51
|
Yoon KB, Cho SY, An SJ, Park KR, Lee HJ, Yoon HS, Lee SM, Kim YC, Han SY. Characterization of the aminopyridine derivative KRC-180 as a JAK2 inhibitor. Oncol Lett 2017; 14:1347-1354. [PMID: 28789350 PMCID: PMC5529941 DOI: 10.3892/ol.2017.6353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 03/07/2017] [Indexed: 01/29/2023] Open
Abstract
Janus kinase 2 (JAK2) is a non-receptor tyrosine kinase that regulates the signal transducer and activator of transcription (STAT) signaling pathway. Deregulation of JAK2 signaling has previously been observed in hematologic malignancies, including erythroleukemia. In the present study, an aminopyridine derivative compound, KRC-180, exhibited direct inhibition of the JAK2 protein at the catalytic site, as demonstrated using in vitro kinase activity assays and docking analyses. In addition, KRC-180 reduced the phosphorylation of STAT3 and STAT5, downstream signaling molecules of JAK2. The growth of HEL92.1.7 erythroleukemia cells harboring a constitutively activated form of JAK2 was suppressed by KRC-180 treatment; KRC-180 induced apoptotic cell death and cell cycle arrest. The results of the present study indicate that KRC-180 is a JAK2 inhibitor with anti-leukemic properties.
Collapse
Affiliation(s)
- Kyoung Bin Yoon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| | - Sung Yun Cho
- Bio-organic Science Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Su Jin An
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| | - Kyeong Ryang Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| | - Hyo Jeong Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| | - Hae Sung Yoon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| | - Sun-Mi Lee
- School of Life Sciences, Gwangju Institute of Science & Technology, Gwangju 61005, Republic of Korea
| | - Yong-Chul Kim
- School of Life Sciences, Gwangju Institute of Science & Technology, Gwangju 61005, Republic of Korea
| | - Sun-Young Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| |
Collapse
|
52
|
Chen Z, Soutto M, Rahman B, Fazili MW, Peng D, Blanca Piazuelo M, Chen H, Kay Washington M, Shyr Y, El-Rifai W. Integrated expression analysis identifies transcription networks in mouse and human gastric neoplasia. Genes Chromosomes Cancer 2017; 56:535-547. [PMID: 28281307 DOI: 10.1002/gcc.22456] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 02/24/2017] [Accepted: 02/27/2017] [Indexed: 12/16/2022] Open
Abstract
Gastric cancer (GC) is a leading cause of cancer-related deaths worldwide. The Tff1 knockout (KO) mouse model develops gastric lesions that include low-grade dysplasia (LGD), high-grade dysplasia (HGD), and adenocarcinomas. In this study, we used Affymetrix microarrays gene expression platforms for analysis of molecular signatures in the mouse stomach [Tff1-KO (LGD) and Tff1 wild-type (normal)] and human gastric cancer tissues and their adjacent normal tissue samples. Combined integrated bioinformatics analysis of mouse and human datasets indicated that 172 genes were consistently deregulated in both human gastric cancer samples and Tff1-KO LGD lesions (P < .05). Using Ingenuity pathway analysis, these genes mapped to important transcription networks that include MYC, STAT3, β-catenin, RELA, NFATC2, HIF1A, and ETS1 in both human and mouse. Further analysis demonstrated activation of FOXM1 and inhibition of TP53 transcription networks in human gastric cancers but not in Tff1-KO LGD lesions. Using real-time RT-PCR, we validated the deregulated expression of several genes (VCAM1, BGN, CLDN2, COL1A1, COL1A2, COL3A1, EpCAM, IFITM1, MMP9, MMP12, MMP14, PDGFRB, PLAU, and TIMP1) that map to altered transcription networks in both mouse and human gastric neoplasia. Our study demonstrates significant similarities in deregulated transcription networks in human gastric cancer and gastric tumorigenesis in the Tff1-KO mouse model. The data also suggest that activation of MYC, STAT3, RELA, and β-catenin transcription networks could be an early molecular step in gastric carcinogenesis.
Collapse
Affiliation(s)
- Zheng Chen
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, 37232.,Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Mohammed Soutto
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, 37232.,Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Bushra Rahman
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Muhammad W Fazili
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - DunFa Peng
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Maria Blanca Piazuelo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Gastroenterology, Hepatology, & Nutrition, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Heidi Chen
- Center of Quantitative Sciences, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN, 37232
| | - M Kay Washington
- Department of Pathology, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN, 37232
| | - Yu Shyr
- Center of Quantitative Sciences, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN, 37232
| | - Wael El-Rifai
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, 37232.,Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| |
Collapse
|
53
|
Sokolova O, Naumann M. NF-κB Signaling in Gastric Cancer. Toxins (Basel) 2017; 9:toxins9040119. [PMID: 28350359 PMCID: PMC5408193 DOI: 10.3390/toxins9040119] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/14/2017] [Accepted: 03/22/2017] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is a leading cause of cancer death worldwide. Diet, obesity, smoking and chronic infections, especially with Helicobacter pylori, contribute to stomach cancer development. H. pylori possesses a variety of virulence factors including encoded factors from the cytotoxin-associated gene pathogenicity island (cagPAI) or vacuolating cytotoxin A (VacA). Most of the cagPAI-encoded products form a type 4 secretion system (T4SS), a pilus-like macromolecular transporter, which translocates CagA into the cytoplasm of the host cell. Only H. pylori strains carrying the cagPAI induce the transcription factor NF-κB, but CagA and VacA are dispensable for direct NF-κB activation. NF-κB-driven gene products include cytokines/chemokines, growth factors, anti-apoptotic factors, angiogenesis regulators and metalloproteinases. Many of the genes transcribed by NF-κB promote gastric carcinogenesis. Since it has been shown that chemotherapy-caused cellular stress could elicit activation of the survival factor NF-κB, which leads to acquisition of chemoresistance, the NF-κB system is recommended for therapeutic targeting. Research is motivated for further search of predisposing conditions, diagnostic markers and efficient drugs to improve significantly the overall survival of patients. In this review, we provide an overview about mechanisms and consequences of NF-κB activation in gastric mucosa in order to understand the role of NF-κB in gastric carcinogenesis.
Collapse
Affiliation(s)
- Olga Sokolova
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, Magdeburg 39120, Germany.
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, Magdeburg 39120, Germany.
| |
Collapse
|
54
|
Cirincione R, Di Maggio FM, Forte GI, Minafra L, Bravatà V, Castiglia L, Cavalieri V, Borasi G, Russo G, Lio D, Messa C, Gilardi MC, Cammarata FP. High-Intensity Focused Ultrasound- and Radiation Therapy-Induced Immuno-Modulation: Comparison and Potential Opportunities. ULTRASOUND IN MEDICINE & BIOLOGY 2017; 43:398-411. [PMID: 27780661 DOI: 10.1016/j.ultrasmedbio.2016.09.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 09/20/2016] [Accepted: 09/21/2016] [Indexed: 05/12/2023]
Abstract
In recent years, high-intensity focused ultrasound (HIFU) has emerged as a new and promising non-invasive and non-ionizing ablative technique for the treatment of localized solid tumors. Extensive pre-clinical and clinical studies have evidenced that, in addition to direct destruction of the primary tumor, HIFU-thermoablation may elicit long-term systemic host anti-tumor immunity. In particular, an important consequence of HIFU treatment includes the release of tumor-associated antigens (TAAs), the secretion of immuno-suppressing factors by cancer cells and the induction of cytotoxic T lymphocyte (CTL) activity. Radiation therapy (RT) is the main treatment modality used for many types of tumors and about 50% of all cancer patients receive RT, often used in combination with surgery and chemotherapy. It is well known that RT can modulate anti-tumor immune responses, modifying micro-environment and stimulating inflammatory factors that can greatly affect cell invasion, bystander effects, radiation tissue complications (such as fibrosis), genomic instability and thus, intrinsic cellular radio-sensitivity. To date, various combined therapeutic strategies (such as immuno-therapy) have been performed in order to enhance RT success in treating locally advanced and recurrent tumors. Recent works suggested the combined use of HIFU and RT treatments to increase the tumor cell radio-sensitivity, in order to synergize the effects reaching the maximum results with minimal doses of ionizing radiation (IR). Here, we highlight the opposite immuno-modulation roles of RT and HIFU, providing scientific reasons to test, by experimental approaches, the use of HIFU immune-stimulatory capacity to improve tumor radio-sensitivity, to reduce the RT induced inflammatory response and to decrease the dose-correlated side effects in normal tissues.
Collapse
Affiliation(s)
| | - Federica Maria Di Maggio
- IBFM CNR, Cefalù, Palermo, Italy; Department of Pathobiology and Medical Biotechnologies, University of Palermo, Palermo, Italy
| | | | | | - Valentina Bravatà
- IBFM CNR, Cefalù, Palermo, Italy; Department of Pathobiology and Medical Biotechnologies, University of Palermo, Palermo, Italy
| | | | - Vincenzo Cavalieri
- Laboratory of Molecular Biology and Functional Genomics, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | | | | | - Domenico Lio
- Department of Pathobiology and Medical Biotechnologies, University of Palermo, Palermo, Italy
| | - Cristina Messa
- IBFM CNR, Cefalù, Palermo, Italy; Department of Health Sciences, Tecnomed Foundation, University of Milano-Bicocca, Milan, Italy; Nuclear Medicine Center, San Gerardo Hospital, Monza, Italy
| | - Maria Carla Gilardi
- IBFM CNR, Cefalù, Palermo, Italy; Department of Health Sciences, Tecnomed Foundation, University of Milano-Bicocca, Milan, Italy; Nuclear Medicine, San Raffaele Scientific Institute, Milan, Italy
| | | |
Collapse
|
55
|
Shi SY, Luk CT, Schroer SA, Kim MJ, Dodington DW, Sivasubramaniyam T, Lin L, Cai EP, Lu SY, Wagner KU, Bazinet RP, Woo M. Janus Kinase 2 (JAK2) Dissociates Hepatosteatosis from Hepatocellular Carcinoma in Mice. J Biol Chem 2017; 292:3789-3799. [PMID: 28100771 DOI: 10.1074/jbc.m116.752519] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 12/29/2016] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma is an end-stage complication of non-alcoholic fatty liver disease (NAFLD). Inflammation plays a critical role in the progression of non-alcoholic fatty liver disease and the development of hepatocellular carcinoma. However, whether steatosis per se promotes liver cancer, and the molecular mechanisms that control the progression in this disease spectrum remain largely elusive. The Janus kinase signal transducers and activators of transcription (JAK-STAT) pathway mediates signal transduction by numerous cytokines that regulate inflammation and may contribute to hepatocarcinogenesis. Mice with hepatocyte-specific deletion of JAK2 (L-JAK2 KO) develop extensive fatty liver spontaneously. We show here that this simple steatosis was insufficient to drive carcinogenesis. In fact, L-JAK2 KO mice were markedly protected from chemically induced tumor formation. Using the methionine choline-deficient dietary model to induce steatohepatitis, we found that steatohepatitis development was completely arrested in L-JAK2 KO mice despite the presence of steatosis, suggesting that JAK2 is the critical factor required for inflammatory progression in the liver. In line with this, L-JAK2 KO mice exhibited attenuated inflammation after chemical carcinogen challenge. This was associated with increased hepatocyte apoptosis without elevated compensatory proliferation, thus thwarting expansion of transformed hepatocytes. Taken together, our findings identify an indispensable role of JAK2 in hepatocarcinogenesis through regulating critical inflammatory pathways. Targeting the JAK-STAT pathway may provide a novel therapeutic option for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Sally Yu Shi
- From the Toronto General Hospital Research Institute, Toronto, Ontario M5G 2C4, Canada.,the Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Cynthia T Luk
- From the Toronto General Hospital Research Institute, Toronto, Ontario M5G 2C4, Canada.,the Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Stephanie A Schroer
- From the Toronto General Hospital Research Institute, Toronto, Ontario M5G 2C4, Canada
| | - Min Jeong Kim
- From the Toronto General Hospital Research Institute, Toronto, Ontario M5G 2C4, Canada.,the Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Korea
| | - David W Dodington
- From the Toronto General Hospital Research Institute, Toronto, Ontario M5G 2C4, Canada
| | - Tharini Sivasubramaniyam
- From the Toronto General Hospital Research Institute, Toronto, Ontario M5G 2C4, Canada.,the Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Lauren Lin
- the Department of Nutritional Sciences, University of Toronto, Toronto, Ontario M5S 3E2, Canada
| | - Erica P Cai
- From the Toronto General Hospital Research Institute, Toronto, Ontario M5G 2C4, Canada.,the Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Shun-Yan Lu
- From the Toronto General Hospital Research Institute, Toronto, Ontario M5G 2C4, Canada
| | - Kay-Uwe Wagner
- the Eppley Institute for Research in Cancer and Allied Diseases and the Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska 68198-6805, and
| | - Richard P Bazinet
- the Department of Nutritional Sciences, University of Toronto, Toronto, Ontario M5S 3E2, Canada
| | - Minna Woo
- From the Toronto General Hospital Research Institute, Toronto, Ontario M5G 2C4, Canada, .,the Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,the Division of Endocrinology, Department of Medicine, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| |
Collapse
|
56
|
Interleukin-27 inhibits malignant behaviors of pancreatic cancer cells by targeting M2 polarized tumor associated macrophages. Cytokine 2017; 89:194-200. [DOI: 10.1016/j.cyto.2015.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 11/20/2015] [Accepted: 12/02/2015] [Indexed: 12/18/2022]
|
57
|
Shen W, Yuan Y, Zhao M, Li J, Xu J, Lou G, Zheng J, Bu S, Guo J, Xi Y. Novel long non-coding RNA GACAT3 promotes gastric cancer cell proliferation through the IL-6/STAT3 signaling pathway. Tumour Biol 2016; 37:14895-14902. [PMID: 27644247 DOI: 10.1007/s13277-016-5372-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 09/08/2016] [Indexed: 12/22/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) play an important role in cancer occurrence and development. We previously demonstrated that lncRNA gastric cancer-associated transcript 3 (GACAT3) was positively correlated with TNM stages, tumor size, and distant metastasis of patients with gastric cancer. However, the role of GACAT3 in gastric cancer remains unclear. In this study, to investigate its function, we synthesized small interference RNAs (siRNAs) against GACTA3 and developed a GACAT3 overexpression vector (pcDNA3-GACAT3), respectively. The siRNA-mediated knockdown of GACAT3 significantly decreased cell proliferation of the gastric cancer HGC-27 cells, in which GACAT3 is overexpressed. Furthermore, GACAT3 overexpression in gastric cancer SGC-7901 cells promoted cell growth. Moreover, GACAT3 expression in HGC-27 cells was greatly upregulated by IL-6 treatment in a concentration-dependent manner. In contrast, siRNA-mediated knockdown of STAT3 decreased GACAT3 expression even in the presence of IL-6. These results demonstrated that as a downstream target of the IL6/STAT3 signaling, lncRNA GACAT3 promotes gastric cancer cell growth suggesting that GACAT3 is an inflammatory response gene and may be served as a valuable potential target for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Wanjing Shen
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China
| | - Yanyan Yuan
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China
| | - Ming Zhao
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China
| | - Jiahui Li
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China
| | - Jin Xu
- Department of Otorhinolaryngology, Ningbo No.7 Hospital, Ningbo, 315211, China
| | - Guoying Lou
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China
| | - Jiachen Zheng
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China
| | - Shizhong Bu
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China
| | - Junming Guo
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China
| | - Yang Xi
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China.
| |
Collapse
|
58
|
Geng L, Li X, Zhou X, Fang X, Yuan D, Wang X. WP1066 exhibits antitumor efficacy in nasal‑type natural killer/T-cell lymphoma cells through downregulation of the STAT3 signaling pathway. Oncol Rep 2016; 36:2868-2874. [PMID: 27633398 DOI: 10.3892/or.2016.5091] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 07/11/2016] [Indexed: 11/06/2022] Open
Abstract
Nasal-type natural killer/T-cell lymphoma (nasal NKTCL) is an aggressive hematological neoplasm with poor prognosis, and its incidence is higher in Asia than in Western countries. Increasing evidence suggests that aberrant activation of signal transducers and activators of transcription 3 (STAT3) is related to numerous malignancies. However, the involvement of STAT3 in the pathogenesis of nasal NKTCL is poorly understood. In this study, immunohistochemistry (IHC) showed that 21/28 (75.0%) nasal NKTCL tissues harbored constitutively expression of phospho‑STAT3 (p‑STAT3) which was positively correlated with the Ki‑67 levels (P﹤0.05). Immunofluorescence (IF) also detected p‑STAT3 expression in SNK6 cells (NKTCL cell line). Furthermore, WP1066 (a novel selective STAT3 inhibitor) was able to inhibit proliferation and induce apoptosis of SNK6 cells. Moreover, western blot analysis and RT‑PCR demonstrated that WP1066 downregulated the protein and mRNA expressions of the pro‑survival molecules (including c‑Myc, cyclin D1, and Bcl‑2) in SNK6 cells. These results suggested that STAT3 activation represents a potential target in nasal NKTCL. WP1066 may be a promising agent in antitumor therapy against nasal NKTCL.
Collapse
Affiliation(s)
- Lingyun Geng
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xinyu Li
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xiaosheng Fang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Dai Yuan
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
59
|
Wu X, Cao Y, Xiao H, Li C, Lin J. Bazedoxifene as a Novel GP130 Inhibitor for Pancreatic Cancer Therapy. Mol Cancer Ther 2016; 15:2609-2619. [PMID: 27535971 DOI: 10.1158/1535-7163.mct-15-0921] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 08/04/2016] [Indexed: 01/06/2023]
Abstract
The IL6/GP130/STAT3 pathway is crucial for tumorigenesis in multiple cancer types, including pancreatic cancer, and presents as a viable target for cancer therapy. We reported Bazedoxifene, which is approved as a selective estrogen modulator by FDA, as a novel inhibitor of IL6/GP130 protein-protein interactions using multiple ligand simultaneous docking and drug repositioning approaches. STAT3 is one of the major downstream effectors of IL6/GP130. Here, we observed Bazedoxifene inhibited STAT3 phosphorylation and STAT3 DNA binding, induced apoptosis, and suppressed tumor growth in pancreatic cancer cells with persistent IL6/GP130/STAT3 signaling in vitro and in vivo In addition, IL6, but not INFγ, rescued Bazedoxifene-mediated reduction of cell viability. Bazedoxifene also inhibited STAT3 phosphorylation induced by IL6 and IL11, but not by OSM or STAT1 phosphorylation induced by INFγ in pancreatic cancer cells, suggesting that Bazedoxifene inhibits the GP130/STAT3 pathway mediated by IL6 and IL11. Furthermore, Bazedoxifene combined with paclitaxel or gemcitabine synergistically inhibited cell viability and cell migration in pancreatic cancer cells. These results indicate that Bazedoxifene is a potential agent and can generate synergism when combined with conventional chemotherapy in human pancreatic cancer cells and tumor xenograft in mice. Therefore, our results support that Bazedoxifene as a novel inhibitor of GP130 signaling and may be a potential and safe therapeutic agent for human pancreatic cancer therapy. Mol Cancer Ther; 15(11); 2609-19. ©2016 AACR.
Collapse
Affiliation(s)
- Xiaojuan Wu
- Department of Pediatric Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Department of Pediatrics, Center for Childhood Cancer and Blood Diseases, the Research Institute at Nationwide Children's Hospital, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Yang Cao
- Department of Pediatrics, Center for Childhood Cancer and Blood Diseases, the Research Institute at Nationwide Children's Hospital, College of Medicine, The Ohio State University, Columbus, Ohio.,Department of Hematology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Xiao
- Department of Pediatrics, Center for Childhood Cancer and Blood Diseases, the Research Institute at Nationwide Children's Hospital, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Chenglong Li
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Jiayuh Lin
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
60
|
Yang A, Fan H, Zhao Y, Zha X, Zhang H, Hu Z, Tu P. Huaier aqueous extract inhibits proliferation and metastasis of tuberous sclerosis complex cell models through downregulation of JAK2/STAT3 and MAPK signaling pathways. Oncol Rep 2016; 36:1491-8. [PMID: 27461043 DOI: 10.3892/or.2016.4969] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/18/2016] [Indexed: 11/06/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is a genetic disorder with formation of benign tumors in many different organs. It has attracted increasing attention from researchers to search for therapeutic drugs for TSC patients. Traditional Chinese medicine (TCM) has become an important source for finding antitumor drugs. Trametes robiniophila Μurr. (Huaier) is a kind of officinal fungi in China and has been applied in TCM for approximately 1,600 years. A large number of clinical applications have revealed that Huaier has good antitumor effect. In this study, we have investigated the effects of Huaier aqueous extract on two TSC cell models, including inhibition of proliferation, induction of apoptosis, cell cycle arrest, and anti-metastasis. We demonstrated that Huaier aqueous extract inhibited JAK2/STAT3 and MAPK signaling pathways in a dose-dependent manner. Therefore, based on the low toxicity and the multi-targets of Huaier treatment, Huaier may be a promising therapeutic drug for TSC.
Collapse
Affiliation(s)
- Ailin Yang
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Haitao Fan
- College of Bioengineering, Beijing Polytechnic, Beijing 100029, P.R. China
| | - Yunfang Zhao
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Xiaojun Zha
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Hongbing Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| | - Zhongdong Hu
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Pengfei Tu
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| |
Collapse
|
61
|
Riquelme I, Saavedra K, Espinoza JA, Weber H, García P, Nervi B, Garrido M, Corvalán AH, Roa JC, Bizama C. Molecular classification of gastric cancer: Towards a pathway-driven targeted therapy. Oncotarget 2016; 6:24750-79. [PMID: 26267324 PMCID: PMC4694793 DOI: 10.18632/oncotarget.4990] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/17/2015] [Indexed: 02/07/2023] Open
Abstract
Gastric cancer (GC) is the third leading cause of cancer mortality worldwide. Although surgical resection is a potentially curative approach for localized cases of GC, most cases of GC are diagnosed in an advanced, non-curable stage and the response to traditional chemotherapy is limited. Fortunately, recent advances in our understanding of the molecular mechanisms that mediate GC hold great promise for the development of more effective treatment strategies. In this review, an overview of the morphological classification, current treatment approaches, and molecular alterations that have been characterized for GC are provided. In particular, the most recent molecular classification of GC and alterations identified in relevant signaling pathways, including ErbB, VEGF, PI3K/AKT/mTOR, and HGF/MET signaling pathways, are described, as well as inhibitors of these pathways. An overview of the completed and active clinical trials related to these signaling pathways are also summarized. Finally, insights regarding emerging stem cell pathways are described, and may provide additional novel markers for the development of therapeutic agents against GC. The development of more effective agents and the identification of biomarkers that can be used for the diagnosis, prognosis, and individualized therapy for GC patients, have the potential to improve the efficacy, safety, and cost-effectiveness for GC treatments.
Collapse
Affiliation(s)
- Ismael Riquelme
- Department of Pathology, School of Medicine, Universidad de La Frontera, CEGIN-BIOREN, Temuco, Chile
| | - Kathleen Saavedra
- Department of Pathology, School of Medicine, Universidad de La Frontera, CEGIN-BIOREN, Temuco, Chile
| | - Jaime A Espinoza
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,UC-Center for Investigational Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Helga Weber
- Department of Pathology, School of Medicine, Universidad de La Frontera, CEGIN-BIOREN, Temuco, Chile
| | - Patricia García
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,UC-Center for Investigational Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bruno Nervi
- UC-Center for Investigational Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Hematology Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcelo Garrido
- UC-Center for Investigational Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Hematology Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandro H Corvalán
- UC-Center for Investigational Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Hematology Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDIS), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Carlos Roa
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,UC-Center for Investigational Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDIS), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina Bizama
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,UC-Center for Investigational Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
62
|
Poh AR, O'Donoghue RJJ, Ernst M, Putoczki TL. Mouse models for gastric cancer: Matching models to biological questions. J Gastroenterol Hepatol 2016; 31:1257-72. [PMID: 26809278 PMCID: PMC5324706 DOI: 10.1111/jgh.13297] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 01/12/2016] [Accepted: 01/14/2016] [Indexed: 02/06/2023]
Abstract
Gastric cancer is the third leading cause of cancer-related mortality worldwide. This is in part due to the asymptomatic nature of the disease, which often results in late-stage diagnosis, at which point there are limited treatment options. Even when treated successfully, gastric cancer patients have a high risk of tumor recurrence and acquired drug resistance. It is vital to gain a better understanding of the molecular mechanisms underlying gastric cancer pathogenesis to facilitate the design of new-targeted therapies that may improve patient survival. A number of chemically and genetically engineered mouse models of gastric cancer have provided significant insight into the contribution of genetic and environmental factors to disease onset and progression. This review outlines the strengths and limitations of current mouse models of gastric cancer and their relevance to the pre-clinical development of new therapeutics.
Collapse
Affiliation(s)
- Ashleigh R Poh
- Department of Medical BiologyUniversity of MelbourneMelbourneVictoriaAustralia
- The Walter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
| | - Robert J J O'Donoghue
- School of Cancer MedicineLa Trobe University, Olivia Newton‐John Cancer Research InstituteMelbourneVictoriaAustralia
| | - Matthias Ernst
- School of Cancer MedicineLa Trobe University, Olivia Newton‐John Cancer Research InstituteMelbourneVictoriaAustralia
| | - Tracy L Putoczki
- Department of Medical BiologyUniversity of MelbourneMelbourneVictoriaAustralia
- The Walter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
| |
Collapse
|
63
|
Role of interleukin-6 in cancer progression and therapeutic resistance. Tumour Biol 2016; 37:11553-11572. [DOI: 10.1007/s13277-016-5098-7] [Citation(s) in RCA: 430] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 05/22/2016] [Indexed: 02/07/2023] Open
|
64
|
|
65
|
Ji HG, Piao JY, Kim SJ, Kim DH, Lee HN, Na HK, Surh YJ. Docosahexaenoic acid inhibits Helicobacter pylori-induced STAT3 phosphorylation through activation of PPARγ. Mol Nutr Food Res 2016; 60:1448-57. [PMID: 27079734 DOI: 10.1002/mnfr.201600009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/08/2016] [Accepted: 03/09/2016] [Indexed: 01/09/2023]
Abstract
SCOPE The health beneficial effects of docosahexaenoic acid (DHA) have been attributed to its anti-inflammatory properties. However, the molecular mechanism underlying anti-inflammatory effects of DHA remains largely elusive. METHODS AND RESULTS In the present study, DHA was found to suppress the phosphorylation and nuclear translocation of signal transducer and activator of transcription 3 (STAT3) induced by Helicobacter pylori infection in human gastric cancer AGS cells. Notably, DHA induced expression of suppressor of cytokine signaling 3 (SOCS3), a negative regulator of STAT3. Knockdown of SOCS3 abolished the suppressive effect of DHA on STAT3(Tyr705) phosphorylation induced by H. pylori infection. DHA also induced nuclear translocation, DNA binding, and transcriptional activities of peroxisome proliferator-activated receptor gamma (PPARγ) in AGS cells. Knockdown of PPARγ inhibited the transcription of SOCS3 and attenuated the suppressive effect of DHA on phosphorylation of STAT3(Tyr705) induced by H. pylori. The PPARγ antagonist bisphenol A diglycidyl ether also mitigated the suppressive effect of DHA on H. pylori-induced phosphorylation of STAT3(Tyr705) . In addition, DHA inhibited the expression of c-Myc, which was attenuated in the AGS cells harboring SOCS3 specific siRNA. DHA also markedly decreased anchorage-independent growth of AGS cells infected by H. pylori. CONCLUSION DHA inhibits H. pylori-induced STAT3 phosphorylation in a PPARγ/SOCS3-dependent manner.
Collapse
Affiliation(s)
- Hyeon-Geun Ji
- Cancer Research Institute, Seoul National University, Seoul, South Korea.,Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Juan-Yu Piao
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Su-Jung Kim
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Sciences, Seoul, South Korea
| | - Do-Hee Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Ha-Na Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Hye-Kyung Na
- Department of Food and Nutrition, College of Human Ecology, Sungshin Women's University, Seoul, South Korea
| | - Young-Joon Surh
- Cancer Research Institute, Seoul National University, Seoul, South Korea.,Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Sciences, Seoul, South Korea
| |
Collapse
|
66
|
Song B, Zhan H, Bian Q, Gu J. Piperlongumine inhibits gastric cancer cells via suppression of the JAK1,2/STAT3 signaling pathway. Mol Med Rep 2016; 13:4475-80. [PMID: 27053336 DOI: 10.3892/mmr.2016.5091] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 03/21/2016] [Indexed: 11/06/2022] Open
Abstract
Piperlongumine (PL), a major active component of long peppers, has been reported to possess anti‑cancer properties; however, its effect on gastric cancer (GC) has remained to be demonstrated. The present study assessed the effects of PL on the MKN45 and AGS GC cell lines and explored the underlying mechanisms. An MTT assay revealed that PL suppressed the proliferation of GC cells, while flow cytometric analysis showed that PL inhibited cell cycle progression. Furthermore, Transwell assays revealed the inhibitory effects of PL on the invasion and migration of GC cells. In addition, PL reduced the phosphorylation of Janus kinase (JAK)1, JAK2 and signal transducer and activator of transcription (STAT)3 in a concentration‑dependent manner, as indicated by western blot analysis, and decreased the expression of STAT3‑dependent tumor‑associated genes in GC cells, as revealed by PCR analysis. In conclusion, the present study was the first, to the best of our knowledge, to reveal the efficacy of PL against GC. The consumption of long peppers is therefore recommended for the prevention and treatment of GC, and PL may be a promising candidate drug for treating GC.
Collapse
Affiliation(s)
- Baoji Song
- Department of General Surgery, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Hongjie Zhan
- Department of Gastric Cancer, Tianjin Cancer Hospital, Key Laboratory of Cancer Prevention and Treatment of Tianjin, Tianjin Medical University, Tianjin 300060, P.R. China
| | - Quan Bian
- Department of General Surgery, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Junping Gu
- Department of General Surgery, Tianjin Hospital, Tianjin 300211, P.R. China
| |
Collapse
|
67
|
Xu Z, Sun Y, Guo Y, Qin G, Mu S, Fan R, Wang B, Gao W, Wu H, Wang G, Zhang Z. NF-YA promotes invasion and angiogenesis by upregulating EZH2-STAT3 signaling in human melanoma cells. Oncol Rep 2016; 35:3630-8. [PMID: 27109360 DOI: 10.3892/or.2016.4761] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 02/22/2016] [Indexed: 11/06/2022] Open
Abstract
The process of angiogenesis is essential for tumor development and metastasis. Vascular endothelial growth factor (VEGF), which is overexpressed in most human cancers, has been demonstrated to be a major modulator of angiogenesis. Thus, inhibition of VEGF signaling has the potential for tumor anti-angiogenic therapy. Signal transducer and activator of transcription-3 (STAT3) is a key regulator for angiogenesis by directly binding to the VEGF promoter to upregulate its transcription. Several factors can enhance STAT3 activity to affect angiogenesis. Here, we found that overexpression of nuclear transcription factor-Y alpha (NF-YA) gene could promote cell invasion and angiogenesis accompanying the increase of STAT3 signaling in human melanoma cells. Moreover, the expression and secretion of VEGF was also found to be upregulated by the overexpression of NF-YA gene in melanoma cells. The STAT3 inhibitor was able to attenuate the upregulation of VEGF induced by NF-YA overexpression. Enhancer of zeste homolog 2 (EZH2), the catalytic subunit of the Polycomb repressive complex 2, enhances STAT3 activity by mediating its lysine methylation. We also showed that NF-YA upregulated the expression of EZH2 and NF-YA‑induced angiogenesis could be inhibited by EZH2 knockdown. Taken together, these findings indicate that overexpression of NF-YA contributes to tumor angiogenesis through EZH2-STAT3 signaling in human melanoma cells, highlighting NF-YA as a potential therapeutic target in human melanoma.
Collapse
Affiliation(s)
- Zihan Xu
- Department of Burns and Plastic Surgery, Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Yaowen Sun
- Department of Burns and Plastic Surgery, Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Yadong Guo
- Department of Burns and Plastic Surgery, Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Gaoping Qin
- Department of Burns and Plastic Surgery, Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Shengzhi Mu
- Department of Burns and Plastic Surgery, Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Ronghui Fan
- Department of Burns and Plastic Surgery, Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Benfeng Wang
- Department of Burns and Plastic Surgery, Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Wenjie Gao
- Department of Burns and Plastic Surgery, Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Hangli Wu
- Department of Burns and Plastic Surgery, Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Guodong Wang
- Department of Burns and Plastic Surgery, Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Zhenxin Zhang
- Department of Burns and Plastic Surgery, Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| |
Collapse
|
68
|
Jácome AA, Coutinho AK, Lima EM, Andrade AC, Santos JSD. Personalized medicine in gastric cancer: Where are we and where are we going? World J Gastroenterol 2016; 22:1160-71. [PMID: 26811654 PMCID: PMC4716027 DOI: 10.3748/wjg.v22.i3.1160] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 09/22/2015] [Accepted: 11/19/2015] [Indexed: 02/06/2023] Open
Abstract
Despite improvements in adjuvant therapies for gastric cancer in recent years, the disease is characterized by high recurrence rates and a dismal prognosis. The major improvement in the treatment of recurrent or metastatic gastric cancer in recent years has been the incorporation of trastuzumab, a monoclonal antibody that inhibits human epidermal growth factor receptor 2 (HER2) heterodimerization, after the demonstrated predictive value of the overexpression and/or amplification of this receptor. Beyond HER2, other genetic abnormalities have been identified, and these mutations may be targetable by tyrosine kinase inhibitors or monoclonal antibodies. The demonstration of four distinct molecular subtypes of gastric cancer by the Cancer Genome Atlas study highlight the enormous heterogeneity of the disease and its complex interplay between genetic and epigenetic alterations and provide a roadmap to implement genome-guided personalized therapy in gastric cancer. In the present review, we aim to discuss, from a clinical point of view, the genomic landscape of gastric cancer described in recent studies, the therapeutic insights derived from these findings, and the clinical trials that have been conducted and those in progress that take into account tailored therapies for gastric cancer.
Collapse
|
69
|
Zhao G, Zhu G, Huang Y, Zheng W, Hua J, Yang S, Zhuang J, Ye J. IL-6 mediates the signal pathway of JAK-STAT3-VEGF-C promoting growth, invasion and lymphangiogenesis in gastric cancer. Oncol Rep 2016; 35:1787-95. [PMID: 26750536 DOI: 10.3892/or.2016.4544] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 12/11/2015] [Indexed: 12/30/2022] Open
Abstract
Gastric cancer shows the highest invasive and metastasis features, especially lymph metastasis, which is closely associated with poor prognosis of gastric cancer. Although there is evidence that interleukin-6 (IL-6) can promote gastric cancer progression, the underlying specific mechanisms and the mechanisms of gastric cancer lymphangiogenesis are largely unknown. In the present study, we explore whether IL-6 could promote the proliferation and invasion activity of gastric cancer cells, and whether IL-6 mediating VEGF-C production affected the lymphangiogenesis in gastric cancer cells. Our results revealed that IL-6 and its receptors (IL-6 and gp130) are broadly expressed in various gastric cancer cell lines including SGC-7901, MGC, MKN-28 and AGS. Exogenous IL-6 increased the ability of gastric cancer cell proliferation and invasion, which could be weakened by AG490. in addition, exogenous IL-6 promoted the VEGF-C production of gastric cancer cells and the lymphangiogenesis of HDLECs. As we expected, AG490 was able to reduce these effects. Western blot analysis showed that IL-6 increased JKA, STAT3, p-STAT3 and VEGF-C protein levels in the gastric cancer cells. However, the JKA, STAT3, p-STAT3 and VEGF-C protein expression levels were inhibited by AG490. Our data suggested that IL-6 mediates the singnal pathway of JAK-STAT3-VEGF-C promoting the growth, invasion and lymphangiogenesis in gastric cancer. Thus, IL-6 and its related signal pathways may be a promising target for treatment of gastric cancer growth and lymphangiogenesis.
Collapse
Affiliation(s)
- Guibin Zhao
- Department of Oncology Surgery, Mindong Hospital Affiliated to Fujian Medical University, Fuan, Fujian, P.R. China
| | - Guangwei Zhu
- Department of Gastrointestinal Surgery, First Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Yongjian Huang
- Department of Gastrointestinal Surgery, First Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Wei Zheng
- Department of Gastrointestinal Surgery, First Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Jin Hua
- Department of Gastrointestinal Surgery, First Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Shugang Yang
- Department of Gastrointestinal Surgery, First Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Jinfu Zhuang
- Department of Gastrointestinal Surgery, First Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Jianxin Ye
- Department of Gastrointestinal Surgery, First Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian, P.R. China
| |
Collapse
|
70
|
Mace TA, Shakya R, Elnaggar O, Wilson K, Komar HM, Yang J, Pitarresi JR, Young GS, Ostrowski MC, Ludwig T, Bekaii-Saab T, Bloomston M, Lesinski GB. Single agent BMS-911543 Jak2 inhibitor has distinct inhibitory effects on STAT5 signaling in genetically engineered mice with pancreatic cancer. Oncotarget 2015; 6:44509-22. [PMID: 26575024 PMCID: PMC4792572 DOI: 10.18632/oncotarget.6332] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 10/22/2015] [Indexed: 12/19/2022] Open
Abstract
The Jak/STAT pathway is activated in human pancreatic ductal adenocarcinoma (PDAC) and cooperates with mutant Kras to drive initiation and progression of PDAC in murine models. We hypothesized that the small-molecule Jak2 inhibitor (BMS-911543) would elicit anti-tumor activity against PDAC and decrease immune suppressive features of the disease. We used an aggressive genetically engineered PDAC model with mutant KrasG12D, tp53R270H, and Brca1 alleles (KPC-Brca1 mice). Mice with confirmed tumor burden were treated orally with vehicle or 30 mg/kg BMS-911543 daily for 14 days. Histologic analysis of pancreata from treated mice revealed fewer foci of adenocarcinoma and significantly decreased Ki67+ cells versus controls. In vivo administration of BMS-911543 significantly reduced pSTAT5 and FoxP3 positive cells within the pancreas, but did not alter STAT3 phosphorylation. Continuous dosing of KPC-Brca1 mice with BMS-911543 resulted in a median survival of 108 days, as compared to a median survival of 87 days in vehicle treated animals, a 23% increase (p = 0.055). In vitro experiments demonstrated that PDAC cell lines were poorly sensitive to BMS-911543, requiring high micromolar concentrations to achieve targeted inhibition of Jak/STAT signaling. Similarly, BMS-911543 had little in vitro effect on the viability of both murine and human PDAC-derived stellate cell lines. However, BMS-911543 potently inhibited phosphorylation of pSTAT3 and pSTAT5 at low micromolar doses in human PBMC and reduced in vitro differentiation of Foxp3+ T regulatory cells. These results indicate that single agent Jak2i deserves further study in preclinical models of PDAC and has distinct inhibitory effects on STAT5 mediated signaling.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/enzymology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/pathology
- Cell Survival/drug effects
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Forkhead Transcription Factors/metabolism
- Genes, BRCA1
- Genes, p53
- Genes, ras
- Genetic Predisposition to Disease
- Heterocyclic Compounds, 3-Ring/pharmacology
- Janus Kinase 2/antagonists & inhibitors
- Janus Kinase 2/metabolism
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Mice, Transgenic
- Molecular Targeted Therapy
- Mutation
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/enzymology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/pathology
- Phenotype
- Phosphorylation
- Protein Kinase Inhibitors/pharmacology
- STAT5 Transcription Factor/metabolism
- Signal Transduction/drug effects
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/pathology
- Time Factors
- Tumor Burden
Collapse
Affiliation(s)
- Thomas A. Mace
- Division of Medical Oncology, Department of Internal Medicine, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Reena Shakya
- Comprehensive Cancer Center, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Omar Elnaggar
- Division of Medical Oncology, Department of Internal Medicine, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Kristin Wilson
- Veterinary Biosciences, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Hannah M. Komar
- Division of Medical Oncology, Department of Internal Medicine, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jennifer Yang
- Division of Medical Oncology, Department of Internal Medicine, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jason R. Pitarresi
- Department of Molecular and Cellular Biochemistry, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Gregory S. Young
- Center for Biostatistics, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Michael C. Ostrowski
- Department of Molecular and Cellular Biochemistry, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Thomas Ludwig
- Department of Molecular and Cellular Biochemistry, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Tanios Bekaii-Saab
- Division of Medical Oncology, Department of Internal Medicine, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Mark Bloomston
- Division of Surgical Oncology, Department of Surgery, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Gregory B. Lesinski
- Division of Medical Oncology, Department of Internal Medicine, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
71
|
Delayed Administration of WP1066, an STAT3 Inhibitor, Ameliorates Radiation-Induced Lung Injury in Mice. Lung 2015; 194:67-74. [PMID: 26563331 DOI: 10.1007/s00408-015-9821-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 10/30/2015] [Indexed: 12/19/2022]
Abstract
PURPOSE The present study was designed to investigate the effects of WP1066, a specific inhibitor of STAT3 signaling, on radiation-induced lung injury in mice. METHODS C57BL/6J mice were subjected to a single thoracic irradiation of 15 Gy X-ray and WP1066 was administrated through intraperitoneal injection. The early and delayed treatment groups were treated with WP1066 during the first 2 weeks and the second 2 weeks, respectively. The therapeutic effects of WP1066 were evaluated by survival analysis, histological examination, and measurement of inflammatory parameters and collagen deposition. The activation of STAT3 pathway was also estimated by immunohistochemical staining and Western blotting. RESULTS Delayed treatment of WP1066, but not early treatment, prolonged survival time and prevented the development of radiation pneumonitis and the subsequent lung fibrosis in mice. WP1066 treatment also significantly suppressed the activation of STAT3 signaling in the irradiated lung tissues. CONCLUSIONS The activation of STAT3 pathway might play an important part in the pathogenesis of radiation-induced lung injury. The protective effects of delayed treatment of WP1066 suggested STAT3 signaling could be a therapeutic target for radiation pneumonitis.
Collapse
|
72
|
Niu Z, Wang M, Zhou L, Yao L, Liao Q, Zhao Y. Elevated GRP78 expression is associated with poor prognosis in patients with pancreatic cancer. Sci Rep 2015; 5:16067. [PMID: 26530532 PMCID: PMC4632002 DOI: 10.1038/srep16067] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 10/06/2015] [Indexed: 01/05/2023] Open
Abstract
Glucose-regulated protein 78 (GRP78) is a member of the heat-shock protein 70 family. We evaluated the expression of GRP78 using tissue microarray-based immunohistochemistry in tumor tissues and adjacent nontumor tissues from 180 pancreatic ductal adenocarcinoma (PDAC) patients. The associations between the expression levels of GRP78, clinicopathological factors, and overall survival were evaluated. The results showed that the expression of GRP78 was significantly higher in PDAC cells than in normal pancreatic duct cells within adjacent nontumor tissues (p < 0.05). The increased expression of GRP78 in the tumor tissues was significantly correlated with a higher T-stage (p < 0.05) and a shorter overall survival (OS, p < 0.05). In an in vitro study, the regulation of GRP78 in the PDAC cell lines affected the proliferation, migration, and invasion of PDAC cells through the regulation of CyclinD1, cyclin-dependent kinase (CDK) 4, CDK6, phospho-signal transducer, activator of transcription 3 (p-STAT3), janus kinase 2 (JAK2), ras homolog gene family member A (RhoA), Rho-associated kinase 1 (ROCK1), and sterile alpha motif domain containing protein 4 (Smad4). The present data suggest that GRP78 plays a crucial role in the proliferation, migration, and invasion of pancreatic cancer cells and may be a suitable prognostic marker in PDAC.
Collapse
Affiliation(s)
- Zheyu Niu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science &Peking Union Medical College, Beijing 100730, China
| | - Mengyi Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science &Peking Union Medical College, Beijing 100730, China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science &Peking Union Medical College, Beijing 100730, China
| | - Lutian Yao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science &Peking Union Medical College, Beijing 100730, China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science &Peking Union Medical College, Beijing 100730, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science &Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
73
|
Evaluation of selected interleukins in patients with different gastric neoplasms: a preliminary report. Sci Rep 2015; 5:14382. [PMID: 26486258 PMCID: PMC4613562 DOI: 10.1038/srep14382] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 08/21/2015] [Indexed: 12/25/2022] Open
Abstract
Abnormal interactions between cytokines may be an overlooked mechanism linking the development of different types of gastric neoplasms. In this study a comprehensive analysis of the systemic levels of interleukins (IL-1,IL-6, IL-8,IL-10 and IL-12) was performed in 75 patients with different gastric neoplasms (cancer, gastrointestinal stromal tumors, neuroendocrine neoplasms, lymphomas) and 40 healthy volunteers. Patients with gastric cancer (GC) have significantly higher IL-6 levels, and lower IL-8 and IL-10 concentrations, in comparison to controls and patients with other gastric neoplasms. Analogous results were observed in terms of IL-6/IL-8 and IL-6/IL-10 ratios, whose values were also higher in GC patients. In GC patients no associations were detected between the systemic levels/values of interleukins (ratios) and TNM staging. IL-6, IL-10, IL-6/IL-8 and IL-6/IL-10 ratios appeared to hold diagnostic potential in confirming/excluding the presence of GC. Their sensitivity/specificity in GC detection/exclusion was approximately 54–72%. In conclusion, disturbed systemic biochemical balance in multiple interleukins exists at the earliest stages of and appears to be specific to GC. The interleukin ratios proposed here seem to be more promising indicators of GC in humans than direct systemic levels of interleukins, and probably possess the potential to be applied as a supporting factor for techniques routinely used.
Collapse
|
74
|
Menheniott TR, Judd LM, Giraud AS. STAT3: a critical component in the response to Helicobacter pylori infection. Cell Microbiol 2015; 17:1570-82. [PMID: 26332850 DOI: 10.1111/cmi.12518] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 08/16/2015] [Accepted: 08/28/2015] [Indexed: 12/15/2022]
Abstract
STAT3 imparts a profound influence on both the epithelial and immune components of the gastric mucosa, and through regulation of key intracellular signal transduction events, is well placed to control inflammatory and oncogenic outcomes in the context of Helicobacter (H.) pylori infection. Here we review the roles of STAT3 in the host immune response to H. pylori infection, from both gastric mucosal and systemic perspectives, as well as alluding more specifically to STAT3-dependent mechanisms that might be exploited as drug targets.
Collapse
Affiliation(s)
- Trevelyan R Menheniott
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Louise M Judd
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew S Giraud
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
75
|
Yan R, Lin F, Hu C, Tong S. Association between STAT3 polymorphisms and cancer risk: a meta-analysis. Mol Genet Genomics 2015; 290:2261-70. [PMID: 26063618 DOI: 10.1007/s00438-015-1074-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 05/28/2015] [Indexed: 01/05/2023]
Abstract
Five polymorphisms, rs2293152, rs4796793, rs12949918, rs6503695, rs744166, in the STAT3 gene have been implicated in susceptibility to cancer, but the results were inconclusive. The aim of this meta-analysis is to investigate the association between the five polymorphisms and cancer risk. All eligible case-control studies published up to March 2015 were identified by searching PubMed, Web of Science, Wanfang, VIP, and CNKI. Effect sizes of odds ratio (OR) and 95 % confidence interval (95 % CI) were calculated by using a fixed- or random-effect model. A total of 15 articles were included. Overall, a significantly decreased risk was found for rs12949918 polymorphism (dominant model: OR = 0.83, 95 % CI: 0.75-0.91, recessive model: OR = 0.77, 95 % CI: 0.68-0.87, TC vs. TT: OR = 0.87, 95 % CI: 0.79-0.96, CC vs. TT: OR = 0.71, 95 % CI: 0.62-0.81), and for rs744166 polymorphism (recessive model: OR = 0.75, 95 % CI: 0.58-0.98; GG vs. AA: OR = 0.68, 95 % CI: 0.51-0.90), while there was no significant association for other three polymorphisms under all genetic models. In subgroup analysis by ethnicity, for rs12949918 polymorphism, similar results were detected among Caucasians, similarly, a significant decreased risk was observed in Asians under dominant and CC vs. TT model; for rs2293152 polymorphism, significant association was detected among Asians under recessive model. This meta-analysis suggests that the STAT3 rs12949918 and rs744166 polymorphisms, but not other three polymorphisms, may be an important protective factor for cancer.
Collapse
Affiliation(s)
- Ruicheng Yan
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.
| | - Fusheng Lin
- Department of General Surgery, Zhongshan Hospital of Xiamen University, Xiamen, 361004, Fujian Province, People's Republic of China
| | - Chao Hu
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Shilun Tong
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| |
Collapse
|
76
|
Targeting JAK kinase in solid tumors: emerging opportunities and challenges. Oncogene 2015; 35:939-51. [DOI: 10.1038/onc.2015.150] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 03/24/2015] [Accepted: 03/24/2015] [Indexed: 02/07/2023]
|
77
|
Di Maggio FM, Minafra L, Forte GI, Cammarata FP, Lio D, Messa C, Gilardi MC, Bravatà V. Portrait of inflammatory response to ionizing radiation treatment. J Inflamm (Lond) 2015; 12:14. [PMID: 25705130 PMCID: PMC4336767 DOI: 10.1186/s12950-015-0058-3] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 01/29/2015] [Indexed: 01/05/2023] Open
Abstract
Ionizing radiation (IR) activates both pro-and anti-proliferative signal pathways producing an imbalance in cell fate decision. IR is able to regulate several genes and factors involved in cell-cycle progression, survival and/or cell death, DNA repair and inflammation modulating an intracellular radiation-dependent response. Radiation therapy can modulate anti-tumour immune responses, modifying tumour and its microenvironment. In this review, we report how IR could stimulate inflammatory factors to affect cell fate via multiple pathways, describing their roles on gene expression regulation, fibrosis and invasive processes. Understanding the complex relationship between IR, inflammation and immune responses in cancer, opens up new avenues for radiation research and therapy in order to optimize and personalize radiation therapy treatment for each patient.
Collapse
Affiliation(s)
- Federica Maria Di Maggio
- />Department of Pathobiology and Medical and Forensic Biotechnologies, University of Palermo, Palermo, Italy
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
| | - Luigi Minafra
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
| | - Giusi Irma Forte
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
| | | | - Domenico Lio
- />Department of Pathobiology and Medical and Forensic Biotechnologies, University of Palermo, Palermo, Italy
| | - Cristina Messa
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
- />Department of Health Sciences, Tecnomed Foundation, University of Milano-Bicocca, Milan, Italy
- />Nuclear Medicine Center, San Gerardo Hospital, Monza, Italy
| | - Maria Carla Gilardi
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
- />Department of Health Sciences, Tecnomed Foundation, University of Milano-Bicocca, Milan, Italy
- />Nuclear Medicine, San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Bravatà
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
| |
Collapse
|
78
|
Role of microRNA 30a targeting insulin receptor substrate 2 in colorectal tumorigenesis. Mol Cell Biol 2015; 35:988-1000. [PMID: 25582198 DOI: 10.1128/mcb.01242-14] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
MicroRNAs (miRNAs) are dysregulated in many types of malignant diseases, including colorectal cancer. miRNA 30a (miR-30a) is a member of the miR-30 family and has been implicated in many types of cancers. In this study, we determined the expression of miR-30a in human colon cancer tissues and cell lines. miR-30a was found to be significantly downregulated in both the tissues and cell lines. Furthermore, overexpression of miR-30a inhibited, while silencing of miR-30a promoted, cell proliferation, migration, and invasion in vitro. Consistently, stable overexpression of miR-30a suppressed the growth of colon cancer cell xenografts in vivo. Moreover, bioinformatic algorithms and luciferase reporter assays revealed that insulin receptor substrate 2 (IRS2) is a direct target of miR-30a. Further functional studies suggested that repression of IRS2 by miR-30a partially mediated the tumor suppressor effect of miR-30a. In addition, miR-30a inhibited constitutive phosphorylation of Akt by targeting IRS2. Additionally, clinicopathological analysis indicated that miR-30a has an inverse correlation with the staging in patients with colon cancer. Taken together, our study provides the first evidence that miR-30a suppressed colon cancer cell growth through inhibition of IRS2. Thus, miR-30a might serve as a promising therapeutic strategy for colon cancer treatment.
Collapse
|