1
|
Malathi N, Rajan ST, Warnakulasuriya S. Natural products and diet for the prevention of oral cancer: Research from south and southeast Asia. Oral Dis 2025; 31:1503-1516. [PMID: 38804557 DOI: 10.1111/odi.15002] [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: 02/28/2024] [Revised: 04/19/2024] [Accepted: 05/06/2024] [Indexed: 05/29/2024]
Abstract
Medicinal plants are of prime importance in the discovery of drugs. They are an inherent source of naturally available phytochemicals that can help in the prevention and treatment of several diseases including cancer. This article reviews the experimental and clinical evidence of phytochemicals available in natural dietary products that are used in everyday life across South Asia and South-East Asia for their perceived effectiveness in the management of Potentially Malignant Disorders and prevention of Oral Cancer. The review also highlights the active phytometabolites, their in vitro anti-proliferative properties and targeted signalling pathways, biological activities in in vivo models and translative potential for clinical trials in humans.
Collapse
Affiliation(s)
- N Malathi
- Department of Oral Pathology, Sri Ramachandra Dental College & Hospital, Sri Ramachandra Institute of Higher Education & Research, Chennai, Tamil Nadu, India
| | - Sharada T Rajan
- Department of Oral Pathology, Sri Ramachandra Dental College & Hospital, Sri Ramachandra Institute of Higher Education & Research, Chennai, Tamil Nadu, India
| | - Saman Warnakulasuriya
- Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
- WHO Collaborating Centre for Oral Cancer, London, UK
| |
Collapse
|
2
|
Gao HQ, Bu XM, Jiang W, Wan YZ, Song W. Compound Taxus exerts marked anti-tumor activity and radiosensitization effect on hepatocellular carcinoma cells. Heliyon 2024; 10:e27345. [PMID: 38495161 PMCID: PMC10940940 DOI: 10.1016/j.heliyon.2024.e27345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 02/24/2024] [Accepted: 02/28/2024] [Indexed: 03/19/2024] Open
Abstract
Background Compound Taxus capsule, as an antineoplastic Chinese patent drug, has been increasingly applied as an adjunctive treatment for the management of non-small-cell lung cancer (NSCLC) and some other malignancies, but research about its antitumor activity and radiosensitization effect on hepatocellular carcinoma (HCC) cells is very rare. Purpose To investigate the antitumor activity and radiosensitization effect of Compound Taxus on HCC cells and to preliminarily explore the possible molecule mechanisms involved. Methods Cell viability, cell cycle distribution, apoptosis, DNA damage repair and protein expression levels were detected by CCK-8 assay, flow cytometry, immunofluorescence staining, western blotting analysis and immunohistochemical staining, respectively. The migration and invasion activities and vasculogenic mimicry (VM) formation and angiogenesis were evaluated by tube formation and VM formation assay. Radiation survival curves were obtained from the colony formation assay in human HCC cell lines, Smmc7721 and Bel7402 cells, pretreated with or without Compound Taxus before receiving X-ray irradiation. A Bel7402 tumor-bearing mouse model was established and the radiosensitization effect of Compound Taxus in vivo was evaluated by analyzing tumor volume and tumor weight in different groups receiving different treatments. Results Compound Taxus decreased viability, induced G2/M arrest, promoted apoptosis, suppressed migration and invasion, and inhibited VM formation and angiogenesis in Smmc7721 and Bel7402 cells. Furthermore, Compound Taxus inhibited irradiation-induced DNA damage repair, enhanced the radiosensitivity of Smmc7721 and Bel7402 cells and improved the anti-tumor therapeutic efficacy of irradiation in Bel7402 tumor-bearing mice. Radiotherapy in combination with Compound Taxus showed the best tumor inhibition compared to that of Compound Taxus alone or irradiation alone. In addition, Compound Taxus significantly down-regulated NF-κB p65, p-NF-κB p65 and Bcl-2, and up-regulated Bax in vitro and in vivo, yet NF-κB p65 overexpression reversed the proapoptotic effect of Taxus on HCC cells, indicating that the NF-κB signaling pathway might be an important signal mediator in the Compound-Taxus-modulated biological responses. Conclusion Our findings suggest that Compound Taxus shows marked antitumor activity and significant radiosensitization effect on HCC cells, making it possible for Compound Taxus to become a promising auxiliary modality for HCC management and a potential radiosensitizer of HCC in the future.
Collapse
Affiliation(s)
- Hui-quan Gao
- Department of Radiotherapy, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiang-mao Bu
- Clinical Laboratory, Qingdao Women and Children's Hospital, Qingdao Women and Children's Hospital Affiliated to Qingdao University, Qingdao, China
| | - Wei Jiang
- Department of Radiotherapy, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Yan-zhen Wan
- Clinical Laboratory, Qingdao Women and Children's Hospital, Qingdao Women and Children's Hospital Affiliated to Qingdao University, Qingdao, China
| | - Wei Song
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
3
|
Boretti A. Evidence for the use of curcumin in radioprotection and radiosensitization. Phytother Res 2024; 38:464-469. [PMID: 36897074 DOI: 10.1002/ptr.7803] [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: 01/11/2023] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/11/2023]
Abstract
Curcumin has antineoplastic properties and is considered a chemotherapeutic and chemopreventive agent. Curcumin may be associated with radiation therapy (RT) as a radiosensitizer for cancer cells and a radioprotector for normal cells. In principle, it may result in a reduction of RT dosage for the same therapeutic effect on cancer cells, and further reduced damage to normal cells. Though the overall level of evidence is modest, limited to in vivo and in vitro experiences and practically no clinical trials, as the risks of adverse effects are extremely low, it is reasonable to promote the general supplementation with curcumin during RT targeting the reduction of side effects through anti-inflammatory mechanisms.
Collapse
|
4
|
Budi HS, Farhood B. Tumor microenvironment remodeling in oral cancer: Application of plant derived-natural products and nanomaterials. ENVIRONMENTAL RESEARCH 2023; 233:116432. [PMID: 37331557 DOI: 10.1016/j.envres.2023.116432] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/08/2023] [Accepted: 06/15/2023] [Indexed: 06/20/2023]
Abstract
Oral cancers consist of squamous cell carcinoma (SCC) and other malignancies in the mouth with varying degrees of invasion and differentiation. For many years, different modalities such as surgery, radiation therapy, and classical chemotherapy drugs have been used to control the growth of oral tumors. Nowadays, studies have confirmed the remarkable effects of the tumor microenvironment (TME) on the development, invasion, and therapeutic resistance of tumors like oral cancers. Therefore, several studies have been conducted to modulate the TME in various types of tumors in favor of cancer suppression. Natural products are intriguing agents for targeting cancers and TME. Flavonoids, non-flavonoid herbal-derived molecules, and other natural products have shown promising effects on cancers and TME. These agents, such as curcumin, resveratrol, melatonin, quercetin and naringinin have demonstrated potency in suppressing oral cancers. In this paper, we will review and discuss about the potential efficacy of natural adjuvants on oral cancer cells. Furthermore, we will review the possible therapeutic effects of these agents on the TME and oral cancer cells. Moreover, the potential of nanoparticles-loaded natural products for targeting oral cancers and TME will be reviewed. The potentials, gaps, and future perspectives for targeting TME by nanoparticles-loaded natural products will also be discussed.
Collapse
Affiliation(s)
- Hendrik Setia Budi
- Department of Oral Biology, Dental Pharmacology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
5
|
Lee SC, Shen CY, Wang WH, Lee YP, Liang KW, Chou YH, Tyan YS, Hwang JJ. Synergistic Effect of Ginsenoside Rh2 Combines with Ionizing Radiation on CT26/ luc Colon Carcinoma Cells and Tumor-Bearing Animal Model. Pharmaceuticals (Basel) 2023; 16:1188. [PMID: 37764996 PMCID: PMC10535731 DOI: 10.3390/ph16091188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/01/2023] [Accepted: 08/11/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND The local tumor control rate of colon cancer by radiotherapy is unsatisfactory due to recurrence and radioresistance. Ginsenoside Rh2 (Rh2), a panoxadiol saponin, possesses various antitumor effects. METHODS CT26/luc murine colon carcinoma cells and a CT26/luc tumor-bearing animal model were used to investigate the therapeutic efficacy of Rh2 combined with ionizing radiation and the underlying mechanisms. RESULTS Rh2 caused cell cycle arrest at the G1 phase in CT26/luc cells; however, when combined with ionizing radiation, the cells were arrested at the G2/M phase. Rh2 was found to suppress the activity of NF-κB induced by radiation by inhibiting the MAPK pathway, consequently affecting the expression of effector proteins. In an in vivo study, the combination treatment significantly increased tumor growth delay time and overall survival. Furthermore, the combination treatment significantly reduced NF-κB and NF-κB-related effector proteins, along with PD-1 receptor expression. Additionally, Rh2 administration led to increased levels of interleukin-12, -18, and interferon-γ in the mice's sera. Importantly, biochemical analysis revealed no toxicities associated with Rh2 alone or combined with radiation. CONCLUSIONS The combination of Rh2 with radiation may have potential as an alternative to improve the therapeutic efficacy of colorectal cancer.
Collapse
Affiliation(s)
- Shan-Chih Lee
- Department of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung 40201, Taiwan; (S.-C.L.); (Y.-H.C.)
| | - Chao-Yu Shen
- Department of Medical Imaging, Chung Shan Medical University Hospital, School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (C.-Y.S.); (K.-W.L.)
| | - Wei-Hsun Wang
- Department of Orthopedic Surgery, Changhua Christian Hospital, Changhua 50044, Taiwan;
| | - Yen-Po Lee
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei Branch, Hsinchu City 30010, Taiwan;
| | - Keng-Wei Liang
- Department of Medical Imaging, Chung Shan Medical University Hospital, School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (C.-Y.S.); (K.-W.L.)
| | - Ying-Hsiang Chou
- Department of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung 40201, Taiwan; (S.-C.L.); (Y.-H.C.)
- Department of Radiation Oncology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Yeu-Sheng Tyan
- Department of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung 40201, Taiwan; (S.-C.L.); (Y.-H.C.)
- Department of Medical Imaging, Chung Shan Medical University Hospital, School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (C.-Y.S.); (K.-W.L.)
| | - Jeng-Jong Hwang
- Department of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung 40201, Taiwan; (S.-C.L.); (Y.-H.C.)
- Department of Medical Imaging, Chung Shan Medical University Hospital, School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (C.-Y.S.); (K.-W.L.)
| |
Collapse
|
6
|
Jiang Y, Zhang J, Shi C, Li X, Jiang Y, Mao R. NF- κB: a mediator that promotes or inhibits angiogenesis in human diseases? Expert Rev Mol Med 2023; 25:e25. [PMID: 37503730 DOI: 10.1017/erm.2023.20] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The nuclear factor of κ-light chain of enhancer-activated B cells (NF-κB) signaling pathway, which is conserved in invertebrates, plays a significant role in human diseases such as inflammation-related diseases and carcinogenesis. Angiogenesis refers to the growth of new capillary vessels derived from already existing capillaries and postcapillary venules. Maintaining normal angiogenesis and effective vascular function is a prerequisite for the stability of organ tissue function, and abnormal angiogenesis often leads to a variety of diseases. It has been suggested that NK-κB signalling molecules under pathological conditions play an important role in vascular differentiation, proliferation, apoptosis and tumourigenesis by regulating the transcription of multiple target genes. Many NF-κB inhibitors are being tested in clinical trials for cancer treatment and their effect on angiogenesis is summarised. In this review, we will summarise the role of NF-κB signalling in various neovascular diseases, especially in tumours, and explore whether NF-κB can be used as an attack target or activation medium to inhibit tumour angiogenesis.
Collapse
Affiliation(s)
- Yijing Jiang
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| | - Jie Zhang
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, 30Tongyang North Road, Pingchao Town, Nantong 226361, Jiangsu, People's Republic of China
| | - Conglin Shi
- Department of Pathogenic Biology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| | - Xingjuan Li
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| | - Yongying Jiang
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| | - Renfang Mao
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| |
Collapse
|
7
|
Mukherjee D, Krishnan A. Therapeutic potential of curcumin and its nanoformulations for treating oral cancer. World J Methodol 2023; 13:29-45. [PMID: 37456978 PMCID: PMC10348080 DOI: 10.5662/wjm.v13.i3.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/14/2023] [Accepted: 04/14/2023] [Indexed: 06/14/2023] Open
Abstract
The global incidence of oral cancer has steadily increased in recent years and is associated with high morbidity and mortality. Oral cancer is the most common cancer in the head and neck region, and is predominantly of epithelial origin (i.e. squamous cell carcinoma). Oral cancer treatment modalities mainly include surgery with or without radiotherapy and chemotherapy. Though proven effective, chemotherapy has significant adverse effects with possibilities of tumor resistance to anticancer drugs and recurrence. Thus, there is an imperative need to identify suitable anticancer therapies that are highly precise with minimal side effects and to make oral cancer treatment effective and safer. Among the available adjuvant therapies is curcumin, a plant polyphenol isolated from the rhizome of the turmeric plant Curcuma longa. Curcumin has been demonstrated to have anti-infectious, antioxidant, anti-inflammatory, and anticarcinogenic properties. Curcumin has poor bioavailability, which has been overcome by its various analogues and nanoformulations, such as nanoparticles, liposome complexes, micelles, and phospholipid complexes. Studies have shown that the anticancer effects of curcumin are mediated by its action on multiple molecular targets, including activator protein 1, protein kinase B (Akt), nuclear factor κ-light-chain-enhancer of activated B cells, mitogen-activated protein kinase, epidermal growth factor receptor (EGFR) expression, and EGFR downstream signaling pathways. These targets play important roles in oral cancer pathogenesis, thereby making curcumin a promising adjuvant treatment modality. This review aims to summarize the different novel formulations of curcumin and their role in the treatment of oral cancer.
Collapse
Affiliation(s)
- Diptasree Mukherjee
- Department of Biochemistry, All India Institute of Medical Sciences, Bhubaneswar 751019, Odisha, India
- Department of Medicine, Apex Institute of Medical Science, Kolkata 700075, West Bengal, India
| | - Arunkumar Krishnan
- Department of Medicine Section of Gastroenterology and Hepatology, West Virginia University School of Medicine, Morgantown, WV 26505, United States
| |
Collapse
|
8
|
Li B, Lv Y, Zhang C, Xiang C. lncRNA HOXA11-AS maintains the stemness of oral squamous cell carcinoma stem cells and reduces the radiosensitivity by targeting miR-518a-3p/PDK1. J Oral Pathol Med 2023; 52:216-225. [PMID: 36661031 DOI: 10.1111/jop.13405] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/26/2022] [Accepted: 01/11/2023] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Oral squamous cell carcinoma (OSCC) is the most prevailing oral malignancy. The lncRNA HOXA11-AS shows prominent roles in OSCC. This study explored the effects of lncRNA HOXA11-AS on regulating OSCC stem cell stemness and radiosensitivity by targeting miR-518a-3p/PDK1. METHODS Human OSCC cell lines SCC9 and SCC15 were selected. CD133+ cancer stem cells (CSCs) were sorted by immunomagnetic beads. CD133 expression in cells and HOXA11-AS expression in SCC9, SCC15, and CD133+ SCC9, CD133+ SCC15 cells were assessed by flow cytometry and RT-qPCR. HOXA11-AS was silenced/overexpressed in SCC9, SCC15, CD133+ SCC9, and CD133+ SCC15 cells. Cell proliferation, radiosensitivity, invasion, and stem cell sphere formation ability were examined by CCK-8, colony formation, Transwell, and stem cell sphere formation. The levels of stemness-related genes (Oct4, Nanog, Sox2), miR-518a-3p, epithelial-mesenchymal transition (EMT)-related proteins (E-cadherin, Vimentin, N-cadherin), and PDK1 were assessed by RT-qPCR and Western blot assay. RESULTS HOXA11-AS was up-regulated in SCC9, SCC15, CD133+ SCC9, and CD133+ SCC15 cells. HOXA11-AS silencing inhibited OSCC proliferation and invasion and enhanced radiosensitivity. HOXA11-AS maintained CSC stemness in OSCC. HOXA11-AS silencing reduced CD133+ SCC9 and CD133+ SCC15 stem cell sphere formation ability, reduced stem cell stemness-related gene levels, and inhibited EMT. HOXA11-AS regulated OSCC stem cell stemness and radiosensitivity by targeting miR-518a-3p. PDK1 overexpression annulled the regulatory effects of HOXA11-AS silencing on OSCC cell stem cell stemness and radiosensitivity. CONCLUSION In vitro lncRNA HOXA11-AS silencing inhibited OSCC stem cell stemness by targeting the miR-518a-3p/PDK1 axis, thus enhancing OSCC cell radiosensitivity.
Collapse
Affiliation(s)
- Baojun Li
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yuanjing Lv
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Cui Zhang
- Department of Medical Ultrasound, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Cheng Xiang
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
9
|
Gao R, Gu Y, Yang Y, He Y, Huang W, Sun T, Tang Z, Wang Y, Yang W. Robust radiosensitization of hemoglobin-curcumin nanoparticles suppresses hypoxic hepatocellular carcinoma. J Nanobiotechnology 2022; 20:115. [PMID: 35248069 PMCID: PMC8898525 DOI: 10.1186/s12951-022-01316-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 02/17/2022] [Indexed: 12/24/2022] Open
Abstract
Background Radioresistance inducing by hypoxic microenvironment of hepatocellular carcinoma is a major obstacle to clinical radiotherapy. Advanced nanomedicine provides an alternative to alleviate the hypoxia extent of solid tumor, even to achieve effective synergistic treatment when combined with chemotherapy or radiotherapy. Results Herein, we developed a self-assembled nanoparticle based on hemoglobin and curcumin for photoacoustic imaging and radiotherapy of hypoxic hepatocellular carcinoma. The fabricated nanoparticles inhibited hepatoma migration and vascular mimics, and enhanced the radiosensitivity of hypoxic hepatoma cells in vitro via repressing cell proliferation and DNA damage repair, as well as inducing apoptosis. Benefit from oxygen-carrying hemoglobin combined with polyphenolic curcumin, the nanoparticles also effectively enhanced the photoacoustic contrast and the efficacy of radiotherapy for hepatocellular carcinoma in vivo. Conclusions Together, the current study offered a radiosensitization platform for optimizing the efficacy of nanomedicines on hypoxic radioresistant tumor. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01316-w.
Collapse
|
10
|
Du C, Tian Y, Duan W, Chen X, Ren W, Deng Q. Curcumin Enhances the Radiosensitivity of Human Urethral Scar Fibroblasts by Apoptosis, Cell Cycle Arrest and Downregulation of Smad4 via Autophagy. Radiat Res 2021; 195:452-462. [PMID: 33755170 DOI: 10.1667/rade-20-00239.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 02/15/2021] [Indexed: 11/03/2022]
Abstract
The goals of this study were to determine whether curcumin can radiosensitize human urethral scar fibroblasts (HUSFs) and inhibit the synthesis of collagen, and to explore the molecular mechanism. Here, HUSFs were established and cultured in vitro and cell counting kit-8 (CCK-8) experiment and plate clone formation assay were performed to determine the appropriate concentration of curcumin and radiation dose. The radiosensitization of curcumin was confirmed by plate clone formation assay. Cell cycle distribution was determined by flow cytometry and apoptosis rate by TdT-mediated dUTP nick-end labeling (TUNEL). Western blot was used to detect the levels of collagen I, collagen III, Smad2, Smad3, Smad4, transforming growth factor-β (TGF-β1), Beclin1 and microtubule-associated protein light chain 3 (LC3), as a means of determining the mechanism. Our findings showed that curcumin enhanced radiosensitivity of HUSFs in vitro (sensitization enhancement ratio = 2.030). Furthermore, curcumin and radiation treatments promoted the apoptosis of HUSFs and blocked the cells in G2/M phase. In addition, curcumin combined with radiation inhibited the synthesis of collagen I and collagen III through Smad4 pathway, with possible involvement of autophagy. These results suggest that curcumin could be a radiosensitizer of HUSFs, inhibit the proliferation of HUSFs and suppress fibrosis by downregulation of Smad4 via autophagy.
Collapse
Affiliation(s)
- Chun Du
- Department of a Urology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | | | - Wanli Duan
- Department of a Urology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Xin Chen
- Department of Radiotherapy, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Wei Ren
- Department of a Urology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Qian Deng
- Department of a Urology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| |
Collapse
|
11
|
Galeaz C, Totis C, Bisio A. Radiation Resistance: A Matter of Transcription Factors. Front Oncol 2021; 11:662840. [PMID: 34141616 PMCID: PMC8204019 DOI: 10.3389/fonc.2021.662840] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/12/2021] [Indexed: 12/14/2022] Open
Abstract
Currently, radiation therapy is one of the standard therapies for cancer treatment. Since the first applications, the field of radiotherapy has constantly improved, both in imaging technologies and from a dose-painting point of view. Despite this, the mechanisms of resistance are still a great problem to overcome. Therefore, a more detailed understanding of these molecular mechanisms will allow researchers to develop new therapeutic strategies to eradicate cancer effectively. This review focuses on different transcription factors activated in response to radiotherapy and, unfortunately, involved in cancer cells’ survival. In particular, ionizing radiations trigger the activation of the immune modulators STAT3 and NF-κB, which contribute to the development of radiation resistance through the up-regulation of anti-apoptotic genes, the promotion of proliferation, the alteration of the cell cycle, and the induction of genes responsible for the Epithelial to Mesenchymal Transition (EMT). Moreover, the ROS-dependent damaging effects of radiation therapy are hampered by the induction of antioxidant enzymes by NF-κB, NRF2, and HIF-1. This protective process results in a reduced effectiveness of the treatment, whose mechanism of action relies mainly on the generation of free oxygen radicals. Furthermore, the previously mentioned transcription factors are also involved in the maintenance of stemness in Cancer Stem Cells (CSCs), a subset of tumor cells that are intrinsically resistant to anti-cancer therapies. Therefore, combining standard treatments with new therapeutic strategies targeted against these transcription factors may be a promising opportunity to avoid resistance and thus tumor relapse.
Collapse
Affiliation(s)
- Chiara Galeaz
- Laboratory of Radiobiology, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Cristina Totis
- Laboratory of Radiobiology, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Alessandra Bisio
- Laboratory of Radiobiology, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| |
Collapse
|
12
|
Effects of orlistat combined with enzalutamide and castration through inhibition of fatty acid synthase in a PC3 tumor-bearing mouse model. Biosci Rep 2021; 41:228631. [PMID: 33974005 PMCID: PMC8164108 DOI: 10.1042/bsr20204203] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/01/2021] [Accepted: 05/11/2021] [Indexed: 01/18/2023] Open
Abstract
Androgen deprivation therapy (ADT) is one of the typical treatments used for patients with prostate cancer (PCa). ADT, however, may fail when PCa develops castration-resistance. Fatty acid synthase (FASN), a critical enzyme involved in fatty acid synthesis, is found to be up-regulated in PCa. Since enzalutamide and ADT are frequently used for the treatment of PCa, the present study aimed to unravel the underlying mechanism of combination of orlistat, an FASN inhibitor, and enzalutamide using PC3 cell line; and orlistat and castration in PC3 tumor-bearing animal model. Cytotoxicity was determined by AlamarBlue assay. Drug effects on the cell cycle and protein expressions were assayed by the flow cytometry and Western blot. Electromobility shift assay was used to evaluate the NF-κB activity. The tumor growth delay, expressions of the signaling-related proteins, and histopathology post treatments of orlistat and castration were evaluated in PC3 tumor-bearing mouse model. The results showed that orlistat arrested the PC3 cells at the G1 phase of the cell cycle and enhanced the cytotoxic effects of enzalutamide synergistically. Pretreatment with orlistat combined with castration inhibited the tumor growth significantly compared with those of castration and orlistat treatments alone in PC3 tumor-bearing mice. Combination treatment reduced both FASN and NF-κB activities and their downstream effector proteins. The present study demonstrated the synergistic effects of orlistat combined with enzalutamide in vitro and castration in vivo on human PCa.
Collapse
|
13
|
Liu T, Long T, Li H. Curcumin suppresses the proliferation of oral squamous cell carcinoma through a specificity protein 1/nuclear factor-κB-dependent pathway. Exp Ther Med 2021; 21:202. [PMID: 33500696 DOI: 10.3892/etm.2021.9635] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 07/17/2020] [Indexed: 12/17/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common cancer of the oral cavity. Curcumin (Cur), a naturally derived compound, is reported to have broad-spectrum anticancer activity and is considered as an effective nuclear factor-κB (NF-κB) inhibitor. The present study aimed to clarify the detailed molecular mechanism though which Cur regulates NF-κB pathway activity in OSCC. The viability of HSC3 and CAL33 cells following treatment with Cur was determined using a Cell Counting Kit-8 assay. The protein and mRNA expression of specificity protein 1 (Sp1), p65 and heat shock factor 1 (HSF1) was determined by western blotting and reverse transcription-quantitative PCR analysis, respectively. The NF-κB activity was measured by Dual-Luciferase reporter assay. Short hairpin RNA targeting Sp1 or control RNA was transfected into HSC3 cells using X-treme GENE HP DNA Transfection System. Colony formation assays were performed using crystal violet staining. The results demonstrated that Cur significantly inhibited the viability and colony formation ability of HSC3 and CAL33 cells. In addition, Cur decreased the expression of Sp1, p65 and HSF1 by suppressing their transcription levels. Cur decreased NF-κB activity in OSCC cells, and Sp1 downregulation enhanced the effect of Cur. The findings from the present study suggested that Cur may inhibit the proliferation of OSCC cells via a Sp1/NF-κB-dependent mechanism.
Collapse
Affiliation(s)
- Tian Liu
- Department of Stomatology, The Central Hospital of Wuhan, Wuhan, Hubei 430000, P.R. China
| | - Tian Long
- Department of Stomatology, The Central Hospital of Wuhan, Wuhan, Hubei 430000, P.R. China
| | - Haosen Li
- Department of Stomatology Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| |
Collapse
|
14
|
DNA-BINDING and DNA-protecting activities of small natural organic molecules and food extracts. Chem Biol Interact 2020; 323:109030. [PMID: 32205154 DOI: 10.1016/j.cbi.2020.109030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/21/2020] [Accepted: 02/26/2020] [Indexed: 01/07/2023]
Abstract
The review summarizes literature data on the DNA-binding, DNA-protecting and DNA-damaging activities of a range of natural human endogenous and exogenous compounds. Small natural organic molecules bind DNA in a site-specific mode, by arranging tight touch with the structure of the major and minor grooves, as well as individual bases in the local duplex DNA. Polyphenols are the best-studied exogenous compounds from this point of view. Many of them demonstrate hormetic effects, producing both beneficial and damaging effects. An attempt to establish the dependence of DNA damage or DNA protection on the concentration of the compound turned out to be successful for some polyphenols, daidzein, genistein and resveratrol, which were DNA protecting in low concentrations and DNA damaging in high concentrations. There was no evident dependence on concentration for quercetin and kaempferol. Probably, the DNA-protecting effect is associated with the affinity to DNA. Caffeine and theophylline are DNA binders; at the same time, they favor DNA repair. Although most alkaloids damage DNA, berberine can protect DNA against damage. Among the endogenous compounds, hormones belonging to the amine class, thyroid and steroid hormones appear to bind DNA and produce some DNA damage. Thus, natural compounds continue to reveal beneficial or adverse effects on genome integrity and provide a promising source of therapeutic activities.
Collapse
|
15
|
Sak K. Radiosensitizing Potential of Curcumin in Different Cancer Models. Nutr Cancer 2019; 72:1276-1289. [PMID: 31648572 DOI: 10.1080/01635581.2019.1681480] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 10/11/2019] [Accepted: 10/12/2019] [Indexed: 12/12/2022]
Abstract
Over the past decades, studies of phytochemicals as modifiers of radiotherapeutic efficacy have become increasingly popular to improve the treatment outcome of human malignancies. In the current comprehensive review article, radiosensitizing effects of curcumin, a yellow-colored polyphenolic constituent of turmeric, in various preclinical cancer models, both In Vitro and In Vivo, are presented. Attenuation of radioadaptation and augmentation of irradiation-induced cancer cell killing are achieved through multifaceted action of curcumin on suppression of prosurvival and antiapoptotic factors. Most importantly, curcumin can block radiation-triggered NF-κB signaling pathway and downregulate downstream effector proteins, thereby conferring potentiation of radioresponses. Based on the elucidated molecular mechanisms but also due to its safety profile and low cost, curcumin might be considered a promising adjuvant agent to enhance radiotherapeutic efficacy in the treatment of various cancer types formed in different human organ systems. Further efforts to translate the current preclinical knowledge to the real application of curcumin in combinatorial radiotherapeutic strategies in clinical settings are necessary.AbbreviationsAKTprotein kinase BARMSalveolar rhabdomyosarcomaATMataxia telangiectasia mutatedBaxBcl-2-associated X proteinBcl-2B-cell lymphoma 2CDC2cyclin-dependent kinase 2Bcl-xLB-cell lymphoma-extra largec-FLIPcellular FLICE-like inhibitory proteinCDDPcisplatinCOX-2cyclooxygenase-2cyt ccytochrome cDNA-PKcsDNA-dependent protein kinaseEGFRepidermal growth factor receptorEMTepithelial-mesenchymal transitionERKextracellular signal-regulated kinaseESEwing`s sarcomaETS2erythroblastosis virus transcription factor 2GBMglioblastoma multiformeHCChepatocellular carcinomaHNSCChead and neck squamous cell carcinomaIAPinhibitor of apoptosis proteinIκBαinhibitor of κB alphaIKKinhibitor of κB kinaseIRionizing radiationlncRNAlong non-coding RNAlucluciferaseMcl-1myeloid cell leukemia-1MDR1multidrug resistance protein 1miRmicroRNAMMP-9matrix metalloproteinase-9mTORmammalian target of rapamycinNBneuroblastomaNF-κBnuclear factor-κBNPCnasopharyngeal carcinomaNSCLCnon-small cell lung cancerOSCCoral squamous cell carcinomaPARPpoly-(ADP-ribose)-polymerasepH2AXphosphorylated histone 2AX-immunoreactivePI3Kphosphatidylinositol 3-kinasePrp4KPre-mRNA processing factor 4 kinaseRCCrenal cell carcinomaROSreactive oxygen speciesSCCsquamous cell carcinomaSLNsolid lipid nanoparticleSOD2superoxide dismutase 2TERTtelomerase reverse transcriptaseTNF-αtumor necrosis factor-αTxnRd1thioredoxin reductase-1VEGFvascular endothelial growth factorXIAPX-linked inhibitor of apoptosis proteinΔΨmmitochondrial membrane potential.
Collapse
|
16
|
Lee MJ, Tsai YJ, Lin MY, You HL, Kalyanam N, Ho CT, Pan MH. Calebin-A induced death of malignant peripheral nerve sheath tumor cells by activation of histone acetyltransferase. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 57:377-384. [PMID: 30831486 DOI: 10.1016/j.phymed.2019.01.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/02/2019] [Indexed: 05/27/2023]
Abstract
BACKGROUND Neurofibromatosis type 1 (NF1) is one of the most common hereditary neurocutaneous disorders. The malignant peripheral nerve sheath tumor (MPNST), transformed from NF1 related plexiform neurofibroma, is a rapidly growing and highly invasive tumor. No effective chemotherapeutic agent is currently available. Calebin-A is a derivative from turmeric Curcuma longa. Given the anti-inflammatory and anticancer potentials of curcumin, whether Calebin-A also had the tumoricidal effect upon MPNST cells is still elusive. PURPOSE To determine whether Calebin-A has the potential for anti-MPNST effect. METHODS The MTT and FACS analysis of normal Schwann (HSC) and MPNST cells have been employed to determine the tumoricidal effect of Calebin-A. The expression of the signal pathway molecules was assessed by Western blotting. The CHIP with quantitative PCR assay was performed to quantify the promoter DNA binding to acetylated histone 3 (acetyl H3). The enzyme activities of histone acetyltransferase (HAT) and deacetylase (HDAC) have been evaluated by commercial kits. The measurements of tumor size of the xenograft mouse model were also performed. RESULTS Calebin-A inhibited the proliferation of MPNST and primary neurofibroma cells in a dose-dependent manner. The flow cytometry analysis of the MPNST cells after treatment of 25 μm of Calebin-A demonstrated an increase of population in the G0/G1 phase but decrease in G2/M phase. Before treatment, the expression of Axl, Tyro3, and acetyl H3 was significantly higher in MPNST cells when compared to HSC. The expression of phosphorylated-AKT, -ERK1/2, survivin, hTERT, and acetyl H3 proteins were reduced after treatment. The CHIP assay shows the promoter DNA copies of survivin (BRIC5) and hTERT genes are significantly reduced post-treatment. The enzyme activity of HAT was significantly reduced, but not that of HDAC. Two HAT inhibitors, epigallocatechin-3-gallate (EGCG) and anacardic acid (AA) have also demonstrated a significant inhibitory effect on MPNST cells. Finally, the measurements of tumor size showed a significant reduction of the xenograft tumors after treatment of Calebin-A. CONCLUSION Both in vitro and in vivo studies showed Calebin-A could inhibit the proliferation of MPNST with suppression of survivin and hTERT. The reduced expression of these two factors might be through the epigenetic histone modification resulting from the decreased activity of HAT.
Collapse
Affiliation(s)
- Ming-Jen Lee
- Department of Neurology, National Taiwan University Hospital, Taipei 10012, Taiwan; Department of Medical Genetics, National Taiwan University Hospital, Taipei 10012, Taiwan.
| | - Yi-Jane Tsai
- Department of Neurology, National Taiwan University Hospital, Taipei 10012, Taiwan
| | - May-Yao Lin
- Department of Neurology, National Taiwan University Hospital, Taipei 10012, Taiwan
| | - Huey-Ling You
- Department of Neurology, National Taiwan University Hospital, Taipei 10012, Taiwan
| | | | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901, USA
| | - Min-Hsiung Pan
- Institute of Food Science and Technology, National Taiwan University, Taipei 10617, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan.
| |
Collapse
|
17
|
Mortezaee K, Najafi M, Farhood B, Ahmadi A, Shabeeb D, Musa AE. NF‐κB targeting for overcoming tumor resistance and normal tissues toxicity. J Cell Physiol 2019; 234:17187-17204. [DOI: 10.1002/jcp.28504] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/22/2019] [Accepted: 03/05/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy School of Medicine, Kurdistan University of Medical Sciences Sanandaj Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department School of Paramedical Sciences, Kermanshah University of Medical Sciences Kermanshah Iran
| | - Bagher Farhood
- Departments of Medical Physics and Radiology Faculty of Paramedical Sciences, Kashan University of Medical Sciences Kashan Iran
| | - Amirhossein Ahmadi
- Pharmaceutical Sciences Research Center Faculty of Pharmacy, Mazandaran University of Medical Sciences Sari Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology College of Medicine, University of Misan Misan Iraq
| | - Ahmed E. Musa
- Department of Medical Physics Tehran University of Medical Sciences (International Campus) Tehran Iran
| |
Collapse
|
18
|
Chen WT, Hsu FT, Liu YC, Chen CH, Hsu LC, Lin SS. Fluoxetine Induces Apoptosis through Extrinsic/Intrinsic Pathways and Inhibits ERK/NF-κB-Modulated Anti-Apoptotic and Invasive Potential in Hepatocellular Carcinoma Cells In Vitro. Int J Mol Sci 2019; 20:ijms20030757. [PMID: 30754643 PMCID: PMC6386946 DOI: 10.3390/ijms20030757] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 01/09/2019] [Accepted: 01/30/2019] [Indexed: 01/06/2023] Open
Abstract
The aim of the present study was to verify the effects of fluoxetine on dysregulation of apoptosis and invasive potential in human hepatocellular carcinoma (HCC) SK-Hep1 and Hep3B cells. Cells were treated with different concentrations of fluoxetine for different times. MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide) assays were used for testing the effects of fluoxetine on cell viability. The regulation of apoptosis signaling, and anti-apoptotic, proliferation, and metastasis-associated proteins after fluoxetine treatment were assayed by flow cytometry and Western blotting assay. The detection of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation after fluoxetine treatment was performed by NF-κB reporter gene assay. The results demonstrated that fluoxetine significantly reduced cell viability, cell migration/invasion, NF-κB, extracellular signal-regulated kinases (ERK) activation, and expression of anti-apoptotic (Cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein (C-FLIP), Myeloid cell leukemia-1 (MCL-1), X-Linked inhibitor of apoptosis protein (XAIP), and Survivin), proliferation (Cyclin-D1), angiogenesis (vascular endothelial growth factor (VEGF)), and metastasis-associated proteins (matrix metalloproteinase-9 (MMP-9)). Fluoxetine also significantly induced apoptosis, unregulated extrinsic (activation of first apoptosis signal protein and ligand (Fas/FasL), and caspase-8) and intrinsic (loss of mitochondrial membrane potential (ΔΨm) pathways and increased Bcl-2 homologous antagonist killer (BAK) apoptosis signaling. Taken together, these results demonstrated that fluoxetine induced apoptosis through extrinsic/intrinsic pathways and diminished ERK/NF-κB-modulated anti-apoptotic and invasive potential in HCC cells in vitro.
Collapse
Affiliation(s)
- Wei-Ting Chen
- Department of Medical Imaging and Radiological Sciences, Central Taiwan University of Science and Technology, Taichung 406, Taiwan.
- Department of Psychiatry, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung 813, Taiwan.
- Department of Physical Therapy, Shu-Zen Junior College of Medicine and Management, Kaohsiung 821, Taiwan.
| | - Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan.
| | - Yu-Chang Liu
- Department of Medical Imaging and Radiological Sciences, Central Taiwan University of Science and Technology, Taichung 406, Taiwan.
- Department of Radiation Oncology, Chang Bing Show Chwan Memorial Hospital, Changhua 505, Taiwan.
- Department of Radiation Oncology, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
| | - Cheng-Hsien Chen
- Department of Surgery, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
| | - Li-Cho Hsu
- Division of Endocrinology and Metabolism, Department of Medicine, National Yang-Ming University Hospital, Yilan 260, Taiwan.
| | - Song-Shei Lin
- Department of Medical Imaging and Radiological Sciences, Central Taiwan University of Science and Technology, Taichung 406, Taiwan.
| |
Collapse
|
19
|
Farhood B, Mortezaee K, Goradel NH, Khanlarkhani N, Salehi E, Nashtaei MS, Najafi M, Sahebkar A. Curcumin as an anti-inflammatory agent: Implications to radiotherapy and chemotherapy. J Cell Physiol 2018; 234:5728-5740. [PMID: 30317564 DOI: 10.1002/jcp.27442] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 08/27/2018] [Indexed: 12/21/2022]
Abstract
Cancer is the second cause of death worldwide. Chemotherapy and radiotherapy are the most common modalities for the treatment of cancer. Experimental studies have shown that inflammation plays a central role in tumor resistance and the incidence of several side effects following both chemotherapy and radiotherapy. Inflammation resulting from radiotherapy and chemotherapy is responsible for adverse events such as dermatitis, mucositis, pneumonitis, fibrosis, and bone marrow toxicity. Chronic inflammation may also lead to the development of second cancer during years after treatment. A number of anti-inflammatory drugs such as nonsteroidal anti-inflammatory agents have been proposed to alleviate chronic inflammatory reactions after radiotherapy or chemotherapy. Curcumin is a well-documented herbal anti-inflammatory agents. Studies have proposed that curcumin can help management of inflammation during and after radiotherapy and chemotherapy. Curcumin targets various inflammatory mediators such as cyclooxygenase-2, inducible nitric oxide synthase, and nuclear factor κB (NF-κB), thereby attenuating the release of proinflammatory and profibrotic cytokines, and suppressing chronic production of free radicals, which culminates in the amelioration of tissue toxicity. Through modulation of NF-κB and its downstream signaling cascade, curcumin can also reduce angiogenesis, tumor growth, and metastasis. Low toxicity of curcumin is linked to its cytoprotective effects in normal tissues. This protective action along with the capacity of this phytochemical to sensitize tumor cells to radiotherapy and chemotherapy makes it a potential candidate for use as an adjuvant in cancer therapy. There is also evidence from clinical trials suggesting the potential utility of curcumin for acute inflammatory reactions during radiotherapy such as dermatitis and mucositis.
Collapse
Affiliation(s)
- Bagher Farhood
- Departments of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Nasser Hashemi Goradel
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Neda Khanlarkhani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ensieh Salehi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Shabani Nashtaei
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Najafi
- Department of Radiology and Nuclear Medicine, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Lee MJ, Tseng WS, Lai JCY, Shieh HR, Chi CW, Chen YJ. Differential Pharmacological Activities of Oxygen Numbers on the Sulfoxide Moiety of Wasabi Compound 6-(Methylsulfinyl) Hexyl Isothiocyanate in Human Oral Cancer Cells. Molecules 2018; 23:E2427. [PMID: 30248933 PMCID: PMC6222327 DOI: 10.3390/molecules23102427] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/15/2018] [Accepted: 09/19/2018] [Indexed: 02/07/2023] Open
Abstract
6-(methylsulfinyl) hexyl isothiocyanate (6-MITC) is a naturally occurring compound isolated from Wasabia japonica (wasabi). The synthetic derivatives, 6-(methylsulfenyl) hexyl isothiocyanate (I7447) and 6-(methylsulfonyl) hexyl isothiocyanate (I7557), were derived from 6-MITC with the deletion and addition of oxygen, respectively. We aimed to evaluate the effect of these synthetic compounds on human oral cancer cells, SAS and OECM-1. All three compounds (I7447, 6-MITC, and I7557) inhibited the viability of SAS and OECM-1 cells using MTT assay. Morphological observations showed various proportions of mitotic arrest and apoptosis in cells treated with these compounds. Cell cycle analysis revealed relatively abundant G2/M arrest in 6-MITC and I7557-treated cells, whereas sub-G1 accumulation was found in I7447-treated cells. In using phosphorylated histone H3 as a marker for mitosis, the addition of 6-MITC and I7557 (excluding I7447) could be shown to arrest cells during mitosis. In contrast, I7447 induced more prominent apoptosis than the 6-MITC or I7557 compounds. The down-regulated expression of the phosphorylated form of CHK1 and Cdc25c was noted in 6-MITC and I7557-treated cells. I7557 could sensitize SAS cells to death by radiation. The wasabi compound, 6-MITC, and its chemical derivatives with different numbers of oxygen may have differential pharmacological effects on human oral cancer cells.
Collapse
Affiliation(s)
- Min-Ju Lee
- Taipei First Girls High School, Taipei 10045, Taiwan.
| | - Wen-Ser Tseng
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan.
| | - Jerry Cheng-Yen Lai
- Department of Medical Research, Taitung MacKay Memorial Hospital, Taitung City 95054, Taiwan.
| | - Hui-Ru Shieh
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan.
| | - Chih-Wen Chi
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan.
- Department of Nursing, MacKay Medical College, New Taipei City 25245, Taiwan.
| | - Yu-Jen Chen
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25160, Taiwan.
- Department of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan.
- Department of Radiation Oncology, MacKay Memorial Hospital, Taipei 10449, Taiwan.
| |
Collapse
|
21
|
Zhang X, Peng L, Liu A, Ji J, Zhao L, Zhai G. The enhanced effect of tetrahydrocurcumin on radiosensitivity of glioma cells. J Pharm Pharmacol 2018; 70:749-759. [PMID: 29492979 DOI: 10.1111/jphp.12891] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 01/13/2018] [Indexed: 12/22/2022]
Abstract
Abstract
Objectives
To evaluate the effects of tetrahydrocurcumin (THC) on the radiosensitivity of glioma cells and the possible molecular mechanism.
Methods
MTT assay, colony forming and wound healing assays were performed to detect the proliferation, radiosensitivity and migration of cells with various treatments. Cell apoptosis, cell cycle and GHS level were determined for exploring potent sensitization mechanism of THC. Meanwhile, protein expressions of cyclin D1 and PCNA were also measured. Furthermore, both orthotopic C6 mouse models and C6 subcutaneously grafted mouse models were established to test the tumour inhibitory effects of combined treatment in vivo.
Key findings
Cells treated with combined THC and radiation demonstrated lower cell viability and higher apoptosis rate as compared to radiation group. Moreover, the intracellular GSH was also decreased in the THC co-treated C6 cells. More importantly, combinatorial treatment group significantly induced G0/G1 cell cycle arrest and a decrease in the S phase cell through the down-regulation of cyclin D1 and PCNA. The in-vivo therapeutic efficacy assay indicated that the growth of tumour was greatly inhibited in combinatorial group.
Conclusions
Tetrahydrocurcumin can synergistically enhance the radiosensitivity of glioma cells by inhibiting the expressions of cyclin D1 and PCNA.
Collapse
Affiliation(s)
- Xingzhen Zhang
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Shandong University, Jinan, China
| | - Lei Peng
- Department of Hepatology and Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Anchang Liu
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan, China
| | - Jianbo Ji
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Shandong University, Jinan, China
| | - Lixia Zhao
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan, China
| | - Guangxi Zhai
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Shandong University, Jinan, China
| |
Collapse
|
22
|
Rajagopal C, Lankadasari MB, Aranjani JM, Harikumar KB. Targeting oncogenic transcription factors by polyphenols: A novel approach for cancer therapy. Pharmacol Res 2018; 130:273-291. [PMID: 29305909 DOI: 10.1016/j.phrs.2017.12.034] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/30/2017] [Accepted: 12/31/2017] [Indexed: 02/06/2023]
Abstract
Inflammation is one of the major causative factor of cancer and chronic inflammation is involved in all the major steps of cancer initiation, progression metastasis and drug resistance. The molecular mechanism of inflammation driven cancer is the complex interplay between oncogenic and tumor suppressive transcription factors which include FOXM1, NF-kB, STAT3, Wnt/β- Catenin, HIF-1α, NRF2, androgen and estrogen receptors. Several products derived from natural sources modulate the expression and activity of multiple transcription factors in various tumor models as evident from studies conducted in cell lines, pre-clinical models and clinical samples. Further combination of these natural products along with currently approved cancer therapies added an additional advantage and they considered as promising targets for prevention and treatment of inflammation and cancer. In this review we discuss the application of multi-targeting natural products by analyzing the literature and future directions for their plausible applications in drug discovery.
Collapse
Affiliation(s)
- Chitra Rajagopal
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
| | - Manendra Babu Lankadasari
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
| | - Jesil Mathew Aranjani
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - K B Harikumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India.
| |
Collapse
|
23
|
Liu YC, Wu RH, Wang WS. Regorafenib diminishes the expression and secretion of angiogenesis and metastasis associated proteins and inhibits cell invasion via NF-κB inactivation in SK-Hep1 cells. Oncol Lett 2017; 14:461-467. [PMID: 28693192 DOI: 10.3892/ol.2017.6142] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 01/12/2017] [Indexed: 01/01/2023] Open
Abstract
The aim of the present study was to investigate the effects of regorafenib on the nuclear factor κ-light-chain-enhancer of activated B cells (NF)-κB-modulated expression of angiogenesis- and metastasis-associated proteins and cell invasion in human hepatocellular carcinoma SK-Hep1 cells. The SK-Hep1 cells were treated with different concentrations of NF-κB inhibitor 4-N-[2-(4-phenoxyphenyl) ethyl] quinazoline-4,6-diamine (QNZ) or regorafenib for 24 or 48 h. The effects of QNZ and regorafenib on cell viability, NF-κB activation, expression and secretion levels of angiogenesis- and metastasis-associated proteins and cell invasion were evaluated with MTT assays, western blotting, ELISA, gelatin zymography and cell invasion assays. The results demonstrated that QNZ and regorafenib significantly reduced the expression and secretion levels of the angiogenesis- and metastasis-associated proteins vascular endothelial growth factor, tumor necrosis factor-α, interleukin (IL)-1β, IL-6, matrix metalloproteinase (MMP)-2 and MMP-9, NF-κB activation and cell invasion. In conclusion, the inhibition of NF-κB activation induces anti-angiogenic and antimetastatic effects in SK-Hep1 cells. Regorafenib reduces the level of expression and secretion of angiogenesis- and metastasis-associated proteins and cell invasion through the suppression of NF-κB activation in SK-Hep1 cells.
Collapse
Affiliation(s)
- Yu-Chang Liu
- Department of Radiation Oncology, National Yang-Ming University Hospital, Yilan 260, Taiwan, R.O.C.,Faculty of Medicine, National Yang-Ming University School of Medicine, Taipei 112, Taiwan, R.O.C.,Cancer Medical Care Center, National Yang-Ming University Hospital, Yilan 260, Taiwan, R.O.C.,Department of Radiological Technology, Central Taiwan University of Science and Technology, Taichung 40601, Taiwan, R.O.C
| | - Reng-Hong Wu
- Department of Radiological Technology, Central Taiwan University of Science and Technology, Taichung 40601, Taiwan, R.O.C.,Department of Medical Imaging, Chi Mei Medical Center, Tainan 710, Taiwan, R.O.C
| | - Wei-Shu Wang
- Faculty of Medicine, National Yang-Ming University School of Medicine, Taipei 112, Taiwan, R.O.C.,Cancer Medical Care Center, National Yang-Ming University Hospital, Yilan 260, Taiwan, R.O.C.,Department of Medicine, National Yang-Ming University Hospital, Yilan 260, Taiwan, R.O.C
| |
Collapse
|
24
|
Verma V. Relationship and interactions of curcumin with radiation therapy. World J Clin Oncol 2016; 7:275-283. [PMID: 27298767 PMCID: PMC4896895 DOI: 10.5306/wjco.v7.i3.275] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 02/11/2016] [Accepted: 03/23/2016] [Indexed: 02/06/2023] Open
Abstract
Curcumin is widely reported to have remarkable medicinal - and antineoplastic - properties. This review details curcumin’s relationship with radiotherapy (RT), principally as a radiosensitizer for various malignancies and a radioprotector for normal tissues. First, examples of radiosensitization are provided for various cancers: Pediatric, lymphoma, sarcoma, prostate, gynecologic, pancreas, liver, colorectal, breast, lung, head/neck, and glioma. It is not the purpose of this article to comprehensively review all radiosensitization data; however, high-quality studies are discussed in relationship to currently-controversial RT questions for many cancers, and thus the importance of developing a natural radiosensitizer. Attention is then shifted to radioprotection, for which supporting research is discussed for the following RT toxicities: Dermatitis, pneumonitis, cataractogenesis, neurocognition, myelosuppression, secondary malignancies, and mucositis/enteritis. Though there is fewer data for radioprotection, the overall quality of clinical evidence is higher, and small clinical trials implicating the efficacy of curcumin for RT toxicities (vs placebo/current therapies) are also detailed. Though the overall level of evidence for curcumin as a radiosensitizer and radioprotector is low, it must be recognized that risks of adverse effects are exceedingly low, and clinicians may need to judge the yet-unproven rewards with low toxicity risks.
Collapse
|
25
|
Borges GÁ, Rêgo DF, Assad DX, Coletta RD, De Luca Canto G, Guerra ENS. In vivoandin vitroeffects of curcumin on head and neck carcinoma: a systematic review. J Oral Pathol Med 2016; 46:3-20. [DOI: 10.1111/jop.12455] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Gabriel Álvares Borges
- Laboratory of Oral Histopathology; Health Sciences Faculty; University of Brasília; Brasília Brazil
| | - Daniela Fortunato Rêgo
- Laboratory of Oral Histopathology; Health Sciences Faculty; University of Brasília; Brasília Brazil
| | - Daniele Xavier Assad
- Laboratory of Oral Histopathology; Health Sciences Faculty; University of Brasília; Brasília Brazil
- Hospital Sírio-Libanês; Brasília Brazil
| | - Ricardo D. Coletta
- Department of Oral Diagnosis; School of Dentistry; University of Campinas; Piracicaba São Paulo Brazil
| | - Graziela De Luca Canto
- Department of Dentistry; Brazilian Centre for Evidence-Based Research; Federal University of Santa Catarina; Florianopolis Brazil
- School of Dentistry; Faculty of Medicine and Dentistry; University of Alberta; Edmonton AB Canada
| | | |
Collapse
|
26
|
Wu SH, Hsiao YT, Kuo CL, Yu FS, Hsu SC, Wu PP, Chen JC, Hsia TC, Liu HC, Hsu WH, Chung JG. Bufalin Inhibits NCI-H460 Human Lung Cancer Cell Metastasis In Vitro by Inhibiting MAPKs, MMPs, and NF-κB Pathways. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2016; 43:1247-64. [PMID: 26446205 DOI: 10.1142/s0192415x15500718] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Bufalin, a component of Chan Su (a traditional Chinese medicine), has been known to have antitumor effects for thousands of years. In this study, we investigated its anti-metastasis effects on NCI-H460 lung cancer cells. Under sub-lethal concentrations (from 25 up to 100 nM), bufalin significantly inhibits the invasion and migration nature of NCI-H460 cells that were measured by Matrigel Cell Migration Assay and Invasion System. Bufalin also suppressed the enzymatic activity of matrix metalloproteinase (MMP)-9, which was examined by gelatin zymography methods. Western blotting revealed that bufalin depressed several key metastasis-related proteins, such as NF-κB, MMP-2, MMP-9, protein kinase C (PKC), phosphatidylinositol 3-kinase (PI3-K), phosphorylated Akt, growth factor receptor-bound protein 2 (GRB2), phosphorylated extracellular signal-regulated kinase (ERK), phosphorylated p38, and phosphorylated c-Jun NH2-terminal kinase (JNK). As evidenced by immunostaining and the electrophoretic mobility shift assay (EMSA), bufalin induced not only a decreased cytoplasmic NF-κB production, but also decreased its nuclear translocation. Several metastasis-related genes, including Rho-associated (Rho A), coiled-coil-containing protein kinase 1 (ROCK1), and focal adhesion kinase (FAK), were down-regulated after bufalin treatment. In conclusion, bufalin is effective in inhibiting the metastatic nature of NCI-H460 cells in low, sub-lethal concentrations. Such an effect involves many mechanisms including MMPs, mitogen-activated protein kinases (MAPKs) and NF-κB systems. Bufalin has a potential to evolve into an anti-metastasis drug for human lung cancer in the future.
Collapse
Affiliation(s)
- Shin-Hwar Wu
- Institute of Clinical Medical Science, China Medical University, Taichung 404, Taiwan.,Division of Critical Care Medicine, Department of Medicine, Changhua Christian Hospital, Changhua 505, Taiwan
| | - Yung-Ting Hsiao
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan
| | - Chao-Lin Kuo
- Department of Chinese Medicine Resources, China Medical University, Taichung 404, Taiwan
| | - Fu-Shun Yu
- School of Dentistry, China Medical University, Taichung 404, Taiwan
| | - Shu-Chun Hsu
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan
| | - Ping-Ping Wu
- School of Pharmacy, China Medical University, Taichung 404, Taiwan
| | - Jaw-Chyun Chen
- Department of Medicinal Botany and Health Applications, Da-Yeh University, Changhua 515, Taiwan
| | - Te-Chun Hsia
- Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Hsin-Chung Liu
- Department of Chinese Medicine Resources, China Medical University, Taichung 404, Taiwan
| | - Wu-Huei Hsu
- Department of Internal Medicine, China Medical University, Taichung 404, Taiwan.,Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Jing-Gung Chung
- Department of Chinese Medicine Resources, China Medical University, Taichung 404, Taiwan.,Department of Biotechnology, Asia University, Taichung 413, Taiwan
| |
Collapse
|
27
|
NF-κB acts as a multifunctional modulator in bone invasion by oral squamous cell carcinoma. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/s1348-8643(15)00038-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
28
|
Hsu FT, Chang B, Chen JCH, Chiang IT, Liu YC, Kwang WK, Hwang JJ. Synergistic Effect of Sorafenib and Radiation on Human Oral Carcinoma in vivo. Sci Rep 2015; 5:15391. [PMID: 26487364 PMCID: PMC4613834 DOI: 10.1038/srep15391] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 09/15/2015] [Indexed: 12/26/2022] Open
Abstract
Oral squamous cell carcinoma often causes bone invasion resulting in poor prognosis and affects the quality of life for patients. Herein, we combined radiation with sorafenib, to evaluate the combination effect on tumor progression and bone erosion in an in situ human OSCC-bearing mouse model. Treatment procedure were arranged as following groups: (a) normal (no tumor); (b) control (with tumor); (c) sorafenib (10 mg/kg/day); (d) radiation (single dose of 6 Gy); (e) pretreatment (sorafenib treatment for 3 days prior to radiation), and (f) concurrent treatment (sorafenib and radiation on the same day). The inhibition of tumor growth and expression level of p65 of NF-κB in tumor tissues were the most significant in the pretreatment group. EMSA and Western blot showed that DNA/NF-κB activity and the expressions of NF-κB-associated proteins were down-regulated. Notably, little to no damage in mandibles and zygomas of mice treated with combination of sorafenib and radiation was found by micro-CT imaging. In conclusion, sorafenib combined with radiation suppresses radiation-induced NF-κB activity and its downstream proteins, which contribute to radioresistance and tumorigenesis. Additionally, bone destruction is also diminished, suggesting that combination treatment could be a potential strategy against human OSCC.
Collapse
Affiliation(s)
- Fei-Ting Hsu
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, No. 155, Sec. 2, Li-Nong St., Bei-tou, Taipei 112, Taiwan
| | - Betty Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, No. 155, Sec. 2, Li-Nong St., Bei-tou, Taipei 112, Taiwan
| | - John Chun-Hao Chen
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, No. 155, Sec. 2, Li-Nong St., Bei-tou, Taipei 112, Taiwan.,Department of Radiation Oncology, Mackay Memorial Hospital, No. 45, Minsheng Rd., Tamsui District, New Taipei City 251, Taiwan
| | - I-Tsang Chiang
- Department of Radiation Oncology, National Yang-Ming University Hospital, I-Lan 260, Taiwan
| | - Yu-Chang Liu
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, No. 155, Sec. 2, Li-Nong St., Bei-tou, Taipei 112, Taiwan.,Department of Radiation Oncology, National Yang-Ming University Hospital, I-Lan 260, Taiwan
| | - Wei-Kang Kwang
- Department of Anatomical Pathology, Cheng-Hsin General Hospital, Taipei 112, Taiwan
| | - Jeng-Jong Hwang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, No. 155, Sec. 2, Li-Nong St., Bei-tou, Taipei 112, Taiwan.,Biophotonics and Molecular Imaging Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
29
|
Curcumin Sensitizes Hepatocellular Carcinoma Cells to Radiation via Suppression of Radiation-Induced NF-κB Activity. BIOMED RESEARCH INTERNATIONAL 2015; 2015:363671. [PMID: 26539482 PMCID: PMC4619792 DOI: 10.1155/2015/363671] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 08/11/2015] [Indexed: 01/30/2023]
Abstract
The effects and possible underlying mechanism of curcumin combined with radiation in human hepatocellular carcinoma (HCC) cells in vitro were evaluated. The effects of curcumin, radiation, and combination of both on cell viability, apoptosis, NF-κB activation, and expressions of NF-κB downstream effector proteins were investigated with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), NF-κB reporter gene, mitochondrial membrane potential (MMP), electrophoretic mobility shift (EMSA), and Western blot assays in Huh7-NF-κB-luc2, Hep3B, and HepG2 cells. Effect of I kappa B alpha mutant (IκBαM) vector, a specific inhibitor of NF-κB activation, on radiation-induced loss of MMP was also evaluated. Results show that curcumin not only significantly enhances radiation-induced cytotoxicity and depletion of MMP but inhibits radiation-induced NF-κB activity and expressions of NF-κB downstream proteins in HCC cells. IκBαM vector also shows similar effects. In conclusion, we suggest that curcumin augments anticancer effects of radiation via the suppression of NF-κB activation.
Collapse
|
30
|
Chen Y, Pan L, Jiang M, Li D, Jin L. Nanostructured lipid carriers enhance the bioavailability and brain cancer inhibitory efficacy of curcumin both in vitro and in vivo. Drug Deliv 2015; 23:1383-92. [PMID: 26066035 DOI: 10.3109/10717544.2015.1049719] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Yuhui Chen
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Lizhen Pan
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Ming Jiang
- School of Life Science and Technology, Tongji University, Shanghai, P.R. China, and
| | - Dong Li
- Department of Clinical Laboratory, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Lingjing Jin
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| |
Collapse
|