1
|
Xiong JX, Li YT, Tan XY, Chen T, Liu BH, Fu L. Targeting PRSS23 with tipranavir induces gastric cancer stem cell apoptosis and inhibits growth of gastric cancer via the MKK3/p38 MAPK-IL24 pathway. Acta Pharmacol Sin 2024; 45:405-421. [PMID: 37814123 PMCID: PMC10789761 DOI: 10.1038/s41401-023-01165-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/04/2023] [Indexed: 10/11/2023]
Abstract
Gastric cancer stem cells (GCSCs) contribute to the refractory features of gastric cancer (GC) and are responsible for metastasis, relapse, and drug resistance. The key factors drive GCSC function and affect the clinical outcome of GC patients remain poorly understood. PRSS23 is a novel serine protease that is significantly up-regulated in several types of cancers and cancer stem cells, and related to tumor progression and drug resistance. In this study, we investigated the role of PRSS23 in GCSCs as well as the mechanism by which PRSS23 regulated the GCSC functions. We demonstrated that PRSS23 was critical for sustaining GCSC survival. By screening a collection of human immunodeficiency virus (HIV) protease inhibitors (PIs), we identified tipranavir as a PRSS23-targeting drug, which effectively killed both GCSC and GC cell lines (its IC50 values were 4.7 and 6.4 μM in GCSC1 cells and GCSC2 cells, respectively). Administration of tipranavir (25 mg·kg-1·d-1, i.p., for 8 days) in GCSC-derived xenograft mice markedly inhibited the growth of subcutaneous GCSC tumors without apparent toxicity. In contrast, combined treatment with 5-FU plus cisplatin did not affect the tumor growth but causing significant weight loss. Furthermore, we revealed that tipranavir induced GCSC cell apoptosis by suppressing PRSS23 expression, releasing MKK3 from the PRSS23/MKK3 complex to activate p38 MAPK, and thereby activating the IL24-mediated Bax/Bak mitochondrial apoptotic pathway. In addition, tipranavir was found to kill other types of cancer cell lines and drug-resistant cell lines. Collectively, this study demonstrates that by targeting both GCSCs and GC cells, tipranavir is a promising anti-cancer drug, and the clinical development of tipranavir or other drugs specifically targeting the PRSS23/MKK3/p38MAPK-IL24 mitochondrial apoptotic pathway may offer an effective approach to combat gastric and other cancers.
Collapse
Affiliation(s)
- Ji-Xian Xiong
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China.
| | - Yu-Ting Li
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Xiang-Yu Tan
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Tie Chen
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Bao-Hua Liu
- Department of Biochemistry, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Li Fu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China.
| |
Collapse
|
2
|
Abstract
Previous studies have shown that interleukin-24 (IL-24) has tumor-suppressing activity by multiple pathways. However, the immunogenicity moderation effect of IL-24 on malignant cells has not been explored extensively. In this study, we investigated the role of IL-24 in immunogenicity modulation of the myelogenous leukemia cells. Data show that myelogenous leukemia cells express low levels of immunogenicity molecules. Treatment with IL-24 could enhance leukemia cell immunogenicity, predominantly regulate leukemia cells to produce immune-associated cytokines, and improve the cytotoxic sensitivity of these cells to immune effector cells. IL-24 expression could retard transplanted leukemia cell tumor growth in vivo in athymic nude mice. Moreover, IL-24 had marked effects on downregulating the expression of angiogenesis-related proteins vascular endothelial growth factor, cluster of differentiation (CD) 31, CD34, collagen IV and metastasis-related factors CD147, membrane type-1 matrix metalloproteinase (MMP), and MMP-2 and MMP-9 in transplanted tumors. These findings indicated novel functions of this antitumor gene and characterized IL-24 as a promising agent for further clinical trial for hematologic malignancy immunotherapy.
Collapse
|
3
|
Valiyari S, Salami M, Mahdian R, Shokrgozar MA, Oloomi M, Mohammadi Farsani A, Bouzari S. sIL-24 peptide, a human interleukin-24 isoform, induces mitochondrial-mediated apoptosis in human cancer cells. Cancer Chemother Pharmacol 2017; 80:451-459. [DOI: 10.1007/s00280-017-3370-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/20/2017] [Indexed: 12/12/2022]
|
4
|
Li YJ, Liu G, Xia L, Xiao X, Liu JC, Menezes ME, Das SK, Emdad L, Sarkar D, Fisher PB, Archer MC, Zacksenhaus E, Ben-David Y. Suppression of Her2/Neu mammary tumor development in mda-7/IL-24 transgenic mice. Oncotarget 2016; 6:36943-54. [PMID: 26460950 PMCID: PMC4741907 DOI: 10.18632/oncotarget.6046] [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: 07/06/2015] [Accepted: 09/23/2015] [Indexed: 12/21/2022] Open
Abstract
Melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24) encodes a tumor suppressor gene implicated in the growth of various tumor types including breast cancer. We previously demonstrated that recombinant adenovirus-mediated mda-7/IL-24 expression in the mammary glands of carcinogen-treated (methylnitrosourea, MNU) rats suppressed mammary tumor development. Since most MNU-induced tumors in rats contain activating mutations in Ha-ras, which arenot frequently detected in humans, we presently examined the effect of MDA-7/IL-24 on Her2/Neu-induced mammary tumors, in which the RAS pathway is induced. We generated tet-inducible MDA-7/IL-24 transgenic mice and crossed them with Her2/Neu transgenic mice. Triple compound transgenic mice treated with doxycycline exhibited a strong inhibition of tumor development, demonstrating tumor suppressor activity by MDA-7/IL-24 in immune-competent mice. MDA-7/IL-24 induction also inhibited growth of tumors generated following injection of Her2/Neu tumor cells isolated from triple compound transgenic mice that had not been treated with doxycycline, into the mammary fat pads of isogenic FVB mice. Despite initial growth suppression, tumors in triple compound transgenic mice lost mda-7/IL-24 expression and grew, albeit after longer latency, indicating that continuous presence of this cytokine within tumor microenvironment is crucial to sustain tumor inhibitory activity. Mechanistically, MDA-7/IL-24 exerted its tumor suppression effect on HER2+ breast cancer cells, at least in part, through PERP, a member of PMP-22 family with growth arrest and apoptosis-inducing capacity. Overall, our results establish mda-7/IL-24 as a suppressor of mammary tumor development and provide a rationale for using this cytokine in the prevention/treatment of human breast cancer.
Collapse
Affiliation(s)
- You-Jun Li
- Department of Anatomy, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin, China
| | - Guodong Liu
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Lei Xia
- Division of Biology, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, China
| | - Xiao Xiao
- Division of Biology, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, China
| | - Jeff C Liu
- Toronto General Research Institute - University Health Network, Toronto, Ontario, Canada
| | - Mitchell E Menezes
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Swadesh K Das
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Luni Emdad
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Paul B Fisher
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Michael C Archer
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Eldad Zacksenhaus
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Toronto General Research Institute - University Health Network, Toronto, Ontario, Canada
| | - Yaacov Ben-David
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Division of Biology, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, China
| |
Collapse
|
5
|
Cao H, Xiang T, Zhang C, Yang H, Jiang L, Liu S, Huang X. MDA7 combined with targeted attenuated Salmonella vector SL7207/pBud-VP3 inhibited growth of gastric cancer cells. Biomed Pharmacother 2016; 83:809-815. [PMID: 27497809 DOI: 10.1016/j.biopha.2016.07.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 07/07/2016] [Accepted: 07/13/2016] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND/AIM To investigate the therapeutic effect of MDA7 combined with apoptin targeted attenuated Salmonella typhimurium vector SL7207/pBud-VP3 on gastric cancer cells. MATERIALS AND METHODS MDA7 was inserted into pBud-VP3 using molecular cloning technology to obtain the eukaryotic expression plasmid pBud-VP3-MDA7 and it was transformed into attenuated Salmonella typhimurium SL7207 by high voltage electroporation to obtain SL7207/pBud-VP3-MDA7. Mice bearing a sarcoma of gastric cancer cells were treated with SL7207/pBud-VP3-MDA7 and the growth-suppressing effect was assessed by measurement of tumor volume. Western blot was used to identify the MDA7 expression products. IL-6, INF-γ, TNF-α and caspase-3, VEGF in tumor tissue were detected by RT-PCR and immunohistochemistry. RESULTS SL7207/pBud-VP3-MDA7 was successfully constructed and expression of the protein MDA7 was identified in tumor tissue. SL7207/pBud-VP3-MDA7 significantly caused tumor inhibition and regression (p<0.05). The level of expression of cytokines IL-6, INF-γ, TNF-α in tumor tissue was significantly higher than in the other groups (p<0.05). The expression of caspase-3 was up-regulated and VEGF was down-regulated (p<0.05). CONCLUSION This study shows that SL7207/pBud-VP3-MDA7 has inhibitory effect on the growth of gastric cancer cells. The mechanism involved is related to the promotion of tumor apoptosis, immunity regulation and inhibition of tumor blood vessels.
Collapse
Affiliation(s)
- Hongdan Cao
- Chongqing Medical and Pharmaceutical Higher specialty College, Road 82, Shapingba District University City, Chongqing 401331, China
| | - Tingxiu Xiang
- Artron BioResearch Inc., 3938 North Fraser Way, Burnaby, BC V5 J 5H6, Canada
| | - Chaohong Zhang
- Chongqing Medical and Pharmaceutical Higher specialty College, Road 82, Shapingba District University City, Chongqing 401331, China
| | - Hong Yang
- Chongqing Medical and Pharmaceutical Higher specialty College, Road 82, Shapingba District University City, Chongqing 401331, China
| | - Lingqun Jiang
- Chongqing Medical and Pharmaceutical Higher specialty College, Road 82, Shapingba District University City, Chongqing 401331, China
| | - Shanli Liu
- Chongqing Medical and Pharmaceutical Higher specialty College, Road 82, Shapingba District University City, Chongqing 401331, China
| | - Xiaolan Huang
- Ph.D Research Center for Medical and Social Development, Chongqing Medical University, Road 1, Yuzhong District School of Medicine, Chongqing 400016, China.
| |
Collapse
|
6
|
Yu X, Xia W, Li S, Blumenfeld J, Zhang B, Yang J, Miao J, Gu ZJ. Antitumor effect and underlying mechanism of RGD-modified adenovirus mediated IL-24 expression on myeloid leukemia cells. Int Immunopharmacol 2015. [DOI: 10.1016/j.intimp.2015.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
7
|
Wei S, Cao H, Zhou X, Wu H, Yang J. Prokaryotically and eukaryotically expressed interleukin-24 induces breast cancer growth suppression via activation of apoptosis and inhibition of tumor angiogenesis. Mol Med Rep 2014; 11:3673-81. [PMID: 25544477 DOI: 10.3892/mmr.2014.3136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Accepted: 10/24/2014] [Indexed: 11/05/2022] Open
Abstract
Melanoma differentiation‑associated‑7 (mda‑7)/interleukin‑24 (IL‑24), a unique cytokine‑tumor suppressor, exerts tumor‑selective killing activity in numerous types of cancer cell. Although eukaryotically and prokaryotically expressed recombinant human (rh)IL‑24 proteins have been previously shown to produce potent antitumor effects, to the best of our knowledge, no side‑by‑side study has been conducted that compares the two proteins directly. In the present study, rhIL‑24 protein was expressed in BL21 Escherichia coli transformed with the pET‑21a(+)‑hIL‑24 plasmid by isopropyl‑β‑D‑1‑thiogalactopyranoside induction. Following a denaturing and renaturing process, the soluble rhIL‑24 was purified using a Q‑Sepharose column. rhIL‑24 protein was also expressed in Chinese hamster ovary mammalian cells stably transfected with the pcDNA3‑hIL‑24 plasmid. The in vitro antitumor efficacies of the two treatments were compared using the MDA‑MB‑231 human breast cancer cell line. Furthermore, the therapeutic efficacies of the bacteria‑derived rhIL‑24 protein and the liposome‑coated pcDNA3‑hIL‑24 naked plasmid were evaluated in athymic nude mice with subcutaneously xenografted MDA‑MB‑231 cell tumors. The prokaryotically expressed/purified rhIL‑24 protein and the eukaryotically expressed rhIL‑24 in the cell supernate were revealed to be capable of efficiently suppressing MDA‑MB‑231 tumor growth in vitro. Similarly, the administration of bacteria‑derived rhIL‑24 protein and pcDNA3‑hIL‑24 naked plasmid also provided therapeutic benefits in the treatment of in vivo MDA‑MB‑231 xenografted tumors. The retarded in vitro and in vivo breast cancer growth elicited by rhIL‑24 was closely associated with the upregulation of the ratio of anti‑apoptotic B cell lymphoma 2 (Bcl‑2) to pro‑apoptotic Bcl‑2‑associated X protein (Bax), as well as the activation of caspase‑3 followed by marked induction of apoptosis, and the notable inhibition of tumor angiogenesis. Thus, the results of the present study indicate that prokaryotically expressed rhIL‑24 protein may be an alternate and promising antitumor agent in human breast cancer or other types of cancer.
Collapse
Affiliation(s)
- Shaohua Wei
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Hua Cao
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Xiaoyan Zhou
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Haorong Wu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Jicheng Yang
- Department of Cell and Molecular Biology, College of Medicine, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| |
Collapse
|
8
|
Jiang C, Niu J, Li M, Teng Y, Wang H, Zhang Y. Tumor vasculature-targeted recombinant mutated human TNF-α enhanced the antitumor activity of doxorubicin by increasing tumor vessel permeability in mouse xenograft models. PLoS One 2014; 9:e87036. [PMID: 24466321 PMCID: PMC3899378 DOI: 10.1371/journal.pone.0087036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 12/03/2013] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Increasing evidence suggests that, when used in combination, tumor necrosis factor-α (TNF-α) synergizes with traditional chemotherapeutic drugs to exert a heightened antitumor effect. The present study investigated the antitumor efficacy of recombinant mutated human TNF-α specifically targeted to the tumor vasculature (RGD-rmhTNF-α) combined with the chemotherapeutic agent doxorubicin in 2 murine allografted tumor models. METHODS Mice bearing hepatoma or sarcoma allografted tumors were treated with various doses of RGD-rmhTNF-α alone or in combination with doxorubicin (2 mg/kg). We then evaluated tumor growth and tumor vessel permeability as well as intratumoral levels of RGD-rmhTNF-α and doxorubicin. RESULTS RGD-rmhTNF-α treatment enhanced the permeability of the tumor vessels and increased intratumoral doxorubicin levels. In addition, intratumoral RGD-rmhTNF-α levels were significantly higher than that of rmhTNF-α. In both of the tested tumor models, administering RGD-rmhTNF-α in combination with doxorubicin resulted in an enhanced antitumor response compared to either treatment alone. Double-agent combination treatment of doxorubicin with 50,000 IU/kg RGD-rmhTNF-α induced stronger antitumor effects on H22 allografted tumor-bearing mice than the single doxorubicin agent alone. Moreover, doxorubicin with 10,000 IU/kg RGD-rmhTNF-α synergized to inhibit tumor growth in S180 allografted tumor-bearing mice. CONCLUSIONS These results suggest that targeted delivery of low doses of RGD-rmhTNF-α into the tumor vasculature increases the antitumor efficacy of chemotherapeutic drugs.
Collapse
Affiliation(s)
- Changli Jiang
- Clinical Laboratory, Army Center for Molecular Biological Analysis, Kunming General Hospital of Chengdu Military Area, Kunming, Yunnna, China
- The State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Junzhou Niu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Meng Li
- The State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yi Teng
- Clinical Laboratory, Army Center for Molecular Biological Analysis, Kunming General Hospital of Chengdu Military Area, Kunming, Yunnna, China
| | - Huixuan Wang
- Clinical Laboratory, Army Center for Molecular Biological Analysis, Kunming General Hospital of Chengdu Military Area, Kunming, Yunnna, China
- * E-mail: (HW); (YZ)
| | - Yingqi Zhang
- The State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi’an, Shaanxi, China
- * E-mail: (HW); (YZ)
| |
Collapse
|
9
|
Enhancement of expression of survivin promoter-driven CD/TK double suicide genes by the nuclear matrix attachment region in transgenic gastric cancer cells. Gene 2013; 534:177-82. [PMID: 24220851 DOI: 10.1016/j.gene.2013.10.064] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 10/27/2013] [Accepted: 10/29/2013] [Indexed: 01/10/2023]
Abstract
This work aimed to study a novel transgenic expression system of the CD/TK double suicide genes enhanced by the nuclear matrix attachment region (MAR) for gene therapy. The recombinant vector pMS-CD/TK containing the MAR-survivin promoter-CD/TK cassette was developed and transfected into human gastric cancer SGC-7901 cells. Expression of the CD/TK genes was detected by quantitative real-time PCR (qPCR) and Western blot. Cell viability and apoptosis were measured using the methyl thiazolyl tetrazolium (MTT) assay and flow cytometry. When the MAR fragment was inserted into the upstream of the survivin promoter, the qPCR result showed that the expression of the CD/TK genes significantly increased 7.7-fold in the transgenic SGC-7901 cells with plasmid pMS-CD/TK compared with that without MAR. MTT and flow cytometry analyses indicated that treatment with the prodrugs (5-FC+GCV) significantly decreased the cellular survival rate and enhanced the cellular apoptosis in the SGC-7901 cells. The expression of the CD/TK double suicide genes driven by the survivin promoter can be enhanced by the MAR fragment in human gastric cancer cells.
Collapse
|
10
|
Zhuo B, Wang R, Zhang H, Qin H, Yin Y, Shi Y. Interleukin-24 inhibits cell migration and invasion in the neuroblastoma cell line SH-SY5Y. Oncol Rep 2013; 30:2749-54. [PMID: 24084981 DOI: 10.3892/or.2013.2756] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Accepted: 09/05/2013] [Indexed: 11/06/2022] Open
Abstract
Neuroblastomas are common pediatric solid tumors with a variable clinical course; approximately 50% of patients present with metastatic disease at diagnosis. The development of metastatic lesions often causes a fatal outcome. Therefore, the prevention of metastases during the early stage of tumor development is critical for the improvement of the prognosis of neuroblastoma patients. We previously observed the suppression of neuroblastoma growth in response to overexpression of interleukin-24 (IL-24) in vitro and in vivo. IL-24 exerts its tumor-suppressive effects by multiple mechanisms, including the balance of Bcl-2 family proteins toward the pro-apoptotic pathway and the activation of the caspase cascade. In this study, we used adenovirus-mediated IL-24 (Ad-IL24) to examine the effect of the ectopic production of IL-24 on cell migration and invasion in human neuroblastoma cells. We found that IL-24 effectively inhibits SH-SY5Y neuroblastoma cell migration and invasion by changing subcellular localization and cellular levels of β-catenin and regulating the levels of proteins associated with cell migration and invasion. Thus, IL-24 should be considered a therapeutic agent that can inhibit primary neuroblastoma growth and that may prevent metastasis.
Collapse
Affiliation(s)
- Baobiao Zhuo
- Department of Surgery, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221006, P.R. China
| | | | | | | | | | | |
Collapse
|
11
|
Yuan L, Zhao H, Zhang L, Liu X. The efficacy of combination therapy using adeno-associated virus-mediated co-expression of apoptin and interleukin-24 on hepatocellular carcinoma. Tumour Biol 2013; 34:3027-34. [PMID: 23907578 DOI: 10.1007/s13277-013-0867-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 05/14/2013] [Indexed: 12/14/2022] Open
Abstract
Multigene-based combination therapy is an effective practice in cancer gene therapy. Apoptin is a chicken anemia virus-derived, p53-independent, Bcl-2-insensitive apoptotic protein with the ability to specifically induce apoptosis in various human tumor cells. Interleukin-24 (IL-24) displays ubiquitous antitumor property and tumor-specific killing activity. Adeno-associated virus (AAV) is a promising gene delivery vehicle due to its advantage of low pathogenicity and long-term gene expression. In this study, we assessed the efficacy of combination therapy using AAV-mediated co-expression of apoptin and interleukin-24 on hepatocellular carcinoma in vitro and in vivo. Our results showed that AAV-mediated co-expression of IL-24 and apoptin significantly suppressed the growth and induced the apoptosis of HepG2 cells in vitro. Furthermore, AAV-mediated combined treatment of IL-24 and apoptin significantly suppressed tumor growth and induced apoptosis of tumor cells in xenograft nude mice. These data suggest that AAV vectors that co-express apoptin and IL-24 have great potential in cancer gene therapy.
Collapse
Affiliation(s)
- Lijie Yuan
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Daqing Campus, Daqing, 163319, People's Republic of China,
| | | | | | | |
Collapse
|
12
|
Li Y, Zhang H, Zhu X, Feng D, Gong J, Han T. Interleukin-24 induces neuroblastoma SH-SY5Y cell differentiation, growth inhibition, and apoptosis by promoting ROS production. J Interferon Cytokine Res 2013; 33:709-14. [PMID: 23692552 DOI: 10.1089/jir.2013.0004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Neuroblastoma is among the most aggressive tumors that occur in childhood and infancy. The clinical prognosis of children with advanced-stage neuroblastoma is still poor. Interleukin-24 (IL-24) is emerging as a new cytokine involved in tumor cellular proliferation, differentiation, and apoptosis and has been widely studied as a tumor inhibitor. However, little is known about this cytokine's role in neuroblastoma. In this study, we investigated the possible effects of IL-24 on inducing neuroblastoma cell differentiation, growth inhibition, and apoptosis in vitro. Our data show that IL-24 promotes neuroblastoma SH-SY5Y cell differentiation, growth inhibition, and apoptosis. Furthermore, we found that the differentiation- and apoptosis-inducing action of IL-24 depends on the accumulation of reactive oxygen species (ROS). These results suggest that IL-24 can induce neuroblastoma cell differentiation and apoptosis and may be a potential therapeutic agent for neuroblastoma.
Collapse
Affiliation(s)
- Yuan Li
- Department of Pediatric Surgery, Xuzhou Children's Hospital , Xuzhou, Jiangsu, China
| | | | | | | | | | | |
Collapse
|
13
|
Zhuo B, Wang R, Yin Y, Zhang H, Ma T, Liu F, Cao H, Shi Y. Adenovirus arming human IL-24 inhibits neuroblastoma cell proliferation in vitro and xenograft tumor growth in vivo. Tumour Biol 2013; 34:2419-26. [PMID: 23609032 DOI: 10.1007/s13277-013-0792-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 04/03/2013] [Indexed: 11/25/2022] Open
Abstract
Data have increasingly shown that interlukin-24 (IL-24) has growth suppression activity and can induce apoptosis in a broad spectrum of tumor cells. However, the therapeutic effect of IL-24 on human neuroblastoma has rarely been explored. In this study, we used a human neuroblastoma cell line (SH-SY5Y) to reveal the effect of adenovirus-mediated IL-24 (Ad-IL24) gene therapy for neuroblastoma. We showed that Ad-IL24 effectively inhibited the proliferation of SH-SY5Y cells in vitro by conspicuously inducing apoptosis. To further explore the molecular mechanism by which Ad-IL24 induced apoptosis in SH-SY5Y tumor cells, we found that Ad-IL24 increased the expression of Bax and promoted the activation of caspase-3, while decreasing Bcl-2 levels. We also demonstrated that Ad-IL24 significantly inhibited tumor growth in vivo in a xenograft neuroblastoma tumor in athymic nude mice. In summary, Ad-IL24 overexpression exerted potent antitumor activity via inducing apoptosis in neuroblastoma cells. Therefore, IL-24 has the potential to serve as an agent for gene therapy in the treatment of neuroblastoma.
Collapse
Affiliation(s)
- Baobiao Zhuo
- Department of Surgery, Xuzhou Children's Hospital, 18 Suti North Road, Xuzhou, Jiangsu, 221006, China.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Whitaker EL, Filippov VA, Duerksen-Hughes PJ. Interleukin 24: Mechanisms and therapeutic potential of an anti-cancer gene. Cytokine Growth Factor Rev 2012; 23:323-31. [DOI: 10.1016/j.cytogfr.2012.08.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 08/20/2012] [Indexed: 12/18/2022]
|
15
|
Zhuang Y, Peng LS, Zhao YL, Shi Y, Mao XH, Guo G, Chen W, Liu XF, Zhang JY, Liu T, Luo P, Yu PW, Zou QM. Increased intratumoral IL-22-producing CD4(+) T cells and Th22 cells correlate with gastric cancer progression and predict poor patient survival. Cancer Immunol Immunother 2012; 61:1965-75. [PMID: 22527243 PMCID: PMC11029610 DOI: 10.1007/s00262-012-1241-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 02/27/2012] [Indexed: 12/20/2022]
Abstract
IL-22-producing CD4(+) T cells (IL-22(+)CD4(+) T cells) and Th22 cells (IL-22(+)IL-17(-)IFN-γ(-)CD4(+) T cells) represent newly discovered T-cell subsets, but their nature, regulation, and clinical relevance in gastric cancer (GC) are presently unknown. In our study, the frequency of IL-22(+)CD4(+) T cells in tumor tissues from 76 GC patients was significantly higher than that in tumor-draining lymph nodes, non-tumor, and peritumoral tissues. Most intratumoral IL-22(+)CD4(+) T cells co-expressed IL-17 and IFN-γ and showed a memory phenotype. Locally enriched IL-22(+)CD4(+) T cells positively correlated with increased CD14(+) monocytes and IL-6 and IL-23 detection ex vivo, and in vitro IL-6 and IL-23 induced the polarization of IL-22(+)CD4(+) T cells in a dose-dependent manner and the polarized IL-22(+)CD4(+) T cells co-expressed of IL-17 and IFN-γ. Moreover, IL-22(+)CD4(+) T-cell subsets (IL-22(+)IL-17(+)CD4(+), IL-22(+)IL-17(-)CD4(+), IL-22(+)IFN-γ(+)CD4(+), IL-22(+)IFN-γ(-)CD4(+), and IL-22(+)IL-17(+)IFN-γ(+)CD4(+) T cells), and Th22 cells were also increased in tumors. Furthermore, higher intratumoral IL-22(+)CD4(+) T-cell percentage and Th22-cell percentage were found in patients with tumor-node-metastasis stage advanced and predicted reduced overall survival. In conclusion, our data indicate that IL-22(+)CD4(+) T cells and Th22 cells are likely important in establishing the tumor microenvironment for GC; increased intratumoral IL-22(+)CD4(+) T cells and Th22 cells are associated with tumor progression and predict poorer patient survival, suggesting that tumor-infiltrating IL-22(+)CD4(+) T cells and Th22 cells may be suitable therapeutic targets in patients with GC.
Collapse
Affiliation(s)
- Yuan Zhuang
- Department of Clinical Microbiology and Immunology, College of Medical Laboratory Science, Third Military Medical University, No.30 Gaotanyan street, Chongqing, 400038 People’s Republic of China
| | - Liu-sheng Peng
- Department of Clinical Microbiology and Immunology, College of Medical Laboratory Science, Third Military Medical University, No.30 Gaotanyan street, Chongqing, 400038 People’s Republic of China
| | - Yong-liang Zhao
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, No.30 Gaotanyan street, Chongqing, 400038 People’s Republic of China
| | - Yun Shi
- Department of Clinical Microbiology and Immunology, College of Medical Laboratory Science, Third Military Medical University, No.30 Gaotanyan street, Chongqing, 400038 People’s Republic of China
| | - Xu-hu Mao
- Department of Clinical Microbiology and Immunology, College of Medical Laboratory Science, Third Military Medical University, No.30 Gaotanyan street, Chongqing, 400038 People’s Republic of China
| | - Gang Guo
- Department of Clinical Microbiology and Immunology, College of Medical Laboratory Science, Third Military Medical University, No.30 Gaotanyan street, Chongqing, 400038 People’s Republic of China
| | - Weisan Chen
- Ludwig Institute for Cancer Research, Austin Hospital, Studley Road, Heidelberg, VIC 3084 Australia
| | - Xiao-fei Liu
- Department of Clinical Microbiology and Immunology, College of Medical Laboratory Science, Third Military Medical University, No.30 Gaotanyan street, Chongqing, 400038 People’s Republic of China
| | - Jin-yu Zhang
- Department of Clinical Microbiology and Immunology, College of Medical Laboratory Science, Third Military Medical University, No.30 Gaotanyan street, Chongqing, 400038 People’s Republic of China
| | - Tao Liu
- Department of Clinical Microbiology and Immunology, College of Medical Laboratory Science, Third Military Medical University, No.30 Gaotanyan street, Chongqing, 400038 People’s Republic of China
| | - Ping Luo
- Department of Clinical Microbiology and Immunology, College of Medical Laboratory Science, Third Military Medical University, No.30 Gaotanyan street, Chongqing, 400038 People’s Republic of China
| | - Pei-wu Yu
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, No.30 Gaotanyan street, Chongqing, 400038 People’s Republic of China
| | - Quan-ming Zou
- Department of Clinical Microbiology and Immunology, College of Medical Laboratory Science, Third Military Medical University, No.30 Gaotanyan street, Chongqing, 400038 People’s Republic of China
| |
Collapse
|
16
|
Nagakawa H, Shimozato O, Yu L, Wada A, Kawamura K, Li Q, Chada S, Tada Y, Takiguchi Y, Tatsumi K, Tagawa M. Expression of a murine homolog of apoptosis-inducing human IL-24/MDA-7 in murine tumors fails to induce apoptosis or produce anti-tumor effects. Cell Immunol 2012; 275:90-7. [PMID: 22475191 DOI: 10.1016/j.cellimm.2012.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2011] [Revised: 01/18/2012] [Accepted: 02/16/2012] [Indexed: 11/30/2022]
Abstract
Expression of human interleukin (IL)-24 in tumors achieved anti-tumor effects through apoptosis. IL-24 also induced secretion of proinflammatory cytokines, suggesting the role in immunity. We showed that murine IL-24 transcripts started from the second initiation codon and that expressed mIL-24 in tumors failed to induce apoptosis. Proliferation of murine cells expressing mIL-24 was the same as that of the parent cells and inoculation of the mIL-24-expressing tumors into syngeneic mice did not produce anti-tumor effects. Secretory mIL-24 did not induce the expression of the IL-6, TNF-α or IFN-γ gene in spleen cells. Expression of mIL-24 receptor subunits, IL-22R and IL-20R1, was undetectable in spleen cells even though they were stimulated by anti-CD3, anti-CD40 antibody or concanavalin A. Transduction of murine tumors with adenoviruses expressing the human IL-24 gene however suppressed the viability and decreased the tumor growth. These data suggest that mIL-24 is functionally irrelevant to the human counterpart.
Collapse
Affiliation(s)
- Hiroyasu Nagakawa
- Division of Pathology and Cell Therapy, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba 260-8717, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Luo XR, Li JS, Niu Y, Miao L. Targeted killing effects of double CD and TK suicide genes controlled by survivin promoter on gastric cancer cell. Mol Biol Rep 2010; 38:1201-7. [PMID: 20574710 DOI: 10.1007/s11033-010-0218-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Accepted: 06/11/2010] [Indexed: 10/25/2022]
Abstract
Suicide genes such as cytosine deaminase (CD) and herpes simplex virus thymidine kinase (TK) encode products that convert nontoxic substances (prodrugs) into toxic metabolites. Studies in recent years indicated that survivin(sur) expression was associated with the biological behaviors of gastric carcinoma. In the present study, targeted killing effects of double CD and TK suicide genes controlled by survivin promoter on gastric cancer cell were investigated, the recombinant pSCT vector containing CD and TK genes driven by sur promoter was constructed and transfected into SGC-7901 cells. After adding the CCV and 5-FC, the effects of double suicide genes on cell growth, cell cycle and proliferation were determined by MTT assay and flow cytometry (FCM). The results showed that sur promoter could specifically drive the expression of double CD/TK gene in SGC-7901 cells, whereas not in the normal GES-1 cell. After using CCV and 5-FC, the growth of SGC-7901 cells was inhibited. G1 phase proportion was significantly higher in SGC-7901 cells transfected with double suicide genes than the untransfected cells. These results suggest that CD and TK double suicide genes driven by sur promoter could provide a new approach for enhancing selective suicide gene therapy of CD/5-FC for the treatment of advanced gastric carcinoma.
Collapse
Affiliation(s)
- Xian-Run Luo
- Department of Gastroenterology, The First Affiliated Hospital of ZhengZhou University, No 1 Jianshe East Road, Zhengzhou, Henan, 450052, People's Republic of China
| | | | | | | |
Collapse
|
18
|
Anticancer genes: inducers of tumour-specific cell death signalling. Trends Mol Med 2010; 16:88-96. [PMID: 20138582 DOI: 10.1016/j.molmed.2009.12.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 12/14/2009] [Accepted: 12/21/2009] [Indexed: 12/11/2022]
Abstract
Recent studies have revealed a new class of genes encoding proteins with specific anticancer activity. Upon ectopic expression, these factors cause cell death specifically in tumour cells by apoptosis, autophagy or mitotic catastrophe, yet normal cells are spared. Some of these genes or their encoded proteins are in clinical development and show promising results, and their signalling pathways are currently under intense investigation. Defining these genes as anticancer genes, we review what is known about their functions, the specific cell death signals they induce and the status of cancer therapy approaches that emulate their function. Systematic screening for such anticancer genes might lead to the identification of a repertoire of signalling pathways directed against cellular alterations that are specific for tumour cells.
Collapse
|