1
|
Alam M, Hasan GM, Eldin SM, Adnan M, Riaz MB, Islam A, Khan I, Hassan MI. Investigating regulated signaling pathways in therapeutic targeting of non-small cell lung carcinoma. Biomed Pharmacother 2023; 161:114452. [PMID: 36878052 DOI: 10.1016/j.biopha.2023.114452] [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: 01/19/2023] [Revised: 02/19/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Non-small cell lung carcinoma (NSCLC) is the most common malignancy worldwide. The signaling cascades are stimulated via genetic modifications in upstream signaling molecules, which affect apoptotic, proliferative, and differentiation pathways. Dysregulation of these signaling cascades causes cancer-initiating cell proliferation, cancer development, and drug resistance. Numerous efforts in the treatment of NSCLC have been undertaken in the past few decades, enhancing our understanding of the mechanisms of cancer development and moving forward to develop effective therapeutic approaches. Modifications of transcription factors and connected pathways are utilized to develop new treatment options for NSCLC. Developing designed inhibitors targeting specific cellular signaling pathways in tumor progression has been recommended for the therapeutic management of NSCLC. This comprehensive review provided deeper mechanistic insights into the molecular mechanism of action of various signaling molecules and their targeting in the clinical management of NSCLC.
Collapse
Affiliation(s)
- Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Sayed M Eldin
- Center of Research, Faculty of Engineering, Future University in Egypt, New Cairo 11835, Egypt
| | - Mohd Adnan
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Muhammad Bilal Riaz
- Faculty of Applied Physics and Mathematics, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdnask, Poland; Department of Computer Science and Mathematics, Lebanese American University, Byblos, Lebanon
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ilyas Khan
- Department of Mathematics, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
2
|
Wang X, Yu G, Yan Z, Wan L, Wang W, Cui L. Lung Cancer Subtype Diagnosis by Fusing Image-Genomics Data and Hybrid Deep Networks. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:512-523. [PMID: 34855599 DOI: 10.1109/tcbb.2021.3132292] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Accurate diagnosis of cancer subtypes is crucial for precise treatment, because different cancer subtypes are involved with different pathology and require different therapies. Although deep learning techniques have made great success in computer vision and other fields, they do not work well on Lung cancer subtype diagnosis, due to the distinction of slide images between different cancer subtypes is ambiguous. Furthermore, they often over-fit to high-dimensional genomics data with limited samples, and do not fuse the image and genomics data in a sensible way. In this paper, we propose a hybrid deep network based approach LungDIG for Lung cancer subtype Diagnosis by fusing Image-Genomics data. LungDIG first tiles the tissue slide image into small patches and extracts the patch-level features by fine-tuning an Inception-V3 model. Since the patches may contain some false positives in non-diagnostic regions, it further designs a patch-level feature combination strategy to integrate the extracted patch features and maintain the diversity between different cancer subtypes. At the same time, it extracts the genomics features from Copy Number Variation data by an attention based nonlinear extractor. Next, it fuses the image and genomics features by an attention based multilayer perceptron (MLP) to diagnose cancer subtype. Experiments on TCGA lung cancer data show that LungDIG can not only achieve higher accuracy for cancer subtype diagnosis than state-of-the-art methods, but also have a high authenticity and good interpretability.
Collapse
|
3
|
Li F, Gu F, Li Q, Zhai C, Gong R, Zhu X. ROR1-AS1 knockdown inhibits growth and invasion and promotes apoptosis in NSCLC cells by suppression of the PI3K/Akt/mTOR pathway. J Biochem Mol Toxicol 2021; 35:e22726. [PMID: 33491326 DOI: 10.1002/jbt.22726] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 11/26/2020] [Accepted: 01/09/2021] [Indexed: 11/07/2022]
Abstract
The role of ROR1-AS1 in non-small-cell lung cancer (NSCLC) remains unclear. Therefore, we aimed to investigate the functional role of ROR1-AS1 in NSCLC and to explore the underlying mechanisms. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide assay was performed to detect cell proliferation. Transwell assay was performed to evaluate cell invasive ability. Cell apoptotic rates and caspase-3/7 activity were determined to evaluate apoptosis. The expression levels of PI3K/Akt/mTOR pathway-related proteins were measured using Western blot analysis. Results showed that ROR1-AS1 expression was upregulated in NSCLC samples. Knockdown of ROR1-AS1 inhibited the viability and invasive ability of NSCLC cells. Knockdown of ROR1-AS1 induced apoptotic rate and caspase-3/7 activity and suppressed xenograft NSCLC tumor growth. In addition, ROR1-AS1 knockdown inhibited the activation of the PI3K/Akt/mTOR pathway in NSCLC cells. However, treatment with 740Y-P prevented the effects of si-ROR1-AS1 on viability, invasive ability, and apoptosis of NSCLC cells. These findings implied that ROR1-AS1 played an oncogenic role in NSCLC via regulating the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Fengbo Li
- Department of Respiratory Medicine, Nanshi Hospital, Nanyang, China
| | - Fengming Gu
- Department of Intensive Care, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an, China
| | - Qian Li
- Department of Respiratory Medicine, Nanshi Hospital, Nanyang, China
| | - Chaoshuan Zhai
- Department of Respiratory Medicine, Nanshi Hospital, Nanyang, China
| | - Rui Gong
- Department of Respiratory Medicine, Nanshi Hospital, Nanyang, China
| | - Xuezhuan Zhu
- Department of Intensive Care, Lianshui People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huai'an, China
| |
Collapse
|
4
|
Liu Y, Tian Y, Cai W, Guo Y, Xue C, Wang J. DHA/EPA-Enriched Phosphatidylcholine Suppresses Tumor Growth and Metastasis via Activating Peroxisome Proliferator-Activated Receptor γ in Lewis Lung Cancer Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:676-685. [PMID: 33406839 DOI: 10.1021/acs.jafc.0c06890] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
In the present study, the antitumor effects of docosahexaenoic acid-phosphatidylcholine (DHA-PC) and eicosapentanoic acid-phosphatidylcholine (EPA-PC) in Lewis lung cancer mice were investigated. As observed, DHA-PC and EPA-PC obviously inhibited the transplanted tumor growth and the positive expression of Ki67. The metastatic nodules and hematoxylin and eosin (HE) staining of the lung indicated that DHA-PC and EPA-PC suppressed lung metastasis. PPARγ has a key role in cell survival, which may be a target for cancer therapy. Further mechanism research indicated that DHA-PC and EPA-PC significantly enhanced the levels of PPARγ and subsequently downregulated the NF-κB pathway. DHA-PC and EPA-PC accelerate cancer cell apoptosis by decreasing NF-κB-mediated antiapoptotic factors Bcl-2 and Bcl-XL, thereby inhibiting tumor growth. In addition, DHA-PC and EPA-PC significantly decreased the levels of NF-κB-mediated matrix metallopeptidase 9 (MMP9) and heparanase (HPA), which block the extracellular matrix (ECM) degradation, thereby suppressing lung metastasis. These findings suggested that DHA-PC and EPA-PC could be used as food supplements and/or functional ingredients for cancer patients.
Collapse
Affiliation(s)
- Yuanyuan Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, China
| | - Yingying Tian
- Marine Biomedical Research Institute of Qingdao, Qingdao 266061, Shandong, China
| | - Weizhen Cai
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, China
| | - Yao Guo
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, China
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao 266003, Shandong, China
| | - Jingfeng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, China
| |
Collapse
|
5
|
Guerriero I, Monaco G, Coppola V, Orlacchio A. Serum and Glucocorticoid-Inducible Kinase 1 (SGK1) in NSCLC Therapy. Pharmaceuticals (Basel) 2020; 13:ph13110413. [PMID: 33266470 PMCID: PMC7700219 DOI: 10.3390/ph13110413] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/21/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) remains the most prevalent and one of the deadliest cancers worldwide. Despite recent success, there is still an urgent need for new therapeutic strategies. It is also becoming increasingly evident that combinatorial approaches are more effective than single modality treatments. This review proposes that the serum and glucocorticoid-inducible kinase 1 (SGK1) may represent an attractive target for therapy of NSCLC. Although ubiquitously expressed, SGK1 deletion in mice causes only mild defects of ion physiology. The frequent overexpression of SGK1 in tumors is likely stress-induced and provides a therapeutic window to spare normal tissues. SGK1 appears to promote oncogenic signaling aimed at preserving the survival and fitness of cancer cells. Most importantly, recent investigations have revealed the ability of SGK1 to skew immune-cell differentiation toward pro-tumorigenic phenotypes. Future studies are needed to fully evaluate the potential of SGK1 as a therapeutic target in combinatorial treatments of NSCLC. However, based on what is currently known, SGK1 inactivation can result in anti-oncogenic effects both on tumor cells and on the immune microenvironment. A first generation of small molecules to inactivate SGK1 has already been already produced.
Collapse
Affiliation(s)
- Ilaria Guerriero
- Biogem Institute for Genetic Research Gaetano Salvatore, Ariano Irpino, 83031 Avellino, Italy; (I.G.); (G.M.)
| | - Gianni Monaco
- Biogem Institute for Genetic Research Gaetano Salvatore, Ariano Irpino, 83031 Avellino, Italy; (I.G.); (G.M.)
| | - Vincenzo Coppola
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: (V.C.); (A.O.); Tel.: +1-614-688-8038 (V.C.); +1-646-552-0641 (A.O.)
| | - Arturo Orlacchio
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: (V.C.); (A.O.); Tel.: +1-614-688-8038 (V.C.); +1-646-552-0641 (A.O.)
| |
Collapse
|
6
|
NF-κB inhibitors in treatment and prevention of lung cancer. Biomed Pharmacother 2020; 130:110569. [PMID: 32750649 DOI: 10.1016/j.biopha.2020.110569] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/12/2020] [Accepted: 07/26/2020] [Indexed: 12/27/2022] Open
Abstract
Intracellular signalling pathways have provided excellent resource for drug development particularly in the development of cancer therapeutics. A wide variety of malignancies common in human exhibit aberrant NF-κB constitutive expression which results in tumorigenic processes and cancer survival in a variety of solid tumour, including pancreatic cancer, lung, cervical, prostate, breast and gastric carcinoma. Numerous evidences indicate that NF-κB signalling mechanism is mainly involved in the progression of several cancers which may intensify an enhanced knowledge on its role in disease particularly lung tumorigenesis. This has led to tremendous research in designing a variety of NF-κB antagonists with enhanced clinical applications through different approaches the most common being suppression of IκB kinase (IKK) beta activity. Many NF-κB inhibitors for lung cancer are now under clinical trials. Preliminary results of clinical trials for several of these agents include small-molecule inhibitors and monoclonal antibodies. A few combinatorial treatment therapies are currently under investigation in the clinics and have shown promise, particularly NF-κB inhibition associated with lung cancer.
Collapse
|
7
|
Shi H, Ji Y, Li W, Zhong Y, Ming Z. PDLIM2 acts as a cancer suppressor gene in non-small cell lung cancer via the down regulation of NF-κB signaling. Mol Cell Probes 2020; 53:101628. [PMID: 32621848 DOI: 10.1016/j.mcp.2020.101628] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/17/2020] [Accepted: 06/30/2020] [Indexed: 12/13/2022]
Abstract
PDZ and LIM domain containing protein 2 (PDLIM2) has been identified as a vital tumor-associated gene that is aberrantly expressed in various types of tumors. Yet, the involvement of PDLIM2 in non-small cell lung cancer (NSCLC) is currently undetermined. The design of the current study was to evaluate whether PDLIM2 plays a role in NSCLC. We found that PDLIM2 expression was commonly decreased in NSCLC tissues. Moreover, low expression of PDLIM2 was also detected in NSCLC cell lines and demethylation treatment restored PDLIM2 expression. The re-expression of PDLIM2 impeded the proliferative, colony-forming, and invasive capabilities of NCLCL cells. In contrast, depletion of PDLIM2 markedly enhanced the malignant behaviors of NSCLC cells. Notably, PDLIM2 overexpression downregulated the expression of nuclear factor (NF)-κB p65 subunit and repressed NF-κB transcription reporter activity in NSCLC cells. The overexpression of p65 significantly reversed PDLIM2-mediated antitumor effects in NSCLC cells. Additionally, the Xenograft tumor formation assay revealed that the overexpression of PDLIM2 markedly restricted the tumor growth of NSCLC in vivo. Overall, our study confirms that PDLIM2 acts as a tumor-inhibitor in NSCLC through the inactivation of NF-κB, suggesting PDLIM2 as a candidate therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Hongyang Shi
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, PR China.
| | - Yuqiang Ji
- Department of Cardiovascular Disease, Xi'an No.1 Hospital, Xi'an, 710002, Shaanxi Province, PR China
| | - Wei Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, PR China
| | - Yujie Zhong
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, PR China
| | - Zongjuan Ming
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, PR China
| |
Collapse
|
8
|
Targeting NF-κB-mediated inflammatory pathways in cisplatin-resistant NSCLC. Lung Cancer 2019; 135:217-227. [PMID: 31446998 DOI: 10.1016/j.lungcan.2019.07.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/25/2019] [Accepted: 07/07/2019] [Indexed: 12/31/2022]
Abstract
OBJECTIVES The majority of patients with non-small cell lung cancer (NSCLC) present with advanced stage disease, at which time chemotherapy is usually the most common treatment option. While somewhat effective, patients treated with platinum-based regimens will eventually develop resistance, with others presenting with intrinsic resistance. Multiple pathways have been implicated in chemo-resistance, however the critical underlying mechanisms have yet to be elucidated. The aim of this project was to determine the role of inflammatory mediators in cisplatin-resistance in NSCLC. MATERIALS AND METHODS Inflammatory mediator, NF-κB, and its associated pathways were investigated in an isogenic model of cisplatin-resistant NSCLC using age-matched parental (PT) and corresponding cisplatin-resistant (CisR) sublines. Pathways were assessed using mass spectrometry, western blot analysis and qRT-PCR. The cisplatin sensitizing potential of an NF-κB small molecule inhibitor, DHMEQ, was also assessed by means of viability assays and western blot analysis. RESULTS Proteomic analysis identified dysregulated NF-κB responsive targets in CisR cells when compared to PT cells, with increased NF-κB expression identified in four out of the five NSCLC sub-types examined (CisR versus PT). DHMEQ treatment resulted in reduced NF-κB expression in the presence of cisplatin, and re-sensitized CisR cells to the cytotoxic effects of the drug. CONCLUSION This study identified NF-ĸB as a potential therapeutic target in cisplatin-resistant NSCLC. Furthermore, inhibition of NF-ĸB using DHMEQ re-sensitized chemo-resistant cells to cisplatin treatment.
Collapse
|
9
|
Mervai Z, Reszegi A, Miklya I, Knoll J, Schaff Z, Kovalszky I, Baghy K. Inhibitory Effect of (2R)-1-(1-Benzofuran-2-yl)-N-propylpentan-2-amine on Lung Adenocarcinoma. Pathol Oncol Res 2019; 26:727-734. [PMID: 30734151 PMCID: PMC7242259 DOI: 10.1007/s12253-019-00603-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 01/15/2019] [Indexed: 12/11/2022]
Abstract
BPAP is a potent enhancer substance with catecholaminergic and serotoninergic activity in the brain. It was discovered that it is also effective against certain types of experimental cancers, showing the most promising results in case of lung cancer. That is why we tested its efficacy in two different doses in a newly developed EGFR wild type mouse lung adenocarcinoma xenograft model. Experiments were conducted on FVB/N and SCID mouse strains treated with low and high dose of BPAP. Body weight, survival, and tumor volumes were recorded. Furthermore, the activity of major signaling pathways of NSCLC such as MAPK and Akt/mTOR as well as cell cycle regulation were determined. Significant inhibition of tumor growth was exerted by both doses, but the mechanism of action was different. High dose directly inhibited, whereas low dose activated the main signaling pathways. Exposure to low dose BPAP resulted in elevated activity of the mTOR pathway together with p16INK-induced cell cycle arrest, a typical feature of geroconversion, a senescent state characterized by loss of cell proliferation. Finally the events culminated in cell cycle inhibition point in case of both doses mirrored by the decrease of cyclin D1, CDK4 and PCNA. In addition, BPAP treatment had a beneficial effect on bodyweight suggesting that the compound at least in part is able to compensate the cancer-related wasting. In view of the low toxicity and confirmed antitumor effect of BPAP against experimental lung adenocarcinoma, this novel compound deserves further attention.
Collapse
Affiliation(s)
- Zsolt Mervai
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26., Budapest, H-1085, Hungary
| | - Andrea Reszegi
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26., Budapest, H-1085, Hungary
| | - Ildikó Miklya
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - József Knoll
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zsuzsa Schaff
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Ilona Kovalszky
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26., Budapest, H-1085, Hungary
| | - Kornélia Baghy
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26., Budapest, H-1085, Hungary.
| |
Collapse
|
10
|
Zhang J, Guan X, Liang N, Li S. Estrogen-related receptor alpha triggers the proliferation and migration of human non-small cell lung cancer via interleukin-6. Cell Biochem Funct 2018; 36:255-262. [PMID: 29862528 DOI: 10.1002/cbf.3337] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/23/2018] [Accepted: 05/11/2018] [Indexed: 12/27/2022]
Abstract
Human non-small cell lung cancer (NSCLC) is one of the leading causes of cancer deaths worldwide. Estrogenic signals have been suggested to be important for the growth and metastasis of NSCLC cells. Our present data showed that estrogen-related receptor alpha (ERRα), while not ERRβ or ERRγ, was significantly elevated in NSCLC cell lines as compared with that in normal bronchial epithelial cell line BEAS-2B. The expression of ERRα in clinical NSCLC tissues was significantly greater than that in their matched normal adjacent tissues. Over expression of ERRα can trigger the proliferation, migration, and invasion of NSCLC cells, while si-ERRα or ERRα inhibitor showed opposite effects. ERRα can increase the mRNA and protein expression of IL-6, while not IL-8, IL-10, IL-22, VEGF, TGF-β, or TNF-α, in NSCLC cells. Silence of IL-6 attenuated ERRα induced proliferation and cell invasion. Furthermore, our data revealed the inhibition of NF-κB, while not ERK1/2 or PI3K/Akt, abolished ERRα induced production of IL-6. This might be due to that overexpression of ERRα can increase the expression and nuclear translocation of p65 in NSCLC cells. Collectively, our data showed that activation of NF-κB/IL-6 is involved in ERRα induced migration and invasion of NSCLC cells. It suggested that ERRα might be a potential target for NSCLC treatment.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Oncology in First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Xiangqian Guan
- Laboratory of the First Affiliated Hospital of University of Science and Technology of China, China
| | - Naixin Liang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
11
|
A Bowman-Birk type chymotrypsin inhibitor peptide from the amphibian, Hylarana erythraea. Sci Rep 2018; 8:5851. [PMID: 29643444 PMCID: PMC5895817 DOI: 10.1038/s41598-018-24206-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 03/28/2018] [Indexed: 01/10/2023] Open
Abstract
The first amphibian skin secretion-derived Bowman-Birk type chymotrypsin inhibitor is described here from the Asian green frog, Hylarana erythraea, and was identified by use of molecular cloning and tandem mass spectrometric amino acid sequencing. It was named Hylarana erythraea chymotrypsin inhibitor (HECI) and in addition to inhibition of chymotrypsin (Ki = 3.92 ± 0.35 μM), the peptide also inhibited the 20 S proteasome (Ki = 8.55 ± 1.84 μM). Additionally, an analogue of HECI, named K9-HECI, in which Phe9 was substituted by Lys9 at the P1 position, was functional as a trypsin inhibitor. Both peptides exhibited anti-proliferation activity against the human cancer cell lines, H157, PC-3 and MCF-7, up to a concentration of 1 mM and possessed a low degree of cytotoxicity on normal cells, HMEC-1. However, HECI exhibited higher anti-proliferative potency against H157. The results indicate that HECI, inhibiting chymotryptic-like activity of proteasome, could provide new insights in treatment of lung cancer.
Collapse
|
12
|
Liu M, Chen YL, Kuo YH, Lu MK, Liao CC. Aqueous extract of Sapindus mukorossi induced cell death of A549 cells and exhibited antitumor property in vivo. Sci Rep 2018; 8:4831. [PMID: 29555954 PMCID: PMC5859273 DOI: 10.1038/s41598-018-23096-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 03/02/2018] [Indexed: 12/15/2022] Open
Abstract
Sapindus mukorossi is a deciduous plant and has recently been recognized to have anticancer property. In the present study, we discovered that S. mukorossi leaf and stem aqueous extract (SaM) contained two polysaccharides mainly made of myo-inositol, galactose, glucose, and fructose and the aim of this study was to investigate the antitumor property the aqueous extract SaM. In vitro treatment of SaM diminished proliferative potential of lung adenocarcinomic cells and induced intracellular oxidative stress, as well as necrotic cell death. Moreover, exposure to SaM attenuated cell migration, demonstrating the effectiveness at reducing invasive property of malignant lung cells. Gene and protein expression studies indicated that SaM treatment altered the expression of proliferation/survival modulator NF-κB, tumor growth modulator ERK2, metastasis-associated molecules MMP9/12, and tumor suppressor p53 in A549 cells. Using model animals bearing Lewis lung cancer cell LL/2, we demonstrated that SaM was antitumoral and did not induce any undesired organ damage, immunotoxicity, and off-target inflammation. This work, to our knowledge, is the first study documents the antitumor bioactivity of aqueous extract riched in polysaccharides from S. mukorossi and provides insights into the potential pharmacological application of SaM as antitumor agent against lung cancer.
Collapse
Affiliation(s)
- Min Liu
- Department of Life Science, Chinese Culture University, Taipei, Republic of China. .,Graduate Institute of Biotechnology, Chinese Culture University, Taipei, Republic of China.
| | - Yen-Lin Chen
- Department of Life Science, Chinese Culture University, Taipei, Republic of China
| | - Yao-Haur Kuo
- Ministry of Health and Welfare, National Research Institute of Chinese Medicine, Taipei, Republic of China.,Graduate Institute of Integrated Medicine, China Medial University, Taichung, Republic of China.,Department of Horticulture and Biotechnology, College of Agriculture, Chinese Culture University, Taipei, Republic of China
| | - Mei-Kuang Lu
- Ministry of Health and Welfare, National Research Institute of Chinese Medicine, Taipei, Republic of China
| | - Chia-Ching Liao
- Department of Biology, Chinese Culture University, Taipei, Republic of China
| |
Collapse
|
13
|
Li J, Li Y, Wang B, Ma Y, Chen P. Id-1 promotes migration and invasion of non-small cell lung cancer cells through activating NF-κB signaling pathway. J Biomed Sci 2017; 24:95. [PMID: 29233161 PMCID: PMC5727929 DOI: 10.1186/s12929-017-0400-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/01/2017] [Indexed: 12/21/2022] Open
Abstract
Background Numerous studies have shown that Id-1 (Inhibitor of differentiation 1) is upregulated in several cancers and associated with tumor malignant characters. However, the clinical significance and biological role of Id-1 in non-small cell lung cancer (NSCLC) remains unclear. Methods We used RT-PCR, Western blot and Immunohistochemistry to measure Id-1 expression in NSCLC tissues and matched adjacent noncancerous tissues. The expression pattern of Id-1 in NSCLC tissues was determined by scoring system of immunohistochemical analysis. The Kaplan-Meier method was used to calculate the survival curve, and log-rank test to determine statistical significance. The Id-1 gene was overexpressed or downreuglated with Lentiviral vectors in NSCLC cells. And, the migration ability of NSCLC cells was tested in a Transwell Boyden Chamber. Results We found that Id-1 is generally expressed higher in NSCLC tissues compared with matched adjacent noncancerous tissues. We also found that high Id-1 expression in tumor tissues is significantly correlated with tumor progression and poor survival in NSCLC patients. Furthermore, our experimental data revealed that knockdown of Id-1 significantly suppressed the proliferation, migration and invasion of NSCLC cells, whereas ectopic expression of Id-1 promoted the malignant phenotype of NSCLC cells. Mechanistic study showed that NF-κB signaling pathway contributed to the effects of Id-1 in NSCLC cells. Moreover, blocking the NF-κB pathway significantly inhibited the tumor-promoting actions of Id-1 in NSCLC cells. Conclusions We identified a tumorigenic role of Id-1 in NSCLC and provided a novel therapeutic target for NSCLC patients.
Collapse
Affiliation(s)
- Jie Li
- Department of Chest Surgery, The General Hospital of The People's Liberation Army, No. 28 Fuxing road, Beijing, 100853, China.
| | - Yingjie Li
- Department of Cardio-thoracic Surgery, First Affiliated Hospital, General Hospital of The People's Liberation Army, Beijing, China
| | - Bin Wang
- Department of Chest Surgery, The General Hospital of The People's Liberation Army, No. 28 Fuxing road, Beijing, 100853, China
| | - Yongfu Ma
- Department of Chest Surgery, The General Hospital of The People's Liberation Army, No. 28 Fuxing road, Beijing, 100853, China
| | - Ping Chen
- Department of Cardio-thoracic Surgery, First Affiliated Hospital, General Hospital of The People's Liberation Army, Beijing, China
| |
Collapse
|
14
|
Rac1 Regulates the Proliferation, Adhesion, Migration, and Differentiation of MDPC-23 Cells. J Endod 2017; 43:580-587. [DOI: 10.1016/j.joen.2016.11.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/23/2016] [Accepted: 11/17/2016] [Indexed: 11/21/2022]
|
15
|
Peng J, Hamanishi J, Matsumura N, Abiko K, Murat K, Baba T, Yamaguchi K, Horikawa N, Hosoe Y, Murphy SK, Konishi I, Mandai M. Chemotherapy Induces Programmed Cell Death-Ligand 1 Overexpression via the Nuclear Factor-κB to Foster an Immunosuppressive Tumor Microenvironment in Ovarian Cancer. Cancer Res 2015; 75:5034-45. [PMID: 26573793 DOI: 10.1158/0008-5472.can-14-3098] [Citation(s) in RCA: 395] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 09/01/2015] [Indexed: 12/13/2022]
Abstract
Emerging evidence has highlighted the host immune system in modulating the patient response to chemotherapy, but the mechanism of this modulation remains unclear. The aim of this study was to analyze the effect of chemotherapy on antitumor immunity in the tumor microenvironment of ovarian cancer. Treatment of ovarian cancer cell lines with various chemotherapeutic agents resulted in upregulated expression of MHC class I and programmed cell death 1 ligand 1 (PD-L1) in a NF-κB-dependent manner and suppression of antigen-specific T-cell function in vitro. In a mouse model of ovarian cancer, treatment with paclitaxel increased CD8(+) T-cell infiltration into the tumor site, upregulated PD-L1 expression, and activated NF-κB signaling. In particular, tumor-bearing mice treated with a combination of paclitaxel and a PD-L1/PD-1 signal blockade survived longer than mice treated with paclitaxel alone. In summary, we found that chemotherapy induces local immune suppression in ovarian cancer through NF-κB-mediated PD-L1 upregulation. Thus, a combination of chemotherapy and immunotherapy targeting the PD-L1/PD-1 signaling axis may improve the antitumor response and offers a promising new treatment modality against ovarian cancer.
Collapse
Affiliation(s)
- Jin Peng
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Noriomi Matsumura
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kaoru Abiko
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kumuruz Murat
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tsukasa Baba
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Naoki Horikawa
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuko Hosoe
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Susan K Murphy
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, North Carolina
| | - Ikuo Konishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masaki Mandai
- Department of Obstetrics and Gynecology, Kinki University, Osaka, Japan
| |
Collapse
|
16
|
Anticancer cardamonin analogs suppress the activation of NF-kappaB pathway in lung cancer cells. Mol Cell Biochem 2014; 389:25-33. [PMID: 24390014 DOI: 10.1007/s11010-013-1923-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 12/06/2013] [Indexed: 12/29/2022]
Abstract
NF-kappaB pathway has been proven to be critical to survival of lung cancer cells, and many natural products from plants were shown to inhibit the activation of this pathway. In this study, we investigated the effects of two cardamonin analogs, 4,4'-dihydroxylchalcone (DHC) and 4,4'-dihydroxy-2'-methoxychalcone (DHMC), on survival of lung cancer cells and the involved mechanisms. MTT assay revealed that the two compounds potently decreased the survival of immortalized and primary lung cancer cells. DHC and DHMC were able to induce apoptosis in A549 and NCI-H460 cells. Immunoblotting, immunofluorescent staining, and luciferase reporter further demonstrated that the two compounds suppressed the activation of NF-kappaB pathway in lung cancer cells. PMA-mediated NF-kappaB reactivation abrogated the effect of DHC and DHMC on lung cancer cells. DHC and DHMC were also shown to suppress the growth of A549 xenograft in mice. Collectively, we verified two cardamonin analogs as novel compounds suppressing NF-kappaB signaling for lung cancer therapy.
Collapse
|
17
|
Rousalova I, Banerjee S, Sangwan V, Evenson K, McCauley JA, Kratzke R, Vickers SM, Saluja A, D'Cunha J. Minnelide: a novel therapeutic that promotes apoptosis in non-small cell lung carcinoma in vivo. PLoS One 2013; 8:e77411. [PMID: 24143232 PMCID: PMC3797124 DOI: 10.1371/journal.pone.0077411] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 09/10/2013] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Minnelide, a pro-drug of triptolide, has recently emerged as a potent anticancer agent. The precise mechanisms of its cytotoxic effects remain unclear. METHODS Cell viability was studied using CCK8 assay. Cell proliferation was measured real-time on cultured cells using Electric Cell Substrate Impedence Sensing (ECIS). Apoptosis was assayed by Caspase activity on cultured lung cancer cells and TUNEL staining on tissue sections. Expression of pro-survival and anti-apoptotic genes (HSP70, BIRC5, BIRC4, BIRC2, UACA, APAF-1) was estimated by qRTPCR. Effect of Minnelide on proliferative cells in the tissue was estimated by Ki-67 staining of animal tissue sections. RESULTS In this study, we investigated in vitro and in vivo antitumor effects of triptolide/Minnelide in non-small cell lung carcinoma (NSCLC). Triptolide/Minnelide exhibited anti-proliferative effects and induced apoptosis in NSCLC cell lines and NSCLC mouse models. Triptolide/Minnelide significantly down-regulated the expression of pro-survival and anti-apoptotic genes (HSP70, BIRC5, BIRC4, BIRC2, UACA) and up-regulated pro-apoptotic APAF-1 gene, in part, via attenuating the NF-κB signaling activity. CONCLUSION In conclusion, our results provide supporting mechanistic evidence for Minnelide as a potential in NSCLC.
Collapse
Affiliation(s)
- Ilona Rousalova
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Identification of proteomic signatures associated with lung cancer and COPD. J Proteomics 2013; 89:227-37. [PMID: 23665002 DOI: 10.1016/j.jprot.2013.04.037] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 04/03/2013] [Accepted: 04/18/2013] [Indexed: 01/30/2023]
Abstract
UNLABELLED Lung cancer (LC) and chronic obstructive pulmonary disease (COPD) commonly coexist in smokers, and the presence of COPD increases the risk of developing LC. The aim of this study was to identify distinct proteomic profiles able to discriminate these two pathological entities. Protein content was assessed in the bronchoalveolar lavage (BAL) of 60 patients classified in four groups: COPD, COPD and LC, LC without COPD, and control with neither COPD nor LC. Proteins were separated into spots by bidimensional polyacrylamide gel electrophoresis (2D-PAGE) and examined by matrix-assisted laser desorption/ionization time of flight mass spectrometry (MALDI-TOF/TOF). A total of 40 proteins were differentially expressed in the LC and/or COPD groups as compared with the control group. Distinct protein profiles were identified and validated for each pathological entity (LC and COPD). The main networks involved were related to inflammatory signalling, free radical scavenging and oxidative stress response, and glycolysis and gluconeogenesis pathways. The most relevant signalling link between LC and COPD was through the NF-κB pathway. In conclusion, the protein profiles identified contribute to elucidate the underlying pathogenic pathways of both diseases, and provide new tools of potential use as biomarkers for the early diagnosis of LC. BIOLOGICAL SIGNIFICANCE Sequence coverage. The protein sequence coverage (95%) was estimated for specific proteins by the percentage of matching amino acids from the identified peptides having confidence greater than or equal to 95% divided by the total number of amino acids in the sequence. Ingenuity Pathways Analysis. Mapping of our proteins onto biological pathways and disease networks demonstrated that 22 proteins were linked to inflammatory signalling (p-value: 1.35 10(-08)-1.42 10(-02)), 15 proteins were associated with free radical scavenging and oxidative stress response (p-value: 4.93 10(-11)-1.27 10(-02)), and 9 proteins were related with glycolysis and gluconeogenesis pathways (p-value: 7.39 10(-09)-1.58 10(-02)).
Collapse
|
19
|
Identification of oxidative stress related proteins as biomarkers for lung cancer and chronic obstructive pulmonary disease in bronchoalveolar lavage. Int J Mol Sci 2013; 14:3440-55. [PMID: 23389041 PMCID: PMC3588051 DOI: 10.3390/ijms14023440] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Revised: 01/23/2013] [Accepted: 01/31/2013] [Indexed: 12/11/2022] Open
Abstract
Lung cancer (LC) and chronic obstructive pulmonary disease (COPD) commonly coexist in smokers, and the presence of COPD increases the risk of developing LC. Cigarette smoke causes oxidative stress and an inflammatory response in lung cells, which in turn may be involved in COPD and lung cancer development. The aim of this study was to identify differential proteomic profiles related to oxidative stress response that were potentially involved in these two pathological entities. Protein content was assessed in the bronchoalveolar lavage (BAL) of 60 patients classified in four groups: COPD, COPD and LC, LC, and control (neither COPD nor LC). Proteins were separated into spots by two dimensional polyacrylamide gel electrophoresis (2D-PAGE) and examined by matrix-assisted laser desorption/ionization time of flight mass spectrometry (MALDI-TOF/TOF). A total of 16 oxidative stress regulatory proteins were differentially expressed in BAL samples from LC and/or COPD patients as compared with the control group. A distinct proteomic reactive oxygen species (ROS) protein signature emerged that characterized lung cancer and COPD. In conclusion, our findings highlight the role of the oxidative stress response proteins in the pathogenic pathways of both diseases, and provide new candidate biomarkers and predictive tools for LC and COPD diagnosis.
Collapse
|
20
|
Tamatani T, Takamaru N, Hara K, Kinouchi M, Kuribayashi N, Ohe G, Uchida D, Fujisawa K, Nagai H, Miyamoto Y. Bortezomib-enhanced radiosensitization through the suppression of radiation-induced nuclear factor-κB activity in human oral cancer cells. Int J Oncol 2013; 42:935-44. [PMID: 23340716 DOI: 10.3892/ijo.2013.1786] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 10/12/2012] [Indexed: 11/05/2022] Open
Abstract
Oral cancer cells have a significantly augmented nuclear factor-κB (NF-κB) activity and the inhibition of this activity suppresses tumor growth. Bortezomib is a proteasome inhibitor and a drug used for molecular-targeted therapy (targets NF-κB). In this study, we investigated whether bortezomib would be effective as an inhibitor of proliferation and a radiosensitizer for the treatment of oral cancer. We demonstrate that bortezomib inhibits NF-κB activity and cell proliferation. The combined treatment with bortezomib and radiation (RT) suppressed NF-κB activity and cell growth in vitro and in vivo compared with RT treatment alone. To investigate the mechanisms by which bortezomib suppresses tumor growth, the expression of signaling molecules downstream of NF-κB were examined by ELISA. The combined treatment significantly inhibited the radiation-induced production of angiogenic factors and decreased the number of blood vessels in the tumor tissues. Although the expression of anti-apoptotic proteins was upregulated by RT, bortezomib downregulated the RT-induced expression of these proteins. Moreover, the expression of cleaved poly(ADP-ribose) polymerase in vitro and in vivo was enhanced by bortezomib, indicating that bortezomib inhibits tumor growth by inducing apoptosis. This study clearly demonstrates that bortezomib significantly inhibits tumor growth and that the combined treatment with bortezomib and RT results in a significant inhibition of tumor growth. The mechanisms underlying the inhibition of tumor growth by bortezomib include the suppression of angiogenesis and the induction of apoptosis. A novel molecular targeting therapy including bortezomib may be effective in the treatment of oral cancer by suppressing NF-κB activity.
Collapse
Affiliation(s)
- Tetsuya Tamatani
- Department of Oral Surgery, Subdivision of Molecular Oral Medicine, Division of Integrated Sciences of Translational Research, Institute of Health Biosciences, the University of Tokushima Graduate School, Tokushima 770-8504, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Yang HT, Chao PC, Yin MC. Riboflavin at High Doses Enhances Lung Cancer Cell Proliferation, Invasion, and Migration. J Food Sci 2013; 78:H343-9. [DOI: 10.1111/1750-3841.12012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 11/04/2012] [Indexed: 11/27/2022]
Affiliation(s)
- Hui-ting Yang
- Dept. of Nutrition; China Medical Univ.; Taichung City; Taiwan
| | - Pei-chun Chao
- Dept. of Nutrition; Chung Shan Medical Univ. Hospital; Taichung City; Taiwan
| | | |
Collapse
|
22
|
Lin CC, Lee IT, Wu WL, Lin WN, Yang CM. Adenosine triphosphate regulates NADPH oxidase activity leading to hydrogen peroxide production and COX-2/PGE2 expression in A549 cells. Am J Physiol Lung Cell Mol Physiol 2012; 303:L401-12. [PMID: 22773695 DOI: 10.1152/ajplung.00090.2012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Non-small cell lung carcinoma (NSCLC) accounts for most of all lung cancers, which is the leading cause of mortality in human beings. High level of cyclooxygenase-2 (COX-2) is one of the features of NSCLC and related to the low survival rate of NSCLC. However, whether extracellular nucleotides releasing from stressed resident tissues contributes to the expression of COX-2 remains unclear. Here, we showed that stimulation of A549 cells by adenosine 5'-O-(3-thiotriphosphate) (ATPγS) led to an increase in COX-2 gene expression and prostaglandin E(2) (PGE(2)) synthesis, revealed by Western blotting, RT-PCR, promoter assay, and enzyme-linked immunosorbent assay. In addition, ATPγS induced intracellular reactive oxygen species (ROS) generation through the activation of NADPH oxidase. The increase of ROS level resulted in activation of the c-Src/epidermal growth factor receptor (EGFR)/phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/nuclear factor (NF)-κB cascade. We also found that activated Akt was translocated into the nucleus and recruited with NF-κB and p300 to form a complex. Thus, activation of p300 modulated the acetylation of histone H4 via the NADPH oxidase/c-Src/EGFR/PI3K/Akt/NF-κB cascade stimulated by ATPγS. Our results are the first to show a novel role of NADPH oxidase-dependent Akt/p65/p300 complex formation that plays a key role in regulating COX-2/PGE(2) expression in ATPγS-treated A549 cells. Taken together, we demonstrated that ATPγS stimulated activation of NADPH oxidase, resulting in generation of ROS, which then activated the downstream c-Src/EGFR/PI3K/Akt/NF-κB/p300 cascade to regulate the expression of COX-2 and synthesis of PGE(2) in A549 cells. Understanding the regulation of COX-2 expression and PGE(2) release by ATPγS on A549 cells may provide potential therapeutic targets of NSCLC.
Collapse
Affiliation(s)
- Chih-Chung Lin
- Dept. of Pharmacology, College of Medicine, Chang Gung Univ., Kwei-San, Tao-Yuan, Taiwan
| | | | | | | | | |
Collapse
|
23
|
Gastonguay A, Berg T, Hauser AD, Schuld N, Lorimer E, Williams CL. The role of Rac1 in the regulation of NF-κB activity, cell proliferation, and cell migration in non-small cell lung carcinoma. Cancer Biol Ther 2012; 13:647-56. [PMID: 22549160 DOI: 10.4161/cbt.20082] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The small GTPase Rac1 regulates many cellular processes, including cytoskeletal reorganization, cell migration, proliferation, and survival. Additionally, Rac1 plays a major role in activating NF-κB-mediated transcription. Both Rac1 and NF-κB regulate many properties of the malignant phenotype, including anchorage-independent proliferation and survival, metastasis, and angiogenesis. Despite these findings, the roles of Rac1and NF-κB in non-small cell lung carcinoma, a leading cause of cancer deaths, have not been thoroughly investigated. Here, we compared the effects of Rac1 siRNA to that of the Rac1 inhibitor NSC23766 on multiple features of the NSCLC malignant phenotype, including NF-κB activity. We show that the siRNA-mediated silencing of Rac1 in lung cancer cells results in decreased cell proliferation and migration. The decrease in proliferation was observed in both anchorage-dependent and anchorage-independent assays. Furthermore, cells with decreased Rac1 expression have a slowed progression through the G 1 phase of the cell cycle. These effects induced by Rac1 siRNA correlated with a decrease in NF-κB transcriptional activity. Additionally, inhibition of NF-κB signaling with BAY 11-7082 inhibited proliferation; indicating that the loss of cell proliferation and migration induced by the silencing of Rac1 expression may be attributed in part to loss of NF-κB activity. Interestingly, treatment with the Rac1 inhibitor NSC23766 strongly inhibits cell proliferation, cell cycle progression, and NF-κB activity in lung cancer cells, to an even greater extent than the inhibition induced by Rac1 siRNA. These findings indicate that Rac1 plays an important role in lung cancer cell proliferation and migration, most likely through its ability to promote NF-κB activity, and highlight Rac1 pathways as therapeutic targets for the treatment of lung cancer.
Collapse
Affiliation(s)
- Adam Gastonguay
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | | | | | | | | |
Collapse
|
24
|
Cieckiewicz E, Angenot L, Gras T, Kiss R, Frédérich M. Potential anticancer activity of young Carpinus betulus leaves. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2012; 19:278-283. [PMID: 22014503 DOI: 10.1016/j.phymed.2011.09.072] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 08/04/2011] [Accepted: 09/16/2011] [Indexed: 05/31/2023]
Abstract
As part of our continuing research for anticancer compounds from the Walloon Region forest, EtOAc extract from Carpinus betulus leaves was phytochemically studied, leading to the bioguided isolation of pheophorbide a, which is responsible of anticancer properties of C. betulus young leaves. This compound was identified using nuclear magnetic resonance and mass spectrophotometric data and comparison with a commercial standard. Evaluation of the growth inhibitory activities of pheophorbide a using MTT colorimetric assay and phase-contrast microscopy in various human cancer cell lines confirmed the photoactivable properties of this compound. Our research showed, for the first time, the presence of pheophorbide a, a chlorophyll derived compound, which we quantified in high quantities in young leaves of C. betulus. This is in contrast with the literature which generally describes pheophorbide a as a catabolic product of chlorophyll, then preferentially present in old leaves.
Collapse
Affiliation(s)
- Ewa Cieckiewicz
- University of Liège, CIRM, Laboratory of Pharmacognosy, Liège, Belgium
| | | | | | | | | |
Collapse
|
25
|
Lamoral-Theys D, Fattorusso E, Mangoni A, Perinu C, Kiss R, Costantino V. Evaluation of the antiproliferative activity of diterpene isonitriles from the sponge Pseudoaxinella flava in apoptosis-sensitive and apoptosis-resistant cancer cell lines. JOURNAL OF NATURAL PRODUCTS 2011; 74:2299-2303. [PMID: 21985105 DOI: 10.1021/np2005055] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
One new (1) and three known (2-4) isonitrile diterpenes, isolated from the Caribbean sponge Pseudoaxinella flava, were assayed in human cancer cell lines in vitro using an MTT colorimetric assay and quantitative videomicroscopy. Compounds 1-4 displayed activity for human PC3 prostate apoptosis-sensitive cancer cell lines. Compounds 3 and 4 demonstrated similar growth inhibitory effects for three apoptosis-sensitive and three apoptosis-resistant cancer cell lines. Quantitative videomicroscopy analysis revealed that compounds 1 and 2 exerted their activity through cytotoxic effects, and compounds 3 and 4 through cytostatic effects. These results identify marine diterpene isonitriles as potential lead compounds for anticancer drug discovery.
Collapse
Affiliation(s)
- Delphine Lamoral-Theys
- The NeaNAT Group, Dipartimento di Chimica delle Sostanze Naturali, Università degli Studi di Napoli Federico II, Via D. Montesano 49, 80131 Napoli, Italy
| | | | | | | | | | | |
Collapse
|
26
|
Liu Y, Mayo MW, Nagji AS, Smith PW, Ramsey CS, Li D, Jones DR. Phosphorylation of RelA/p65 promotes DNMT-1 recruitment to chromatin and represses transcription of the tumor metastasis suppressor gene BRMS1. Oncogene 2011; 31:1143-54. [PMID: 21765477 PMCID: PMC3219802 DOI: 10.1038/onc.2011.308] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The majority of patients with lung cancer present with metastatic disease. Chronic inflammation and subsequent activation of NF-κB have been associated the development of cancers. The RelA/p65 subunit of NF-κB is typically associated with transcriptional activation. In this report we show that RelA/p65 can function as an active transcriptional repressor through enhanced methylation of the BRMS1 metastasis suppressor gene promoter via direct recruitment of DNMT-1 to chromatin in response to TNF. TNF-mediated phosphorylation of S276 on RelA/p65 is required for RelA/p65-DNMT-1 interactions, chromatin loading of DNMT-1, and subsequent BRMS1 promoter methylation and transcriptional repression. The ability of RelA/65 to function as an active transcriptional repressor is promoter specific as the NF-κB-regulated gene cIAP2 is transcriptionally activated while BRMS1 is repressed under identical conditions. Small molecule inhibition of either of the minimal interacting domains between RelA/p65-DNMT-1 and RelA/p65-BRMS1 promoter abrogates BRMS1 methylation and its transcriptional repression. The ability of RelA/p65 to directly recruit DNMT-1 to chromatin resulting in promoter-specific methylation and transcriptional repression of tumor metastasis suppressor gene BRMS1 highlights a new mechanism through which NF-κB can regulate metastatic disease, and offers a potential target for newer generation epigenetic oncopharmaceuticals.
Collapse
Affiliation(s)
- Y Liu
- Department of Surgery, University of Virginia, Charlottesville, VA 22908-0679, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Zhang XY, Dong QG, Huang JS, Huang AM, Shi CL, Jin B, Sha HF, Feng JX, Geng Q, Zhou J, Xu HL, Han BH. The expression of stem cell-related indicators as a prognostic factor in human lung adenocarcinoma. J Surg Oncol 2011; 102:856-62. [PMID: 20818602 DOI: 10.1002/jso.21718] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
INTRODUCTION The purpose of the present study was to detect the presence of BASC-like stem cell-related indicators, such as clara cell secretory protein (CCSP), Octamer-4 (OCT4) and Bmi-1, and evaluate their implications in the prognosis of patients with lung adenocarcinoma. METHODS Specimens of 134 cases of lung adenocarcinoma were collected after radical surgery from January 1999 to June 2004. RESULTS One hundred and twenty-six cases showed cells that were positive for CCSP, 99 cases positive for OCT4, 91 cases simultaneous expression of CCSP and OCT4 and 74 cases positive for Bmi-1. Bmi-1 was significantly higher in patients at stage III compared to patients at stages I and II. The pattern of survival curves showed that Bmi-1 was a significant prognostic factor of poor overall survival in lung adenocarcinoma patients (P = 0.0000), and the patients with OCT4(+) expression showed a greater increase in mortality than OCT4(-) patients (P = 0.0103). The results of univariate and multivariate Cox analysis revealed that the pathological stages of tumor node metastases (P = 0.037), OCT4 (P = 0.046) and Bmi-1 expression (P = 0.001) were independent prognostic factors. CONCLUSIONS OCT4 and Bmi-1 may be good biomarkers to predict the prognosis of patients with completely resected lung adenocarcinoma.
Collapse
Affiliation(s)
- Xue-Yan Zhang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Chen W, Li Z, Bai L, Lin Y. NF-kappaB in lung cancer, a carcinogenesis mediator and a prevention and therapy target. Front Biosci (Landmark Ed) 2011; 16:1172-85. [PMID: 21196225 DOI: 10.2741/3782] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Lung cancer ranks as the first malignant tumor killer worldwide. Despite the knowledge that carcinogens from tobacco smoke and the environment constitute the main causes of lung cancer, the mechanisms for lung carcinogenesis are still elusive. Cancer development and progression depend on the balance between cell survival and death signals. Common cell survival signaling pathways are activated by carcinogens as well as by inflammatory cytokines, which contribute substantially to cancer development. As a major cell survival signal, nuclear factor-kappaB (NF-kappaB) is involved in multiple steps in carcinogenesis and in cancer cell's resistance to chemo- and radio-therapy. Recent studies with animal models and cell culture systems have established the links between NF-kappaB and lung carcinogenesis, highlighting the significance of targeting NF-kappa signaling pathway for lung cancer treatment and chemoprevention. In this review, we summarize progresses in understanding the NF-kappaB pathway in lung cancer development as well as in modulating NF-kappaB for lung cancer prevention and therapy.
Collapse
Affiliation(s)
- Wenshu Chen
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest DR SE, Albuquerque, NM 87108, USA
| | | | | | | |
Collapse
|
29
|
Balde ES, Andolfi A, Bruyère C, Cimmino A, Lamoral-Theys D, Vurro M, Damme MV, Altomare C, Mathieu V, Kiss R, Evidente A. Investigations of fungal secondary metabolites with potential anticancer activity. JOURNAL OF NATURAL PRODUCTS 2010; 73:969-71. [PMID: 20415482 DOI: 10.1021/np900731p] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Fourteen metabolites, isolated from phytopathogenic and toxigenic fungi, were evaluated for their in vitro antigrowth activity for six distinct cancer cell lines, using the MTT colorimetric assay. Bislongiquinolide (1) and dihydrotrichodimerol (5), which belong to the bisorbicillinoid structural class, displayed significant growth inhibitory activity against the six cancer cell lines studied, while the remaining compounds displayed weak or no activity. The data show that 1 and 5 have similar growth inhibitory activities with respect to those cancer cell lines that display certain levels of resistance to pro-apoptotic stimuli or those that are sensitive to apoptosis. Quantitative videomicroscopy analysis revealed that 1 and 5 exert their antiproliferative effect through cytostatic and not cytotoxic activity. The preliminary results from the current study have stimulated further structure-activity investigations with respect to the growth inhibitory activity of compounds belonging to the bisorbicillinoid group.
Collapse
Affiliation(s)
- ElHadj Saidou Balde
- Laboratoire de Toxicologie and Laboratoire de Chimie Analytique, Toxicologie et Chimie Physique Appliquee, Institut de Pharmacie, Universite Libre de Bruxelles (ULB), Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Smyrniotopoulos V, Vagias C, Bruyère C, Lamoral-Theys D, Kiss R, Roussis V. Structure and in vitro antitumor activity evaluation of brominated diterpenes from the red alga Sphaerococcus coronopifolius. Bioorg Med Chem 2010; 18:1321-30. [DOI: 10.1016/j.bmc.2009.12.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 12/02/2009] [Accepted: 12/08/2009] [Indexed: 10/20/2022]
|
31
|
Scagliotti GV, Germonpré P, Bosquée L, Vansteenkiste J, Gervais R, Planchard D, Reck M, De Marinis F, Lee JS, Park K, Biesma B, Gans S, Ramlau R, Szczesna A, Makhson A, Manikhas G, Morgan B, Zhu Y, Chan KC, von Pawel J. A randomized phase II study of bortezomib and pemetrexed, in combination or alone, in patients with previously treated advanced non-small-cell lung cancer. Lung Cancer 2009; 68:420-6. [PMID: 19692142 DOI: 10.1016/j.lungcan.2009.07.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Revised: 07/17/2009] [Accepted: 07/19/2009] [Indexed: 11/15/2022]
Abstract
BACKGROUND This is a phase II randomized study to evaluate the efficacy and safety of bortezomib and pemetrexed alone or in combination, in patients with previously treated advanced non-small-cell lung cancer (NSCLC). The primary end point was assessment of response rate. METHODS A total of 155 patients were randomized (1:1:1) to pemetrexed (500mg/m(2)) on day 1 plus bortezomib (1.6mg/m(2)) on days 1 and 8 (Arm A) or pemetrexed (500mg/m(2)) on day 1 (Arm B) or bortezomib (1.6mg/m(2)) on days 1 and 8 (Arm C) of a 21 day cycle. Response rate was assessed by investigators using Response Evaluation Criteria In Solid Tumors (RECIST) criteria and toxicity assessed by the National Cancer Institute-Common Terminology Criteria for Adverse Events (NCI-CTCAE) grading system. RESULTS Response rate was 7% in Arm A, 4% in Arm B, and 0% in Arm C; disease control rates were 73%, 62%, and 43%, respectively. Median overall survival was 8.6 months in Arm A, 12.7 months in Arm B, and 7.8 months in Arm C; time to progression was 4.0 months, 2.9 months, and 1.4 months, respectively. Most common reported adverse events >/=grade 3 were neutropenia (19%), thrombocytopenia (15%), and dyspnea (13%) in Arm A, neutropenia (10%) in Arm B, and dyspnea (13%) and fatigue (10%) in Arm C. CONCLUSION In previously treated NSCLC the addition of bortezomib to pemetrexed was well tolerated but offered no statistically significant response or survival advantage versus pemetrexed alone, while bortezomib alone showed no clinically significant activity.
Collapse
Affiliation(s)
- Giorgio V Scagliotti
- University of Torino, Department of Clinical & Biological Sciences, Thoracic Oncology Unit, S. Luigi Hospital, Regione Gonzole 10, Orbassano (Torino) 10043, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Davies AM, Chansky K, Lara PN, Gumerlock PH, Crowley J, Albain KS, Vogel SJ, Gandara DR. Bortezomib plus gemcitabine/carboplatin as first-line treatment of advanced non-small cell lung cancer: a phase II Southwest Oncology Group Study (S0339). J Thorac Oncol 2009; 4:87-92. [PMID: 19096312 PMCID: PMC3024911 DOI: 10.1097/jto.0b013e3181915052] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Bortezomib is a small-molecule proteasome inhibitor with single-agent activity in patients with non-small cell lung carcinoma (NSCLC) and synergy with gemcitabine in preclinical studies. This phase II study of bortezomib in combination with gemcitabine/carboplatin was conducted in chemotherapy-naive advanced NSCLC patients to assess efficacy and safety. METHODS Patients with selected stage IIIB/IV NSCLC, performance status 0-1, and no history of brain metastasis received up to six 21-day cycles of gemcitabine 1000 mg/m, days 1 and 8, carboplatin area under curve 5.0, day 1, and bortezomib 1.0 mg/m, days 1, 4, 8, and 11. RESULTS One-hundred-fourteen patients (52% adenocarcinoma, 85% stage IV) received a median of 3.6 treatment cycles. Median follow-up was >3 years. Median overall survival was 11 months; 1-year and 2-year survival rates were 47% and 19%, respectively. Median progression-free survival was 5 months; 1-year progression-free survival rate was 7%. Response rate was 23%, and disease control rate (responses + stable disease) was 68%. The most common grade 3/4 toxicities were thrombocytopenia (63%) and neutropenia (52%). One patient experienced febrile neutropenia. Grade 3/4 neuropathy occurred in 4%, and a further 6% experienced grade 2 sensory neuropathy. CONCLUSIONS Bortezomib plus gemcitabine/carboplatin resulted in a notable survival benefit in patients with advanced NSCLC, with the anticipated primary toxicity of myelosuppression. Further studies designed to investigate the role of bortezomib in advanced NSCLC are warranted.
Collapse
Affiliation(s)
- Angela M Davies
- Department of Hematology/Oncology, University of California, Davis Cancer Center, Sacramento, California, USA.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Vrzalikova K, Skarda J, Ehrmann J, Murray PG, Fridman E, Kopolovic J, Knizetova P, Hajduch M, Klein J, Kolek V, Radova L, Kolar Z. Prognostic value of Bmi-1 oncoprotein expression in NSCLC patients: a tissue microarray study. J Cancer Res Clin Oncol 2008; 134:1037-42. [PMID: 18264721 DOI: 10.1007/s00432-008-0361-y] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2007] [Accepted: 01/09/2008] [Indexed: 11/29/2022]
Abstract
PURPOSE Bmi-1 is a Polycomb group member which participates in many physiological processes as well as in a wide spectrum of cancers. The aim of this study was to investigate Bmi-1 expression in non-small cell lung cancer (NSCLC) in respect to clinicopathological features and therapeutic outcomes. METHODS Immunohistochemical staining for Bmi-1 was performed on tissue microarrays (TMAs) constructed from 179 formalin-fixed and paraffin-embedded NSCLC samples (106 squamous, 58 adeno-, and 15 large cell carcinomas). Data were subject to statistical analysis by SPSS. RESULTS Overall evaluation of all tumor cases showed that 20 (11.43%) were negative, 37 (21.14%) showed weak, 65 (37.14%) moderate and 57 (32.57%) strong nuclear positivity for Bmi-1. Statistical analysis of our data revealed that the expression of Bmi-1 was significantly higher in stage III (P = 10(-6)) and stage IV (P = 10(-5)) tumors compared to stages I and II tumors. The administration of adjuvant chemotherapy significantly increased DFS at stage I and II patients who did not express Bmi-1 when compared to their Bmi-1 positive counterparts (P = 0.05). CONCLUSIONS Our results suggest that Bmi-1 is significantly associated with progression of NSCLC and might serve as a prognostic marker of adverse disease outcome.
Collapse
Affiliation(s)
- Katerina Vrzalikova
- Laboratory of Molecular Pathology, Department of Pathology, Palacky University, Olomouc, Czech Republic.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Tew GW, Lorimer EL, Berg TJ, Zhi H, Li R, Williams CL. SmgGDS regulates cell proliferation, migration, and NF-kappaB transcriptional activity in non-small cell lung carcinoma. J Biol Chem 2007; 283:963-76. [PMID: 17951244 DOI: 10.1074/jbc.m707526200] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Non-small cell lung carcinoma (NSCLC) is promoted by the increased activities of several small GTPases, including K-Ras4B, Rap1A, Rap1B, RhoC, and Rac1. SmgGDS is an unusual guanine nucleotide exchange factor that activates many of these small GTPases, and thus may promote NSCLC development or progression. We report here that SmgGDS protein levels are elevated in NSCLC tumors, compared with normal lung tissue from the same patients or from individuals without cancer. To characterize SmgGDS functions in NSCLC, we tested the effects of silencing SmgGDS expression by transfecting cultured NSCLC cells with SmgGDS small interfering RNA (siRNA). Cells with silenced SmgGDS expression form fewer colonies in soft agar, do not proliferate in culture due to an arrest in G(1) phase, and exhibit disrupted myosin organization and reduced cell migration. The transcriptional activity of NF-kappaB in NSCLC cells is diminished by transfecting the cells with SmgGDS siRNA, and enhanced by transfecting the cells with a cDNA encoding SmgGDS. Because RhoA is a major substrate for SmgGDS, we investigated whether diminished RhoA expression mimics the effects of diminished SmgGDS expression. Silencing RhoA expression with RhoA siRNA disrupts myosin organization, but only moderately decreases cell proliferation and does not inhibit migration. Our finding that the aggressive NSCLC phenotype is more effectively suppressed by silencing SmgGDS than by silencing RhoA is consistent with the ability of SmgGDS to regulate multiple small GTPases in addition to RhoA. These results demonstrate that SmgGDS promotes the malignant NSCLC phenotype and is an intriguing therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Gaik Wei Tew
- Department of Pharmacology and Toxicology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226, USA
| | | | | | | | | | | |
Collapse
|
35
|
Vegeler RC, Yip-Schneider MT, Ralstin M, Wu H, Crooks PA, Neelakantan S, Nakshatri H, Sweeney CJ, Schmidt CM. Effect of celecoxib and novel agent LC-1 in a hamster model of lung cancer. J Surg Res 2007; 143:169-76. [PMID: 17950089 DOI: 10.1016/j.jss.2007.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2007] [Indexed: 11/29/2022]
Abstract
BACKGROUND Lung cancer is the leading cause of cancer deaths in the United States. Inflammatory molecules, cyclooxygenase-2 (COX-2) and nuclear factor kappa B (NF-kappaB) have been implicated in lung carcinogenesis. The therapeutic potential of celecoxib, a COX-2 selective inhibitor, and LC-1, a pro-apoptotic drug with accompanying inhibition of NF-kappaB, were investigated. MATERIALS AND METHODS Syrian golden hamsters (n = 140) underwent N-nitroso-bis(2-oxopropyl)amine (BOP) injection weekly for 6 wk. Hamsters were randomized into seven groups: placebo and low/high doses of LC-1, celecoxib, and LC-1/celecoxib. Treatments were given via orogastric lavage for 32 wk. Immunohistochemistry was used to determine COX-2 expression and NF-kappaB activity. Ki-67 labeling was used as an index of proliferation. COX activity was measured by prostaglandin E(2) enzyme-linked immunosorbent assay. RESULTS BOP successfully induced lung adenocarcinoma in 63% of placebo animals. Lung tumors strongly expressed COX-2 and NF-kappaB. Prostaglandin E(2) levels were decreased in celecoxib compared with placebo groups (P < 0.05) reflecting suppression of COX activity, but no decrease in NF-kappaB was seen as measured by immunohistochemistry in the tumors. There was no significant difference in tumor size, tumor incidence, or tumor proliferation index between placebo and treatment groups. CONCLUSIONS Carcinogen exposure results in increased COX-2 and NF-kappaB expression and suggests a role in carcinogenesis. Celecoxib and LC-1 did not have any effect in preventing lung cancer development when co-administered with and continued after the carcinogen BOP. Higher doses that can result in suppression of NF-kappaB activity will need to be explored to determine the viability of this approach to prevent lung cancer development.
Collapse
Affiliation(s)
- Reid C Vegeler
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kashiwagi H, McDunn JE, Goedegebuure PS, Gaffney MC, Chang K, Trinkaus K, Piwnica-Worms D, Hotchkiss RS, Hawkins WG. TAT-Bim Induces Extensive Apoptosis in Cancer Cells. Ann Surg Oncol 2007; 14:1763-71. [PMID: 17206479 DOI: 10.1245/s10434-006-9298-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2006] [Revised: 11/07/2006] [Accepted: 11/09/2006] [Indexed: 11/18/2022]
Abstract
BACKGROUND Suppression of apoptosis is central to the development of cancer and is associated with resistance to modern adjuvant treatments. Therefore, molecules and pathways of apoptotic processes are critical targets for the development of anti-cancer therapeutics. Since apoptosis is executed by intracellular proteins, molecular approaches must incorporate a method to deliver the treatment into the tumor cells. METHODS We utilized a peptide that contains two domains, a peptide transduction domain derived from the HIV-1 TAT protein and a biological effector domain, the BH3 domain from the pro-apoptotic Bcl-2 family member Bim. We examined whether this construct (TAT-Bim) induced apoptosis in several cancer cell lines (T-cell lymphoma (EL4), pancreatic cancer (Panc-02), and melanoma (B16)) and whether TAT-Bim treatment synergized with radiation. A mutant TAT-Bim peptide with no biologic activity (TAT-Bim-inactive) was used as a control. C57/BL6 mice were challenged with syngeneic cancer cell lines and the effects of intratumoral TAT-Bim injection on tumor growth and host survival were determined. RESULTS TAT-Bim was internalized by all cancer cells within two hours. TAT-Bim resulted in apoptosis in a dose dependent fashion in all cell lines and sublethal irradiation augmented the effects of TAT-Bim induced apoptosis. TAT-Bim significantly slowed tumor growth in murine models of pancreatic cancer and melanoma. CONCLUSION TAT-Bim exemplifies a strategy for cancer therapy that involves inducing apoptosis by antagonizing the endogenous anti-apoptotic machinery. Small peptide therapeutics, in combination with traditional adjuvant therapies such as radiation, may provide a valuable 'second hit' and drive tumor cells into programmed cell death.
Collapse
Affiliation(s)
- Hiroyuki Kashiwagi
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Petersen RP, D'Amico TA. Molecular and Genetic Markers in Thoracic Surgery. Ann Thorac Surg 2006; 82:2335-6. [PMID: 17131544 DOI: 10.1016/j.athoracsur.2006.06.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Rebecca P Petersen
- Albert Thoracic Oncology Program, Division of Thoracic Surgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | |
Collapse
|
38
|
Nadkar A, Pungaliya C, Drake K, Zajac E, Singhal SS, Awasthi S. Therapeutic resistance in lung cancer. Expert Opin Drug Metab Toxicol 2006; 2:753-77. [PMID: 17014393 DOI: 10.1517/17425255.2.5.753] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Despite considerable progress over the last 25 years in the systemic therapy of lung cancer, intrinsic and acquired resistance to chemotherapeutic agents and radiation remains a vexing problem. The number of mechanisms of therapeutic resistance in lung cancer has expanded considerably over the past three decades, and the crucial role of stress resistance pathways is increasingly recognised as a cause of intrinsic and acquired chemo- and radiotherapy resistance. This paper reviews recent evidence for stress defence proteins, particularly RALBP1/RLIP76, in mediating intrinsic and acquired chemotherapy and radiation resistance in human lung cancer.
Collapse
Affiliation(s)
- Aalok Nadkar
- Department of Chemistry and Biochemistry, University of Texas at Arlington, 700 Planetarium Place, CPB # 351, 76019-0065, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Liu Y, Denlinger CE, Rundall BK, Smith PW, Jones DR. Suberoylanilide Hydroxamic Acid Induces Akt-mediated Phosphorylation of p300, Which Promotes Acetylation and Transcriptional Activation of RelA/p65. J Biol Chem 2006. [DOI: 10.1016/s0021-9258(19)84048-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
40
|
Liu Y, Denlinger CE, Rundall BK, Smith PW, Jones DR. Suberoylanilide Hydroxamic Acid Induces Akt-mediated Phosphorylation of p300, Which Promotes Acetylation and Transcriptional Activation of RelA/p65. J Biol Chem 2006; 281:31359-68. [PMID: 16926151 DOI: 10.1074/jbc.m604478200] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have previously demonstrated that the transcription factor NF-kappaB is activated by histone deacetylase inhibitors in a PI3K/Akt-dependent manner. The molecular mechanisms governing this process have not been well described. By virtue of their inhibitory action, it is unclear whether the addition of histone deacetylase inhibitors simply preserves the acetylation status of RelA/p65 or whether they actively stimulate signaling cascades that result in increased acetylation and transcription of NF-kappaB. Here we provide evidence that suberoylanilide hydroxamic acid stimulates NF-kappaB transcription through a signaling cascade that involves activation of both the serine/threonine kinase Akt and the p300 acetyltransferase. Using newly developed phosphospecific antibodies to p300 (pSer(1834)), and site-directed mutant proteins, we find that suberoylanilide hydroxamic acid stimulates Akt activity, which is required to phosphorylate p300 at Ser(1834). Akt-mediated phosphorylation of p300 dramatically increases its acetyltransferase activity as measured by an increased acetylation of RelA/p65 at Lys(310), a modification that is required for full NF-kappaB transcription. Importantly, coordinate activation of Akt/p300 pathway by suberoylanilide hydroxamic acid occurs at the chromatin level, resulting in recruitment of activated Akt (pSer(473)), p300 (pSer(1834)), acetylated RelA/p65 (Lys(310)), and RNA polymerase II to the NF-kappaB-dependent cIAP-2 and Bfl-1/A1 promoters. These studies provide evidence that histone deacetylase inhibitors, such as suberoylanilide hydroxamic acid, not only inhibit deacetylase activity but also stimulate active NF-kappaB transcription and cell survival through signaling pathways involving Akt and increased p300 acetyltransferase activity.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
| | | | | | | | | |
Collapse
|
41
|
Zhang Z, Ma J, Li N, Sun N, Wang C. Expression of Nuclear Factor-κB and Its Clinical Significance in Nonsmall-Cell Lung Cancer. Ann Thorac Surg 2006; 82:243-8. [PMID: 16798222 DOI: 10.1016/j.athoracsur.2006.01.049] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2005] [Revised: 01/07/2006] [Accepted: 01/10/2006] [Indexed: 11/26/2022]
Abstract
BACKGROUND It has been known that transcription factor nuclear factor (NF)-kappaB plays an important role in cell proliferation and oncogenesis. The aims of this study were to evaluate expression levels of NF-kappaB in nonsmall-cell lung cancer (NSCLC) and to elucidate its clinical significance and prognostic value for patients with NSCLC. METHODS Using 45 tumor tissue specimens from 45 patients with NSCLC who underwent surgery, we investigated the expression of NF-kappaB using Western blotting analysis. Apoptotic rate of NSCLC cells with different expression of NF-kappaB was determined by TUNEL (terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end-labeling) assay. Paraffin-embedded tissue blocks from 71 consecutive patients with NSCLC were obtained for immunohistochemical staining. RESULTS The expression level of NF-kappaB in poorly or moderately differentiated lung cancer cells was higher than that in well-differentiated ones (p = 0.001). The apoptotic rate was lower for lung cancer cells with higher NF-kappaB expression than for those with lower NF-kappaB expression (p = 0.0238). Furthermore, expression of NF-kappaB was correlated with caspase-3, cyclooxygenase-2, and p53 expression in lung cancer cells that were examined. Most NSCLC cells showed nuclear staining pattern and the nuclear positive rate was 67.6% (48 of 71 specimens). Immunohistochemical NF-kappaB expression in patients with NSCLC was an independent prognostic factor for overall survival. CONCLUSIONS The results of this study suggest that expression of NF-kappaB may correlate with lung cancer differentiation. Overexpression of NF-kappaB inhibits tumor cell apoptosis and indicates an unfavorable prognosis for overall survival in some patients with NSCLC.
Collapse
Affiliation(s)
- Zhenfa Zhang
- Department of Lung Cancer, Affiliated Cancer Hospital, Tian Jin Medical University, Tian Jin City, China
| | | | | | | | | |
Collapse
|
42
|
Scagliotti G. Proteasome inhibitors in lung cancer. Crit Rev Oncol Hematol 2006; 58:177-89. [PMID: 16427303 DOI: 10.1016/j.critrevonc.2005.12.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2005] [Revised: 11/30/2005] [Accepted: 12/06/2005] [Indexed: 01/12/2023] Open
Abstract
Proteasome inhibition is a novel therapeutic approach that is being investigated in non-small cell and small cell lung cancer (NSCLC and SCLC). Proteasome inhibition affects a range of intracellular signals and disrupts the levels of numerous proteins, causing apoptosis via multiple pathways. Importantly, malignant cells are more sensitive to proteasome inhibition than normal cells. A number of proteasome inhibitors have demonstrated activity in preclinical studies, both as single agents and in combination with conventional and novel antineoplastic agents. However, only bortezomib, a dipeptide boronic acid analog, has been investigated in lung cancer clinical trials, in which it has shown activity as a single agent and in combination regimens. Numerous preclinical and clinical studies are ongoing in both NSCLC and SCLC. Proteasome inhibition could potentially play a significant role in the future management of these conditions.
Collapse
Affiliation(s)
- Giorgio Scagliotti
- University of Torino, Department of Clinical and Biological Sciences, Thoracic Oncology Unit, San Luigi Hospital-Regione Gonzole 10, 10043 Orbassano, Torino, Italy.
| |
Collapse
|
43
|
Toloza EM, D'Amico TA. Targeted therapy for non-small cell lung cancer. Semin Thorac Cardiovasc Surg 2006; 17:199-204. [PMID: 16253823 DOI: 10.1053/j.semtcvs.2005.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2005] [Indexed: 11/11/2022]
Abstract
The overall survival for the treatment of lung cancer patients is less than 15%, despite advances in chemotherapy, radiation therapy, and surgery, due to the inability to control metastatic disease. Over the past three decades, the genetics of lung cancer has been progressively delineated. Small molecule drugs or monoclonal antibodies have been developed that target and inactivate specific cancer-related proteins, such as growth factor receptors or their kinases. This article will review the therapeutic implications of molecular changes associated with non-small cell lung cancer and the status of targeted therapies in its treatment.
Collapse
Affiliation(s)
- Eric M Toloza
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA.
| | | |
Collapse
|
44
|
Abstract
More effective therapies are needed for non-small-cell lung cancer (NSCLC) and small-cell lung cancer (SCLC). Proteasome inhibitors are one class of molecularly targeted antineoplastic agents being investigated for these diseases. These agents block the activity of the 26S proteasome, which is responsible for the degradation of the vast majority of intracellular proteins and thus affect multiple signaling pathways within cells. Bortezomib is the first proteasome inhibitor to be evaluated in human studies and is approved for use in multiple myeloma. Bortezomib is now being investigated as a potential treatment for NSCLC and SCLC. Preclinical studies have shown that single-agent bortezomib causes growth inhibition and apoptosis in numerous NSCLC cell lines in vitro and has antitumor activity in vivo. Bortezomib affects the levels of several proteins known to be of significance in lung cancers. Studies of bortezomib in combination with other antitumor agents in vitro and in vivo demonstrate that these combination regimens can offer additive/synergistic effects compared with the single agents. Bortezomib has been investigated in combination with taxanes, gemcitabine, carboplatin, histone deactylase inhibitors, and other molecularly targeted agents in various NSCLC cell lines. The sequence of administration of the agents in preclinical combination regimens in vitro and in vivo has been shown to be of significance; further elucidation of the mechanism of efficacy of bortezomib in lung cancer is required. Numerous clinical studies have been carried out or are ongoing. Bortezomib has the potential to play a significant role in the future management of NSCLC and SCLC.
Collapse
|
45
|
Abstract
After more than three decades of its declaration, the war against cancer still appears far from being won. Although there have been decisive victories in a few battles, such as the one against testicular cancer, the overall result is sobering. Hopes for an imminent cure had been raised among the public by the promises of molecular biology, combinatorial chemistry and high-throughput screening. These promises have manifested themselves in the widely proclaimed strategy of rationally targeted anticancer drug discovery, which may be summarized as the 'one-gene-one target-one drug' approach. Over the years, however, it has gradually become clear that, in most cases, treatment of cancer with a single drug may at best delay progression of the disease but is unlikely to lead to a cure. Thus, it appears that rationally targeted monotherapy will have to be replaced by rationally targeted combination therapy. Inhibitors of NF-kappaB look likely to become an important weapon in the anticancer combination therapy arsenal.
Collapse
Affiliation(s)
- Burkhard Haefner
- Department of Oncology, Johnson & Johnson Pharmaceutical Research and Development, Beerse, Belgium
| |
Collapse
|
46
|
Denlinger CE, Rundall BK, Jones DR. Inhibition of phosphatidylinositol 3-kinase/Akt and histone deacetylase activity induces apoptosis in non-small cell lung cancer in vitro and in vivo. J Thorac Cardiovasc Surg 2005; 130:1422-9. [PMID: 16256798 DOI: 10.1016/j.jtcvs.2005.06.051] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2005] [Revised: 06/06/2005] [Accepted: 06/16/2005] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Resistance to histone deacetylase inhibitors in non-small cell lung cancer is mediated in part through activation of nuclear factor-kappaB through a phosphatidylinositol 3-kinase/Akt-dependent pathway. We hypothesize that inhibition of phosphatidylinositol 3-kinase/Akt will sensitize non-small cell lung cancer cells to histone deacetylase inhibitor-induced apoptosis. METHODS Tumorigenic non-small cell lung cancer cell lines H157, H358, H460, and A549 were treated with nothing, the histone deacetylase inhibitor butyrate, the phosphatidylinositol 3-kinase/Akt inhibitor LY294002, or both compounds. Nuclear factor-kappaB activity was assessed by reporter gene assays and reverse transcriptase-polymerase chain reaction of the nuclear factor-kappaB dependent genes cIAP-2, Bfl/A1, and MnSOD. Whole cell extracts were immunoblotted for phospho-Akt, Akt, and phospho-ser/thr-Akt substrate. Cell death and apoptosis were measured by crystal violet staining, caspase-3 activity, and DNA fragmentation. A549 non-small cell lung cancer xenografts were created in athymic nude mice, and tumor growth was assessed after treatments as noted above. Explanted tumors underwent terminal deoxynucleotide transferase-mediated dUTP nick-end labeling and Western blot analyses for apoptosis assessment and drug target validation, respectively. RESULTS Butyrate activated nuclear factor-kappaB-dependent transcription, and LY294002 abrogated this effect. Combined treatment induced more apoptosis and cell death in vitro compared with either drug alone as measured by caspase-3, DNA fragmentation, and clonogenic survival. Combined butyrate and LY294002 was tumoristatic in vivo, but all other xenografts grew. This decreased tumor growth correlated with more apoptosis in the xenografts treated with combined therapy. Tumor levels of phospho-Akt and acetylated histone H3 were decreased and increased, respectively, in xenografts treated with combined therapy. CONCLUSIONS Combined histone deacetylase inhibitor and phosphatidylinositol 3-kinase/Akt pathway inhibition sensitized non-small cell lung cancer xenografts to apoptosis. Further investigations of this combined therapy are warranted as new pharmacologic phosphatidylinositol 3-kinase/Akt pathway inhibitors are developed.
Collapse
Affiliation(s)
- Chadrick E Denlinger
- Department of Surgery, University of Virginia, Charlottesville, Va 22908-0679, USA
| | | | | |
Collapse
|
47
|
Gibson AD, Stewart J, Reddy GK, Gibson TB. 2005 highlights from: 41st Annual Meeting of the American Society of Clinical Oncology, Orlando, FL, May 2005. Clin Lung Cancer 2005; 7:11-6. [PMID: 16098238 DOI: 10.1016/s1525-7304(11)70384-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
48
|
Denlinger CE, Rundall BK, Jones DR. Proteasome inhibition sensitizes non-small cell lung cancer to histone deacetylase inhibitor-induced apoptosis through the generation of reactive oxygen species. J Thorac Cardiovasc Surg 2005; 128:740-8. [PMID: 15514602 DOI: 10.1016/j.jtcvs.2004.07.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
OBJECTIVES The histone deacetylase inhibitor suberoylanilide hydroxamic acid induces apoptosis in some malignancies through mitochondrial injury and generation of reactive oxygen species. Histone deacetylase inhibitors also activate the antiapoptotic transcription factor nuclear factor kappaB. We hypothesize that proteasome inhibition with bortezomib (Velcade; Millennium Pharmaceuticals, Inc, Cambridge, Mass)will inhibit nuclear factor kappaB activation, enhance suberoylanilide hydroxamic acid-induced mitochondrial injury, and sensitize non-small cell lung cancer cells to apoptosis. METHODS Four tumorigenic non-small cell lung cancer cell lines were treated with nothing, suberoylanilide hydroxamic acid, bortezomib, or both drugs. Nuclear factor kappaB-dependent transcription was determined by reporter gene assays and endogenous interleukin 8 transcription. Reactive oxygen species were quantified by using the fluorophore H 2 DCFDA. Cell viability was determined on the basis of clonogenic survival, and apoptosis was measured by quantifying caspase-3 activity and DNA fragmentation. Apoptosis and cell-survival assays were repeated in similarly treated cells incubated in the presence or absence of N-acetyl cysteine. Statistical significance was determined by means of analysis of variance. RESULTS Suberoylanilide hydroxamic acid significantly enhanced interleukin 8 and nuclear factor kappaB-dependent reporter gene transcription, and these effects were inhibited by bortezomib ( P < or = .01). Combined treatment with suberoylanilide hydroxamic acid and bortezomib induced greater reactive oxygen species generation, more apoptosis ( P < or = .02), and more cell death ( P < or = .001) than either drug alone. N-acetyl cysteine diminished the induction of apoptosis and enhanced cell survival ( P < or = .04). CONCLUSIONS Suberoylanilide hydroxamic acid and bortezomib synergistically induce reactive oxygen species generation in non-small cell lung cancer, and this plays a critical role in the induction of apoptosis after treatment. Combined treatment with suberoylanilide hydroxamic acid and bortezomib might be an effective treatment strategy for non-small cell lung cancer.
Collapse
Affiliation(s)
- Chadrick E Denlinger
- Department of Thoracic and Cardiovascular Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
49
|
Denlinger CE, Rundall BK, Keller MD, Jones DR. Proteasome Inhibition Sensitizes Non–Small-Cell Lung Cancer to Gemcitabine-Induced Apoptosis. Ann Thorac Surg 2004; 78:1207-14; discussion 1207-14. [PMID: 15464472 DOI: 10.1016/j.athoracsur.2004.04.029] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/05/2004] [Indexed: 01/10/2023]
Abstract
BACKGROUND My colleagues and I have previously shown that chemotherapy activates the antiapoptotic transcription factor nuclear factor (NF)-kappaB in non-small-cell lung cancer (NSCLC). We hypothesized that inhibition of NF-kappaB by using the proteasome inhibitor bortezomib (Velcade) would sensitize NSCLC to gemcitabine-induced apoptosis. METHODS Tumorigenic NSCLC cell lines (H157 and A549) were treated with nothing, gemcitabine, bortezomib, or both compounds. NF-kappaB activity was determined by nuclear p65 protein levels, electrophoretic mobility shift assays, and reverse transcription-polymerase chain reaction of the NF-kappaB-regulated genes interleukin-8, c-IAP2, and Bcl-xL. The p21 and p53 protein levels were determined in similarly treated cells. Cell-cycle dysregulation was assessed by fluorescence-activated cell sorting analysis. Cell death and apoptosis were quantified by clonogenic assays, caspase-3 activation, and DNA fragmentation. NSCLC A549 xenografts were generated and treated as noted previously. Tumor growth was assessed over a 4-week treatment period. Statistical analysis was performed with analysis of variance. RESULTS Gemcitabine enhanced nuclear p65 levels, NF-kappaB binding to DNA, and transcription of all NF-kappaB-regulated genes. Bortezomib inhibited each of these effects. Combined gemcitabine and bortezomib enhanced p21 and p53 expression and induced S-phase and G2/M cell-cycle arrests, respectively. Combined treatment killed 80% of the NSCLC cells and induced apoptosis, as determined by caspase-3 activation (p = 0.05) and DNA fragmentation (p = 0.02). NSCLC xenografts treated with combination therapy grew significantly slower than xenografts treated with gemcitabine alone (p = 0.02). CONCLUSIONS Bortezomib inhibits gemcitabine-induced activation of NF-kappaB and sensitizes NSCLC to death in vitro and in vivo. This combined treatment strategy warrants further investigation and may represent a reasonable treatment strategy for select patients with NSCLC given the current clinical availability of both drugs.
Collapse
Affiliation(s)
- Chadrick E Denlinger
- Department of Thoracic and Cardiovascular Surgery, University of Virginia, Charlottesville, Virginia 22908-0679, USA
| | | | | | | |
Collapse
|