1
|
Shaikh SB, Gouda MM, Khandhal I, Rahman T, Shetty A, Bhandary YP. Alterations in mRNA Expression Levels of Tight Junction Proteins in the Blood Cells of Smokers with or without COPD. Endocr Metab Immune Disord Drug Targets 2023; 23:389-395. [PMID: 35642119 DOI: 10.2174/1871530322666220531121609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/24/2022] [Accepted: 03/17/2022] [Indexed: 11/22/2022]
Abstract
AIM This study aimed to assess the role of Tight junction proteins (TJPs) and claudins in smokers with and without COPD compared to healthy individuals. BACKGROUND Chronic obstructive pulmonary disease (COPD) is a complex chronic respiratory disease, including various inflammatory mediators. The prime etiological element in the development of COPD is cigarette smoking. The lung airway epithelium comprises beneficial immunological barriers to draw in insults, such as environmental particulates, cigarette smoke, etc. Tight junctions (TJ) connected by transmembrane proteins determine epithelial permeability. Cigarette smoke is indicated to defect TJ integrity. The possible involvement of the airway epithelium in the pathogenesis of COPD has recently become apparent; however, its detailed mechanisms remain elusive. The integrity of airway epithelium is crucial for airway homeostasis; defective airway barrier activity contributes to COPD. OBJECTIVE In the present study, the objective was to investigate mRNA expression levels of TJP's like TJP-1, TJP-2, TJP-3, Tight junction-associated proteins-1, claudin-1, claudin-3, claudin-4, claudin-7, claudin-10, claudin-15, claudin-19, and claudin-25 from blood samples of smokers with COPD and compared them with smokers without COPD and healthy individuals. METHODS The mRNA expressions were evaluated by the quantitative PCR method. RESULTS The gene expressions of these TJPs were significantly down-regulated, specifically in COPD patients with a history of smoking (Smokers with COPD). Besides, FEV% was also established for these patients. Similarly, smokers with COPD showed a significant increase in the expression levels of transcription factors, like ZEB-1, ZEB-2, PDGFA, and HDGF, compared to COPD patients without a history of smoking (smokers without COPD) and the healthy subjects. CONCLUSION In conclusion, cigarette smoke disrupts TJ of the human airway epithelium, and the transcriptional factors counteract this smoke-induced COPD. Thus, TJPs may serve as protective elements for airway epithelial homeostasis during COPD.
Collapse
Affiliation(s)
- Sadiya Bi Shaikh
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore -575018, Karnataka, India
- Department of Molecular, Cell and Systems Biology, Institute for Integrative Genome Biology, University of California at Riverside, Riverside, CA 92521, USA
| | - Mahesh Manjunath Gouda
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore -575018, Karnataka, India
- Rahman Lab, Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, United States of America
| | - Irfan Khandhal
- Department of Pulmonary Medicine, Yenepoya Medical College (Deemed to be University), Deralakatte, Mangalore, Karnataka 575018, India
| | - Tanyeem Rahman
- Department of Anatomy, Yenepoya Medical College (Deemed to be University), Deralakatte, Mangalore, Karnataka, 575018, India
| | - Ashwini Shetty
- Department of Anatomy, Yenepoya Medical College (Deemed to be University), Deralakatte, Mangalore, Karnataka, 575018, India
| | | |
Collapse
|
2
|
Brisebarre A, Ancel J, Ponchel T, Loeffler E, Germain A, Dalstein V, Dormoy V, Durlach A, Delepine G, Deslée G, Polette M, Nawrocki-Raby B. Transcriptomic FHIT low/pHER2 high signature as a predictive factor of outcome and immunotherapy response in non-small cell lung cancer. Front Immunol 2022; 13:1058531. [PMID: 36544755 PMCID: PMC9760670 DOI: 10.3389/fimmu.2022.1058531] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/18/2022] [Indexed: 12/11/2022] Open
Abstract
Introduction In recent decades, the development of immunotherapy and targeted therapies has considerably improved the outcome of non-small cell lung cancer (NSCLC) patients. Despite these impressive clinical benefits, new biomarkers are needed for an accurate stratification of NSCLC patients and a more personalized management. We recently showed that the tumor suppressor fragile histidine triad (FHIT), frequently lost in NSCLC, controls HER2 receptor activity in lung tumor cells and that tumor cells from NSCLC patients harboring a FHITlow/pHER2high phenotype are sensitive to anti-HER2 drugs. Here, we sought to identify the transcriptomic signature of this phenotype and evaluate its clinical significance. Materials and methods We performed RNA sequencing analysis on tumor cells isolated from NSCLC (n=12) according to FHIT/pHER2 status and a functional analysis of differentially regulated genes. We also investigated the FHITlow/pHER2high signature in The Cancer Genome Atlas (TCGA) lung adenocarcinoma (LUAD) (n=489) and lung squamous cell carcinoma (LUSC) (n=493) cohorts and used the tumor immune dysfunction and exclusion (TIDE) model to test the ability of this signature to predict response to immune checkpoint inhibitors (ICI). Results We showed that up-regulated genes in FHITlow/pHER2high tumors were associated with cell proliferation, metabolism and metastasis, whereas down-regulated genes were related to immune response. The FHITlow/pHER2high signature was associated with the higher size of tumors, lymph node involvement, and late TNM stages in LUAD and LUSC cohorts. It was identified as an independent predictor of overall survival (OS) in LUAD cohort. FHITlow/pHER2high tumors were also predictive of poor response to ICI in both LUAD and LUSC cohorts. Conclusion These data suggest that ICI might not be a relevant option for NSCLC patients with FHITlow/pHER2high tumors and that anti-HER2 targeted therapy could be a good therapeutic alternative for this molecular subclass with poorer prognosis.
Collapse
Affiliation(s)
- Audrey Brisebarre
- INSERM, Université de Reims Champagne-Ardenne, P3Cell, UMR-S 1250, SFR CAP Santé, Reims, France
| | - Julien Ancel
- INSERM, Université de Reims Champagne-Ardenne, P3Cell, UMR-S 1250, SFR CAP Santé, Reims, France,CHU Reims, Hôpital Maison Blanche, Service de Pneumologie, Reims, France
| | - Théophile Ponchel
- INSERM, Université de Reims Champagne-Ardenne, P3Cell, UMR-S 1250, SFR CAP Santé, Reims, France
| | - Emma Loeffler
- INSERM, Université de Reims Champagne-Ardenne, P3Cell, UMR-S 1250, SFR CAP Santé, Reims, France
| | - Adeline Germain
- INSERM, Université de Reims Champagne-Ardenne, P3Cell, UMR-S 1250, SFR CAP Santé, Reims, France
| | - Véronique Dalstein
- INSERM, Université de Reims Champagne-Ardenne, P3Cell, UMR-S 1250, SFR CAP Santé, Reims, France,CHU Reims, Pôle de Biologie Territoriale, Service de Pathologie, Reims, France
| | - Valérian Dormoy
- INSERM, Université de Reims Champagne-Ardenne, P3Cell, UMR-S 1250, SFR CAP Santé, Reims, France
| | - Anne Durlach
- INSERM, Université de Reims Champagne-Ardenne, P3Cell, UMR-S 1250, SFR CAP Santé, Reims, France,CHU Reims, Pôle de Biologie Territoriale, Service de Pathologie, Reims, France
| | - Gonzague Delepine
- INSERM, Université de Reims Champagne-Ardenne, P3Cell, UMR-S 1250, SFR CAP Santé, Reims, France,CHU Reims, Hôpital Robert Debré, Service de Chirurgie cardio-vasculaire et thoracique, Reims, France
| | - Gaëtan Deslée
- INSERM, Université de Reims Champagne-Ardenne, P3Cell, UMR-S 1250, SFR CAP Santé, Reims, France,CHU Reims, Hôpital Maison Blanche, Service de Pneumologie, Reims, France
| | - Myriam Polette
- INSERM, Université de Reims Champagne-Ardenne, P3Cell, UMR-S 1250, SFR CAP Santé, Reims, France,CHU Reims, Pôle de Biologie Territoriale, Service de Pathologie, Reims, France
| | - Béatrice Nawrocki-Raby
- INSERM, Université de Reims Champagne-Ardenne, P3Cell, UMR-S 1250, SFR CAP Santé, Reims, France,*Correspondence: Béatrice Nawrocki-Raby,
| |
Collapse
|
3
|
Jeon Y, Kang H, Yang Y, Park D, Choi B, Kim J, Kim J, Nam K. A Novel Selective Axl/Mer/CSF1R Kinase Inhibitor as a Cancer Immunotherapeutic Agent Targeting Both Immune and Tumor Cells in the Tumor Microenvironment. Cancers (Basel) 2022; 14:cancers14194821. [PMID: 36230744 PMCID: PMC9563311 DOI: 10.3390/cancers14194821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/25/2022] [Accepted: 09/29/2022] [Indexed: 12/03/2022] Open
Abstract
Simple Summary Immune checkpoint blockade has had great success over the past decade, but many patients with cancer do not benefit because most immune checkpoint inhibitors only target T cells. Targeting non-T cell populations in the tumor microenvironment (TME) has been shown to affect responses to them. Simultaneous inhibition of Axl, Mer and CSF1R by a novel receptor tyrosine kinase inhibitor Q702 induces antitumor immunity by reducing the number of M2 macrophages and MDSCs and inducing M1 macrophages and cytotoxic CD8 T cells in the TME, and increasing the expression of MHC-I and E-cadherin in tumor cells. Our data indicate that therapy targeting both immune cells and cancer cells in the TME by Q702 can induce more effective clinical responses in patients with cancer. Abstract Although immune checkpoint blockade (ICB) represents a major breakthrough in cancer immunotherapy, only a limited number of patients with cancer benefit from ICB-based immunotherapy because most immune checkpoint inhibitors (ICIs) target only T cell activation. Therefore, targeting non-T cell components in the tumor microenvironment (TME) can help subvert resistance and increase the applications of ICB-based therapy. Axl and Mer are involved in the carcinogenesis of multiple types of cancer by modulating immune and biological behaviors within tumors. Colony stimulating factor 1 receptor (CSF1R) mediates tumorigenesis in the TME by enhancing tumor associated macrophage (TAM) and myeloid-derived suppressor cell (MDSC) infiltration, facilitating immune escape. Therefore, the simultaneous inhibition of Axl, Mer, and CSF1R kinases may improve therapeutic efficacy by targeting non-T cell components in the TME. Here, we present Q702, a selective, potent small molecule inhibitor targeting Axl, Mer, and CSF1R, for oral administration. Q702 induced antitumor activity in syngeneic tumor mouse models by: remodeling the TME toward immune stimulation; expanding M1 macrophage and CD8 T cell populations and decreasing M2 macrophage and MDSC populations in the TME; and increasing MHC class I and E-cadherin expression in tumor cells. Thus, Q702 may have great potential to broaden the coverage of populations benefiting from ICB-based immunotherapy.
Collapse
|
4
|
Ushio R, Murakami S, Saito H. Predictive Markers for Immune Checkpoint Inhibitors in Non-Small Cell Lung Cancer. J Clin Med 2022; 11:1855. [PMID: 35407463 PMCID: PMC9000007 DOI: 10.3390/jcm11071855] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically improved the outcomes of non-small cell lung cancer patients and have increased the possibility of long-term survival. However, few patients benefit from ICIs, and no predictive biomarkers other than tumor programmed cell death ligand 1 (PD-L1) expression have been established. Hence, the identification of biomarkers is an urgent issue. This review outlines the current understanding of predictive markers for the efficacy of ICIs, including PD-L1, tumor mutation burden, DNA mismatch repair deficiency, microsatellite instability, CD8+ tumor-infiltrating lymphocytes, human leukocyte antigen class I, tumor/specific genotype, and blood biomarkers such as peripheral T-cell phenotype, neutrophil-to-lymphocyte ratio, interferon-gamma, and interleukin-8. A tremendous number of biomarkers are in development, but individual biomarkers are insufficient. Tissue biomarkers have issues in reproducibility and accuracy because of intratumoral heterogeneity and biopsy invasiveness. Furthermore, blood biomarkers have difficulty in reflecting the tumor microenvironment and therefore tend to be less predictive for the efficacy of ICIs than tissue samples. In addition to individual biomarkers, the development of composite markers, including novel technologies such as machine learning and high-throughput analysis, may make it easier to comprehensively analyze multiple biomarkers.
Collapse
Affiliation(s)
| | - Shuji Murakami
- Kanagawa Cancer Center, Department of Thoracic Oncology, 2-3-2 Nakao, Asahi, Yokohama 241-8515, Japan; (R.U.); (H.S.)
| | | |
Collapse
|
5
|
Bravaccini S, Bronte G, Petracci E, Puccetti M, D'Arcangelo M, Ravaioli S, Tumedei MM, Maltoni R, Delmonte A, Cappuzzo F, Crinò L. The Expression of Programmed Death Ligand 1 and Vimentin in Resected Non-Metastatic Non-Small-Cell Lung Cancer: Interplay and Prognostic Effects. Front Cell Dev Biol 2021; 9:772216. [PMID: 34917615 PMCID: PMC8669606 DOI: 10.3389/fcell.2021.772216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/12/2021] [Indexed: 12/25/2022] Open
Abstract
Programmed death ligand 1 (PD-L1) is an immune checkpoint with a role in cancer-related immune evasion. It is a target for cancer immunotherapy and its expression is detected for the use of some immune checkpoint inhibitors in advanced non-small cell lung cancer patients (NSCLC). Vimentin is a key component of the epithelial-to-mesenchymal transition phenotype. Its expression has negative prognostic effects in NSCLC. In this study, we retrospectively evaluated PD-L1 and vimentin expression in tumor cells, immune infiltrate and PD-L1 positive immune infiltrate via immunohistochemistry in tissue samples from resected non-metastatic NSCLC patients. We explored the interplay between PD-L1 and vimentin expression through Spearman’s correlation test. We performed univariate analysis through the Cox models for demographic and clinico-pathological variables, and also for dichotomized biomarkers, i.e., PD-L1 and vimentin in tumor cells, both with 1 and 50% cutoffs. We used Kaplan-Meier method to estimate the overall survival, comparing both vimentin and PD-L1 positive patients with all the others. We found a weak positive correlation between PD-L1 and vimentin expressions in tumor cells (r = 0.25; p = 0.001). We also observed a statistically not significant trend towards a shorter overall survival in patients with both PD-L1 and vimentin expression >1% (HR = 1.36; 95% CI: 0.96–1.93, p = 0.087). In conclusion, these findings suggest that interplay between PD-L1 and vimentin may exist in non-metastatic NSCLC patients and the positivity of both markers in tumor tissue is associated with a trend towards a worse prognosis.
Collapse
Affiliation(s)
- Sara Bravaccini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giuseppe Bronte
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Elisabetta Petracci
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | | | | | - Sara Ravaioli
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Maria Maddalena Tumedei
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Roberta Maltoni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Angelo Delmonte
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | | | - Lucio Crinò
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
6
|
Zhang H, Qin G, Zhang C, Yang H, Liu J, Hu H, Wu P, Liu S, Yang L, Chen X, Zhao X, Wang L, Zhang Y. TRAIL promotes epithelial-to-mesenchymal transition by inducing PD-L1 expression in esophageal squamous cell carcinomas. J Exp Clin Cancer Res 2021; 40:209. [PMID: 34167551 PMCID: PMC8223376 DOI: 10.1186/s13046-021-01972-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 05/05/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Tumor necrosis factor-associated apoptosis-inducing ligand (TRAIL) was initially considered an immunity guard; however, its function remains controversial. Besides immune cells, lung and colon cancer cells have also been reported to express TRAIL, which can promote tumor invasion and metastasis. However, the biological function and underlying mechanism of action of TRAIL in esophageal squamous cell carcinoma (ESCC) remain poorly elucidated. METHODS The ESCC cells stemness, migration, and proliferation ability was assessed by sphere formation, Transwell, and CCK8 assay. The stemness- and epithelial-mesenchymal transition (EMT)- related genes expression levels were analyzed by Western blot and RT-qPCR. The signal activation was conducted by Western blot. The xenograft mouse experiments and lung metastasis model were performed to confirm our findings in vitro. RESULTS Herein, we found that TRAIL is a negative predictor in patients with ESCC. To further investigate the biological function of TRAIL, we established TRAIL knockdown and overexpression ESCC cell lines and found that TRAIL induced EMT and promoted tumor aggressiveness. Furthermore, we demonstrated that TRAIL- overexpressing cells upregulated PD-L1 expression, which was dependent on the p-ERK/STAT3 signaling pathway. We obtained similar results when using recombinant human TRAIL. Finally, we validated the biological role and mechanism of action of TRAIL in vivo. CONCLUSIONS These findings demonstrate that TRAIL promotes ESCC progression by enhancing PD-L1 expression, which induces EMT. This may explain the failure of TRAIL preclinical trials.
Collapse
Affiliation(s)
- Huanyu Zhang
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan, 450052, Zhengzhou, China
| | - Guohui Qin
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan, 450052, Zhengzhou, China
| | - Chaoqi Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Huiyun Yang
- School of Life Sciences, Zhengzhou University, 450052, Zhengzhou, China
| | - Jinyan Liu
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China
| | - Hongwei Hu
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China
| | - Peng Wu
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China
| | - Shasha Liu
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China
| | - Li Yang
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China
| | - Xinfeng Chen
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan, 450052, Zhengzhou, China
| | - Xueke Zhao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan, 450052, Zhengzhou, China
| | - Lidong Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan, 450052, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center & Cancer Center, the First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Henan, 450052, Zhengzhou, China.
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Henan, 450052, Zhengzhou, China.
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Henan Key Laboratory for Tumor Immunology and Biotherapy, 450052, Zhengzhou, China.
| |
Collapse
|
7
|
Li HJ, Ke FY, Lin CC, Lu MY, Kuo YH, Wang YP, Liang KH, Lin SC, Chang YH, Chen HY, Yang PC, Wu HC. ENO1 Promotes Lung Cancer Metastasis via HGFR and WNT Signaling-Driven Epithelial-to-Mesenchymal Transition. Cancer Res 2021; 81:4094-4109. [PMID: 34145039 DOI: 10.1158/0008-5472.can-20-3543] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/06/2021] [Accepted: 06/15/2021] [Indexed: 11/16/2022]
Abstract
ENO1 (α-enolase) expression is significantly correlated with reduced survival and poor prognosis in many cancer types, including lung cancer. However, the function of ENO1 in carcinogenesis remains elusive. In this study, we found that high expression of ENO1 is present in metastatic lung cancer cell lines and malignant tumors and is associated with poor overall survival of patients with lung cancer. Knockdown of ENO1 decreased cancer cell proliferation and invasiveness, whereas overexpression of ENO1 enhanced these processes. Moreover, ENO1 expression promoted tumor growth in orthotopic models and enhanced lung tumor metastasis in tail-vein injection models. These effects were mediated by upregulation of mesenchymal markers N-cadherin and vimentin and the epithelial-to-mesenchymal transition regulator SLUG, along with concurrent downregulation of E-cadherin. Mechanistically, ENO1 interacted with hepatocyte growth factor receptor (HGFR) and activated HGFR and Wnt signaling via increased phosphorylation of HGFR and the Wnt coreceptor LRP5/6. Activation of these signaling axes decreased GSK3β activity via Src-PI3K-AKT signaling and inactivation of the β-catenin destruction complex to ultimately upregulate SLUG and β-catenin. In addition, we generated a chimeric anti-ENO1 mAb (chENO1-22) that can decrease cancer cell proliferation and invasion. chENO1-22 attenuated cancer cell invasion by inhibiting ENO1-mediated GSK3β inactivation to promote SLUG protein ubiquitination and degradation. Moreover, chENO1-22 prevented lung tumor metastasis and prolonged survival in animal models. Taken together, these findings illuminate the molecular mechanisms underlying the function of ENO1 in lung cancer metastasis and support the therapeutic potential of a novel antibody targeting ENO1 for treating lung cancer. SIGNIFICANCE: This study shows that ENO1 promotes lung cancer metastasis via HGFR and WNT signaling and introduces a novel anti-ENO1 antibody for potential therapeutic use in lung cancer.
Collapse
Affiliation(s)
- Hsin-Jung Li
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Feng-Yi Ke
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Chia-Ching Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Mei-Yi Lu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Huei Kuo
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Ping Wang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.,Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | - Kang-Hao Liang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.,Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, Taiwan
| | - Shin-Chang Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.,Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, Taiwan
| | - Ya-Hsuan Chang
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Hsuan-Yu Chen
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Pan-Chyr Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan. .,Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, Taiwan
| |
Collapse
|
8
|
Gómez-Valenzuela F, Escobar E, Pérez-Tomás R, Montecinos VP. The Inflammatory Profile of the Tumor Microenvironment, Orchestrated by Cyclooxygenase-2, Promotes Epithelial-Mesenchymal Transition. Front Oncol 2021; 11:686792. [PMID: 34178680 PMCID: PMC8222670 DOI: 10.3389/fonc.2021.686792] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 05/13/2021] [Indexed: 12/11/2022] Open
Abstract
The tumor microenvironment (TME) corresponds to a complex and dynamic interconnection between the extracellular matrix and malignant cells and their surrounding stroma composed of immune and mesenchymal cells. The TME has constant cellular communication through cytokines that sustain an inflammatory profile, which favors tumor progression, angiogenesis, cell invasion, and metastasis. Although the epithelial-mesenchymal transition (EMT) represents a relevant metastasis-initiating event that promotes an invasive phenotype in malignant epithelial cells, its relationship with the inflammatory profile of the TME is poorly understood. Previous evidence strongly suggests that cyclooxygenase-2 (COX-2) overexpression, a pro-inflammatory enzyme related to chronic unresolved inflammation, is associated with common EMT-signaling pathways. This review article summarizes how COX-2 overexpression, within the context of the TME, orchestrates the EMT process and promotes initial metastatic-related events.
Collapse
Affiliation(s)
- Fernán Gómez-Valenzuela
- Department of Hematology-Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Enrico Escobar
- Department of Oral Pathology and Medicine, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Ricardo Pérez-Tomás
- Department of Pathology and Experimental Therapy - Bellvitge, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Viviana P Montecinos
- Department of Hematology-Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
9
|
Bronte G, Puccetti M, Petracci E, Landi L, Cravero P, Scodes S, Ulivi P, Ravaioli S, Tumedei MM, Burgio MA, Cappuzzo F, Delmonte A, Crinò L, Bravaccini S. The Interplay Between Programmed Death Ligand 1 and Vimentin in Advanced Non-Small-Cell Lung Cancer. Front Oncol 2021; 11:669839. [PMID: 34017688 PMCID: PMC8130554 DOI: 10.3389/fonc.2021.669839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/09/2021] [Indexed: 12/26/2022] Open
Abstract
Background Current therapy for non-small-cell lung cancer (NSCLC) frequently includes immune checkpoint inhibitors, such as pembrolizumab, and programmed death ligand 1 (PD-L1) positivity is mandatory for its use in this setting. Vimentin plays a role in carcinogenesis through the activation of the epithelial-to-mesenchymal transition (EMT) process. Its prognostic impact in NSCLC has been investigated in numerous studies but little data are available on its relation with PD-L1 expression. Patients and Methods We retrospectively retrieved data on patients with advanced NSCLC consecutively enrolled in a clinical trial at our institute. PD-L1 and vimentin expression were determined by immunohistochemistry. Correlations between variables were assessed using the Spearman correlation coefficient. The Kaplan-Meier method was used to estimate overall survival (OS) and the Log-rank test was used to compare survival curves. The association between demographic, clinical and biomarker information and survival was investigated with the Cox model. Results Fifty-three patients were included in the study. A weak positive correlation was observed between the PD-L1 and vimentin (ρ=0.41, P=0.003). Patients with PD-L1 values <1% showed a slightly better OS than those with higher values (HR=2.07; 95% CI: 0.92-4.65), but the difference was not significant (P=0.080). No difference in overall survival (OS) was observed on the basis of vimentin expression (HR=1.25; 95% CI: 0.59-2.66; P=0.554). Patients harboring both vimentin and PD-L1 negative expression (<1%) showed a trend towards better survival than those with ≥1% expression (HR=2.31; 95% CI: 0.87-6.17, P=0.093). No significant associations were observed between gender, age at diagnosis, stage at diagnosis, histology, KRAS or EGFR status, radical surgery or immunotherapy and OS. Conclusions The weak positive association between PD-L1 and vimentin suggests a potential interplay between these biomarkers. Further research is warranted to evaluate EMT and immune escape as two components of the same process.
Collapse
Affiliation(s)
- Giuseppe Bronte
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Maurizio Puccetti
- Pathology Unit, Azienda Unitá Sanitaria Locale (AUSL) Imola, Imola, Italy
| | - Elisabetta Petracci
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Lorenza Landi
- AUSL Romagna, Ospedale Santa Maria delle Croci, Ravenna, Italy
| | - Paola Cravero
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Simona Scodes
- AUSL Romagna, Ospedale Santa Maria delle Croci, Ravenna, Italy
| | - Paola Ulivi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sara Ravaioli
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Maria Maddalena Tumedei
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Marco Angelo Burgio
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | | | - Angelo Delmonte
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Lucio Crinò
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sara Bravaccini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
10
|
Chakafana G, Shonhai A. The Role of Non-Canonical Hsp70s (Hsp110/Grp170) in Cancer. Cells 2021; 10:254. [PMID: 33525518 PMCID: PMC7911927 DOI: 10.3390/cells10020254] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/21/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Although cancers account for over 16% of all global deaths annually, at present, no reliable therapies exist for most types of the disease. As protein folding facilitators, heat shock proteins (Hsps) play an important role in cancer development. Not surprisingly, Hsps are among leading anticancer drug targets. Generally, Hsp70s are divided into two main subtypes: canonical Hsp70 (Escherichia coli Hsp70/DnaK homologues) and the non-canonical (Hsp110 and Grp170) members. These two main Hsp70 groups are delineated from each other by distinct structural and functional specifications. Non-canonical Hsp70s are considered as holdase chaperones, while canonical Hsp70s are refoldases. This unique characteristic feature is mirrored by the distinct structural features of these two groups of chaperones. Hsp110/Grp170 members are larger as they possess an extended acidic insertion in their substrate binding domains. While the role of canonical Hsp70s in cancer has received a fair share of attention, the roles of non-canonical Hsp70s in cancer development has received less attention in comparison. In the current review, we discuss the structure-function features of non-canonical Hsp70s members and how these features impact their role in cancer development. We further mapped out their interactome and discussed the prospects of targeting these proteins in cancer therapy.
Collapse
Affiliation(s)
| | - Addmore Shonhai
- Department of Biochemistry, University of Venda, Private Bag X5050, 0950 Thohoyandou, South Africa
| |
Collapse
|
11
|
Shrestha R, Bridle KR, Crawford DHG, Jayachandran A. Immune checkpoint molecules are regulated by transforming growth factor (TGF)- β1-induced epithelial-to-mesenchymal transition in hepatocellular carcinoma. Int J Med Sci 2021; 18:2466-2479. [PMID: 34104078 PMCID: PMC8176170 DOI: 10.7150/ijms.54239] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer with a high mortality rate. Epithelial-to-mesenchymal transition (EMT) confers cancer cells with immune evasive ability by modulating the expression of immune checkpoints in many cancers. Thus, the aim of our study is to examine the interplay between EMT and immune checkpoint molecules in HCC. A reversible EMT model was utilised with transforming growth factor (TGF)-β1 as an EMT inducer for HCC cell lines Hep3B and PLC/PRF/5. HCC cells were treated with TGF-β1 for 72 h and the EMT status and immune checkpoint expression were examined. In addition, the migratory ability of HCC cells were examined using wound healing and transwell migration assays in the reversible EMT model. siRNA-mediated knockdown of immune checkpoint molecule, B7-H3, was further utilised to validate the association between TGF-β1-mediated EMT and immune checkpoint expression in HCC. In addition, a web-based platform, SurvExpress, was utilised to evaluate the association between expression of TGF-β1 in combination with immune checkpoint molecules and overall survival in HCC patients. We observed induction of EMT upon treatment of HCC cells with TGF-β1 revealed by reduced expression of epithelial markers along with increased expression of mesenchymal markers. Withdrawal of TGF-β1 reversed the process of EMT with elevated expression of epithelial markers and reduced expression of mesenchymal markers. TGF-β1 treatment elevated the migratory potential of HCC cells which was reversed following reversal assay. Notably, during TGF-β1-induced EMT, there was upregulation of immune checkpoint molecules PD-L1 and B7-H3. However, the reversal of EMT decreased the expression of PD-L1 and B7-H3. In addition, TGF-β1 driven EMT was reversed following knockdown of B7-H3 in both HCC cells further validating the interplay between TGF-β1-mediated EMT and immune checkpoint expression in HCC. Furthermore, the coordinate expression of TGF-β1 with PD-L1 (p=0.01487) and B7-H3 (p=0.009687) was correlated with poor overall survival in 422 HCC patients. Our study has demonstrated a close association between TGF-β1-mediated EMT and regulation of immune checkpoints in HCC.
Collapse
Affiliation(s)
- Ritu Shrestha
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, Queensland, Australia
| | - Kim R Bridle
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, Queensland, Australia
| | - Darrell H G Crawford
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, Queensland, Australia
| | - Aparna Jayachandran
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, Queensland, Australia.,Fiona Elsey Cancer Research Institute, Ballarat, Victoria, Australia
| |
Collapse
|
12
|
Moradpoor R, Salimi M. Crosstalk between Tumor Cells and Immune System Leads to Epithelial-Mesenchymal Transition Induction and Breast Cancer Progression. IRANIAN BIOMEDICAL JOURNAL 2021; 25:1-7. [PMID: 33129234 PMCID: PMC7748115 DOI: 10.29252/ibj.25.1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023]
Abstract
Herein, we review the current findings of how a variety of accessory cells could participate in shaping the tumor microenvironment and supporting the mechanisms by which cancer cells undertake the epithelial-mesenchymal transition (EMT). EMT, a complex of phenotypic changes, promotes cancer cell invasion and creates resistance to chemotherapies. Among the accessory cells present in the EMT, immune cells (both native and adaptive) can reciprocally influence the tumor cells features, promote EMT and negatively regulate the anticancer immune response. In this review, we look over the role of EMT in crosstalk between tumor cells and the immune system, with specific emphasis on breast tumors. Finally, we suggest that understanding the role of immune cells in cancer progression could create new opportunities for diagnostic and therapeutic interventions in cancer combination therapy.
Collapse
Affiliation(s)
- Raheleh Moradpoor
- Department of Basic Sciences, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Salimi
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
13
|
Qin S, Jiang J, Lu Y, Nice EC, Huang C, Zhang J, He W. Emerging role of tumor cell plasticity in modifying therapeutic response. Signal Transduct Target Ther 2020; 5:228. [PMID: 33028808 PMCID: PMC7541492 DOI: 10.1038/s41392-020-00313-5] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/25/2020] [Accepted: 08/30/2020] [Indexed: 02/07/2023] Open
Abstract
Resistance to cancer therapy is a major barrier to cancer management. Conventional views have proposed that acquisition of resistance may result from genetic mutations. However, accumulating evidence implicates a key role of non-mutational resistance mechanisms underlying drug tolerance, the latter of which is the focus that will be discussed here. Such non-mutational processes are largely driven by tumor cell plasticity, which renders tumor cells insusceptible to the drug-targeted pathway, thereby facilitating the tumor cell survival and growth. The concept of tumor cell plasticity highlights the significance of re-activation of developmental programs that are closely correlated with epithelial-mesenchymal transition, acquisition properties of cancer stem cells, and trans-differentiation potential during drug exposure. From observations in various cancers, this concept provides an opportunity for investigating the nature of anticancer drug resistance. Over the years, our understanding of the emerging role of phenotype switching in modifying therapeutic response has considerably increased. This expanded knowledge of tumor cell plasticity contributes to developing novel therapeutic strategies or combination therapy regimens using available anticancer drugs, which are likely to improve patient outcomes in clinical practice.
Collapse
Affiliation(s)
- Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, People's Republic of China
| | - Jingwen Jiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, People's Republic of China
| | - Yi Lu
- School of Medicine, Southern University of Science and Technology Shenzhen, Shenzhen, Guangdong, 518055, People's Republic of China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen, Guangdong, People's Republic of China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, People's Republic of China.
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Road, 611137, Chengdu, People's Republic of China.
| | - Jian Zhang
- School of Medicine, Southern University of Science and Technology Shenzhen, Shenzhen, Guangdong, 518055, People's Republic of China.
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen, Guangdong, People's Republic of China.
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China.
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, People's Republic of China.
| |
Collapse
|
14
|
Hofman P. New insights into the interaction of the immune system with non-small cell lung carcinomas. Transl Lung Cancer Res 2020; 9:2199-2213. [PMID: 33209644 PMCID: PMC7653157 DOI: 10.21037/tlcr-20-178] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The basis of current and future lung cancer immunotherapy depends mainly on our knowledge of the molecular mechanisms of interactions between cancer and immune cells (ICs), as well as on interactions occurring between the different populations of intra-tumor ICs. These interactions are very complex, as virtually all immune cell types, including macrophages, neutrophils, mast cells, natural killer (NK) cells, dendritic cells and T and B lymphocytes can infiltrate lung cancer tissues at the same time. Moreover these interactions lead to progressive emergence of an imbalance in ICs. Initially ICs have an anti-tumor effect but then induce immune tolerance and eventually tumor progression and dissemination. All the cells of innate and adaptive intra-tumor immunity engage in this progressive phenotypic switch. A majority of non-small cell lung carcinoma (NSCLC) patients do not benefit from the expected positive responses associated with current immunotherapy. Thus, there is urgent need to better understand the different roles of the associated cancer ICs. This review summarizes some of the new insights into this domain, with particular focus on: the myeloid cell population associated with tumors, the tertiary lymphoid structures (TLSs), the role of the P2 purinergic receptors (P2R) and ATP, and the new concept of the “liquid microenvironment” implying blood circulating ICs.
Collapse
Affiliation(s)
- Paul Hofman
- CHU Nice, FHU OncoAge, Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Université Côte d'Azur, Nice, France.,CNRS, INSERM, IRCAN, FHU OncoAge, Team 4, Hospital-Integrated Biobank, Université Côte d'Azur, Nice, France.,CHU Nice, FHU OncoAge, Hospital-Integrated Biobank, Université Côte d'Azur, Nice, France
| |
Collapse
|
15
|
Ancel J, Dewolf M, Deslée G, Nawrocky-Raby B, Dalstein V, Gilles C, Polette M. Clinical Impact of the Epithelial-Mesenchymal Transition in Lung Cancer as a Biomarker Assisting in Therapeutic Decisions. Cells Tissues Organs 2020; 211:91-109. [PMID: 32750701 DOI: 10.1159/000510103] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/11/2020] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is one of the most common solid cancers and represents the leading cause of cancer death worldwide. Over the last decade, research on the epithelial-mesenchymal transition (EMT) in lung cancer has gained increasing attention. Here, we review clinical and histological features of non-small-cell lung cancer associated with EMT. We then aimed to establish potential clinical implications of EMT in current therapeutic options, including surgery, radiation, targeted therapy against oncogenic drivers, and immunotherapy.
Collapse
Affiliation(s)
- Julien Ancel
- Inserm, Université de Reims Champagne Ardenne, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France.,Service de Pneumologie, Hôpital Maison Blanche, CHU de Reims, Reims, France
| | - Maxime Dewolf
- Service de Pneumologie, Hôpital Maison Blanche, CHU de Reims, Reims, France
| | - Gaëtan Deslée
- Inserm, Université de Reims Champagne Ardenne, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France.,Service de Pneumologie, Hôpital Maison Blanche, CHU de Reims, Reims, France
| | - Béatrice Nawrocky-Raby
- Inserm, Université de Reims Champagne Ardenne, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France
| | - Véronique Dalstein
- Inserm, Université de Reims Champagne Ardenne, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France.,Laboratoire de Pathologie, Hôpital Maison Blanche, CHU de Reims, Reims, France
| | - Christine Gilles
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Liège, Belgium,
| | - Myriam Polette
- Inserm, Université de Reims Champagne Ardenne, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France.,Laboratoire de Pathologie, Hôpital Maison Blanche, CHU de Reims, Reims, France
| |
Collapse
|
16
|
Yang X, Wu W, Pan Y, Zhou Q, Xu J, Han S. Immune-related genes in tumor-specific CD4 + and CD8 + T cells in colon cancer. BMC Cancer 2020; 20:585. [PMID: 32571262 PMCID: PMC7310260 DOI: 10.1186/s12885-020-07075-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/15/2020] [Indexed: 12/17/2022] Open
Abstract
Background Immune escape is an immunological mechanism underlying tumorigenesis, and T cells play an important role in this process. In this study, immune-related genes were evaluated in tumor-infiltrating CD4+ and CD8+ T cells in colon cancer. Methods ESTIMATE was used to calculate stromal and immune scores for tumor datasets downloaded from The Cancer Genome Atlas–Colon Cancer (COAD). Differentially expressed genes (DEGs) between samples with high and low stromal and immune scores were screened, followed by a functional enrichment analysis of the overlapping DEGs. The DEGs related to CD4+ and the CD8+ T cells were then screened. Predicted miRNA–mRNA and lncRNA–miRNA pairs were used to construct a competing endogenous RNA (ceRNA) network. Furthermore, chemical–gene interactions were predicted for genes in the ceRNA network. Kaplan–Meier survival curves were also plotted. Results In total, 83 stromal-related DEGs (5 up-regulated and 78 down-regulated) and 1270 immune-related DEGs (807 up-regulated and 293 down-regulated genes) were detected. The 79 overlapping DEGs were enriched for 39 biological process terms. Furthermore, 79 CD4+ T cell-related genes and 8 CD8+ T cell-related genes, such as ELK3, were screened. Additionally, ADAD1 and DLG3, related to CD4+ T cells, were significantly associated with the prognosis of patients with colon cancer. The chr22-38_28785274–29,006,793.1–miR-106a-5p-DDHD1 and chr22-38_28785274–29,006,793.1–miR-4319-GRHL1 axes obtained from CD4+ and CD8+ T cell-related ceRNAs were identified as candidates for further studies. Conclusion ELK3 is a candidate immune-related gene in colon cancer. The chr22-38_28785274–29,006,793.1–miR-106a-5p-DDHD1 and chr22-38_28785274–29,006,793.1–miR-4319-GRHL1 axes may be related to CD4+ and CD8+ T cell infiltration in colon cancer.
Collapse
Affiliation(s)
- Xi Yang
- Department of Oncology, Huzhou Cent Hosp, Affiliated Cent Hops HuZhou University, No. 198 Hongqi Road, Huzhou, 313000, Zhejiang Province, China
| | - Wei Wu
- Department of Gastroenterology, Huzhou Cent Hosp, Affiliated Cent Hops HuZhou University, No.198 Hongqi Road, Huzhou, 313000, Zhejiang Province, China
| | - Yuefen Pan
- Department of Oncology, Huzhou Cent Hosp, Affiliated Cent Hops HuZhou University, No. 198 Hongqi Road, Huzhou, 313000, Zhejiang Province, China
| | - Qing Zhou
- Department of Critical Care Medicine, Huzhou Cent Hosp, Affiliated Cent Hops HuZhou University, No. 198 Hongqi Road, Huzhou, 313000, Zhejiang Province, China
| | - Jiamin Xu
- Graduate School of Nursing, Huzhou University, No. 1 Bachelor Road, Huzhou, 313000, Zhejiang Province, China
| | - Shuwen Han
- Department of Oncology, Huzhou Cent Hosp, Affiliated Cent Hops HuZhou University, No. 198 Hongqi Road, Huzhou, 313000, Zhejiang Province, China.
| |
Collapse
|
17
|
Mao W, Yin H, Chen W, Zhao T, Wu S, Jin H, Du B, Tan Y, Zhang R, He Y. Network Pharmacology and Experimental Evidence Reveal Dioscin Suppresses Proliferation, Invasion, and EMT via AKT/GSK3b/mTOR Signaling in Lung Adenocarcinoma. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:2135-2147. [PMID: 32546976 PMCID: PMC7266311 DOI: 10.2147/dddt.s249651] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Purpose Dioscin, a natural glycoside derived from many plants, has been proved to exert anti-cancer activity. Several studies have found that it reverses TGF-β1-induced epithelial–mesenchymal transition (EMT). Whether dioscin can reverse EMT by pathways other than TGF-β is still unknown. Methods We used network-based pharmacological methods to systematically explore the potential mechanisms by which dioscin acts on lung cancer. Cell Counting Kit-8 assay, scratch healing, Transwell assay, Matrigel invasion assay, immunofluorescence assay, and Western blotting were employed to confirm the prediction of key targets and the effects of dioscin on EMT. Results Here, using network-based pharmacological methods, we found 42 possible lung cancer-related targets of dioscin, which were assigned to 98 KEGG pathways. Among the 20 with the lowest p-values, the PI3K-AKT signaling pathway is involved and significantly related to EMT. AKT1 and mTOR, with high degrees (reflecting higher connectivity) in the compound-target analysis, participate in the PI3K-AKT signaling pathway. Molecular docking indicated the occurrence of dioscin-AKT1 and dioscin-mTOR binding. Functional experiments demonstrated that dioscin suppressed the proliferation, migration, invasion, and EMT of human lung adenocarcinoma cells in a dose-dependent manner, without TGF-β stimulation. Furthermore, we determined that dioscin downregulated p-AKT, p-mTOR and p-GSK3β in human lung adenocarcinoma cells without affecting their total protein levels. The PI3K inhibitor LY294002 augmented these changes. Conclusion Dioscin suppressed proliferation, invasion and EMT of lung adenocarcinoma cells via the inactivation of AKT/mTOR/GSK3β signaling, probably by binding to AKT and mTOR, and inhibiting their phosphorylation.
Collapse
Affiliation(s)
- Wenli Mao
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China
| | - Heng Yin
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China
| | - Wenya Chen
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China
| | - Tingxiu Zhao
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China
| | - Shaofeng Wu
- Research Center for Integrative Medicine of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China
| | - He Jin
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China
| | - Biaoyan Du
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China
| | - Yuhui Tan
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China
| | - Ren Zhang
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China.,Research Center for Integrative Medicine of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China
| | - Yanli He
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China.,Research Center for Integrative Medicine of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China
| |
Collapse
|