1
|
Araújo D, Ribeiro E, Amorim I, Vale N. Repurposed Drugs in Gastric Cancer. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010319. [PMID: 36615513 PMCID: PMC9822219 DOI: 10.3390/molecules28010319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/21/2022] [Accepted: 12/25/2022] [Indexed: 01/04/2023]
Abstract
Gastric cancer (GC) is one of the major causes of death worldwide, ranking as the fifth most incident cancer in 2020 and the fourth leading cause of cancer mortality. The majority of GC patients are in an advanced stage at the time of diagnosis, presenting a poor prognosis and outcome. Current GC treatment approaches involve endoscopic detection, gastrectomy and chemotherapy or chemoradiotherapy in an adjuvant or neoadjuvant setting. Drug development approaches demand extreme effort to identify molecular mechanisms of action of new drug candidates. Drug repurposing is based on the research of new therapeutic indications of drugs approved for other pathologies. In this review, we explore GC and the different drugs repurposed for this disease.
Collapse
Affiliation(s)
- Diana Araújo
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Eduarda Ribeiro
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Irina Amorim
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Correspondence: ; Tel.: +351-220426537
| |
Collapse
|
2
|
Ni Y, Low JT, Silke J, O’Reilly LA. Digesting the Role of JAK-STAT and Cytokine Signaling in Oral and Gastric Cancers. Front Immunol 2022; 13:835997. [PMID: 35844493 PMCID: PMC9277720 DOI: 10.3389/fimmu.2022.835997] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
When small proteins such as cytokines bind to their associated receptors on the plasma membrane, they can activate multiple internal signaling cascades allowing information from one cell to affect another. Frequently the signaling cascade leads to a change in gene expression that can affect cell functions such as proliferation, differentiation and homeostasis. The Janus kinase-signal transducer and activator of transcription (JAK-STAT) and the tumor necrosis factor receptor (TNFR) are the pivotal mechanisms employed for such communication. When deregulated, the JAK-STAT and the TNF receptor signaling pathways can induce chronic inflammatory phenotypes by promoting more cytokine production. Furthermore, these signaling pathways can promote replication, survival and metastasis of cancer cells. This review will summarize the essentials of the JAK/STAT and TNF signaling pathways and their regulation and the molecular mechanisms that lead to the dysregulation of the JAK-STAT pathway. The consequences of dysregulation, as ascertained from founding work in haematopoietic malignancies to more recent research in solid oral-gastrointestinal cancers, will also be discussed. Finally, this review will highlight the development and future of therapeutic applications which modulate the JAK-STAT or the TNF signaling pathways in cancers.
Collapse
Affiliation(s)
- Yanhong Ni
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jun T. Low
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - John Silke
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Lorraine A. O’Reilly
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
3
|
Dai L, Li Z, Tao Y, Liang W, Hu W, Zhou S, Fu X, Wang X. Emerging roles of suppressor of cytokine signaling 3 in human cancers. Biomed Pharmacother 2021; 144:112262. [PMID: 34607102 DOI: 10.1016/j.biopha.2021.112262] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/22/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023] Open
Abstract
As a member of the suppressor of cytokine signaling (SOCS) family, SOCS3 is a cytokine-inducible protein that inhibits cytokine signaling in a variety of signaling pathways. Increasing evidence shows that SOCS3 regulates tumor development through multiple pathological and physiological processes. It is worth mentioning that SOCS3 negatively regulates JAK/STAT signaling by binding to JAK/cytokine receptors or phosphorylation docking sites on STAT receptors, thus preventing tumor cell proliferation and inhibiting tumor cell invasion and metastasis. The kinase inhibitory region KIR of SOCS3 is the key to JAK inhibition. In addition, SOCS3 may also regulate tumor progression through other molecules or signaling pathways, such as microRNAs (miRNAs), IL-6 and NF-κB signaling pathway. MicroRNAs inhibit SOCS3 expression by binding to the 3' untranslated region of SOCS3 mRNA, thus regulating tumor development processes, including tumor cell proliferation, invasion, metastasis, differentiation, cell cycle and apoptosis, as well as tumor metastasis and chemotherapy resistance. On the whole, SOCS3 acts as an inhibitor of the majority of tumors through various pathways. In the present review, the role of SOCS3 in multitudinous tumors was comprehensively summarized, the molecular mechanisms and modes of action of SOCS3 in tumors were discussed, and the association between SOCS3 expression and the clinical characteristics of patients with cancer were emphasized.
Collapse
Affiliation(s)
- Lirui Dai
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China; Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, China
| | - Zian Li
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China; Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, China
| | - Yiran Tao
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China; Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, China
| | - Wulong Liang
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, China
| | - Weihua Hu
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, China
| | - Shaolong Zhou
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China; Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, China
| | - Xudong Fu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China; Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, China
| | - Xinjun Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China; Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, China.
| |
Collapse
|
4
|
Thilakasiri P, Huynh J, Poh AR, Tan CW, Nero TL, Tran K, Parslow AC, Afshar-Sterle S, Baloyan D, Hannan NJ, Buchert M, Scott AM, Griffin MD, Hollande F, Parker MW, Putoczki TL, Ernst M, Chand AL. Repurposing the selective estrogen receptor modulator bazedoxifene to suppress gastrointestinal cancer growth. EMBO Mol Med 2020; 11:emmm.201809539. [PMID: 30885958 PMCID: PMC6460354 DOI: 10.15252/emmm.201809539] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Excessive signaling through gp130, the shared receptor for the interleukin (IL)6 family of cytokines, is a common hallmark in solid malignancies and promotes their progression. Here, we established the in vivo utility of bazedoxifene, a steroid analog clinically approved for the treatment of osteoporosis, to suppress gp130‐dependent tumor growth of the gastrointestinal epithelium. Bazedoxifene administration reduced gastric tumor burden in gp130Y757F mice, where tumors arise exclusively through excessive gp130/STAT3 signaling in response to the IL6 family cytokine IL11. Likewise, in mouse models of sporadic colon and intestinal cancers, which arise from oncogenic mutations in the tumor suppressor gene Apc and the associated β‐catenin/canonical WNT pathway, bazedoxifene treatment reduces tumor burden. Consistent with the proposed orthogonal tumor‐promoting activity of IL11‐dependent gp130/STAT3 signaling, tumors of bazedoxifene‐treated Apc‐mutant mice retain excessive nuclear accumulation of β‐catenin and aberrant WNT pathway activation. Likewise, bazedoxifene treatment of human colon cancer cells harboring mutant APC did not reduce aberrant canonical WNT signaling, but suppressed IL11‐dependent STAT3 signaling. Our findings provide compelling proof of concept to support the repurposing of bazedoxifene for the treatment of gastrointestinal cancers in which IL11 plays a tumor‐promoting role.
Collapse
Affiliation(s)
- Pathum Thilakasiri
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Jennifer Huynh
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Ashleigh R Poh
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Chin Wee Tan
- The Walter and Eliza Hall Institute, Melbourne, Vic., Australia
| | - Tracy L Nero
- ACRF Rational Drug Discovery Centre, St Vincent's Institute, Melbourne, Vic., Australia.,Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, Melbourne, Vic., Australia
| | - Kelly Tran
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Adam C Parslow
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia.,Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Vic., Australia
| | - Shoukat Afshar-Sterle
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - David Baloyan
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Natalie J Hannan
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Vic., Australia
| | - Michael Buchert
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Andrew Mark Scott
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia.,Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Vic., Australia.,Department of Medicine, University of Melbourne, Melbourne, Vic., Australia
| | - Michael Dw Griffin
- Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, Melbourne, Vic., Australia
| | - Frederic Hollande
- Department of Clinical Pathology, University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, University of Melbourne, Melbourne, Vic., Australia
| | - Michael W Parker
- ACRF Rational Drug Discovery Centre, St Vincent's Institute, Melbourne, Vic., Australia.,Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, Melbourne, Vic., Australia
| | | | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Ashwini L Chand
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| |
Collapse
|
5
|
Luo Y, Huang X, Yang J, Huang L, Li R, Wu Q, Jiang X. Proteomics analysis of G protein-coupled receptor kinase 4-inhibited cellular growth of HEK293 cells. J Proteomics 2019; 207:103445. [DOI: 10.1016/j.jprot.2019.103445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/25/2019] [Accepted: 07/14/2019] [Indexed: 12/12/2022]
|
6
|
Li N, Dou Z, Liu J, Chai B, Li Y, An X, Chu P, Zhang X. Therapeutic Effect of HGF on NASH Mice Through HGF/c-Met and JAK2-STAT3 Signalling Pathway. Ann Hepatol 2019; 17:501-510. [PMID: 29735799 DOI: 10.5604/01.3001.0011.7395] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND AIM Hepatocyte growth factor (HGF) has been shown to ameliorate liver inflammation and fibrosis; however, the mechanism underlying its effects in non-alcoholic steatohepatitis (NASH) is unclear. This study aimed to analyse the relationship between the JAK2-STAT3 signalling pathway and the ameliorating effect of HGF on NASH. MATERIAL AND METHODS Mice were fed a high-fat diet (HFD) for 16 weeks, and then plasma and hepatic tissues were collected. Histological and clinical chemistry assays were performed to assess liver disease. The mRNA and protein levels of JAK2, STAT3, and c-Met were assessed by real-time PCR and western blotting, respectively. RESULTS Serum ALT, AST, and TG levels were increased in NASH mice. Histological analysis showed different degrees of steatosis, inflammatory infiltrates, and fibrosis in HFD animals. Exogenous administration of recombinant human (rh) HGF via the tail vein for 14 days markedly decreased ALT and AST to levels lower than those in the control group. Compared with the levels in HFD mice, c-Met, p-c-Met, JAK2, p-JAK2, and p-STAT3 levels were increased in mice that were administered HGF (P < 0.05). Furthermore, silencing of HGF or blocking of its receptor c-Met affected JAK2 and STAT3 protein phosphorylation. CONCLUSIONS Excess HGF highly probable improved NASH liver function. Combined with its ligand, c-Met, HGF may promote the phosphorylation of JAK2-STAT3 and inhibit inflammation in NASH. Therefore, it may be potentially useful treatment for NASH.
Collapse
Affiliation(s)
- Ning Li
- Department of Pathology, First hospital of Shanxi Medical University, Taiyuan, Shanxi, P.R. China
| | - Zhangfeng Dou
- Department of Gastroenterology, First hospital of Shanxi Medical University, Taiyuan, Shanxi, P.R. China
| | - Jinchun Liu
- Department of Gastroenterology, First hospital of Shanxi Medical University, Taiyuan, Shanxi, P.R. China
| | - Bao Chai
- Department of Gastroenterology, Shanxi Academy of Medical Science, Shanxi DaYi Hospital, Shanxi, Taiyuan, P.R. China
| | - Yue Li
- Department of Gastroenterology, First hospital of Shanxi Medical University, Taiyuan, Shanxi, P.R. China
| | - Xiuqin An
- Department of Gastroenterology, First hospital of Shanxi Medical University, Taiyuan, Shanxi, P.R. China
| | - Peiling Chu
- Department of Gastroenterology, First hospital of Shanxi Medical University, Taiyuan, Shanxi, P.R. China
| | - Xiaolan Zhang
- Department of Gastroenterology, First hospital of Shanxi Medical University, Taiyuan, Shanxi, P.R. China
| |
Collapse
|
7
|
Wang X, Li W, Zhang N, Zheng X, Jing Z. Opportunities and challenges of co-targeting epidermal growth factor receptor and autophagy signaling in non-small cell lung cancer. Oncol Lett 2019; 18:499-506. [PMID: 31289521 DOI: 10.3892/ol.2019.10372] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 04/11/2019] [Indexed: 12/17/2022] Open
Abstract
Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) are a standard therapy for patients with non-small cell lung cancer (NSCLC) with sensitive mutations. However, acquired resistance emerges following a median of 6-12 months. Several studies demonstrated that EGFR-TKI-induced tumor microenvironment stresses and autophagy are important causes of resistance. The current review summarizes the molecular mechanisms involved in EGFR-mediated regulation of autophagy. The role of autophagy in EGFR-TKI treatment, which may serve a role in protection or cell death, was discussed. Furthermore, co-inhibiting EGFR and autophagy signaling as a rational therapeutic strategy in the treatment of patients with NSCLC was explored.
Collapse
Affiliation(s)
- Xiaoju Wang
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, P.R. China
| | - Wenxin Li
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, P.R. China
| | - Ni Zhang
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, P.R. China
| | - Xiaoli Zheng
- Cancer Research Institute, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, P.R. China
| | - Zhao Jing
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, P.R. China
| |
Collapse
|
8
|
Zhang M, Sun H, Deng Y, Su M, Wei S, Wang P, Yu L, Liu J, Guo J, Wang X, Han X, He Q, Shen L. COPI-Mediated Nuclear Translocation of EGFRvIII Promotes STAT3 Phosphorylation and PKM2 Nuclear Localization. Int J Biol Sci 2019; 15:114-126. [PMID: 30662352 PMCID: PMC6329918 DOI: 10.7150/ijbs.28679] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 09/26/2018] [Indexed: 12/17/2022] Open
Abstract
As a non-ligand-dependent activation protein, EGFRvIII is the most common mutant of EGFR, and its existence or especially its nuclear translocation in tumors can exacerbate the malignancy. Compared with the nuclear translocation of EGFR, which has been studied extensively, the specific mechanism by which EGFRvIII undergoes nuclear translocation has not yet been reported. Here, we found that EGFRvIII eventually reached the nucleus with the involvement of the Golgi and endoplasmic reticulum (ER) in glioma cells. In this process, syntaxin-6 was responsible for the identification and transport of EGFRvIII on Golgi. We also demonstrated that COPI mediated the reverse transport of EGFRvIII from the Golgi to ER, which process was also important for EGFRvIII's nuclear accumulation. EGFRvIII's nuclear translocation can significantly promote STAT3 phosphorylation and PKM2 nuclear localization. Finally, we showed that EGFRvIII's nuclear translocation obviously induced the growth of gliomas in an intracranial xenotransplantation experiment. These data suggested that searching methods that inhibit EGFRvIII entry into the nucleus will be effective glioma treatments.
Collapse
Affiliation(s)
- Mingzhi Zhang
- Department of Cell Biology, Stem Cell Research Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People's Republic of China
| | - Haojie Sun
- Neuroscience Research Institute, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People's Republic of China
| | - Yue Deng
- Beijing Cellonis Biotechnologies Co. Ltd, Zhongguancun Bio-Medicine Park, Beijing, People's Republic of China
| | - Ming Su
- Department of Cell Biology, Stem Cell Research Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People's Republic of China
| | - Shiruo Wei
- Department of Cell Biology, Stem Cell Research Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People's Republic of China
| | - Peipei Wang
- Beijing Cellonis Biotechnologies Co. Ltd, Zhongguancun Bio-Medicine Park, Beijing, People's Republic of China
| | - Lanlan Yu
- Beijing DongFang YaMei Gene Science and Technology Research Institute, Beijing, People's Republic of China
| | - Jinwen Liu
- Beijing DongFang YaMei Gene Science and Technology Research Institute, Beijing, People's Republic of China
| | - Jinhai Guo
- Beijing DongFang YaMei Gene Science and Technology Research Institute, Beijing, People's Republic of China
| | - Xuan Wang
- Research Center of Artificial Organ and Tissue Engineering, Second Department of Hepatobiliary Surgery, ZhuJiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Xu Han
- Research Center of Artificial Organ and Tissue Engineering, Second Department of Hepatobiliary Surgery, ZhuJiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Qihua He
- Medical and Health Analytical Center, Peking University Health Science Center, People's Republic of China
| | - Li Shen
- Department of Cell Biology, Stem Cell Research Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People's Republic of China
| |
Collapse
|
9
|
Overexpressed C14orf166 associates with disease progression and poor prognosis in non-small-cell lung cancer. Biosci Rep 2018; 38:BSR20180479. [PMID: 30126850 PMCID: PMC6137245 DOI: 10.1042/bsr20180479] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/12/2018] [Accepted: 08/09/2018] [Indexed: 01/05/2023] Open
Abstract
Chromosome 14 ORF 166 (C14orf166), a protein involved in the regulation of RNA transcription and translation, has been reported to possess the potency to promote tumorigenesis; however, the role of C14orf166 in non-small-cell lung cancer (NSCLC) remains unknown. The purpose of the present study was to assess C14orf166 expression and its clinical significance in NSCLC. Immunohistochemical staining, quantitative real-time PCR (qRT-PCR), and Western blotting were used to detect the C14orf166 protein and mRNA expression levels in NSCLC tissues compared with adjacent normal tissues, as well as in NSCLC cells lines compared with normal human bronchial epithelial cells (HBE). Then, the correlations between the C14orf166 expression levels and the clinicopathological features of NSCLC were analyzed. Additionally, the Cox proportional hazard model was used to evaluate the prognostic significance of C14orf166. We found that C14orf166 expression increased in carcinoma tissues compared with their adjacent normal tissues at the protein (P<0.001) and mRNA levels (P<0.001). High expression of C14orf166 was significantly associated with the T stage (P=0.006), lymph node metastasis (P=0.001), advanced TNM stage (P<0.001), and chemotherapy (P<0.001). Moreover, according to the survival analysis, patients with overexpressed C14orf166 were inclined to experience a shorter overall survival and disease-free survival time (P<0.001). Multivariate COX analysis implied that C14orf166 was an independent prognostic biomarker. Taken together, our findings indicate that the overexpression of C14orf166 may contribute to the disease progression of NSCLC, represent a novel prognostic predictor and help high-risk patients make better decisions for subsequent therapy.
Collapse
|
10
|
Kuo CL, Liu ST, Chang YL, Wu CC, Huang SM. Zac1 regulates IL-11 expression in osteoarthritis. Oncotarget 2018; 9:32478-32495. [PMID: 30197757 PMCID: PMC6126702 DOI: 10.18632/oncotarget.25980] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/29/2018] [Indexed: 01/24/2023] Open
Abstract
Interleukin (IL)-11, a member of the IL-6 family of cytokines, exerts pleiotropic effects under normal and various disease conditions. We assessed IL-11 expression regulation and the IL-11/IL-6 ratio in osteoarthritis (OA) to better guide clinical therapeutic decision-making. Our findings suggest that Zac1, a zinc finger protein that regulates apoptosis and cell cycle arrest, is a transcription factor regulating IL-11 expression. Zac1 overexpression or knockdown respectively induced or suppressed IL-11 expression in HeLa cells. Zac1 acted synergistically with AP-1, human papillomavirus E2, and hypoxia inducible factor 1 alpha (HIF1α). IL-11 expression under various conditions, including hypoxia or treatment with phorbol 12-myristate 13-acetate or copper sulfate. Recombinant IL-11-induced phosphorylation of signal transducer and activator of transcription 3 at tyrosine 705 was reduced in a dose-dependent manner in HeLa cells. Cross-talk between Zac1, IL-11, p53, and suppressor of cytokine signaling 3 was differentially affected by copper sulfate, digoxin, and caffeine. Finally, aggressive vs. conventional treatment of OA patients was primarily determined by IL-6 levels. However, we suggest that OA patients with higher IL-11 levels may respond well to conventional treatments, even in the presence of high IL-6.
Collapse
Affiliation(s)
- Chun-Lin Kuo
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taiwan, Republic of China
- Department of Orthopaedic Surgery, Tri-Service General Hospital, National Defense Medical Center, Taiwan, Republic of China
| | - Shu-Ting Liu
- Department of Biochemistry, National Defense Medical Center, Taiwan, Republic of China
| | - Yung-Lung Chang
- Department of Biochemistry, National Defense Medical Center, Taiwan, Republic of China
| | - Chia-Chun Wu
- Department of Orthopaedic Surgery, Tri-Service General Hospital, National Defense Medical Center, Taiwan, Republic of China
| | - Shih-Ming Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taiwan, Republic of China
- Department of Biochemistry, National Defense Medical Center, Taiwan, Republic of China
| |
Collapse
|
11
|
Zhou C, Ma J, Su M, Shao D, Zhao J, Zhao T, Song Z, Meng Y, Jiao P. Down-regulation of STAT3 induces the apoptosis and G1 cell cycle arrest in esophageal carcinoma ECA109 cells. Cancer Cell Int 2018; 18:53. [PMID: 29636641 PMCID: PMC5883295 DOI: 10.1186/s12935-018-0549-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 03/23/2018] [Indexed: 12/20/2022] Open
Abstract
Background Signal transducer and activator of transcription 3 (STAT3) is persistently activated in a wide variety of epithelial cancers. Aberrant activity of STAT3 correlates with tumor growth, invasion and metastasis, which makes it a potential therapeutic target of cancer. To explore the biological role of STAT3 in esophageal cancer, we used small hairpin RNA to knockdown the expression of the STAT3 gene in the esophageal carcinoma ECA109 cell line and the cell apoptosis, cell cycle and cell migration were investigated. Methods The cell apoptosis was tested using DNA ladder, mitochondrial membrane potential assay, TUNEL assay, annexin V-PI staining. Cell cycle phases were estimated using flow cytometry analysis. The mRNA and proteins related to apoptosis and cell cycle were examined by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot, respectively. And cell migration was investigated by in vitro Transwell assay. The data were analyzed with two-sample Student’s t test and ANOVA followed by the LSD post hoc test. Results Our results showed that knockdown of STAT3 in ECA109 cells induced noticeable apoptotic morphological changes like cell shrinkage, apoptotic vacuoles, membrane blebbing time-dependently. In addition, DNA ladder, TUNEL assay, Annexin V-PI staining and declined level of cleaved Caspase-3 indicated that down-regulation of STAT3 could induce apoptosis in ECA109 cells. Flow cytometry analysis displayed the induction of G1-phase cell cycle arrest of ECA109 cells by STAT3 decreasing, consistent with the descend of c-Myc and cyclin D1 in protein levels. Furthermore, STAT3 knockdown suppressed the expression of matrix metalloproteinases-9, sushi domain containing 2 and urokinase plasminogen activator in ECA109 cells and inhibited cell migration ability. Conclusions Knockdown of STAT3 could induce the apoptosis and G1 cell cycle arrest in esophageal carcinoma ECA109 cells, and inhibit the migration ability of cells as well.
Collapse
Affiliation(s)
- Chao Zhou
- 1School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021 People's Republic of China
| | - Jie Ma
- 1School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021 People's Republic of China
| | - Mengyuan Su
- 1School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021 People's Republic of China
| | - Dan Shao
- 2The First Hospital of Jilin University, 1163 Xinmin Street, Changchun, Jilin 130021 People's Republic of China
| | - Jianan Zhao
- 1School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021 People's Republic of China
| | - Tongjian Zhao
- 1School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021 People's Republic of China
| | - Zhuoyao Song
- 1School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021 People's Republic of China
| | - Yan Meng
- 1School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021 People's Republic of China
| | - Ping Jiao
- 1School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021 People's Republic of China
| |
Collapse
|
12
|
Ollila S, Domènech-Moreno E, Laajanen K, Wong IP, Tripathi S, Pentinmikko N, Gao Y, Yan Y, Niemelä EH, Wang TC, Viollet B, Leone G, Katajisto P, Vaahtomeri K, Mäkelä TP. Stromal Lkb1 deficiency leads to gastrointestinal tumorigenesis involving the IL-11-JAK/STAT3 pathway. J Clin Invest 2017; 128:402-414. [PMID: 29202476 DOI: 10.1172/jci93597] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 10/24/2017] [Indexed: 12/12/2022] Open
Abstract
Germline mutations in the gene encoding tumor suppressor kinase LKB1 lead to gastrointestinal tumorigenesis in Peutz-Jeghers syndrome (PJS) patients and mouse models; however, the cell types and signaling pathways underlying tumor formation are unknown. Here, we demonstrated that mesenchymal progenitor- or stromal fibroblast-specific deletion of Lkb1 results in fully penetrant polyposis in mice. Lineage tracing and immunohistochemical analyses revealed clonal expansion of Lkb1-deficient myofibroblast-like cell foci in the tumor stroma. Loss of Lkb1 in stromal cells was associated with induction of an inflammatory program including IL-11 production and activation of the JAK/STAT3 pathway in tumor epithelia concomitant with proliferation. Importantly, treatment of LKB1-defcient mice with the JAK1/2 inhibitor ruxolitinib dramatically decreased polyposis. These data indicate that IL-11-mediated induction of JAK/STAT3 is critical in gastrointestinal tumorigenesis following Lkb1 mutations and suggest that targeting this pathway has therapeutic potential in Peutz-Jeghers syndrome.
Collapse
Affiliation(s)
- Saara Ollila
- Research Programs Unit, Faculty of Medicine and.,HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.,Division of Digestive and Liver Diseases, Department of Medicine, Irving Cancer Research Center, Columbia University Medical Center, New York, New York, USA
| | - Eva Domènech-Moreno
- Research Programs Unit, Faculty of Medicine and.,HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Kaisa Laajanen
- Research Programs Unit, Faculty of Medicine and.,HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Iris Pl Wong
- Research Programs Unit, Faculty of Medicine and.,HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Sushil Tripathi
- Research Programs Unit, Faculty of Medicine and.,HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Nalle Pentinmikko
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Yajing Gao
- Research Programs Unit, Faculty of Medicine and.,HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Yan Yan
- Research Programs Unit, Faculty of Medicine and.,HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Elina H Niemelä
- Research Programs Unit, Faculty of Medicine and.,HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Irving Cancer Research Center, Columbia University Medical Center, New York, New York, USA
| | - Benoit Viollet
- INSERM, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, France
| | - Gustavo Leone
- Department of Cancer Biology and Genetics, College of Medicine, Department of Molecular Genetics, College of Biological Sciences, and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Pekka Katajisto
- HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.,Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.,Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|
13
|
Shao D, Ma J, Zhou C, Zhao JN, Li LL, Zhao TJ, Ai XL, Jiao P. STAT3 down-regulation induces mitochondria-dependent G2/M cell cycle arrest and apoptosis in oesophageal carcinoma cells. Clin Exp Pharmacol Physiol 2017; 44:413-420. [PMID: 27896845 DOI: 10.1111/1440-1681.12708] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 11/25/2016] [Accepted: 11/25/2016] [Indexed: 12/21/2022]
Abstract
STAT3 is persistently activated in a wide variety of human tumours, and aberrant STAT3 activity promotes tumour growth, invasion and metastasis. To explore STAT3 down-regulation in human oesophageal cancer cells, cell proliferation, apoptosis and mitochondrial mechanisms were explored in oesophageal carcinoma TE1 cell cultures. We demonstrate for the first time that STAT3 down-regulation by RNAi is sufficient to inhibit oesophageal cancer cell proliferation inducing cell apoptosis. Further, we demonstrate that mitochondrial transmembrane potential is impaired thereby leading to collapsed mitochondrial membrane potential, abnormal mitochondrial membrane depolarization, nuclear DNA fragmentation and cell cycle G2/M arrest under the conditions of STAT3 down-regulation. Thus, our results suggest that STAT3 inhibition is a valid approach to induce oesophageal carcinoma cell mitochondrial-dependent apoptosis in therapeutic strategies against oesophageal cancers.
Collapse
Affiliation(s)
- Dan Shao
- School of Pharmaceutical Sciences, Jilin University, Changchun, China.,The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Jie Ma
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Chao Zhou
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Jia-Nan Zhao
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Lu-Lu Li
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Tong-Jian Zhao
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Xi-Lei Ai
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Ping Jiao
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| |
Collapse
|
14
|
Interleukin-6 as a Multifunctional Regulator: Inflammation, Immune Response, and Fibrosis. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2017. [DOI: 10.5301/jsrd.5000265] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Interleukin 6 (IL-6) is a 184-amino acid protein cytokine that is produced by many types of cells and is expressed during states of cellular stress, such as inflammation, infection, wound sites, and cancer. IL-6 levels may increase several thousand-fold in these states and may help to coordinate the response to dysregulation of tissue homeostasis. IL-6 acts through a membrane-bound IL-6 receptor (mIL-6R), which, together with a second receptor, glycoprotein 130 (gp130), leads to the initiation of intracellular signaling (classic signaling). Given that IL-6R is expressed on only a few types of cells, though all cells express gp130, direct stimulation by IL-6 is limited to cells that express mIL-6R. However, IL-6R is also produced as a soluble, secreted protein that, together with IL-6, can stimulate all gp130-expressing cells by a process termed IL-6 trans-signaling. IL-6 trans-signaling can be blocked without affecting IL-6 classic signaling through mIL-6R. IL-6 has major effects on the adaptive and innate immune system and on mesenchymal and stromal responses during inflammation. It promotes the development of pathogenic T-helper 17 T cells and the maturation of B lymphocytes. Many innate immune cells, neutrophils, and monocytes/macrophages produce and respond to IL-6, resulting in autocrine feedback loops that amplify inflammation. IL-6 has been implicated in the pathogenesis of fibrotic diseases in which IL-6 trans-signaling has been shown to stimulate the proliferation of fibroblasts and the release of procollagen and fibronectin.
Collapse
|
15
|
Wang M, Zhang C, Song Y, Wang Z, Wang Y, Luo F, Xu Y, Zhao Y, Wu Z, Xu Y. Mechanism of immune evasion in breast cancer. Onco Targets Ther 2017; 10:1561-1573. [PMID: 28352189 PMCID: PMC5359138 DOI: 10.2147/ott.s126424] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Breast cancer (BC) is the most common malignant tumor among women, with high morbidity and mortality. Its onset, development, metastasis, and prognosis vary among individuals due to the interactions between tumors and host immunity. Many diverse mechanisms have been associated with BC, with immune evasion being the most widely studied to date. Tumor cells can escape from the body’s immune response, which targets abnormal components and foreign bodies, using different approaches including modification of surface antigens and modulation of the surrounding environment. In this review, we summarize the mechanisms and factors that impact the immunoediting process and analyze their functions in detail.
Collapse
Affiliation(s)
| | - Changwang Zhang
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yongxi Song
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Zhenning Wang
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | | | | | | | - Yi Zhao
- Department of Breast Surgery
| | - Zhonghua Wu
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | | |
Collapse
|
16
|
Alorro MG, Pierce TP, Eissmann MF, Dijkstra C, Dickins RA, Ernst M, Buchert M, Masson F. Generation of an inducible mouse model to reversibly silence Stat3. Genesis 2017; 55. [DOI: 10.1002/dvg.23023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/19/2017] [Accepted: 02/01/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Mariah G. Alorro
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine; Heidelberg Victoria 3084 Australia
| | - Thomas P. Pierce
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine; Heidelberg Victoria 3084 Australia
| | - Moritz F. Eissmann
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine; Heidelberg Victoria 3084 Australia
| | - Christine Dijkstra
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine; Heidelberg Victoria 3084 Australia
| | - Ross A. Dickins
- Dickins Laboratory, Australian Centre for Blood Diseases, Monash University; Melbourne Victoria 3004 Australia
| | - Matthias Ernst
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine; Heidelberg Victoria 3084 Australia
| | - Michael Buchert
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine; Heidelberg Victoria 3084 Australia
| | - Frederic Masson
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine; Heidelberg Victoria 3084 Australia
| |
Collapse
|
17
|
Zhou YW, Li R, Duan CJ, Gao Y, Cheng YD, He ZW, Zeng JX, Zhang CF. Expression and clinical significance of C14orf166 in esophageal squamous cell carcinoma. Mol Med Rep 2016; 15:605-612. [PMID: 28000881 PMCID: PMC5364856 DOI: 10.3892/mmr.2016.6056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 11/04/2016] [Indexed: 01/05/2023] Open
Abstract
C14orf166, a 28 kD protein regulating RNA transcription and translation, may serve a critical role in oncogenesis. The aim of the current study was to explore the association between C14orf166 expression and esophageal squamous cell carcinoma (ESCC) and to draw attention to the association between C14orf166 and the initiation, progression and prognosis of ESCC. C14orf166 expression in ESCC and paired normal tissues was detected by immunohistochemical staining, western blotting and reverse transcription-quantitative polymerase chain reaction, and the association between C14orf166 expression and clinicopathological characters of ESCC was analyzed. Survival analysis was used to assess the prognostic significance of C14orf166 and it was observed that C14orf166 expression was higher in the ESCC tissues when compared with adjacent non-cancerous tissues at protein (P<0.001) and mRNA levels (P<0.001). There was a significant difference in T stage, lymph node metastasis and TNM stage in patients categorized according to different C14orf166 expression levels. The overexpression of C14orf166 was associated with a shorter overall survival and disease-free survival, and multivariate analysis indicated that C14orf166 was an independent prognostic indicator. The present study indicates that the expression of C14orf166 is elevated in ESCC, and is potentially a valuable prognostic predictor for ESCC.
Collapse
Affiliation(s)
- Yan-Wu Zhou
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Rong Li
- Department of Gastroenterology, Xiangya Third Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Chao-Jun Duan
- Institute of Medical Science, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yang Gao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yuan-Da Cheng
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Zhi-Wei He
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jun-Xian Zeng
- Department of Clinical Medicine, Hunan Xiangnan College, Chenzhou, Hunan 423043, P.R. China
| | - Chun-Fang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
18
|
Khanna P, Chua PJ, Bay BH, Baeg GH. The JAK/STAT signaling cascade in gastric carcinoma (Review). Int J Oncol 2015; 47:1617-26. [PMID: 26398764 DOI: 10.3892/ijo.2015.3160] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 08/03/2015] [Indexed: 11/06/2022] Open
Abstract
Gastric carcinoma remains one of the most prevalent forms of cancer worldwide, despite the decline in incidence rates, increased awareness of the disease and advancement in treatment strategies. Helicobacter pylori infection, dietary factors, lifestyle influences and various genetic aberrations have been shown to contribute to the development and progression of gastric cancer. Recent studies on the genomic landscape of gastric adenocarcinoma have identified several key signaling molecules, including epidermal growth factor receptor family (ErbB) members, vascular endothelial growth factor receptor family (VEGFR) members and PI3K/Akt/mTOR pathway components, that have been implicated in the molecular pathogenesis of gastric cancers. However, clinical trials with compounds that target these molecules have failed to show a significant improvement in overall survival rates when supplemented with conventional therapies. Therefore, it is essential to identify effective prognostic and/or diagnostic biomarkers and develop molecular targeted therapies. The JAK/STAT cascade is a principal signal transduction pathway in cytokine and growth factor signaling, regulating various cellular processes such as cell proliferation, differentiation, migration and survival. Numerous in vivo and in vitro studies have shown that dysregulated JAK/STAT signaling is a driving force in the pathogenesis of various solid cancers as well as hematopoietic malignancies. Hence, a large number of preclinical and clinical studies of drugs targeting this pathway are currently underway. Notably, aberrant JAK/STAT signaling has also been implicated in gastric cancers. In this review, we focus on the ongoing research on the JAK/STAT cascade in gastric carcinoma and discuss the therapeutic potential of targeting JAK/STAT signaling for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Puja Khanna
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117 597, Republic of Singapore
| | - Pei Jou Chua
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117 597, Republic of Singapore
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117 597, Republic of Singapore
| | - Gyeong Hun Baeg
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117 597, Republic of Singapore
| |
Collapse
|
19
|
Rocha GA, Rocha AMC, Gomes AD, Faria CL, Melo FF, Batista SA, Fernandes VC, Almeida NBF, Teixeira KN, Brito KS, Queiroz DMM. STAT3 polymorphism and Helicobacter pylori CagA strains with higher number of EPIYA-C segments independently increase the risk of gastric cancer. BMC Cancer 2015; 15:528. [PMID: 26186918 PMCID: PMC4506573 DOI: 10.1186/s12885-015-1533-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/02/2015] [Indexed: 12/18/2022] Open
Abstract
Background Because to date there is no available study on STAT3 polymorphism and gastric cancer in Western populations and taking into account that Helicobacter pylori CagA EPIYA-C segment deregulates SHP-2/ERK-JAK/STAT3 pathways, we evaluated whether the two variables are independently associated with gastric cancer. Methods We included 1048 subjects: H. pylori-positive patients with gastric carcinoma (n = 232) and with gastritis (n = 275) and 541 blood donors. Data were analyzed using logistic regression model. Results The rs744166 polymorphic G allele (p = 0.01; OR = 1.76; 95 % CI = 1.44-2.70), and CagA-positive (OR = 12.80; 95 % CI = 5.58-19.86) status were independently associated with gastric cancer in comparison with blood donors. The rs744166 polymorphism (p = 0.001; OR = 1.64; 95 % CI = 1.16-2.31) and infection with H. pylori CagA-positive strains possessing higher number of EPIYA-C segments (p = 0.001; OR = 2.28; 95 % CI = 1.41-3.68) were independently associated with gastric cancer in comparison with gastritis. The association was stronger when host and bacterium genotypes were combined (p < 0.001; OR = 3.01; 95 % CI = 2.29-3.98). When stimulated with LPS (lipopolysaccharide) or Pam3Cys, peripheral mononuclear cells of healthy carriers of the rs744166 GG and AG genotypes expressed higher levels of STAT3 mRNA than those carrying AA genotype (p = 0.04 for both). The nuclear expression of phosphorylated p-STAT3 protein was significantly higher in the antral gastric tissue of carriers of rs744166 GG genotype than in carriers of AG and AA genotypes. Conclusions Our study provides evidence that STAT3 rs744166 G allele and infection with CagA-positive H. pylori with higher number of EPIYA-C segments are independent risk factors for gastric cancer. The odds ratio of having gastric cancer was greater when bacterium and host high risk genotypes were combined.
Collapse
Affiliation(s)
- Gifone A Rocha
- Laboratory of Research in Bacteriology, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Alfredo Balena, 190 s/216, 30130-100, Belo Horizonte, Brazil.
| | - Andreia M C Rocha
- Laboratory of Research in Bacteriology, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Alfredo Balena, 190 s/216, 30130-100, Belo Horizonte, Brazil.
| | - Adriana D Gomes
- Laboratory of Research in Bacteriology, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Alfredo Balena, 190 s/216, 30130-100, Belo Horizonte, Brazil.
| | - César Ll Faria
- Laboratory of Research in Bacteriology, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Alfredo Balena, 190 s/216, 30130-100, Belo Horizonte, Brazil.
| | - Fabrício F Melo
- Laboratory of Research in Bacteriology, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Alfredo Balena, 190 s/216, 30130-100, Belo Horizonte, Brazil.
| | - Sérgio A Batista
- Laboratory of Research in Bacteriology, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Alfredo Balena, 190 s/216, 30130-100, Belo Horizonte, Brazil.
| | - Viviane C Fernandes
- Laboratory of Research in Bacteriology, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Alfredo Balena, 190 s/216, 30130-100, Belo Horizonte, Brazil.
| | - Nathálie B F Almeida
- Laboratory of Research in Bacteriology, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Alfredo Balena, 190 s/216, 30130-100, Belo Horizonte, Brazil.
| | - Kádima N Teixeira
- Laboratory of Research in Bacteriology, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Alfredo Balena, 190 s/216, 30130-100, Belo Horizonte, Brazil.
| | - Kátia S Brito
- Laboratory of Research in Bacteriology, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Alfredo Balena, 190 s/216, 30130-100, Belo Horizonte, Brazil.
| | - Dulciene Maria Magalhães Queiroz
- Laboratory of Research in Bacteriology, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Alfredo Balena, 190 s/216, 30130-100, Belo Horizonte, Brazil.
| |
Collapse
|
20
|
Esquivel-Velázquez M, Ostoa-Saloma P, Palacios-Arreola MI, Nava-Castro KE, Castro JI, Morales-Montor J. The role of cytokines in breast cancer development and progression. J Interferon Cytokine Res 2015; 35:1-16. [PMID: 25068787 PMCID: PMC4291218 DOI: 10.1089/jir.2014.0026] [Citation(s) in RCA: 335] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 05/23/2014] [Indexed: 12/12/2022] Open
Abstract
Cytokines are highly inducible, secretory proteins that mediate intercellular communication in the immune system. They are grouped into several protein families that are referred to as tumor necrosis factors, interleukins, interferons, and colony-stimulating factors. In recent years, it has become clear that some of these proteins as well as their receptors are produced in the organisms under physiological and pathological conditions. The exact initiation process of breast cancer is unknown, although several hypotheses have emerged. Inflammation has been proposed as an important player in tumor initiation, promotion, angiogenesis, and metastasis, all phenomena in which cytokines are prominent players. The data here suggest that cytokines play an important role in the regulation of both induction and protection in breast cancer. This knowledge could be fundamental for the proposal of new therapeutic approaches to particularly breast cancer and other cancer-related disorders.
Collapse
Affiliation(s)
- Marcela Esquivel-Velázquez
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, México
| | - Pedro Ostoa-Saloma
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, México
| | | | - Karen E. Nava-Castro
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, SSA, Cuernavaca, Morelos, México
| | - Julieta Ivonne Castro
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, SSA, Cuernavaca, Morelos, México
| | - Jorge Morales-Montor
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, México
| |
Collapse
|
21
|
O'Sullivan KE, Reynolds JV, O'Hanlon C, O'Sullivan JN, Lysaght J. Could signal transducer and activator of transcription 3 be a therapeutic target in obesity-related gastrointestinal malignancy? J Gastrointest Cancer 2014; 45:1-11. [PMID: 24163144 DOI: 10.1007/s12029-013-9555-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION A large body of evidence has implicated the signal transducer and activator of transcription (STAT) family and particularly the ubiquitously expressed STAT3 protein in the pathogenesis of colorectal, hepatocellular, gastric and pancreatic carcinoma. DISCUSSION Concomitantly, an increasing body of epidemiological evidence has linked obesity and its associated pro-inflammatory state with the development of gastrointestinal cancers. Visceral adipose tissue is no longer considered inert and is known to secrete a number of adipocytokines such as leptin, interleukin (IL)-6, IL-8, IL-1β and tumour necrosis factor-alpha (TNF-α) into the surrounding environment. Interestingly, these adipocytokines are strongly linked with the Janus kinase (JAK)/STAT pathway of signal transduction and there is experimental evidence linking IL-1β, IL-8 and TNF-α to JAK/STAT signaling in other tissues. The result is an up-regulation of a wide range of anti-apoptotic, pro-metastatic and pro-angiogenic genes and processes. This is particularly relevant for gastrointestinal malignancy as these factors have the potential to signal adjacent endothelial cells in a paracrine manner. CONCLUSION This review examines the potential role of the STAT3 signaling pathway in the pathogenesis of obesity-related gastrointestinal malignancy and the potential therapeutic role of STAT3 blockade given its status as a signaling hub for a number of inflammatory adipocytokines.
Collapse
Affiliation(s)
- Katie E O'Sullivan
- Department of Surgery, Institute of Molecular Medicine, St. James Hospital, Dublin 8, Ireland,
| | | | | | | | | |
Collapse
|
22
|
SOCS3 methylation in synergy with Reg3A overexpression promotes cell growth in pancreatic cancer. J Mol Med (Berl) 2014; 92:1257-69. [PMID: 24996521 DOI: 10.1007/s00109-014-1184-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 06/03/2014] [Accepted: 06/16/2014] [Indexed: 01/05/2023]
Abstract
UNLABELLED Pancreatic cancer (PaC) is the fifth leading cause of cancer death in the world, but the molecular mechanisms for its development remain unclear. Regenerating islet-derived protein 3-alpha (Reg3A) has been reported overexpressed in pancreatic inflammation and associated with PaC malignancies, thus believed as a potential target in inflammation-linked pancreatic carcinogenesis. Silencing of suppressor of cytokine signaling SOCS3, a well-known feedback inhibitor of cell proliferation, has been found in many human cancers. Here, we identified that SOCS3 was aberrantly methylated in its CpG island in 3/5 human PaC cell lines and 11/36 cancer tissue samples. SOCS3 restoration by a demethylating agent, 5-aza-2'-deoxycytidine, remarkably suppressed cell proliferation and induced apoptosis of methylated PaC cells. Moreover, we also have shown that Reg3A was highly expressed in PaC cells and tissue samples. Assessment of potential relationship between SOCS3 and Reg3A aberrations in vitro revealed that SOCS3 worked downstream of Reg3A and modulated Reg3A-linked pro-tumor functions. siRNA-mediated SOCS3 knock-down in normal pancreatic epithelial cells and plasmid-transfected SOCS3 overexpression in PaC cells, respectively, resulted in the obvious promotion and inhibition of Reg3A-induced cell proliferation, thereby suggesting SOCS3 negatively regulating Reg3A-mediated PaC progression. In addition, our findings also revealed that JAK/STAT3/NF-κB appear involved in the effect of SOCS3-Reg3A interaction on pancreatic cell growth. In summary, SOCS3 inactivation by methylation was demonstrated to act in synergy with Reg3A overexpression to promote PaC cell growth and maybe the progress of inflammation-linked pancreatic carcinogenesis. KEY MESSAGES Reg3A overexpression promoted cell growth in pancreatic cancer. SOCS3 is a key target in cancer by inhibiting cell growth and inducing apoptosis. SOCS3 negatively regulated Reg3A-mediated cell growth in pancreatic cancer. SOCS3 methylation act in synergy with Reg3A overexpression to promote pancreatic cancer cell growth.
Collapse
|
23
|
|
24
|
Blakely AM, Heffernan DS, McPhillips J, Cioffi WG, Miner TJ. Elevated C-reactive protein as a predictor of patient outcomes following palliative surgery. J Surg Oncol 2014; 110:651-5. [DOI: 10.1002/jso.23682] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 05/19/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Andrew M. Blakely
- Department of Surgery; Rhode Island Hospital; Warren Alpert Medical School of Brown University; Providence Rhode Island
| | - Daithi S. Heffernan
- Department of Surgery; Rhode Island Hospital; Warren Alpert Medical School of Brown University; Providence Rhode Island
| | - Jane McPhillips
- Department of Surgery; Rhode Island Hospital; Warren Alpert Medical School of Brown University; Providence Rhode Island
| | - William G. Cioffi
- Department of Surgery; Rhode Island Hospital; Warren Alpert Medical School of Brown University; Providence Rhode Island
| | - Thomas J. Miner
- Department of Surgery; Rhode Island Hospital; Warren Alpert Medical School of Brown University; Providence Rhode Island
| |
Collapse
|
25
|
Abubaker K, Luwor RB, Escalona R, McNally O, Quinn MA, Thompson EW, Findlay JK, Ahmed N. Targeted Disruption of the JAK2/STAT3 Pathway in Combination with Systemic Administration of Paclitaxel Inhibits the Priming of Ovarian Cancer Stem Cells Leading to a Reduced Tumor Burden. Front Oncol 2014; 4:75. [PMID: 24782986 PMCID: PMC3988380 DOI: 10.3389/fonc.2014.00075] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 03/25/2014] [Indexed: 12/15/2022] Open
Abstract
Chemotherapy resistance associated with recurrent disease is the major cause of poor survival of ovarian cancer patients. We have recently demonstrated activation of the JAK2/STAT3 pathway and the enhancement of a cancer stem cell (CSC)-like phenotype in ovarian cancer cells treated in vitro with chemotherapeutic agents. To elucidate further these mechanisms in vivo, we used a two-tiered paclitaxel treatment approach in nude mice inoculated with ovarian cancer cells. In the first approach, we demonstrate that a single intraperitoneal administration of paclitaxel in mice 7 days after subcutaneous transplantation of the HEY ovarian cancer cell line resulted in a significant increase in the expression of CA125, Oct4, and CD117 in mice xenografts compared to control mice xenografts which did not receive paclitaxel. In the second approach, mice were administered once weekly with paclitaxel and/or a daily dose of the JAK2-specific inhibitor, CYT387, over 4 weeks. Mice receiving paclitaxel only demonstrated a significant decrease in tumor volume compared to control mice. At the molecular level, mouse tumors remaining after paclitaxel administration showed a significant increase in the expression of Oct4 and CD117 coinciding with a significant activation of the JAK2/STAT3 pathway compared to control tumors. The addition of CYT387 with paclitaxel resulted in the suppression of JAK2/STAT3 activation and abrogation of Oct4 and CD117 expression in mouse xenografts. This coincided with significantly smaller tumors in mice administered CYT387 in addition to paclitaxel, compared to the control group and the group of mice receiving paclitaxel only. These data suggest that the systemic administration of paclitaxel enhances Oct4- and CD117-associated CSC-like marker expression in surviving cancer cells in vivo, which can be suppressed by the addition of the JAK2-specific inhibitor CYT387, leading to a significantly smaller tumor burden. These novel findings have the potential for the development of CSC-targeted therapy to improve the treatment outcomes of ovarian cancer patients.
Collapse
Affiliation(s)
- Khalid Abubaker
- Women's Cancer Research Centre, Royal Women's Hospital , Parkville, VIC , Australia ; Department of Surgery, St Vincent Hospital, University of Melbourne , Melbourne, VIC , Australia
| | - Rodney B Luwor
- Department of Surgery, Royal Melbourne Hospital, University of Melbourne , Melbourne, VIC , Australia
| | - Ruth Escalona
- Women's Cancer Research Centre, Royal Women's Hospital , Parkville, VIC , Australia
| | - Orla McNally
- Women's Cancer Research Centre, Royal Women's Hospital , Parkville, VIC , Australia ; Department of Obstetrics and Gynaecology, University of Melbourne , Melbourne, VIC , Australia
| | - Michael A Quinn
- Women's Cancer Research Centre, Royal Women's Hospital , Parkville, VIC , Australia ; Department of Obstetrics and Gynaecology, University of Melbourne , Melbourne, VIC , Australia
| | - Erik W Thompson
- Department of Surgery, St Vincent Hospital, University of Melbourne , Melbourne, VIC , Australia ; St Vincent's Institute , Fitzroy, VIC , Australia
| | - Jock K Findlay
- Women's Cancer Research Centre, Royal Women's Hospital , Parkville, VIC , Australia ; Department of Obstetrics and Gynaecology, University of Melbourne , Melbourne, VIC , Australia ; Prince Henry's Institute of Medical Research , Clayton, VIC , Australia
| | - Nuzhat Ahmed
- Women's Cancer Research Centre, Royal Women's Hospital , Parkville, VIC , Australia ; Department of Surgery, St Vincent Hospital, University of Melbourne , Melbourne, VIC , Australia ; Department of Obstetrics and Gynaecology, University of Melbourne , Melbourne, VIC , Australia ; Prince Henry's Institute of Medical Research , Clayton, VIC , Australia
| |
Collapse
|
26
|
Jung YS, Kim DH, Hwang JY, Yun NY, Lee YH, Han SB, Hwang BY, Lee MS, Jeong HS, Hong JT. Anti-inflammatory effect of tricin 4'-O-(threo-β-guaiacylglyceryl) ether, a novel flavonolignan compound isolated from Njavara on in RAW264.7 cells and in ear mice edema. Toxicol Appl Pharmacol 2014; 277:67-76. [PMID: 24631338 DOI: 10.1016/j.taap.2014.03.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 02/26/2014] [Accepted: 03/01/2014] [Indexed: 12/22/2022]
Abstract
Although recent study has shown tricin 4'-O-(threo-β-guaiacylglyceryl) ether (TTGE), an isolated compound from Njavara rice, to have the most potent anti-inflammatory effects, the action mechanism has not been fully understood. Here, we examined the effect of TTGE on the inflammation and elucidated the potential mechanism. We demonstrated that TTGE significantly inhibited LPS-induced NO and ROS generation in RAW264.7 cells, which was correlated with the down-regulating effect of TTGE on the iNOS and COX-2 expression via NF-κB and STAT3. TPA-induced ear edema was also efficiently inhibited by the TTGE treatment. TTGE blocked the induction of iNOS and COX-2 through the regulation of NF-κB and STAT3, which could explain the reduced TPA-induced edema symptoms. Moreover, the introduction of ERK inhibitor abrogated the anti-inflammatory effect of TTGE via the recovery of NF-κB and STAT3 signalings. Taken together, these results suggest that TTGE has anti-inflammatory properties through down-regulation of NF-κB and STAT3 pathways.
Collapse
Affiliation(s)
- Young-Suk Jung
- College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Dae Hwan Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, Chungbuk 361-763, Republic of Korea
| | - Jae Yeon Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Chungbuk 361-763, Republic of Korea
| | - Na Young Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, Chungbuk 361-763, Republic of Korea
| | - Yun-Hee Lee
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Sang Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Chungbuk 361-763, Republic of Korea
| | - Bang Yeon Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Chungbuk 361-763, Republic of Korea
| | - Moon Soon Lee
- College of Agriculture, Life and Environments, Chungbuk National University, Chungbuk 361-763, Republic of Korea
| | - Heon-Sang Jeong
- Department of Food Science and Technology, Chungbuk National University, Chungbuk 361-763, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Chungbuk 361-763, Republic of Korea.
| |
Collapse
|
27
|
The IL-6/JAK/Stat3 feed-forward loop drives tumorigenesis and metastasis. Neoplasia 2014; 15:848-62. [PMID: 23814496 DOI: 10.1593/neo.13706] [Citation(s) in RCA: 358] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 04/19/2013] [Accepted: 04/22/2013] [Indexed: 02/07/2023] Open
Abstract
We have investigated the importance of interleukin-6 (IL-6) in promoting tumor growth and metastasis. In human primary breast cancers, increased levels of IL-6 were found at the tumor leading edge and positively correlated with advanced stage, suggesting a mechanistic link between tumor cell production of IL-6 and invasion. In support of this hypothesis, we showed that the IL-6/Janus kinase (JAK)/signal transducer and activator of transcription 3 (Stat3) pathway drives tumor progression through the stroma and metastatic niche. Overexpression of IL-6 in tumor cell lines promoted myeloid cell recruitment, angiogenesis, and induced metastases. We demonstrated the therapeutic potential of interrupting this pathway with IL-6 receptor blockade or by inhibiting its downstream effectors JAK1/2 or Stat3. These clinically relevant interventions did not inhibit tumor cell proliferation in vitro but had profound effects in vivo on tumor progression, interfering broadly with tumor-supportive stromal functions, including angiogenesis, fibroblast infiltration, and myeloid suppressor cell recruitment in both the tumor and pre-metastatic niche. This study provides the first evidence for IL-6 expression at the leading edge of invasive human breast tumors and demonstrates mechanistically that IL-6/JAK/Stat3 signaling plays a critical and pharmacologically targetable role in orchestrating the composition of the tumor microenvironment that promotes growth, invasion, and metastasis.
Collapse
|
28
|
Ernst M, Thiem S, Nguyen PM, Eissmann M, Putoczki TL. Epithelial gp130/Stat3 functions: an intestinal signaling node in health and disease. Semin Immunol 2014; 26:29-37. [PMID: 24434062 DOI: 10.1016/j.smim.2013.12.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 12/23/2013] [Indexed: 12/16/2022]
Abstract
A contiguous intestinal epithelial barrier safeguards against aberrant activation of the immune system and therefore requires molecular mechanisms that ensure effective wound-healing responses. During this processes cytokine-producing myeloid cells serve as rheostats that link the degree of wounding and local inflammation to the epithelial repair response. Likewise, intestinal inflammation is an important factor by which the microenvironment promotes tumorigenesis and the progression of established cancers by facilitating neoplastic cell survival and proliferation. Among the cytokines and chemokines orchestrating this process, those comprising the interleukin (IL) IL6, IL10/IL22 and IL17/IL23 families play a prominent role by virtue of converging on the latent Signal Transducer and Activator of Transcription (Stat)-3. Accordingly, aberrant and persistent Stat3 activation is a frequent observation in cancers of the gastrointestinal tract where it promotes "cancer hallmark capabilities" in the malignant epithelium and suppresses the anti-tumor response of innate and adaptive immune cells. Here, we discuss recent insights arising from situations where persistent activation of the gp130/Stat3 signaling cascades result from excessive abundance of IL6 family cytokines. In particular, we highlight novel and unique roles for IL11 in promoting intestinal wound-healing and, in its corrupted form, enabling and facilitating growth of inflammation-associated and sporadic gastrointestinal tumors.
Collapse
Affiliation(s)
- Matthias Ernst
- The Walter and Eliza Hall Institute for Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Australia.
| | - Stefan Thiem
- The Walter and Eliza Hall Institute for Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Australia
| | - Paul M Nguyen
- The Walter and Eliza Hall Institute for Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Australia
| | - Moritz Eissmann
- The Walter and Eliza Hall Institute for Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Australia
| | - Tracy L Putoczki
- The Walter and Eliza Hall Institute for Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Australia
| |
Collapse
|
29
|
Nguyen AV, Wu YY, Liu Q, Wang D, Nguyen S, Loh R, Pang J, Friedman K, Orlofsky A, Augenlicht L, Pollard JW, Lin EY. STAT3 in epithelial cells regulates inflammation and tumor progression to malignant state in colon. Neoplasia 2013; 15:998-1008. [PMID: 24027425 PMCID: PMC3769879 DOI: 10.1593/neo.13952] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 06/26/2013] [Accepted: 07/02/2013] [Indexed: 01/05/2023]
Abstract
Chronic inflammation is an important risk factor for the development of colorectal cancer; however, the mechanism of tumorigenesis especially tumor progression to malignancy in the inflamed colon is still unclear. Our study shows that epithelial signal transducer and activator of transcription 3 (STAT3), persistently activated in inflamed colon, is not required for inflammation-induced epithelial overproliferation and the development of early-stage tumors; however, it is essential for tumor progression to advanced malignancy. We found that one of the mechanisms that epithelial STAT3 regulates in tumor progression might be to modify leukocytic infiltration in the large intestine. Activation of epithelial STAT3 promotes the infiltration of the CD8+ lymphocyte population but inhibits the recruitment of regulatory T (Treg) lymphocytes. The loss of Stat3 in epithelial cells promoted the expression of cytokines/chemokines including CCL19, CCL28, and RANTES, which are known to be able to recruit Treg lymphocytes. Linked to these changes was the pathway mediated by sphingosine 1-phosphate receptor 1 and sphingosine 1-phosphate kinases, which is activated in colonic epithelial cells in inflamed colon with functional STAT3 but not in epithelial cells deleted of STAT3. Our data suggest that epithelial STAT3 plays a critical role in inflammation-induced tumor progression through regulation of leukocytic recruitment especially the infiltration of Treg cells in the large intestine.
Collapse
Affiliation(s)
- Andrew V Nguyen
- Department of Biological Sciences and Geology, Queensborough-The City University of New York, Bayside, NY
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Schultz NA, Christensen IJ, Werner J, Giese N, Jensen BV, Larsen O, Bjerregaard JK, Pfeiffer P, Calatayud D, Nielsen SE, Yilmaz MK, Holländer NH, Wøjdemann M, Bojesen SE, Nielsen KR, Johansen JS. Diagnostic and Prognostic Impact of Circulating YKL-40, IL-6, and CA 19.9 in Patients with Pancreatic Cancer. PLoS One 2013; 8:e67059. [PMID: 23840582 PMCID: PMC3694124 DOI: 10.1371/journal.pone.0067059] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Accepted: 05/13/2013] [Indexed: 12/11/2022] Open
Abstract
Purpose We tested the hypothesis that high plasma YKL-40 and IL-6 associate with pancreatic cancer and short overall survival. Patients and Methods In all, 559 patients with pancreatic cancer from prospective biomarker studies from Denmark (n = 448) and Germany (n = 111) were studied. Plasma YKL-40 and IL-6 were determined by ELISAs and serum CA 19.9 by chemiluminescent immunometric assay. Results Odds ratios (ORs) for prediction of pancreatic cancer were significant for all biomarkers, with CA 19.9 having the highest AUC (CA 19.9: OR = 2.28, 95% CI 1.97 to 2.68, p<0.0001, AUC = 0.94; YKL-40: OR = 4.50, 3.99 to 5.08, p<0.0001, AUC = 0.87; IL-6: OR = 3.68, 3.08 to 4.44, p<0.0001, AUC = 0.87). Multivariate Cox analysis (YKL-40, IL-6, CA 19.9, age, stage, gender) in patients operated on showed that high preoperative IL-6 and CA 19.9 (dichotomized according to normal values) were independently associated with short overall survival (CA 19.9: HR = 2.51, 1.22–5.15, p = 0.013; IL-6: HR = 2.03, 1.11 to 3.70, p = 0.021). Multivariate Cox analysis of non-operable patients (Stage IIB-IV) showed that high pre-treatment levels of each biomarker were independently associated with short overall survival (YKL-40: HR = 1.30, 1.03 to 1.64, p = 0.029; IL-6: HR = 1.71, 1.33 to 2.20, p<0.0001; CA 19.9: HR = 1.54, 1.06 to 2.24, p = 0.022). Patients with preoperative elevation of both IL-6 and CA 19.9 had shorter overall survival (p<0.005) compared to patients with normal levels of both biomarkers (45% vs. 92% alive after 12 months). Conclusions Plasma YKL-40 and IL-6 had less diagnostic impact than CA 19.9. Combination of pretreatment YKL-40, IL-6, and CA 19.9 may have clinical value to identify pancreatic cancer patients with the poorest prognosis.
Collapse
Affiliation(s)
- Nicolai A. Schultz
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Oncology Copenhagen University Hospital at Herlev, Denmark
- Department of Surgical Gastroenterology, Copenhagen University Hospital at Herlev, Herlev, Denmark
- Department of Medicine, Copenhagen University Hospital at Herlev, Herlev, Denmark
- * E-mail:
| | - Ib J. Christensen
- The Finsen Laboratory, Rigshospitalet, and Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Jens Werner
- Department of General, Visceral, and Transplant Surgery, University of Heidelberg, Heidelberg, Germany
| | - Nathalia Giese
- Department of General, Visceral, and Transplant Surgery, University of Heidelberg, Heidelberg, Germany
| | - Benny V. Jensen
- Department of Oncology Copenhagen University Hospital at Herlev, Denmark
| | - Ole Larsen
- Department of Oncology Copenhagen University Hospital at Herlev, Denmark
| | | | - Per Pfeiffer
- Department of Oncology Odense University Hospital, Odense, Denmark
| | - Dan Calatayud
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | - Mette K. Yilmaz
- Department of Oncology Aalborg University Hospital, Aalborg, Denmark
| | | | - Morten Wøjdemann
- Department of Surgical Gastroenterology, Copenhagen University Hospital at Herlev, Herlev, Denmark
| | - Stig E. Bojesen
- Department of Clinical Biochemistry, Copenhagen University Hospital at Herlev, Herlev, Denmark
| | - Kaspar R. Nielsen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Julia S. Johansen
- Department of Oncology Copenhagen University Hospital at Herlev, Denmark
- Department of Medicine, Copenhagen University Hospital at Herlev, Herlev, Denmark
| |
Collapse
|
31
|
Interleukin-10-induced neutrophil gelatinase-associated lipocalin production in macrophages with consequences for tumor growth. Mol Cell Biol 2012; 32:3938-48. [PMID: 22851691 DOI: 10.1128/mcb.00413-12] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Tumor cell-derived factors, such as interleukin 10 (IL-10), polarize macrophages toward a regulatory M2 phenotype, characterized by the expression of anti-inflammatory cytokines and protumorigenic mediators. Here we explored molecular mechanisms allowing IL-10 to upregulate the protumorigenic protein NGAL in primary human macrophages. Reporter assays of full-length or deletion constructs of the NGAL promoter provided evidence that NGAL production is STAT3 dependent, activated downstream of the IL-10-Janus kinase (Jak) axis, as well as being C/EBPβ dependent. The involvement of STAT3 and C/EBPβ was shown by chromatin immunoprecipitation (ChIP) and ChIP-Western analysis, as well as decoy oligonucleotides scavenging both STAT3 and C/EBPβ in human macrophages. Furthermore, the production of NGAL in macrophages in response to IL-10 induces cellular growth and proliferation of MCF-7 breast cancer cells. We conclude that both STAT3 and C/EBPβ are needed to elicit IL-10-mediated NGAL expression in primary human macrophages. Macrophage-secreted NGAL shapes the protumorigenic macrophage phenotype to promote growth of MCF-7 breast cancer cells. Our data point to a macrophage-dependent IL-10-STAT3-NGAL axis that might contribute to tumor progression.
Collapse
|
32
|
Liu JP, Xu J. Introduction: Understanding the signalling mechanisms in molecular physiology and diseases. Clin Exp Pharmacol Physiol 2012; 39:658-60. [PMID: 22831391 DOI: 10.1111/j.1440-1681.2012.05740.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|