1
|
Bashir B, Sethi P, Panda S, Manikyam HK, Vishwas S, Singh SK, Singh K, Jain D, Chaitanya MVNL, Coutinho HDM. Unravelling the epigenetic based mechanism in discovery of anticancer phytomedicine: Evidence based studies. Cell Signal 2025; 131:111743. [PMID: 40107479 DOI: 10.1016/j.cellsig.2025.111743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Epigenetic mechanisms play a crucial role in the normal development and maintenance of tissue-specific gene expression patterns in mammals. Disruption of these processes can result in changes to gene function and the transformation of cells into a malignant state. Cancer is characterized by widespread alterations in the epigenetic landscape, revealing that it involves not only genetic mutations but also epigenetic abnormalities. Recent progress in the field of cancer epigenetics has demonstrated significant reprogramming of various components of the epigenetic machinery in cancer, such as DNA methylation, modifications to histones, positioning of nucleosomes, and the expression of non-coding RNAs, particularly microRNAs. The ability to reverse epigenetic abnormalities has given rise to the hopeful field of epigenetic therapy, which has shown advancement with the recent approval by the FDA of three drugs targeting epigenetic mechanisms for the treatment of cancer. In the present manuscript, a comprehensive review has been presented about the role of understanding the epigenetic link between cancer and mechanisms by which phytomedicine offers treatment avenues. Further, this review deciphers the significance of natural products in the identification of epigenetic therapeutics, the diversity of their molecular targets, the use of nanotechnology, and the creation of new strategies for overcoming the inherent clinical challenges associated with developing these drug leads.
Collapse
Affiliation(s)
- Bushra Bashir
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Pranshul Sethi
- Department of Pharmacology, College of Pharmacy, Shri Venkateshwara University, Gajraula, Uttar Pradesh, India
| | - Satyajit Panda
- Department of Pharmaceutics, Institute of Pharmacy and Technology, Salipur, Cuttack, Odisha 754202, India
| | - Hemanth Kumar Manikyam
- Department of Chemistry, Faculty of science, North East Frontier Technical University, Arunachal Pradesh 791001, India
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Kuldeep Singh
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| | - Divya Jain
- Department of Microbiology, School of Applied and Life sciences, Uttaranchal University, Dehradun, Uttarakhand 248007, India.
| | - M V N L Chaitanya
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India.
| | | |
Collapse
|
2
|
Tu L, Xing B, Ma S, Zou Z, Wang S, Feng J, Cheng M, Jin Y. A review on polysaccharide-based tumor targeted drug nanodelivery systems. Int J Biol Macromol 2025; 304:140820. [PMID: 39933669 DOI: 10.1016/j.ijbiomac.2025.140820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/13/2025]
Abstract
The tumor-targeted drug delivery system (TTDNS) uses nanocarriers to transport chemotherapeutic agents to target tumor cells or tissues precisely. This innovative approach considerably increases the effective concentration of these drugs at the tumor site, thereby enhancing their therapeutic efficacy. Many chemotherapeutic agents face challenges, such as low bioavailability, high cytotoxicity, and inadequate drug resistance. To address these obstacles, TTDNS comprising natural polysaccharides have gained increasing popularity in the field of nanotechnology owing to their ability to improve safety, bioavailability, and biocompatibility while reducing toxicity. In addition, it enhances permeability and allows for controlled drug delivery and release. This review focuses on the sources of natural polysaccharides and their direct and indirect mechanisms of anti-tumor activity. We also explored the preparation of various polysaccharide-based nanocarriers, including nanoparticles, nanoemulsions, nanohydrogels, nanoliposomes, nanocapsules, nanomicelles, nanocrystals, and nanofibers. Furthermore, this review delves into the versatile applications of polysaccharide-based nanocarriers, elucidating their capabilities for in vivo targeting, controlled release, and responsiveness to endogenous and exogenous stimuli, such as pH, reactive oxygen species, glutathione, light, ultrasound, and magnetic fields. This sophisticated design substantially enhances the chemotherapeutic efficacy of the encapsulated drugs at tumor sites and provides a basis for preclinical and clinical research. However, the in vivo stability, drug loading, and permeability of these preparations into tumor tissues still need to be improved. Most of the currently developed biomarker-sensitive polysaccharide nanocarriers are still in the laboratory stage, more innovative delivery mechanisms and clinical studies are needed to develop commercial nanocarriers for medical use.
Collapse
Affiliation(s)
- Liangxing Tu
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Banghuai Xing
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Shufei Ma
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Zijian Zou
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Siying Wang
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Jianfang Feng
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China; Guangxi University of Chinese Medicine, Nanning 530200, PR China.
| | - Meng Cheng
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China.
| | - Yi Jin
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China.
| |
Collapse
|
3
|
Ferdushi R, Kim D, Sriramulu DK, Hwang Y, Park K, Key J. Computational insights into fucoidan-receptor binding: Implications for fucoidan-based targeted drug delivery. Drug Discov Today 2025; 30:104315. [PMID: 39984116 DOI: 10.1016/j.drudis.2025.104315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 01/31/2025] [Accepted: 02/13/2025] [Indexed: 02/23/2025]
Abstract
Fucoidan, a polysaccharide from seaweed, holds promise as a drug delivery system and immune modulator; however, its exact mechanism of action remains unclear. As various carbohydrates play key roles in immune responses by binding to carbohydrate-binding proteins like lectins, fucoidan is hypothesized to interact with immune receptors, potentially driving its anticancer activities. However, structural variability, extraction-induced heterogeneity, and weak binding affinities pose challenges to research. Computational tools offer valuable insights into fucoidan-receptor interactions, addressing these challenges and enabling the design of more effective therapies. This review examines fucoidan's therapeutic activities, drug delivery potential, and receptor interactions, emphasizing computational approaches to advance immune modulation and anticancer applications using carbohydrate polymers.
Collapse
Affiliation(s)
- Rumana Ferdushi
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| | - Dohyeon Kim
- Center for Natural Product Systems Biology, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea; Department of Bioinformatics and Life Science, Soongsil University, Seoul 06978, Republic of Korea
| | - Dinesh Kumar Sriramulu
- Center for Natural Product Systems Biology, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Yoonho Hwang
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| | - Keunwan Park
- Center for Natural Product Systems Biology, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea; Department of YM-KIST Bio-Health Convergence, Yonsei University, Wonju 26493, Republic of Korea.
| | - Jaehong Key
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea.
| |
Collapse
|
4
|
Biswas I, Precilla S D, Kuduvalli SS, K B, R S, T S A. Ultrastructural and immunohistochemical insights on the anti-glioma effects of a dual-drug cocktail in an in vivo experimental model. J Chemother 2024; 36:593-606. [PMID: 38240036 DOI: 10.1080/1120009x.2024.2302741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/27/2023] [Accepted: 01/03/2024] [Indexed: 10/23/2024]
Abstract
Glioma coined as 'butterfly tumor' exhibits intense heterogeneity at the molecular and cellular levels. Although, Temozolomide exerted a long-ranging and prevailing therapeutic effect against glioma, albeit it has provided modest survival outcome. Fucoidan, (marine brown algal derivative) has demonstrated potent anti-tumor effects including glioma. Nevertheless, there is paucity of studies conducted on Fucoidan to enhance the anti-glioma efficacy of Temozolomide. The present study aimed to explore the plausible synergistic anti-glioma efficacy of Fucoidan in combination with Temozolomide in an in vivo experimental model. The dual-drug combination significantly inhibited tumor growth in in vivo and prolonged the survival rate when compared with the other treatment and tumor-control groups, via down-regulation of inflammatory cascade- IL-6/T LR4 and JAK/STAT3 as per the immunohistochemistry findings. Furthermore, the ultrastructural analysis indicated that the combinatorial treatment had restored the normal neuronal architecture of glioma-induced rats. Overall, the dual-drug cocktail might enhance the therapeutic outcome in glioma patients.
Collapse
Affiliation(s)
- Indrani Biswas
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry, India
| | - Daisy Precilla S
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry, India
| | - Shreyas S Kuduvalli
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry, India
| | - Bhavani K
- Department of Pathology, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry, India
| | | | - Anitha T S
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry, India
| |
Collapse
|
5
|
Yang J, Zhao H, Qu S. Therapeutic potential of fucoidan in central nervous system disorders: A systematic review. Int J Biol Macromol 2024; 277:134397. [PMID: 39097066 DOI: 10.1016/j.ijbiomac.2024.134397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024]
Abstract
Central nervous system (CNS) disorders have a complicated pathogenesis, and to date, no single mechanism can fully explain them. Most drugs used for CNS disorders primarily aim to manage symptoms and delay disease progression, and none have demonstrated any pathological reversal. Fucoidan is a safe, sulfated polysaccharide from seaweed that exhibits multiple pharmacological effects, and it is anticipated to be a novel treatment for CNS disorders. To assess the possible clinical uses of fucoidan, this review aims to provide an overview of its neuroprotective mechanism in both in vivo and in vitro CNS disease models, as well as its pharmacokinetics and safety. We included 39 articles on the pharmacology of fucoidan in CNS disorders. In vitro and in vivo experiments demonstrate that fucoidan has important roles in regulating lipid metabolism, enhancing the cholinergic system, maintaining the functional integrity of the blood-brain barrier and mitochondria, inhibiting inflammation, and attenuating oxidative stress and apoptosis, highlighting its potential for CNS disease treatment. Fucoidan has a protective effect against CNS disorders. With ongoing research on fucoidan, it is expected that a natural, highly effective, less toxic, and highly potent fucoidan-based drug or nutritional supplement targeting CNS diseases will be developed.
Collapse
Affiliation(s)
- Jing Yang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, 110004 Shenyang, Liaoning, PR China.
| | - He Zhao
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, 110004 Shenyang, Liaoning, PR China.
| | - Shengtao Qu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, 110004 Shenyang, Liaoning, PR China.
| |
Collapse
|
6
|
Lakshmana Senthil S. A comprehensive review to assess the potential, health benefits and complications of fucoidan for developing as functional ingredient and nutraceutical. Int J Biol Macromol 2024; 277:134226. [PMID: 39074709 DOI: 10.1016/j.ijbiomac.2024.134226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 07/21/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024]
Abstract
Polysaccharides from seaweeds or macroalgae are garnering significant interest from pharmaceutical and food industries due to their bioactivities and promising therapeutic effects. Among the diverse agal polysaccharides, fucoidan is a well-documented and stands out as a well-researched sulphated heteropolysaccharide found in brown seaweeds. It primarily consists of l-fucose and sulfate ester groups, along with other monosaccharides like xylose, mannose, uronic acid, rhamnose, arabinose, and galactose. Recent scientific investigations have unveiled the formidable inhibitory prowess of fucoidan against SARS-CoV-2, offering a promising avenue for therapeutic intervention in our current landscape. Moreover, fucoidan has demonstrated remarkable abilities in safeguarding the gastrointestinal tract, regulating angiogenesis, mitigating metabolic syndrome, and fortifying bone health. Despite the abundance of studies underscoring fucoidan's potential as a vital component sourced from nature, its exploitation remains constrained by inherent limitations. Thus, the primary objective of this article is to furnish a comprehensive discourse on the structural attributes, health-enhancing properties, safety parameters, and potential toxicity associated with fucoidan. Furthermore, the discourse extends to elucidating the practical applications and developmental prospects of fucoidan as a cornerstone in the realm of functional foods and nutraceuticals.
Collapse
|
7
|
Zhang W, Park HB, An EK, Kim SJ, Ryu D, Kim D, Lim D, Hwang J, Kwak M, You S, Lee PCW, Jin JO. Fucoidan from Durvillaea Antarctica enhances the anti-cancer effect of anti-PD-L1 antibody by activating dendritic cells and T cells. Int J Biol Macromol 2024; 280:135922. [PMID: 39322135 DOI: 10.1016/j.ijbiomac.2024.135922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/08/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
Immune checkpoint inhibitors are showing groundbreaking results in tumor immunotherapy. However, there are cases where treatment efficiency is insufficient due to limitations in immune activity, and various trials to overcome this are being studied. In this study, we investigated the immune activation ability of fucoidan extracted from Durvillaea antarctica (FDA) and whether it can enhance the anti-cancer effects of immune checkpoint inhibitors. FDA treatment resulted in an elevation of co-stimulator and major histocompatibility complex molecule expression, as well as the production of pro-inflammatory cytokines in bone marrow-derived and splenic dendritic cells (DCs). Administration of 50 mg/kg FDA increased the number of splenic CD8 T cells by >1.4-fold compared to PBS administration. Additionally, 50 mg/kg FDA increased the production of IFN-γ in CD4 and CD8 T cells by 4.3-fold and 7.2-fold, respectively, compared to the PBS control. FDA promoted immune cell activation was TLR4 dependent. Furthermore, anti-PD-L1 antibody administration inhibited CT-26 tumor growth by approximately 3-fold compared to the PBS control group, whereas combined treatment with FDA and anti-PD-L1 antibody showed an 8.4-fold tumor growth inhibition effect compared to the PBS control group. Therefore, FDA may be used to enhance the anti-cancer effects of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Wei Zhang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Hae-Bin Park
- Department of Microbiology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Eun-Koung An
- Department of Microbiology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - So-Jung Kim
- Department of Microbiology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Dayoung Ryu
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, 05505, South Korea
| | - Dayoung Kim
- Department of Microbiology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Daeun Lim
- Department of Microbiology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Juyoung Hwang
- Department of Chemistry, Pukyong National University, Busan 48513, South Korea
| | - Minseok Kwak
- Department of Chemistry, Pukyong National University, Busan 48513, South Korea
| | - SangGuan You
- Department of Marine Food Science and Technology, Gangneung-Wonju National University, 120 Gangneung Daehangno, Gangneung, Gangwon 210-702, South Korea
| | - Peter C W Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, 05505, South Korea
| | - Jun-O Jin
- Department of Microbiology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea.
| |
Collapse
|
8
|
Mary Martin T, K MS. Seaweeds and Their Secondary Metabolites: A Promising Drug Candidate With Novel Mechanisms Against Cancers and Tumor Angiogenesis. Cureus 2024; 16:e66662. [PMID: 39262521 PMCID: PMC11387980 DOI: 10.7759/cureus.66662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/12/2024] [Indexed: 09/13/2024] Open
Abstract
Cancer continually remains a severe threat to public health and requires constant demand for novel therapeutic drug candidates. Due to their multi-target orientation, lesser toxicity, and easy availability, natural compounds attract more attention from current scientific research interest than synthetic drug molecules. The plants and microorganisms produce a huge variety of secondary metabolites because of their physiological diversification, and the seaweeds occupy a prominent position as effective drug resources. Seaweeds comprise microscopic or macroscopic photosynthetic, multicellular, eukaryotic marine algae that commonly inhabit the coastal regions. Several molecules (such as polysaccharides, lipids, proteinaceous fractions, phenolic compounds, and alkaloids) are derived from seaweeds, and those small molecules are well attractive and more effective in cancer research programs. Their structural variation, derivative diversity, and quantity vary with seaweed species and geographical origin. Their smaller molecular weight, unique derivatives, hydrophobicity, and degree of sulfation are reported to be causes of their crucial role against different cancer cells in vitro. Several reports showed that those compounds selectively discriminate between normal and cancer cells based on receptor variations, enzyme deficiency, and structural properties. The present review aimed to give a concise explanation regarding their structural diversity, extractability, and mechanism of action related to their anti-cancer activities based on recently published data.
Collapse
Affiliation(s)
- Taniya Mary Martin
- Zebrafish Facility, Department of Anatomy, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS) Saveetha University, Chennai, IND
| | - Meenakshi Sundaram K
- Zebrafish Facility, Department of Anatomy, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS) Saveetha University, Chennai, IND
| |
Collapse
|
9
|
Visuddho V, Halim P, Helen H, Muhar AM, Iqhrammullah M, Mayulu N, Surya R, Tjandrawinata RR, Ribeiro RIMA, Tallei TE, Taslim NA, Kim B, Syahputra RA, Nurkolis F. Modulation of Apoptotic, Cell Cycle, DNA Repair, and Senescence Pathways by Marine Algae Peptides in Cancer Therapy. Mar Drugs 2024; 22:338. [PMID: 39195454 DOI: 10.3390/md22080338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
Marine algae, encompassing both macroalgae and microalgae, have emerged as a promising and prolific source of bioactive compounds with potent anticancer properties. Despite their significant therapeutic potential, the clinical application of these peptides is hindered by challenges such as poor bioavailability and susceptibility to enzymatic degradation. To overcome these limitations, innovative delivery systems, particularly nanocarriers, have been explored. Nanocarriers, including liposomes, nanoparticles, and micelles, have demonstrated remarkable efficacy in enhancing the stability, solubility, and bioavailability of marine algal peptides, ensuring controlled release and prolonged therapeutic effects. Marine algal peptides encapsulated in nanocarriers significantly enhance bioavailability, ensuring more efficient absorption and utilization in the body. Preclinical studies have shown promising results, indicating that nanocarrier-based delivery systems can significantly improve the pharmacokinetic profiles and therapeutic outcomes of marine algal peptides. This review delves into the diverse anticancer mechanisms of marine algal peptides, which include inducing apoptosis, disrupting cell cycle progression, and inhibiting angiogenesis. Further research focused on optimizing nanocarrier formulations, conducting comprehensive clinical trials, and continued exploration of marine algal peptides holds great promise for developing innovative, effective, and sustainable cancer therapies.
Collapse
Affiliation(s)
- Visuddho Visuddho
- Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Princella Halim
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Helen Helen
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Adi Muradi Muhar
- Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Muhammad Iqhrammullah
- Postgraduate Program of Public Health, Universitas Muhammadiyah Aceh, Banda Aceh 23123, Indonesia
| | - Nelly Mayulu
- Department of Nutrition, Faculty of Health Science, Muhammadiyah Manado University, Manado 95249, Indonesia
| | - Reggie Surya
- Department of Food Technology, Faculty of Engineering, Bina Nusantara University, Jakarta 11480, Indonesia
| | - Raymond Rubianto Tjandrawinata
- Department of Biotechnology, Faculty of Biotechnology, Atma Jaya Catholic University of Indonesia, Jakarta 12930, Indonesia
| | | | - Trina Ekawati Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado 95115, Indonesia
| | - Nurpudji Astuti Taslim
- Division of Clinical Nutrition, Department of Nutrition, Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Fahrul Nurkolis
- Department of Biological Sciences, Faculty of Sciences and Technology, State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta 55281, Indonesia
| |
Collapse
|
10
|
Shi C, Zhao S, Mi L, Niu D, Hu F, Han W, Li B. Fucoidan MF4 from Fucus vesiculosus inhibits Lewis lung cancer via STING-TBK1-IRF3 pathway. Int J Biol Macromol 2024; 267:131336. [PMID: 38583840 DOI: 10.1016/j.ijbiomac.2024.131336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 03/28/2024] [Accepted: 04/01/2024] [Indexed: 04/09/2024]
Abstract
Fucoidan, a sulfated polysaccharide of marine origin found in brown algae and sea cucumbers, has been identified as a neuroprotective compound. In this study, a novel fucoidan MF4 was extracted from Fucus vesiculosus and isolated using Q-Sepharose fast-flow ion-exchange chromatography. The physicochemical properties of MF4 were characterized. MF4 is primarily composed of fucose, xylose, galactose, glucose, and mannose in a molar ratio of 12.3: 4.9: 1.1: 1.0: 1.1, with an average molecular weight of 67.7 kDa. Notably, MF4 demonstrated suppression of LLC tumor growth in vivo. RNA-sequencing analysis revealed that MF4 enhanced the expression of type I interferon-associated downstream genes in macrophages. Furthermore, MF4 increased the levels of phosphorylated TBK1 and IRF3 proteins in vitro. By activating the STING-TBK1-IRF3 signaling pathway, MF4 may enhance the antitumor activity of macrophages. Taken together, MF4 has promising potential as an antitumor and immunomodulatory agent.
Collapse
Affiliation(s)
- Chuanqin Shi
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266003, China; Center of Translational Medicine, Zibo Central Hospital, Zibo 255020, China
| | - Shihua Zhao
- Department of Endocrinology, Zibo Central Hospital, Zibo 255020, China
| | - Liyan Mi
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 261400, China
| | - Deying Niu
- Department of Neurosurgery, Zibo Central Hospital, Zibo 255020, China
| | - Fanwen Hu
- Departmet of Pharmacy, Jinan Dermatosis Prevention and Contorl Hospital, Jinan 250000, China
| | - Wenwei Han
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao 266071, China.
| | - Bing Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266003, China; Department of Dermatology, The Affiliated Haici Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
11
|
Gagliardi A, Chiarella E, Voci S, Ambrosio N, Celano M, Cristina Salvatici M, Cosco D. DIFUCOSIN: DIclofenac sodium salt loaded FUCOidan-SericIN nanoparticles for the management of chronic inflammatory diseases. Int J Pharm 2024; 655:124034. [PMID: 38531433 DOI: 10.1016/j.ijpharm.2024.124034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/07/2024] [Accepted: 03/20/2024] [Indexed: 03/28/2024]
Abstract
The current investigation emphasizes the use of fucoidan and sericin as dual-role biomaterials for obtaining novel nanohybrid systems for the delivery of diclofenac sodium (DS) and the potential treatment of chronic inflammatory diseases. The innovative formulations containing 4 mg/ml of fucoidan and 3 mg/ml of sericin showed an average diameter of about 200 nm, a low polydispersity index (0.17) and a negative surface charge. The hybrid nanosystems demonstrated high stability at various pHs and temperatures, as well as in both saline and glucose solutions. The Rose Bengal assay evidenced that fucoidan is the primary modulator of relative surface hydrophobicity with a two-fold increase of this parameter when compared to sericin nanoparticles. The interaction between the drug and the nanohybrids was confirmed through FT-IR analysis. Moreover, the release profile of DS from the colloidal systems showed a prolonged and constant drug leakage over time both at pH 5 and 7. The DS-loaded nanohybrids (DIFUCOSIN) induced a significant decrease of IL-6 and IL-1β with respect to the active compound in human chondrocytes evidencing a synergistic action of the individual components of nanosystems and the drug and demonstrating the potential application of the proposed nanomedicine for the treatment of inflammation.
Collapse
Affiliation(s)
- Agnese Gagliardi
- Department of Health Sciences, University "Magna Græcia", 88100 Catanzaro, Italy
| | - Emanuela Chiarella
- Department of Experimental and Clinical Medicine, University "Magna Græcia", 88100 Catanzaro, Italy
| | - Silvia Voci
- Department of Health Sciences, University "Magna Græcia", 88100 Catanzaro, Italy
| | - Nicola Ambrosio
- Department of Health Sciences, University "Magna Græcia", 88100 Catanzaro, Italy
| | - Marilena Celano
- Department of Health Sciences, University "Magna Græcia", 88100 Catanzaro, Italy
| | - Maria Cristina Salvatici
- Institute of Chemistry of Organometallic Compounds (ICCOM)-Electron Microscopy Centre (Ce.M.E.), National Research Council (CNR), 50019, Sesto Fiorentino, Firenze, Italy
| | - Donato Cosco
- Department of Health Sciences, University "Magna Græcia", 88100 Catanzaro, Italy.
| |
Collapse
|
12
|
Geng H, Chen M, Guo C, Wang W, Chen D. Marine polysaccharides: Biological activities and applications in drug delivery systems. Carbohydr Res 2024; 538:109071. [PMID: 38471432 DOI: 10.1016/j.carres.2024.109071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024]
Abstract
The ocean is the common home of a large number of marine organisms, including plants, animals, and microorganisms. Researchers can extract thousands of important bioactive components from the oceans and use them extensively to treat and prevent diseases. In contrast, marine polysaccharide macromolecules such as alginate, carrageenan, Laminarin, fucoidan, chitosan, and hyaluronic acid have excellent physicochemical properties, good biocompatibility, and high bioactivity, which ensures their wide applications and strong therapeutic potentials in drug delivery. Drug delivery systems (DDS) based on marine polysaccharides and modified marine polysaccharide molecules have emerged as an innovative technology for controlling drug distribution on temporal, spatial, and dosage scales. They can detect and respond to external stimuli such as pH, temperature, and electric fields. These properties have led to their wide application in the design of novel drug delivery systems such as hydrogels, polymeric micelles, liposomes, microneedles, microspheres, etc. In addition, marine polysaccharide-based DDS not only have smart response properties but also can combine with the unique biological properties of the marine polysaccharide base to exert synergistic therapeutic effects. The biological activities of marine polysaccharides and the design of marine polysaccharide-based DDS are reviewed. Marine polysaccharide-based responsive DDS are expected to provide new strategies and solutions for disease treatment.
Collapse
Affiliation(s)
- Hongxu Geng
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, School of Pharmacy, Yantai University, Yantai, 264005, PR China.
| | - Meijun Chen
- Yantai Muping District Hospital of Traditional Chinese Medicine, No.505, Government Street, Muping District, Yantai, 264110, PR China.
| | - Chunjing Guo
- College of Marine Life Science, Ocean University of China, 5# Yushan 10 Road, Qingdao, 266003, PR China.
| | - Wenxin Wang
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, School of Pharmacy, Yantai University, Yantai, 264005, PR China.
| | - Daquan Chen
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, School of Pharmacy, Yantai University, Yantai, 264005, PR China.
| |
Collapse
|
13
|
Vafaei S, Alkhrait S, Yang Q, Ali M, Al-Hendy A. Empowering Strategies for Lifestyle Interventions, Diet Modifications, and Environmental Practices for Uterine Fibroid Prevention; Unveiling the LIFE UP Awareness. Nutrients 2024; 16:807. [PMID: 38542717 PMCID: PMC10975324 DOI: 10.3390/nu16060807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/28/2024] [Accepted: 03/07/2024] [Indexed: 04/01/2024] Open
Abstract
Uterine fibroids (UFs) are the most common prevalent benign tumor among women of reproductive age, disproportionately affecting women of color. This paper introduces an innovative management strategy for UFs, emphasizing the curbing of disease prevention and progression. Traditionally, medical intervention is deferred until advanced stages, necessitating invasive surgeries such as hysterectomy or myomectomy, leading to high recurrence rates and increased healthcare costs. The strategy, outlined in this review, emphasizes UF disease management and is named LIFE UP awareness-standing for Lifestyle Interventions, Food Modifications, and Environmental Practices for UF Prevention. These cost-effective, safe, and accessible measures hold the potential to prevent UFs, improve overall reproductive health, reduce the need for invasive procedures, and generate substantial cost savings for both individuals and healthcare systems. This review underscores the importance of a proactive UF management method, paving the way for future research and policy initiatives in this domain.
Collapse
Affiliation(s)
| | | | | | - Mohamed Ali
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (S.V.); (S.A.); (Q.Y.)
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (S.V.); (S.A.); (Q.Y.)
| |
Collapse
|
14
|
Antonisamy AJ, Rajendran K, Dhanaraj P. Network pharmacology integrated molecular docking of fucoidan against oral cancer and in vitro evaluation- A study using GEO datasets. J Biomol Struct Dyn 2024:1-24. [PMID: 38385359 DOI: 10.1080/07391102.2024.2316771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/02/2024] [Indexed: 02/23/2024]
Abstract
Oral cancer is a widespread health concern in rural India due to a lack of awareness, delayed diagnosis and limited access to affordable treatment options. The current chemotherapy has notable side effects, underscoring the need for new drug candidates with improved bioavailability and specificity. In this current research, fucoidan, a sulphated polysaccharide, was extracted from the brown algae Spatoglossum asperum, and shown to be cytotoxic in vitro against oral cancer cells (KB cell line) at an IC50 of 107.76 µg/ml, suggesting its potential as a drug candidate. This study further aimed to explore the potential therapeutic implications of fucoidan in managing oral cancer using network pharmacology. PharmMapper, Comparative Toxicogenomics Database and SuperPred were initially used to identify fucoidan protein targets. The identified targets were further screened against Gene Expression Omnibus (GSE23558, GSE25099 and GSE146483), OMIM, TCGA and GeneCards datasets to identify oral cancer-specific protein targets. The interactions between the selected proteins were visualised using STRING and Cytoscape. Subsequently, Database for Annotation, Visualization and Integrated Discovery was used for gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis of candidate targets. The cancer-related network was assessed using CancerGeneNet, while life expectancy based on the expression of the top 10 CytoHubba ranked hub genes was evaluated using Kaplan-Meier plots. Finally, EGFR, AKT1, HSP90AA1 and SRC were selected for docking and molecular dynamics simulation with fucoidan, using Maestro and GROMACS, respectively.
Collapse
Affiliation(s)
- Arul Jayanthi Antonisamy
- Department of Biotechnology, Mepco Schlenk Engineering College (Autonomous), Sivakasi, Tamil Nadu, India
| | - Karthikeyan Rajendran
- Department of Biotechnology, Mepco Schlenk Engineering College (Autonomous), Sivakasi, Tamil Nadu, India
| | - Premnath Dhanaraj
- Department of Biotechnology, School of Agriculture and Bio sciences, Karunya Institute of Technology and Science, Coimbatore, Tamil Nadu, India
| |
Collapse
|
15
|
Nguyen AN, Van Ngo Q, Quach TTM, Ueda S, Yuguchi Y, Matsumoto Y, Kitamura S, Ho CD, Thanh TTT. Fucoidan from brown seaweed Tubinaria decurrens: Structure and structure - anticancer activity relationship. Int J Biol Macromol 2024; 259:129326. [PMID: 38218264 DOI: 10.1016/j.ijbiomac.2024.129326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/26/2023] [Accepted: 01/06/2024] [Indexed: 01/15/2024]
Abstract
The aims of this study are to determine the structure of a fucoidan from brown seaweed Turbinaria decurrens, to investigate its anticancer activity and structure-activity relationship. SEC-MALLS, IR, ESI-MS and NMR spectra analysis indicated that dominant structure of the fucoidan, with a Mw 122.6 KDa, has a backbone of (1 → 3)- and (1 → 4)-α-L-Fucp residues, branched at C-4, sulfate groups are attached at C-2, C-3 and C-4; branches are (1 → 4)-β-D-Galp residues and sulfated at C-2. The fucoidan was hydrolyzed by HCl aqueous solution to obtain hydrolyzed fucoidans. It is assumed that native and hydrolyzed fucoidans have a rod-like conformation in solution with cross-sectional radius of gyration (Rgc) ranged from 0.53 to 1.52 nm as estimated from SAXS measurements. The fucoidans show great anticancer activity against HT29 human colon cancer cell line with IC50 ranging from 5.41 ± 0.36 to 73.52 ± 2.54 μg/mL. Anticancer activity of the fucoidan could be significantly improved by lowering molecular weight, furthermore, fucoidan required small molecular weight, small molecular weight distribution and rod-like structure with a short branch length for high anticancer activity.
Collapse
Affiliation(s)
- Anh Ngoc Nguyen
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Viet Nam
| | - Quang Van Ngo
- Institute of Chemistry, Vietnam Academy of Science and Technology, Viet Nam
| | - Thu Thi Minh Quach
- Institute of Chemistry, Vietnam Academy of Science and Technology, Viet Nam.
| | - Suzuno Ueda
- Faculty of Engineering, Osaka Electro-Communication University, Japan.
| | - Yoshiaki Yuguchi
- Faculty of Engineering, Osaka Electro-Communication University, Japan.
| | - Yuki Matsumoto
- International Polysaccharide Engineering Inc., Center for Research and Development of Bioresources, Osaka Metropolitan University, Japan.
| | - Shinichi Kitamura
- Organization for Research Promotion, Osaka Metropolitan University, Japan.
| | - Cuong Duc Ho
- Hanoi University of Science and Technology, Viet Nam.
| | - Thuy Thi Thu Thanh
- Institute of Chemistry, Vietnam Academy of Science and Technology, Viet Nam.
| |
Collapse
|
16
|
Biswas I, Precilla DS, Kuduvalli SS, Ramachandran MA, Akshaya S, Raman V, Prabhu D, Anitha TS. Unveiling the anti-glioma potential of a marine derivative, Fucoidan: its synergistic cytotoxicity with Temozolomide-an in vitro and in silico experimental study. 3 Biotech 2023; 13:397. [PMID: 37974928 PMCID: PMC10645720 DOI: 10.1007/s13205-023-03814-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 10/16/2023] [Indexed: 11/19/2023] Open
Abstract
Glioma coined as a "butterfly" tumor associated with a dismal prognosis. Marine algal compounds with the richest sources of bioactive components act as significant anti-tumor therapeutics. However, there is a paucity of studies conducted on Fucoidan to enhance the anti-glioma efficacy of Temozolomide. Therefore, the present study aimed to evaluate the synergistic anti-proliferative, anti-inflammatory and pro-apoptotic effects of Fucoidan with Temozolomide in in vitro and in silico experimental setup. The anti-proliferative effects of Temozolomide and Fucoidan were evaluated on C6 glioma cells by MTT and migration assay. Modulation of inflammatory markers and apoptosis induction was affirmed at the morphological and transcriptional level by dual staining and gene expression. Molecular docking (MD) and molecular dynamics simulation (MDS) studies were performed against the targets to rationalize the inhibitory effect. The dual-drug combination significantly reduced the cell viability and migration of glioma cells in a synergistic dose-dependent manner. At the molecular level, the dual-drug combination significantly down-regulated inflammatory genes with a concomitant upregulation of pro-apoptotic marker. In consensus with our in vitro findings, molecular docking and simulation studies revealed that the anti-tumor ligands: Temozolomide, Fucoidan with 5-(3-Methy1-trizeno)-imidazole-4-carboxamide (MTIC), and 4-amino-5-imidazole-carboxamide (AIC) had the potency to bind to the inflammatory proteins at their active sites, mediated by H-bonds and other non-covalent interactions. The dual-drug combinatorial treatment synergistically inhibited the proliferation, migration of glioma cells and promoted apoptosis; conversely with the down-regulation of inflammatory genes. However, pre-clinical experimental evidence is warranted for the possible translation of this combination. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-023-03814-6.
Collapse
Affiliation(s)
- Indrani Biswas
- Mahatma Gandhi Medical Advanced Research Institute, Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry, 607402 India
| | - Daisy S. Precilla
- Mahatma Gandhi Medical Advanced Research Institute, Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry, 607402 India
| | - Shreyas S. Kuduvalli
- Mahatma Gandhi Medical Advanced Research Institute, Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry, 607402 India
| | | | - S. Akshaya
- Jeppiaar College of Engineering, Chennai, Tamil Nadu 600119 India
| | - Venkat Raman
- Thiruvalluvar University, Vellore, Tamil Nadu 632115 India
| | - Dhamodharan Prabhu
- Centre for Drug Discovery, Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, 641021 India
| | - T. S. Anitha
- Mahatma Gandhi Medical Advanced Research Institute, Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry, 607402 India
- Present Address: Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, 605014 India
| |
Collapse
|
17
|
Laeliocattleya RA, Yunianta Y, Risjani Y, Wulan SN. In silico molecular docking, molecular dynamics, ADMET analysis of fucoidan against receptor frizzled-8 and coreceptor LRP6 in Wnt/β-Catenin pathway and in vitro analysis of fucoidan extract from Sargassum echinocarpum as β-catenin inhibitor in breast cancer cell line (MCF-7). J Biomol Struct Dyn 2023; 42:11828-11843. [PMID: 37811743 DOI: 10.1080/07391102.2023.2265488] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/24/2023] [Indexed: 10/10/2023]
Abstract
This study aimed to investigate the effect of fucoidan on the Wnt/β-Catenin pathway using both in-silico molecular docking, molecular dynamics, ADMET analysis (in frizzled-8 receptor and LRP6 coreceptor) and in-vitro experiments using MCF-7 breast cancer cells. Through the molecular docking analysis, the binding energies on the frizzled-8 receptor were -5.6, -5.1, -9.4, and -8.8 kcal/mol, respectively. Meanwhile, those on the LRP6 receptor, were -7.3, -6.2, -10.0, and -9.8 kcal/mol, respectively. The results showed that fucoidan had a favorable binding affinity for both receptors. Furthermore, it was discovered to reduce the interaction and binding affinity between Wnt agonists to frizzled-8 and LRP6 receptors. This reduction was reflected in the change in the binding energy of the fucoidan-Wnt agonist-frizzled 8 and fucoidan-Wnt agonist-LRP6 complexes, which exhibited decreases of -7.0 kcal/mol and -7.8 kcal/mol, respectively. Fucoidan was found stable in complexes with frizzled-8 receptor and co-receptor LRP6. ADMET study showed it's non-carcinogenic and can be distributed in the body. Fucoidan effectively inhibited β-catenin production, a critical factor in the Wnt/β-catenin pathway. The MCF-7 breast cancer cells were treated with fucoidan extract from S. echinocarpum at incubation times of 24, 48, and 72 h, resulting in a reduction of β-catenin levels by 95.19%, 83.88%, and 80.88%, respectively. Fucoidan also shows no significant difference in value compared to fucoidan standard (F. vesiculosus) and doxorubicin. Fucoidan exhibited antiproliferative effects against breast cancer cells, specifically through its modulation of the Wnt/β-Catenin pathway, and held great potential as an herbal anticancer agent.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Yunianta Yunianta
- Department of Food Science and Biotechnology, Faculty of Agricultural Technology, Brawijaya University, Malang, Indonesia
- AlgaEn Research Center, Brawijaya University, Malang, Indonesia
| | - Yenny Risjani
- AlgaEn Research Center, Brawijaya University, Malang, Indonesia
- Department of Aquatic Resources Management, Faculty of Fisheries and Marine Sciences, Brawijaya University, Malang, Indonesia
| | - Siti Narsito Wulan
- Department of Food Science and Biotechnology, Faculty of Agricultural Technology, Brawijaya University, Malang, Indonesia
- AlgaEn Research Center, Brawijaya University, Malang, Indonesia
| |
Collapse
|
18
|
Du H, Jin X, Jin S, Zhang D, Chen Q, Jin X, Wang C, Qian G, Ding H. Anti-Leukemia Activity of Polysaccharide from Sargassum fusiforme via the PI3K/AKT/BAD Pathway In Vivo and In Vitro. Mar Drugs 2023; 21:289. [PMID: 37233483 PMCID: PMC10221275 DOI: 10.3390/md21050289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/06/2023] [Accepted: 05/06/2023] [Indexed: 05/27/2023] Open
Abstract
Studies have shown that Sargassum fusiforme and its extracts are effective herbal treatments for leukemia. We previously found that a polysaccharide from Sargassum fusiforme, SFP 2205, stimulated apoptosis in human erythroleukemia (HEL) cells. However, the structural characterization and antitumoral mechanisms of SFP 2205 remain uncertain. Here, we studied the structural characteristics and anticancer mechanisms of SFP 2205 in HEL cells and a xenograft mouse model. The results demonstrated that SFP 2205, with a molecular weight of 41.85 kDa, consists of mannose, rhamnose, galactose, xylose, glucose, and fucose with monosaccharides composition of 14.2%, 9.4%, 11.8%, 13.7%, 11.0%, and 38.3%, respectively. On animal assays, SFP 2205 significantly inhibited growth of HEL tumor xenografts with no discernible toxicity to normal tissues. Western blotting showed that SFP 2205 therapy improved Bad, Caspase-9, and Caspase-3 protein expression, and ultimately induced HEL tumor apoptosis, indicating mitochondrial pathway involvement. Furthermore, SFP 2205 blocked the PI3K/AKT signaling pathway and 740 Y-P, an activator of the PI3K/AKT pathway, rescued the effects of SFP 2205 on HEL cell proliferation and apoptosis. Overall, SFP 2205 may be a potential functional food additive or adjuvant for preventing or treating leukemia.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Guoying Qian
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, China; (H.D.); (X.J.); (S.J.); (D.Z.); (Q.C.); (X.J.); (C.W.)
| | - Haomiao Ding
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, China; (H.D.); (X.J.); (S.J.); (D.Z.); (Q.C.); (X.J.); (C.W.)
| |
Collapse
|
19
|
Liyanage NM, Nagahawatta DP, Jayawardena TU, Jeon YJ. The Role of Seaweed Polysaccharides in Gastrointestinal Health: Protective Effect against Inflammatory Bowel Disease. Life (Basel) 2023; 13:life13041026. [PMID: 37109555 PMCID: PMC10143107 DOI: 10.3390/life13041026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/08/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a prominent global public health issue. Anti-inflammatory medications, immunosuppressants, and biological therapies are currently used as treatments. However, they are often unsuccessful and have negative consequences on human health. Thus, there is a tremendous demand for using natural substances, such as seaweed polysaccharides, to treat IBD's main pathologic treatment targets. The cell walls of marine algae are rich in sulfated polysaccharides, including carrageenan in red algae, ulvan in green algae, and fucoidan in brown algae. These are effective candidates for drug development and functional nutrition products. Algal polysaccharides treat IBD through therapeutic targets, including inflammatory cytokines, adhesion molecules, intestinal epithelial cells, and intestinal microflora. This study aimed to systematically review the potential therapeutic effects of algal polysaccharides on IBD while providing the theoretical basis for a nutritional preventive mechanism for IBD and the restoration of intestinal health. The results suggest that algal polysaccharides have significant potential in complementary IBD therapy and further research is needed for fully understanding their mechanisms of action and potential clinical applications.
Collapse
Affiliation(s)
- N M Liyanage
- Department of Marine Life Sciences, Jeju National University, Jeju 690-756, Republic of Korea
| | - D P Nagahawatta
- Department of Marine Life Sciences, Jeju National University, Jeju 690-756, Republic of Korea
| | - Thilina U Jayawardena
- Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, QC G8Z 4M3, Canada
| | - You-Jin Jeon
- Department of Marine Life Sciences, Jeju National University, Jeju 690-756, Republic of Korea
- Marine Science Institute, Jeju National University, Jeju 63333, Republic of Korea
| |
Collapse
|
20
|
Mabate B, Daub CD, Pletschke BI, Edkins AL. Comparative Analyses of Fucoidans from South African Brown Seaweeds That Inhibit Adhesion, Migration, and Long-Term Survival of Colorectal Cancer Cells. Mar Drugs 2023; 21:203. [PMID: 37103342 PMCID: PMC10144773 DOI: 10.3390/md21040203] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/28/2023] Open
Abstract
Human colorectal cancer (CRC) is a recurrent, deadly malignant tumour with a high incidence. The incidence of CRC is of increasing alarm in highly developed countries, as well as in middle to low-income countries, posing a significant global health challenge. Therefore, novel management and prevention strategies are vital in reducing the morbidity and mortality of CRC. Fucoidans from South African seaweeds were hot water extracted and structurally characterised using FTIR, NMR and TGA. The fucoidans were chemically characterised to analyse their composition. In addition, the anti-cancer properties of the fucoidans on human HCT116 colorectal cells were investigated. The effect of fucoidans on HCT116 cell viability was explored using the resazurin assay. Thereafter, the anti-colony formation potential of fucoidans was explored. The potency of fucoidans on the 2D and 3D migration of HCT116 cells was investigated by wound healing assay and spheroid migration assays, respectively. Lastly, the anti-cell adhesion potential of fucoidans on HCT116 cells was also investigated. Our study found that Ecklonia sp. Fucoidans had a higher carbohydrate content and lower sulphate content than Sargassum elegans and commercial Fucus vesiculosus fucoidans. The fucoidans prevented 2D and 3D migration of HCT116 colorectal cancer cells to 80% at a fucoidan concentration of 100 µg/mL. This concentration of fucoidans also significantly inhibited HCT116 cell adhesion by 40%. Moreover, some fucoidan extracts hindered long-term colony formation by HCT116 cancer cells. In summary, the characterised fucoidan extracts demonstrated promising anti-cancer activities in vitro, and this warrants their further analyses in pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Blessing Mabate
- Enzyme Science Programme (ESP), Department of Biochemistry and Microbiology, Faculty of Science, Rhodes University, Makhanda 6140, South Africa
| | - Chantal Désirée Daub
- Enzyme Science Programme (ESP), Department of Biochemistry and Microbiology, Faculty of Science, Rhodes University, Makhanda 6140, South Africa
| | - Brett Ivan Pletschke
- Enzyme Science Programme (ESP), Department of Biochemistry and Microbiology, Faculty of Science, Rhodes University, Makhanda 6140, South Africa
| | - Adrienne Lesley Edkins
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry and Microbiology, Rhodes University, Makhanda 6139, South Africa
| |
Collapse
|
21
|
Anti-Proliferative and Pro-Apoptotic vLMW Fucoidan Formulas Decrease PD-L1 Surface Expression in EBV Latency III and DLBCL Tumoral B-Cells by Decreasing Actin Network. Mar Drugs 2023; 21:md21020132. [PMID: 36827173 PMCID: PMC9963441 DOI: 10.3390/md21020132] [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: 01/26/2023] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Epstein-Barr virus (EBV) infects 95% of the world's population and persists latently in the body. It immortalizes B-cells and is associated with lymphomas. LCLs (lymphoblastoid cell lines, EBV latency III B-cells) inhibit anti-tumoral T-cell response following PD-L1 overexpression (programmed death-ligand 1 immune checkpoint). Many cancer cells, including some DLBCLs (diffuse large B-cell lymphomas), also overexpress PD-L1. Immunotherapies are based on inhibition of PD-L1/PD-1 interactions but present some dose-dependent toxicities. We aim to find new strategies to improve their efficiency by decreasing PD-L1 expression. Fucoidan, a polysaccharide extracted from brown seaweed, exhibits immunomodulatory and anti-tumor activities depending on its polymerization degree, but data are scarce on lymphoma cells or immune checkpoints. LCLs and DLBCLs cells were treated with native fucoidan (Fucus vesiculosus) or original very-low-molecular-weight fucoidan formulas (vLMW-F). We observed cell proliferation decrease and apoptosis induction increase with vLMW-F and no toxicity on normal B- and T-cells. We highlighted a decrease in transcriptional and PD-L1 surface expression, even more efficient for vLMW than native fucoidan. This can be explained by actin network alteration, suggesting lower fusion of secretory vesicles carrying PD-L1 with the plasma membrane. We propose vLMW-F as potential adjuvants to immunotherapy due to their anti-proliferative and proapoptotic effects and ability to decrease PD-L1 membrane expression.
Collapse
|
22
|
Immunopotentiating Activity of Fucoidans and Relevance to Cancer Immunotherapy. Mar Drugs 2023; 21:md21020128. [PMID: 36827169 PMCID: PMC9961398 DOI: 10.3390/md21020128] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/05/2023] [Accepted: 02/11/2023] [Indexed: 02/18/2023] Open
Abstract
Fucoidans, discovered in 1913, are fucose-rich sulfated polysaccharides extracted mainly from brown seaweed. These versatile and nontoxic marine-origin heteropolysaccharides have a wide range of favorable biological activities, including antitumor, immunomodulatory, antiviral, antithrombotic, anticoagulant, antithrombotic, antioxidant, and lipid-lowering activities. In the early 1980s, fucoidans were first recognized for their role in supporting the immune response and later, in the 1990s, their effects on immune potentiation began to emerge. In recent years, the understanding of the immunomodulatory effects of fucoidan has expanded significantly. The ability of fucoidan(s) to activate CTL-mediated cytotoxicity against cancer cells, strong antitumor property, and robust safety profile make fucoidans desirable for effective cancer immunotherapy. This review focusses on current progress and understanding of the immunopotentiation activity of various fucoidans, emphasizing their relevance to cancer immunotherapy. Here, we will discuss the action of fucoidans in different immune cells and review how fucoidans can be used as adjuvants in conjunction with immunotherapeutic products to improve cancer treatment and clinical outcome. Some key rationales for the possible combination of fucoidans with immunotherapy will be discussed. An update is provided on human clinical studies and available registered cancer clinical trials using fucoidans while highlighting future prospects and challenges.
Collapse
|
23
|
Lin X, Wang J, Wu X, Luo Y, Wang Y, Zhao Y. Marine‐Derived Hydrogels for Biomedical Applications. ADVANCED FUNCTIONAL MATERIALS 2023; 33. [DOI: 10.1002/adfm.202211323] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Indexed: 04/01/2025]
Abstract
Marine organisms provide novel and broad sources for the preparations and applications of biomaterials. Since the urgent requirement of bio‐hydrogels to mimic tissue extracellular matrix (ECM), the natural biomacromolecule hydrogels derived from marine sources have received increasing attention. Benefiting from their outstanding bioactivity and biocompatibility, many attempts have been made to reconstruct ECM components by applying marine‐derived natural hydrogels. Moreover, marine hydrogels have been successfully applied in biomedicine by means of microfluidics, electrospray, and bioprinting. In this review, the classification and characteristics of marine‐derived hydrogels are summarized. In particular, their role in the development of biomaterials is also introduced. Then, the recent advances in bio‐fabrication strategies for various hydrogel materials are focused upon. Besides, the influences of hydrogel types on their functions in biomedical applications are discussed in depth. Finally, critical reflections on the limitations and future development of marine‐derived hydrogels are presented.
Collapse
Affiliation(s)
- Xiang Lin
- Department of Rheumatology and Immunology Nanjing Drum Tower Hospital School of Biological Science and Medical Engineering Southeast University Nanjing 210096 China
- State Key Laboratory of Toxicology and Medical Countermeasures Beijing Institute of Pharmacology and Toxicology Beijing 100850 China
| | - Jinglin Wang
- Department of Rheumatology and Immunology Nanjing Drum Tower Hospital School of Biological Science and Medical Engineering Southeast University Nanjing 210096 China
| | - Xiangyi Wu
- Department of Rheumatology and Immunology Nanjing Drum Tower Hospital School of Biological Science and Medical Engineering Southeast University Nanjing 210096 China
| | - Yuan Luo
- State Key Laboratory of Toxicology and Medical Countermeasures Beijing Institute of Pharmacology and Toxicology Beijing 100850 China
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures Beijing Institute of Pharmacology and Toxicology Beijing 100850 China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology Nanjing Drum Tower Hospital School of Biological Science and Medical Engineering Southeast University Nanjing 210096 China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine Vision and Brain Health) Wenzhou Institute University of Chinese Academy of Sciences Wenzhou Zhejiang 325001 China
| |
Collapse
|
24
|
V. K. AD, Udduttula A, Jaiswal AK. Unveiling the secrets of marine-derived fucoidan for bone tissue engineering-A review. Front Bioeng Biotechnol 2023; 10:1100164. [PMID: 36698636 PMCID: PMC9868180 DOI: 10.3389/fbioe.2022.1100164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 12/19/2022] [Indexed: 01/10/2023] Open
Abstract
Biomedical uses for natural polysaccharides of marine origin are growing in popularity. The most prevalent polysaccharides, including alginates, agar, agarose and carrageenan, are found in seaweeds. One among these is fucoidan, which is a sulfated polysaccharide derived from brown algae. Compared to many of the biomaterials of marine origin currently in research, it is more broadly accessible and less expensive. This polysaccharide comes from the same family of brown algae from which alginate is extracted, but has garnered less research compared to it. Although it was the subject of research beginning in the 1910's, not much has been done on it since then. Few researchers have focused on its potential for biomedical applications; nevertheless, a thorough knowledge of the molecular mechanisms behind its diverse features is still lacking. This review provides a quick outline of its history, sources, and organization. The characteristics of this potential biomaterial have also been explored, with a thorough analysis concentrating on its use in bone tissue engineering. With the preclinical research completed up to this point, the fucoidan research status globally has also been examined. Therefore, the study might be utilized as a comprehensive manual to understand in depth the research status of fucoidan, particularly for applications related to bone tissue engineering.
Collapse
Affiliation(s)
- Anupama Devi V. K.
- Tissue Engineering Group, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India,School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Anjaneyulu Udduttula
- School of Engineering, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Amit Kumar Jaiswal
- Tissue Engineering Group, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India,*Correspondence: Amit Kumar Jaiswal,
| |
Collapse
|
25
|
Chauhan PS, Yadav D, Jin JO. The Therapeutic Potential of Algal Nanoparticles: A Brief Review. Comb Chem High Throughput Screen 2022; 25:2443-2451. [PMID: 34477514 DOI: 10.2174/1386207324666210903143832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 01/27/2023]
Abstract
Recently, the green synthesis of metallic nanoparticles (NPs) has received tremendous attention as a simple approach. The green pathway of biogenic synthesis of metallic NPs through microbes may provide a sustainable and environmentally friendly protocol. Green technology is the most innovative technology for various biological activities and lacks toxic effects. Reports have shown the algae-mediated synthesis of metal NPs. Algae are widely used for biosynthesis as they grow fast; they produce biomass on average ten times that of plants and are easily utilized experimentally. In the future, the production of metal NPs by different microalgae and their biological activity can be explored in diverse areas such as catalysis, medical diagnosis, and anti-biofilm applications.
Collapse
Affiliation(s)
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea
| | - Jun O Jin
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea.,Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| |
Collapse
|
26
|
Yadav D, Song M. Therapeutic Applications of Fucoidans and their Potential to Act Against COVID-19. Curr Pharm Des 2022; 28:3671-3676. [PMID: 36475344 DOI: 10.2174/1381612829666221207093215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022]
Abstract
In this review article, we present the updated evidence of therapeutic applications of fucoidan (a seaweed polysaccharide) and its novel potential to treat infectious diseases such as coronavirus disease (COVID-19). Because of their many biological activities, seaweeds have been identified as a rich and useful source of bioactive chemicals. Sulfated polysaccharides from the sea are considered a source of physiologically active chemicals that might be used in medication development. Antitumor, antiviral, antioxidant, antibacterial, anticoagulant, and immune-inflammatory properties have all been described for these compounds. By interfering at various phases of viral infection, marine sulfated polysaccharide has a virucidal effect. As a result, it opens the door to the development of antiviral treatments. Virus entry into host cells is an initial process, avoiding this type of entry makes any precautionary measure effective. The inhibitory action of certain marine sulfated polysaccharides against coronavirus was tested, and fucoidan, iota-carrageenan, and sea cucumber sulfated polysaccharides all showed a substantial antiviral impact. Fucoidan is one of the useful sulfated polysaccharides that has been widely studied and explored in various research. There are different sources of fucoidans, which have been used in the treatment of viral infection. Additionally, we highlight the mechanism of action of fuocidan against COVID-19. Hence, we could suggest that COVID-19 might be prevented and treated using these sulfated polysaccharides. This review thus highlights ample evidence to support the hypothesis that a large number of drugs have been developed from powerful compounds isolated from marine seaweeds.
Collapse
Affiliation(s)
- Dhananjay Yadav
- Department of Life Science, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Minseok Song
- Department of Life Science, Yeungnam University, Gyeongsan, 38541, South Korea
| |
Collapse
|
27
|
Wu Y, Li Y, Guo W, Liu J, Lao W, Hu P, Lin Y, Chen H. Laminaria japonica Peptides Suppress Liver Cancer by Inducing Apoptosis: Possible Signaling Pathways and Mechanism. Mar Drugs 2022; 20:704. [PMID: 36355026 PMCID: PMC9698768 DOI: 10.3390/md20110704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 01/10/2024] Open
Abstract
The anticancer properties of Laminaria japonica peptides (LJPs) have never been studied. Here, we extracted LJPs from fresh seaweed and explored their anti-liver cancer activity (in vivo and in vitro). LJPs were isolated/purified by HPLC-ESI-MS. HepG2 cell apoptosis and cell cycle were evaluated. MTT assays were used to examine the cytotoxicity of LJPs. Caspase activation of caspases 3 and 9, cleaved caspases 3 and 9, and cleaved PARP was examined by Western blotting. The PI3K/AKT pathway and the phosphorylation states of MAPKs (p38 and JNK) were examined. We found that the LJP-1 peptide had the most antiproliferative activity in H22 cells in vitro. LJP-1 blocked H22 cells in the G0/G1 phase, accompanied by inhibition of cyclin expression. LJP-1 induced apoptosis through caspase activation and regulation of the ASK1/MAPK pathway. Concurrent in vivo studies demonstrated that LJP-1 significantly inhibited tumor growth and induced tumor cell apoptosis/necrosis. In conclusion, LJPs, particularly LJP-1, exert strong inhibitory effects on liver cancer growth in vivo and in vitro. LJP-1 induces HCC cell apoptosis through the caspase-dependent pathway and G0/G1 arrest. LJP-1 induces caspase-dependent apoptosis, in part by inhibiting PI3K, MAPK signaling pathways, and cell cycle proteins. LJP-1 has the potential to be a novel candidate for human liver cancer therapeutics.
Collapse
Affiliation(s)
- Yingzi Wu
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China
| | - Yuanhui Li
- National Marketing Center, Sinopharm Group Pharmaceutical Co., Ltd., Guangzhou 510010, China
| | - Wenhai Guo
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Jie Liu
- State Key Laboratory of Respiratory Disease for Allergy and Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen 518060, China
| | - Weiguo Lao
- Department of Biochemistry, Douglass Hanly Moir Pathology, Macquarie Park, NSW 2113, Australia
| | - Penghui Hu
- Department of Oncology, Jiangmen Central Hospital, Jiangmen 529030, China
| | - Yiguang Lin
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- School of Life Sciences, University of Technology Sydney, Broadway, NSW 2007, Australia
| | - Hongjie Chen
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
28
|
Zaitseva OO, Sergushkina MI, Khudyakov AN, Polezhaeva TV, Solomina ON. Seaweed sulfated polysaccharides and their medicinal properties. ALGAL RES 2022. [DOI: 10.1016/j.algal.2022.102885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
29
|
Anti-angiogenic properties of sulfated polysaccharides fucoidans and their analogs. Russ Chem Bull 2022. [DOI: 10.1007/s11172-022-3680-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
30
|
Kiselevskiy MV, Anisimova NY, Ustyuzhanina NE, Vinnitskiy DZ, Tokatly AI, Reshetnikova VV, Chikileva IO, Shubina IZ, Kirgizov KI, Nifantiev NE. Perspectives for the Use of Fucoidans in Clinical Oncology. Int J Mol Sci 2022; 23:11821. [PMID: 36233121 PMCID: PMC9569813 DOI: 10.3390/ijms231911821] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 11/17/2022] Open
Abstract
Fucoidans are natural sulfated polysaccharides that have a wide range of biological functions and are regarded as promising antitumor agents. The activity of various fucoidans and their derivatives has been demonstrated in vitro on tumor cells of different histogenesis and in experiments on mice with grafted tumors. However, these experimental models showed low levels of antitumor activity and clinical trials did not prove that this class of compounds could serve as antitumor drugs. Nevertheless, the anti-inflammatory, antiangiogenic, immunostimulating, and anticoagulant properties of fucoidans, as well as their ability to stimulate hematopoiesis during cytostatic-based antitumor therapy, suggest that effective fucoidan-based drugs could be designed for the supportive care and symptomatic therapy of cancer patients. The use of fucoidans in cancer patients after chemotherapy and radiation therapy might promote the rapid improvement of hematopoiesis, while their anti-inflammatory, immunomodulatory, and anticoagulant effects have the potential to improve the quality of life of patients with advanced cancer.
Collapse
Affiliation(s)
- Mikhail V. Kiselevskiy
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoe Sh., Moscow 115478, Russia
- Center for Biomedical Engineering, National University of Science and Technology MISIS, Leninsky Prospect 4, Moscow 119049, Russia
| | - Natalia Yu. Anisimova
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoe Sh., Moscow 115478, Russia
- Center for Biomedical Engineering, National University of Science and Technology MISIS, Leninsky Prospect 4, Moscow 119049, Russia
| | - Nadezhda E. Ustyuzhanina
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Av., 47, Moscow 119991, Russia
| | - Dmitry Z. Vinnitskiy
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Av., 47, Moscow 119991, Russia
| | - Alexandra I. Tokatly
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Av., 47, Moscow 119991, Russia
| | - Vera V. Reshetnikova
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoe Sh., Moscow 115478, Russia
| | - Irina O. Chikileva
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoe Sh., Moscow 115478, Russia
| | - Irina Zh. Shubina
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoe Sh., Moscow 115478, Russia
| | - Kirill I. Kirgizov
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoe Sh., Moscow 115478, Russia
| | - Nikolay E. Nifantiev
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Av., 47, Moscow 119991, Russia
| |
Collapse
|
31
|
Seaweeds in the Oncology Arena: Anti-Cancer Potential of Fucoidan as a Drug—A Review. Molecules 2022; 27:molecules27186032. [PMID: 36144768 PMCID: PMC9506145 DOI: 10.3390/molecules27186032] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Marine natural products are a discerning arena to search for the future generation of medications to treat a spectrum of ailments. Meanwhile, cancer is becoming more ubiquitous over the world, and the likelihood of dying from it is rising. Surgery, radiation, and chemotherapy are the mainstays of cancer treatment worldwide, but their extensive side effects limit their curative effect. The quest for low-toxicity marine drugs to prevent and treat cancer is one of the current research priorities of researchers. Fucoidan, an algal sulfated polysaccharide, is a potent therapeutic lead candidate against cancer, signifying that far more research is needed. Fucoidan is a versatile, nontoxic marine-origin heteropolysaccharide that has received much attention due to its beneficial biological properties and safety. Fucoidan has been demonstrated to exhibit a variety of conventional bioactivities, such as antiviral, antioxidant, and immune-modulatory characteristics, and anticancer activity against a wide range of malignancies has also recently been discovered. Fucoidan inhibits tumorigenesis by prompting cell cycle arrest and apoptosis, blocking metastasis and angiogenesis, and modulating physiological signaling molecules. This review compiles the molecular and cellular aspects, immunomodulatory and anticancer actions of fucoidan as a natural marine anticancer agent. Specific fucoidan and membranaceous polysaccharides from Ecklonia cava, Laminaria japonica, Fucus vesiculosus, Astragalus, Ascophyllum nodosum, Codium fragile serving as potential anticancer marine drugs are discussed in this review.
Collapse
|
32
|
Oligo-Fucoidan supplementation enhances the effect of Olaparib on preventing metastasis and recurrence of triple-negative breast cancer in mice. J Biomed Sci 2022; 29:70. [PMID: 36109724 PMCID: PMC9479298 DOI: 10.1186/s12929-022-00855-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/08/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Seaweed polysaccharides have been recommended as anticancer supplements and for boosting human health; however, their benefits in the treatment of triple-negative breast cancers (TNBCs) and improving immune surveillance remain unclear. Olaparib is a first-in-class poly (ADP-ribose) polymerase inhibitor. Oligo-Fucoidan, a low-molecular-weight sulfated polysaccharide purified from brown seaweed (Laminaria japonica), exhibits significant bioactivities that may aid in disease management. METHODS Macrophage polarity, clonogenic assays, cancer stemness properties, cancer cell trajectory, glucose metabolism, the TNBC 4T1 cells and a 4T1 syngeneic mouse model were used to inspect the therapeutic effects of olaparib and Oligo-Fucoidan supplementation on TNBC aggressiveness and microenvironment. RESULTS Olaparib treatment increased sub-G1 cell death and G2/M arrest in TNBC cells, and these effects were enhanced when Oligo-Fucoidan was added to treat the TNBC cells. The levels of Rad51 and programmed death-ligand 1 (PD-L1) and the activation of epidermal growth factor receptor (EGFR) and adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) facilitate drug resistance and TNBC metastasis. However, the combination of olaparib and Oligo-Fucoidan synergistically reduced Rad51 and PD-L1 levels, as well as the activity of EGFR and AMPK; consistently, TNBC cytotoxicity and stemness were inhibited. Oligo-Fucoidan plus olaparib better inhibited the formation of TNBC stem cell mammospheroids with decreased subpopulations of CD44high/CD24low and EpCAMhigh cells than monotherapy. Importantly, Oligo-Fucoidan plus olaparib repressed the oncogenic interleukin-6 (IL-6)/p-EGFR/PD-L1 pathway, glucose uptake and lactate production. Oligo-Fucoidan induced immunoactive and antitumoral M1 macrophages and attenuated the side effects of olaparib, such as the promotion on immunosuppressive and protumoral M2 macrophages. Furthermore, olaparib plus Oligo-Fucoidan dramatically suppressed M2 macrophage invasiveness and repolarized M2 to the M0-like (F4/80high) and M1-like (CD80high and CD86high) phenotypes. In addition, olaparib- and Oligo-Fucoidan-pretreated TNBC cells resulted in the polarization of M0 macrophages into CD80(+) M1 but not CD163(+) M2 macrophages. Importantly, olaparib supplemented with oral administration of Oligo-Fucoidan in mice inhibited postsurgical TNBC recurrence and metastasis with increased cytotoxic T cells in the lymphatic system and decreased regulatory T cells and M2 macrophages in tumors. CONCLUSION Olaparib supplemented with natural compound Oligo-Fucoidan is a novel therapeutic strategy for reprogramming cancer stemness, metabolism and the microenvironment to prevent local postsurgical recurrence and distant metastasis. The combination therapy may advance therapeutic efficacy that prevent metastasis, chemoresistance and mortality in TNBC patients.
Collapse
|
33
|
Luo J, Li L, Zhu Z, Chang B, Deng F, Wang D, Lu X, Zuo D, Chen Q, Zhou J. Fucoidan inhibits EGFR redistribution and potentiates sorafenib to overcome sorafenib-resistant hepatocellular carcinoma. Biomed Pharmacother 2022; 154:113602. [PMID: 36029544 DOI: 10.1016/j.biopha.2022.113602] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths worldwide. Although sorafenib is a standard first-line molecule-targeted drug against advanced HCC, the drug resistance development and adverse side effects usually limit its efficacy. This study investigated the effect of fucoidan on the sorafenib sensitivity of sorafenib-resistant human HCC cell line HepG2-SR established by long-time exposure of HepG2 to sorafenib. We demonstrated fucoidan combined with sorafenib synergistically promoted apoptosis and cell cycle arrest whereas inhibited cell migration in HepG2-SR cells. This combination treatment effectively suppressed the cellular epithelial growth factor receptor (EGFR) nuclear distribution and downstream gene transcription. Interestingly, fucoidan bound the cell surface EGFR, dampening EGFR translocation to lipid raft and further nuclear distribution, restoring the sorafenib sensitivity in HepG2-SR cells. Blocking fucoidan-EGFR interaction using EGFR antibody restrained the enhanced anti-tumor effects upon the combined administration. Besides, EGFR knockdown abolished the combination treatment-improved anti-tumor efficacy. This combination also suppressed in vivo xenograft tumor growth in nude mice. Our present study uncovered that fucoidan overcame sorafenib resistance in HCC via its interaction with cell membrane EGFR and further suppression of EGFR redistribution and downstream signaling in sorafenib-resistant cells. Overall, current results suggest that simultaneous treatment of fucoidan and sorafenib might serve as a potential therapeutic strategy against sorafenib-resistant HCC.
Collapse
Affiliation(s)
- Jialiang Luo
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Lei Li
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Zhengyumeng Zhu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Bo Chang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Fan Deng
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Di Wang
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Xiao Lu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Daming Zuo
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Qingyun Chen
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, PR China.
| | - Jia Zhou
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China; Key Laboratory of Functional Proteomics of Guangdong Province, Guangzhou, Guangdong, PR China.
| |
Collapse
|
34
|
Sun Y, Xu M, Wang C, Guan S, Wang L, Cong B, Zhu W, Xu Y. Low-molecular-weight fucoidan bidirectionally regulates lipid uptake and cholesterol efflux through the p38 MAPK phosphorylation. Int J Biol Macromol 2022; 220:371-384. [PMID: 35970372 DOI: 10.1016/j.ijbiomac.2022.08.059] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/26/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022]
Abstract
Atherosclerosis (AS) is the pathological basis of many cardiovascular and cerebrovascular diseases, in which macrophage-derived foam cells are the critical step and a typical pathological feature of early atherosclerosis. We previously confirmed that low-molecular-weight fucoidan (LMWF) had a good anti-AS effect, but the mechanism is still unclear. Here with aim to investigate the inhibitory effect of LMWF on foam cells and its molecular mechanism. Oil red O staining showed that LMWF effectively alleviated lipid accumulation and the formation of foam cells. Flow cytometry detection showed that LMWF promoted foam cells apoptosis. In addition, immunofluorescence showed that LMWF inhibited macrophage scavenger receptor A1 (SR-A1)-mediated lipid uptake and promoted ATP-binding cassette transporter A1 (ABCA1)-mediated cholesterol outflow. Western blot showed that LMWF downregulated SR-A1 protein expression and upregulated ABCA1 protein expression by inhibiting p38 mitogen activated protein kinase (p38MAPK) phosphorylation. Moreover, the mRNA transcriptions of Stat1, Elk-1, and Myc were downregulated when treated with LMWF. It concluded that, LMWF achieved bidirectional regulation of SR-A1 and ABCA1, then prevented the formation of foam cells, finally ameliorated the development of AS.
Collapse
Affiliation(s)
- Yu Sun
- Medical College, Qingdao University, Qingdao 266071, China
| | - Ming Xu
- Medical College, Qingdao University, Qingdao 266071, China
| | - Changxin Wang
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Shulong Guan
- Department of Surgery, Qingdao Shinan District People's Hospital, Qingdao 266520, China
| | - Lina Wang
- Department of Blood Transfusion, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shangdong University, Qingdao 266035, China
| | - Beibei Cong
- Central Laboratory, Qingdao Stomatological Hospital, Qingdao 266001, China.
| | - Wenlong Zhu
- Business School, Qingdao University of Technology, Qingdao 266520, China.
| | - Yingjie Xu
- Central Laboratory, Qingdao Stomatological Hospital, Qingdao 266001, China.
| |
Collapse
|
35
|
Immunostimulatory effects of a polysaccharide from Pimpinella anisum seeds on RAW264.7 and NK-92 cells. Int J Biol Macromol 2022; 213:546-554. [PMID: 35660044 DOI: 10.1016/j.ijbiomac.2022.05.174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/14/2022] [Accepted: 05/26/2022] [Indexed: 12/23/2022]
Abstract
Polysaccharides from Pimpinella anisum were isolated using water at elevated temperature and DEAE Sepharose FF chromatography to examine their chemical structure and activation capacity on immune cells. P. anisum fractions (PAF1, PAF2 and PAF3) were mainly composed of neutral sugars (84.0-98.2%) and uronic acids (2.1-11.8%) with weight average molecular weight (Mw) ranging from 186.6 to 5474.5 × 103 g/mol. Polysaccharides induced a significant inflammatory response in RAW264.7 murine macrophage cells releasing nitric oxide and expressing TNF-α, IL-1β, IL-6 and IL-10 cytokines. The induction of NK-92 natural killer cells resulted in TNF-α and IFN-γ production and activation of GrB/perforin-, NKG2D- and FasL-mediated cytotoxicity. Polysaccharides triggered the phosphorylation of NF-κB, ERK, JNK and p38 proteins in RAW264.7 and NK-92 cells indicating the involvement of NF-κB and MAPKs signaling pathways. The most active polysaccharide was a galactoarabinan with complex structure.
Collapse
|
36
|
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
|
37
|
Wang WN, Li T, Li Y, Zhang Y, Wu HL, Xiang WZ, Li AF. Exopolysaccharides from the Energy Microalga Strain Botryococcus braunii: Purification, Characterization, and Antioxidant Activity. Foods 2022; 11:foods11010110. [PMID: 35010236 PMCID: PMC8750022 DOI: 10.3390/foods11010110] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/24/2021] [Accepted: 12/29/2021] [Indexed: 11/26/2022] Open
Abstract
Botryococcus braunii, a prestigious energy microalga, has recently received widespread attention because it can secrete large amounts of exopolysaccharides (EPS) with potential applications in food, cosmetics, and nutraceuticals. Unfortunately, the insufficiency of research on the bioactivity and structure–activity relationship of B. braunii EPS has impeded the downstream applications. In the present study, alcohol precipitation, deproteinization, and DEAE-cellulose column chromatography were used to extract and purify B. braunii SCS-1905 EPS. It was found that B. braunii SCS-1905 EPS were high-molecular-weight heteropolysaccharides containing uronic acid (7.43–8.83%), protein (2.30–4.04%), and sulfate groups (1.52–1.95%). Additionally, the EPS primarily comprised galactose (52.34–54.12%), glucose (34.60–35.53%), arabinose (9.41–10.32%), and minor amounts of fucose (1.80–1.99%), with the presence of a pyranose ring linked by a β-configurational glycosidic bond. Notably, the antioxidant activity of crude exopolysaccharides (CEPS) was stronger, and the half maximal inhibitory concentration (IC50) for ABTS and hydroxyl radicals was significantly lower than that of deproteinized exopolysaccharides (DEPS). Overall, this study indicated a potential application of B. braunii SCS-1905 EPS as a natural antioxidant. In summary, B. braunii EPS could be used as a potential feedstock for the production of antioxidant health foods.
Collapse
Affiliation(s)
- Wei-Nan Wang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Institution of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; (W.-N.W.); (T.L.); (H.-L.W.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tao Li
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Institution of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; (W.-N.W.); (T.L.); (H.-L.W.)
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China
| | - Yi Li
- Engineering Research Center for Tropical and Subtropical Aquatic Ecological Engineering, Ministry of Education, Jinan University, Guangzhou 510632, China; (Y.L.); (Y.Z.)
| | - Ying Zhang
- Engineering Research Center for Tropical and Subtropical Aquatic Ecological Engineering, Ministry of Education, Jinan University, Guangzhou 510632, China; (Y.L.); (Y.Z.)
| | - Hua-Lian Wu
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Institution of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; (W.-N.W.); (T.L.); (H.-L.W.)
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China
| | - Wen-Zhou Xiang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Institution of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; (W.-N.W.); (T.L.); (H.-L.W.)
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China
- Correspondence: (W.-Z.X.); (A.-F.L.); Tel.: +86-20-89023223 (W.-Z.X.); +86-20-85224366 (A.-F.L.)
| | - Ai-Fen Li
- Engineering Research Center for Tropical and Subtropical Aquatic Ecological Engineering, Ministry of Education, Jinan University, Guangzhou 510632, China; (Y.L.); (Y.Z.)
- Correspondence: (W.-Z.X.); (A.-F.L.); Tel.: +86-20-89023223 (W.-Z.X.); +86-20-85224366 (A.-F.L.)
| |
Collapse
|
38
|
Hwang J, Yadav D, Lee PC, Jin JO. Immunomodulatory effects of polysaccharides from marine algae for treating cancer, infectious disease, and inflammation. Phytother Res 2021; 36:761-777. [PMID: 34962325 DOI: 10.1002/ptr.7348] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 12/16/2022]
Abstract
A significant rise in the occurrence and severity of adverse reactions to several synthetic drugs has fueled considerable interest in natural product-based therapeutics. In humans and animals, polysaccharides from marine microalgae and seaweeds have immunomodulatory effects. In addition, these polysaccharides may possess antiviral, anticancer, hypoglycemic, anticoagulant, and antioxidant properties. During inflammatory diseases, such as autoimmune diseases and sepsis, immunosuppressive molecules can serve as therapeutic agents. Similarly, molecules that participate in immune activation can induce immune responses against cancer and infectious diseases. We aim to discuss the chemical composition of the algal polysaccharides, namely alginate, fucoidan, ascophyllan, and porphyran. We also summarize their applications in the treatment of cancer, infectious disease, and inflammation. Recent applications of nanoparticles that are based on algal polysaccharides for the treatment of cancer and inflammatory diseases have also been addressed. In conclusion, these applications of marine algal polysaccharides could provide novel therapeutic alternatives for several diseases.
Collapse
Affiliation(s)
- Juyoung Hwang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China.,Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Republic of Korea.,Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Peter Cw Lee
- Department of Biomedical Sciences, University of Ulsan College of Medicine, ASAN Medical Center, Seoul, South Korea
| | - Jun-O Jin
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China.,Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Republic of Korea.,Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| |
Collapse
|
39
|
Park AY, Nafia I, Stringer DN, Karpiniec SS, Fitton JH. Fucoidan Independently Enhances Activity in Human Immune Cells and Has a Cytostatic Effect on Prostate Cancer Cells in the Presence of Nivolumab. Mar Drugs 2021; 20:12. [PMID: 35049864 PMCID: PMC8779234 DOI: 10.3390/md20010012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/14/2021] [Accepted: 12/20/2021] [Indexed: 12/18/2022] Open
Abstract
Fucoidan compounds may increase immune activity and are known to have cancer inhibitory effects in vitro and in vivo. In this study, we aimed to investigate the effect of fucoidan compounds on ex vivo human peripheral blood mononuclear cells (PBMCs), and to determine their cancer cell killing activity both solely, and in combination with an immune-checkpoint inhibitor drug, Nivolumab. Proliferation of PBMCs and interferon gamma (IFNγ) release were assessed in the presence of fucoidan compounds extracted from Fucus vesiculosus, Undaria pinnatifida and Macrocystis pyrifera. Total cell numbers and cell killing activity were assessed using a hormone resistant prostate cancer cell line, PC3. All fucoidan compounds activated PBMCs, and increased the effects of Nivolumab. All fucoidan compounds had significant direct cytostatic effects on PC3 cells, reducing cancer cell numbers, and PBMCs exhibited cell killing activity as measured by apoptosis. However, there was no fucoidan mediated increase in the cell killing activity. In conclusion, fucoidan compounds promoted proliferation and activity of PBMCs and added to the effects of Nivolumab. Fucoidan compounds all had a direct cytostatic effect on PC3 cells, as shown through their proliferation reduction, while their killing was not increased.
Collapse
Affiliation(s)
- Ah Young Park
- Marinova Pty Ltd., Cambridge, TAS 7170, Australia; (D.N.S.); (S.S.K.); (J.H.F.)
| | - Imane Nafia
- Explicyte Immuno-Oncology, 33000 Bordeaux, France;
| | - Damien N. Stringer
- Marinova Pty Ltd., Cambridge, TAS 7170, Australia; (D.N.S.); (S.S.K.); (J.H.F.)
| | - Samuel S. Karpiniec
- Marinova Pty Ltd., Cambridge, TAS 7170, Australia; (D.N.S.); (S.S.K.); (J.H.F.)
| | - J. Helen Fitton
- Marinova Pty Ltd., Cambridge, TAS 7170, Australia; (D.N.S.); (S.S.K.); (J.H.F.)
- RDadvisor, Hobart, TAS 7006, Australia
| |
Collapse
|
40
|
Gao Y, Li Y, Niu Y, Ju H, Chen R, Li B, Song X, Song L. Chemical Characterization, Antitumor, and Immune-Enhancing Activities of Polysaccharide from Sargassum pallidum. Molecules 2021; 26:7559. [PMID: 34946640 PMCID: PMC8709291 DOI: 10.3390/molecules26247559] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 12/24/2022] Open
Abstract
Searching for natural products with antitumor and immune-enhancing activities is an important aspect of cancer research. Sargassum pallidum is an edible brown alga that has been used in Chinese traditional medicine for the treatment of tumors. However, the purification and application of its active components are still insufficient. In the present study, the polysaccharides from S. pallidum (SPPs) with antitumor and immune-enhancing activities were isolated and purified, and five polysaccharide fractions (SPP-0.3, SPP-0.5, SPP-0.7, SPP-1, and SPP-2) were obtained. The ratio of total saccharides, monosaccharide composition, and sulfated contents was determined, and their structures were analyzed by Fourier transform infrared spectroscopy. Moreover, bioactivity analysis showed that all five fractions had significant antitumor activity against three types of cancer cells (A549, HepG2, and B16), and can induce cancer cell apoptosis. In addition, the results indicated that SPPs can enhance the proliferation of immune cells and improve the expression levels of serum cytokines (IL-6, IL-1β, iNOS, and TNF-α). SPP-0.7 was identified as the most active fraction and selected for further purification, and its physicochemical properties and antitumor mechanism were further analyzed. Transcriptome sequencing result showed that SPP-0.7 can significantly induce the cell apoptosis, cytokine secretion, and cellular stress response process, and inhibit the normal physiological processes of cancer cells. Overall, SPPs and SPP-0.7 may be suitable for use as potential candidate agents for cancer therapy.
Collapse
Affiliation(s)
- Yi Gao
- College of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; (Y.G.); (B.L.)
| | - Yizhen Li
- College of Life Sciences, Qingdao Agricultural University, Qingdao 266109, China; (Y.L.); (Y.N.); (H.J.); (R.C.)
| | - Yunze Niu
- College of Life Sciences, Qingdao Agricultural University, Qingdao 266109, China; (Y.L.); (Y.N.); (H.J.); (R.C.)
| | - Hao Ju
- College of Life Sciences, Qingdao Agricultural University, Qingdao 266109, China; (Y.L.); (Y.N.); (H.J.); (R.C.)
| | - Ran Chen
- College of Life Sciences, Qingdao Agricultural University, Qingdao 266109, China; (Y.L.); (Y.N.); (H.J.); (R.C.)
| | - Bin Li
- College of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; (Y.G.); (B.L.)
| | - Xiyun Song
- College of Agronomy, Qingdao Agricultural University, Qingdao 266109, China;
| | - Lin Song
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
- Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao 266042, China
| |
Collapse
|
41
|
The Use of Oligo Fucoidan in Cancer Bearing Dogs Undergoing Chemotherapy: A Double-Blinded Study. Top Companion Anim Med 2021; 46:100616. [PMID: 34864255 DOI: 10.1016/j.tcam.2021.100616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/01/2021] [Accepted: 11/29/2021] [Indexed: 11/21/2022]
Abstract
We evaluated the effect of oligo fucoidan (Laminina Japonica) derived from oceanic brown seaweed on the quality of life in dogs with cancer undergoing chemotherapy in a double-blinded case control study. Included in this prospective study were 100 dogs with a confirmed diagnosis of cancer that were being treated with chemotherapy. Dogs were randomly assigned to be treated with oligo fucoidan (treated group; n = 68) or placebo (placebo group; n = 32). Dogs were evaluated every 2-3 weeks for 3 months with a complete blood count (CBC) and serum biochemistry profile, and a complete history and physical examination by blinded clinicians at The Veterinary Cancer Center. The owners of the dogs enrolled in the study were required at each visit to complete a Quality-of-Life Questionnaire specifically designed for cancer-bearing veterinary patients. The owners were also blinded as to whether their dog was receiving oligo fucoidan or placebo. There were no significant differences between the CBC parameters or the serum biochemical parameters of the dogs in the treated and placebo-controlled groups. There was no significant difference in the median quality of life scores between the 2 cohorts, however, when evaluating the individual quality of life metrics, 5 out of the 23 metrics showed statistically significant improvement, and none of the quality-of-life metrics declined in the oligo fucoidan group as compared to the placebo group. All of the dogs that had a positive change in overall quality of life scores were dogs that received oligo fucoidan. There were minimal adverse side effects of giving the oligo fucoidan to dogs. Treatment with oligo fucoidan was safe and improved some of the quality-of-life metrics in dogs who were being treated with chemotherapy for cancer.
Collapse
|
42
|
Intranasal Administration of Codium fragile Polysaccharide Elicits Anti-Cancer Immunity against Lewis Lung Carcinoma. Int J Mol Sci 2021; 22:ijms221910608. [PMID: 34638944 PMCID: PMC8508762 DOI: 10.3390/ijms221910608] [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: 09/06/2021] [Revised: 09/26/2021] [Accepted: 09/28/2021] [Indexed: 01/02/2023] Open
Abstract
Natural polysaccharides have shown promising effects on the regulation of immunity in animals. In this study, we examined the immune stimulatory effect of intranasally administered Codium fragile polysaccharides (CFPs) in mice. Intranasal administration of CFPs in C57BL/6 mice induced the upregulation of surface activation marker expression in macrophages and dendritic cells (DCs) in the mediastinal lymph node (mLN) and the production of interleukin-6 (IL-6), IL-12p70, and tumor necrosis factor-α in bronchoalveolar lavage fluid. Moreover, the number of conventional DCs (cDCs) was increased in the mLNs by the upregulation of C-C motif chemokine receptor 7 expression, and subsets of cDCs were also activated following the intranasal administration of CFP. In addition, the intranasal administration of CFPs promoted the activation of natural killer (NK) and T cells in the mLNs, which produce pro-inflammatory cytokines and cytotoxic mediators. Finally, daily administration of CFPs inhibited the infiltration of Lewis lung carcinoma cells into the lungs, and the preventive effect of CFPs on tumor growth required NK and CD8 T cells. Furthermore, CFPs combined with anti-programmed cell death-ligand 1 (PD-L1) antibody (Ab) improved the therapeutic effect of anti-PD-L1 Ab against lung cancer. Therefore, these data demonstrated that the intranasal administration of CFP induced mucosal immunity against lung cancer.
Collapse
|
43
|
Zhang W, Hwang J, Yadav D, An EK, Kwak M, Lee PCW, Jin JO. Enhancement of Immune Checkpoint Inhibitor-Mediated Anti-Cancer Immunity by Intranasal Treatment of Ecklonia cava Fucoidan against Metastatic Lung Cancer. Int J Mol Sci 2021; 22:9125. [PMID: 34502035 PMCID: PMC8431244 DOI: 10.3390/ijms22179125] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/11/2021] [Accepted: 08/20/2021] [Indexed: 11/16/2022] Open
Abstract
Although fucoidan, a well-studied seaweed-extracted polysaccharide, has shown immune stimulatory effects that elicit anticancer immunity, mucosal adjuvant effects via intranasal administration have not been studied. In this study, the effect of Ecklonia cava-extracted fucoidan (ECF) on the induction of anti-cancer immunity in the lung was examined by intranasal administration. In C57BL/6 and BALB/c mice, intranasal administration of ECF promoted the activation of dendritic cells (DCs), natural killer (NK) cells, and T cells in the mediastinal lymph node (mLN). The ECF-induced NK and T cell activation was mediated by DCs. In addition, intranasal injection with ECF enhanced the anti-PD-L1 antibody-mediated anti-cancer activities against B16 melanoma and CT-26 carcinoma tumor growth in the lungs, which were required cytotoxic T lymphocytes and NK cells. Thus, these data demonstrated that ECF functioned as a mucosal adjuvant that enhanced the immunotherapeutic effect of immune checkpoint inhibitors against metastatic lung cancer.
Collapse
Affiliation(s)
- Wei Zhang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai 201508, China; (W.Z.); (J.H.)
| | - Juyoung Hwang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai 201508, China; (W.Z.); (J.H.)
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (D.Y.); (E.-K.A.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (D.Y.); (E.-K.A.)
| | - Eun-Koung An
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (D.Y.); (E.-K.A.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| | - Minseok Kwak
- Department of Chemistry, Pukyong National University, Busan 48513, Korea;
| | - Peter Chang-Whan Lee
- ASAN Medical Center, Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Jun-O Jin
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai 201508, China; (W.Z.); (J.H.)
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (D.Y.); (E.-K.A.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| |
Collapse
|