1
|
Li T, Jiang S, Li T, Xu H, Zhang X, Yan R, Wu X, Jin Y, Wang Z. Exploring the Potential of Cyclic Peptidyl Antitumor Agents Derived from Natural Macrocyclic Peptide Phakellistatin 13. J Med Chem 2024; 67:11789-11813. [PMID: 38990190 DOI: 10.1021/acs.jmedchem.4c00393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
The exploration of novel anticancer compounds based on natural cyclopeptides has emerged as a pivotal paradigm in the contemporary advancement of macrocyclic pharmaceuticals. Phakellistatin 13 is a cycloheptapeptide derived from the brown snubby sponge and exhibits remarkable antitumor activity. In this study, we have designed and synthesized a series of chiral cyclopeptides incorporating the rigid isoindolinone moiety at various sites within the natural cycloheptapeptide Phakellistatin 13, with the aim of investigating conformationally constrained cyclopeptides as potential antitumor agents. Cyclopeptide 3, comprising alternating l-/d-amino acid residues, exhibited promising antihepatocellular carcinoma effects. Detailed biological experiments have revealed that Phakellistatin 13 analogs effectively inhibit the proliferation of tumor cells and induce apoptosis and autophagy, while also causing cell cycle arrest through the modulation of the p53 and mitogen-activated protein kinase (MAPK) signaling pathway. This study not only provides valuable insights into chemical structural modifications but also contributes to a deeper understanding of the biological mechanisms underlying the development of natural cyclopeptide-based drugs.
Collapse
Affiliation(s)
- Tong Li
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Shitian Jiang
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Tingting Li
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Hongyu Xu
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Xiong Zhang
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Rui Yan
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Xiaodan Wu
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Yingxue Jin
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Zhiqiang Wang
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin 150025, China
| |
Collapse
|
2
|
Hong R, Tong Y, Liu H, Chen P, Liu R. Edge-based relative entropy as a sensitive indicator of critical transitions in biological systems. J Transl Med 2024; 22:333. [PMID: 38576021 PMCID: PMC10996174 DOI: 10.1186/s12967-024-05145-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/29/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Disease progression in biosystems is not always a steady process but is occasionally abrupt. It is important but challenging to signal critical transitions in complex biosystems. METHODS In this study, based on the theoretical framework of dynamic network biomarkers (DNBs), we propose a model-free method, edge-based relative entropy (ERE), to identify temporal key biomolecular associations/networks that may serve as DNBs and detect early-warning signals of the drastic state transition during disease progression in complex biological systems. Specifically, by combining gene‒gene interaction (edge) information with the relative entropy, the ERE method converts gene expression values into network entropy values, quantifying the dynamic change in a biomolecular network and indicating the qualitative shift in the system state. RESULTS The proposed method was validated using simulated data and real biological datasets of complex diseases. The applications show that for certain diseases, the ERE method helps to reveal so-called "dark genes" that are non-differentially expressed but with high ERE values and of essential importance in both gene regulation and prognosis. CONCLUSIONS The proposed method effectively identified the critical transition states of complex diseases at the network level. Our study not only identified the critical transition states of various cancers but also provided two types of new prognostic biomarkers, positive and negative edge biomarkers, for further practical application. The method in this study therefore has great potential in personalized disease diagnosis.
Collapse
Affiliation(s)
- Renhao Hong
- School of Mathematics, South China University of Technology, Guangzhou, 510640, China
| | - Yuyan Tong
- School of Mathematics, South China University of Technology, Guangzhou, 510640, China
| | - Huisheng Liu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Pei Chen
- School of Mathematics, South China University of Technology, Guangzhou, 510640, China.
| | - Rui Liu
- School of Mathematics, South China University of Technology, Guangzhou, 510640, China.
| |
Collapse
|
3
|
Manandhar B, Paudel KR, Clarence DD, De Rubis G, Madheswaran T, Panneerselvam J, Zacconi FC, Williams KA, Pont LG, Warkiani ME, MacLoughlin R, Oliver BG, Gupta G, Singh SK, Chellappan DK, Hansbro PM, Dua K. Zerumbone-incorporated liquid crystalline nanoparticles inhibit proliferation and migration of non-small-cell lung cancer in vitro. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:343-356. [PMID: 37439806 PMCID: PMC10771618 DOI: 10.1007/s00210-023-02603-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 06/25/2023] [Indexed: 07/14/2023]
Abstract
Lung cancer is the second most prevalent type of cancer and is responsible for the highest number of cancer-related deaths worldwide. Non-small-cell lung cancer (NSCLC) makes up the majority of lung cancer cases. Zerumbone (ZER) is natural compound commonly found in the roots of Zingiber zerumbet which has recently demonstrated anti-cancer activity in both in vitro and in vivo studies. Despite their medical benefits, ZER has low aqueous solubility, poor GI absorption and oral bioavailability that hinders its effectiveness. Liquid crystalline nanoparticles (LCNs) are novel drug delivery carrier that have tuneable characteristics to enhance and ease the delivery of bioactive compounds. This study aimed to formulate ZER-loaded LCNs and investigate their effectiveness against NSCLC in vitro using A549 lung cancer cells. ZER-LCNs, prepared in the study, inhibited the proliferation and migration of A549 cells. These inhibitory effects were superior to the effects of ZER alone at a concentration 10 times lower than that of free ZER, demonstrating a potent anti-cancer activity of ZER-LCNs. The underlying mechanisms of the anti-cancer effects by ZER-LCNs were associated with the transcriptional regulation of tumor suppressor genes P53 and PTEN, and metastasis-associated gene KRT18. The protein array data showed downregulation of several proliferation associated proteins such as AXL, HER1, PGRN, and BIRC5 and metastasis-associated proteins such as DKK1, CAPG, CTSS, CTSB, CTSD, and PLAU. This study provides evidence of potential for increasing the potency and effectiveness of ZER with LCN formulation and developing ZER-LCNs as a treatment strategy for mitigation and treatment of NSCLC.
Collapse
Affiliation(s)
- Bikash Manandhar
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2050, Australia
| | - Dvya Delilaa Clarence
- School of Postgraduate Studies, International Medical University (IMU), 57000, Kuala Lumpur, Malaysia
| | - Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Thiagarajan Madheswaran
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Jithendra Panneerselvam
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Flavia C Zacconi
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, 7820436, Macul, Santiago, Chile
- Centro de Investigación en Nanotecnología y Materiales Avanzados, CIEN-UC, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, 7820436, Macul, Santiago, Chile
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Kylie A Williams
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Lisa G Pont
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales, Australia
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Ronan MacLoughlin
- Research and Development, Aerogen Limited, IDA Business Park, Galway, Connacht, H91 HE94, Ireland
- School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Leinster, D02 YN77, Ireland
- School of Pharmacy & Pharmaceutical Sciences, Trinity College, Dublin, Leinster, D02 PN40, Ireland
| | - Brian Gregory Oliver
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, 2137, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jaipur, Rajasthan, India
- Center for Transdisciplinary Research, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, 144411, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, 57000, Kuala Lumpur, Malaysia.
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2050, Australia.
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia.
- Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia.
| |
Collapse
|
4
|
Paudel KR, Rajput R, De Rubis G, Raju Allam VSR, Williams KA, Singh SK, Gupta G, Salunke P, Hansbro PM, Gerlach J, Dua K. In vitro anti-cancer activity of a polyherbal preparation, VEDICINALS®9, against A549 human lung adenocarcinoma cells. Pathol Res Pract 2023; 250:154832. [PMID: 37774532 DOI: 10.1016/j.prp.2023.154832] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/20/2023] [Accepted: 09/23/2023] [Indexed: 10/01/2023]
Abstract
PURPOSE Non-small cell lung cancer (NSCLC) is among the leading causes of morbidity and mortality worldwide. Despite the availability of several treatment options, the five-year survival rate of NSCLC is extremely low (<20%). This underlines the necessity of more effective therapeutic alternatives. In this context, plant-derived extracts and bioactive molecules extracted from plants, known collectively as phytoceuticals, represent an extremely variegated source of bioactive compounds with potent anticancer potential. In the present study, we tested the in vitro anticancer activity of a polyherbal preparation, VEDICINALS®9, containing nine different bioactive principles extracted by medicinal plants. METHODS The anticancer activity of VEDICINALS®9 was investigated by measuring its impact on A549 human NSCLC cell proliferation (MTT assay and trypan blue staining), migration (wound healing assay and transwell chamber assay) and by measuring the impact on the expression of cancer-related proteins (Human XL Oncology Protein Array). RESULTS We show that VEDICINALS®9 at a concentration of 0.2% v/v has potent anticancer effect, significantly inhibiting A549 cell proliferation and migration. Mechanistically, this was achieved by downregulating the expression of proteins involved in cancer cell proliferation (Axl, FGF basic, enolase 2, progranulin, survivin) and migration (Dkk-1, cathepsins B and D, BCL-x, amphiregulin, CapG, u-plasminogen activator). Furthermore, treatment with VEDICINALS®9 resulted in increased expression of the oncosuppressor protein p53 and of the angiogenesis inhibitor endostatin. CONCLUSIONS Taken together, our results provide proof of principle of the potent anticancer activity of the polyherbal preparation VEDICINALS®9, highlighting its enormous potential as an alternative or adjuvant therapy for lung cancer.
Collapse
Affiliation(s)
- Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia
| | - Rashi Rajput
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Venkata Sita Rama Raju Allam
- Department of Medical Biochemistry and Microbiology, Biomedical Centre (BMC), Uppsala University, Uppsala, Sweden
| | - Kylie Anne Williams
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Sachin Kumar Singh
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jaipur, Rajasthan, India; Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India
| | | | - Philip Michael Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia
| | | | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia.
| |
Collapse
|
5
|
De Rubis G, Paudel KR, Liu G, Agarwal V, MacLoughlin R, de Jesus Andreoli Pinto T, Singh SK, Adams J, Nammi S, Chellappan DK, Oliver BGG, Hansbro PM, Dua K. Berberine-loaded engineered nanoparticles attenuate TGF-β-induced remodelling in human bronchial epithelial cells. Toxicol In Vitro 2023; 92:105660. [PMID: 37591407 DOI: 10.1016/j.tiv.2023.105660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/05/2023] [Accepted: 08/14/2023] [Indexed: 08/19/2023]
Abstract
Airway remodelling occurs in chronic respiratory diseases (CRDs) such as asthma and chronic obstructive pulmonary disease (COPD). It is characterized by aberrant activation of epithelial reparation, excessive extracellular matrix (ECM) deposition, epithelial-to-mesenchymal transition (EMT), and airway obstruction. The master regulator is Transforming Growth Factor-β (TGF-β), which activates tissue repair, release of growth factors, EMT, increased cell proliferation, and reduced nitric oxide (NO) secretion. Due to its fundamental role in remodelling, TGF-β is an emerging target in the treatment of CRDs. Berberine is a benzylisoquinoline alkaloid with antioxidant, anti-inflammatory, and anti-fibrotic activities whose clinical application is hampered by poor permeability. To overcome these limitations, in this study, berberine was encapsulated in monoolein-based liquid crystalline nanoparticles (BM-LCNs). The potential of BM-LCNs in inhibiting TGF-β-induced remodelling features in human bronchial epithelial cells (BEAS-2B) was tested. BM-LCNs significantly inhibited TGF-β-induced migration, reducing the levels of proteins upregulated by TGF-β including endoglin, thrombospondin-1, basic fibroblast growth factor, vascular-endothelial growth factor, and myeloperoxidase, and increasing the levels of cystatin C, a protein whose expression was downregulated by TGF-β. Furthermore, BM-LCNs restored baseline NO levels downregulated by TGF-β. The results prove the in vitro therapeutic efficacy of BM-LCNs in counteracting TGF-β-induced remodelling features. This study supports the suitability of berberine-loaded drug delivery systems to counteract airway remodelling, with potential application as a treatment strategy against CRDs.
Collapse
Affiliation(s)
- Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia
| | - Gang Liu
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia
| | - Vipul Agarwal
- Cluster for Advanced Macromolecular Design (CAMD), School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Ronan MacLoughlin
- Aerogen, IDA Business Park, H91 HE94 Galway, Connacht, Ireland; School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Leinster, Ireland; School of Pharmacy & Pharmaceutical Sciences, Trinity College, D02 PN40 Dublin, Leinster, Ireland
| | | | - Sachin Kumar Singh
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Jon Adams
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Srinivas Nammi
- School of Science, Western Sydney University, Penrith, NSW 2751, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Brian Gregory George Oliver
- Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia; School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Philip Michael Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India.
| |
Collapse
|
6
|
Zhang Q, Zhao M, Lin S, Han Q, Ye H, Peng F, Li L. Prediction of prognosis and immunotherapy response in lung adenocarcinoma based on CD79A, DKK1 and VEGFC. Heliyon 2023; 9:e18503. [PMID: 37534013 PMCID: PMC10392102 DOI: 10.1016/j.heliyon.2023.e18503] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 07/14/2023] [Accepted: 07/19/2023] [Indexed: 08/04/2023] Open
Abstract
Background Tumor immune microenvironment (TIME) is crucial for tumor initiation, progression, and metastasis; however, its relationship with lung adenocarcinoma (LUAD) is unknown. Traditional predictive models screen for biomarkers that are too general and infrequently associated with immune genes. Methods RNA sequencing data of LUAD patients and immune-related gene sets were retrieved from public databases. Using the common genes shared by The Cancer Genome Atlas (TCGA) and Immunology Database and Analysis Portal (ImmPort), differential gene expression analysis, survival analysis, Lasso regression analysis, and univariate and multivariate Cox regression analyses were performed to generate a novel risk score model. LUAD cohort in International Cancer Genome Consortium (ICGC), GSE68465 cohort in Gene Expression Omnibus (GEO) and an immunohistochemical assay were used to validate the key genes constructed risk score. The LUAD-related prognosis, clinical indicators, immune infiltrate characteristics, response to immunotherapy, and response to chemotherapeutic agents in different risk groups were evaluated by CIBERSORT, ImmuCellAI, pRRophetic and other tools. Results The risk score model was constructed using CD79a molecule (CD79A), Dickkopf WNT signaling pathway inhibitor 1 (DKK1), and vascular endothelial growth factor C (VEGFC). High risk score was identified as a negative predictor for overall survival (OS) in subgroup analyses with tumor stage, TNM classification, therapy outcome, and ESTIMATE scores (P < 0.05). Low risk score was positively associated with plasma cells, memory B cells, CD8 T cells, regulatory T cells and γδT cells (P < 0.05). In low-risk group, programmed cell death 1 receptor (PD1), cytotoxic T-lymphocyte associated protein 4 (CTLA4), and lymphocyte activating 3 (LAG3) and indoleamine 2,3-dioxygenase (IDO) were more robustly expressed (P < 0.05). The treatment responses of immune checkpoint blockade (ICB) therapy and chemotherapy were likewise superior in low-risk group (P < 0.05). In immunohistochemical analysis, the tumor group had significantly higher levels of CD79A, DKK1, and VEGFC than the adjacent normal group (P < 0.01). Conclusions CD79A, DKK1 and VEGFC are important differential genes related to LUAD, risk score could reliably predict prognosis, composition of TIME and immunotherapy responses in LUAD patients. The excellent performance of the risk model shows its strong and broad application potential.
Collapse
Affiliation(s)
- Qilong Zhang
- Department of Pharmacy, Zhejiang Hospital, Hangzhou, Zhejiang 310007, China
| | - Mingyuan Zhao
- Department of Pathology, Zhejiang Hospital, Hangzhou, Zhejiang 310007, China
| | - Shuangyan Lin
- Department of Pathology, Zhejiang Hospital, Hangzhou, Zhejiang 310007, China
| | - Qi Han
- Department of Pharmacy, Zhejiang Hospital, Hangzhou, Zhejiang 310007, China
| | - He Ye
- Department of Pharmacy, Zhejiang Hospital, Hangzhou, Zhejiang 310007, China
| | - Fang Peng
- Department of Pathology, Zhejiang Hospital, Hangzhou, Zhejiang 310007, China
| | - Li Li
- Department of Pharmacy, Zhejiang Hospital, Hangzhou, Zhejiang 310007, China
| |
Collapse
|
7
|
He X, Su Y, Liu P, Chen C, Chen C, Guan H, Lv X, Guo W. Machine learning-based immune prognostic model and ceRNA network construction for lung adenocarcinoma. J Cancer Res Clin Oncol 2023; 149:7379-7392. [PMID: 36939925 DOI: 10.1007/s00432-023-04609-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/27/2023] [Indexed: 03/21/2023]
Abstract
PURPOSE Lung adenocarcinoma (LUAD) is a malignant tumor with a high lethality rate. Immunotherapy has become a breakthrough in cancer treatment and improves patient survival and prognosis. Therefore, it is necessary to find new immune-related markers. However, the current research on immune-related markers in LUAD is not sufficient. Therefore, there is a need to find new immune-related biomarkers to help treat LUAD patients. METHODS In this study, a bioinformatics approach combined with a machine learning approach screened reliable immune-related markers to construct a prognostic model to predict the overall survival (OS) of LUAD patients, thus promoting the clinical application of immunotherapy in LUAD. The experimental data were obtained from The Cancer Genome Atlas (TCGA) database, including 535 LUAD and 59 healthy control samples. Firstly, the Hub gene was screened using a bioinformatics approach combined with the Support Vector Machine Recursive Feature Elimination algorithm; then, a multifactorial Cox regression analysis by constructing an immune prognostic model for LUAD and a nomogram to predict the OS rate of LUAD patients. Finally, the regulatory mechanism of Hub genes in LUAD was analyzed by ceRNA. RESULTS Five genes, ADM2, CDH17, DKK1, PTX3, and AC145343.1, were screened as potential immune-related genes in LUAD. Among them, ADM2 and AC145343.1 had a good prognosis in LUAD patients (HR < 1) and were novel markers. The remaining three genes screened were associated with poor prognosis in LUAD patients (HR > 1). In addition, the experimental results showed that patients in the low-risk group had better OS rates than those in the high-risk group (P < 0.001). CONCLUSION In this paper, we propose an immune prognostic model to predict OS rate in LUAD patients and show the correlation between five immune genes and the level of immune-related cell infiltration. It provides new markers and additional ideas for immunotherapy in patients with LUAD.
Collapse
Affiliation(s)
- Xiaoqian He
- College of Information Science and Engineering, Xinjiang University, Urumqi, 830046, China
| | - Ying Su
- College of Information Science and Engineering, Xinjiang University, Urumqi, 830046, China
| | - Pei Liu
- College of Information Science and Engineering, Xinjiang University, Urumqi, 830046, China
| | - Cheng Chen
- College of Software, Xinjiang University, Urumqi, 830046, China.
| | - Chen Chen
- College of Information Science and Engineering, Xinjiang University, Urumqi, 830046, China
| | - Haoqin Guan
- College of Information Science and Engineering, Xinjiang University, Urumqi, 830046, China
| | - Xiaoyi Lv
- College of Software, Xinjiang University, Urumqi, 830046, China.
| | - Wenjia Guo
- Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830011, China.
| |
Collapse
|
8
|
Li X, Zheng T, Zhang Y, Zhao Y, Liu F, Dai S, Liu X, Zhang M. Dickkopf-1 promotes vascular smooth muscle cell foam cell formation and atherosclerosis development through CYP4A11/SREBP2/ABCA1. FASEB J 2023; 37:e23048. [PMID: 37389895 DOI: 10.1096/fj.202300295r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 05/20/2023] [Accepted: 06/08/2023] [Indexed: 07/01/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are considered to be a crucial source of foam cells in atherosclerosis due to their low expression level of cholesterol exporter ATP-binding cassette transporter A1 (ABCA1) intrinsically. While the definite regulatory mechanisms are complicated and have not yet been fully elucidated, we previously reported that Dickkopf-1 (DKK1) mediates endothelial cell (EC) dysfunction, thereby aggravating atherosclerosis. However, the role of smooth muscle cell (SMC) DKK1 in atherosclerosis and foam cell formation remains unknown. In this study, we established SMC-specific DKK1-knockout (DKK1SMKO ) mice by crossbreeding DKK1flox/flox mice with TAGLN-Cre mice. Then, DKK1SMKO mice were crossed with APOE-/- mice to generate DKK1SMKO /APOE-/- mice, which exhibited milder atherosclerotic burden and fewer SMC foam cells. In vitro loss- and gain-of-function studies of DKK1 in primary human aortic smooth muscle cells (HASMCs) have proven that DKK1 prevented oxidized lipid-induced ABCA1 upregulation and cholesterol efflux and promoted SMC foam cell formation. Mechanistically, RNA-sequencing (RNA-seq) analysis of HASMCs as well as chromatin immunoprecipitation (ChIP) experiments showed that DKK1 mediates the binding of transcription factor CCAAT/enhancer-binding protein delta (C/EBPδ) to the promoter of cytochrome P450 epoxygenase 4A11 (CYP4A11) to regulate its expression. In addition, CYP4A11 as well as its metabolite 20-HETE-promoted activation of transcription factor sterol regulatory element-binding protein 2 (SREBP2) mediated the DKK1 regulation of ABCA1 in SMC. Furthermore, HET0016, the antagonist of CYP4A11, has also shown an alleviating effect on atherosclerosis. In conclusion, our results demonstrate that DKK1 promotes SMC foam cell formation during atherosclerosis via a reduction in CYP4A11-20-HETE/SREBP2-mediated ABCA1 expression.
Collapse
Affiliation(s)
- Xiao Li
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Tengfei Zheng
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yu Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yachao Zhao
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Fengming Liu
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Shen Dai
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xiaolin Liu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Mei Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
9
|
Yang RH, Qin J, Cao JL, Zhang MZ, Li YY, Wang MQ, Fang D, Xie SQ. Dickkopf-1 drives tumor immune evasion by inducing PD-L1 expression in hepatocellular carcinoma. Biochem Pharmacol 2023; 208:115378. [PMID: 36513141 DOI: 10.1016/j.bcp.2022.115378] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Understanding the mechanisms regulating PD-L1 expression in hepatocellular carcinoma (HCC) is important to improve the response rate to PD-1/PD-L1 blockade therapy. Here, we show that DKK1 expression is positively associated with PD-L1 expression and inversely correlated with CD8+ T cell infiltration in human HCC tumor specimens. In a subcutaneous xenograft tumor model, overexpression of DKK1 significantly promotes tumor growth, tumoral PD-L1 expression, but reduces tumoral CD8+ T cell infiltration; whereas knockdown of DKK1 has opposite effects. Moreover, enforced expression of DKK1 dramatically promotes PD-L1 expression, Akt activation, β-catenin phosphorylation and total protein expression in HCC cells. By contrast, knockdown of DKK1 inhibits all, relative to controls. In addition, CKAP4 depletion, Akt inhibition, or β-catenin depletion remarkably abrogates DKK1 overexpression-induced transcriptional expression of PD-L1 in HCC cells. Reconstituted expression of the active Akt1 largely increased PD-L1 transcriptional expression in HCC cells. Similarly, expression of WT β-catenin, but not the phosphorylation-defective β-catenin S552A mutant, significantly promotes PD-L1 expression. Correlation analysis of human HCC tumor specimens further revealed that DKK1 and PD-L1 expression were positively correlated with p-β-catenin expression. Together, our findings revealed that DKK1 promotes PD-L1 expression through the activation of Akt/β-catenin signaling, providing a potential strategy to enhance the clinical efficacy of PD-1/PD-L1 blockade therapy in HCC patients.
Collapse
Affiliation(s)
- Ruo-Han Yang
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng 475004, China; The Academy for Advanced Interdisplinary Studies, Henan University, N. Jinming Ave, Kaifeng 475004, China
| | - Jia Qin
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng 475004, China
| | - Jin-Lan Cao
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng 475004, China
| | - Ming-Zhu Zhang
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng 475004, China
| | - Ying-Ying Li
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng 475004, China
| | - Meng-Qing Wang
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng 475004, China
| | - Dong Fang
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng 475004, China; The Academy for Advanced Interdisplinary Studies, Henan University, N. Jinming Ave, Kaifeng 475004, China; Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, Kaifeng 475004, China.
| | - Song-Qiang Xie
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng 475004, China; The Academy for Advanced Interdisplinary Studies, Henan University, N. Jinming Ave, Kaifeng 475004, China.
| |
Collapse
|
10
|
Jiang Z, Wang X, Huang J, Li G, Li S. Pyroptosis-based risk score predicts prognosis and drug sensitivity in lung adenocarcinoma. Open Med (Wars) 2023; 18:20230663. [PMID: 36941988 PMCID: PMC10024350 DOI: 10.1515/med-2023-0663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/06/2023] [Accepted: 01/19/2023] [Indexed: 03/14/2023] Open
Abstract
Pyroptosis is a recently identified form of programmed cell death; however, its role in lung adenocarcinoma (LUAD) remains unclear. Therefore, we set out to explore the prognostic potential of pyroptosis-related genes in LUAD. The pyroptosis-related risk score (PRRS) was developed by least absolute shrinkage and selection operator Cox regression and multivariate Cox regression. We found that PRRS was an independent prognostic factor for LUAD. LUAD patients in the high-PRRS group showed a significantly shorter overall survival (OS) and enriched in cell proliferation-related pathways. Then pathway enrichment analyses, mutation profile, tumor microenvironment, and drug sensitivity analysis were further studied in PRRS stratified LUAD patients. Tumor purity (TP) analyses revealed that L-PRRS LUAD patients had a lower TP, and patients in L-TP + L-PRRS subgroup had the most prolonged OS. Mutation analyses suggested that the L-PRRS LUAD patients had a lower tumor mutation burden (TMB), and patients in H-TMB + L-PRRS subgroup had the most prolonged OS. Drug sensitivity analyses showed that PRRS was significantly negatively correlated with the sensitivity of cisplatin, besarotene, etc., while it was significantly positively correlated with the sensitivity of kin001-135. Eventually, a nomogram was constructed based on PRRS and clinical characters of LUAD. Overall, the pyroptosis-related signature is helpful for prognostic prediction and in guiding treatment for LUAD patients.
Collapse
Affiliation(s)
- Zhengsong Jiang
- Department of Laboratory Medicine, The First Hospital of Jiujiang, Jiujiang, Jiangxi, China
| | - Xiang Wang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | | | - Guoyin Li
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi’an, 710061, China
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, China
| | - Shangfu Li
- Department of Oncology, Yueyang Second People’s Hospital, Yueyang, Hunan, 414022, China
| |
Collapse
|
11
|
Cai S, Hu T, Venkatesan M, Allam M, Schneider F, Ramalingam SS, Sun SY, Coskun AF. Multiplexed protein profiling reveals spatial subcellular signaling networks. iScience 2022; 25:104980. [PMID: 36093051 PMCID: PMC9460555 DOI: 10.1016/j.isci.2022.104980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 05/25/2022] [Accepted: 08/16/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Shuangyi Cai
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Thomas Hu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Mythreye Venkatesan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Mayar Allam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Frank Schneider
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, GA 30322, USA
- Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Suresh S. Ramalingam
- Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shi-Yong Sun
- Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ahmet F. Coskun
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Corresponding author
| |
Collapse
|
12
|
Tang X, Qi C, Zhou H, Liu Y. A novel metabolic-immune related signature predicts prognosis and immunotherapy response in lung adenocarcinoma. Heliyon 2022; 8:e10164. [PMID: 36016532 PMCID: PMC9396642 DOI: 10.1016/j.heliyon.2022.e10164] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/19/2022] [Accepted: 07/29/2022] [Indexed: 11/24/2022] Open
Abstract
Background Lung adenocarcinoma (LUAD) is one of the most frequent types of lung cancer, with a high mortality and recurrence rate. This study aimed to design a RiskScore to predict the prognosis and immunotherapy response of LUAD patients due to a lack of metabolic and immune-related prognostic models. Methods To identify prognostic genes and generate a RiskScore, we conducted differential gene expression analysis, bulk survival analysis, Lasso regression analysis, and univariate and multivariate Cox regression analysis using TCGA-LUAD as a training subset. GSE31210 and GSE50081 were used as validation subsets to validate the constructed RiskScore. Following that, we explored the connection between RiskScore and clinicopathological characteristics, immune cells infiltration, and immunotherapy. In addition, we investigated into RiskScore's biological roles and constructed a Nomogram model. Results A RiskScore was identified consisting of five genes (DKK1, CCL20, NPAS2, GNPNAT1 and MELTF). In the RiskScore-high group, LUAD patients showed decreased overall survival rates and shorter progression-free survival. Multiple clinicopathological characteristics and immune cells infiltration in TME, in particular, have been linked to RiskScore. Of note, RiskScore-related genes have been implicated to substance metabolism, carcinogenesis, and immunological pathways, among other things. Finally, the C-index of the RiskScore-based Nomogram model was 0.804 (95% CI: 0.783-0.825), and time-dependent ROC predicted probabilities of 1-, 3- and 5-year survival for LUAD patients were 0.850, 0.848 and 0.825, respectively. Conclusion The RiskScore, which integrated metabolic and immunological features with DKK1, CCL20, NPAS2, GNPNAT1, and MELTF, could reliably predict prognosis and immunotherapy response in LUAD patients. Moreover, the RiskScore-based Nomogram model had a promising clinical application.
Collapse
Affiliation(s)
- Xiaolong Tang
- Department of Clinical Laboratory Diagnostics, Binzhou Medical University, Binzhou, Shandong, 256603, China
| | - Chumei Qi
- Department of Clinical Laboratory, Dazhou Women and Children's Hospital, Dazhou, Sichuan, 635000, China
| | - Honghong Zhou
- Key Laboratory of RNA Biology, Center for Big Data Research in Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yongshuo Liu
- Department of Clinical Laboratory, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China.,Biomedical Pioneering Innovation Center (BIOPIC), Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| |
Collapse
|
13
|
Evaluation of the Cytotoxic Activity and Anti-Migratory Effect of Berberine–Phytantriol Liquid Crystalline Nanoparticle Formulation on Non-Small-Cell Lung Cancer In Vitro. Pharmaceutics 2022; 14:pharmaceutics14061119. [PMID: 35745691 PMCID: PMC9228615 DOI: 10.3390/pharmaceutics14061119] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 02/04/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) is the most common form of lung cancer, which is a leading cause of cancer-related deaths worldwide. Berberine is an isoquinoline alkaloid that is commercially available for use as a supplement for the treatment of diabetes and cardiovascular diseases. However, the therapeutic benefits of berberine are limited by its extremely low bioavailability and toxicity at higher doses. Increasing evidence suggests that the incorporation of drug compounds in liquid crystal nanoparticles provides a new platform for the safe, effective, stable, and controlled delivery of the drug molecules. This study aimed to formulate an optimized formulation of berberine–phytantriol-loaded liquid crystalline nanoparticles (BP-LCNs) and to investigate the in vitro anti-cancer activity in a human lung adenocarcinoma A549 cell line. The BP-LCN formulation possessing optimal characteristics that was used in this study had a favorable particle size and entrapment efficiency rate (75.31%) and a superior drug release profile. The potential mechanism of action of the formulation was determined by measuring the mRNA levels of the tumor-associated genes PTEN, P53, and KRT18 and the protein expression levels with a human oncology protein array. BP-LCNs decreased the proliferation, migration, and colony-forming activity of A549 cells in a dose-dependent manner by upregulating the mRNA expression of PTEN and P53 and downregulating the mRNA expression of KRT18. Similarly, BP-LCNs also decreased the expression of proteins related to cancer cell proliferation and migration. This study highlights the utility of phytantriol-based LCNs in incorporating drug molecules with low GI absorption and bioavailability to increase their pharmacological effectiveness and potency in NSCLC.
Collapse
|
14
|
Seo HY, Kim SC, Roh WL, Shin YK, Kim S, Kim DW, Kim TM, Ku JL. Culture and multiomic analysis of lung cancer patient-derived pleural effusions revealed distinct druggable molecular types. Sci Rep 2022; 12:6345. [PMID: 35428753 PMCID: PMC9012760 DOI: 10.1038/s41598-022-10318-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/04/2022] [Indexed: 12/12/2022] Open
Abstract
Malignant pleural effusion (MPE) is an independent determinant of poor prognostic factor of non-small cell lung cancer (NSCLC). The course of anchorage independent growth within the pleural cavity likely reforms the innate molecular characteristics of malignant cells, which largely accounts for resistance to chemotherapy and poor prognosis after the surgical resection. Nevertheless, the genetic and transcriptomic features with respect to various drug responses of MPE-complicated NSCLC remain poorly understood. To obtain a clearer overview of the MPE-complicated NSCLC, we established 28 MPE-derived lung cancer cell lines which were subjected to genomic, transcriptomic and pharmacological analysis. Our results demonstrated MPE-derived NSCLC cell lines recapitulated representative driver mutations generally found in the primary NSCLC. It also exhibited the presence of distinct translational subtypes in accordance with the mutational profiles. The drug responses of several targeted chemotherapies accords with both genomic and transcriptomic characteristics of MPE-derived NSCLC cell lines. Our data also suggest that the impending drawback of mutation-based clinical diagnosis in evaluating MPE-complicated NSCLS patient responses. As a potential solution, our work showed the importance of comprehending transcriptomic characteristics in order to defy potential drug resistance caused by MPE.
Collapse
Affiliation(s)
- Ha-Young Seo
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 103, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.,Cancer Research Institute, Seoul National University, Seoul, 03080, Korea
| | - Soon-Chan Kim
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 103, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.,Cancer Research Institute, Seoul National University, Seoul, 03080, Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Woo-Lee Roh
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 103, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Young-Kyoung Shin
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 103, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Soyeon Kim
- Cancer Research Institute, Seoul National University, Seoul, 03080, Korea
| | - Dong-Wan Kim
- Cancer Research Institute, Seoul National University, Seoul, 03080, Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, 03080, Korea
| | - Tae Min Kim
- Cancer Research Institute, Seoul National University, Seoul, 03080, Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, 03080, Korea
| | - Ja-Lok Ku
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 103, Daehak-ro, Jongno-gu, Seoul, 03080, Korea. .,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea. .,Cancer Research Institute, Seoul National University, Seoul, 03080, Korea. .,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, South Korea.
| |
Collapse
|
15
|
A Novel Immune-Related Gene Signature Predicts Prognosis of Lung Adenocarcinoma. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4995874. [PMID: 35437508 PMCID: PMC9013292 DOI: 10.1155/2022/4995874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/12/2021] [Accepted: 02/27/2022] [Indexed: 12/25/2022]
Abstract
Background Lung adenocarcinoma (LUAD) is the most common form of lung cancer, accounting for 30% of all cases and 40% of all non-small-cell lung cancer cases. Immune-related genes play a significant role in predicting the overall survival and monitoring the status of the cancer immune microenvironment. The present study was aimed at finding an immune-related gene signature for predicting LUAD patient outcomes. Methods First, we chose the TCGA-LUAD project in the TCGA database as the training cohort for model training. For model validating, we found the datasets of GSE72094 and GSE68465 in the GEO database and took them as the candidate cohorts. We obtained 1793 immune-related genes from the ImmPort database and put them into a univariate Cox proportional hazard model to initially look for the genes with potential prognostic ability using the data of the training cohort. These identified genes then entered into a random survival forests-variable hunting algorithm for the best combination of genes for prognosis. In addition, the LASSO Cox regression model tested whether the gene combination can be further shrinkage, thereby constructing a gene signature. The Kaplan-Meier, Cox model, and ROC curve were deployed to examine the gene signature's prognosis in both cohorts. We conducted GSEA analysis to study further the mechanisms and pathways that involved the gene signature. Finally, we performed integrating analyses about the 22 TICs, fully interpreted the relationship between our signature and each TIC, and highlighted some TICs playing vital roles in the signature's prognostic ability. Results A nine-gene signature was produced from the data of the training cohort. The Kaplan-Meier estimator, Cox proportional hazard model, and ROC curve confirmed the independence and predictive ability of the signature, using the data from the validation cohort. The GSEA analysis results illustrated the gene signature's mechanism and emphasized the importance of immune-related pathways for the gene signature. 22 TICs immune infiltration analysis revealed resting mast cells' key roles in contributing to gene signature's prognostic ability. Conclusions This study discovered a novel immune-related nine-gene signature (BTK, CCR6, S100A10, SEMA3C, GPI, SCG2, TNFRSF11A, CCL20, and DKK1) that predicts LUAD prognosis precisely and associates with resting mast cells strongly.
Collapse
|
16
|
Zeybek A, Öz N, Kalemci S, Tosun K, Edgünlü TG, Kızıltuğ MT, Tekin L, Erdal ME. The role of Wnt pathway antagonists in early-stage lung adenocarcinoma. Mol Biol Rep 2021; 49:9-17. [PMID: 34779987 DOI: 10.1007/s11033-021-06759-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 09/20/2021] [Indexed: 01/08/2023]
Abstract
PURPOSE We aimed to examine the expression levels of the genes encoding adenomatous polyposis coli (APC) 1, APC-2, Dickkopf related protein (DKK)-1, DKK-3, secreted frizzled-related protein (SFRP)-2, SFRP-4, and SFRP-5, which play roles in the Wnt signaling pathway, in lung adenocarcinoma and adjacent normal lung tissues and to evaluate their relationships with clinicopathologic factors. MATERIALS AND METHODS The expression levels of genes in formalin-fixed paraffin-embedded samples of tumor tissue and adjacent intact lung tissue from 57 patients who underwent surgery for lung adenocarcinoma between 2011 and 2018 were determined by real-time PCR analysis. RESULTS The expression levels of the DKK-1 in tumor tissue, especially in stage I-II tumor tissue, were significantly suppressed compared to those in normal tissue (p < 0.025). Whereas DKK-1 expression was suppressed in the tumor tissue of patients with early-stage lung adenocarcinoma, expression of the SFRP-5 in these patients was significantly higher in tumor tissue than in normal tissue (p < 0.039). CONCLUSION In our study, opposing regulation was found between the SFRP-5 and DKK-1, which are known to be extracellular antagonists of the Wnt signaling pathway. The SFRP-5 was found to have an oncogenic role in adenocarcinoma development. Studies of the opposing regulation between these genes in early-stage lung adenocarcinoma may shed light on the mechanisms associated with the development of carcinogenesis. The relationships or interactions of these genes may serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Arife Zeybek
- Department of Thoracic Surgery, Medical Faculty, School of Medicine, Mugla Sıtkı Kocman University, Mugla, Turkey.
| | - Necdet Öz
- Department of Thoracic Surgery, Private Antalya Med-Star Hospital, Antalya, Turkey
| | - Serdar Kalemci
- Department of Chest Disease, Kocaeli Medikal Park Hospital, Kocaeli, Turkey
| | | | - Tuba Gökdoğan Edgünlü
- Department of Medical Biology, Medical Faculty, Mugla Sıtkı Kocman University, Mugla, Turkey
| | | | - Leyla Tekin
- Department of Medical Pathology, Medical Faculty, Mugla Sıtkı Kocman University, Mugla, Turkey
| | - Mehmet Emin Erdal
- Department of Medical Biology, Mersin University Medical Faculty, Mersin, Turkey
| |
Collapse
|
17
|
Ding G, Lu W, Zhang Q, Li K, Zhou H, Wang F, Zhao C, Fan C, Wang J. ZBTB38 suppresses prostate cancer cell proliferation and migration via directly promoting DKK1 expression. Cell Death Dis 2021; 12:998. [PMID: 34697293 PMCID: PMC8546125 DOI: 10.1038/s41419-021-04278-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 12/24/2022]
Abstract
Prostate cancer is still one of the most common malignancies in men all around the world. The mechanism of how prostate cancer initiates and develops is still not clear. Here in this study, we show that tumor suppressor ZBTB38 could suppress the migration and proliferation of prostate cancer cells. We find lower ZBTB38 expression in prostate cancer tissues, which also strongly predicts a poorer prognosis of prostate cancer. ZBTB38 binds DKK1 (Dickkopf WNT signaling pathway inhibitor 1) locus and promotes DKK1 expression in prostate cancer cell lines. Consistently, reduction of DKK1 expression significantly restores ZBTB38-mediated suppression of migration and proliferation of prostate cancer cell lines. Mechanistically, we find that ZBTB38 primarily binds the promoters of target genes, and differentially regulates the expression of 1818 genes. We also identify PRKDC (protein kinase, DNA-activated, catalytic subunit) as a ZBTB38-interacting protein that could repress the function of ZBTB38 in suppressing migration and proliferation of prostate cancer cells. Taken together, our results indicate that ZBTB38 could repress cell migration and proliferation in prostate cancer via promoting DKK1 expression, and also provide evidence supporting ZBTB38 as a potential prognosis marker for prostate cancer.
Collapse
Affiliation(s)
- Guanxiong Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei Lu
- School of Nursing, Suzhou Vocational Health College, Suzhou, China
| | - Qing Zhang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Kai Li
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Huihui Zhou
- Department of pathology, Affiliated Yuhuangding Hospital of Qingdao University, Qingdao, China
| | - Fei Wang
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Chunchun Zhao
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Caibin Fan
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Jianqing Wang
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
18
|
Zheng TF, Liu XL, Li X, Wang QQ, Zhao YC, Li X, Li MM, Zhang Y, Zhang M, Zhang WC, Zhang C, Zhang Y, Zhang M. Dickkopf-1 promotes Vascular Smooth Muscle Cell proliferation and migration through upregulating UHRF1 during Cyclic Stretch application. Int J Biol Sci 2021; 17:1234-1249. [PMID: 33867842 PMCID: PMC8040467 DOI: 10.7150/ijbs.56247] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/10/2021] [Indexed: 12/28/2022] Open
Abstract
Dickkopf-1 (DKK1) was recently shown to play an important role in cardiovascular disease. The aim of this work was to assess the role of DKK1 in the regulation of smooth muscle cell function by mechanical stretch and the mechanisms underlying this process. Methods: Wild-type C57BL/6J mice were subjected to sham or abdominal aortic constriction (AAC) surgery. The expression level of DKK1 was examined by immunohistochemical staining and Western blotting. Analyses of DKK1 function in vascular smooth muscle cell (VSMC) proliferation and migration were performed. Transcriptome sequencing analysis was performed to identify the differentially expressed genes and pathways regulated by DKK1. Smooth muscle-specific Dkk1 knockout mice were used to confirm the function of DKK1 in vivo. Chromatin immunoprecipitation (ChIP) was used to confirm DNA-protein interactions. Promoter luciferase analysis was used to detect transcription factor activity. Results: We found that AAC significantly increased DKK1 protein levels in the thoracic aorta and coronary artery in vivo. In vitro, high-level stretch (18%) induced the expression of DKK1 in VSMCs. Knocking down DKK1 inhibited VSMC proliferation and migration under high-level stretch (18%). We identified ubiquitin-like containing PHD and RING finger domains 1 (UHRF1) as a target gene of DKK1. Knockdown of UHRF1 with small interfering RNAs partially reversed the regulatory effect of recombinant DKK1 on VSMCs. Specific deletion of DKK1 in VSMCs was sufficient to attenuate the AAC-induced upregulation of UHRF1, thickening of arterial media and increase in VSMC proliferation. Furthermore, we found that DKK1 regulated UHRF1 expression through the YAP-TEAD pathway. TEAD1 and TEAD4 bound directly to the promoter of UHRF1, and blocking the YAP-TEAD interaction inhibited UHRF1 upregulation due to DKK1. Conclusions: This study reveals that DKK1 mediates the mechanical stretch regulation of smooth muscle cell function by modulating UHRF1 expression through the YAP-TEAD pathway.
Collapse
Affiliation(s)
- Teng-Fei Zheng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University,107 Wenhuaxi Road, 250012 Jinan, China
| | - Xiao-Lin Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University,107 Wenhuaxi Road, 250012 Jinan, China
| | - Xiao Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University,107 Wenhuaxi Road, 250012 Jinan, China
| | - Qian-Qian Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University,107 Wenhuaxi Road, 250012 Jinan, China
| | - Ya-Chao Zhao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University,107 Wenhuaxi Road, 250012 Jinan, China
| | - Xuan Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University,107 Wenhuaxi Road, 250012 Jinan, China
| | - Meng-Meng Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University,107 Wenhuaxi Road, 250012 Jinan, China
| | - Yu Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University,107 Wenhuaxi Road, 250012 Jinan, China
| | - Meng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University,107 Wenhuaxi Road, 250012 Jinan, China
| | - Wen-Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University,107 Wenhuaxi Road, 250012 Jinan, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University,107 Wenhuaxi Road, 250012 Jinan, China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University,107 Wenhuaxi Road, 250012 Jinan, China
| | - Mei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University,107 Wenhuaxi Road, 250012 Jinan, China
| |
Collapse
|
19
|
Zengin T, Önal-Süzek T. Comprehensive Profiling of Genomic and Transcriptomic Differences between Risk Groups of Lung Adenocarcinoma and Lung Squamous Cell Carcinoma. J Pers Med 2021; 11:154. [PMID: 33672117 PMCID: PMC7926392 DOI: 10.3390/jpm11020154] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/11/2021] [Accepted: 02/19/2021] [Indexed: 12/17/2022] Open
Abstract
Lung cancer is the second most frequently diagnosed cancer type and responsible for the highest number of cancer deaths worldwide. Lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) are subtypes of non-small-cell lung cancer which has the highest frequency of lung cancer cases. We aimed to analyze genomic and transcriptomic variations including simple nucleotide variations (SNVs), copy number variations (CNVs) and differential expressed genes (DEGs) in order to find key genes and pathways for diagnostic and prognostic prediction for lung adenocarcinoma and lung squamous cell carcinoma. We performed a univariate Cox model and then lasso-regularized Cox model with leave-one-out cross-validation using The Cancer Genome Atlas (TCGA) gene expression data in tumor samples. We generated 35- and 33-gene signatures for prognostic risk prediction based on the overall survival time of the patients with LUAD and LUSC, respectively. When we clustered patients into high- and low-risk groups, the survival analysis showed highly significant results with high prediction power for both training and test datasets. Then, we characterized the differences including significant SNVs, CNVs, DEGs, active subnetworks, and the pathways. We described the results for the risk groups and cancer subtypes separately to identify specific genomic alterations between both high-risk groups and cancer subtypes. Both LUAD and LUSC high-risk groups have more downregulated immune pathways and upregulated metabolic pathways. On the other hand, low-risk groups have both up- and downregulated genes on cancer-related pathways. Both LUAD and LUSC have important gene alterations such as CDKN2A and CDKN2B deletions with different frequencies. SOX2 amplification occurs in LUSC and PSMD4 amplification in LUAD. EGFR and KRAS mutations are mutually exclusive in LUAD samples. EGFR, MGA, SMARCA4, ATM, RBM10, and KDM5C genes are mutated only in LUAD but not in LUSC. CDKN2A, PTEN, and HRAS genes are mutated only in LUSC samples. The low-risk groups of both LUAD and LUSC tend to have a higher number of SNVs, CNVs, and DEGs. The signature genes and altered genes have the potential to be used as diagnostic and prognostic biomarkers for personalized oncology.
Collapse
Affiliation(s)
- Talip Zengin
- Department of Molecular Biology and Genetics, Muğla Sıtkı Koçman University, 48000 Muğla, Turkey;
- Department of Bioinformatics, Muğla Sıtkı Koçman University, 48000 Muğla, Turkey
| | - Tuğba Önal-Süzek
- Department of Bioinformatics, Muğla Sıtkı Koçman University, 48000 Muğla, Turkey
- Department of Computer Engineering, Muğla Sıtkı Koçman University, 48000 Muğla, Turkey
| |
Collapse
|
20
|
Kong W, Chen Y, Zhao Z, Zhang L, Lin X, Luo X, Wang S, Song Z, Lin X, Lai G, Yu Z. EXT1 methylation promotes proliferation and migration and predicts the clinical outcome of non-small cell lung carcinoma via WNT signalling pathway. J Cell Mol Med 2021; 25:2609-2620. [PMID: 33565239 PMCID: PMC7933929 DOI: 10.1111/jcmm.16277] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 12/29/2020] [Accepted: 12/31/2020] [Indexed: 01/15/2023] Open
Abstract
DNA methylation is important for lung cancer prognosis. In this work, it is aimed to seek novel biomarkers with DNA methylation‐expression‐pathway pattern and explore its underlying mechanism. Prognostic DNA methylation sites and mRNAs were screened in NSCLC data set from TCGA, and further validated using the samples retrospectively collected, and EXT1 was identified as a potential target. Gene body methylation of three CpG sites (cg03276982, cg11592677, cg16286281) on EXT1 was significantly associated with clinical outcome, and the EXT1 gene expression also predicted prognosis. The expression level of EXT1 was also correlated with its DNA methylation level. This observation was further validated in a new data set consist of 170 samples. Knocking down of EXT1 resulted in decreased proliferation and migration. EXT1 targets were analysed using GSEA. It is found that the WNT signalling is the potential downstream target of EXT1. Further analyses revealed that the EXT1 targets the beta‐catenin and effect migration rate of NSCLC cell lines. The WNT signalling inhibitor, XAV‐939, effectively disrupted the migration promotion effect induced by EXT1. In summary, EXT1 methylation regulates the gene expression, effects the proliferation and migration via WNT pathway and predicted a poor prognosis for NSCLC.
Collapse
Affiliation(s)
- Wencui Kong
- Department of Respiratory Medicine and Critical Care Medicine, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, China
| | - Ying Chen
- Department of Respiratory Medicine and Critical Care Medicine, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, China
| | - Zhongquan Zhao
- Department of Respiratory Medicine and Critical Care Medicine, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, China
| | - Lei Zhang
- Department of Respiratory Medicine and Critical Care Medicine, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, China
| | - Xiandong Lin
- Laboratory of Radiation Oncology and Radiobiology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Xingguang Luo
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, West Haven, CT, USA
| | - Shuiliang Wang
- Department of Urology, 900th Hospital of the Joint Logistics Team Support Force, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Transplant Biology, Affiliated Dongfang Hospital, Xiamen University School of Medicine, Fuzhou, China
| | - Zhengbo Song
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, China.,Department of Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, China.,Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xiangwu Lin
- Medical Oncology, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, China
| | - Guoxiang Lai
- Department of Respiratory Medicine and Critical Care Medicine, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, China
| | - Zongyang Yu
- Department of Respiratory Medicine and Critical Care Medicine, The 900th Hospital of Joint Logistic Support Force, PLA, Fujian Medical University,Affiliated Dongfang Hospital, Xiamen University School of Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
21
|
Li C, Long Q, Zhang D, Li J, Zhang X. Identification of a four-gene panel predicting overall survival for lung adenocarcinoma. BMC Cancer 2020; 20:1198. [PMID: 33287749 PMCID: PMC7720456 DOI: 10.1186/s12885-020-07657-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 11/17/2020] [Indexed: 12/24/2022] Open
Abstract
Background Lung cancer is the most frequently diagnosed carcinoma and the leading cause of cancer-related mortality. Although molecular targeted therapy and immunotherapy have made great progress, the overall survival (OS) is still poor due to a lack of accurate and available prognostic biomarkers. Therefore, in this study we aimed to establish a multiple-gene panel predicting OS for lung adenocarcinoma. Methods We obtained the mRNA expression and clinical data of lung adenocarcinoma (LUAD) from TCGA database for further integrated bioinformatic analysis. Lasso regression and Cox regression were performed to establish a prognosis model based on a multi-gene panel. A nomogram based on this model was constructed. The receiver operating characteristic (ROC) curve and the Kaplan–Meier curve were used to assess the predicted capacity of the model. The prognosis value of the multi-gene panel was further validated in TCGA-LUAD patients with EGFR, KRAS and TP53 mutation and a dataset from GEO. Gene set enrichment analysis (GSEA) was performed to explore potential biological mechanisms of a novel prognostic gene signature. Results A four-gene panel (including DKK1, GNG7, LDHA, MELTF) was established for LUAD prognostic indicator. The ROC curve revealed good predicted performance in both test cohort (AUC = 0.740) and validation cohort (AUC = 0.752). Each patient was calculated a risk score according to the model based on the four-gene panel. The results showed that the risk score was an independent prognostic factor, and the high-risk group had a worse OS compared with the low-risk group. The nomogram based on this model showed good prediction performance. The four-gene panel was still good predictors for OS in LUAD patients with TP53 and KRAS mutations. GSEA revealed that the four genes may be significantly related to the metabolism of genetic material, especially the regulation of cell cycle pathway. Conclusion Our study proposed a novel four-gene panel to predict the OS of LUAD, which may contribute to predicting prognosis accurately and making the clinical decisions of individual therapy for LUAD patients.
Collapse
Affiliation(s)
- Chunyu Li
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Qizhong Long
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Danni Zhang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Jun Li
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Xianming Zhang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| |
Collapse
|
22
|
Zhou X, Wang Y, Li Q, Ma D, Nie A, Shen X. LncRNA Linc-PINT inhibits miR-523-3p to hamper retinoblastoma progression by upregulating Dickkopf-1 (DKK1). Biochem Biophys Res Commun 2020; 530:47-53. [PMID: 32828314 DOI: 10.1016/j.bbrc.2020.06.120] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 06/24/2020] [Indexed: 02/08/2023]
Abstract
Emerging evidences indicated that long non-coding RNAs (LncRNAs) regulated the pathogenesis of retinoblastoma (RB). However, up until now, the role of LncRNA Linc-PINT in the regulation of RB progression is still largely unknown. The present study identified LncRNA Linc-PINT as a tumor suppressor to hinder RB development by regulating miR-523-3p/Dickkopf-1 (DKK1) axis. Mechanistically, Linc-PINT was low-expressed, while miR-523-3p was high-expressed in RB cells, compared to the normal retinal epithelial cells (ARPE-19). Further gain- and loss-function experiments verified that both upregulation of Linc-PINT and miR-523-3p downregulation slowed down cell growth, invasion and migration, and promoted cell apoptosis in RB cells, but Linc-PINT ablation and miR-523-3p overexpression promoted malignant phenotypes in RB cells. In addition, the dual-luciferase reporter gene system and RNA pull-down assay validated that Linc-PINT positively regulated DKK1 expressions by sponging miR-523-3p, and Linc-PINT inhibited RB progression by regulating miR-523-3p/DKK1 axis. Functionally, we found that both miR-523-3p overexpression and DKK1 silence abrogated the anti-cancer effects of overexpressed Linc-PINT on RB cells. Finally, Linc-PINT inhibited tumorigenicity of RB cells in xenograft mice models. In general, analysis of the data suggested that Linc-PINT inhibited miR-523-3p to upregulate DKK1, resulting in the inhibition of RB, and we demonstrated that Linc-PINT and miR-523-3p could be utilized as potential diagnostic and therapeutic biomarkers for RB in clinic.
Collapse
Affiliation(s)
- Xiaoping Zhou
- Department of Ophthalmology, the First People's Hospital of Chenzhou, Youth Avenue No.8, Chenzhou, 423000, Hunan, China
| | - Yongping Wang
- Department of Ophthalmology, the First Affiliated Hospital of Dalian Medical University, Zhongshan Road No.222, Dalian, 116011, China
| | - Qiang Li
- Department of Ophthalmology, Shenzhen People's Hospital Affiliated to Jinan University, Dongmen North Road No. 1017, Shenzhen 518020, Guangdong, China
| | - Dahui Ma
- Department of Ophthalmology, Shenzhen Eye Hospital, Shenzhen Eye Institute, School of Optometry, Shenzhen University Department of Ophthalmology, Zetian Road No. 18, Shenzhen, 518040, Guangdong, China
| | - Aiqing Nie
- Department of Ophthalmology, Shenzhen People's Hospital Affiliated to Jinan University, Dongmen North Road No. 1017, Shenzhen 518020, Guangdong, China
| | - Xiaoli Shen
- Department of Ophthalmology, Shenzhen Eye Hospital, Shenzhen Eye Institute, School of Optometry, Shenzhen University Department of Ophthalmology, Zetian Road No. 18, Shenzhen, 518040, Guangdong, China.
| |
Collapse
|
23
|
Yamazaki M, Maruyama S, Abé T, Tsuneki M, Kato H, Izumi K, Tanuma JI, Cheng J, Saku T. Rac1-dependent phagocytosis of apoptotic cells by oral squamous cell carcinoma cells: A possible driving force for tumor progression. Exp Cell Res 2020; 392:112013. [PMID: 32320683 DOI: 10.1016/j.yexcr.2020.112013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 03/30/2020] [Accepted: 04/15/2020] [Indexed: 01/13/2023]
Abstract
Apoptotic cell death frequently occurs in human cancer tissues including oral squamous cell carcinoma (SCC), wherein apoptotic tumor cells are phagocytosed not only by macrophages but also by neighboring tumor cells. We previously reported that the engulfment of apoptotic SCC cells by neighboring SCC cells frequently occurs at the invading front. Therefore, we hypothesized that the phagocytosis of these apoptotic cells by tumor cells contributes to disease progression. Herein, using cultured oral SCC cells, we aimed to confirm whether tumor cells actually phagocytose apoptotic cells and to examine whether cellular activities are regulated by the phagocytosis of apoptotic cells. Co-culture experiments showed that living cells could ingest apoptotic cells into phagolysosomes. NSC23766, an inhibitor of Rac1, which is a key regulator of phagocytic cup formation in professional phagocytes, dramatically suppressed the phagocytosis of apoptotic cells by living cells. Additionally, cell migration and the secretion of DKK1, a tumor-promoting protein, were enhanced by co-culture with apoptotic cells, whereas NSC23766 inhibited these effects. These results show that tumor cells can actively phagocytose apoptotic neighbors in a Rac1-dependent manner and that such activity increases their migration. The regulation of apoptotic cell phagocytosis thus represents new directions for therapeutic intervention for oral cancer.
Collapse
Affiliation(s)
- Manabu Yamazaki
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| | - Satoshi Maruyama
- Oral Pathology Section, Department of Surgical Pathology, Niigata University Hospital, Niigata, Japan
| | - Tatsuya Abé
- Division of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Masayuki Tsuneki
- Oral Pathology Section, Department of Surgical Pathology, Niigata University Hospital, Niigata, Japan
| | - Hiroko Kato
- Division of Biomimetics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Research Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata University, Japan
| | - Kenji Izumi
- Division of Biomimetics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Jun-Ichi Tanuma
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Jun Cheng
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takashi Saku
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Fukuoka Dental College, Fukuoka, Japan
| |
Collapse
|
24
|
Luo Q, Li B, Li G. Mannose Suppresses the Proliferation and Metastasis of Lung Cancer by Targeting the ERK/GSK-3β/β-Catenin/SNAIL Axis. Onco Targets Ther 2020; 13:2771-2781. [PMID: 32308412 PMCID: PMC7135191 DOI: 10.2147/ott.s241816] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/18/2020] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION It has been found that mannose exerts antitumoural properties in vitro and in animal models. Whether mannose has potential anti-proliferative and anti-metastatic properties against non-small-cell lung cancer (NSCLC) is still unclear. METHODS Here, we performed ex vivo experiments and established a nude mouse model to evaluate the anticancer effects of mannose on NSCLC cells and its effects on the ERK/GSK-3β/β-catenin/SNAIL axis. A CCK-8 assay was conducted to evaluate the effects of mannose on lung cancer cells (A549 and HCC827) and normal lung cells (HPAEpiC). Transwells were used to examine the motility of cancer cells. qRT-PCR was used to evaluate the effects of mannose on the mRNA expression of β-catenin. Western blotting was conducted to explore the effects of mannose on the ERK/GSK-3β/β-catenin/SNAIL axis and nuclear accumulation of β-catenin. An animal model was established to evaluate the antitumoural effect of mannose on hepatic metastasis in vivo. RESULTS In this study, we found that mannose inhibited the proliferation of A549 and HCC827 cells in vitro both time- and dose-dependently. However, it exerted only a slight influence on the viability of normal lung cells in vitro. Moreover, mannose also inhibited the migrating and invading capacity of NSCLC cells in vitro. Using Western blotting, we observed that mannose reduced SNAIL and β-catenin expression and ERK activation and promoted phospho-GSK-3β expression. The ERK agonist LM22B-10 promoted the metastatic ability of NSCLC cells and increased SNAIL and β-catenin expression in cancer cells, which could be reversed by mannose. Furthermore, ERK-mediated phosphorylation of the β-catenin-Tyr654 residue might participate in the nuclear accumulation of β-catenin and its transcriptional function. The results from animal experiments showed that mannose effectively reduced hepatic metastasis of A549 cells in vivo. Furthermore, mannose inhibited ERK/GSK-3β/β-catenin/SNAIL in tumour tissues obtained from nude mice. DISCUSSION Collectively, these findings suggest that mannose exerts anti-metastatic activity against NSCLC by inhibiting the activation of the ERK/GSK-3β/β-catenin/SNAIL axis, which indicates the potential anticancer effects of mannose.
Collapse
Affiliation(s)
- Qingsong Luo
- Thoracic Surgery, Sichuan Academy Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, Sichuan610072, People’s Republic of China
| | - Bei Li
- Thoracic Surgery, Sichuan Academy Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, Sichuan610072, People’s Republic of China
| | - Gang Li
- Thoracic Surgery, Sichuan Academy Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, Sichuan610072, People’s Republic of China
| |
Collapse
|
25
|
Fezza M, Moussa M, Aoun R, Haber R, Hilal G. DKK1 promotes hepatocellular carcinoma inflammation, migration and invasion: Implication of TGF-β1. PLoS One 2019; 14:e0223252. [PMID: 31568519 PMCID: PMC6768474 DOI: 10.1371/journal.pone.0223252] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 09/17/2019] [Indexed: 12/24/2022] Open
Abstract
Dickkopf-1 (DKK1), an inhibitor of the most frequently impaired signaling pathway in hepatocellular carcinoma (HCC), the Wnt/beta-catenin pathway, seems to fulfill contradictory functions in the process of tumorigenesis, acting either as an oncogenic promoter of metastasis or as a tumor suppressor. Elevated serum levels of DKK1 have been reported in HCC; however, little is known about its functional significance. In the current study, we treated HepG2/C3A and PLC/PRF/5 with the recombinant protein DKK1. Cytotoxicity was first determined by the WST-8 assay. AFP expression was measured at both the mRNA and protein levels. Expression of the oncogenes MYC, CCND1, hTERT, and MDM2 and the tumor suppressor genes TP53, P21 and RB was assessed. Western blot analysis of non-phosphorylated ẞ-catenin and Sanger sequencing were performed to explain the functional differences between the two cell lines. Subsequently, inflammation, migration and invasion were evaluated by qPCR, ELISA, the Boyden chamber assay, zymography, and MMP-2 and MMP-9 western blot analysis. Knockdown of DKK1 and TGF-β1 were also performed. Our results suggest that DKK1 exerts an oncogenic effect on HepG2/C3A cell line by upregulating the expression of oncogenes and downregulating that of tumor suppressor genes, whereas the opposite effect was demonstrated in PLC/PRF/5 cells. This differential impact of DKK1 can be explained by the mutations that affect the canonical Wnt pathway that were detected in exon 3 of the CTNNB1 gene in the HepG2 cell line. We further confirmed that DKK1 promotes inflammation, tumor invasion and migration in both cell types. The canonical pathway was not responsible for the DKK1 proinvasive effect, as indicated by the active ẞ-catenin levels in the two cell lines upon DKK1 treatment. Interestingly, knockdown of TGF-β1 negatively affected the DKK1 proinvasive effect. Taken together, DKK1 appears to facilitate tumor invasion and migration through TGF- β1 by remodeling the tumor microenvironment and inducing inflammation. This finding endorses the relevance of TGF-β1 as a therapeutic target.
Collapse
Affiliation(s)
- Maha Fezza
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Mayssam Moussa
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Rita Aoun
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Rita Haber
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - George Hilal
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
- * E-mail:
| |
Collapse
|
26
|
Song Z, Wang H, Zhang S. Negative regulators of Wnt signaling in non-small cell lung cancer: Theoretical basis and therapeutic potency. Biomed Pharmacother 2019; 118:109336. [PMID: 31545260 DOI: 10.1016/j.biopha.2019.109336] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 07/20/2019] [Accepted: 08/05/2019] [Indexed: 02/05/2023] Open
Abstract
Significant advances in the treatment of non-small cell lung cancer (NSCLC) have been made over the past decade, and they predominantly involve molecular targets such as epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase (ALK) rearrangements. However, despite the initial good response, drug resistance eventually develops. The Wnt signaling pathway has recently been considered important in embryonic development and tumorigenesis in many cancers, particularly NSCLC. Moreover, the aberrant Wnt pathway plays a significant role in NSCLC and is associated with cancer cell proliferation, metastasis, invasion and drug resistance, and the suppression of canonical or noncanonical Wnt signaling through various biological or pharmacological negative regulators has been proven to produce specific anticancer effects. Thus, blocking the Wnt pathway via its negative regulators may overcome the resistance of current treatment methods and lead to new treatment strategies for NSCLC. Therefore, in this review, we summarize recent studies on the role of negative regulators in Wnt signaling in NSCLC and the therapeutic potency of these molecules as agents and targets for NSCLC treatments.
Collapse
Affiliation(s)
- Zikuan Song
- West China School of Basic Medical Science and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Haoyu Wang
- West China School of Basic Medical Science and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shuang Zhang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
27
|
Magruder S, Carter E, Williams MA, English J, Akyalcin S, Letra A. Further evidence for the role of WNT10A, WNT10B
and GREM2
as candidate genes for isolated tooth agenesis. Orthod Craniofac Res 2018; 21:258-263. [DOI: 10.1111/ocr.12248] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/05/2018] [Accepted: 09/16/2018] [Indexed: 01/08/2023]
Affiliation(s)
- Sonia Magruder
- Department of Orthodontics; UTHealth School of Dentistry; Houston Texas
| | - Emily Carter
- Department of Orthodontics; UTHealth School of Dentistry; Houston Texas
| | - Meredith A. Williams
- Department of Orthodontics; UTHealth School of Dentistry; Houston Texas
- Center for Craniofacial Research; UTHealth School of Dentistry; Houston Texas
| | - Jeryl English
- Department of Orthodontics; UTHealth School of Dentistry; Houston Texas
| | - Sercan Akyalcin
- Department of Orthodontics; Tufts University School of Dental Medicine; Boston Massachusetts
| | - Ariadne Letra
- Center for Craniofacial Research; UTHealth School of Dentistry; Houston Texas
- Department of Diagnostic and Biomedical Sciences; UTHealth School of Dentistry; Houston Texas
| |
Collapse
|
28
|
Tang Y, Zhang Z, Tang Y, Chen X, Zhou J. Identification of potential target genes in pancreatic ductal adenocarcinoma by bioinformatics analysis. Oncol Lett 2018; 16:2453-2461. [PMID: 30013637 PMCID: PMC6036577 DOI: 10.3892/ol.2018.8912] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/22/2018] [Indexed: 12/29/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most complicated and fatally pathogenic human malignancies. Therefore, there is an urgent need to improve our understanding of the underlying molecular mechanism that drives the initiation, progression, and metastasis of PDAC. The aim of the present study was to identify the key genes and signaling pathways associated with PDAC using bioinformatics analysis. Four transcriptome microarray datasets (GSE15471, GSE55643, GSE62165 and GSE91035) were acquired from Gene Expression Omnibus datasets, which included 226 PDAC samples and 65 normal pancreatic tissue samples. We screened differentially expressed genes (DEGs) with GEO2R and investigated their biological function by Gene Ontology (GO) and Kyoto Encyclopedia of Genes (KEGG) analysis. The overall survival data was obtained from UALCAN, which calculated the data shared with The Cancer Genome Atlas. In addition, a protein-protein interaction (PPI) network of the DEGs was constructed by STRING and Cytoscape software. The four sets of DEGs exhibited an intersection consisting of 205 genes (142 up-regulated and 63 down-regulated), which may be associated with PDAC. GO analysis showed that the 205 DEGs were significantly enriched in the plasma membrane, cell adhesion molecule activity and the Energy pathways, and glycine, serine, threonine metabolism were the most enriched pathways according to KEGG pathway analysis. Kaplan-Meier survival analysis revealed that 22 of 205 common genes were significantly associated with the overall survival of pancreatic cancer patients. In the PPI network and sub-network, DKK1 and HMGA2 were considered as hub genes with high connectivity degrees. DKK1 and HMGA2 are strongly associated with WNT3A and TP53 separately, which indicates that they may play an important role in the Wnt and P53 signaling pathways. Using integrated bioinformatics analysis, we identified DKK1 and HMGA2 as candidate genes in PDAC, which may improve our understanding of the mechanisms of the pathogenesis and integration; the two genes may be therapeutic targets and prognostic markers for PDAC.
Collapse
Affiliation(s)
- Yuchen Tang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China.,Pancreatic Disease Research Center, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Zixiang Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China.,Pancreatic Disease Research Center, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yaocheng Tang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Xinyu Chen
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jian Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China.,Pancreatic Disease Research Center, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|