1
|
Ge L, Rui Y, Wang C, Wu Y, Wang H, Wang J. The RNA m 6A reader IGF2BP3 regulates NFAT1/IRF1 axis-mediated anti-tumor activity in gastric cancer. Cell Death Dis 2024; 15:192. [PMID: 38448411 PMCID: PMC10917814 DOI: 10.1038/s41419-024-06566-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024]
Abstract
N6-methyladenosine (m6A) and its associated reader protein insulin like growth factor 2 mRNA binding protein 3 (IGF2BP3) are involved in tumor initiation and progression via regulating RNA metabolism. This study aims to investigate the biological function and clinical significance of IGF2BP3 in gastric cancer (GC). The clinical significance of IGF2BP3 was evaluated using tumor related databases and clinical tissues. The biological role and molecular mechanism of IGF2BP3 in GC progression were investigated by multi-omics analysis including Ribosome sequence (Ribo-seq), RNA sequence (RNA-seq) and m6A sequence (m6A-seq) combined with gain- and loss- of function experiments. IGF2BP3 expression is significantly elevated in GC tissues and associated with poor prognosis of GC patients. Knockdown of IGF2BP3 significantly weakens the migration and clonogenic ability, promotes the apoptosis, inhibits translation, and suppresses in vitro growth and progression of GC cells. Mechanistically, IGF2BP3 regulates the mRNA stability and translation of the nuclear factor of activated T cells 1(NFAT1) in a m6A dependent manner. Then NFAT1 induced by IGF2BP3 acts as a transcription factor (TF) to negatively regulates the promoter activities of interferon regulatory factor 1 (IRF1) to inhibit its expression. Inhibition of IGF2BP3-induced expression of IRF1 activates interferon (IFN) signaling pathway and then exerts its anti-tumor effect. Elevated IGF2BP3 promotes in vivo and in vitro GC progression via regulation of NFAT1/IRF1 pathways. Targeted inhibition of IGF2BP3 might be a potential therapeutic approach for GC treatment.
Collapse
Affiliation(s)
- Lichen Ge
- Department of Clinical Laboratory, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yalan Rui
- Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Cheng Wang
- Department of Clinical Laboratory, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Yingmin Wu
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550009, China
| | - Hongsheng Wang
- Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Junjun Wang
- Department of Clinical Laboratory, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China.
| |
Collapse
|
2
|
Gibson AR, Sateriale A, Dumaine JE, Engiles JB, Pardy RD, Gullicksrud JA, O’Dea KM, Doench JG, Beiting DP, Hunter CA, Striepen B. A genetic screen identifies a protective type III interferon response to Cryptosporidium that requires TLR3 dependent recognition. PLoS Pathog 2022; 18:e1010003. [PMID: 35584177 PMCID: PMC9154123 DOI: 10.1371/journal.ppat.1010003] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 05/31/2022] [Accepted: 04/11/2022] [Indexed: 11/18/2022] Open
Abstract
Cryptosporidium is a leading cause of severe diarrhea and diarrheal-related death in children worldwide. As an obligate intracellular parasite, Cryptosporidium relies on intestinal epithelial cells to provide a niche for its growth and survival, but little is known about the contributions that the infected cell makes to this relationship. Here we conducted a genome wide CRISPR/Cas9 knockout screen to discover host genes that influence Cryptosporidium parvum infection and/or host cell survival. Gene enrichment analysis indicated that the host interferon response, glycosaminoglycan (GAG) and glycosylphosphatidylinositol (GPI) anchor biosynthesis are important determinants of susceptibility to C. parvum infection and impact on the viability of host cells in the context of parasite infection. Several of these pathways are linked to parasite attachment and invasion and C-type lectins on the surface of the parasite. Evaluation of transcript and protein induction of innate interferons revealed a pronounced type III interferon response to Cryptosporidium in human cells as well as in mice. Treatment of mice with IFNλ reduced infection burden and protected immunocompromised mice from severe outcomes including death, with effects that required STAT1 signaling in the enterocyte. Initiation of this type III interferon response was dependent on sustained intracellular growth and mediated by the pattern recognition receptor TLR3. We conclude that host cell intrinsic recognition of Cryptosporidium results in IFNλ production critical to early protection against this infection.
Collapse
Affiliation(s)
- Alexis R. Gibson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Adam Sateriale
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jennifer E. Dumaine
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Julie B. Engiles
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Pathobiology, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ryan D. Pardy
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jodi A. Gullicksrud
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Keenan M. O’Dea
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - John G. Doench
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Daniel P. Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
3
|
Schaalan M, Mohamed W, Fathy S. MiRNA-200c, MiRNA-139 and ln RNA H19; new predictors of treatment response in H-pylori- induced gastric ulcer or progression to gastric cancer. Microb Pathog 2020; 149:104442. [PMID: 32795593 DOI: 10.1016/j.micpath.2020.104442] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023]
Abstract
Recent evidence indicates that the pathogenesis of gastric ulcer and progression to gastric cancer could be attributed to altered inflammatory/immunological response and associated differential non-coding RNAs expression signatures. However, co-expression profiling of lncRNA-miRNAs in GU/GC patients are scarcely focused on. Therefore, in the present study the expression of H19 and related miRNAs including miR-139, and miR-200 were assayed in the plasma samples of treatment responsive GU vs nonresponsive GC patients. This study is a case-control study carried out on 130 subjects recruited from the Gastrointestinal Endoscopy Unit in Al-Kasr Al-Aini Hospital, in Egypt. All recruited patients were diagnosed with H-pylori infection, 50 of them were gastric cancer patients (GC), with previous H-pylori induced gastric ulcer but were treatment non-respondent. Real-time PCR was performed to evaluate the expression level of serum non-coding RNA; miRNA-200c, miR-139, Ln RNA H19 in patients with peptic ulcer treatment non-respondent, who progressed to GC vs non-progressed gastric ulcer patients (GU) (n = 50), and compared to early diagnosed H-pylori-gastric ulcer patients (n = 30). The association between these miRNAs and the FGF-18/FGF-R signaling indicators of H-pylori-GC pathogenesis were then investigated. RESULTS: showed that the H19 level was significantly elevated while miR-139 and miR-200c expression were significantly down-regulated in GC patients, compared to GU participants (P < 0.01). The herein investigated ncRNAs are correlated to the disease duration with Ln H19 being significantly correlated with all inflammatory markers; TNF-α, INF-γ, TAC, MMP-9, and FGF18/FGFR2. A significant correlation was also observed between miRNA 200c and each of miRNA 139 and FGFR2. Moreover, ROC analysis revealed that miRNA 200c showed the highest AUC (0.906) and 81.2% sensitivity and 100% specificity. Moreover, the combined analysis of miRNA 200c/miRNA 139 revealed superior AUC (0.96) and 93% sensitivity and 100% specificity, than each separately. As for discriminative accuracy between stages III to IV of gastric cancer, LncRNA H19 showed the highest diagnostic accuracy (95.5%), specificity (100%), and sensitivity (90.9%). The current study demonstrated that the combination of serum miRNA 200c/miRNA 139 expression levels (down-regulation) could provide a new potential prognostic panel for GU predictive response and potential sequelae. In conclusion, LncRNA H19 and related miRNAs, miRNA 200c/miRNA 139, could serve as a potential diagnostic biomarker for early gastric cancer diagnosis.
Collapse
Affiliation(s)
- Mona Schaalan
- Department of Clinical Pharmacy, Clinical and Translational Research Unit, Faculty of Pharmacy, Misr International University, Cairo, Egypt.
| | - Waleed Mohamed
- Department of Internal Medicine, Kasr El Aini Teaching Hospitals, Cairo University, Cairo, Egypt.
| | - Shimaa Fathy
- Department of Clinical Pharmacy, Clinical and Translational Research Unit, Faculty of Pharmacy, Misr International University, Cairo, Egypt.
| |
Collapse
|
4
|
Lam T, Aguirre‐Ghiso JA, Geller MA, Aksan A, Azarin SM. Immobilization rapidly selects for chemoresistant ovarian cancer cells with enhanced ability to enter dormancy. Biotechnol Bioeng 2020; 117:3066-3080. [DOI: 10.1002/bit.27479] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/24/2020] [Accepted: 06/24/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Tiffany Lam
- Department of Chemical Engineering and Materials Science University of Minnesota Minneapolis Minnesota
| | - Julio A. Aguirre‐Ghiso
- Division of Hematology and Oncology, Department of Medicine, Tisch Cancer Institute, Black Family Stem Cell Institute, Precision Immunology Institute Icahn School of Medicine at Mount Sinai New York New York
| | - Melissa A. Geller
- Department of Obstetrics, Gynecology and Women's Health, Division of Gynecologic Oncology University of Minnesota Minneapolis Minnesota
| | - Alptekin Aksan
- Department of Mechanical Engineering University of Minnesota Minneapolis Minnesota
| | - Samira M. Azarin
- Department of Chemical Engineering and Materials Science University of Minnesota Minneapolis Minnesota
| |
Collapse
|
5
|
Zheng Y, Guo C, Zhang X, Wang X, Ma A. Garcinol acts as an antineoplastic agent in human gastric cancer by inhibiting the PI3K/AKT signaling pathway. Oncol Lett 2020; 20:667-676. [PMID: 32565991 PMCID: PMC7285879 DOI: 10.3892/ol.2020.11585] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 04/15/2020] [Indexed: 12/13/2022] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies worldwide; however, treatment options other than surgery remain limited. Neoadjuvant chemotherapy has the potential to suppress of gastric tumorigenesis. Garcinol has been reported to exert inhibitory effects on the progression of numerous carcinomas. However, its effects in GC remain unclear. Therefore, the aim of the present study was to investigate the effects of garcinol on the proliferation, invasion and apoptosis of gastric carcinoma cells and then to explore the underlying mechanisms. Garcinol significantly decreased the proliferation and invasion of GC cells and increased apoptosis in a dose-dependent manner. Additionally, the expression of AKTp-Thr308, cyclin D1, Bcl-2, BAX, matrix metalloprotease (MMP-2) and MMP-9 in HGC-27 cells following treatment with garcinol. The results obtained in the present study suggested that garcinol may inhibit gastric tumorigenesis by suppressing the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Yuanyuan Zheng
- Department of Gastroenterology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang 312000, P.R. China
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai 200072, P.R. China
| | - Xiaoping Zhang
- Department of Gastroenterology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang 312000, P.R. China
| | - Xiaoli Wang
- Department of Gastroenterology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang 312000, P.R. China
| | - A'Huo Ma
- Department of Gastroenterology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang 312000, P.R. China
| |
Collapse
|
6
|
Mohamed WA, Schaalan MF, Ramadan B. The expression profiling of circulating miR-204, miR-182, and lncRNA H19 as novel potential biomarkers for the progression of peptic ulcer to gastric cancer. J Cell Biochem 2019; 120:13464-13477. [PMID: 30945348 DOI: 10.1002/jcb.28620] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/17/2019] [Accepted: 02/28/2019] [Indexed: 12/17/2022]
Abstract
Deregulation of noncoding RNAs, microRNAs (miRNAs) and long noncoding RNA (lncRNA), are implicated in the initiation and progression of gastric cancer (GC). This study is a pilot case-control study carried out on 75 subjects, 40 of them were Helicobacter pylori-gastric ulcer patients and 35 were GC patients recruited from the Gastrointestinal Endoscopy Unit in Al-Kasr Al-Aini Hospital, Cairo University in Egypt. Real-time PCR was performed to evaluate the expression level of serum miR-204, miR-182, and lncRNA H19 in patients with peptic ulcer-progressed GC vs nonprogressed peptic ulcer patients. Fibroblast growth factor 18 (FGF-18)/FGF receptor 2 (FGFR2) expression and their downstream immunological and inflammatory signaling markers were assessed and their association with the addressed noncoding RNAs investigated. As regards miR-204 and miR-182, they were significantly increased (12.5 and 2.6 folds, respectively) in GU samples, compared with those of healthy control levels. The elevated levels of these miRNAs were significantly de-escalated in GC samples compared with GU and the fold decrease valued 2.2 fold for miR-204 and 1.8 folds for miR-182. On the other hand, the significant escalation in the level of lnRNA H19 in GU recorded a 16.6 fold increase and further elevation in its levels was evident in GC samples. The herein assessed miRNAs are correlated with disease duration and FGFR2 with miR-182 being significantly correlated with all inflammatory markers, TAC, INF-γ, matrix metallopeptidase 9, and FGF-18. In terms of diagnostic accuracy of assessed miRNAs (stages III to IV), the receiver operating characteristic analysis indicated that serum lncRNA H19 showed the highest diagnostic accuracy (95.5%), specificity (100%), and sensitivity (90.9%), compared with miR-204 and miR-182, which showed the same specificity (60%), sensitivity (72.7%), and diagnostic accuracy (68.8%). Our findings conclude that lnRNA H19, miR-204, and miR-182 may function as novel prospective plasma biomarkers to detect GC and its progression from H. pylori-peptic ulcer, which would be helpful to improve the theranostics of GC.
Collapse
Affiliation(s)
- Waleed A Mohamed
- Department of Chemistry, Kasr El Aini Teaching Hospital, Cairo University, Cairo, Egypt
| | - Mona F Schaalan
- Department of Clinical Pharmacy and Pharmacy Practice, Clinical and Translational Research Unit, Faculty of Pharmacy, Misr International University, Cairo, Egypt
| | - Basma Ramadan
- Department of Physiology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
7
|
Li CJ, Liao WT, Wu MY, Chu PY. New Insights into the Role of Autophagy in Tumor Immune Microenvironment. Int J Mol Sci 2017; 18:1566. [PMID: 28753959 PMCID: PMC5536054 DOI: 10.3390/ijms18071566] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 07/11/2017] [Accepted: 07/13/2017] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment is a complex system that is affected by various factors, including hypoxia, acidosis, and immune and inflammatory responses, which have significant effects on tumor adhesion, invasion, metastasis, angiogenesis, and autophagy. In this hostile tumor microenvironment, autophagy of tumor cells can promote tumor growth and metastasis. As autophagy is a double-edged sword in tumors, treatment of cancer via regulation of autophagy is extremely complicated. Therefore, understanding the relationship between tumor autophagy and the tumor microenvironment is extremely important. As the immune milieu plays an important role in tumor development, immunotherapy has become a promising form of cancer therapy. A multi-pronged treatment approach using immunotherapy and molecular targets may become the major direction for future cancer treatments. This article reviews existing knowledge regarding the immune factors in the tumor microenvironment and the status of tumor autophagy research.
Collapse
Affiliation(s)
- Chia-Jung Li
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
| | - Wan-Ting Liao
- Chinese Medicine Department, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
- Graduate Institute of Chinese Medicine, China Medical University, Taichung 404, Taiwan.
| | - Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan.
| | - Pei-Yi Chu
- Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan.
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| |
Collapse
|
8
|
Kelm NE, Zhu Z, Ding VA, Xiao H, Wakefield MR, Bai Q, Fang Y. The role of IL-29 in immunity and cancer. Crit Rev Oncol Hematol 2016; 106:91-8. [PMID: 27637354 PMCID: PMC7129698 DOI: 10.1016/j.critrevonc.2016.08.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 07/27/2016] [Accepted: 08/09/2016] [Indexed: 02/06/2023] Open
Abstract
Interleukin-29 (IL-29) is a new member of the recently discovered interferon λ (IFNλ) family. It is produced predominantly by maturing dendritic cells and macrophages. It has been implicated in numerous immunological responses and has shown antiviral activity similar to the Type I interferons, although its target cell population is more limited than the Type I interferons. In recent years, the role of IL-29 in the pathogenesis of various cancers has also been extensively studied. In this review, we will discuss the recent advances of IL-29 in immunological processes and the pathogenesis of various cancer.
Collapse
Affiliation(s)
- Noah E Kelm
- Department of Microbiology, Immunology & Pathology, Des Moines University, Des Moines, IA 50312, United States
| | - Ziwen Zhu
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, United States
| | - Vivi A Ding
- Department of Microbiology, Immunology & Pathology, Des Moines University, Des Moines, IA 50312, United States
| | - Huaping Xiao
- Department of Microbiology, Immunology & Pathology, Des Moines University, Des Moines, IA 50312, United States; The Affiliated Hospital of Xiangnan University, Chenzhou, Hunan, China
| | - Mark R Wakefield
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, United States
| | - Qian Bai
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, United States
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, Des Moines University, Des Moines, IA 50312, United States; Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, United States.
| |
Collapse
|
9
|
Bu X, Wang M, Zhang J, Liu J, Jia L, Liang B, Yan Y. Recombinant adenovirus expressing hIFN-λ1 inhibits gastric adenocarcinoma cell line SGC-7901 proliferation. Oncol Lett 2015; 11:287-292. [PMID: 26870205 DOI: 10.3892/ol.2015.3890] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 09/24/2015] [Indexed: 01/03/2023] Open
Abstract
The aim of the present study is to investigate the effect of a recombinant plasmid adenovirus (pAd) expressing human interferon-λ1 (hIFN-λ1) on the proliferation of the gastric adenocarcinoma cell line SGC-7901. For this purpose, human gastric adenocarcinoma SGC-7901 cells were infected with recombinant pAd-hIFN-λ1, pAd-LacZ and phosphate-buffered saline (PBS), respectively, and the subsequent effects on the proliferation of the infected cells were compared. Cell proliferation was evaluated by MTT assay, while mRNA and protein expression of hIFN-λ1 were detected by reverse transcription-polymerase chain reaction analysis and immunofluorescence assay, respectively. In addition, terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling assay and flow cytometry were conducted to analyze the rate of cell apoptosis. The results indicated that the proliferation of gastric adenocarcinoma SGC-7901 cells was significantly inhibited by pAd-hIFN-λ1. Furthermore, the apoptosis rate and the mRNA and protein expression levels of hIFN-λ1 were higher in pAd-hIFN-λ1-transfected cells, compared with the pAd-LacZ and PBS control groups. In conclusion, recombinant pAd-hIFN-λ1 induced the expression of hIFN-λ1 in gastric adenocarcinoma SGC-7901 cells, and significantly inhibited cell proliferation by promoting apoptosis in these cancer cells.
Collapse
Affiliation(s)
- Xuefeng Bu
- Department of General Surgery, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Mubin Wang
- Department of Surgical Medicine, Clinical Medicine College of Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Jie Zhang
- Department of Surgical Medicine, Clinical Medicine College of Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Jun Liu
- Department of Surgical Medicine, Clinical Medicine College of Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Lijuan Jia
- Department of Internal Medicine, Clinical Medicine College of Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Bing Liang
- Department of Internal Medicine, Clinical Medicine College of Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yulan Yan
- Department of Respiratory Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| |
Collapse
|
10
|
Silencing stathmin-modulating efficiency of chemotherapy for esophageal squamous cell cancer with paclitaxel. Cancer Gene Ther 2015; 22:115-21. [PMID: 25572118 DOI: 10.1038/cgt.2014.74] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 11/20/2014] [Accepted: 11/21/2014] [Indexed: 01/04/2023]
Abstract
Paclitaxel (PTX) is broadly considered the drug of choice for treating human esophageal squamous cell cancer (ESCC). However, PTX resistance often ultimately leads to treatment failure. stathmin, or Op18, is a ubiquitously expressed 19-kDa cytosolic phosphoprotein that can integrate various cellular regulatory signals. stathmin overexpression could lead to resistance to chemotherapeutic agents. In this study we investigated the effect of stathmin gene silencing, using small interfering RNA (stathmin siRNA), on the efficacy of PTX in ESCC. Transfection of stathmin siRNA could significantly inhibit stathmin mRNA and protein levels in ESCC cell lines EC9706 and Eca-109. The silencing of stathmin combined with PTX significantly inhibited the proliferation of EC9706 and Eca-109 cells, with a significantly higher proportion of cells at G2/M phase and this antiproliferative effect was accompanied by an increase in apoptosis rates and morphology changes of EC9706 and Eca-109. Thus, combined chemotherapeutic agent PTX and stathmin siRNA could potentially enhance the therapeutic outcomes of PTX in treating ESCC.
Collapse
|