1
|
Wu ZB, Wang GY, Wang B, Peng X, Wang JS, Zhang MC, Geng W, Xie HT. IFNα2b/5-FU inhibits proliferation and cell cycle of squamous carcinoma cell line Cal27. Int J Ophthalmol 2025; 18:565-574. [PMID: 40256022 PMCID: PMC11947539 DOI: 10.18240/ijo.2025.04.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/18/2024] [Indexed: 04/22/2025] Open
Abstract
AIM To investigate the pathological features of ocular surface squamous neoplasia (OSSN) and evaluate the synergistic therapeutic effects of interferon-α2b (IFNα2b) and 5-fluorouracil (5-FU) on cellular proliferation, migration, apoptosis, and cell cycle of human oral squamous carcinoma cell line Cal27. METHODS Tissue specimens from OSSN were processed with hematoxylin-eosin (HE) and immunofluorescence (IF) staining to characterize pathological changes. We analyzed the expression levels of four pivotal proteins involved in 5-FU metabolism: interferon alpha receptor (IFNAR), thymidylate synthase (TS), thymidine phosphorylase (TP), and dihydropyrimidine dehydrogenase (DPD). Cal27 cell lines were treated with a spectrum of concentrations of IFNα2b and 5-FU, either in isolation or in combination. Then, cell activity was measured utilizing CCK-8 assay and dose-effect curves were calculated, while tumor cell migration was detected by cell scratch experiments. Cal27 cells were added with IFNα2b and 5-FU in a non-constant ratio drug combination design and the corresponding combination index (CI) and fraction affected (Fa) were calculated with CompuSyn software. Western blot assay was conducted to quantify the expression of TP, TS, and DPD. Cell cycle and apoptosis were measured with flow cytometry and terminal deoxynucleotidyl transferase-mediated dUTP nick and labeling (TUNEL) assay. RESULTS Treatment with both IFNα2b and 5-FU inhibited cell proliferation. Except for the lowest and highest doses of 5-FU, CI values for all other groups were below 1, suggesting a synergistic interaction. Low concentrations of IFNα2b and 5-FU both diminished the relative mobility of Cal27 cells, instead, a stronger inhibitory effect was observed when the two drugs were co-applied. The expression levels of TP and DPD in Cal27 cells were dose-dependently increased at a low concentration of IFNα2b. Low-dose IFNα2b combined with 5-FU significantly inhibited cell proliferation in G0/G1 phase compared to 5-FU monotherapy. Medium and high doses of IFNα2b and all concentrations of 5-FU could induce apoptosis in a concentration-dependent manner. The susceptibility to 5-FU treatment and apoptosis rates of tumor cells were elevated with low doses of IFNα2b. CONCLUSION Both IFNα2b and 5-FU, when administered individually or in combination, effectively suppress the proliferation and migration of Cal27 tumor cells, induce cell apoptosis and arrest cell cycle. Low doses of IFNα2b increase the antitumor effects of 5-FU on Cal27 potentially through up-regulating the expression of TP, demonstrating a synergistic effect between IFNα2b and 5-FU.
Collapse
Affiliation(s)
- Zong-Bo Wu
- Department of Ophthalmology, Jingshan Union Hospital, Union Hospital, Huazhong University of Science and Technology, Jingshan 431800, Hubei Province, China
| | - Gong-Yue Wang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Bei Wang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Xi Peng
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Jia-Song Wang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Ming-Chang Zhang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Wen Geng
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Hua-Tao Xie
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| |
Collapse
|
2
|
Li W, Chen G, Peng H, Zhang Q, Nie D, Guo T, Zhu Y, Zhang Y, Lin M. Research Progress on Dendritic Cells in Hepatocellular Carcinoma Immune Microenvironments. Biomolecules 2024; 14:1161. [PMID: 39334927 PMCID: PMC11430656 DOI: 10.3390/biom14091161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Dendritic cells (DCs) are antigen-presenting cells that play a crucial role in initiating immune responses by cross-presenting relevant antigens to initial T cells. The activation of DCs is a crucial step in inducing anti-tumor immunity. Upon recognition and uptake of tumor antigens, activated DCs present these antigens to naive T cells, thereby stimulating T cell-mediated immune responses and enhancing their ability to attack tumors. It is particularly noted that DCs are able to cross-present foreign antigens to major histocompatibility complex class I (MHC-I) molecules, prompting CD8+ T cells to proliferate and differentiate into cytotoxic T cells. In the malignant progression of hepatocellular carcinoma (HCC), the inactivation of DCs plays an important role, and the activation of DCs is particularly important in anti-HCC immunotherapy. In this review, we summarize the mechanisms of DCs activation in HCC, the involved regulatory factors and strategies to activate DCs in HCC immunotherapy. It provides a basis for the study of HCC immunotherapy through DCs activation.
Collapse
Affiliation(s)
- Wenya Li
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
- Graduate School, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Guojie Chen
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
- Medical School, Nantong University, Nantong 226019, China
| | - Hailin Peng
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
| | - Qingfang Zhang
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
| | - Dengyun Nie
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
- Graduate School, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ting Guo
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
- Graduate School, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yinxing Zhu
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
| | - Yuhan Zhang
- The First School of Clinical Medicine Southern Medical University, Guangzhou 510515, China
| | - Mei Lin
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
- Graduate School, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
3
|
Li J, Xuan S, Dong P, Xiang Z, Gao C, Li M, Huang L, Wu J. Immunotherapy of hepatocellular carcinoma: recent progress and new strategy. Front Immunol 2023; 14:1192506. [PMID: 37234162 PMCID: PMC10206122 DOI: 10.3389/fimmu.2023.1192506] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Abstract
Due to its widespread occurrence and high mortality rate, hepatocellular carcinoma (HCC) is an abhorrent kind of cancer. Immunotherapy is a hot spot in the field of cancer treatment, represented by immune checkpoint inhibitors (ICIs), which aim to improve the immune system's ability to recognize, target and eliminate cancer cells. The composition of the HCC immune microenvironment is the result of the interaction of immunosuppressive cells, immune effector cells, cytokine environment, and tumor cell intrinsic signaling pathway, and immunotherapy with strong anti-tumor immunity has received more and more research attention due to the limited responsiveness of HCC to ICI monotherapy. There is evidence of an organic combination of radiotherapy, chemotherapy, anti-angiogenic agents and ICI catering to the unmet medical needs of HCC. Moreover, immunotherapies such as adoptive cellular therapy (ACT), cancer vaccines and cytokines also show encouraging efficacy. It can significantly improve the ability of the immune system to eradicate tumor cells. This article reviews the role of immunotherapy in HCC, hoping to improve the effect of immunotherapy and develop personalized treatment regimens.
Collapse
Affiliation(s)
- Jiarui Li
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shihai Xuan
- Department of Laboratory Medicine, The People’s Hospital of Dongtai City, Dongtai, China
| | - Peng Dong
- Hangzhou Institute of Cardiovascular Diseases, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
| | - Ze Xiang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ce Gao
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Mo Li
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Lan Huang
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Jian Wu
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| |
Collapse
|
4
|
Xiang D, Yang W, Fang Z, Mao J, Yan Q, Li L, Tan J, Yu C, Qian J, Tang D, Pan X, Cheng H, Sun D. Agrimol B inhibits colon carcinoma progression by blocking mitochondrial function through the PGC-1α/NRF1/TFAM signaling pathway. Front Oncol 2022; 12:1055126. [PMID: 36591497 PMCID: PMC9794846 DOI: 10.3389/fonc.2022.1055126] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022] Open
Abstract
Background The activation of peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) stimulates the transcription of the downstream target proteins, mitochondrial transcription factor A (TFAM) and nuclear respiratory factor 1 (NRF1), which induces mitochondrial biogenesis and promotes colorectal tumorigenesis. Agrimol B (Agr) is a constituent of Agrimonia pilosa Ledeb. that exerts anticancer effects. Herein, we aimed to investigate the antitumor activity of Agr and its mechanism of action. Methods The interaction between Agr and PGC-1α was predicted by molecular docking. After the treatment with different concentrations of Agr (0, 144, 288, and 576 nM), the cell viability, migration rate, proliferation rate, and apoptosis rate of human colon cancer HCT116 cells were determined. Mitochondrial activity, cellular reactive oxygen species (ROS), and mitochondrial membrane potential were assessed to measure the regulatory effect of Agr on mitochondrial function. Western blotting (WB) assay was used to examine the expression of PGC-1α, NRF1, and TFAM, as well as of the pro-apoptotic proteins, Bax and Caspase-3, and the antiapoptotic protein (Bcl-2). Finally, subcutaneous tumor xenograft model mice were used to evaluate the effect of Agr on colorectal cancer (CRC) in vivo. Results The molecular docking results revealed a high likelihood of Agr interacting with PGC-1α. Agr inhibited the proliferation and migration of HCT116 cells, promoted ROS production and mitochondrial oxidative stress, inhibited mitochondrial activity, and decreased mitochondrial membrane potential. Agr induced cell apoptosis and, in combination with PGC-1α, impaired mitochondrial biogenesis and suppressed the expression of NRF1 and TFAM. Agr also suppressed the expression of Bcl-2 and Cleaved-Caspase-3 and increased the expression of Bax and Caspase-3. In addition, the in vivo antitumor effect and mechanism of Agr were confirmed by using a subcutaneous tumor xenograft mouse model. Conclusions Our findings demonstrated that Agr regulates the expression of PGC-1α, thereby inducing mitochondrial dysfunction and promoting tumor cell apoptosis. This work highlights the potential of Agr as a promising therapeutic candidate in CRC.
Collapse
Affiliation(s)
- Dongyang Xiang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenjuan Yang
- Oncology Department, Kunshan Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan, China
| | - Zihan Fang
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jialei Mao
- Oncology Department, Kunshan Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan, China
| | - Qiuying Yan
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liu Li
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China,The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiani Tan
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China,The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chengtao Yu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China,The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jun Qian
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China,The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China,Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Dongxin Tang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xiaoting Pan
- Oncology Department, Kunshan Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan, China,*Correspondence: Haibo Cheng, ; Xiaoting Pan, ; Dongdong Sun,
| | - Haibo Cheng
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China,The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China,Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China,*Correspondence: Haibo Cheng, ; Xiaoting Pan, ; Dongdong Sun,
| | - Dongdong Sun
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China,School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China,*Correspondence: Haibo Cheng, ; Xiaoting Pan, ; Dongdong Sun,
| |
Collapse
|
5
|
Mousa AM, Aldebasi YH. L-carnosine mitigates interleukin-1α-induced dry eye disease in rabbits via its antioxidant, anti-inflammatory, antiapoptotic, and antifibrotic effects. Cutan Ocul Toxicol 2021; 40:241-251. [PMID: 34056995 DOI: 10.1080/15569527.2021.1935995] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To elucidate the implications of L-carnosine on interleukin-1α (IL-1α)-induced inflammation of lacrimal glands (LGs). MATERIALS AND METHODS Forty rabbits were divided equally into four groups: control group (G1), IL-1α (G2), L-carnosine (G3), and L-carnosine plus IL-1α (G4). Several clinical, histopathological, immunohistochemical, morphometric, and biochemical investigations were performed, followed by statistical analysis to diagnose the presence of dry eye disease (DED). RESULTS The LGs of G2 rabbits showed degeneration of the acinar cells, increased deposition of collagen fibers, and marked immunoexpression of FasL; elevated levels of interferon-γ, tumor necrosis factor-α, transforming growth factor-β1, and malondialdehyde; and decreased levels of glutathione peroxidase, superoxide dismutase, catalase, and reactive oxygen species compared with those of G1 rabbits. In contrast, administration of L-carnosine to G4 rabbits revealed marked improvement of all previously harmful changes in G2 rabbits, indicating the cytoprotective effects of L-carnosine against IL-1α-induced inflammation of LGs. CONCLUSIONS IL-1α induced inflammation of LGs and eye dryness via oxidative stress, proinflammatory, apoptotic, and profibrotic effects, whereas L-carnosine mitigated DED through antioxidant, anti-inflammatory, antiapoptotic, and antifibrotic effects on LGs. Therefore, this work demonstrates for the first time that L-carnosine may be used as adjuvant therapy for the preservation of visual integrity in patients with DED.HighlightsIL-1α induced dry eye disease through its oxidative stress, proinflammatory, apoptotic and profibrotic effects on the lacrimal glands of rabbit.L-carnosine has antioxidant, anti-inflammatory, antiapoptotic and antifibrotic effects.L-carnosine mitigated IL-1α induced dry eye disease via elevating the levels of FasL, IFN-γ, TNF-α, TGFβ1 and MDA as well as reducing the levels of antioxidants (GPx, SOD, and catalase) and ROS in the lacrimal glands of rabbit.L-carnosine could be used as a novel adjuvant therapy for the treatment of dry eye disease.
Collapse
Affiliation(s)
- Ayman M Mousa
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia.,Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Yousef H Aldebasi
- Department of Optometry, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
6
|
Engineering Chlamydomonas reinhardtii for Expression of Functionally Active Human Interferon-α. Mol Biotechnol 2018; 61:134-144. [DOI: 10.1007/s12033-018-0143-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
7
|
Effect of silencing NEK2 on biological behaviors of HepG2 in human hepatoma cells and MAPK signal pathway. Tumour Biol 2015; 37:2023-35. [DOI: 10.1007/s13277-015-3993-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 08/25/2015] [Indexed: 12/11/2022] Open
|
8
|
Motawi TK, El-Boghdady NA, El-Sayed AM, Helmy HS. Comparative study of the effects of PEGylated interferon-α2a versus 5-fluorouracil on cancer stem cells in a rat model of hepatocellular carcinoma. Tumour Biol 2015; 37:1617-25. [PMID: 26304505 DOI: 10.1007/s13277-015-3920-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 08/11/2015] [Indexed: 12/20/2022] Open
Abstract
Cancer stem cells (CSCs) in hepatocellular carcinoma (HCC) possess tumor-initiating, metastatic, and drug resistance properties. This study was conducted to evaluate the effects of PEGylated interferon-α2a (PEG-IFN-α2a) and 5-fluorouracil (5-FU) on the expression of CSC markers and on specific pathways that contribute to the propagation of CSCs in HCC. HCC was initiated in rats using a single intraperitoneal dose of diethylnitrosamine (DENA) (200 mg/kg) and promoted by weekly subcutaneous injections of carbon tetrachloride (CCl4) for 6 weeks. After the appearance of dysplastic nodules, the animals received PEG-IFN-α2a or 5-FU for 8 weeks. CSC markers (OV6, CD90) and molecules related to transforming growth factor β (TGF-β) and other signaling pathways were assessed in hepatic tissues. The PEG-IFN-α2a treatment effectively suppressed the hepatic expression of OV6 and CD90, ameliorated the diminished hepatic expression of TGF-β receptor II (TGF-βRII) and β2-spectrin (β2SP), and significantly reduced the elevated hepatic expression of TGF-β1, interleukin6 (IL6), signal transducer and activator of transcription3 (STAT3), and vascular endothelial growth factor (VEGF). In contrast, the 5-FU treatment failed to reduce the overexpression of CSC markers and barely affected the disrupted TGF-β signaling. Furthermore, it had no effect on angiogenesis or nitrosative stress. PEG-IFN-α2a, but not 5-FU, could reduce the propagation of CSCs during the progression of HCC by upregulating the disrupted TGF-β signaling, suppressing the IL6/STAT3 pathway and reducing angiogenesis.
Collapse
Affiliation(s)
- Tarek Kamal Motawi
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | | | - Abeer Mostafa El-Sayed
- Department of Pathology, National Cancer Institute, Cairo University, Cairo, 11796, Egypt
| | - Hebatullah Samy Helmy
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
9
|
Yi BR, Kim SU, Choi KC. Additional effects of engineered stem cells expressing a therapeutic gene and interferon-β in a xenograft mouse model of endometrial cancer. Int J Oncol 2015; 47:171-8. [PMID: 25963746 DOI: 10.3892/ijo.2015.2999] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 05/04/2015] [Indexed: 11/06/2022] Open
Abstract
Endometrial cancer is the most common gynecologic malignancy in women worldwide. In the present study, we evaluated the effects of neural stem cell-directed enzyme/prodrug therapy (NDEPT) designed to more selectively target endometrial cancer. For this, we employed two different types of neural stem cells (NSCs), HB1.F3.CD and HB1.F3.CD.IFN-β cells. Cytosine deaminase (CD) can convert the non-toxic prodrug, 5-fluorocytosine (5-FC), into a toxic agent, 5-fluorouracil (5-FU), which inhibits DNA synthesis. IFN-β is a powerful cytotoxic cytokine that is released by activated immune cells or lymphocytes. In an animal model xenografted with endometrial Ishikawa cancer cells, the stem cells stained with CM-DiI were injected into nearby tumor masses and 5-FC was delivered by intraperitoneal injection. Co-expression of CD and IFN-β significantly inhibited the growth of cancer (~50-60%) in the presence of 5-FC. Among migration-induced factors, VEGF gene was highly expressed in endometrial cancer cells. Histological analysis showed that the aggressive nature of cancer was inhibited by 5-FC in the mice treated with the therapeutic stem cells. Furthermore, PCNA expression was more decreased in HB1.F3.CD.IFN-β treated mice rather than HB1.F3.CD treated mice. To confirm the in vitro combined effects of 5-FU and IFN-β, 5-FU was treated in Ishikawa cells. 5-FU increased the IFN-β/receptor 2 (IFNAR2) and BXA levels, indicating that 5-FU increased sensitivity of endometrial cancer cells to IFN-β, leading to apoptosis of cancer cells. Taken together, these results provide evidence for the efficacy of therapeutic stem cell-based immune therapy involving the targeted expression of CD and IFN-β genes at endometrial cancer sites.
Collapse
Affiliation(s)
- Bo-Rim Yi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Seung U Kim
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|
10
|
Wang J, Xu F, Zhu D, Duan Y, Chen J, Sun X, He X, Li P, Sun W, Feng J. Schistosoma japonicum soluble egg antigens facilitate hepatic stellate cell apoptosis by downregulating Akt expression and upregulating p53 and DR5 expression. PLoS Negl Trop Dis 2014; 8:e3106. [PMID: 25144704 PMCID: PMC4140669 DOI: 10.1371/journal.pntd.0003106] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 07/09/2014] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND The induction of hepatic stellate cell (HSC) apoptosis has potential as a potent strategy to diminish the progression of liver fibrosis. Previous studies have demonstrated the ability of soluble egg antigens (SEA) from schistosomes to inhibit HSC activation and to induce apoptosis in vitro. In this study, we aimed to explore the mechanism of SEA-induced apoptosis in HSCs. METHODOLOGY/PRINCIPAL FINDINGS In this study, we found that SEA could upregulate p53 and DR5 and downregulate the p-Akt. The apoptosis of HSCs induced by SEA could be reduced in HSCs that were treated with p53-specific siRNA and in HSCs that were treated with DR5-specific shRNA. In addition, GW501516, which enhances the expression of Akt, could also decrease the SEA-induced HSC apoptosis. We also found that the increased expression of p53 and DR5 induced by SEA through Mdm2 were reduced by GW501516. CONCLUSIONS/SIGNIFICANCE Our data suggest that SEA can induce HSC apoptosis by downregulating Akt expression and upregulating p53-dependent DR5 expression.
Collapse
Affiliation(s)
- Jianxin Wang
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Feifan Xu
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Dandan Zhu
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Yinong Duan
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
- * E-mail:
| | - Jinling Chen
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Xiaolei Sun
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Xue He
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Pan Li
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Wei Sun
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Jinrong Feng
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| |
Collapse
|
11
|
Rajan M, Raj V, Al-Arfaj AA, Murugan A. Hyaluronidase enzyme core-5-fluorouracil-loaded chitosan-PEG-gelatin polymer nanocomposites as targeted and controlled drug delivery vehicles. Int J Pharm 2013; 453:514-22. [DOI: 10.1016/j.ijpharm.2013.06.030] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 06/08/2013] [Accepted: 06/12/2013] [Indexed: 11/24/2022]
|
12
|
Ortiz R, Prados J, Melguizo C, Rama AR, Alvarez PJ, Rodríguez-Serrano F, Caba O, Boulaiz H, Aranega A. Gef gene therapy enhances the therapeutic efficacy of cytotoxics in colon cancer cells. Biomed Pharmacother 2012; 66:563-567. [PMID: 22770988 DOI: 10.1016/j.biopha.2012.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 05/28/2012] [Indexed: 02/04/2023] Open
Abstract
The potential use of gene therapy to improve the response of patients with advanced cancer is being intensively analyzed. We evaluated the cytotoxic impact of the gef gene, a suicide gene, which has a demonstrated antiproliferative activity in tumor cells, in colon carcinoma cells in order to improve the antitumour effect of chemotherapeutic drugs used as first line treatment in the management of advanced colon cancer. We found that the gef gene induced a marked decrease in cell viability (50% in 24h) in T-84 cells through cell death by apoptosis. Interestingly, when gef gene expression was combined with drugs of choice in the clinical treatment of colon cancer (5-fluorouracil, oxaliplatin and irinotecan), a strong synergistic effect was observed with approximately a 15-20% enhancement of the antiproliferative effect. Our data demonstrate, for the first time, that gef gene expression induces significant growth arrest in colon cancer cells and that it is able to enhance the effect of some cytotoxic drugs compared with a single therapeutic approach. These results indicate the potential therapeutic value of the gef gene in colon cancer combination therapy.
Collapse
Affiliation(s)
- Raúl Ortiz
- Department of Health Sciences, University of Jaén, Jaén, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Ortiz R, Prados J, Melguizo C, Arias JL, Ruiz MA, Álvarez PJ, Caba O, Luque R, Segura A, Aránega A. 5-Fluorouracil-loaded poly(ε-caprolactone) nanoparticles combined with phage E gene therapy as a new strategy against colon cancer. Int J Nanomedicine 2012; 7:95-107. [PMID: 22275826 PMCID: PMC3260954 DOI: 10.2147/ijn.s26401] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Indexed: 01/22/2023] Open
Abstract
This work aimed to develop a new therapeutic approach to increase the efficacy of 5-fluorouracil (5-FU) in the treatment of advanced or recurrent colon cancer. 5-FU-loaded biodegradable poly(ε-caprolactone) nanoparticles (PCL NPs) were combined with the cytotoxic suicide gene E (combined therapy). The SW480 human cancer cell line was used to assay the combined therapeutic strategy. This cell line was established from a primary adenocarcinoma of the colon and is characterized by an intrinsically high resistance to apoptosis that correlates with its resistance to 5-FU. 5-FU was absorbed into the matrix of the PCL NPs during synthesis using the interfacial polymer disposition method. The antitumor activity of gene E from the phage ϕX174 was tested by generating a stable clone (SW480/12/E). In addition, the localization of E protein and its activity in mitochondria were analyzed. We found that the incorporation of 5-FU into PCL NPs (which show no cytotoxicity alone), significantly improved the drug's anticancer activity, reducing the proliferation rate of colon cancer cells by up to 40-fold when compared with the nonincorporated drug alone. Furthermore, E gene expression sensitized colon cancer cells to the cytotoxic action of the 5-FU-based nanomedicine. Our findings demonstrate that despite the inherent resistance of SW480 to apoptosis, E gene activity is mediated by an apoptotic phenomenon that includes modulation of caspase-9 and caspase-3 expression and intense mitochondrial damage. Finally, a strongly synergistic antiproliferative effect was observed in colon cancer cells when E gene expression was combined with the activity of the 5-FU-loaded PCL NPs, thereby indicating the potential therapeutic value of the combined therapy.
Collapse
Affiliation(s)
- Raúl Ortiz
- Institute of Biopathology and Regenerative Medicine (IBIMER)
- Department of Health Science, University of Jaén, Jaén, Spain
| | - José Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER)
| | | | - José L Arias
- Department of Pharmacy and Pharmaceutical Technology, University of Granada, Granada, Spain
| | - M Adolfina Ruiz
- Department of Pharmacy and Pharmaceutical Technology, University of Granada, Granada, Spain
| | - Pablo J Álvarez
- Institute of Biopathology and Regenerative Medicine (IBIMER)
| | - Octavio Caba
- Institute of Biopathology and Regenerative Medicine (IBIMER)
- Department of Health Science, University of Jaén, Jaén, Spain
| | - Raquel Luque
- Service of Medical Oncology, Virgen de las Nieves Hospital, Granada, Spain
| | - Ana Segura
- CSIC-Estacion Experimental del Zaidin, Department of Environmental Protection, Granada, Spain
| | - Antonia Aránega
- Institute of Biopathology and Regenerative Medicine (IBIMER)
| |
Collapse
|
14
|
Nagano H, Wada H, Kobayashi S, Marubashi S, Eguchi H, Tanemura M, Tomimaru Y, Osuga K, Umeshita K, Doki Y, Mori M. Long-term outcome of combined interferon-α and 5-fluorouracil treatment for advanced hepatocellular carcinoma with major portal vein thrombosis. Oncology 2011; 80:63-9. [PMID: 21659784 DOI: 10.1159/000328281] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Accepted: 02/07/2011] [Indexed: 12/17/2022]
Abstract
BACKGROUND/AIM We previously reported the beneficial effects of a combination therapy of interferon (IFN)-α/5-fluorouracil (FU) for advanced hepatocellular carcinoma (HCC) with tumor thrombi in the major portal branches. This report describes the results of longer follow-up and includes more than twice the number of patients relative to the previous report; it also evaluates the clinical predictor on the response to the combination therapy and long-term survival. METHODS The study subjects were 102 patients with advanced HCC and tumor thrombi in the major branches of the portal vein (Vp3 or 4). They were treated with at least 2 courses of IFN-α/5-FU. RESULTS No major treatment-related complications were noted. In the 102 patients, 40 (39.2%) showed objective response [11 (10.8%) showed complete response, 29 (28.4%) partial response], 8 (7.9%) showed no response and 54 (52.9%) showed progressive disease. CONCLUSION IFN-α/5-FU combination therapy is a promising modality for advanced HCC with tumor thrombi in the major portal branches.
Collapse
Affiliation(s)
- Hiroaki Nagano
- Department of Surgery, Graduate School of Medicine, Osaka University, Suita, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Yin H, Xie F, Zhang J, Yang Y, Deng B, Sun J, Wang Q, Qu X, Mao H. Combination of interferon-α and 5-fluorouracil induces apoptosis through mitochondrial pathway in hepatocellular carcinoma in vitro. Cancer Lett 2011; 306:34-42. [PMID: 21474236 DOI: 10.1016/j.canlet.2011.02.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 02/14/2011] [Accepted: 02/15/2011] [Indexed: 11/30/2022]
Abstract
Many clinical reports have proven that the combination therapy of interferon-alpha plus 5-fluorouracil is remarkably effective for advanced hepatocellular carcinoma (HCC). However, the mechanism of this therapy is not well understood. Here, we demonstrated that the combination therapy synergistically inhibited the growth of Fas-negative HCC cells, arrested cell-cycle progression and induced apoptosis. Moreover, the combination therapy significantly increased the protein expression of caspase-8, activated Bid and cytochrome c. Meanwhile, the expression of anti-apoptotic gene Bcl-xL was reduced and intracellular calcium elevated obviously during the early stage of treatment. Therefore, mitochondrial pathway was involved in the apoptosis of Fas-negative HCC cells induced by IFN-α/5-FU and Ca(2+) partially promoted the beneficial effect against HCC.
Collapse
Affiliation(s)
- Haipeng Yin
- Institute of Basic Medical Sciences, Qi Lu Hospital, Shandong University, Jinan, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Hagiwara S, Kudo M, Ueshima K, Chung H, Yamaguchi M, Takita M, Haji S, Kimura M, Arao T, Nishio K, Park AM, Munakata H. The cancer stem cell marker CD133 is a predictor of the effectiveness of S1+ pegylated interferon α-2b therapy against advanced hepatocellular carcinoma. J Gastroenterol 2011; 46:212-21. [PMID: 20683621 DOI: 10.1007/s00535-010-0294-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Accepted: 07/08/2010] [Indexed: 02/04/2023]
Abstract
BACKGROUND Combination therapy with the oral fluoropyrimidine anticancer drug S1 and interferon is reportedly effective for the treatment of advanced hepatocellular carcinoma (HCC), but selection criteria for this therapy have not been clarified. In this study, we attempted to identify factors predicting the effectiveness of this combination therapy. METHODS Pathological specimens of HCC were collected before treatment from 31 patients with advanced HCC who underwent S1+ pegylated-interferon (PEG-IFN) α-2b therapy between January 2007 and January 2009. In these pathological specimens, the expression levels of CD133, thymidylate synthase (TS), dihydropyrimidine dehydrogenase (DPD), and interferon-receptor 2 (IFNR2) proteins were determined by Western blot assay. The presence or absence of p53 gene mutations was determined by direct sequencing. The relationships between these protein expression levels and the response rate (RR), progression-free survival (PFS), and overall survival (OS) were evaluated. RESULTS The CD133 protein expression level was significantly lower in the responder group than in the nonresponder group. Comparing the PFS and OS between high- and low-level CD133 expression groups (n = 13 and 18, respectively) revealed that both parameters were significantly prolonged in the latter group. The expression levels of TS, DPD, and IFNR2 protein and the presence of p53 gene mutations did not correlate with the RR. CONCLUSIONS CD133 was identified as a predictor of the therapeutic effect of S1+ PEG-IFN α-2b therapy against advanced HCC.
Collapse
Affiliation(s)
- Satoru Hagiwara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kinki University School of Medicine, 377-2 Ohno-Higashi, Ōsakasayama, Osaka 589-8511, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Zheng T, Wang J, Song X, Meng X, Pan S, Jiang H, Liu L. Nutlin-3 cooperates with doxorubicin to induce apoptosis of human hepatocellular carcinoma cells through p53 or p73 signaling pathways. J Cancer Res Clin Oncol 2010; 136:1597-604. [PMID: 20174822 DOI: 10.1007/s00432-010-0817-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Accepted: 02/01/2010] [Indexed: 11/30/2022]
Abstract
PURPOSE Despite recent advances in chemotherapeutic agents for Hepatocellular carcinoma (HCC) treatment, the results of chemotherapy remain unsatisfactory. Doxorubicin (DOX) still represents the cornerstone in HCC chemotherapy, but resistance and toxicity to normal cells are major obstacles to successful chemotherapy. Therefore, new active agents in HCC chemotherapy and agents that increase the chemosensitivity of HCC cells to DOX are still urgently required. Nutlin-3 is a small-molecule inhibitor that acts to inhibit murine double minute-2 (MDM2) binding to p53 or p73, and subsequently activates p53- or p73-dependent apoptosis signaling pathway. This study was designed to investigate whether Nutlin-3 alters cell toxicity to HCC cells following DNA damage and to assess the suitability of DOX/Nutlin-3 as a chemotherapeutic combination in HCC chemotherapy. METHODS Four human HCC cells were analyzed using cell proliferation assay, apoptosis assay, western blotting, co-immunoprecipitation and siRNA experiments. Anti-tumoral effects of Nutlin-3/DOX targeting the p53/MDM2 and p73/MDM2 pathways were evaluated in HCC cell lines. RESULTS Nutlin-3 enhances the growth inhibition by DOX and potentates the apoptotic effect in all HCC cell lines with different p53 types. Nutlin-3 acts through the disruption of p53-MDM2 binding in HepG2, and the disruption of p73-MDM2 in Huh-7 and Hep3B cell lines with subsequent activation of the apoptotic pathway, which leads to the increase in chemosensitivity to DOX in HCC cells. CONCLUSIONS Taken together, our findings suggest that Nutlin-3 will be active in the treatment of HCC and offers new prospects for overcoming DOX resistance.
Collapse
Affiliation(s)
- Tongsen Zheng
- Department of General Surgery, First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin 150001, Heilongjiang Province, China
| | | | | | | | | | | | | |
Collapse
|
18
|
Taura M, Fukuda R, Suico MA, Eguma A, Koga T, Shuto T, Sato T, Morino-Koga S, Kai H. TLR3 induction by anticancer drugs potentiates poly I:C-induced tumor cell apoptosis. Cancer Sci 2010; 101:1610-7. [PMID: 20367642 PMCID: PMC11159143 DOI: 10.1111/j.1349-7006.2010.01567.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Toll-like receptor 3 (TLR3) has gained recognition as a novel molecular target for cancer therapy because TLR3 activation by its synthetic ligand poly I:C directly causes tumor cell death. Recently, we reported that tumor suppressor p53 increases the expression of TLR3 in several tumor cell lines. Another study also showed that interferon-alpha (IFN-alpha) up-regulates TLR3 expression. We thus hypothesized that various anticancer drugs such as p53-activating reagents and IFNs may potentiate poly I:C-induced tumor cell death through the up-regulation of TLR3 expression. Here, we screened several anticancer drugs that, together with poly I:C, effectively cause tumor cell death in colon carcinoma HCT116 cells. We found that the DNA-damaging reagent 5-fluorouracil (5-FU) increased TLR3 mRNA expression and potentiated poly I:C-induced apoptosis in HCT116 p53(+/+) cells but had only minimal effect in p53(-/-) cells, indicating a p53-dependent pathway. On the other hand, IFN-alpha increased poly I:C-induced apoptosis and the TLR3 mRNA level in HCT116 p53(+/+) and p53(-/-) cell lines. Furthermore, the combination of poly I:C, 5-FU and IFN-alpha induced the highest apoptosis in HCT116 p53(+/+) and p53(-/-) cells. Taken together, these data suggest that the anticancer drugs increased TLR3 expression and subsequently potentiated poly I:C-induced apoptosis likely via p53-dependent and -independent pathways. Considering that the p53 status in malignant cells is heterogeneous, this combination approach may provide a highly effective tumor therapy.
Collapse
Affiliation(s)
- Manabu Taura
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Global COE Cell Fate Regulation Research and Education Unit, Kumamoto University, Kumamoto, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Hepatitis C virus core protein and cellular protein HAX-1 promote 5-fluorouracil-mediated hepatocyte growth inhibition. J Virol 2009; 83:9663-71. [PMID: 19605487 DOI: 10.1128/jvi.00872-09] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Hepatitis C virus (HCV) often causes chronic infection and may lead to hepatocellular carcinoma (HCC). We have shown previously that HCV core protein has pleiotropic functions, including transcriptional regulation of a number of cellular genes, although the mechanism for gene regulation remains unclear. In this study, a mammalian two-hybrid screen identified a novel binding partner, HS1-associated protein X-1 (HAX-1), for HCV core protein from a human liver cDNA library. An association between HAX-1 and HCV core protein was further verified by confocal microscopy and coimmunoprecipitation in HepG2 cells expressing HCV core or full-length (FL) gene. Both HCV core protein and a chemotherapeutic agent for HCC, 5-flouorouracil (5-FU), are known to modulate p53. We examined here whether an association between core and HAX-1 has any functional relevance to p53 modulation in 5-FU-treated cells. For this, the role of HAX-1 on 5-FU treatment was examined in HepG2 cells expressing HCV core or FL gene using cell proliferation, p53 expression, and caspase activation analysis. Cells expressing HCV-core or FL gene were more susceptible to 5-FU-induced growth inhibition than control cells, whereas cell survival was enhanced after suppression of HAX-1 by small interfering RNA. Further, 5-FU-mediated p53 expression was reduced with concurrent HAX-1 suppression in core- or polyprotein-expressing cells compared to control HepG2 cells, and caspase-2 and -7 activities were diminished. On the other hand, HCV core protein did not play a detectable role in 5-FU-mediated caspase-7 activation in the absence of functional p53 in Hep3B or Huh-7 cells. These observations underscore an association between HCV core and HAX-1, which promotes 5-FU mediated p53-dependent caspase-7 activation and hepatocyte growth inhibition.
Collapse
|
20
|
Yang JQ, Pan GD, Chu GP, Liu Z, Liu Q, Xiao Y, Yuan L. Interferon-alpha restrains growth and invasive potential of hepatocellular carcinoma induced by hepatitis B virus X protein. World J Gastroenterol 2008; 14:5564-9; discussion 5568. [PMID: 18810776 PMCID: PMC2746345 DOI: 10.3748/wjg.14.5564] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of interferon-alpha (IFN-α) to restrain the growth and invasive potential of hepatocellular carcinoma (HCC) induced by hepatitis B virus (HBV) X protein.
METHODS: The pcDNA3.1-HBx plasmid was transfected into Chang cells by Lipofectamine in vitro, and Chang/HBx was co-cultured with IFN-α. Cell survival growth curve and clonogenicity assay were used to test the growth potential of Chang/pcDNA3.1, Chang/HBx and IFN-α-Chang/HBx in vitro. Growth assay in nude mice was used to detect the growth potential of Chang/pcDNA3.1, Chang/HBx and IFN-α-Chang/HBx in vivo. Wound healing and transwell migration assays were used to detect the invasive ability of Chang/pcDNA3.1, Chang/HBx and IFN-α-Chang/HBx.
RESULTS: Compared with CCL13 cells transfected with pcDNA3.1, CCL13 with stable expression of hepatitis B virus X protein showed the characteristics of malignant cells with high capability of growth and invasion by detecting their growth curves, colony forming efficiency, wound healing , transwell migration assays and growth assays in nude mice. Its capability of growth and invasion could be controlled by IFN-α.
CONCLUSION: IFN-α can restrain the growth and invasive potential of HCC cells induced by HBx protein, which has provided an experimental basis for IFN-α therapy of HCC.
Collapse
|