1
|
Lei M, Huang H, Li J, Zhang J, Yu G, Jin X, Liu J, Kang F, Liu Z. Design, synthesis and biological evaluation of coumarin-containing 2,4-diphenylpyrimidine derivatives as novel focal adhesion kinase inhibitors for treatment of non-small cell lung cancer. Bioorg Med Chem Lett 2025; 123:130240. [PMID: 40228675 DOI: 10.1016/j.bmcl.2025.130240] [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: 01/21/2025] [Revised: 03/16/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025]
Abstract
A series of hybrids (8a-h and 11a-h) containing 2,4-diphenylpyrimidine scaffold and coumarin moiety were designed and synthesized as novel focal adhesion kinase (FAK) inhibitors for the intervention of non-small-cell lung cancer (NSCLC). Most compounds effectively suppressed the proliferative of NSCLC cells, and compound 8a was identified as the most active compound with IC50 value of 0.28 μM in H1299 cells, superior to TAE226 (IC50 = 2.28 μM). In addition, 8a was also found to inhibit the invasion and migration of NSCLC cells. Furthermore, 8a exhibited potent kinase inhibitory activity of FAK (IC50 = 4.968 nM) with a considerable selectivity profile against various kinase families, subsequently resulting in cell cycle arrest, apoptosis- inducing as well as the decrease of MMP-2 and MMP-9 expression in H1299 cells dose-dependently. Moreover, 8a was relatively safe to mice and inhibited the growth of implanted NSCLC tumors more potently than TAE226 in mice. Therefore, 8a may be a promising candidate for the treatment of NSCLC.
Collapse
Affiliation(s)
- Mengrong Lei
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Engineering Research Center for applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, PR China
| | - Hanxue Huang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Engineering Research Center for applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, PR China
| | - Jiayi Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, PR China
| | - Jinlin Zhang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, PR China
| | - Geng Yu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, PR China
| | - Xin Jin
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Engineering Research Center for applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, PR China
| | - Junyan Liu
- Department of orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Fenghua Kang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, PR China.
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Engineering Research Center for applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, PR China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, PR China.
| |
Collapse
|
2
|
Verma S, Magazzù G, Eftekhari N, Lou T, Gilhespy A, Occhipinti A, Angione C. Cross-attention enables deep learning on limited omics-imaging-clinical data of 130 lung cancer patients. CELL REPORTS METHODS 2024; 4:100817. [PMID: 38981473 PMCID: PMC11294841 DOI: 10.1016/j.crmeth.2024.100817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 04/18/2024] [Accepted: 06/17/2024] [Indexed: 07/11/2024]
Abstract
Deep-learning tools that extract prognostic factors derived from multi-omics data have recently contributed to individualized predictions of survival outcomes. However, the limited size of integrated omics-imaging-clinical datasets poses challenges. Here, we propose two biologically interpretable and robust deep-learning architectures for survival prediction of non-small cell lung cancer (NSCLC) patients, learning simultaneously from computed tomography (CT) scan images, gene expression data, and clinical information. The proposed models integrate patient-specific clinical, transcriptomic, and imaging data and incorporate Kyoto Encyclopedia of Genes and Genomes (KEGG) and Reactome pathway information, adding biological knowledge within the learning process to extract prognostic gene biomarkers and molecular pathways. While both models accurately stratify patients in high- and low-risk groups when trained on a dataset of only 130 patients, introducing a cross-attention mechanism in a sparse autoencoder significantly improves the performance, highlighting tumor regions and NSCLC-related genes as potential biomarkers and thus offering a significant methodological advancement when learning from small imaging-omics-clinical samples.
Collapse
Affiliation(s)
- Suraj Verma
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, UK
| | | | | | - Thai Lou
- Gateshead Health NHS Foundation Trust, Gateshead, UK
| | - Alex Gilhespy
- South Tyneside and Sunderland NHS Foundation Trust, Sunderland, UK
| | - Annalisa Occhipinti
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, UK; Centre for Digital Innovation, Teesside University, Middlesbrough, UK; National Horizons Centre, Teesside University, Darlington, UK
| | - Claudio Angione
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, UK; Centre for Digital Innovation, Teesside University, Middlesbrough, UK; National Horizons Centre, Teesside University, Darlington, UK.
| |
Collapse
|
3
|
Flockerzi FA, Hohneck J, Langer F, Bohle RM, Stahl PR. THSD7A Positivity Predicts Poor Survival and Is Linked to High FAK Expression and FGFR1-Wildtype in Female Patients with Squamous Cell Carcinoma of the Lung. Int J Mol Sci 2023; 24:10639. [PMID: 37445817 DOI: 10.3390/ijms241310639] [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: 05/21/2023] [Revised: 06/14/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths in the western world, with squamous cell carcinoma being one of the most common histological subtypes. Prognostic and predictive markers are still largely missing for squamous cell carcinoma of the lung (LSCC). Several studies indicate that THSD7A might at least play a role in the prognosis of different tumors. FAK seems to play an important role in lung cancer and is discussed as a potential therapeutic target. In addition, there is evidence that FAK-dependent signaling pathways might be affected by THSD7A. For that reason, we investigated the role of THSD7A as a potential tumor marker in LSCC and whether THSD7A expression has an impact on the expression level of FAK. A total of 101 LSCCs were analyzed by immunohistochemistry using tissue microarrays. THSD7A positivity was associated with poor overall survival in female patients and showed a relation to high FAK expression in this subgroup. To our knowledge, we are the first to report these correlations in lung cancer. The results might be proof of the assumed activation of FAK-dependent signaling pathways by THSD7A and that as a membrane-associated protein, THSD7A might serve as a putative therapeutic target in LSCC.
Collapse
Affiliation(s)
| | - Johannes Hohneck
- Department of Pathology, Saarland University Medical Center, 66424 Homburg, Germany
| | - Frank Langer
- Department of Thoracic and Cardiovascular Surgery, Saarland University Medical Center, 66424 Homburg, Germany
| | - Rainer Maria Bohle
- Department of Pathology, Saarland University Medical Center, 66424 Homburg, Germany
| | - Phillip Rolf Stahl
- Department of Pathology, Saarland University Medical Center, 66424 Homburg, Germany
| |
Collapse
|
4
|
Zhang Z, Li J, Jiao S, Han G, Zhu J, Liu T. Functional and clinical characteristics of focal adhesion kinases in cancer progression. Front Cell Dev Biol 2022; 10:1040311. [PMID: 36407100 PMCID: PMC9666724 DOI: 10.3389/fcell.2022.1040311] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022] Open
Abstract
Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase and an adaptor protein that primarily regulates adhesion signaling and cell migration. FAK promotes cell survival in response to stress. Increasing evidence has shown that at the pathological level, FAK is highly expressed in multiple tumors in several systems (including lung, liver, gastric, and colorectal cancers) and correlates with tumor aggressiveness and patient prognosis. At the molecular level, FAK promotes tumor progression mainly by altering survival signals, invasive capacity, epithelial-mesenchymal transition, the tumor microenvironment, the Warburg effect, and stemness of tumor cells. Many effective drugs have been developed based on the comprehensive role of FAK in tumor cells. In addition, its potential as a tumor marker cannot be ignored. Here, we discuss the pathological and pre-clinical evidence of the role of FAK in cancer development; we hope that these findings will assist in FAK-based clinical studies.
Collapse
Affiliation(s)
- Zhaoyu Zhang
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jinlong Li
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Simin Jiao
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Guangda Han
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jiaming Zhu
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Tianzhou Liu
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
5
|
Chen F, Zhong Z, Zhang C, Lu Y, Chan YT, Wang N, Zhao D, Feng Y. Potential Focal Adhesion Kinase Inhibitors in Management of Cancer: Therapeutic Opportunities from Herbal Medicine. Int J Mol Sci 2022; 23:13334. [PMID: 36362132 PMCID: PMC9659249 DOI: 10.3390/ijms232113334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/27/2022] [Accepted: 10/29/2022] [Indexed: 08/15/2024] Open
Abstract
Focal adhesion kinase (FAK) is a multifunctional protein involved in cellular communication, integrating and transducing extracellular signals from cell-surface membrane receptors. It plays a central role intracellularly and extracellularly within the tumor microenvironment. Perturbations in FAK signaling promote tumor occurrence and development, and studies have revealed its biological behavior in tumor cell proliferation, migration, and adhesion. Herein we provide an overview of the complex biology of the FAK family members and their context-dependent nature. Next, with a focus on cancer, we highlight the activities of FAK signaling in different types of cancer and how knowledge of them is being used for screening natural compounds used in herbal medicine to fight tumor development.
Collapse
Affiliation(s)
- Feiyu Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Zhangfeng Zhong
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yuanjun Lu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yau-Tuen Chan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Di Zhao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
6
|
Peng K, Li S, Li Q, Zhang C, Yuan Y, Liu M, Zhang L, Wang Y, Yu S, Zhang H, Liu T. Positive Phospho-Focal Adhesion Kinase in Gastric Cancer Associates With Poor Prognosis After Curative Resection. Front Oncol 2022; 12:953938. [PMID: 35982966 PMCID: PMC9379279 DOI: 10.3389/fonc.2022.953938] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
Gastric cancer (GC) is the fifth most commonly diagnosed cancer and usually has a dismal prognosis. Our previous study highlights the contribution of focal adhesion kinase (FAK) in the tumorigenesis of diffuse gastric cancer (DGC), a subtype of GC according to Lauren classification. The prognostic value of phosphorylated FAK (pFAK) in GC remains to be explored. To explore the prognostic value of pFAK, we retrospectively collected 176 formalin-fixed paraffin-embedded (FFPE) tumor tissues from GC patients who underwent D2 gastrectomy without neoadjuvant treatment. The immunohistochemistry (IHC) staining of pFAK was performed. Survival analysis was performed by Kaplan–Meier and risk factors were evaluated by Cox regression analysis. A pFAK-based nomogram was also constructed for the prediction of overall survival (OS). We demonstrated that the prognosis of pFAK-positive patients was worse than that of the pFAK-negative patients in GC (p = 0.010; hazard ratio [HR] = 1.777, 95% CI 1.131 to 2.791; median OS, 46.6 vs. 86.3 months, respectively), and positive pFAK was also an independent risk factor for the worse prognosis of GC (p = 0.0054; HR = 1.89, 95% CI 1.21–2.96). Moreover, the nomogram based on pFAK and other independent risk factors could improve predictive accuracy for prognosis of GC. In conclusion, through analysis of a large collection of clinically annotated GC samples, we demonstrate that pFAK is a negative prognostic factor in GC, and a nomogram integrating pFAK could help predict OS for GC patients.
Collapse
Affiliation(s)
- Ke Peng
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Center of Evidence-Based Medicine, Fudan University, Shanghai, China
| | - Suyao Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Center of Evidence-Based Medicine, Fudan University, Shanghai, China
| | - Qian Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Center of Evidence-Based Medicine, Fudan University, Shanghai, China
| | - Chenlu Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yitao Yuan
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Center of Evidence-Based Medicine, Fudan University, Shanghai, China
| | - Menglin Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Center of Evidence-Based Medicine, Fudan University, Shanghai, China
| | - Lei Zhang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yichen Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shan Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Center of Evidence-Based Medicine, Fudan University, Shanghai, China
| | - Haisheng Zhang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Tianshu Liu, ; Haisheng Zhang,
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Center of Evidence-Based Medicine, Fudan University, Shanghai, China
- *Correspondence: Tianshu Liu, ; Haisheng Zhang,
| |
Collapse
|
7
|
Gunn SA, Kreps LM, Zhao H, Landon K, Ilacqua JS, Addison CL. Focal Adhesion Kinase Inhibitors Prevent Osteoblast Mineralization in Part Due to Suppression of Akt-mediated stabilization of Osterix. J Bone Oncol 2022; 34:100432. [PMID: 35620245 PMCID: PMC9126966 DOI: 10.1016/j.jbo.2022.100432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 04/06/2022] [Accepted: 04/19/2022] [Indexed: 11/17/2022] Open
Abstract
Pharmacological blockade of FAK results in reduced ALP expression and mineralization by differentiated osteoblasts. Although FAK inhibition resulted in increased levels of BMP2, Wnt3a and Mdm2, and decreased p53, alteration of these pathways was unable to restore mineralization in the presence of FAK tyrosine kinase inhibitors. FAK tyrosine kinase inhibitors resulted in decreased levels of phospho-S473 Akt which led to increased levels of active GSK3β which in turn inhibited Runx2 activity that could contribute to the observed reduced ALP levels. FAK tyrosine kinase inhibitors blocked Akt-mediated stabilization of osterix leading to decreased overall levels of osterix and impaired mineralization in MC3T3-E1 cells differentiated into osteoblasts.
Focal Adhesion Kinase (FAK) is an important regulator of tumor cell proliferation, survival and metastasis. As such it has become a therapeutic target of interest in cancer. Previous studies suggested that use of FAK tyrosine kinase inhibitors (TKIs) blocks osteolysis in in vivo models of bone metastasis. However, from these studies it was not clear whether FAK TKIs blocked bone degradation by osteoclasts or also promoted bone formation by osteoblasts. In this study we evaluated whether use of the FAK TKI PF-562,271 affected the differentiation of pre-osteoblasts, or activity of mature differentiated osteoblasts. MC3T3-E1 pre-osteoblastic cells were treated with various doses of PF-562,271 following 3 or 10 days of differentiation which led to the inhibition of alkaline phosphatase (ALP) expression and reduced viable cell numbers in a dose-dependent manner. MC3T3-E1 cells which had been differentiated for 21 days prior to treatment with PF-562,271 showed a dose dependent decrease in mineralization as assessed by Alizarin Red staining, with concomitant decreased expression of ALP which is known to facilitate the bone mineralization activity of osteoblasts, however mRNA levels of the transcription factors RUNX2 and osterix which are important for osteoblast maturation and mineralization appeared unaffected at this time point. We speculated that this may be due to altered function of RUNX2 protein due to inhibitory phosphorylation by GSK3β. We found treatment with PF-562,271 resulted in increased GSK3β activity as measured by reduced levels of phospho-Ser9-GSK3β which would result in phosphorylation and inhibition of RUNX2. Treatment of 21 day differentiated MC3T3-E1 cells with PF-562,271 in combination with GSK3β inhibitors partially restored mineralization however this was not statistically significant. As we observed that FAK TKI also resulted in suppression of Akt, which is known to alter osterix protein stability downstream of RUNX2, we examined protein levels by western blot and found a dose-dependent decrease in osterix in FAK TKI treated differentiated MC3T3-E1 cells which is likely responsible for the reduced mineralization observed. Taken together our results suggest that use of FAK TKIs as therapeutics in the bone metastatic setting may block new bone formation as an off-target effect and thereby exacerbate the defective bone regulation that is characteristic of the bone metastatic environment.
Collapse
|
8
|
Lees DM, Reynolds LE, Pedrosa AR, Roy-Luzarraga M, Hodivala-Dilke KM. Phosphorylation of pericyte FAK-Y861 affects tumour cell apoptosis and tumour blood vessel regression. Angiogenesis 2021; 24:471-482. [PMID: 33730293 PMCID: PMC8292267 DOI: 10.1007/s10456-021-09776-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 02/25/2021] [Indexed: 12/20/2022]
Abstract
Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that is overexpressed in many cancer types and in vivo studies have shown that vascular endothelial cell FAK expression and FAK-phosphorylation at tyrosine (Y) 397, and subsequently FAK-Y861, are important in tumour angiogenesis. Pericytes also play a vital role in regulating tumour blood vessel stabilisation, but the specific involvement of pericyte FAK-Y397 and FAK-Y861 phosphorylation in tumour blood vessels is unknown. Using PdgfrβCre + ;FAKWT/WT, PdgfrβCre + ;FAKY397F/Y397F and PdgfrβCre + ;FAKY861F/Y861F mice, our data demonstrate that Lewis lung carcinoma tumour growth, tumour blood vessel density, blood vessel perfusion and pericyte coverage were affected only in late stage tumours in PdgfrβCre + ;FAKY861F/Y861F but not PdgfrβCre + ;FAKY397F/Y397F mice. Further examination indicates a dual role for pericyte FAK-Y861 phosphorylation in the regulation of tumour vessel regression and also in the control of pericyte derived signals that influence apoptosis in cancer cells. Overall this study identifies the role of pericyte FAK-Y861 in the regulation of tumour vessel regression and tumour growth control and that non-phosphorylatable FAK-Y861F in pericytes reduces tumour growth and blood vessel density.
Collapse
Affiliation(s)
- Delphine M Lees
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Microenvironment, Barts Cancer Institute - a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Louise E Reynolds
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Microenvironment, Barts Cancer Institute - a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Ana Rita Pedrosa
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Microenvironment, Barts Cancer Institute - a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Marina Roy-Luzarraga
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Microenvironment, Barts Cancer Institute - a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Kairbaan M Hodivala-Dilke
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Microenvironment, Barts Cancer Institute - a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
9
|
Jiang W, Xu Z, Yu L, Che J, Zhang J, Yang J. MicroRNA-144-3p suppressed TGF-β1-induced lung cancer cell invasion and adhesion by regulating the Src-Akt-Erk pathway. Cell Biol Int 2020; 44:51-61. [PMID: 31038242 DOI: 10.1002/cbin.11158] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/28/2019] [Indexed: 01/24/2023]
Abstract
Lung cancer remains a leading cause to cancer-related death worldwide. The anti-cancer ability of microRNA-144-3p has been reported in many cancer types. This study focused on the mechanisms underlying miR-144-3p in inhibiting lung cancer. The expression levels of miR-144-3p and steroid receptor coactivator (Src) in different lung cancer cell lines and those in bronchial epithelial cells (16HBE) were compared. miR-144-3p mimic and siSrc were transfected into A549 cells. Under the conditions of transforming growth factor-β1 (TGF-β1). Small interfering transfection or TGF-β1 treatment, cell invasive and adhesive abilities were analyzed by Transwell and cell adhesion assays. miR-144-3p inhibitor and siSrc were co-transfected into A549 cells and the changes in cell invasion and adhesion were detected. The activation of Src-protein kinase B-extracellular-regulated protein kinases (Src-Akt-Erk) pathway was determined using Western blot. The downregulated miR-144-3p and upregulated Src were generally detected in lung cancer cell lines and were the most significant genes in A549 cells. Both miR-144-3p overexpression and Src inhibition could obviously inhibit the invasion and adhesion abilities of A549 cells in the presence or absence of the effects of TGF-β1. The inhibition of Src could block the promotive effects of miR-144-3p inhibitor and TGF-β1 on cell invasion and adhesion. Furthermore, we found that miR-144-3p could negatively regulate the phosphorylation levels of Akt and Erk. Our data indicated the essential role of Src in the mechanisms underlying TGF-β1-induced cell invasion and adhesion of lung cancer, and that miR-144-3p could effectively suppress TGF-β1-induced aggressive lung cancer cells by regulating Src expression.
Collapse
Affiliation(s)
- Wensheng Jiang
- Department of Cardiothoracic Surgery, Yantaishan Hospital of Yantai City, No. 91 Jiefang Road, Yantai, Shandong Province, 264000, China
| | - Zhiying Xu
- Department of Nuclear Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20 Yuhuangding East Road, Zhifu District, Yantai, Shandong Province, 264000, China
| | - Lili Yu
- Department of Breast Surgery, Yantaishan Hospital of Yantai City, No. 91 Jiefang Road, Yantai, Shandong Province, 264000, China
| | - Jianpeng Che
- Department of Cardiothoracic Surgery, Yantaishan Hospital of Yantai City, No. 91 Jiefang Road, Yantai, Shandong Province, 264000, China
| | - Jie Zhang
- Department of Cardiothoracic Surgery, Yantaishan Hospital of Yantai City, No. 91 Jiefang Road, Yantai, Shandong Province, 264000, China
| | - Jun Yang
- Department of Cardiothoracic Surgery, Yantaishan Hospital of Yantai City, No. 91 Jiefang Road, Yantai, Shandong Province, 264000, China
| |
Collapse
|
10
|
Shen T, Guo Q. EGFR signaling pathway occupies an important position in cancer-related downstream signaling pathways of Pyk2. Cell Biol Int 2020; 44:2-13. [PMID: 31368612 PMCID: PMC6973235 DOI: 10.1002/cbin.11209] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 07/27/2019] [Indexed: 01/24/2023]
Abstract
Proline-rich tyrosine kinase 2 (Pyk2) is a member of focal adhesion kinase (FAK) non-receptor tyrosine kinase family and has been found to promote cancer cell survival, proliferation, migration, invasion, and metastasis. Pyk2 takes part in different carcinogenic signaling pathways to promote cancer progression, including epidermal growth factor receptor (EGFR) signaling pathway. EGFR signaling pathway is a traditional carcinogenic signaling pathway, which plays a critical role in tumorigenesis and tumor progression. FAK inhibitors have been reported to fail to get the ideal anti-cancer outcomes because of activation of EGFR signaling pathway. Better understanding of Pyk2 downstream targets and interconnectivity between Pyk2 and carcinogenic EGFR signaling pathway will help finding more effective targets for clinical anti-cancer combination therapies. Thus, the interconnectivity between Pyk2 and EGFR signaling pathway, which regulates tumor development and metastasis, needs to be elucidated. In this review, we summarized the downstream targets of Pyk2 in cancers, focused on the connection between Pyk2 and EGFR signaling pathway in different cancer types, and provided a new overview of the roles of Pyk2 in EGFR signaling pathway and cancer development.
Collapse
Affiliation(s)
- Ting Shen
- Medical SchoolKunming University of Science and TechnologyKunming650500YunnanChina,Department of Gastroenterology, The Affiliated Hospital of Kunming University of Science and TechnologyThe First People's Hospital of Yunnan ProvinceKunming650032YunnanChina
| | - Qiang Guo
- Medical SchoolKunming University of Science and TechnologyKunming650500YunnanChina,Department of Gastroenterology, The Affiliated Hospital of Kunming University of Science and TechnologyThe First People's Hospital of Yunnan ProvinceKunming650032YunnanChina
| |
Collapse
|
11
|
Masraksa W, Tanasawet S, Hutamekalin P, Wongtawatchai T, Sukketsiri W. Luteolin attenuates migration and invasion of lung cancer cells via suppressing focal adhesion kinase and non-receptor tyrosine kinase signaling pathway. Nutr Res Pract 2019; 14:127-133. [PMID: 32256987 PMCID: PMC7075744 DOI: 10.4162/nrp.2020.14.2.127] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/16/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND/OBJECTIVES Non-small cell lung cancer is mostly recognized among other types of lung cancer with a poor prognosis by cause of chemotherapeutic resistance and increased metastasis. Luteolin has been found to decrease cell metastasis. However, its underlying mechanisms remain unresolved. The objective of this study was to examine the effect (and its mechanism) of luteolin on the migration and invasion of human non-small cell lung cancer A549 cells. MATERIALS/METHODS Cell viability was investigated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Wound healing and transwell assays were evaluated to assess migration and invasion, respectively. Western blot analysis and immunofluorescence were further performed to investigate the role of luteolin and its mechanisms of action. RESULTS Administration with up to 40 µM luteolin showed no cytotoxic activity on lung cancer A549 cells or non-cancer MRC-5 cells. Additionally, luteolin at 20-40 µM significantly suppressed A549 cells' migration, invasion, and the formation of filopodia in a concentration-dependent manner at 24 h. This is similar with western blot analysis, which revealed diminished the phosphorylated focal adhesion kinase (pFAK), phosphorylated non-receptor tyrosine kinase (pSrc), Ras-related C3 botulinum toxin substrate 1 (Rac1), cell division control protein 42 (Cdc42), and Ras homolog gene family member A (RhoA) expression levels. CONCLUSIONS Overall, our data indicate that luteolin plays a role in controlling lung cancer cells' migration and invasion via Src/FAK and its downstream Rac1, Cdc42, and RhoA pathways. Luteolin might be considered a promising candidate for suppressing invasion and metastasis of lung cancer cells.
Collapse
Affiliation(s)
- Wuttipong Masraksa
- Department of Pharmacology, Faculty of Science, Prince of Songkla University, Songkhla, 90110, Thailand
| | - Supita Tanasawet
- Department of Anatomy, Faculty of Science, Prince of Songkla University, Songkhla, 90110, Thailand
| | - Pilaiwanwadee Hutamekalin
- Department of Physiology, Faculty of Science, Prince of Songkla University, Songkhla, 90110, Thailand
| | - Tulaporn Wongtawatchai
- Department of Anatomy, Faculty of Science, Prince of Songkla University, Songkhla, 90110, Thailand
| | - Wanida Sukketsiri
- Department of Pharmacology, Faculty of Science, Prince of Songkla University, Songkhla, 90110, Thailand
| |
Collapse
|
12
|
Aboubakar Nana F, Vanderputten M, Ocak S. Role of Focal Adhesion Kinase in Small-Cell Lung Cancer and Its Potential as a Therapeutic Target. Cancers (Basel) 2019; 11:E1683. [PMID: 31671774 PMCID: PMC6895835 DOI: 10.3390/cancers11111683] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/22/2019] [Accepted: 10/24/2019] [Indexed: 02/07/2023] Open
Abstract
Small-cell lung cancer (SCLC) represents 15% of all lung cancers and it is clinically the most aggressive type, being characterized by a tendency for early metastasis, with two-thirds of the patients diagnosed with an extensive stage (ES) disease and a five-year overall survival (OS) as low as 5%. There are still no effective targeted therapies in SCLC despite improved understanding of the molecular steps leading to SCLC development and progression these last years. After four decades, the only modest improvement in OS of patients suffering from ES-SCLC has recently been shown in a trial combining atezolizumab, an anti-PD-L1 immune checkpoint inhibitor, with carboplatin and etoposide, chemotherapy agents. This highlights the need to pursue research efforts in this field. Focal adhesion kinase (FAK) is a non-receptor protein tyrosine kinase that is overexpressed and activated in several cancers, including SCLC, and contributing to cancer progression and metastasis through its important role in cell proliferation, survival, adhesion, spreading, migration, and invasion. FAK also plays a role in tumor immune evasion, epithelial-mesenchymal transition, DNA damage repair, radioresistance, and regulation of cancer stem cells. FAK is of particular interest in SCLC, being known for its aggressiveness. The inhibition of FAK in SCLC cell lines demonstrated significative decrease in cell proliferation, invasion, and migration, and induced cell cycle arrest and apoptosis. In this review, we will focus on the role of FAK in cancer cells and their microenvironment, and its potential as a therapeutic target in SCLC.
Collapse
Affiliation(s)
- Frank Aboubakar Nana
- Institut de Recherche Expérimentale et Clinique (IREC), Pôle de Pneumologie, ORL et Dermatologie (PNEU), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
- Division of Pneumology, Cliniques Universitaires St-Luc, UCL, 1200 Brussels, Belgium.
| | - Marie Vanderputten
- Institut de Recherche Expérimentale et Clinique (IREC), Pôle de Pneumologie, ORL et Dermatologie (PNEU), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
| | - Sebahat Ocak
- Institut de Recherche Expérimentale et Clinique (IREC), Pôle de Pneumologie, ORL et Dermatologie (PNEU), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
- Division of Pneumology, CHU UCL Namur (Godinne Site), UCL, 5530 Yvoir, Belgium.
| |
Collapse
|
13
|
Aboubakar Nana F, Hoton D, Ambroise J, Lecocq M, Vanderputten M, Sibille Y, Vanaudenaerde B, Pilette C, Bouzin C, Ocak S. Increased Expression and Activation of FAK in Small-Cell Lung Cancer Compared to Non-Small-Cell Lung Cancer. Cancers (Basel) 2019; 11:cancers11101526. [PMID: 31658694 PMCID: PMC6827365 DOI: 10.3390/cancers11101526] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Focal adhesion kinase (FAK) plays a crucial role in cancer development and progression. FAK is overexpressed and/or activated and associated with poor prognosis in various malignancies. However, in lung cancer, activated FAK expression and its prognostic value are unknown. METHODS FAK and activated FAK (phospho-FAK Y397) expressions were analyzed by multiplex immunofluorescence staining in formalin-fixed paraffin-embedded tissues from 95 non-small-cell lung cancer (NSCLC) and 105 small-cell lung cancer (SCLC) patients, and 37 healthy donors. The FAK staining score was defined as the percentage (%) of FAK-stained tumor area multiplied by (×) FAK mean intensity and phospho-FAK staining score as the (% of phospho-FAK-stained area of low intensity × 1) + (% of phospho-FAK-stained area of medium intensity × 2) + (% of the phospho-FAK-stained area of high intensity × 3). FAK and phospho-FAK staining scores were compared between normal, NSCLC, and SCLC tissues. They were also tested for correlations with patient characteristics and clinical outcomes. RESULTS The median follow-up time after the first treatment was 42.5 months and 6.4 months for NSCLC and SCLC patients, respectively. FAK and phospho-FAK staining scores were significantly higher in lung cancer than in normal lung and significantly higher in SCLC compared to NSCLC tissues (p < 0.01). Moreover, the ratio between phospho-FAK and FAK staining scores was significantly higher in SCLC than in NSCLC tissues (p < 0.01). However, FAK and activated FAK expression in lung cancer did not correlate with recurrence-free and overall survival in NSCLC and SCLC patients. CONCLUSIONS Total FAK and activated FAK expressions are significantly higher in lung cancer than in normal lung, and significantly higher in SCLC compared to NSCLC, but are not prognostic biomarkers in this study.
Collapse
Affiliation(s)
- Frank Aboubakar Nana
- Pole of Pneumology, ENT, and Dermatology (PNEU), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
- Division of Pneumology, Cliniques Universitaires St-Luc, UCLouvain, 1200 Brussels, Belgium.
| | - Delphine Hoton
- Department of Pathology, Cliniques Universitaires Saint-Luc, UCLouvain, 1200 Brussels, Belgium.
| | - Jérôme Ambroise
- Centre de Technologies Moléculaires Appliquées, IREC, UCLouvain, 1200 Brussels, Belgium.
| | - Marylène Lecocq
- Pole of Pneumology, ENT, and Dermatology (PNEU), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
| | - Marie Vanderputten
- Pole of Pneumology, ENT, and Dermatology (PNEU), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
| | - Yves Sibille
- Pole of Pneumology, ENT, and Dermatology (PNEU), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
- Division of Pneumology, CHU UCL Namur (Godinne Site), UCLouvain, 5530 Yvoir, Belgium.
| | - Bart Vanaudenaerde
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium.
| | - Charles Pilette
- Pole of Pneumology, ENT, and Dermatology (PNEU), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
- Division of Pneumology, Cliniques Universitaires St-Luc, UCLouvain, 1200 Brussels, Belgium.
| | | | - Sebahat Ocak
- Pole of Pneumology, ENT, and Dermatology (PNEU), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
- Division of Pneumology, CHU UCL Namur (Godinne Site), UCLouvain, 5530 Yvoir, Belgium.
| |
Collapse
|
14
|
Zeng Y, Cao Y, Liu L, Zhao J, Zhang T, Xiao L, Jia M, Tian Q, Yu H, Chen S, Cai Y. SEPT9_i1 regulates human breast cancer cell motility through cytoskeletal and RhoA/FAK signaling pathway regulation. Cell Death Dis 2019; 10:720. [PMID: 31558699 PMCID: PMC6763430 DOI: 10.1038/s41419-019-1947-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 08/09/2019] [Accepted: 08/26/2019] [Indexed: 02/08/2023]
Abstract
Increasing cell mobility is the basis of tumor invasion and metastasis, and is therefore a therapeutic target for preventing the spread of many types of cancer. Septins are a family of cytoskeletal proteins with GTPase activity, and play a role in many important cellular functions, including cell migration. SEPT9 isoform 1 protein (SEPT9_i1) has been associated with breast tumor development and the enhancement of cell migration; however, the exact mechanism of how SEPT9_i1 might affect breast cancer progression remains to be elucidated. Here, we report that the expression of SEPT9_i1 positively correlated with paxillin, and both were significantly upregulated in invasive breast cancer tissues of patients with lymph node metastases. Lentivirus-mediated shRNA knockdown of SEPT9 in MCF-7 cells diminished tumor cell migration, focal adhesion (FA) maturation and the expression of β-actin, β-tubulin, Cdc42, RhoA, and Rac, whereas overexpression of SEPT9_i1 in SEPT9-knockdown MCF-7 cells promoted cell migration, FA maturation and relevant protein expression. Furthermore, overexpression of SEPT9_i1 in MCF-7 cells markedly increased FAK/Src/paxillin signaling, at least in part through RhoA/ROCK1 upstream activation. Transcriptome profiling suggested that SEPT9_i1 may directly affect “Focal adhesion” and “Regulation of actin cytoskeleton” signaling mechanisms. Finally, overexpression of SEPT9_i1 markedly enhanced lung metastases in vivo 6 weeks after tumor inoculation. These findings suggest that a mechanism of Septin-9-induced aberrant cancer cell migration is through cytoskeletal regulation and FA modulation, and encourages the use of SEPT9 as novel therapeutic target in the prevention of tumor metastasis.
Collapse
Affiliation(s)
- Yongqiu Zeng
- Key Laboratory of Obstetric, Gynecologic, and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, Sichuan, China. .,Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China.
| | - Yang Cao
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Lan Liu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Jiao Zhao
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Ting Zhang
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Lifan Xiao
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Man Jia
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Qiang Tian
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Hong Yu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Shaokun Chen
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Yansen Cai
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
15
|
Andisha NM, McMillan DC, Gujam FJA, Roseweir A, Edwards J. The relationship between phosphorylation status of focal adhesion kinases, molecular subtypes, tumour microenvironment and survival in patients with primary operable ductal breast cancer. Cell Signal 2019; 60:91-99. [PMID: 30981841 DOI: 10.1016/j.cellsig.2019.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/01/2019] [Accepted: 04/10/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Despite advances in therapies to treat breast cancer, over 100,000 patients die in the UK of this disease per year, highlighting the need to develop effect predictive and prognostic markers for patients with primary operable ductal breast cancer. Therefore, the aim of the present study was to examine the relationship between membranous, cytoplasmic and nuclear expression of focal adhesion kinase (phosphorylated at Y 397, Y 861 and Y 925), molecular subtypes, tumour microenvironment and survival in patients with primary operable ductal breast cancer. METHODS Four hundred and seventy-four patients presenting between 1995 and 1998 with primary operable ductal breast cancer were included in this study. Using tissue microarrays expression of membranous, cytoplasmic and nuclear tumour cell phosphorylation of FAK at Y397, Y861 and Y925 was assessed, and associations with clinicopathological characteristics, tumour microenvironment and cancer-specific survival (CSS) were examined. RESULTS No significant association was observed for ph-FAK Y861 with survival at all sites. However, high expression of membranous ph-FAK Y397 was associated with increased tumour grade (P < .001), molecular subtypes (P < .001), increased tumour necrosis (P < .001), high Klintrup-Mäkinen grade (P < .001), increased CD138+ plasma cells (P = .031), endocrine therapy (P = .001) and poor cancer specific survival (P = .040). Similarly, high expression of nuclear ph-FAK Y397 was associated with decreased age (P = .042), increased CD138+ plasma cells (P = .001) and poor cancer specific survival (P = .003). Furthermore, high expression of cytoplasmic ph-FAK Y925 was associated with decreased tumour grade (P < .001), less involved lymph node (P = .020), molecular subtypes (P < .001), decreased tumour necrosis (P < .001), low Klintrup-Mäkinen grade (P < .001), decreased CD4+ T-cells (P = .006), decreased CD138+ plasma cells (P = .034), endocrine therapy (P < .001), chemotherapy (P = .048), and improved cancer specific survival (P = .044). On multivariate analysis, high expression of nuclear ph-FAK Y397 was independently associated with reduced cancer specific survival (P = .017). CONCLUSION The results of the present study show that membranous and nuclear ph-FAK Y397 and cytoplasmic ph-FAK Y925 were associated with prognosis in patients with primary operable ductal breast cancer. In addition, high expression of nuclear ph-FAK Y397 was an independent prognostic factor in patients with primary operable ductal breast cancer and could be incorporated into clinical practice.
Collapse
Affiliation(s)
- Najla M Andisha
- Academic Unit of Surgery, College of Medical, Veterinary and Life Sciences-University of Glasgow, Royal Infirmary, Glasgow, UK; Unit of Gastrointestinal cancer and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences-University of Glasgow, Glasgow, UK.
| | - Donald C McMillan
- Academic Unit of Surgery, College of Medical, Veterinary and Life Sciences-University of Glasgow, Royal Infirmary, Glasgow, UK
| | - Fadia J A Gujam
- Academic Unit of Surgery, College of Medical, Veterinary and Life Sciences-University of Glasgow, Royal Infirmary, Glasgow, UK; Unit of Gastrointestinal cancer and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences-University of Glasgow, Glasgow, UK
| | - Antonia Roseweir
- Unit of Gastrointestinal cancer and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences-University of Glasgow, Glasgow, UK
| | - Joanne Edwards
- Unit of Gastrointestinal cancer and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences-University of Glasgow, Glasgow, UK
| |
Collapse
|
16
|
Li W, Yu X, Ma X, Xie L, Xia Z, Liu L, Yu X, Wang J, Zhou H, Zhou X, Yang Y, Liu H. Deguelin attenuates non-small cell lung cancer cell metastasis through inhibiting the CtsZ/FAK signaling pathway. Cell Signal 2018; 50:131-141. [PMID: 30018008 DOI: 10.1016/j.cellsig.2018.07.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 06/09/2018] [Accepted: 07/05/2018] [Indexed: 01/13/2023]
Abstract
Lung cancer is the leading cause of cancer-related death among both men and women every year, mainly due to metastasis. Although natural compound deguelin has been reported to inhibited cell migration and invasion in various cancer cells, the details of this regulation progress remain to be fully elucidated. In this study, we investigated the underlying mechanism of deguelin-suppressed metastasis of non-small cell lung cancer (NSCLC) cells. Our results demonstrate that deguelin inhibits NSCLC cell migration, invasion, and metastasis both in vitro and in vivo. These inhibitory effects of deguelin were mediated by suppressing of Cathepsin Z (CtsZ) expression and interrupting the interaction of CtsZ with integrin β3. Moreover, deguelin inhibits the activation of CtsZ downstream FAK/Src/Paxillin signaling. Knockdown of CtsZ mimicked the effect of deguelin on NSCLC cells migration and invasion. Our study reveals that deguelin exerts its anti-metastatic effect both in vitro and in vivo is partly dependent on the suppression of CtsZ signaling. Deguelin would be a potential anti-metastasis agent against NSCLC.
Collapse
Affiliation(s)
- Wei Li
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Xinfang Yu
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Xiaolong Ma
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Li Xie
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhenkun Xia
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Lijun Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xinyou Yu
- Shangdong Lvdu Bio-Industry Co., Ltd., Binzhou, Shangdong 256600, China
| | - Jian Wang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Huiling Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xinmin Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Yifeng Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Haidan Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
17
|
Zhang R, Zhang TT, Zhai GQ, Guo XY, Qin Y, Gan TQ, Zhang Y, Chen G, Mo WJ, Feng ZB. Evaluation of the HOXA11 level in patients with lung squamous cancer and insights into potential molecular pathways via bioinformatics analysis. World J Surg Oncol 2018; 16:109. [PMID: 29914539 PMCID: PMC6006563 DOI: 10.1186/s12957-018-1375-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 03/26/2018] [Indexed: 01/11/2023] Open
Abstract
Background This study was carried out to discover the underlying role that HOXA11 plays in lung squamous cancer (LUSC) and uncover the potential corresponding molecular mechanisms and functions of HOXA11-related genes. Methods Twenty-three clinical paired LUSC and non-LUSC samples were utilized to examine the level of HOXA11 using quantitative real-time polymerase chain reaction (qRT-PCR). The clinical significance of HOXA11 was systematically analyzed based on 475 LUSC and 18 non-cancerous adjacent tissues from The Cancer Genome Atlas (TCGA) database. A total of 102 LUSC tissues and 121 non-cancerous tissues were available from Oncomine to explore the expressing profiles of HOXA11 in LUSC. A meta-analysis was carried out to further assess the differential expression of HOXA11 in LUSC, including in-house qRT-PCR data, expressing data extracted from TCGA and Oncomine databases. Moreover, the enrichment analysis and potential pathway annotations of HOXA11 in LUSC were accomplished via Gene Oncology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). The expression of hub genes and according correlations with HOXA11 were assessed to further explore the biological role of HOXA11 in LUSC. Results HOXA11 expression in LUSC had a tendency to be upregulated in comparison to adjacent non-cancerous tissues by qRT-PCR. TCGA data displayed that HOXA11 was remarkably over-expressed in LUSC compared with that in non-LUSC samples, and the area under curves (AUC) was 0.955 (P < 0.001). A total of 1523 co-expressed genes were sifted for further analysis. The most significant term enriched in the KEGG pathway was focal adhesion. Among the six hub genes of HOXA11, including PARVA, ILK, COL4A1, COL4A2, ITGB1, and ITGA5, five (with the exception of COL4A1) were significantly decreased compared with the normal lung tissues. Moreover, the expression of ILK was negatively related to HOXA11 (r = − 0.141, P = 0.002). Conclusion High HOXA11 expression may lead to carcinogenesis and the development of LUSC. Furthermore, co-expressed genes might affect the prognosis of LUSC.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | | | - Gao-Qiang Zhai
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xian-Yu Guo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Yuan Qin
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Ting-Qing Gan
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Yu Zhang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Wei-Jia Mo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.
| | - Zhen-Bo Feng
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.
| |
Collapse
|
18
|
Han J, Liu S, Zhang Y, Xu Y, Jiang Y, Zhang C, Li C, Li X. MiRSEA: Discovering the pathways regulated by dysfunctional MicroRNAs. Oncotarget 2018; 7:55012-55025. [PMID: 27474169 PMCID: PMC5342398 DOI: 10.18632/oncotarget.10839] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/10/2016] [Indexed: 12/17/2022] Open
Abstract
Recent studies have shown that dysfunctional microRNAs (miRNAs) are involved in the progression of various cancers. Dysfunctional miRNAs may jointly regulate their target genes and further alter the activities of canonical biological pathways. Identification of the pathways regulated by a group of dysfunctional miRNAs could help uncover the pathogenic mechanisms of cancer and facilitate development of new drug targets. Current miRNA-pathway analyses mainly use differentially-expressed miRNAs to predict the shared pathways on which they act. However, these methods fail to consider the level of differential expression level, which could improve our understanding of miRNA function. We propose a novel computational method, MicroRNA Set Enrichment Analysis (MiRSEA), to identify the pathways regulated by dysfunctional miRNAs. MiRSEA integrates the differential expression levels of miRNAs with the strength of miRNA pathway associations to perform direct enrichment analysis using miRNA expression data. We describe the MiRSEA methodology and illustrate its effectiveness through analysis of data from hepatocellular cancer, gastric cancer and lung cancer. With these analyses, we show that MiRSEA can successfully detect latent biological pathways regulated by dysfunctional miRNAs. We have implemented MiRSEA as a freely available R-based package on CRAN (https://cran.r-project.org/web/packages/MiRSEA/).
Collapse
Affiliation(s)
- Junwei Han
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, PR China
| | - Siyao Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, PR China
| | - Yunpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, PR China
| | - Yanjun Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, PR China
| | - Ying Jiang
- College of Basic Medical Science, Heilongjiang University of Chinese Medicine, Harbin 150040, PR China
| | - Chunlong Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, PR China
| | - Chunquan Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Harbin, 150081, PR China
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, PR China
| |
Collapse
|
19
|
Zhou LN, Li SC, Li XY, Ge H, Li HM. Identification of differential protein-coding gene expressions in early phase lung adenocarcinoma. Thorac Cancer 2017; 9:234-240. [PMID: 29266838 PMCID: PMC5792719 DOI: 10.1111/1759-7714.12569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/01/2017] [Accepted: 11/02/2017] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The diagnosis of early phase lung adenocarcinoma (LADC) is associated with therapeutic strategy, effect, and survival time. However, the sensitive biomarkers of early phase LADC are still unclear. This study aimed to identify protein-coding genes that can be used as biomarkers of early stage LADC. METHODS Gene microarray analysis was performed to identify key hub genes that show different expression in lung adenocarcinoma compared to normal tissues. The microarray data of lung adenocarcinoma in stages IA, IB, IIA, IIB, and normal tissues (GSE10072) were downloaded from a free online database, Gene Expression Omnibus (GEO). RESULTS A total of 572 differentially expressed genes (DEGs) were identified between early phase lung adenocarcinoma and normal tissues using R software. Database for Annotation, Visualization and Integrated Discovery online tools were used to obtain Gene Ontology analysis and the Kyoto Encyclopedia of Genes and Genomes was used to analyze DEGs. Cytoscape software was used to express the protein-protein interaction network. We found that some cancer-related Gene Ontology terms and pathways (e.g. cell adhesion, cell surface receptor signaling pathway, PI3K-Akt signaling pathway) were significantly enriched in DEGs. CONCLUSION Protein-coding genes JUN, FYN, CAV1, and SFN may play vital roles in the progress of early-stage lung adenocarcinoma. Consequently, through bioinformatics analysis, the key genes could be established to provide more potential references for the therapeutic targets of lung adenocarcinoma.
Collapse
Affiliation(s)
- Li-Na Zhou
- Department of Cancer, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shi-Cheng Li
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xue-Ying Li
- Department of Cancer, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong Ge
- Department of Cancer, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong-Mei Li
- Department of Cancer, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
20
|
Li M, Hou F, Zhao J, Zhang T, Li D, Wu W, Liu X, Xu L. Focal adhesion kinase is overexpressed in thymic epithelial tumors and may serve as an independent prognostic biomarker. Oncol Lett 2017; 15:3001-3007. [PMID: 29435030 PMCID: PMC5778861 DOI: 10.3892/ol.2017.7676] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 11/02/2017] [Indexed: 01/16/2023] Open
Abstract
Focal adhesion kinase (FAK) has long been considered to be a key regulator of growth factor receptor- and integrin-mediated signals, with pivotal roles in tumor cells through its kinase activity and scaffolding function. Increased FAK expression and activity has been observed in tumors of various origins and is often associated with a poor prognosis. However, there have been no studies on the aberrant expression of FAK in thymic epithelial tumors to date. The aim of the present study was to evaluate FAK expression in thymic epithelial tumors and to explore the prognostic significance of FAK. FAK expression was investigated in 100 formalin-fixed, paraffin-embedded human thymic epithelial tumor (TET) specimens using immunohistochemical analysis with FAK-specific monoclonal antibody 4.47, and the associations between FAK expression and clinicopathological parameters (including sex, age, tumor size, myasthenia gravis, World Health Organization classification and Masaoka-Koga stage) were analyzed. FAK was significantly overexpressed in TETs compared with in normal thymus tissues (P<0.001). Additionally, FAK overexpression was significantly associated with advanced tumor stages (stages III or IV; P<0.001) and highly aggressive TET subtypes (type B2 and B3 thymomas and thymic carcinomas; P<0.001). Furthermore, FAK overexpression was significantly associated with a worse 10-year overall survival, as determined by univariate analysis (P<0.001). Multivariate analysis revealed that FAK overexpression was an independent prognostic factor for patients with TETs (P=0.034). The results of the present study suggest that FAK serves an important role in the tumorigenesis and progression of TETs. Therefore, FAK may serve as a prognostic biomarker and is a potential therapeutic target for the treatment of TETs.
Collapse
Affiliation(s)
- Meng Li
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Feng Hou
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Jie Zhao
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Tianqiang Zhang
- Department of Cardiothoracic Surgery, The Municipal Hospital of Zaozhuang, Zaozhuang, Shandong 277100, P.R. China
| | - Dongfei Li
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Wensi Wu
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Xiaotong Liu
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Linhao Xu
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
21
|
Thanapprapasr K, Nartthanarung A, Thanapprapasr D, Jinawath A. pFAK-Y397 overexpression as both a prognostic and a predictive biomarker for patients with metastatic osteosarcoma. PLoS One 2017; 12:e0182989. [PMID: 28846700 PMCID: PMC5573209 DOI: 10.1371/journal.pone.0182989] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/27/2017] [Indexed: 11/18/2022] Open
Abstract
Focal adhesion kinase (FAK) is important for tumor cell survival and metastasis in various cancers. However, its expression and prognostic value in patients with metastatic osteosarcoma remain unknown. We investigated the expression of FAK and its phosphorylated form (pFAK-Y397) in osteosarcoma tissues from 53 patients by immunohistochemistry and evaluated their correlations with clinicopathologic characteristics and outcomes. The prognostic values were assessed using Kaplan-Meier survival and Cox regression analyses. Total FAK and pFAK-Y397 were overexpressed in 48 (90.6%) and 33 (62.3%) cases, respectively. pFAK-Y397 overexpression was correlated with poor histologic response after neoadjuvant chemotherapy in patients with osteosarcoma regardless of the presence of metastasis or not. Kaplan-Meier curve showed that patients with metastatic osteosarcoma with pFAK-Y397 overexpression had significantly worse overall survival (OS) than those with non-overexpression (P = 0.044). Multivariate Cox regression analysis confirmed pFAK-Y397 overexpression as an independent prognostic predictor for OS and post metastases OS (PMOS) (P = 0.017, P = 0.006, respectively). Age at diagnosis was also an independent indicator for PMOS (P = 0.003). However, total FAK expression was not correlated with any clinicopathologic characteristics or OS in patients with metastatic osteosarcoma. In conclusion, our findings identified FAK as a common aberrant protein overexpression in various subtypes of osteosarcoma. pFAK-Y397 overexpression can be used as a prognostic biomarker predicting poor OS for patients with metastatic osteosarcoma, and the expression of pFAK-Y397 differentiated good and poor responders to neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Kamolrat Thanapprapasr
- Biomedical Engineering Research Unit, National Metal and Materials Technology Center, National Science and Technology Development Agency, Pathumthani, Thailand
- * E-mail: (AJ); (KT)
| | - Adisak Nartthanarung
- Musculoskeletal Oncology Unit, Department of Orthopaedics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Duangmani Thanapprapasr
- Surgical Oncology Unit, Wattanosoth Cancer Hospital, Bangkok Hospital Medical Center, Bangkok, Thailand
| | - Artit Jinawath
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- * E-mail: (AJ); (KT)
| |
Collapse
|
22
|
Zhou B, Wang GZ, Wen ZS, Zhou YC, Huang YC, Chen Y, Zhou GB. Somatic Mutations and Splicing Variants of Focal Adhesion Kinase in Non–Small Cell Lung Cancer. J Natl Cancer Inst 2017; 110:4565750. [PMID: 29087503 DOI: 10.1093/jnci/djx157] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 06/30/2017] [Indexed: 12/17/2022] Open
Affiliation(s)
- Bo Zhou
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences and University of Chinese Academy of Sciences, Beijing, China
| | - Gui-Zhen Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences and University of Chinese Academy of Sciences, Beijing, China
| | - Zhe-Sheng Wen
- Department of Thoracic Surgery, the Cancer Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yong-Chun Zhou
- Department of Thoracic Surgery, the Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yun-Chao Huang
- Department of Thoracic Surgery, the Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ying Chen
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Guang-Biao Zhou
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences and University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
23
|
Chikara S, Lindsey K, Borowicz P, Christofidou-Solomidou M, Reindl KM. Enterolactone alters FAK-Src signaling and suppresses migration and invasion of lung cancer cell lines. Altern Ther Health Med 2017; 17:30. [PMID: 28068967 PMCID: PMC5223372 DOI: 10.1186/s12906-016-1512-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 12/03/2016] [Indexed: 11/26/2022]
Abstract
Background Systemic toxicity of chemotherapeutic agents and the challenges associated with targeting metastatic tumors are limiting factors for current lung cancer therapeutic approaches. To address these issues, plant-derived bioactive components have been investigated for their anti-cancer properties because many of these agents are non-toxic to healthy tissues. Enterolactone (EL) is a flaxseed-derived mammalian lignan that has demonstrated anti-migratory properties for various cancers, but EL has not been investigated in the context of lung cancer, and its anticancer mechanisms are ill-defined. We hypothesized that EL could inhibit lung cancer cell motility by affecting the FAK-Src signaling pathway. Methods Non-toxic concentrations of EL were identified for A549 and H460 human lung cancer cells by conducting 3-(4, 5-Dimethylthiazol-2-yl)-2, 5-Dephenyltetrazolium Bromide (MTT) assays. The anti-migratory and anti-invasive potential of EL for lung cancer cell lines was determined by scratch wound healing and Matrigel® invasion assays. Changes in filamentous actin (F-actin) fiber density and length in EL-treated cells were determined using phalloidin-conjugated rhodamine dye and fluorescent microscopy. Vinculin expression in focal adhesions upon EL treatment was determined by immunocytochemistry. Gene and protein expression levels of FAK-Src signaling molecules in EL-treated lung cancer cells were determined using PCR arrays, qRT-PCR, and western blotting. Results Non-toxic concentrations of EL inhibited lung cancer cell migration and invasion in a concentration- and time-dependent manner. EL treatment reduced the density and number of F-actin fibers in lung cancer cell lines, and reduced the number and size of focal adhesions. EL decreased phosphorylation of FAK and its downstream targets, Src, paxillin, and decreased mRNA expression of cell motility-related genes, RhoA, Rac1, and Cdc42 in lung cancer cells. Conclusions Our data suggest that EL suppresses lung cancer cell motility and invasion by altering FAK activity and subsequent activation of downstream proteins needed for focal adhesion formation and cytoskeletal rearrangement. Therefore, administration of EL may serve as a safe and complementary approach for inhibiting lung tumor cell motility, invasion, and metastasis. Electronic supplementary material The online version of this article (doi:10.1186/s12906-016-1512-3) contains supplementary material, which is available to authorized users.
Collapse
|
24
|
Dragoj M, Milosevic Z, Bankovic J, Tanic N, Pesic M, Stankovic T. Targeting CXCR4 and FAK reverses doxorubicin resistance and suppresses invasion in non-small cell lung carcinoma. Cell Oncol (Dordr) 2016; 40:47-62. [DOI: 10.1007/s13402-016-0304-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2016] [Indexed: 12/12/2022] Open
|
25
|
Prognostic Value of Focal Adhesion Kinase (FAK) in Human Solid Carcinomas: A Meta-Analysis. PLoS One 2016; 11:e0162666. [PMID: 27637100 PMCID: PMC5026375 DOI: 10.1371/journal.pone.0162666] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 08/26/2016] [Indexed: 01/08/2023] Open
Abstract
Background Recently, the number of reports on focal adhesion kinase (FAK) as a vital therapeutic target in solid carcinomas has increased; however, the prognostic role of FAK status remains poorly understood. This study aims to evaluate the prognostic effect of FAK by means of a meta-analysis. Methods We performed a systematic literature search in order to examine the correlation between expression of FAK and overall survival(OS). The hazard ratio (HR) of OS was used to measure survival. A random-effects model was used to pool study statistics. Sensitivity and publication bias analyses were also conducted. Results Thirty eligible studies involving 4702 patients were included. The median expression rate of FAK was 54%. Meta-analysis of the HRs demonstrated that high FAK expression was associated with worse OS (average HR = 2.073, 95%confidence interval[CI]:1.712–2.510, p = 0.000). Regarding cancer type, FAK was associated with worse OS in gastric cancer (HR = 2.646,95% CI:1.743–4.017, p = 0.000), hepatocellular carcinoma (HR = 1.788,95% CI:1.228–2.602, p = 0.002), ovarian cancer (HR = 1.815, 95% CI: 1.193–2.762, p = 0.005), endometrial cancer (HR = 4.149, 95% CI:2.832–6.079, p = 0.000), gliomas (HR = 2.650, 95% CI: 1.205–5.829, p = 0.015), and squamous cell carcinoma (HR = 1,696, 95% CI: 1.030–2.793, p = 0.038). No association was found between HR and disease staging according to our meta-regression analysis. Conclusions Our study shows that high expression of FAK is associated with a worse OS in patients with carcinomas, but the association between FAK and prognosis varies according to cancer type. The value of FAK status in clinical prognosis in cancer needs further research.
Collapse
|
26
|
Pawełczyk K, Marciniak M, Błasiak P. Evaluation of new classifications of N descriptor in non-small cell lung cancer (NSCLC) based on the number and the ratio of metastatic lymph nodes. J Cardiothorac Surg 2016; 11:68. [PMID: 27079794 PMCID: PMC4832480 DOI: 10.1186/s13019-016-0456-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 04/05/2016] [Indexed: 11/17/2022] Open
Abstract
Background The aim of the study was to evaluate the prognostic power of new classifications of N descriptor created basing on the number (NLN) and the ratio of metastatic lymph nodes (RLN) in NSCLC compared to the current classification (CLN). Methods The data of 529 patients with NSCLC operated with the intention of radical resection, were analyzed. The new categories of N descriptor were created as follows: 1) NLN - median number of metastatic nodes was 3, thus in NLN0 the number of metastatic nodes =0, in NLN1 1-2, in NLN2 ≥ 3, 2) RLN - median ratio (number of metastatic lymph nodes to all nodes removed) was 12.4 %, thus in RLN0 the ratio was 0, in RLN1 < 13 %, in RLN2 > 13 %. The prognostic value of each classification was calculated on the basis of hazard ratios defined in multivariate Cox proportional hazard model. Results The new classifications of N descriptor turned out to be an independent strong prognostic factor (p <0.001) with a 5-year survival rate NLN0-62 %, NLN1-39 %, NLN2-26 % and RLN0-62 %, RLN1-37 % and RLN2-26 %. For 5-year survival rates in CLN0-62 %, CLN1-42 %, CLN2-24 % (p < 0.001), a higher prognostic value of new classifications was not demonstrated, the hazard ratio amounted to 2.22, 2.08, 2.49 for NLN2, RLN2 and CLN2 respectively. Conclusion Despite the significantly high prognostic power, the new classifications cannot be considered superior over CLN. There are some deficiencies in the current classification, therefore further studies on its improvement are needed.
Collapse
Affiliation(s)
- Konrad Pawełczyk
- Department of General Thoracic Surgery, Wroclaw Thoracic Surgery Centre, Wroclaw Medical University, Wroclaw, Poland.
| | - Marek Marciniak
- Department of General Thoracic Surgery, Wroclaw Thoracic Surgery Centre, Wroclaw Medical University, Wroclaw, Poland
| | - Piotr Błasiak
- Department of General Thoracic Surgery, Wroclaw Thoracic Surgery Centre, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
27
|
KANG YU, PU TAO, CAI QINGQING, HONG SHANSHAN, ZHANG MINGXING, LI GUILING, ZHU ZHILING, XU CONGJIAN. Identification of lymphatic metastasis-associated genes in a metastatic ovarian cancer cell line. Mol Med Rep 2015; 12:2741-8. [DOI: 10.3892/mmr.2015.3743] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 03/24/2015] [Indexed: 11/06/2022] Open
|
28
|
Hung CH, Huang CC, Hsu LS, Kao SH, Wang CJ. Apple polyphenol inhibits colon carcinoma metastasis via disrupting Snail binding to focal adhesion kinase. J Funct Foods 2015. [DOI: 10.1016/j.jff.2014.10.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
29
|
Dy GK, Ylagan L, Pokharel S, Miller A, Brese E, Bshara W, Morrison C, Cance WG, Golubovskaya VM. The prognostic significance of focal adhesion kinase expression in stage I non-small-cell lung cancer. J Thorac Oncol 2014; 9:1278-84. [PMID: 25122425 PMCID: PMC4133746 DOI: 10.1097/jto.0000000000000248] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Focal adhesion kinase (FAK) plays a significant role in cancer cell survival signaling and is overexpressed in various malignancies, including lung cancer. Previous studies suggest that FAK overexpression is an independent factor predicting poor prognosis in non-small-cell lung cancer (NSCLC). The aim of this study is to confirm these findings specifically in stage I NSCLC. METHODS A retrospective tissue microarray (TMA) analysis of FAK protein expression by immunohistochemistry was performed in 157 surgically resected stage I NSCLC specimen and in the corresponding matched normal lung tissue. The FAK 4.47 monoclonal antibody was used for FAK immunostaining. The scoring system of triplicate tumor cores included intensity of staining plus extent of staining for a composite score that ranged from 0 to 6. The association between FAK score and survival was evaluated. RESULTS There were 103 stage IA and 54 stage IB patients, with mean follow-up of 5.5 years. Normal lung alveoli and interstitial tissue had mean FAK score of 0 (median score 0, range 0 to 2). Tumor samples had mean FAK score 3.1 (median score 3.5, range 0-6), with 57% of the samples having FAK score ≥ 3. Continuous FAK score was not associated with demographic data, tumor histology, or grade, nor survival in this cohort of stage I NSCLC patients. CONCLUSIONS FAK is expressed in more than 50% of stage I NSCLC lung cancer but not in normal lung alveoli and interstitial tissue. FAK expression is not associated with survival outcome in this North American cohort.
Collapse
Affiliation(s)
- Grace K. Dy
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, 14263 USA
| | - Lourdes Ylagan
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, 14263 USA
| | - Saraswati Pokharel
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, 14263 USA
| | - Austin Miller
- Department of Biostatistics, Roswell Park Cancer Institute, Buffalo, NY, 14263USA
| | - Elizabeth Brese
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, 14263 USA
| | - Wiam Bshara
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, 14263 USA
| | - Carl Morrison
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, 14263 USA
| | - William G. Cance
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY, 14263 USA
| | - Vita M. Golubovskaya
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY, 14263 USA
| |
Collapse
|
30
|
HU YONG, XU SHENGLIN, JIN WENSEN, YI QIYI, WEI WEI. Effect of the PTEN gene on adhesion, invasion and metastasis of osteosarcoma cells. Oncol Rep 2014; 32:1741-7. [PMID: 25069680 DOI: 10.3892/or.2014.3362] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 07/04/2014] [Indexed: 11/05/2022] Open
|
31
|
Alexopoulou AN, Ho-Yen CM, Papalazarou V, Elia G, Jones JL, Hodivala-Dilke K. Tumour-associated endothelial-FAK correlated with molecular sub-type and prognostic factors in invasive breast cancer. BMC Cancer 2014; 14:237. [PMID: 24693876 PMCID: PMC3997837 DOI: 10.1186/1471-2407-14-237] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 03/25/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Breast cancer is a heterogeneous disease that can be classified into one of 4 main molecular sub-types: luminal A, luminal B, Her2 over-expressing and basal-like (BL). These tumour sub-types require different treatments and have different risks of disease progression. BL cancers can be considered a sub-group of Triple negative (TN) cancers since they lack estrogen (ER), progesterone (PR) and Her2 expression. No targeted treatment currently exists for TN/BL cancers. Thus it is important to identify potential therapeutic targets and describe their relationship with established prognostic factors. Focal adhesion kinase (FAK) is upregulated in several human cancers and also plays a functional role in tumour angiogenesis. However, the association between breast cancer sub-types and tumour endothelial-FAK expression is unknown. METHODS Using immunofluorescence, we quantified FAK expression in tumour endothelial and tumour cell compartments in 149 invasive breast carcinomas and correlated expression with clinical, pathological and molecular parameters. RESULTS Low endothelial-FAK expression was independently associated with luminal A tumours at univariate (p < 0.001) and multivariate (p = 0.001) analysis. There was a positive correlation between FAK expression in the vascular and tumour cell compartments (Spearman's correlation co-efficient = 0.394, p < 0.001). Additionally, endothelial and tumour cell FAK expression were significantly increased in TN tumours (p = 0.043 and p = 0.033 respectively), in tumours with negative ER and PR status, and in high grade tumours at univariate analysis. CONCLUSION Our findings establish a relationship between endothelial-FAK expression levels and the molecular sub-type of invasive breast cancer, and suggest that endothelial-FAK expression is potentially more clinically relevant than tumour cell FAK expression in breast cancer.
Collapse
Affiliation(s)
- Annika N Alexopoulou
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Biology, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Colan M Ho-Yen
- Breast Group, Centre for Tumour Biology, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Vassilis Papalazarou
- Vascular Adhesion Lab, BSRC Al. Fleming, 34 Fleming str., 166 72 Vari Athens, Greece
| | - George Elia
- Breast Group, Centre for Tumour Biology, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - J Louise Jones
- Breast Group, Centre for Tumour Biology, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Kairbaan Hodivala-Dilke
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Biology, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
32
|
Lee HW, Park YM, Lee SJ, Cho HJ, Kim DH, Lee JI, Kang MS, Seol HJ, Shim YM, Nam DH, Kim HH, Joo KM. Alpha-smooth muscle actin (ACTA2) is required for metastatic potential of human lung adenocarcinoma. Clin Cancer Res 2013; 19:5879-89. [PMID: 23995859 DOI: 10.1158/1078-0432.ccr-13-1181] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Metastatic relapse of primary lung cancer leads to therapeutic resistance and unfavorable clinical prognosis; therefore, identification of key molecules associated with metastatic conversion has significant clinical implications. We previously identified a link between early brain metastasis of lung adenocarcinoma and amplification of the α-smooth muscle actin (ACTA2) gene. The aim of present study was to investigate the prognostic and functional significance of ACTA2 expression in cancer cells for the metastatic potential of lung adenocarcinomas. EXPERIMENTAL DESIGN ACTA2 expression was analyzed in tumor cells from 263 patients with primary lung adenocarcinomas by immunohistochemistry, and was correlated with clinicopathologic parameters. The expression of ACTA2 in human lung adenocarcinoma cells was modulated with short hairpin RNAs (shRNA) and siRNAs specifically targeting ACTA2. RESULTS The patients with lung adenocarcinomas with high ACTA2 expression in tumor cells showed significantly enhanced distant metastasis and unfavorable prognosis. ACTA2 downregulation remarkably impaired in vitro migration, invasion, clonogenicity, and transendothelial penetration of lung adenocarcinoma cells without affecting proliferation. Consistent with the in vitro results, depletion of ACTA2 in human lung adenocarcinoma PC14PE6 cells significantly reduced their metastatic potential without altering their tumorigenic potential. Expression of c-MET and FAK in lung adenocarcinoma cells was also reduced by ACTA2-targeting siRNAs and shRNAs, and was accompanied by a loss of mesenchymal characteristics. CONCLUSIONS These findings indicate that ACTA2 regulates c-MET and FAK expression in lung adenocarcinoma cells, which positively and selectively influence metastatic potential. Therefore, ACTA2 could be a promising prognostic biomarker and/or therapeutic target for metastatic lung adenocarcinoma.
Collapse
Affiliation(s)
- Hye Won Lee
- Authors' Affiliations: Cancer Stem Cell Research Center and Department of Neurosurgery, Samsung Medical Center; Graduate School, Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University; Samsung Biomedical Research Institute, Department of Anatomy and Cell Biology, Department of Molecular Cell Biology, Center for Genome Research, and Department of Thoracic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|