavorable overall survival in LUAD patients. Combined the analysis of protein-protein interaction, it implies that KLRG1, BTK, CCR2 and SCML4 may influence the LUAD development through immune system process. The detailed mechanisms of these four factors involved in LUAD development are intriguing questions needed to be investigated.

Conclusions

Altogether, the results presented here indicate that KLRG1, BTK, CCR2 and SCML4 play important roles in the development of lung adenocarcinoma, and their expression can be effective biomarkers to predict LUAD patients' survival. Notably, the expression of KLRG1 is also a good biomarker to predict the treatment response of immune-checkpoint inhibitors.

Acknowledgements

No.

Authors' contributions

Idea and design: XY, ZH, XZ. Data collection: XY, YZ. Data analysis: XY, YZ, ZH, XZ. Manuscript writing: XY. Manuscript revision: XY, YZ, ZH, XZ. All authors read and approved the version of the manuscript to be published. All authors take responsibility for appropriate content.

Funding

Not applicable.

Availability of data and materials

All data are included in this article.

Declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Supplementary Information

Graph: Additional file 1: Supplementary Figure 1. Uncropped western blots used in Fig. 3b. The figure shows all original uncropped blots. The western blots of KLRG1-shRNA3-5 were not shown in the manuscript and Fig. 3b, because they did not shown significant knockdown efficacy. Blots were cropped where indicated by the red lines. Supplementary Figure 2. Uncropped western blots used in Fig. 3e. The figure shows all original uncropped blots. Blots were cropped where indicated by the red lines.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References 1 Tan WL, Jain A, Takano A, Newell EW, Iyer NG, Lim WT, Tan EH, Zhai W, Hillmer AM, Tam WL, Tan DSW. Novel therapeutic targets on the horizon for lung cancer. Lancet Oncol. 2016; 17; 8: e347-e362. 1:CAS:528:DC%2BC28Xht1ygsrbF. 10.1016/S1470-2045(16)30123-1. 27511159 2 Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019; 69; 1: 7-34. 10.3322/caac.21551 3 Barnett R. Lung cancer. Lancet. 2017; 390; 10098: 928. 10.1016/S0140-6736(17)32243-2. 28872022 4 de Groot P, Munden RF. Lung cancer epidemiology, risk factors, and prevention. Radiol Clin N Am. 2012; 50; 5: 863-876. 10.1016/j.rcl.2012.06.006. 22974775. 22974775 5 Relli V, Trerotola M, Guerra E, Alberti S. Abandoning the notion of non-small cell lung Cancer. Trends Mol Med. 2019; 25; 7: 585-594. 10.1016/j.molmed.2019.04.012. 31155338 6 Gibney GT, Weiner LM, Atkins MB. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016; 17; 12: e542-e551. 1:CAS:528:DC%2BC28XhvF2gsL7M. 10.1016/S1470-2045(16)30406-5. 27924752. 5702534 7 Ahn BC, Pyo KH, Xin CF, Jung D, Shim HS, Lee CY, Park SY, Yoon HI, Hong MH, Cho BC, Kim HR. Comprehensive analysis of the characteristics and treatment outcomes of patients with non-small cell lung cancer treated with anti-PD-1 therapy in real-world practice. J Cancer Res Clin Oncol. 2019; 145; 6: 1613-1623. 1:CAS:528:DC%2BC1MXhtFWnurvP. 10.1007/s00432-019-02899-y. 30911841. 6527531 8 Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, Ferrucci PF, Hill A, Wagstaff J, Carlino MS, Haanen JB, Maio M, Marquez-Rodas I, McArthur GA, Ascierto PA, Long GV, Callahan MK, Postow MA, Grossmann K, Sznol M, Dreno B, Bastholt L, Yang A, Rollin LM, Horak C, Hodi FS, Wolchok JD. Combined Nivolumab and Ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015; 373; 1: 23-34. 1:CAS:528:DC%2BC2MXht12isr7M. 10.1056/NEJMoa1504030. 5698905. 5698905 9 Yoon J, Suh YJ, Han K, Cho H, Lee HJ, Hur J, Choi BW. Utility of CT radiomics for prediction of PD-L1 expression in advanced lung adenocarcinomas. Thorac Cancer. 2020; 11; 4: 993-1004. 1:CAS:528:DC%2BB3cXmtFSgsL8%3D. 10.1111/1759-7714.13352. 32043309. 7113038 Kim R, Keam B, Kim S, Kim M, Kim SH, Kim JW, Kim YJ, Kim TM, Jeon YK, Kim DW, Chung DH, Lee JS, Heo DS. Differences in tumor microenvironments between primary lung tumors and brain metastases in lung cancer patients: therapeutic implications for immune checkpoint inhibitors. BMC Cancer. 2019; 19; 1: 19. 10.1186/s12885-018-5214-8. 30616523. 6322302 Cho JH, Jung HA, Lee SH, Ahn JS, Ahn MJ, Park K, Sun JM. Impact of EGFR mutation on the clinical efficacy of PD-1 inhibitors in patients with pulmonary adenocarcinoma. J Cancer Res Clin Oncol. 2019; 145; 5: 1341-1349. 1:CAS:528:DC%2BC1MXhtFWntbfJ. 10.1007/s00432-019-02889-0. 30900155 Liao J, Shen J, Leng Q, Qin M, Zhan M, Jiang F. MicroRNA-based biomarkers for diagnosis of non-small cell lung cancer (NSCLC). Thorac Cancer. 2020; 11; 3: 762-768. 1:CAS:528:DC%2BB3cXktFamsrk%3D. 10.1111/1759-7714.13337. 31994346. 7049510 Jiang T, Xu XF, Qiao M, Li XF, Zhao C, Zhou F, Gao GH, Wu FY, Chen XX, Su CX, Ren S, Zhai C, Zhou C. Comprehensive evaluation of NT5E/CD73 expression and its prognostic significance in distinct types of cancers. BMC Cancer. 2018; 18; 1: 267. 1:CAS:528:DC%2BC1cXisFWnt7fJ. 10.1186/s12885-018-4073-7. 29514610. 5842577 Tang Z, Kang B, Li C, Chen T, Zhang Z. GEPIA2: an enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res. 2019; 47; W1: W556-W560. 1:CAS:528:DC%2BB3cXisVCmur0%3D. 10.1093/nar/gkz430. 31114875. 6602440 Cline MS, Craft B, Swatloski T, Goldman M, Ma S, Haussler D, Zhu JC. Exploring TCGA pan-Cancer data at the UCSC Cancer genomics browser. Sci Rep-Uk. 2013; 3: 1-6 Gao JJ, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun YC, Jacobsen A, Sinha R, Larsson E. integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013; 6: 269. 1:CAS:528:DC%2BC3sXhsVyhur3O. 10.1126/scisignal.2004088 Cerami E, Gao JJ, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, Antipin Y, Reva B, Goldberg AP, Sander C, Schultz N. The cBio Cancer genomics portal: an open platform for exploring multidimensional Cancer genomics data. Cancer Discov. 2012; 2; 5: 401-404. 10.1158/2159-8290.CD-12-0095. 22588877 Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP, Kuhn M, Bork P, Jensen LJ, von Mering C. STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 2015; 43; D1: D447-D452. 1:CAS:528:DC%2BC2sXhtVymt7bE. 10.1093/nar/gku1003. 25352553 Prat A, Navarro A, Pare L, Reguart N, Galvan P, Pascual T, Martinez A, Nuciforo P, Comerma L, Alos L. Immune-related gene expression profiling after PD-1 blockade in non-small cell lung carcinoma, head and neck squamous cell carcinoma, and melanoma. Cancer Res. 2017; 77; 13: 3540-3550. 1:CAS:528:DC%2BC2sXhtFSmsrfK. 10.1158/0008-5472.CAN-16-3556. 28487385 Henson SM, Akbar AN. KLRG1-more than a marker for T cell senescence. Age. 2009; 31; 4: 285-291. 1:CAS:528:DC%2BC3cXpslCmug%3D%3D. 10.1007/s11357-009-9100-9. 19479342. 2813054 Xu R, Abramson J, Fridkin M, Pecht I. SH2 domain-containing inositol polyphosphate 5 '-phosphatase is the main mediator of the inhibitory action of the mast cell function-associated antigen. J Immunol. 2001; 167; 11: 6394-6402. 1:CAS:528:DC%2BD3MXovVCisbo%3D. 10.4049/jimmunol.167.11.6394. 11714805 Tessmer MS, Fugere C, Stevenaert F, Naidenko OV, Chong HJ, Leclercq G, Brossay L. KLRG1 binds cadherins and preferentially associates with SHIP-1. Int Immunol. 2007; 19; 4: 391-400. 1:CAS:528:DC%2BD2sXjslektbg%3D. 10.1093/intimm/dxm004. 17307799 Greenberg SA, Kong SW, Thompson E, Gulla SV. Co-inhibitory T cell receptor KLRG1: human cancer expression and efficacy of neutralization in murine cancer models. Oncotarget. 2019; 10; 14: 1399-1406. 10.18632/oncotarget.26659. 30858925. 6402715 Schwartzkopff S, Grundemann C, Schweier O, Rosshart S, Karjalainen KE, Becker KF, Pircher H. Tumor-associated E-cadherin mutations affect binding to the killer cell lectin-like receptor G1 in humans. J Immunol. 2007; 179; 2: 1022-1029. 1:CAS:528:DC%2BD2sXnt1Wnsr4%3D. 10.4049/jimmunol.179.2.1022. 17617594 Henson SM, Franzese O, Macaulay R, Libri V, Azevedo RI, Kiani-Alikhan S, Plunkett FJ, Masters JE, Jackson S, Griffiths SJ, Pircher HP, Soares MVD, Akbar AN. KLRG1 signaling induces defective Akt (ser(473)) phosphorylation and proliferative dysfunction of highly differentiated CD8(+) T cells. Blood. 2009; 113; 26: 6619-6628. 1:CAS:528:DC%2BD1MXotlWru7k%3D. 10.1182/blood-2009-01-199588. 19406987 Mohamed AJ, Yu L, Backesjo CM, Vargas L, Faryal R, Aints A, Christensson B, Berglof A, Vihinen M, Nore BF. Bruton's tyrosine kinase (Btk): function, regulation, and transformation with special emphasis on the PH domain. Immunol Rev. 2009; 228; 1: 58-73. 1:CAS:528:DC%2BD1MXhsFGls7rO. 10.1111/j.1600-065X.2008.00741.x. 19290921 Singh SP, Dammeijer F, Hendriks RW. Role of Bruton's tyrosine kinase in B cells and malignancies. Mol Cancer. 2018; 17: 1-23. 1:CAS:528:DC%2BC1MXisFyqtrs%3D. 10.1186/s12943-017-0753-1 Hong D, Rasco D, Veeder M, Luke JJ, Chandler J, Balmanoukian A, George TJ, Munster P, Berlin JD, Gutierrez M, Mita A, Wakelee H, Samakoglu S, Guan S, Dimery I, Graef T, Borazanci E. A phase 1b/2 study of the Bruton tyrosine kinase inhibitor Ibrutinib and the PD-L1 inhibitor Durvalumab in patients with pretreated solid tumors. Oncology. 2019; 97; 2: 102-111. 1:CAS:528:DC%2BC1MXhsFKjs7%2FL. 10.1159/000500571. 31230047 Lim SY, Yuzhalin AE, Gordon-Weeks AN, Muschel RJ. Targeting the CCL2-CCR2 signaling axis in cancer metastasis. Oncotarget. 2016; 7; 19: 28697-28710. 10.18632/oncotarget.7376. 26885690. 5053756 Lanca T, Costa MF, Goncalves-Sousa N, Rei M, Grosso AR, Penido C, Silva-Santos B. Protective role of the inflammatory CCR2/CCL2 chemokine pathway through recruitment of type 1 cytotoxic gamma delta T lymphocytes to tumor beds. J Immunol. 2013; 190; 12: 6673-6680. 1:CAS:528:DC%2BC3sXptFensbg%3D. 10.4049/jimmunol.1300434. 23686489 Molon B, Ugel S, Del Pozzo F, Soldani C, Zilio S, Avella D, De Palma A, Mauri P, Monegal A, Rescigno M. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J Exp Med. 2011; 208; 10: 1949-1962. 1:CAS:528:DC%2BC3MXht1KjsbnI. 10.1084/jem.20101956. 21930770. 3182051 Rollins BJ, Sunday ME. Suppression of tumor formation in vivo by expression of the JE gene in malignant cells. Mol Cell Biol. 1991; 11; 6: 3125-3131. 1:CAS:528:DyaK3MXksVOmsbk%3D. 10.1128/mcb.11.6.3125-3131.1991. 2038321. 360158 Li Y, Wang DW, Chen YD, Chen C, Guo J, Zhang S, Sun ZJ, Ding H, Yao Y, Zhou L, Xu K, Song C, Yang F, Zhao B, Yan H, Wang WJ, Wu C, Lu X, Yang X, Dong J, Zheng G, Tian S, Cui Y, Jin L, Liu G, Cui H, Wang S, Jiang F, Wang C, Erdmann J, Zeng L, Huang S, Zhong J, Ma Y, Chen W, Sun J, Lei W, Chen S, Rao S, Gu D, Schunkert H, Tian XL. Genome-wide association and functional studies identify SCML4 and THSD7A as novel susceptibility genes for coronary artery disease. Arterioscl Throm Vas. 2018; 38; 4: 964-975. 1:CAS:528:DC%2BC1cXltFenuro%3D. 10.1161/ATVBAHA.117.310594

By Xiaodong Yang; Yuexin Zheng; Zhihai Han and Xiliang Zhang

Reported by Author; Author; Author; Author

Titel:
Functions and clinical significance of KLRG1 in the development of lung adenocarcinoma and immunotherapy
Autor/in / Beteiligte Person: Han, Zhihai ; Yang, Xiaodong ; Zhang, Xiliang ; Zheng, Yuexin
Link:
Zeitschrift: BMC Cancer, Jg. 21 (2021), Heft 1, S. 1-16
Veröffentlichung: BMC, 2021
Medientyp: unknown
ISSN: 1471-2407 (print)
Schlagwort:
  • Male
  • 0301 basic medicine
  • KLRG1
  • Lung adenocarcinoma
  • Cancer Research
  • CCR2
  • Lung Neoplasms
  • medicine.medical_treatment
  • Adenocarcinoma of Lung
  • KLRG1 Gene
  • 03 medical and health sciences
  • Chemokine receptor
  • 0302 clinical medicine
  • Genetics
  • medicine
  • Humans
  • Bruton's tyrosine kinase
  • Lectins, C-Type
  • Receptors, Immunologic
  • RC254-282
  • Cell Proliferation
  • Gene knockdown
  • biology
  • Gene Expression Profiling
  • Neoplasms. Tumors. Oncology. Including cancer and carcinogens
  • Immunotherapy
  • medicine.disease
  • Prognosis
  • 030104 developmental biology
  • Oncology
  • Cell culture
  • 030220 oncology & carcinogenesis
  • biology.protein
  • Cancer research
  • Adenocarcinoma
  • Female
  • Research Article
Sonstiges:
  • Nachgewiesen in: OpenAIRE
  • Sprachen: English
  • Language: English
  • Rights: OPEN

Klicken Sie ein Format an und speichern Sie dann die Daten oder geben Sie eine Empfänger-Adresse ein und lassen Sie sich per Email zusenden.

oder
oder

Wählen Sie das für Sie passende Zitationsformat und kopieren Sie es dann in die Zwischenablage, lassen es sich per Mail zusenden oder speichern es als PDF-Datei.

oder
oder

Bitte prüfen Sie, ob die Zitation formal korrekt ist, bevor Sie sie in einer Arbeit verwenden. Benutzen Sie gegebenenfalls den "Exportieren"-Dialog, wenn Sie ein Literaturverwaltungsprogramm verwenden und die Zitat-Angaben selbst formatieren wollen.

xs 0 - 576
sm 576 - 768
md 768 - 992
lg 992 - 1200
xl 1200 - 1366
xxl 1366 -