Next Article in Journal
Staphylococcus aureus from Minas Artisanal Cheeses: Biocide Tolerance, Antibiotic Resistance and Enterotoxin Genes
Next Article in Special Issue
Physiological Effects of Red-Colored Food-Derived Bioactive Compounds on Cardiovascular and Metabolic Diseases
Previous Article in Journal
Efficacy of Two Toothpaste in Preventing Tooth Erosive Lesions Associated with Gastroesophageal Reflux Disease
Previous Article in Special Issue
Physiological Effects of Bioactive Compounds Derived from Whole Grains on Cardiovascular and Metabolic Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Potential of Seaweed-Derived Bioactive Compounds for Cardiovascular Disease Treatment

1
Korea Food Research Institute, 245 Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Korea
2
Department of Marine Life Science, Jeju National University, Jeju 63243, Korea
3
Department of Food Biotechnology, Korea University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea
4
Marine Science Institute, Jeju National University, Jeju 63333, Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Appl. Sci. 2022, 12(3), 1025; https://doi.org/10.3390/app12031025
Submission received: 29 December 2021 / Revised: 10 January 2022 / Accepted: 10 January 2022 / Published: 19 January 2022
(This article belongs to the Special Issue Bioactive Compounds for Cardiovascular and Metabolic Diseases)

Abstract

:
Cardiovascular diseases are closely related to hypertension, type 2 diabetes mellitus, obesity, and hyperlipidemia. Many studies have reported that an unhealthy diet and sedentary lifestyle are critical factors that enhance these diseases. Recently, many bioactive compounds isolated from marine seaweeds have been studied for their benefits in improving human health. In particular, several unique bioactive metabolites such as polyphenols, polysaccharides, peptides, carotene, and sterol are the most effective components responsible for these activities. This review summarizes the current in vitro, in vivo, and clinical studies related to the protective effects of bioactive compounds isolated from seaweeds against cardiovascular disorders, including anti-diabetic, anti-hypertensive, anti-hyperlipidemia, and anti-obesity effects. Therefore, this present review summarizes these concepts and provides a basis for further in-depth research.

1. Introduction

Cardiovascular disease (CVD) is known as the primary cause of death globally, and it is estimated that approximately 17.7 million people die from CVD, accounting for 31% of global deaths. In recent decades, the increasing prevalence of CVD has deteriorated children’s and adults’ physical and mental health, affecting their quality of life [1]. The main risk factors linked to CVD are hypertension (HTN), elevated blood low-density cholesterol, type 2 diabetes mellitus (T2DM), endothelial dysfunction, overweightness or obesity, high triglyceride levels, and dietary patterns [2].
Previous evidence has shown that hyperinsulinemia and obesity increase sympathetic nerve traffic, promoting salt reabsorption in the renal tubules and activating the renin–angiotensin system (RAS) [3,4]. Additionally, endothelial dysfunction and vascular oxidative stress were observed in the development of obesity. These reactions would amplify the reactive oxygen species (ROS), decrease the availability of nitric oxide (NO), and further affect the vascular tone [5,6]. The progression of the pathophysiological mechanisms of HTN and obesity-related T2DM are closely connected. Thus, it is necessary to find effective treatments to reduce risks and help decrease the epidemic levels of cardiovascular-related deaths.
Currently, multiple biological mechanisms underlying CVDs have been confirmed, providing evidence for the direct development of pharmacological tools and treatments. The most common medical treatments include calcium channel blockers, diuretics, and inhibitors of angiotensin-converting enzyme (ACE). ACE, which is a metalloproteinase, a strong vasoconstrictor implicated in the pathophysiology of hypertension, is important in regulating blood pressure by catalyzing the conversion of angiotensin I to angiotensin II [7,8]. As a result, the inhibition of ACE activity has become a prominent target for HTN management. Conversely, in vitro, the biochemical pathways between vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) are vital and are involved in regulating the vascular tone [9]. Therefore, we reviewed numerous in vitro and in vivo studies to summarize the newest therapeutic strategies.
The growing interest in natural products to promote human health has resulted in seaweeds becoming popular due to their high bioactive compound contents, especially those exhibiting effects related to cardiovascular protection and metabolism regulation. Because of their high nutritional value, seaweeds have been traditionally consumed as a healthy food in many Asian countries since ancient times [10]. Brown seaweeds (Ochrophyta), red seaweeds (Rhodophyta), and green seaweeds (Chlorophyta) are the three primary classifications or species of seaweeds [11,12]. Different types of brown seaweeds, such as Undaria, Laminaria, Gim, and Hiziki have been used in traditional Asian meals for a long time. They contain low levels of lipids and high levels of polysaccharides, fibers, and polyunsaturated fatty acids (PUFAs), which are considered high-quality nutritional components for food preparation. The consumption of seaweed in western countries is relatively low compared with Asian countries because of their food habits. However, over the last few decades, interest in seaweeds has gradually increased because of their various properties as food ingredients and the identification of their invaluable health effects [13]. Due to its chemical diversity and unique components, seaweed has been of interest in many studies and is widely used in the medical, nutraceutical, and cosmetic industries. The primary seaweed metabolites include proteins, polysaccharides, and lipids, whereas secondary metabolites contain phenolic compounds, halogenated compounds, sterols, terpenes, and small peptides among other bioactive compounds produced in seaweed tissues [12,14]. Seaweed not only contains various primary and secondary metabolites but can also be used as a latent material for bioactive oligopeptides and oligosaccharides via bioconversion processes such as fermentation and enzyme hydrolysis [15]. These bioactive substances derived from seaweeds have numerous therapeutic roles in metabolic disease prevention, with functional properties such as anti-oxidant, anti-bacterial, anti-cancer, anti-diabetic, anti-tumor, anti-inflammatory, and cardiovascular protection activities. Recent studies have reported evidence of their effects on human health, and mechanisms of biological activity have been reported [13]. Seaweeds are photosynthetic eukaryotes that possess simple reproductive structures with diverse forms and sizes. Seaweeds exhibit rapid adaptabilities to survive due to the complex ocean environment, such as changes in salinity, temperature variations, nutrient-deficient habitats, and UV irradiation, which are attributed to their bioactive secondary metabolites which cannot be found in other terrestrial organisms [16]. In addition to its bioactive properties, other important advantages are their easy cultivation, rapid growth, and availability to produce high value-added bioactive compounds by manipulating environmental conditions, as well as through gene modification and mutagenesis.
Based on these viewpoints, we reviewed the available data from animal studies and clinical trials regarding the pathophysiological mechanisms under bioactive compound treatment and the links between cardiovascular and metabolic disorders (Figure 1). Despite numerous efforts to improve the industrial use of bioactive seaweed compounds to prevent CVD, there have been many limitations. The nutraceutical industry demands more rigorous quality, standardization of components, and a clearer bioactivity mechanism than previously provided for seaweeds. To overcome this problem, it is necessary to secure information on the characteristics and diversity of seaweed’s bioactive substances and systematically understand their overall biological activities in the metabolic disorders closely related to CVD. We believe this review can provide useful insights and open up great potential for many research groups considering the huge interest regarding seaweed’s bioactive compounds in the prevention of CVD.

2. Seaweed-Derived Phlorotannins with Therapeutic Potential against CVD

Phlorotannins are polyphenol molecules generated as secondary metabolites by seaweed and consist of polymerization of phloroglucinol (1,3,5-tryhydroxybenzene) monomer units, which are biosynthesized via the acetate-malonate pathway, also known as the polyketide pathway [17]. In particular, some phlorotannins can be found exclusively in brown seaweeds, such as Ecklonia species [18]. Phlorotannins can be classified into four subclasses according to the type of linkage between the phloroglucinol units. Fuhalols and phlorethols, as well as phloroglucinol units, are linked by aryl ether bonds. Fucols are composed of phloroglucinol units linked with aryl-aryl bonds. Fucophloroethols are linked to ether and phenyl bonds. Eckols and carmalols are characterized by the presence of dibenzodioxin units and differ from carmalol in that they generally have a lower molecular weight and a phenoxyl moiety at C4 [19,20]. Many researchers have revealed various beneficial biological activities of phlorotannins (mainly brown seaweed such as Ecklonia and Ishige species), including anti-oxidant, anti-cancer, anti-bacterial, anti-allergic, anti-mutagenic, anti-diabetic, anti-inflammatiory, anti-proliferatiive, anti-hypertensive, and anti-obesity effects [18,21,22,23,24]. Therefore, phlorotannins have been recognized for many years as promising bioactive compounds with health benefits for preventing and treating various human diseases.
Hypertension is known as a major risk factor for cardiovascular disease (CVD). Normally, the anti-hypertensive ability is determined as the IC50 value, which represents the angiotensin-converting enzyme (ACE) inhibitor concentration causing 50% inhibition of ACE activity. Captopril, a well-known ACE inhibitor, exhibits competitive inhibition [25]. Additionally, several previous studies have confirmed that phlorotannins such as eckol [26,27], dieckol [26,27], 6,6′-bieckol [28], phloroglucinol [26,27], phlorofucofuroeckol A [27], triphlorethol-A [26,27], eckstolonol [26,27], fucosterol [27], and octaphlorethol A [29], isolated from Ecklonia cava, Ecklonia stolonifera, and Ishige foliacea, showed non-competitive ACE inhibition (Table 1). These phlorotannins demonstrated ACE inhibitory activity similar to or even higher than that of captopril.
In addition to ACE inhibition, vasodilators also contribute greatly to the anti-hypertensive properties. NO is a well-known vessel-relaxing factor produced from L-arginine by endothelial nitric oxide synthase (eNOS) in the presence of oxygen and the cofactors Ca2+ and calmodulin (CaM) [30]. A previous study has indicated that genetically deficient eNOS mice are hypertensive with lower circulating NO levels, thus indicating the critical role of eNOS and NO in CVD [5,9]. It has also been reported that multiple mechanisms control NO production via eNOS activation [31].
The phosphatidylinositol 3-kinase (PI3K) pathway members, including their downstream molecule, protein kinase B (Akt), are essential regulators. Activated Akt would directly phosphorylate ser1177 on eNOS, enhancing the [Ca2+]/CaM complex [32]. Second, the concentration of intracellular Ca2+ ([Ca2+]i) in ECs and VSMCs is closely related to the vascular tone and influences blood pressure [33]. Third, the L-type calcium channel is one of the critical ion channels that regulates vasoconstriction and vasodilation. Nitrodilators promote vasodilation by increasing soluble guanylyl cyclase (cGMP) and decreasing the [Ca2+]i levels in VSMCs. These actions would reduce the phosphorylation of the Ca2+-sensitive myosin light chains, resulting in vasodilation [34]. One study demonstrated that Lu et al. used the EA.hy926 cells and zebrafish model to systematically establish the potential mechanisms of the vasodilation produced by diekol and diphlorethohydroxycarmalol isolated from Ecklonia cava (E. cava) and Ishige okamurae (I. okamurae) [35,36] (Table 1). In addition, four major phlorotannins—dieckol, 2,7-phloroglucinol-6,6-bieckol, phlorofucofuroeckol A, and pyrogallol-phloroglucinol-6,6-bieckol—isolated from Ecklonia cava effectively inhibited monocyte-associated vascular inflammation and dysfunction by suppressing monocyte migration and protecting monocyte-associated endothelial cell death [37]. Moreover, pyrogallol-phloroglucinol-6,6′-bieckol and dieckol isolated from E. Cava have been shown to improve blood circulation in mice fed diets to induce obesity and hypertension [36,38]. Therefore, while numerous factors contribute to hypertension, it is well recognized that an increased vascular tone is always the ultimate goal.
The evidence mentioned above has great pharmaceutical potential, with some already being used in the clinical trial phase. Clinical studies have shown cardiovascular protection, supporting the benefits of polyphenols extracted from land plants [39]. Nevertheless, only a few epidemiological studies have discussed the association between the consumption of specific components of seaweed and blood pressure. Despite investigating the benefits of single compounds from seaweed, the general components, including minerals, fiber, and peptides, were more popular in clinical trials. This might be because high doses of these compounds have comparatively low side effects. A case control study demonstrated the anti-hypertensive effect of Undaria pinnatifida powder (5 g/capsule/day) by significantly reducing blood pressure in elderly Japanese patients following 8 weeks of administration [40]. However, Murray et al. reported that no reduction in blood pressure occurred in healthy adults who were administered the Fucus vesiculosus extract [41]. As mentioned above, several phlorotannins have been confirmed to possess effects against hypertension by ACE inhibition, calcium regulation, antagonism of L-type calcium channels, or activation of the critical pathway (PI3K/Akt/eNOS). Despite these mechanisms being verified, many other physiological signaling pathways in vascular regulation remain unclear. It will be helpful to explore the cardiovascular protection or anti-hypertension properties of phlorotannin via its use as a potassium channel opener and angiotensin receptor blocker.
Diabetes mellitus (DM) is a prime risk factor for dramatically increasing CVD, contributing to more than 3 million cardiovascular deaths worldwide each year [42]. Type 2 diabetes mellitus (T2DM) is a complex metabolic disorder involving insulin resistance, impaired insulin signaling and β-cell dysfunction, and abnormal glucose and lipid metabolism. Hyperglycemia is the most important criterion for all types of diabetes and is the cause of diabetic complications such as CVD. Therefore, it is important to prevent or delay the onset of diabetes by controlling blood glucose levels in diabetic patients, as hyperglycemia increases the risk of developing CVD long before clinical diabetes begins [43]. In humans, α-glucosidase and α-amylase in the small intestine play an important role in the digesting dietary carbohydrates into glucose. Therefore, reducing postprandial hyperglycemia by delaying the absorption of glucose in the body through the inhibition of α-amylase and α-glucosidase is an important approach for treating T2DM [44,45]. The enzyme inhibitors used for this action suppress the digestion of carbohydrates and consequently slow the postprandial plasma glucose rise, thereby delaying the rate of glucose absorption [46]. The inhibition of starch-digesting enzymes using synthetic drugs exhibiting antidiabetic effects with α-glucosidase inhibitory properties, such as acarbose, voglibose, miglitol, and emiglitate, is an important clinical strategy for controlling postprandial hyperglycemia. However, these synthetic drugs have been reported to effectively lower postprandial blood glucose levels but cause serious side effects, such as liver disorders [45,47]. The use of enzyme inhibitors derived from natural products (terrestrial plants or seaweeds) with a lower risk of the potential side effects caused by synthetic enzyme inhibitors is recommended. Therefore, to avoid or reduce the side effects caused by currently used synthetase inhibitors, the use of enzyme inhibitors (α-amylase and α-glucosidase) from natural products is considered the best alternative. It has been reported that minor phlorotannin derivatives such as eckol, 2-phloroeckol, 8,8′-bieckol, 6,8′-bieckol, and 2-O-(2,4,6-trihydroxyphenyl)-6,6′-bieckol isolated from E. cava showed α-glucosidase inhibitory activity, with an IC50 value ranging from 2.3 to 59.8 μM (Table 1). Among the minor phlorotannin derivatives, 8,8′-bieckol and 6,8′-bieckol exhibited the strongest α-glucosidase inhibitory activity. In addition, molecular docking studies revealed that both phlorotannins act as α-glucosidase inhibitors by competitive inhibition [48]. In addition, several researchers have found that phlorotannins such as fucodiphloroethol G, dieckol, 6,6′-bieckol, 7-phloroeckol, phlorofucofuroeckol A, and 2,7′-phloroglucinol-6,6′-bieckol isolated from Ecklonia cava have inhibitory activity against α-glucosidase and α-amylase [49,50,51]. Moon et al. revealed that phlorotannins, such as phloroglucinol, dioxinodehydroeckol, eckol, phlorofucofuroeckol-A, dieckol, and 7-phloroeckol isolated from Ecklonia stolinifera (E. stolinifera) have anti-diabetic effects by inhibiting α-glucosidase. In addition, it was confirmed that these phlorotannins effectively inhibit protein tyrosine phosphatase 1 B, an enzyme that plays an important role in the development of insulin resistance, thereby preventing a rapid increase in postprandial blood glucose levels [52]. In addition, diphlorethohydroxycarmalol (DPHC), ishophloroglucin A (IPA), and octaphlorethol A (OPA) were isolated from I. okamurae and Ishige foliacea (I. foliacea) from the Ishigeaceae family and showed α-glucosidase and α-amylase inhibitory activity [53,54]. In particular, oral administration of 100 mg/kg DPHC significantly suppressed the postprandial blood glucose levels in streptozotocin-induced diabetic mice [55]. According to Lee et al., molecular docking analysis revealed that OPA interacts with amino acid residues in the region close to the active site of α-glucosidase. Hence, OPA has the potential to be used as a non-competitive inhibitor with a high-affinity binding site for α-glucosidase [54].
Insulin resistance and impaired glucose metabolism are the most common factors promoting the development of type 2 diabetes mellitus. Type 2 diabetes mellitus is typically caused by two factors: impaired insulin production by pancreatic β cells and a failure of insulin-sensitive tissues to respond appropriately to insulin [56]. Signaling pathways involved in insulin secretion in β-cells under physiological conditions can be divided into a few stages. First, insulin release is primarily triggered by high glucose concentrations, mainly via glucose transporter 2 (GLUT2). When glucose catabolism is stimulated, intracellular ATP levels increase and close the cell membrane potassium channels, increasing intracellular Ca2+ concentrations and amplifying insulin secretion [57,58]. Furthermore, the AMP-activated protein kinase (AMPK) system simulates the effect of insulin on glucose transport in the muscle and glucose production in the liver. Because the AMPK system plays an important role in glucose homeostasis, it is a key target for discovering anti-diabetic agents [59]. Some evidence has demonstrated that 2,7″-phloroglucinol-6,6′-bieckol isolated from E. cava protects against high glucose-induced glucotoxicity and apoptotic cell death in INS-1 cells [60] (Table 1). In addition, in C57BL/KsJ-db/db mice, the oral administration of 20 mg/kg dieckol isolated from E. cava significantly reduced blood glucose levels, serum insulin levels, and body weight [61]. Yang et al. demonstrated that oral administration of ishophloroglucin A (IPA) and oxtaphlorethol A (OPA) isolated from I. okamurae and I. foliacea significantly ameliorated glucose intolerance and the fasting glucose levels in high-fat diet (HFD)-fed mice, thereby reducing fasting and 2 h blood glucose levels, as well as stimulated GLUT4 in HFD mouse muscle [62,63]. Furthermore, phlorotannins such as DPHC and IPA isolated from I. okamurae treatment exhibited an anti-angiogenic effect by interfering with the VEGFR-2 signaling pathway [64,65]. These phlorotannin compounds isolated from various seaweeds exhibit anti-diabetic efficacy through various mechanisms in vivo and in vitro, demonstrating the potential as natural agents that can replace synthetic anti-diabetic treatments. Despite the evidence suggesting that type 2 diabetes mellitus might be caused by more complex molecular pathways implicated in cell biology, the main concepts of treatments still mainly focus on the pathway we mentioned above, and clinical trials using single compounds isolated from seaweeds have not been reported.
Despite its relatively simple definition, obesity, defined as excess body fat, is a complex condition resulting from a chronic positive energy balance when the dietary energy intake exceeds energy expenditure. Excess energy is converted to triglycerides and stored in adipose tissue depots, expanding in size, producing weight gain, and increasing body fat. The adipose tissue includes several types of cells, such as mature adipocytes, pre-adipocytes, fibroblasts, endothelial cells, and immune cells [66]. Adipose tissue undergoes dynamic histological changes as obesity progresses, including adipocyte enlargement, enhanced angiogenesis, immune cell infiltration, and extracellular matrix overproduction [67]. In contrast, adipocytes undergo differentiation, and specific proteins, including sterol regulatory element-binding protein 1 (SREBP1c), CCAAT/enhancer-binding protein (C/EBPα), and peroxisome proliferator-activated receptor γ (PPARγ) are involved in all processes [68,69]. Several lines of evidence indicate that the phlorotannins contained in seaweed such as E. cava (eckol, dieckol, triphlorethol-A, 6,6′-bieckol, and phlorofucoeckol A), Ecklonia stolonifera (phloroglucinol, eckol, dieckol, dioxinodehydroeckol, and phlorofucofuroeckol A), Eisenia bicyclis (6,6′-bieckol, 6,8′-bieckol, 8,8′-bieckol, and dieckol, phlorofucofuroeckol A), and I. okamurae (diphlorethohydroxycarmalol) exhibit anti-obesity properties by inhibiting intracellular lipid accumulation, suppressing adipogenesis in 3T3-L1 cells via inhibiting the expression of PPARγ, C/EBPα, SREBP-1, and FABP4, and activating AMP-activated protein kinase (AMPK) and ACC phosphorylation [70,71,72,73,74,75,76,77] (Table 1). In addition to these mechanisms, some phlorotannins isolated from brown seaweed exhibited anti-obesity effects by reducing leptin resistance or inhibiting pancreatic lipase [78,79]. Additionally, oral administration of phlorotannins such as eckol and dieckol isolated from E. cava and Ecklonia stolonifera showed antihyperlipidemic effects by decreasing the total cholesterol levels, triglyceride levels, and low-density lipoprotein levels and increasing the high-density lipoprotein in the serum of high-fat diet mice [80,81].

3. Seaweed-Derived Polysaccharides with Therapeutic Potential against CVD

In terms of economic value, seaweed polysaccharides are the most important products derived from seaweeds. Several studies have demonstrated the pharmacological activities of polysaccharides obtained from seaweeds. They are the multi-component mixtures made up of one or more monosaccharides linked by one, three, or four glycosidic linkages distributed between and inside seaweed cells [89]. In recent years, the most famous polysaccharides such as ulvan (sulfated glucuronoxylorhamnans), fucoidan (l-fucose and sulfate ester groups), and carrageenan (sulfated galactans) extracted from green, brown, and red seaweeds, respectively, have been extensively investigated for their health-promoting effects [90,91,92,93].
Among the risk factors for cardiovascular disease, seaweed-derived polysaccharides have been the focus of anti-diabetic research, and studies using fucoidan are the most widely reported. Fucoidan derived from Sargassum wightii, Sargassum thumbergii, Sragassum honeri, Sargassum ringgoldianum, Sargassum, siliquastrum, Sargassum graminifolium, Sargassum kjellmanianum, Fucus vesiculosus, Ascophyllum nodosum, Saccharina longicruris, Cystoseira crinite, Ecklonia maxima, and Turbinaria conoides exhibit anti-diabetic activity through the inhibition of α-amylase and α-glucosidase alone or both enzymes [94,95,96,97,98,99,100,101,102] (Table 2). Interestingly, Kim et al. revealed that the α-amylase and α-glucosidase inhibition of fucoidan differed depending on the species from which the fucoidan was extracted and by the month and year of collection [98]. In addition, the oral administration of Sargassum fusiforme-derived fucoidan decreased the fasting blood glucose levels in high-fat diet/streptozotocin-induced diabetic mice model and reduced diabetes-related intestinal bacteria, which may potentially aid diabetes [103,104,105]. Many studies have demonstrated cardioprotection by polysaccharides. Sulfated polysaccharides from Padina tetrastromatica exerted cardioprotective effects by activating the PI3K/Akt/Nrf2 signaling pathway in in vitro and Sprague Dawley rat models [106]. Fucoidan is a well-known l-fucose-enriched sulfate. Low molecular weight fucoidan (MW = 7000 Da) produced from Laminaria japonica (L. japonica) could induce endothelium-dependent vasodilation via the eNOS expression upregulation in cerebral microvascular endothelial cells and effectively improve blood pressure and local blood flow in rats [107]. Cystoseira crinite-derived fucoidan exhibits ACE inhibitory activity in vitro and in vivo [108]. In addition, Li et al. suggested that fucoidan (100 mg/kg/day) from Undaria pinnatifida prevents vascular dysfunction through PI3K/Akt/eNOS-dependent mechanisms in a hypertensive rat model [109]. Furthermore, continuous feeding with kappa-carrageenan produced from Kappaphycus alvarezii or iota-carrageenan from Sarconema filiform to high-fat diet-fed rats attenuated the parameters defining cardiovascular and metabolic health such as body weight, abdominal and liver fat, systolic blood pressure, plasma total cholesterol concentrations, and the plasma activities of alanine transaminase and aspartate transaminase [110]. In green seaweeds, ulvan is one of the major polysaccharide structures isolated from Ulva pertusa which had anti-hyperlipidemic effects via the modulation of lipid levels and the mRNA expression of FXR, LXR, and PPARγ in an in vivo rat model [111]. However, the anti-hypertensive effect of ulvan is not clear. Thus, further detailed investigations are required.
These functional compounds from seaweed protect the muscle and platelets and stimulate blood vessel dilatation [112]. In human trials, one previous report demonstrated that daily fucoidan consumption from L. japonica could remarkably suppress thrombus formation and further protect cardiovascular health [113]. However, there have been limited studies on the anti-hypertensive effects in human subjects. Most of the related research is closely associated with cardioprotective effects, such as anticoagulant polysaccharides in in vitro and in vivo models [114,115,116]. Therefore, due to the bioactivity of polysaccharides, more detailed human trials are important for verifying and developing alternative therapies soon.

4. Seaweed-Derived Peptides with Therapeutic Potential against CVD

As the importance of marine organisms as sources of novel bioactive substances is growing, marine bioactive peptides have received much attention recently. Bioactive peptides are usually 2−20 amino acid residues [123]. Depending on the amino acid sequence, they may be involved in various biological functions such as antioxidant, anti-cancer, opioid agonists or antagonists, immunomodulatory, anti-thrombotic, anti-atherosclerotic, and antimicrobial activities, in addition to nutrient utilization [123]. A pepsin-hydrolyzed peptide (VECYGPNRPQF) from Chlorella vulgaris protein waste possessed potent antioxidant activity against various free radicals and exhibited gastrointestinal enzyme resistance. Still, no cytotoxicity was observed in human lung fibroblast cell lines (WI-38) [124]. The antitumor polypeptide Y2 was obtained from the trypsin digest of Spirulina platensis proteins [125]. Cian et al. found that enzymatic hydrolysates from a phycobili protein byproduct of Porphyra columbina exhibited immunosuppressive effects on rat splenocytes by enhancing IL-10 production and inhibiting the production of TNF-α and IFN-γ [126].
Several bioactive peptides have been identified from Undaria pinnatifida (U. pinnatifida) and Palmaria palmate (P. palmate), which exerted an anti-hypertensive effect in vitro by showing potent ACE inhibition activity and a significant reduction in blood pressure observed in an oral feeding rat model [127,128,129,130,131]. In particular, bioactive peptides derived from various seaweeds (brown seaweed 10, red seaweed 10, and green seaweed 10) have antihypertensive effects through ACE inhibition (Table 3). One mg/kg BW of Leu-Trp, Val-Tyr, Ile-Tyr, Phe-Tyr, and Ile-Tyr from U. pinnatifida decreased the systolic blood pressure in spontaneously hypertensive rats (SHRs) [127]. Furthermore, Suetsuna et al. reported that both single administration and repeated oral administration of synthetic dipeptides (Tyr-His, Lys-Tyr, Phe-Tyr, and Ile-Tyr) from U. pinnatifida significantly decreased the blood pressure in spontaneously hypertensive rats (SHRs) [129]. The α and β subunits of phycoerythrin (a major light-harvesting protein pigment of red seaweed) from P. palmate showed ACE inhibition activity [130]. Ile-Leu-Ala-Pro, Leu-Leu-Ala-Pro, and Met-Ala-Gly-Val-Asp-His-Ile purified from P. palmate inhibited DPP-Ⅳ, a novel biomarker of ischemic cardiovascular disease, and had insulinotropic potency [131,132]. Ile-Pro and Ala-Phe-Leu isolated from Chlorophyta U. rigida exhibited ACE inhibition activity. Ko et al. [25] demonstrated significant systolic blood pressure reduction in hypertensive rats following 10 mg/kg body weight of Val-Glu-Gly-Tyr administered orally. Gracilariopsis lemaneiformis inhibited ACE-I with IC50 values of 9.64. 23.94, and 474.36 μM for the peptides FQIN [M(O)] CILR, TGAPCR, and Gln-Val-Glu-Tyr, respectively [133,134]. The administration of 10 mg/kg of FQIN [M(O)] CILR and TGAPCR had antihypertensive effects in spontaneously hypertensive rats [133].

5. Seaweed-Derived Carotenoids and Other Components with Therapeutic Potential against CVD

All carotenoids, including fucoxanthin, carotene, lycopene, and siphonaxanthin, are bioactive substances from seaweeds. Green seaweed extracts rich in carotenoids exhibit significant antigenotoxic activities [147]. Fucoxanthin is a recognized secondary metabolite found in macroalgae, and its biological properties are well established [23]. Fucoxanthin from brown seaweed exerts prebiotic and anti-inflammatory activities in human intestinal epithelial cells [148]. Fucoxanthin upregulates the expression of UCP1 in white adipose tissue (WAT) in KK-Ay mice [149]. Fucoxanthin and siphonaxanthin inhibit angiogenesis by downregulating the FGF-2-mediated intracellular signals in vascular endothelial cells [150]. Siphonaxanthin has been reported to have anti-angiogenic and anti-inflammatory effects [151]. Siphonaxanthin induces apoptosis by decreasing Bcl-2 expression and activating caspase-3 in human leukemia (HL-60) cells [152].
In Ae’s study, fucoxanthin extracted from Undaria pinnatifida (U. pinnatifida) increased the serum HDL. It decreased the triglyceride levels in high-fat diet-fed rats at a 0.2% diet dose for 4 weeks [153] (Table 4). In addition, the α-amylase and α-glucosidase inhibitory activities of fucoxanthin were observed in Kawee-Ai’s study [154]. The administration of a 3-g/kg 9-cis β-carotene diet disrupted the increases in plasma cholesterol and LDL in LDL-receptor knockout mice [155]. Feeding 8% 9-Cis β-carotene feed for 23 days disrupted triglyceride elevation in 5-week-old female db/db mice [156]. Other carotenoids have also been shown to have anti-atherogenic effects. Zhuo et al. showed that siphonaxanthin extracted from Codium fragile disrupts the elevation of the serum total cholesterol, triglyceride, and HDL in KK-Ay mice [157]. In addition, (all-E)-lutein and (9-Z)-zeaxanthin displayed anti-diabetes mellitus potency via α-glucosidase inhibition [158].
Seaweeds have a variety of physiologically active ingredients in addition to phloro-tannins, polysaccharides, peptides, and carotenoids. It has been reported that seaweeds contain various sterol compounds [159]. Sterols are an important family of lipids present in most eukaryotic cells. They are categorized into the steroid group, which contains the same fused four-ring core structure and has different biological roles than hormones and signaling molecules. The search for natural bioactive sterols as safe alternatives from marine seaweed is important in the food industry. Fucosterol isolated from the marine seaweed Pelvetia siliquosa causes a significant elevation of free radical scavenging enzyme activities, such as superoxide dismutase (SOD), catalase, and glutathione peroxidase (GSH-px) [160]. Moreover, 3 g,28 ξ-dihydroxy-24-ethylcholesta-5,23 Z-dien and 24 ξ-hydroperoxy-24-vinylcholesterol isolated from the brown seaweed Sargassum carpophyllum showed cytotoxic activity against human promyelocytic leukemia cells [161].
Zhen and Su studied bioactive compounds isolated from the edible brown seaweed Sargassum fusiforme (S. fusiforme), and 24(S)-saringosterol showed the strongest LXRβ-mediated transactivity among the seven phytosterols isolated from S. fusiforme containing fucosterol, 24-hydroperoxy-24-vinyl-cholesterol, 29-hydroperoxy-stigmasta-5,24(28)-dien-3β-ol, 24-methylenecholesterol, 24-keto-cholesterol, and 5α,8α-epidioxyergosta-6,22-dien-3β-ol [162], while 18α-glycyrrhetinic acid and 18β-glycyrrhetinic acid were extracted from S. fusiforme-inhibited α-glucosidase. Other compounds, including unsaturated FAs C20:4 (Δ7,9,11,13), C17:3 (Δ8,11,14), neolignan, and trace amines also potently inhibited α-glucosidase [163]. Two plastoquinones (sargachromenol and sargaquinoic acid) isolated from the active n-hexane fraction of Sargassum serratifolium produced concentration-dependent inhibition against α-glucosidase [164].
These physiologically active dietary components derived from seaweeds have been actively studied at the in vitro and in vivo levels to inhibit or alleviate the symptoms of diabetes, obesity, hypertension, and hyperglycemia and hyperlipidemia, which are closely related to the pathogenesis of CVD. However, research on the clinical trial stage using the seaweeds and bioactive dietary substances is insufficient. Bioactive dietary components derived from food are known to affect epigenetic gene expression in humans through various mechanisms, such as alteration of the chromatin structure or non-coding RNA, activation of transcription factors by signaling cascades, and direct ligand binding to nuclear receptors. Studies on the influence of polyphenols, PUFAs, and vitamins, which are bioactive dietary components contained in terrestrial plant resources, on epigenetic gene expression in humans through epigenetic change in the genome, miRNA level change, IncRNA level change, and transcription factor activity change are continuously being conducted [165]. By clearly analyzing the mechanisms of interaction between these dietary components and human genome structure and gene activity, a better understanding of these relationships will help design dietary guidelines that can help maintain good health by providing more effective dietary recommendations for individuals. Therefore, it is considered necessary to study not only the various bioactive dietary substances contained in seaweeds but also their influence on epigenetic changes in parallel.

6. Conclusions

The active compounds extracted from seaweed have been widely used for a long time, owing to their many benefits for improving human health. Increasing evidence has indicated that increasing the consumption of seaweeds helps decrease the risks of CVD and other related diseases by modulating different biological signaling pathways in vivo and in vitro. Further detailed human research is necessary to verify the clinical relevance of bioactive compounds isolated from seaweeds. In summary, this review lists the bioactive compounds isolated from different seaweeds to ameliorate CVD. This review will help summarize these concepts and promote further in-depth research.

Author Contributions

Conceptualization, C.-H.C. and Y.-A.L.; writing—original draft preparation, C.-H.C., Y.-A.L. and M.-Y.K.; writing—review and editing, Y.-J.J. and S.-H.L.; visualization, C.-H.C. and M.-Y.K.; supervision, Y.-J.J. and S.-H.L.; project administration, S.-H.L.; funding acquisition, S.-H.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by a National Research Foundation of Korea (NRF) grant funded by the Korean government (Ministry of Science and ICT, MSIT) (No. NRF-2020R1A2C2012608).

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. Roth, G.A.; Johnson, C.; Abajobir, A.; Abd-Allah, F.; Abera, S.F.; Abyu, G.; Ahmed, M.; Aksut, B.; Alam, T.; Alam, K.; et al. Global, Regional, and National Burden of Cardiovascular Diseases for 10 Causes, 1990 to 2015. J. Am. Coll. Cardiol. 2017, 70, 1–25. [Google Scholar] [CrossRef] [PubMed]
  2. Pace, R.; Brazeau, A.-S.; Meltzer, S.; Rahme, E.; Dasgupta, K. Conjoint Associations of Gestational Diabetes and Hypertension with Diabetes, Hypertension, and Cardiovascular Disease in Parents: A Retrospective Cohort Study. Am. J. Epidemiol. 2017, 186, 1115–1124. [Google Scholar] [CrossRef] [PubMed]
  3. Seravalle, G.; Grassi, G. Obesity and hypertension. Pharmacol. Res. 2017, 122, 1–7. [Google Scholar] [CrossRef]
  4. Leggio, M.; Lombardi, M.; Caldarone, E.; Severi, P.; D’emidio, S.; Armeni, M.; Bravi, V.; Bendini, M.G.; Mazza, A. The relationship between obesity and hypertension: An updated comprehensive overview on vicious twins. Hypertens. Res. 2017, 40, 947–963. [Google Scholar] [CrossRef]
  5. Gheibi, S.; Jeddi, S.; Kashfi, K.; Ghasemi, A. Regulation of vascular tone homeostasis by NO and H2S: Implications in hypertension. Biochem. Pharmacol. 2018, 149, 42–59. [Google Scholar] [CrossRef] [PubMed]
  6. Gutierrez, J.; Alloubani, A.; Mari, M.; Alzaatreh, M. Cardiovascular disease risk factors: Hypertension, diabetes mellitus and obesity among Tabuk citizens in Saudi Arabia. Open Cardiovasc. Med. J. 2018, 12, 41–49. [Google Scholar] [CrossRef] [PubMed]
  7. Leenen, F.H.H.; Nwachuku, C.E.; Black, H.R.; Cushman, W.C.; Davis, B.R.; Simpson, L.M.; Alderman, M.H.; Atlas, S.A.; Basile, J.N.; Cuyjet, A.B.; et al. Chlinical events in high-risk hypertensive patients randomly assigned to calcium channel blocker versus angiotensin-converting enzyme inhibitor in the antihypertensive and lipid-lowering treatment to prevent heart attack trial. Hypertension 2006, 48, 374–384. [Google Scholar] [CrossRef]
  8. Haller, H. Effective management of hypertension with dihydropyridine calcium channel blocker-based combination therapy in patients at high cardiovascular risk. Int. J. Clin. Pract. 2008, 62, 781–790. [Google Scholar] [CrossRef] [Green Version]
  9. Lin, Q.; Zhao, L.; Jing, R.; Trexler, C.; Wang, H.; Li, Y.; Tang, H.; Huang, F.; Zhang, F.; Fang, X.; et al. Inositol 1,4,5-trisphosphate receptors in endothelial cells play an essential role in vasodilation and blood pressure regulation. J. Am. Heart Assoc. 2019, 8, e011704. [Google Scholar] [CrossRef]
  10. Dhargalkar, V.K.; Pereira, N. Seaweed: Promising plant of the millennium. Sci. Cult. 2005, 71, 60–66. [Google Scholar]
  11. O’Sullivan, L.; Murphy, B.; McLoughlin, P.; Duggan, P.; Lawlor, P.G.; Hughes, H.; Gardiner, G.E. Prebiotics from marine macroalgae for human and animal health applications. Mar. Drugs 2010, 8, 2038–2064. [Google Scholar] [CrossRef] [Green Version]
  12. Rengasamy, K.R.; Mahomoodally, M.F.; Aumeeruddy, M.Z.; Zengin, G.; Xiao, J.; Kim, D.H. Bioactive compounds in seaweeds: An overview of their biological properties and safety. Food Chem. Toxicol. 2020, 135, 111013. [Google Scholar] [CrossRef] [PubMed]
  13. Ganesan, A.R.; Tiwari, U.; Rajauria, G. Seaweed nutraceuticals and their therapeutic role in disease prevention. Food Sci. Hum. Wellness 2019, 8, 252–263. [Google Scholar] [CrossRef]
  14. Fernando, I.P.S.; Ryu, B.; Ahn, G.; Yeo, I.-K.; Jeon, Y.-J. Therapeutic potential of algal natural products against metabolic syndrome: A review of recent developments. Trends Food Sci. Technol. 2020, 97, 286–299. [Google Scholar] [CrossRef]
  15. Samarakoon, K.; Jeon, Y.-J. Bio-vunctionalities of proteins derived from marine algae—A review. Food Res. Int. 2012, 48, 948–960. [Google Scholar] [CrossRef]
  16. Plaza, M.; Cifuentes, A.; Ibáñez, E. In the search of new functional food ingredients from algae. Trends Food Sci. Technol. 2008, 19, 31–39. [Google Scholar] [CrossRef] [Green Version]
  17. Meslet-Cladière, L.; Delage, L.; Leroux, C.J.-J.; Goulitquer, S.; Leblanc, C.; Creis, E.; Gall, E.A.; Stiger-Pouvreau, V.; Czjzek, M.; Potin, P. Structure/function analysis of a type III polyketide synthase in the brown alga Ectocarpus Siliculosus reveals a biochemical pathway in phlorotannin monomer biosynthesis. Plant. Cell 2013, 25, 3089–3103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Wikesekara, 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]
  19. Singh, I.P.; Bharate, S.B. Phloroglucinol compounds of natural origin. Nat. Prod. Rep. 2005, 23, 558–591. [Google Scholar] [CrossRef] [PubMed]
  20. Arbenz, A.; Avérous, L. Chemical modification of tannins to elaborate aromatic biobased macromolecular architectures. Green Chem. 2015, 17, 2626–2646. [Google Scholar] [CrossRef] [Green Version]
  21. Meng, W.; Sun, T.M.H.; Garcia-Vaquero, M. Phlorotannins: A review of extraction methods, structural characteristics, bioactivities, bioavalability, and future trends. Algal Res. 2021, 60, 102484. [Google Scholar] [CrossRef]
  22. Shrestha, S.; Zhang, W.; Smid, S. Phlorotannins: A review on biosynthesis, chemistry and bioactivity. Food Biosci. 2021, 39, 100832. [Google Scholar] [CrossRef]
  23. Seca, A.M.L.; Pinto, D.C.G.A. Overview on the antihypertensive and anti-obesity effects of secondary metabolites from seaweeds. Mar. Drugs 2018, 16, 237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Wijesinghe, W.A.J.P.; Jeon, Y.-J. Exploiting biological activities of brown seaweed Ecklonia cava for potential industrial applications: A review. Int. J. Food Sci. Nutr. 2012, 63, 225–235. [Google Scholar] [CrossRef]
  25. Cushman, D.W.; Ondetti, M.A. History of the design of captopril and related inhibitors of angiotensin converting enzyme. Hypertension 1991, 17, 589–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Wijesinghe, W.A.J.P.; Ko, S.-C.; Jeon, Y.-J. Effect of phlorotannins isolated from Ecklonia cava on angiotensin I-convering enzyme (ACE) inhibitory activity. Nutr. Res. Pract. 2011, 5, 93–100. [Google Scholar] [CrossRef] [Green Version]
  27. Jung, H.A.; Hyun, S.K.; Kim, H.R.; Choi, J.S. Angiotensin-converting enzyme I inhibitory activity of phlorotannins from Ecklonia stolonifera. Fish. Sci. 2006, 72, 1292–1299. [Google Scholar] [CrossRef]
  28. Ko, S.-C.; Kang, M.C.; Kang, N.; Kim, H.-S.; Lee, S.-H.; Ahn, G.; Jung, W.-K.; Jeon, Y.-J. Effect of angiotensin I-convering enzyme (ACE) inhibition and nitric oxide (NO) production of 6,6’-bieckol, a marine algal polyphenol and its anti-hypertensive effect in spontaneously hypertensive rats. Process. Biochem. 2017, 58, 326–332. [Google Scholar] [CrossRef]
  29. Ko, S.-C.; Jung, W.-K.; Kang, S.-M.; Lee, S.-H.; Kang, M.C.; Heo, S.-J.; Kang, K.-H.; Kim, Y.-T.; Park, S.-J.; Jeong, Y.; et al. Angiotensin I-converting enzyme (ACE) inhibition and nitric oxide (NO)-mediated antihypertensive effect of oxtaphorethol A isolated from Ishige sinicola: In vitro molecular mechanism and in vivo SHR model. J. Funct. Foods 2015, 18, 289–299. [Google Scholar] [CrossRef]
  30. Szabo, C. Hydrogen sulfide, an enhancer of vascular nitric oxide signaling: Mechanisms and implications. Am. J. Physiol. Cell Physiol. 2017, 312, C3–C15. [Google Scholar] [CrossRef]
  31. Devika, N.T.; Ali, B.M.J. Analysing calcium dependent and independent regulation of eNOS in endothelium triggered by extracelluar signalling events. Mol. BioSystems 2013, 9, 2653–2664. [Google Scholar] [CrossRef]
  32. Dudzinski, D.M.; Michel, T. Life history of eNOS: Partners and pathways. Cardiovasc. Res. 2007, 75, 247–260. [Google Scholar] [CrossRef] [PubMed]
  33. Raouf, A.K. Modulators of the vascular endothelin receptor in blood pressure regulation and hypertension. Curr. Mol. Pharmacol. 2011, 4, 176–186. [Google Scholar]
  34. Nakamura, K.; Koga, Y.; Sakai, H.; Homma, K.; Ikebe, M. cGMP-dependent relaxation of smooth muscle is coupled with the change in the phosphorylatin of myosin phophatase. Circ. Res. 2007, 101, 712–722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Lu, Y.-A.; Je, J.-G.; Hwang, J.; Jeon, Y.-J.; Ryu, B. Ecklonia cava extract and its derivative dieckol promote vasodilation by modulating calcium signaling and PI3K/AKT/eNOS pathway in in vitro and in vivo model. Biomedicines 2021, 9, 438. [Google Scholar] [CrossRef] [PubMed]
  36. Lu, Y.A.; Jiang, Y.; Yang, H.-W.; Hwang, J.; Jeon, Y.-J.; Ryu, B. Diphlorethohydroxycarmalol isolated from Ishige okamurae exerts vasodilatory effects via calcium signaling and PI3K/Akt/eNOS pathway. Int. J. Mol. Sci. 2021, 22, 1610. [Google Scholar] [CrossRef]
  37. Oh, S.; Son, M.; Lee, H.S.; Kim, H.-S.; Jeon, Y.-J.; Byun, K. Protective effect of pyrogallol-phloroglucinol-6,6-bieckol from Ecklonia cava on monocyte-associated vascular dysfunction. Mar. Drugs 2018, 16, 441. [Google Scholar] [CrossRef] [Green Version]
  38. Son, M.; Oh, S.; Lee, H.S.; Ryu, B.; Jiang, Y.; Jang, J.T.; Jeon, Y.-J.; Byun, K. Pyrogallol-phloroglucinol-6,6’-bieckol from Ecklonia cava improved blood circulation in diet-induced obese and diet-induced hypertension mouse model. Mar. Drugs 2019, 17, 272. [Google Scholar] [CrossRef] [Green Version]
  39. Gross, M. Flavonoids and cardiovascular disease. Pharm. Biol. 2004, 42, 21–35. [Google Scholar] [CrossRef] [Green Version]
  40. Hata, Y.; Nakajima, K.; Uchida, J.-I.; Hidaka, H.; Nakano, T. Clinical effects of brown seaweed, Undaria pinnatifida (wakame), on blood pressure in hypertensive subjects. J. Clin. Biochem. Nutr. 2001, 30, 43–53. [Google Scholar] [CrossRef] [Green Version]
  41. Murray, M.; Dordevic, A.L.; Ryan, L.; Bonham, M.P. The impact of a single dose of a polyphenol-rich seaweed extract on postprandial glycaemic control in healthy adults: A randomised cross-over trial. Nutrients 2018, 10, 270. [Google Scholar] [CrossRef] [Green Version]
  42. Danaei, G.; Lawes, C.M.; Hoorn, S.V.; Murray, C.J.; Ezzati, M. Global and regional mortality from ischaemic heart disease and stroke attributable to higher-than-optimum blood glucose concentration: Comparative risk assessment. Lancet 2006, 368, 1651–1659. [Google Scholar] [CrossRef]
  43. Bahadoran, Z.; Mirmiran, P.; Azizi, F. Dietary polyphenols as potential nutraceuticals in management of diabetes: A review. J. Diabetes Metab. Disord. 2013, 12, 43. [Google Scholar] [CrossRef] [Green Version]
  44. Asgar, A. Anti-diabetic potential of phenolic compounds: A review. Int. J. Food Prop. 2013, 16, 91–103. [Google Scholar] [CrossRef]
  45. Ghani, U. Re-exploring promising α-glucosidase inhibitors for potential development into oral anti-diabetic drugs: Finding needle in the haystack. Eur. J. Med. Chem. 2015, 103, 133–162. [Google Scholar] [CrossRef]
  46. Hossain, U.; Das, A.K.; Ghosh, S.; Sil, P.C. An overview on the role of bioactive α-glucosidase inhibitors in ameliorating diabetic complications. Food Chem. Toxicol. 2020, 145, 111738. [Google Scholar] [CrossRef] [PubMed]
  47. Dash, R.P.; Babu, R.J.; Srinivas, N.R. Reappraisal and perspectives of clinical drug-drug interaction potential of α-glucosidase inhibitors such as acarbose, voglibose and miglitol in the treatment of type 2 diabetes mellitus. Xenobiotica 2018, 48, 89–108. [Google Scholar] [CrossRef]
  48. Park, S.R.; Kim, J.H.; Jang, H.D.; Yang, S.Y.; Kim, Y.H. Inhibitory activity of minor phlorotannins from Ecklonia cava on α-glucosidase. Food Chem. 2018, 257, 128–134. [Google Scholar] [CrossRef] [PubMed]
  49. Lee, S.-H.; Karadeniz, F.; Kim, M.M.; Kim, S.K. α-Glucosidase and α-amylase inhibitory activities of phloroglucinal derivatives from edible marine brown alga, Ecklonia cava. J. Sci. Food Agric. 2008, 89, 1552–1558. [Google Scholar] [CrossRef]
  50. Lee, H.-A.; Lee, J.-H.; Han, J.-S. A phlorotannin constituent of Ecklonia cava alleviates postprandial hyperglycemia in diabetic mice. Pharm. Biol. 2017, 55, 1149–1154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Lee, S.-H.; Park, M.-H.; Heo, S.-J.; Kang, S.-M.; Ko, S.-C.; Han, J.-S.; Jeon, Y.-J. Dieckol isolated from Ecklonia cava inhibits α-glucosidase and α-amylase in vitro and alleviates postprandial hyperglycemia in streptozotocin-induced diabetic mice. Food Chem. Toxicol. 2010, 48, 2633–2637. [Google Scholar] [CrossRef]
  52. Moon, H.E.; Islam, M.N.; Ahn, B.R.; Chowdhury, S.S.; Sohn, H.S.; Jung, H.A.; Choi, J.S. Protein tyrosine phosphatase 1B and α-glucosidase inhibitory phlorotannins from edible brown algae, Ecklonia stolonifera and Eisenia bicyclis. Biosci. Biotechnol. Biochem. 2011, 75, 1472–1480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Ryu, B.; Jiang, Y.; Kim, H.-S.; Hyun, J.-M.; Lim, S.-B.; Li, Y.; Jeon, Y.-J. Ishophloroglucin A, a novel phlorotannin for standardizing the anti-α-glucosidase activity of Ishige okamurae. Mar. Drugs 2018, 16, 436. [Google Scholar] [CrossRef] [Green Version]
  54. Lee, S.-H.; Kang, S.-M.; Ko, S.-C.; Moon, S.-H.; Jeon, B.-T.; Lee, D.H.; Jeon, Y.-J. Octaphlorethol A: A potent α-glucosidase inhibitor isolated from Ishige foliacea shows an anti-hyperglycemic effect in mice with streptozotocin-induced diabetes. Food Funct. 2014, 5, 2602–2608. [Google Scholar] [CrossRef] [PubMed]
  55. Heo, S.-J.; Hwang, J.-Y.; Choi, J.-I.; Han, J.-S.; Kim, H.-J.; Jeon, Y.-J. Diphlorethohydroxycarmalol isolated from Ishige okamurae, a brown algae, a potent α-glucosidase and α-amylase inhibitor, alleviates postprandial hyperglycemia in diabetic mice. Eur. J. Pharmacol. 2009, 615, 252–256. [Google Scholar] [CrossRef] [PubMed]
  56. Galicia-Garcia, U.; Benito-Vicente, A.; Jebari, S.; Larrea-Sebal, A.; Siddiqi, H.; Uribe, K.B.; Ostolaza, H.; Martín, C. Pathophysiology of type 2 diabetes mellitus. Int. J. Mol. Sci. 2020, 21, 6275. [Google Scholar] [CrossRef]
  57. Doland, B.B.; Rhodes, C.J.; Grimsby, J.S. The dynamic plasticity of insulin production in β-cells. Mol. Metab. 2017, 6, 958–973. [Google Scholar]
  58. Fu, Z.; Gilbert, E.R.; Liu, D. Regulation of insulin synthesis and secretion and pancreatic beta-cell dysfunction in diabetes. Curr. Diabetes Rev. 2013, 9, 25–53. [Google Scholar] [CrossRef]
  59. Viollet, B.; Mounier, R.; Leclerc, J.; Yazigi, A.; Foretz, M.; Andreelli, F. Targeting AMP-activated protein kinase as a novel therapeutic approach for the treatment of metabolic disorders. Diabetes Metab. 2007, 33, 395–402. [Google Scholar] [CrossRef]
  60. Lee, H.-A.; Lee, J.-H.; Han, J.-S. 2,7”-Phloroglucinol-6,6’-bieckol pretects INS-1 cells against high glucose-induced apoptosis. Biomed. Pharmacother. 2018, 103, 1473–1481. [Google Scholar] [CrossRef]
  61. Kang, M.-C.; Wijesinghe, W.; 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]
  62. Yang, H.-W.; Son, M.; Choi, J.; Oh, S.; Jeon, Y.-J.; Byun, K.; Ryu, B. Effect of ishophloroglucin A, a component of Ishige okamurae, on glucose homeostasis in the pancreas and muscle of high fat diet-fed mice. Mar. Drugs 2019, 17, 608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Lee, S.-H.; Ko, S.-C.; Kang, M.-C.; Lee, D.H.; Jeon, Y.-J. Octaphlorethol A, a marine algae product, exhibits antidiabetic effects in type 2 diabetic mice by activating AMP-activated protein kinase and upregulating the expression of glucose transporter 4. Food Chem. Toxicol. 2016, 91, 58–64. [Google Scholar] [CrossRef] [PubMed]
  64. Fernando, K.H.N.; Yang, H.-W.; Jiang, Y.; Jeon, Y.-J.; Ryu, B. Diphlorethohydroxycarmalol isolated from Ishige okamurae represses high glucose-induced angiogenesis in vitro and in vivo. Mar. Drugs 2018, 16, 375. [Google Scholar] [CrossRef] [Green Version]
  65. Fernando, K.H.N.; Yang, H.-W.; Jiang, Y.; Jeon, Y.-J.; Ryu, B. Ishige okamurae extract and its constituent ishophloroglucin A attenuated in vitro and in vivo high glucose-induced angiogenesis. Int. J. Mol. Sci. 2019, 20, 5542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Tanaka, M.; Itoh, M.; Ogawa, Y.; Suganami, T. Molecular mechanism of obesity-induced ‘metabolic’ tissue remodeling. J. Diabetes Investig. 2017, 9, 256–261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Sun, K.; Kusminski, C.M.; Scherer, P.E. Adipose tissue remodeling and obesity. J. Clin. Investig. 2011, 121, 2094–2101. [Google Scholar] [CrossRef] [Green Version]
  68. Kuri-harcuch, W.; Velez-delValle, C.; Vazquez-Sandoval, A.; Hernández-Mosqueira, C.; Fernandez-Sanchez, V. A cellular perspective of adipogenesis transcriptional regulation. J. Cell. Physiol. 2019, 234, 1111–1129. [Google Scholar] [CrossRef]
  69. Choi, K.-M.; Jeon, Y.S.; Kim, W.; Lee, A.; Kim, Y.-G.; Lee, J.H.; Kang, Y.E.; Jung, J.-C.; Lee, J.; Min, B.; et al. Xanthigen attenuates high-fat diet-induced obesity through down-regulation of PPARγ and activation of the AMPK pathway. Food Sci. Biotechnol. 2014, 23, 931–935. [Google Scholar] [CrossRef]
  70. Karadeniz, F.; Ahn, B.-N.; Kim, J.-A.; Seo, Y.; Jang, M.-S.; Nam, K.-H.; Kim, M.; Lee, S.-H.; Kong, C.-S. Phlorotannins suppress adipogensis in pre-adipocytes while enhancing osteoblastogenesis in pre-osteoblasts. Arch. Pharmacal Res. 2015, 38, 2172–2182. [Google Scholar] [CrossRef]
  71. Ko, S.-C.; Lee, M.; Lee, J.-H.; Lee, S.-H.; Lim, Y.; Jeon, Y.-J. Dieckol, a phlorotannin isolated from a brown seaweed, Ecklonia cava, inhibits adipogenesis through AMP-activated protein kinase (AMPK) activation in 3T3-L1 preadipocytes. Environ. Toxicol. Pharmacol. 2013, 36, 1253–1260. [Google Scholar] [CrossRef]
  72. Choi, H.-S.; Jeon, H.J.; Lee, O.H.; Lee, B.Y. Dieckol, a major phlorotannin in Ecklonia cava, suppresses lipid accumuation in the adipocytes of high-fat diet-fed zebrafish and mice: Inhibition of early adipogensis via cell-cycle arrest and AMPKα activation. Mol. Nutr. Food Res. 2015, 59, 1458–1471. [Google Scholar] [CrossRef] [PubMed]
  73. Jung, H.A.; Jung, H.J.; Jeong, H.Y.; Kwon, H.J.; Ali, M.Y.; Choi, J.S. Phlorotannins isolated from the edible brown alga Ecklonia stolonifera exert anti-adipogenic activity on 3T3-L1 adipocytes by downregulating C/EBPα and PPARγ. Fitoterapia 2014, 92, 260–269. [Google Scholar] [CrossRef]
  74. Kwon, T.-H.; Wu, Y.X.; Kim, J.S.; Woo, J.H.; Park, K.T.; Kwon, O.J.; Seo, H.-J.; Kim, T.; Park, N.-H. 6,6’-Bieckol inhibits adipocyte differentiation through downregulation of adipogenesis and lipogenesis in 3T3-L1 cells. J. Sci. Food Agric. 2014, 95, 1830–1837. [Google Scholar] [CrossRef] [PubMed]
  75. Park, M.H.; Jeon, Y.J.; Kim, H.J.; Han, J.S. Effect of diphlorethohydroxycarmalol isolated from Ishige okamurae on apoptosis in 3T3-L1 preadipocytes. Phytother. Res. 2013, 27, 931–936. [Google Scholar] [CrossRef] [PubMed]
  76. Jeon, Y.; Song, S.; Kim, H.; Cheon, Y.-P. Diphlorethohydroxycarmalol of Ishige okamurae and caffeine modified the expression of extracellular fibrillars during adipogenesis of mouse subcutaneous adipose derived stem cell. Dev. Reprod. 2013, 17, 275–287. [Google Scholar] [CrossRef] [Green Version]
  77. Kang, M.-C.; Ding, Y.; Kim, H.-S.; Jeon, Y.-J.; Lee, S.-H. Inhibition of adipogenesis by diphlorethohydroxycarmalol (DPHC) through AMPK activation in adipocytes. Mar. Drugs 2019, 17, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Eom, S.-H.; Lee, M.S.; Lee, E.W.; Kim, Y.M.; Kim, T.H. Pancreatic lipase inhibitory activity of phlorotannins isolated from Eisenia bicyclis. Phytother. Res. 2013, 27, 148–151. [Google Scholar] [CrossRef] [PubMed]
  79. Oh, S.; Son, M.; Choi, J.; Choi, C.H.; Park, K.Y.; Son, K.H. Phlorotannins from Ecklonia cava attenuates plamitate-induced endoplasmic reticulum stress and leptin resistance in hypothalamic neurons. Mar. Drugs 2019, 17, 570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Moon, H.E.; Ahn, B.R.; Jung, H.A.; Choi, J.S. Inhibitory activity of Ecklonia stolonifera and its isolated phlorotannins against Cu2+-induced low-density lipoprotein oxidation. Fish. Sci. 2012, 78, 927–934. [Google Scholar] [CrossRef]
  81. Yoon, N.Y.; Kim, H.R.; Chung, H.Y.; Choi, J.S. Anti-hyperlipidemic effect of an edible brown algae, Ecklonia stolonifera, and its constituents on polyxamer 407-induced hyperlipidemic and choloesterol-fed rats. Arch. Pharmacal Res. 2008, 31, 1564–1571. [Google Scholar] [CrossRef]
  82. Yeo, A.-R.; Lee, J.; Tae, I.H.; Park, S.-R.; Cho, Y.H.; Lee, B.H. Anti-hyperlipidemic effect of polyphenol extract (SeapolynolTM) and dieckol isolated from Ecklonia cava in in vivo and in vitro models. Prev. Nutr. Food Sci. 2012, 17, 1–7. [Google Scholar] [CrossRef]
  83. Shibata, T.; Yamaguchi, K.; Nagayama, K.; Kawaguchi, S.; Nakamura, T. Inhibitory activity of brown algal phlorotannins against glycosidase from the viscera of the turban shell Turbo cornutus. Eur. J. Phycol. 2002, 37, 493–500. [Google Scholar] [CrossRef] [Green Version]
  84. Heo, S.-J.; Hwang, J.-Y.; Choi, J.-I.; Lee, S.-H.; Park, P.-J.; Kang, D.-H.; Oh, C.; Kim, D.-W.; Han, J.-S.; Jeon, Y.-J.; et al. Protective effect of diphlorethohydroxycarmalol isolated from Ishige okamurae against high glucose-induced-oxidative stress in human umbilical vein endothelial cells. Food Chem. Toxicol. 2010, 48, 1448–1454. [Google Scholar] [CrossRef]
  85. Lee, S.-H.; Choi, J.-I.; Heo, S.-J.; Park, M.-H.; Park, P.-J.; Jeon, B.-T.; Se-Kwon Kim, S.-K.; Han, J.-S.; Jeon, Y.-J. Diphlorethohydroxycarmalol isolated from Pae (Ishige okamurae) protects high glucose0induced damage in RINm5F pancreatic β cells via its antioxidant effects. Food Sci. Biotechnol. 2012, 21, 239–246. [Google Scholar] [CrossRef]
  86. Ding, Y.; Wang, L.; Im, S.; Hwang, O.; Kim, H.-S.; Kang, M.-C.; Lee, S.-H. Anti-obesity effect of diphlorethohydroxycarmalol isolated from brown alga Ishige okamurae in high-fat diet-induced obese mice. Mar. Drugs 2019, 17, 637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Lee, S.-H.; Kang, N.; Kim, E.-A.; Heo, S.-J.; Moon, S.-H.; Jeon, B.-T.; Jeon, Y.-J. Antidiabetogenic and antioxidant effect of octaphlorethol A isolated from the brown algae Ishige foliacea in streptozotocin-induced diabetic mice. Food Sci. Biotechnol. 2014, 23, 1261–1266. [Google Scholar] [CrossRef]
  88. Lee, S.-H.; Kang, S.-M.; Ko, S.-C.; Kang, M.-C.; Jeon, Y.-J. Octaphlorethol A, a novel phenolic compound isolated from Ishige foliacea, protects against streptozotocin-induced pancreatic β cell damage by reducing oxidative stress and apoptosis. Food Chem. Toxicol. 2013, 59, 643–649. [Google Scholar] [CrossRef] [PubMed]
  89. Holdt, S.L.; Kraan, S. Bioactive compounds inseaweed: Functional food applications and legislation. J. Appl. Phycol. 2011, 23, 543–597. [Google Scholar] [CrossRef]
  90. Mo’o, F.R.C.; Wilar, G.; Devkota, H.P.; Wathoni, N. Ulvan, a polysaccharide from Macroalga Ulva sp.: A review of chemistry, biological activities and potential for food and biomedical applications. Appl. Sci. 2020, 10, 5488. [Google Scholar] [CrossRef]
  91. Shen, P.; Yin, Z.; Qu, G.; Wang, C. Fucoidan and its health benefits. Bioact. Seaweeds Food Appl. Nat. Ingred. Healthy Diets 2018, 223–238. [Google Scholar]
  92. Wijesekara, I.; Pangestuti, R.; Kim, S.-K. Biological activities and potential health benefits of sulfated polysaccharides derived from marine algae. Carbohydr. Polym. 2011, 84, 14–21. [Google Scholar] [CrossRef]
  93. Pangestuti, R.; Kim, S.-K. Chapter Seven-Biological Activities of Carrageenan. Adv. Food Nutr. Res. 2014, 72, 113–124. [Google Scholar] [PubMed]
  94. Maneesh, A.; Chakraborty, K. Pharmacological potential of sulfated polygalactopyranosyl-fucopyranan from the brown seaweed Sragassum wightii. J. Appl. Phycol. 2018, 30, 1971–1988. [Google Scholar] [CrossRef]
  95. Senthil, S.L.; Kumar, T.V.; Geetharmani, D.; Suja, G.; Yesudas, R.; Chacko, A. Fucoidan-An α-amylase inhibitor from Sargassum wightii with relevance to NIDDM. Int. J. Biol. Marcromol. 2015, 81, 644–647. [Google Scholar] [CrossRef] [PubMed]
  96. Kumar, T.V.; Lakshmanasenthil, S.; Geetharamani, D.; Marudhupandi, T.; Suja, G.; Suganya, P. Fucoidan-A α-D-glucosidase inhibitor from Sargassum wightii with relevance to type 2 diabetes mellitus therapy. Int. J. Biol. Marcromol. 2015, 72, 1044–1047. [Google Scholar] [CrossRef] [PubMed]
  97. Kim, K.-T.; Rioux, L.-E.; Turgeon, S.L. Molecular weight and sulfate content modulate the inhibition of α-amylase by fucoidan relevant for type 2 diabetes management. PharmaNutrition 2015, 3, 108–114. [Google Scholar] [CrossRef]
  98. Kim, K.-T.; Rioux, L.-E.; Turgeon, S.L. Alpha-amylase and alpha-glucosidase inhibition is differentially modulated by fucoidan obtained from Fucus vesiculosus and Ascophyllum nodosum. Phytochemistry 2014, 98, 27–33. [Google Scholar] [CrossRef]
  99. Baub, C.D.; Mabate, B.; Malgas, S.; Pletschke, B.I. Fucoidan from Eclonia maxima is a powerful inhibitor of the diabetes-related enzyme, α-glucosidase. Int. J. Biol. Marcromol. 2020, 151, 412–420. [Google Scholar]
  100. Shan, X.; Liu, X.; Hao, J.; Cai, C.; Fan, F.; Dun, Y.; Zhao, X.; Liu, X.; Li, C.; Yu, G. In vitro and in vivo hypoglycemic effects of brown algal fucoidans. Int. J. Biol. Marcromol. 2016, 82, 249–255. [Google Scholar] [CrossRef]
  101. Wang, J.; Jin, W.; Zhang, W.; Hou, Y.; Zhang, H.; Zhang, Q. Hypoglycemic property of acidic polysaccharide extracted from Saccharina japonica and its potential mechanism. Carbohydr. Polym. 2013, 95, 143–147. [Google Scholar] [CrossRef]
  102. Senthil, S.L.; Chandrasekaran, R.; Arjun, H.A.; Anantharaman, P. In vitro and in silico inhibition properties of fucoidan against α-amylase and α-D-glucosidase with relevance to type 2 diabetes mellitus. Carbohydr. Polym. 2019, 209, 350–355. [Google Scholar]
  103. Cheng, Y.; Sibusiso, L.; Hou, L.; Jiang, H.; Chen, P.; Zhang, X.; Wu, M.; Tong, H. Sargassum fusiforme fucoidan modifies the gut microbiota during alleivation of streptozotocin-induced hyperglycemia in mice. Int. J. Biol. Marcromol. 2019, 131, 1162–1170. [Google Scholar] [CrossRef]
  104. Zhang, Y.; Zuo, J.; Yan, L.; Cheng, Y.; Li, Q.; Wu, S.; Chen, L.; Thring, R.W.; Yang, Y.; Gao, Y.; et al. Sargassum fusiforme fucoidan alleviates high-fat diet-induced obesity and insulin resistance associated with the improvement of hepatic oxidative stress and gut microbiota profile. J. Agric. Food Chem. 2020, 68, 10626–10638. [Google Scholar] [CrossRef] [PubMed]
  105. Wu, Q.; Wu, S.; Cheng, Y.; Zhang, Z.; Mao, G.; Li, S.; Yang, Y.; Zhang, X.; Wu, M.; Tong, H. Sargassum fusiforme fucoidan modified gut microbiota and intestinal metabolites during alleviation of hyperglycemia in type 2 diabetic mice. Food Funct. 2021, 12, 3572. [Google Scholar] [CrossRef]
  106. Lekshmi, V.S.; Arun, A.; Rauf, G.; Muraleedhara, K. Sulfated polysaccharides from the edible marine algae Padina tetrastromatica attenuates isoproterenol-induced oxidative damage via activation of PI3K/Akt/Nrf2 signaling pathway-An in vitro and in vivo approach. Chem. -Biol. Interact. 2019, 308, 258–268. [Google Scholar]
  107. Cui, W.; Zheng, Y.; Zhang, Q.; Wang, J.; Wang, L.; Yang, W.; Guo, C.; Gao, W.; Wang, X.; Luo, D. Low-molecular-weight fucoidan protects endothelial function and ameliorates basal hypertension in diabetic Goto-Kakizaki rats. Lab. Investig. 2014, 94, 382–393. [Google Scholar] [CrossRef]
  108. Gara, A.B.; Kolsi, R.B.A.K.; Jardak, N.; Chaaben, R.; El-Feki, A.; Fki, L.; Belghith, H.; Belghith, K. Inhibitory activities of Cystoseira crinita sulfated polysaccharide on key enzymes related to diabetes and hypertension: In vitro and animal study. Arch. Physiol. Biochem. 2017, 123, 31–42. [Google Scholar] [CrossRef]
  109. Li, X.; Li, J.; Li, Z.; Sang, Y.; Niu, Y.; Zhang, Q.; Ding, H.; Yin, S. Fucoidan from Undaria pinnatifida prevents vascular dysfunction through PI3K/Akt/eNOS-dependent mechanisms in the L-NAME-induced hypertensive rat model. Food Funct. 2016, 7, 2398–2408. [Google Scholar] [CrossRef] [PubMed]
  110. Preez, R.D.; Paul, N.; Mouatt, P.; Majzoub, M.E.; Thomas, T.; Panchal, S.K.; Brown, L. Carrageenans from the red seaweed Sarconema filiforme attenuate symptoms of diet-induced metablic syndrome in rats. Mar. Drugs 2020, 18, 97. [Google Scholar] [CrossRef] [Green Version]
  111. Qi, H.; Sheng, J. The antihyperlipidemic mechanism of high sulfate content ulvan in rats. Mar. Drugs 2015, 13, 3407–3421. [Google Scholar] [CrossRef] [PubMed]
  112. Oak, M.-H.; Auger, C.; Belcastro, E.; Park, S.-H.; Lee, H.-H.; Shini-Kerth, V.B. Potential mechanisms underlying cardiovascular protection by polyphenols: Role of the endothelium. Free Radic. Biol. Med. 2018, 122, 161–170. [Google Scholar] [CrossRef] [PubMed]
  113. Ren, R.; Azuma, Y.; Ojima, T.; Hashimoto, T.; Mizuno, M.; Nishitani, Y.; Yoshida, M.; Azuma, T.; Kanazawa, K. Modulation of platelet aggregation-related eicosanoid production by dietary F-fucoidan from brown alga Laminaria japonica in human subjects. Br. J. Nutr. 2013, 110, 880–890. [Google Scholar] [CrossRef] [Green Version]
  114. Faggio, C.; Pagano, M.; Morabito, M.; Minicante, S.A.; Arfuso, F.; Genovese, G. In vitro assessment of the effect of Undaria pinnatifida extracts on erythrocytes membrane integrity and blood coagulation parameters of Equus Caballus. J. Coast. Life Med. 2014, 2, 614–616. [Google Scholar]
  115. Favaloro, E.J.; Lippi, G.; Koutts, J. Laboratory testing of anticoagulants: The present and the future. Pathology 2011, 43, 682–692. [Google Scholar] [CrossRef] [PubMed]
  116. Casella, S.; Giannetto, C.; Giudice, E.; Marafioti, S.; Fazio, F.; Assenza, A.; Piccione, G. ADP-induced platelet aggregation after addition of tramadol in vitro in fed and fasted horses plasma. Res. Vet. Sci. 2013, 94, 325–330. [Google Scholar] [CrossRef]
  117. Yu, W.-C.; Chen, Y.-L.; Hwang, P.-A.; Chen, T.-H.; Chou, T.-C. Fucoidan ameliorates pancreatic β -cell death and impaired insulin synthesis in streptozotocin-treated β cells and mice via a Sirt-1-dependent manner. Mol. Nutr. Food Res. 2017, 61, 1700136. [Google Scholar] [CrossRef] [PubMed]
  118. Kim, K.-J.; Yoon, J.-Y.; Lee, B.-Y. Fucoidan regulate blood glucose homeostasis in C57BL/KSJ m+/+db and C57BL/KSJ db/db mice. Fitoterapia 2012, 83, 1105–1109. [Google Scholar] [CrossRef]
  119. Sim, S.-Y.; Shin, Y.-E.; Kim, H.-K. Fucoidan from Undaria pinnatifida has anti-diabetic effects by stimulation of glucose uptake and reduction of basal lipolysis in 3T3-L1 adipocytes. Nutr. Res. Pract. 2019, 65, 54–62. [Google Scholar] [CrossRef]
  120. Kui-Jin; Lee, B.-Y. Fucoidan from the sporophyll of Undaria pinnatifida suppresses adipocyte differentiation by inhibition of inflammation-related cytokines in 3T3-L1 cells. Nutr. Res. 2012, 32, 439–447. [Google Scholar]
  121. Juang, X.; Yu, J.; Ma, Z.; Zhang, H.; Xie, F. Effects of fucoidan on insulin stimulation and pancreatic protection via the cAMP signaling pathway in vivo and in vitro. Mol. Med. Rep. 2015, 12, 4501–4507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Lekshmi, V.S.; Kurup, G.M. Sulfated polysaccharides from the edible marine algae Padina tetrastromatica protects heart by ameliorating hyperlipidemia, endothelial dysfunction and inflammation in isoproterenol induced experimental myocardial infarction. J. Funct. Foods 2019, 54, 22–31. [Google Scholar] [CrossRef]
  123. Fan, X.; Bai, L.; Zhu, L.; Yang, L.; Zhang, X. Marine algae-derived bioactive peptides for human nutrition and health. J. Agric. Food Chem. 2014, 62, 9211–9222. [Google Scholar] [CrossRef]
  124. Sheih, I.-C.; Wu, T.-K.; Fang, T.J. Antioxidant properties of a new antioxidative peptide from algae protein waste hydrolysate in different oxidation systems. Bioresour. Technol. 2009, 100, 3419–3425. [Google Scholar] [CrossRef] [PubMed]
  125. Zhang, B.; Zhang, X. Separation and nanoencapsulation of antitumor polypeptide from Spirulina platensis. Biotechnol. Prog. 2013, 29, 1230–1238. [Google Scholar] [CrossRef] [PubMed]
  126. Cian, R.E.; Martínez-Augustin, O.; Drago, S.R. Bioactive properties of peptides obtained by enzymatic hydrolysis from protein byproducts of Porphyra columbina. Food Res. Int. 2012, 49, 364–372. [Google Scholar] [CrossRef]
  127. Sato, M.; Hosokawa, T.; Yamaguchi, T.; Nakano, T.; Muramoto, K.; Kahara, T.; Funayama, K.; Kobayashi, A.; Nakano, T. Angiotensin I-converting enzyme inhibitory peptides derived from wakame (Undaria pinnatifida) and their antihypertensive effect in spontaneously hypertensive rats. J. Agric. Food Chem. 2002, 50, 6245–6252. [Google Scholar] [CrossRef] [PubMed]
  128. Suetsuna, K.; Nakano, T. Identification of an antihypertensive peptide from peptic digest of wakame (Undaria pinnatifida). J. Nutr. Biochem. 2000, 11, 450–454. [Google Scholar] [CrossRef]
  129. Suetsuna, K.; Maekawa, K.; Chen, J.-R. Antihypertensive effects of Undaria pinnatifida (wakame) peptide on blood pressure in spontaneously hypertensive rats. J. Nutr. Biochem. 2004, 15, 267–272. [Google Scholar] [CrossRef]
  130. Furuta, T.; Miyabe, Y.; Yasui, H.; Kinoshita, Y.; Kishimura, H. Angiotensin I converting enzyme inhibitory peptides derived from phycobiliproteins of dulse Palmaria palmata. Mar. Drugs 2016, 14, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Harnedy, P.A.; O’Keeffe, M.B.; FitzGerald, R.J. Purification and identification of dipeptidyl peptidase (DPP) IV inhibitory peptides from the macroalga Palmaria palmata. Food Chem. 2015, 172, 400–406. [Google Scholar] [CrossRef]
  132. Lei, Y.; Hu, L.; Yang, G.; Piao, L.; Jin, M.; Cheng, X. Dipeptidyl peptidase-IV inhibition for the treatment of cardiovascular disease-Recent insights focusing on angiogenesis and neovascularization. Circ. J. 2017, 81, 770–776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Deng, Z.; Liu, Y.; Wang, J.; Wu, S.; Geng, L.; Sui, Z.; Zhang, Q. Antihypertensive Effects of Two Novel Angiotensin I-Converting Enzyme (ACE) Inhibitory Peptides from Gracilariopsis lemaneiformis (Rhodophyta) in Spontaneously Hypertensive Rats (SHRs). Mar. Drugs 2018, 16, 299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Cao, D.; Lv, X.; Xu, X.; Yu, H.; Sun, X.; Xu, N. Purification and identification of a novel ACE inhibitory peptide from marine alga Gracilariopsis lemaneiformis protein hydrolysate. Eur. Food Res. Technol. 2017, 243, 1829–1837. [Google Scholar] [CrossRef]
  135. Carrizzo, A.; Conte, G.M.; Sommella, E.; Damato, A.; Ambrosio, M.; Sala, M.; Scala, M.C.; Aquino, R.P.; Lucia, M.D.; Madonna, M.; et al. Novel potent decameric peptide of Spirulina platensis reduces blood pressure levels through a PI3K/AKT/eNOS-dependent mechanism. Hypertension 2019, 73, 449–457. [Google Scholar] [CrossRef] [PubMed]
  136. Kumagai, Y.; Kitade, Y.; Kobayashi, M.; Watanabe, K.; Kurita, H.; Takeda, H.; Yasui, H.; Kishimura, H. Identification of ACE inhibitory peptides from red alga Mazzaella japonica. Eur. Food Res. Technol. 2020, 246, 2225–2231. [Google Scholar] [CrossRef]
  137. Suetsuna, K. Purification and identification of angiotensin I-converting enzyme inhibitors from the red alga Porphyra yezoensis. J. Mar. Biotechnol. 1998, 6, 163–167. [Google Scholar]
  138. Suetsuna, K. Separation and identification of angiotensin I-converting enzyme inhibitory peptides from peptic digest of Hizikia fusiformis protein. Nippon Suisan Gakkaishi 1998, 64, 862–866. [Google Scholar] [CrossRef]
  139. Sun, S.; Xu, X.; Sung, X.; Zhang, X.; Chen, X.; Xu, N. Preparation and identification of ACE inhibitory peptides from the marine macroalga Ulva intestinalis. Mar. Drugs 2019, 17, 179. [Google Scholar] [CrossRef] [Green Version]
  140. Pan, S.; Wang, S.; Jing, L.; Yao, D. Purification and characterisation of a novel angiotensin-I converting enzyme (ACE)-inhibitory peptide derived from the enzymatic hydrolysate of Enteromorpha clathrata protein. Food Chem. 2016, 211, 423–430. [Google Scholar] [CrossRef] [PubMed]
  141. Kumagai, Y.; Toji, K.; Katsukura, S.; Morikawa, R.; Uji, T.; Yasui, H.; Shimizu, T.; Kishimura, H. Characterization of ACE Inhibitory Peptides Prepared from Pyropia pseudolinearis Protein. Mar. Drugs 2021, 19, 200. [Google Scholar] [CrossRef]
  142. Chen, J.C.; Wang, J.; Zheng, B.-D.; Pang, J.; Chen, L.-J.; Lin, H.-T.; Guo, X. Simultaneous Determination of 8 Small Antihypertensive Peptides with Tyrosine at the C-Terminal in L aminaria japonica Hydrolysates by RP-HPLC Method. J. Food Process. Preserv. 2016, 40, 492–501. [Google Scholar] [CrossRef]
  143. Wu, Q.; Cai, Q.-F.; Yoshida, A.; Sun, L.-C.; Liu, Y.-X.; Liu, G.-M.; Su, W.-J.; Cao, M.-J. Purification and characterization of two novel angiotensin I-converting enzyme inhibitory peptides derived from r-phycoerythrin of red algae (Bangia fusco-purpurea). Eur. Food Res. Technol. 2017, 243, 779–789. [Google Scholar] [CrossRef]
  144. Admassu, H.; Gasmalla, M.A.A.; Yang, R.; Zhao, W. Identification of bioactive peptides with α-amylase inhibitory potential from enzymatic protein hydrolysates of red seaweed (Porphyra spp). J. Agric. Food Chem. 2018, 66, 4872–4882. [Google Scholar] [CrossRef] [PubMed]
  145. Joel, C.H.; Sutopo, C.C.Y.; Prajitno, A.; Su, J.-H.; Hsu, J.-L. Screening of angiotensin-I converting enzyme inhibitory peptides derived from Caulerpa lentillifera. Molecules 2018, 23, 3005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Cermeño, M.; Stack, J.; Tobin, P.R.; O’Keeffe, M.B.; Harnedy, P.A.; Stengel, D.B.; FitzGerald, R.J. Peptide identification from a Porphyra dioica protein hydrolysate with antioxidant, angiotensin converting enzyme and dipeptidyl peptidase IV inhibitory activities. Food Funct. 2019, 10, 3421–3429. [Google Scholar] [CrossRef] [PubMed]
  147. Bhagavathy, S.; Sumathi, P. Evaluation of antigenotoxic effects of carotenoids from green algae Chlorococcum humicola using human lymphocytes. Asian Pac. J. Trop. Biomed. 2012, 2, 109–117. [Google Scholar] [CrossRef] [Green Version]
  148. Hwang, P.-A.; Phan, N.N.; Lu, W.-J.; Hieu, B.T.N.; Lin, Y.-C. Low-molecular-weight fucoidan and high-stability fucoxanthin from brown seaweed exert prebiotics and anti-inflammatory activities in Caco-2 cells. Food Nutr. Res. 2016, 60, 32033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Maeda, H.; Hosokawa, M.; Sashima, T.; Funayama, K.; Miyashita, K. Fucoxanthin from edible seaweed, Undaria pinnatifida, shows antiobesity effect through UCP1 expression in white adipose tissues. Biochem. Biophys. Res. Commun. 2005, 332, 392–397. [Google Scholar] [CrossRef]
  150. Ganesan, P.; Matsubara, K.; Sugawara, T.; Hirata, T. Marine algal carotenoids inhibit angiogenesis by down-regulating FGF-2-mediated intracellular signals in vascular endothelial cells. Mol. Cell. Biochem. 2013, 380, 1–9. [Google Scholar] [CrossRef]
  151. Sugawara, T.; Ganesan, P.; Li, Z.; Manabe, Y.; Hirata, T. Siphonaxanthin, a green algal carotenoid, as a novel functional compound. Mar. Drugs 2014, 12, 3660–3668. [Google Scholar] [CrossRef] [Green Version]
  152. Ganesan, P.; Noda, K.; Manabe, Y.; Ohkubo, T.; Tanaka, Y.; Maoka, T.; Sugawara, T.; Hirata, T. Siphonaxanthin, a marine carotenoid from green algae, effectively induces apoptosis in human leukemia (HL-60) cells. Biochim. Biophys. Acta Gen. Subj. 2011, 1810, 497–503. [Google Scholar] [CrossRef]
  153. Ha, A.W.; Kim, W.K. The effect of fucoxanthin rich power on the lipid metabolism in rats with a high fat diet. Nutr. Res. Pract. 2013, 7, 287–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Kawee-Ai, A.; Kim, A.T.; Kim, S.M. Inhibitory activities of microalgal fucoxanthin against α-amylase, α-glucosidase, and glucose oxidase in 3T3-L1 cells linked to type 2 diabetes. J. Oceanol. Limnol. 2019, 37, 928–937. [Google Scholar] [CrossRef]
  155. Harari, A.; Harats, D.; Marko, D.; Cohen, H.; Barshack, I.; Kamari, Y.; Gonen, A.; Gerber, Y.; Ben-Amotz, A.; Shaish, A. A 9-cis β-carotene–enriched diet inhibits atherogenesis and fatty liver formation in LDL receptor knockout mice. J. Nutr. 2008, 138, 1923–1930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Harari, A.; Harats, D.; Marko, D.; Cohen, H.; Barshack, I.; Gonen, A.; Ben-Shushan, D.; Kamari, Y.; Ben-Amotz, A.; Shaish, A. Supplementation with 9-cis β-carotene-rich alga Dunaliella improves hyperglycemia and adipose tissue inflammation in diabetic mice. J. Appl. Phycol. 2013, 25, 687–693. [Google Scholar] [CrossRef]
  157. Li, Z.-S.; Noda, K.; Fujita, E.; Manabe, Y.; Hirata, T.; Sugawara, T. The green algal carotenoid siphonaxanthin inhibits adipogenesis in 3T3-L1 preadipocytes and the accumulation of lipids in white adipose tissue of KK-Ay mice. J. Nutr. 2015, 145, 490–498. [Google Scholar] [CrossRef] [Green Version]
  158. Qi, J.; Kim, S.M. α-Glucosidase inhibitory activities of lutein and zeaxanthin purified from green alga Chlorella ellipsoidea. J. Ocean Univ. China 2018, 17, 983–989. [Google Scholar] [CrossRef]
  159. Sánchez-Machado, D.I.; López-Cervantes, J.; López-Hernández, J.; Paseiro-Losada, P. Fatty acids, total lipid, protein and ash contents of processed edible seaweeds. Food Chem. 2004, 85, 439–444. [Google Scholar] [CrossRef]
  160. Lee, S.; Lee, Y.S.; Kang, S.S.; Shin, K.H. Anti-oxidant activities of fucosterol from the marine algae Pelvetia siliquosa. Arch. Pharmacal Res. 2003, 26, 719–722. [Google Scholar] [CrossRef] [PubMed]
  161. Tang, H.-F.; Yi, Y.-H.; Yao, X.-S.; Xu, Q.-Z.; Zhang, S.-Y.; Lin, H.-W. Bioactive steroids from the brown alga Sargassum carpophyllum. J. Asian Nat. Prod. Res. 2002, 4, 95–101. [Google Scholar] [CrossRef] [PubMed]
  162. Chen, Z.; Liu, J.; Fu, Z.; Ye, C.; Zhang, R.; Song, Y.; Zhang, Y.; Li, H.; Ying, H.; Liu, H. 24 (S)-Saringosterol from edible marine seaweed Sargassum fusiforme is a novel selective LXRβ agonist. J. Agric. Food Chem. 2014, 62, 6130–6137. [Google Scholar] [CrossRef] [PubMed]
  163. Seong, S.H.; Nguyen, D.H.; Wagle, A.; Woo, M.H.; Jug, H.A.; Choi, J.S. Experimental and computational study to reveal the potential of non-polar constituents from Hizikia fusiformis as dual protein tyrosine phosphatase 1B and α-glucosidase inhibitors. Mar. Drugs 2019, 17, 302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Ali, M.; Kim, D.H.; Seong, S.H.; Kim, H.-R.; Jung, H.A.; Choi, J.S. α-Glucosidase and protein tyrosine phosphatase 1B inhibitory activity of plastoquinones from marine brown alga Sargassum serratifolium. Mar. Drugs 2017, 15, 368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Mierziak, J.; Kostyn, K.; Boba, A.; Czemplik, M.; Kulma, A.; Wojtasik, W. Influence of the bioactive diet components on the gene expression regulation. Nutrients 2021, 13, 3673. [Google Scholar] [CrossRef]
Figure 1. The preventive potential of various seaweed-derived natural components associated with cardiovascular disease (CVD) pathogenesis, such as diabetes mellitus, obesity, hypertension, and hyperlipidemia.
Figure 1. The preventive potential of various seaweed-derived natural components associated with cardiovascular disease (CVD) pathogenesis, such as diabetes mellitus, obesity, hypertension, and hyperlipidemia.
Applsci 12 01025 g001
Table 1. Phlorotannin compounds isolated from seaweeds with therapeutic potential against cardiovascular disease.
Table 1. Phlorotannin compounds isolated from seaweeds with therapeutic potential against cardiovascular disease.
SourceTarget Disease/MechanismBioactive ComponentModel UsedBiological EffectReference
Ecklonia cavaDiabetes mellitus/α-glucosidase and α-amylase inhibitionFucodiphloroethol G, dieckol, 6,6′-bieckol, 7-phloroeckol, phlorofucofuroeckol Ain vitro– α-Glucosidase inhibitory activity (IC50): fucodiphloroethol G (19.52 μM/L−1), dieckol (10.79 μM/L−1), 6,6′-bieckol (22.22 μM/L−1), 7-phloroeckol G (49.49 μM/L−1), phlorofucofuroeckol A (19.71 μM/L−1)
– α-Amylase inhibitory activity (IC50): fucodiphloroethol G (>500 μM/L−1), dieckol (124.98 μM/L−1), 6,6′-bieckol (>500 μM/L−1), 7-phloroeckol G (250.02 μM/L−1), phlorofucofuroeckol A (>500 μM/L−1)
[49]
Diabetes mellitus/α-glucosidase inhibitionEckol, 2-phloroeckol, 8,8′-bieckol, 6,8′-bieckol, 2-O-(2,4,6-trihydroxyphenyl)-6,6′-bieckolin vitro– α-Glucosidase inhibitory activity (IC50): Eckol (59.8 ± 0.8 μM), 2-phloroeckol (32.5 ± 2.1 μM), 8,8′-bieckol (12.5 ± 3.1 μM, competitive), 6,8′-bieckol (2.3 ± 1.2 μM, competitive), 2-O-(2,4,6-trihydroxyphenyl)-6,6′-bieckol (123.1 ± 2.4 μM, competitive)[48]
Diabetes mellitus/α-glucosidase and α-amylase inhibition, INS-1 cell protection against glucotoxicity2,7″-phloroglucinol-6,6′-bieckolin vitro and in vivo– α-Glucosidase inhibitory activity (IC50): 2,7″-phloroglucinol-6,6′-bieckol (23.35 μM)
– α-Amylase inhibitory activity (IC50): 2,7″-phloroglucinol-6,6′-bieckol (6.94 μM)
- Glucose response curve: 2,7″-phloroglucinol-6,6′-bieckol (2349.3 mmol·min/L)
– 2,7″-phloroglucinol-6,6′-bieckol protects pancreatic β cells against high glucose-induced apoptosis
[50,60]
Diabetes mellitus/α-glucosidase and α-amylase inhibition, postprandial hyperglycemia inhibitionDieckolin vitro and in vivo– α-Glucosidase inhibitory activity (IC50): dieckol (0.24 mM)
– α-amylase inhibitory activity (IC50): dieckol (0.66 mM)
– Area under the curve of postprandial glucose responses in streptozotocin-induced diabetic mice: diabetic mice (483 mmol·min/L), dieckol 100 mg/kg body weight (259 mmol·min/L)
[51]
Ecklonia cavaDiabetes mellitus/activation of both AMPK and Akt signaling pathwaysDieckolin vivo– Administration of 20 mg/kg body weight dieckol was reduced blood glucose, serum insulin level, and body weight[61]
Obesity/anti-adipogenesisTriphlorethol-A, eckol, dieckolin vitro– Triphlorethol-A, eckol, dieckol (20 μM): decreased intracellular lipid accumulation and increased intracellular calcification with an intervention in differentiation pathways at 3T3-L1 and MC3T3-E1 cell lines, respectively[70]
Obesity/inhibition of adipogenesisDieckol, 6,6′-bieckol, phlorofucoeckol Ain vitro– Phlorotannin compounds such as dieckol, 6,6′-bieckol, and phlorofucoeckol A in Ecklonia cava inhibit intracellular lipid accumulation. In particular, dieckol suppressed adipogenesis in 3T3-L1 cells by suppressing the expression of PPARγ, C/EBPα, SREBP-1, and FABP4.[71]
Obesity/suppresses lipid accumulation and adipogenesisDieckolin vitro and in vivo– Dieckol inhibits early adipogenic events by suppressing cell cycle progression, and plays important roles in regulating AMPKα, ERK, and AKT signaling to inhibit lipid accumulation on high-fat diet-fed zebrafish, mice and 3T3-L1 models[72]
Obesity/reduced leptin resistanceDieckol, 2,7-phloroglucinol-6,6-bieckol, pyrogallol-phloroglucinol-6,6-bieckol, phlorofucofuroeckol Ain vitro– Phlorotannin compounds such as dieckol, 2,7-phloroglucinol-6,6-bieckol, pyrogallol-phloroglucinol-6,6-bieckol, and phlorofucofuroeckol A isolated from Ecklonia cava had the most potent effect on attenuating leptin resistance[79]
Hypertension/ACE inhibition6,6′-Bieckolin vitro– ACE inhibitory activity (IC50): 0.42 mM[28]
Hypertension/ACE inhibitionPhloroglucinol, Triphlorethol-A, eckol, dieckol, eckstolonolin vitro– ACE inhibitory activity (IC50): phloroglucinol (2.57 mM), triphlorethol-A (2.01 mM), eckol (2.27 mM), dieckol (1.47 mM), eckstolonol (2.95 mM)[26]
Hypertension/vascular smooth muscle cell proliferation and migrationDieckol, 2,7-phloroglucinol-6,6-bieckol, phlorofucofuroeckol A, pyrogallol-phloroglucinol-6,6-bieckolin vitro– Inhibits monocyte migration and differentiation to inflammatory macrophages and monocyte associated vascular cell dysfunction[37]
Ecklonia cavaHypertension/improved blood circulationPyrogallol-phloroglucinol-6,6′-bieckol (PPB)in vitro and in vivo– PPB improved blood circulation, including reduced adhesion molecule expression, endothelial cell death, excessive vascular smooth muscle cell proliferation, and migration– PPB remarkably reduced blood pressure, serum cholesterol, and lipoprotein levels in vivo[38]
Hypertension/promotion of vasodilationDieckolin vitro and in vivo– Dieckol effectively promoted endothelial-dependent NO production by activating the PI3K/Akt/eNOS pathway and [Ca2+]cytosol regulation
– Dieckol promotes vasodilation by increasing the DA diameter, further regulating blood-flow velocity in a zebrafish model
[35]
Hyperlipidemia/reduction of total cholesterol, triglyceride, low-density lipoproteinDieckolin vitro and in vivo– In vitro: 200 μg/mL dieckol inhibited adipocyte differentiation, intracellular triglyceride accumulation, and lipid accumulation in 3T3-L1 cells
– In vivo: administration of dieckol reduced total cholesterols, triglycerides and low-density lipoproteins in the serum of high-fat diet mice
[82]
Ecklonia stoloniferaDiabetes mellitus/protein tyrosine phosphatase 1B, α-glucosidase inhibitionPhloroglucinol, dioxinodehydroeckol, eckol, phlorofucofuroeckol-A, dieckol, 7-phloroeckolin vitro– Protein tyrosine phosphatase 1B inhibitory activity (IC50): phloroglucinol (55.48 μM), dioxinodehydroeckol (29.97 μM), eckol (2.64 μM), phlorofucofuroeckol-A (0.56 μM), dieckol (1.18 μM), 7-phloroeckol (2.09 μM)
– α-Glucosidase inhibitory activity (IC50): phloroglucinol (141.18 μM), dioxinodehydroeckol (34.60 μM), eckol (22.78 μM), phlorofucofuroeckol-A (1.37 μM), dieckol (1.61 μM), 7-phloroeckol (6.13 μM)
[52]
Obesity/inhibition of lipid accumulation and adipocyte differentiation, modulation of adipocyte marker gene expressionPhloroglucinol, eckol, dieckol, dioxinodehydroeckol, phlorofucofuroeckol Ain vitro– Phlorotannins, such as phloroglucinol, eckol, dieckol, dioxinodehydroeckol, and phlorofucofuroeckol A isolated from Ecklonia stolonifera was reduced lipid accumulation in 3T3-L1 cell line
– These phlorotannin compounds suppressed adipocyte differentiation through inhibiting C/EBPα and PPARγ expression
[73]
Hypertension/ACE inhibitionPhloroglucinol, eckstolonol, eckol, phlorofucofuroeckol A, dieckol, Triphlorethol-A, fucosterolin vitro– ACE inhibitory activity (IC50) of phloroglucinol: (N.A.), eckstolonol (410.12 μM), eckol (70.82 μM), phlorofucofuroeckol A (12.74 μM), dieckol (34.25 μM), Triphlorethol-A (700.9 μM), fucosterol (N.A.)[27]
Ecklonia stoloniferaHyperlipidemia/reduction of Cu2+-induced LDL oxidationPhloroglucinol, dioxinodehydroeckol, eckol, phlorofucofuroeckol-A, dieckol, 7-phloroeckolin vitro– Cu2+-induced LDL oxidation inhibitory activity (IC50): phloroglucinol (87.30 μM), dioxinodehydroeckol (16.57 μM), eckol (7.47 μM), phlorofucofuroeckol-A (4.34 μM), dieckol (3.10 μM), 7-phloroeckol (9.07 μM)[80]
Hyperlipidemia/reduction of total cholesterol, triglyceride, low-density lipoprotein-cholesterol, and atherogenic indexEckol, dieckolin vivo– Poloxamer 407-induced hyperlipidemic rats model:
20 mg/kg BW eckol—TC level (255.6 mg/dL → 157.0 mg/dL), TG level (240.2 mg/dL → 174.9 mg/dL), LDL-C level (145.1 mg/dL →63.1 mg/dL), AI (3.47 → 1.77)
20 mg/kg BW dieckol—TC level (255.6 mg/dL → 144.7 mg/dL), TG level (240.2 mg/dL → 165.7 mg/dL), LDL-C level (145.1 mg/dL →35.5 mg/dL), AI (3.47 → 0.95)
– High-cholesterol diet rats model:
20 mg/kg BW eckol—TC level (239.9 mg/dL → 226.3 mg/dL), TG3 level (271.1 mg/dL → 256.7 mg/dL), LDL-C3 level (160.6 mg/dL → 146.8 mg/dL), AI (7.55 → 7.14)
20 mg/kg BW dieckol—TC level (239.9 mg/dL → 200.7 mg/dL), TG level (271.1 mg/dL → 219.8 mg/dL), LDL-C level (160.6 mg/dL → 125.4 mg/dL), AI (7.55 → 5.53)
[81]
Eisenia bicyclisDiabetes mellitus/α-fucosidase, β-galactosidase, β-mannosidase inhibitionPhloroglucinol, phloroglucinol tetramer, eckol, phlorofucofuroeckol A, dieckol, 8,8′-bieckolin vitro– Among the 6 phlorotannin compounds isolated from Eisenia bicyclis, phlorofucofuroeckol A, dieckol, and 8,8′-bieckol showed α-fucosidase, β-galactosidase, and β-mannosidase inhibitory activity. On the other hand, phloroglucinol, phloroglucinol tetramer, and eckol showed a weak activity of inhibiting these enzymes.
– Dieckol was exhibited as a competitive inhibitor of α-fucosidase with an inhibition constant (K1) of 0.12 mM
[83]
Obesity/pancreatic lipase inhibitory activityEckol, fucofuroeckol A, 7-phloroeckol, dioxindehydroeckol, phlorofucofuroeckol A, dieckolin vitro– Pancreatic lipase inhibitory activity (IC50): eckol (76.6 μM), fucofuroeckol A (37.2 μM), 7-phloroeckol (12.7 μM), dioxindehydroeckol (>200 μM), phlorofucofuroeckol A (>200 μM), dieckol (99.3 μM)[78]
Obesity/inhibition of lipid accumulation and adipocyte differentiation6,6′-bieckol, 6,8′-bieckol, 8,8′-bieckol, dieckol, phlorofucofuroeckol-Ain vitro– Phlorotannin compounds such as 6,6′-bieckol, 6,8′-bieckol, 8,8′-bieckol, dieckol, and phlorofucofuroeckol-A isolated from Eisenia bicyclis showed suppressed differentiation of 3T3-L1 adipocyte through downregulation of adipogenesis and lipogenesis[74]
Ishige okamuraeDiabetes mellitus/improve glucose homeostasisIshophloroglucin A
(IPA)
in vivo– Administration of 1.35 mg/kg BW IPA improved glucose homeostasis in high-fat diet-fed mice
– IPA ameliorated glucose intolerance, reducing fasting glucose levels and 2 h glucose levels in high-fat diet-fed mice
– IPA protect pancreatic function in high-fat diet-fed mice through pancreatic β-cells and C-peptide
– Administration of IPA improves glucose homeostasis by increasing glucose transporter 4 levels in the muscles of high-fat diet-fed mice
[62]
Diabetes mellitus/human umbilical vein endothelial cell protection against high glucose-induced oxidative stressDiphlorethohydroxycarmalol
(DPHC)
in vitro– DPHC prevented human umbilical vein endothelial cells from high glucose-induced damage through restoring cell viability, suppressed lipid peroxidation, reduced intracellular reactive oxygen species, and nitric oxide level[84]
Diabetes mellitus/α-glucosidase and α-amylase inhibitionDiphlorethohydroxycarmalol
(DPHC)
in vitro and in vivo– α-Glucosidase inhibitory activity (IC50): 0.16 mM
– α-Amylase inhibitory activity (IC50): 0.53 mM
– Administration of 100 mg/kg DPHC was reduced blood glucose level in streptozotocin-induced diabetic mice
– Postprandial glucose response: normal mice (965 mmol·min/L), diabetic mice (2210 mmol·min/L), DPHC treated mice (1964 mmol·min/L)
[55]
Diabetes mellitus/α-glucosidase inhibitionIshophloroglucin A (IPA), diphlorethohydroxycarmalol
(DPHC)
in vitro– α-Glucosidase inhibitory activity (IC50): IPA (54.97 μM), DPHC (175.78 μM)[53]
Diabetes mellitus/inhibition of abnormal angiogenesis, vascular dysfunctionDiphlorethohydroxycarmalol
(DPHC)
in vitro and in vitro– DPHC treatment suppressed the phosphorylation of VEGFR-2 and down-regulation of angiogenesis-related key mechanisms[64]
Diabetes mellitus/protect RINm5F pancreatic β cells from high glucose-induced damageDiphlorethohydroxycarmalol
(DPHC)
in vivo– DPHC treatment inhibited the apoptotic cell death of RINm5F pancreatic β cell via decrease of thiobarbituric acid reactive substances, intracellular reactive oxygen species generation, and nitric oxide level[85]
Ishige okamuraeDiabetes mellitus/anti-angiogenic effectIshophloroglucin A
(IPA)
in vitro– IPA effectively inhibited high glucose-induced endothelial cell proliferation, migration, and capillary formation, and exhibited an anti-angiogenic effect by interfering with the VEGFR-2 signaling pathway[65]
Obesity/reduction of total cholesterol, triglyceride, low-density lipoprotein-cholesterol, and atherogenic indexDiphlorethohydroxycarmalol
(DPHC)
in vivo– Triglyceride levels: high-fat diet mice 137.88 mg/dL → 50 mg/kg BW 86.73 mg/dL
– High-density lipoprotein cholesterol levels: high-fat diet mice 50.49 mg/dL → 50 mg/kg BW 72.71 mg/dL
– Low-density lipoprotein cholesterol levels: high-fat diet mice 22.24 mg/dL → 50 mg/kg BW 16.82 mg/dL
– Leptin levels: high-fat diet mice 2.04 ng/dL → 50 mg/kg BW 1.23 ng/dL
[86]
Obesity/induces apoptosis in 3T3-L1 preadipocytesDiphlorethohydroxycarmalol
(DPHC)
in vitro– DPHC treatment increased the number of early and late apoptotic cells in 3T3-L1 pre-adipocytes
– DPHC mediated apoptotic cell death via the activation of caspase-3, caspase-8, and Bax
[75]
Obesity/anti-adipogenesisDiphlorethohydroxycarmalol
(DPHC)
in vitro– DPHC showed an anti-adipogenic effect via regulation of ECM during adipogenesis
– DPHC treatment positively affects normal adipose tissue generation and acts as a suppressor of abnormal ECM structures
[76]
Obesity/inhibition of lipid accumulation and suppressed adipogenesis via AMPK activationDiphlorethohydroxycarmalol
(DPHC)
in vitro– DPHC treatment inhibited the fat accumulation by activating AMPK and ACC phosphorylation in 3T3-L1 adipocytes[77]
Hypertension/vasodilatory effect through increasing calcium intake levelDiphlorethohydroxycarmalol
(DPHC)
in vitro and in vivo– DPHC stimulated NO production by increasing calcium levels and endothelial nitric oxide synthase expression
– DPHC modulated Ca2+ levels by activating AchR and VEGFR2
– DPHC modulated calcium transit through AchR and VEGFR2, increasing endothelial-dependent NO production in a zebrafish model
[36]
Ishige foliaceaDiabetes mellitus/anti-diabetogenic effectOctaphlorethol A
(OPA)
in vivo– Administration of 10 mg/kg BW OPA increased anti-apoptotic (Bcl-xL) and pro-apoptotic (Bax) protein expression level and increased antioxidant enzymes (SOD, CAT, GSH).[87]
Ishige foliaceaDiabetes mellitus/impaired glucose tolerance improvementOctaphlorethol A
(OPA)
in vivo– OPA treatment significantly decreased postprandial blood glucose levels in db/db mice
– OPA supplements significantly improved fasting blood glucose levels and impaired glucose tolerance, decreased serum insulin levels, augmented the activation of AMPK, and increased the expression of GLUT4 in skeletal muscle
[63]
Diabetes mellitus/pancreatic β cells protectionOctaphlorethol A
(OPA)
in vitro– Pretreatment with 50 μg/mL OPA decreased the streptozotocin-induced pancreatic β cells damage by reducing the thiobarbituric acid reactive substances and intracellular ROS generation
– OPA treatment increased the activity of antioxidant enzymes such as CAT, SOD, GSH in STZ-treated pancreatic β cells
[88]
Diabetes mellitus/α-glucosidase inhibitionOctaphlorethol A
(OPA)
in vitro and in vivo– α-glucosidase inhibitory activity (IC50): OPA (0.11 mM)
– α-glucosidase molecular docking: binding energy (−140.98 kcal mol−1)
– OPA interacts with Phe575, His600, Arg526, Met444, Asp542, Tyr605, Ser448, Asp203, Lys480, and Phe450
– OPA treatment suppressed increases in postprandial blood glucose levels
[54]
Ishige sinicolaHypertension/ACE inhibitionOctaphlorethol A
(OPA)
in vitro– ACE inhibitory activity (IC50): OPA (59 μM)
– OPA exhibited an anti-hypertensive effect via AMPK and Akt activation in endothelial cells
[29]
ACE, angiotensin I-converting enzyme; DA, dorsal aorta; TC, total cholesterol; TG, triglyceride; LDL-C, low-density lipoprotein-cholesterol; AI, atherogenic index.
Table 2. Polysaccharides isolated from seaweeds with therapeutic potential against cardiovascular disease.
Table 2. Polysaccharides isolated from seaweeds with therapeutic potential against cardiovascular disease.
SourceTarget Disease/MechanismBioactive ComponentModel UsedBiological EffectReference
Sargassum wightiiDiabetes mellitus/α-glucosidase and α-amylase inhibition
Hypertension/ACE inhibition
Sulfated
polygalactofucan
in vitro– α-Glucosidase activity (IC50): 1.48 mg/mL
– α-Amylase activity (IC50): 0.93 mg/mL
– ACE inhibitory activity (IC50): 0.22 mg/mL
[94]
Diabetes mellitus/α-amylase inhibitionFucoidanin vitro– α-Amylase activity (IC50): 103.83 μg[95]
Diabetes mellitus/α-glucosidase inhibitionFucoidanin vitro– α-Glucosidase activity (IC50): 132 μg[96]
Sargassum fusiformeDiabetes mellitus/inhibition of hyperglycemia, altered the composition of gut microbiotaFucoidanin vivo– Administration of fucoidan decreased fasting blood glucose levels, dietary and water intake, and alleviated pathological changes in the heart and liver
– Fucoidan supplements altered the composition of gut microbiota in STZ-induced diabetic mice
[103]
Diabetes mellitus/inhibition of hyperglycemia, altered the composition of gut microbiotaFucoidanin vivo– Administration of fucoidan decreased fasting blood glucose, food consumption, water intake, and serum lipid levels in high-fat diet/STZ-induced diabetic mice
– Administration of fucoidan altered the composition of gut microbiota and increased the levels of carnitine and choline in the colon
[105]
Diabetes mellitus/improved insulin sensitivity, altered the composition of gut microbiotaFucoidanin vivo– Administration of fucoidan reduced fasting blood glucose and insulin resistance indexes and improved glucose tolerance
– Administration of fucoidan increased the abundance and diversity of gut microbiota in obese mice and improved intestinal integrity
[104]
Sargassum hemiphyllumDiabetes mellitus/prevention of pancreatic β cell damage and dysfunctionFucoidanin vitro and in vivo– Fucoidan treatment attenuated pancreatic β cell death, pancreatic islet mass loss, and dysfunction
– Fucoidan treatment increased insulin synthesis via activation of Sirt-1-dependent upregulation of PDX and GLP-1R
[117]
Undaria pinnatifidaDiabetes mellitus/regulation of blood glucose homeostasisFucoidanin vivo– Serum insulin (μIU/mL): db/db mice (41.6) → db/db mice + fucoidan (37.7)
– Fasting blood glucose (mg/dL): db/db mice (445) → db/db mice + fucoidan (257)
[118]
Diabetes mellitus/improved insulin-stimulated glucose uptakeFucoidanin vitro– Fucoidan treatment stimulated glucose uptake and inhibited basal lipolysis in hypertrophied insulin resistance[119]
Hypertension/vascular dysfunction preventionFucoidanin vivo– Fucoidan treatment induce NO release and eNOS activation
– In L-NAME-induced hypertensive rats, administration of fucoidan attenuated elevated blood pressure, increased endothelium-dependent vasodilation, and improved vascular elasticity
[109]
Undaria pinnatifidaObesity/inhibition of adipocyte differentiationFucoidanin vitro– Fucoidan treatment suppressed adipogenesis by inhibiting proliferator-activated receptor γ, CCAAR/enhancer-binding protein α, adipocyte protein 2, and lipid accumulation in 3T3-L1 cells[120]
Saccharina longicrurisDiabetes mellitus/α-amylase inhibitionFucoidanin vitro– α-amylase activity (%):fucoidan (1 mg/mL) inhibited α-amylase activity by 80.3%[97]
Fucus vesiculosusDiabetes mellitus/α-amylase inhibitionFucoidanin vitro– α-amylase activity (%): harvested in October 2002: NA, commercial fucoidan from F. vesiculosus: NA[97]
Diabetes mellitus/α-glucosidase inhibitionFucoidanin vitro– α-glucosidase activity (IC50): 0.049 mg/mL[98]
Diabetes mellitus/insulin stimulation and pancreatic protectionFucoidanin vitro and in vivo– Fucoidan supplements increase insulin secretion and provide pancreatic protection via the cAMP signaling pathway[121]
Diabetes mellitus/α-amylase inhibitionFucoidanin vitro– α-amylase inhibitory activity (IC50): 0.04 mg/mL[99]
Diabetes mellitus/α-glucosidase inhibitionFucoidanin vitro– α-glucosidase inhibitory activity (IC50): 67.9 μg/mL[100]
Ascophyllum nodosumDiabetes mellitus/α-amylase inhibitionFucoidanin vitro– α-amylase activity (%):fucoidan (1 mg/mL) inhibited α-amylase activity by 83.2%[97]
Diabetes mellitus/α-glucosidase inhibitionFucoidanin vitro– α-glucosidase activity according to harvest seasons (IC50): May (0.047 mg/mL), June (0.037 mg/mL), July (0.014–0.036 mg/mL), August (0.017–0.046 mg/mL), September (0.026–0.029 mg/mL), October (0.013 mg/mL), and November (0.014 mg/mL)[98]
Diabetes mellitus/α-glucosidase inhibitionFucoidanin vitro– α-glucosidase inhibitory activity (IC50): 165.4 μg/mL[100]
Cystoseira crinitaDiabetes mellitus/α-amylase inhibitionSulfated polysaccharidein vitro and in vivo– α-amylase inhibitory activity (IC50): 39.16 μg/mL
– α-amylase inhibitory activity (IC50): in serum (23%), in pancreas (44.38%), and intestine (45%)
[108]
Hypertenstion/ACE inhibitionSulfated polysaccharidein vitro and in vivo– ACE inhibitory activity (IC50): 58.35 μg/mL[108]
Saccharina japonicaDiabetes mellitus/reduced hyperglycemiaFucoidanin vivo– Administration of 1200 mg/kg BW fucoidan reduced the blood glucose level by 34%
– Increased serum insulin levels
– Fucoidan supplements alter plasma lipid levels by lowering cholesterol, triglyceride, and low-density lipoprotein concentrations
[101]
Ecklonia maximaDiabetes mellitus/α-glucosidase inhibitionFucoidanin vitro– α-amylase inhibitory activity (IC50): 0.29 mg/mL[99]
Sargassum thumbergiiDiabetes mellitus/α-glucosidase inhibitionFucoidanin vitro– α-glucosidase inhibitory activity (IC50): 376.7 μg/mL[100]
Sargassum honeriDiabetes mellitus/α-glucosidase inhibitionFucoidanin vitro– α-glucosidase inhibitory activity (IC50): 351.0 μg/mL[100]
Sargassum ringgoldianumDiabetes mellitus/α-glucosidase inhibitionFucoidanin vitro– α-glucosidase inhibitory activity (IC50): 172.9 μg/mL[100]
Sargassum siliquastrumDiabetes mellitus/α-glucosidase inhibitionFucoidanin vitro– α-glucosidase inhibitory activity (IC50): 399.6 μg/mL[100]
Sargassum graminifoliumDiabetes mellitus/α-glucosidase inhibitionFucoidanin vitro– α-glucosidase inhibitory activity (IC50): 271.7 μg/mL[100]
Sargassum kjellmanianumDiabetes mellitus/α-glucosidase inhibitionFucoidanin vitro– α-glucosidase inhibitory activity (IC50): 415.2 μg/mL[100]
Turbinaria conoidesDiabetes mellitus/α-amylase and α-d-glucosidase inhibitionFucoidanin vitro– α-amylase inhibitory activity (IC50): 1.07 μM
– α-d-glucosidase inhibitory activity (IC50): 0.68 μM
[102]
Laminaria japonicaDiabetes mellitus/protects endothelial functionLow-molecular-weight fucoidanin vivo– Administration of 200 mg/kg/day fucoidan protects vasoendothelial function and reduces basal blood pressure in type 2 diabetes rats[107]
Padina tetrastromaticaHypertension/cardioprotective effectSulfated polysaccharidesin vitro and in vivo– Treatment of sulfated polysaccharides isolated from Padina tetrastromatica reduced isoproterenol-induced cardiac damage via activation of PI3K/Akt/Nrf2 signaling pathway[106]
Atherosclerosis/reduced hyperlipidemia, endothelial dysfunctionSulfated polysaccharidesin vitro and in vivo– Sulfated polysaccharides treatment maintained lipid homeostasis by regulating the expressions of SREBP-2 and LDL-R
– Administration of sulfated polysaccharides normalized ISO-induced oxidative damage, hyperlipidemia, endothelial dysfunction, and inflammation in a rat model.
[122]
Sarconema
filiforme
Modulation of cardiovascular and metabolic health parametersCarrageenanin vivo– Administration of 5% carrageenan in high-fat diet-fed rats attenuated cardiovascular diseases and metabolic health parameters[110]
Ulva
Pertusa
Hyperlipidemia/modulating hyperlipidemia related parametersUlvanin vivo– Ulvan decreased total cholesterol and low-density lipoprotein levels in high cholesterol-fed rats
– Ulvan treatment improved lipid profiles via regulating FXR, PPARγ, and LXR expression levels
[111]
ACE, angiotensin I-converting enzyme; STZ, streptozotocin.
Table 3. Peptides isolated from seaweeds with therapeutic potential against cardiovascular disease.
Table 3. Peptides isolated from seaweeds with therapeutic potential against cardiovascular disease.
SourceTarget Disease/MechanismBioactive ComponentModel UsedBiological EffectReference
Undaria pinnatifidaHypertension/ACE inhibition, lower SBPVal-Tyr, Ile-Tyr, Ala-Trp, Phe-Tyr, Val-Trp, Ile-Trp, and Leu-Trp, Val-Tyr, Ile-Tyr, Phe-Tyr, and Ile-Tyrin vitro– ACE inhibitory activity (IC50): Val-Tyr (35.2 μM), Ile-Tyr (6.1 μM), Ala-Trp (18.8 μM), Phe-Tyr (42.3 μM), Val-Trp (3.3 μM), Ile-Trp (1.5 μM) and Leu-Trp (23.6 μM)
– Lower SBP (Val-Tyr; 206.7 mmHg, Ile-Tyr;184.3 mmHg, Phe-Tyr; 193.0 mmHg, and Ile-Trp;199.5 mmHg)
[127]
Hypertension/ACE inhibitionAla-Ile-Tyr-Lys, Tyr-Lys-Tyr-Tyr, Lys-Phe-Tyr-Gly, and Tyr-Asn-Lys-Leuin vitro– ACE inhibitory activity (IC50): Ala-Ile-Tyr-Lys (213 μM), Tyr-Lys-Tyr-Tyr (64.2 μM), Lys-Phe-Tyr-Gly (90.5 μM) and Tyr-Asn-Lys-Leu (21 μM)[128]
Undaria pinnatifidaHypertension/ACE inhibitionTyr-His, Lys-Tyr, Phe-Tyr, and Ile-Tyrin vitro– ACE inhibitory activity (IC50): Tyr-His (5.1 μM), Lys-Tyr (7.7 μM), Phe-Tyr (3.7 μM) and Ile-Tyr (2.7 μM)[129]
Gracilariopsis lemaneiformisHypertension/ACE inhibitionFQIN [M(O)] CILR and TGAPCRin vitro and in vivo– ACE inhibitory activity (IC50): FQIN [M(O)] CILR (9.64 μM) and TGAPCR(23.94 μM)
– Decrements in SBP: FQIN [M(O)] CILR (27 mmHg) and TGAPCR (25 mmHg)
[133]
Hypertension/ACE inhibitionGln-Val-Glu-Tyrin vitro– ACE inhibitory activity (IC50): Gln-Val-Glu-Tyr (474.36 μM)[134]
Chlorophyta U. rigidaHypertension/ACE inhibitionIle-Pro and Ala-Phe-Leuin vitro– ACE inhibitory activity (IC50): Ile-Pro (87.6 μM), Ala-Phe-Leu (65.9 μM)[135]
Sargassum maclureiHypertension/ACE inhibitionRWDISQPYin vitro– ACE inhibitory activity (IC50): 72.24 μM
– Endothelin-1 suppressing capacity: 26.21% at 1.5 mg/mL
[136]
Mazzaella japonicaHypertension/ACE inhibitionYRDin vitro– ACE inhibitory activity (IC50): YRD (320 μM)[137]
Sargassum fusiformeHypertension/ACE inhibitionly-Lys-Tyr, Ser-Val-Tyr and Ser-Lys-Thr-Tyrin vitro– ACE inhibitory effect (IC50): Gly-Lys-Tyr (3.92 μM) Ser-Val-Tyr (8.12 μM) and Ser-Lys-Tyr-Tyr (11.07 μM)[138]
Ulva intestinalisHypertension/ACE inhibitionPhe-Gly-Met-Pro-Leu-Asp-Arg and Met-Glu-Leu-Val-Leu-Argin vitro– ACE inhibitory activity (IC50): Phe-Gly-Met-Pro-Leu-Asp-Arg (219.35 μM) and Met-Glu-Leu-Val-Leu-Arg (236.85 μM)[139]
Enteromorpha clathrateHypertension/ACE inhibitionPro-Ala-Phe-Glyin vitro– ACE inhibitory activity (IC50): Pro-Ala-Phe-Gly(35.9 μM)[140]
Pyropia PseudolinearisHypertension/ACE inhibitionLRMin vitro– ACE inhibitory activity (IC50): LRM (0.15 μM)[141]
Palmaria palmataHypertension/ACE inhibitionα subunits of PE (rPEα and rPEβ)in vitro– ACE inhibitory effect (%): rPEα (94.4%) and rPEβ (87.0%)[130]
Diabetes mellitus/DPP-Ⅳ inhibitory activityIle-Leu-Ala-Pro, Leu-Leu-Ala-Pro and Met-Ala-Gly-Val-Asp-His-Ilein vitro– DPP-Ⅳ inhibitory activity (IC50): Ile-Leu-Ala-Pro (17.90 μg/mL), Leu-Leu-Ala-Pro (22.14 μg/mL), and Met-Ala-Gly-Val-Asp-His-Ile (118.23 μg/mL)[131]
Laminaria JaponicaHypertension/ACE inhibitionKY, GKY, SKTY, AKY, AKYSY, KKFY, FY and KFKYin vitro– ACE inhibitory activity (IC50): KY (5.24 μM), GKY (7.94 μM), SKTY (20.63 μM), AKY (7.52 μM), AKYSY (2.42 μM), KKFY (15.33 μM), FY (4.83 μM), and KFKY (10.73 μM)[142]
Bangia fusco-purpureaHypertension/ACE inhibitionr-phycoerythrin, ALLAGDPSVLEDR and VVGGTGPVDEWGIAGARin vitro– ACE inhibitory activity (IC50): r-phycoerythrin (191.1 μg/mL), ALLAGDPSVLEDR (57.2 μg/mL), and VVGGTGPVDEWGIAGAR (66.2 μg/mL)[143]
Porphyra spp.
Caulerpa lentillifera
Diabetes mellitus/Gly-Gly-Ser-Lys and Glu-Leu-Serin vitro– α-amylase inhibitory activity (IC50): Gly-Gly-Ser-Lys (2.58 mM) and Glu-Leu-Ser (2.62 mM)[144]
Hypertension/ACE inhibitionFDGIP and AIDPVRAin vitro– ACE inhibitory activity (IC50): FDGIP (58.89 μM) and AIDPVRA (65.76 μM)[145]
Porphyra dioicaHypertension/ACE inhibitionThr-Tyr-Ile-Ala and Tyr-Leu-Val-Alain vitro– ACE inhibitory activity (IC50): Thr-Tyr-Ile-Ala (197.5 μM), Tyr-Leu-Val-Ala (259.7 μM), and Asp-Tyr-Tyr-Lys-Arg (628.9 μM)
– DPP-Ⅳ inhibitory activity (IC50): Tyr-Leu-Val-Ala (439.5 μM)
[146]
ACE, angiotensin I-converting enzyme; SBP, streptozotocin; PE, phycoerythrin; DPP-Ⅳ, dipeptidyl peptidase-Ⅳ.
Table 4. Carotenoids and other bioactive substances isolated from seaweeds with therapeutic potential against cardiovascular disease.
Table 4. Carotenoids and other bioactive substances isolated from seaweeds with therapeutic potential against cardiovascular disease.
SourceTarget Disease/MechanismBioactive ComponentModel UsedBiological EffectReference
Undaria PinnatifidaAtherosclerosis/decreased in serum TG levelsFucoxanthinin vivo– Decreased tryglycerides in plasma: 19.4 mg/100 mL
– Enhancement of HDL-cholesterol in plasma: 7.5 mg/100 mL
[153]
Codium fragileObesity/altered in serum lipidSiphonaxanthinin vivo– Decreased serum lipid: total-C (21 mg/dL) and tryglyceride (16 mg/dL)[157]
Phaeodactylum tricornutumDiabetes mellitus/α-amylase and α-glucosidase inhibitionFucoxanthinin vitro– Enzyme inhibitory activity (Ki): α-amylase (0.13 mM) and α-glucosidase (0.05 mM)[154]
Sargassum fusiformeAtherosclerosis/LXRβ agonist activity24(S)-Saringosterolin vitro– LXRβ-mediated transactivation: 3.50-fold vs 1.63-fold compared with control[162]
Diabetes mellitus/α-glucosidase inhibition18α-glycyrrhetinic acid,18β-glycyrrhetinic acid, FAs C20:4 (Δ7,9,11,13), C17:3 (Δ8,11,14), neolignan, and tracein vitro– α-glucosidase inhibitory activity (IC50): 18α-glycyrrhetinic acid (113.30 μM), 18β-glycyrrhetinic acid (128.72 μM), FAs C20:4 (Δ7,9,11,13) (34.85 μM), C17:3 (Δ8,11,14) (43.90 μM), neolignan (133.84 μM), and trace amine (273.23 μM)[163]
Sargassum
Serratifolium
Diabetes mellitus/α-glucosidase inhibitionSargachromenol and sargaquinoic acidin vitro– α-glucosidase inhibitory activity (IC50): sargachromenol (42.41 μM) and sargaquinoic acid (96.17 μM)[164]
TG, triglyceride; LXR, liver X receptor.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cho, C.-H.; Lu, Y.-A.; Kim, M.-Y.; Jeon, Y.-J.; Lee, S.-H. Therapeutic Potential of Seaweed-Derived Bioactive Compounds for Cardiovascular Disease Treatment. Appl. Sci. 2022, 12, 1025. https://doi.org/10.3390/app12031025

AMA Style

Cho C-H, Lu Y-A, Kim M-Y, Jeon Y-J, Lee S-H. Therapeutic Potential of Seaweed-Derived Bioactive Compounds for Cardiovascular Disease Treatment. Applied Sciences. 2022; 12(3):1025. https://doi.org/10.3390/app12031025

Chicago/Turabian Style

Cho, Chi-Heung, Yu-An Lu, Ming-Yeong Kim, You-Jin Jeon, and Sang-Hoon Lee. 2022. "Therapeutic Potential of Seaweed-Derived Bioactive Compounds for Cardiovascular Disease Treatment" Applied Sciences 12, no. 3: 1025. https://doi.org/10.3390/app12031025

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