1
|
Kaur M, Shitanaka T, Surendra KC, Khanal SK. Macroalgae-derived bioactive compounds for functional food and pharmaceutical applications-a critical review. Crit Rev Food Sci Nutr 2024:1-23. [PMID: 39078214 DOI: 10.1080/10408398.2024.2384643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
The rising demand for global food resources, combined with an overreliance on land-based agroecosystems, poses a significant challenge for the sustainable production of food products. Macroalgae cultivation is a promising approach to mitigate impending global food insecurities due to several key factors: independence from terrestrial farming, rapid growth rates, unique biochemical makeup, and carbon capture potential. Furthermore, macroalgae are rich in vitamins, minerals, essential amino acids, polyunsaturated fatty acids and fiber, demonstrating significant potential as sustainable alternatives for enhancing dietary diversity and fulfilling nutritional requirements. This review provides an overview of the nutritional composition and functional properties of commercially cultivated macroalgae species, with emphasis on their viability as value additions to the functional food market. Furthermore, the review discusses the technological aspects of integrating macroalgae into food products, covering both innovative solutions and existing challenges. Macroalgae, beyond being nutritional powerhouses, contain a plethora of bioactive compounds with varied biological activities, including anti-diabetic, anti-cancer, cardioprotective, and neuroprotective properties, making them excellent candidates in developing novel pharmaceuticals. Thus, this review also summarizes the pharmaceutical applications of macroalgae, identifies research gaps and proposes potential strategies for incorporating macroalgae-derived bioactive compounds into therapeutic products.
Collapse
Affiliation(s)
- Manpreet Kaur
- Department of Molecular Biosciences and Bioengineering, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - Ty Shitanaka
- Department of Molecular Biosciences and Bioengineering, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - K C Surendra
- Department of Molecular Biosciences and Bioengineering, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - Samir Kumar Khanal
- Department of Molecular Biosciences and Bioengineering, University of Hawai'i at Mānoa, Honolulu, HI, USA
- Department of Environmental Engineering, Korea University Sejong Campus, Sejong, Korea
| |
Collapse
|
2
|
Chen J, Ding Z. Natural products as potential drug treatments for acute promyelocytic leukemia. Chin Med 2024; 19:57. [PMID: 38566147 PMCID: PMC10988969 DOI: 10.1186/s13020-024-00928-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024] Open
Abstract
Acute promyelocytic leukemia (APL), which was once considered one of the deadliest types of leukemia, has become a curable malignancy since the introduction of all-trans retinoic acid (ATRA) and arsenic trioxide (ATO) as clinical treatments. ATO, which has become the first-line therapeutic agent for APL, is derived from the natural mineral product arsenic, exemplifying an important role of natural products in the treatment of APL. Many other natural products, ranging from small-molecule compounds to herbal extracts, have also demonstrated great potential for the treatment and adjuvant therapy of APL. In this review, we summarize the natural products and representative components that have demonstrated biological activity for the treatment of APL. We also discuss future directions in better exploring their medicinal value, which may provide a reference for subsequent new drug development and combination therapy programs.
Collapse
Affiliation(s)
- Jiaxin Chen
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, China
| | - Zuoqi Ding
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, China.
- Editorial Board of Chinese Journal of Natural Medicines, Nanjing, China.
| |
Collapse
|
3
|
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
|
4
|
Carvajal-Barriga EJ, Fields RD. Sulfated polysaccharides as multi target molecules to fight COVID 19 and comorbidities. Heliyon 2023; 9:e13797. [PMID: 36811015 PMCID: PMC9936785 DOI: 10.1016/j.heliyon.2023.e13797] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/19/2023] Open
Abstract
The majority of research to combat SARS-CoV-2 infection exploits the adaptive immune system, but innate immunity, the first line of defense against pathogenic microbes, is equally important in understanding and controlling infectious diseases. Various cellular mechanisms provide physiochemical barriers to microbe infection in mucosal membranes and epithelia, with extracellular polysaccharides, particularly sulfated polysaccharides, being among the most widespread and potent extracellular and secreted molecules blocking and deactivating bacteria, fungi, and viruses. New research reveals that a range of polysaccharides effectively inhibits COV-2 infection of mammalian cells in culture. This review provides an overview of sulfated polysaccharides nomenclature, its significance as immunomodulators, antioxidants, antitumors, anticoagulants, antibacterial, and as potent antivirals. It summarizes current research on various interactions of sulfated polysaccharide with a range of viruses, including SARS-CoV-2, and their application for potential treatments for COVID-19. These molecules interact with biochemical signaling in immune cell responses, by actions in oxidative reactions, cytokine signaling, receptor binding, and through antiviral and antibacterial toxicity. These properties provide the potential for the development of novel therapeutic treatments for SARS-CoV-2 and other infectious diseases from modified polysaccharides.
Collapse
Affiliation(s)
- Enrique Javier Carvajal-Barriga
- Pontificia Universidad Católica Del Ecuador, Neotropical Center for the Biomass Research, Quito, Ecuador.,The Eunice Kennedy Shriver National Institutes of Health, National Institute of Children and Human Development, Bethesda, MD, USA
| | - R Douglas Fields
- The Eunice Kennedy Shriver National Institutes of Health, National Institute of Children and Human Development, Bethesda, MD, USA
| |
Collapse
|
5
|
Characteristics of Marine Biomaterials and Their Applications in Biomedicine. Mar Drugs 2022; 20:md20060372. [PMID: 35736175 PMCID: PMC9228671 DOI: 10.3390/md20060372] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/21/2022] [Accepted: 05/27/2022] [Indexed: 02/04/2023] Open
Abstract
Oceans have vast potential to develop high-value bioactive substances and biomaterials. In the past decades, many biomaterials have come from marine organisms, but due to the wide variety of organisms living in the oceans, the great diversity of marine-derived materials remains explored. The marine biomaterials that have been found and studied have excellent biological activity, unique chemical structure, good biocompatibility, low toxicity, and suitable degradation, and can be used as attractive tissue material engineering and regenerative medicine applications. In this review, we give an overview of the extraction and processing methods and chemical and biological characteristics of common marine polysaccharides and proteins. This review also briefly explains their important applications in anticancer, antiviral, drug delivery, tissue engineering, and other fields.
Collapse
|
6
|
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
|
7
|
Sun T, Xue M, Yang J, Pei Z, Zhang N, Qin K, Liang H. Metabolic regulation mechanism of fucoidan via intestinal microecology in diseases. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:4456-4463. [PMID: 33682122 DOI: 10.1002/jsfa.11202] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/23/2021] [Accepted: 03/07/2021] [Indexed: 06/12/2023]
Abstract
The intestinal microecology is an extremely complex ecosystem consisting of gut microbiota, intestinal mucosa and the intestinal immune system. The intestinal microecology performs several important functions and is considered to be an essential 'organ' because it plays an important role in regulating human metabolism. Fucoidan contains a large amount of fucose and galactose residues, as well as various other neutral and acidic monosaccharides. Fucoidan particularly effects tumors, inflammatory bowel disease, diabetes and obesity by repairing intestinal mucosal damage and improving the intestinal microecological environment. It has been proposed that fucoidan could be used as a prebiotic agent for pharmaceutical and functional foods. In this review, we elucidate the potential mechanisms of the metabolic regulation of fucoidan with respect to the intestinal microecology of diseases. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Ting Sun
- Basic Medical College, Qingdao University of Medicine, Qingdao, China
| | - Meilan Xue
- Basic Medical College, 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
| | - Nan Zhang
- Basic Medical College, Qingdao University of Medicine, Qingdao, China
| | - Kunpeng Qin
- Basic Medical College, Qingdao University of Medicine, Qingdao, China
| | - Hui Liang
- Department of Human Nutrition, College of Public Health, Qingdao University of Medicine, Qingdao, China
| |
Collapse
|
8
|
Yang J, Yang X, Pan W, Wang M, Lu Y, Zhang J, Fang Z, Zhang X, Ji Y, Bei JX, Dong J, Wu Y, Pan C, Yu G, Zhou P, Li B. Fucoidan-Supplemented Diet Potentiates Immune Checkpoint Blockage by Enhancing Antitumor Immunity. Front Cell Dev Biol 2021; 9:733246. [PMID: 34434936 PMCID: PMC8382313 DOI: 10.3389/fcell.2021.733246] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 07/20/2021] [Indexed: 11/19/2022] Open
Abstract
Immune checkpoint blockade (ICB) therapies such as PD-1 antibodies have produced significant clinical responses in treating a variety of human malignancies, yet only a subset of cancer patients benefit from such therapy. To improve the ICB efficacy, combinations with additional therapeutics were under intensive investigation. Recently, special dietary compositions that can lower the cancer risk or inhibit cancer progression have drawn significant attention, although few were reported to show synergistic effects with ICB therapies. Interestingly, Fucoidan is naturally derived from edible brown algae and exhibits antitumor and immunomodulatory activities. Here we discover that fucoidan-supplemented diet significantly improves the antitumor activities of PD-1 antibodies in vivo. Specifically, fucoidan as a dietary ingredient strongly inhibits tumor growth when co-administrated with PD-1 antibodies, which effects can be further strengthened when fucoidan is applied before PD-1 treatments. Immune analysis revealed that fucoidan consistently promotes the activation of tumor-infiltrating CD8+ T cells, which support the evident synergies with ICB therapies. RNAseq analysis suggested that the JAK-STAT pathway is critical for fucoidan to enhance the effector function of CD8+ T cells, which could be otherwise attenuated by disruption of the T-cell receptor (TCR)/CD3 complex on the cell surface. Mechanistically, fucoidan interacts with this complex and augments TCR-mediated signaling that cooperate with the JAK-STAT pathway to stimulate T cell activation. Taken together, we demonstrated that fucoidan is a promising dietary supplement combined with ICB therapies to treat malignancies, and dissected an underappreciated mechanism for fucoidan-elicited immunomodulatory effects in cancer.
Collapse
Affiliation(s)
- Juan Yang
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangdong, China
| | - Xianzhi Yang
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Wenfeng Pan
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Mingshuo Wang
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Yuxiong Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangdong, China.,Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jianeng Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangdong, China
| | - Ziqian Fang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangdong, China
| | - Xiaomin Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangdong, China
| | - Yin Ji
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Simcere Diagnostics Co., Ltd., Jiangsu, China
| | - Jin-Xin Bei
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangdong, China.,Center for Precision Medicine, Sun Yat-sen University, Guangdong, China
| | - Jiajun Dong
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Yi Wu
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Chaoyun Pan
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Guangli Yu
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Shandong, China.,Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Shandong, China
| | - Penghui Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangdong, China
| | - Bo Li
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangdong, China.,Center for Precision Medicine, Sun Yat-sen University, Guangdong, China
| |
Collapse
|
9
|
Brown Seaweed Food Supplementation: Effects on Allergy and Inflammation and Its Consequences. Nutrients 2021; 13:nu13082613. [PMID: 34444774 PMCID: PMC8398742 DOI: 10.3390/nu13082613] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/15/2021] [Accepted: 07/27/2021] [Indexed: 12/11/2022] Open
Abstract
Multiple health benefits have been ascribed to brown seaweeds that are used traditionally as dietary component mostly in Asia. This systematic review summarizes information on the impact of brown seaweeds or components on inflammation, and inflammation-related pathologies, such as allergies, diabetes mellitus and obesity. We focus on oral supplementation thus intending the use of brown seaweeds as food additives. Despite the great diversity of experimental systems in which distinct species and compounds were tested for their effects on inflammation and immunity, a remarkably homogeneous picture arises. The predominant effects of consumption of brown seaweeds or compounds can be classified into three categories: (1) inhibition of reactive oxygen species, known to be important drivers of inflammation; (2) regulation, i.e., in most cases inhibition of proinflammatory NF-κB signaling; (3) modulation of adaptive immune responses, in particular by interfering with T-helper cell polarization. Over the last decades, several inflammation-related diseases have increased substantially. These include allergies and autoimmune diseases as well as morbidities associated with lifestyle and aging. In this light, further development of brown seaweeds and seaweed compounds as functional foods and nutriceuticals might contribute to combat these challenges.
Collapse
|
10
|
Fonseca RJC, Mourão PAS. Pharmacological Activities of Sulfated Fucose-Rich Polysaccharides after Oral Administration: Perspectives for the Development of New Carbohydrate-Based Drugs. Mar Drugs 2021; 19:425. [PMID: 34436263 PMCID: PMC8400256 DOI: 10.3390/md19080425] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/08/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Marine organisms are a source of active biomolecules with immense therapeutic and nutraceutical potential. Sulfated fucose-rich polysaccharides are present in large quantities in these organisms with important pharmacological effects in several biological systems. These polysaccharides include sulfated fucan (as fucoidan) and fucosylated chondroitin sulfate. The development of these polysaccharides as new drugs involves several important steps, among them, demonstration of the effectiveness of these compounds after oral administration. The oral route is the more practical, comfortable and preferred by patients for long-term treatments. In the past 20 years, reports of various pharmacological effects of these polysaccharides orally administered in several animal experimental models and some trials in humans have sparked the possibility for the development of drugs based on sulfated polysaccharides and/or the use of these marine organisms as functional food. This review focuses on the main pharmacological effects of sulfated fucose-rich polysaccharides, with an emphasis on the antidislipidemic, immunomodulatory, antitumor, hypoglycemic and hemostatic effects.
Collapse
Affiliation(s)
- Roberto J. C. Fonseca
- Laboratório de Tecido Conjuntivo, Hospital Universitário Clementino Fraga Filho, Rio de Janeiro 21941-913, Brazil;
- Centro de Ciências da Saúde, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Paulo A. S. Mourão
- Laboratório de Tecido Conjuntivo, Hospital Universitário Clementino Fraga Filho, Rio de Janeiro 21941-913, Brazil;
- Centro de Ciências da Saúde, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| |
Collapse
|
11
|
Jin JO, Chauhan PS, Arukha AP, Chavda V, Dubey A, Yadav D. The Therapeutic Potential of the Anticancer Activity of Fucoidan: Current Advances and Hurdles. Mar Drugs 2021; 19:265. [PMID: 34068561 PMCID: PMC8151601 DOI: 10.3390/md19050265] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/04/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023] Open
Abstract
Several types of cancers share cellular and molecular behaviors. Although many chemotherapy drugs have been designed to weaken the defenses of cancer cells, these drugs may also have cytotoxic effects on healthy tissues. Fucoidan, a sulfated fucose-based polysaccharide from brown algae, has gained much attention as an antitumor drug owing to its anticancer effects against multiple cancer types. Among the anticancer mechanisms of fucoidan are cell cycle arrest, apoptosis evocation, and stimulation of cytotoxic natural killer cells and macrophages. Fucoidan also protects against toxicity associated with chemotherapeutic drugs and radiation-induced damage. The synergistic effect of fucoidan with existing anticancer drugs has prompted researchers to explore its therapeutic potential. This review compiles the mechanisms through which fucoidan slows tumor growth, kills cancer cells, and interacts with cancer chemotherapy drugs. The obstacles involved in developing fucoidan as an anticancer agent are also discussed in this review.
Collapse
Affiliation(s)
- Jun-O. Jin
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 201508, China
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| | - Pallavi Singh Chauhan
- Amity Institute of Biotechnology, Amity University Madhya Pradesh, Gwalior 474005, India;
| | - Ananta Prasad Arukha
- Comparative Diagnostic and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA;
| | - Vishal Chavda
- Division of Anaesthesia, Sardar Women’s Hospital, Ahmedabad 380004, Gujarat, India;
| | - Anuj Dubey
- Department of Chemistry, ITM Group of Institutions, Gwalior 475005, India;
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea
| |
Collapse
|
12
|
Kokoulin MS, Kuzmich AS, Romanenko LA, Chikalovets IV. Structure and in vitro antiproliferative activity of the acidic capsular polysaccharide from the deep-sea bacterium Psychrobacter submarinus KMM 225 T. Carbohydr Polym 2021; 262:117941. [PMID: 33838818 DOI: 10.1016/j.carbpol.2021.117941] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/27/2022]
Abstract
Psychrobacter submarinus KMM 225T is a Gram-negative bacterium isolated from a sea-water sample collected at a depth of 300 m in the Northwest Pacific Ocean. Here we report the structure of the capsular polysaccharide from P. submarinus KMM 225T and its effect on the viability and colony formation of cancer cells. The glycopolymer was purified by ultracentrifugation and chromatography methods, and the structure was elucidated using NMR spectroscopy and composition analyses. The following structure of the acidic capsular polysaccharide, containing 2-acetamido-2,4,6-trideoxy-4-[(S)-3-hydroxybutyramido]-d-glucose [d-QuipNAc4N(S-Hb)] and 4,6-O-[(S)-1-carboxyethylidene]-2-acetamido-2-deoxy-d-glucose [d-GlcpNAc4,6(S-Pyr)] was established: The capsular polysaccharide slightly reduced the viability but effectively suppressed the colony formation of different types of cancer cells, of which the most pronounced inhibition was shown for the human chronic myelogenous leukemia K-562 cells.
Collapse
Affiliation(s)
- Maxim S Kokoulin
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159/2, Prospect 100 let Vladivostoku, Vladivostok, 690022, Russia.
| | - Alexandra S Kuzmich
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159/2, Prospect 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Lyudmila A Romanenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159/2, Prospect 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Irina V Chikalovets
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159/2, Prospect 100 let Vladivostoku, Vladivostok, 690022, Russia; Far Eastern Federal University, 8, Sukhanova Str., Vladivostok, 690950, Russia
| |
Collapse
|
13
|
Chantree P, Na-Bangchang K, Martviset P. Anticancer Activity of Fucoidan via Apoptosis and Cell Cycle Arrest on Cholangiocarcinoma Cell. Asian Pac J Cancer Prev 2021; 22:209-217. [PMID: 33507701 PMCID: PMC8184191 DOI: 10.31557/apjcp.2021.22.1.209] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/15/2021] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Many previous studies reported that fucoidan has antitumor activities. The objective of the present study was to determine the cytotoxic effects and related mechanisms of cell death induced by fucoidan extracted from Fucus vesiculosus on CL-6 cholangiocarcinoma cell. METHODS CL-6 and OUMS cells were treated with 0, 100, 200, and 300 μg/mL of fucoidan. MTT assay was used to determine cytotoxicity. Flow cytometry-based assay was used to examine the distribution of apoptosis and cell cycle. The changes in nuclear morphology were determined using Hoechst 33,342 staining. Mitochondrial membrane potential (ΔΨm) was evaluated using the JC-1 kit. The apoptotic, anti-apoptotic, and cell cycle-related proteins study were examined by Western blot analysis. RESULTS The relative viable cell number of treated CL-6 cells was decreased but no effect was observed in OUMS normal cells. Furthermore, treated cells were arrested in the G0/G1 phase with down-regulation of cyclin D1 and CDK4. Annexin V/PI staining with flow cytometry analysis suggested that fucoidan could induce apoptosis in CL-6 cells. Western blot study revealed the up-regulation of apoptotic markers including Bax, cleaved PARP, cleaved caspase-3, but down-regulation of anti-apoptotic markers, cl-2. Moreover, fucoidan could induce nuclear fragmentation and chromatin condensation with alteration of ΔΨm. Conclusion: Fucoidan exerts antitumor properties against CL-6 cholangiocarcinoma cells illustrated by the induction of apoptosis and cell cycle arrest. .
Collapse
Affiliation(s)
- Pathanin Chantree
- Division of Anatomy, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand.
- Research Unit in Nutraceuticals and Food Safety, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand.
| | - Kesara Na-Bangchang
- Graduate Program in Bioclinical Sciences, Chulabhorn International College of Medicine, Thammasat University, Pathumthani, 12120, Thailand.
- Center of Excellence in Molecular Biology and Pharmacology of Malaria and Cholangiocarcinoma, Thammasat University, Pathumthani, 12120, Thailand.
| | - Pongsakorn Martviset
- Center of Excellence in Molecular Biology and Pharmacology of Malaria and Cholangiocarcinoma, Thammasat University, Pathumthani, 12120, Thailand.
- Division of Parasitology, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand.
| |
Collapse
|
14
|
Luo T, Gao J, Lin N, Wang J. Effects of Two Kinds of Iron Nanoparticles as Reactive Oxygen Species Inducer and Scavenger on the Transcriptomic Profiles of Two Human Leukemia Cells with Different Stemness. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1951. [PMID: 33007950 PMCID: PMC7600526 DOI: 10.3390/nano10101951] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/27/2020] [Accepted: 09/28/2020] [Indexed: 12/26/2022]
Abstract
Leukemia is a common and lethal disease. In recent years, iron-based nanomedicines have been developed as a new ferroptosis inducer to leukemia. However, the cytotoxicity of iron nanoparticles to leukemia cells at the transcriptomic level remains unclear. This study investigated the effects of two kinds of iron nanoparticles, 2,3-Dimercaptosuccinic acid (DMSA)-coated Fe3O4 nanoparticles (FeNPs) as a reactive oxygen species (ROS) inducer and Prussian blue nanoparticles (PBNPs) as an ROS scavenger, on the transcriptomic profiles of two leukemia cells (KG1a and HL60) by RNA-Seq. As a result, 470 and 1690 differentially expressed genes (DEGs) were identified in the FeNP-treated HL60 and KG1a cells, respectively, and 2008 and 2504 DEGs were found in the PBNP-treated HL60 and KG1a cells, respectively. Among them, 14 common upregulated and 4 common downregulated DEGs were found, these genes were representative genes that play key roles in lipid metabolism (GBA and ABCA1), iron metabolism (FTL, DNM1, and TRFC), antioxidation (NQO1, GCLM, and SLC7A11), vesicle traffic (MCTP2, DNM1, STX3, and BIN2), and innate immune response (TLR6, ADGRG3, and DDX24). The gene ontology revealed that the mineral absorption pathway was significantly regulated by PBNPs in two cells, whereas the lipid metabolism and HIF-1 signaling pathways were significantly regulated by FeNPs in two cells. This study established the gene signatures of two kinds of nanoparticles in two leukemia cells, which revealed the main biological processes regulated by the two kinds of iron nanoparticles. These data shed new insights into the cytotoxicity of iron nanoparticles that differently regulate ROS in leukemia cells with variant stemness.
Collapse
Affiliation(s)
| | | | | | - Jinke Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China; (T.L.); (J.G.); (N.L.)
| |
Collapse
|
15
|
Chantree P, Surarak T, Sangpairoj K, Aguilar P, Hitakomate E. Antitumor Effects of Fucoidan Via Apoptotic and Autophagic Induction on HSC-3 Oral Squamous CellCarcinoma. Asian Pac J Cancer Prev 2020; 21:2469-2477. [PMID: 32856880 PMCID: PMC7771925 DOI: 10.31557/apjcp.2020.21.8.2469] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
Objective: Many studies suggested that fucoidan has anticancer potential. The objective of the present study was to determine the cytotoxic effects and mechanism of cell death induced by fucoidan extracted from Fucus vesiculosus on HSC-3 oral squamous cell carcinoma. Methods: HSC-3 cells were treated with 0, 100, 200, and 400 μg/mL of fucoidan. Cell viability was measured using MTT assay. Apoptosis and cell cycle were measured with a flow cytometry-based assay. Chromatin condensation and nuclear fragmentation were determined using Hoechst 33342 staining. Mitochondrial membrane potential (ΔΨm) was determined using the JC-1 kit. The apoptotic, anti-apoptotic, and autophagic markers study were done by western blot analysis. Results: the viable cell number of treated HSC-3 cells was decreased. Moreover, treated cells were arrested in the G0/G1 phase. Annexin V/PI staining revealed that fucoidan could induce apoptosis in HSC-3 cells. Western blot analysis suggested the up-regulation of apoptotic markers including cleaved caspase-3, cleaved PARP, Bax, and autophagic markers including LC3-II and Beclin-1 but down-regulation of anti-apoptotic markers, Bcl-2. Fucoidan could disturb ΔΨm and induce chromatin condensation with nuclear fragmentation. Conclusion: fucoidan has potential in anticancer properties against HSC-3 cells manifested by the induction of apoptosis, cell cycle arrest, and autophagy.
Collapse
Affiliation(s)
- Pathanin Chantree
- Division of Anatomy, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand
| | - Thanakorn Surarak
- Division of Pharmacology, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand
| | - Kant Sangpairoj
- Division of Anatomy, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand
| | - Panuroot Aguilar
- Faculty of Dentistry, Thammasat University, Pathumthani, 12120, Thailand
| | - Ekarat Hitakomate
- Faculty of Dentistry, Thammasat University, Pathumthani, 12120, Thailand
| |
Collapse
|
16
|
Lin Y, Qi X, Liu H, Xue K, Xu S, Tian Z. The anti-cancer effects of fucoidan: a review of both in vivo and in vitro investigations. Cancer Cell Int 2020; 20:154. [PMID: 32410882 PMCID: PMC7206694 DOI: 10.1186/s12935-020-01233-8] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/23/2020] [Indexed: 01/16/2023] Open
Abstract
Fucoidan is a kind of the polysaccharide, which comes from brown algae and comprises of sulfated fucose residues. It has shown a large range of biological activities in basic researches, including many elements like anti-inflammatory, anti-cancer, anti-viral, anti-oxidation, anticoagulant, antithrombotic, anti-angiogenic and anti-Helicobacter pylori, etc. Cancer is a multifactorial disease of multiple causes. Most of the current chemotherapy drugs for cancer therapy are projected to eliminate the ordinary deregulation mechanisms in cancer cells. Plenty of wholesome tissues, however, are also influenced by these chemical cytotoxic effects. Existing researches have demonstrated that fucoidan can directly exert the anti-cancer actions through cell cycle arrest, induction of apoptosis, etc., and can also indirectly kill cancer cells by activating natural killer cells, macrophages, etc. Fucoidan is used as a new anti-tumor drug or as an adjuvant in combination with an anti-tumor drug because of its high biological activity, wide source, low resistance to drug resistance and low side effects. This paper reviews the mechanism by which fucoidan can eliminate tumor cells, delay tumor growth and synergize with anticancer chemotherapy drugs in vitro, in vivo and in clinical trials.
Collapse
Affiliation(s)
- Yuan Lin
- The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Shinan Disrtict, Qingdao, China
| | - Xingsi Qi
- The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Shinan Disrtict, Qingdao, China
| | - Hengjian Liu
- The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Shinan Disrtict, Qingdao, China
| | - Kuijin Xue
- The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Shinan Disrtict, Qingdao, China
| | - Shan Xu
- The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Shinan Disrtict, Qingdao, China
| | - Zibin Tian
- The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Shinan Disrtict, Qingdao, China
| |
Collapse
|
17
|
Li L, Xu Y, Wang Y, Liu M, Deng S, Yu X, Cong B, Wang W. Effects of Low Molecular Weight Fucoidan on the Proliferation and Apoptosis of Dysplastic Oral Keratinocyte and Oral Squamous Cell Carcinoma Cells. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20921681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most common malignant tumors with high incidence, rapid progress, and high mortality. There are still some defects in the treatments of OSCC, which seriously affect the quality of patients’ life. Therefore, it is urgent to find a safer and more effective treatment for OSCC. Low molecular weight fucoidan (LMWF) has various biological activities, such as antitumor, anti-inflammatory, and antithrombotic, and has no obvious side effects, but the effect of LMWF on OSCC has not been reported. In this study, the effects of LMWF on dysplastic oral keratinocyte (DOK) and OSCC cells SCC-9 and SCC-25 were detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, flow cytometry, TUNEL, colony formation, and wound healing assay, and the expression levels of Notch-1 and Jagged-1 treated with LMWF were detected by RT-qPCR. The results showed that LMWF could inhibit the proliferation and migration of DOK and SCC-9, and promote apoptosis. Low molecular weight fucoidan upregulated the expression of Notch-1 and Jagged-1 in DOK and SCC-9 cells. It indicated that LMWF might promote the apoptosis of DOK and SCC-9 by upregulating the Notch signal pathway, thereby inhibiting cell proliferation and playing an antitumor role. It provided theoretical basis to develop LMWF as a novel therapeutic drug for oral cancer.
Collapse
Affiliation(s)
- Lulu Li
- School of Stomatology of Qingdao University, Shandong, China
| | - Yingjie Xu
- Department of Oral Medicine, Qingdao Stomatological Hospital, Shandong, China
| | - Yixue Wang
- School of Stomatology of Nanjing Medical University, Jiangsu, China
| | - Mengjia Liu
- School of Stomatology of Qingdao University, Shandong, China
| | - Songsong Deng
- School of Stomatology of Qingdao University, Shandong, China
| | - Xixi Yu
- Department of Oral Medicine, Qingdao Stomatological Hospital, Shandong, China
| | - Beibei Cong
- Department of Oral Medicine, Qingdao Stomatological Hospital, Shandong, China
| | - Wanchun Wang
- Department of Oral Medicine, Qingdao Stomatological Hospital, Shandong, China
| |
Collapse
|
18
|
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
|
19
|
Micro RNA Expression after Ingestion of Fucoidan; A Clinical Study. Mar Drugs 2020; 18:md18030143. [PMID: 32121066 PMCID: PMC7143719 DOI: 10.3390/md18030143] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 02/26/2020] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Fucoidans are a class of fucose-rich sulfated polysaccharides derived from brown macroalgae that exert a range of biological activities in vitro and in vivo. To generate an unbiased assessment of pathways and processes affected by fucoidan, a placebo-controlled double-blind pilot study was performed in healthy volunteers. Blood samples were taken immediately before and 24 h after ingestion of a single dose of 1 g of Undaria pinnatifida fucoidan (UPF) or placebo. Levels of isolated miRNAs were analyzed using Taqman Open Array Human MicroRNA panels. Out of 754 miRNAs screened, UPF affected a total of 53 miRNAs. Pathway analysis using the TALOS data analysis tool predicted 29 different pathways and processes that were largely grouped into cell surface receptor signaling, cancer-related pathways, the majority of which were previously associated with fucoidans. However, this analysis also identified nine pathways and processes that have not been associated with fucoidans before. Overall, this study illustrates that even a single dose of fucoidans has the potential to affect the expression of genes related to fundamental cellular processes. Moreover, it confirms previous data that fucoidans influence immunity, cancer cells, inflammation, and neurological function.
Collapse
|
20
|
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
|
21
|
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
|
22
|
Bobiński M, Okła K, Łuszczki J, Bednarek W, Wawruszak A, Moreno-Bueno G, Dmoszyńska-Graniczka M, Tarkowski R, Kotarski J. Isobolographic Analysis Demonstrates the Additive and Synergistic Effects of Gemcitabine Combined with Fucoidan in Uterine Sarcomas and Carcinosarcoma Cells. Cancers (Basel) 2019; 12:cancers12010107. [PMID: 31906221 PMCID: PMC7017062 DOI: 10.3390/cancers12010107] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/28/2019] [Accepted: 12/30/2019] [Indexed: 12/31/2022] Open
Abstract
Background: Uterine sarcomas and carcinosarcoma are associated with unfavorable prognosis. The regimens that are used in chemotherapy are associated with high incidence of side effects and usually do not significantly increase patients’ survival rates. In this study we investigated the activity and interactions between gemcitabine and fucoidan, the natural compound known for its anti-tumor properties, in human sarcomas and carcinosarcoma cell models. Methods: SK-UT-1, SK-UT1-B (carcinosarcoma), MES-SA (leiomyosarcoma), and ESS-1 (endometrial stromal sarcoma) cell lines were used for the experiments. Cells were incubated in the presence of gemcitabine, fucoidan, and mixtures, after the incubation the MTT tests were performed. In order to assess the interactions between tested compounds isobolographic analysis was performed. Additional assessments of apoptosis and cell cycle were done. Results: Additive effect of combined treatment with gemcitabine and fucoidan was observed in ESS-1 and SK-UT-1 cell line. Although the supra-additive (synergistic) effect noticed in SK-UT-1B cell line. It was not possible to determine the interactions of fucoidan and gemcitabine in MES-SA cell line due to insufficient response to treatment. Addition of fucoidan to gemcitabine enhances its proapoptotic activity, what was observed especially in ESS-1 and SK-UT-1B cell lines. The arrest of cell cycle induced by mixture of gemcitabine and fucoidan, superior comparing gemcitabine alone was observed in SK-UT-1B. Conclusions: Obtained data showed that a combination of fucoidan and gemcitabine in uterine endometrial stromal sarcoma and carcinosarcoma cell lines has additive or even synergistic effect in decreasing cell viability. Furthermore, this drug combination induces apoptosis and arrest of cell cycle. The resistance of uterine leiomyosarcoma cell line, justifies searching for other drugs combinations to improve therapy efficacy.
Collapse
Affiliation(s)
- Marcin Bobiński
- I Chair and Department of Gynaecological Oncology and Gynaecology, Medical University of Lublin, 20-081 Lublin, Poland
- Correspondence: ; Tel.: +48-81-53-27-847
| | - Karolina Okła
- I Chair and Department of Gynaecological Oncology and Gynaecology, Medical University of Lublin, 20-081 Lublin, Poland
| | - Jarogniew Łuszczki
- I Chair and Department of Pathophisiology, Medical University of Lublin, 20-081 Lublin, Poland
| | - Wiesława Bednarek
- I Chair and Department of Gynaecological Oncology and Gynaecology, Medical University of Lublin, 20-081 Lublin, Poland
| | - Anna Wawruszak
- Chair and Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-081 Lublin, Poland
| | - Gema Moreno-Bueno
- Laboratorio de Investigación Traslacional, MD Anderson Cancer Centre Madrid, Calle de Arturo Soria, 270 28033 Madrid, Spain
| | | | - Rafał Tarkowski
- I Chair and Department of Gynaecological Oncology and Gynaecology, Medical University of Lublin, 20-081 Lublin, Poland
| | - Jan Kotarski
- I Chair and Department of Gynaecological Oncology and Gynaecology, Medical University of Lublin, 20-081 Lublin, Poland
| |
Collapse
|
23
|
Nagamine T, Kadena K, Tomori M, Nakajima K, Iha M. Activation of NK cells in male cancer survivors by fucoidan extracted from Cladosiphon okamuranus. Mol Clin Oncol 2019; 12:81-88. [PMID: 31814980 DOI: 10.3892/mco.2019.1943] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 09/25/2019] [Indexed: 01/11/2023] Open
Abstract
Cancer survivors are highly motivated to seek information about the use of dietary supplements and complementary nutritional therapies to improve their quality of life. Fucoidan, a sulfated polysaccharide extracted from brown marine alga, exhibits a wide range of bioactivities, including anticancer activity. As natural killer (NK) cells serve an important role in defenses against tumor cells, the present study examined the effects of fucoidan extracted from Cladosiphon Okamuranus on NK cell activity in cancer survivors. A prospective, open-label clinical study was conducted on cancer survivors treated with fucoidan via oral administration; 11 cancer survivors with a performance status of 0 or 1 participated and consumed 3 g of fucoidan for 6 months. No significant changes were observed in the mean activities of NK cells in total subjects following the ingestion of fucoidan. An analysis of each sex revealed that NK cell activity was significantly increased by the ingestion of fucoidan in male, yet not female subjects. Serum fucoidan levels were markedly increased following the ingestion of fucoidan and the peak levels ranged between 30 and 198 ng/ml. Tumor markers remained within the reference range during the trial period in subjects, in whom primary tumors were eradicated by treatment. The basal values of tumor markers were elevated in three cases; tumor markers were increased in two cases and decreased in one by the ingestion of fucoidan. These findings suggest that fucoidan enhances the activation of NK cells in male cancer survivors.
Collapse
Affiliation(s)
- Takeaki Nagamine
- Department of Nutrition, Takasaki University of Health and Welfare, Takasaki, Gunma 370-0036, Japan
| | - Kizuku Kadena
- South Product Co., Ltd., Uruma, Okinawa 904-2234, Japan
| | - Makoto Tomori
- South Product Co., Ltd., Uruma, Okinawa 904-2234, Japan
| | - Katsuyuki Nakajima
- Department of Clinical Laboratory Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8514, Japan
| | - Masahiko Iha
- South Product Co., Ltd., Uruma, Okinawa 904-2234, Japan
| |
Collapse
|
24
|
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
|
25
|
Bittkau KS, Dörschmann P, Blümel M, Tasdemir D, Roider J, Klettner A, Alban S. Comparison of the Effects of Fucoidans on the Cell Viability of Tumor and Non-Tumor Cell Lines. Mar Drugs 2019; 17:E441. [PMID: 31357497 PMCID: PMC6722501 DOI: 10.3390/md17080441] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/15/2019] [Accepted: 07/23/2019] [Indexed: 12/13/2022] Open
Abstract
Fucoidans extracted from brown algae exert manifold biological activities paving the way for the development of numerous applications including treatments outside tumor therapy such as age-related macular degeneration or tissue engineering. In this study, we investigated the antiproliferative effects of fucoidans extracted from six different algae (Fucus vesiculosus, F. serratus, F. distichus subsp. evanescens, Dictyosiphon foeniculaceus, Laminaria digitata, Saccharina latissima) as well as three reference compounds (Sigma fucoidan, heparin, enoxaparin) on tumor (HL-60, Raji, HeLa, OMM-1, A-375, HCT-116, Hep G2) and non-tumor (ARPE-19, HaCaT) cell lines. All fucoidans were extracted according to a standardized procedure and tested in a commercially available MTS assay. Cell viability was measured after 24 h incubation with test compounds (1-100 µg/mL). Apart from few exceptions, fucoidans and heparins did not impair cell viability. In contrast, fucoidans significantly increased cell viability of suspension cell lines, but not of adherent cells. Fucoidans slightly increased viability of tumor cells and had no impact on the viability of non-tumor cells. The cell viability of HeLa and ARPE-19 cells negatively correlated with protein content and total phenolic content (TPC) of fucoidans, respectively. In summary, none of the tested fucoidans turned out to be anti-proliferative, rendering them interesting for future studies and applications.
Collapse
Affiliation(s)
- Kaya Saskia Bittkau
- Pharmaceutical Institute, Kiel University, Gutenbergstraße 76, 24118 Kiel, Germany
| | - Philipp Dörschmann
- University of Kiel, University Medical Center, Department of Ophthalmology, Arnold-Heller-Str. 3, Haus 25, 24105 Kiel, Germany
| | - Martina Blümel
- GEOMAR Centre for Marine Biotechnology (GEOMAR-Biotech), Research Unit Marine Natural Products Chemistry, GEOMAR Helmholtz Centre for Ocean Research Kiel, Am Kiel-Kanal 44, 24106 Kiel, Germany
| | - Deniz Tasdemir
- GEOMAR Centre for Marine Biotechnology (GEOMAR-Biotech), Research Unit Marine Natural Products Chemistry, GEOMAR Helmholtz Centre for Ocean Research Kiel, Am Kiel-Kanal 44, 24106 Kiel, Germany
- Faculty of Mathematics and Natural Sciences, Kiel University, Christian-Albrechts-Platz 4, 24118 Kiel, Germany
| | - Johann Roider
- University of Kiel, University Medical Center, Department of Ophthalmology, Arnold-Heller-Str. 3, Haus 25, 24105 Kiel, Germany
| | - Alexa Klettner
- University of Kiel, University Medical Center, Department of Ophthalmology, Arnold-Heller-Str. 3, Haus 25, 24105 Kiel, Germany.
| | - Susanne Alban
- Pharmaceutical Institute, Kiel University, Gutenbergstraße 76, 24118 Kiel, Germany.
| |
Collapse
|
26
|
Bobiński M, Okła K, Bednarek W, Wawruszak A, Dmoszyńska-Graniczka M, Garcia-Sanz P, Wertel I, Kotarski J. The Effect of Fucoidan, a Potential New, Natural, Anti-Neoplastic Agent on Uterine Sarcomas and Carcinosarcoma Cell Lines: ENITEC Collaborative Study. Arch Immunol Ther Exp (Warsz) 2019; 67:125-131. [PMID: 30659312 PMCID: PMC6420609 DOI: 10.1007/s00005-019-00534-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 01/04/2019] [Indexed: 01/04/2023]
Abstract
The aim of the study was to assess the activity of fucoidan on the uterine sarcomas (MES-SA and ESS-1) and carcinosarcoma cell lines (SK-UT-1 and SK-UT-1B) and its toxicity on the human skin fibroblasts (HSF). Two uterine sarcomas and two carcinosarcoma cell lines were examined, as a control HSF were used. Cell viability was assessed with MTT test, apoptosis with caspase-3 activity and cell cycle by assessment of DNA synthesis. Fucoidan significantly decreases cell viability in SK-UT-1, SK-UT-1B, and ESS-1 cell lines, such effect was not observed in MES-SA. Fucoidan was not substantially affecting proliferation among normal cells. The tested agent induced apoptosis in all cell cultures used in the experiment. Fucoidan affects cell cycle of all tested cell lines except MES-SA by increasing percentage of cells in G0/sub-G1/G1 phase. Fucoidan do not only affect proliferation but induces apoptosis in selected uterine sarcoma and carcinosarcoma cell lines, so it has potential to be used as cytotoxic agent. Fucoidan seems to be promising anti-cancer agent for endometrial stromal sarcoma and carcinosarcoma.
Collapse
Affiliation(s)
- Marcin Bobiński
- 1st Chair and Department of Gynecological Oncology and Gynecology, Medical University of Lublin, Staszica 16, 20-081, Lublin, Poland.
| | - Karolina Okła
- 1st Chair and Department of Gynecological Oncology and Gynecology, Medical University of Lublin, Staszica 16, 20-081, Lublin, Poland
| | - Wiesława Bednarek
- 1st Chair and Department of Gynecological Oncology and Gynecology, Medical University of Lublin, Staszica 16, 20-081, Lublin, Poland
| | - Anna Wawruszak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | | | - Pablo Garcia-Sanz
- Laboratory of Translational Research, MD Anderson Cancer Center, Madrid, Spain
| | - Iwona Wertel
- 1st Chair and Department of Gynecological Oncology and Gynecology, Medical University of Lublin, Staszica 16, 20-081, Lublin, Poland
| | - Jan Kotarski
- 1st Chair and Department of Gynecological Oncology and Gynecology, Medical University of Lublin, Staszica 16, 20-081, Lublin, Poland
| |
Collapse
|
27
|
van Weelden G, Bobiński M, Okła K, van Weelden WJ, Romano A, Pijnenborg JMA. Fucoidan Structure and Activity in Relation to Anti-Cancer Mechanisms. Mar Drugs 2019; 17:E32. [PMID: 30621045 PMCID: PMC6356449 DOI: 10.3390/md17010032] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/29/2018] [Accepted: 01/02/2019] [Indexed: 02/06/2023] Open
Abstract
Fucoidan is a natural derived compound found in different species of brown algae and in some animals, that has gained attention for its anticancer properties. However, the exact mechanism of action is currently unknown. Therefore, this review will address fucoidans structure, the bioavailability, and all known different pathways affected by fucoidan, in order to formulate fucoidans structure and activity in relation to its anti-cancer mechanisms. The general bioactivity of fucoidan is difficult to establish due to factors like species-related structural diversity, growth conditions, and the extraction method. The main pathways influenced by fucoidan are the PI3K/AKT, the MAPK pathway, and the caspase pathway. PTEN seems to be important in the fucoidan-mediated effect on the AKT pathway. Furthermore, the interaction with VEGF, BMP, TGF-β, and estrogen receptors are discussed. Also, fucoidan as an adjunct seems to have beneficial effects, for both the enhanced effectiveness of chemotherapy and reduced toxicity in healthy cells. In conclusion, the multipotent character of fucoidan is promising in future anti-cancer treatment. However, there is a need for more specified studies of the structure⁻activity relationship of fucoidan from the most promising seaweed species.
Collapse
Affiliation(s)
- Geert van Weelden
- Faculty of Science, (Medical) Biology, Radboud University, 6525 XZ Nijmegen, The Netherlands.
- The First Department of Gynecologic Oncology and Gynecology, Medical University of Lublin, 20-081 Lublin, Poland.
| | - Marcin Bobiński
- The First Department of Gynecologic Oncology and Gynecology, Medical University of Lublin, 20-081 Lublin, Poland.
| | - Karolina Okła
- The First Department of Gynecologic Oncology and Gynecology, Medical University of Lublin, 20-081 Lublin, Poland.
| | - Willem Jan van Weelden
- Department of Obstetrics & Gynecology, Radboud University Nijmegen, Medical Centre, 6525 GA Nijmegen, The Netherlands.
| | - Andrea Romano
- Department of Obstetrics and Gynecology, GROW-School for Oncology and Developmental Biology Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands.
| | - Johanna M A Pijnenborg
- Department of Obstetrics & Gynecology, Radboud University Nijmegen, Medical Centre, 6525 GA Nijmegen, The Netherlands.
| |
Collapse
|
28
|
Atashrazm F, Lowenthal RM, Dickinson JL, Holloway AF, Woods GM. Fucoidan enhances the therapeutic potential of arsenic trioxide and all-trans retinoic acid in acute promyelocytic leukemia, in vitro and in vivo. Oncotarget 2018; 7:46028-46041. [PMID: 27329592 PMCID: PMC5216779 DOI: 10.18632/oncotarget.10016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/23/2016] [Indexed: 12/14/2022] Open
Abstract
The morbidity and mortality associated with current therapies for acute promyelocytic leukemia (APL) remain a significant clinical concern, despite improvements in patient survival. Consequently, the development of adjuvant therapies that increase efficacy while reducing morbidities is important. Reducing the concentration of the toxic drugs in adjuvant therapy has the potential to reduce unwanted side effects. Therefore, this study aimed to determine the synergistic effects of fucoidan, an anti-tumor agent, with current APL therapies.When the human APL cell line, NB4, was treated in vitro with fucoidan plus ATO and ATRA at therapeutic and sub-therapeutic doses, there was an increase in sub-G0/G1 cells, annexin V/PI-positive-apoptotic cells and DNA fragmentation. This reduction in proliferation and increase in apoptosis was accompanied by enhanced myeloid differentiation as indicated by an increased expression of CD11b. This was not observed with the AML cell line Kasumi-1, suggesting specificity for APL.In vivo treatment of APL-bearing mice with fucoidan+ATRA or fucoidan+ATO delayed tumor growth, induced differentiation and increased tumor volume doubling time. The differentiated APL cells derived from the excised tumor mass exhibited decreased CD44 expression in fucoidan+ATRA treated mice. This could translate to decreased cell migration in APL patients.Our findings provide evidence supporting the use of fucoidan as an adjuvant therapeutic agent in the treatment of APL.
Collapse
Affiliation(s)
- Farzaneh Atashrazm
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Ray M Lowenthal
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Joanne L Dickinson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Adele F Holloway
- School of Medicine, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Gregory M Woods
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia.,School of Medicine, University of Tasmania, Hobart, Tasmania 7000, Australia
| |
Collapse
|
29
|
Sanjeewa KKA, Lee JS, Kim WS, Jeon YJ. The potential of brown-algae polysaccharides for the development of anticancer agents: An update on anticancer effects reported for fucoidan and laminaran. Carbohydr Polym 2017; 177:451-459. [PMID: 28962791 DOI: 10.1016/j.carbpol.2017.09.005] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/31/2017] [Accepted: 09/04/2017] [Indexed: 12/24/2022]
Abstract
In recent decades, attention to cancer-preventive treatments and studies on the development of anticancer drugs have sharply increased owing to the increase in cancer-related death rates in every region of the world. However, due to the adverse effects of synthetic drugs, much attention has been given to the development of anticancer drugs from natural sources because of fewer side effects of natural compounds than those of synthetic drugs. Recent studies on compounds and crude extracts from marine algae have shown promising anticancer properties. Among those compounds, polysaccharides extracted from brown seaweeds play a principal role as anticancer agents. Especially, a number of studies have revealed that polysaccharides isolated from brown seaweeds, such as fucoidan and laminaran, have promising effects against different cancer cell types in vitro and in vivo. Herein, we reviewed in vitro and in vivo anticancer properties reported for fucoidan and laminaran toward various cancer cells from 2013 to 2016.
Collapse
Affiliation(s)
- K K Asanka Sanjeewa
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Republic of Korea
| | - Jung-Suck Lee
- Research Center for Industrial Development of Seafood, Gyeongsang National University, Republic of Korea.
| | - Won-Suck Kim
- College of Medical and Life Sciences, Silla University, Busan 46958, Republic of Korea
| | - You-Jin Jeon
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Republic of Korea.
| |
Collapse
|
30
|
Induction of p53-Independent Apoptosis and G1 Cell Cycle Arrest by Fucoidan in HCT116 Human Colorectal Carcinoma Cells. Mar Drugs 2017; 15:md15060154. [PMID: 28555064 PMCID: PMC5484104 DOI: 10.3390/md15060154] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/16/2017] [Accepted: 05/22/2017] [Indexed: 12/24/2022] Open
Abstract
It is well known that fucoidan, a natural sulfated polysaccharide present in various brown algae, mediates anticancer effects through the induction of cell cycle arrest and apoptosis. Nevertheless, the role of tumor suppressor p53 in the mechanism action of fucoidan remains unclear. Here, we investigated the anticancer effect of fucoidan on two p53 isogenic HCT116 (p53+/+ and p53-/-) cell lines. Our results showed that inhibition of cell viability, induction of apoptosis and DNA damage by treatment with fucoidan were similar in two cell lines. Flow cytometric analysis revealed that fucoidan resulted in G1 arrest in the cell cycle progression, which correlated with the inhibition of phosphorylation of retinoblastoma protein (pRB) and concomitant association of pRB with the transcription factor E2Fs. Furthermore, treatment with fucoidan obviously upregulated the expression of cyclin-dependent kinase (CDK) inhibitors, such as p21WAF1/CIP1 and p27KIP1, which was paralleled by an enhanced binding with CDK2 and CDK4. These events also commonly occurred in both cell lines, suggesting that fucoidan triggered G1 arrest and apoptosis in HCT116 cells by a p53-independent mechanism. Thus, given that most tumors exhibit functional p53 inactivation, fucoidan could be a possible therapeutic option for cancer treatment regardless of the p53 status.
Collapse
|
31
|
Zhang F, Shi JJ, Thakur K, Hu F, Zhang JG, Wei ZJ. Anti-Cancerous Potential of Polysaccharide Fractions Extracted from Peony Seed Dreg on Various Human Cancer Cell Lines Via Cell Cycle Arrest and Apoptosis. Front Pharmacol 2017; 8:102. [PMID: 28316571 PMCID: PMC5334287 DOI: 10.3389/fphar.2017.00102] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 02/20/2017] [Indexed: 12/24/2022] Open
Abstract
In this study, four homo/heterogenous polysaccharides (HBSS, CHSS, DASS, and CASS) extracted from peony seed dreg with respective molecular weights of 3467, 4677, 229, and 56 kDa were evaluated for anti-cancerous attributes in prostate cancer cells (Pc-3), colon cancer cells (HCT-116), human breast cancer cells (MCF-7), cervical cancer (Hela cells) and human embryonic kidney 293 (HEK 293) cells as control. Among them, CASS and DASS extracted by alkali, consisted of 34.43% Gal, 26.39% Ara, 21.80% Glc and 35.77% Ara, 19.35% Gal, 17.77% Man, respectively. CASS fraction had the most significant inhibitory effects on all the cell lines used whereas HBSS had least effect. The CASS shown remarkable inhibition and cytotoxic effects in Hela cells followed by other cell lines as compared to 5-fluorouracil (5-FU). CASS arrested cell cycle in G0/G1 phase except MCF-7 cells and increased apoptotic cells percentage varied in different treated cells. CASS down regulated the expression of Cyclin A/B1/D1/E1, CDK-1/2/4/6 and p15/16/21/27 excluding p53. The notable change in expression of proteins (Cytochrome C, Bax, Bcl-2, p-Caspase-3, -8, -9, and PARP) was observed followed by Apaf-1 and Survivin. These findings indicated that CASS has an anti-cancerous potential in the treatment of human cancers which make it a potent candidate in functional foods.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhao-Jun Wei
- School of Food Science and Engineering, Hefei University of TechnologyHefei, China
| |
Collapse
|
32
|
Wei J, Zhang L, Ren L, Zhang J, Yu Y, Wang J, Duan J, Peng C, Sun Z, Zhou X. Endosulfan inhibits proliferation through the Notch signaling pathway in human umbilical vein endothelial cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 221:26-36. [PMID: 27939630 DOI: 10.1016/j.envpol.2016.08.083] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 08/20/2016] [Accepted: 08/30/2016] [Indexed: 06/06/2023]
Abstract
Our previous research showed that endosulfan triggers the extrinsic coagulation pathway by damaging endothelial cells and causes hypercoagulation of blood. To identify the mechanism of endosulfan-impaired endothelial cells, we treated human umbilical vein endothelial cells (HUVECs) with different concentrations of endosulfan, with and without an inhibitor for Notch, N-[N-(3, 5-difluorophenacetyl)-1-alanyl]S-Phenylglycinet-butylester (DAPT, 20 μM), or a reactive oxygen species (ROS) scavenger, N-Acetyl-l-cysteine (NAC, 3 mM), for 24 h. The results showed that endosulfan could inhibit cell viability/proliferation by increasing the release of lactate dehydrogenase (LDH), arresting the cell cycle in both S and G2/M phases, and inducing apoptosis in HUVECs. We also found that endosulfan can damage microfilaments, microtubules, and nuclei; arrest mitosis; remarkably increase the expressions of Dll4, Notch1, Cleaved-Notch1, Jagged1, Notch4, Hes1, and p21; and significantly induce ROS and malondialdehyde production in HUVECs. The presence of DAPT antagonized the above changes of cycle arrest, proliferation inhibition, and expressions of Dll4, Notch1, Cleaved-Notch1, Hes1, and p21 caused by endosulfan; however, NAC could attenuate LDH release; ROS and malondialdehyde production; apoptosis; and the expression levels of Dll4, Notch1, Cleaved-Notch1, Notch4, and Hes1 induced by endosulfan. These results demonstrated that endosulfan inhibited proliferation through the Notch signaling pathway as a result of oxidative stress. In addition, endosulfan can damage the cytoskeleton and block mitosis, which may add another layer of toxic effects on endothelial cells.
Collapse
Affiliation(s)
- Jialiu Wei
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing China
| | - Lianshuang Zhang
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing China
| | - Lihua Ren
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing China
| | - Jin Zhang
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing China
| | - Yang Yu
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing China
| | - Ji Wang
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing China
| | - Junchao Duan
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing China
| | - Cheng Peng
- National Research Centre for Environmental Toxicology (Entox), Member of Queensland Alliance for Environmental Health Science (QAEHS), The University of Queensland, Coopers Plains, 4108, Brisbane, QLD, Australia
| | - Zhiwei Sun
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing China
| | - Xianqing Zhou
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, 100069, Beijing China.
| |
Collapse
|