Next Article in Journal
Brown Macroalgae (Phaeophyceae): A Valuable Reservoir of Antimicrobial Compounds on Northern Coast of Spain
Next Article in Special Issue
Immunomodulatory, Antioxidant, Anticancer, and Pharmacokinetic Activity of Ulvan, a Seaweed-Derived Sulfated Polysaccharide: An Updated Comprehensive Review
Previous Article in Journal
Conus regius-Derived Conotoxins: Novel Therapeutic Opportunities from a Marine Organism
Previous Article in Special Issue
Carbonic Anhydrase Inhibitors from Marine Natural Products
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Marine Polyphenol Phlorotannins as a Natural Sleep Aid for Treatment of Insomnia: A Review of Sedative–Hypnotic Effects and Mechanism of Action

1
Department of Food Science and Technology, Institute of Food Science, Pukyong National University, Busan 48513, Republic of Korea
2
Research Division of Food Functionality, Korea Food Research Institute, Wanju 55365, Republic of Korea
3
Department of Seafood Science and Technology, The Institute of Marine Industry, Gyeongsang National University, 38 Cheondaegukchi-gil, Tongyeong-si 53064, Republic of Korea
4
Department of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan 47227, Republic of Korea
5
Anti-Aging Research Center and Core-Facility Center for Tissue Regeneration, Dong-Eui University, Busan 47340, Republic of Korea
*
Author to whom correspondence should be addressed.
Mar. Drugs 2022, 20(12), 774; https://doi.org/10.3390/md20120774
Submission received: 19 November 2022 / Revised: 4 December 2022 / Accepted: 9 December 2022 / Published: 10 December 2022
(This article belongs to the Special Issue The Mechanisms of Action of Bioactive Marine Natural Products)

Abstract

:
Insomnia is a common sleep disorder. Natural sleep aids are gaining worldwide popularity as alternatives to prescription drugs for improving sleep. Recently, numerous studies have investigated the sedative–hypnotic effects of the polyphenols of terrestrial plants. The hypnotic effects of marine polyphenols have also been studied in recent years. Phlorotannins are marine polyphenols that are found only in brown algae. Phlorotannins exert sedative–hypnotic effects via the gamma-aminobutyric acid type A-benzodiazepine receptor. In addition, the brown seaweed Ecklonia cava supplement containing phlorotannins has been approved by the Ministry of Food and Drug Safety as a health-functional ingredient that helps improve sleep quality. Currently, it is meaningful to deal with the sedative–hypnotic effects of phlorotannins as natural sleep aids. The current review comprehensively presents the sedative–hypnotic effects in animal models and human clinical trials as well as their mechanism of action, extraction, purification, and safety.

Graphical Abstract

1. Introduction

Polyphenols are one of the most common classes of secondary metabolites found in terrestrial and marine plants [1]. Polyphenols from terrestrial plants and marine algae have different chemical structures [2]. Phlorotannins are a major polyphenolic class found only in brown algae, whereas red and green algae contain the most phenolic compounds, such as flavonoids, phenolic acids, and bromophenols [3]. Phlorotannins are oligomers and polymers of the monomeric unit phloroglucinol (1,3,5-tri-hydroxybenzene) with molecular weights in the range of 250–1738 Da [4,5]. They are an extremely diverse group, and individual phlorotannin compounds are structurally similar [5]. To date, approximately 150 phlorotannins have been isolated from various brown seaweeds [6,7].
In the last three decades, phlorotannins have been extensively investigated and shown to possess various biological properties including antioxidative, antidiabetic, anti-aging, anti-inflammatory, anti-allergic, neuroprotective, and memory-enhancing properties [7,8,9,10,11,12,13,14,15,16]. However, the sedative–hypnotic effects of phlorotannins have only recently been studied [4,17,18,19]. It has been demonstrated that phlorotannins from brown seaweeds have hypnotic effects in in vitro and in vivo studies as well as in clinical trials. Studies on the hypnotic effects of phlorotannins have shown their characteristics as agonists for gamma-aminobutyric acid type A (GABAA)-benzodiazepine (BZD) receptors. Several researchers have reviewed the biological properties of marine polyphenol phlorotannins; however, their hypnotic effects have not been reviewed.
Sleep deprivation and disorders, such as insomnia, are now associated with numerous serious health problems and are appraised as emerging global epidemics that cause social and financial burdens [20]. As insomnia becomes more common, herbal sleep aids are gaining popularity worldwide as alternatives to prescription drugs to treat insomnia or improve sleep quality [21,22]. Most sedative–hypnotic drugs have numerous side effects, such as impairment of memory, cognitive function, and general daytime performance; therefore, their use is generally not recommended beyond 4 weeks [23,24]. In addition, long-term administration typically results in dependence and tolerance [25]. Thus, sedative–hypnotic effects of herbal plants or their phytochemicals have been widely reported, such as valerian (Valeriana officinalis), St. John’s wort (Hypericum perforatum), kava kava (Piper methysticum), passion flower (Passiflora incarnata), and hops (Humulus lupulus) [19,24]. Although numerous studies have been conducted on the hypnotic effects of herbal plants, few studies have investigated marine polyphenol phlorotannins. Currently, it is noteworthy to mention the sedative–hypnotic effects of the marine polyphenol phlorotannins. This review aims to present the extraction, purification, safety, sedative–hypnotic effects, and mechanism of action of phlorotannins.

2. Extraction and Purification of Phlorotannins

Phlorotannins from brown seaweeds have been extracted using traditional extraction techniques (Soxhlet, solid–liquid, and liquid–liquid extractions), enzymatic hydrolysis, and solvent extraction [26,27]. Currently, the solvents used in extraction methods should be non-toxic and inexpensive [28]. Ethanol extraction is the preferred method in the food industry because of its safety for human consumption, the convenience of processing, and low cost [29]. The correct selection of the extraction solvent, solvent concentration, temperature, and time are variables that directly influence the yield of biologically active compounds. To determine the scale-up, it is important to alter different parameters to optimize the extraction process [30]. One of the most consistent multivariate techniques in analytical optimization is response surface methodology [31]. According to a report by Yoon et al. [26], in which the sedative–hypnotic compound was extracted from Ecklonia cava, the active total phlorotannin content, yield of phlorotannins, and sleep duration were independent variables [26]. Sleep duration and total phlorotannin content were highly correlated (R2 = 0.9102), and the optimal conditions for extraction time, extraction temperature, and ethanol concentration were 22.8 h, 80 °C, and 95.0%, respectively [26]. The optimal conditions for the yield of phlorotannins were 24.0 h extraction time, 80 °C, and 88.3% humidity. There were several differences between the hypnotic effect under optimal conditions and the sleep effect under other conditions. Following optimization, the total phlorotannins and yield of phlorotannins were approximately 570 mg phloroglucinol equivalents per gram (mg PGE/g) and 7.5%, respectively, which were 1.8- and 1.5-fold higher than the 315.4 mg PGE/g and 4.9% obtained under the conditions that showed the lowest results.
Brown algae products are considered a major safety concern for arsenic [32]. In particular, brown algae have the highest arsenic concentration, whereas red and green algae have phlorotannin-chelating activity [33]. According to the Ministry of Food and Drug Safety (MFDS), the acceptable daily intake (ADI) of arsenic is 150 μg for a person weighing 60 kg. High arsenic intake can cause numerous health concerns, including skin and lung cancers [34], hyperkeratosis [35], diabetes [36,37], and vascular diseases [38]. In 2014, Kim et al. [39] reported that 1 g of crude phlorotannin extract included 180 μg arsenic. These results show that, according to the MFDS, the crude phlorotannin extract exceeded the ADI of arsenic. Several efforts have been made to reduce arsenic through purification [40]. Macroporous adsorption resins have been extensively used to purify phytochemicals and bioactive compounds from food and plant extracts due to their high adsorption capacity, easy recyclability, and various functional groups [41]. These resins can be used for the absorption of organic constituents because of their weak polar and hydrophobic properties [42]. A previous study showed that the arsenic content of the final phlorotannin product was 48 μg/g, which was 3.75-fold lower than that of the crude phlorotannin extract purified using HP-20 resin [39]. These results suggest that the purification of phlorotannins using HP-20 resin is effective for arsenic removal.

3. Safety and Toxicity of Phlorotannins

3.1. In Vitro

In human and animal cell lines, such as human epidermal (HaCaT), Henrietta Lacks (HeLa), human colon adenocarcinoma (Caco-2), highly tumorigenic (HT1080), HepG2, B16F10 melanoma, KU812, RBL-2H3, MRC-5, HT-29, human fibroblast cells, and rat vibrissae immortalized dermal papilla cell line [43,44,45,46,47,48,49,50,51,52,53,54,55,56,57], phlorotannins decreased the generation of reactive oxygen species (ROS), malondialdehyde levels, deoxyribonucleic acid (DNA) damage, and ultraviolet B (UVB) radiation-induced damage. In addition to these activities, phlorotannins also reduce binding between immunoglobulin E (IgE) and the high-affinity IgE receptor as well as the expression of several genes, including tumor necrosis factor alpha, interleukin-1β (IL-1β), IL-6, IL-8β expression, prostaglandin E2 (PGE2) release, cyclooxygenase 1 (COX-1), COX-2, microsomal prostaglandin E synthase-1 (mPGES-1), nuclear factor-kappa B, activator protein-1 reporter, the mitogen-activated protein kinase (MAPK) signaling pathway, and melanin synthesis. Moreover, phlorotannins have been reported to inhibit the growth of HeLa, A549, HT1080, and HT29 tumor cells. Phlorotannins also inhibit 5-reductase activity and increase cell viability and glutathione concentration. Notably, to the best of our knowledge, studies have reported that phlorotannins exhibit biological activities without toxicity in human and animal cell lines.

3.2. In Vivo

In animals, the safety and toxicity of phlorotannins have been evaluated in fish, such as seabream (Pagrus major), tiger puffer (Fugu rubripes) [58], zebrafish (Danio rerio) embryos [59], and zebrafish [60]; in rodents, such as Institute of Cancer Research (ICR) mice [60,61], HR-1 hairless male mice [46], and Sprague–Dawley (SD) rats [61,62]; and in Beagle dogs [63]. In fish, phlorotannins reduced ROS levels, cell death, generation of thiobarbituric acid reactive substances, and adipogenic factors, such as peroxisome proliferator-activated receptors (PPAR), CCAAT-enhancer-binding proteins (C/EBP), fatty acid-binding protein 11a (FABP11a), and sterol regulatory element-binding factor-1 (SREBF-1) with minor side effects including writhing and gasping for several seconds (after which the fish calmed down) and some discharged oral mucus. However, the survival rate of these fish was 100% [58,59,60]. In rodents, phlorotannins reduce the final body weight, the high-fat diet-induced elevation of liver fat, low-density lipoprotein cholesterol [60], lipid peroxidation, protein carbonylation, epidermal height, and MAPK expression [46]. Phlorotannins also increased the levels of plasma triglycerides, total cholesterol [60], and α-amylase to the normal range [62] and increased the survival rate of rodents until the end of the experiments. In Beagles, mild side effects such as soft stool and diarrhea were reported after phlorotannin treatment. However, the survival rate of the Beagles was 100% at the end of the treatment [63]. Further research is needed to confirm the potential for phlorotannins as health-functional feed agents and in veterinary medicine for various animal species.

3.3. Clinical Human Studies

In humans, phlorotannins can be used as food supplements and functional food ingredients. Phlorotannins have been reported to possess numerous advantages [64,65,66] and mild side effects [64]. A study by Paradis et al. [66] found that phlorotannins isolated from Fucus vesiculosus and Ascophyllum nodosum decreased the incremental areas under the curve for plasma insulin, the post-load plasma insulin concentration, the plasma glucose area under the curve, and the postprandial insulin concentration in 23 participants following treatment with 250 mg/capsule. Moreover, phlorotannins elevated the level of a surrogate marker for insulin sensitivity in all participants.
A study by Baldrick et al. [65] reported that phlorotannins extracted from A. nodosum decreased DNA damage and did not significantly improve C-reactive protein, antioxidant status, or inflammatory cytokines in 80 participants between 30 and 65 years old following administration of 100 mg/capsule for 8 weeks. Similarly, Shin et al. [64] reported that phlorotannins isolated from E. cava decreased the total cholesterol/high-density lipoprotein cholesterol level, body fat ratio, atherogenic index, total cholesterol/low-density lipoprotein cholesterol level, body mass index, waist circumference, and waist/hip ratio in 107 participants (138 men and 69 women) following administration of 72 and 144 mg/capsule. In another study, phlorotannins successfully increased sleep duration scores and inhibited the onset of wakefulness after sleep [67]. However, phlorotannins showed no serious adverse effects, such as mild fatigue, dizziness, nausea, and abdominal distension [67]. The mechanisms of action of other classes of phlorotannins that have not been tested should be further investigated to evaluate their potential as novel pharmaceutical agents for humans.

3.4. The Regulation of Phlorotannins as Human Supplements

The European Food Safety Authority Panel on Dietetic Products, Nutrition, and Allergies, pursuant to Regulation No. 258/97, announced that novel food supplements from phlorotannins (marketed as SeaPolynolTM) are safe for human consumption [68]. The application of phlorotannins as food supplements and functional food ingredients was reported by Turck et al. [68] and Catarino et al. [69]. As a food supplement, the daily intake of phlorotannins depends on the age of the consumer. In adolescents (12–14 years of age), the maximum daily intake was 163 mg/day. For those above 14 years of age and adults, the daily intakes were 230 mg/day and 263 mg/day, respectively.
E. cava extract is the main ingredient of Seanol-F sold by Simply Healthy LLC. (Leander, TX, USA) and was reported as a New Dietary Ingredient by the US Food and Drug Administration (FDA) in 2008 [70]. Daily intake was 47 mg/day for those aged >12 years. In 2015, the MFDS recognized E. cava extract (No. 2015-6) as a functional ingredient in health-functional foods that helps improve sleep quality [71].

4. Sedative–Hypnotic Effects of Phlorotannins in Animal Models

4.1. Phlorotannin Preparations

Various phlorotannin preparations, including ethanol and enzymatic extracts, and purified phlorotannin supplements have been investigated to evaluate their sedative–hypnotic effects [17,72,73]. Additionally, several solvent fractions from E. cava ethanol extracts have been shown to have hypnotic effects [17]. Ethanol [17] and enzymatic [72] extracts decreased sleep latency and increased sleep duration, respectively, in a pentobarbital-induced sleep test in mice. In a study by Cho et al. [73], phlorotannin supplementation with 90% phlorotannin potentiated sleep induced by pentobarbital in a dose-dependent manner. Among the solvent fractions (hexane, ethyl acetate, and butanol), the ethyl acetate fraction, which was characterized as a polyphenol-rich fraction, showed the best hypnotic effect. These results indicate that phlorotannins are responsible for the sedative–hypnotic effects of brown seaweed extracts or phlorotannin supplementation.
The pentobarbital-induced sleep test is a well-known method to assess suspected sedative–hypnotic activity [74,75] (Figure 1). However, it is difficult to identify only pentobarbital-induced sleep tests because the hypnotic effects of compounds can be induced by toxicity or other side effects [76]. In addition, this method only evaluates sleep quantity, such as sleep latency and sleep duration. Meanwhile, an analysis of sleep structure based on polygraphic recordings can verify sleep quality, including delta activity during non-rapid eye movement sleep (NREMS), and sleep–wake profiles [77] (Figure 1) (Table 1).
In the polygraphic recordings, E. cava ethanol extract at 500 mg/kg significantly increased the amount of NREMS by 71.4% during the first 2 h after oral administration [78]. In addition, phlorotannin supplementation at doses of 250 and 500 mg/kg significantly increased the amount of NREMS 2 h immediately after oral administration [4]. The phlorotannin supplement (500 mg/kg) showed sleep-promoting effects similar to those of diazepam (6 mg/kg). However, while diazepam reduced the electroencephalogram (EEG) power density of NREMS (frequency range, 0.5–4 Hz), phlorotannin supplementation did not show any significant difference. These results suggest that phlorotannin supplementation induces natural sleep without adverse effects following the onset of sleep [79].
Sedative–hypnotic effects can be evaluated using a caffeine-induced sleep disruption model [8]. Caffeine promotes wakefulness by blocking the activation of the adenosine A2A receptor [80,81]. Oral administration (500 mg/kg) of the phlorotannin supplement attenuated caffeine (25 mg/kg)-induced sleep disruption, and its effects were comparable to those of the hypnotic drug zolpidem (10 mg/kg). This result implies that phlorotannin supplementation may be useful in relieving the transitory insomnia symptoms caused by caffeine consumption.

4.2. Individual Phlorotannin Compounds

Phlorotannins are an extremely heterogeneous group, and approximately 150 different phlorotannin compounds have been isolated from various brown seaweeds [6,7,82]. Among the phlorotannin constituents, the six major phlorotannins (dieckol, eckstolonol, eckol, triphlorethol A, fucodiphlorethol G, and 6,6′-bieckol) were found to have sedative-hypnotics (Figure 2) [4]. All six phlorotannin compounds (50 mg/kg) significantly increased the sleep duration in mice treated with a hypnotic dose of pentobarbital (Table 2).
In particular, dieckol is the most abundant phlorotannin preparation from brown seaweeds and has been considered an indicator compound [39]. Yoon et al. [19] reported that dieckol has sleep-enhancing effects by analyzing its effects on the sleep–wake profiles of C57BL/6N mice using the recorded EEG and electromyogram (EMG). Dieckol administration increased NREMS duration dose-dependently. Dieckol (100 and 150 mg/kg) significantly increased NREMS levels 2 h after administration. In particular, there were no significant differences in NREMS or sleep latency between dieckol (150 mg/kg) and zolpidem (10 mg/kg). In addition, there were no significant differences in EEG power density (0–20 Hz) and delta activity (frequency range of 0.5–4 Hz) of NREMS between dieckol and the vehicle, whereas zolpidem decreased delta activity. These results imply that dieckol increases sleep quantity without inducing any adverse effects. Eckstolonol and triphlorethol A were also analyzed for their effects on sleep–wake profiles [4,83]. Eckstolonol (50 mg/kg) and triphlorethol A (50 mg/kg) significantly decreased sleep latency and increased the amount of NREMS in C57BL/6N mice, without affecting delta activity (0.5–4 Hz), similar to dieckol. Eckstolonol induced sleep effects via a GABAergic mechanism; however, the inducing effects in NREMS were moderate compared to diazepam (6 mg/kg). Triphlorethol A (50 mg/kg) showed no significant difference from zolpidem (10 mg/kg) in NREMS (Table 2).
Phlorotannins are oligomers and polymers of phloroglucinol (1,3,5-tri-hydroxybenzene), and approximately 150 phlorotannins have been isolated from various brown seaweeds. However, among the individual phlorotannin compounds, in vitro and in vivo studies have only been conducted on the six major phlorotannins (dieckol, eckstolonol, eckol, triphlorethol A, fucodiphlorethol G, and 6,6′-bieckol). Therefore, it is necessary to investigate the hypnotic effects of phloroglucinol, which is the basic structural unit of a phlorotannin, and to study further the synergistic effects of phlorotannin compounds.

5. Sleep-Promoting Effects of Phlorotannins in Clinical Trials

The promising sleep-promoting effects of phlorotannins have also been observed in humans. A clinical case study demonstrated the effects of acupuncture therapy and the phlorotannin-rich E. cava extract (500 mg/day) on sleep disturbance in patients with amyotrophic lateral sclerosis (ALS) [84]. After 5 months of combined treatment, the Pittsburgh Sleep Quality Index (PSQI) score decreased from 13 to 8 in patients with ALS. Additionally, in a randomized, double-blind, placebo-controlled trial, the effectiveness and safety of phlorotannins at a dose of 500 mg/day for 7 d in adults with self-reported sleep disturbances were investigated [67]. Sleep parameters were assessed at baseline and 1 week using the PSQI and polysomnography (PSG). Um et al. reported that phlorotannin supplementation significantly increased the “Sleep duration” scores compared to those in the placebo group. However, there were no significant differences in total PSQI scores. PSG recordings revealed that wakefulness after sleep onset, total wake time, and the respiratory disturbance index during supine rapid eye movement sleep (REMS) were significantly lower in the phlorotannin group than in the placebo group. There were no serious adverse effects, and some side effects did not correlate with the consumption of phlorotannins. However, because this sample size was small and the treatment period was limited to 7 days, a large-scale controlled/long-term clinical trial is required. In addition, metabolomics studies are required to explain how phlorotannins affect sleep after intake in the human brain. Finally, this evidence provides insights into the physiological function of phlorotannins, suggesting that they might be used as a natural sleep agent.

6. Action Mechanism of Phlorotannins

Previous in vitro and in vivo studies have demonstrated that both preparations and the individual constituents of phlorotannins exert sedative–hypnotic effects via a GABAergic (gamma-aminobutyric acid-mediated) mechanism (Figure 3). The BZD-binding site of the GABAA receptor has been considered the most important molecular target for the development of sedative–hypnotic drugs [85,86]. Currently, BZD agonists are the most commonly prescribed hypnotics. These agents act as positive allosteric modulators as BZD ligands; potentiate GABA-mediated inhibitory neurotransmission, which results in membrane hyperpolarization by allowing chloride anion (Cl-) influx; and, subsequently, exhibit sedative–hypnotic effects [87,88]. Similar to BZD agonists, phlorotannins induce sleep by acting as allosteric agonists of GABAA receptors and binding to the BZD-binding site of GABAA receptors.
The in vitro GABAergic mechanism of phlorotannins was demonstrated by the ligand-binding and functional assays of GABAA receptors. In the GABAA-BZD receptor-binding assay, ethanol, methanol, and enzymatic extracts from the brown seaweed E. cava were significantly displaced [3H]-flumazenil binding [17,72]. Among the three extracts containing phlorotannins, the ethanol extract had the lowest half-maximal inhibitory concentration (IC50) (0.127 mg/mL) [17]. The binding affinities (IC50) of the ethyl acetate fraction [17] and purified phlorotannin supplement [4] from the E. cava ethanol extract were 0.019 and 0.012 mg/mL, respectively. In addition, four phlorotannin compounds (eckstolonol, eckol, triphlorethol-A, and dieckol) were identified as ligands for the BZD-binding site of GABAA receptors [17]. The significant results of the binding assay imply that phlorotannins act as BZD ligands to GABAA receptors; however, they cannot provide information to demonstrate phlorotannins are BZD agonists (positive allosteric modulators). This can be demonstrated using a functional assay based on electrophysiological measurements. Purified phlorotannin supplementation and dieckol potentiated the GABA-mediated inward current cultured neurons, and their activities were blocked by the co-application of a BZD antagonist flumazenil, such as the BZD agonist diazepam [18]. Flumazenil inhibits the sedative–hypnotic activity of diazepam by blocking the binding of diazepam to the BZD site of GABAA receptors [89]. These results provide direct evidence that purified phlorotannins and dieckol act as positive allosteric modulators of GABAA receptors. It has been demonstrated that eckstolonol is a partial BZD agonist based on electrophysiological measurements and pharmacophore modeling [4]. The GABAergic mechanism of phlorotannins has also been demonstrated through in vivo animal assays. The hypnotic activities of all phlorotannin preparations (except butanol and hexane fractions) and the individual constituents shown in Table 3 were completely blocked by the BZD antagonist flumazenil [4,17,18,72].

7. Conclusions

Numerous studies on the biological properties of phlorotannins and their constituents have been performed. Recently, marine polyphenol phlorotannins have been demonstrated to have sedative–hypnotic effects in animal models and clinical trials. In Korea, the MFDS has approved E. cava supplementation as a functional ingredient for health foods that help improve sleep quality. The sedative–hypnotic effects of phlorotannins suggest that they possess other neuropharmacological activities. It is necessary to demonstrate their anxiolytic or antidepressant effects and possible mechanisms of action. Red and green seaweeds contain non-phlorotannin polyphenol compounds. To date, the sedative–hypnotic effects of red or green seaweeds have not yet been reported. Therefore, these seaweeds could be promising raw materials for finding sedative–hypnotic compounds with novel structures or mechanisms.

Author Contributions

Conceptualization, S.K. and S.C.; investigation, S.K., D.K., M.Y.U., J.-S.C., Y.H.C., M.Y. and S.C.; writing—original draft preparation, S.K. and S.C.; writing—review and editing, S.K., D.K., M.Y.U., J.-S.C., Y.H.C. and S.C.; supervision, S.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Korea Institute of Marine Science & Technology Promotion (KIMST), funded by the Ministry of Oceans and Fisheries (20220488, 20220128).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lee, S.H.; Jeon, Y.J. Anti-Diabetic Effects of Brown Algae Derived Phlorotannins, Marine Polyphenols through Diverse Mechanisms. Fitoterapia 2013, 86, 129–136. [Google Scholar] [CrossRef] [PubMed]
  2. Shibata, T.; Fujimoto, K.; Nagayama, K.; Yamaguchi, K.; Nakamura, T. Inhibitory Activity of Brown Algal Phlorotannins against Hyaluronidase. Int. J. Food Sci. Technol. 2002, 37, 703–709. [Google Scholar] [CrossRef]
  3. Chen, L.; Liu, R.; He, X.; Pei, S.; Li, D. Effects of Brown Seaweed Polyphenols, a Class of Phlorotannins, on Metabolic Disorders: Via Regulation of Fat Function. Food Funct. 2021, 12, 2378–2388. [Google Scholar] [CrossRef]
  4. Cho, S.; Yoon, M.; Pae, A.N.; Jin, Y.H.; Cho, N.C.; Takata, Y.; Urade, Y.; Kim, S.; Kim, J.S.; Yang, H.; et al. Marine Polyphenol Phlorotannins Promote Non-Rapid Eye Movement Sleep in Mice via the Benzodiazepine Site of the GABAA Receptor. Psychopharmacology 2014, 231, 2825–2837. [Google Scholar] [CrossRef] [PubMed]
  5. Isaza Martínez, J.H.; Torres Castañeda, H.G. Preparation and Chromatographic Analysis of Phlorotannins. J. Chromatogr. Sci. 2013, 51, 825–838. [Google Scholar] [CrossRef] [Green Version]
  6. Khan, F.; Jeong, G.J.; Khan, M.S.A.; Tabassum, N.; Kim, Y.M. Seaweed-Derived Phlorotannins: A Review of Multiple Biological Roles and Action Mechanisms. Mar. Drugs 2022, 20, 384. [Google Scholar] [CrossRef]
  7. Rajan, D.K.; Mohan, K.; Zhang, S.; Ganesan, A.R. Dieckol: A Brown Algal Phlorotannin with Biological Potential. Biomed. Pharmacother. 2021, 142, 111988. [Google Scholar] [CrossRef]
  8. Kwon, S.; Yoon, M.; Lee, J.; Moon, K.D.; Kim, D.; Kim, S.B.; Cho, S. A Standardized Phlorotannin Supplement Attenuates Caffeine-Induced Sleep Disruption in Mice. Nutrients 2019, 11, 556. [Google Scholar] [CrossRef] [Green Version]
  9. Wijesekara, I.; Yoon, N.Y.; Kim, S.K. Phlorotannins from Ecklonia Cava (Phaeophyceae): Biological Activities and Potential Health Benefits. BioFactors 2010, 36, 408–414. [Google Scholar] [CrossRef]
  10. Lee, S.; Youn, K.; Kim, D.H.; Ahn, M.R.; Yoon, E.; Kim, O.Y.; Jun, M. Anti-Neuroinflammatory Property of Phlorotannins from Ecklonia Cava on Aβ25-35-Induced Damage in PC12 Cells. Mar. Drugs 2018, 17, 7. [Google Scholar] [CrossRef]
  11. Kang, M.C.; Wijesinghe, W.A.J.P.; Lee, S.H.; Kang, S.M.; Ko, S.C.; Yang, X.; Kang, N.; Jeon, B.T.; Kim, J.; Lee, D.H.; et al. Dieckol Isolated from Brown Seaweed Ecklonia Cava Attenuates Type II Diabetes in Db/Db Mouse Model. Food Chem. Toxicol. 2013, 53, 294–298. [Google Scholar] [CrossRef] [PubMed]
  12. Li, Y.; Qian, Z.J.; Ryu, B.M.; Lee, S.H.; Kim, M.M.; Kim, S.K. Chemical Components and Its Antioxidant Properties in Vitro: An Edible Marine Brown Alga, Ecklonia Cava. Bioorg. Med. Chem. 2009, 17, 1963–1973. [Google Scholar] [CrossRef] [PubMed]
  13. Jang, S.K.; Yu, J.M.; Kim, S.T.; Kim, G.H.; Park, D.W.; Lee, D.I.; Joo, S.S. An Aβ42 Uptake and Degradation via Rg3 Requires an Activation of Caveolin, Clathrin and Aβ-Degrading Enzymes in Microglia. Eur. J. Pharmacol. 2015, 758, 1–10. [Google Scholar] [CrossRef] [PubMed]
  14. Wang, C.H.; Li, X.F.; Jin, L.F.; Zhao, Y.; Zhu, G.J.; Shen, W.Z. Dieckol Inhibits Non-Small–Cell Lung Cancer Cell Proliferation and Migration by Regulating the PI3K/AKT Signaling Pathway. J. Biochem. Mol. Toxicol. 2019, 33, e22346. [Google Scholar] [CrossRef] [Green Version]
  15. Lee, A.J.; Ashkar, A.A. The Dual Nature of Type I and Type II Interferons. Front. Immunol. 2018, 9, 2061. [Google Scholar] [CrossRef] [Green Version]
  16. Um, M.Y.; Lim, D.W.; Son, H.J.; Cho, S.; Lee, C. Phlorotannin-Rich Fraction from Ishige Foliacea Brown Seaweed Prevents the Scopolamine-Induced Memory Impairment via Regulation of ERK-CREB-BDNF Pathway. J. Funct. Foods 2018, 40, 110–116. [Google Scholar] [CrossRef]
  17. Cho, S.; Yang, H.; Jeon, Y.J.; Lee, C.J.; Jin, Y.H.; Baek, N.I.; Kim, D.; Kang, S.M.; Yoon, M.; Yong, H.; et al. Phlorotannins of the Edible Brown Seaweed Ecklonia Cava Kjellman Induce Sleep via Positive Allosteric Modulation of Gamma-Aminobutyric Acid Type A-Benzodiazepine Receptor: A Novel Neurological Activity of Seaweed Polyphenols. Food Chem. 2012, 132, 1133–1142. [Google Scholar] [CrossRef]
  18. Kwon, S.; Jung, J.H.; Cho, S.; Moon, K.D.; Lee, J. Dieckol Is a Natural Positive Allosteric Modulator of GABAA-Benzodiazepine Receptors and Enhances Inhibitory Synaptic Activity in Cultured Neurons. Nutr. Neurosci. 2021, 24, 835–842. [Google Scholar] [CrossRef]
  19. Yoon, M.; Kim, J.S.; Seo, S.; Lee, K.; Um, M.Y.; Lee, J.; Jung, J.; Cho, S. Dieckol, a Major Marine Polyphenol, Enhances Non-Rapid Eye Movement Sleep in Mice via the GABAA-Benzodiazepine Receptor. Front. Pharmacol. 2020, 11, 1–10. [Google Scholar] [CrossRef]
  20. McCarthy, B.; O’Neill, G.; Abu-Ghannam, N. Potential Psychoactive Effects of Microalgal Bioactive Compounds for the Case of Sleep and Mood Regulation: Opportunities and Challenges. Mar. Drugs 2022, 20, 493. [Google Scholar] [CrossRef]
  21. Hu, Z.; Oh, S.; Ha, T.W.; Hong, J.T.; Oh, K.W. Sleep-Aids Derived from Natural Products. Biomol. Ther. 2018, 26, 343–349. [Google Scholar] [CrossRef] [PubMed]
  22. Neubauer, D.N. New and Emerging Pharmacotherapeutic Approaches for Insomnia. Int. Rev. Psychiatry 2014, 26, 214–224. [Google Scholar] [CrossRef] [PubMed]
  23. Roth, T.; Drake, C. Evolution of Insomnia: Current Status and Future Direction. Sleep Med. 2004, 5, 23–30. [Google Scholar] [CrossRef] [PubMed]
  24. Cho, S.; Shimizu, M. Natural sleep aids and polyphenols as treatments for insomnia. In Bioactive Nutraceuticals and Dietary Supplements in Neurological and Brain Disease; Elsevier: Amsterdam, The Netherlands, 2015; pp. 141–151. [Google Scholar]
  25. Fang, X.S.; Hao, J.F.; Zhou, H.Y.; Zhu, L.X.; Wang, J.H.; Song, F.Q. Pharmacological studies on the sedative-hypnotic effect of Semen ziziphi spinosae (Suanzaoren) and Radix et Rhizoma Salviae miltiorrhizae (Danshen) extracts and the synergistic effect of their combinations. Phytomedicine 2010, 17, 75–80. [Google Scholar] [CrossRef]
  26. Yoon, M.; Kim, J.S.; Um, M.Y.; Yang, H.; Kim, J.; Kim, Y.T.; Lee, C.; Kim, S.B.; Kwon, S.; Cho, S. Extraction Optimization for Phlorotannin Recovery from the Edible Brown Seaweed Ecklonia Cava. J. Aquat. Food Prod. Technol. 2017, 26, 801–810. [Google Scholar] [CrossRef]
  27. Kadam, S.U.; Tiwari, B.K.; O’Donnell, C.P. Application of Novel Extraction Technologies for Bioactives from Marine Algae. J. Agric. Food Chem. 2013, 61, 4667–4675. [Google Scholar] [CrossRef]
  28. Cotas, J.; Leandro, A.; Monteiro, P.; Pacheco, D.; Figueirinha, A.; Goncąlves, A.M.M.; da Silva, G.J.; Pereira, L. Seaweed Phenolics: From Extraction to Applications. Mar. Drugs 2020, 18, 384. [Google Scholar] [CrossRef]
  29. Shi, J.; Nawaz, H.; Pohorly, J.; Mittal, G.; Kakuda, Y.; Jiang, Y. Extraction of Polyphenolics from Plant Material for Functional Foods—Engineering and Technology. Food Rev. Int. 2005, 21, 139–166. [Google Scholar] [CrossRef]
  30. Sridhar, A.; Vaishampayan, V.; Senthil Kumar, P.; Ponnuchamy, M.; Kapoor, A. Extraction Techniques in Food Industry: Insights into Process Parameters and Their Optimization. Food Chem. Toxicol. 2022, 166, 113207. [Google Scholar] [CrossRef]
  31. Bezerra, M.A.; Santelli, R.E.; Oliveira, E.P.; Villar, L.S.; Escaleira, L.A. Response Surface Methodology (RSM) as a Tool for Optimization in Analytical Chemistry. Talanta 2008, 76, 965–977. [Google Scholar] [CrossRef]
  32. Huang, Z.; Bi, R.; Musil, S.; Pétursdóttir, Á.H.; Luo, B.; Zhao, P.; Tan, X.; Jia, Y. Arsenic Species and Their Health Risks in Edible Seaweeds Collected along the Chinese Coastline. Sci. Total Environ. 2022, 847, 157429. [Google Scholar] [CrossRef] [PubMed]
  33. Šlejkovec, Z.; Kápolna, E.; Ipolyi, I.; van Elteren, J.T. Arsenosugars and Other Arsenic Compounds in Littoral Zone Algae from the Adriatic Sea. Chemosphere 2006, 63, 1098–1105. [Google Scholar] [PubMed]
  34. Hong, Y.S.; Song, K.H.; Chung, J.Y. Health Effects of Chronic Arsenic Exposure. J. Prev. Med. Public Health 2014, 47, 245–252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Mandal, P. An Insight of Environmental Contamination of Arsenic on Animal Health. Emerg. Contam. 2017, 3, 17–22. [Google Scholar]
  36. Chen, C.J.; Wang, S.L.; Chiou, J.M.; Tseng, C.H.; Chiou, H.Y.; Hsueh, Y.M.; Chen, S.Y.; Wu, M.M.; Lai, M.S. Arsenic and Diabetes and Hypertension in Human Populations: A Review. Toxicol. Appl. Pharmacol. 2007, 222, 298–304. [Google Scholar] [CrossRef]
  37. Singh, A.P.; Goel, R.K.; Kaur, T. Mechanisms Pertaining to Arsenic Toxicity. Toxicol. Int. 2011, 18, 87–93. [Google Scholar]
  38. Engel, R.R.; Hopenhayn-Rich, C.; Receveur, O.; Smith, A.H. Vascular Effects of Chronic Arsenic Exposure: A Review. Epidemiol. Rev. 1994, 16, 184–209. [Google Scholar]
  39. Kim, J.; Yoon, M.; Yang, H.; Jo, J.; Han, D.; Jeon, Y.J.; Cho, S. Enrichment and Purification of Marine Polyphenol Phlorotannins Using Macroporous Adsorption Resins. Food Chem. 2014, 162, 135–142. [Google Scholar] [CrossRef]
  40. Pal, P.; Sen, M.; Manna, A.; Pal, J.; Pal, P.; Roy, S.; Roy, P. Contamination of Groundwater by Arsenic: A Review of Occurrence, Causes, Impacts, Remedies and Membrane-Based Purification. J. Integr. Environ. Sci. 2009, 6, 295–316. [Google Scholar]
  41. Jiang, Z.; Wang, Y. Stepwise Elution by High-Speed Counter-Current Chromatography Combined with a Modified Macroporous Resin to Isolate and Purify Antioxidant Phenolics from Discarded Jackfruit (Artocarpusheterophyllus Lam.) Peels. Anal Methods 2020, 12, 4674–4681. [Google Scholar] [CrossRef]
  42. Ren, J.; Zheng, Y.; Lin, Z.; Han, X.; Liao, W. Macroporous Resin Purification and Characterization of Flavonoids from Platycladus Orientalis (L.) Franco and Their Effects on Macrophage Inflammatory Response. Food Funct. 2017, 8, 86–95. [Google Scholar] [CrossRef]
  43. Wijesekara, I.; Kim, S.K.; Li, Y.; Li, Y.X. Phlorotannins as Bioactive Agents from Brown Algae. Process Biochem. 2011, 46, 2219–2224. [Google Scholar]
  44. Joe, M.-J.; Kim, S.-N.; Choi, H.-Y.; Shin, W.-S.; Park, G.-M.; Kang, D.-W.; Kim, Y.K. The Inhibitory Effects of Eckol and Dieckol from Ecklonia Stolonifera on the Expression of Matrix Metalloproteinase-1 in Human Dermal Fibroblasts. Biol. Pharm. Bull. 2006, 29, 1735–1739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Lee, S.H.; Kang, S.M.; Sok, C.H.; Hong, J.T.; Oh, J.Y.; Jeon, Y.J. Cellular Activities and Docking Studies of Eckol Isolated from Ecklonia Cava (Laminariales, Phaeophyceae) as Potential Tyrosinase Inhibitor. Algae 2015, 30, 163–170. [Google Scholar]
  46. Zhen, A.X.; Hyun, Y.J.; Piao, M.J.; Sameera Madushan Fernando, P.D.; Kang, K.A.; Ahn, M.J.; Yi, J.M.; Kang, H.K.; Koh, Y.S.; Lee, N.H.; et al. Eckol Inhibits Particulate Matter 2.5-Induced Skin Keratinocyte Damage via MAPK Signaling Pathway. Mar. Drugs 2019, 17, 444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Ha, J.W.; Song, H.; Hong, S.S.; Boo, Y.C. Marine Alga Ecklonia Cava Extract and Dieckol Attenuate Prostaglandin E2 Production in HaCaT Keratinocytes Exposed to Airborne Particulate Matter. Antioxidants 2019, 8, 190. [Google Scholar] [CrossRef] [PubMed]
  48. Heo, S.J.; Ko, S.C.; Cha, S.H.; Kang, D.H.; Park, H.S.; Choi, Y.U.; Kim, D.; Jung, W.K.; Jeon, Y.J. Effect of Phlorotannins Isolated from Ecklonia Cava on Melanogenesis and Their Protective Effect against Photo-Oxidative Stress Induced by UV-B Radiation. Toxicol. Vitro 2009, 23, 1123–1130. [Google Scholar] [CrossRef]
  49. Ko, S.C.; Cha, S.H.; Heo, S.J.; Lee, S.H.; Kang, S.M.; Jeon, Y.J. Protective Effect of Ecklonia Cava on UVB-Induced Oxidative Stress: In Vitro and in Vivo Zebrafish Model. J. Appl. Phycol. 2011, 23, 697–708. [Google Scholar] [CrossRef]
  50. Le, Q.T.; Li, Y.; Qian, Z.J.; Kim, M.M.; Kim, S.K. Inhibitory Effects of Polyphenols Isolated from Marine Alga Ecklonia Cava on Histamine Release. Process Biochem. 2009, 44, 168–176. [Google Scholar] [CrossRef]
  51. Lee, J.W.; Seok, J.K.; Boo, Y.C. Ecklonia Cava Extract and Dieckol Attenuate Cellular Lipid Peroxidation in Keratinocytes Exposed to PM10. Evid.-Based Complement. Altern. Med. 2018, 2018, 8248323. [Google Scholar] [CrossRef] [Green Version]
  52. Zhang, C.; Li, Y.; Shi, X.; Kim, S.-K. Inhibition of the expression on MMP-2, 9 and morphological changes via human fibrosarcoma cell line by 6,6′-bieckol from marine alga Ecklonia cava. BMB Rep. 2010, 43, 62–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Yoon, N.Y.; Eom, T.K.; Kim, M.M.; Kim, S.K. Inhibitory Effect of Phlorotannins Isolated from Ecklonia Cava on Mushroom Tyrosinase Activity and Melanin Formation in Mouse B16F10 Melanoma Cells. J. Agric. Food Chem. 2009, 57, 4124–4129. [Google Scholar] [CrossRef] [PubMed]
  54. O’Sullivan, A.M.; O’Callaghan, Y.C.; O’Grady, M.N.; Queguineur, B.; Hanniffy, D.; Troy, D.J.; Kerry, J.P.; O’Brien, N.M. In Vitro and Cellular Antioxidant Activities of Seaweed Extracts Prepared from Five Brown Seaweeds Harvested in Spring from the West Coast of Ireland. Food Chem. 2011, 126, 1064–1070. [Google Scholar] [CrossRef]
  55. Quéguineur, B.; Goya, L.; Ramos, S.; Martín, M.A.; Mateos, R.; Guiry, M.D.; Bravo, L. Effect of Phlorotannin-Rich Extracts of Ascophyllum Nodosum and Himanthalia Elongata (Phaeophyceae) on Cellular Oxidative Markers in Human HepG2 Cells. J. Appl. Phycol. 2013, 25, 1–11. [Google Scholar] [CrossRef] [Green Version]
  56. Zhen, A.X.; Piao, M.J.; Hyun, Y.J.; Kang, K.A.; Fernando, P.D.S.M.; Cho, S.J.; Ahn, M.J.; Hyun, J.W. Diphlorethohydroxycarmalol Attenuates Fine Particulate Matter-Induced Subcellular Skin Dysfunction. Mar. Drugs 2019, 17, 95. [Google Scholar] [CrossRef] [Green Version]
  57. Kang, J., II; Kim, S.C.; Kim, M.K.; Boo, H.J.; Jeon, Y.J.; Koh, Y.S.; Yoo, E.S.; Kang, S.M.; Kang, H.K. Effect of Dieckol, a Component of Ecklonia Cava, on the Promotion of Hair Growth. Int. J. Mol. Sci. 2012, 13, 6407–6423. [Google Scholar] [CrossRef]
  58. Nagayama, K.; Shibata, T.; Fujimoto, K.; Honjo, T.; Nakamura, T. Algicidal Effect of Phlorotannins from the Brown Alga Ecklonia Kurome on Red Tide Microalgae. Aquaculture 2003, 218, 601–611. [Google Scholar] [CrossRef]
  59. Kang, M.C.; Cha, S.H.; Wijesinghe, W.A.J.P.; Kang, S.M.; Lee, S.H.; Kim, E.A.; Song, C.B.; Jeon, Y.J. Protective Effect of Marine Algae Phlorotannins against AAPH-Induced Oxidative Stress in Zebrafish Embryo. Food Chem. 2013, 138, 950–955. [Google Scholar] [CrossRef]
  60. Choi, H.S.; Jeon, H.J.; Lee, O.H.; Lee, B.Y. Dieckol, a Major Phlorotannin in Ecklonia Cava, Suppresses Lipid Accumulation in the Adipocytes of High-Fat Diet-Fed Zebrafish and Mice: Inhibition of Early Adipogenesis via Cell-Cycle Arrest and AMPKα Activation. Mol. Nutr. Food Res. 2015, 59, 1458–1471. [Google Scholar] [CrossRef]
  61. Hwang, H.; Terada, M.; Shin, H.-C. Single Dose Oral Toxicity and 4-Weeks Repeated Oral Toxicity Studies of Ecklonia Cava Extract. Seikatsu Eisei 2008, 52, 282–289. [Google Scholar]
  62. Zaragozá, M.C.; López, D.; Sáiz, M.P.; Poquet, M.; Pérez, J.; Puig-Parellada, P.; Màrmol, F.; Simonetti, P.; Gardana, C.; Lerat, Y.; et al. Toxicity and Antioxidant Activity in Vitro and in Vivo of Two Fucus Vesiculosus Extracts. J. Agric. Food Chem. 2008, 56, 7773–7780. [Google Scholar] [CrossRef]
  63. Yang, H.; Yoon, M.; Kim, J.; Cho, S. Acute Oral Toxicity of Phlorotannins in Beagle Dogs. Kor. J. Fish Aquat. Sci. 2014, 47, 356–362. [Google Scholar]
  64. Shin, H.C.; Kim, S.H.; Park, Y.; Lee, B.H.; Hwang, H.J. Effects of 12-Week Oral Supplementation of Ecklonia Cava Polyphenols on Anthropometric and Blood Lipid Parameters in Overweight Korean Individuals: A Double-Blind Randomized Clinical Trial. Phytother. Res. 2012, 26, 363–368. [Google Scholar] [CrossRef] [PubMed]
  65. Baldrick, F.R.; McFadden, K.; Ibars, M.; Sung, C.; Moffatt, T.; Megarry, K.; Thomas, K.; Mitchell, P.; Wallace, J.M.W.; Pourshahidi, L.K.; et al. Impact of a (Poly)Phenol-Rich Extract from the Brown Algae Ascophyllum Nodosum on DNA Damage and Antioxidant Activity in an Overweight or Obese Population: A Randomized Controlled Trial. Am. J. Clin. Nutr. 2018, 108, 688–700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Paradis, M.E.; Couture, P.; Lamarche, B. A Randomised Crossover Placebo-Controlled Trial Investigating the Effect of Brown Seaweed (Ascophyllum Nodosum and Fucus Vesiculosus) on Postchallenge Plasma Glucose and Insulin Levels in Men and Women. Appl. Physiol. Nutr. Metab. 2011, 36, 913–919. [Google Scholar] [CrossRef] [PubMed]
  67. Um, M.Y.; Kim, J.Y.; Han, J.K.; Kim, J.; Yang, H.; Yoon, M.; Kim, J.; Kang, S.W.; Cho, S. Phlorotannin Supplement Decreases Wake after Sleep Onset in Adults with Self-Reported Sleep Disturbance: A Randomized, Controlled, Double-Blind Clinical and Polysomnographic Study. Phytother. Res. 2018, 32, 698–704. [Google Scholar] [CrossRef]
  68. Turck, D.; Bresson, J.; Burlingame, B.; Dean, T.; Fairweather-Tait, S.; Heinonen, M.; Hirsch-Ernst, K.I.; Mangelsdorf, I.; McArdle, H.J.; Naska, A.; et al. Safety of Ecklonia Cava Phlorotannins as a Novel Food Pursuant to Regulation (EC) No 258/97. EFSA J. 2017, 15, e05003. [Google Scholar]
  69. Catarino, M.D.; Silva, A.M.S.; Mateus, N.; Cardoso, S.M. Optimization of Phlorotannins Extraction from Fucus Vesiculosus and Evaluation of Their Potential to Prevent Metabolic Disorders. Mar. Drugs 2019, 17, 162. [Google Scholar] [CrossRef] [Green Version]
  70. U.S. Food & Drug Administration (FDA). Recent Updates to the Notifications for New Dietary Ingredients. 2022. Available online: https://www.fda.gov/food/new-dietary-ingredients-ndi-notification-process/submitted-75-day-premarket-notifications-new-dietary-ingredients (accessed on 21 October 2022).
  71. Ministry of Food and Drug Safety (MFDS). Functional Ingredients for Health Functional Foods That Help Improve Sleep Quality. 2022. Available online: https://www.foodsafetykorea.go.kr/portal/healthyfoodlife/searchHomeHFDetail.do?prdlstReportLedgNo=2015021000031576 (accessed on 21 October 2022).
  72. Cho, S.; Han, D.; Kim, S.B.; Yoon, M.; Yang, H.; Jin, Y.H.; Jo, J.; Yong, H.; Lee, S.H.; Jeon, Y.J.; et al. Depressive Effects on the Central Nervous System and Underlying Mechanism of the Enzymatic Extract and Its Phlorotannin-Rich Fraction from Ecklonia Cava Edible Brown Seaweed. Biosci. Biotechnol. Biochem. 2012, 76, 163–168. [Google Scholar] [CrossRef]
  73. Cho, S.; Yang, H.; Yoon, M.; Kim, J.; Kim, D.; Kim, J.; Kim, S.B. Arousal Inhibitory Effect of Phlorotannins on Caffeine in Pentobarbital-Induced Mice. Fish Aquatic. Sci. 2014, 17, 13–18. [Google Scholar] [CrossRef] [Green Version]
  74. Askari, V.R.; Rahimi, V.B.; Ghorbani, A.; Rakhshandeh, H. Hypnotic Effect of Ocimum Basilicum on Pentobarbital-Induced Sleep in Mice. Iran Red. Crescent Med. J. 2016, 18, e24261. [Google Scholar] [CrossRef] [Green Version]
  75. Rakhshandeh, H.; Heidari, A.; Pourbagher-Shahri, A.M.; Rashidi, R.; Forouzanfar, F. Hypnotic Effect of A. Absinthium Hydroalcoholic Extract in Pentobarbital-Treated Mice. Neurol. Res. Int. 2021, 2021, 5521019. [Google Scholar] [CrossRef]
  76. Rahimi, V.B.; Askari, V.R.; Tajani, A.S.; Hosseini, A.; Rakhshandeh, H. Evaluation of the Sleep-Prolonging Effect of Lagenaria Vulgaris and Cucurbita Pepo Extracts on Pentobarbital-Induced Sleep and Possible Mechanisms of Action. Medicina 2018, 54, 55. [Google Scholar] [CrossRef] [Green Version]
  77. Cho, S.; Han, D.; Kim, J.; Yoon, M.; Yang, H.; Kim, J. Potential claims and evaluation methods for sleep-promoting effects of foods. Food Sci. Ind. 2013, 46, 8–22. [Google Scholar]
  78. Yoon, M.; Kim, J.S.; Jo, J.; Han, D.; Cho, S. Sleep-Promoting Effect of Ecklonia Cava: Ethanol Extract Promotes Non-Rapid Eye Movement Sleep in C57BL/6N Mice. Fish Aquatic. Sci. 2014, 17, 19–25. [Google Scholar] [CrossRef] [Green Version]
  79. Masaki, M.; Aritake, K.; Tanaka, H.; Shoyama, Y.; Huang, Z.L.; Urade, Y. Crocin Promotes Non-Rapid Eye Movement Sleep in Mice. Mol. Nutr. Food Res. 2012, 56, 304–308. [Google Scholar] [CrossRef]
  80. Huang, Z.L.; Qu, W.M.; Eguchi, N.; Chen, J.F.; Schwarzschild, M.A.; Fredholm, B.B.; Urade, Y.; Hayaishi, O. Adenosine A2A, but Not A1, Receptors Mediate the Arousal Effect of Caffeine. Nat. Neurosci. 2005, 8, 858–859. [Google Scholar] [CrossRef]
  81. Revel, F.G.; Gottowik, J.; Gatti, S.; Wettstein, J.G.; Moreau, J.L. Rodent Models of Insomnia: A Review of Experimental Procedures That Induce Sleep Disturbances. Neurosci. Biobehav. Rev. 2009, 33, 874–899. [Google Scholar] [CrossRef]
  82. Fernando, I.P.S.; Lee, W.W.; Ahn, G. Marine Algal Flavonoids and Phlorotannins; an Intriguing Frontier of Biofunctional Secondary Metabolites. Crit. Rev. Biotechnol. 2022, 42, 23–45. [Google Scholar] [CrossRef]
  83. Yoon, M.; Cho, S. Triphlorethol A, a Dietary Polyphenol from Seaweed, Decreases Sleep Latency and Increases Non-Rapid Eye Movement Sleep in Mice. Mar. Drugs 2018, 16, 139. [Google Scholar] [CrossRef]
  84. Lee, S.M.; Jeong, H.H.; Lee, J.C.; Park, M.Y.; Kim, S.C. A Clinical Case Study on the Effects of Acupuncture Therapy and Ecklonia Cava Extract on Sleep Disturbances in ALS Patients. J. Acupunct. Res. 2013, 30, 247–252. [Google Scholar] [CrossRef]
  85. Borja, N.L.; Daniel, K.L. Ramelteon for the treatment of insomnia. Clin. Ther. 2006, 28, 1540–1555. [Google Scholar] [CrossRef]
  86. Ebert, B.; Wafford, K.A.; Deacon, S. Treating Insomnia: Current and Investigational Pharmacological Approaches. Pharmacol. Ther. 2006, 112, 612–629. [Google Scholar] [CrossRef]
  87. Trevor, A.J.; Way, W.L. Sedative-Hypnotic Drugs. In Basic and Clinical Pharmacology, 12th ed.; Katzung, B.G., Ed.; McGraw-Hill Medical: New York, NY, USA, 2007; pp. 373–388. ISBN 978-0-07-176402-5. [Google Scholar]
  88. Erman, M.K. Therapeutic options in the treatment of insomnia. J. Clin. Psychiatr. 2005, 66, 18–23. [Google Scholar]
  89. Brogden, R.N.; Goa, K.L. Flumazenil. A Reappraisal of Its Pharmacological Properties and Therapeutic Efficacy as a Benzodiazepine Antagonist. Drugs 1991, 42, 1061–1089. [Google Scholar] [CrossRef]
Figure 1. Schematic illustrations of (a) pentobarbital-induced sleep test and (b) polygraphic recordings. Abbreviations: p.o., post-oral injection; i.p., intraperitoneal injection; EEG, electroencephalogram; EMG, electromyogram; FFT, fast Fourier transform.
Figure 1. Schematic illustrations of (a) pentobarbital-induced sleep test and (b) polygraphic recordings. Abbreviations: p.o., post-oral injection; i.p., intraperitoneal injection; EEG, electroencephalogram; EMG, electromyogram; FFT, fast Fourier transform.
Marinedrugs 20 00774 g001
Figure 2. Molecular structure and molecular weight of the individual constituents of phlorotannins.
Figure 2. Molecular structure and molecular weight of the individual constituents of phlorotannins.
Marinedrugs 20 00774 g002
Figure 3. Sleep-inducing mechanism of phlorotannins.
Figure 3. Sleep-inducing mechanism of phlorotannins.
Marinedrugs 20 00774 g003
Table 1. In vivo evaluation methods for assessing hypnotic effects.
Table 1. In vivo evaluation methods for assessing hypnotic effects.
MethodsPentobarbital-Induced Sleep TestPolygraphic Recordings
AnimalICR mice or SD ratsC57BL/6N mice or SD rats
MeasurementsRighting reflexEEG and EMG
Evaluation markersSleep latency, sleep duration, and sleep onsetSleep latency, amount of NREMS and REMS, delta activity, sleep–wake episodes
AdvantagesShort assay time, possible to screen many samplesAssessment of both sleep quantity and quality
DisadvantagesImpossible to evaluate sleep qualityLong assay time, high cost
Abbreviations: ICR, imprinting control region; SD, Sprague–Dawley; EEG, electroencephalogram; EMG, electromyogram; NREMS, non-rapid eye movement sleep; REMS, rapid eye movement sleep.
Table 2. Results from the pentobarbital-induced sleep test and polygraphic recordings studies on individual constituents of phlorotannins.
Table 2. Results from the pentobarbital-induced sleep test and polygraphic recordings studies on individual constituents of phlorotannins.
CompoundMethods (Dose) and Activities
EckolPentobarbital-induced sleep test (50 mg/kg) duration ↑ [4]
EckstolonolPentobarbital-induced sleep test (50 mg/kg) duration ↑ [4]
Polygraphic recordings (50 mg/kg) NREMS ↑, latency ↓ Delta activity − [4]
DieckolPentobarbital-induced sleep test (50 mg/kg) duration ↑ [4]
Polygraphic recordings (150 mg/kg) NREMS ↑, latency ↓ Delta activity − [19]
Triphlorethol APentobarbital-induced sleep test (50 mg/kg) duration ↑ [4]
Polygraphic recordings (50 mg/kg) NREMS ↑, latency ↓ Delta activity − [83]
Fucodiphlorethol GPentobarbital-induced sleep test (50 mg/kg) duration ↑ [4]
6,6′-BieckolPentobarbital-induced sleep test (50 mg/kg) duration ↑ [4]
NREMS, non-rapid eye movement sleep; −, not significant; ↑, increase; ↓, decrease.
Table 3. Results from in vitro GABAergic mechanism studies on preparations and individual constituents of phlorotannins.
Table 3. Results from in vitro GABAergic mechanism studies on preparations and individual constituents of phlorotannins.
SamplesBinding Affinity to the BZD Binding Site (IC50)Functional Assay for the GABAA Receptors
Preparations from Ecklonia cava
 Enzymatic extract1.409 mg/mL [72]-
 Methanol extract0.392 mg/mL [17]-
 Ethanol extract (EE)0.127 mg/mL [17]-
 Ethyl acetate fraction from EE0.019 mg/mL [17]-
 Butanol fraction from EE 0.103 mg/mL [17]-
 Hexane fraction from EE0.141 mg/mL [17]-
 Purified phlorotannin supplement0.012 mg/mL [4]Positive allosteric activation to the GABAA receptors [18]
Individual phlorotannin compounds
 Eckstolonol2.422 μM [17]Positive allosteric activation to the GABAA receptors [4]
 Eckol1.739 μM [17]-
 Triphlorethol-A7.180 μM [17]-
 Dieckol4.991 μM [17]Positive allosteric activation to the GABAA receptors [18]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kim, S.; Kim, D.; Um, M.Y.; Yoon, M.; Choi, J.-S.; Choi, Y.H.; Cho, S. Marine Polyphenol Phlorotannins as a Natural Sleep Aid for Treatment of Insomnia: A Review of Sedative–Hypnotic Effects and Mechanism of Action. Mar. Drugs 2022, 20, 774. https://doi.org/10.3390/md20120774

AMA Style

Kim S, Kim D, Um MY, Yoon M, Choi J-S, Choi YH, Cho S. Marine Polyphenol Phlorotannins as a Natural Sleep Aid for Treatment of Insomnia: A Review of Sedative–Hypnotic Effects and Mechanism of Action. Marine Drugs. 2022; 20(12):774. https://doi.org/10.3390/md20120774

Chicago/Turabian Style

Kim, Seonghui, Duhyeon Kim, Min Young Um, Minseok Yoon, Jae-Suk Choi, Yung Hyun Choi, and Suengmok Cho. 2022. "Marine Polyphenol Phlorotannins as a Natural Sleep Aid for Treatment of Insomnia: A Review of Sedative–Hypnotic Effects and Mechanism of Action" Marine Drugs 20, no. 12: 774. https://doi.org/10.3390/md20120774

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop