1
|
Cheng F, Zhao S, Li J, Niu Y, Huang H, Yang J, Ma S, Liu J, Sun P. Enhanced effect of recombinant adenoviruses co‐expression of
ING4
and
OSM
on anti‐tumour activity of laryngeal cancer. J Cell Mol Med 2022; 26:1556-1566. [PMID: 35075768 PMCID: PMC8899183 DOI: 10.1111/jcmm.17192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 01/02/2022] [Accepted: 01/08/2022] [Indexed: 11/29/2022] Open
Abstract
The inhibitor of growth family member 4 (ING4) is one of the ING family genes, serves as a repressor of angiogenesis or tumour growth and suppresses loss of contact inhibition. Oncostatin M (OSM) is a multifunctional cytokine that belongs to the interleukin (IL)‐6 subfamily with several biological activities. However, the role of recombinant adenoviruses co‐expressing ING4 and OSM (Ad‐ING4‐OSM) in anti‐tumour activity of laryngeal cancer has not yet been identified. Recombinant Ad‐ING4‐OSM was used to evaluate their combined effect on enhanced anti‐tumour activity in Hep‐2 cells of laryngeal cancer in vivo. Moreover, in vitro function assays of co‐expression of Ad‐ING4‐OSM were performed to explore impact of co‐expression of Ad‐ING4‐OSM on biological phenotype of laryngeal cancer cell line, that is Hep‐2 cells. In vitro, Ad‐ING4‐OSM significantly inhibited the growth, enhanced apoptosis, altered cell cycle with G1 and G2/M phase arrest, and upregulated the expression of P21, P27, P53 and downregulated survivin in laryngeal cancer Hep‐2 cells. Furthermore, in vivo functional experiments of co‐expressing of Ad‐ING4‐OSM demonstrated that solid tumours in the nude mouse model were significantly suppressed, and the co‐expressing Ad‐ING4‐OSM showed a significant upregulation expression of P21, P53, Bax and Caspase‐3 and a downregulation of Cox‐2, Bcl‐2 and CD34. This study for the first time demonstrated the clinical value and the role of co‐expressing Ad‐ING4‐OSM in biological function of laryngeal cancer. This work suggested that co‐expressing Ad‐ING4‐OSM might serve as a potential therapeutic target for laryngeal cancer patients.
Collapse
Affiliation(s)
- Fuwei Cheng
- Department of Otolaryngology The First Affiliated Hospital of Soochow University Suzhou China
| | - Shuangping Zhao
- Department of Otolaryngology The First Affiliated Hospital of Soochow University Suzhou China
| | - Jiachen Li
- Department of Otolaryngology The First Affiliated Hospital of Soochow University Suzhou China
| | - Yuyu Niu
- Department of Otolaryngology The First Affiliated Hospital of Soochow University Suzhou China
| | - Haiping Huang
- Department of Otolaryngology The First Affiliated Hospital of Soochow University Suzhou China
| | - Jicheng Yang
- Cell and Molecular Biology Institute College of Medicine Soochow University Suzhou China
| | - Shiyin Ma
- Department of Otolaryngology The First Affiliated Hospital of Bengbu Medical College Bengbu China
| | - Jisheng Liu
- Department of Otolaryngology The First Affiliated Hospital of Soochow University Suzhou China
| | - Peng Sun
- Department of Otolaryngology The First Affiliated Hospital of Soochow University Suzhou China
| |
Collapse
|
2
|
Shatnawi A, Abu Rabe DI, Frigo DE. Roles of the tumor suppressor inhibitor of growth family member 4 (ING4) in cancer. Adv Cancer Res 2021; 152:225-262. [PMID: 34353439 DOI: 10.1016/bs.acr.2021.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Inhibitor of growth family member 4 (ING4) is best known as a tumor suppressor that is frequently downregulated, deleted, or mutated in many cancers. ING4 regulates a broad array of tumor-related processes including proliferation, apoptosis, migration, autophagy, invasion, angiogenesis, DNA repair and chromatin remodeling. ING4 alters local chromatin structure by functioning as an epigenetic reader of H3K4 trimethylation histone marks (H3K4Me3) and regulating gene transcription through directing histone acetyltransferase (HAT) and histone deacetylase (HDAC) protein complexes. ING4 may serve as a useful prognostic biomarker for many cancer types and help guide treatment decisions. This review provides an overview of ING4's central functions in gene expression and summarizes current literature on the role of ING4 in cancer and its possible use in therapy.
Collapse
Affiliation(s)
- Aymen Shatnawi
- Department of Pharmaceutical and Administrative Sciences, University of Charleston School of Pharmacy, Charleston, WV, United States.
| | - Dina I Abu Rabe
- Integrated Bioscience Program, North Carolina Central University, Durham, NC, United States
| | - Daniel E Frigo
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, United States; Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
3
|
The essential role of tumor suppressor gene ING4 in various human cancers and non-neoplastic disorders. Biosci Rep 2019; 39:BSR20180773. [PMID: 30643005 PMCID: PMC6356015 DOI: 10.1042/bsr20180773] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 12/19/2018] [Accepted: 01/13/2019] [Indexed: 12/21/2022] Open
Abstract
Inhibitor of growth 4 (ING4), a member of the ING family discovered in 2003, has been shown to act as a tumor suppressor and is frequently down-regulated in various human cancers. Numerous published in vivo and in vitro studies have shown that ING4 is responsible for important cancer hallmarks such as pathologic cell cycle arrest, apoptosis, autophagy, contact inhibition, and hypoxic adaptation, and also affects tumor angiogenesis, invasion, and metastasis. These characteristics are typically associated with regulation through chromatin acetylation by binding histone H3 trimethylated at lysine 4 (H3K4me3) and through transcriptional activity of transcription factor P53 and NF-κB. In addition, emerging evidence has indicated that abnormalities in ING4 expression and function play key roles in non-neoplastic disorders. Here, we provide an overview of ING4-modulated chromosome remodeling and transcriptional function, as well as the functional consequences of different genetic variants. We also present the current understanding concerning the role of ING4 in the development of neoplastic and non-neoplastic diseases. These studies offer inspiration for pursuing novel therapeutics for various cancers.
Collapse
|
4
|
Wu Y, Mou X, Wang S, Liu XE, Sun X. ING4 expressing oncolytic vaccinia virus promotes anti-tumor efficiency and synergizes with gemcitabine in pancreatic cancer. Oncotarget 2017; 8:82728-82739. [PMID: 29137298 PMCID: PMC5669924 DOI: 10.18632/oncotarget.21095] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/26/2017] [Indexed: 12/20/2022] Open
Abstract
With no effective treatments available for most pancreatic cancer patients, pancreatic cancer continues to be one of the most difficult malignancies to treat. Oncolytic virus mediated-gene therapy has exhibited ubiquitous antitumor potential. In this study, we constructed a novel oncolytic vaccinia virus harboring the inhibitor of growth family member 4 gene (VV-ING4) to investigate its therapeutic efficacy alone or in combination with gemcitabine against pancreatic cancer cells in vitro and in vivo. ING4 expression was determined via quantitative real-time polymerase chain reaction (qPCR) and western blot. The cytotoxicity of VV-ING4 was measured using a cell proliferation assay. Both flow cytometry and western blot were applied to analyze the cell cycle and apoptosis. Furthermore, the combination inhibitory effect of VV-ING4 and gemcitabine was assessed using Chou-Talalay analysis in vitro and a BLAB/c mice model in vivo. We found that VV-ING4 significantly increases ING4 expression, displayed greater cytotoxic efficiency, and induced pancreatic cancer cell apoptosis and G2/M phase arrest. Additionally, the combination of VV-ING4 and gemcitabine synergistically effect in vitro and in vivo. Taken together, our data implicate VV-ING4 as a conceivable pancreatic cancer therapeutic candidate alone or in combination with gemcitabine.
Collapse
Affiliation(s)
- Yinfang Wu
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, P. R. China.,The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, P. R. China.,Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, P. R. China
| | - Xiaozhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, P. R. China.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou 310014, P. R. China
| | - Shibing Wang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, P. R. China.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou 310014, P. R. China
| | - Xing-E Liu
- Department of Medical Oncology, Zhejiang Hospital, Hangzhou 310007, P. R. China
| | - Xiaodong Sun
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, P. R. China.,The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, P. R. China.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou 310014, P. R. China
| |
Collapse
|
5
|
Park KM, Teoh JP, Wang Y, Broskova Z, Bayoumi AS, Tang Y, Su H, Weintraub NL, Kim IM. Carvedilol-responsive microRNAs, miR-199a-3p and -214 protect cardiomyocytes from simulated ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol 2016; 311:H371-83. [PMID: 27288437 PMCID: PMC5005281 DOI: 10.1152/ajpheart.00807.2015] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 06/01/2016] [Indexed: 12/24/2022]
Abstract
The nonselective β-adrenergic receptor antagonist (β-blocker) carvedilol has been shown to protect against myocardial injury, but the detailed underlying mechanisms are unclear. We recently reported that carvedilol stimulates the processing of microRNA (miR)-199a-3p and miR-214 in the heart via β-arrestin1-biased β1-adrenergic receptor (β1AR) cardioprotective signaling. Here, we investigate whether these β-arrestin1/β1AR-responsive miRs mediate the beneficial effects of carvedilol against simulated ischemia/reperfusion (sI/R). Using cultured cardiomyocyte cell lines and primary cardiomyocytes, we demonstrate that carvedilol upregulates miR-199a-3p and miR-214 in both ventricular and atrial cardiomyocytes subjected to sI/R. Overexpression of the two miRs in cardiomyocytes mimics the effects of carvedilol to activate p-AKT survival signaling and the expression of a downstream pluripotency marker Sox2 in response to sI/R. Moreover, carvedilol-mediated p-AKT activation is abolished by knockdown of either miR-199a-3p or miR-214. Along with previous studies to directly link the cardioprotective actions of carvedilol to upregulation of p-AKT/stem cell markers, our findings suggest that the protective roles of carvedilol during ischemic injury are in part attributed to activation of these two protective miRs. Loss of function of miR-199a-3p and miR-214 also increases cardiomyocyte apoptosis after sI/R. Mechanistically, we demonstrate that miR-199a-3p and miR-214 repress the predictive or known apoptotic target genes ddit4 and ing4, respectively, in cardiomyocytes. These findings suggest pivotal roles for miR-199a-3p and miR-214 as regulators of cardiomyocyte survival and contributors to the functional benefits of carvedilol therapy.
Collapse
Affiliation(s)
- Kyoung-Mi Park
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Jian-Peng Teoh
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Yongchao Wang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Zuzana Broskova
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Ahmed S Bayoumi
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Yaoliang Tang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Huabo Su
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Neal L Weintraub
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Il-Man Kim
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
6
|
Rakshit N, Yang S, Zhou W, Xu Y, Deng C, Yang J, Yu H, Wei W. Adenovirus-mediated co-expression of ING4 and PTEN cooperatively enhances their antitumor activity in human hepatocellular carcinoma cells. Acta Biochim Biophys Sin (Shanghai) 2016; 48:704-13. [PMID: 27421660 DOI: 10.1093/abbs/gmw062] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/24/2016] [Indexed: 11/13/2022] Open
Abstract
Both inhibitor of growth 4 (ING4) and phosphatase and tensin homolog deleted on chromosome 10 (PTEN) are well known as tumor suppressors that are closely related to tumor occurrence and progression. It was reported that ING4 and PTEN showed synergistic antitumor activities in nasopharyngeal carcinoma cells. The two tumor suppressors demonstrated synergistic effect on growth inhibition and apoptosis activation. In this study, we investigated their therapeutic potential in hepatocellular carcinoma (HCC) cells. Recombinant adenoviruses co-expressing ING4 and PTEN (Ad-ING4-PTEN) were constructed, and the antitumor effect on SMMC-7721 and HepG2 HCC cells was evaluated. Ad-ING4-PTEN cooperatively inhibited cell growth, stimulated apoptosis, and suppressed invasion in both HCC cells, and regulated cell cycle in SMMC-7721. Further studies showed that the combination of ING4 and PTEN by Ad-ING4-PTEN cooperatively enhanced the alteration of the expression of cell cycle-related proteins (p53, p21, and cyclin D1) and apoptotic factors (Bad, Bcl-2, Bcl-XL, and Bax), which are involved in the regulation of cell cycle and the activation of apoptotic pathways, leading to the synergistic antitumor effect. These results indicate that the combination of ING4 and PTEN may provide an effective therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Nargis Rakshit
- Department of Cell Biology, School of Medicine, Soochow University, Suzhou 215123, China
| | - Sijun Yang
- School of Life Science, Shangrao Normal University, Shangrao 334001, China
| | - Wei Zhou
- Department of Cell Biology, School of Medicine, Soochow University, Suzhou 215123, China
| | - Yi Xu
- Department of Cell Biology, School of Medicine, Soochow University, Suzhou 215123, China
| | - Chenghui Deng
- Department of Cell Biology, School of Medicine, Soochow University, Suzhou 215123, China
| | - Jiecheng Yang
- Department of Cell Biology, School of Medicine, Soochow University, Suzhou 215123, China
| | - Huijun Yu
- Department of Cell Biology, School of Medicine, Soochow University, Suzhou 215123, China
| | - Wenxiang Wei
- Department of Cell Biology, School of Medicine, Soochow University, Suzhou 215123, China
| |
Collapse
|
7
|
Pan X, Wang R, Bian H, De W, Zhang P, Wei C, Wang Z. Overexpression of Inhibitor of Growth 4 Enhances Radiosensitivity in Non-Small Cell Lung Cancer Cell Line SPC-A1. Technol Cancer Res Treat 2016; 16:533-545. [PMID: 27381846 DOI: 10.1177/1533034616656315] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Inhibitor of growth 4 is a member of the inhibitor of growth family proteins, which is involved in cell apoptosis, migration, invasion, and cell cycle progress. In this study, we investigated the inhibitor of growth 4 level in non-small cell lung cancer tissues and explored the antitumor activity of inhibitor of growth 4 in vitro and in vivo using non-small cell lung cancer cell line SPC-A1 and its underlying molecular mechanisms. We also explored its role on the radiosensitivity in SPC-A1 cells. The level of inhibitor of growth 4 protein was significantly decreased in 28 cases of non-small cell lung cancer tissues in comparison with corresponding noncancerous lung epithelial tissues. Upregulation of inhibitor of growth 4 by plasmid pcDNA3.1-ING4 delivery could suppress proliferation and increase apoptosis of SPC-A1 cells both in vitro and in vivo. Additionally, we found that overexpression of inhibitor of growth 4 in SPC-A1 cell line could lead to a higher Bcl-2/Bax ratio, which might be an important factor in the apoptosis regulation. Furthermore, overexpression of inhibitor of growth 4 enhanced the radiosensitivity of SPC-A1 cells to irradiation. Inhibitor of growth 4 upregulation plus radiotherapy induced synergistic tumor suppression in SPC-A1 xenografts implanted in athymic nude mice. Thus, the restoration of inhibitor of growth 4 function might provide a potential strategy for non-small cell lung cancer radiosensitization.
Collapse
Affiliation(s)
- Xuan Pan
- 1 Department of Medical Oncology, Nanjing Medical University Affiliated Cancer Hospital of Jiangsu Province, Cancer Institution of Jiangsu Province, Nanjing, China
- 2 Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Wang
- 3 Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Haibo Bian
- 2 Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei De
- 4 Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Ping Zhang
- 5 Department of Pathology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chenchen Wei
- 2 Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhaoxia Wang
- 2 Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
8
|
Ashshi AM, El-Shemi AG, Dmitriev IP, Kashentseva EA, Curiel DT. Combinatorial strategies based on CRAd-IL24 and CRAd-ING4 virotherapy with anti-angiogenesis treatment for ovarian cancer. J Ovarian Res 2016; 9:38. [PMID: 27349517 PMCID: PMC4924320 DOI: 10.1186/s13048-016-0248-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 06/22/2016] [Indexed: 01/10/2023] Open
Abstract
Background A major hurdle incurrent to the human clinical application of conditionally replicative adenovirus (CRAd)-based virotherapy agents is their limited therapeutic efficacy. In this study we evaluated whether arming our previously reported Ad5/3Δ24 CRAd vector containing a 24-base pair deletion in the E1A conserved region 2, which allows selective replication within Rb-p16-deficient tumor cells, to express therapeutic genes could improve oncolytic virus potency in ovarian cancer cells. We choose to assess the therapeutic benefits achieved by virus-mediated expression of interleukin 24 (IL-24), a cytokine-like protein of the IL-10 family, and the inhibitor of growth 4 (ING4) tumor suppressor protein. Results The generated CRAd-IL24 and CRAd-ING4 vectors were tested in ovarian cancer cell lines in vitro to compare their replication, yield, and cytotoxic effects with control CRAd Ad5/3∆24 lacking the therapeutic gene. These studies showed that CRAd-IL24 infection resulted in significantly increased yield of infectious particles, which translated to a marked enhancement of virus-induced cytotoxic effects as compared to CRAd-ING4 and non-armed CRAd. Testing CRAd-IL24 and CRAd-ING4 vectors combined together did not revealed synergistic effects exceeding oncolytic potency of single CRAD-IL24 vector. Both CRAds were also tested along with anti-VEGF monoclonal antibody Avastin and showed no significant augmentation of viral cytolysis by anti-angiogenesis treatment in vitro. Conclusions Our studies validated that arming with these key immunomodulatory genes was not deleterious to virus-mediated oncolysis. These findings thus, warrant further preclinical studies of CRAd-IL24 tumoricidal efficacy in murine ovarian cancer models to establish its potential utility for the virotherapy of primary and advanced neoplastic diseases.
Collapse
Affiliation(s)
- Ahmad Mohammad Ashshi
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, PO Box 7607, Holy Makkah, Saudi Arabia
| | - Adel Galal El-Shemi
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, PO Box 7607, Holy Makkah, Saudi Arabia.,Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Igor P Dmitriev
- The Division of Cancer Biology and Biologic Therapeutic Center, Department of Radiation Oncology, School of Medicine, Washington University in Saint Louis, 660 South Euclid Avenue, Campus Box 8224, St. Louis, MO, 63110, USA
| | - Elena A Kashentseva
- The Division of Cancer Biology and Biologic Therapeutic Center, Department of Radiation Oncology, School of Medicine, Washington University in Saint Louis, 660 South Euclid Avenue, Campus Box 8224, St. Louis, MO, 63110, USA
| | - David T Curiel
- The Division of Cancer Biology and Biologic Therapeutic Center, Department of Radiation Oncology, School of Medicine, Washington University in Saint Louis, 660 South Euclid Avenue, Campus Box 8224, St. Louis, MO, 63110, USA.
| |
Collapse
|
9
|
Qi R, Qiao T, Zhuang X. Small interfering RNA targeting S100A4 sensitizes non-small-cell lung cancer cells (A549) to radiation treatment. Onco Targets Ther 2016; 9:3753-62. [PMID: 27382312 PMCID: PMC4922784 DOI: 10.2147/ott.s106557] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Objective This study aimed to investigate the impact of S100A4-small interfering RNA (S100A4-siRNA) on apoptosis and enhanced radiosensitivity in non-small-cell lung cancer (A549) cells. We also explored the mechanisms of radiosensitization and identified a new target to enhance radiosensitivity and gene therapy for non-small-cell lung cancer. Methods RNA interference is a powerful tool for gene silencing. In this study, we constructed an effective siRNA to knock down S100A4. A549 cells were randomly divided into three groups: blank, negative control, and S100A4-siRNA. To investigate the effect of S100A4-siRNA, the expression of S100A4, E-cadherin, and p53 proteins and their messenger RNA (mRNA) was detected by Western blot and quantitative real-time polymerase chain reaction. Transwell chambers were used to assess cell invasion. Cell cycle and apoptosis were analyzed by flow cytometry. Radiosensitivity was determined by colony formation ability. Results Our results demonstrate that S100A4-siRNA effectively silenced the S100A4 gene. When siRNA against S100A4 was used, S100A4 protein expression was downregulated, whereas the expressions of E-cadherin and p53 were upregulated. In addition, a clear reduction in S100A4 mRNA levels was noted compared with the blank and negative control groups, whereas E-cadherin and p53 mRNA levels increased. Transfection with S100A4-siRNA significantly reduced the invasiveness of A549 cells. S100A4 silencing induced immediate G2/M arrest in cell cycle studies and increased apoptosis rates in A549 cells. In clonogenic assays, we used a multitarget, single-hit model to detect radiosensitivity after S100A4 knockdown. All parameters (D0, Dq, α, β) indicated that the downregulation of S100A4 enhanced radiosensitivity in A549 cells. Furthermore, S100A4-siRNA upregulated p53 expression, suggesting that S100A4 may promote A549 cell proliferation, invasion, and metastasis by regulating the expression of other proteins. Therefore, siRNA-directed S100A4 knockdown may represent a viable clinical therapy for lung cancer. Conclusion S100A4 downregulation potentially enhances the sensitivity of human A549 cells to radiotherapy.
Collapse
Affiliation(s)
- Ruixue Qi
- Department of Oncology, Affiliated Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Tiankui Qiao
- Department of Oncology, Affiliated Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Xibing Zhuang
- Department of Oncology, Affiliated Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
10
|
Zhao D, Liu X, Zhang Y, Ding Z, Dong F, Xu H, Wang B, Wang W. Reduced expression and prognostic implication of inhibitor of growth 4 in human osteosarcoma. Oncol Lett 2016; 11:2869-2874. [PMID: 27073567 DOI: 10.3892/ol.2016.4324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 02/16/2016] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma is the most prevalent type of primary malignant bone tumor. Inhibitor of growth 4 (ING4) has been demonstrated to function as a tumor suppressor through multiple pathways, and is its expression is understood to be suppressed or reduced in various malignancies. The present study aimed to investigate the expression of ING4 and to determine its prognostic value in osteosarcoma tissue. Formalin-fixed, paraffin-embedded tissue microarrays were analyzed, and contained 41 osteosarcoma specimens and 11 normal bone tissue specimens with duplicate cores. ING4 expression was evaluated by immunohistochemical staining. The association between ING4 expression in the osteosarcoma and normal bone tissues was analyzed, in addition to the association between ING4 expression and Enneking classification of the osteosarcoma tissues. A significant statistical difference was observed in the ING4 immunohistochemical staining score between the osteosarcoma and normal bone tissues (P<0.001). Furthermore, a significant negative correlation was detected between the ING4 immunohistochemical staining scores and the Enneking classification results of the 41 osteosarcoma tissues (P=0.002). Low expression of ING4 was observed in the osteosarcoma specimens, and this reduced expression of ING4 was negatively correlated with Enneking classification. Thus, the results of the present study indicate that ING4 may serve as a promising prognostic marker in osteosarcoma.
Collapse
Affiliation(s)
- Dahang Zhao
- The Third Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xiangjie Liu
- The Third Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yunge Zhang
- The Third Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Zhaoming Ding
- The Third Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Feng Dong
- The Third Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Hongwei Xu
- The Third Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Baoxin Wang
- The Third Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Wenbo Wang
- The Third Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
11
|
Yuan S, Jin J, Shi J, Hou Y. Inhibitor of growth-4 is a potential target for cancer therapy. Tumour Biol 2016; 37:4275-9. [PMID: 26803518 DOI: 10.1007/s13277-016-4842-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 01/12/2016] [Indexed: 12/11/2022] Open
Abstract
The inhibitor of growth-4 (ING-4) belongs to the inhibitor of growth (ING) family that is a type II tumor suppressor gene including five members (ING1-5). As a tumor suppressor, ING4 inhibits tumor growth, invasion, and metastasis by multiple signaling pathways. In addition to that, ING4 can facilitate cancer cell sensitivity to chemotherapy and radiotherapy. Although ING4 loss is observed for many types of cancers, increasing evidences show that ING4 can be used for gene therapy. In this review, the recent progress of ING4 regulating tumorigenesis is discussed.
Collapse
Affiliation(s)
- Shuping Yuan
- Department of Oncology, Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, 212017, People's Republic of China.,Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, People's Republic of China
| | - Jianhua Jin
- Department of Oncology, Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, 212017, People's Republic of China
| | - Juanjuan Shi
- Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, People's Republic of China.
| | - Yongzhong Hou
- Department of Oncology, Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, 212017, People's Republic of China. .,Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, People's Republic of China.
| |
Collapse
|
12
|
Wan J, Zhang X, Liu T, Zhang X. Strategies and developments of immunotherapies in osteosarcoma. Oncol Lett 2015; 11:511-520. [PMID: 26834853 DOI: 10.3892/ol.2015.3962] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 10/27/2015] [Indexed: 12/14/2022] Open
Abstract
Osteosarcoma (OS) is a frequently observed primary malignant tumor. Current therapy for osteosarcoma consists of comprehensive treatment. The long-term survival rate of patients exhibiting nonmetastatic OS varies between 65-70%. However, a number of OS cases have been observed to be resistant to currently used therapies, leading to disease recurrence and lung metastases, which are the primary reasons leading to patient mortality. In the present review, a number of pieces of evidence provide support for the potential uses of immunotherapy, including immunomodulation and vaccine therapy, for the eradication of tumors via upregulation of the immune response. Adoptive T-cell therapy and oncolytic virotherapy have been used to treat OS and resulted in objective responses. Immunologic checkpoint blockade and targeted therapy are also potentially promising therapeutic tools. Immunotherapy demonstrates significant promise with regard to improving the outcomes for patients exhibiting OS.
Collapse
Affiliation(s)
- Jia Wan
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Xianghong Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Tang Liu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Xiangsheng Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
13
|
Wang Y, Wang T, Han Y, Wu H, Zhao W, Tong D, Wei L, Zhong Z, An R, Wang Y. Reduced ING4 Expression Is Associated with the Malignancy of Human Bladder. Urol Int 2015; 94:464-71. [DOI: 10.1159/000364832] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 05/25/2014] [Indexed: 11/19/2022]
Abstract
Introduction: Inhibitor of growth 4 (ING4) is a tumor suppressor. However the role of ING4 in human bladder malignancy is unknown. In this study, ING4 expression in human bladder cancer and its potential effects were studied. Materials and Methods: ING4 expression in 47 human bladder cancer tissues and paired adjacent normal tissues was detected by Western blotting, quantitative reverse transcription-polymerase chain reaction, and immunohistochemistry. The migration and cell cycle progression of SV-HUC-1 and T24 cells with aberrant ING4 expression were examined. Results: ING4 protein and mRNA were significantly decreased in bladder cancer tissues. ING4 protein level was significantly lower in the group of patients over 50 years of age. ING4 knockdown caused more rapid cell migration and increased the population of SV-HUC-1 and T24 cells in the G2-M phase. Conclusion: Our data suggest a close connection between aberrant ING4 expression and the carcinogenesis of human bladder cells. ING4 may be a potential target for bladder cancer chemotherapy.
Collapse
|
14
|
Guérillon C, Bigot N, Pedeux R. The ING tumor suppressor genes: Status in human tumors. Cancer Lett 2014; 345:1-16. [DOI: 10.1016/j.canlet.2013.11.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 11/27/2013] [Accepted: 11/29/2013] [Indexed: 12/18/2022]
|
15
|
Keep-ING balance: tumor suppression by epigenetic regulation. FEBS Lett 2014; 588:2728-42. [PMID: 24632289 DOI: 10.1016/j.febslet.2014.03.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 03/06/2014] [Indexed: 12/26/2022]
Abstract
Cancer cells accumulate genetic and epigenetic changes that alter gene expression to drive tumorigenesis. Epigenetic silencing of tumor suppressor, cell cycle, differentiation and DNA repair genes contributes to neoplastic transformation. The ING (inhibitor of growth) proteins (ING1-ING5) have emerged as a versatile family of growth regulators, phospholipid effectors, histone mark sensors and core components of HDAC1/2 - and several HAT chromatin-modifying complexes. This review will describe the characteristic pathways by which ING family proteins differentially affect the Hallmarks of Cancer and highlight the various epigenetic mechanisms by which they regulate gene expression. Finally, we will discuss their potentials as biomarkers and therapeutic targets in epigenetic treatment strategies.
Collapse
|
16
|
黄 锦, 杨 吉, 凌 春, 赵 大, 谢 宇, 由 振. [The mechanism of inhibition effect of adenovirus-mediated ING4 on human lung adenocarcinoma xenografts in nude mice]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2014; 17:142-7. [PMID: 24581166 PMCID: PMC6000057 DOI: 10.3779/j.issn.1009-3419.2014.02.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Revised: 11/05/2013] [Indexed: 11/05/2022]
Abstract
BACKGROUND AND OBJECTIVE The inhibitor of growth 4 (ING4) is an important tumor suppressive gene.It has been proven that ING4 could inhibite the proliferation of many tumors. e aim of this study is to investigate the inhibitory effect and anti-cancer mechanism of adenovirus-mediated ING4 gene on SPC-A1 human lung adenocarcinoma in nude mice. METHODS A human lung adenocarcinoma xenograft model was established with SPC-A1 cells in nude mice. A total of 15 tumor-bearing nude mice were randomly divided into three groups, namely, PBS, Ad-GFP, and Ad-ING4. e mice in the three groups were intratumorally injected every other day. Their tumor volumes were continually recorded. The treatment tumors were then removed from the mice and weighed. Tumor inhibition rates were calculated. Cell apoptosis was examined by TUNEL method. Caspase-3, COX-2, Fas, and FasL expressions were investigated by immunohistochemistry SP assay. RESULTS Both tumor weight and volume in the Ad-ING4 group were significantly decreased. The tumor inhibition rate of the mice in the Ad-ING4 group (33.17% ± 5.24%) was statistically different from that of the mice in the Ad-GFP group (1.31% ± 0.31%; P<0.05). The apoptotic index of the mice in the Ad-ING4 group (69.23% ± 6.53%) was also significantly different from those in PBS (17.04% ± 1.10%) and Ad-GFP groups (18.81% ± 1.93%; P<0.05). Based on immunohistochemistry SP assay, the results showed that Ad-ING4 may not only upregulate the expressions of caspase-3, Fas, and FasL but also downregulate the expression of COX-2. CONCLUSION ING4 gene elicited a remarkable growth inhibitory e-ect on human lung adenocarcinoma xenografts in nude mice. e mechanism is possibly related to an increase in tumor cell apoptosis.
Collapse
Affiliation(s)
- 锦宏 黄
- 215500 常熟,常熟市第二人民医院呼吸内科Department of Respiratory, Second People's Hospital of Changshu, Changshu 215500, China
| | - 吉成 杨
- 215123 苏州,苏州大学医学部基础医学与生物科学学院细胞与分子生物学教研室Department of Cell and Molecular Biology, College of Medicine, Soochow University, Suzhou 215123, China
| | - 春华 凌
- 215006 苏州,苏州大学附属第一医院呼吸内科Department of Respiratory Medicine, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - 大国 赵
- 215006 苏州,苏州大学附属第一医院呼吸内科Department of Respiratory Medicine, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - 宇锋 谢
- 215123 苏州,苏州大学医学部基础医学与生物科学学院细胞与分子生物学教研室Department of Cell and Molecular Biology, College of Medicine, Soochow University, Suzhou 215123, China
| | - 振华 由
- 215500 常熟,常熟市第二人民医院呼吸内科Department of Respiratory, Second People's Hospital of Changshu, Changshu 215500, China
| |
Collapse
|
17
|
RegulatING chromatin regulators: post-translational modification of the ING family of epigenetic regulators. Biochem J 2013; 450:433-42. [DOI: 10.1042/bj20121632] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The five human ING genes encode at least 15 splicing isoforms, most of which affect cell growth, differentiation and apoptosis through their ability to alter gene expression by epigenetic mechanisms. Since their discovery in 1996, ING proteins have been classified as type II tumour suppressors on the basis of reports describing their down-regulation and mislocalization in a variety of cancer types. In addition to their regulation by transcriptional mechanisms, understanding the range of PTMs (post-translational modifications) of INGs is important in understanding how ING functions are fine-tuned in the physiological setting and how they add to the repertoire of activities affected by the INGs. In the present paper we review the different PTMs that have been reported to occur on INGs. We discuss the PTMs that modulate ING function under normal conditions and in response to a variety of stresses. We also describe the ING PTMs that have been identified by several unbiased MS-based PTM enrichment techniques and subsequent proteomic analysis. Among the ING PTMs identified to date, a subset has been characterized for their biological significance and have been shown to affect processes including subcellular localization, interaction with enzymatic complexes and ING protein half-life. The present review aims to highlight the emerging role of PTMs in regulating ING function and to suggest additional pathways and functions where PTMs may effect ING function.
Collapse
|
18
|
Radiosensitivity by ING4–IL-24 bicistronic adenovirus-mediated gene cotransfer on human breast cancer cells. Cancer Gene Ther 2012; 20:38-45. [DOI: 10.1038/cgt.2012.82] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
19
|
Zhang L, Wang Y, Zhang F, Wang Y, Zhang Q. Correlation between tumor suppressor inhibitor of growth family member 4 expression and microvessel density in breast cancer. Hum Pathol 2012; 43:1611-7. [DOI: 10.1016/j.humpath.2011.11.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 11/17/2011] [Accepted: 11/18/2011] [Indexed: 10/28/2022]
|
20
|
ING4 is negatively correlated with microvessel density in colon cancer. Tumour Biol 2012; 33:2357-64. [PMID: 23055189 DOI: 10.1007/s13277-012-0498-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 08/16/2012] [Indexed: 10/27/2022] Open
Abstract
ING4 is a novel tumor suppressor which is downregulated in a number of cancers. In this study, we investigated the role of ING4 in tumor angiogenesis in colorectal carcinoma (CRC) patients. Semi-quantitative RT-PCR, western blots, and immunohistochemistry were used to determine ING4 mRNA and protein expression in CRC and normal tissue from 60 CRC specimens and 30 colonic adenoma specimens. The correlation between ING4 expression and clinical stage, histological grade as well as lymph node metastasis was evaluated. Immunohistochemistry was performed to explore the correlation between ING4 expression and microvessel density (MVD) in CRC. CRC tissue had significantly lower levels of ING4 mRNA and protein compared to colonic adenoma and normal intestinal tissue. Immunostaining showed ING4 expression in 38 (63.3 %), 30 (100 %), and 60 (100 %) cases of normal colonic mucosa, adenoma, and normal intestinal mucosal tissue, respectively. Lower ING4 levels correlated with higher clinical stage and histological grade. ING4 mRNA and protein levels were significantly lower in CRC patients with lymph node metastasis compared to patients without lymph node metastasis (0.41 ± 0.30 vs. 0.91 ± 0.29 and 0.60 ± 0.21 vs. 0.87 ± 0.27, respectively; p < 0.001). Importantly, ING4 mRNA and protein levels were negatively correlated with MVD in CRC patients (p < 0.001). Our data suggest that ING4 levels are a potential biomarker of CRC progression and that ING4 may inhibit tumor growth by modulating angiogenesis in CRC.
Collapse
|
21
|
Zhao Y, Li Z, Sheng W, Miao J, Yang J. Adenovirus-mediated ING4/IL-24 double tumor suppressor gene co-transfer enhances antitumor activity in human breast cancer cells. Oncol Rep 2012; 28:1315-24. [PMID: 22842937 DOI: 10.3892/or.2012.1930] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 07/04/2012] [Indexed: 11/06/2022] Open
Abstract
Cancer gene therapy represents a new and promising therapeutic modality for various types of cancer. Two or more anti-oncogenes carried by a single vector could theoretically improve treatment efficacy, reduce side-effects from vectors, and have a satisfactory clinical application prospect; however, this has seldom been studied in breast cancer. The inhibitor of growth 4 (ING4), as a member of the inhibitor of growth tumor suppressor family has potent inhibitory effects on a variety of tumors. Interleukin‑24 (IL-24) has also shown broad spectrum and tumor-specific antitumor activities. In this study, we aimed to prove the enhanced antitumor activity of adenovirus-mediated ING4/IL-24 double tumor suppressor gene co-transfer in human breast cancer cells. We assessed the combined effect of the ING4/IL-24 bicistronic adenovirus (Ad-ING4-IL-24) in vitro and in vivo on MDA-MB-231 human breast cancer cells by detecting and comparing the apoptotic status in the bicistronic anti-oncogene group (Ad-ING4-IL-24) and in the ING4 or IL-24 single anti-oncogene groups, and also investigated the possible underlying mechanism. Our results showed that the bicistronic adenovirus-mediated ING4 and IL-24 co-expression induced additive growth suppression and apoptosis as well as an overlapping effect on the upregulation of p21, p27 and Bax, and the downregulation of Bcl-2 and survivin in MDA-MB‑231 human breast cancer cells in vitro or in vivo. Moreover, Ad-ING4-IL-24 treatment additively reduced CD34 expression and the microvessel density in MDA-MB-231 xenografted tumors in athymic nude mice, which correlated with the decreased expression of the vascular endothelial growth factor. The enhanced antitumor activity on breast cancer elicited by Ad-ING4-IL-24 was closely associated with the activation of the apoptotic pathways and the additive inhibition of tumor angiogenesis.
Collapse
Affiliation(s)
- Yaodong Zhao
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, PR China
| | | | | | | | | |
Collapse
|
22
|
Expression of inhibitor of growth 4 by HSV1716 improves oncolytic potency and enhances efficacy. Cancer Gene Ther 2012; 19:499-507. [PMID: 22595793 DOI: 10.1038/cgt.2012.24] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
We have isolated and characterized a novel variant of the replication-competent oncolytic HSV1716 that expresses inhibitor of growth 4 (Ing4) (HSV1716Ing4). We demonstrate that Ing4 expression enhances progeny output during HSV1716 infection of human tumor cells both in vitro and in vivo, thereby significantly augmenting its oncolytic potency. In tissue culture, compared with HSV1716, HSV1716Ing4 produced significantly higher numbers of infectious progeny in human squamous cell carcinoma (SCC), breast, ovarian, prostate and colorectal cancer cell lines. Immediate-early expression of Ing4 was crucial for this effect and an intact Ing4 was required as there was no enhanced progeny production with HSV1716 variants that expressed Ing4 mutants lacking the C-terminal plant homeodomain domain or conserved nuclear localization signals. In mouse xenograft models of SCC, ovarian and breast cancer, HSV1716Ing4 was significantly more efficacious than HSV1716 with at least 1000-fold more infectious virus found in tumors after HSV1716Ing4 treatment compared with tumors from HSV1716 treatment. Using a sensitive herpes simplex virus type 1 (HSV-1) PCR, virus DNA was only detected in tumors and was not detected in the DNA extracted from any organs of the injected mice demonstrating that, like HSV1716, HSV1716Ing4 replication is exclusively restricted to tumor cells. Our results suggest that the potential for enhanced tumor destruction by oncolytic HSV expressing Ing4 merits clinical investigation.
Collapse
|
23
|
Synergistic antitumor effect of adenovirus-mediated hING4 gene therapy and 125I radiation therapy on pancreatic cancer. Cancer Lett 2012; 316:211-8. [DOI: 10.1016/j.canlet.2011.11.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 10/31/2011] [Accepted: 11/01/2011] [Indexed: 02/05/2023]
|
24
|
Xie Y, Lv H, Sheng W, Miao J, Xiang J, Yang J. Synergistic Tumor Suppression by Adenovirus-Mediated Inhibitor of Growth 4 and Interleukin-24 Gene Cotransfer in Hepatocarcinoma Cells. Cancer Biother Radiopharm 2011; 26:681-95. [DOI: 10.1089/cbr.2011.1047] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Yufeng Xie
- Cell and Molecular Biology Institute, College of Medicine, Soochow University, Suzhou, China
- Department of Oncology and Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Haitao Lv
- Department of Internal Medicine, Children's Hospital Affiliated to Soochow University, Suzhou, China
| | - Weihua Sheng
- Cell and Molecular Biology Institute, College of Medicine, Soochow University, Suzhou, China
| | - Jingcheng Miao
- Cell and Molecular Biology Institute, College of Medicine, Soochow University, Suzhou, China
| | - Jim Xiang
- Department of Oncology and Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Jicheng Yang
- Cell and Molecular Biology Institute, College of Medicine, Soochow University, Suzhou, China
| |
Collapse
|
25
|
Wang L, Sun L, Huang J, Jiang M. Cyclin-dependent kinase inhibitor 3 (CDKN3) novel cell cycle computational network between human non-malignancy associated hepatitis/cirrhosis and hepatocellular carcinoma (HCC) transformation. Cell Prolif 2011; 44:291-9. [PMID: 21535270 DOI: 10.1111/j.1365-2184.2011.00752.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The relationship of cyclin-dependent kinase inhibitor 3 (CDKN3) with tumours has previously been presented in a number of publications. However, the molecular network and interpretation of CDKN3 through the cell cycle between non-malignancy associated hepatitis/cirrhosis and hepatocellular carcinoma (HCC) have remained to be elucidated. Here, we have constructed and analysed significant high expression gene CDKN3 activated and inhibited cell cycle networks from 25 HCC versus 25 non-malignancy associated hepatitis/cirrhosis patients (viral infection HCV or HBV) in GEO Dataset GSE10140-10141, by combination of a gene regulatory network inference method based on linear programming, and decomposition procedure using CapitalBio MAS 3.0 software, based on integration of public databases including Gene Ontology, KEGG, BioCarta, GenMapp, Intact, UniGene, OMIM, and others. Comparing the same and differently activated and inhibited CDKN3 networks with GO analysis, between non-malignancy associated hepatitis/cirrhosis and HCC, our results suggest a CDKN3 cell cycle network (i) with stronger DNA replication and with weaker ubiquitin-dependent protein catabolism as common characteristics in both non-malignancy associated hepatitis/cirrhosis and HCC; (ii) with more cell division and weaker mitotic G2 checkpoint in non-malignancy associated hepatitis/cirrhosis; (iii) with stronger cell cycle and weaker cytokinesis, as a result forming multinucleate cells in HCC. Thus, it is useful to identify CDKN3 cell cycle networks for comprehension of molecular mechanism between non-malignancy associated hepatitis/cirrhosis and HCC transformation.
Collapse
Affiliation(s)
- L Wang
- Biomedical Center, School of Electronic Engineering, Beijing University of Posts and Telecommunications, Beijing, China.
| | | | | | | |
Collapse
|
26
|
Enhanced tumor suppression by an ING4/IL-24 bicistronic adenovirus-mediated gene cotransfer in human non-small cell lung cancer cells. Cancer Gene Ther 2011; 18:627-36. [PMID: 21660060 DOI: 10.1038/cgt.2011.31] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
ING4 as a member of inhibitor of growth (ING) tumor suppressor family has potent inhibitory effects on a variety of tumors. Interleukin-24 (IL-24), a cytokine-tumor suppressor, also shows broad-spectrum and tumor-specific antitumor activities. In this report, we constructed an ING4/IL-24 bicistronic adenovirus (Ad-ING4-IL-24) and assessed its combined effect on in vitro and in vivo A549 human non-small cell lung cancer cells. We demonstrated that ING4 and IL-24 combination treatment by adenovirus-mediated ING4 and IL-24 coexpression induced additive growth suppression and apoptosis as well as an overlapping effect on upregulation of P21, P27, Fas, Bax and cleaved Caspases-8, 9, 3 and downregulation of Bcl-2 in in vitro A549 lung carcinoma cells. Moreover, Ad-ING4-IL-24 treatment additively inhibited in vivo A549 lung carcinoma subcutaneous (s.c.) xenografted tumor growth and reduced CD34 and microvessel density in A549 xenografted tumors in athymic nude mice. The enhanced antitumor activity elicited by Ad-ING4-IL-24 was closely associated with the coordinate activation of extrinsic and intrinsic apoptotic pathways and additive inhibition of tumor angiogenesis. Thus, our results indicate that cancer gene therapy combining two or more tumor suppressors such as ING4 and IL-24 may constitute a novel and effective therapeutic strategy for lung carcinoma and other cancers.
Collapse
|
27
|
Fillat C, Jose A, Bofill-Deros X, Mato-Berciano A, Maliandi MV, Sobrevals L. Pancreatic cancer gene therapy: from molecular targets to delivery systems. Cancers (Basel) 2011; 3:368-95. [PMID: 24212620 PMCID: PMC3756366 DOI: 10.3390/cancers3010368] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 01/05/2011] [Accepted: 01/13/2011] [Indexed: 02/08/2023] Open
Abstract
The continuous identification of molecular changes deregulating critical pathways in pancreatic tumor cells provides us with a large number of novel candidates to engineer gene-targeted approaches for pancreatic cancer treatment. Targets—both protein coding and non-coding—are being exploited in gene therapy to influence the deregulated pathways to facilitate cytotoxicity, enhance the immune response or sensitize to current treatments. Delivery vehicles based on viral or non-viral systems as well as cellular vectors with tumor homing characteristics are a critical part of the design of gene therapy strategies. The different behavior of tumoral versus non-tumoral cells inspires vector engineering with the generation of tumor selective products that can prevent potential toxic-associated effects. In the current review, a detailed analysis of the different targets, the delivery vectors, the preclinical approaches and a descriptive update on the conducted clinical trials are presented. Moreover, future possibilities in pancreatic cancer treatment by gene therapy strategies are discussed.
Collapse
Affiliation(s)
- Cristina Fillat
- Programa Gens i Malaltia, Centre de Regulació Genòmica-CRG, UPF, Parc de Recerca Biomèdica de Barcelona-PRBB and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain.
| | | | | | | | | | | |
Collapse
|
28
|
Enhanced antitumor activity by combining an adenovirus harboring ING4 with cisplatin for hepatocarcinoma cells. Cancer Gene Ther 2010; 18:176-88. [PMID: 21052098 PMCID: PMC3047817 DOI: 10.1038/cgt.2010.67] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The inhibitor of growth (ING) family proteins have been defined as candidate tumor suppressors. ING4 as a novel member of the ING family has potential tumor-suppressive effects. In this study, we explored the combined effect of adenovirus-mediated ING4 (Ad-ING4) gene transfer plus chemotherapy drug cisplatin (CDDP) on SMMC-7721 human hepatocarcinoma cells in vitro and in vivo, and its underlying mechanism. We demonstrated that Ad-ING4 plus CDDP induced synergistic growth inhibition, enhanced apoptosis, and had an additive effect on upregulation of Fas, Bax, Bak, cleaved Bid, cleaved caspase-8, caspase-9, caspase-3 and cleaved PARP, and on downregulation of Bcl-2 and Bcl-XL in SMMC-7721 hepatocarcinoma cells. Moreover, Ad-ING4 plus CDDP synergistically suppressed in vivo SMMC-7721 hepatocarcinoma subcutaneous (s.c.) xenografted tumor growth and reduced tumor vessel CD34 expression and microvessel density (MVD) in athymic nude mice. Most importantly, Ad-ING4 plus CDDP did not have overlapping toxicities in HL-7702 normal human liver cells and normal liver tissues of mice. The in vitro and in vivo enhanced antitumor effect elicited by Ad-ING4 plus CDDP was closely associated with the cooperative regulation of extrinsic and intrinsic apoptotic pathways and synergistic inhibition of tumor angiogenesis. Thus, our results indicate that Ad-ING4 plus CDDP is a potential combined treatment strategy for hepatocarcinoma.
Collapse
|
29
|
ING Genes Work as Tumor Suppressor Genes in the Carcinogenesis of Head and Neck Squamous Cell Carcinoma. JOURNAL OF ONCOLOGY 2010; 2011:963614. [PMID: 21052543 PMCID: PMC2968421 DOI: 10.1155/2011/963614] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2010] [Accepted: 10/01/2010] [Indexed: 12/24/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer in the world. The evolution and progression of HNSCC are considered to result from multiple stepwise alterations of cellular and molecular pathways in squamous epithelium. Recently, inhibitor of growth gene (ING) family consisting of five genes, ING1 to ING5, was identified as a new tumor suppressor gene family that was implicated in the downregulation of cell cycle and chromatin remodeling. In contrast, it has been shown that ING1 and ING2 play an oncogenic role in some cancers, this situation being similar to TGF-β. In HNSCC, the ING family has been reported to be downregulated, and ING translocation from the nucleus to the cytoplasm may be a critical event for carcinogenesis. In this paper, we describe our recent results and briefly summarize current knowledge regarding the biologic functions of ING in HNSCC.
Collapse
|
30
|
Zha L, Qiao T, Yuan S, Lei L. Enhancement of radiosensitivity by CpG-oligodeoxyribonucleotide-7909 in human non-small cell lung cancer A549 cells. Cancer Biother Radiopharm 2010; 25:165-70. [PMID: 20423229 DOI: 10.1089/cbr.2009.0686] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
CpG-oligodeoxyribonucleotides (CpG-ODNs), which induce signaling through the toll-like receptor 9, are currently under investigation as immunity stimulators against cancer. It has recently been suggested that CpG-ODNs may also enhance sensitivity to traditional therapies including chemotherapy in certain cancer-cell lines. The purpose of this study was to define the activity of CpG-ODN7909 in increasing radiosensitivity of the human non-small cell lung cancer cell line A549 in vitro. First, a dose- and time-dependent inhibitory effect on cell viability was observed after A549 cells were treated with different concentrations of CpG-ODN7909 (5, 10, 30, and 60 microg/mL). Second, decreased cell clonogenic survival, enhanced cell apoptotic index, accumulated percentage of cells in the G2/M phase, and increased tumor necrosis factor (TNF)-alpha secretion were found after combined treatments with 10 microg/mL of CpG-ODN7909 and radiation compared to either treatment alone (p < 0.05). Furthermore, the toll-like receptor 9 mRNA was found to express in A549. The results suggest that CpG-ODN7909 can increase the radiosensitivity of human non-small cell lung cancer A549 cells, which may be associated with reduced cell clonogenic survival, enhanced apoptosis, prolonged cell-cycle arrest in G2/M, and stimulation of TNF-alpha secretion.
Collapse
Affiliation(s)
- Lin Zha
- Department of Radiotherapy Oncology, Jinshan Hospital, Medical Center of Fudan University, Shanghai, People's Republic of China
| | | | | | | |
Collapse
|
31
|
Li Z, Xie Y, Sheng W, Miao J, Xiang J, Yang J. Tumor-Suppressive Effect of Adenovirus-Mediated Inhibitor of Growth 4 Gene Transfer in Breast Carcinoma Cells In Vitro and In Vivo. Cancer Biother Radiopharm 2010; 25:427-37. [DOI: 10.1089/cbr.2010.0778] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Affiliation(s)
- Zhengyi Li
- Cell and Molecular Biology Institute, College of Medicine, Soochow University, Suzhou, China
| | - Yufeng Xie
- Cell and Molecular Biology Institute, College of Medicine, Soochow University, Suzhou, China
- Department of Oncology and Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Weihua Sheng
- Cell and Molecular Biology Institute, College of Medicine, Soochow University, Suzhou, China
| | - Jingcheng Miao
- Cell and Molecular Biology Institute, College of Medicine, Soochow University, Suzhou, China
| | - Jim Xiang
- Department of Oncology and Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Jicheng Yang
- Cell and Molecular Biology Institute, College of Medicine, Soochow University, Suzhou, China
| |
Collapse
|