Arghavan Hosseinpouri, Mehdi Mohammadi, Elham Ehsandoost, Paria Sharafi-Badr, Narges Obeidi. Chemical identification, antioxidant, cholinesterase inhibitory, and cytotoxic properties of fucoidan extracted from Persian Gulf Sargassum angustifolium[J]. Acta Oceanologica Sinica, 2022, 41(12): 133-141. doi: 10.1007/s13131-021-1961-5
Citation: Arghavan Hosseinpouri, Mehdi Mohammadi, Elham Ehsandoost, Paria Sharafi-Badr, Narges Obeidi. Chemical identification, antioxidant, cholinesterase inhibitory, and cytotoxic properties of fucoidan extracted from Persian Gulf Sargassum angustifolium[J]. Acta Oceanologica Sinica, 2022, 41(12): 133-141. doi: 10.1007/s13131-021-1961-5

Chemical identification, antioxidant, cholinesterase inhibitory, and cytotoxic properties of fucoidan extracted from Persian Gulf Sargassum angustifolium

doi: 10.1007/s13131-021-1961-5
Funds:  The Iran National Science Foundation under contract No. 96015033.
More Information
  • Corresponding author: E-mail: mohammadim@pgu.ac.ir
  • Received Date: 2020-08-24
  • Accepted Date: 2021-11-18
  • Available Online: 2022-08-30
  • Publish Date: 2022-12-30
  • Marine macroalgal sulfated fucose-containing polysaccharides, like fucoidan, have drawn significant attention due to their biotechnological potentials, such as anti-cancer, antioxidant, and anti-cholinesterase activities. The fucoidan derived from brown macroalgae Sargassum angustifolium species (FSA) was investigated for its cytotoxic effects and alterations in cell proliferation, and cell cycle-related gene expression in the present study occurred on NB4 cell line. The results showed that FSA would induce p53, p21, pro-apoptotic genes and increase expression of the p15 gene as a cell arrest marker. Also, FSA inhibited the anti-apoptotic effect of the Bcl-2 gene and decreased dnmt-1 gene expression. FSA significantly exhibited potent 2, 2-diphenyl-1-picrylhydrazyl (DPPH) radical scavenging activity (p<0.05) with an IC50 value of 0.157 mg/mL and showed moderate anti-acetylcholinesterase activity with an IC50 value of 1.20 μg/mL. These results indicated the potential of FSA for the development of therapeutic or preventive agents of cancer and Alzheimer’s disease mainly through cytotoxic effect and AChE (acetylcholinesterase) inhibition as well as additional antioxidant capacities.
  • Natural products, including plants, microbes, and marine animals, are primary resources for treating human diseases (Harvey, 2008; Ratcliffe et al., 2011). Seaweeds or marine algae are found to be an important source of therapeutics and various biological products. Polysaccharides exhibited numerous applications with different health benefits and great potential applications in many sectors such as nutraceutical, pharmaceutical, and functional food industries (Molinski et al., 2009; Martins et al., 2014). Fucoidan, natural sulfated polysaccharide (poly-L-fucopyranose), isolated from cell walls of brown seaweeds, is an important food source that possesses immunomodulatory, anti-tumor, anti-viral, antioxidant, and anti-cholinesterase inhibitory activities in cognitive diseases (Atashrazm et al., 2015; Gao et al., 2012; Wang et al., 2019; Fitton et al., 2015; Kandasamy et al., 2015). Acute Myeloid Leukemia (AML), a type of cancer associated with the proliferation of bone marrow, colonization of cells, and abnormal differentiation (Adams and Nassiri, 2015), was classified according to the World Health Organization classification of blood and lymphoid tumors (Sobin and Fleming, 1997). The AML studies had been performed to identify the principles of genetic organization that control the onset and progression of disease (Lindsley et al., 2015). Acute Promyelocytic Leukemia (APL) was found to be associated with coagulation disorders following expression of Alpha Trans Retinoic Acid (ATRA) receptor protein fusion in AML, the neoplastic proliferation of promyelocytic phenotype bone marrow cells, and blast cells translocation mutations (Warrell et al., 1991, 1994; Grimwade et al., 2000; Redner, 2002; Kakizuka et al., 1991). Patients confront multiple laboratory disorders, including D-Dimer, fibrinogen depletion, and prolonged bleeding time (Kakizuka et al., 1991). Many chemical drugs have been used as anti-leukemia agents in recent decades, especially against APL (Barbui et al., 1998). One of the most potent APL treatment drugs is arsenic trioxide (As2O3), which cannot induce apoptosis in low doses, but might induce adverse effects in high doses in patients (Shen et al., 2004). Some other studies had shown the ability of ATRA in the differentiation of abnormal promyelocytes to mature granulocytes as well as reduction of relapse incidence with the addition of ATRA to chemotherapy protocol in APL (Chen et al., 1991; Castaigne et al., 1990; Fenaux, 1999; Degos et al., 1990; Bernard et al., 1973). Also, ATRA was treated coagulation disorder in some patients with ATRA syndrome without causing aplasia (Warrell et al., 1991, 1994; Frankel et al., 1992). Anyway, the medications used for this type of leukemia have not been effective in all patients according to numerous side effects. Design of the novel drugs with high effectiveness in reducing chemotherapy and radiotherapy-induced side effects from natural products as well as a description of the possible mechanisms of action can improve the prevention treatment strategies for dealing with diseases such as APL (Zhang et al., 2018; Castaigne et al., 1990; Chen et al., 1991; Bernard et al., 1973). Various species of Sargassum (Sargassaceae), tropical brown macroalgae (seaweed), have been found to possess anti-tumor, anti-cholinesterase as well as antioxidant activities (Yende et al., 2014; Hu et al., 2016; Natarajan et al., 2009). The Persian Gulf, especially around the seashores of Bushehr and Hormozgan provinces in the southern parts of Iran, has potent sources of marine seaweeds with a wide range of biological activities (Ahmady-Asbchin and Mohammadi, 2011). The pharmaceutical and nutritional properties of the Persian Gulf seaweeds are entirely unknown (Sohrabipour and Rabiei, 1999, 2007; Vaziri Zadeh et al., 2012). In the present study, fucoidan extracted from the Sargassum angustifolium (FSA), brown algae of the Persian Gulf, was isolated and characterized. Then the cytotoxic activity of FSA on reduction of NB4 (human acute myeloid leukemia cells) cell death in APL was investigated. Also, antioxidant and anti-cholinesterase inhibition activities of FSA were assessed.

    Sargassum angustifolium was collected during summer from the intertidal zone of the Persian Gulf. The macroalgae were identified by the department of biotechnology, Persian Gulf Algae Development Technology, located in Bushehr province. The seaweed sample was washed with fresh water, then air-dried to obtain the dry biomass, and stored in plastic bags.

    Extraction was carried out as described by Sellimi et al. (2014), Benslima et al. (2021), Bahramzadeh et al. (2019), Borazjani et al. (2018) and Vaziri Zadeh et al. (2012) with slight modifications. Briefly, S. angustifolium powder was de-pigmented using 95% ethanol. The sample was suspended in a solution of 90% ethanol which included 10% formaldehyde for 8 h at 30℃ under constant stirring (200 r/min). This was done to polymerize the phenolic metabolites. Solvent filtered off, and the powder was extracted three times in succession at room temperature with 95% ethanol to remove excess formaldehyde, pigments, lipids, and other interfering phenolic metabolites. The remaining ethanol was evaporated, and the sample was brought into total dryness. Depigmented powders were air-dried and then treated in 1 L 0.1 mol/L HCl (pH=3) for 2 h at 60℃ under constant stirring (250 r/min) for the extraction. The mixture was cooled at room temperature and then centrifuged for 20 min at 4 000× g at 4℃ in a centrifuge. The resulting extract was mixed with 2 volumes of absolute ethanol and left for 12 h at 4℃. In addition, the fucoidan collected in the pellet by centrifugation (16 000× g for 10 min, 4℃) was redissolved in distilled water and dialyzed using 14 kDa cutoff dialysis membrane from Sigma-Aldrich (St. Louis, MO, USA). The supernatant was lyophilized and used for further analysis.

    A quantitative determination of the monosaccharide composition of FSA was performed by High-Performance Liquid Chromatography (HPLC) method using the isocratic HPLC (Knauer, Berlin, Germany) method. The HPLC system was equipped with a Knauer smart line RI detector and a Eurospher column (4.6 mm×250 mm, 5 µm, Knauer). The extract was passed through a 0.45 μm filter (Millipore, Westboro, MA, USA) before being injected into the HPLC. Before HPLC analysis, the fucoidan (6 mg) was hydrolyzed for 90 min in 2 mol/L Trifluoroacetic acid (TFA) at 120℃. After removing TFA, the hydrolyzed polysaccharide sample was injected into the HPLC system. A 90:10 v/v mixture of acetonitrile and deionized water was used as the mobile phase at a 2 mL/min flow rate. The resolution peaks were recorded on the HPLC chart according to the retention times of standards. The monosaccharide standard was L-fucose, galactose, mannose, glucose, and glucuronic acid (Farvin and Jacobsen, 2013).

    For structure identification analysis of FSA, Fourier transforms infrared spectroscopy (PerkinElmer FT-IR, model Spectrum RXI, Texas, USA) technique was used. Two milligrams of sample was ground with KBr until a fine particle size was produced. The signals automatically were collected using 60 scans over the range of 400–4 000 cm−1 at a resolution of 32 cm−1 and were compared to a background spectrum collected from the KBr alone at room temperature (Borazjani et al., 2018).

    The NB4 were purchased from the hematology department of Bushehr University of Medical Sciences of Iran (Bushehr) and were maintained in RPMI-1640 medium (Bioidea, Tehran, Iran) supplemented with 10% heat-inactivated Fetal Bovine Serum (FBS; Gibco, Carlsbad, USA), 100 U/mL penicillin and 100 mg/mL streptomycin, and incubated at 37℃ in a humidified 5% CO2 incubator.

    The NB4 cells (5×103 cells/well) were seeded at the 96-well plate in triplicate and after 12 h, were treated with 100 μL of various concentrations of FSA (62 µg/mL, 125 µg/mL, 250 µg/mL, 500 µg/mL, 1 000 µg/mL, and 2 000 µg/mL) for 24–72 h incubation time. The cell viability was measured by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) (Biotech, Tehran, Iran). After incubation time, 100 μL of MTT (5 mg/mL in phosphate-buffered saline (PBS)) was added to each well and then incubated at 37℃ for an additional 3 h. The supernatant was aspirated, and 100 μL DMSO (Merck, Darmstadt, Germany) was added to dissolve formazan crystals. The absorbance was measured by a microplate reader (BioTech, Munich, Germany) at 570 nm (Borazjani et al., 2018). The results were calculated as percentages of the growth inhibition by the following equation:

    $$\rm {\rm{Cell}}\;{\rm{viability}}=({\rm{ODtest}}/{\rm{ODcontrol}}) \times 100 \% . $$ (1)

    Total RNA extraction and cyclic DNA (cDNA) synthesis examined cells (5×104 cells/well) were treated with FSA (62.5 µg/mL) for 24 h using a Takara Primescript RT reagent kit (Cat. No. RR037Q, Tokyo, Japan). Total RNA was transcribed reversely into cDNA using a YTA Real-Time kit protocol (YT2551, Iran). Primer sequences of HPRT, Dnmt1, p15, p21, p53, and Bcl-2, as well as the reference gene, were listed in Table 1. The mRNA levels of Real-time quantitative PCR (qRT-PCR) were conducted with the Corbett RotorGene 3000 (Sydney, Australia), using SYBR Green I Master (Cat. No. RR820Q, Takara, Tokyo, Japan) master mix. qRT-PCR was performed in a reaction according to the manufacturer’s instructions. Briefly, the main protocols were described as follows: (1) activation at 37℃ for 15 min; (2) denaturation at 85℃ for 5 s; (3) annealing/extension at 4℃ for 5 min and agarose gel electrophoresis for evaluation of purity of isolated DNA by determining the spectrophotometric absorbance of the samples using a UV-DOC Spectrophotometer (BioRAD). All samples and control were run in triplicate. The qRT-PCR data were analyzed by a comparative threshold (Ct) method. The fold inductions of the samples were compared with the untreated samples. HPRT was an internal reference gene. The Ct cycle was determined the expression level in control cells and NB4 cells treated with FSA for 24 h. The relative expression levels were measured by the following formula: ΔΔCtCt (treated)−ΔCt (control). The expression levels were presented as n-fold differences relative to the calibrator. The value was used to plot the expression of apoptotic genes utilizing the expression of $2^{{-\Delta}{\Delta{{{C}}_{\rm{t}}}}} $.

    Table  1.  Sequences of primers used in qRT-PCR
    Base pairForward (5'–3') Reverse (5'–3')Gene
    111TGGACAGGACTGAACGTCTTGCCAGCAGGTCAGCAAAGAATTTAHPRT
    90CGGTGGGGTCATTTGTGTGCGGTTCAGGTACTCAGTCATCCBcl-2
    118ACTCATCCGATTTGGCTCTTTCCCTAGCCCCAGGATTACAAGDnmt1
    82GATGTGCAAGCGACGACAGAGAGCAAAGGCCAGCATCCTp15
    130CCTCTCACTGTCTTGTACCCTGCGTTTGGAGTGGTAGAAATCTp21
    121TAACAGTTCCTGCATGGGCGGCAGGACAGGCACAAACACGCACCp53
     | Show Table
    DownLoad: CSV

    Following the Declaration of Helsinki, 10 mL of fresh and normal citrated blood was obtained from healthy donors who gave written consent to Bushehr University of Medical Sciences (Jenny et al., 2011). According to Lonza Lymphocyte Separation Protocol (INST-17-829-2 09/11), Peripheral blood mononuclear cells (PBMCs) were isolated with Ficoll-Hypaque gradient centrifugation (Sigma, Burlington, MA, USA) (Nauseef, 2014) and mixed with an equal volume of RPMI-1640 medium containing 10% FBS. The PBMCs obtained were divided to expose to the different concentrations of FSA with the MTT assay. The cells were counted with a hemocytometer slide and counted at 0.2×106 for every 96 well plates and treated for 24 h, 48 h, and 72 h incubation in an atmosphere of 5% CO2 at 37℃, with concentrations of FSA (62.5 μg/mL, 125 μg/mL, 250 μg/mL, 500 μg/mL, 1 000 μg/mL and 2 000 μg/mL). In each of the incubations, the MTT solution was added to each well. After 3 h incubation, dimethyl sulfoxide was added, followed by shaking for 10 min to dissolve the formazan crystals. Then plates were put on a microplate reader (BioTek, Germany) to evaluate optical absorption at 570 nm.

    To evaluate the hemolytic effect of FSA, 5 mL of fresh and normal human citrated blood sample was prepared by the hematology laboratory of Bushehr University of Medical Sciences. According to Devi et al. (2014) blood was prepared and treated with 62.5 µg/mL, 125 µg/mL, 250 µg/mL, 500 µg/mL, 1 000 µg/mL, 2 000 µg/mL concentrations of FSA. Triton X-100, a known hemolytic agent, was used as positive and normal saline as a negative control. The supernatant of each sample was extracted and transferred to 96 well plates. Then, their optical absorption was read by an ELISA reader (BioTek, Munich, Germany) at 570 nm. The absorption was calculated according to the below formula, and the amount of RBC lysis was investigated.

    $$ \begin{split}&\rm H{\rm{emolysis}} (\%)=\\ &\frac{\mathrm{a}\mathrm{b}\mathrm{s}\mathrm{o}\mathrm{r}\mathrm{b}\mathrm{a}\mathrm{n}\mathrm{c}\mathrm{e}\;\mathrm{o}\mathrm{f}\;\mathrm{s}\mathrm{a}\mathrm{m}\mathrm{p}\mathrm{l}\mathrm{e}-\mathrm{a}\mathrm{b}\mathrm{s}\mathrm{o}\mathrm{r}\mathrm{b}\mathrm{a}\mathrm{n}\mathrm{c}\mathrm{e}\;\mathrm{o}\mathrm{f}\;\mathrm{n}\mathrm{e}\mathrm{g}\mathrm{a}\mathrm{t}\mathrm{i}\mathrm{v}\mathrm{e}\;\mathrm{c}\mathrm{o}\mathrm{n}\mathrm{t}\mathrm{r}\mathrm{o}\mathrm{l}}{\mathrm{a}\mathrm{b}\mathrm{s}\mathrm{o}\mathrm{r}\mathrm{b}\mathrm{a}\mathrm{n}\mathrm{c}\mathrm{e}\;\mathrm{o}\mathrm{f}\;\mathrm{p}\mathrm{o}\mathrm{s}\mathrm{i}\mathrm{t}\mathrm{i}\mathrm{v}\mathrm{e}\;\mathrm{c}\mathrm{o}\mathrm{n}\mathrm{t}\mathrm{r}\mathrm{o}\mathrm{l}-\mathrm{a}\mathrm{b}\mathrm{s}\mathrm{o}\mathrm{r}\mathrm{b}\mathrm{a}\mathrm{n}\mathrm{c}\mathrm{e}\;\mathrm{o}\mathrm{f}\;\mathrm{n}\mathrm{e}\mathrm{g}\mathrm{a}\mathrm{t}\mathrm{i}\mathrm{v}\mathrm{e}\;\mathrm{c}\mathrm{o}\mathrm{n}\mathrm{t}\mathrm{r}\mathrm{o}\mathrm{l}}.\end{split} $$ (2)

    2, 2-diphenyl-1-picrylhydrazyl (DPPH) radical scavenging activity of FSA was determined according to the method proposed by Palanisamy et al. (2017) and Yamaguchi et al. (1998). Briefly, 100 μL of the sample concentration in a range of 0.5 mg/mL to 8 mg/mL was mixed with 400 μL of 0.1 mmol/L DPPH in methyl alcohol and incubated in the dark place. After 30 min, the absorbance of the sample was measured at 517 nm using a UV–Vis spectrometer. Positive control was ascorbic acid, and 0.1 mmol/L DPPH in methyl alcohol was used as blank. The following equation calculated the percentage of DPPH scavenging activity,

    $$\begin{split} {\rm{Scavenging}}\;{\rm{activity}} =&({{A}}_0\; {\rm{sample}}-{{A}}_1\; {\rm{control}})/\\ &{{A}}_0\; {\rm{sample}}\times 100\%, \end{split}$$ (3)

    where A1 control represents the absorbance of the methanol DPPH solution without the samples, and the A0 sample is the absorbance of the methanol DPPH solution with the tested samples.

    The AChE inhibitory activity of the samples was measured by 96-well microplate colorimetric assay based on the modified method developed by Adhami et al. (2011) and Ellman et al. (1961). One hundred and twenty-five microliters of 3 mmol/L 5,5-dithiobis-2-nitrobenzoic acid (DTNB), 25 μL of 15 mmol/L acetylthiocholine iodide (ATCI) (Sigma-Aldrich Corporation, St. Louis, MO, USA), and 50 μL of Tris-HCl buffer (pH 8.0) were added to a 96 well plate followed by 25 μL of a sample (different concentrations (0.01 µg/mL to 100 µg/mL)) of FSA dissolved in 10% DMSO/buffer. The absorbance was measured by a microplate reader (Synergy MX Biotek, San Leandro, CA, USA) at 405 nm five times every 13 s. Then 25 μL of AChE (0.22 U/mL in buffer solution) was added, and after 10 min incubation, the absorbance was measured again at the same wavelength eight times every 13 s. Physostigmine and sample solvent (10% DMSO in buffer) were used as positive control and blank. The percentage of inhibition was calculated as below formula:

    $$ {\rm{AChE}}\;{\rm{inhibitory}}=100\% -(\Delta_{{\rm{SM}}}/\Delta_{{\rm{BL}}}) \times 100\% , $$ (4)

    where ΔSM is difference of sample absorption before and after adding enzyme (AChE); ΔBL is difference of blank absorption before and after adding enzyme (AChE).

    One-way analysis of variance (ANOVA) was used for MTT data analysis. The student’s T-test was used for the qRT-PCR study to compare the differences between various treatment groups. All statistical analyses were performed using SPSS 21.0 package (SPSS Inc., Chicago, IL, USA) and GraphPad Prism 5.01 (San Diego, CA, USA). The data were presented as mean±SD, and 95% confident levels (p<0.05) were statistically significant.

    Overall, sugar analysis showed that fucose was the major sugar with significant glucose and little mannose, galactose, and uronic acids, respectively (Table 2). Other monosaccharides such as rhamnose and xylose were not detected. Fucoidan polymers in the literature have demonstrated considerable variations in their monosaccharide compositions. In general, the chemical composition of fucoidan polymers is significantly different depending on species, anatomical regions, growing conditions, and extraction procedures.

    Table  2.  Sugar analysis of the polysaccharide from Sargassum angustifolium
    SugarComposition/%
    Fucose53±2.0
    Galactose0.2±0.9
    Mannose0.3±0.7
    Glucose12.6±0.4
    Uronic acids0.36±0.5
    Note: Each value represents the percentage of monosaccharides obtained in the fraction.
     | Show Table
    DownLoad: CSV

    FTIR results indicated that FSA exhibited typical absorption bands of the fucoidan spectrum. The guluronic units produced a band in the range of approximately 11 025 cm–1, and the mannuronic units created a band of about 1 100 cm–1. Therefore, the FTIR spectrum can compare the guluronic to manoronic acid concentration ratios using their two peak intensities ratios. Bands of 1 022 cm–1 and 1 074 cm–1 represented the units of guluronic acid and mannuronic acid, respectively. Peaks between 1 034 cm–1 and 1 041 cm–1 belonged to the sulfate groups. These bands are characteristics of sulfated polysaccharides. The strong band at 1 400 cm–1 represented the deformed CH2 groups. C-O-C and C-O-H tensile states represented bands in the regions of 1 125–1 250 cm–1 and 1 000–1 025 cm–1, respectively.

    The broadband at 3 424 cm–1 was related to the O-H tensile vibration. The 2 926 cm–1 and 1 652 cm–1 were related to C-H tensile vibration and O-C-O asymmetric tensile carboxylate. This finding was in support of the conclusions from Mathlouti and Koening in 1986. They reported that the absorption at 1 438 cm–1 was a sign of modified C-OH vibration with the participation of symmetric tensile vibration of the carboxylate group. The bands generated at 1 022 cm–1 and 1 074 cm–1 could be attributed to the C-O tensile vibration and the pyranose’s C-O and C-C tensile vibration. The anomeric region (750–950 cm–1) was the most widely discussed carbohydrate. The band at 947 cm–1 indicated uronic acid residues by C-O tensile vibration. The weak band at 873 cm–1 showed the vibration signal of C-H deformation of the β-mannuronic acid residues. The other weak band at 2 138 cm˗1 was the characteristic of the mannuronic acid residues.

    Further signals at 1 669 cm–1 to 800 cm–1 indicated the vibration of the asymmetric α–L glucopyranuronic ring and sulfated groups, respectively.

    As shown in Fig. 1, reduction of the concentration of FSA had decreased the inhibitory effect, therefore, at 62 µg/mL, the inhibitory effect reached to (40±0.024) µg/mL within 72 h, while its inhibitory effect after 48 h decreased to (37±0.116) µg/mL. These results indicated that FSA inhibited the survival of NB4 cells in a dose and time-dependent manner.

    Figure  1.  Survival of NB4 cell viability under fucoidan treatment with concentrations of 62 μg/mL, 125 μg/mL, 250 μg/mL, 500 μg/mL, 1 000 μg/mL and 2 000 μg/mL after 24 h, 48 h and 72 h incubation (**p<0.01, ***p<0.005 and ****p<0.000 5).

    The changes in expression levels of apoptosis-related genes from cancer cells after treatment with FSA were shown in Fig. 2. Gene expression analysis using GraphPad Prism (T-test, unpaired) revealed that the expression of p53, p21, p15, Dnmt1, and Bcl-2 genes was affected by 62 µg/mL FSA treatment after 24 h. The gene expression of pro-apoptotic genes p53, p21, p15, and Dnmt1, responsible for DNA methylation and anti-apoptotic Bcl-2, was decreased (p<0.000 1).

    Figure  2.  Real-time PCR diagram of fucoidan extracted from Sargassum angustifolium ( **p<0.01 and ****p<0.000 5).

    In the correlation analysis of genes (Fig. 3), it was found that the expression of tumor suppressor genes under the influence of FSA was significantly increased, and the expression of anti-apoptosis-inducible genes was decreased considerably. The experimental results at the mRNA level showed that p53 significantly increased fourfold than the control. At the same time, the expression of Dnmt1 and Bcl-2 genes was reduced compared to control (p<0.5). According to the gene expression diagram, p15 had the highest correlation with p21. After that, p53, Dnmt1 and Bcl-2 had also been shown to exhibit a correlation of 0.500, which may indicate the coexistence of these genes in the process of cell proliferation. These results can be used to investigate further the signalling pathways underlying FSA and their specificity of function.

    Figure  3.  Diagram of correlation analysis of apoptosis-inducing and inhibitory genes.

    The level of FSA below the Roc chart showed a 60% real positive. This indicated low sensitivity of the target genes to the FSA. The specificity of Bcl-2, Dnmt1, and p53 genes was 100%, and the specificity of p21 and p15 genes was shown to be zero. The sensitivity of Bcl-2, Dnmt1, p53 genes was 20%, 40%, and 60%, respectively, and this value for p21 and p15 genes was 60% and 80%, respectively (Fig. 4).

    Figure  4.  Diagram of gene susceptibility evaluation to fucoidan extracted from Sargassum angustifolium.

    The results showed that with increasing concentration, cell viability was changed from low to high. According to Fig. 5, PBMC survival was approximately 100% after 48 h and 72 h incubations compared to the control sample. In low concentrations, due to the decrease in glucose concentration, the effect of FSA decreased. Therefore, cell viability at low concentrations was at a minimum level.

    Figure  5.  Diagram of PBMC cells survival after treatment with concentrations of 2 000 µg/mL, 1 000 µg/mL, 500 µg/mL, 250 µg/mL, 125 µg/mL, 62.5 µg/mL related to fucoidan extracted from Sargassum angustifolium at 24h , 48 h and 72 h incubations (**p<0.01 and ****p<0.000 5).

    RBC lysis test is an important method for assessing hemolytic properties of natural materials and evaluating their hemoglobin release in the plasma upon treatment of RBCs with substances. The effect of FSA on RBC was found that FSA did not induce RBC lyses in the mentioned concentrations compared to positive control treated with Triton X-100, implying the biocompatibility of the FSA. RBCs incubated with normal saline were used as negative control and showed no hemolysis.

    DPPH method was used to measure the antioxidant activity of FSA. According to the test, when the DPPH solution (a stable free radical) was mixed with the FSA sample, it became a donor of the hydrogen atom and a non-radical form of DPPH obtained by simultaneous change of the color from purple to yellow. The length of the reaction depends on the ability of the antioxidants to hydrogenate in the FSA. Concentrations of 0.5 μg/mL, 1 μg/mL, 2 μg/mL, 4 μg/mL, 5 μg/mL, 7 μg/mL, and 10 μg/mL of FSA were used, and 50% inhibition activity of free fucoidan radicals (IC50) was between 2 μg/mL and 4 µg/mL. The antioxidant activity of FSA is related to the sulfur functional group, which refers to their ability to donate protons to free radicals. This test was repeated with mean±SD three times and was statistically significant compared with the control group (*p<0.05, **p<0.01, and ***p<0.001).

    According to “Amyloid Hypothesis”, Cholinesterase inhibitors (ChEIs) are the most promising therapeutic agents for Alzheimer (AD) disease since they could cause progress in a β-amyloid deposition in the form of senile plaques as well as the modulation of amyloid precursor protein (APP) processing (Bolognesi et al., 2009). AChE inhibition plays a key role in enhancing the cholinergic neurotransmission in the brain and decreases the aggregation of β-amyloid peptides in AD (Hodges, 2006). Different concentrations of FSA (0.01 µg/mL, 0.1 µg/mL, 1 µg/mL, 10 µg/mL, and 100 µg/mL) were tested for AChE inhibitory activity according to the microplate assay discussed in the methods, and the results were shown in Fig. 6. AChE inhibitory activity of fucoidan (p< 0.05) compared with the control group had an IC50 value of (1.20±0.445) μg/mL. FSA inhibited the enzyme by 10%–20% at lower doses (0.01 μg/mL to 0.1 μg/mL). However, higher doses, i.e., 10–100 μg/mL, induced more than 50% inhibition indicated that FSA exhibited anti-cholinesterase activity in a dose-dependent manner. Physostigmine served as a positive control, and the IC50 value for this compound was determined as 0.98 μg/mL, which was in accordance with published values (Carpinella et al., 2010; Mukherjee et al., 2007).

    Figure  6.  Diagram of the anti-cholinesterase activity of fucoidan extracted from sargassum angustifolium. The anti-cholinesterase activity of fucoidan in concentrations of 0.01 μg/mL, 0.1 μg/mL, 1 μg/mL, 10 μg/mL, and 100 μg/mL compared with the control group (10% DMSO in buffer). The values are presented as mean±SEM of 3 independent experiments (n=3) and analyzed by one-way analysis of variance (ANOVA) (*p<0.05, **p<0.01 and ***p<0.001 compared with the control group).

    Acute myeloid leukemia is a horrible hematological disorder. Although many patients with AML receive common remedies, their side effects might impress survival chances (Degos et al., 1990). In decades, brown algae have been used in the health and production of medicines (Fitton et al., 2015; Choo et al., 2016; Nagamine et al., 2009; Yuan et al., 2015). Fucoidan is a sulfated polysaccharide extracted from brown algae and has been recognized as a promising medicine for preventing and treating diseases like cancer (Wei et al., 2015). First, the present study investigated the anti-cancer property extracted from native Persian Gulf algae, Sargassum angustifolium. The previous studies have been shown the effects of standard fucoidan inhibition activities on PC-3 cell line at a concentration of 10 μg/mL to 100 μg/mL (Boo et al., 2013), MCF-7 cell line at a concentration of 80 μg/mL to 820 μg/mL (Zhang et al., 2011) and U937 cells at a concentration of 20 μg/mL to 100 μg/mL (Park et al., 2013). The FSA extracted from the native species of Sargassum angustifolium had an inhibitory effect at concentrations of 62.5 μg/mL, 125 μg/mL, 250 μg/mL, 500 μg/mL, 1 000 μg/mL, and 2 000 μg/mL on the proliferation of NB4 cells. This was similar to the effects of the fucoidan inhibitors of other species. Based on the obtained results, FSA acted in a dose and time-dependent manner, inhibiting growth and inducing apoptosis. The greatest inhibitory effect of FSA was observed at a concentration of 2 000 μg/mL at 72 h when cell growth was zero. With concentration reduction, the inhibitory effect of FSA decreased. Therefore at concentration of 62 µg/mL, IC50 was (40±0.023 9) μg/mL and (37±0.116 1) μg/mL within 48 h and 72 h, respectively. In comparison to Jin et al. (2010), fucoidan at a concentration of 62 µg/mL caused cell death, while in the study of Jin, the concentration of 150 µg/mL resulted in cell cycle arrest within 48 h. However, the cell death rate in this study was similar to those reported by Jin et al. (2010). In the meantime, multiple gene families play a role in maintaining or induction of apoptosis (Saitoh et al., 2009). Fucoidan, as previously described, induces apoptosis by various mechanisms, including activation of caspases and activation of mitochondrial signaling pathways (Park et al., 2013, 2015). In this study, the effect of fucoidan on the expression of p53, p21, and p15 pro-apoptotic genes showed that fucoidan at 62 µg/mL increased p53, p21, and p15 genes expression within 24 h. Also, in this concentration, the expression of the Bcl-2 gene decreased, which is an apoptotic inhibitor gene. The results indicated that the treatment of NB4 cells with FSA increased the expression of p53 and p21 genes that activated the mitochondrial pathway of apoptosis, and this was similar to the results of the work by Jin et al. (2010). Also, they indicated that in the NB4 cell line, fucoidan has a strong apoptotic effect. By increasing p15 expression, cellular arrest in the G0/G1 phases increases, and Bcl-2 expression reduction happens, which prevents its anti-apoptotic effect (Jin et al., 2010). In this study, the expression level of the Dnmt1, the responsible gene for DNA methylation, decreased, which also has an inhibitory effect on tumor suppressor genes. Yan et al. (2015) have also reported that fucoidan might suppress the expression of methylation genes. The correlation analysis of the present study showed an inverse relationship between the expression of Dnmt1 and p53 under the influence of FSA. In the study conducted by Wong et al., the oncogenic effect of the Dnmt1 gene on inhibition of tumor suppressor genes in AML has been described (Wong et al., 2019). According to Fig. 3, it seems that diminished expression of Dnmt1 treated with FSA increased p53 gene expression and downstream of the p21 gene. Due to the assessment of the FSA effect on PBMC, it was found that FSA had no effect on normal cells and even stimulated normal cell growth. A study by Dinesh et al. had not reported any adverse effects of fucoidan on PBMC (Wong et al., 2019). It has also been reported by Yang and colleagues that fucoidan induced mononuclear cell maturation (Yang et al., 2008). Irhimeh indicated that oral use of fucoidan increased CD34+ as a marker of T-cell growth (Irhimeh et al., 2007). Since the hemolysis rate should be in the range of 10% to 100% (Devi et al.,2014), the study of the effect of fucoidan on RBC lyses of fucoidan did not cause in the specified concentrations. Further study by Veena et al. (2007) explained increasing RBC unity and decreased membrane damage due to increased oxalate. Activation and enhancement in p53 expression indicate that fucoidan is an apoptotic component through mitochondrial pathways. Some investigations have reported the independent relation of p53 mediated apoptosis, but as shown in the present study, fucoidan increased the expression level of p53 in cancer cells. However, it has to be determined whether the fucoidan directly induces DNA damage in the S phase, leading to cell arrest, or whether it interferes with DNA regeneration by destroying the replication. These data indicated that the anti-cancer activity of fucoidan occurs through multiple pathways. Therefore, the fucoidan of Sargassum angustifolium, native to the Persian Gulf, could be suggested as a promising treatment for APL. It can be concluded that FSA was a complex molecule with a similar structure to the standard fucoidan and was capable of absorbing free radicals comparing to ascorbic acid. For the first time, the present study elucidated the anti-cholinesterase properties of the Persian Gulf brown seaweed Sargassum angustifolium. This study suggested that FSA had powerful AChE inhibitory activity, which may help prevent or slow down AD progress. The reason for their potential ChEI activity was plastoquinones and farnesylacetone derivatives (Choi et al., 2007; Natarajan et al., 2009). However, further research is underway to isolate the active compounds responsible for different biological activities of the native Persian Gulf Sargassum angustifolium.

    Acknowledgements: We thank Ahmad Ghasemi for his assistance in RT-PCR. We are also grateful for the support received from the Persian Gulf Research Institute of Persian Gulf University and the Department of Hematology and Cell Culture of Bushehr University of Medical Sciences.
  • Adams J, Nassiri M. 2015. Acute promyelocytic leukemia: a review and discussion of variant translocations. Archives of Pathology & Laboratory Medicine, 139(10): 1308–1313
    Adhami H R, Farsam H, Krenn L. 2011. Screening of medicinal plants from Iranian traditional medicine for acetylcholinesterase inhibition. Phytotherapy Research, 25(8): 1148–1152. doi: 10.1002/ptr.3409
    Ahmady-Asbchin S, Mohammadi M. 2011. Biosorption of copper ions by marine brown alga Fucus vesiculosus. Journal of Biological and Environmental Sciences, 5(15): 121–127
    Atashrazm F, Lowenthal R M, Woods G M, et al. 2015. Fucoidan and cancer: a multifunctional molecule with anti-tumor potential. Marine Drugs, 13(4): 2327–2346. doi: 10.3390/md13042327
    Bahramzadeh S, Tabarsa M, You S G, et al. 2019. Purification, structural analysis and mechanism of murine macrophage cell activation by sulfated polysaccharides from Cystoseira indica. Carbohydrate Polymers, 205: 261–270. doi: 10.1016/j.carbpol.2018.10.022
    Barbui T, Finazzi G, Falanga A. 1998. The impact of all-trans-retinoic acid on the coagulopathy of acute promyelocytic leukemia. Blood, 91(9): 3093–3102. doi: 10.1182/blood.V91.9.3093
    Benslima A, Sellimi S, Hamdi M, et al. 2021. Brown seaweed Cystoseira schiffneri as a promising source of sulfated fucans: seasonal variability of structural, chemical, and antioxidant properties. Food Science & Nutrition, 9(3): 1551–1563
    Bernard J, Weil M, Boiron M, et al. 1973. Acute promyelocytic leukemia: results of treatment by daunorubicin. Blood, 41(4): 489–496. doi: 10.1182/blood.V41.4.489.489
    Bolognesi M L, Matera R, Minarini A, et al. 2009. Alzheimer’s disease: new approaches to drug discovery. Current Opinion in Chemical Biology, 13(3): 303–308. doi: 10.1016/j.cbpa.2009.04.619
    Boo H J, Hong J Y, Kim S C, et al. 2013. The anticancer effect of fucoidan in PC-3 prostate cancer cells. Marine Drugs, 11(8): 2982–2999. doi: 10.3390/md11082982
    Borazjani N J, Tabarsa M, You S G, et al. 2018. Purification, molecular properties, structural characterization, and immunomodulatory activities of water soluble polysaccharides from Sargassum angustifolium. International Journal of Biological Macromolecules, 109: 793–802. doi: 10.1016/j.ijbiomac.2017.11.059
    Carpinella M C, Andrione D G, Ruiz G, et al. 2010. Screening for acetylcholinesterase inhibitory activity in plant extracts from Argentina. Phytotherapy Research, 24(2): 259–263. doi: 10.1002/ptr.2923
    Castaigne S, Chomienne C, Daniel M T, et al. 1990. All-trans retinoic acid as a differentiation therapy for acute promyelocytic leukemia. I. clinical results. Blood, 76(9): 1704–1709. doi: 10.1182/blood.V76.9.1704.1704
    Chen Zixing, Xue Yongquan, Zhang Ri, et al. 1991. A clinical and experimental study on all-trans retinoic acid-treated acute promyelocytic leukemia patients. Blood, 78(6): 1413–1419. doi: 10.1182/blood.V78.6.1413.1413
    Choi B W, Ryu G, Park S H, et al. 2007. Anticholinesterase activity of plastoquinones from Sargassum sagamianum: lead compounds for Alzheimer’s disease therapy. Phytotherapy Research, 21(5): 423–426. doi: 10.1002/ptr.2090
    Choo G S, Lee H N, Shin S A, et al. 2016. Anticancer effect of fucoidan on DU-145 prostate cancer cells through inhibition of PI3K/Akt and MAPK pathway expression. Marine Drugs, 14(7): 126. doi: 10.3390/md14070126
    Degos L, Chomienne C, Daniel M T, et al. 1990. Treatment of first relapse in acute promyelocytic leukaemia with all-trans retinoic acid. Lancet, 336(8728): 1440–1441
    Devi L B, Das S K, Mandal A B. 2014. Impact of surface functionalization of AgNPs on binding and conformational change of hemoglobin (Hb) and hemolytic behavior. The Journal of Physical Chemistry C, 118(51): 29739–29749. doi: 10.1021/jp5075048
    Ellman G L, Courtney K D, Andres Jr V, et al. 1961. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochemical Pharmacology, 7(2): 88–90, IN1, 91–95
    Farvin K H S, Jacobsen C. 2013. Phenolic compounds and antioxidant activities of selected species of seaweeds from Danish coast. Food Chemistry, 138(2–3): 1670–1681
    Fenaux P, Chastang C, Chevret S, et al. 1999. A randomized comparison of all transretinoic acid (ATRA) followed by chemotherapy and ATRA plus chemotherapy and the role of maintenance therapy in newly diagnosed acute promyelocytic leukemia. Blood, 94(4): 1192–2000. doi: 10.1182/blood.V94.4.1192
    Fitton J H, Stringer D N, Karpiniec S S. 2015. Therapies from fucoidan: an update. Marine Drugs, 13(9): 5920–5946. doi: 10.3390/md13095920
    Frankel S R, Eardley A, Lauwers G, et al. 1992. The “retinoic acid syndrome” in acute promyelocytic leukemia. Annals of Internal Medicine, 117(4): 292–296. doi: 10.7326/0003-4819-117-4-292
    Gao Yonglin, Li Chunmei, Yin Jungang, et al. 2012. Fucoidan, a sulfated polysaccharide from brown algae, improves cognitive impairment induced by infusion of Aβ peptide in rats. Environmental Toxicology and Pharmacology, 33(2): 304–311. doi: 10.1016/j.etap.2011.12.022
    Grimwade D, Biondi A, Mozziconacci M J, et al. 2000. Characterization of acute promyelocytic leukemia cases lacking the classic t (15; 17): results of the European Working Party. Blood, 96(4): 1297–1308
    Harvey A L. 2008. Natural products in drug discovery. Drug Discovery Today, 13(19–20): 894–901
    Hodges J R. 2006. Alzheimer’s centennial legacy: origins, landmarks and the current status of knowledge concerning cognitive aspects. Brain, 129(11): 2811–2822. doi: 10.1093/brain/awl275
    Hu Pei, Li Zhixiong, Chen Mingcang, et al. 2016. Structural elucidation and protective role of a polysaccharide from Sargassum fusiforme on ameliorating learning and memory deficiencies in mice. Carbohydrate Polymers, 139: 150–158. doi: 10.1016/j.carbpol.2015.12.019
    Irhimeh M R, Fitton J H, Lowenthal R M. 2007. Fucoidan ingestion increases the expression of CXCR4 on human CD34+ cells. Experimental Hematology, 35(6): 989–994. doi: 10.1016/j.exphem.2007.02.009
    Jenny M, Klieber M, Zaknun D, et al. 2011. In vitro testing for anti-inflammatory properties of compounds employing peripheral blood mononuclear cells freshly isolated from healthy donors. Inflammation Research, 60(2): 127–135. doi: 10.1007/s00011-010-0244-y
    Jin J O, Song M G, Kim Y N, et al. 2010. The mechanism of fucoidan-induced apoptosis in leukemic cells: involvement of ERK1/2, JNK, glutathione, and nitric oxide. Molecular Carcinogenesis, 49(8): 771–782
    Kakizuka A, Miller Jr W H, Umesono K, et al. 1991. Chromosomal translocation t (1.5; 17) in human acute promyelocytic leukemia fuses RARα with a novel putative transcription factor, PML. Cell, 66(4): 663–674. doi: 10.1016/0092-8674(91)90112-C
    Kandasamy S, Khan W, Kulshreshtha G, et al. 2015. The fucose containing polymer (FCP) rich fraction of Ascophyllum nodosum (L. ) Le Jol. protects Caenorhabditis elegans against Pseudomonas aeruginosa by triggering innate immune signaling pathways and suppression of pathogen virulence factors. Algae, 30(2): 147–161
    Lindsley R C, Mar B G, Mazzola E, et al. 2015. Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood, 125(9): 1367–1376. doi: 10.1182/blood-2014-11-610543
    Martins A, Vieira H, Gaspar H, et al. 2014. Marketed marine natural products in the pharmaceutical and cosmeceutical industries: tips for success. Marine Drugs, 12(2): 1066–1101. doi: 10.3390/md12021066
    Molinski T F, Dalisay D S, Lievens S L, et al. 2009. Drug development from marine natural products. Nature Reviews Drug Discovery, 8(1): 69–85. doi: 10.1038/nrd2487
    Mukherjee P K, Kumar V, Houghton P J. 2007. Screening of Indian medicinal plants for acetylcholinesterase inhibitory activity. Phytotherapy Research, 21(12): 1142–1145. doi: 10.1002/ptr.2224
    Nagamine T, Hayakawa K, Kusakabe T, et al. 2009. Inhibitory effect of fucoidan on Huh7 hepatoma cells through downregulation of CXCL12. Nutrition and Cancer, 61(3): 340–347. doi: 10.1080/01635580802567133
    Natarajan S, Shanmugiahthevar K P, Kasi P D. 2009. Cholinesterase inhibitors from Sargassum and Gracilaria gracilis: seaweeds inhabiting South Indian coastal areas (Hare Island, Gulf of Mannar). Natural Product Research, 23(4): 355–369. doi: 10.1080/14786410802156036
    Nauseef W M. 2014. Isolation of human neutrophils from venous blood. In: Quinn M T, DeLeo F R, eds. Neutrophil Methods and Protocols. Totowa: Humana Press, 13–18
    Palanisamy S, Vinosha M, Marudhupandi T, et al. 2017. Isolation of fucoidan from Sargassum polycystum brown algae: structural characterization, in vitro antioxidant and anticancer activity. International Journal of Biological Macromolecules, 102: 405–412. doi: 10.1016/j.ijbiomac.2017.03.182
    Park H Y, Choi I W, Kim G Y, et al. 2015. Fucoidan induces G1 arrest of the cell cycle in EJ human bladder cancer cells through down-regulation of pRB phosphorylation. Revista Brasileira de Farmacognosia, 25(3): 246–251. doi: 10.1016/j.bjp.2015.03.011
    Park H S, Hwang H J, Kim G Y, et al. 2013. Induction of apoptosis by fucoidan in human leukemia U937 cells through activation of p38 MAPK and modulation of Bcl-2 family. Marine Drugs, 11(7): 2347–2364. doi: 10.3390/md11072347
    Ratcliffe N A, Mello C B, Garcia E S, et al. 2011. Insect natural products and processes: new treatments for human disease. Insect Biochemistry and Molecular Biology, 41(10): 747–769. doi: 10.1016/j.ibmb.2011.05.007
    Redner R L. 2002. Variations on a theme: the alternate translocations in APL. Leukemia, 16(10): 1927–1932. doi: 10.1038/sj.leu.2402720
    Saitoh Y, Nagai Y, Miwa N. 2009. Fucoidan-Vitamin C complex suppresses tumor invasion through the basement membrane, with scarce injuries to normal or tumor cells, via decreases in oxidative stress and matrix metalloproteinases. International Journal of Oncology, 35(5): 1183–1189
    Sellimi S, Kadri N, Barragan-Montero V, et al. 2014. Fucans from a Tunisian brown seaweed Cystoseira barbata: structural characteristics and antioxidant activity. International Journal of Biological Macromolecules, 66: 281–288. doi: 10.1016/j.ijbiomac.2014.02.041
    Shen Zhixiang, Shi Zhanzhong, Fang Jing, et al. 2004. All-trans retinoic acid/As2O3 combination yields a high quality remission and survival in newly diagnosed acute promyelocytic leukemia. Proceedings of the National Academy of Sciences of the United States of America, 101(15): 5328–5335. doi: 10.1073/pnas.0400053101
    Sobin L H, Fleming I D. 1997. TNM classification of malignant tumors, fifth edition (1997). Union internationale contre le cancer and the American joint committee on cancer. Cancer, 80(9): 1803–1804. doi: 10.1002/(SICI)1097-0142(19971101)80:9<1803::AID-CNCR16>3.0.CO;2-9
    Sohrabipour J, Rabiei R. 1999. A list of marine algae of seashores of Persian Gulf and Oman Sea in the Hormozgan Province. Iranian Journal of Botany, 8(1): 131–162
    Sohrabipour J, Rabiei R. 2007. The checklist of green algae of the Iranian coastal lines of the Persian Gulf and Gulf of Oman. The Iranian Journal of Botany, 13(2): 146–149
    Vaziri Zadeh A, Mohammadi M, Fakhri A. 2012. Ecological assessment of mollusc communities in the rocky shores of Bushehr Province. Journal of Oceanography, 3(9): 55–61
    Veena C K, Josephine A, Preetha S, et al. 2007. Effect of sulphated polysaccharides on erythrocyte changes due to oxidative and nitrosative stress in experimental hyperoxaluria. Human & Experimental Toxicology, 26(12): 923–932
    Wang Yu, Xing Maochen, Cao Qi, et al. 2019. Biological activities of fucoidan and the factors mediating its therapeutic effects: a review of recent studies. Marine Drugs, 17(3): 183. doi: 10.3390/md17030183
    Warrell Jr R P, Frankel S R, Miller Jr W H, et al. 1991. Differentiation therapy of acute promyelocytic leukemia with tretinoin (all-trans-retinoic acid). New England Journal of Medicine, 324(20): 1385–1393. doi: 10.1056/NEJM199105163242002
    Warrell Jr R P, Maslak P, Eardley A, et al. 1994. Treatment of acute promyelocytic leukemia with all-trans retinoic acid: an update of the New York experience. Leukemia, 8(6): 929–933
    Wei Chunmei, Xiao Qing, Kuang Xingyi, et al. 2015. Fucoidan inhibits proliferation of the SKM-1 acute myeloid leukaemia cell line via the activation of apoptotic pathways and production of reactive oxygen species. Molecular Medicine Reports, 12(5): 6649–6655. doi: 10.3892/mmr.2015.4252
    Wong K K, Lawrie C H, Green T M. 2019. Oncogenic roles and inhibitors of DNMT1, DNMT3A, and DNMT3B in acute myeloid leukaemia. Biomarker Insights, 14: 1–12. doi: 10.1177/1177271919846454
    Yamaguchi T, Takamura H, Matoba T, et al. 1998. HPLC method for evaluation of the free radical-scavenging activity of foods by using 1, 1-diphenyl-2-picrylhydrazyl. Bioscience, Biotechnology, and Biochemistry, 62(6): 1201–1204
    Yan Mingde, Yao C J, Chow J M, et al. 2015. Fucoidan elevates microRNA-29b to regulate DNMT3B-MTSS1 axis and inhibit EMT in human hepatocellular carcinoma cells. Marine Drugs, 13(10): 6099–6116. doi: 10.3390/md13106099
    Yang Meixiang, Ma Chunhong, Sun Jintang, et al. 2008. Fucoidan stimulation induces a functional maturation of human monocyte-derived dendritic cells. International Immunopharmacology, 8(13–14): 1754–1760
    Yende S R, Harle U N, Chaugule B B. 2014. Therapeutic potential and health benefits of Sargassum species. Pharmacognosy Review, 8(15): 1–7. doi: 10.4103/0973-7847.125514
    Yuan Xiumei, Zeng Yawei, Nie Kaiying, et al. 2015. Extraction optimization, characterization and bioactivities of a major polysaccharide from Sargassum thunbergii. PLoS ONE, 10(12): e0144773. doi: 10.1371/journal.pone.0144773
    Zhang Zhongyuan, Teruya K, Eto H, et al. 2011. Fucoidan extract induces apoptosis in MCF-7 cells via a mechanism involving the ROS-dependent JNK activation and mitochondria-mediated pathways. PLoS ONE, 6(11): e27441. doi: 10.1371/journal.pone.0027441
    Zhang Qingyu, Wang Feixuan, Jia Keke, et al. 2018. Natural product interventions for chemotherapy and radiotherapy-induced side effects. Frontiers in Pharmacology, 9: 1253. doi: 10.3389/fphar.2018.01253
  • Relative Articles

  • Cited by

    Periodical cited type(4)

    1. Elham Ehsandoost, Mohammad-Taghi Golmakani, Mohammad Hadi Eskandari, et al. Effect of lecithin on the antioxidant mechanism of action of fucoidan in stripped bulk sunflower oil and respective oil in water emulsion. International Journal of Biological Macromolecules, 2025, 289: 138837. doi:10.1016/j.ijbiomac.2024.138837
    2. Abbas Sadeghi, Ali Rajabiyan, Najme Meygoli Nezhad, et al. A review on Persian Gulf brown algae as potential source for anticancer drugs. Algal Research, 2024, 79: 103446. doi:10.1016/j.algal.2024.103446
    3. Nuohan Zhang, Wenwei Chen, Xinyu Li, et al. Enzyme-Assisted Ultrasonic Extraction and Antioxidant Activities of Polysaccharides from Schizochytrium limacinum Meal. Foods, 2024, 13(6): 880. doi:10.3390/foods13060880
    4. Melis Cokdinleyen, Luana Cristina dos Santos, Cristiano José de Andrade, et al. A Narrative Review on the Neuroprotective Potential of Brown Macroalgae in Alzheimer’s Disease. Nutrients, 2024, 16(24): 4394. doi:10.3390/nu16244394

    Other cited types(0)

  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Figures(6)  / Tables(2)

    Article Metrics

    Article views (528) PDF downloads(30) Cited by(4)
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return