1
|
Jen CI, Ng LT. F2-sulfated polysaccharides of Laetiporus sulphureus suppress triple-negative breast cancer cell proliferation and metastasis through the EGFR-mediated signaling pathway. Int J Biol Macromol 2025; 306:141407. [PMID: 39993674 DOI: 10.1016/j.ijbiomac.2025.141407] [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: 11/22/2024] [Revised: 01/20/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
Sulfated polysaccharides (SPS) are a unique secondary metabolite isolated from Laetiporus sulphureus. This study examined the detailed molecular mechanisms of action of F2, a medium molecular weight SPS of L. sulphureus, on breast cancer MDA-MB-231 cell proliferation and metastasis. Results showed that the sulfate and protein content of F2 were 2.1 % and 15.6 %, respectively. F2 had a molecular weight of 23.8 kDa and did not contain a triple helix conformation. The monosaccharide composition of F2 was mannose, galactose, glucose, and fucose. F2 inhibited MDA-MB-231 cell proliferation mainly by blocking the cell cycle at the G0/G1 phase, which was attributed to the down-regulation of CDK4 and cyclin D1 and the up-regulation of p21 protein expression. F2 suppressed epidermal growth factor receptor (EGFR)-mediated intracellular signaling events, such as phosphorylation of ERK1/2, Akt, and GSK-3β and activation of NF-κB and β-catenin, resulting in the cell cycle arrest. Moreover, F2 significantly reduced the EGFR phosphorylation and expression, and the level of mutant p53 protein. F2 also effectively inhibited breast cancer cell migration and invasion through down-regulating MMP-9 and MMP-2 protein expression. In conclusion, this study demonstrated that F2 exhibited anti-proliferative and anti-metastatic activities against MDA-MB-231 cells by inhibiting the activation of EGFR-mediated signaling pathways.
Collapse
Affiliation(s)
- Chia-I Jen
- Department of Agricultural Chemistry, National Taiwan University, No.1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan
| | - Lean-Teik Ng
- Department of Agricultural Chemistry, National Taiwan University, No.1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan.
| |
Collapse
|
2
|
Ma Z, Wang H, Zhou Z, Lu C, Zhang M, Mu R, Zhang C, Yi Z, Deng Z, Zhao Y, Zhu J, Wen G, Jin H, An J, Tuo B, Yuan P, Liu X, Li T. SLC26A9 promotes the initiation and progression of breast cancer by activating the PI3K/AKT signaling pathway. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119912. [PMID: 39880129 DOI: 10.1016/j.bbamcr.2025.119912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 01/19/2025] [Accepted: 01/20/2025] [Indexed: 01/31/2025]
Abstract
SLC26A9 is a member of the Slc26a family of multifunctional anion transporters that function as Cl- channels in the stomach. We reported for the first time that SLC26A9 is involved in gastric tumorigenesis. However, the role of SLC26A9 in breast cancer has not yet been investigated. We first demonstrated that the upregulation of SLC26A9 is associated with the clinicopathological progression and poor prognosis of patients with breast cancer and is positively correlated with HER2 amplification. SLC26A9 alters the proliferation, migration, and invasion potential of breast cancer cells by regulating the PI3K/AKT signaling pathway. SLC26A9 acts as an oncogene in the development of breast cancer. These findings provide valuable insights for the development of future diagnostic and therapeutic strategies for BC.
Collapse
Affiliation(s)
- Zhiyuan Ma
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China; Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China; Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hu Wang
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China; Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhengxing Zhou
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China; Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chengli Lu
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China; Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Minglin Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Renmin Mu
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China; Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chengmin Zhang
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China; Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhiqiang Yi
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zilin Deng
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yingying Zhao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiaxing Zhu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Guorong Wen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hai Jin
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiaxing An
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Peng Yuan
- The Affiliated Tumor Hospital of China Academy of Medical Science, Beijing, China
| | - Xuemei Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| | - Taolang Li
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China; Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
3
|
Chu X, Wang X, Feng K, Bi Y, Xin Y, Liu S. Fucoidan ameliorates lipid accumulation, oxidative stress, and NF-κB-mediated inflammation by regulating the PI3K/AKT/Nrf2 signaling pathway in a free fatty acid-induced NAFLD spheroid model. Lipids Health Dis 2025; 24:55. [PMID: 39962463 PMCID: PMC11831825 DOI: 10.1186/s12944-025-02483-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 02/12/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease worldwide. Previous studies have reported that fucoidan can relieve obesity and hepatic steatosis in vivo, although the molecular mechanism remains unclear. This study aimed to explore the effect and potential molecular mechanism of fucoidan in NAFLD using the free fatty acid (FFA)-induced NAFLD spheroid model. MATERIALS AND METHODS The spheroids were constructed by fusing the HepG2 and LX-2 cells. Spheroids and HepG2 cells were stimulated with FFAs and fucoidan, then the intracellular lipid contents and the oxidative stress levels (ROS/MDA/GSH/GR/GPx/NQO1/GCLC/HO-1) were detected. Furthermore, the regulation of PI3K/AKT/Nrf2 pathway and the expression of inflammatory factors (TNF-α and IL-6) were measured. RESULTS Fucoidan markedly reduced FFA-induced intracellular lipid accumulation in spheroids and HepG2 cells. Notably, fucoidan relieved FFA-induced oxidative stress by reducing the levels of ROS and MDA, and elevating the levels of GSH, GR, and GPx. Furthermore, fucoidan reduced FFA-induced oxidative stress by activating the PI3K/AKT/Nrf2 signaling pathway and by inhibiting ROS-induced P65 NF-κB activation and inflammatory responses via Nrf2 pathway activation. CONCLUSIONS Our results demonstrated that fucoidan ameliorated FFA-induced lipid accumulation, oxidative stress, and NF-κB-mediated inflammation through the PI3K/AKT/Nrf2 signaling pathway in the spheroid and HepG2 cells model of NAFLD. These results provided new evidence for the clinical use of fucoidan in the treatment of NAFLD and its potential molecular mechanism of action.
Collapse
Affiliation(s)
- Xueru Chu
- School of Medicine and Pharmacy, Ocean University of China, Department of Infectious Disease, Qingdao Municipal Hospital, Qingdao, 266011, China
| | - Xuan Wang
- School of Medicine and Pharmacy, Ocean University of China, Department of Infectious Disease, Qingdao Municipal Hospital, Qingdao, 266011, China
| | - Keqing Feng
- School of Medicine and Pharmacy, Ocean University of China, Department of Infectious Disease, Qingdao Municipal Hospital, Qingdao, 266011, China
| | - Yanzhen Bi
- Department of Infectious Disease, Qingdao Municipal Hospital, 1 Jiaozhou Road, Qingdao, Shandong Province, 266011, China
| | - Yongning Xin
- School of Medicine and Pharmacy, Ocean University of China, Department of Infectious Disease, Qingdao Municipal Hospital, Qingdao, 266011, China.
- Department of Infectious Disease, Qingdao Municipal Hospital, 1 Jiaozhou Road, Qingdao, Shandong Province, 266011, China.
| | - Shousheng Liu
- Clinical Research Center, Qingdao Municipal Hospital, 5 Donghaizhong Road, Qingdao, Shandong Province, 266011, China.
| |
Collapse
|
4
|
Wang Z, Lai Y, Zhang N, Yang H, Huang Y, Yang Y, Zhang X, Ye J, Xiao M. Fucoidan treats chemotherapy-induced alopecia and helps cyclophosphamide treat tumors. Int J Biol Macromol 2025; 287:138321. [PMID: 39638216 DOI: 10.1016/j.ijbiomac.2024.138321] [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: 07/26/2024] [Revised: 10/30/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Chemotherapy-induced alopecia (CIA) represents one of the most common side effects of cancer treatment. Currently, scalp cooling systems are utilized to treat CIA, but their safety and effectiveness remain limited. The objective of this study was to investigate the effect of fucoidan on CIA and to elucidate the possible mechanism of fucoidan in treating CIA. The results showed that when the dosage of fucoidan was 100 mg/kg·d, it could effectively alleviate CIA induced by cyclophosphamide and promote hair recovery. Altering the dosage affected the therapeutic effect. A lower dosage (50 mg/kg·d) could not effectively prevent the hair from falling off, and the regrown hair was sparse, while an increased dosage led to slow hair growth, although the hair regrown was thick and black. It was also found that with the increase in dosage, key CIA proteins P53 and Fas were down-regulated. However, the cyclin was decreased when the dose was too high. In addition, fucoidan proved beneficial to cyclophosphamide treatment, which further inhibited tumor growth, aggravated tumor necrosis, and reduced the side effects of cyclophosphamide, especially at high doses. These results demonstrate that fucoidan has a therapeutic effect on CIA and does not compromise the effect of chemotherapy.
Collapse
Affiliation(s)
- Zhiyan Wang
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian 361021, China
| | - Yanbin Lai
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian 361021, China
| | - Na Zhang
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian 361021, China; Xiamen Engineering and Technological Research Center for Comprehensive Utilization of Marine Biological Resources, Xiamen 361021, China.
| | - Hongjie Yang
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian 361021, China; Xiamen Engineering and Technological Research Center for Comprehensive Utilization of Marine Biological Resources, Xiamen 361021, China
| | - Yayan Huang
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian 361021, China; Xiamen Engineering and Technological Research Center for Comprehensive Utilization of Marine Biological Resources, Xiamen 361021, China
| | - Yucheng Yang
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian 361021, China; Xiamen Engineering and Technological Research Center for Comprehensive Utilization of Marine Biological Resources, Xiamen 361021, China
| | - Xueqin Zhang
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian 361021, China; Xiamen Engineering and Technological Research Center for Comprehensive Utilization of Marine Biological Resources, Xiamen 361021, China
| | - Jing Ye
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian 361021, China; Xiamen Engineering and Technological Research Center for Comprehensive Utilization of Marine Biological Resources, Xiamen 361021, China
| | - Meitian Xiao
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian 361021, China; Xiamen Engineering and Technological Research Center for Comprehensive Utilization of Marine Biological Resources, Xiamen 361021, China
| |
Collapse
|
5
|
Behera M, Singh L, Pradhan B, Behera KC. Seaweed-Derived Bioactive Compounds: Potent Modulators in Breast Cancer Therapy. Chem Biodivers 2024:e202401613. [PMID: 39652742 DOI: 10.1002/cbdv.202401613] [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: 07/07/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 12/18/2024]
Abstract
Cancer remains a major global health concern, with breast cancer being particularly challenging. To address this, new therapeutic strategies are being explored, including natural alternatives. Seaweeds, rich in bioactive compounds, have gained attention for their therapeutic potential. Traditionally valued for their nutritional content, seaweed-derived compounds such as polysaccharides, polyphenols, sterols, vitamins, minerals, and carotenoids have shown anticancer properties. These compounds can modulate key cellular processes like apoptosis, angiogenesis, and inflammation-crucial in cancer progression. Their antioxidant, anti-inflammatory, and immunomodulatory effects make them promising candidates for complementary cancer therapies. Key bioactive components like fucoidans, laminarins, phlorotannins, and carotenoids exhibit antiproliferative, proapoptotic, antiangiogenic, and antimetastatic properties. Recent studies focus on the ability of these compounds to induce apoptosis in cancer cells. This review highlights the chemical constituents of various seaweed species with antitumor activity, their mechanisms of action, and the potential for integration into cancer treatments. It also addresses challenges in clinical applications and outlines future research directions for leveraging these marine resources in breast cancer therapy.
Collapse
Affiliation(s)
- Maheswari Behera
- Department of Botany, College of Basic Science and Humanities, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha, India
| | - Lakshmi Singh
- Department of Botany, College of Basic Science and Humanities, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha, India
| | | | | |
Collapse
|
6
|
Chi Y, Li Y, Ding C, Liu X, Luo M, Wang Z, Bi Y, Luo S. Structural and biofunctional diversity of sulfated polysaccharides from the genus Codium (Bryopsidales, Chlorophyta): A review. Int J Biol Macromol 2024; 263:130364. [PMID: 38401579 DOI: 10.1016/j.ijbiomac.2024.130364] [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: 10/16/2023] [Revised: 01/14/2024] [Accepted: 02/20/2024] [Indexed: 02/26/2024]
Abstract
It is believed that polysaccharides will become a focal point for future production of food, pharmaceuticals, and materials due to their ubiquitous and renewable nature, as well as their exceptional properties that have been extensively validated in the fields of nutrition, healthcare, and materials. Sulfated polysaccharides derived from seaweed sources have attracted considerable attention owing to their distinctive structures and properties. The genus Codium, represented by the species C. fragile, holds significance as a vital economic green seaweed and serves as a traditional Chinese medicinal herb. To date, the cell walls of the genus Codium have been found to contain at least four types of sulfated polysaccharides, specifically pyruvylated β-d-galactan sulfates, sulfated arabinogalactans, sulfated β-l-arabinans, and sulfated β-d-mannans. These sulfated polysaccharides exhibit diverse biofunctions, including anticoagulant, immune-enhancing, anticancer, antioxidant activities, and drug-carrying capacity. This review explores the structural and biofunctional diversity of sulfated polysaccharides derived from the genus Codium. Additionally, in addressing the impending challenges within the industrialization of these polysaccharides, encompassing concerns regarding scale-up production and quality control, we outline potential strategies to address these challenges from the perspectives of raw materials, extraction processes, purification technologies, and methods for quality control.
Collapse
Affiliation(s)
- Yongzhou Chi
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, Jiangsu 223003, China.
| | - Yang Li
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, Jiangsu 223003, China
| | - Chengcheng Ding
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, Jiangsu 223003, China
| | - Xiao Liu
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, Jiangsu 223003, China
| | - Meilin Luo
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, Jiangsu 223003, China
| | - Zhaoyu Wang
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, Jiangsu 223003, China
| | - Yanhong Bi
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, Jiangsu 223003, China
| | - Si Luo
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, Jiangsu 223003, China
| |
Collapse
|
7
|
Yu H, Zhang Q, Farooqi AA, Wang J, Yue Y, Geng L, Wu N. Opportunities and challenges of fucoidan for tumors therapy. Carbohydr Polym 2024; 324:121555. [PMID: 37985117 DOI: 10.1016/j.carbpol.2023.121555] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/22/2023]
Abstract
The large-scale collections, screening and discovery of biologically active and pharmacologically significant marine-derived natural products have garnered tremendous attraction. Edible brown algae are rich in fucoidan. Importantly, fucoidan has been reported to inhibit carcinogenesis and metastasis mainly through the regulation of deregulated cell signaling pathways. This review summarizes the structural features of fucoidan, including monosaccharide type, sulfate content, and main chain structure. We have set spotlight on fucoidan-mediated tumor suppressive effects in cell cultures studies and tumor-bearing rodent models. Fucoidan exerts anti-tumor effects primarily through the inhibition of tumor cell viability, proliferation and metastatic dissemination of cancer cells from primary tumor sites to distant secondary sites. Fucoidan not only promotes immunological responses in tumor microenvironment but also induces apoptotic death in cancer cells. In addition, fucoidan can be used as a dietary supplement for preventive purposes, in combination with other drugs as complementary and alternative medicine or with nanoparticle modifications will be the future of fucoidan use. Cutting-edge research related to fucoidan has catalyzed the transition of fucoidan from preclinical studies to different phases of clinical trials. Rationally designed clinical trials for the critical evaluation of fucoidan against different cancers will be valuable to reap full benefits.
Collapse
Affiliation(s)
- Haoyu Yu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Department of Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Quanbin Zhang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ammad Ahmad Farooqi
- Department of Molecular Oncology, Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 54000, Pakistan
| | - Jing Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Yue
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Lihua Geng
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Ning Wu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Department of Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
8
|
Zayed A, Al-Saedi DA, Mensah EO, Kanwugu ON, Adadi P, Ulber R. Fucoidan's Molecular Targets: A Comprehensive Review of Its Unique and Multiple Targets Accounting for Promising Bioactivities Supported by In Silico Studies. Mar Drugs 2023; 22:29. [PMID: 38248653 PMCID: PMC10820140 DOI: 10.3390/md22010029] [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: 11/22/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Fucoidan is a class of multifunctional polysaccharides derived from marine organisms. Its unique and diversified physicochemical and chemical properties have qualified them for potential and promising pharmacological uses in human diseases, including inflammation, tumors, immunity disorders, kidney diseases, and diabetes. Physicochemical and chemical properties are the main contributors to these bioactivities. The previous literature has attributed such activities to its ability to target key enzymes and receptors involved in potential disease pathways, either directly or indirectly, where the anionic sulfate ester groups are mainly involved in these interactions. These findings also confirm the advantageous pharmacological uses of sulfated versus non-sulfated polysaccharides. The current review shall highlight the molecular targets of fucoidans, especially enzymes, and the subsequent responses via either the upregulation or downregulation of mediators' expression in various tissue abnormalities. In addition, in silico studies will be applied to support the previous findings and show the significant contributors. The current review may help in understanding the molecular mechanisms of fucoidan. Also, the findings of this review may be utilized in the design of specific oligomers inspired by fucoidan with the purpose of treating life-threatening human diseases effectively.
Collapse
Affiliation(s)
- Ahmed Zayed
- Institute of Bioprocess Engineering, Rheinland-Pfälzische Technische Universität Kaiserslautern-Landau, Gottlieb-Daimler-Straße 49, 67663 Kaiserslautern, Germany
- Department of Pharmacognosy, College of Pharmacy, Tanta University, El-Guish Street (Medical Campus), Tanta 31527, Egypt
| | - Dalal A. Al-Saedi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Emmanuel Ofosu Mensah
- Faculty of Ecotechnology, ITMO University, Lomonosova Street 9, Saint Petersburg 191002, Russia;
| | - Osman Nabayire Kanwugu
- Institute of Chemical Engineering, Ural Federal University, Mira Street 28, Yekaterinburg 620002, Russia;
- ARC Centre of Excellence in Synthetic Biology, School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Parise Adadi
- Department of Food Science, University of Otago, Dunedin 9054, New Zealand;
| | - Roland Ulber
- Institute of Bioprocess Engineering, Rheinland-Pfälzische Technische Universität Kaiserslautern-Landau, Gottlieb-Daimler-Straße 49, 67663 Kaiserslautern, Germany
| |
Collapse
|
9
|
Ma S, Zheng Y, Ma J, Zhang X, Qu D, Song N, Sang C, Hui L. Lappaconitine sulfate inhibits proliferation and induces mitochondrial-mediated apoptosis via regulating PI3K/AKT/GSK3β signaling pathway in HeLa cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3695-3705. [PMID: 37306713 DOI: 10.1007/s00210-023-02564-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/30/2023] [Indexed: 06/13/2023]
Abstract
Lappaconitine (LA), a diterpenoid alkaloid extracted from the root of Aconitum sinomontanum Nakai, exhibits broad pharmacological effects, including anti-tumor activity. The inhibitory effect of lappaconitine hydrochloride (LH) on HepG2 and HCT-116 cells and the toxicity of lappaconitine sulfate (LS) on HT-29, A549, and HepG2 cells have been described. But the mechanisms of LA against human cervical cancer HeLa cells still need to be clarified. This study was designed to investigate the effects and molecular mechanisms of lappaconitine sulfate (LS) on the growth inhibition and apoptosis in HeLa cells. The cell viability and proliferation were evaluated using the Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2´-deoxyuridine (EdU) assay, respectively. The cell cycle distribution and apoptosis were detected by flow cytometry analysis and 4', 6-diamidino-2-phenylindole (DAPI) staining. The mitochondrial membrane potential (MMP) was determined through the 5, 5', 6, 6'-tetrachloro-1, 1', 3, 3'-tetraethylbenzimi-dazolyl carbocyanine iodide (JC-1) staining. The cell cycle arrest-, apoptosis-, and the phosphatidylinositol-3-kinase/protein kinase B/glycogen synthase kinase 3β (PI3K/AKT/GSK3β) pathway-related proteins were estimated by western blot analysis. LS markedly reduced the viability and suppressed the proliferation of HeLa cells. LS induced G0/G1 cell cycle arrest through the inhibition of Cyclin D1, p-Rb, and induction of p21 and p53. Furthermore, LS triggered apoptosis through the activation of mitochondrial-mediated pathway based on decrease of Bcl-2/Bax ratio and MMP and activation of caspase-9/7/3. Additionally, LS led to constitutive downregulation of the PI3K/AKT/GSK3β signaling pathway. Collectively, LS inhibited cell proliferation and induced apoptosis through mitochondrial-mediated pathway by suppression of the PI3K/AKT/GSK3β signaling pathway in HeLa cells.
Collapse
Affiliation(s)
- Shaocheng Ma
- College of Life Science, Northwest Normal University, Lanzhou, 730070, China
| | - Yidan Zheng
- College of Life Science, Northwest Normal University, Lanzhou, 730070, China
| | - Junyi Ma
- College of Life Science, Northwest Normal University, Lanzhou, 730070, China.
| | - Xuemei Zhang
- College of Life Science, Northwest Normal University, Lanzhou, 730070, China
| | - Danni Qu
- College of Life Science, Northwest Normal University, Lanzhou, 730070, China
| | - Na Song
- College of Life Science, Northwest Normal University, Lanzhou, 730070, China
| | - Chunyan Sang
- Key Laboratory of Stem Cells and Gene Drug of Gansu Province, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, 730050, China.
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
| | - Ling Hui
- Key Laboratory of Stem Cells and Gene Drug of Gansu Province, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, 730050, China.
| |
Collapse
|
10
|
Huang X, Li S, Ding R, Li Y, Li C, Gu R. Antitumor effects of polysaccharides from medicinal lower plants: A review. Int J Biol Macromol 2023; 252:126313. [PMID: 37579902 DOI: 10.1016/j.ijbiomac.2023.126313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
Cancer is one of the leading causes of death worldwide, yet the drugs currently approved for cancer treatment are associated with significant side effects, making it urgent to develop alternative drugs with low side effects. Polysaccharides are natural polymers with ketone or aldehyde groups, which are widely found in plants and have various biological activities such as immunomodulation, antitumor and hypolipidemic. The lower plants have attracted much attention for their outstanding anticancer effects, and many studies have shown that medicinal lower plant polysaccharides (MLPPs) have antitumor activity against various cancers and are promising alternatives with potential development in the food and pharmaceutical fields. Therefore, this review describes the structure and mechanism of action of MLPPs with antitumor activity. In addition, the application of MLPPs in cancer treatment is discussed, and the future development of MLPPs is explored.
Collapse
Affiliation(s)
- Xi Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Si Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong Ding
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Canlin Li
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rui Gu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China; School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
11
|
Silva JL, Santos EA, Alvarez-Leite JI. Are We Ready to Recommend Capsaicin for Disorders Other Than Neuropathic Pain? Nutrients 2023; 15:4469. [PMID: 37892544 PMCID: PMC10609899 DOI: 10.3390/nu15204469] [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: 09/25/2023] [Revised: 10/15/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Capsaicin, a lipophilic, volatile compound, is responsible for the pungent properties of chili peppers. In recent years, a significant increase in investigations into its properties has allowed the production of new formulations and the development of tools with biotechnological, diagnostic, and potential therapeutic applications. Most of these studies show beneficial effects, improving antioxidant and anti-inflammatory status, inducing thermogenesis, and reducing white adipose tissue. Other mechanisms, including reducing food intake and improving intestinal dysbiosis, are also described. In this way, the possible clinical application of such compound is expanding every year. This opinion article aims to provide a synthesis of recent findings regarding the mechanisms by which capsaicin participates in the control of non-communicable diseases such as obesity, diabetes, and dyslipidemia.
Collapse
Affiliation(s)
| | | | - Jacqueline I. Alvarez-Leite
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte 30161-970, MG, Brazil; (J.L.S.); (E.A.S.)
| |
Collapse
|
12
|
Yang S, Li D, Liu W, Chen X. Polysaccharides from marine biological resources and their anticancer activity on breast cancer. RSC Med Chem 2023; 14:1049-1059. [PMID: 37360387 PMCID: PMC10285744 DOI: 10.1039/d3md00035d] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 04/03/2023] [Indexed: 06/28/2023] Open
Abstract
In recent decades, natural products from marine organisms have been widely studied for the treatment of various breast cancers. Among them, polysaccharides have been favored by researchers because of their good effects and safety. In this review, polysaccharides from marine algae including macroalgae and microalgae, chitosan, microorganisms such as marine bacteria and fungi, and starfish are addressed. Their anticancer activities on different breast cancers and action mechanisms are discussed in detail. In general, polysaccharides from marine organisms are potential sources of low side-effect and high efficiency anticancer drugs for development. However, further research on animals and clinical research are needed.
Collapse
Affiliation(s)
- Shengfeng Yang
- Department of Oncology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital China
| | - Dacheng Li
- Department of Nuclear Medicine, Affiliated Hospital of Qingdao University China
| | - Weili Liu
- Department of Nuclear Medicine, Affiliated Hospital of Qingdao University China
| | - Xiaolin Chen
- Institute of Oceanology, Chinese Academy of Sciences China
| |
Collapse
|
13
|
Thilagavathi R, Priyankha S, Kannan M, Prakash M, Selvam C. Compounds from diverse natural origin against triple-negative breast cancer: A comprehensive review. Chem Biol Drug Des 2023; 101:218-243. [PMID: 36323650 DOI: 10.1111/cbdd.14172] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/25/2022] [Accepted: 10/29/2022] [Indexed: 11/05/2022]
Abstract
Triple-negative breast cancer (TNBC) is caused due to the lack of estrogen receptors (ER), progesterone receptors (PR), and human epidermal growth factor 2 (HER2) expression. Triple-negative breast cancer is the most aggressive heterogeneous disease that is capable of producing different clones and mutations. Tumorigenesis in TNBC is caused due to the mutation or overexpression of tumor suppressor genes. It is also associated with mutations in the BRCA gene which is linked to hereditary breast cancer. In addition, PARP proteins and checkpoint proteins also play a crucial function in causing TNBC. Many cell signaling pathways are dysregulated in TNBC. Even though chemotherapy and immunotherapy are good options for TNBC treatment, the response rates are still low in general. Many phytochemicals that are derived from natural compounds have shown very good inhibitions for TNBC. Natural compounds have the great advantage of being less toxic, having lesser side effects, and being easily available. The secondary metabolites such as alkaloids, terpenoids, steroids, and flavonoids in natural products make them promising inhibitors of TNBC. Their compositions also offer vital insights into inhibitory action, which could lead to new cancer-fighting strategies. This review can help in understanding how naturally occurring substances and medicinal herbs decrease specific tumors and pave the way for the development of novel and extremely efficient antitumor therapies.
Collapse
Affiliation(s)
- Ramasamy Thilagavathi
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Coimbatore, India
| | - Sridhar Priyankha
- Department of Chemistry, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu, India
| | - Manivel Kannan
- Faculty of Pharmacy, Karpagam Academy of Higher Education, Coimbatore, India
| | - Muthuramalingam Prakash
- Department of Chemistry, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu, India
| | - Chelliah Selvam
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas, USA
| |
Collapse
|
14
|
Velayutham M, Sarkar P, Sudhakaran G, Al-Ghanim KA, Maboob S, Juliet A, Guru A, Muthupandian S, Arockiaraj J. Anti-Cancer and Anti-Inflammatory Activities of a Short Molecule, PS14 Derived from the Virulent Cellulose Binding Domain of Aphanomyces invadans, on Human Laryngeal Epithelial Cells and an In Vivo Zebrafish Embryo Model. Molecules 2022; 27:7333. [PMID: 36364155 PMCID: PMC9654460 DOI: 10.3390/molecules27217333] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/17/2022] [Accepted: 10/25/2022] [Indexed: 08/20/2023] Open
Abstract
In this study, the anti-cancer and anti-inflammatory activities of PS14, a short peptide derived from the cellulase binding domain of pathogenic fungus, Aphanomyces invadans, have been evaluated, in vitro and in vivo. Bioinformatics analysis of PS14 revealed the physicochemical properties and the web-based predictions, which indicate that PS14 is non-toxic, and it has the potential to elicit anti-cancer and anti-inflammatory activities. These in silico results were experimentally validated through in vitro (L6 or Hep-2 cells) and in vivo (zebrafish embryo or larvae) models. Experimental results showed that PS14 is non-toxic in L6 cells and the zebrafish embryo, and it elicits an antitumor effect Hep-2 cells and zebrafish embryos. Anticancer activity assays, in terms of MTT, trypan blue and LDH assays, showed a dose-dependent inhibitory effect on cell proliferation. Moreover, in the epithelial cancer cells and zebrafish embryos, the peptide challenge (i) caused significant changes in the cytomorphology and induced apoptosis; (ii) triggered ROS generation; and (iii) showed a significant up-regulation of anti-cancer genes including BAX, Caspase 3, Caspase 9 and down-regulation of Bcl-2, in vitro. The anti-inflammatory activity of PS14 was observed in the cell-free in vitro assays for the inhibition of proteinase and lipoxygenase, and heat-induced hemolysis and hypotonicity-induced hemolysis. Together, this study has identified that PS14 has anti-cancer and anti-inflammatory activities, while being non-toxic, in vitro and in vivo. Future experiments can focus on the clinical or pharmacodynamics aspects of PS14.
Collapse
Affiliation(s)
- Manikandan Velayutham
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Chennai 603 203, Tamil Nadu, India
| | - Purabi Sarkar
- Department of Molecular Medicine, School of Allied Healthcare and Sciences, Jain Deemed-to-be University, Bangalore 560 066, Karnataka, India
| | - Gokul Sudhakaran
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Chennai 603 203, Tamil Nadu, India
| | | | - Shahid Maboob
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Annie Juliet
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Ajay Guru
- Department of Conservative Dentistry and Endodontics, Saveetha Dental College and Hospitals, SIMATS, Chennai 600 077, Tamil Nadu, India
| | - Saravanan Muthupandian
- AMR and Nanomedicine Lab, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciencess (SIMATS), Chennai 600 077, Tamil Nadu, India
| | - Jesu Arockiaraj
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Chennai 603 203, Tamil Nadu, India
| |
Collapse
|
15
|
A Review on Nutrients, Phytochemicals, and Health Benefits of Green Seaweed, Caulerpa lentillifera. Foods 2022; 11:foods11182832. [PMID: 36140958 PMCID: PMC9498133 DOI: 10.3390/foods11182832] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/27/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Caulerpa lentillifera is a type of green seaweed widely consumed as a fresh vegetable, specifically in Southeast Asia. Interestingly, this green seaweed has recently gained popularity in the food sector. Over the last two decades, many studies have reported that C. lentillifera is rich in polyunsaturated fatty acids, minerals, vitamins, and bioactive compounds that contribute many health benefits. On the other hand, there is currently hardly any article dedicated specifically to C. lentillifera regarding nutritional composition and recent advancements in its potential health benefits. Hence, this study will summarise the findings on the nutritional content of C. lentillifera and compile recently discovered beneficial properties throughout the past decade. From the data compiled in this review paper, it can be concluded that the nutrient and phytochemical profile of C. lentillifera differs from one region to another depending on various external factors. As a result, this paper will offer researchers the groundwork to develop food products based on C. lentillifera. The authors of this paper are hopeful that a more systematic review could be done in the future as currently, existing data is still scarce.
Collapse
|
16
|
Brevilaterin B from Brevibacillus laterosporus has selective antitumor activity and induces apoptosis in epidermal cancer. World J Microbiol Biotechnol 2022; 38:201. [PMID: 35999383 DOI: 10.1007/s11274-022-03372-8] [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: 10/02/2021] [Accepted: 07/28/2022] [Indexed: 10/15/2022]
Abstract
Brevilaterins as antimicrobial peptides (AMPs) secreted by a newly discovered species Brevibacillus laterosporus, had been demonstrated to display excellent antibacterial and antifungal activities; however, very limited information about their new bioactivity was ever developed. Herein, we discovered Brevilaterin B, an AMP produced by Br. laterosporus S62-9, exhibited a new anticancer activity and investigated its anticancer details. Proliferation, membrane permeability and apoptotic rate of cell lines were studied by methods of CCK-8 Assay, LDH Assay and Annexin V-FITC/PI Kits, respectively. ROS levels and mitochondrial membrane potential of tested cells were further detected through the fluorescent probes DCFH-DA and JC-1. Brevilaterin B exhibited broad-spectrum anticancer activity in a dose-dependent manner. It selectively inhibited the proliferation of epidermal cancer cell A431 but had no effect on its control normal cells in a dose of 2.0 µg/mL. In comparision, typical morphological characteristics of apoptosis and an apoptotic ratio of 71.0% in A431 were observed after treatment by 2.0-3.0 µg/mL of Brevilaterin B. The ROS levels increased by 21.3% and mitochondrial membrane potential reduced by 48.8% from A431 were further occurred, indicating Brevilaterin B's anticancer action was mainly focus on the mitochondrion of cancer cells. In total, Brevilaterin B we reported above maybe believed to be a potential application as an anticancer medicament, increasing its commercial value.
Collapse
|
17
|
Shiau JP, Chuang YT, Cheng YB, Tang JY, Hou MF, Yen CY, Chang HW. Impacts of Oxidative Stress and PI3K/AKT/mTOR on Metabolism and the Future Direction of Investigating Fucoidan-Modulated Metabolism. Antioxidants (Basel) 2022; 11:911. [PMID: 35624775 PMCID: PMC9137824 DOI: 10.3390/antiox11050911] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/05/2022] [Indexed: 12/22/2022] Open
Abstract
The critical factors for regulating cancer metabolism are oxidative stress and phosphoinositide-3-kinase/AKT serine-threonine kinase/mechanistic target of the rapamycin kinase (PI3K/AKT/mTOR). However, the metabolic impacts of oxidative stress and PI3K/AKT/mTOR on individual mechanisms such as glycolysis (Warburg effect), pentose phosphate pathway (PPP), fatty acid synthesis, tricarboxylic acid cycle (TCA) cycle, glutaminolysis, and oxidative phosphorylation (OXPHOS) are complicated. Therefore, this review summarizes the individual and interacting functions of oxidative stress and PI3K/AKT/mTOR on metabolism. Moreover, natural products providing oxidative stress and PI3K/AKT/mTOR modulating effects have anticancer potential. Using the example of brown algae-derived fucoidan, the roles of oxidative stress and PI3K/AKT/mTOR were summarized, although their potential functions within diverse metabolisms were rarely investigated. We propose a potential application that fucoidan may regulate oxidative stress and PI3K/AKT/mTOR signaling to modulate their associated metabolic regulations. This review sheds light on understanding the impacts of oxidative stress and PI3K/AKT/mTOR on metabolism and the future direction of metabolism-based cancer therapy of fucoidan.
Collapse
Affiliation(s)
- Jun-Ping Shiau
- Department of Surgery, Kaohsiung Municipal Siaogang Hospital, Kaohsiung 81267, Taiwan;
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Ya-Ting Chuang
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Yuan-Bin Cheng
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan;
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaoshiung Medical University, Kaohsiung 80708, Taiwan
| | - Ming-Feng Hou
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Ching-Yu Yen
- Department of Oral, Maxillofacial Surgery Chi-Mei Medical Center, Tainan 71004, Taiwan
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
18
|
Velayutham M, Sarkar P, Rajakrishnan R, Kuppusamy P, Juliet A, Arockiaraj J. Antiproliferation of MP12 derived from a fungus, Aphanomyces invadans virulence factor, cysteine-rich trypsin inhibitor on human laryngeal epithelial cells, and in vivo zebrafish embryo model. Toxicon 2022; 210:100-108. [PMID: 35217022 DOI: 10.1016/j.toxicon.2022.02.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/18/2022] [Accepted: 02/19/2022] [Indexed: 11/19/2022]
Abstract
Peptide-based drug development is an emerging and promising approach in cancer therapeutics. The present study focuses on understanding the mechanism of MP12 peptide (MDNHVCIPLCPP) derived from cysteine-rich trypsin inhibitor protein of virulence factor of pathogenic fungus Aphanomyces invadans. MP12 is involved in antiproliferative activity against the human laryngeal epithelial cell (Hep-2), demonstrated in this study. MP12 sequence showed a significant binding score and has multiple hydrogen bond interactions with the proteins that play a vital role in apoptotic pathways such as Bcl-2, caspase-3, caspase-7, and XIAP. Based on the bioinformatics characterization and molecular docking result, further study was focused on MP12 antiproliferative activity. The peptide showed a dose-dependent inhibition against Hep-2 cell line proliferation, analyzed over MTT and neutral red uptake assays. The IC50 value of the MP12 peptide was calculated based on the antiproliferative property (24.7 ± 0.34 μM). MP12 treated Hep-2 cells showed significant shrinkage in cell morphology compared to untreated cells, inhibiting the cell cycle. The gene expression analysis validated that the MP12 significantly upregulates the caspase-3, caspase-7, and caspase-9 genes. The developmental toxicity study using zebrafish embryos as in vivo model proved that the MP12 is nontoxic. Based on the obtained results, we proposed that the peptide MP12 derived from cysteine-rich trypsin inhibitor protein of virulence molecule of pathogenic fungus have a potential antiproliferative activity. However, further clinical trials need to be focused on the mechanism and therapeutic application against laryngeal cancer.
Collapse
Affiliation(s)
- Manikandan Velayutham
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603 203, Chennai, Tamil Nadu, India
| | - Purabi Sarkar
- School of Allied Healthcare and Sciences, Jain Deemed-to-be University, Whitefield, Bangalore, 560 066, Karnataka, India
| | - R Rajakrishnan
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Palaniselvam Kuppusamy
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju, 54896, South Korea
| | - Annie Juliet
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, 1 University Station A4800, Austin, TX, 78712, USA
| | - Jesu Arockiaraj
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603 203, Chennai, Tamil Nadu, India.
| |
Collapse
|
19
|
Jian Y, Kong L, Xu H, Shi Y, Huang X, Zhong W, Huang S, Li Y, Shi D, Xiao Y, Yang M, Li S, Chen X, Ouyang Y, Hu Y, Chen X, Song L, Ye R, Wei W. Protein phosphatase 1 regulatory inhibitor subunit 14C promotes triple-negative breast cancer progression via sustaining inactive glycogen synthase kinase 3 beta. Clin Transl Med 2022; 12:e725. [PMID: 35090098 PMCID: PMC8797469 DOI: 10.1002/ctm2.725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/28/2021] [Accepted: 01/17/2022] [Indexed: 11/21/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is fast-growing and highly metastatic with the poorest prognosis among the breast cancer subtypes. Inactivation of glycogen synthase kinase 3 beta (GSK3β) plays a vital role in the aggressiveness of TNBC; however, the underlying mechanism for sustained GSK3β inhibition remains largely unknown. Here, we find that protein phosphatase 1 regulatory inhibitor subunit 14C (PPP1R14C) is upregulated in TNBC and relevant to poor prognosis in patients. Overexpression of PPP1R14C facilitates cell proliferation and the aggressive phenotype of TNBC cells, whereas the depletion of PPP1R14C elicits opposite effects. Moreover, PPP1R14C is phosphorylated and activated by protein kinase C iota (PRKCI) at Thr73. p-PPP1R14C then represses Ser/Thr protein phosphatase type 1 (PP1) to retain GSK3β phosphorylation at high levels. Furthermore, p-PPP1R14C recruits E3 ligase, TRIM25, toward the ubiquitylation and degradation of non-phosphorylated GSK3β. Importantly, the blockade of PPP1R14C phosphorylation inhibits xenograft tumorigenesis and lung metastasis of TNBC cells. These findings provide a novel mechanism for sustained GSK3β inactivation in TNBC and suggest that PPP1R14C might be a potential therapeutic target.
Collapse
Affiliation(s)
- Yunting Jian
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
- Department of Pathology, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Key Laboratory for Major Obstetric Diseases of Guangdong ProvinceThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Lingzhi Kong
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Hongyi Xu
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
- Department of Breast SurgerySun Yat‐sen University Cancer CenterGuangzhouChina
| | - Yawei Shi
- Department of Thyroid and Breast SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Xinjian Huang
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Wenjing Zhong
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
- Department of Breast SurgerySun Yat‐sen University Cancer CenterGuangzhouChina
| | - Shumei Huang
- Department of Biochemistry, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Yue Li
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Dongni Shi
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Yunyun Xiao
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Muwen Yang
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Siqi Li
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
- Department of Breast SurgerySun Yat‐sen University Cancer CenterGuangzhouChina
| | - Xiangfu Chen
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Ying Ouyang
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Yameng Hu
- Department of Biochemistry, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Xin Chen
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences; Guangzhou Institute of OncologyTumor Hospital, Guangzhou Medical UniversityGuangzhouChina
| | - Libing Song
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Runyi Ye
- Department of Thyroid and Breast SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Weidong Wei
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
- Department of Breast SurgerySun Yat‐sen University Cancer CenterGuangzhouChina
| |
Collapse
|
20
|
Zhang N, Xue M, Wang Q, Liang H, Yang J, Pei Z, Qin K. Inhibition of fucoidan on breast cancer cells and potential enhancement of their sensitivity to chemotherapy by regulating autophagy. Phytother Res 2021; 35:6904-6917. [PMID: 34687482 DOI: 10.1002/ptr.7303] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 12/24/2022]
Abstract
Fucoidan is a marine-origin sulfated polysaccharide that has gained attention for its anticancer activities. However, the inhibitory effect of fucoidan on breast cancers by regulating autophagy and its mechanism are not clear, and the chemotherapeutic sensitization of fucoidan is largely unknown. In the present study, the anticancer potential of fucoidan was revealed in MCF-7 and MDA-MB-231 cells. Additionally, we also studied the chemotherapeutic sensitization of fucoidan by combining chemotherapeutic drugs doxorubicin (ADM) and cisplatin (DDP) with fucoidan on breast cancer cells. In the two kinds of human breast cancer cells, cell viability was determined by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay. Apoptosis was examined with flow cytometry. Transfection assay was used to examine autophagy flow. Western blot was used to examine the expressions of related proteins. Results suggested that fucoidan could induce autophagy and might enhance the sensitivity of breast cancer cells to chemotherapeutic drugs. Mechanistically, fucoidan induced autophagy in breast cancer cells by down-regulating m-TOR/p70S6K/TFEB pathway. In conclusion, our research revealed that fucoidan could induce autophagy of breast cancer cells by mediating m-TOR/p70S6K/TFEB pathway, thus inhibiting tumor development. Furthermore, fucoidan might enhance the sensitivity of breast cancer cells to ADM and DDP, and this enhancement was related to autophagy.
Collapse
Affiliation(s)
- Nan Zhang
- Basic Medical College, Qingdao University of Medicine, Qingdao, China
| | - Meilan Xue
- Department of Biochemistry and Molecular Biology, Basic Medical College, Qingdao University of Medicine, Qingdao, China
| | - Qing Wang
- Department of Ophthalmology, Affiliated Hospital of Qingdao, Qingdao, China
| | - Hui Liang
- The Institute of Human Nutrition, Qingdao University of Medicine, Qingdao, China
| | - Jia Yang
- Basic Medical College, Qingdao University of Medicine, Qingdao, China
| | - Zhongqian Pei
- Basic Medical College, Qingdao University of Medicine, Qingdao, China
| | - Kunpeng Qin
- Basic Medical College, Qingdao University of Medicine, Qingdao, China
| |
Collapse
|
21
|
Begum R, Howlader S, Mamun-Or-Rashid ANM, Rafiquzzaman SM, Ashraf GM, Albadrani GM, Sayed AA, Peluso I, Abdel-Daim MM, Uddin MS. Antioxidant and Signal-Modulating Effects of Brown Seaweed-Derived Compounds against Oxidative Stress-Associated Pathology. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9974890. [PMID: 34336128 PMCID: PMC8289617 DOI: 10.1155/2021/9974890] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/07/2021] [Accepted: 06/15/2021] [Indexed: 11/25/2022]
Abstract
The biological and therapeutic properties of seaweeds have already been well known. Several studies showed that among the various natural marine sources of antioxidants, seaweeds have become a potential source of antioxidants because of their bioactive compounds. Most of the metabolic diseases are caused by oxidative stress. It is very well known that antioxidants have a pivotal role in the treatment of those diseases. Recent researches have revealed the potential activity of seaweeds as complementary medicine, which have therapeutic properties for health and disease management. Among the seaweeds, brown seaweeds (Phaeophyta) and their derived bioactive substances showed excellent antioxidant properties than other seaweeds. This review focuses on brown seaweeds and their derived major bioactive compounds such as sulfated polysaccharide, polyphenol, carotenoid, and sterol antioxidant effects and molecular mechanisms in the case of the oxidative stress-originated disease. Antioxidants have a potential role in the modification of stress-induced signaling pathways along with the activation of the oxidative defensive pathways. This review would help to provide the basis for further studies to researchers on the potential antioxidant role in the field of medical health care and future drug development.
Collapse
Affiliation(s)
- Rahima Begum
- Department of Environmental Medical Biology, Wonju College of Medicine, Yonsei University, Wonju, Gangwon-do, 26426, Republic of Korea
| | - Saurav Howlader
- Department of Pharmacology and Pharmaco Genomics Research Centre (PGRC), Inje University College of Medicine, Busan, Republic of Korea
| | - A. N. M. Mamun-Or-Rashid
- Anti-Aging Medical Research Center and Glycative Stress Research Center, Graduate School of Life and Medical Sciences, Doshisha University, 1-3 Tatara Miyakodani, Kyotanabe, Kyoto 610-0394, Japan
| | - S. M. Rafiquzzaman
- Department of Fisheries Biology & Aquatic Environment, Bangabandhu Sheikh Mujibur Rahman Agricultural University (BSMRAU), Gazipur 1706, Bangladesh
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghadeer M. Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia
| | - Amany A. Sayed
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Ilaria Peluso
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), 00142 Rome, Italy
| | - Mohamed M. Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Md. Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| |
Collapse
|
22
|
Farghadani R, Naidu R. Curcumin: Modulator of Key Molecular Signaling Pathways in Hormone-Independent Breast Cancer. Cancers (Basel) 2021; 13:cancers13143427. [PMID: 34298639 PMCID: PMC8307022 DOI: 10.3390/cancers13143427] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 06/27/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Breast cancer remains the most commonly diagnosed cancer and the leading cause of cancer death among females worldwide. It is a highly heterogeneous disease, classified according to hormone and growth factor receptor expression. Patients with triple negative breast cancer (TNBC) (estrogen receptor-negative/progesterone receptor-negative/human epidermal growth factor receptor (HER2)-negative) and hormone-independent HER2 overexpressing subtypes still represent highly aggressive behavior, metastasis, poor prognosis, and drug resistance. Thus, new alternative anticancer agents based on the use of natural products have been receiving enormous attention. In this regard, curcumin is a promising lead in cancer drug discovery due its ability to modulate a diverse range of molecular targets and signaling pathways. The current review has emphasized the underlying mechanism of curcumin anticancer action mediated through the modulation of PI3K/Akt/mTOR, JAK/STAT, MAPK, NF-ĸB, p53, Wnt/β-catenin, apoptosis, and cell cycle pathways in hormone-independent breast cancer, providing frameworks for future studies and insights to improve its efficiency in clinical practice. Abstract Breast cancer is the most frequently diagnosed cancer and the leading cause of cancer death among women worldwide. Despite the overall successes in breast cancer therapy, hormone-independent HER2 negative breast cancer, also known as triple negative breast cancer (TNBC), lacking estrogens and progesterone receptors and with an excessive expression of human epidermal growth factor receptor 2 (HER2), along with the hormone-independent HER2 positive subtype, still remain major challenges in breast cancer treatment. Due to their poor prognoses, aggressive phenotype, and highly metastasis features, new alternative therapies have become an urgent clinical need. One of the most noteworthy phytochemicals, curcumin, has attracted enormous attention as a promising drug candidate in breast cancer prevention and treatment due to its multi-targeting effect. Curcumin interrupts major stages of tumorigenesis including cell proliferation, survival, angiogenesis, and metastasis in hormone-independent breast cancer through the modulation of multiple signaling pathways. The current review has highlighted the anticancer activity of curcumin in hormone-independent breast cancer via focusing on its impact on key signaling pathways including the PI3K/Akt/mTOR pathway, JAK/STAT pathway, MAPK pathway, NF-ĸB pathway, p53 pathway, and Wnt/β-catenin, as well as apoptotic and cell cycle pathways. Besides, its therapeutic implications in clinical trials are here presented.
Collapse
|
23
|
Kumar Y, Tarafdar A, Badgujar PC. Seaweed as a Source of Natural Antioxidants: Therapeutic Activity and Food Applications. J FOOD QUALITY 2021; 2021:1-17. [DOI: 10.1155/2021/5753391] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Seaweed is a valuable source of bioactive compounds, polysaccharides, antioxidants, minerals, and essential nutrients such as fatty acids, amino acids, and vitamins that could be used as a functional ingredient. The variation in the composition of biologically active compounds in seaweeds depends on the environmental growth factors that make seaweed of the same species compositionally different across the globe. Nevertheless, all seaweeds exhibit extraordinary antioxidant potential which can be harnessed for a broad variety of food applications such as in preparation of soups, pasta, salads, noodles, and other country specific dishes. This review highlights the nutritional and bioactive compounds occurring in different classes of seaweeds while focusing on their therapeutic activities including but not limited to blood cell aggregation, antiviral, antitumor, anti-inflammatory, and anticancer properties. The review also explores the existing and potential application of seaweeds as a source of natural antioxidant in food products. Seaweed-derived compounds have great potential for being used as a supplement in functional foods due to their high stability as well as consumer demand for antioxidant-rich foods.
Collapse
Affiliation(s)
- Yogesh Kumar
- Department of Food Science and Technology, National Institute of Food Technology Entrepreneurship and Management, Kundli, Sonipat 131028, Haryana, India
| | - Ayon Tarafdar
- Department of Food Engineering, National Institute of Food Technology Entrepreneurship and Management, Kundli, Sonipat 131028, Haryana, India
- Livestock Production and Management Section, ICAR-Indian Veterinary Research Institute, Izzatnagar, Bareilly 243 122, Uttar Pradesh, India
| | - Prarabdh C. Badgujar
- Department of Food Science and Technology, National Institute of Food Technology Entrepreneurship and Management, Kundli, Sonipat 131028, Haryana, India
| |
Collapse
|
24
|
Ouyang Y, Qiu Y, Liu Y, Zhu R, Chen Y, El-Seedi HR, Chen X, Zhao C. Cancer-fighting potentials of algal polysaccharides as nutraceuticals. Food Res Int 2021; 147:110522. [PMID: 34399500 DOI: 10.1016/j.foodres.2021.110522] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 01/03/2023]
Abstract
Cancer has been listed as one of the world's five incurable diseases by the World Health Organization and causes tens of thousands of deaths every year. Unfortunately, anticancer agents either show limited efficacy or show serious side effects. The algae possess high nutritional value and their polysaccharides have a variety of biological activities, especially anti-cancer and immunomodulatory properties. Algal polysaccharides exert anti-cancer effects by inducing apoptosis, cell cycle arrest, anti-angiogenesis, and regulating intestinal flora and immune function. Algal polysaccharides can be combined with nanoparticles and other drugs to reduce the side effects caused by chemotherapy and increase the anticancer effects. This review shows the signal pathways related to the anti-cancer mechanisms of algal polysaccharides, including their influence on intestinal flora and immune regulation, the application of nanoparticles, and the effects on combination therapy and clinical trials of cancer treatments.
Collapse
Affiliation(s)
- Yuezhen Ouyang
- Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yinghui Qiu
- Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yuning Liu
- Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ruiyu Zhu
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
| | - Yihan Chen
- Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Hesham R El-Seedi
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China; Pharmacognosy Group, Department of Pharmaceutical Biosciences, BMC, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Xinhua Chen
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Chao Zhao
- Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China.
| |
Collapse
|
25
|
Corso CR, Mulinari Turin de Oliveira N, Moura Cordeiro L, Sauruk da Silva K, da Silva Soczek SH, Frota Rossato V, Fernandes ES, Maria-Ferreira D. Polysaccharides with Antitumor Effect in Breast Cancer: A Systematic Review of Non-Clinical Studies. Nutrients 2021; 13:2008. [PMID: 34200897 PMCID: PMC8230509 DOI: 10.3390/nu13062008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/02/2021] [Accepted: 06/05/2021] [Indexed: 12/30/2022] Open
Abstract
Purpose: To review the effects of polysaccharides and their proposed mechanisms of action in breast cancer experimental models. Data sources, selection, and extraction: Articles were selected by using PubMed, ScienceDirect, Scopus, and Medline, assessed from 1 May 2019 to 1 July 2020. The systematic review was registered in the International Prospective Register of Systematic Reviews (Prospero) under the number CRD42020169103. Results: Most of the studies explore algae polysaccharides (43.2%), followed by mushrooms (13.5%), plants (13.5%), fruits (10.8%), fungus (2.7%), bacteria, (2.7%), and sea animals (2.7%). A total of 8.1% investigated only in vitro models, 62.1% evaluated only in vivo models, and 29.7% evaluated in vitro and in vivo models. The mechanism of action involves apoptosis, inhibition of cellular proliferation, angiogenesis, and antimetastatic effects through multiple pathways. Conclusions: Findings included here support further investigations on the anti-tumor effect of polysaccharides. Some polysaccharides, such as fucoidan and β-glucans, deserve detailed and structured studies aiming at translational research on breast tumors, since they are already used in the clinical practice of other proposals of human health.
Collapse
Affiliation(s)
- Claudia Rita Corso
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (C.R.C.); (N.M.T.d.O.); (L.M.C.); (K.S.d.S.); (S.H.d.S.S.); (V.F.R.); (E.S.F.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Natalia Mulinari Turin de Oliveira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (C.R.C.); (N.M.T.d.O.); (L.M.C.); (K.S.d.S.); (S.H.d.S.S.); (V.F.R.); (E.S.F.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Leonardo Moura Cordeiro
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (C.R.C.); (N.M.T.d.O.); (L.M.C.); (K.S.d.S.); (S.H.d.S.S.); (V.F.R.); (E.S.F.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Karien Sauruk da Silva
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (C.R.C.); (N.M.T.d.O.); (L.M.C.); (K.S.d.S.); (S.H.d.S.S.); (V.F.R.); (E.S.F.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Suzany Hellen da Silva Soczek
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (C.R.C.); (N.M.T.d.O.); (L.M.C.); (K.S.d.S.); (S.H.d.S.S.); (V.F.R.); (E.S.F.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Virgilio Frota Rossato
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (C.R.C.); (N.M.T.d.O.); (L.M.C.); (K.S.d.S.); (S.H.d.S.S.); (V.F.R.); (E.S.F.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Elizabeth Soares Fernandes
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (C.R.C.); (N.M.T.d.O.); (L.M.C.); (K.S.d.S.); (S.H.d.S.S.); (V.F.R.); (E.S.F.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Daniele Maria-Ferreira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (C.R.C.); (N.M.T.d.O.); (L.M.C.); (K.S.d.S.); (S.H.d.S.S.); (V.F.R.); (E.S.F.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| |
Collapse
|
26
|
Liang R, Zhu X. UC2288 induces cell apoptosis of nasopharyngeal carcinoma cells via inhibiting EGFR/ERK pathway. J Cancer 2021; 12:988-995. [PMID: 33442398 PMCID: PMC7797659 DOI: 10.7150/jca.48282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 11/30/2020] [Indexed: 02/04/2023] Open
Abstract
Radiotherapy and chemotherapy are the standard care for patients with nasopharyngeal carcinoma (NPC). These treatments cause some severe toxicity and about 30% of patients develop recurrence and metastases after treatment. UC2288 is structurally similar to sorafenib, a multikinase inhibitor. However, studies about the effects of UC2288 on tumors are few. Here, UC2288 inhibited proliferation and induced apoptosis of NPC cells in a dose- and time-dependent manner. Using western blot and immunofluorescence assay, we found that UC2288 promoted DNA damage. In addition, UC2288 decreased the phosphorylation of EGFR and ERK. Moreover, pretreatment with EGF partially rescued cell viability suppressed by UC2288. In conclusion, UC2288 suppressed the growth of NPC via inhibiting EGFR/ERK pathway and it may be a promising therapeutic option for NPC.
Collapse
Affiliation(s)
- Renba Liang
- Department of Oncology, Wuming Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China.,Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, P.R. China
| | - Xiaodong Zhu
- Department of Oncology, Wuming Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China.,Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, P.R. China.,Key Laboratory of Early Prevention and Treatment for Regional High-Incidence-Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, P.R. China
| |
Collapse
|
27
|
Pradhan B, Nayak R, Patra S, Jit BP, Ragusa A, Jena M. Bioactive Metabolites from Marine Algae as Potent Pharmacophores against Oxidative Stress-Associated Human Diseases: A Comprehensive Review. Molecules 2020; 26:E37. [PMID: 33374738 PMCID: PMC7793479 DOI: 10.3390/molecules26010037] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/13/2022] Open
Abstract
In addition to cancer and diabetes, inflammatory and ROS-related diseases represent one of the major health problems worldwide. Currently, several synthetic drugs are used to reduce oxidative stress; nevertheless, these approaches often have side effects. Therefore, to overcome these issues, the search for alternative therapies has gained importance in recent times. Natural bioactive compounds have represented, and they still do, an important source of drugs with high therapeutic efficacy. In the ''synthetic'' era, terrestrial and aquatic photosynthetic organisms have been shown to be an essential source of natural compounds, some of which might play a leading role in pharmaceutical drug development. Marine organisms constitute nearly half of the worldwide biodiversity. In the marine environment, algae, seaweeds, and seagrasses are the first reported sources of marine natural products for discovering novel pharmacophores. The algal bioactive compounds are a potential source of novel antioxidant and anticancer (through modulation of the cell cycle, metastasis, and apoptosis) compounds. Secondary metabolites in marine Algae, such as phenolic acids, flavonoids, and tannins, could have great therapeutic implications against several diseases. In this context, this review focuses on the diversity of functional compounds extracted from algae and their potential beneficial effects in fighting cancer, diabetes, and inflammatory diseases.
Collapse
Affiliation(s)
- Biswajita Pradhan
- Algal Biotechnology and Molecular Systematic Laboratory, Post Graduate Department of Botany, Berhampur University, Brahmapur 760007, India; (B.P.); (R.N.)
| | - Rabindra Nayak
- Algal Biotechnology and Molecular Systematic Laboratory, Post Graduate Department of Botany, Berhampur University, Brahmapur 760007, India; (B.P.); (R.N.)
| | - Srimanta Patra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela 769001, India;
| | - Bimal Prasad Jit
- Department of Biochemistry, All India Institute of Medical Science, Ansari Nagar, New Delhi 110023, India;
| | - Andrea Ragusa
- Department of Biological and Environmental Sciences and Technologies, Campus Ecotekne, University of Salento, via Monteroni, 73100 Lecce, Italy
- CNR-Nanotec, Institute of Nanotechnology, via Monteroni, 73100 Lecce, Italy
| | - Mrutyunjay Jena
- Algal Biotechnology and Molecular Systematic Laboratory, Post Graduate Department of Botany, Berhampur University, Brahmapur 760007, India; (B.P.); (R.N.)
| |
Collapse
|
28
|
Present Status, Limitations and Future Directions of Treatment Strategies Using Fucoidan-Based Therapies in Bladder Cancer. Cancers (Basel) 2020; 12:cancers12123776. [PMID: 33333858 PMCID: PMC7765304 DOI: 10.3390/cancers12123776] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/08/2020] [Accepted: 12/13/2020] [Indexed: 02/06/2023] Open
Abstract
Bladder cancer (BC) is a common urological cancer, with poor prognosis for advanced/metastatic stages. Various intensive treatments, including radical cystectomy, chemotherapy, immune therapy, and radiotherapy are commonly used for these patients. However, these treatments often cause complications and adverse events. Therefore, researchers are exploring the efficacy of natural product-based treatment strategies in BC patients. Fucoidan, derived from marine brown algae, is recognized as a multi-functional and safe substrate, and has been reported to have anti-cancer effects in various types of malignancies. Additionally, in vivo and in vitro studies have reported the protective effects of fucoidan against cancer-related cachexia and chemotherapeutic agent-induced adverse events. In this review, we have introduced the anti-cancer effects of fucoidan extracts in BC and highlighted its molecular mechanisms. We have also shown the anti-cancer effects of fucoidan therapy with conventional chemotherapeutic agents and new treatment strategies using fucoidan-based nanoparticles in various malignancies. Moreover, apart from the improvement of anti-cancer effects by fucoidan, its protective effects against cancer-related disorders and cisplatin-induced toxicities have been introduced. However, the available information is insufficient to conclude the clinical usefulness of fucoidan-based treatments in BC patients. Therefore, we have indicated the aspects that need to be considered regarding fucoidan-based treatments and future directions for the treatment of BC.
Collapse
|
29
|
Webb MJ, Kukard C. A Review of Natural Therapies Potentially Relevant in Triple Negative Breast Cancer Aimed at Targeting Cancer Cell Vulnerabilities. Integr Cancer Ther 2020; 19:1534735420975861. [PMID: 33243021 PMCID: PMC7705812 DOI: 10.1177/1534735420975861] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We reviewed the research into the mechanisms of growth of triple negative breast cancer (TNBC) based on laboratory pre-clinical studies that have shaped understanding of the disease over the past decade. In response to these findings, we propose an approach to potentially prevent cancer metabolic adaptation and recurrence. This paper collates pre-clinical results, first to determine the tumor’s mechanisms of growth and then to source natural substances that could potentially suppress those mechanisms. The results from in vivo and in vitro studies of TNBC were combined first to select 10 primary mechanisms (Hypoxia-inducible factor 1α, Hedgehog, MAPK, MTAP, NF-κ B, Notch, P13K, STAT3, and Wnt signaling pathways plus p53 and POL2A gene expression) that promote TNBC growth, and second to propose a treatment array of 21 natural compounds that suppress laboratory models of TNBC via these mechanisms. We included BRCA mutations in the review process, but only pathways with the most preclinical studies utilizing natural products were included. Then we outlined potential biomarkers to assess the changes in the micro-environment and monitor biochemical pathway suppression. This suppression-centric aim targets these mechanisms of growth with the goal of potentially halting tumor growth and preventing cancer cell metabolic adaptation. We chose TNBC to demonstrate this 5-step strategy of supplementary therapy, which may be replicated for other tumor types.
Collapse
Affiliation(s)
| | - Craig Kukard
- University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
30
|
Chang LC, Chiang YF, Chen HY, Huang YJ, Liu AC, Hsia SM. The Potential Effect of Fucoidan on Inhibiting Epithelial-to-Mesenchymal Transition, Proliferation, and Increase in Apoptosis for Endometriosis Treatment: In Vivo and In Vitro Study. Biomedicines 2020; 8:E528. [PMID: 33266505 PMCID: PMC7700274 DOI: 10.3390/biomedicines8110528] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/16/2020] [Accepted: 11/21/2020] [Indexed: 12/18/2022] Open
Abstract
Endometriosis is common in reproductive-age women and its pathology is to increase proliferation and migration to enhance epithelial-to-mesenchymal transition progression (EMT). However, treatments are currently limited, so it is important to explore new therapeutic drugs. Hence, in this study, we investigate the therapeutic effect of fucoidan (FC) on the progression and mechanisms of endometriosis. The cell viability of endometrial cell lines End1/E6E7 and Vk2/E6E7 treated with different concentrations of FC were assessed by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and cell counting. Cell migration was evaluated using wound-healing assay. In an in vivo experiment, female Balb/c mice received surgically induced endometriosis followed by different concentrations of fucoidan for 6 weeks. High-frequency ultrasound imaging was applied to detect subsequent lesion growth. The results demonstrated that fucoidan inhibited the viability and migration ability of End1/E6E7 and Vk2/E6E7 cells. Additionally, the administration of fucoidan reduced the volume and weight of endometriotic lesions, decreased inflammatory cytokines and vascular endothelial growth factor (VEGF) of serum and lesions, and improved EMT proliferation and apoptosis-related protein expression. For the first time, fucoidan indicated anti-proliferative and anti-inflammatory effects as well as inhibited EMT progression and induced apoptosis, improving endometriosis.
Collapse
Affiliation(s)
- Li-Chun Chang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (L.-C.C.); (Y.-F.C.); (H.-Y.C.); (Y.-J.H.)
| | - Yi-Fen Chiang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (L.-C.C.); (Y.-F.C.); (H.-Y.C.); (Y.-J.H.)
| | - Hsin-Yuan Chen
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (L.-C.C.); (Y.-F.C.); (H.-Y.C.); (Y.-J.H.)
| | - Yun-Ju Huang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (L.-C.C.); (Y.-F.C.); (H.-Y.C.); (Y.-J.H.)
| | - An-Chieh Liu
- Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei 11031, Taiwan;
| | - Shih-Min Hsia
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (L.-C.C.); (Y.-F.C.); (H.-Y.C.); (Y.-J.H.)
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
- School of Food and Safety, Taipei Medical University, Taipei 11031, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
| |
Collapse
|
31
|
Zhang Y, Du H, Yu X, Zhu J. Fucoidan attenuates hyperoxia-induced lung injury in newborn rats by mediating lung fibroblasts differentiate into myofibroblasts. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1501. [PMID: 33313246 PMCID: PMC7729344 DOI: 10.21037/atm-20-6601] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Hyperoxia-induced lung injury is one of the most common and frequent diseases in premature infants and may develop into bronchopulmonary dysplasia (BPD). Fucoidan, extracted from brown seaweed and brown algae, has anti-apoptosis, antioxidative and anti-fibrosis effects. This study aimed to explore whether fucoidan could alleviate hyperoxia-induced lung injury in newborn rats. Methods Lung wet-weight/dry-weight (W/D) ratio, total protein (TP) content, total cell counts, and lactate dehydrogenase (LDH) levels are used to evaluate lung injury. Masson staining is used to evaluate lung fibrotic. Tunnel assay and Hoechst 33258 assay were used to evaluate apoptosis. The levels of serum superoxide dismutase (SOD), malondialdehyde (MDA), and glutathione (GSH) were measured using ELISA to assess oxidative stress. Western blot assay was used to detect apoptosis-related proteins Bcl-1, Bax, and myofibroblast proteins α-SMA. Results The data indicating fucoidan treatment remarkably reduces the lung W/D ratio and TP content, total cell counts, and LDH levels in bronchoalveolar lavage fluid (BALF). Also, fucoidan treatment significantly inhibited cell apoptosis with the elevated expression of Bcl-2/Bax in cultured lung fibroblasts. Moreover, treatment with fucoidan suppressed the levels of MDA significantly and elevated the level of SOD and GSH, showing that oxidative stress was restrained by fucoidan. Furthermore, the decreased expression levels of α-SMA and collagen I was detected in fibroblast treated with fucoidan. Conclusions These data suggest fucoidan may protect the lung from hyperoxia via suppressing cell apoptosis, mitigating oxidative stress, and inhibiting lung fibroblasts from differentiating into myofibroblasts.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Geriatric Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University Hospital of Electronic Science & Technology, Chengdu, China
| | - Hengjian Du
- Department of Geriatric Infectious Diseases, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University Hospital of Electronic Science & Technology, Chengdu, China
| | - Xuelian Yu
- Department of Geriatric Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University Hospital of Electronic Science & Technology, Chengdu, China
| | - Jiang Zhu
- Department of Respiratory and Critical Care Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University Hospital of Electronic Science & Technology, Chengdu, China
| |
Collapse
|
32
|
Protein kinases as targets for developing anticancer agents from marine organisms. Biochim Biophys Acta Gen Subj 2020; 1865:129759. [PMID: 33038451 DOI: 10.1016/j.bbagen.2020.129759] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/03/2020] [Accepted: 10/03/2020] [Indexed: 01/11/2023]
Abstract
Protein kinases play a fundamental role in the intracellular transduction because of their ability to phosphorylate plethora of proteins. Over the past three decades, numerous protein kinase inhibitors have been identified and are being used clinically successfully. The biodiversity of marine organisms provides a rich source for the discovery and development of novel anticancer agents in the treatment of human malignancies and a lot of bioactive ingredients from marine organisms display anticancer effects by affecting the protein kinases-mediated pathways. In the present mini-review, anticancer compounds from marine source were reviewed and discussed in context of their targeted pathways associated with protein kinases and the progress of these compounds as anticancer agents in recent five years were emphasized. The molecular entities and their modes of actions were presented. We focused on protein kinases-mediated signaling pathways including PI3K/Akt/mTOR, p38 MAPK, and EGFR. The marine compounds targeting special pathways of protein kinases were highlighted. We have also discussed the existing challenges and prospects related to design and development of novel protein kinase inhibitors from marine sources.
Collapse
|
33
|
de Oliveira Filho JWG, Andrade TDJADS, de Lima RMT, Dos Reis AC, Silva DHS, Santos JVDO, de Menezes AAPM, da Mata AMO, Dias ACS, de Alencar MVOB, Paz MFCJ, Moreno LCGEAI, Islam MT, Mubarak MS, Sousa JMDCE, Melo Cavalcante AADC. Citrinin against breast cancer: A cytogenotoxicological study. Phytother Res 2020; 35:504-516. [PMID: 32869401 DOI: 10.1002/ptr.6830] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/30/2020] [Accepted: 07/11/2020] [Indexed: 11/10/2022]
Abstract
Breast cancer is one of the most lethal types of cancer and a leading cause of mortality among Women worldwide. Citrinin (CIT), a polyketide extracted from the fungus Penicillium citrinum, exhibits a wide range of biological activities such as antibacterial, antifungal, and cytotoxic effects. The aim of the current study was to evaluate the antitumoral effects of CIT against 7,12-dimethylbenzanthracene (DMBA)-induced mammary carcinoma in Swiss mice For this, CIT, DMBA and the standard cyclophosphamide (CPA) induced behavioral changes in experimental animals, and these changes were screened by using the rota rod and open field tests. Additionally, hematological, biochemical, immuno-histochemical, and histopathological analyses were carried out. Results suggest that CIT did not alter behavioral, hematological, and biochemical parameters in mice. DMBA induced invasive mammary carcinoma and showed genotoxic effects in the breasts, bone marrow, lymphocytes, and hepatic cells. It also caused mutagenic effects in the formation of micronuclei, bridges, shoots, and binucleate cells in bone marrow and liver. CIT and CPA genotoxic effects were observed after 3 weeks of therapy, where CIT exhibited a repair capacity and induced significant apoptotic damage in mouse lymphocytes. In conclusion, CIT showed antitumoral effects in Swiss mice, possibly through induction of apoptosis.
Collapse
Affiliation(s)
- José Williams Gomes de Oliveira Filho
- Northeast Biotechnology Network (RENORBIO), Postgraduate Program in Biotechnology, Federal University of Piauí - UFPI, Teresina, Piauí, Brazil.,Laboratory of Research in Toxicological Genetics - LAPGENIC, Federal University of Piauí, Teresina, Piauí, Brazil.,Federal Institute of Piauí (IFPI), Teresina, Piauí, Brazil
| | | | - Rosália Maria Tôrres de Lima
- Northeast Biotechnology Network (RENORBIO), Postgraduate Program in Biotechnology, Federal University of Piauí - UFPI, Teresina, Piauí, Brazil
| | - Antonielly Campinho Dos Reis
- Laboratory of Research in Toxicological Genetics - LAPGENIC, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Dulce Helena Siqueira Silva
- Nucleus of Bioassays, Biosynthesis and Ecophysiology of Natural Products (NuBBE), Department of Organic Chemistry, Institute of Chemistry, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | | | | | - Ana Maria Oliveira da Mata
- Laboratory of Research in Toxicological Genetics - LAPGENIC, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Ana Carolina Soares Dias
- Laboratory of Genetics and Molecular Biology, Federal University of Maranhão, São Luís, Maranhão, Brazil
| | | | | | | | - Muhammad Torequl Islam
- Laboratory of Theoretical and Computational Biophysics, Ton Duc Thang University, Ho Chi Minh City, Vietnam.,Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | | | - João Marcelo de Castro E Sousa
- Northeast Biotechnology Network (RENORBIO), Postgraduate Program in Biotechnology, Federal University of Piauí - UFPI, Teresina, Piauí, Brazil.,Laboratory of Pharmaceutical Nanosystems - NANOSFAR, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Ana Amélia de Carvalho Melo Cavalcante
- Northeast Biotechnology Network (RENORBIO), Postgraduate Program in Biotechnology, Federal University of Piauí - UFPI, Teresina, Piauí, Brazil.,Laboratory of Pharmaceutical Nanosystems - NANOSFAR, Federal University of Piauí, Teresina, Piauí, Brazil
| |
Collapse
|
34
|
Mayor ABR, Guevarra LA, Santiago-Bautista MR, Santiago LA. Phlogiellus bundokalbo spider venom: cytotoxic fractions against human lung adenocarcinoma (A549) cells. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20190104. [PMID: 32788916 PMCID: PMC7401667 DOI: 10.1590/1678-9199-jvatitd-2019-0104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Spider venom is a potential source of pharmacologically important compounds. Previous studies on spider venoms reported the presence of bioactive molecules that possess cell-modulating activities. Despite these claims, sparse scientific evidence is available on the cytotoxic mechanisms in relation to the components of the spider venom. In this study, we aimed to determine the cytotoxic fractions of the spider venom extracted from Phlogiellus bundokalbo and to ascertain the possible mechanism of toxicity towards human lung adenocarcinoma (A549) cells. Methods Spider venom was extracted by electrostimulation. Components of the extracted venom were separated by reversed-phase high performance liquid chromatography (RP-HPLC) using a linear gradient of 0.1% trifluoroacetic acid (TFA) in water and 0.1% TFA in 95% acetonitrile (ACN). Cytotoxic activity was evaluated by the MTT assay. Apoptotic or necrotic cell death was assessed by microscopic evaluation in the presence of Hoechst 33342 and Annexin V, Alexa FluorTM 488 conjugate fluorescent stains, and caspase activation assay. Phospholipase A2 (PLA2) activity of the cytotoxic fractions were also measured. Results We observed and isolated six fractions from the venom of P. bundokalbo collected from Aurora, Zamboanga del Sur. Four of these fractions displayed cytotoxic activities. Fractions AT5-1, AT5-3, and AT5-4 were found to be apoptotic while AT5-6, the least polar among the cytotoxic components, was observed to induce necrosis. PLA2 activity also showed cytotoxicity in all fractions but presented no relationship between specific activity of PLA2 and cytotoxicity. Conclusion The venom of P. bundokalbo spider, an endemic tarantula species in the Philippines, contains components that were able to induce either apoptosis or necrosis in A549 cells.
Collapse
Affiliation(s)
- Anna Beatriz R Mayor
- The Graduate School, University of Santo Tomas, Manila, Philippines.,Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| | - Leonardo A Guevarra
- Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines.,Department of Biochemistry, Faculty of Pharmacy, University of Santo Tomas, Manila, Philippines
| | - Myla R Santiago-Bautista
- The Graduate School, University of Santo Tomas, Manila, Philippines.,Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines.,Department of Biochemistry, Faculty of Pharmacy, University of Santo Tomas, Manila, Philippines
| | - Librado A Santiago
- The Graduate School, University of Santo Tomas, Manila, Philippines.,Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines.,Department of Biochemistry, Faculty of Pharmacy, University of Santo Tomas, Manila, Philippines
| |
Collapse
|
35
|
Liu S, Yang J, Peng X, Li J, Zhu C. The Natural Product Fucoidan Inhibits Proliferation and Induces Apoptosis of Human Ovarian Cancer Cells: Focus on the PI3K/Akt Signaling Pathway. Cancer Manag Res 2020; 12:6195-6207. [PMID: 32884336 PMCID: PMC7434378 DOI: 10.2147/cmar.s254784] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/16/2020] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Ovarian cancer (OC) is the leading cause of death among gynecological tumors; however, no effective treatment is currently available. Fucoidan, which is extracted from marine algae, has significant anti-cancer effects. The aim of this study was to determine the effects of fucoidan on the proliferation and apoptosis of OC cells through inhibition of the PI3K/Akt signaling pathway. METHODS Human ovarian normal epithelial cells (IOSE80) and human OC cells (SKOV-3, A2780, OVCAR-3, TOV-112D, and Caov-3) were selected to verify the safety of fucoidan at various doses in SKOV-3 and Caov-3 cells as well as a xenograft mouse model using various molecular biology techniques. RESULTS Fucoidan had no significant effect on normal ovarian epithelial cells, but had significantly inhibited the proliferation of OC cells, induced cell cycle arrest at the G0/G1 phase, increased the proportion of apoptotic cells and expression of pro-apoptotic proteins, and inhibited the expression of PI3K and phosphorylation of Akt, which could be partly rescued by IGF-1. CONCLUSION Fucoidan had anti-tumor effects both in vivo and in vitro via a mechanism that is related to the inhibition of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Shuhan Liu
- Department of Reproductive Medicine, Jining First People’s Hospital, Jining272000, People’s Republic of China
| | - Jing Yang
- Department of Gynecology, Jining First People’s Hospital, Jining272000, People’s Republic of China
| | - Xudong Peng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao266000, People’s Republic of China
| | - Jingjing Li
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao266000, People’s Republic of China
| | - Cunjing Zhu
- Department of Gynecology, Jining First People’s Hospital, Jining272000, People’s Republic of China
| |
Collapse
|
36
|
Li S, Zhou G, Liu W, Ye J, Yuan F, Zhang Z. Curcumol Inhibits Lung Adenocarcinoma Growth and Metastasis via Inactivation of PI3K/AKT and Wnt/-Catenin Pathway. Oncol Res 2020; 28:685-700. [PMID: 32886059 PMCID: PMC8420902 DOI: 10.3727/096504020x15917007265498] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Curcumol (Cur), isolated from the Traditional Chinese Medical plant Rhizoma Curcumae, is the bioactive component of sesquiterpene reported to possess antitumor activity. However, its bioactivity and mechanisms against lung adenocarcinoma are still unclear. We investigated its effect on lung adenocarcinoma and elucidated its underlying molecular mechanisms. In vitro, Cur effectively suppressed proliferation, migration, and invasion of lung adenocarcinoma cells A549 and H460, which were associated with the altered expressions of signaling molecules, including p-AKT, p-PI3K, p-LRP5/6, AXIN, APC, GSK3 and p--catenin, matrix metalloproteinase (MMP)-2, and MMP-9. Furthermore, Cur significantly induced cell apoptosis of A549 and H460 by promoting the expression of Bax, caspase 3, and caspase 9 and suppressing the expression of Bcl-2, and arrested the cell cycle at the G0/G1 phase by lowering the levels of cyclin D1, CDK1, and CDK4. In vivo experiment revealed that Cur could inhibit lung tumor growth and lung metastasis, which were consistent with these in vitro results. In xenograft model mice, Cur strongly decreased tumor weight and tumor volume, which may be related to the downregulation of p-AKT and p-PI3K by immunofluorescence analysis. In addition, a lung metastasis model experiment suggested that Cur dramatically decreased the ratio of lung/total weight, tumor metastatic nodules, and the expressions of MMP-2 and MMP-9 in lung tissues compared with the control. Overall, these data suggested that the inhibitory activity of Cur on lung adenocarcinoma via the inactivation of PI3K/Akt and Wnt/-catenin pathways, at least in part, indicates that curcumol may be a potential antitumor agent for lung adenocarcinoma therapy.
Collapse
Affiliation(s)
- Sheng Li
- Department of Chemotherapy, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer HospitalNanjingP.R. China
| | - Guoren Zhou
- Department of Chemotherapy, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer HospitalNanjingP.R. China
| | - Wei Liu
- Department of Radiotherapy, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer HospitalNanjingP.R. China
| | - Jinjun Ye
- Department of Radiotherapy, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer HospitalNanjingP.R. China
| | - Fangliang Yuan
- Department of Thoracic Surgery, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer HospitalNanjingP.R. China
| | - Zhi Zhang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer HospitalNanjingP.R. China
| |
Collapse
|
37
|
Jo G, Kwon MJ, Kim JN, Kim BJ. Radix Sophorae Flavescentis induces apoptosis through by Caspase, MAPK Activation and ROS Signaling Pathways in 5637 Human Bladder Cancer Cells. Int J Med Sci 2020; 17:1474-1481. [PMID: 32669949 PMCID: PMC7359395 DOI: 10.7150/ijms.45831] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/01/2020] [Indexed: 12/19/2022] Open
Abstract
The anti-cancer mechanisms of Radix Sophorae Flavescentis were investigated in 5637 bladder cancer cells. Radix Sophorae Flavescentis extract (RSF) (50‑400 µg/ml) inhibited the proliferation of 5637 cells and increased sub‑G1 phase ratios. RSF‑induced cell death was associated with the down-regulation of B‑cell lymphoma 2 (Bcl‑2) and the up-regulation of Bcl‑2 X‑associated protein (Bax). RSF also activated caspase‑3 and -9 and regulated the activations of mitogen-activated protein kinases (MAPKs). In addition, RSF increased intracellular reactive oxygen species (ROS) levels and depolarized the mitochondrial membrane potential. These findings suggest RSF induces apoptosis in 5637 bladder cancer cells and that it has potential use as a novel anti-cancer drug for bladder cancer.
Collapse
Affiliation(s)
| | | | | | - Byung Joo Kim
- Division of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan 50612, Republic of Korea
| |
Collapse
|
38
|
Ran L, Chen F, Zhang J, Mi J, Lu L, Yan Y, Cao Y. Antitumor effects of pollen polysaccharides from Chinese wolfberry on DU145 cells via the PI3K/AKT pathway in vitro and in vivo. Int J Biol Macromol 2020; 152:1164-1173. [DOI: 10.1016/j.ijbiomac.2019.10.206] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/13/2019] [Accepted: 10/24/2019] [Indexed: 12/15/2022]
|
39
|
Zubair H, Khan MA, Anand S, Srivastava SK, Singh S, Singh AP. Modulation of the tumor microenvironment by natural agents: implications for cancer prevention and therapy. Semin Cancer Biol 2020; 80:237-255. [PMID: 32470379 PMCID: PMC7688484 DOI: 10.1016/j.semcancer.2020.05.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 05/10/2020] [Accepted: 05/14/2020] [Indexed: 02/07/2023]
Abstract
The development of cancer is not just the growth and proliferation of a single transformed cell, but its surrounding environment also coevolves with it. Indeed, successful cancer progression depends on the ability of the tumor cells to develop a supportive tumor microenvironment consisting of various types of stromal cells. The interactions between the tumor and stromal cells are bidirectional and mediated through a variety of growth factors, cytokines, metabolites, and other biomolecules secreted by these cells. Tumor-stromal crosstalk creates optimal conditions for the tumor growth, metastasis, evasion of immune surveillance, and therapy resistance, and its targeting is being explored for clinical management of cancer. Natural agents from plants and marine life have been at the forefront of traditional medicine. Numerous epidemiological studies have reported the health benefits imparted on the consumption of certain fruits, vegetables, and their derived products. Indeed, a significant majority of anti-cancer drugs in clinical use are either naturally occurring compounds or their derivatives. In this review, we describe fundamental cellular and non-cellular components of the tumor microenvironment and discuss the significance of natural compounds in their targeting. Existing literature provides hope that novel prevention and therapeutic approaches will emerge from ongoing scientific efforts leading to the reduced tumor burden and improve clinical outcomes in cancer patients.
Collapse
Affiliation(s)
- Haseeb Zubair
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Mohammad Aslam Khan
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Shashi Anand
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Sanjeev Kumar Srivastava
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Seema Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Ajay Pratap Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, USA.
| |
Collapse
|
40
|
Bai X, Wang Y, Hu B, Cao Q, Xing M, Song S, Ji A. Fucoidan Induces Apoptosis of HT-29 Cells via the Activation of DR4 and Mitochondrial Pathway. Mar Drugs 2020; 18:E220. [PMID: 32326052 PMCID: PMC7231298 DOI: 10.3390/md18040220] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/04/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023] Open
Abstract
Fucoidan has a variety of pharmacological activities, but the understanding of the mechanism of fucoidan-induced apoptosis of colorectal cancer cells remains limited. The results of the present study demonstrated that the JNK signaling pathway is involved in the activation of apoptosis in colorectal cancer-derived HT-29 cells, and fucoidan induces apoptosis by activation of the DR4 at the transcriptional and protein levels. The survival rate of HT-29 cells was approximately 40% in the presence of 800 μg/mL of fucoidan, but was increased to 70% after DR4 was silenced by siRNA. Additionally, fucoidan has been shown to reduce the mitochondrial membrane potential and destroy the integrity of mitochondrial membrane. In the presence of an inhibitor of cytochrome C inhibitor and DR4 siRNA or the presence of cytochrome C inhibitor only, the cell survival rate was significantly higher than when cells were treated with DR4 siRNA only. These data indicate that both the DR4 and the mitochondrial pathways contribute to fucoidan-induced apoptosis of HT-29 cells, and the extrinsic pathway is upstream of the intrinsic pathway. In conclusion, the current work identified the mechanism of fucoidan-induced apoptosis and provided a novel theoretical basis for the future development of clinical applications of fucoidan as a drug.
Collapse
Affiliation(s)
- Xu Bai
- Marine College, Shandong University, Weihai 264209, China; (X.B.); (Y.W.); (B.H.); (Q.C.); (M.X.)
| | - Yu Wang
- Marine College, Shandong University, Weihai 264209, China; (X.B.); (Y.W.); (B.H.); (Q.C.); (M.X.)
| | - Bo Hu
- Marine College, Shandong University, Weihai 264209, China; (X.B.); (Y.W.); (B.H.); (Q.C.); (M.X.)
| | - Qi Cao
- Marine College, Shandong University, Weihai 264209, China; (X.B.); (Y.W.); (B.H.); (Q.C.); (M.X.)
| | - Maochen Xing
- Marine College, Shandong University, Weihai 264209, China; (X.B.); (Y.W.); (B.H.); (Q.C.); (M.X.)
| | - Shuliang Song
- Marine College, Shandong University, Weihai 264209, China; (X.B.); (Y.W.); (B.H.); (Q.C.); (M.X.)
| | - Aiguo Ji
- Marine College, Shandong University, Weihai 264209, China; (X.B.); (Y.W.); (B.H.); (Q.C.); (M.X.)
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| |
Collapse
|
41
|
Hsu WJ, Lin MH, Kuo TC, Chou CM, Mi FL, Cheng CH, Lin CW. Fucoidan from Laminaria japonica exerts antitumor effects on angiogenesis and micrometastasis in triple-negative breast cancer cells. Int J Biol Macromol 2020; 149:600-608. [PMID: 32004612 DOI: 10.1016/j.ijbiomac.2020.01.256] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 01/22/2020] [Accepted: 01/25/2020] [Indexed: 02/07/2023]
Abstract
Fucoidan is a fucose-rich polysaccharide that has gained attention for its various anticancer properties. However, the effect and underlying mechanism of fucoidan on triple-negative breast cancer (TNBC) are still unknown. Herein, we investigated the anticancer potential of fucoidan from Laminaria japonica. We found that fucoidan showed modest antiproliferative activity against TNBC cells, while it effectively reduced migratory and invasive capacities. Mechanistically, fucoidan suppressed activation of MAPK and PI3K followed by inhibition of AP-1 and NF-κB signaling in TNBC. Additionally, fucoidan downregulated expressions of proangiogenic factors in TNBC cells, and fucoidan blocked tumor-elicited tube formation by human umbilical vascular endothelial cells (HUVECs). We also observed that fucoidan blocked tumor adhesion and invasion towards HUVECs. Surprisingly, fucoidan robustly suppressed tube formation on HUVECs. Moreover, fucoidan inhibited in vivo angiogenesis and micrometastasis in a transgenic zebrafish model. Together, L. japonica fucoidan exhibits potent antitumor effects by its attenuation of invasiveness and proangiogenesis in TNBC.
Collapse
Affiliation(s)
- Wen-Jing Hsu
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Mei-Hsiang Lin
- Graduate Institute of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Tai-Chih Kuo
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Ming Chou
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Fwu-Long Mi
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Hsiung Cheng
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Cheng-Wei Lin
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
42
|
Gupta D, Silva M, Radziun K, Martinez DC, Hill CJ, Marshall J, Hearnden V, Puertas-Mejia MA, Reilly GC. Fucoidan Inhibition of Osteosarcoma Cells Is Species and Molecular Weight Dependent. Mar Drugs 2020; 18:E104. [PMID: 32046368 PMCID: PMC7074035 DOI: 10.3390/md18020104] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 12/11/2022] Open
Abstract
Fucoidan is a brown algae-derived polysaccharide having several biomedical applications. This study simultaneously compares the anti-cancer activities of crude fucoidans from Fucus vesiculosus and Sargassum filipendula, and effects of low (LMW, 10-50 kDa), medium (MMW, 50-100 kDa) and high (HMW, >100 kDa) molecular weight fractions of S. filipendula fucoidan against osteosarcoma cells. Glucose, fucose and acid levels were lower and sulphation was higher in F. vesiculosus crude fucoidan compared to S. filipendula crude fucoidan. MMW had the highest levels of sugars, acids and sulphation among molecular weight fractions. There was a dose-dependent drop in focal adhesion formation and proliferation of cells for all fucoidan-types, but F. vesiculosus fucoidan and HMW had the strongest effects. G1-phase arrest was induced by F. vesiculosus fucoidan, MMW and HMW, however F. vesiculosus fucoidan treatment also caused accumulation in the sub-G1-phase. Mitochondrial damage occurred for all fucoidan-types, however F. vesiculosus fucoidan led to mitochondrial fragmentation. Annexin V/PI, TUNEL and cytochrome c staining confirmed stress-induced apoptosis-like cell death for F. vesiculosus fucoidan and features of stress-induced necrosis-like cell death for S. filipendula fucoidans. There was also variation in penetrability of different fucoidans inside the cell. These differences in anti-cancer activity of fucoidans are applicable for osteosarcoma treatment.
Collapse
Affiliation(s)
- Dhanak Gupta
- Department of Materials Science and Engineering, University of Sheffield, Sheffield S1 3JD, UK; (D.G.); (K.R.); (D.C.M.); (V.H.)
- INSIGNEO Institute for in Silico Medicine, University of Sheffield, Sheffield S1 3JD, UK;
| | - Melissa Silva
- Institute of Chemistry, University of Antioquia, Medellín A.A.1226, Colombia; (M.S.); (M.A.P.-M.)
| | - Karolina Radziun
- Department of Materials Science and Engineering, University of Sheffield, Sheffield S1 3JD, UK; (D.G.); (K.R.); (D.C.M.); (V.H.)
- INSIGNEO Institute for in Silico Medicine, University of Sheffield, Sheffield S1 3JD, UK;
- Cell Bank, Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Diana C. Martinez
- Department of Materials Science and Engineering, University of Sheffield, Sheffield S1 3JD, UK; (D.G.); (K.R.); (D.C.M.); (V.H.)
| | - Christopher J. Hill
- Department of Molecular Biology and Biotechnology (MBB), University of Sheffield, Sheffield S10 2TN, UK;
| | - Julie Marshall
- INSIGNEO Institute for in Silico Medicine, University of Sheffield, Sheffield S1 3JD, UK;
| | - Vanessa Hearnden
- Department of Materials Science and Engineering, University of Sheffield, Sheffield S1 3JD, UK; (D.G.); (K.R.); (D.C.M.); (V.H.)
| | - Miguel A. Puertas-Mejia
- Institute of Chemistry, University of Antioquia, Medellín A.A.1226, Colombia; (M.S.); (M.A.P.-M.)
| | - Gwendolen C. Reilly
- Department of Materials Science and Engineering, University of Sheffield, Sheffield S1 3JD, UK; (D.G.); (K.R.); (D.C.M.); (V.H.)
- INSIGNEO Institute for in Silico Medicine, University of Sheffield, Sheffield S1 3JD, UK;
| |
Collapse
|
43
|
Bae H, Lee JY, Yang C, Song G, Lim W. Fucoidan Derived from Fucus vesiculosus Inhibits the Development of Human Ovarian Cancer via the Disturbance of Calcium Homeostasis, Endoplasmic Reticulum Stress, and Angiogenesis. Mar Drugs 2020; 18:E45. [PMID: 31936539 PMCID: PMC7024155 DOI: 10.3390/md18010045] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/05/2020] [Accepted: 01/06/2020] [Indexed: 12/22/2022] Open
Abstract
Marine organisms are sources of several natural compounds with potential clinical use. However, only a few marine-based pharmaceuticals have been approved for use due to limited knowledge on their biological activities. Here, we identified the functional role of fucoidan extracted from Fucus vesiculosus on ovarian cancer. Fucoidan increased the death of ES-2 and OV-90 cells, through a reduction in proliferation, cell cycle arrest, releases of cytochrome c, reactive oxygen species (ROS) generation, and endoplasmic reticulum (ER) stress. Additionally, fucoidan increased the concentration of cytosolic and mitochondrial calcium in both cells. The decrease of cell proliferation was controlled by the inactivation of PI3K and MAPK signaling cascades in ES-2 and OV-90 cells. In a toxicity assay with normal zebrafish larvae, fucoidan did not induce toxicity, cardiotoxicity, development, kinesis, and apoptosis at different concentrations. However, it disrupted tumor formation and vascular development in a zebrafish xenograft model and angiogenesis transgenic (Tg, fli1-eGFP) model, respectively. Collectively, the results indicate that fucoidan may be a novel pharmaceutical for the management of human ovarian cancer.
Collapse
Affiliation(s)
- Hyocheol Bae
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea; (H.B.); (C.Y.)
| | - Jin-Young Lee
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Changwon Yang
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea; (H.B.); (C.Y.)
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea; (H.B.); (C.Y.)
| | - Whasun Lim
- Department of Food and Nutrition, College of Science and Technology, Kookmin University, Seoul 02707, Korea
| |
Collapse
|
44
|
Torres M, Flórez-Fernández N, Simón-Vázquez R, Giménez-Abián J, Díaz J, González-Fernández Á, Domínguez H. Fucoidans: The importance of processing on their anti-tumoral properties. ALGAL RES 2020. [DOI: 10.1016/j.algal.2019.101748] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
45
|
Delma CR, Thirugnanasambandan S, Srinivasan GP, Raviprakash N, Manna SK, Natarajan M, Aravindan N. Fucoidan from marine brown algae attenuates pancreatic cancer progression by regulating p53 - NFκB crosstalk. PHYTOCHEMISTRY 2019; 167:112078. [PMID: 31450091 DOI: 10.1016/j.phytochem.2019.112078] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 06/10/2023]
Abstract
Poor pancreatic cancer (PC) prognosis has been attributed to its resistance to apoptosis and propensity for early systemic dissemination. Existing therapeutic strategies are often circumvented by the molecular crosstalk between cell-signalling pathways. p53 is mutated in more than 50% of PC and NFκB is constitutively activated in therapy-resistant residual disease; these mutations and activations account for the avoidance of cell death and metastasis. Recently, we demonstrated the anti-PC potential of fucoidan extract from marine brown alga, Turbinaria conoides (J. Agardh) Kützing (Sargassaceae). In this study, we aimed to characterize the active fractions of fucoidan extract to identify their select anti-PC efficacy, and to define the mechanism(s) involved. Five fractions of fucoidan isolated by ion exchange chromatography were tested for their potential in genetically diverse human PC cell lines. All fractions exerted significant dose-dependent and time-dependent regulation of cell survival. Fucoidans induced apoptosis, activated caspase -3, -8 and -9, and cleaved Poly ADP ribose polymerase (PARP). Pathway-specific transcriptional analysis recognized inhibition of 57 and 38 nuclear factor κB (NFκB) pathway molecules with fucoidan-F5 in MiaPaCa-2 and Panc-1 cells, respectively. In addition, fucoidan-F5 inhibited both the constitutive and Tumor necrosis factor-α (TNFα)-mediated NFκB DNA-binding activity in PC cells. Upregulation of cytoplasmic IκB levels and significant reduction of NFκB-dependent luciferase activity further substantiate the inhibitory potential of seaweed fucoidans on NFκB. Moreover, fucoidan(s) treatment increased cellular p53 in PC cells and reverted NFκB forced-expression-related p53 reduction. The results suggest that fucoidan regulates PC progression and that fucoidans may target p53-NFκB crosstalk and dictate apoptosis in PC cells.
Collapse
Affiliation(s)
- Caroline R Delma
- Centre of Advanced Study in Marine Biology, Faculty of Marine Sciences, Annamalai University, Parangipettai, TN, India; Department of Pathology, University of Texas Health Sciences Center at San Antonio, TX, USA.
| | | | - Guru Prasad Srinivasan
- Centre of Advanced Study in Marine Biology, Faculty of Marine Sciences, Annamalai University, Parangipettai, TN, India
| | - Nune Raviprakash
- Laboratory of Immunology, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, AP, India
| | - Sunil K Manna
- Laboratory of Immunology, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, AP, India
| | - Mohan Natarajan
- Department of Pathology, University of Texas Health Sciences Center at San Antonio, TX, USA
| | - Natarajan Aravindan
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
46
|
Juárez-Portilla C, Olivares-Bañuelos T, Molina-Jiménez T, Sánchez-Salcedo JA, Moral DID, Meza-Menchaca T, Flores-Muñoz M, López-Franco Ó, Roldán-Roldán G, Ortega A, Zepeda RC. Seaweeds-derived compounds modulating effects on signal transduction pathways: A systematic review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 63:153016. [PMID: 31325683 DOI: 10.1016/j.phymed.2019.153016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Recently, the study of marine natural products has gained interest due to their relevant biological activities. Specially, seaweeds produce bioactive compounds that could act as modulators of cell signaling pathways involved in a plethora of diseases. Thereby, the description of the molecular mechanisms by which seaweeds elicit its biological functions will certainly pave the way to the pharmacological development of drugs. AIM This review describes the molecular mechanisms by which seaweeds act and its possible utilization in the design of new drugs. METHODS This review was conducted according to the PRISMA-P guidelines for systematic reviews. Two independent authors searched into four different databases using combinations of keywords. Two more authors selected the articles following the eligibility criteria. Information extraction was conducted by two separated authors and entered into spreadsheets. Methodological quality and risk of bias were determined applying a 12-question Risk of Bias criteria tool. RESULTS AND DISCUSSION We found 2360 articles (SCOPUS: 998; PubMed: 678; Wiley: 645 and EBSCO: 39) using the established keywords, of which 113 articles fit the inclusion criteria and were included in the review. This work comprises studies in cell lines, and animal models, any clinical trial was excluded. The articles were published from 2005 up to March 31st 2018. The biggest amount of articles was published in 2017. Furthermore, the seaweeds tested in the studies were collected in 15 countries, mainly in Eastern countries. We found that the main modulated signaling pathways by seaweeds-derivate extracts and compounds were: L-Arginine/NO, TNF-α, MAPKs, PI3K/AKT/GSK, mTOR, NF-κB, extrinsic and intrinsic apoptosis, cell cycle, MMPs and Nrf2. Finally, the articles we analyzed showed moderate risk of bias in almost all the parameters evaluated. However, the studies fail to describe the place and characteristics of sample collection, the sample size, and the blindness of the experimental design. CONCLUSION In this review we identified and summarized relevant information related to seaweed-isolated compounds and extracts having biological activity; their role in different signal pathways to better understand their potential to further development of cures for cancer, diabetes, and inflammation-related diseases.
Collapse
Affiliation(s)
- Claudia Juárez-Portilla
- Centro de Investigaciones Biomédicas, Universidad Veracruzana. Av. Dr. Luis Castelazo Ayala s/n. Col. Industrial Ánimas, C.P. 91190, Xalapa, Veracruz, México
| | - Tatiana Olivares-Bañuelos
- Instituto de Investigaciones Oceanológicas, Universidad Autónoma de Baja California. Km 103 autopista Tijuana-Ensenada, A.P. 453. Ensenada, Baja California, México
| | - Tania Molina-Jiménez
- Facultad de Química Farmacéutica Biológica, Universidad Veracruzana. Circuito Gonzalo Aguirre Beltrán s/n. Zona Universitaria, C.P. 91000, Xalapa, Veracruz, México
| | - José Armando Sánchez-Salcedo
- Programa de Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana. Av. San Rafael Atlixco No. 186, Col. Vicentina, C.P. 09340, Iztapalapa, Ciudad de México
| | - Diana I Del Moral
- Programa de Doctorado en Ciencias Biomédicas, Universidad Veracruzana. Av. Dr. Luis Castelazo Ayala s/n. Col. Industrial Ánimas, C.P. 91190, Xalapa, Veracruz, México
| | - Thuluz Meza-Menchaca
- Laboratorio de Genómica Humana, Facultad de Medicina, Universidad Veracruzana. Médicos y Odontólogos s/n. Col. Unidad del Bosque, C.P. 91010, Xalapa, Veracruz, México
| | - Mónica Flores-Muñoz
- Instituto de Ciencias de la Salud, Universidad Veracruzana. Av. Dr. Luis Castelazo Ayala s/n. Col. Industrial Ánimas, C.P. 91190, Xalapa, Veracruz, México
| | - Óscar López-Franco
- Instituto de Ciencias de la Salud, Universidad Veracruzana. Av. Dr. Luis Castelazo Ayala s/n. Col. Industrial Ánimas, C.P. 91190, Xalapa, Veracruz, México
| | - Gabriel Roldán-Roldán
- Laboratorio de Neurobiología Conductual, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Arturo Ortega
- Laboratorio de Neurotoxicología, Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, A.P. 14-740, 07300, Ciudad de México, México
| | - Rossana C Zepeda
- Centro de Investigaciones Biomédicas, Universidad Veracruzana. Av. Dr. Luis Castelazo Ayala s/n. Col. Industrial Ánimas, C.P. 91190, Xalapa, Veracruz, México.
| |
Collapse
|
47
|
Wang J, Zhang Y, Liu X, Wang J, Li B, Liu Y, Wang J. Alantolactone enhances gemcitabine sensitivity of lung cancer cells through the reactive oxygen species-mediated endoplasmic reticulum stress and Akt/GSK3β pathway. Int J Mol Med 2019; 44:1026-1038. [PMID: 31524219 PMCID: PMC6657978 DOI: 10.3892/ijmm.2019.4268] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 06/18/2019] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is one of the leading causes of cancer‑associated mortality in China and globally. Gemcitabine (GEM), as a first‑line therapeutic drug, has been used to treat lung cancer, but GEM resistance poses a major limitation on the efficacy of GEM chemotherapy. Alantolactone (ALT), a sesquiterpene lactone compound isolated from Inula helenium, has been identified to exert anticancer activity in various types of cancer, including breast, pancreatic, lung squamous and colorectal cancer. However, the underlying mechanisms of the anticancer activity of ALT in lung cancer remain to be fully elucidated. The present study aimed to determine whether ALT enhances the anticancer efficacy of GEM in lung cancer cells and investigated the underlying mechanisms. The cell viability was assessed with a Cell Counting Kit‑8 assay. The cell cycle, apoptosis and the level of reactive oxygen species (ROS) were assessed by flow cytometry, and the expression of cell cycle‑associated and apoptosis‑associated proteins were determined by western blot analysis. The results demonstrated that ALT inhibited cell growth and induced S‑phase arrest and cell apoptosis in A549 and NCI‑H520 cells. Furthermore, ALT increased the level of ROS, inhibited the Akt/glycogen synthase kinase (GSK)3β pathway and induced endoplasmic reticulum (ER) stress in A549 and NCI‑H520 cells. Additionally, ALT treatment sensitized lung cancer cells to GEM. Analysis of the molecular mechanisms further revealed that ALT enhanced the anticancer effects of GEM via ROS‑mediated activation of the Akt/GSK3β and ER stress pathways. In conclusion, combined treatment with ALT and GEM may have potential as a clinical strategy for lung cancer treatment.
Collapse
Affiliation(s)
| | | | - Xu Liu
- Department of Thoracic Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061
| | - Jizhao Wang
- Department of Thoracic Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061
| | - Bin Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Yongkang Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Jiansheng Wang
- Department of Thoracic Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061
| |
Collapse
|
48
|
S-ethyl ethanethiosulfinate, a derivative of allicin, induces metacaspase-dependent apoptosis through ROS generation in Penicillium chrysogenum. Biosci Rep 2019; 39:BSR20190167. [PMID: 31142631 PMCID: PMC6567679 DOI: 10.1042/bsr20190167] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 05/15/2019] [Accepted: 05/23/2019] [Indexed: 01/14/2023] Open
Abstract
Allicin can be used as fumigant to protect food and cultural relics from fungal contamination because of its strong antifungal activity and the characteristics of high volatility and no residues. However, the obvious disadvantages such as high minimal inhibitory concentration and instability prevent it from wide application. In this study, a stable derivative of allicin, S-ethyl ethanethiosulfinate (ALE), was synthesized. We further explored its antifungal activity and apoptosis-inducing effect, as well as the underlying mechanism. ALE had an excellent capability of inhibiting spore germination and mycelial growth of Penicillium chrysogenum observed by inverted microscope and scanning electron microscopy. XTT colorimetric assay indicated ALE could reduce the cell viability obviously and IC50 was 0.92 μg/ml, only 1/42 of allicin (38.68 μg/ml). DHR 123 ROS Assay Kit, flow cytometry assay and confocal immunofluorescence revealed intercellular ROS generation and metacaspase-dependent apoptosis triggered by ALE, while antioxidant tocopherol could reverse ALE-induced cytotoxicity effect and metacaspase activation. These results indicate that ALE induces metacaspase-dependent apoptosis through ROS generation, thus possesses an effective antifungal activity. This new derivative of allicin might be developed as a high efficient alternative to the conventional fungicides for food storage and cultural relic protection.
Collapse
|
49
|
Li W, Xue D, Xue M, Zhao J, Liang H, Liu Y, Sun T. Fucoidan inhibits epithelial-to-mesenchymal transition via regulation of the HIF-1α pathway in mammary cancer cells under hypoxia. Oncol Lett 2019; 18:330-338. [PMID: 31289504 DOI: 10.3892/ol.2019.10283] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 03/07/2019] [Indexed: 01/02/2023] Open
Abstract
This study examined the effects of fucoidan on epithelial-to-mesenchymal transition (EMT) in a human triple-negative breast cancer (TNBC) cell line in a hypoxic microenvironment. Transwell and wound-healing assays were performed to analyze the invasion and migration of MDA-MB-231 human mammary cancer cells, respectively. The expression levels of EMT markers and hypoxia-inducible factor-1α (HIF-1α) were detected through western blotting. Under hypoxia, fucoidan treatment inhibited proliferation of breast cancer cells. Fucoidan also suppressed the invasion and migration of MDA-MB-231 cells. Western blotting revealed that fucoidan treatment significantly reduced the protein expression levels of HIF-1α and HIF-1 target genes. Furthermore, the nuclear translocation and activity of HIF-1α were reduced. Fucoidan treatment significantly downregulated the expression levels of mesenchymal markers (N-cadherin and vimentin), but upregulated the expression levels of the epithelial markers zonula occludens-1 and E-cadherin. In addition, overexpression of HIF1-α protected cells from fucoidan-mediated suppression of migration and invasion. These data suggested that fucoidan may inhibit EMT in human TNBC cells via downregulation of the HIF1-α signaling pathway.
Collapse
Affiliation(s)
- Weiwei Li
- Department of Biochemistry and Molecular Biology, Basic Medical College, Qingdao University of Medicine, Qingdao, Shandong 266021, P.R. China
| | - Dingshan Xue
- Department of Senior Grade Three, Qingdao West Coast District No. 1 Senior High School, Qingdao, Shandong 266555, P.R. China
| | - Meilan Xue
- Department of Biochemistry and Molecular Biology, Basic Medical College, Qingdao University of Medicine, Qingdao, Shandong 266021, P.R. China
| | - Jinglan Zhao
- Department of Cardiothoracic Surgery of Qingdao Center Medical Group, Qingdao, Shandong 266042, P.R. China
| | - Hui Liang
- The Institute of Human Nutrition, Qingdao University of Medicine, Qingdao, Shandong 266021, P.R. China
| | - Ying Liu
- Department of Biochemistry and Molecular Biology, Basic Medical College, Qingdao University of Medicine, Qingdao, Shandong 266021, P.R. China
| | - Ting Sun
- Department of Biochemistry and Molecular Biology, Basic Medical College, Qingdao University of Medicine, Qingdao, Shandong 266021, P.R. China
| |
Collapse
|
50
|
Hong Y, Fan D. Ginsenoside Rk1 induces cell cycle arrest and apoptosis in MDA-MB-231 triple negative breast cancer cells. Toxicology 2019; 418:22-31. [DOI: 10.1016/j.tox.2019.02.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/01/2019] [Accepted: 02/19/2019] [Indexed: 12/13/2022]
|